Skip to main content
Dialogues in Clinical Neuroscience logoLink to Dialogues in Clinical Neuroscience
. 2008 Dec;10(4):419–430. doi: 10.31887/DCNS.2008.10.4/jlenze

Incomplete response in late-life depression: getting to remission

Respuesta incomplete en la depresión del anciano: alcanzando la remisión

Réponse partielle dans la dépression du sujet âgé: vers la rémission

Eric J Lenze 1, Meera Sheffrin 2, Henry C Driscoll 3, Benoit H Mulsant 4, Bruce G Pollock 5, Mary Amanda Dew 6, Frank Lotrich 7, Bernie Devlin 8, Robert Bies 9, Charles F Reynolds III 10,*
PMCID: PMC3181898  PMID: 19170399

Abstract

Incomplete response in the treatmen tof late-life depression is a large public health challenge: at least 50% of older people fail to respond adequately to first-line antidepressant pharmacotherapy, even under optimal treatment conditions. Treatment-resistant late-life depression (TRLLD) increases risk for early relapse, undermines adherence to treatment for coexisting medical disorders, amplifies disability and cognitive impairment, imposes greater burden on family caregivers, and increases the risk for early mortality, including suicide, Gettinq to and sustaininq remission is the primary goal of treatment yet there is a paucity of empirical data on how best to manage TRLLD. A pilot study by our group on aripiprazole augmentation in 24 incomplete responders to sequential SSRI and SRNI pharmacotherapy found that 50% remitted over 12 weeks with the addition of aripiprazole, and that remission was sustained in all participants during 6 months of continuation treatment In addition to controlled assessment, evidence is needed to support personalized treatment by testing the moderating role of clinical (eg, comorbid anxiety, medical burden, and executive impairment) and genetic (eg, selected polymorphisms in serotonin, norepinephrine, and dopamine genes) variables, while also controlling for variability in drug exposure. Such studies may advance us toward the goal of personalized treatment in late-life depression.

Keywords: old-age depression, incomplete response, remission, pharmacologic augmentation, aripiprazole

Incomplete response in late-life depression is an important public health problem

The aging of the US population is expected to increase the number of persons aged 65 and older from 35 million (in 2000) to more than 86 million by 2050.1 These data, together with longer life expectancy and increased depression rates in recent cohorts,2 predict an epidemic of late-life depression (LLD). LLD complicates medical illnesses3-7 and increases mortality,8 disability,9 and health care utilization.10 LLD often has poor acute outcome and brittle long-term outcome with antidepressant. treatment.11 Thus, new treatment approaches are needed to increase remission from LLD and to support, evidence -based selection of appropriate interventions at different points in the course of illness (ie, the right, treatment at the right, time).

Treatment-resistant depression (TRD) has been defined as failure to achieve remission with one antidepressant medication trial,12-14 or two trials,15 of adequate dose and duration. Rates of treatment resistance in randomized controlled trials in LLD are as high as 77% using selective serotonin reuptake inhibitors (SSRIs)16 and range from 55% to 81 % using serotonin/norepinephrine reuptake inhibitors (SNRIs).16-19 Treatment resistance must be distinguished from inadequate treatment (eg, short treatment duration preventing late responders from achieving remission), and misdiagnosis (eg, failing to recognize dementia, psychosis, or bipolar disorder). Treatment resistance is particularly germane to LLD, for three reasons. First, high rates of comorbid anxiety and medical illness contribute to treatment failure. Second, older adults may have greater pharmacodynamic variability as a result, of genetic variability (eg, at. serotonin receptors20) and ageor medical illness-related changes in brain structure or function (eg, decline in serotonin receptors21,22), interruptions in neurocircuitry integrity from cerebrovascular disease or prodromal Alzheimer's disease.23,24 Third, older adults may have greater pharmacokinetic variability, as a result of poor adherence (eg, due to cognitive impairment.) and metabolic variability (eg, due to age-related changes in drug metabolism).25

The serious consequences of persistent depressive symptoms in elderly persons include relapse and recurrence,26-29 functional disability,30 and cognitive decline, owing in part to the impact of long periods of untreated depression on hippocampal volume.31 Persisting LLD is also associated with an increased mortality,32 including suicide. Risk for suicide can be reduced with successful treatment.33,34 Finally, treatment-resistant late-life depression (TRLLD) is associated with increased caregiver burden in family members of depressed elders (Martire L, personal communication, 2008). In these ways, incomplete response in late-life depression and the need to get to remission are major public health challenges.

Despite this challenge, almost no data exist to guide the treatment of TRLLD. The best, current evidence guiding intervention for treatment-resistant depression comes from the Sequenced Treatment Alternatives to Relieve Depression (STAR*D study35). However, only a small minority of subjects who participated in STAR*D were elderly. Our collaborative group has carried out several examinations of treatment strategies for TRLLD, including open studies of switching from an SSRI to nortriptyline,36 venlafaxine,37 or duloxetine,38 a stepwise strategy of bupropion, nortriptyline, or lithium augmentation of SSRI,39,40 and electroconvulsive therapy.41,42 Our findings suggest that, a significant proportion (40% to 50%) of SSRI nonresponders will respond to these strategies, consistent with a prior open sequential trial.43 In the only published placebo-controlled pharmacotherapy trial for TRLLD, Sunderland et al44 found that the monoamine oxidase inhibitor (MAOI) selegiline was efficacious. However, in a recent randomized comparison of lithium augmentation and the MAOI phenelzine for TRLLD, one third of those receiving lithium remitted versus none receiving phenelzine.19 These two controlled studies suffer from small sample size, short, duration, and inclusion of subjects with psychosis. Thus, beyond the intuitive step of switching from SSRI to SNRI, there appear to be almost no controlled data to inform the treatment of TRLLD in old age.

The relationship of anxiety, comorbid medical illness, and executive dysfunction to TRLLD

Literature reviews have suggested that anxiety, medical illness, and executive dysfunction may be key clinical predictors of treatment resistance in LLD.37,45

Anxiety

Anxiety is a common cotraveler with LLD. Several studies have found an increased time to remission, and reduced remission rate, in LLD when there are either high levels of anxiety symptoms46-52 or a comorbid anxiety disorder such as generalized anxiety.53 Despite numerous studies establishing anxiety as a predictor of treatment resistance in LLD, this relationship is poorly understood. Mechanisms that may explain this relation-ship include reduced tolerance of, and adherence to, medication, or a more severe subtype of depression. Anxiety in late life is multidimensional, encompassing worry, panic/fear, somatization, and personality factors54; the differential impact, of these dimensions on treatment resistance is largely unstudied. Along these lines, we have found preliminarily that symptoms of worry, and not fear or panic, predict both poor short-term outcome in LLD and poor long-term stability of remission (Andreescu C, personal communication, 2008). Needed is a treatment trial incorporating examinations of these multiple dimensions that will shed light on the anxiety-depression interface in late life.

Medical burden

Several studies have demonstrated that LLD patients with greater medical burden have a lower, and slower, treatment response in LLD (eg, refs 55-57). Although some studies have not supported a link between medical burden and treatment outcome,58,59 our group found that greater medical burden predicted poorer acute outcome to antidepressant augmentation (primarily with bupropion or nortriptyline40) and poorer maintenance outcomes.60 One reason may be that medical illnesses seen in patients with LLD (eg, hypertension, high cholesterol, diabetes, endocrinologie disease) induce pharmacodynamic or structural central nervous system changes that reduce the efficacy of standard antidepressants. Other possibilities are that medical burden interferes with antidepressant adherence and/or increases variability of drug exposure, thus reducing the impact of antidepressants.

Impairment of executive functioning

Neuropsychological impairment, particularly in executive functioning, is common and clinically significant in LLD.61 Several studies have noted a relationship of cognitive impairment, with lower antidepressant response rates,62-64 though other studies have not found this relationship.65-67 The discrepancy may result, from the variability between studies in measuring executive functioning, and the current consensus in the field is that executive dysfunction is associated with poorer LLD treatment outcomes with antidepressants. Treatment resistance in the context of executive dysfunction is thought to be due to alterations in neurocircuitry integrity that disrupt the pharmacodynamics of antidepressants.68 In summary, the above clinical variables predict poor antidepressant outcomes in LLD. However, there is insufficient understanding of how they contribute to poorer outcomes, and so their clinical utility is limited. This lack of understanding is part of the gap between personalized medicine (matching treatment, to patients based upon patient characteristics) and the current trialand-error approach to LLD management.

The relationship of genetic and drug exposure variability to TRLLD

Functional genetic polymorphisms change the pharmacodynamics of antidepressant medications; therefore, it is posited that antidepressant outcomes in LLD can be predicted by genetic variation in their homologous receptor targets.69 In other words, functional genetic variation of the 5-HTT is expected to affect. SSRI response, while variation in the norepinephrine transporter (NET) is expected to affect. SNRI response. One example is the serotonin transporter linked polymorphic region (5-HTTLPR) in the promoter of the gene that encodes for the serotonin transporter (5-HTF), the primary target of SSRIs. A deletion polymorphism in 5-HTTLPR, the s allele (s=“short” vs l=“long”), appears to be functional: it reduces expression of 5-HTT so that individuals with the s allele have fewer 5-HTTs than those with 1/1 genotype. The association of the s allele with poorer SSRI outcomes has been demonstrated in LLD,70 including a study from our group that, was the first to report this association in LLD.20 The association appears specific to SSRIs and was not found with mirtazapine71 or nortriptyline.70 In addition, we think that measures of drug exposure are needed to interpret clinical and genetic findings.72 Specifically, we think that, pharmacokinetic modeling is important in pharmacogenetic analyses. Supporting this contention, Lotrich et al73 found that the 5-HTTLPR s allele predicted poorer treatment outcome at lower concentrations of paroxetine but not at. higher concentrations.

Following up on this observation, Lotrich examined depressed elderly subjects who were treated in an openlabel paroxetine study and who were genotyped (n=110). Again, there was an interaction between paroxetine concentration and 5-HTTLPR genotype on symptomatic improvement over 12 weeks (F(18,59.5)=1.8; P<0.05): paroxetine concentrations were correlated with change in the Hamilton Depression Rating Scale (HAM-D) in subjects with the s allele, but not. in subjects homozygous for the 1 allele. In other words, the s allele moderated the impact of the drug. 'ITtiesc data demonstrate the importance of pharmacokinetic data for conducting meaningful pharmacogenetic analyses. This issue is particularly relevant to geriatrics, as age-related changes in drug elimination amplify drug concentration differences for a given dose.

Relevance of anxiety, medical burden, executive dysfunction, and genetic variability for augmentation strategies in TRLLD

Comorbid anxiety, medical burden, and executive dysfunction are highly prevalent in TRLLD patients. Because these variables are associated with poor outcomes using standard antidepressants, they may identify patients likely to require more aggressive strategies including augmentation (as opposed to “staying the course”). As such, these variables are expected to moderate the efficacy of augmentation (ie, increase drugplacebo difference). This is consistent with research from PROSPECT (Prevention of Suicide in Primary Care Elderly: Collaborative Trial) in which executive dysfunction moderated the difference between aggressive LLD management and usual care.74 A similar moderation effect, has been found with medical comorbidity60,75 and comorbid anxiety.9 Thus, we hypothesize that anxiety, medical burden, and executive dysfunction are clinical markers of need for augmentation.

Conversely, it is possible that these variables predict treatment nonadherence or increased metabolic variability resulting in poor outcomes regardless of treatment.40 This possibility underscores the importance of measuring drug exposure in studies of TRLLD. For example, by controlling for both the average drug concentration and the variability of drug exposure, it. is possible to determine the contribution of comorbid medical illness to treatment efficacy while accounting for drug exposure. The same logic applies for patients with highly prevalent genetic polymorphisms. Thus, by using drug exposure data the effect of clinical and genetic moderators can be more precisely examined, ultimately reducing the gap between the potential of personalized medicine and the current empiric approach for LLD management. In the next section, we present for heuristic purposes our work with aripiprazole as a candidate augmentation strategy for managing incomplete response in LLD and getting to remission. We present, first a pharmacologic and clinical rationale, followed by pilot data. Finally, we describe the design of a randomized controlled trial informed by those data.

Aripiprazole as a potential treatment for TRLLD

Aripiprazole is an atypical antipsychotic (or “atypical”) approved by the Food and Drug Administration to treat schizophrenia and mania. It has a high 13, receptor affinity, and as a partial agonist, it has a higher affinity for the G protein-coupled state of the D2 receptor, ie, its active state.76 With partial D2 agonist properties it. is conceived as a dopamine system stabilizer: in high dopaminergic states it. acts as an antagonist, and in low dopaminergic states it. acts as an agonist.77 This may explain why it is unlikely to cause extrapyramidal side effects or prolactin elevation even at. high D2 receptor occupancy.78-80 Aripiprazole also has high affinity for the D3 receptor and is an antagonist at the 5-HT2a receptor.81 ft has only moderate affinity to the adrenergic alpha-1 receptor and histamine H1 receptor, and negligible affinity to the muscarinic receptor.82 As a result, orthostatic hypotension and antihistaminergic or anticholinergic adverse effects are less likely to occur than with other atypicals. Also, increases in mean QTc interval are not. observed. Finally, as hyperprolactinemia can contribute to osteoporosis, aripiprazole's lack of this side effect, reduces this concern. These pharmacodynamic features make aripiprazole attractive for use in older patients.

A meta-analysis of the use of atypicals as augmentation treatment for depression found pooled response rates of 57% vs 35% for placebo.83 The meta-analysis utilized data from 10 double-blind, placebo-controlled studies of augmentation of an antidepressant with an atypical antipsychotic agent. Augmentation with olanzapine, risperidone, and quetiapine was found to be efficacious for treatment-resistant depression. 'this meta-analysis did not include data on aripiprazole or from geriatric samples. In part, the efficacy of atypicals in this context, seems to stem from their benefit for anxiety,84,85 which is a marker for poor outcomes in MDD. Their 5-HT2a receptor antagonism would be expected to increase serotonin and norepinephrine release, thus augmenting the effect of SSRIs and SNRIs.86,87 In the case of aripiprazole, antidepressant and antianxiety actions could also stem from its D2 partial antagonism88 or its high affinity for D3 receptors. A novel neurobiological paradigm views anxiety and depression in the context, of the amygdala-prefrontal circuit, with amygdala hyperactivity coinciding with prefrontal hypoactivity89 and both coinciding with imbalances in dopamine.90 Aripiprazole, through its dopamine partial agonism, may promote equilibrium in this circuit, and provide benefits for anxiety and depression. However, this neurobiological argument, requires further testing.

Two large, industry-initiated, placebo-controlled trials of nongeriatric adults have recently demonstrated the efficacy of aripiprazole as an augmentation treatment for depression incompletely responsive to SSRIs and SNRIs.91,92 Based on these regulatory trials, the FDA has approved an indication for the use of aripiprazole to augment SSRIs and SNRIs for treatment-resistant depression. The one published trial showed a higher rate of remission (as measured with the Montgomery-Åsberg Depression Rating Scale) in the aripiprazole group than in the placebo group. Few adverse events leading to about 3% discontinuation in each group.91,92 Two limitations of this study were the short duration of the augmentation trial (6 weeks) and the high placebo remission rate (37%) suggesting that the criteria for treatment resistance (failure to respond to one 8-week antidepressant lead-in phase that did not maximize dosage) were not stringent enough.

Aripiprazole has been examined preliminarily in LLD as an augmentation for SSRI nonresponders,93 and the Pittsburgh group has examined its effect and tolerability in 24 SNRI nonresponders94 (data presented below). As described below, these two small open-label studies have similar results: 50% of older nonresponders converted to remitters, and adverse events requiring treatment cessation were infrequent. These preliminary results are encouraging but, to our knowledge, no placebo-controlled examination of aripiprazole for TRLLD has been carried out.

Safety issues with atypicals in older adults

Atypicals have come under scrutiny due to the metabolic disturbances they may cause and safety issues uncovered in older patients with dementia. Weight, gain and related metabolic disturbances such as glucose intolerance and dyslipidemia occur more frequently in psychiatric patients than the general population, with the totality of risk related not only to medication effects but. to under

lying characteristics of the patient, population (eg, baseline overweight, and obesity, high fat/high caloric diet, poor medical care).95 The Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) confirmed other reports demonstrating risks for metabolic disturbances with atypicals, although that study did not. examine aripiprazole. Among the atypicals, risk of weight gain, dyslipidemia, and diabetes is highest with clozapine and olanzapine; more modest, weight, gain is generally observed with quetiapine and risperidone, along with lower insulin resistance risk, variably lower dyslipidemia risk, and largely negative if somewhat discrepant results concerning diabetes risk. The lowest, risk of weight gain, as well as little or no risk for dyslipidemia or diabetes, is observed with aripiprazole and ziprasidone.95-97 These metabolic risks have not been consistently reported in the elderly, where some studies indicate little or no weight gain, even on higher-risk agents (eg refs 98-100). However, there are limited data in elderly samples, and available reports that include a placebo group often find weight loss, consistent with progressive reductions in lean muscle mass. Thus, in elderly persons, measuring weight gain alone with antipsychotics could miss treatmentrelated increases in adiposity. Direct measures of adiposity such as dual-energy X-ray absorptiometry (DEXA) as well as sensitive and reliable measures of insulin resistance, lipid metabolism, and glucose control, are needed in research studies of these medications to examine metabolic risk.

A meta-analysis found a higher mortality with atypicals compared with placebo in older patients with dementia, resulting in a black -box warning for the entire class of atypicals. It remains unclear what the increased mortality resulted from, though possibilities include the sedating properties of these agents (leading to falls or aspiration pneumonia), QT prolongation (leading to arrhythmias and sudden cardiac death), venous thromboembolism leading to pulmonary embolism, and other cardiovascular or cerebrovascular events.101,102 It. is unknown whether these risks apply to nondemented elderly patients. A third type of adverse event, involves extrapyramidal side effects and other (nonvascular) neurological problems. Age-associated reductions in dopamine and D2 receptors make the elderly more sensitive to antipsychotics, although aripiprazole's partial agonism at the D2 receptor could reduce such effects. Thus, a placebo-controlled clinical trial is needed to further investigate the tolerability and safety of aripiprazole augmentation in LLD. The lack of such a trial is a significant gap in our knowledge base.

In summary, TRLLD is a common and potentially devastating condition, yet we have an extremely limited evidence basis for its management. Clinicians do not have data to guide them regarding which augmentation agent to use, in whom, how, or with which risk:bencfit ratio. Needed is a randomized placebo-controlled trial to support, the value of a modern pharmacologic treatment for TRLLD, to establish a new approach to TRLLD, to lead to a greater understanding of treatment response variability and ultimately to personalized treatment, for LLD. Also needed is a multidimensional approach to treatment resistance, in which key clinical features in LLD (anxiety, medical comorbidity, and executive dysfunction) are examined as hypothesized moderators for augmentation outcomes. An examination of genetic variability at the drug target molecules, with a goal to predict those in whom specific treatment, strategics (eg, high-dose venlafaxine, aripiprazole augmentation) are more robust is also needed to personalize treatment. Finally, a detailed examination of the sources of treatment, resistance using state-of-the-art pharmacokinetic methods is necessary. For illustrative purposes, we now present work in progress with aripiprazole as a candidate augmentation strategy for incomplete response to antidepressant pharmacotherapy.

Aripiprazole augmentation data: pilot study and design of a controlled trial

To examine the acceptability, feasibility, and safety of aripiprazole as an augmentation agent, for incomplete response in LLD, we carried out a 12-week open-label pilot study in 24 elderly patients.94 Patients aged 65+ with current major depressive disorder, with an initial HAMD score ≥5 were first, treated with escitalopram for 16 weeks. Those who failed to respond (HAM-D≥15,N=19) or responded partially (HAM-D=11-14, N=5) were switched to either duloxetine up to 120mg/d or venlafaxine up to 225 mg/day (depending on tolerability and prior medication history) and treated for 12 weeks. Those with partial or nonrcsponse to the SNRI were started on 2.5 mg/day of adjunctive aripiprazole, titrated weekly in 2.5mg increments to 15 mg, as tolerated and as needed to reach remission.

The 24 subjects had a mean age of 74 (range 65 to 91); 58% were female; 8% were African-American. Nineteen of 24 (79%) patients completed all 1 2 weeks of augmentation with aripiprazole, and 12/24 (50%) met criteria for remission (defined as 2 consecutive weeks of HAMD≤10).

Tolerability and side effects

Three of 24 (13%) discontinued prior to week 12 due to failure to improve or withdrawal of consent, and 2/24 (8%) discontinued due to side effects (one each: sedation, akathisia). Side effects were also examined via the UKU side effects scale.103 Overall UKU scores showed a decline (indicating fewer reports of somatic complaints compared to baseline). However, the mean score of the UKU-Neurologic subscalc increased. Six of 24 (25%) subjects had a positive score on the UKU-akathisia item on at least one time point; however, in all but. one case, these were mild and/or transient.

We also examined metabolic changes and weight gain during the 12-week period of pharmacotherapy augmentation. One subject had a significant increase in lipids, and none had a significant increase in blood sugar, suggesting that metabolic effects were infrequent with aripiprazole. Weight gain was highly variable: 9/15 (60%) gained <2 kg (mean [range] 0.8 [-0.7- 1.8]) while 6/15 (40%) gained >3 kg (mean [range] 4.7 [3.2-6.4]), suggesting that an examination of sources of weight gain variability would be useful. Two possibilities from the literature are genetic variation at. the 5-HT2C receptor (posited as the receptor responsible for weight gain with aripiprazole) and baseline body mass index (BMI). Also, we were not. able to determine whether weight gain represented an increase in adiposity vs an increase in lean body mass with remission from depression. Thus, we determined that a controlled study should include: (i) a more precise examination of changes in adiposity, including DEXA scans which would provide quantitative measures of body fat; (ii) an examination of moderators of weight gain (including baseline BMI and 5-HT2C genotyping); and (iii) a continuation phase, allowing longer duration to observe weight, changes.

Pilot study of continuation phase pharmacotherapy

Of the 24 participants who received acute-phase adjunctive aripiprazole, 12 met study criteria for complete response (remission) and entered continuation phase pharmacotherapy, on an average daily dose of 10 mg of aripiprazole (as an adjunct to their primary antidepressant, pharmacotherapy). The 12 participants in the feasibility study of continuation-phase pharmacotherapy had a mean age of 72.7 (SD: 6.2); 9 were women, and 10 were white (2 were African-American).

Outcomes

Depressive relapse during continuation-phase pharmacotherapy

Over a median duration of 27.6 weeks (range: 2-106) of continuation-phase combined pharmacotherapy (antidepressant. + aripiprazole), none of the 12 participants experienced relapse of a major depressive episode.

Retention

One of 12 participants was noncompliant with study procedure (due to respondent burden and other treatment preferences) and exited the study.

Side effects

UKU side effect, scores remained stable (9.4[3.2] at start of continuation-phase pharmacotherapy [n = 12] and 7.9[2.8] at. 6 months [n = 7]). No participant left the study due to treatment-emergent adverse events.

Metabolic data

Body mass index was stable over 6 months (29.8 [6.1] at start of continuation phase pharmacotherapy [n = 12] and 30.1 [6.1] at 6 months [n = 7]).

Figure 1 depicts individual participants' patterns of change in the metabolic data between baseline and 6 months, after an overnight fast for glucose, triglyderides, cholesterol, HDL, and LDL. In general, values were stable over time. One person each had a spike in glucose, triglycerides, and cholesterol/LDL. After the 6-month follow-up, this last person was started on a statin prescribed by their primary care physician, who judged that the benefit of continuing treatment with aripiprazole in the study was substantial, and that metabolic changes could be managed medically.

Figure 1. Metabolic profile of individual patients (each indicated by a separate symbol) during continuation treatment with aripiprazoie over a median period of 6 months.

Figure 1.

In general, glucose and triglycerides showed minimal change, suggesting that aripiprazole does not cause insulin resistance as do some other atypicals do (eg, olanzapine). In a comprehensive review of this topic, Newcomer showed that generally a lipid signal with atypicals will be seen in triglycerides; thus the lack of a signal in these pilot, data suggest that aripiprazole will be a safe treatment in older adults with respect, to metabolic effects.97 We plan to closely control the collection procedures in subjects, so that pre -post differences are not due to variability in fasting, stasis-venous collection, etc. The lack of clinically informative data on this in the elderly is striking in light of the high cardiovascular mortality in mentally ill persons generally and underscores the need for this research.104

These data from acute and continuation open pharmacotherapy illustrate three points.

  1. Further investigation should evaluate both the benefits and the costs (eg, adverse effects, metabolic changes) of adjunctive aripiprazole pharmacotherapy, using a double-blind, randomized, placebo-controlled design.

  2. These data show the feasibility and safety of treating participants (i) during acute-phase pharmacotherapy (n=24), to determine change from incomplete to complete response; and (ii) during continuation -phase pharmacotherapy (n=12), to determine stability of remission and rates of depressive relapse.

  3. These data also underscore the importance of examining risks, as well as benefits, in a large randomized, double-blind, placebo-controlled study. The cost-benefit ratio and the ensuing clinical conclusions may be very different when benefit and harm are conjointly considered, from what they are when benefit, and harm are considered separately (as the post-marketing experience with COX-2 inhibitors and oral hypoglycemic agents teaches us, vis-à-vis heart disease). We believe that this is the most appropriate approach scientifically and ethically to a treatment study of frail older depressed patients who have responded only partially to antidepressant pharmacotherapy.

Our conclusions from this pilot study

  1. In older adults with MDD having incomplete response to an SSRI followed by an SNRI, remission was obtained in 50% during aripiprazole augmentation.

  2. In most subjects who remitted, the improvements in depression were stable throughout 6-month continuation pharmacotherapy.

  3. Aripiprazole was well-tolerated, with a low rate of dropout due to side effects and a high completion rate, but restlessness and weight gain were not uncommon. Overall, a larger, placebo-controlled study is needed to test hypotheses related to remission, tolerability, safety, and outcome predictors. These pilot data support the feasibility of such a trial. In Figure 2, we show the design of a placebo-controlled randomized clinical trial which we plan to conduct.

Figure 2. Representation of proposed multisite study of aripiprazoie (aripip) augmentation for treatment-resistant late-life depression. Venla, venlafaxine .

Figure 2.

The planned trial calls for enrolling 500 patients aged 60 and older with major depressive disorder and treating them openly for 12 weeks with venlafaxine XR (up to 225 mg/d) to prospectively determine incomplete response (phase 1). Participants meeting criteria for incomplete response estimated (n=200) will be randomly assigned to receive either aripiprazoie (2.5-15 mg/d; target dose: 10 mg/d) or placebo augmentation of venlafaxine for 12 weeks (phase 2), with the goal of achieving remission (Montgomery-Åsberg DRS<10 for two consecutive assessments). Those who remit in phase 2 will receive continuation treatment, with the same doubleblinded intervention to which they were randomly assigned (phase 3), for 12 weeks to determine the stability of remission. Based on efficacy and tolerability data, we will estimate number needed to treat and number needed to harm, providing a clinically informative estimate of benefits and risks of aripiprazoie augmentation for TRLLD.

Conclusion

In summary, the public health importance of TRLLD studies is great, but. there are no data from controlled studies to guide practice. Data are needed to not only examine the overall efficacy of adjunctive treatments but also examine in whom such treatments are most, efficacious and safe, thus moving the treatment of LLD into the arena of personalized medicine.

Acknowledgments

P30 MH07 1944, R37 MH43832, R01 MH3786769, T32 MH19986, RR 024153, and the John A. Hartford Foundation Center of Excellence in Geriatric Psychiatry

Selected abbreviations and acronyms

LLD

late-life depression

SRNI

serotonin/norepinephrine reuptake inhibitor

SSRI

selective serotonin reuptake inhibitor

TRD

treatment-resistant depression

TRLLD

treatment-resistant late-life depression

Contributor Information

Eric J. Lenze, Washington University School of Medicine Department of Psychiatry, St Louis, MO, USA.

Meera Sheffrin, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Henry C Driscoll, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Benoit H. Mulsant, Center for Addictions and Mental Health, University of Toronto, Canada.

Bruce G. Pollock, Center for Addictions and Mental Health, University of Toronto, Canada.

Mary Amanda Dew, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Frank Lotrich, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Bernie Devlin, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Robert Bies, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

Charles F. Reynolds III, Advanced Center for Interventions and Services Research for Late-Life Mood Disorders, University of Pittsburgh, PA, USA.

REFERENCES

  • 1.United States Census Bureau. Projected Population of the United States, by Race and Hispanic Origin: 2000 to 2050. Available at: http://www.census.gov/ipc/www/usinterimprol/. 2004 [Google Scholar]
  • 2.Lewinsohn PM, Rohde P, Seeley JR, Fischer SA. Age-cohort changes in the lifetime occurrence of depression and other mental disorders. J Abnorrn Psychol. 1993;102:110–120. doi: 10.1037//0021-843x.102.1.110. [DOI] [PubMed] [Google Scholar]
  • 3.Sinyor D, Amato P, Kaloupek DG, Becker R, Goldenberg M, Coopersmith H. Post-stroke depression: relationships to functional impairment, coping strategies, and rehabilitation outcome. Stroke. 1986;17:1102–1107. doi: 10.1161/01.str.17.6.1102. [DOI] [PubMed] [Google Scholar]
  • 4.Palinkas LA, Wingard DL, Barrett-Connor E. Chronic illness and depressive symptoms in the elderly: a population-based study. J Clin Epidemiol. 1990;43:1131–1141. doi: 10.1016/0895-4356(90)90014-g. [DOI] [PubMed] [Google Scholar]
  • 5.Carney RM, Freedland KE, Eisen SA, Rich MW, Jaffe AS. Major depression and medication adherence in elderly patients with coronary artery disease. Health Psychol. 1995;14:88–90. doi: 10.1037//0278-6133.14.1.88. [DOI] [PubMed] [Google Scholar]
  • 6.Michelson D, Stratakis C, Hill L, Reynolds J, et al. Bone mineral density in women with depression. N Engl J Med. 1996;335:1176–1181. doi: 10.1056/NEJM199610173351602. [DOI] [PubMed] [Google Scholar]
  • 7.Musselman DL, Evans DL, Nemeroff CB. The relationship of depression to cardiovascular disease: epidemiology, biology, and treatment. Arch Gen Psychiatry. 1998;55:580–592. doi: 10.1001/archpsyc.55.7.580. [DOI] [PubMed] [Google Scholar]
  • 8.Rovner BW, German PS, Brant LJ, Clark R, Burton L, Folstein MF. Depression and mortality in nursing homes. JAMA. 1991;265:993–996 [published erratum appears in JAMA. 1991;265:2672]. doi: 10.1001/jama.265.8.993. [DOI] [PubMed] [Google Scholar]
  • 9.Lenze EJ, Rogers JC, Martire LM, et al. The association of late-life depression and anxiety with physical disability: a review of the literature and prospectus for future research. Am J Geriatr Psychiatry. 2001;9:113–135. [PubMed] [Google Scholar]
  • 10.Katon WJ, Lin E, Russo J, Unutzer J. Increased medical costs of a population-based sample of depressed elderly patients. Arch Gen Psychiatry. 2003;60:897–903. doi: 10.1001/archpsyc.60.9.897. [DOI] [PubMed] [Google Scholar]
  • 11.Charney DS, Nemeroff C, Lewis L, et al. National depressive manicdepressive association consensus statement on the use of placebo in clinical trials of mood disorders. Arch Gen Psychiatry. 2002;59:262–270. doi: 10.1001/archpsyc.59.3.262. [DOI] [PubMed] [Google Scholar]
  • 12.Fava M. Diagnosis and definition of treatment-resistant depression. Biol Psychiatry. 2003;53:649–659. doi: 10.1016/s0006-3223(03)00231-2. [DOI] [PubMed] [Google Scholar]
  • 13.Kornstein SG, Schneider RK. Clinical features of treatment-resistant depression. J Clin Psychiatry. 2001;62(suppl 16):18–25. [PubMed] [Google Scholar]
  • 14.Tew JD, Mulsant BH, Houck PR, et al. Impact of prior inadequate treatment exposure on response to antidepressant treatment in late life. Am J Geriatr Psychiatry. 2006;14:957–965. doi: 10.1097/01.JGP.0000222311.70424.85. [DOI] [PubMed] [Google Scholar]
  • 15.Souery D, Papakostas Gl, Trivedi MH. Treatment-resistant depression. J Clin Psychiatry. 2006;67(suppl 6):16–22. [PubMed] [Google Scholar]
  • 16.Allard P, Gram L, Timdahl K, Behnke K, Hanson M, Sogaard J. Efficacy and tolerability of venlafaxine in geriatric outpatients with major depression: a double-blind, randomised 6-month comparative trial with citalopram. Int J Geriatr Psychiatry. 2004;19:1123–1130. doi: 10.1002/gps.1190. [DOI] [PubMed] [Google Scholar]
  • 17.Schatzberg A, Roose S. A double-blind, placebo-controlled study of venlafaxine and fluoxetine in geriatric outpatients with major depression. Am J Geriatr Psychiatry. 2006;14:361–370. doi: 10.1097/01.JGP.0000194645.70869.3b. [DOI] [PubMed] [Google Scholar]
  • 18.Raskin J, Wiltse CG, Siegal A, et al. Efficacy of duloxetine on cognition, depression, and pain in elderly patients with major depressive disorder: an 8-week, double-blind, placebo-controlled trial. An J Psychiatiy. 2007;164:900–909. doi: 10.1176/ajp.2007.164.6.900. [DOI] [PubMed] [Google Scholar]
  • 19.Kok RM, Nolen WA, Heeren TJ. Venlafaxine versus nortriptyline in the treatment of elderly depressed inpatients: a randomised, double-blind, controlled trial. Int J Geriatr Psychiatry. 2007;22:1247–1254. doi: 10.1002/gps.1823. [DOI] [PubMed] [Google Scholar]
  • 20.Pollock BG, Ferrell RE, Mulsant BH, et al. Allelic variation in the serotonin transporter promoter affects onset of paroxetine treatment response in late-life depression. Neuropsychopharmacology. 2000;23:587–590. doi: 10.1016/S0893-133X(00)00132-9. [DOI] [PubMed] [Google Scholar]
  • 21.Sheline Yl, Mintun MA, Barch DM, Wilkins C, Snyder AZ, Moerlein SM. Decreased hippocampal 5-HT(2A) receptor binding in older depressed patients using [18F]altanserin positron emission tomography. Neuropsychopharmacology. 2004;29:2235–2241. doi: 10.1038/sj.npp.1300555. [DOI] [PubMed] [Google Scholar]
  • 22.Meltzer CC, Price JC, Mathis CA, et al. Serotonin 1A receptor binding and treatment response in late-life depression. Neuropsychopharmacology. 2004;29:2258–2265. doi: 10.1038/sj.npp.1300556. [DOI] [PubMed] [Google Scholar]
  • 23.Bae JN, MacFall JR. Krishnan KR, Payne ME, Steffens DC, Taylor WD. Dorsolateral prefrontal cortex and anterior cingulate cortex white matter alterations in late-life depression. Biol Psychiatry. 2006;60:1356–1363. doi: 10.1016/j.biopsych.2006.03.052. [DOI] [PubMed] [Google Scholar]
  • 24.Sweet RA, Hamilton RL, Butters MA, et al. Neuropathology correlates of late-onset major depression. Neuropsychopharmacology. 2004;29:2242–2250. doi: 10.1038/sj.npp.1300554. [DOI] [PubMed] [Google Scholar]
  • 25.Mulsant BH, Pollock B. Treatment-resistant depression in late-life. J Geriatr Psychiatry Neurol. 1998;11:186–193. doi: 10.1177/089198879901100404. [DOI] [PubMed] [Google Scholar]
  • 26.Dew MA, Reynolds CF, Mulsant B, et al. Initial recovery patterns may predict which maintenance therapies for depression will keep older adults well. J Affect Disord. 2001;65:155–166. doi: 10.1016/s0165-0327(00)00280-9. [DOI] [PubMed] [Google Scholar]
  • 27.Steffens DC, McQuoid DR, Krishnan KR. Partial response as a predictor of outcome in geriatric depression. Am J Geriatr Psychiatry. 2003;11:340–348. [PubMed] [Google Scholar]
  • 28.Taylor WD, McQuoid DR, Steffens DC, Krishnan KR. Is there a definition s of remission in late-life depression that predicts later relapse? Neuropsychopharmacology. 2004;29:2272–2277. doi: 10.1038/sj.npp.1300549. [DOI] [PubMed] [Google Scholar]
  • 29.Dombrovski AY, Mulsant BH, Houck PR, et al. Residual symptoms and recurrence during maintenance treatment of late-life depression. J Affect Disord. 2007;103:77–82. doi: 10.1016/j.jad.2007.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lenze EJ, Schulz R, Martire LM, et al. The course of functional decline in older people with persistently elevated depressive symptoms: longitudinal findings from the Cardiovascular Health Study. J Am Geriatr Soc. 2005;53:569–575. doi: 10.1111/j.1532-5415.2005.53202.x. [DOI] [PubMed] [Google Scholar]
  • 31.Sheline Yl, Gado MH, Kraemer HC. Untreated depression and hippocampal volume loss. Am J Psychiatry. 2003;160:1516–1518. doi: 10.1176/appi.ajp.160.8.1516. [DOI] [PubMed] [Google Scholar]
  • 32.Ganguli M, Dodge HH, Mulsant BH. Rates and predictors of mortality in an aging, rural, community-based cohort: the role of depression. Arch Gen Psychiatry. 2002;59:1046–1052. doi: 10.1001/archpsyc.59.11.1046. [DOI] [PubMed] [Google Scholar]
  • 33.Gallo JJ, Bogner HR, Morales KH, Post EP, Lin JY, Bruce ML. The effect of a primary care practice-based depression intervention on mortality in older adults: a randomized trial. Ann Intern Med. 2007;146:689–698. doi: 10.7326/0003-4819-146-10-200705150-00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Nelson JC, Delucchi K, Schneider L. Suicidal thinking and behavior during treatment with sertraline in late-life depression. Am J Geriatr Psychiatry. 2007;15:573–580. doi: 10.1097/JGP.0b013e318050c9c2. [DOI] [PubMed] [Google Scholar]
  • 35.Rush AJ, Trivedi MH, Wisniewski SR, et al. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry. 2006;163:1905–1917. doi: 10.1176/ajp.2006.163.11.1905. [DOI] [PubMed] [Google Scholar]
  • 36.Houck PR, Mazumdar S, Mulsant BH, Pollock BG, Dew MA, Reynolds CF. An intent-to-treat method for enhancing analysis of clinical trials with rescue medication: A mixed-model approach. Psychopharmacol Bull. 2003;37:79–89. [PubMed] [Google Scholar]
  • 37.Whyte EM, Basinski J, Farhi P, et al. Geriatric depression treatment in SSRI non-responders. J Clin Psychiatry. 2004;65:1634–1641. doi: 10.4088/jcp.v65n1208. [DOI] [PubMed] [Google Scholar]
  • 38.Karp JF, Whyte EM, Lenze EJ, et al. Rescue pharmacotherapy with duloxetine for selective serotonin reuptake inhibitor nonresponders in late-life depression: outcome and tolerability. J Clin Psychiatry. In press. doi: 10.4088/jcp.v69n0317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Mulsant BH, Alexopoulos GS, Reynolds CF, et al., the PROSPECT Study Group. Pharmacological treatment of depression in elderly primary care patients: The PROSPECT algorithm. Int J Geriatr Psychiatry. 2001;16:585–592. doi: 10.1002/gps.465. [DOI] [PubMed] [Google Scholar]
  • 40.Dew MA, Whyte EM, Lenze EJ, et al. Recovery from major depression in older adults receiving augmentation of antidepressant pharmacotherapy. Am J Psychiatry. 2007;164:892–899. doi: 10.1176/ajp.2007.164.6.892. [DOI] [PubMed] [Google Scholar]
  • 41.Tew JD, Mulsant BH, Haskett RF, et al. Acute efficacy of ECT in the treatment of major depression in the old-old. Am J Psychiatry. 1999;156:1865–1870. doi: 10.1176/ajp.156.12.1865. [DOI] [PubMed] [Google Scholar]
  • 42.Dombrovski AY, Mulsant BH, Haskett RF, Prudic J, Begley AE, Sackeim HA. Predictors of remission after electroconvulsive therapy in unipolar major depression. J Clin Psychiatry. 2005;66:1043–1049. doi: 10.4088/jcp.v66n0813. [DOI] [PubMed] [Google Scholar]
  • 43.Flint AJ, Rifat SL. A prospective study of lithium augmentation in antidepressant-resistant geriatric depression. J Clin Psychopharmacol. 1994;14:353–356. [PubMed] [Google Scholar]
  • 44.Sunderland T, Cohen RM, Molchan S, et al. High-dose selegiline in treatment-resistant older depressive patients. Arch Gen Psychiatry. 1994;51:607–615. doi: 10.1001/archpsyc.1994.03950080019003. [DOI] [PubMed] [Google Scholar]
  • 45.Driscoll HC, Karp JF, Dew MA, Reynolds CF. Getting better, getting well: Understanding and managing partial and non-response to pharmacological treatment of non-psychotic major depression in old age. Drugs Aging. 2007;24:801–814. doi: 10.2165/00002512-200724100-00002. [DOI] [PubMed] [Google Scholar]
  • 46.Mulsant BH, Reynolds CF, Shear MK, Sweet RA, Miller MD. Comorbid anxiety disorders in late-life depression. Anxiety. 1996;2:242–247. doi: 10.1002/(SICI)1522-7154(1996)2:5<242::AID-ANXI6>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 47.Dew MA, Reynolds CF, Houck PR, et al. Temporal profiles of the course of depression during treatment: Predictors of pathways toward recovery in the elderly. Arch Gen Psychiatry. 1997;54:1016–1024. doi: 10.1001/archpsyc.1997.01830230050007. [DOI] [PubMed] [Google Scholar]
  • 48.Lenze EJ, Mulsant BH, Dew MA, et al. Good treatment outcomes in late-life depression with comorbid anxiety. J Affect Disord. 2003;77:247–254. doi: 10.1016/s0165-0327(02)00177-5. [DOI] [PubMed] [Google Scholar]
  • 49.Alexopoulos GS, Katz IR, Bruce ML, et al. Remission in depressed geriatric primary care patients: A report from the PROSPECT study. Am J Psychiatry. 2005;162:718–724. doi: 10.1176/appi.ajp.162.4.718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Andreescu C, Lenze EJ, Dew MA, et al. Effect of comorbid anxiety on treatment response and relapse risk in late-life depression: Controlled study. Br J Psychiatry. 2007;190:344–349. doi: 10.1192/bjp.bp.106.027169. [DOI] [PubMed] [Google Scholar]
  • 51.Saghafi R, Brown C, Butters MA, et al. Predicting 6-week treatment response to escitalopram pharmacotherapy in late-life major depressive disorder. Int J Geriatr Psychiatry. 2007;22:1141–1146. doi: 10.1002/gps.1804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Flint AJ, Rifat SL. Anxious depression in elderly patients. Response to antidepressant treatment. Am J Geriatr Psychiatry. 1997;5:107–115. [PubMed] [Google Scholar]
  • 53.Steffens DC, McQuoid DR. Impact of symptoms of generalized anxiety disorder on the course of late-life depression. Am J Geriatr Psychiatry. 2005;13:40–47. doi: 10.1176/appi.ajgp.13.1.40. [DOI] [PubMed] [Google Scholar]
  • 54.Wetherell JL, Le Roux H, Gatz M. DSM-IV criteria for generalized anxiety disorder in older adults: Distinguishing the worried from the well. Psychol Aging. 2003;18:622–627. doi: 10.1037/0882-7974.18.3.622. [DOI] [PubMed] [Google Scholar]
  • 55.Katon W, Russo J, Frank E, et al. Predictors of nonresponse to treatment in primary care patients with dysthymia. Gen Hosp Psychiatry. 2002;24:20–27. doi: 10.1016/s0163-8343(01)00171-2. [DOI] [PubMed] [Google Scholar]
  • 56.Alexopoulos GS, Kiosses DN, Murphy C, Heo M. Executive dysfunction, heart disease burden, and remission of geriatric depression. Neuropsychopharmacology. 2004;29:2278–2284. doi: 10.1038/sj.npp.1300557. [DOI] [PubMed] [Google Scholar]
  • 57.Oslin DW, Datto CJ, Kalian MJ, Katz IR, Edell WS, TenHave T. Association between medical comorbidity and treatment outcomes in late-life depression. J Am Geriatr Soc. 2002;50:823–828. doi: 10.1046/j.1532-5415.2002.50206.x. [DOI] [PubMed] [Google Scholar]
  • 58.Miller MD, Lenze EJ, Dew MA, et al. Effect of cerebrovascular risk factors on depression treatment outcome in later life. Am J Geriatr Psychiatry. 2002;10:592–598. [PubMed] [Google Scholar]
  • 59.Wise TN, Wiltse CG, losifescu DV, Sheridan M, Xu JY, Raskin J. The safety and tolerability of duloxetine in depressed elderly patients with and without medical comorbidity. Int J Clin Pract. 2007;61:1283–1293. doi: 10.1111/j.1742-1241.2007.01476.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Reynolds CF, Dew MA, Pollock BG, et al. Maintenance treatment of major depression in old age. N Engl J Med. 2006;354:1130–118. doi: 10.1056/NEJMoa052619. [DOI] [PubMed] [Google Scholar]
  • 61.Butters MA, Whyte EM, Nebes RD, et al. The nature and determinants of neuropsychological functioning in late-life depression. Arch Gen Psychiatry. 2004;61:587–595. doi: 10.1001/archpsyc.61.6.587. [DOI] [PubMed] [Google Scholar]
  • 62.Magni G, Palazzolo O, Bianchin G. The course of depression in elderly outpatients. Can J Psychiatry. 1988;33:21–24. doi: 10.1177/070674378803300106. [DOI] [PubMed] [Google Scholar]
  • 63.Kalayam B, Alexopoulos GS. Prefrontal dysfunction and treatment response in geriatric depression. Arch Gen Psychiatry. 1999;56:713–718. doi: 10.1001/archpsyc.56.8.713. [DOI] [PubMed] [Google Scholar]
  • 64.Alexopoulos GS, Meyers BS, Young RC, et al. Executive dysfunction and longterm outcomes of geriatric depression. Arch Gen Psychiatry. 2000;57:285–290. doi: 10.1001/archpsyc.57.3.285. [DOI] [PubMed] [Google Scholar]
  • 65.Burvill PW, Hall WD, Stampfer HG. The prognosis of depression in old age. Br J Psychiatry . 1991;158:64–71. doi: 10.1192/bjp.158.1.64. [DOI] [PubMed] [Google Scholar]
  • 66.Baldwin RC, Benbow SM, Marriott A, Tomenson B. Depression in old age. A reconsideration of cerebral disease in relation to outcome. Br J Psychiatry. 1993;163:82–90. doi: 10.1192/bjp.163.1.82. [DOI] [PubMed] [Google Scholar]
  • 67.Butters MA, Bhalla RK, Mulsant BH, et al. Executive functioning, illness course, and relapse/recurrence in continuation and maintenance treatment of late-life depression: Istherea relationship? Am J Geriatr Psychiatry. 2004;12:387–394. doi: 10.1176/appi.ajgp.12.4.387. [DOI] [PubMed] [Google Scholar]
  • 68.Alexopoulos GS, Murphy CF, Gunning-Dixon FM, et al. Microstructural white matter abnormalities and remission of geriatric depression. Am J Psychiatry. 2008;165:238–244. doi: 10.1176/appi.ajp.2007.07050744. [DOI] [PubMed] [Google Scholar]
  • 69.Lotrich F, Pollock BG, Ferrell RE. Polymorphism of the serotonin transporter: Implications for the use of specific serotonin reuptake inhibitors. Am J Pharmacogenomics. 2001;1:153–164. doi: 10.2165/00129785-200101030-00001. [DOI] [PubMed] [Google Scholar]
  • 70.Kim H, Lim SW, Kim S, et al. Monoamine transporter gene polymorphisms and antidepressant response in koreans with late-life depression. JAMA. 2006;296:1609–1618. doi: 10.1001/jama.296.13.1609. [DOI] [PubMed] [Google Scholar]
  • 71.Murphy GM, Hollander SB, Rodrigues HE, Kremer C, Schatzberg AF. Effects of the serotonin transporter gene promoter polymorphism on mirtazapine and paroxetine efficacy and adverse events in geriatric major depression. Arch Gen Psychiatry. 2004;61:1163–1169. doi: 10.1001/archpsyc.61.11.1163. [DOI] [PubMed] [Google Scholar]
  • 72.Lotrich FE, Pollock BG, Kirshner M, Ferrell RF, Reynolds CF. Serotonin transporter genotype interacts with paroxetine plasma levels to influence depression response in geriatric patients. J Psychiatry Neurosci. In press. [PMC free article] [PubMed] [Google Scholar]
  • 73.Lotrich FE, Pollock BG, Reynolds C, Ferrell RE. SSRI levels interact with serotonin transporter genotype (5-HTTLPR) to influence antidepressant treatment response. Int J Neuropsychopharacol. 2006;9(suppl 1):110. [Google Scholar]
  • 74.Bogner HR, Bruce ML, Reynolds CF, et al. The PROSPECT Group. The effects of memory, attention, and executive dysfunction on outcomes of depression in a primary care intervention trial: The PROSPECT Study. Int J Geriatr Psychiatry. 2007;22:922–929. doi: 10.1002/gps.1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bogner HR, Cary MS, Bruce ML, et al., and the PROSPECT Group. The role of medical comorbidity on outcome of major depression in primary care: The PROSPECT study. Am J Geriatr Psychiatry. 2005;13:861–868. doi: 10.1176/appi.ajgp.13.10.861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Burris KD, Molski TF, Xu C, et al. Aripiprazoie, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther. 2002;302:381–389. doi: 10.1124/jpet.102.033175. [DOI] [PubMed] [Google Scholar]
  • 77.Coward D, Dixon K, Enz A, et al. Partial brain dopamine D2 receptor agonists in the treatment of schizophrenia. Psychopharmacol Bull. 1989;25:393–397. [PubMed] [Google Scholar]
  • 78.Yokoi F, Grunder G, Biziere K, et al. Dopamine D2 and D3 receptor occupancy in normal humans treated with the antipsychotic drug aripiprazoie (OPC 14597): a study using positron emission tomography and [11C]raclopride. Neuropsychopharmacology. 2002;27:248–259. doi: 10.1016/S0893-133X(02)00304-4. [DOI] [PubMed] [Google Scholar]
  • 79.Kane JM, Carson WH, Saha AR, et al. Efficacy and safety of aripiprazoie and haloperidol versus placebo in patients with schizophrenia and schizoaffective disorder. J Clin Psychiatry. 2002;63:763–771. doi: 10.4088/jcp.v63n0903. [DOI] [PubMed] [Google Scholar]
  • 80.Mamo D, Graff A, Mizrahi R, Shammi CM, Romeyer F, Kapur S. Differential effects of aripiprazoie on D, 5-HT, and 5-HT(1A) receptor occupancy in patients with schizophrenia: a triple tracer PET study. Am J Psychiatry. 2007;164:1411–1417. doi: 10.1176/appi.ajp.2007.06091479. [DOI] [PubMed] [Google Scholar]
  • 81.Hirose T, Uwahodo Y, Yamada S, et al. Mechanism of action of aripiprazoie predicts clinical efficacy and a favourable side-effect profile. J Psychopharmacol. 2004;18:375–383. doi: 10.1177/026988110401800308. [DOI] [PubMed] [Google Scholar]
  • 82.Chew ML, Mulsant BH, Pollock BG, et al. A model of anticholinergic activity of atypical antipsychotic medications. Schizophr Res. 2006;88:63–72. doi: 10.1016/j.schres.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 83.Papakostas Gl, Shelton RC, Smith J, Fava M. Augmentation of antidepressants with atypical antipsychotic medications for treatment-resistant major depressive disorder: a meta-analysis. J Clin Psychiatry. 2007;68:826–831. doi: 10.4088/jcp.v68n0602. [DOI] [PubMed] [Google Scholar]
  • 84.Carson WH, Kitagawa H, Nemeroff CB. Drug development for anxiety disorders: new roles for atypical antipsychotics. Psychopharmacol Bull. 2004;38 Suppl 1:38–45. [PubMed] [Google Scholar]
  • 85.Menza MA, Dobkin RD, Marin H. An open-label trial of aripiprazoie augmentation for treatment-resistant generalized anxiety disorder. J Clin Psychopharmacol. 2007;27:207–210. doi: 10.1097/01.jcp.0000248620.34541.bc. [DOI] [PubMed] [Google Scholar]
  • 86.Meller E, Goldstein M, Bohmaker K. Receptor reserve for 5-hydroxytryptaminelA-mediated inhibition of serotonin synthesis: possible relationship to anxiolytic properties of 5-hydroxytryptamine1A agonists. Mol Pharmacol. 1990;37:231–237. [PubMed] [Google Scholar]
  • 87.Blier P, Szabo ST. Potential mechanisms of action of atypical antipsychotic medications in treatment-resistant depression and anxiety. J Clin Psychiatry. 2005;66(suppl 8):30–40. [PubMed] [Google Scholar]
  • 88.Gao K, Muzina D, Gajwani P, Calabrese JR. Efficacy of typical and atypical antipsychotics for primary and comorbid anxiety symptoms or disorders: a review. J Clin Psychiatry. 2006;67:1327–1340. doi: 10.4088/jcp.v67n0902. [DOI] [PubMed] [Google Scholar]
  • 89.Hariri AR, Drabant EM, Weinberger DR. Imaging genetics: perspectives from studies of genetically driven variation in serotonin function and corticolimbic affective processing. Biol Psychiatry. 2006;59:888–897. doi: 10.1016/j.biopsych.2005.11.005. [DOI] [PubMed] [Google Scholar]
  • 90.Davidson RJ. Affective style, psychopathology, and resilience: brain mechanisms and plasticity. Am Psychol. 2000;55:1196–1214. doi: 10.1037//0003-066x.55.11.1196. [DOI] [PubMed] [Google Scholar]
  • 91.Marcus RN, McQuade RD, Carson WH, et al. The efficacy and safety of aripiprazoie as adjunctive therapy in major depressive disorder: a second multicenter, randomized, double-blind, placebo-controlled study. J Clin Psychopharmacol. 2008;28:156–165. doi: 10.1097/JCP.0b013e31816774f9. [DOI] [PubMed] [Google Scholar]
  • 92.Berman RM, Marcus RN, Swanink R, et al. The efficacy and safety of aripiprazoie as adjunctive therapy in major depressive disorder: a multicenter, randomized, double-blind, placebo-controlled study. Clin Psychiatry. 2007;68:843–853. doi: 10.4088/jcp.v68n0604. [DOI] [PubMed] [Google Scholar]
  • 93.Rutherford B, Sneed J, Miyazaki M, et al. An open trial of aripiprazoie augmentation for SSRI non-remitters with late-life depression. Int J Geriatr Psychiatry. 2007;22:986–991. doi: 10.1002/gps.1775. [DOI] [PubMed] [Google Scholar]
  • 94.Sheffrin M, Driscoll HC, Lenze EJ, et al. Getting to remission: use of aripiprazoie for incomplete response in late-life depression. J Clin Psychiatry. In press. doi: 10.4088/jcp.07m03805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Newcomer JW. Metabolic considerations in the use of antipsychotic medications: a review of recent evidence. J Clin Psychiatry. 2007;68 (suppl) 1:20–27. [PubMed] [Google Scholar]
  • 96.American Diabetes Association. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27:596–601. doi: 10.2337/diacare.27.2.596. [DOI] [PubMed] [Google Scholar]
  • 97.Newcomer JW, Haupt DW. The metabolic effects of antipsychotic medications. Can J Psychiatry. 2006;51:480–491. doi: 10.1177/070674370605100803. [DOI] [PubMed] [Google Scholar]
  • 98.Barak Y, Aizenberg D. Effects of olanzapine on lipid abnormalities in elderly psychotic patients. Drugs Aging. 2003;20:893–896. doi: 10.2165/00002512-200320120-00003. [DOI] [PubMed] [Google Scholar]
  • 99.Etminan M, Streiner DL, Rochon PA. Exploring the association between atypical neuroleptic agents and diabetes mellitus in older adults. Pharmacotherapy. 2003;23:1411–1415. doi: 10.1592/phco.23.14.1411.31939. [DOI] [PubMed] [Google Scholar]
  • 100.Micca JL, Hoffmann VP, Lipkovich I, Ahl J, Baker RW, Hardy TA. Retrospective analysis of diabetes risk in elderly patients with dementia in olanzapine clinical trials. Am J Geriatr Psychiatry. 2006;14:62–70. doi: 10.1097/01.JGP.0000192477.05061.96. [DOI] [PubMed] [Google Scholar]
  • 101.Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebocontrolled trials. JAMA. 2005;294:1934–1943. doi: 10.1001/jama.294.15.1934. [DOI] [PubMed] [Google Scholar]
  • 102.Gill SS, Bronskill SE, Normand SL, et al. Antipsychotic drug use and mortality in older adults with dementia. Ann intern Med. 2007;146:775–786. doi: 10.7326/0003-4819-146-11-200706050-00006. [DOI] [PubMed] [Google Scholar]
  • 103.Lingjaerde O, Ahlfors UG, Bech P, Dencker SJ, Elgen K. The UKU side effect rating scale. A new comprehensive rating scale for psychotropic drugs and a cross-sectional study of side effects in neuroleptic-treated patients. Acta Psychiatr Scand Suppl. 1987;334:1–100. doi: 10.1111/j.1600-0447.1987.tb10566.x. [DOI] [PubMed] [Google Scholar]
  • 104.Newcomer JW, Hennekens CH. Severe mental illness and risk of cardiovascular disease. JAMA. 2007;298:1794–1796. doi: 10.1001/jama.298.15.1794. [DOI] [PubMed] [Google Scholar]

Articles from Dialogues in Clinical Neuroscience are provided here courtesy of Taylor & Francis

RESOURCES