Jump to content

Antidepressant: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
Reverted 1 good faith edit by Larsbieber99 using STiki
tweak cites
(One intermediate revision by the same user not shown)
Line 4: Line 4:
'''Antidepressants''' are drugs used for the treatment of [[major depressive disorder]] and other conditions, including [[dysthymia]], [[anxiety disorder]]s, [[obsessive compulsive disorder]], [[eating disorder]]s, [[chronic pain]], [[neuropathic pain]] and, in some cases, [[dysmenorrhoea]], snoring, [[migraine]]s, [[attention-deficit hyperactivity disorder]] (ADHD), [[substance abuse]] and [[sleep disorder]]s. They can be used alone or in combination with other medications.
'''Antidepressants''' are drugs used for the treatment of [[major depressive disorder]] and other conditions, including [[dysthymia]], [[anxiety disorder]]s, [[obsessive compulsive disorder]], [[eating disorder]]s, [[chronic pain]], [[neuropathic pain]] and, in some cases, [[dysmenorrhoea]], snoring, [[migraine]]s, [[attention-deficit hyperactivity disorder]] (ADHD), [[substance abuse]] and [[sleep disorder]]s. They can be used alone or in combination with other medications.


The most important classes of antidepressants are the [[selective serotonin reuptake inhibitors]] (SSRIs), [[serotonin–norepinephrine reuptake inhibitors]] (SNRIs), [[tricyclic antidepressants]] (TCAs) and [[monoamine oxidase inhibitors]] (MAOIs). Other drugs used or proposed for the treatment of depression include [[buprenorphine]],<ref>{{cite journal|doi=10.1097/00004714-199502000-00008 |title=Buprenorphine Treatment of Refractory Depression |year=1995 |last1=Bodkin |first1=JA |last2=Zornberg |first2=GL |last3=Lukas |first3=SE |last4=Cole |first4=JO |journal=Journal of Clinical Psychopharmacology |volume=15 |issue=1 |pages=49–57 |pmid=7714228}}</ref> [[tryptophan]],<ref name="pmid9716275">{{cite journal|pmid=9716275 |year=1998 |last1=Ghadirian |first1=AM |last2=Murphy |first2=BE |last3=Gendron |first3=MJ |title=Efficacy of light versus tryptophan therapy in seasonal affective disorder |volume=50 |issue=1 |pages=23–7 |journal=Journal of Affective Disorders |doi=10.1016/S0165-0327(98)00053-6}}</ref> low-dose [[antipsychotics]],<ref>{{cite journal|doi=10.4088/JCP.v64n0512 |title=Conventional Antipsychotic Prescription in Unipolar Depression, I |year=2003 |last1=Vega |first1=JAW |last2=Mortimer |first2=AM |last3=Tyson |first3=PJ |journal=The Journal of Clinical Psychiatry |volume=64 |issue=5 |pages=568–74 |pmid=12755661}}</ref> and [[Hypericum perforatum|St John's wort]].<ref name="Hypericum">{{cite journal|last1=Linde |first1=K |last2=Berner |first2=MM |last3=Kriston |first3=L |title=St John's wort for major depression |journal=Cochrane Database of Systematic Reviews |volume= |issue=4 |pages=CD000448 |year=2008 |pmid=18843608 |doi=10.1002/14651858.CD000448.pub3 |editor1-last=Linde |editor1-first=Klaus}}</ref>
The most important classes of antidepressants are the [[selective serotonin reuptake inhibitors]] (SSRIs), [[serotonin–norepinephrine reuptake inhibitors]] (SNRIs), [[tricyclic antidepressants]] (TCAs) and [[monoamine oxidase inhibitors]] (MAOIs). Other drugs used or proposed for the treatment of depression include [[buprenorphine]],<ref>{{cite journal | author = Bodkin JA, Zornberg GL, Lukas SE, Cole JO | title = Buprenorphine Treatment of Refractory Depression | journal = Journal of Clinical Psychopharmacology | volume = 15 | issue = 1 | pages = 49–57 | year = 1995 | pmid = 7714228 | doi = 10.1097/00004714-199502000-00008 }}</ref> [[tryptophan]],<ref name="pmid9716275">{{cite journal | author = Ghadirian AM, Murphy BE, Gendron MJ | title = Efficacy of light versus tryptophan therapy in seasonal affective disorder | journal = J Affect Disord | volume = 50 | issue = 1 | pages = 23–7 | year = 1998 | pmid = 9716275 | doi = 10.1016/S0165-0327(98)00053-6 }}</ref> low-dose [[antipsychotics]],<ref name="pmid12755661">{{cite journal | author = Wheeler Vega JA, Mortimer AM, Tyson PJ | title = Conventional antipsychotic prescription in unipolar depression, I: an audit and recommendations for practice | journal = J Clin Psychiatry | volume = 64 | issue = 5 | pages = 568–74 | year = 2003 | pmid = 12755661 | doi = 10.4088/JCP.v64n0512 }}</ref> and [[Hypericum perforatum|St John's wort]].<ref name="pmid18843608">{{cite journal | author = Linde K, Berner MM, Kriston L | title = St John's wort for major depression | journal = Cochrane Database Syst Rev | volume = | issue = 4 | pages = CD000448 | year = 2008 | pmid = 18843608 | doi = 10.1002/14651858.CD000448.pub3 }}</ref>


==Efficacy==
==Efficacy==


To establish [[efficacy]], an antidepressant must show that it can produce a therapeutic effect for the condition for which it is taken. An antidepressant should be more efficacious than placebo to justify the risk associated with side effects. For depression, the [[Hamilton Depression Rating Scale]] (HAM-D) is often used to measure the severity of depression.<ref>{{cite journal|doi=10.1136/jnnp.23.1.56 |title=A Rating Scale for Depression |year=1960 |last1=Hamilton |first1=M |journal=Journal of Neurology, Neurosurgery & Psychiatry |volume=23 |issue=1 |pages=56–62 |pmid=14399272 |pmc=495331}}</ref> The maximum score for the 17-item HAM-D questionnaire is 52; the higher the score, the more severe the depression. What constitute a sufficient response to a drug has not been well established, but total remission or virtual elimination of all depression symptoms is the goal, however, remission rates are rarely published. For placebo, the percentage of symptom reduction is approximately 31 to 38%, compared to 46 to 54% for antidepressants.<ref>{{cite journal|doi=10.1371/journal.pone.0041778 |title=A Systematic Review of Comparative Efficacy of Treatments and Controls for Depression |year=2012 |editor1-last=Holscher |editor1-first=Christian |last1=Khan |first1=A |last2=Faucett |first2=J |last3=Lichtenberg |first3=P |last4=Kirsch |first4=I |last5=Brown |first5=WA |journal=PLoS ONE |volume=7 |issue=7 |pages=e41778 |pmid=22860015 |pmc=3408478 |bibcode=2012PLoSO...741778K}}</ref>
To establish [[efficacy]], an antidepressant must show that it can produce a therapeutic effect for the condition for which it is taken. An antidepressant should be more efficacious than placebo to justify the risk associated with side effects. For depression, the [[Hamilton Depression Rating Scale]] (HAM-D) is often used to measure the severity of depression.<ref name="pmid14399272">{{cite journal | author = Hamilton M | title = A rating scale for depression | journal = J. Neurol. Neurosurg. Psychiatr. | volume = 23 | issue = | pages = 56–62 | year = 1960 | pmid = 14399272 | pmc = 495331 | doi = 10.1136/jnnp.23.1.56 }}</ref> The maximum score for the 17-item HAM-D questionnaire is 52; the higher the score, the more severe the depression. What constitute a sufficient response to a drug has not been well established, but total remission or virtual elimination of all depression symptoms is the goal, however, remission rates are rarely published. For placebo, the percentage of symptom reduction is approximately 31 to 38%, compared to 46 to 54% for antidepressants.<ref name="pmid22860015">{{cite journal | author = Khan A, Faucett J, Lichtenberg P, Kirsch I, Brown WA | title = A systematic review of comparative efficacy of treatments and controls for depression | journal = PLoS ONE | volume = 7 | issue = 7 | pages = e41778 | year = 2012 | pmid = 22860015 | pmc = 3408478 | doi = 10.1371/journal.pone.0041778 | bibcode = 2012PLoSO...741778K }}</ref>


On the basis of 234 studies, no clinically relevant superiority of one antidepressant over another was detected for the treatment of acute, continuation, and maintenance phases of depression, taking into account age, sex, ethnicity, or [[comorbidity|comorbid conditions]]. Individual drugs differed in onset of action, adverse events, and some measures of health-related quality of life.<ref>{{cite journal|last1=Gartlehner |first1=G |last2=Hansen |first2=RA |last3=Morgan |first3=LC |last4=Thaler |first4=K |last5=Lux |first5=L |last6=Van Noord |first6=M |last7=Mager |first7=U |last8=Thieda |first8=P |last9=Gaynes |first9=BN |last10=Wilkins |first10=T |last11=Strobelberger |first11=M |last12=Lloyd |first12=S |last13=Reichenpfader |first13=U |last14=Lohr |first14=KN |displayauthors=9 |title=Comparative Benefits and Harms of Second-Generation Antidepressants for Treating Major Depressive Disorder: An Updated Meta-analysis |journal=Annals of Internal Medicine |pmid=22147715 |url=http://www.annals.org/article.aspx?volume=155&page=772 |doi=10.7326/0003-4819-155-11-201112060-00009 |year=2011 |volume=155 |issue=11 |pages=772–85}}</ref>
On the basis of 234 studies, no clinically relevant superiority of one antidepressant over another was detected for the treatment of acute, continuation, and maintenance phases of depression, taking into account age, sex, ethnicity, or [[comorbidity|comorbid conditions]]. Individual drugs differed in onset of action, adverse events, and some measures of health-related quality of life.<ref>{{cite journal | author = Gartlehner G, Hansen RA, Morgan LC, Thaler K, Lux L, Van Noord M, Mager U, Thieda P, Gaynes BN, Wilkins T, Strobelberger M, Lloyd S, Reichenpfader U, Lohr KN | title = Comparative Benefits and Harms of Second-Generation Antidepressants for Treating Major Depressive Disorder: An Updated Meta-analysis | journal = Annals of Internal Medicine | volume = 155 | issue = 11 | pages = 772–85 | year = 2011 | pmid = 22147715 | doi = 10.7326/0003-4819-155-11-201112060-00009 | url = http://www.annals.org/article.aspx?volume=155&page=772 }}</ref>


The largest and most expensive study conducted to date, on the effectiveness of pharmacological treatment for depression, was commissioned by the [[National Institute of Mental Health]].<ref>{{cite web|title=Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Study |url=http://www.nimh.nih.gov/trials/practical/stard/index.shtml |publisher=National Institute of Mental Health |accessdate=28 November 2012|archiveurl = http://www.webcitation.org/6EszRRSYD |archivedate = 2013-03-05|deadurl=yes}}</ref> The study was dubbed "[[STAR*D|The Sequenced Treatment Alternatives to Relieve Depression]]" (STAR*D) Study. The results<ref>{{cite journal|last1=Fava |first1=M |last2=Rush |first2=AJ |last3=Wisniewski |first3=SR |last4=Nierenberg |first4=AA |last5=Alpert |first5=JE |last6=McGrath |first6=PJ |last7=Thase |first7=ME |last8=Warden |first8=D |last9=Biggs |first9=M |last10=Luther |first10=JF |last11=Niederehe |first11=G |last12=Ritz |first12=L |last13=Trivedi |first13=MH |displayauthors=9 |title=A Comparison of Mirtazapine and Nortriptyline Following Two Consecutive Failed Medication Treatments for Depressed Outpatients: A STAR*D Report |journal=The American Journal of Psychiatry |pmid=16816220 |year=2006 |volume=163 |issue=7 |pages=1161–72 |doi=10.1176/appi.ajp.163.7.1161}}</ref><ref name="pmid16554526" /> are summarized here.
The largest and most expensive study conducted to date, on the effectiveness of pharmacological treatment for depression, was commissioned by the [[National Institute of Mental Health]].<ref>{{cite web|title=Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Study |url=http://www.nimh.nih.gov/trials/practical/stard/index.shtml |publisher=National Institute of Mental Health |accessdate=28 November 2012|archiveurl = http://www.webcitation.org/6EszRRSYD |archivedate = 2013-03-05|deadurl=yes}}</ref> The study was dubbed "[[STAR*D|The Sequenced Treatment Alternatives to Relieve Depression]]" (STAR*D) Study. The results<ref>{{cite journal | author = Fava M, Rush AJ, Wisniewski SR, Nierenberg AA, Alpert JE, McGrath PJ, Thase ME, Warden D, Biggs M, Luther JF, Niederehe G, Ritz L, Trivedi MH | title = A Comparison of Mirtazapine and Nortriptyline Following Two Consecutive Failed Medication Treatments for Depressed Outpatients: A STAR*D Report | journal = The American Journal of Psychiatry | volume = 163 | issue = 7 | pages = 1161–72 | year = 2006 | pmid = 16816220 | doi = 10.1176/appi.ajp.163.7.1161 | displayauthors = 9 }}</ref><ref name="pmid16554526" /> are summarized here.
* After the first course of treatment, 27.5% of the 2,876 participants reached remission with a HAM-D score of 7 or less. 21% dropped out.<ref name="pmid16390886">{{cite journal|doi=10.1176/appi.ajp.163.1.28 |title=Evaluation of Outcomes with Citalopram for Depression Using Measurement-Based Care in STAR*D: Implications for Clinical Practice |year=2006 |last1=Trivedi |first1=MH |journal=American Journal of Psychiatry |volume=163 |issue=1 |pages=28–40 |pmid=16390886 |last2=Rush |first2=AJ |last3=Wisniewski |first3=SR |last4=Nierenberg |first4=AA |last5=Warden |first5=D |last6=Ritz |first6=L |last7=Norquist |first7=G |last8=Howland |first8=RH |last9=Lebowitz |first9=B |displayauthors=9 |last10=McGrath |first10=PJ |last11=Shores-Wilson |first11=K |last12=Biggs |first12=MM |last13=Balasubramani |first13=GK |last14=Fava |first14=M |last15=Stard Study Team}}</ref>
* After the first course of treatment, 27.5% of the 2,876 participants reached remission with a HAM-D score of 7 or less. 21% dropped out.<ref name="pmid16390886">{{cite journal | author = Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, Norquist G, Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK, Fava M | title = Evaluation of Outcomes with Citalopram for Depression Using Measurement-Based Care in STAR*D: Implications for Clinical Practice | journal = American Journal of Psychiatry | volume = 163 | issue = 1 | pages = 28–40 | year = 2006 | pmid = 16390886 | doi = 10.1176/appi.ajp.163.1.28 | displayauthors = 9 }}</ref>
* After the second course of treatment, 21 to 30% of the remaining 1,439 participants remitted. Only 310 participants were willing or available to continue the study.<ref name="pmid16554526">{{cite journal|doi=10.1056/NEJMoa052964 |title=Medication Augmentation after the Failure of SSRIs for Depression |year=2006 |last1=Trivedi |first1=MH |last2=Fava |first2=M |last3=Wisniewski |first3=SR |last4=Thase |first4=ME |last5=Quitkin |first5=F |last6=Warden |first6=D |last7=Ritz |first7=L |last8=Nierenberg |first8=AA |last9=Lebowitz |first9=BD |last10=Biggs |first10=MM |last11=Luther |first11=JF |last12=Shores-Wilson |first12=K |last13=Rush |first13=AJ |last14=Stard Study Team |displayauthors=9 |journal=New England Journal of Medicine |volume=354 |issue=12 |pages=1243–52 |pmid=16554526}}</ref> Switching medications can achieve remission in about 25% of patients.<ref name="pmid16554525">{{cite journal|doi=10.1056/NEJMoa052963 |title=Bupropion-SR, Sertraline, or Venlafaxine-XR after Failure of SSRIs for Depression |year=2006 |last1=Rush |first1=AJ |last2=Trivedi |first2=MH |last3=Wisniewski |first3=SR |last4=Stewart |first4=JW |last5=Nierenberg |first5=AA |last6=Thase |first6=ME |last7=Ritz |first7=L |last8=Biggs |first8=MM |last9=Warden |first9=D |last10=Luther |first10=JF |last11=Shores-Wilson |first11=K |last12=Niederehe |first12=G |last13=Fava |first13=M |last14=Stard Study Team |displayauthors=9 |journal=New England Journal of Medicine |volume=354 |issue=12 |pages=1231–42 |pmid=16554525}}</ref>
* After the second course of treatment, 21 to 30% of the remaining 1,439 participants remitted. Only 310 participants were willing or available to continue the study.<ref name="pmid16554526">{{cite journal | author = Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, Ritz L, Nierenberg AA, Lebowitz BD, Biggs MM, Luther JF, Shores-Wilson K, Rush AJ | title = Medication Augmentation after the Failure of SSRIs for Depression | journal = New England Journal of Medicine | volume = 354 | issue = 12 | pages = 1243–52 | year = 2006 | pmid = 16554526 | doi = 10.1056/NEJMoa052964 | displayauthors = 9 }}</ref> Switching medications can achieve remission in about 25% of patients.<ref name="pmid16554525">{{cite journal | author = Rush AJ, Trivedi MH, Wisniewski SR, Stewart JW, Nierenberg AA, Thase ME, Ritz L, Biggs MM, Warden D, Luther JF, Shores-Wilson K, Niederehe G, Fava M | title = Bupropion-SR, Sertraline, or Venlafaxine-XR after Failure of SSRIs for Depression | journal = New England Journal of Medicine | volume = 354 | issue = 12 | pages = 1231–42 | year = 2006 | pmid = 16554525 | doi = 10.1056/NEJMoa052963 | displayauthors = 9 }}</ref>
* After the third course of treatment, 17.8% of the remaining 310 participants remitted.
* After the third course of treatment, 17.8% of the remaining 310 participants remitted.
* After the fourth and last course of treatment, 10.1% of the remaining 109 participants remitted.
* After the fourth and last course of treatment, 10.1% of the remaining 109 participants remitted.
* After a one year follow-up, of the 1085 remitted participants, 93% participants had either relapsed or dropped out of the study.
* After a one year follow-up, of the 1085 remitted participants, 93% participants had either relapsed or dropped out of the study.


There were no statistical or meaningful clinical differences in remission rates, response rates, or times to remission or response among any of the medications compared in this study.<ref>{{cite journal|pmid=18221624 |year=2007 |last1=Warden |first1=D |last2=Rush |first2=AJ |last3=Trivedi |first3=MH |last4=Fava |first4=M |last5=Wisniewski |first5=SR |title=The STAR*D Project results: A comprehensive review of findings |volume=9 |issue=6 |pages=449–59 |journal=Current psychiatry reports |doi=10.1007/s11920-007-0061-3}}</ref> These included bupropion sustained release, bupropion, citalopram, lithium, mirtazapine, nortriptyline, sertraline, triiodothyronine, tranylcypromine, and venlafaxine extended release.{{medical citation needed|date=March 2013}}
There were no statistical or meaningful clinical differences in remission rates, response rates, or times to remission or response among any of the medications compared in this study.<ref>{{cite journal | author = Warden D, Rush AJ, Trivedi MH, Fava M, Wisniewski SR | title = The STAR*D Project results: A comprehensive review of findings | journal = Current psychiatry reports | volume = 9 | issue = 6 | pages = 449–59 | year = 2007 | pmid = 18221624 | doi = 10.1007/s11920-007-0061-3 }}</ref> These included bupropion sustained release, bupropion, citalopram, lithium, mirtazapine, nortriptyline, sertraline, triiodothyronine, tranylcypromine, and venlafaxine extended release.{{medical citation needed|date=March 2013}}


A 2008 review of [[randomized controlled trial]]s concluded that symptomatic improvement with SSRIs was greatest by the end of the first week of use, but that some improvement continued for at least 6 weeks.<ref>{{cite journal|doi=10.1001/archpsyc.63.11.1217 |title=Early Onset of Selective Serotonin Reuptake Inhibitor Antidepressant Action: Systematic Review and Meta-analysis |year=2006 |last1=Taylor |first1=MJ |last2=Freemantle |first2=N |last3=Geddes |first3=JR |last4=Bhagwagar |first4=Z |journal=Archives of General Psychiatry |volume=63 |issue=11 |pages=1217–23 |pmid=17088502 |pmc=2211759}}</ref>
A 2008 review of [[randomized controlled trial]]s concluded that symptomatic improvement with SSRIs was greatest by the end of the first week of use, but that some improvement continued for at least 6 weeks.<ref>{{cite journal | author = Taylor MJ, Freemantle N, Geddes JR, Bhagwagar Z | title = Early Onset of Selective Serotonin Reuptake Inhibitor Antidepressant Action: Systematic Review and Meta-analysis | journal = Archives of General Psychiatry | volume = 63 | issue = 11 | pages = 1217–23 | year = 2006 | pmid = 17088502 | pmc = 2211759 | doi = 10.1001/archpsyc.63.11.1217 }}</ref>


===Clinical guidelines===
===Clinical guidelines===
Line 29: Line 29:


===Limitations and strategies===
===Limitations and strategies===
Between 30% and 50% of individuals treated with a given antidepressant do not show a response.<ref>{{cite journal|doi=10.2174/138161206775474422 |title=Recent Progress in Pharmacological and Non-Pharmacological Treatment Options of Major Depression |year=2006 |last1=Baghai |first1=Thomas |last2=Möller |first2=Hans-Jurgen |last3=Rupprecht |first3=Rainer |journal=Current Pharmaceutical Design |volume=12 |issue=4 |pages=503–15 |pmid=16472142}}</ref><ref name="SSRIswitch">{{cite journal|doi=10.4088/JCP.v67n1203 |title=Switching Antidepressants After a First Selective Serotonin Reuptake Inhibitor in Major Depressive Disorder |year=2006 |last1=Ruhé |first1=HG |last2=Huyser |first2=J |last3=Swinkels |first3=JA |last4=Schene |first4=AH |journal=The Journal of Clinical Psychiatry |volume=67 |issue=12 |pages=1836–55 |pmid=17194261}}</ref> In clinical studies, approximately one-third of patients achieve a full [[remission (medicine)|remission]], one-third experience a response and one-third are nonresponders. Partial remission is characterized by the presence of poorly defined residual symptoms. These symptoms typically include depressed mood, psychic anxiety, sleep disturbance, fatigue and diminished interest or pleasure. It is currently unclear which factors predict partial remission. However, it is clear that residual symptoms are powerful predictors of relapse, with relapse rates 3–6 times higher in patients with residual symptoms than in those who experience full remission.<ref>{{cite journal|pmid=12174733 |year=2002 |last1=Tranter |first1=R |last2=O'Donovan |first2=C |last3=Chandarana |first3=P |last4=Kennedy |first4=S |title=Prevalence and outcome of partial remission in depression |volume=27 |issue=4 |pages=241–7 |pmc=161658 |journal=Journal of psychiatry & neuroscience}}</ref> In addition, antidepressant drugs tend to lose efficacy over the course of treatment.<ref>{{cite journal|doi=10.4088/JCP.v59n0602 |title=Loss of Antidepressant Efficacy During Maintenance Therapy |year=1998 |last1=Byrne |first1=SE |last2=Rothschild |first2=AJ |journal=The Journal of Clinical Psychiatry |volume=59 |issue=6 |pages=279–88 |pmid=9671339}}</ref> A number of strategies are used in clinical practice to try to overcome these limits and variations.<ref>{{cite journal|pmid=10900529 |year=2000 |last1=Mischoulon |first1=D |last2=Nierenberg |first2=AA |last3=Kizilbash |first3=L |last4=Rosenbaum |first4=JF |last5=Fava |first5=M |title=Strategies for managing depression refractory to selective serotonin reuptake inhibitor treatment: A survey of clinicians |volume=45 |issue=5 |pages=476–81 |journal=Canadian journal of psychiatry}}</ref> They include switching medication, augmentation, and combination.
Between 30% and 50% of individuals treated with a given antidepressant do not show a response.<ref>{{cite journal | author = Baghai TC, Möller HJ, Rupprecht R | title = Recent Progress in Pharmacological and Non-Pharmacological Treatment Options of Major Depression | journal = Current Pharmaceutical Design | volume = 12 | issue = 4 | pages = 503–15 | year = 2006 | pmid = 16472142 | doi = 10.2174/138161206775474422 }}</ref><ref name="SSRIswitch">{{cite journal | author = Ruhé HG, Huyser J, Swinkels JA, Schene AH | title = Switching Antidepressants After a First Selective Serotonin Reuptake Inhibitor in Major Depressive Disorder | journal = The Journal of Clinical Psychiatry | volume = 67 | issue = 12 | pages = 1836–55 | year = 2006 | pmid = 17194261 | doi = 10.4088/JCP.v67n1203 }}</ref> In clinical studies, approximately one-third of patients achieve a full [[remission (medicine)|remission]], one-third experience a response and one-third are nonresponders. Partial remission is characterized by the presence of poorly defined residual symptoms. These symptoms typically include depressed mood, psychic anxiety, sleep disturbance, fatigue and diminished interest or pleasure. It is currently unclear which factors predict partial remission. However, it is clear that residual symptoms are powerful predictors of relapse, with relapse rates 3–6 times higher in patients with residual symptoms than in those who experience full remission.<ref>{{cite journal | author = Tranter R, O'Donovan C, Chandarana P, Kennedy S | title = Prevalence and outcome of partial remission in depression | journal = Journal of psychiatry & neuroscience | volume = 27 | issue = 4 | pages = 241–7 | year = 2002 | pmid = 12174733 | pmc = 161658 }}</ref> In addition, antidepressant drugs tend to lose efficacy over the course of treatment.<ref>{{cite journal | author = Byrne SE, Rothschild AJ | title = Loss of Antidepressant Efficacy During Maintenance Therapy | journal = The Journal of Clinical Psychiatry | volume = 59 | issue = 6 | pages = 279–88 | year = 1998 | pmid = 9671339 | doi = 10.4088/JCP.v59n0602 }}</ref> A number of strategies are used in clinical practice to try to overcome these limits and variations.<ref>{{cite journal | author = Mischoulon D, Nierenberg AA, Kizilbash L, Rosenbaum JF, Fava M | title = Strategies for managing depression refractory to selective serotonin reuptake inhibitor treatment: A survey of clinicians | journal = Canadian journal of psychiatry | volume = 45 | issue = 5 | pages = 476–81 | year = 2000 | pmid = 10900529 }}</ref> They include switching medication, augmentation, and combination.


===="Trial and error" switching====
===="Trial and error" switching====
The [[American Psychiatric Association]] 2000 Practice Guideline advises that where no response is achieved following six to eight weeks of treatment with an antidepressant, to switch to an antidepressant in the same class, then to a different class of antidepressant. The remission rate reported by the STAR*D study was 21% using this method.{{medical citation needed|date=March 2013}}
The [[American Psychiatric Association]] 2000 Practice Guideline advises that where no response is achieved following six to eight weeks of treatment with an antidepressant, to switch to an antidepressant in the same class, then to a different class of antidepressant. The remission rate reported by the STAR*D study was 21% using this method.{{medical citation needed|date=March 2013}}


A 2006 meta-analysis review found wide variation in the findings of prior studies; for patients who had failed to respond to an SSRI antidepressant, between 12% and 86% showed a response to a new drug. However, the more antidepressants an individual had already tried, the less likely they were to benefit from a new antidepressant trial.<ref name="SSRIswitch"/> However, a later meta-analysis found no difference between switching to a new drug and staying on the old medication; although 34% of treatment resistant patients responded when switched to the new drug, 40% responded without being switched.<ref>{{cite journal|doi=10.1111/j.1600-0447.2009.01458.x |title=No evidence for switching the antidepressant: Systematic review and meta-analysis of RCTs of a common therapeutic strategy |year=2010 |last1=Bschor |first1=T |last2=Baethge |first2=C |journal=Acta Psychiatrica Scandinavica |volume=121 |issue=3 |pages=174–9 |pmid=19703121}}</ref> Thus, the clinical response to the new drug might be a [[placebo effect]] associated with the belief that one is receiving a different medication.{{medical citation needed|date=March 2013}}
A 2006 meta-analysis review found wide variation in the findings of prior studies; for patients who had failed to respond to an SSRI antidepressant, between 12% and 86% showed a response to a new drug. However, the more antidepressants an individual had already tried, the less likely they were to benefit from a new antidepressant trial.<ref name="SSRIswitch"/> However, a later meta-analysis found no difference between switching to a new drug and staying on the old medication; although 34% of treatment resistant patients responded when switched to the new drug, 40% responded without being switched.<ref>{{cite journal | author = Bschor T, Baethge C | title = No evidence for switching the antidepressant: Systematic review and meta-analysis of RCTs of a common therapeutic strategy | journal = Acta Psychiatrica Scandinavica | volume = 121 | issue = 3 | pages = 174–9 | year = 2010 | pmid = 19703121 | doi = 10.1111/j.1600-0447.2009.01458.x }}</ref> Thus, the clinical response to the new drug might be a [[placebo effect]] associated with the belief that one is receiving a different medication.{{medical citation needed|date=March 2013}}


====Augmentation and combination====
====Augmentation and combination====
For a partial response, the American Psychiatric Association guidelines suggest augmentation, or adding a drug from a different class. These include: [[Lithium (medication)|lithium]] and [[thyroid]] augmentation, [[dopamine agonists]], [[sex steroids]], [[Norepinephrine reuptake inhibitor|NRIs]], [[glucocorticoid]]-specific agents, or the newer [[anticonvulsant]]s.<ref name="augment">{{cite journal|doi=10.1007/s11920-005-0064-x |title=Update on augmentation of antidepressant response in resistant depression |year=2005 |last1=Debattista |first1=C |last2=Lembke |first2=A |journal=Current Psychiatry Reports |volume=7 |issue=6 |pages=435–40 |pmid=16318821}}</ref>
For a partial response, the American Psychiatric Association guidelines suggest augmentation, or adding a drug from a different class. These include: [[Lithium (medication)|lithium]] and [[thyroid]] augmentation, [[dopamine agonists]], [[sex steroids]], [[Norepinephrine reuptake inhibitor|NRIs]], [[glucocorticoid]]-specific agents, or the newer [[anticonvulsant]]s.<ref name="augment">{{cite journal | author = DeBattista C, Lembke A | title = Update on augmentation of antidepressant response in resistant depression | journal = Current Psychiatry Reports | volume = 7 | issue = 6 | pages = 435–40 | year = 2005 | pmid = 16318821 | doi = 10.1007/s11920-005-0064-x }}</ref>


A combination strategy involves adding an additional antidepressant, usually from a different class so as to have effect on other mechanisms. Although this may be used in clinical practice, there is little evidence for the relative efficacy or adverse effects of this strategy.<ref>{{cite journal|doi=10.4088/JCP.v63n0805 |title=Combining Antidepressants for Treatment-Resistant Depression |year=2002 |last1=Lam |first1=RW |last2=Wan |first2=DDC |last3=Cohen |first3=NL |last4=Kennedy |first4=SH |journal=The Journal of Clinical Psychiatry |volume=63 |issue=8 |pages=685–93 |pmid=12197448}}</ref>
A combination strategy involves adding an additional antidepressant, usually from a different class so as to have effect on other mechanisms. Although this may be used in clinical practice, there is little evidence for the relative efficacy or adverse effects of this strategy.<ref>{{cite journal | author = Lam RW, Wan DD, Cohen NL, Kennedy SH | title = Combining Antidepressants for Treatment-Resistant Depression | journal = The Journal of Clinical Psychiatry | volume = 63 | issue = 8 | pages = 685–93 | year = 2002 | pmid = 12197448 | doi = 10.4088/JCP.v63n0805 }}</ref>


Opponents of switching, augmentation and combination argue that treatment may also propel the illness to a malignant and treatment-unresponsive course with [[wikt:iatrogenic|iatrogenic]] psychiatric-like symptoms and treatment resistance or episode acceleration.<ref>{{cite journal|doi=10.1159/000112883 |title=Atypical Antipsychotics: CATIE Study, Drug-Induced Movement Disorder and Resulting Iatrogenic Psychiatric-Like Symptoms, Supersensitivity Rebound Psychosis and Withdrawal Discontinuation Syndromes |year=2008 |last1=Chouinard |first1=G |last2=Chouinard |first2=Virginie-Anne |journal=Psychotherapy and Psychosomatics |volume=77 |issue=2 |pages=69–77 |pmid=18230939}}</ref>
Opponents of switching, augmentation and combination argue that treatment may also propel the illness to a malignant and treatment-unresponsive course with [[wikt:iatrogenic|iatrogenic]] psychiatric-like symptoms and treatment resistance or episode acceleration.<ref>{{cite journal | author = Chouinard G, Chouinard VA | title = Atypical Antipsychotics: CATIE Study, Drug-Induced Movement Disorder and Resulting Iatrogenic Psychiatric-Like Symptoms, Supersensitivity Rebound Psychosis and Withdrawal Discontinuation Syndromes | journal = Psychotherapy and Psychosomatics | volume = 77 | issue = 2 | pages = 69–77 | year = 2008 | pmid = 18230939 | doi = 10.1159/000112883 }}</ref>


====Long-term use====
====Long-term use====
Line 104: Line 104:
|-
|-


| <ref name="GG">{{cite isbn|9780071624428}}</ref><ref name="UpToDate">{{cite web|title=Side effects of antidepressant medications |work=UpToDate |publisher=Wolters Kluwer Health |accessdate=24 October 2013 |url=http://www.uptodate.com/contents/image?imageKey=PC%2F62488&source=image_view&view=print&topicKey=PSYCH/85816&source=see_link&elapsedTimeMs=2|archiveurl=http://web.archive.org/web/20131102183135/http://www.uptodate.com/contents/image?imageKey=PC%2F62488&source=image_view&view=print&topicKey=PSYCH/85816&source=see_link&elapsedTimeMs=2 |archivedate=2013-11-02 }}</ref><ref name="Martindale">{{cite book|title=MARTINDALE – The Complete Drug Reference |publisher=Pharmaceutical Press |author=Royal Pharmaceutical Society of Great Britain |accessdate=31 October 2013 |url=https://www.medicinescomplete.com/mc/martindale/current/login.htm?uri=http%3A%2F%2Fwww.medicinescomplete.com%2Fmc%2Fmartindale%2Fcurrent%2F|archiveurl=http://www.webcitation.org/6IOEuW8Pn |archivedate=2013-07-26 }}</ref>
| <ref name="GG">{{cite book | author = Brunton, L; Chabner, B; Knollman, B | title = Goodman and Gilman's The Pharmacological Basis of Therapeutics | language = English | year = 2010 | publisher = McGraw-Hill Professional | isbn = 978-0-07-162442-8 | edition = 12th | location = New York }}</ref><ref name="UpToDate">{{cite web|title=Side effects of antidepressant medications |work=UpToDate |publisher=Wolters Kluwer Health |accessdate=24 October 2013 |url=http://www.uptodate.com/contents/image?imageKey=PC%2F62488&source=image_view&view=print&topicKey=PSYCH/85816&source=see_link&elapsedTimeMs=2|archiveurl=http://web.archive.org/web/20131102183135/http://www.uptodate.com/contents/image?imageKey=PC%2F62488&source=image_view&view=print&topicKey=PSYCH/85816&source=see_link&elapsedTimeMs=2 |archivedate=2013-11-02 }}</ref><ref name="Martindale">{{cite book|title=MARTINDALE – The Complete Drug Reference |publisher=Pharmaceutical Press |author=Royal Pharmaceutical Society of Great Britain |accessdate=31 October 2013 |url=https://www.medicinescomplete.com/mc/martindale/current/login.htm?uri=http%3A%2F%2Fwww.medicinescomplete.com%2Fmc%2Fmartindale%2Fcurrent%2F|archiveurl=http://www.webcitation.org/6IOEuW8Pn |archivedate=2013-07-26 }}</ref>
|| <ref name = Martindale/><ref name="Lancet">{{cite journal | author = Cipriani A, Furukawa TA, Salanti G, Geddes JR, Higgins JP, Churchill R, Watanabe N, Nakagawa A, Omori IM, McGuire H, Tansella M, Barbui C | title = Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis | journal = Lancet | volume = 373 | issue = 9665 | pages = 746–58 | year = 2009 | pmid = 19185342 | doi = 10.1016/S0140-6736(09)60046-5 }}</ref>
|| <ref name = Martindale/><ref name="Lancet">{{cite pmid|19185342}}</ref>
|| <ref name = GG/><ref name = UpToDate/><ref name = "isbn 9780470979488" /><br><ref name = Martindale/><ref name =Lancet/>
|| <ref name = GG/><ref name = UpToDate/><ref name = "isbn 9780470979488" /><br><ref name = Martindale/><ref name =Lancet/>
|| <ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="Overdoses">{{cite pmid|19031375}}</ref>
|| <ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="Overdoses">{{cite journal | author = White N, Litovitz T, Clancy C | title = Suicidal antidepressant overdoses: a comparative analysis by antidepressant type | journal = J Med Toxicol | volume = 4 | issue = 4 | pages = 238–50 | year = 2008 | pmid = 19031375 | pmc = 3550116 | doi = 10.1007/BF03161207 }}</ref>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488">{{cite isbn|9780470979488}}</ref>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488">{{cite book | last1 = Taylor | first1 = D | last2 = | first2 = | last3 = | first3 = | title = The Maudsley prescribing guidelines in psychiatry | language = English | year = 2012 | url = | isbn = 978-0-470-97948-8 | author2 = Paton, C; Shitij, K | editor1-last = | editor1-first = | oclc = | editor2-link = | location = West Sussex | publisher = Wiley-Blackwell }}</ref>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="AMH">{{cite isbn|978-0-9805790-9-3}}</ref>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="AMH">{{cite book | title = Australian Medicines Handbook | year = 2013 | publisher = The Australian Medicines Handbook Unit Trust | isbn = 978-0-9805790-9-3 | edition = 2013 | place = Adelaide | editor = Rossi, S }}</ref>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="AMH"/>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" /><ref name="AMH"/>
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" />
|| <ref name = UpToDate/><ref name = Martindale/><ref name = "isbn 9780470979488" />
Line 853: Line 853:
[[Imipramine]]:
[[Imipramine]]:
First marketed [[Tricyclic antidepressant|TCA]]. Somewhat selective for serotonin reuptake (26x over norepinephrine).<br>
First marketed [[Tricyclic antidepressant|TCA]]. Somewhat selective for serotonin reuptake (26x over norepinephrine).<br>
Relative efficacy: 3 <ref name="pmid8912401">{{cite journal|last1=Bruijn |first1=JA |last2=Moleman |first2=P |last3=Mulder |first3=PG |last4=van den Broek |first4=WW |last5=van Hulst |first5=AM |last6=van der Mast |first6=RC |last7=van de Wetering |first7=BJ |title=A double-blind, fixed blood-level study comparing mirtazapine with imipramine in depressed in-patients |journal=Psychopharmacology (Berl.) |volume=127 |issue=3 |pages=231?7 |date=October 1996 |pmid=8912401 |doi=10.1007/BF02246131}}</ref><ref name="pmid10463374">{{cite journal|last1=Bruijn |first1=JA |last2=Moleman |first2=P |last3=Mulder |first3=PG |last4=van den Broek |first4=WW |title=Depressed in-patients respond differently to imipramine and mirtazapine |journal=Pharmacopsychiatry |volume=32 |issue=3 |pages=87?92 |date=May 1999 |pmid=10463374 |doi=10.1055/s-2007-979200}}</ref>
Relative efficacy: 3<ref name="pmid8912401">{{cite journal | author = Bruijn JA, Moleman P, Mulder PG, van den Broek WW, van Hulst AM, van der Mast RC, van de Wetering BJ | title = A double-blind, fixed blood-level study comparing mirtazapine with imipramine in depressed in-patients | journal = Psychopharmacology (Berl.) | volume = 127 | issue = 3 | pages = 231–7 | year = 1996 | month = October | pmid = 8912401 | doi = 10.1007/BF02246131 }}</ref><ref name="pmid10463374">{{cite journal | author = Bruijn JA, Moleman P, Mulder PG, van den Broek WW | title = Depressed in-patients respond differently to imipramine and mirtazapine | journal = Pharmacopsychiatry | volume = 32 | issue = 3 | pages = 87–92 | year = 1999 | pmid = 10463374 | doi = 10.1055/s-2007-979200 }}</ref>


[[Lofepramine]]:
[[Lofepramine]]:
Line 905: Line 905:


[[Fluvoxamine]]:
[[Fluvoxamine]]:
Not [[Food and Drug Administration|FDA]] approved for major depression; FDA approved for [[Obsessive-compulsive disorder|OCD]]. Has the highest affinity of any SSRI towards the [[sigma-1 receptor]] at which it serves as an agonist.<ref name="sig">{{cite journal|title=Sigma receptors: Potential targets for a new class of antidepressant drug |journal=Pharmacology & Therapeutics |issue=3 |pages=271?282 |date=September 2010 |doi=10.1016/j.pharmthera.2010.04.003 |pmid=20438757 |last1=Fishback |first1=JA |last2=Robson |first2=MJ |last3=Xu |first3=YT |last4=Matsumoto |first4=RR |volume=127}}</ref><ref name =psychdep/>
Not [[Food and Drug Administration|FDA]] approved for major depression; FDA approved for [[Obsessive-compulsive disorder|OCD]]. Has the highest affinity of any SSRI towards the [[sigma-1 receptor]] at which it serves as an agonist.<ref name="sig">{{cite journal | author = Fishback JA, Robson MJ, Xu YT, Matsumoto RR | title = Sigma receptors: potential targets for a new class of antidepressant drug | journal = Pharmacol. Ther. | volume = 127 | issue = 3 | pages = 271–82 | year = 2010 | pmid = 20438757 | pmc = 3993947 | doi = 10.1016/j.pharmthera.2010.04.003 }}</ref><ref name =psychdep/>


[[Paroxetine]]:
[[Paroxetine]]:
Only SSRI that's not Australian pregnancy category C but is rather category D due to an increased risk of [[Persistent fetal circulation|Persistent Pulmonary Hypertension of the Newborn]]. The [[Food and Drug Administration|FDA]] of the US has placed it in category D. It is associated with a higher risk of sexual dysfunction, weight gain, anticholinergic side effects and drowsiness than the other SSRIs. Has a short half life compared to other SSRIs and hence is the most prone to causing withdrawal effects whenever a dose is missed. Paroxetine has the lowest affinity for the sigma-1 receptors of all the SSRIs.<ref name = sig/> It also possesses the highest propensity of any SSRI for causing [[extrapyramidal symptoms]].<ref name="CPT">{{cite isbn|9780443102851}}</ref>
Only SSRI that's not Australian pregnancy category C but is rather category D due to an increased risk of [[Persistent fetal circulation|Persistent Pulmonary Hypertension of the Newborn]]. The [[Food and Drug Administration|FDA]] of the US has placed it in category D. It is associated with a higher risk of sexual dysfunction, weight gain, anticholinergic side effects and drowsiness than the other SSRIs. Has a short half life compared to other SSRIs and hence is the most prone to causing withdrawal effects whenever a dose is missed. Paroxetine has the lowest affinity for the sigma-1 receptors of all the SSRIs.<ref name = sig/> It also possesses the highest propensity of any SSRI for causing [[extrapyramidal symptoms]].<ref name="CPT">{{cite book | title = Clinical Pharmacy and Therapeutics | year = 2007 | publisher = Churchill Livingstone Elsevier | isbn = 978-0-7020-4293-5 | edition = 4th | location = Edinburgh | origyear = 1994 | editor = Walker, R; Whittlesea, C }}</ref>


[[Sertraline]]:
[[Sertraline]]:
Highest risk of psychiatric side effects (e.g. mania, suicidal behavior/ideation, psychosis, etc.)<ref name =Martindale/> Has slight (but clinically significant) inhibitory effects on dopamine reuptake.<ref>{{cite journal|title=[The effect of sertraline on cognitive functions in patients with obsessive-compulsive disorder] |journal=Psychiatria Polska |date=November–December 2002 |volume=36 |issue=6 suppl |pages=289?295 |last1=Borkowska |first1=A |last2=Pilaczy?ska |first2=E |last3=Araszkiewicz |first3=A |last4=Rybakowski |first4=J |language=Polish |pmid=12647451}}</ref><ref>{{cite journal|title=Additional dopamine reuptake inhibition attenuates vigilance impairment induced by serotonin reuptake inhibition in man |journal=Journal of Psychopharmacology |volume=16 |issue=3 |pages=207?214 |pmid=12236626 |doi=10.1177/026988110201600303 |date=September 2002 |last1=Schmitt |first1=JAJ |last2=Ramaekers |first2=JG |last3=Kruizinga |first3=MJ |last4=Van Boxtel |first4=MPJ |last5=Vuurman |first5=EFPM |last6=Riedel |first6=WJ}}</ref> Has the second highest affinity of the SSRIs towards the sigma-1 receptor where it may serve as a sigma-1 receptor antagonist.<ref name="psychdep">{{cite journal|title=The opposite effects of fluvoxamine and sertraline in the treatment of psychotic major depression: a case report |journal=Annals of General Psychiatry |date=May 2010 |volume=9 |issue=1 |pages=23?25 |doi=10.1186/1744-859X-9-23 |pmid=20492642 |pmc=2881105 |last1=Kishimoto |first1=Akira |last2=Todani |first2=Ayako |last3=Miura |first3=Junko |last4=Kitagaki |first4=Tetsuno |last5=Hashimoto |first5=Kenji}}</ref><br>
Highest risk of psychiatric side effects (e.g. mania, suicidal behavior/ideation, psychosis, etc.)<ref name =Martindale/> Has slight (but clinically significant) inhibitory effects on dopamine reuptake.<ref>{{cite journal | author = Borkowska A, Pilaczyńska E, Araszkiewicz A, Rybakowski J | title = [The effect of sertraline on cognitive functions in patients with obsessive-compulsive disorder] | language = Polish | journal = Psychiatr. Pol. | volume = 36 | issue = 6 Suppl | pages = 289–95 | year = 2002 | pmid = 12647451 | doi = }}</ref><ref>{{cite journal | author = Schmitt JA, Ramaekers JG, Kruizinga MJ, van Boxtel MP, Vuurman EF, Riedel WJ | title = Additional dopamine reuptake inhibition attenuates vigilance impairment induced by serotonin reuptake inhibition in man | journal = J. Psychopharmacol. (Oxford) | volume = 16 | issue = 3 | pages = 207–14 | year = 2002 | pmid = 12236626 | doi = 10.1177/026988110201600303 }}</ref> Has the second highest affinity of the SSRIs towards the sigma-1 receptor where it may serve as a sigma-1 receptor antagonist.<ref name="psychdep">{{cite journal | author = Kishimoto A, Todani A, Miura J, Kitagaki T, Hashimoto K | title = The opposite effects of fluvoxamine and sertraline in the treatment of psychotic major depression: a case report | journal = Ann Gen Psychiatry | volume = 9 | issue = | pages = 23 | year = 2010 | pmid = 20492642 | pmc = 2881105 | doi = 10.1186/1744-859X-9-23 }}</ref><br>
GI toxicity: 2 (mostly<br>diarrhoea)<ref name = "isbn 9780470979488" />
GI toxicity: 2 (mostly<br>diarrhoea)<ref name = "isbn 9780470979488" />


Line 933: Line 933:


[[Mianserin]]:
[[Mianserin]]:
Not licensed for use in the US and Canada. Licensed for use in Australia and the UK. Can cause blood dyscrasias (including [[agranulocytosis]]) and consequently both the [[British National Formulary|BNF]] and [[Australian Medicines Handbook|AMH]] recommend regular complete blood count monitoring.<ref name="AMH" /><ref name="BNF">{{cite isbn|9780857110848}}</ref>
Not licensed for use in the US and Canada. Licensed for use in Australia and the UK. Can cause blood dyscrasias (including [[agranulocytosis]]) and consequently both the [[British National Formulary|BNF]] and [[Australian Medicines Handbook|AMH]] recommend regular complete blood count monitoring.<ref name="AMH" /><ref name="BNF">{{cite book | last1 = Joint Formulary Committee | title = British National Formulary (BNF) | year = 2013 | isbn = 978-0-85711-084-8 | edition = 65 | location = London, UK | publisher = Pharmaceutical Press }}</ref>


[[Mirtazapine]]:
[[Mirtazapine]]:
Line 960: Line 960:
[[Agomelatine]]:
[[Agomelatine]]:
Not licensed in the US or Canada. Licensed in Australia and the UK.<br>
Not licensed in the US or Canada. Licensed in Australia and the UK.<br>
Relative efficacy: 2 <ref>{{cite journal|title=Clinical Studies on the Efficacy of Agomelatine on Depressive Symptoms |journal=CNS Drugs |volume=23 |issue=Suppl. 2 |pages=35?39 |year=2009 |doi=10.2165/11318650-000000000-00000 |last=Goodwin |first=GM |pmid=19708724 |url=http://link.springer.com/article/10.2165%2F11318650-000000000-00000 |subscription=yes}}</ref>
Relative efficacy: 2 <ref name="pmid19708724">{{cite journal | author = Goodwin GM | title = Clinical studies on the efficacy of agomelatine on depressive symptoms | journal = CNS Drugs | volume = 23 Suppl 2 | issue = | pages = 35–9 | year = 2009 | pmid = 19708724 | doi = 10.2165/11318650-000000000-00000 }}</ref>


[[Bupropion]]:
[[Bupropion]]:
Line 976: Line 976:
===General===
===General===
{{Main|Serotonin syndrome|MAOIs}}
{{Main|Serotonin syndrome|MAOIs}}
For bipolar depression, antidepressants (most frequently SSRIs) can exacerbate or trigger symptoms of [[hypomania]] and [[mania]].<ref>{{cite journal|doi=10.1016/S0165-0327(97)00082-7 |title=Antidepressant-associated hypomania in outpatient depression: A 203-case study in private practice |year=1997 |last1=Benazzi |first1=F |journal=Journal of Affective Disorders |volume=46 |pages=73–7 |pmid=9387089 |issue=1}}</ref>
For bipolar depression, antidepressants (most frequently SSRIs) can exacerbate or trigger symptoms of [[hypomania]] and [[mania]].<ref name="pmid9387089">{{cite journal | author = Benazzi F | title = Antidepressant-associated hypomania in outpatient depression: a 203-case study in private practice | journal = J Affect Disord | volume = 46 | issue = 1 | pages = 73–7 | year = 1997 | pmid = 9387089 | doi = 10.1016/S0165-0327(97)00082-7 }}</ref>


Almost any medication involved with serotonin regulation has the potential to cause [[serotonin syndrome|serotonin toxicity]] (also known as ''serotonin syndrome''){{spaced ndash}} an excess of serotonin that can induce mania, restlessness, agitation, [[emotional lability]], insomnia and confusion as its primary symptoms.<ref>{{cite journal|title=At the Bedside: Serotonin syndrome: a brief review |journal=Canadian Medical Association Journal |year=2003 |volume=168 |issue=11 |pages=1439–1442 |accessdate=29 November 2013 |last1=Birmes |first1=P |last2=Coppin |first2=D |last3=Schmitt |first3=L |last4=Lauque |first4=D |pmc=155963 |pmid=12771076}}</ref><ref>{{cite journal|title=Current Concepts: The Serotonin Syndrome |journal=New England Journal of Medicine |year=2005 |volume=352 |issue=11 |pages=1112–1120 |doi=10.1056/NEJMra041867 |pmid=15784664 |url=http://toxicology.ucsd.edu/art%203%20serotonin%20syndrome.pdf |format=PDF |accessdate=29 November 2013 |last1=Boyer |first1=EW |last2=Shannon |first2=M|archiveurl=http://web.archive.org/web/20130618053344/http://toxicology.ucsd.edu/art%203%20serotonin%20syndrome.pdf |archivedate=2013-06-18 }}</ref> Although the condition is serious, it is not particularly common, generally only appearing at high doses or while on other medications. Assuming proper medical intervention has been taken (within about 24&nbsp;hours) it is rarely fatal.<ref>{{cite journal|url=http://meta.wkhealth.com/pt/pt-core/template-journal/lwwgateway/media/landingpage.htm?issn=0025-7974&volume=79&issue=4&spage=201 |title=Serotonin syndrome. Presentation of 2 cases and review of the literature |journal=Medicine (Baltimore) |last1=Mason |first1=PJ |last2=Morris |first2=VA |last3=Balcezak |first3=TJ |year=2000 |volume=79 |issue=4 |pages=201–209 |doi=10.1097/00005792-200007000-00001 |pmid=10941349 |subscription=yes |pmc=}}</ref><ref>{{cite journal|title=Serotonin syndrome: potentially fatal but difficult to recognize |last1=Sampson |first1=E |last2=Warner |first2=JP |pmid=10818648 |year=1999 |volume=49 |issue=448 |pages=867–868 |journal=British Journal of General Practice |pmc=1313553 |doi=10.1097/00005792-200007000-00001}}</ref>
Almost any medication involved with serotonin regulation has the potential to cause [[serotonin syndrome|serotonin toxicity]] (also known as ''serotonin syndrome''){{spaced ndash}} an excess of serotonin that can induce mania, restlessness, agitation, [[emotional lability]], insomnia and confusion as its primary symptoms.<ref name="pmid12771076">{{cite journal | author = Birmes P, Coppin D, Schmitt L, Lauque D | title = Serotonin syndrome: a brief review | journal = CMAJ | volume = 168 | issue = 11 | pages = 1439–42 | year = 2003 | pmid = 12771076 | pmc = 155963 | doi = }}</ref><ref name="pmid15784664">{{cite journal | author = Boyer EW, Shannon M | title = The serotonin syndrome | journal = N. Engl. J. Med. | volume = 352 | issue = 11 | pages = 1112–20 | year = 2005 | pmid = 15784664 | doi = 10.1056/NEJMra041867 | url = http://toxicology.ucsd.edu/art%203%20serotonin%20syndrome.pdf |format=PDF | archiveurl = http://web.archive.org/web/20130618053344/http://toxicology.ucsd.edu/art%203%20serotonin%20syndrome.pdf }}</ref> Although the condition is serious, it is not particularly common, generally only appearing at high doses or while on other medications. Assuming proper medical intervention has been taken (within about 24&nbsp;hours) it is rarely fatal.<ref name="pmid10941349">{{cite journal | author = Mason PJ, Morris VA, Balcezak TJ | title = Serotonin syndrome. Presentation of 2 cases and review of the literature | journal = Medicine (Baltimore) | volume = 79 | issue = 4 | pages = 201–9 | year = 2000 | month = July | pmid = 10941349 | doi = 10.1097/00005792-200007000-00001 }}</ref><ref name="pmid10818648">{{cite journal | author = Sampson E, Warner JP | title = Serotonin syndrome: potentially fatal but difficult to recognize | journal = Br J Gen Pract | volume = 49 | issue = 448 | pages = 867–8 | year = 1999 | month = November | pmid = 10818648 | pmc = 1313553 | doi = 10.1097/00005792-200007000-00001 }}</ref>


[[MAOIs]] tend to have pronounced (sometimes fatal) interactions with a wide variety of medications and [[over-the-counter drug]]s. If taken with foods that contain very high levels of [[tyramine]] (e.g., mature cheese, cured meats, or yeast extracts), they may cause a potentially lethal [[hypertensive crisis]]. At lower doses the person may be bothered by only a headache due to an increase in blood pressure.<ref>{{cite journal|title=Hypertensive crisis and cheese |journal=Indian Journal of Psychiatry |year=2009 |pages=65–66 |volume=51 |issue=1 |doi=10.4103/0019-5545.44910 |pmid=19742203 |last1=Yeragani |first1=V |last2=Yeragani |first2=TS |pmc=2738414}}</ref><!--
[[MAOIs]] tend to have pronounced (sometimes fatal) interactions with a wide variety of medications and [[over-the-counter drug]]s. If taken with foods that contain very high levels of [[tyramine]] (e.g., mature cheese, cured meats, or yeast extracts), they may cause a potentially lethal [[hypertensive crisis]]. At lower doses the person may be bothered by only a headache due to an increase in blood pressure.<ref name="pmid19742203">{{cite journal | author = Sathyanarayana Rao TS, Yeragani VK | title = Hypertensive crisis and cheese | journal = Indian J Psychiatry | volume = 51 | issue = 1 | pages = 65–6 | year = 2009 | pmid = 19742203 | pmc = 2738414 | doi = 10.4103/0019-5545.44910 }}</ref><!--


commented out 29 November 2013; no citation given, contributes little
commented out 29 November 2013; no citation given, contributes little
Line 986: Line 986:
Although these reactions may be lethal, the total number of deaths due to interactions and dietary concerns is comparable to over-the-counter medications.-->
Although these reactions may be lethal, the total number of deaths due to interactions and dietary concerns is comparable to over-the-counter medications.-->


In response to these adverse effects, a different type of MAOI has been developed: the [[reversible inhibitor of monoamine oxidase A]] (RIMA) class of drugs. Their primary advantage is that they do not require the person to follow a special diet, while being purportedly effective as SSRIs and tricyclics in treating depressive disorders.<ref name="pmid7717091">{{cite journal|doi=10.1111/j.1600-0447.1995.tb05920.x |title=Clinical efficacy of reversible and selective inhibitors of monoamine oxidase a in major depression |year=1995 |last1=Paykel |first1=E S. |journal=Acta Psychiatrica Scandinavica |volume=91 |pages=22–7 |pmid=7717091}}</ref>
In response to these adverse effects, a different type of MAOI has been developed: the [[reversible inhibitor of monoamine oxidase A]] (RIMA) class of drugs. Their primary advantage is that they do not require the person to follow a special diet, while being purportedly effective as SSRIs and tricyclics in treating depressive disorders.<ref name="pmid7717091">{{cite journal | author = Paykel ES | title = Clinical efficacy of reversible and selective inhibitors of monoamine oxidase A in major depression | journal = Acta Psychiatr Scand Suppl | volume = 386 | issue = | pages = 22–7 | year = 1995 | pmid = 7717091 | doi = 10.1111/j.1600-0447.1995.tb05920.x }}</ref>


===Pregnancy===
===Pregnancy===
Line 996: Line 996:
===Suicide===
===Suicide===
{{main|Antidepressants and suicide risk}}
{{main|Antidepressants and suicide risk}}
The relationship between antidepressant use and suicide risk is uncertain, complicated, and the target of medical research. Some studies have shown that the use of some antidepressants correlate with an increased risk of suicide in some patients and especially youth.<ref>{{cite doi|10.1002/14651858.CD008324.pub2}}</ref> This problem has been serious enough to warrant government interventions in some places to label greater likelihood of suicide as a risk of using antidepressants.<ref>{{cite doi| 10.1056/NEJMp078015}}</ref> The circumstances under which this can happen are not clear, and other studies show that antidepressants treat [[suicidal ideation]].
The relationship between antidepressant use and suicide risk is uncertain, complicated, and the target of medical research. Some studies have shown that the use of some antidepressants correlate with an increased risk of suicide in some patients and especially youth.<ref name="pmid23152255">{{cite journal | author = Cox GR, Callahan P, Churchill R, Hunot V, Merry SN, Parker AG, Hetrick SE | title = Psychological therapies versus antidepressant medication, alone and in combination for depression in children and adolescents | journal = Cochrane Database Syst Rev | volume = 11 | issue = | pages = CD008324 | year = 2012 | pmid = 23152255 | doi = 10.1002/14651858.CD008324.pub2 }}</ref> This problem has been serious enough to warrant government interventions in some places to label greater likelihood of suicide as a risk of using antidepressants.<ref name="pmid17485726">{{cite journal | author = Friedman RA, Leon AC | title = Expanding the black box - depression, antidepressants, and the risk of suicide | journal = N. Engl. J. Med. | volume = 356 | issue = 23 | pages = 2343–6 | year = 2007 | pmid = 17485726 | doi = 10.1056/NEJMp078015 }}</ref> The circumstances under which this can happen are not clear, and other studies show that antidepressants treat [[suicidal ideation]].


===Sexual===
===Sexual===
{{Details|Post-SSRI sexual dysfunction}}
{{Details|Post-SSRI sexual dysfunction}}


Sexual side-effects are also common with SSRIs, such as loss of [[sexual drive]], [[anorgasmia|failure to reach orgasm]], and [[erectile dysfunction]].<ref>{{cite book|last=Potenza|first=edited by Jon E. Grant, Marc N.|title=The Oxford handbook of impulse control disorders|year=2012|publisher=Oxford University Press|location=Oxford|isbn=978-0-19-538971-5}}</ref> Although usually reversible, these sexual side-effects can, in rare cases, last for months or years after the drug has been completely withdrawn. This is referred to as [[Post SSRI Sexual Dysfunction]].<ref>{{cite journal|last=Csoka|first=AB|author2=Bahrick, A|author3= Mehtonen, OP|title=Persistent sexual dysfunction after discontinuation of selective serotonin reuptake inhibitors.|journal=The journal of sexual medicine|date=Jan 2008|volume=5|issue=1|pages=227–33|pmid=18173768|doi=10.1111/j.1743-6109.2007.00630.x}}</ref>
Sexual side-effects are also common with SSRIs, such as loss of [[sexual drive]], [[anorgasmia|failure to reach orgasm]], and [[erectile dysfunction]].<ref>{{cite book|last=Potenza|first=edited by Jon E. Grant, Marc N.|title=The Oxford handbook of impulse control disorders|year=2012|publisher=Oxford University Press|location=Oxford|isbn=978-0-19-538971-5}}</ref> Although usually reversible, these sexual side-effects can, in rare cases, last for months or years after the drug has been completely withdrawn. This is referred to as [[Post SSRI Sexual Dysfunction]].<ref name="pmid18173768">{{cite journal | author = Csoka AB, Csoka A, Bahrick A, Mehtonen OP | title = Persistent sexual dysfunction after discontinuation of selective serotonin reuptake inhibitors | journal = J Sex Med | volume = 5 | issue = 1 | pages = 227–33 | year = 2008 | pmid = 18173768 | doi = 10.1111/j.1743-6109.2007.00630.x }}</ref>


In a study of 1022 outpatients, overall sexual dysfunction with all antidepressants averaged 59.1%<ref>{{cite journal|pmid=11229449 |year=2001 |last1=Montejo |first1=AL |last2=Llorca |first2=G |last3=Izquierdo |first3=JA |last4=Rico-Villademoros |first4=F |title=Incidence of sexual dysfunction associated with antidepressant agents: A prospective multicenter study of 1022 outpatients. Spanish Working Group for the Study of Psychotropic-Related Sexual Dysfunction |volume=62 |issue=Suppl 3 |pages=10–21 |journal=The Journal of clinical psychiatry}}</ref> with SSRIs values between 57 and 73%, mirtazapine 24%, nefazodone 8%, amineptine 7% and moclobemide 4%. [[Moclobemide]], a selective reversible MAO-A inhibitor, does not cause sexual dysfunction,<ref name="pmid19440080">{{cite journal|doi=10.1097/JCP.0b013e3181a5233f |title=Treatment-Emergent Sexual Dysfunction Related to Antidepressants |year=2009 |last1=Serretti |first1=Alessandro |last2=Chiesa |first2=Alberto |journal=Journal of Clinical Psychopharmacology |volume=29 |issue=3 |pages=259–66 |pmid=19440080}}</ref> and can actually lead to an improvement in all aspects of sexual function.<ref name="pmid9696909">{{cite journal|pmid=9696909 |year=1998 |last1=Chebili |first1=S |last2=Abaoub |first2=A |last3=Mezouane |first3=B |last4=Le Goff |first4=JF |title=Antidepressants and sexual stimulation: The correlation |volume=24 |issue=3 |pages=180–4 |journal=L'Encephale}}</ref>
In a study of 1022 outpatients, overall sexual dysfunction with all antidepressants averaged 59.1%<ref name="pmid11229449">{{cite journal | author = Montejo AL, Llorca G, Izquierdo JA, Rico-Villademoros F | title = Incidence of sexual dysfunction associated with antidepressant agents: a prospective multicenter study of 1022 outpatients. Spanish Working Group for the Study of Psychotropic-Related Sexual Dysfunction | journal = J Clin Psychiatry | volume = 62 Suppl 3 | issue = | pages = 10–21 | year = 2001 | pmid = 11229449 | doi = }}</ref> with SSRIs values between 57 and 73%, mirtazapine 24%, nefazodone 8%, amineptine 7% and moclobemide 4%. [[Moclobemide]], a selective reversible MAO-A inhibitor, does not cause sexual dysfunction,<ref name="pmid19440080">{{cite journal | author = Serretti A, Chiesa A | title = Treatment-emergent sexual dysfunction related to antidepressants: a meta-analysis | journal = J Clin Psychopharmacol | volume = 29 | issue = 3 | pages = 259–66 | year = 2009 | pmid = 19440080 | doi = 10.1097/JCP.0b013e3181a5233f }}</ref> and can actually lead to an improvement in all aspects of sexual function.<ref name="pmid9696909">{{cite journal | author = Chebili S, Abaoub A, Mezouane B, Le Goff JF | title = [Antidepressants and sexual stimulation: the correlation] | language = French | journal = Encephale | volume = 24 | issue = 3 | pages = 180–4 | year = 1998 | pmid = 9696909 | doi = }}</ref>


Biochemical mechanisms suggested as causative include increased serotonin, particularly affecting [[5-HT2 receptor|5-HT<sub>2</sub>]] and [[5HT3 receptor|5-HT<sub>3</sub> receptors]]; decreased [[dopamine]]; decreased [[norepinephrine]]; blockade of [[cholinergic receptor|cholinergic]] and [[alpha-1 adrenergic receptor|α<sub>1</sub>adrenergic receptors]]; inhibition of [[nitric oxide synthase|nitric oxide synthetase]]; and elevation of [[prolactin]] levels.<ref>{{cite journal|doi=10.1111/j.1744-6163.2002.tb00665.x |title=Biological Perspectives |year=2009 |last1=Keltner |first1=Norman L. |last2=McAfee |first2=Kelly M. |last3=Taylor |first3=Carey L. |journal=Perspectives in Psychiatric Care |volume=38 |issue=3 |pages=111–6 |pmid=12385082}}</ref> [[Mirtazapine]] is reported to have fewer sexual side-effects, most likely because it antagonizes 5-HT<sub>2</sub> and 5-HT<sub>3</sub> receptors and may, in some cases, reverse sexual dysfunction induced by SSRIs by the same mechanism.<ref>{{cite journal|doi=10.1002/hup.929 |title=Mirtazapine augmentation in depressed patients with sexual dysfunction due to selective serotonin reuptake inhibitors |year=2008 |last1=Ozmenler |first1=NK |last2=Karlidere |first2=T |last3=Bozkurt |first3=A |last4=Yetkin |first4=S |last5=Doruk |first5=A |last6=Sutcigil |first6=L |last7=Cansever |first7=A |last8=Uzun |first8=O |last9=Ozgen |first9=F |displayauthors=9 |last10=Ozsahin |first10=A |journal=Human Psychopharmacology: Clinical and Experimental |volume=23 |issue=4 |page=321}}</ref>
Biochemical mechanisms suggested as causative include increased serotonin, particularly affecting [[5-HT2 receptor|5-HT<sub>2</sub>]] and [[5HT3 receptor|5-HT<sub>3</sub> receptors]]; decreased [[dopamine]]; decreased [[norepinephrine]]; blockade of [[cholinergic receptor|cholinergic]] and [[alpha-1 adrenergic receptor|α<sub>1</sub>adrenergic receptors]]; inhibition of [[nitric oxide synthase|nitric oxide synthetase]]; and elevation of [[prolactin]] levels.<ref>{{cite journal|doi=10.1111/j.1744-6163.2002.tb00665.x |title=Biological Perspectives |year=2009 |last1=Keltner |first1=Norman L. |last2=McAfee |first2=Kelly M. |last3=Taylor |first3=Carey L. |journal=Perspectives in Psychiatric Care |volume=38 |issue=3 |pages=111–6 |pmid=12385082}}</ref> [[Mirtazapine]] is reported to have fewer sexual side-effects, most likely because it antagonizes 5-HT<sub>2</sub> and 5-HT<sub>3</sub> receptors and may, in some cases, reverse sexual dysfunction induced by SSRIs by the same mechanism.<ref>{{cite journal|doi=10.1002/hup.929 |title=Mirtazapine augmentation in depressed patients with sexual dysfunction due to selective serotonin reuptake inhibitors |year=2008 |last1=Ozmenler |first1=NK |last2=Karlidere |first2=T |last3=Bozkurt |first3=A |last4=Yetkin |first4=S |last5=Doruk |first5=A |last6=Sutcigil |first6=L |last7=Cansever |first7=A |last8=Uzun |first8=O |last9=Ozgen |first9=F |displayauthors=9 |last10=Ozsahin |first10=A |journal=Human Psychopharmacology: Clinical and Experimental |volume=23 |issue=4 |page=321}}</ref>
Line 1,070: Line 1,070:
==Pharmacology==
==Pharmacology==
{{Main|Pharmacology of antidepressants}}
{{Main|Pharmacology of antidepressants}}
The earliest and probably most widely accepted scientific theory of antidepressant action is the [[monoamine hypothesis]] (which can be traced back to the 1950s), which states that depression is due to an imbalance (most often a deficiency) of the [[monoamine neurotransmitter]]s (namely [[serotonin]], [[norepinephrine]] and [[dopamine]]).<ref name = GG/> It was originally proposed based on the observation that certain hydrazine anti-tuberculosis agents produce antidepressant effects, which was later linked to their inhibitory effects on [[monoamine oxidase]], the enzyme that catalyses the breakdown of the monoamine neurotransmitters.<ref name = GG/> All currently marketed antidepressants have the monoamine hypothesis as their theoretical basis, with the possible exception of [[agomelatine]] which acts on a dual [[Melatonin|melatonergic]]-[[Serotonin|serotonergic]] pathway.<ref name = GG/> Despite the success of the monoamine hypothesis it has a number of limitations: for one, all monoaminergic antidepressants have a delayed onset of action of at least a week; and secondly, there are a sizeable portion (>40%) of depressed patients that do not adequately respond to monoaminergic antidepressants.<ref name="Infl">{{cite pmid|19085093}}</ref><ref name="glut">{{cite pmid|21827775}}</ref> Further evidence to the contrary of the monoamine hypothesis are the recent findings that a single intravenous infusion with [[ketamine]], an antagonist of the [[NMDA receptor]]&nbsp;— a type of [[glutamate]] receptor&nbsp;— produces rapid (within 2&nbsp;hours), robust and sustained (lasting for up to a fortnight) antidepressant effects.<ref name = glut/> To overcome these flaws with the monoamine hypothesis a number of alternative hypotheses have been proposed, including the glutamate, neurogenic, [[Epigenetics|epigenetic]], cortisol hypersecretion and inflammatory hypotheses.<ref name = Infl/><ref name = glut/><ref name="Epig">{{cite pmid|22681167}}</ref><ref name="Epig2">{{cite pmid|23020296}}</ref>
The earliest and probably most widely accepted scientific theory of antidepressant action is the [[monoamine hypothesis]] (which can be traced back to the 1950s), which states that depression is due to an imbalance (most often a deficiency) of the [[monoamine neurotransmitter]]s (namely [[serotonin]], [[norepinephrine]] and [[dopamine]]).<ref name = GG/> It was originally proposed based on the observation that certain hydrazine anti-tuberculosis agents produce antidepressant effects, which was later linked to their inhibitory effects on [[monoamine oxidase]], the enzyme that catalyses the breakdown of the monoamine neurotransmitters.<ref name = GG/> All currently marketed antidepressants have the monoamine hypothesis as their theoretical basis, with the possible exception of [[agomelatine]] which acts on a dual [[Melatonin|melatonergic]]-[[Serotonin|serotonergic]] pathway.<ref name = GG/> Despite the success of the monoamine hypothesis it has a number of limitations: for one, all monoaminergic antidepressants have a delayed onset of action of at least a week; and secondly, there are a sizeable portion (>40%) of depressed patients that do not adequately respond to monoaminergic antidepressants.<ref name="Infl">{{cite journal | author = Maes, M; Yirmyia, R; Noraberg, J; Brene, S; Hibbeln, J; Perini, G; Kubera, M; Bob, P; Lerer, B; Maj, M | title = The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression | journal = Metabolic Brain Disease | volume = 24 | issue = 1 | pages = 27-53 | date = March 2009 | pmid = 19085093 | doi = 10.1007/s11011-008-9118-1 }}</ref><ref name="glut">{{cite journal | author = Sanacora, G; Treccani, G; Popoli, M | title = Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders | journal = Neuropharmacology | volume = 62 | issue = 1 | pages = 63-77 | date = January 2012 | pmid = 21827775 | doi = 10.1016/j.neuropharm.2011.07.036 | url = http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3205453/pdf/nihms318867.pdf | accessdate = | format = PDF }}</ref> Further evidence to the contrary of the monoamine hypothesis are the recent findings that a single intravenous infusion with [[ketamine]], an antagonist of the [[NMDA receptor]]&nbsp;— a type of [[glutamate]] receptor&nbsp;— produces rapid (within 2&nbsp;hours), robust and sustained (lasting for up to a fortnight) antidepressant effects.<ref name = glut/> To overcome these flaws with the monoamine hypothesis a number of alternative hypotheses have been proposed, including the glutamate, neurogenic, [[Epigenetics|epigenetic]], cortisol hypersecretion and inflammatory hypotheses.<ref name = Infl/><ref name = glut/><ref name="Epig">{{cite journal | author = Menke A, Klengel T, Binder EB | title = Epigenetics, depression and antidepressant treatment | journal = Current Pharmaceutical Design | volume = 18 | issue = 36 | pages = 5879-5889 | year = 2012 | pmid = 22681167 | doi = 10.2174/138161212803523590 | url = | accessdate = | format = }}</ref><ref name="Epig2">{{cite journal | author = Vialou, V; Feng, J; Robison, AJ; Nestler, EJ | title = Epigenetic mechanisms of depression and antidepressant action | journal = Annual Review of Pharmacology and Toxicology | volume = 53 | issue = 1 | pages = 59-87 | date = January 2013 | pmid = 23020296 | doi = 10.1146/annurev-pharmtox-010611-134540 | url = | accessdate = | format = }}</ref>
A recent study was done to discover more of the processes that are influenced by antidepressant medications. In this study mononuclear cells (MNC’s) were extracted from patients’ blood, because they have similar processes and protein structures as the brain, and tested it before medication was taken and then again after the medication was taken for six weeks. The antidepressants that the patients took varied slightly based on the need of the patient. The data collected from this experiment showed that different aspects of the integrin signal pathways were expressed differently after the medications. The integrin pathway in the central nervous system deal with synaptic plasticity and glutamatergic transmission, which essentially controls learning, memory, and anxiety behaviors. The conclusion showed that anti-depressants are most successful when protein expression is decreased. Granted, different antidepressants affect different proteins and signal pathways, but over all it can be seen that antidepressants seem to affect biological processes in the body in opposite directions.<ref>Martins-de-Souza, Daniel. "Blood Mononuclear Cell Proteome Suggests Integrin and Ras Signaling as Critical Pathways for Antidepressant Treatment Response." Rev. of Society of Biological Psychiatry. 06 Feb. 2014: E1-E3. Print.</ref>
A recent study was done to discover more of the processes that are influenced by antidepressant medications. In this study mononuclear cells (MNC’s) were extracted from patients’ blood, because they have similar processes and protein structures as the brain, and tested it before medication was taken and then again after the medication was taken for six weeks. The antidepressants that the patients took varied slightly based on the need of the patient. The data collected from this experiment showed that different aspects of the integrin signal pathways were expressed differently after the medications. The integrin pathway in the central nervous system deal with synaptic plasticity and glutamatergic transmission, which essentially controls learning, memory, and anxiety behaviors. The conclusion showed that anti-depressants are most successful when protein expression is decreased. Granted, different antidepressants affect different proteins and signal pathways, but over all it can be seen that antidepressants seem to affect biological processes in the body in opposite directions.<ref>Martins-de-Souza, Daniel. "Blood Mononuclear Cell Proteome Suggests Integrin and Ras Signaling as Critical Pathways for Antidepressant Treatment Response." Rev. of Society of Biological Psychiatry. 06 Feb. 2014: E1-E3. Print.</ref>


Line 1,108: Line 1,108:
Early studies have shown that [[ketamine]] may be effective in [[treatment-resistant depression]], though experts have stated that it is not yet ready for clinical practice but rather may lead to the development of novel medications in the future.<ref name="Rush2013">{{cite journal|last1=Rush |first1=A John |title=Ketamine for Treatment-Resistant Depression: Ready or Not for Clinical Use? |journal=American Journal of Psychiatry |volume=170 |issue=10 |year=2013 |pages=1079–81 |issn=0002-953X |doi=10.1176/appi.ajp.2013.13081034 |pmid=23982324}}</ref><ref>{{cite journal|last=Diazgranados |first=N |first2=Lobna |first3=Nancy E. |first4=Andrew |first5=Phillip |first6=Sami |first7=William A. |first8=Zenaide |first9=David A. |first10=Giacomo |first11=Rodrigo |first12=Husseini K. |first13=Carlos A. |title=A Randomized Add-on Trial of an N-methyl-d-aspartate Antagonist in Treatment-Resistant Bipolar Depression |journal=Archives of General Psychiatry |volume=67 |issue=8 |pages=793–802 |date=August 2010 |pmid=20679587 |doi=10.1001/archgenpsychiatry.2010.90 |pmc=3000408}}</ref> It produces a rapid antidepressant effect, acting within two hours as opposed to the several weeks taken by typical antidepressants to work.<ref>{{cite journal|author=Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, Kavalali ET, Monteggia LM |title=NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses |journal=Nature |volume=475 |issue=7354 |pages=91–95 |date=7 July 2011 |url=http://www.nature.com/nature/journal/vaop/ncurrent/full/nature10130.html |doi=10.1038/nature10130 |pmid=21677641 |pmc=3172695|archiveurl=http://web.archive.org/web/20110623092306/http://www.nature.com/nature/journal/vaop/ncurrent/full/nature10130.html |archivedate=2011-06-23 }} </ref>
Early studies have shown that [[ketamine]] may be effective in [[treatment-resistant depression]], though experts have stated that it is not yet ready for clinical practice but rather may lead to the development of novel medications in the future.<ref name="Rush2013">{{cite journal|last1=Rush |first1=A John |title=Ketamine for Treatment-Resistant Depression: Ready or Not for Clinical Use? |journal=American Journal of Psychiatry |volume=170 |issue=10 |year=2013 |pages=1079–81 |issn=0002-953X |doi=10.1176/appi.ajp.2013.13081034 |pmid=23982324}}</ref><ref>{{cite journal|last=Diazgranados |first=N |first2=Lobna |first3=Nancy E. |first4=Andrew |first5=Phillip |first6=Sami |first7=William A. |first8=Zenaide |first9=David A. |first10=Giacomo |first11=Rodrigo |first12=Husseini K. |first13=Carlos A. |title=A Randomized Add-on Trial of an N-methyl-d-aspartate Antagonist in Treatment-Resistant Bipolar Depression |journal=Archives of General Psychiatry |volume=67 |issue=8 |pages=793–802 |date=August 2010 |pmid=20679587 |doi=10.1001/archgenpsychiatry.2010.90 |pmc=3000408}}</ref> It produces a rapid antidepressant effect, acting within two hours as opposed to the several weeks taken by typical antidepressants to work.<ref>{{cite journal|author=Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, Kavalali ET, Monteggia LM |title=NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses |journal=Nature |volume=475 |issue=7354 |pages=91–95 |date=7 July 2011 |url=http://www.nature.com/nature/journal/vaop/ncurrent/full/nature10130.html |doi=10.1038/nature10130 |pmid=21677641 |pmc=3172695|archiveurl=http://web.archive.org/web/20110623092306/http://www.nature.com/nature/journal/vaop/ncurrent/full/nature10130.html |archivedate=2011-06-23 }} </ref>


Some research has attributed the effect to [[ketamine]] being an [[N-methyl-D-aspartate receptor|NMDA]] [[receptor antagonist]],<ref>{{cite journal|author=Zarate CA, Singh JB, Carlson PJ, ''et al.'' |title=A randomized trial of an N-methyl-D-aspartate antagonist in treatment-resistant major depression |journal=Archives of General Psychiatry |volume=63 |issue=8 |pages=856–64 |date=August 2006 |pmid=16894061 |doi=10.1001/archpsyc.63.8.856}}</ref><ref>{{cite journal|author=Berman RM, Cappiello A, Anand A, ''et al.'' |title=Antidepressant effects of ketamine in depressed patients |journal=Biological Psychiatry |volume=47 |issue=4 |pages=351–4 |date=February 2000 |pmid=10686270 |doi=10.1016/S0006-3223(99)00230-9}}</ref> though others have suggested that blocking the NMDA receptor is an intermediate step that increases the activity of another receptor, [[AMPA receptor|AMPA]], which is what is responsible for ketamine's rapid antidepressant actions.<ref>{{cite pmid|21669235}}</ref><ref>{{cite web|url=http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml |title=Faster-Acting Antidepressants Closer to Becoming a Reality |publisher=[[National Institute of Mental Health|NIMH]] |date=25 July 2007|archiveurl=http://web.archive.org/web/20130410040237/http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml |archivedate=2013-04-10 }}</ref>
Some research has attributed the effect to [[ketamine]] being an [[N-methyl-D-aspartate receptor|NMDA]] [[receptor antagonist]],<ref name="pmid10686270">{{cite journal | author = Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH | title = Antidepressant effects of ketamine in depressed patients | journal = Biol. Psychiatry | volume = 47 | issue = 4 | pages = 351–4 | year = 2000 | month = February | pmid = 10686270 | doi = 10.1001/archpsyc.63.8.856 }}</ref> though others have suggested that blocking the NMDA receptor is an intermediate step that increases the activity of another receptor, [[AMPA receptor|AMPA]], which is what is responsible for ketamine's rapid antidepressant actions.<ref name="pmid21669235">{{cite journal | author = Koike H, Iijima M, Chaki S | title = Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression | journal = Behav. Brain Res. | volume = 224 | issue = 1 | pages = 107–11 | year = 2011 | pmid = 21669235 | doi = 10.1016/j.bbr.2011.05.035 }}</ref><ref>{{cite web|url=http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml |title=Faster-Acting Antidepressants Closer to Becoming a Reality |publisher=[[National Institute of Mental Health|NIMH]] |date=25 July 2007 | archiveurl = http://web.archive.org/web/20130410040237/http://www.nimh.nih.gov/science-news/2007/faster-acting-antidepressants-closer-to-becoming-a-reality.shtml |archivedate=2013-04-10 }}</ref>


===Nutrition===
===Nutrition===


[[Omega-3 fatty acids]] have been proposed as a treatment for depression, alone or in combination with other treatments. One small pilot study of childhood depression (ages 6–12) suggested omega 3 fatty acids may have therapeutic benefits for treating childhood depression.<ref>{{cite journal|doi=10.1176/appi.ajp.163.6.1098 |title=Omega-3 Treatment of Childhood Depression: A Controlled, Double-Blind Pilot Study |year=2006 |last1=Nemets |first1=H |last2=Nemets |first2=B |last3=Apter |first3=A |last4=Bracha |first4=Z |last5=Belmaker |first5=RH |journal=American Journal of Psychiatry |volume=163 |issue=6 |pages=1098–100 |pmid=16741212}}</ref> A 2005 review article that included [[double-blind studies]], [[randomized control trials]], and epidemiological studies linking omega-3 fatty acids consumption and depression found that low fish consumption (the primary source of omega-3 fatty acids) correlated to increased rates of depression. Additionally, case-control and cohort studies of unipolar and [[postpartum depression]] indicated low blood levels of omega-3 fatty acids in depressed patients.<ref>{{cite journal|doi=10.1016/j.ypmed.2005.11.005 |title=Ω-3 polyunsaturated fatty acids and depression: A review of the evidence and a methodological critique |year=2006 |last1=Sontrop |first1=Jessica |last2=Campbell |first2=M Karen |journal=Preventive Medicine |volume=42 |issue=1 |pages=4–13 |pmid=16337677}}</ref>
[[Omega-3 fatty acids]] have been proposed as a treatment for depression, alone or in combination with other treatments. One small pilot study of childhood depression (ages 6–12) suggested omega 3 fatty acids may have therapeutic benefits for treating childhood depression.<ref name="pmid16741212">{{cite journal | author = Nemets H, Nemets B, Apter A, Bracha Z, Belmaker RH | title = Omega-3 treatment of childhood depression: a controlled, double-blind pilot study | journal = Am J Psychiatry | volume = 163 | issue = 6 | pages = 1098–100 | year = 2006 | pmid = 16741212 | doi = 10.1176/appi.ajp.163.6.1098 }}</ref> A 2005 review article that included [[double-blind studies]], [[randomized control trials]], and epidemiological studies linking omega-3 fatty acids consumption and depression found that low fish consumption (the primary source of omega-3 fatty acids) correlated to increased rates of depression. Additionally, case-control and cohort studies of unipolar and [[postpartum depression]] indicated low blood levels of omega-3 fatty acids in depressed patients.<ref name="pmid16337677">{{cite journal | author = Sontrop J, Campbell MK | title = Omega-3 polyunsaturated fatty acids and depression: a review of the evidence and a methodological critique | journal = Prev Med | volume = 42 | issue = 1 | pages = 4–13 | year = 2006 | pmid = 16337677 | doi = 10.1016/j.ypmed.2005.11.005 }}</ref>


A 2008 review of clinical studies of the effectiveness of omega-3 fatty acids on depression has shown somewhat inconsistent results: "Of the evaluated studies, 13 showed a significant positive association between omega-3 and depression, while six studies did not show a relationship between the referred variables."<ref>{{cite journal|doi=10.1586/ern.10.77 |title=What is the effectiveness of the use of polyunsaturated fatty acid omega-3 in the treatment of depression? |year=2010 |last1=Rocha Araujo |first1=Daniele Marano |last2=Vilarim |first2=Marina Machado |last3=Nardi |first3=Antonio E |journal=Expert Review of Neurotherapeutics |volume=10 |issue=7 |pages=1117–29 |pmid=20586692}}</ref> To be read with caution because of limited data, a 2008 [[Cochrane Library|Cochrane]] systematic review found in the one eligible study that omega-3 fatty acids are an effective [[adjunctive therapy]] for depressed but not manic symptoms in [[bipolar disorder]]. The authors found an "acute need" for more randomized, controlled trials.<ref>{{cite journal|doi=10.1002/14651858.CD005169.pub2 |title=Omega-3 fatty acids for bipolar disorder |journal=Cochrane Database of Systematic Reviews |year=2008 |last1=Montgomery |first1=Paul |last2=Richardson |first2=Alexandra J |editor1-last=Montgomery |editor1-first=Paul |pmid=18425912 |issue=2 |pages=CD005169}}</ref>
A 2008 review of clinical studies of the effectiveness of omega-3 fatty acids on depression has shown somewhat inconsistent results: "Of the evaluated studies, 13 showed a significant positive association between omega-3 and depression, while six studies did not show a relationship between the referred variables."<ref>{{cite journal|doi=10.1586/ern.10.77 |title=What is the effectiveness of the use of polyunsaturated fatty acid omega-3 in the treatment of depression? |year=2010 |last1=Rocha Araujo |first1=Daniele Marano |last2=Vilarim |first2=Marina Machado |last3=Nardi |first3=Antonio E |journal=Expert Review of Neurotherapeutics |volume=10 |issue=7 |pages=1117–29 |pmid=20586692}}</ref> To be read with caution because of limited data, a 2008 [[Cochrane Library|Cochrane]] systematic review found in the one eligible study that omega-3 fatty acids are an effective [[adjunctive therapy]] for depressed but not manic symptoms in [[bipolar disorder]]. The authors found an "acute need" for more randomized, controlled trials.<ref>{{cite journal|doi=10.1002/14651858.CD005169.pub2 |title=Omega-3 fatty acids for bipolar disorder |journal=Cochrane Database of Systematic Reviews |year=2008 |last1=Montgomery |first1=Paul |last2=Richardson |first2=Alexandra J |editor1-last=Montgomery |editor1-first=Paul |pmid=18425912 |issue=2 |pages=CD005169}}</ref>

Revision as of 20:08, 22 May 2014

refer to caption
Fluoxetine (Prozac), an SSRI
chemical structure of the SNRI drug venlafaxine
The chemical structure of venlafaxine (Effexor), an SNRI

Antidepressants are drugs used for the treatment of major depressive disorder and other conditions, including dysthymia, anxiety disorders, obsessive compulsive disorder, eating disorders, chronic pain, neuropathic pain and, in some cases, dysmenorrhoea, snoring, migraines, attention-deficit hyperactivity disorder (ADHD), substance abuse and sleep disorders. They can be used alone or in combination with other medications.

The most important classes of antidepressants are the selective serotonin reuptake inhibitors (SSRIs), serotonin–norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs) and monoamine oxidase inhibitors (MAOIs). Other drugs used or proposed for the treatment of depression include buprenorphine,[1] tryptophan,[2] low-dose antipsychotics,[3] and St John's wort.[4]

Efficacy

To establish efficacy, an antidepressant must show that it can produce a therapeutic effect for the condition for which it is taken. An antidepressant should be more efficacious than placebo to justify the risk associated with side effects. For depression, the Hamilton Depression Rating Scale (HAM-D) is often used to measure the severity of depression.[5] The maximum score for the 17-item HAM-D questionnaire is 52; the higher the score, the more severe the depression. What constitute a sufficient response to a drug has not been well established, but total remission or virtual elimination of all depression symptoms is the goal, however, remission rates are rarely published. For placebo, the percentage of symptom reduction is approximately 31 to 38%, compared to 46 to 54% for antidepressants.[6]

On the basis of 234 studies, no clinically relevant superiority of one antidepressant over another was detected for the treatment of acute, continuation, and maintenance phases of depression, taking into account age, sex, ethnicity, or comorbid conditions. Individual drugs differed in onset of action, adverse events, and some measures of health-related quality of life.[7]

The largest and most expensive study conducted to date, on the effectiveness of pharmacological treatment for depression, was commissioned by the National Institute of Mental Health.[8] The study was dubbed "The Sequenced Treatment Alternatives to Relieve Depression" (STAR*D) Study. The results[9][10] are summarized here.

  • After the first course of treatment, 27.5% of the 2,876 participants reached remission with a HAM-D score of 7 or less. 21% dropped out.[11]
  • After the second course of treatment, 21 to 30% of the remaining 1,439 participants remitted. Only 310 participants were willing or available to continue the study.[10] Switching medications can achieve remission in about 25% of patients.[12]
  • After the third course of treatment, 17.8% of the remaining 310 participants remitted.
  • After the fourth and last course of treatment, 10.1% of the remaining 109 participants remitted.
  • After a one year follow-up, of the 1085 remitted participants, 93% participants had either relapsed or dropped out of the study.

There were no statistical or meaningful clinical differences in remission rates, response rates, or times to remission or response among any of the medications compared in this study.[13] These included bupropion sustained release, bupropion, citalopram, lithium, mirtazapine, nortriptyline, sertraline, triiodothyronine, tranylcypromine, and venlafaxine extended release.[medical citation needed]

A 2008 review of randomized controlled trials concluded that symptomatic improvement with SSRIs was greatest by the end of the first week of use, but that some improvement continued for at least 6 weeks.[14]

Clinical guidelines

The UK National Institute for Clinical Excellence (NICE) 2004 guidelines indicate that antidepressants should not be used for the initial treatment of mild depression, because the risk-benefit ratio is poor; that for moderate or severe depression an SSRI is more likely to be tolerated than a tricyclic; and that antidepressants for severe depression should be combined with a psychological treatment such as Cognitive Behavioral Therapy.[15]

The American Psychiatric Association 2000 Practice Guideline for the Treatment of Patients with major depressive disorder indicates that, if preferred by the patient, antidepressant medications may be provided as an initial primary treatment for mild major depressive disorder; antidepressant medications should be provided for moderate to severe major depressive disorder unless electroconvulsive therapy is planned; and a combination of antipsychotic and antidepressant medications or electroconvulsive therapy should be used for psychotic depression. It states that efficacy is generally comparable between classes and within classes and that the initial selection will largely be based on the anticipated side-effects for an individual patient, patient preference, quantity and quality of clinical trial data regarding the medication, and its cost.[16]

Limitations and strategies

Between 30% and 50% of individuals treated with a given antidepressant do not show a response.[17][18] In clinical studies, approximately one-third of patients achieve a full remission, one-third experience a response and one-third are nonresponders. Partial remission is characterized by the presence of poorly defined residual symptoms. These symptoms typically include depressed mood, psychic anxiety, sleep disturbance, fatigue and diminished interest or pleasure. It is currently unclear which factors predict partial remission. However, it is clear that residual symptoms are powerful predictors of relapse, with relapse rates 3–6 times higher in patients with residual symptoms than in those who experience full remission.[19] In addition, antidepressant drugs tend to lose efficacy over the course of treatment.[20] A number of strategies are used in clinical practice to try to overcome these limits and variations.[21] They include switching medication, augmentation, and combination.

"Trial and error" switching

The American Psychiatric Association 2000 Practice Guideline advises that where no response is achieved following six to eight weeks of treatment with an antidepressant, to switch to an antidepressant in the same class, then to a different class of antidepressant. The remission rate reported by the STAR*D study was 21% using this method.[medical citation needed]

A 2006 meta-analysis review found wide variation in the findings of prior studies; for patients who had failed to respond to an SSRI antidepressant, between 12% and 86% showed a response to a new drug. However, the more antidepressants an individual had already tried, the less likely they were to benefit from a new antidepressant trial.[18] However, a later meta-analysis found no difference between switching to a new drug and staying on the old medication; although 34% of treatment resistant patients responded when switched to the new drug, 40% responded without being switched.[22] Thus, the clinical response to the new drug might be a placebo effect associated with the belief that one is receiving a different medication.[medical citation needed]

Augmentation and combination

For a partial response, the American Psychiatric Association guidelines suggest augmentation, or adding a drug from a different class. These include: lithium and thyroid augmentation, dopamine agonists, sex steroids, NRIs, glucocorticoid-specific agents, or the newer anticonvulsants.[23]

A combination strategy involves adding an additional antidepressant, usually from a different class so as to have effect on other mechanisms. Although this may be used in clinical practice, there is little evidence for the relative efficacy or adverse effects of this strategy.[24]

Opponents of switching, augmentation and combination argue that treatment may also propel the illness to a malignant and treatment-unresponsive course with iatrogenic psychiatric-like symptoms and treatment resistance or episode acceleration.[25]

Long-term use

The therapeutic effects of antidepressants typically do not continue once the course of medication ends, resulting in a high rate of relapse. A recent meta-analysis of 31 placebo-controlled antidepressant trials, mostly limited to studies covering a period of one year, found that 18% of patients who had responded to an antidepressant relapsed while still taking it, compared to 41% whose antidepressant was switched for a placebo.[26]

In a five-year follow up, relapse rates was 23% greater for users greater than one year, but not different for 6 or 12 months users.[27] In addition, gradual loss of therapeutic benefit occurs during the course of treatment.[28] A strategy involving the use of pharmacotherapy in the treatment of the acute episode, followed by psychotherapy in its residual phase, has been suggested by some studies.[29][30]

Antidepressant-induced mania

Another possible problem with antidepressants is the chance of antidepressant-induced mania in patients with bipolar disorder. Many cases of bipolar depression are very similar to those of unipolar depression. Therefore, the patient can be misdiagnosed with unipolar depression and be given antidepressants. Studies have shown that antidepressant-induced mania can occur in 20–40% of bipolar patients.[31]

Controversy: the active placebo theory

Various researchers have contested the pharmacological activity of antidepressants in the relief of depression, and state that the evidence is most consistent a role as active placebos.[32]

Controversy regarding the efficacy of antidepressants has arisen due to studies showing that antidepressants fail to provide significantly greater efficacy than placebo in some studies. A 2002 study claimed that the difference between antidepressants and placebo is close to negligible.[32]

Through a Freedom of Information Act request, two psychologists obtained 47 studies used by the FDA for approval of the six antidepressants prescribed most widely between 1987-99. Overall, antidepressant pills worked 18% better than placebos, a statistically significant difference, "but not meaningful for people in clinical settings", says psychologist Irving Kirsch, lead author of the study. He and co-author Thomas Moore released their findings in "Prevention and Treatment", an e-journal of the American Psychological Association.[33] In a later publication, Kirsch concluded that the overall effect of new-generation antidepressant medication is below recommended criteria for clinical significance.[34]

A meta-analysis done by two psychologists led them to believe that although the drugs did help people, the difference between the pills and placebo was not meaningful for patients; a later publication by the same author concluded newer-generation medicines were below the criteria of clinical significance.[35][34] Another study focusing on paroxetine (Paxil) and imipramine found that antidepressant drugs were hardly better than placebo in cases of mild or moderate depression they surveyed.[36]

A study published in the Journal of the American Medical Association (JAMA) demonstrated that the magnitude of the placebo effect in clinical trials of depression have been growing over time, while the effect size of tested drugs has remained relatively constant. The authors suggest that one possible explanation for the growing placebo effect in clinical trials is the inclusion of larger number of participants with shorter term, mild, or spontaneously remitting depression as a result of decreasing stigma associated with antidepressant use.[37]


The Cochrane Collaboration recently performed a systematic review of clinical trials of the generic antidepressant amitriptyline. The study concluded that in spite of moderate evidence for publication bias, there is strong evidence that the efficacy of amitriptyline is superior to placebo.[38]

A review commissioned by the National Institute for Clinical Excellence concluded that there is strong evidence that SSRIs have greater efficacy than placebo on achieving a 50% reduction in depression scores in moderate and severe major depression, and that there is some evidence for a similar effect in mild depression. The treatment guidelines developed in conjunction with this review suggest that antidepressants should be considered in patients with moderate to severe depression and those with mild depression that is persistent or resistant to other treatment modalities.[39]

In 2005, antidepressants became the most prescribed drug in the United States, causing more debate over the issue. Some doctors believe this is a positive sign that people are finally seeking help for their issues. Others disagree, saying that this shows that people are becoming too dependent on antidepressants.[40]

In 2012, Aimee Hunter and her team used electroencephalography (EEG) and showed that taking placebo decreased pre-frontal brain activity in those subjects who had prior use of an antidepressants, similar to the expected antidepressant response, but increased brain activity in antidepressant-naive subjects. She attributes this antidepressant response of placebo, in repeat users, to a memory effect.[41]

However, the later experiment conducted by John H. Krystal at Yale University School of Medicine to assess whether growth mixture modeling can provide insights into antidepressant and placebo responses in clinical trials of patients with major depression showed that Duloxetine and SSRI did not differ in efficacy, and compared with placebo they significantly decreased the odds of following the nonresponder trajectory. Antidepressant responders had significantly better Hamilton Depression Rating Scale (HAM-D) scores over time than placebo-treated patients, but antidepressant nonresponders had significantly worse HAM-D scores over time than the placebo-treated patients.[42]

Evidence of pharmaceutical company deception

A review of antidepressant trials submitted to the FDA by the industry for drug approval revealed that when a trial was successful, the results of the trial was published 94% of the time, however, when the trial was not found to be more effective than placebo, it was only published 50% of the time. This demonstrated a measure of bias in reporting by industry. Combined, 51% of all studies showed efficacy.[43] The difference in effect between active placebos and several antidepressants appeared small and strongly affected by publication bias.[43]

Comparative efficacy and tolerability

Adverse effects

Difficulty tolerating adverse effects is the most common reason for antidepressant discontinuation.[medical citation needed]

General

For bipolar depression, antidepressants (most frequently SSRIs) can exacerbate or trigger symptoms of hypomania and mania.[62]

Almost any medication involved with serotonin regulation has the potential to cause serotonin toxicity (also known as serotonin syndrome) – an excess of serotonin that can induce mania, restlessness, agitation, emotional lability, insomnia and confusion as its primary symptoms.[63][64] Although the condition is serious, it is not particularly common, generally only appearing at high doses or while on other medications. Assuming proper medical intervention has been taken (within about 24 hours) it is rarely fatal.[65][66]

MAOIs tend to have pronounced (sometimes fatal) interactions with a wide variety of medications and over-the-counter drugs. If taken with foods that contain very high levels of tyramine (e.g., mature cheese, cured meats, or yeast extracts), they may cause a potentially lethal hypertensive crisis. At lower doses the person may be bothered by only a headache due to an increase in blood pressure.[67]

In response to these adverse effects, a different type of MAOI has been developed: the reversible inhibitor of monoamine oxidase A (RIMA) class of drugs. Their primary advantage is that they do not require the person to follow a special diet, while being purportedly effective as SSRIs and tricyclics in treating depressive disorders.[68]

Pregnancy

Pregnancy can trigger a range of emotions that make it more difficult to cope with depression. The risk of medication discontinuation and relapse have to be weighed against the risk to the developing fetus and baby. Some antidepressants have lower risk for the baby during pregnancy, but the FDA advises for the risk of birth defects with the use of Paxil[69] and the MAOI should be avoided. The levels of protein expression and constant change of hormones in woman is very common especially when they are taking antidepressant medications. So when a woman becomes pregnant her hormone levels and other vitals must be carefully watched if these medications are going to be continued. Many medications such as the ones mentioned above can lead to low birth weights of newborns as well as preterm birth. Therefore, it is vital that if these medications are to be continued during pregnancy maintenance and therapy must be done to insure the safety of the woman and the child. Timing and the length of exposure for drugs, “during certain trimesters may influence the antidepressants effect on fetal development and subsequent birth outcomes".[70] A neonate (infant less than 28 days old) may experience a withdrawal syndrome from abrupt discontinuation of the antidepressant at birth. The use of antidepressants during pregnancy is associated with an increased risk of spontaneous abortion,[71] birth defects,[72] and developmental delays.[73] Antidepressants have been shown to be present in varying amounts in breast milk, but their effects on infants are currently unknown.[74]

Moreover, SSRIs inhibit nitric oxide synthesis, which leads to vasoconstriction. This is significant in pregnancy as SSRIs have been associated with the development of hypertension (high blood pressure) and pre-eclampsia of pregnancy. This in turn can lead to fetal prematurity.[75]

Suicide

The relationship between antidepressant use and suicide risk is uncertain, complicated, and the target of medical research. Some studies have shown that the use of some antidepressants correlate with an increased risk of suicide in some patients and especially youth.[76] This problem has been serious enough to warrant government interventions in some places to label greater likelihood of suicide as a risk of using antidepressants.[77] The circumstances under which this can happen are not clear, and other studies show that antidepressants treat suicidal ideation.

Sexual

Sexual side-effects are also common with SSRIs, such as loss of sexual drive, failure to reach orgasm, and erectile dysfunction.[78] Although usually reversible, these sexual side-effects can, in rare cases, last for months or years after the drug has been completely withdrawn. This is referred to as Post SSRI Sexual Dysfunction.[79]

In a study of 1022 outpatients, overall sexual dysfunction with all antidepressants averaged 59.1%[80] with SSRIs values between 57 and 73%, mirtazapine 24%, nefazodone 8%, amineptine 7% and moclobemide 4%. Moclobemide, a selective reversible MAO-A inhibitor, does not cause sexual dysfunction,[81] and can actually lead to an improvement in all aspects of sexual function.[82]

Biochemical mechanisms suggested as causative include increased serotonin, particularly affecting 5-HT2 and 5-HT3 receptors; decreased dopamine; decreased norepinephrine; blockade of cholinergic and α1adrenergic receptors; inhibition of nitric oxide synthetase; and elevation of prolactin levels.[83] Mirtazapine is reported to have fewer sexual side-effects, most likely because it antagonizes 5-HT2 and 5-HT3 receptors and may, in some cases, reverse sexual dysfunction induced by SSRIs by the same mechanism.[84]

Bupropion, a weak NDRI and nicotinic antagonist, may be useful in treating reduced libido as a result of SSRI treatment.[85] However, these results are preliminary, and as such must be taken cum grano salis.

REM Sleep

All major antidepressant drugs – except trimipramine, bupropion,[86] mirtazapine, and nefazodone – suppress REM sleep, and it has been proposed that the clinical efficacy of these drugs largely derives from their suppressant effects on REM sleep. The three major classes of antidepressant drugs (MAOIs, TCAs, and SSRIs), profoundly suppress REM sleep.[87] Mirtazapine either has no effect on REM sleep or increases it slightly.[88] The MAOIs almost completely suppress REM sleep, while the TCAs and SSRIs have been shown to produce immediate (40–85%) and sustained (30–50%) reductions in REM sleep. This effect often causes increased fatigue in patients who take large doses of antidepressants for extended periods of time. Such fatigue can occasionally interfere with a patient's everyday activities. Abrupt discontinuation of MAOIs can cause a temporary phenomenon known as "REM rebound" in which the patient experiences extremely vivid dreams and nightmares.[89]

Changes in weight

Changes in appetite or weight are common among antidepressants, but largely drug-dependent and are related to which neurotransmitters they affect. Mirtazapine and paroxetine, for example, have the effect of weight gain and/or increased appetite,[90][91][92] while others (such as bupropion and venlafaxine) achieve the opposite effect.[93][94]

The antihistaminic properties of certain TCA- and TeCA-class antidepressants have been shown to contribute to the common side-effects of increased appetite and weight gain associated with these classes of medication.

Withdrawal symptoms

If an SSRI is suddenly discontinued, it frequently produces an event of "SSRI discontinuation syndrome" that has a both a bodily and psychological withdrawal component.[95]

Withdrawal syndromes have been reported with TCAs,[96] MOAIs,[97] SNRIs,[98] and with SSRIs. Researchers from the Nordic Cochrane Center in Denmark compared the signs and symptoms of SSRI discontinuation to those of the benzodiazepine withdrawal syndrome[99] and concluded that the withdrawal reactions were so similar that both withdrawal reactions indicated a dependence syndrome. Elsewhere, concerns have been raised that SSRIs cause dependence.[100]

When treatment is prolonged over 6–9 months, processes oppose the initial effects of antidepressant drugs (loss of clinical effects). When drug treatment ends, these processes may be unopposed and yield withdrawal symptoms and increased vulnerability to relapse. Such processes are not necessarily reversible. The more antidepressants are switched or potentiated, the more likely oppositional tolerance can take place.[28]

Some of the withdrawal symptoms of SSRI discontinuation include: nausea, chills, muscles aches, dizziness, anxiety, irritability, insomnia, fatigue,[101] and, in some patients, electric shock sensations.[102][103]

Moreover, when changes in antidepressant dosage occur, whether up or down, a doubling of the risk of suicide is seen.[104]

To minimize the intensity of withdrawal and rebound effects[105] antidepressants should be discontinued over a period of several weeks or months depending on a person's response to reductions. A suggested regimen is a decrease in the SSRI by about 25% per week.[101] This is a guideline; the actual amount of time required to withdraw from a given antidepressant is unique to the drug. Certain antidepressants may have long half-lives and remain in the person's system for a period of time long enough to prevent a sudden "drop" in concentration, meaning that withdrawal or rebound effects are unlikely or less pronounced.

Most cases of discontinuation syndrome last between one and four weeks but a substantial minority, perhaps up to 15% of users, have persistent withdrawal symptoms evident one year post-withdrawal.[106] Paroxetine and venlafaxine[97][102][107][108][109][110][111] seem to be particular difficult to discontinue and prolonged withdrawal syndrome lasting over 18 months have been reported with paroxetine.[95] Peer-support groups exist to help patients taper off of their antidepressants.[112]

Environmental impacts

There is good reason to believe that much of the large amounts of antidepressants consumed end up back into the ecosystem. Michael Thomas of Idaho State University in Pocatello reports that around 80% of these drugs are passed right through our bodies and end up in our water supplies.[113] Fluoxetine is exceted from humans unchanged or as glucuronide.[114] Because most antidepressants function by inhibiting the reuptake of neurotransmitters serotonin, dopamine, and norepinepherine[115] these drugs can interfere with natural neurotransmitter levels in other organisms impacted by indirect exposure.[116]

Antidepressants fluoxetine and sertraline have been detected in aquatic organisms residing in effluent dominated streams.[117] The presence of antidepressants in surface waters and aquatic organisms has caused concern because ecotoxicological effects to aquatic organisms due to fluoxetine exposure have been demonstrated.[118] Coral reef fish have been demonstrated to modulate aggressive behavior through serotonin.[119] Exposure to fluoxetine has been demonstrated to increase serotonergic activity in fish, subsequently reducing aggressive behavior.[120] Artificially increasing serotonin levels in crustaceans can temporarily reverse social status and turn subordinates into aggressive and territorial dominant males.[121] Perinatal exposure to fluoxetine at relevant environmental concentrations has been shown to lead to significant modifications of memory processing in 1-month-old cuttlefish[122] This impairment may disadvantage cuttlefish and decrease their survival.

Pharmacology

The earliest and probably most widely accepted scientific theory of antidepressant action is the monoamine hypothesis (which can be traced back to the 1950s), which states that depression is due to an imbalance (most often a deficiency) of the monoamine neurotransmitters (namely serotonin, norepinephrine and dopamine).[44] It was originally proposed based on the observation that certain hydrazine anti-tuberculosis agents produce antidepressant effects, which was later linked to their inhibitory effects on monoamine oxidase, the enzyme that catalyses the breakdown of the monoamine neurotransmitters.[44] All currently marketed antidepressants have the monoamine hypothesis as their theoretical basis, with the possible exception of agomelatine which acts on a dual melatonergic-serotonergic pathway.[44] Despite the success of the monoamine hypothesis it has a number of limitations: for one, all monoaminergic antidepressants have a delayed onset of action of at least a week; and secondly, there are a sizeable portion (>40%) of depressed patients that do not adequately respond to monoaminergic antidepressants.[123][124] Further evidence to the contrary of the monoamine hypothesis are the recent findings that a single intravenous infusion with ketamine, an antagonist of the NMDA receptor — a type of glutamate receptor — produces rapid (within 2 hours), robust and sustained (lasting for up to a fortnight) antidepressant effects.[124] To overcome these flaws with the monoamine hypothesis a number of alternative hypotheses have been proposed, including the glutamate, neurogenic, epigenetic, cortisol hypersecretion and inflammatory hypotheses.[123][124][125][126] A recent study was done to discover more of the processes that are influenced by antidepressant medications. In this study mononuclear cells (MNC’s) were extracted from patients’ blood, because they have similar processes and protein structures as the brain, and tested it before medication was taken and then again after the medication was taken for six weeks. The antidepressants that the patients took varied slightly based on the need of the patient. The data collected from this experiment showed that different aspects of the integrin signal pathways were expressed differently after the medications. The integrin pathway in the central nervous system deal with synaptic plasticity and glutamatergic transmission, which essentially controls learning, memory, and anxiety behaviors. The conclusion showed that anti-depressants are most successful when protein expression is decreased. Granted, different antidepressants affect different proteins and signal pathways, but over all it can be seen that antidepressants seem to affect biological processes in the body in opposite directions.[127]

Types

Adjuncts

Adjunct medications are an umbrella term used to describe substances that increase the potency or "enhance" antidepressants.[128] They work by affecting variables very close to the antidepressant, sometimes affecting a completely different mechanism of action. This is may be attempted when depression treatments have not been successful in the past.

Types of adjunct medication techniques generally fall into the following categories:

  • Two or more antidepressants taken together
    • From the same class (affecting the same area of the brain, often at a much higher level)
    • From different classes (affecting multiple parts of the brain not covered simultaneously by either drug alone)
  • A stimulant with an antidepressant (e.g., amphetamine and fluoxetine)
  • An antipsychotic, particularly atypical antipsychotics, for reasons not clearly understood

A review article published in 2007 found psychostimulants may be effective in treatment-resistant depression with concomitant antidepressant therapy. A more certain conclusion could not be drawn due to substantial deficiencies in the studies available for consideration, and the somewhat contradictory nature of their results.[129]

Chronic nicotine intake via nicotine patches results in an increased response to standard antidepressants. Similarly varenicline has been shown to augment sub-therapeutic doses of SSRIs to produce an antidepressant effect.[130]

Atypical antipsychotics such as aripiprazole (Abilify), quetiapine (Seroquel), olanzapine, and risperidone are also popular adjuncts and appear to be an effective adjunctive treatment option in this indication.[131] Lithium may also be used as an adjunct in major depressive disorder.[131][132][133] Triiodothyronine (T3) has also been successfully used as an adjunct in major depressive disorder.[131][134]

Nicotine

Nicotine is believed to act as an antidepressant,[135] by stimulating the release of dopamine and norepinephrine; in addition, nicotine is believed to exert an antidepressant effect due to the desensitisation of nicotinic receptors, which occurs as a result of tolerance.[136] Clinical trials have demonstrated nicotine (administered using a dermal nicotine patch) exerts an antidepressant effect in both depressed nonsmokers and smokers, and can be considered for treatment-resistant depression. The proposed mechanism of chronic nicotine use causing desensitisation of nicotinic receptors – thereby leading to an antidepressant effect – is consistent with the theory first proposed over 30 years ago and subsequent research that confirmed excessive acetylcholine activity in the brain leads to depressive symptoms. Varenicline, a nicotinic receptor-acting drug used to wean people off of nicotine dependence, has also demonstrated antidepressant properties.[130]

Caffeine

Individuals using caffeine at moderate doses (fewer than 6 cups of coffee per day), have a reduced incidence of depressive symptoms and an overall reduced risk of suicide. Anxiety is an important side-effect of caffeine that occurs more commonly in individuals suffering from panic disorder or social phobia or when taken in excessive amounts.[137]

Ketamine

Early studies have shown that ketamine may be effective in treatment-resistant depression, though experts have stated that it is not yet ready for clinical practice but rather may lead to the development of novel medications in the future.[138][139] It produces a rapid antidepressant effect, acting within two hours as opposed to the several weeks taken by typical antidepressants to work.[140]

Some research has attributed the effect to ketamine being an NMDA receptor antagonist,[141] though others have suggested that blocking the NMDA receptor is an intermediate step that increases the activity of another receptor, AMPA, which is what is responsible for ketamine's rapid antidepressant actions.[142][143]

Nutrition

Omega-3 fatty acids have been proposed as a treatment for depression, alone or in combination with other treatments. One small pilot study of childhood depression (ages 6–12) suggested omega 3 fatty acids may have therapeutic benefits for treating childhood depression.[144] A 2005 review article that included double-blind studies, randomized control trials, and epidemiological studies linking omega-3 fatty acids consumption and depression found that low fish consumption (the primary source of omega-3 fatty acids) correlated to increased rates of depression. Additionally, case-control and cohort studies of unipolar and postpartum depression indicated low blood levels of omega-3 fatty acids in depressed patients.[145]

A 2008 review of clinical studies of the effectiveness of omega-3 fatty acids on depression has shown somewhat inconsistent results: "Of the evaluated studies, 13 showed a significant positive association between omega-3 and depression, while six studies did not show a relationship between the referred variables."[146] To be read with caution because of limited data, a 2008 Cochrane systematic review found in the one eligible study that omega-3 fatty acids are an effective adjunctive therapy for depressed but not manic symptoms in bipolar disorder. The authors found an "acute need" for more randomized, controlled trials.[147]

History

refer to caption
St John's wort

Before the 1950s, opioids, amphetamine, and methamphetamine were commonly used as antidepressants. Their use was later restricted due to their addictive nature and side effects.[148] Extracts from the herb St John's wort had been used as a "nerve tonic" to alleviate depression.[149]

Isoniazid, iproniazid, and imipramine

In 1951, Irving Selikoff and Edward Robitzek, working out of Sea View Hospital on Staten Island, began clinical trials on two new anti-tuberculosis agents developed by Hoffman-LaRoche, isoniazid and iproniazid. Only patients with a poor prognosis were initially treated; nevertheless, their condition improved dramatically. Selikoff and Robitzek noted "a subtle general stimulation ... the patients exhibited renewed vigor and indeed this occasionally served to introduce disciplinary problems."[150] The promise of a cure for tuberculosis in the Sea View Hospital trials was excitedly discussed in the mainstream press.

In 1952, learning of the stimulating side effects of isoniazid, the Cincinnati psychiatrist Max Lurie tried it on his patients. In the following year, he and Harry Salzer reported that isoniazid improved depression in two thirds of their patients and coined the term antidepressant to describe its action.[151] A similar incident took place in Paris, where Jean Delay, head of psychiatry at Sainte-Anne Hospital, heard of this effect from his pulmonology colleagues at Cochin Hospital. In 1952 (before Lurie and Salzer), Delay, with the resident Jean-Francois Buisson, reported the positive effect of isoniazid on depressed patients.[152] For reasons unrelated to its efficacy, isoniazid as an antidepressant was soon overshadowed by the more toxic iproniazid,[151] although it remains a mainstay of tuberculosis treatment. The mode of antidepressant action of isoniazid is still unclear. It is speculated that its effect is due to the inhibition of diamine oxidase, coupled with a weak inhibition of monoamine oxidase A.[153]

Selikoff and Robitzek also experimented with another anti-tuberculosis drug, iproniazid; it showed a greater psychostimulant effect, but more pronounced toxicity.[154] Later, Jackson Smith, Gordon Kamman, George Crane, and Frank Ayd, described the psychiatric applications of iproniazid. Ernst Zeller found iproniazid to be a potent monoamine oxidase inhibitor.[155] Nevertheless, iproniazid remained relatively obscure until Nathan Kline, the influential and flamboyant head of research at Rockland State Hospital, began to popularize it in the medical and popular press as a "psychic energizer".[155][156] Roche put a significant marketing effort behind iproniazid, including promoting its off-label use for depression.[155] Its sales grew until it was recalled in 1961, due to reports of lethal hepatotoxicity.[155]

The antidepressant effect of a tricyclic, a three ringed compound, was first discovered in 1957 by Roland Kuhn in a Swiss psychiatric hospital. Antihistamine derivatives were used to treat surgical shock and later as neuroleptics. Although in 1955 reserpine was shown to be more effective than placebo in alleviating anxious depression, neuroleptics were being developed as sedatives and antipsychotics.[medical citation needed]

Attempting to improve the effectiveness of chlorpromazine, Kuhn – in conjunction with the Geigy Pharmaceutical Company – discovered the compound "G 22355", later renamed imipramine. Imipramine had a beneficial effect in patients with depression who showed mental and motor retardation. Kuhn described his new compound as a "thymoleptic" "taking hold of the emotions," in contrast with neuroleptics, "taking hold of the nerves" in 1955–56. These gradually became established, resulting in the patent and manufacture in the US in 1951 by Häfliger and SchinderA.[157]

Second generation antidepressants

Antidepressants became prescription drugs in the 1950s. It was estimated that no more than 50 to 100 individuals per million suffered from the kind of depression that these new drugs would treat, and pharmaceutical companies were not enthusiastic in marketing for this small market. Sales through the 1960s remained poor compared to the sales of tranquilizers,[158] which were being marketed for different uses.[159] Imipramine remained in common use and numerous successors were introduced. The use of monoamine oxidase inhibitors (MAOI) increased after the development and introduction of "reversible" forms affecting only the MAO-A subtype of inhibitors, making this drug safer to use.[159][160]

By the 1960s, it was thought that the mode of action of tricyclics was to inhibit norepinephrine reuptake. However, norepinephrine reuptake became associated with stimulating effects. Later tricyclics were thought to affect serotonin as proposed in 1969 by Carlsson and Lindqvist as well as Lapin and Oxenkrug.[medical citation needed]

Researchers began a process of rational drug design to isolate antihistamine-derived compounds that would selectively target these systems. The first such compound to be patented was zimelidine in 1971, while the first released clinically was indalpine. Fluoxetine was approved for commercial use by the US Food and Drug Administration (FDA) in 1988, becoming the first blockbuster SSRI. Fluoxetine was developed at Eli Lilly and Company in the early 1970s by Bryan Molloy, Klaus Schmiegel, David Wong and others.[161][162] SSRIs became known as "novel antidepressants" along with other newer drugs such as SNRIs and NRIs with various selective effects.[163]

St John's wort fell out of favor in most countries through the 19th and 20th centuries, except in Germany, where Hypericum extracts were eventually licensed, packaged and prescribed. Small-scale efficacy trials were carried out in the 1970s and 1980s, and attention grew in the 1990s following a meta-analysis.[164] It remains an over-the-counter drug (OTC) supplement in most countries. Research continues to investigate its active component hyperforin, and to further understand its mode of action.[165][166]

Society and culture

In the United Kingdom, the use of antidepressants increased by 234% in the 10 years up to 2002.[167] In the US a 2005 independent report stated that 11% of women and 5% of men in the non-institutionalized population (2002) take antidepressants.[168] A 1998 survey found that 67% of patients diagnosed with depression were prescribed an antidepressant.[169] A 2007 study suggested that 25% of Americans were overdiagnosed with depression, regardless of any medical intervention.[170] The findings were based on a national survey of 8,098 people.

A 2002 survey found that about 3.5% of all people in France were being prescribed antidepressants, compared to 1.7% in 1992, often for conditions other than depression and often not in line with authorizations or guidelines.[171] Between 1996 and 2004 in British Columbia, antidepressant use increased from 3.4% to 7.2% of the population.[172] Data from 1992 to 2001 from the Netherlands indicated an increasing rate of prescriptions of SSRIs, and an increasing duration of treatment.[173] Surveys indicate that antidepressant use, particularly of SSRIs, has increased rapidly in most developed countries, driven by an increased awareness of depression together with the availability and commercial promotion of new antidepressants.[174] Antidepressants are also increasingly used worldwide for non-depressive patients as studies continue to show the potential of immunomodulatory, analgesic and anti-inflammatory properties in antidepressants.[medical citation needed]

The choice of a particular antidepressant is reported to be based, in the absence of research evidence of differences in efficacy, on seeking to avoid certain side-effects, and taking into account comorbid (co-occurring) psychiatric disorders, specific clinical symptoms and prior treatment history.[175]

It is also reported that, despite equivocal evidence of a significant difference in efficacy between older and newer antidepressants, clinicians perceive the newer drugs, including SSRIs and SNRIs, to be more effective than the older drugs (tricyclics and MAOIs).[176] Currently, the most commonly prescribed antidepressants are selective serotonin reuptake inhibitors (SSRIs), even though a Cochrane systematic review found no major difference in efficacy between SSRIs and tricyclic antidepressants.[177] A survey in the UK found that male general physicians were more likely to prescribe antidepressants than female doctors.[178]

The number of antidepressants prescribed by the National Health Service (NHS) in the UK almost doubled during one decade, authorities reported in 2010. Furthermore the number increased sharply in 2009 when 39.1 million prescriptions were issued, compared to 20.1 million issued in 1999. Also, physicians issued 3.18 million more prescriptions in 2009 than in 2008. Health authorities believed the increase was partly linked to the recession. However, other reasons include a diagnosis improvement, a reduction of the stigma on mental ill-health, and more distress caused by the economic crisis. Furthermore, physicians' concern is that some people who exhibit milder symptoms of depression are being prescribed drugs unnecessarily due to the lack of other options including talk therapies, counseling and cognitive behavioral therapy. One more factor that may be increasing the consumption of antidepressants is the fact that these medications now are used for other conditions including social anxiety and post traumatic stress.[179]

The use of antidepressants in the US doubled over one decade, from 1996 to 2005. Antidepressant drugs were prescribed to 13 million in 1996 and to 27 million people by 2005. In 2008, more than 164 million prescriptions were written. During this period, patients were less likely to undergo psychotherapy.[180]

Most commonly prescribed

Structural formula of the SSRI escitalopram, in its free base form.

United States: The most commonly prescribed antidepressants in the US retail market in 2010[181] were:

Sertraline Zoloft SSRI 33,409,838
Citalopram Celexa SSRI 27,993,635
Fluoxetine Prozac SSRI 24,473,994
Escitalopram Lexapro SSRI 23,000,456
Trazodone Desyrel SARI 18,786,495
Duloxetine Cymbalta SNRI 14,591,949
Paroxetine Paxil SSRI 12,979,366
Amitriptyline Elavil TCA 12,611,254
Venlafaxine XR Effexor XR SNRI 7,603,949
Bupropion XL Wellbutrin NDRI 7,317,814
Mirtazapine Remeron TeCA 6,308,288
Venlafaxine ER Effexor SNRI 5,526,132
Bupropion SR NDRI 4,588,996
Desvenlafaxine Pristiq SNRI 3,412,354
Nortriptyline Sensoval TCA 3,210,476
Bupropion ER NDRI 3,132,327
Venlafaxine Effexor SNRI 2,980,525
Bupropion Wellbutrin XL NDRI 753,516

Germany: The most commonly prescribed antidepressant in Germany is reported to be (concentrated extracts of) Hypericum perforatum (St John's wort).[182]

Netherlands: In the Netherlands, paroxetine, marketed as Seroxat among generic preparations, is the most prescribed antidepressant, followed by the tricyclic antidepressant amitriptyline, citalopram and venlafaxine.[183]

MAOIs can be as effective as tricyclic antidepressants, although they generally are used less frequently because they have a higher incidence of dangerous side effects and interactions.

Litigation

  • 2012 GlaxoSmithKline 3 billion dollars (US) – Paxil, The company touted Paxil for off-label use in children and adolescents, despite data that failed to show it was effective for these age groups, – Wellbutrin for marketing its antidepressant for off-label uses, including weight loss, substance abuse and sexual dysfunction and the seizure drug Lamictal.[184]
  • 2012 Johnson & Johnson $1.6 and $2.2 billion Risperdal off-label marketing
  • 2012 Abbott Laboratories $698 million for off-label marketing Depakote for use in dementia patients who became agitated or aggressive, despite lack of evidence that the drug was effective for that use.[185]
  • 2009 Eli Lilly $1.4 billion for marketing Zyprexa for children and the elderly dementia patients, both off-label uses. It is only approved to treat two disorders, schizophrenia and bipolar disorder.[186]
  • 2010 AstraZeneca $520 million for off-label marketing of Seroquel.[187]
  • 2009 Pfizer $301 million for off-label marketing of Geodon.[188]
  • 2007 Bristol-Myers Squibb $515 million for off-label marketing of Abilify for children and adolescents, and geriatric patients suffering from dememtia.[189]
  • 2004 Pfizer $430 million for off-label marketing of Neurontin.[190]
  • Ely Lilly In one of the only three cases to ever go to trial for SSRI indication in suicide, Eli Lilly and Company was caught corrupting the judicial process by making a deal with the plaintiff's attorney to throw the case, in part by not disclosing damaging evidence to the jury. The case, known as the Fentress Case, involved a Kentucky man, Joseph Wesbecker, on Prozac, who went to his workplace and opened fire with an assault rifle, killing 8 people (including Fentress), and injuring 12 others before turning the gun on himself. The jury returned a 9-to-3 verdict in favor of Lilly. The judge, in the end, took the matter to the Kentucky Supreme Court, which found that "there was a serious lack of candor with the trial court and there may have been deception, bad faith conduct, abuse of judicial process and, perhaps even fraud." The judge later revoked the verdict and instead recorded the case as settled. The value of the secret settlement deal has never been disclosed, but was reportedly "tremendous".[191]

Publication of research findings

  • Ghost writing of studies for industry-sponsored drug trials is common. Of all 44 trials approved between 1994 and 1995 by the ethics committees of Copenhagen and Frederiksberg in Denmark, up to 91%, that is 40 of the 44 trials, has some form of ghostwriting, and for the most part, the ghost was a statistician.[192] Cases relating to gabapentin,[193] paroxetine,[194] sertraline,[195] fenfluramine/phentermine (fen-phen)[196] are well documented, while many others, relating to olanzapine, quetiapine, remain under seal by the courts.[medical citation needed]
  • Publication bias. Trials for which results were unfavorable were less likely to be published. Published data suggest a favorable risk-benefit profile for some SSRIs; however, addition of unpublished data indicates that risks could outweigh benefits of these drugs to treat depression in children and young people.[197] Of 90 drugs approved by the FDA between 1998 and 2000, trials that did not show statistically significant results were 34% less likely to have been published;[198] on the other hand, positive results are often published more than once. Based on Healy's examination of the data produced from the Cochrane study of Olanzapine for schizophrenia, the four initial trials of Zyprexa gave rise to 234 publications, most of which were ghost written.[199]
  • Lack of access to raw data, data suppression, misrepresentation, and manipulation has eroded value of trial results. Demands on access to data has been met by resistance on the part of industry.[200]
  • A meta-analysis by UK, US and Canadian researchers was published in 2008, surveying all pharmaceutical company-sponsored drug trials on the six most widely prescribed new-generation antidepressants submitted for approval to the FDA between 1987 and 1999. The results showed that the difference in efficacy between antidepressants and placebo was minimal, but that it increased from virtually no difference at moderate levels of initial depression to a relatively small difference for patients with very severe depression. The difference reached conventional criteria for clinical significance for patients at the upper end of the very severely depressed category, due to a reduction in the efficacy of placebo.[34] The study received widespread media coverage in some countries, but was met with criticism from the professional community.[201]
Eli Lilly and Company responded by highlighting that the study did not take into account more recent studies on its product, Prozac, and that it was proud of the difference Prozac has made to millions of people. GlaxoSmithKline warned that this one study should not be used to cause unnecessary alarm and concern for patients. Two leading UK psychiatrists/pharmacologists, with financial and professional links to pharmaceutical companies, argued that short-term approval trials are not very suitable for evaluating effectiveness, that the unpublished trials are of poorer quality, that the meta-analysis authors came from a "psychology background" rather than drug testing background, and that the media and "elements of the medico/scientific community [sic]" have "a down on antidepressants" and that the media do not appreciate the seriousness of depression, and blame and stigmatize sufferers.[202] Wyeth pointed out that the data were good enough for FDA approval of the drugs.[203]

Social Science Perspective

In looking at the issue of antidepressant use, some academics have highlighted the need to examine the use of antidepressants and other medical treatments in cross cultural terms, due to the fact that often various cultures prescribe and observe different manifestations, symptoms, meanings and associations of depression and other medical conditions within their populations.[204][205] These cross-cultural discrepancies, it has been argued, then have implications on the perceived efficacy and use of antidepressants and other strategies in the treatment of depression in these different cultures.[204][205] This is exemplified in the work of Stefan Ecks, who asserts that in India antidepressants are largely seen as tools to combat marginality, promising the individual the ability to re-integrate themself into society through their use, a view and association not observed in the West.[204]

See also

References

  1. ^ Bodkin JA, Zornberg GL, Lukas SE, Cole JO (1995). "Buprenorphine Treatment of Refractory Depression". Journal of Clinical Psychopharmacology. 15 (1): 49–57. doi:10.1097/00004714-199502000-00008. PMID 7714228.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  2. ^ Ghadirian AM, Murphy BE, Gendron MJ (1998). "Efficacy of light versus tryptophan therapy in seasonal affective disorder". J Affect Disord. 50 (1): 23–7. doi:10.1016/S0165-0327(98)00053-6. PMID 9716275.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  3. ^ Wheeler Vega JA, Mortimer AM, Tyson PJ (2003). "Conventional antipsychotic prescription in unipolar depression, I: an audit and recommendations for practice". J Clin Psychiatry. 64 (5): 568–74. doi:10.4088/JCP.v64n0512. PMID 12755661.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  4. ^ Linde K, Berner MM, Kriston L (2008). "St John's wort for major depression". Cochrane Database Syst Rev (4): CD000448. doi:10.1002/14651858.CD000448.pub3. PMID 18843608.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  5. ^ Hamilton M (1960). "A rating scale for depression". J. Neurol. Neurosurg. Psychiatr. 23: 56–62. doi:10.1136/jnnp.23.1.56. PMC 495331. PMID 14399272.
  6. ^ Khan A, Faucett J, Lichtenberg P, Kirsch I, Brown WA (2012). "A systematic review of comparative efficacy of treatments and controls for depression". PLoS ONE. 7 (7): e41778. Bibcode:2012PLoSO...741778K. doi:10.1371/journal.pone.0041778. PMC 3408478. PMID 22860015.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  7. ^ Gartlehner G, Hansen RA, Morgan LC, Thaler K, Lux L, Van Noord M, Mager U, Thieda P, Gaynes BN, Wilkins T, Strobelberger M, Lloyd S, Reichenpfader U, Lohr KN (2011). "Comparative Benefits and Harms of Second-Generation Antidepressants for Treating Major Depressive Disorder: An Updated Meta-analysis". Annals of Internal Medicine. 155 (11): 772–85. doi:10.7326/0003-4819-155-11-201112060-00009. PMID 22147715.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  8. ^ "Sequenced Treatment Alternatives to Relieve Depression (STAR*D) Study". National Institute of Mental Health. Archived from the original on 5 March 2013. Retrieved 28 November 2012. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  9. ^ Fava M, Rush AJ, Wisniewski SR, Nierenberg AA, Alpert JE, McGrath PJ, Thase ME, Warden D, Biggs M, Luther JF, Niederehe G, Ritz L, Trivedi MH (2006). "A Comparison of Mirtazapine and Nortriptyline Following Two Consecutive Failed Medication Treatments for Depressed Outpatients: A STAR*D Report". The American Journal of Psychiatry. 163 (7): 1161–72. doi:10.1176/appi.ajp.163.7.1161. PMID 16816220. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)CS1 maint: multiple names: authors list (link)
  10. ^ a b Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F, Warden D, Ritz L, Nierenberg AA, Lebowitz BD, Biggs MM, Luther JF, Shores-Wilson K, Rush AJ (2006). "Medication Augmentation after the Failure of SSRIs for Depression". New England Journal of Medicine. 354 (12): 1243–52. doi:10.1056/NEJMoa052964. PMID 16554526. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)CS1 maint: multiple names: authors list (link)
  11. ^ Trivedi MH, Rush AJ, Wisniewski SR, Nierenberg AA, Warden D, Ritz L, Norquist G, Howland RH, Lebowitz B, McGrath PJ, Shores-Wilson K, Biggs MM, Balasubramani GK, Fava M (2006). "Evaluation of Outcomes with Citalopram for Depression Using Measurement-Based Care in STAR*D: Implications for Clinical Practice". American Journal of Psychiatry. 163 (1): 28–40. doi:10.1176/appi.ajp.163.1.28. PMID 16390886. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)CS1 maint: multiple names: authors list (link)
  12. ^ Rush AJ, Trivedi MH, Wisniewski SR, Stewart JW, Nierenberg AA, Thase ME, Ritz L, Biggs MM, Warden D, Luther JF, Shores-Wilson K, Niederehe G, Fava M (2006). "Bupropion-SR, Sertraline, or Venlafaxine-XR after Failure of SSRIs for Depression". New England Journal of Medicine. 354 (12): 1231–42. doi:10.1056/NEJMoa052963. PMID 16554525. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)CS1 maint: multiple names: authors list (link)
  13. ^ Warden D, Rush AJ, Trivedi MH, Fava M, Wisniewski SR (2007). "The STAR*D Project results: A comprehensive review of findings". Current psychiatry reports. 9 (6): 449–59. doi:10.1007/s11920-007-0061-3. PMID 18221624.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  14. ^ Taylor MJ, Freemantle N, Geddes JR, Bhagwagar Z (2006). "Early Onset of Selective Serotonin Reuptake Inhibitor Antidepressant Action: Systematic Review and Meta-analysis". Archives of General Psychiatry. 63 (11): 1217–23. doi:10.1001/archpsyc.63.11.1217. PMC 2211759. PMID 17088502.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  15. ^ "Depression". National Institute for Health and Clinical Excellence. December 2004. Archived from the original on 15 November 2008. Retrieved 20 March 2013. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  16. ^ "Practice guideline for the treatment of patients with major depressive disorder". National Guideline Clearinghouse. 2010. Archived from the original on 28 October 2008. Retrieved 20 March 2013. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  17. ^ Baghai TC, Möller HJ, Rupprecht R (2006). "Recent Progress in Pharmacological and Non-Pharmacological Treatment Options of Major Depression". Current Pharmaceutical Design. 12 (4): 503–15. doi:10.2174/138161206775474422. PMID 16472142.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  18. ^ a b Ruhé HG, Huyser J, Swinkels JA, Schene AH (2006). "Switching Antidepressants After a First Selective Serotonin Reuptake Inhibitor in Major Depressive Disorder". The Journal of Clinical Psychiatry. 67 (12): 1836–55. doi:10.4088/JCP.v67n1203. PMID 17194261.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  19. ^ Tranter R, O'Donovan C, Chandarana P, Kennedy S (2002). "Prevalence and outcome of partial remission in depression". Journal of psychiatry & neuroscience. 27 (4): 241–7. PMC 161658. PMID 12174733.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  20. ^ Byrne SE, Rothschild AJ (1998). "Loss of Antidepressant Efficacy During Maintenance Therapy". The Journal of Clinical Psychiatry. 59 (6): 279–88. doi:10.4088/JCP.v59n0602. PMID 9671339.
  21. ^ Mischoulon D, Nierenberg AA, Kizilbash L, Rosenbaum JF, Fava M (2000). "Strategies for managing depression refractory to selective serotonin reuptake inhibitor treatment: A survey of clinicians". Canadian journal of psychiatry. 45 (5): 476–81. PMID 10900529.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  22. ^ Bschor T, Baethge C (2010). "No evidence for switching the antidepressant: Systematic review and meta-analysis of RCTs of a common therapeutic strategy". Acta Psychiatrica Scandinavica. 121 (3): 174–9. doi:10.1111/j.1600-0447.2009.01458.x. PMID 19703121.
  23. ^ DeBattista C, Lembke A (2005). "Update on augmentation of antidepressant response in resistant depression". Current Psychiatry Reports. 7 (6): 435–40. doi:10.1007/s11920-005-0064-x. PMID 16318821.
  24. ^ Lam RW, Wan DD, Cohen NL, Kennedy SH (2002). "Combining Antidepressants for Treatment-Resistant Depression". The Journal of Clinical Psychiatry. 63 (8): 685–93. doi:10.4088/JCP.v63n0805. PMID 12197448.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  25. ^ Chouinard G, Chouinard VA (2008). "Atypical Antipsychotics: CATIE Study, Drug-Induced Movement Disorder and Resulting Iatrogenic Psychiatric-Like Symptoms, Supersensitivity Rebound Psychosis and Withdrawal Discontinuation Syndromes". Psychotherapy and Psychosomatics. 77 (2): 69–77. doi:10.1159/000112883. PMID 18230939.
  26. ^ Geddes, JR; Carney, SM; Davies, C; Furukawa, TA; Kupfer, DJ; Frank, E; Goodwin, GM (2003). "Relapse prevention with antidepressant drug treatment in depressive disorders: A systematic review". The Lancet. 361 (9358): 653–61. doi:10.1016/S0140-6736(03)12599-8. PMID 12606176.
  27. ^ Gardarsdottir, H; Van Geffen, ECG; Stolker, JJ; Egberts, TCG; Heerdink, ER (2009). "Does the Length of the First Antidepressant Treatment Episode Influence Risk and Time to a Second Episode?". Journal of Clinical Psychopharmacology. 29 (1): 69–72. doi:10.1097/JCP.0b013e31819302b1. PMID 19142111.
  28. ^ a b Fava, Giovanni A.; Offidani, Emanuela (2011). "The mechanisms of tolerance in antidepressant action". Progress in Neuro-Psychopharmacology and Biological Psychiatry. 35 (7): 1593–602. doi:10.1016/j.pnpbp.2010.07.026. PMID 20728491.
  29. ^ Fava, GA; Park, SK; Sonino, N (2006). "Treatment of recurrent depression". Expert Review of Neurotherapeutics. 6 (11): 1735–40. doi:10.1586/14737175.6.11.1735. PMID 17144786.
  30. ^ Petersen, TJ (2006). "Enhancing the efficacy of antidepressants with psychotherapy". Journal of Psychopharmacology. 20 (3 suppl): 19–28. doi:10.1177/1359786806064314. PMID 16644768.
  31. ^ Goldberg, JF; Truman, CJ (2003). "Antidepressant-induced mania: An overview of current controversies". Bipolar Disorders. 5 (6): 407–20. doi:10.1046/j.1399-5618.2003.00067.x. PMID 14636364.
  32. ^ a b Kirsch, I; Moore, TJ; Scoboria, A; Nicholls, SS (2002). "The emperor's new drugs: An analysis of antidepressant medication data submitted to the U.S. Food and Drug Administration". Prevention & Treatment. 5. doi:10.1037/1522-3736.5.1.523a.
  33. ^ "Study: Antidepressant barely better than placebo". USA Today. 7 July 2002. Retrieved 6 November 2008.
  34. ^ a b c Kirsch, I; Deacon, BJ; Huedo-Medina, TB; Scoboria, A; Moore, TJ; Johnson, BT (2008). "Initial Severity and Antidepressant Benefits: A Meta-Analysis of Data Submitted to the Food and Drug Administration". PLoS Medicine. 5 (2): e45. doi:10.1371/journal.pmed.0050045. PMC 2253608. PMID 18303940.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  35. ^ "Study: Antidepressant barely better than placebo". USA Today. 7 July 2002. Retrieved 6 November 2008.[unreliable medical source?]
  36. ^ Langreth, Robert (5 January 2010). "Study Undermines Case for Antidepressants". Forbes. Archived from the original on 3 December 2013. Retrieved 1 July 2010.
  37. ^ Walsh, BT; Seidman, SN; Sysko, R; Gould, M (2002). "Placebo Response in Studies of Major Depression: Variable, Substantial, and Growing". JAMA. 287 (14): 1840–7. doi:10.1001/jama.287.14.1840. PMID 11939870.
  38. ^ Leucht, C; Huhn, M; Leucht, S (2012). Leucht, C (ed.). "Amitriptyline versus placebo for major depressive disorder". Cochrane Database of Systematic Reviews. 12: CD009138. doi:10.1002/14651858.CD009138.pub2. PMID 23235671.
  39. ^ "Depression in Adults (update)" (PDF). National Collaborating Centre for Mental Health Commissioned by the National Institute for Health and Clinical Excellence. www.nice.org.uk. pp. 282–292. Archived from the original (PDF) on 12 June 2013. Retrieved 20 November 2013.
  40. ^ "CDC: Antidepressants most prescribed drugs in U.S". CNN. 9 July 2007. Archived from the original on 11 December 2013. Retrieved 21 May 2011.
  41. ^ "'Remembering' prior antidepressant use affects the brain's response to new drugs". Yahoo! India News. ANI. 27 March 2012. Archived from the original on 4 December 2013.
  42. ^ Gueorguieva, R; Mallinckrodt, C; Krystal, JH (2011). "Trajectories of Depression Severity in Clinical Trials of Duloxetine: Insights into Antidepressant and Placebo Responses". Archives of General Psychiatry. 68 (12): 1227–37. doi:10.1001/archgenpsychiatry.2011.132. PMC 3339151. PMID 22147842.
  43. ^ a b Turner, EH; Matthews, AM; Linardatos, E; Tell, RA; Rosenthal, R (2008). "Selective Publication of Antidepressant Trials and Its Influence on Apparent Efficacy". New England Journal of Medicine. 358 (3): 252–60. doi:10.1056/NEJMsa065779. PMID 18199864.
  44. ^ a b c d e f g h i j Brunton, L; Chabner, B; Knollman, B (2010). Goodman and Gilman's The Pharmacological Basis of Therapeutics (12th ed.). New York: McGraw-Hill Professional. ISBN 978-0-07-162442-8.{{cite book}}: CS1 maint: multiple names: authors list (link)
  45. ^ a b c d e f g h i j k "Side effects of antidepressant medications". UpToDate. Wolters Kluwer Health. Archived from the original on 2 November 2013. Retrieved 24 October 2013.
  46. ^ a b c d e f g h i j k l m n o Royal Pharmaceutical Society of Great Britain. MARTINDALE – The Complete Drug Reference. Pharmaceutical Press. Archived from the original on 26 July 2013. Retrieved 31 October 2013.
  47. ^ a b c Cipriani A, Furukawa TA, Salanti G, Geddes JR, Higgins JP, Churchill R, Watanabe N, Nakagawa A, Omori IM, McGuire H, Tansella M, Barbui C (2009). "Comparative efficacy and acceptability of 12 new-generation antidepressants: a multiple-treatments meta-analysis". Lancet. 373 (9665): 746–58. doi:10.1016/S0140-6736(09)60046-5. PMID 19185342.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  48. ^ a b c d e f g h i j k l Taylor, D; Paton, C; Shitij, K (2012). The Maudsley prescribing guidelines in psychiatry. West Sussex: Wiley-Blackwell. ISBN 978-0-470-97948-8.{{cite book}}: CS1 maint: multiple names: authors list (link)
  49. ^ a b White N, Litovitz T, Clancy C (2008). "Suicidal antidepressant overdoses: a comparative analysis by antidepressant type". J Med Toxicol. 4 (4): 238–50. doi:10.1007/BF03161207. PMC 3550116. PMID 19031375.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  50. ^ a b c d e f g h Rossi, S, ed. (2013). Australian Medicines Handbook (2013 ed.). Adelaide: The Australian Medicines Handbook Unit Trust. ISBN 978-0-9805790-9-3.
  51. ^ a b van Moffaert, M; de Wilde, J; Vereecken, A; Dierick, M; Evrard, JL; Wilmotte, J; Mendlewicz, J (March 1995). "Mirtazapine is more effective than trazodone: a double-blind controlled study in hospitalized patients with major depression". Int Clin Psychopharmacol. 10 (1): 3?9. doi:10.1097/00004850-199503000-00001. PMID 7622801.
  52. ^ "AMOXAPINE tablet [Watson Laboratories, Inc.]". DailyMed. Watson Laboratories, Inc. August 2010. Archived from the original on 2 November 2013. Retrieved 30 October 2013.
  53. ^ a b Walker, R; Whittlesea, C, ed. (2007) [1994]. Clinical Pharmacy and Therapeutics (4th ed.). Edinburgh: Churchill Livingstone Elsevier. ISBN 978-0-7020-4293-5.{{cite book}}: CS1 maint: multiple names: editors list (link)
  54. ^ Bruijn JA, Moleman P, Mulder PG, van den Broek WW, van Hulst AM, van der Mast RC, van de Wetering BJ (1996). "A double-blind, fixed blood-level study comparing mirtazapine with imipramine in depressed in-patients". Psychopharmacology (Berl.). 127 (3): 231–7. doi:10.1007/BF02246131. PMID 8912401. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  55. ^ Bruijn JA, Moleman P, Mulder PG, van den Broek WW (1999). "Depressed in-patients respond differently to imipramine and mirtazapine". Pharmacopsychiatry. 32 (3): 87–92. doi:10.1055/s-2007-979200. PMID 10463374.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  56. ^ a b Fishback JA, Robson MJ, Xu YT, Matsumoto RR (2010). "Sigma receptors: potential targets for a new class of antidepressant drug". Pharmacol. Ther. 127 (3): 271–82. doi:10.1016/j.pharmthera.2010.04.003. PMC 3993947. PMID 20438757.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  57. ^ a b Kishimoto A, Todani A, Miura J, Kitagaki T, Hashimoto K (2010). "The opposite effects of fluvoxamine and sertraline in the treatment of psychotic major depression: a case report". Ann Gen Psychiatry. 9: 23. doi:10.1186/1744-859X-9-23. PMC 2881105. PMID 20492642.{{cite journal}}: CS1 maint: multiple names: authors list (link) CS1 maint: unflagged free DOI (link)
  58. ^ Borkowska A, Pilaczyńska E, Araszkiewicz A, Rybakowski J (2002). "[The effect of sertraline on cognitive functions in patients with obsessive-compulsive disorder]". Psychiatr. Pol. (in Polish). 36 (6 Suppl): 289–95. PMID 12647451.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  59. ^ Schmitt JA, Ramaekers JG, Kruizinga MJ, van Boxtel MP, Vuurman EF, Riedel WJ (2002). "Additional dopamine reuptake inhibition attenuates vigilance impairment induced by serotonin reuptake inhibition in man". J. Psychopharmacol. (Oxford). 16 (3): 207–14. doi:10.1177/026988110201600303. PMID 12236626.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  60. ^ Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. ISBN 978-0-85711-084-8.
  61. ^ Goodwin GM (2009). "Clinical studies on the efficacy of agomelatine on depressive symptoms". CNS Drugs. 23 Suppl 2: 35–9. doi:10.2165/11318650-000000000-00000. PMID 19708724.
  62. ^ Benazzi F (1997). "Antidepressant-associated hypomania in outpatient depression: a 203-case study in private practice". J Affect Disord. 46 (1): 73–7. doi:10.1016/S0165-0327(97)00082-7. PMID 9387089.
  63. ^ Birmes P, Coppin D, Schmitt L, Lauque D (2003). "Serotonin syndrome: a brief review". CMAJ. 168 (11): 1439–42. PMC 155963. PMID 12771076.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  64. ^ Boyer EW, Shannon M (2005). "The serotonin syndrome" (PDF). N. Engl. J. Med. 352 (11): 1112–20. doi:10.1056/NEJMra041867. PMID 15784664 (PDF). {{cite journal}}: |archive-url= requires |archive-date= (help)
  65. ^ Mason PJ, Morris VA, Balcezak TJ (2000). "Serotonin syndrome. Presentation of 2 cases and review of the literature". Medicine (Baltimore). 79 (4): 201–9. doi:10.1097/00005792-200007000-00001. PMID 10941349. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  66. ^ Sampson E, Warner JP (1999). "Serotonin syndrome: potentially fatal but difficult to recognize". Br J Gen Pract. 49 (448): 867–8. doi:10.1097/00005792-200007000-00001. PMC 1313553. PMID 10818648. {{cite journal}}: Unknown parameter |month= ignored (help)
  67. ^ Sathyanarayana Rao TS, Yeragani VK (2009). "Hypertensive crisis and cheese". Indian J Psychiatry. 51 (1): 65–6. doi:10.4103/0019-5545.44910. PMC 2738414. PMID 19742203.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  68. ^ Paykel ES (1995). "Clinical efficacy of reversible and selective inhibitors of monoamine oxidase A in major depression". Acta Psychiatr Scand Suppl. 386: 22–7. doi:10.1111/j.1600-0447.1995.tb05920.x. PMID 7717091.
  69. ^ "FDA Advising of Risk of Birth Defects with Paxil" (Press release). U.S. Food and Drug Administration. Archived from the original on 3 December 2013. Retrieved 29 November 2012.
  70. ^ Huang, Hsiang. "A Meta-analysis of the Relationship between Anti-depressant Use in Pregnancy and the Risk of Preterm Birth and Low Birth Weight." General Hospital Psychiarty 36.1 (2014): 13-18. Science Direct. 01 Jan. 2014. Web. 20 Apr. 2014. <http://www.sciencedirect.com/science/article/pii/S0163834313002399>.
  71. ^ Nakhai-Pour, HR; Broy, P; Bérard, A (2010). "Use of antidepressants during pregnancy and the risk of spontaneous abortion". Canadian Medical Association Journal. 182 (10): 1031–7. doi:10.1503/cmaj.091208. PMC 2900326. PMID 20513781.
  72. ^ Louik, C; Lin, AE; Werler, MM; Hernández-Díaz, Sonia; Mitchell, Allen A. (2007). "First-Trimester Use of Selective Serotonin-Reuptake Inhibitors and the Risk of Birth Defects". New England Journal of Medicine. 356 (26): 2675–83. doi:10.1056/NEJMoa067407. PMID 17596601.
  73. ^ Pedersen, L H.; Henriksen, T B.; Olsen, J (2010). "Fetal Exposure to Antidepressants and Normal Milestone Development at 6 and 19 Months of Age". Pediatrics. 125 (3): e600–8. doi:10.1542/peds.2008-3655. PMID 20176667.
  74. ^ Lanza Di Scalea, T; Wisner, KL (2009). "Antidepressant Medication Use During Breastfeeding". Clinical Obstetrics and Gynecology. 52 (3): 483–97. doi:10.1097/GRF.0b013e3181b52bd6. PMC 2902256. PMID 19661763.
  75. ^ Sivagnanam, G (2012). "Antidepressants". Journal of Pharmacology and Pharmacotherapeutics. 3 (3): 287–8.
  76. ^ Cox GR, Callahan P, Churchill R, Hunot V, Merry SN, Parker AG, Hetrick SE (2012). "Psychological therapies versus antidepressant medication, alone and in combination for depression in children and adolescents". Cochrane Database Syst Rev. 11: CD008324. doi:10.1002/14651858.CD008324.pub2. PMID 23152255.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  77. ^ Friedman RA, Leon AC (2007). "Expanding the black box - depression, antidepressants, and the risk of suicide". N. Engl. J. Med. 356 (23): 2343–6. doi:10.1056/NEJMp078015. PMID 17485726.
  78. ^ Potenza, edited by Jon E. Grant, Marc N. (2012). The Oxford handbook of impulse control disorders. Oxford: Oxford University Press. ISBN 978-0-19-538971-5. {{cite book}}: |first= has generic name (help)CS1 maint: multiple names: authors list (link)
  79. ^ Csoka AB, Csoka A, Bahrick A, Mehtonen OP (2008). "Persistent sexual dysfunction after discontinuation of selective serotonin reuptake inhibitors". J Sex Med. 5 (1): 227–33. doi:10.1111/j.1743-6109.2007.00630.x. PMID 18173768.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  80. ^ Montejo AL, Llorca G, Izquierdo JA, Rico-Villademoros F (2001). "Incidence of sexual dysfunction associated with antidepressant agents: a prospective multicenter study of 1022 outpatients. Spanish Working Group for the Study of Psychotropic-Related Sexual Dysfunction". J Clin Psychiatry. 62 Suppl 3: 10–21. PMID 11229449.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  81. ^ Serretti A, Chiesa A (2009). "Treatment-emergent sexual dysfunction related to antidepressants: a meta-analysis". J Clin Psychopharmacol. 29 (3): 259–66. doi:10.1097/JCP.0b013e3181a5233f. PMID 19440080.
  82. ^ Chebili S, Abaoub A, Mezouane B, Le Goff JF (1998). "[Antidepressants and sexual stimulation: the correlation]". Encephale (in French). 24 (3): 180–4. PMID 9696909.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  83. ^ Keltner, Norman L.; McAfee, Kelly M.; Taylor, Carey L. (2009). "Biological Perspectives". Perspectives in Psychiatric Care. 38 (3): 111–6. doi:10.1111/j.1744-6163.2002.tb00665.x. PMID 12385082.
  84. ^ Ozmenler, NK; Karlidere, T; Bozkurt, A; Yetkin, S; Doruk, A; Sutcigil, L; Cansever, A; Uzun, O; Ozgen, F; Ozsahin, A (2008). "Mirtazapine augmentation in depressed patients with sexual dysfunction due to selective serotonin reuptake inhibitors". Human Psychopharmacology: Clinical and Experimental. 23 (4): 321. doi:10.1002/hup.929. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)
  85. ^ Labbate, LA; Grimes, JB; Hines, A; Pollack, MH (December 1997). "Bupropion treatment of serotonin reuptake antidepressant-associated sexual dysfunction". Annals of clinical psychiatry : official journal of the American Academy of Clinical Psychiatrists. 9 (4): 241–5. doi:10.3109/10401239709147804. PMID 9511948.
  86. ^ Ott, GE; Rao, U; Lin, KM; Gertsik, L; Poland, RE (September 2004). "Effect of treatment with bupropion on EEG sleep: relationship to antidepressant response". The international journal of neuropsychopharmacology / official scientific journal of the Collegium Internationale Neuropsychopharmacologicum (CINP). 7 (3): 275–81. doi:10.1017/S1461145704004298. PMID 15122973.
  87. ^ Vertes, Robert P.; Eastman, Kathleen E. (2000). "The case against memory consolidation in REM sleep". Behavioral and Brain Sciences. 23 (6): 867–76, discussion 904–1121. doi:10.1017/S0140525X00004003. PMID 11515146.
  88. ^ "Mirtazapine Regulates Stress Hormones, Improves Sleep In Depressed Patients: Presented at ISP". Pharmaclub.com.[unreliable medical source?]
  89. ^ Cohen, RM; Pickar, D; Garnett, D; Lipper, S; Gillin, JC; Murphy, DL (1982). "REM sleep suppression induced by selective monoamine oxidase inhibitors". Psychopharmacology. 78 (2): 137–40. doi:10.1007/bf00432251. PMID 6817370.
  90. ^ Stimmel, GL; Dopheide, JA; Stahl, SM (1997). "Mirtazapine: An antidepressant with noradrenergic and specific serotonergic effects". Pharmacotherapy. 17 (1): 10–21. PMID 9017762.
  91. ^ "mirtazapine (Rx) – Remeron, Remeron SolTab". Medscape. WebMD. Archived from the original on 29 October 2013. Retrieved 19 November 2013.
  92. ^ Papakostas, GI (2008). "Tolerability of modern antidepressants". J Clin Psychiatry. 69 (Suppl E1): 8–13. PMID 18494538.
  93. ^ Li Z, Maglione M, Tu W, Mojica W, Arterburn D, Shugarman LR, Hilton L, Suttorp M, Solomon V, Shekelle PG, Morton SC (April 2005). "Meta-analysis: pharmacologic treatment of obesity". Ann. Intern. Med. 142 (7): 532–46. doi:10.7326/0003-4819-142-7-200504050-00012. PMID 15809465.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  94. ^ "Effexor Medicines Data Sheet". Wyeth Pharmaceuticals Inc. 2006. Archived from the original on 17 September 2006. Retrieved 17 September 2006.
  95. ^ a b Tamam, L; Ozpoyraz, N (2002). "Selective serotonin reuptake inhibitor discontinuation syndrome: A review". Advances in Therapy. 19 (1): 17–26. doi:10.1007/BF02850015. PMID 12008858.
  96. ^ Kramer, JC; Klein, DF; Fink, M (1961). "Withdrawal symptoms followingdicontin of imipramine therapy". The American Journal of Psychiatry. 118: 549–50. PMID 14459296.
  97. ^ a b Haddad, Peter M. (2001). "Antidepressant Discontinuation Syndromes". Drug Safety. 24 (3): 183–97. doi:10.2165/00002018-200124030-00003. PMID 11347722.
  98. ^ Parker, G; Blennerhassett, J (1998). "Withdrawal reactions associated with venlafaxine". Aust N Z J Psychiatry. 32 (2): 291–4. doi:10.3109/00048679809062742. PMID 9588310.
  99. ^ Nielsen, Margrethe; Hansen, Ebba Holme; Gøtzsche, Peter C. (2012). "What is the difference between dependence and withdrawal reactions? A comparison of benzodiazepines and selective serotonin re-uptake inhibitors". Addiction. 107 (5): 900–8. doi:10.1111/j.1360-0443.2011.03686.x. PMID 21992148.
  100. ^ Medawar, C (2004). Medicines out of Control. The Netherlands: Aksant.[page needed]
  101. ^ a b "Depressive Disorders". Merck Manual Online. Archived from the original on 9 January 2014. Retrieved 9 January 2014. Continued therapy with an antidepressant for 6 to 12 mo (up to 2 yr in patients > 50) is usually needed to prevent relapse. Most antidepressants, especially SSRIs, should be tapered off (by decreasing the dose by about 25%/wk) rather than stopped abruptly; stopping SSRIs abruptly may result in discontinuationsyndrome [sic] (nausea, chills, muscles aches, dizziness, anxiety, irritability, insomnia, fatigue). The likelihood and severity of withdrawal varies inversely with the half-life of the SSRI.[failed verification] [dead link]
  102. ^ a b Baboolal, Nelleen S. (2004). "Venlafaxine Withdrawal Syndrome". Journal of Clinical Psychopharmacology. 24 (2): 229–31. doi:10.1097/01.jcp.0000117427.05703.f2. PMID 15206672.
  103. ^ Reeves, RR; Mack, JE; Beddingfield, JJ (2003). "Shock-Like Sensations During Venlafaxine Withdrawal". Pharmacotherapy. 23 (5): 678–81. doi:10.1592/phco.23.5.678.32198. PMID 12741444.
  104. ^ Valuck, Robert J.; Orton, Heather D.; Libby, Anne M. (2009). "Antidepressant Discontinuation and Risk of Suicide Attempt". The Journal of Clinical Psychiatry. 70 (8): 1069–77. doi:10.4088/JCP.08m04943. PMID 19758520.
  105. ^ "Antidepressants and Addiction". Biopsychiatry.com. Archived from the original on 2 May 2012. Retrieved 30 November 2012.
  106. ^ Fava, Giovanni A.; Bernardi, Manuela; Tomba, Elena; Rafanelli, Chiara (2007). "Effects of gradual discontinuation of selective serotonin reuptake inhibitors in panic disorder with agoraphobia". The International Journal of Neuropsychopharmacology. 10 (6): 835–8. doi:10.1017/S1461145706007462. PMID 17224089.
  107. ^ Fava, M; Mulroy, R; Alpert, J; Nierenberg, AA; Rosenbaum, JF (1997). "Emergence of adverse events following discontinuation of treatment with extended-release venlafaxine". The American Journal of Psychiatry. 154 (12): 1760–2. PMID 9396960.
  108. ^ Parker, Gordon; Blennerhassett, Jenny (1998). "Withdrawal reactions associated with venlafaxine". Australian and New Zealand Journal of Psychiatry. 32 (2): 291–4. doi:10.3109/00048679809062742. PMID 9588310.
  109. ^ Van Noorden, MS; Vergouwen, AC; Koerselman, GF (2002). "Delirium during withdrawal of venlafaxine". Nederlands tijdschrift voor geneeskunde. 146 (26): 1236–7. PMID 12132141.
  110. ^ Nissen, C; Feige, B; Nofzinger, E; Riemann, D; Berger, M; Voderholzer, U (2005). "Transient narcolepsy-cataplexy syndrome after discontinuation of the antidepressant venlafaxine". Journal of Sleep Research. 14 (2): 207–8. doi:10.1111/j.1365-2869.2005.00447.x. PMID 15910521.
  111. ^ "Paxil Progress". Paxilprogress.org. Archived from the original on 8 February 2014. Retrieved 27 November 2012.
  112. ^ Reardon, Sara. "Antidepressants in water trigger autism genes in fish."NewScientist. N.p., 7 June 2012. Web. 28 Feb. 2014. <http://www.newscientist.com/article/dn21882-antidepressants-in-water-trigger-autism-genes-in-fish.html#.UxCThr9dX9I>.
  113. ^ G. Nentwig Effects of pharmaceuticals on aquatic invertebrates. Part II: The antidepressant drug fluoxetine Arch. Environ. Con. Tox., 52 (2007), pp. 163–170
  114. ^ LeuT-Desipramine Structure Reveals How Antidepressants Block Neurotransmitter Reuptake Zheng Zhou, Juan Zhen, Nathan K. Karpowich, Regina M. Goetz, Christopher J. Law,Maarten E. A. Reith, and Da-Neng Wang 7 September 2007: 317 (5843), 1390–1393.Published online 9 August 2007[DOI:10.1126/science.1147614]
  115. ^ Antidepressants in Aquatic Organisms: A Wide Range of Effects Peter P. Fong Pharmaceuticals and Care Products in the Environment. July 30, 2001, 264–281
  116. ^ B.W. Brooks, C.K. Chambliss, J.K. Stanley, A. Ramirez, K.E. Banks, R.D. Johnson, R.J. Lewis Determination of select antidepressants in fish from an effluent-dominated stream Environ. Toxicol. Chem., 24 (2005), pp. 464–469
  117. ^ Fent K (2006). Ecotoxicology of human pharmaceuticals. Aquatic toxicology. 76(2), 122–159.
  118. ^ S. Winberg, C.G. Carter, J.D. McCarthy, Z.Y. He, G.E. Nilsson, D.F. Houlihan Feeding rank and brain serotonergic activity in rainbow trout Onchorhynchus mykiss J. Exp. Biol., 179 (1993), pp. 197–211
  119. ^ Perreault, H. A. N., Semsar, K., & Godwin, J. (2003). Fluoxetine treatment decreases territorial aggression in a coral reef fish. Physiology & Behavior, 79(4–5), 719–724.
  120. ^ R. Huber, K. Smith, A. Delago, K. Isaksson, E.A. Kravitz Serotonin and aggressive motivation in crustaceans: altering the decision to retreat Proc. Natl. Acad. Sci. U.S.A., 94 (1997), pp. 5939–5942
  121. ^ Di Poi, C., Darmaillacq, A., Dickel, L., Boulouard, M., & Bellanger, C. (2013). Effects of perinatal exposure to waterborne fluoxetine on memory processing in the cuttlefish sepia officinalis. Aquatic Toxicology, 132–133(0), 84–91.
  122. ^ a b Maes, M; Yirmyia, R; Noraberg, J; Brene, S; Hibbeln, J; Perini, G; Kubera, M; Bob, P; Lerer, B; Maj, M (March 2009). "The inflammatory & neurodegenerative (I&ND) hypothesis of depression: leads for future research and new drug developments in depression". Metabolic Brain Disease. 24 (1): 27–53. doi:10.1007/s11011-008-9118-1. PMID 19085093.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  123. ^ a b c Sanacora, G; Treccani, G; Popoli, M (January 2012). "Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders" (PDF). Neuropharmacology. 62 (1): 63–77. doi:10.1016/j.neuropharm.2011.07.036. PMID 21827775.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  124. ^ Menke A, Klengel T, Binder EB (2012). "Epigenetics, depression and antidepressant treatment". Current Pharmaceutical Design. 18 (36): 5879–5889. doi:10.2174/138161212803523590. PMID 22681167.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  125. ^ Vialou, V; Feng, J; Robison, AJ; Nestler, EJ (January 2013). "Epigenetic mechanisms of depression and antidepressant action". Annual Review of Pharmacology and Toxicology. 53 (1): 59–87. doi:10.1146/annurev-pharmtox-010611-134540. PMID 23020296.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  126. ^ Martins-de-Souza, Daniel. "Blood Mononuclear Cell Proteome Suggests Integrin and Ras Signaling as Critical Pathways for Antidepressant Treatment Response." Rev. of Society of Biological Psychiatry. 06 Feb. 2014: E1-E3. Print.
  127. ^ "Depressive Disorders". Merck Manual. Archived from the original on 5 December 2013. Retrieved 30 November 2012.
  128. ^ Orr, Katy; Taylor, David (2007). "Psychostimulants in the Treatment of Depression". CNS Drugs. 21 (3): 239–57. doi:10.2165/00023210-200721030-00004. PMID 17338594.
  129. ^ a b Mineur, Yann S.; Picciotto, Marina R. (2010). "Nicotine receptors and depression: Revisiting and revising the cholinergic hypothesis". Trends in Pharmacological Sciences. 31 (12): 580–6. doi:10.1016/j.tips.2010.09.004. PMC 2991594. PMID 20965579.
  130. ^ a b c Taylor, D; Carol, P; Shitij, K (2012). The Maudsley prescribing guidelines in psychiatry. West Sussex: Wiley-Blackwell. ISBN 978-0-470-97969-3.
  131. ^ Australian Medicines Handbook 2013. Adelaide: Australian Medicines Handbook Pty Ltd. 2013. ISBN 978-0-9805790-9-3.
  132. ^ Bauer, M; Dopfmer, S (October 1999). "Lithium augmentation in treatment-resistant depression: meta-analysis of placebo-controlled studies". Journal of Clinical Psychopharmacology. 19 (5): 427–434. doi:10.1097/00004714-199910000-00006. PMID 10505584.{{cite journal}}: CS1 maint: date and year (link)
  133. ^ Aronson, R; Offman, HJ; Joffe, RT; Naylor, CD (September 1996). "Triiodothyronine augmentation in the treatment of refractory depression. A meta-analysis". Archives of General Psychiatry. 53 (9): 842–848. doi:10.1001/archpsyc.1996.01830090090013. PMID 8792761.{{cite journal}}: CS1 maint: date and year (link)
  134. ^ Ischaki, E; Gratziou, C (2009). "Smoking and depression: Is smoking cessation effective?". Therapeutic Advances in Respiratory Disease. 3 (1): 31–8. doi:10.1177/1753465809102662. PMID 19293201.
  135. ^ Benowitz, Neal L. (2009). "Pharmacology of Nicotine: Addiction, Smoking-Induced Disease, and Therapeutics". Annual Review of Pharmacology and Toxicology. 49: 57–71. doi:10.1146/annurev.pharmtox.48.113006.094742. PMC 2946180. PMID 18834313.
  136. ^ Lara, Diogo R. (2010). "Caffeine, Mental Health, and Psychiatric Disorders". Journal of Alzheimer's Disease. 20 (Suppl 1): S239–48. doi:10.3233/JAD-2010-1378. PMID 20164571. {{cite journal}}: Unknown parameter |doi_brokendate= ignored (|doi-broken-date= suggested) (help) [dead link]
  137. ^ Rush, A John (2013). "Ketamine for Treatment-Resistant Depression: Ready or Not for Clinical Use?". American Journal of Psychiatry. 170 (10): 1079–81. doi:10.1176/appi.ajp.2013.13081034. ISSN 0002-953X. PMID 23982324.
  138. ^ Diazgranados, N (August 2010). "A Randomized Add-on Trial of an N-methyl-d-aspartate Antagonist in Treatment-Resistant Bipolar Depression". Archives of General Psychiatry. 67 (8): 793–802. doi:10.1001/archgenpsychiatry.2010.90. PMC 3000408. PMID 20679587. {{cite journal}}: |first10= missing |last10= (help); |first11= missing |last11= (help); |first12= missing |last12= (help); |first13= missing |last13= (help); |first2= missing |last2= (help); |first3= missing |last3= (help); |first4= missing |last4= (help); |first5= missing |last5= (help); |first6= missing |last6= (help); |first7= missing |last7= (help); |first8= missing |last8= (help); |first9= missing |last9= (help)
  139. ^ Autry AE, Adachi M, Nosyreva E, Na ES, Los MF, Cheng PF, Kavalali ET, Monteggia LM (7 July 2011). "NMDA receptor blockade at rest triggers rapid behavioural antidepressant responses". Nature. 475 (7354): 91–95. doi:10.1038/nature10130. PMC 3172695. PMID 21677641. Archived from the original on 23 June 2011.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  140. ^ Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, Krystal JH (2000). "Antidepressant effects of ketamine in depressed patients". Biol. Psychiatry. 47 (4): 351–4. doi:10.1001/archpsyc.63.8.856. PMID 10686270. {{cite journal}}: Unknown parameter |month= ignored (help)CS1 maint: multiple names: authors list (link)
  141. ^ Koike H, Iijima M, Chaki S (2011). "Involvement of AMPA receptor in both the rapid and sustained antidepressant-like effects of ketamine in animal models of depression". Behav. Brain Res. 224 (1): 107–11. doi:10.1016/j.bbr.2011.05.035. PMID 21669235.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  142. ^ "Faster-Acting Antidepressants Closer to Becoming a Reality". NIMH. 25 July 2007. Archived from the original on 10 April 2013.
  143. ^ Nemets H, Nemets B, Apter A, Bracha Z, Belmaker RH (2006). "Omega-3 treatment of childhood depression: a controlled, double-blind pilot study". Am J Psychiatry. 163 (6): 1098–100. doi:10.1176/appi.ajp.163.6.1098. PMID 16741212.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  144. ^ Sontrop J, Campbell MK (2006). "Omega-3 polyunsaturated fatty acids and depression: a review of the evidence and a methodological critique". Prev Med. 42 (1): 4–13. doi:10.1016/j.ypmed.2005.11.005. PMID 16337677.
  145. ^ Rocha Araujo, Daniele Marano; Vilarim, Marina Machado; Nardi, Antonio E (2010). "What is the effectiveness of the use of polyunsaturated fatty acid omega-3 in the treatment of depression?". Expert Review of Neurotherapeutics. 10 (7): 1117–29. doi:10.1586/ern.10.77. PMID 20586692.
  146. ^ Montgomery, Paul; Richardson, Alexandra J (2008). Montgomery, Paul (ed.). "Omega-3 fatty acids for bipolar disorder". Cochrane Database of Systematic Reviews (2): CD005169. doi:10.1002/14651858.CD005169.pub2. PMID 18425912.
  147. ^ Weber, MM; Emrich, HM (1988). "Current and Historical Concepts of Opiate Treatment in Psychiatric Disorders". International Clinical Psychopharmacology. 3 (3): 255–66. doi:10.1097/00004850-198807000-00007. PMID 3153713.
  148. ^ Czygan, Franz-C. (2003). "Kulturgeschichte und Mystik des Johanniskrauts: Vom 2500 Jahre alten Apotropaikum zum aktuellen Antidepressivum". Pharmazie in unserer Zeit (in German). 32 (3): 184–90. doi:10.1002/pauz.200390062. PMID 12784538. {{cite journal}}: Unknown parameter |trans_title= ignored (|trans-title= suggested) (help)
  149. ^ Selikoff, Irving J.; Robitzek, EH (1952). "Tuberculosis Chemotherapy with Hydrazine Derivatives of Isonicotinic Acid". CHEST Journal. 21 (4): 385–438. doi:10.1378/chest.21.4.385. PMID 14906149.
  150. ^ a b Healy, D (2001). "The Antidepressant Drama". In Weissman, M.M. (ed.). The treatment of depression: bridging the 21st century. American Psychiatric Pub. pp. 10–11. ISBN 978-0-88048-397-1. Retrieved 28 May 2009. {{cite book}}: External link in |chapterurl= (help); Unknown parameter |chapterurl= ignored (|chapter-url= suggested) (help)
  151. ^ Healy, David (1996). The psychopharmacologists: interviews. London: Chapman and Hall. p. 8. ISBN 978-1-86036-008-4.
  152. ^ Healy, David (1998). The Psychopharmacologists: Volume 2. A Hodder Arnold Publication. pp. 132–4. ISBN 978-1-86036-010-7.
  153. ^ Robitzek, EH; Selikoff, IJ; Mamlok, E; Tendlau, A (1953). "Isoniazid and Its Isopropyl Derivative in the Therapy of Tuberculosis in Humans: Comparative Therapeutic and Toxicologic Properties". CHEST Journal. 23 (1): 1–15. doi:10.1378/chest.23.1.1. PMID 12998444.
  154. ^ a b c d López-Muñoz, Francisco; Alamo, Cecilio; Juckel, Georg; Assion, Hans-Jörg (2007). "Half a Century of Antidepressant Drugs". Journal of Clinical Psychopharmacology. 27 (6): 555–9. doi:10.1097/jcp.0b013e3181bb617. PMID 18004120.
  155. ^ "Psychic Energizer". Time. 15 April 1957. Archived from the original on 11 August 2013. Retrieved 28 May 2009.
  156. ^ Kuhn, R (1958). "The treatment of depressive states with G 22355 (imipramine hydrochloride)". The American Journal of Psychiatry. 115 (5): 459–64. PMID 13583250.
  157. ^ "Tranquilizers". Cumberland Moutain Community Services. www.cmcsb.com. Archived from the original on 16 September 2012. Retrieved 20 November 2013.[unreliable medical source?]
  158. ^ a b Healy, David (1999). "The Three Faces of the Antidepressants: A Critical Commentary on the Clinical-Economic Context of Diagnosis". The Journal of Nervous & Mental Disease. 187 (3): 174–80. doi:10.1097/00005053-199903000-00007. PMID 10086474.
  159. ^ Pletscher, A (1991). "The discovery of antidepressants: A winding path". Experientia. 47 (1): 4–8. doi:10.1007/BF02041242. PMID 1999242.
  160. ^ Domino, EF (1999). "History of modern psychopharmacology: A personal view with an emphasis on antidepressants". Psychosomatic Medicine. 61 (5): 591–8. PMID 10511010.
  161. ^ Wong, DT; Bymaster, FP; Horng, JS; Molloy, BB (1975). "A new selective inhibitor for uptake of serotonin into synaptosomes of rat brain: 3-(p-trifluoromethylphenoxy). N-methyl-3-phenylpropylamine". The Journal of Pharmacology and Experimental Therapeutics. 193 (3): 804–11. PMID 1151730.
  162. ^ Freeman, H (1996). "Tolerability and safety of novel antidepressants". European Psychiatry. 11: 206s. doi:10.1016/0924-9338(96)88597-X.
  163. ^ Linde, K; Ramirez, G; Mulrow, CD; Pauls, A; Weidenhammer, W; Melchart, D (1996). "St John's wort for depression—an overview and meta-analysis of randomised clinical trials". BMJ. 313 (7052): 253–8. doi:10.1136/bmj.313.7052.253. PMC 2351679. PMID 8704532.
  164. ^ Müller, W (2003). "Current St. John's wort research from mode of action to clinical efficacy". Pharmacological Research. 47 (2): 101–9. doi:10.1016/S1043-6618(02)00266-9. PMID 12543057.
  165. ^ Nathan, P J. (2001). "Hypericum perforatum (St John's Wort): A non-selective reuptake inhibitor? A review of the recent advances in its pharmacology". Journal of Psychopharmacology. 15 (1): 47–54. doi:10.1177/026988110101500109. PMID 11277608.
  166. ^ (National Institute for Clinical Excellence, 2004)
  167. ^ Stagnitti, M (2005) [study done in 2002]. "Antidepressant Use in the US Civilian Non-Institutionalised Population". US Department of Health and Human Services. Rockville(MD): Medical Expenditure Panel, Agency for Healthcare Research and Quality. Archived from the original on 5 December 2013.
  168. ^ Sleath, B; Tina Shih, Ya-Chen (2003). "Sociological influences on antidepressant prescribing". Social Science & Medicine. 56 (6): 1335–44. doi:10.1016/S0277-9536(02)00132-6. PMID 12600369.
  169. ^ "Depression could be overdiagnosed". HeraldNet. The Washington Post. April 2007. Archived from the original on 3 December 2013. Retrieved 28 May 2009.[unreliable medical source?]
  170. ^ Olié, JP; Elomari, F; Spadone, C; Lépine, JP (2002). "Résultats d'une enquête sur l'usage des antidépresseurs en population générale française". L'Encephale (in French). 28 (5 Pt 1): 411–7. PMID 12386542. {{cite journal}}: Unknown parameter |trans_title= ignored (|trans-title= suggested) (help)
  171. ^ Raymond, CB; Morgan, SG; Caetano, PA (2007). "Antidepressant Utilization in British Columbia from 1996 to 2004: Increasing Prevalence but Not Incidence". Psychiatric Services. 58 (1): 79–84. doi:10.1176/appi.ps.58.1.79. PMID 17215416.
  172. ^ Meijer, WEE; Heerdink, ER; Leufkens, HGM; Herings, RMC; Egberts, ACG; Nolen, WA (2004). "Incidence and determinants of long-term use of antidepressants". European Journal of Clinical Pharmacology. 60 (1): 57–61. doi:10.1007/s00228-004-0726-3. PMID 14985889.
  173. ^ McManus, Peter; Mant, Andrea; Mitchell, Philip B; Montgomery, William S; Marley, John; Auland, Merran E (2000). "Recent trends in the use of antidepressant drugs in Australia, 1990–1998". The Medical journal of Australia. 173 (9): 458–61. PMID 11149300. Archived from the original on 9 September 2011.
  174. ^ Zimmerman, M; Posternak, M; Friedman, M; Attiullah, N; Baymiller, S; Boland, R; Berlowitz, S; Rahman, S; Uy, K; Singer, S (2004). "Which Factors Influence Psychiatrists' Selection of Antidepressants?". American Journal of Psychiatry. 161 (7): 1285–9. doi:10.1176/appi.ajp.161.7.1285. PMID 15229063. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)
  175. ^ Petersen, T; Dording, C; Neault, NB; Kornbluh, R; Alpert, JE; Nierenberg, AA; Rosenbaum, JF; Fava, M (2002). "A survey of prescribing practices in the treatment of depression". Progress in Neuro-Psychopharmacology and Biological Psychiatry. 26 (1): 177–87. doi:10.1016/S0278-5846(01)00250-0. PMID 11853110.
  176. ^ Barbui, C; Hotopf, M; Freemantle, N; Boynton, J; Churchill, R; Eccles, MP; Geddes, JR; Hardy, R; Lewis, G; Mason, JM (2000). Barbui, Corrado (ed.). "Treatment discontinuation with selective serotonin reuptake inhibitors (SSRIs) versus tricyclic antidepressants (TCAs)". Cochrane Database of Systematic Reviews (3): CD002791. doi:10.1002/14651858.CD002791. PMID 17636706. {{cite journal}}: Unknown parameter |displayauthors= ignored (|display-authors= suggested) (help)[unreliable medical source?]
  177. ^ "Male GPs depression pills 'bias'". BBC News. 21 July 2005. Archived from the original on 24 June 2006. Retrieved 29 May 2009.
  178. ^ Davis, Rowenna (11 June 2010). "Antidepressant Use Rises as Recession Feeds Wave of Worry". The Guardian. London. Archived from the original on 15 June 2010. Retrieved 1 July 2010. {{cite news}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  179. ^ "Antidepressant Use Doubles in U.S., Study Finds". Reuters. 4 August 2009. Archived from the original on 3 July 2010. Retrieved 1 July 2010. {{cite news}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  180. ^ "Top 200 generic drugs by units in 2010" (PDF). Archived from the original (PDF) on 15 December 2012."Top 200 brand drugs by units in 2010" (PDF). Archived from the original (PDF) on 22 April 2012.
  181. ^ Tyler, VE (1999). "Herbs Affecting the Central Nervous System". In Janick J (ed.). Perspectives on New Crops and New Uses. ASHS Press. p. 528. ISBN 978-0-9615027-0-6. Archived from the original on 27 April 2009. Retrieved 29 May 2009. {{cite book}}: External link in |chapterurl= (help); Unknown parameter |chapterurl= ignored (|chapter-url= suggested) (help); Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  182. ^ "GIPdatabank". Gipdatabank.nl. Archived from the original on 6 December 2008. Retrieved 6 November 2008. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  183. ^ "USDOJ: GlaxoSmithKline to Plead Guilty and Pay $3 Billion to Resolve Fraud Allegations and Failure to Report Safety Data". Justice.gov. 2 July 2012. Archived from the original on 19 April 2014. Retrieved 30 November 2012.
  184. ^ "USDOJ: Abbott Laboratories Sentenced for Misbranding Drug". Justice.gov. 2 October 2012. Archived from the original on 13 April 2014. Retrieved 30 November 2012.
  185. ^ "Pharmaceutical Company Eli Lilly To Pay Record $1.415 Billion For Off-label Drug Marketing" (PDF). United States Department of Justice. www.justice.gov. 15 January 2009. Archived from the original (PDF) on 30 August 2010. Retrieved 20 November 2013. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  186. ^ "Pharmaceutical Giant AstraZeneca to Pay $520 Million for Off-label Drug Marketing". Justice.gov. 27 April 2010. Archived from the original on 13 April 2014. Retrieved 30 November 2012.
  187. ^ "Pharmaceutical Company Pfizer, Inc. To Pay $301 Million For Off-label Drug Marketing" (PDF). United States Department of Justice. www.justice.gov. 2 September 2009. Archived from the original (PDF) on 20 November 2013. Retrieved 20 November 2013. {{cite web}}: Unknown parameter |deadurl= ignored (|url-status= suggested) (help)
  188. ^ "#07-782: 09–28–07 Bristol-Myers Squibb to Pay More Than $515 Million to Resolve Allegations of Illegal Drug Marketing and Pricing". Justice.gov. 28 September 2007. Archived from the original on 6 December 2013. Retrieved 30 November 2012.
  189. ^ "#322: 05–13–04 WARNER-LAMBERT TO PAY $430 MILLION TO RESOLVE CRIMINAL & CIVIL HEALTH CARE LIABILITY RELATING TO OFF-LABEL PROMOTION". Justice.gov. 13 May 2004. Archived from the original on 13 July 2013. Retrieved 30 November 2012.
  190. ^ Zitrin, Richard; Langford, Carol M. (1999). "Hide and Secrets in Louisville". The Moral Compass of the American Lawyer. Ballantine Books.[page needed]
  191. ^ Gøtzsche, PC; Hróbjartsson, A; Johansen, HK; Haahr, MT; Altman, DG; Chan, An-Wen (2007). "Ghost Authorship in Industry-Initiated Randomised Trials". PLoS Medicine. 4 (1): e19. doi:10.1371/journal.pmed.0040019. PMC 1769411. PMID 17227134.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  192. ^ Steinman, Michael A.; Bero, Lisa A.; Chren, Mary–Margaret; Landefeld, C Seth (2006). "Narrative Review: The Promotion of Gabapentin: An Analysis of Internal Industry Documents". Annals of Internal Medicine. 145 (4): 284–93. doi:10.7326/0003-4819-145-4-200608150-00008. PMID 16908919.
  193. ^ Healy, David; Cattell, Dinah (2003). "Interface between authorship, industry and science in the domain of therapeutics". The British Journal of Psychiatry. 183: 22–7. doi:10.1192/bjp.02-235. PMID 12835239. {{cite journal}}: Unknown parameter |doi_brokendate= ignored (|doi-broken-date= suggested) (help); Unknown parameter |subscription= ignored (|url-access= suggested) (help)
  194. ^ McHenry, L; Jureidini, J (2008). "Industry-Sponsored Ghostwriting in Clinical Trial Reporting: A Case Study". Accountability in Research. 15 (3): 152–67. doi:10.1080/08989620802194384. PMID 18792536.
  195. ^ Mundy, A (2001). Dispensing with the truth: the victims, the drug companies, and the dramatic story behind the battle over Fen-Phen. New York: St. Martin's Press. ISBN 978-0-312-25324-0.[page needed]
  196. ^ Whittington, Craig J; Kendall, Tim; Fonagy, Peter; Cottrell, David; Cotgrove, Andrew; Boddington, Ellen (2004). "Selective serotonin reuptake inhibitors in childhood depression: Systematic review of published versus unpublished data". The Lancet. 363 (9418): 1341–5. doi:10.1016/S0140-6736(04)16043-1. PMID 15110490.
  197. ^ Lee, Kirby; Bacchetti, Peter; Sim, Ida (2008). Clarke, Mike (ed.). "Publication of Clinical Trials Supporting Successful New Drug Applications: A Literature Analysis". PLoS Medicine. 5 (9): e191. doi:10.1371/journal.pmed.0050191. PMC 2553819. PMID 18816163.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  198. ^ Duggan, L; Fenton, M; Dardennes, J; El-Dosoky, R; Indran, A; Indran, S (2005). Duggan, Lorna (ed.). "Olanzapine for schizophrenia". Cochrane Database of Systematic Reviews (2): CD001359. doi:10.1002/14651858.CD001359.pub2. PMID 10796640.
  199. ^ Bian, Zhao-Xiang; Wu, Tai-Xiang (2010). "Legislation for trial registration and data transparency". Trials. 11: 64. doi:10.1186/1745-6215-11-64. PMC 2882906. PMID 20504337.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  200. ^ "Antidepressants do they work? A Review of the Review". Doctors Lounge. 21 September 2009. Archived from the original on 14 February 2014. Retrieved 30 November 2012.[self-published source?]
  201. ^ Nutt, D; Malizia, A (2008). "Why does the world have such a 'down' on antidepressants?". Journal of Psychopharmacology. 22 (3): 223–6. doi:10.1177/0269881108091877. PMID 18541622.
  202. ^ Blue, L (26 February 2008). "Antidepressants Hardly Help". Time. Archived from the original on 26 August 2013.
  203. ^ a b c [ Ecks, S 2005, 'Pharmaceutical Citizenship: Antidepressant Marketing and the Promise of Demarginalization in India', Anthropology & Medicine, 12, 3, pp. 239-254]
  204. ^ a b [ Lock, M. & Nguyen, V. 2010, "Local Biologies and Human Difference" in An Anthropology of Biomedicine [electronic resource], 1st edn, Hoboken : Wiley, 2010, , pp. 83-109. ]

Additional reading

Template:Link GA Template:Link GA