5
 Escitalopram Treatment of Depression in Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome A Randomized, Double-Blind, Placebo-Controlled Study  Jacqueline Hoare, MD, FCPsych, Mphil,* P aul Carey , PhD, Þ  John A. Joska, MMed, FCPsych,*  Henri Carrar a, MPH,þ  Katherine Sorsdahl, PhD,* and Dan J. Stein, FCPsych, PhD* Abstract:  Depression can be a chronic and impairing illness in people with human immunod efici enc y virus (HIV)/acquired immunodeficiency syn- drome. Large randomized studies of newer selective serotonin reuptake in- hibitors such as escitalopram in the treatment of depression in HIV , examining comparative treatment efficacy and safety, have yet to be done in HIV-positive  patients. This was a f ixed-dose, pl acebo-controlled, randomized, double-blind study to inves tigate the efficac y of escital opram in HIV -serop ositiv e subject s with Diagnostic and Statistical Manual of Mental Disord ers, Fou rth Edition, major depressiv e disorde r. One hundred two particip ants were randomly assigned to either 10 mg of escitalopram or placebo for 6 weeks. An analysis of covar ianc e of the comple ter s found that ther e was no adv anta ge for escital opram over placebo on the Montgomery-As berg Depression Rating Sca le ( p  = 0.93). Sixty- two per cent res pond ed to esc ital opra m and 59% responded to placebo on the Clinical Global Impression Scale. Given the relatively high placebo response, future trials in this area need to be selective in partici pant recruitment and to be adequately power ed. Key Words:  Randomized controlled study, depression, HIV (  J Nerv Ment Dis 2014;202: 133  Y 137) D epression can be a chronic and impairing illness in people living with human immunodeficiency virus (HIV)/acquired immuno- deficiency syndrome (AIDS). Depression occurs in almost twice as many people with HIV when compared with the general population in the developed world (Ciesla and Roberts, 2001). In lower- and middle-income countries, indications are that depression affects up- ward of 30% of HIV clinic attendees (Olley et al., 2004). In a South African primary care context, identification of de-  pression was generally poor (Carey et al., 2003) and perhaps even worse in clinics focused on chronic medical illnesses including HIV (Evans et al., 1996). Later in the illness, overlap with HIV symptoms (Jaco bsbe rg and Perry, 1992 ) and side eff ects from antiretr oviral treatment in more advanced disease may serve to complicate diag- nosis (Kalichman et al., 2000). There is little doubt that a significant  proportion of patients with comorbid depression and HIV go unrecognized and untreated. Depression in HIV is associated with greater delays initiating antiretroviral treatment (Fairfield et al., 1999) and poorer adherence to anti retr ovi ral medi catio n (Gor dillo et al., 1999). Depr essio n is important not only because of its psychological impact but also be- cause of its impact on the course of HIV. Evid ence suggest s that depr essio n accel erates HIV disea se pro gressi on (Lese rman et al., 1999) and is independently associated with increased HIV mortality and a more rapid decline in CD4 + lymphocyte counts (Ickovics et al., 2001). Compliant selective serotonin reuptake inhibitor (SSRI) use has been associated with improved antiretroviral adherence and lab- oratory parameters (Horberg et al., 2008). It is possible that early and effective treatment of depression in HIV may significantly improve quality of life and that longer-term use may have an influence on HIV disease progression and associated morbidity. Expert consensus suggests that selective SSRIs are first-line tre atment s of dep res sio n in HI V (Caba lle ro and Nah ata , 2004) . SSRIs may be as effective as and better tolerated than tricyclic anti- depressants (TCAs) in HIV-positive adults (Caballero and Nahata, 2004). However, published controlled studies in this area are limited to trials with fluoxetine versus placebo (Rabkin et al., 1999) and  paroxetin e versus imipramin e (Elliott et al., 1998), with comparable response rates as in depressed cohorts without HIV . A recent meta- analysis of the use of SSRIs for depression in HIV argued that the lack of data prevented conclusions being drawn on the most useful SSRI s in this context (Ferra ndo and Fre yber g, 2008 ). Becau se of fluoxetine’s properties as an inhibitor of cytochrome P450 isoforms 2D6 and 3A4 and its long half-life, interactions with antiretrovirals may occur. Newer SSRIs such as citalopram and escitalopram offer a theoretically lower potential for drug-drug interactions, but large ran- domized studies examining comparative treatment efficacy and safety have yet to be done in HIV-positive patients. A recent meta-analysis of 12 new- gener ation antidepr essant drugs in non- HIV cohorts in the treatment of depression found, for both efficacy and acceptability, in favor of escitalopram and sertraline (Cipriani et al., 2009). Escitalopram, the most selective SSRI available (Owens et al., 2001), may offer a particular advantage for treating depression in HIV. Aside from the established efficacy of escitalopram in non-HIV dep res sion (Burk e et al., 2002; Lepol a et al., 200 3; Wade et al., 2002), escitalopram mediates a more rapid and robust treatment re- sponse than its predecessor citalopram (Gorman et al., 2002) and has only weak or negligible effects on hepatic enzymes crucial to the meta boli sm of most medicine s inclu ding those freq uent ly used in highly acti ve antiretroviral ther apy (Bro sen and Nar anjo, 2001; Gutierrez et al., 2003; von Moltke et al., 2001). This drug-drug inter- action remains relevant in South Africa because patients are still pre- scribed protease inhibitors (PIs). The South African antiretroviral roll out programme consists of a nonnucleoside reverse transcriptase inhibitor (NNRTI) based first-line regimen and a ritonavir-boosted  protease inhibitor (PI) containing second-line regimen. T en milli- grams of escitalopram per day is effective and well tolerated in the treatment of depression in primary care (Burke et al., 2002). Therefore, the present study aimed to investigate the efficacy and tolerability of escitalopram for the treatment of  Diagnostic and ORIGINAL ARTICLE The Journal of Nervous and Mental Disease  &  Volume 202, Number 2, February 2014 www.jonmd.com  133 *Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa; Department of Psychiatry, Faculty of Health Sciences, Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa; and School of Public Health and Family Medicine, Universi ty of Cape Town , Cape Town, South Africa. Send reprint requests to Jacqueline Hoare, MD, FCPsych, Mphil, Department of Psychiatry and Mental Health, University of Cape Town, Anzio Road Obser- vatory , 7925, Cape To wn, South Africa. E-mail:  [email protected]. Copyright  *  2014 by Lippincott Williams & Wilkins ISSN: 0022-3018/14/20202  Y 0133 DOI: 10.1097/NMD.0000000000000082 Copyright © 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Escitalopram Treatment of Depression in HIV and AIDS

Embed Size (px)

DESCRIPTION

Escitalopram Treatment of Depression in HIV AND AIDS.pdf

Citation preview

  • Escitalopram Treatment of Depression in HumanImmunodeficiency Virus/Acquired Immunodeficiency Syndrome

    A Randomized, Double-Blind, Placebo-Controlled Study

    Jacqueline Hoare, MD, FCPsych, Mphil,* Paul Carey, PhD, John A. Joska, MMed, FCPsych,*Henri Carrara, MPH, Katherine Sorsdahl, PhD,* and Dan J. Stein, FCPsych, PhD*

    Abstract: Depression can be a chronic and impairing illness in people withhuman immunodeficiency virus (HIV)/acquired immunodeficiency syn-drome. Large randomized studies of newer selective serotonin reuptake in-hibitors such as escitalopram in the treatment of depression in HIV, examiningcomparative treatment efficacy and safety, have yet to be done in HIV-positivepatients. This was a fixed-dose, placebo-controlled, randomized, double-blindstudy to investigate the efficacy of escitalopram in HIV-seropositive subjectswith Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition,major depressive disorder. One hundred two participants were randomlyassigned to either 10 mg of escitalopram or placebo for 6 weeks. An analysisof covariance of the completers found that there was no advantage forescitalopram over placebo on the Montgomery-Asberg Depression RatingScale ( p = 0.93). Sixty-two percent responded to escitalopram and 59%responded to placebo on the Clinical Global Impression Scale. Given therelatively high placebo response, future trials in this area need to be selectivein participant recruitment and to be adequately powered.

    Key Words: Randomized controlled study, depression, HIV

    (J Nerv Ment Dis 2014;202: 133Y137)

    Depression can be a chronic and impairing illness in people livingwith human immunodeficiency virus (HIV)/acquired immuno-deficiency syndrome (AIDS). Depression occurs in almost twice asmany people with HIV when compared with the general populationin the developed world (Ciesla and Roberts, 2001). In lower- andmiddle-income countries, indications are that depression affects up-ward of 30% of HIV clinic attendees (Olley et al., 2004).

    In a South African primary care context, identification of de-pression was generally poor (Carey et al., 2003) and perhaps evenworse in clinics focused on chronic medical illnesses including HIV(Evans et al., 1996). Later in the illness, overlap with HIV symptoms(Jacobsberg and Perry, 1992) and side effects from antiretroviraltreatment in more advanced disease may serve to complicate diag-nosis (Kalichman et al., 2000). There is little doubt that a significantproportion of patients with comorbid depression and HIV gounrecognized and untreated.

    Depression in HIV is associated with greater delays initiatingantiretroviral treatment (Fairfield et al., 1999) and poorer adherence

    to antiretroviral medication (Gordillo et al., 1999). Depression isimportant not only because of its psychological impact but also be-cause of its impact on the course of HIV. Evidence suggests thatdepression accelerates HIV disease progression (Leserman et al.,1999) and is independently associated with increased HIV mortalityand a more rapid decline in CD4+ lymphocyte counts (Ickovics et al.,2001). Compliant selective serotonin reuptake inhibitor (SSRI) usehas been associated with improved antiretroviral adherence and lab-oratory parameters (Horberg et al., 2008). It is possible that early andeffective treatment of depression in HIV may significantly improvequality of life and that longer-term use may have an influence on HIVdisease progression and associated morbidity.

    Expert consensus suggests that selective SSRIs are first-linetreatments of depression in HIV (Caballero and Nahata, 2004).SSRIs may be as effective as and better tolerated than tricyclic anti-depressants (TCAs) in HIV-positive adults (Caballero and Nahata,2004). However, published controlled studies in this area are limitedto trials with fluoxetine versus placebo (Rabkin et al., 1999) andparoxetine versus imipramine (Elliott et al., 1998), with comparableresponse rates as in depressed cohorts without HIV. A recent meta-analysis of the use of SSRIs for depression in HIV argued that thelack of data prevented conclusions being drawn on the most usefulSSRIs in this context (Ferrando and Freyberg, 2008). Because offluoxetines properties as an inhibitor of cytochrome P450 isoforms2D6 and 3A4 and its long half-life, interactions with antiretroviralsmay occur. Newer SSRIs such as citalopram and escitalopram offer atheoretically lower potential for drug-drug interactions, but large ran-domized studies examining comparative treatment efficacy and safetyhave yet to be done in HIV-positive patients. A recent meta-analysis of12 new-generation antidepressant drugs in non-HIV cohorts in thetreatment of depression found, for both efficacy and acceptability, infavor of escitalopram and sertraline (Cipriani et al., 2009).

    Escitalopram, the most selective SSRI available (Owens et al.,2001), may offer a particular advantage for treating depression inHIV. Aside from the established efficacy of escitalopram in non-HIVdepression (Burke et al., 2002; Lepola et al., 2003; Wade et al.,2002), escitalopram mediates a more rapid and robust treatment re-sponse than its predecessor citalopram (Gorman et al., 2002) andhas only weak or negligible effects on hepatic enzymes crucial to themetabolism of most medicines including those frequently used inhighly active antiretroviral therapy (Brosen and Naranjo, 2001;Gutierrez et al., 2003; von Moltke et al., 2001). This drug-drug inter-action remains relevant in South Africa because patients are still pre-scribed protease inhibitors (PIs). The South African antiretroviralroll out programme consists of a nonnucleoside reverse transcriptaseinhibitor (NNRTI) based first-line regimen and a ritonavir-boostedprotease inhibitor (PI) containing second-line regimen. Ten milli-grams of escitalopram per day is effective and well tolerated in thetreatment of depression in primary care (Burke et al., 2002).

    Therefore, the present study aimed to investigate the efficacyand tolerability of escitalopram for the treatment of Diagnostic and

    ORIGINAL ARTICLE

    The Journal of Nervous and Mental Disease & Volume 202, Number 2, February 2014 www.jonmd.com 133

    *Department of Psychiatry and Mental Health, University of Cape Town, CapeTown, South Africa; Department of Psychiatry, Faculty of Health Sciences,Stellenbosch University, Tygerberg Hospital, Cape Town, South Africa; andSchool of Public Health and Family Medicine, University of Cape Town,Cape Town, South Africa.

    Send reprint requests to Jacqueline Hoare, MD, FCPsych, Mphil, Department ofPsychiatry and Mental Health, University of Cape Town, Anzio Road Obser-vatory, 7925, Cape Town, South Africa. E-mail: [email protected].

    Copyright * 2014 by Lippincott Williams & WilkinsISSN: 0022-3018/14/20202Y0133DOI: 10.1097/NMD.0000000000000082

    Copyright 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

  • Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV ),major depressive disorder (MDD) in people with stage III HIVinfection.

    METHODSThis study was a fixed-dose, placebo-controlled, randomized,

    double-blind study to investigate the efficacy of escitalopram in HIV-seropositive subjects with DSM-IV MDD. Patients meeting inclusionand exclusion criteria were recruited from primary and secondarylevel clinics and then referred to Tygerberg Hospital, University ofStellenbosch South Africa, to participate in this study. Participantselection was a semirandom, convenience sample of consecutivelyscreened patients attending primary care antiretroviral therapy clinicsin Tygerberg Hospital and in the community clinics. Protocol wasapproved by the ethics committee of the University of Stellenbosch,and the patients signed informed written consent.

    ParticipantsEligible participants were HIV seropositive (World Health

    Organization stage III); ranged in age from 18 to 60 years; and metcriteria for a DSM-IV diagnosis of a current major depressive episodewithout psychosis, according to the Mini-international neuropsychi-atric interview-plus neuropsychiatric interview. Exclusion criteriaincluded patients who currently or in the last 6 months have fulfilledcriteria for DSM-IV alcohol or substance abuse/dependence, lifetimehistory of bipolar disorder, history of schizophrenia, or other psy-chotic disorders or dementia; patients presenting at screening with ev-idence of mild cognitive impairment and who were unable to score 10or higher on the HIV Dementia Scale and 23 or higher on the Mini-Mental State Examination (MMSE); and patients with a positiveurine screen for any prohibited substance (including amphetamines,barbiturates, cocaine, marijuana, methadone, opiates, phencyclidine, orpropoxyphene) or medication (benzodiazepines) at the screening visit.

    ProcedureThe participants received an initial screening and evaluation,

    including a review of medical and psychiatric history. This first visitwas followed by 7 days (T3) of single-blind placebo to exclude earlyplacebo responders and to monitor compliance. Thereafter, the sub-jects were randomized to treatment of 10 mg per day of eitherescitalopram or matching placebo for the full 6 weeks of the study.

    Clinical examination (including full systemic examination,weight, blood pressure, pulse, and electrocardiogram) and laboratorytests (full blood count, electrolytes, renal function, liver function,thyroid function tests, pregnancy screen, and urine drug screen) wereconducted at screening. HIV disease stage (CD4, CD8) was done atbaseline and at successful completion of the study as a final assess-ment, whereas efficacy measures were collected every week. Safetyand tolerability measures were done at weeks 1, 2, 4, and 6 by thestudy nurse. Adherence was monitored in dispensing packs weeklyand with patient report.

    Measures

    Primary Outcome MeasureMontgomery-Asberg depression rating scale

    The 10-item MADRS (Montgomery and Asberg, 1979) iswidely used and validated for assessing symptom severity in depres-sion. It has been shown to be a reliable measure to detect depression inHIV-positive subjects (Cockram et al., 1999) and has been found torespond robustly to effective pharmacotherapy in a number of condi-tions with neurovegetative symptoms, including Parkinsons disease(with a preponderance of subcortical cognitive decline; Leentjens et al.,2003, 2000), headache (Walker et al., 1998), and cognitive impairment(Gabryelewicz et al., 2004).

    Secondary Outcome MeasuresHospital anxiety and depression scale

    The Hospital Anxiety and Depression Scale (Zigmond andSnaith, 1983) is a reliable and well-validated self-report measure ofdepressive and anxiety symptom severity in medically ill populationsincluding HIV (Savard et al., 1998).

    Hamilton depression rating scaleThe Hamilton Depression Rating Scale (Hamilton, 1960) is an

    observer-rated 17-item measure of depression severity that reliablydifferentiates depressed from nondepressed subjects in HIV disease(Cockram et al., 1999) and other medical illnesses (Leentjens et al.,2000) and has been widely used in treatment studies of depression inHIV (Elliott et al., 1998; Rabkin et al., 1999; Zisook et al., 1998).

    Clinical global impression scale of severity and improvementThe Clinical Global Impression Scale of Severity (CGI-S) and

    Improvement (CGI-I; Guy, 1976) is a well-validated, clinician-ratedglobal assessment of disease severity (CGI-S) and improvement.CGI responder status is defined as a week 6 score of 2 (much im-proved) or 1 (very much improved) in the global assessment of im-provement (CGI-I) from baseline.

    Sheehan disability scaleThe Sheehan Disability Scale (SDS; Sheehan, 1983) is a

    patient-rated three-item visual analog scale for assessing impairmentin the domains of work, family, and social life.

    HIV dementia scaleThe HIV Dementia Scale (HDS; Berghuis et al., 1999; Power

    et al., 1995) is a brief and reliable measure of global cognitivefunction that is reliably able to identify early cognitive decline of asubcortical pattern.

    Mini-Mental State ExaminationThe MMSE (Folstein et al., 1975) is a widely used and vali-

    dated screening tool for cognitive impairment that was used in ad-dition to the HDS. The MMSE is a reliable brief screening tool usedto identify cognitive decline of a cortical pattern. The pattern of HIV-associated brain loss may be changing from a subcortical to a corticaldisease among HIV-positive patients (Cohen et al., 2010). Assess-ment of cognitive functioning on all subjects at screening was designedto exclude significant cognitive impairment that may negatively influ-ence the ability to comply with the study protocol.

    Blood was taken to measure CD4 and CD8 counts.

    AnalysisAll analyses were conducted using the Statistical Package for

    the Social Sciences and Stata version 11.1 (StataCorp LP, 4905Lakeway Dr, College Station, TX 77845). A sample size of 56 pa-tients per treatment group was required to detect a 3-point differenceon MADRS scores at week 6 between the treatment and placebogroups, with significance at 5% level, assuming a 20% dropout rate.An intention-to-treat analysis was not conducted because of the verylow dropout rate. The efficacy of escitalopram over placebo was testedusing an analysis of covariance (ANCOVA) model including thebaseline score as a covariate. CD4 counts were square root transformedto achieve a normal distribution, and all other outcome measures wereassessed to be normally distributed using the Shapiro-Wilks test. TheFriedmans test was conducted on theMMSE and the international HIVdementia scale (IHDS) because the data were not normally distribu-ted and could not be transformed to achieve a normal distribution.Treatment-emergent adverse events (hereafter referred to as adverseevents) were analyzed. The chi-square test was performed for the in-cidence of adverse events in the escitalopram group versus that in theplacebo group.

    Hoare et al. The Journal of Nervous and Mental Disease & Volume 202, Number 2, February 2014

    134 www.jonmd.com * 2014 Lippincott Williams & Wilkins

    Copyright 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

  • RESULTSA total of 105 participants, predominantly women (86%),

    participated in the study and were randomly assigned to either thetreatment or control groups. With three participants lost to follow-upin the first 2 weeks (in the treatment group), a total of 102 patientscompleted baseline and all weekly assessments (51 in the treatmentgroup and 51 in the control group). All participants included in thestudy were 100% adherent to treatment. The dropout rate was 2.9%.Reasons for loss to follow-up were unrelated to side effects, and allthree participants withdrew within the first 2 weeks of the first studyvisit. Both groups were well matched regarding the baseline demo-graphic characteristics and the mean group scores on the outcomemeasures used in the study, with the exception of the CGI severitymeasure, in which the mean for the treatment group (5.09) was sig-nificantly higher than that for the control group (4.74; Table 1).

    The results of the ANCOVA models are presented in Table 2.The primary efficacy analysis showed an adjusted mean change inMADRS total score from baseline to week 6 of j14 points in theescitalopram group andj13 points in the placebo group. The treatmentdifference was not statistically significant ( p = 0.93). Given that theMADRS was the primary outcome of interest, we further exploredthis variable using cross-sectional time series regression analysis toaccount for changes during the six visits. The results of this analysis didnot alter the conclusion that there was no statistically significant dif-ference according to treatment group ( p = 0.2). TheMADRS improvedover time in both the placebo and escitalopram groups, p G 0.001(see Fig. 1). Similar findings were also reported for changes in SDS( p 9 0.001) and HDS ( p 9 0.001) scores in both groups.

    In addition, after controlling for baseline scores, there was nosignificant effect of escitalopram on the remaining secondary out-come measures. Overall, 62% responded to escitalopram and 59%responded to placebo on the CGI. Furthermore, the proportion of theescitalopram-treated patients in complete remission (28%) was notstatistically significantly higher than that of the placebo-treated pa-tients at week 6 (35%; p = 0.47).

    Treatment-emergent side effects were relatively uncommon, andthe only one that differed between the treatment groups was nausea and

    vomiting. Nausea and vomiting was more frequently reported in thosein taking escitalopram than the placebo group ( p = 0.012).

    DISCUSSIONThis study found no statistically significant advantage for

    escitalopram over placebo in the treatment of depression in HIV. Ahigh placebo response (59%) was evident. There was also no sig-nificant effect of escitalopram on the remaining secondary outcomemeasures. Treatment-emergent side effects were relatively uncom-mon, with nausea and vomiting more frequently reported in thosetaking escitalopram than the placebo group. In terms of safety, thestudy did not find a negative effect of escitalopram on CD4 cellcounts. However CD4 cell counts did not decline significantly overtime, and no positive effects on CD4 cell count were observed.

    High placebo response rates have been reported in a numberof SSRI double-blind randomized controlled trials in HIV. In a pre-vious randomized, double-blind, placebo-controlled prospective trialexamining the efficacy of fluoxetine in treating depression in HIV-positive subjects, Rabkin et al. (1999), in an intent-to-treat analysis,found the difference between the fluoxetine and placebo groups inimproving depression to be nonsignificant. The failure of these dif-ferences to be ultimately significant was attributed to an unusuallyhigh placebo response. Elliott et al. (1998), in a randomized, blinded,placebo-controlled prospective trial comparing the efficacy of anSSRI, paroxetine, versus a TCA, imipramine, showed that althoughboth paroxetine and imipramine were effective through week 8 of thestudy, paroxetine was not superior to placebo at week 12; this findingmay be explained by an unusually high placebo response at week 12.A range of factors may be contributing to the placebo response, in-cluding sex, empathy and compassion of the clinical staff, setting of thetrial, frequency of the visits, and severity of symptoms (Stein et al.,2006). Of note is that the study participants in this trial were receivingcare that was significantly better than treatment as usual. Inadequateservices exist for the diagnosis, treatment, and management of mentalhealth problems in many clinics specializing in treatment of peopleliving with HIV. It is also notable that attrition of 2.9% was lower thanthat reported in other studies of HIV-positive patients. It is our clinical

    TABLE 1. Group Characteristics at Baseline

    Escitalopram Control

    Variable n n (%) n n (%) p

    Sex 0.22Male 51 4 (7.8) 51 8 (15.7)Female 51 47 (92.2) 51 43 (84.3)

    Age, median (range) 51 34 (23Y56) 51 34 (24Y54) 0.82Race 51 51 0.26Black 35 (68.6) 28 (54.9)White 1 (1.96) 0 (0)Colored 12 (23.53) 21 (41.2)Indian 1 (1.96) 0 (0)

    MADRS, mean (SD) 51 31.73 (4.74) 51 30.11 (5.58) 0.12HAM-D, mean (SD) 51 21.33 (5.20) 51 19.64 (5.51) 0.11MMSE, median (range) 51 29 (23Y30) 50 29 (23Y30) 0.37IHDS, median (range) 51 11 (10Y12) 51 11.5 (10Y12) 0.55SDS total, mean (SD) 50 20.80 (6.27) 51 18.76 (6.86) 0.12CD4 count, median (range) 48 425.5 (144Y1072) 48 350.5 (3Y1144) 0.37CD8 count, mean (SD) 47 1278.64 (535.86) 47 1135.94 (526.69) 0.2CGI severity, mean (SD) 51 4.80 (0.80) 51 5.16 (0.73) 0.02*

    *Significant difference at baseline.HAM-D indicates 17-item Hamilton Depression Rating Scale.

    The Journal of Nervous and Mental Disease & Volume 202, Number 2, February 2014 Escitalopram in HIV Depression

    * 2014 Lippincott Williams & Wilkins www.jonmd.com 135

    Copyright 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

  • impression that the excellent supportive care given by the study nurse tothe participants was crucial in retaining participants in the study;however, possibly, it also responsible for the high placebo response rate.If placebo responses in clinical trials are due to frequent patient eval-uation and increased attention, then high placebo response rates un-derscore the power of the expectancy effect created by the relationshipwith the trial team and the therapeutic context (Stein et al., 2006). Areviewof the placebo response in escitalopram placebo-controlled trialsfound that a higher placebo response rate was predicted by decreasedbaseline disorder severity (Stein et al., 2006).

    Our study is noteworthy in so far as it is one of the firstconducted in a lower- and middle-income country that comprisespredominantly African women. The participants were recruited fromprimary and secondary level clinics specializing in treatment ofpeople living with HIV and were invited if they had a diagnosis ofdepression. Recent studies showed that HIV-positive women may bemore likely to be on antiretroviral therapy (antiretroviral therapy;Nattrass, 2006). For example, in South Africa, men who present forantiretroviral therapy are sicker on average than women, suggestingdelays by men in getting tested or care seeking once tested (Bekkeret al., 2006). A study of HIV-positive men in the Cape Town area foundthat additional barriers included the notion that clinics are places forwomen and HIV/AIDS is awomens issue (Kagee et al., 2011). In across-sectional study examining the correlates of depression in a publicsector antiretroviral program, most of the study participants were fe-male (75.7%; Pappin et al., 2012). Previous studies on depression inHIV have overrepresented gay/bisexual male participants and un-derrepresented female and African HIV-positive participants, pos-sibly because of the fact that most studies have been conducted inhigher-income countries. Factors that may contribute to depression

    in this HIV-infected population include stigmatization, lack of socialsupport, and the death of a loved one from HIV/AIDS. Individualsmay therefore respond well to a supportive intervention of the kindprovided by a clinical research trial.

    CONCLUSIONSFuture studies could focus on a longer treatment duration with

    escitalopram or be more selective in participant recruitment. In thiscase, a more selective approach would be to include only severe/persistent MDD.

    DISCLOSURESThis study was funded by Lundbeck. Lundbeck South Africa

    (SA) is a subsidiary of H. Lundbeck A/S, an international, Danish,research-based pharmaceutical company focusing on the centralnervous system. Jacqueline Hoare is funded by the Discovery Foun-dation. Dan J. Stein is funded by the Medical Research Council ofSouth Africa (MRC). John A. Joska is funded by the National Re-search Foundation of South Africa.

    The authors declare no conflict of interest.

    REFERENCESBekker LG, Myer L, Wood R (2006) Impact of HIV Infection on the Epidemi-

    ology of Tuberculosis in a Peri-Urban Community in South Africa: The Needfor Age-Specific Interventions. Clin Infect Dis. 42:1040Y1047.

    Berghuis JP, Uldall KK, Lalonde B (1999) Validity of two scales in identifyingHIV-associated dementia. J Acquir Immune Defic Syndr. 21:134Y140.

    Brosen K, Naranjo CA (2001) Review of pharmacokinetic and pharmacodynamicinteraction studies with citalopram. Eur Neuropsychopharmacol. 11:275Y283.

    Burke WJ, Gergel I, Bose A (2002) Fixed-dose trial of the single isomer SSRIescitalopram in depressed outpatients. J Clin Psychiatry. 63:331Y336.

    Caballero J, Nahata MC (2004) Use of selective serotonin-reuptake inhibitors inthe treatment of depression in adults with HIV. Ann Pharmacother. 30:1065.

    Carey PD, Stein DJ, Zungu-Dirwayi N, Seedat S (2003) Trauma and posttraumaticstress disorder in an urban Xhosa primary care population: Prevalence, comor-bidity, and service use patterns. J Nerv Ment Dis. 191:230Y236.

    Ciesla JA, Roberts JE (2001) Meta-analysis of the relationship between HIV infec-tion and risk for depressive disorders. Am J Psychiatry. 158:725Y730.

    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 antide-pressants: A multiple-treatments meta-analysis. Lancet. 373:746Y758.

    Cockram A, Judd FK, Mijch A, Norman T (1999) The evaluation of depression ininpatients with HIV disease. Aust N Z J Psychiatry. 33:344Y352.

    Cohen RA, Harezlak J, Schifitto G, Hana G, Clark U, Gongvatana A, Paul R,Taylor M, Thompson P, Alger J, Brown M, Zhong J, Campbell T, Singer E,

    TABLE 2. ANCOVA of Follow-up Scores as a Function of Treatment Baseline Scores as Covariate

    Placebo Escitalopram

    Outcome nBaseline,Mean (SD) n

    Follow-up,Mean (SD)

    Change FromBaseline n

    Baseline,Mean (SD) n

    Follow-up,Mean (SD)

    Change FromBaseline

    TreatmentDiff (SE) p

    MADRS 51 30.1 (5.6) 50 17.0 (10.5) j13.2 51 31.7 (4.7) 50 17.8 (9.5) j14 0.9 (2.0) 0.93HAM-D 51 19.6 (5.5) 50 11.8 (8.4) j8 51 21.3 (5.2) 48 12.3 (1.0) j9 1.1 (1.7) 0.98MMSEa 50 29 (23Y30) 43 29 (23Y30) 0 51 29 (23Y30) 44 29 (26Y30) 0 0.0 0.7IHDSa 51 11.5 (10Y12) 45 12 (9Y12) 0.5 51 11 (10Y12) 45 12 (10Y12) 1 0.5 0.86SDS total 51 18.8 (6.9) 49 13.9 (8.1) j4.6 50 20.8 (6.3) 51 12.84 (7.23) j8.1 3.5 (1.8) 0.3CD4 count 48 404.1 (218.5) 41 436.8 (210.6) 43.3b 48 454.3 (29.5) 37 433.6 (214.9) j10.7 54.0 (35.6) 0.2CD8 count 47 1135.9 (526.7) 41 1159.8 (486.1) j13c 47 1278.6 (535.9) 37 1205.8 (448.05) j54.6 41.5 (90.4) 0.96

    aNot normally distributed; therefore, median and range were reported.bComplete data on 39 patients.cComplete data on 38 patients.diff indicates difference.

    FIGURE 1. MADRS improvement over time in both placeboand treatment group.

    Hoare et al. The Journal of Nervous and Mental Disease & Volume 202, Number 2, February 2014

    136 www.jonmd.com * 2014 Lippincott Williams & Wilkins

    Copyright 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

  • Daar E, McMahon D, Tso Y, Yiannoutsos CT, Navia B (2010) Effects of nadirCD4 count and duration of human immunodeficiency virus infection on brainvolumes in the highly active antiretroviral therapy era. J Neurovirol. 16:25Y32.

    Elliott AJ, Uldall KK, Bergam K, Russo J, Claypoole K, Roy-Byrne PP (1998)Randomized, placebo-controlled trial of paroxetine versus imipramine in de-pressed HIV-positive outpatients. Am J Psychiatry. 155:367Y372.

    Evans DL, Staab J, Ward H, Leserman J, Perkins DO, Golden RN, Petitto JM(1996) Depression in the medically ill: Management considerations. DepressAnxiety. 4:199Y208.

    Fairfield KM, Libman H, Davis RB, Eisenberg DM (1999) Delays in protease in-hibitor use in clinical practice. J Gen Intern Med. 14:395Y401.

    Ferrando SJ, Freyberg Z (2008) Treatment of depression in HIV positive individ-uals: A critical review. Int Rev Psychiatry. 20:61Y71.

    Folstein MF, Folstein SE, McHugh PR (1975) Mini-mental state. A practicalmethod for grading the cognitive state of patients for the clinician. J PsychiatrRes. 12:189Y198.

    Gabryelewicz T, Styczynska M, Pfeffer A, Wasiak B, Barczak A, Luczywek E,Androsiuk W, Barcikowska M (2004) Prevalence of major and minordepression in elderly persons with mild cognitive impairmentVMADRSfactor analysis. Int J Geriatr Psychiatry. 19:1168Y1172.

    Gordillo V, del AJ, Soriano V, Gonzalez-Lahoz J (1999) Sociodemographic andpsychological variables influencing adherence to antiretroviral therapy. AIDS.13:1763Y1769.

    Gorman JM, Korotzer A, Su G (2002) Efficacy comparison of escitalopram andcitalopram in the treatment of major depressive disorder: Pooled analysis ofplacebo-controlled trials. CNS Spectr. 7:40Y44.

    Gutierrez MM, Rosenberg J, Abramowitz W (2003) An evaluation of the potentialfor pharmacokinetic interaction between escitalopram and the cytochromeP450 3A4 inhibitor ritonavir. Clin Ther. 25:1200Y1210.

    Guy W (1976) ECDU assessment manual for psychopharmacology. Rockville:National Institute of Mental Health, Research Branch.

    Hamilton M (1960) A rating scale for depression. J Neurol Neurosurg Psychiatry.23:56Y62.

    Horberg M, Silverberg M, Hurley L, Delorenze G, Quesenberry C (2008) Influ-ence of prior antiretroviral experience on adherence and responses to new highlyactive antiretroviral therapy regimens. AIDS Patient Care STDS. 22:301Y312.

    Ickovics JR, Hamburger ME, Vlahov D, Schoenbaum EE, Schuman P, Boland RJ,Moore J (2001) Mortality, CD4 cell count decline, and depressive symptomsamong HIV-seropositive women: Longitudinal analysis from the HIV Epide-miology Research Study. JAMA. 285:1466Y1474.

    Jacobsberg LB, Perry S (1992) Medical management of AIDS patients. Psychiat-ric disturbances. Med Clin North Am. 76:99Y106.

    Kagee A, Remien RH, Berkman A, Hoffman S, Campos L, Swartz L (2011)Structural barriers to ART adherence in Southern Africa: challenges and po-tential ways forward. Glob Public Health. 6:83Y97.

    Kalichman SC, Rompa D, Cage M (2000) Distinguishing between overlapping so-matic symptoms of depression and HIV disease in people living with HIV-AIDS. J Nerv Ment Dis. 188:662Y670.

    Leentjens AF, Marinus J, Van Hilten JJ, Lousberg R, Verhey FR (2003) The con-tribution of somatic symptoms to the diagnosis of depressive disorder inParkinsons disease: A discriminant analytic approach. J Neuropsychiatry ClinNeurosci. 15:74Y77.

    Leentjens AF, Verhey FR, Lousberg R, Spitsbergen H, Wilmink FW (2000) Thevalidity of the Hamilton and Montgomery-Asberg depression rating scalesas screening and diagnostic tools for depression in Parkinsons disease. Int JGeriatr Psychiatry. 15:644Y649.

    Lepola UM, Loft H, Reines EH (2003) Escitalopram (10Y20 mg/day) is effectiveand well tolerated in a placebo-controlled study in depression in primary care.Int Clin Psychopharmacol. 18:211Y217.

    Leserman J, Jackson ED, Petitto JM, Golden RN, Silva SG, Perkins DO, Cai J,Folds JD, Evans DL (1999) Progression to AIDS: The effects of stress, depres-sive symptoms, and social support. Psychosom Med. 61:397Y406.

    Montgomery SA, Asberg M (1979) A new depression scale designed to be sensi-tive to change. Br J Psychiatry. 134:382Y389.

    Nattrass N (2006) Trading off income and health?: AIDS and the disability grantin South Africa. J Soc Policy. 35:3Y19.

    Olley BO, Seedat S, Stein DJ (2004) Psychopathology and coping in recently di-agnosed HIV/AIDS patients. S Afr Med J. 94:720, 722.

    Owens MJ, Knight DL, Nemeroff CB (2001) Second-generation SSRIs: Humanmonoamine transporter binding profile of escitalopram and R-fluoxetine. BiolPsychiatry. 50:345Y350.

    Pappin M, Wouters E, Booysen F (2012) Anxiety and depression amongst patientsenrolled in a public sector antiretroviral treatment programme in South Africa:a cross-sectional study. BMC Public Health. 12:244.

    Power C, Selnes OA, Grim JA, McArthur JC (1995) HIV Dementia Scale: Arapid screening test. J Acquir Immune Defic Syndr Hum Retrovirol. 8:273Y278.

    Rabkin JG, Wagner GJ, Rabkin R (1999) Fluoxetine treatment for depression inpatients with HIV and AIDS: A randomized, placebo-controlled trial. Am JPsychiatry. 156:101Y107.

    Savard J, Laberge B, Gauthier JG, Ivers H, Bergeron MG (1998) Evaluating anx-iety and depression in HIV-infected patients. J Pers Assess. 71:349Y367.

    Sheehan DV (1983) The anxiety disease. New York: Scribners.

    Stein DJ, Baldwin DS, Dolberg OT, Despiegel N, Bandelow B (2006) Which fac-tors predict placebo response in anxiety disorders and major depression? Ananalysis of placebo-controlled studies of escitalopram. J Clin Psychiatry.67:1741Y1746.

    von Moltke LL, Greenblatt DJ, Giancarlo GM, Granda BW, Harmatz JS, ShaderRI (2001) Escitalopram (S-citalopram) and its metabolites in vitro: Cyto-chromes mediating biotransformation, inhibitory effects, and comparison toR-citalopram. Drug Metab Dispos. 29:1102Y1109.

    Wade A, Michael LO, Bang HK (2002) Escitalopram 10 mg/day is effective andwell tolerated in a placebo-controlled study in depression in primary care.Int Clin Psychopharmacol. 17:95Y102.

    Walker Z, Walker RW, Robertson MM, Stansfeld S (1998) Antidepressant treat-ment of chronic tension-type headache: A comparison between fluoxetineand desipramine. Headache. 38:523Y528.

    Zigmond AS, Snaith RP (1983) The Hospital Anxiety and Depression Scale. ActaPsychiatr Scand. 67:361Y370.

    Zisook S, Peterkin J, Goggin KJ, Sledge P, Atkinson JH, Grant I (1998) Treatmentof major depression in HIV-seropositive men. HIV Neurobehavioral ResearchCenter Group. J Clin Psychiatry. 59:217Y224.

    The Journal of Nervous and Mental Disease & Volume 202, Number 2, February 2014 Escitalopram in HIV Depression

    * 2014 Lippincott Williams & Wilkins www.jonmd.com 137

    Copyright 2014 Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.