Next Article in Journal
Visualization of the Dynamic Brain Activation Pattern during a Decision-Making Task
Previous Article in Journal
Cannabis-Based Medicinal Products in the Management of Emotionally Unstable Personality Disorder (EUPD): A Narrative Review and Case Series
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Vortioxetine Improves Depressive Symptoms and Cognition in Parkinson’s Disease Patients with Major Depression: An Open-Label Prospective Study

by
Diego Santos García
1,*,
Maria Gema Alonso Losada
2,
Icíar Cimas Hernando
3,
Iria Cabo López
4,
Rosa Yáñez Baña
5,
Ruben Alonso Redondo
6,
Jose Manuel Paz González
1,
Carlos Cores Bartolomé
1,
Maria José Feal Painceiras
1,
Maria Cristina Íñiguez Alvarado
1,
Carmen Labandeira
2 and
Iago García Díaz
1
1
Department of Neurology, Hospital Universitario de A Coruña (HUAC), CHUAC (Complejo Hospitalario Universitario de A Coruña), C/As Xubias 84, 15006 A Coruña, Spain
2
CHUVI (Complejo Hospitalario Universitario de Vigo), 36312 Vigo, Spain
3
Hospital Povisa, 36211 Vigo, Spain
4
CHOP (Complejo Hospitalario de Pontevedra), 36071 Pontevedra, Spain
5
CHUO (Complejo Hospitalario Universitario de Orense), 32005 Ourense, Spain
6
Hospital Lucus Augusti, 27003 Lugo, Spain
*
Author to whom correspondence should be addressed.
Brain Sci. 2022, 12(11), 1466; https://doi.org/10.3390/brainsci12111466
Submission received: 15 September 2022 / Revised: 15 October 2022 / Accepted: 26 October 2022 / Published: 29 October 2022
(This article belongs to the Section Neurodegenerative Diseases)

Abstract

:
Depression is frequent in Parkinson’s disease (PD) patients, but the evidence for many antidepressant agents to treat it in PD is insufficient. The aim of the present prospective open-label single-arm study (VOPARK, an open-label study of the effectiveness and safety of VOrtioxetine in PARKinson’s disease patients with depression) was to analyze the effectiveness of vortioxetine on depressive symptoms in PD patients with major depression. The primary efficacy outcome was the change from baseline (VB) at the end of the observational period (12 weeks ± 14 days; V12w) in the 17-item Hamilton Depression Rating Scale (HAM-D17) total score. At VB, all patients had a HAM-D17 total score ≥16. A total of 30 patients (age 66.23 ± 10.27; 73.3% males) were included between February 2021 (first patient, 12/FEB/21) and March 2022 (last patient, 14/MAR/22). At 12 weeks, 27 patients completed the follow-up (90%). The total HAM-D17 total score was reduced by 52.7% (from 21.5 ± 4.75 at VB to 10.44 ± 7.54 at V12w; Cohen’s effect size = −2.5; p < 0.0001) and the response and remission rates were 50% and 43.3%, respectively. Apathy (Apathy Scale; p < 0.0001), cognition (PD-Cognitive Rating Scale; p = 0.007), fatigue (Fatigue Severity Scale; p = 0.014), and quality of life (PDQ-39 (p = 0.001) and EUROHIS-QOL8 (p < 0.0001)) improved at 3 weeks as well. A total of 11 adverse events in 10 patients (33.3%) were reported, one of which was severe (vomiting related to vortioxetine with full recovery after drug withdrawal). Vortioxetine was safe and well tolerated and improved depressive symptoms and other non-motor symptoms in PD patients.

1. Introduction

Depression has been strongly associated with Parkinson’s disease (PD), with previous studies estimating the prevalence rate to be between 2.7 to 90% [1]. A systematic review found the weighted prevalence of major depressive disorder to be 17% in PD patients, that of minor depression to be 22%, and that of dysthymia to be 13% [2]. Clinically significant depressive symptoms were present in 35% of patients. Depression is a key determinant of a reduced quality of life (QoL) in PD [3]. Depression has also been associated with sleep disturbances, fatigue, cognitive dysfunction, and decreased functional ability with impairment of activities of daily living (ADL) [4]. Despite the magnitude and impact of depression in PD, there is a shortage of properly conducted large, randomized, clinical trials of antidepressants in PD. The studies reported are limited by their sample size, use of different scales, and heterogeneous patient populations. The main groups of drugs that have been evaluated are dopamine agonists, tricyclic antidepressants (TCAs), and selective serotonergic and norepinephrine reuptake inhibitors (SSRIs and SNRIs) [5]. There is a lack of evidence for recommendations about how to treat depression in PD patients, with evidence being insufficient for many antidepressant agents [6].
In September 2013, vortioxetine was approved by the FDA for the treatment of major depressive disorder in adults in United States. Vortioxetine has different mechanisms of action; it is a SERT inhibitor; antagonist of 5-HT3, 5-HT7, 5-HTID; partial agonist of 5-HT1B; and a full agonist of 5-HT1A [7]. Moreover, vortioxetine does not appear to interact significantly with the norepinephrine transporters or dopamine transporters, but its administration has been shown to increase extracellular levels of norepinephrine, dopamine, and non-monoamine neurotransmitters including acetylcholine. These effects are also thought to be related to the interaction between vortioxetine and various serotonin receptors [8]. Vortioxetine has been demonstrated to be efficacious, safe, and well-tolerated in many randomized, double-blind, placebo controlled, and/or active treatment-referenced clinical trials [9]. Moreover, vortioxetine seems to be safe when it is administered to elderly patients and, interestingly, it could improve cognitive function compared to other antidepressants [10,11,12,13,14]. Other symptoms such as anxiety, pain, or sleep have been reported to improve with vortioxetine as well [15,16,17]. In PD, some non-motor symptoms (NMS) can be related to decreased levels of dopamine (apathy, anhedonia, cognitive problems, etc.) as well as other neurotransmitters such as serotonin (depression, etc.), noradrenaline (orthostatic hypotension, pain, etc.), or acetylcholine (dementia, etc.) [18]. For all this, vortioxetine could be a useful antidepressant agent with interesting possibilities in the treatment of depression in PD patients. However, there is limited evidence on the use of vortioxetine in patients with PD [19,20,21,22]. Some data suggest that vortioxetine may improve depression severity without significant worsening of motor symptoms and with a remarkable safety profile in PD patients [19,20].
The aim of the present prospective open-label single-arm study (VOPARK, an open-label study of the effectiveness and safety of VOrtioxetine in PARKinson’s disease patients with depression) was to analyze the effectiveness of vortioxetine on depressive symptoms in PD patients with major depression (dPD). Secondary objectives were to analyze the effectiveness of vortioxetine on apathy, cognitive function, fatigue, QoL, and functional capacity for ADL in dPD patients, as well as its safety and tolerability.

2. Materials and Methods

VOPARK is a single-country (Spain), multicenter, observational (phase IV), prospective open-label follow-up study. Eight neurology sites from Galicia (Spain) that deal with dPD participated. A total of 40 consecutive dPD patients were expected to be included in the study, corresponding to 5 patients/site. The inclusion criteria were: (1) a diagnosis of Parkinson’s disease according to the United Kingdom Parkinson’s Disease Society Brain Bank criteria [23]; (2) a diagnosis of major depression according to Diagnostic and Statistical Manual of Mental Health Disorders, 5th Edition (DSM-5) criteria [24]; (3) a 17-item Hamilton Depression Rating Scale (HAM-D17) score ≥ 16 [25,26]; (4) undergoing stable dopaminergic treatment and no expectations of dose or drug changes in the next 3 months; (5) no dementia criteria [27]; (6) older than 40 years old; and (7) wishing to voluntarily participate. The exclusion criteria were: (1) contraindication to being treated with vortioxetine according to the product data; (2) to receive at baseline evaluation or to have received up to 15 days before the baseline evaluation a SSRI and/or SNRI (tricyclics such as amitriptyline and heterocyclics such as mirtazapine or trazodone antidepressant agents were allowed); (3) being pregnant and/or breastfeeding; (4) an incapacity to complete the questionnaires adequately; (5) other disabling concomitant neurological diseases (stroke, severe head trauma, neurodegenerative disease, etc.); (6) other severe and disabling concomitant non-neurological diseases (oncological, autoimmune, etc.); (7) expected impossibility of long-term follow-up; or (8) patients who were already participating in a clinical trial and/or other type of study.
The study visits included 3 visits: (1) VB (baseline); (2) V4w (4 weeks ± 7 days; telephonic visit); and (3) V12w (12 weeks ± 14 days; end of the Observational Period). At Baseline (VB), after providing written informed consent to participate in the study, subjects completed an assessment that included motor symptoms (Hoehn and Yahr (H&Y) [28]; Unified Parkinson’s Disease Rating Scale (UPDRS) part III and part IV [29]), mood (HAM-D17) [25]), apathy (Apathy Scale (AS) [30]), cognition (Parkinson’s Disease Cognitive Rating Scale (PD-CRS) [31]), fatigue (Fatigue Severity Scale (FSS) [32], health-related QoL (the 39-item Parkinson’s disease Questionnaire (PDQ-39) [33]) and global QoL (EUROHIS-QOL8 item index [34]), and autonomy for ADL (Schwab and England Activities of Daily Living Scale (ADLS) [35]. In all the scales/questionnaires, a higher score indicates a more severe affectation—with the exception of the PD-CRS, ADLS, and EUROHIS-QOL8, in which the opposite is the case (Supplementary Material Table S1). The V4w was conducted by phone to check the safety and tolerability of the drug and also to adjust the dose according to the neurologist’s criteria. The Patient and doctor Global Impression of Change (CGI-C) scale [36] together with the same scales administered in the baseline visit—except for the H&Y and UPDRS-IV—were assessed at V12w. All visits were conducted with the patient during the ON-state (after taking his/her medication for PD). Information on sociodemographic aspects, factors related to PD, comorbidities, and treatments was collected.
The primary objective of the study was to analyze the effectiveness of vortioxetine on depressive symptoms at 12 weeks in dPD patients. A reduced version with 17 items [25] of the original version of the 21-item Hamilton Depression Rating Scale [37] was administered, like the rest of the scales, by a neurologist. All the neurologists who participated in the study of each center were experts on PD/movement disorders. According to the major depression DSM-5 criteria, the individual has to be experiencing five or more symptoms during the same 2-week period—with at least one of the symptoms being either (1) a depressed mood or (2) a loss of interest or pleasure—including: (1) depressed mood most of the day, nearly every day; (2) markedly diminished interest or pleasure in all, or almost all, activities most of the day, nearly every day; (3) significant weight loss when not dieting or weight gain, or a decrease or increase in appetite nearly every day; (4) a slowing down of thought and a reduction in physical movement (observable by others, not merely subjective feelings of restlessness or being slowed down); (5) fatigue or loss of energy nearly every day; (6) feelings of worthlessness or excessive or inappropriate guilt nearly every day; (7) a diminished ability to think or concentrate, or indecisiveness, nearly every day; or (8) recurrent thoughts of death, recurrent suicidal ideation without a specific plan, a suicide attempt, or a specific plan for committing suicide. To receive a diagnosis of depression (which was given by the neurologist), these symptoms had to cause the individual clinically significant distress or impairment in social, occupational, or other important areas of functioning. The symptoms also had to not be the result of substance abuse or another medical condition.
Secondary objectives included: (1) an analysis of the effectiveness of vortioxetine on apathy (AS), cognitive function (PD-CRS), and fatigue (FSS); (2) an analysis of the effectiveness of vortioxetine on QoL (health-related (PDQ-39) and global (EUROHIS-QOL8 item index)) and functional capacity for ADL (ADLS); (3) an assessment of the clinical global impression of change according to the patient (PGI-C) and the clinician (CGI-C); and (4) an analysis of the safety and security of vortioxetine in PD patients.
Vortioxetine was administered as a once-daily 10 mg pill, with the possibility of increasing the dose at the neurologist’s indication. However, in patients aged 65 years or older, according to the product data sheet or other considerations made by the neurologist, the first dose should be 5 mg. A dose adjustment could be made after the VB visit as well (from 5–10 mg to 15 and/or 20 mg/day). During the follow-up, any medications other than vortioxetine could not be changed (regimen, doses, etc.)—except if the neurologist considered these changes absolutely necessary. All changes, including PD and non-PD-related medications and the levodopa equivalent daily dose (LEDD), [38] were recorded.

2.1. Data Analysis

Data were processed using SPSS (Statistical Package for Social Sciences) 20.0 for Windows. Continuous variables were expressed as the mean ± SD or median and quartiles, depending on whether they were normally distributed. Each domain of the PDQ-39 was expressed as a percentage: (score/total score) × 100. Relationships between variables were evaluated using the Student’s t-test, the Mann–Whitney U test, or the Spearman’s or Pearson’s correlation coefficient, as appropriate (the distribution of the variables was verified by a one-sample Kolmogorov–Smirnov test). Correlations were considered weak for coefficient values ≤0.29, moderate for values between 0.30 and 0.59, and strong for values ≥0.60.
The primary efficacy outcome was the change from baseline (VB) to the end of the observational period (12 weeks) in the HAM-D17 total score. According to the HAM-D17 total score (ranging from 0 to 52), patients were classified at VB and V12w as not depressed (0–7 points); with mild/minor depression (8–13 points); with moderate depression (14–18 points); with severe depression (19–22 points); or with very severe depression (>23 points). Moreover, other aspects were analyzed at V12w: the completion rate (% of patients receiving vortioxetine); response rate (% of patients with a 50% or greater reduction in the HAM-D17 total score); and remission rate (% of patients without criteria for depression (HAM-D17 total score, 0–7)).
Changes from the VB at V12w in the total scores of the UPDRS-III, AS, PD-CRS, FSS, PDQ-39SI, EUROHIS-QOL8 item index, and ADLS were the secondary efficacy outcome variables. Analyses on efficacy variables were performed with the ITT data set (all subjects who received at least 1 pill of vortioxetine and had a baseline and treatment observation for the primary efficacy outcome measure). A paired-sample t-test or Wilcoxon’s rank sum, McNemar, or marginal homogeneity tests were performed when appropriate for testing changes from the baseline. Cohen’s d formula was applied for measuring the effect size, which was considered to be absent, <0.2; small, 0.2–<0.5; moderate, 0.5–<0.8; large, 0.8–1.3; or very large, ≥1.3. Values of p < 0.05 were considered significant.
The safety data set consists of all subjects for whom the study device was initiated. Safety analyses were assessed by adverse events (AEs). All AEs were coded using the current version of the Medical Dictionary for Regulatory Activities (MedDRA). The number and percentage of subjects with treatment-emergent AEs—ordered according to the MedDRA system organ class and preferred term, their severity, and their relationship to the study treatment, as assessed by the investigator—was provided for all subjects.
Standard protocol approvals, registrations, and patient consent were obtained.
For this study, we received approval from the Comité de Ética de la Investigación Clínica de Galicia, Spain (2020/129; 31/MAR/2020). Written informed consent from all participants in this study was obtained before the beginning of the study. VOPARK was classified by the AEMPS (Agencia Española del Medicamento y Productos Sanitarios) as a Post-authorization Prospective Follow-up study, with the code DSG-VOR-2020-01.

2.2. Data Availability

The protocol and the statistical analysis plan are available on request. De-identified participant data are not available for legal and ethical reasons.

3. Results

A total of 30 patients (aged 66.23 ± 10.27; 73.3% males) were included between February 2021 (first patient, 12/FEB/21) and March 2022 (last patient, 14/MAR/22). The percentage of patients recruited with respect to the expected sample size was 75% (30/40). Two centers did not recruit any patients and two patients failed the screening (neither met the criteria for major depression). Data on sociodemographic aspects, comorbidities, antiparkinsonian drugs, and other therapies are shown in Table 1. The mean time from symptom onset in PD was 4.16 ± 3.11 years. All patients were receiving an antiparkinsonian treatment (levodopa 96.7%; MAO-B inhibitor 76.7%; dopamine agonist 60%; COMT inhibitor 23.3%) and three were undertaking a second-line therapy (one deep brain stimulation; one subcutaneous apomorphine infusion; one enteral levodopa/carbidopa infusion). Benzodiazepines, antidepressant agents (allowed by the protocol), and analgesic drugs were taken by 43.3%, 20%,and 20% of the patients, respectively. The mean LEDD was 765.25 ± 477.63 (ranging from 100 to 2150 mg).
At baseline (VB), the mean HAM-D17 total score was 21.5 ± 4.75 (ranging from 16 to 33) and 70% of the patients had severe or very severe depression (Figure 1B). At 12 weeks, 27 patients completed the follow-up (90%). The mean vortioxetine starting dose was 5.5 ± 1.52 mg/day (5 mg, N = 27; 10 mg, N = 3) whereas the final dose at V12w was 9.61 ± 3.13 (5 mg, N = 5; 10 mg, N = 20; 15 mg, N = 1; 20 mg, N = 1). The total HAM-D17 total score was reduced by 52.7% (from 21.5 ± 4.75 at VB to 10.44 ± 7.54 at V12w; Cohen’s effect size = −2.5; p < 0.0001; Table 2 and Figure 1A) and the response and remission rates were 50% and 43.3%, respectively. Only five patients (18.5%) had severe or very severe depression, whereas 19 out of 27 (70.3%) had no depression or minor depression at the end of follow-up (Figure 1B). By scale items, the effect of improvement was very large on “Depressed mood” (Cohen’s effect size = −2.23; p < 0.0001) and “Work and activities” (Cohen’s effect size = −1.51; p < 0.0001; Supplementary Material Table S2).
A significant change from VB to V12w, indicating an improvement, was observed with other scales (AS, PD-CRS, FSS, PDQ-39, EUROHIS-QOL8; Table 2). No significant changes were observed in the UPDRS-III score (p = 0.483). Regarding cognitive-specific aspects, only a significant improvement in “Delayed verbal memory” was observed (from 4.4 ± 2.67 at VB vs 4.96 ± 2.54; Cohen’s effect size = +0.54; p = 0.047), but a trend of significance in the fronto-subcortical (p = 0.104) and cortical-posterior (p = 0.098) domains was detected (Table 2). Both health-related and global QoL improved after the 12-week follow-up: PDQ-39, from 49.56 ± 19.39 to 28.25 ± 22.6 (Cohen’s effect size = −0.78; p = 0.001); EUROHIS-QOL8, from 25.1 ± 4.99 to 38.25 ± 28.19 ± 4.38 (Cohen’s effect size = +1.35; p < 0.0001) (Figure 2). By domains, a significant improvement was detected in “Emotional well-being” (Cohen’s effect size = −1.28; p < 0.0001) and “Cognition” (Cohen’s effect size = −0.6; p = 0.033), as well as in all domains of the EUROHIS-QOL8 except “Economic capacity” and “Habitat” (Table 2 and Figure 2). After the end follow-up (change from VB to V12w), the improvement detected in mood (HAM-D17 total score) correlated with the improvement observed in apathy (AS; r = 0.465; p = 0.015), fatigue (FSS; r = 0.497; p = 0.008), and the health-related QoL (PDQ39; r = 0.406; p = 0.036) (Table 3). At V12w, 23 patients out of 27 (85.2%) felt better regarding the PGI-C: eight very much improved; nine much improved; six minimally improved; two no changes; and two minimally worse. Similar results were recorded with the CGI-C (Figure 3).
A total of 11 adverse events in 10 patients (33.3%) were reported—one of which was severe (vomiting related to vortioxetine, with full recovery 6 days later after drug withdrawal; Table 4). Gastrointestinal symptoms (nausea, N = 5; vomiting, N = 1; 20%) and dizziness (6.6%) were the most frequent. A full recovery was observed in 9 out of 11 (81.8%) adverse events. Three patients discontinued due to different reasons: personal decision of the patient (N = 1); changes in treatment by a psychiatrist; or an SAE (severe adverse event; N = 1).

4. Discussion

The present study observed that PD patients with major depression improved in terms of their depressive symptoms 3 months after starting with vortioxetine. Moreover, vortioxetine was safe and well-tolerated, and other aspects such as cognitive function, apathy, fatigue, and QoL improved after the 3-month follow-up as well. No motor impairments were detected. Importantly, this is the first prospective, published study specifically designed to analyze the effects of vortioxetine on depressive symptoms and other NMSs in PD.
Depression occurs in around 35% of patients with PD and is often persistent [39]. Many cross-sectional and prospective longitudinal studies have demonstrated that depression is a key factor impacting not only patients’ QoL, but also principal caregivers’ QoL too [3,40,41,42]. The underlying mechanisms of depression in PD are not known in detail, but changes in brain structure, signaling by neurotransmitters, and levels of inflammatory and neurotrophic factors are all suggested to contribute to its development [39,43]. Psychosocial factors, sleep problems, and pain could also have roles in depression [39]. Changes in dopaminergic, noradrenergic, and serotonergic systems in patients with PD might help to explain the incidence of depression in PD patients—so dopaminergic drugs and antidepressants with serotonergic and/or noradrenergic effects could be used to treat depression in PD. However, pramipexole [44] and nortriptyline [45] are the only agents that have shown antidepressant effects in placebo-controlled clinical trials in patients with PD, and evidence regarding recommendations on how to treat depression in PD is poor [6]. Moreover, PD patients very frequently develop other NMSs (apathy, fatigue, pain, cognitive impairment, etc.) that can be related to the diversity of pathways and neurotransmitters (dopamine, serotonin, noradrenaline, acetylcholine, etc.) involved in PD [46]; in this context, vortioxetine—an antidepressant agent with a unique pharmacological profile and a multimodal mechanism of action, with the possibility of increasing levels in the brain of five neurotransmitters (serotonin, dopamine, histamine, noradrenaline, and acetylcholine) [47]—could be a good option for trying to improve not only depressive symptoms, but also other NMSs in patients with PD. Despite this, the evidence on the use of vortioxetine in patients with PD is very scarce [19,20,21,22]—this was the justification for proposing the VOPARK study.
In the VOPARK study, the mean reduction in the HAM-D17 total score was 53% (−10.9 points), the response rate was 50%, and complete remission was observed in 43.3% of patients at the 3-month follow-up. Many randomized, placebo-controlled trials have demonstrated the efficacy of vortioxetine for the treatment of major depressive disorder in adults [48,49,50]. Our results are similar to those reported in patients randomized to vortioxetine in double-blind trials using the HAM-D, with reductions ranging from −11.08 points at 8 weeks with 5 mg/day vortioxetine to −16.23 points at 8 weeks with 10 mg/day vortioxetine [51,52,53,54]. Open-label extension studies with vortioxetine have reported response and remission rates of up to 94% and 83%, respectively [55]. However, the evidence on the use of vortioxetine in patients with PD is very poor, with only five entries in PubMed with the search terms “Parkinson” and “vortioxetine” (September 2022). Russo et al. [19] observed in 150 PD patients treated with vortioxetine—with a mean score at baseline on the HAM-D of 18 points—a reduction on the scale of eight points with 10 mg and seven points with 20 mg, without any severe side effects. However, the data are not published [19]. Miliukhina (article written in Russian) observed in 150 PD patients with mild to moderate depression treated with vortioxetine, a significant improvement in depressive symptoms and anxiety after 12 weeks of treatment—with good tolerability and without motor function impairment [20]. A phase IV trial on the “Tolerability, Safety and Efficacy of Vortioxetine” (VorDe-PD; NCT04301492) in patients with a diagnosis of sustained depression and a HAM-D17 ≥ 14 is ongoing.
In our study, patients improved not only in terms of their depressive symptoms, but also in terms of cognitive function, apathy, and fatigue. Previous studies on elderly patients with major depression have detected that vortioxetine could have a multi-domain beneficial effect on cognitive performance—including executive function, attention/speed of processing, and memory [51,56,57]. Changes in EEG (decreased theta power and increased beta power) in patients with major depression treated with vortioxetine have been reported [58]. A beneficial effect on cognition has been observed in elderly patients with Alzheimer’s disease (AD) as well [11], although a recent randomized, double-blind, placebo-controlled study in 100 AD patients was negative [59]. The efficacy of vortioxetine on cognition has been mainly linked to its action on synaptic serotonin levels via the inhibition of the serotonin (5-HT) transporter (SERT), but its cognition-enhancing properties are hypothesized to also be mediated by other mechanisms such as the release of acetylcholine [60]. Regarding the “dual syndrome” hypothesis of cognitive impairment in PD, there are independent contributions of both dopaminergic denervation on fronto-striatal cognition—including executive impairment—and cholinergic denervation on visuospatial and other attentional impairments in PD [61]. The small sample size could explain why the effect on both cognitive sub-scores in our cohort was not significant, although a significant effect on delayed verbal memory and a clear trend in the posterior cortical sub-score together with the lack of correlation between cognitive function improvement and mood improvement might suggest a possible effect of vortioxetine related to cholinergic function. On the contrary, improvements in apathy and fatigue correlated with improvements in depressive symptoms. Although the pathophysiology of fatigue and apathy in PD is clearly multifactorial, in a proportion of PD patients, these symptoms are associated with depression, dopaminergic depletion in the mesocorticolimbic structures, and disruption of the prefrontal cortex–basal ganglia axis [62]—so, improving depressive symptoms with vortioxetine might produce improvement in apathy and fatigue. A previous meta-analysis found that compared with a placebo, vortioxetine improved physical symptoms in 2105 adults with major depression [63]. Our results agree with these findings—with improvement in insomnia, anxiety, and gastrointestinal and general somatic symptoms according to the HAM-D17. Symptoms such as anxiety, pain, or sleep have previously been reported to improve with vortioxetine [15,16,17]. Moreover, the improvement in depressive symptoms and other NMSs in PD patients in our study was accompanied by an improvement in the patient’s health-related and global QoL. Data from previous studies conducted in adults with major depression demonstrates that vortioxetine improves the patient’s QoL after 6 to 8 weeks of treatment [64].
Vortioxetine was not only effective but also safe and well-tolerated, with the drug maintenance rate at 6 months being very high (90%). In line with the literature [65], gastrointestinal symptoms (nausea and/or vomiting) were the most frequent adverse event, being present in one out of five patients. According to an analysis of data pooled from 11 randomized, placebo-controlled acute treatment studies (3018 patients treated with vortioxetine; six studies included venlafaxine as an active reference) and five open-label, long-term extension studies (2457 patients treated for up to 52 weeks with vortioxetine), the most common treatment-emergent adverse events associated with vortioxetine were nausea (20.9–31.2%) and vomiting (2.9–6.5%)—the incidence of which reached a plateau at 15 mg/day 20 [66]. This complication must be taken into account in PD patients, who are sensitive to the effects of dopaminergic medication. Our recommendation to prevent this complication is to take vortioxetine after eating on a full stomach or before going to bed, and always starting with 5 mg per day for a few days. In PD patients, previous, very limited data suggest that vortioxetine is safe and well-tolerated [19,20] and that it does not seem to worsen motor symptoms [19]. In fact, a very recent study conducted in rats reported that vortioxetine ameliorated motor impairments in rotenone-induced PD via the targeting of TLR-2-mediated neuroinflammation [67].
Our study has some important limitations. The most important is related to the study design itself (open-label study), and since there is not a comparative arm with a placebo, the results should be interpreted with caution. Second, the sample size is small (30 patients out of the 40 initially proposed) and it is possible that the changes observed in some variables (e.g., cognitive sub-scores) were not significant due to this. In fact, due to different problems (i.e., the COVID-19 pandemic and the identification of patients with PD and major depression not taking antidepressant medication), the study was closed before reaching the initially planned sample size (N = 40). Third, the data were collected from patients with PD and major depression, so it is necessary to be cautious when extrapolating the results to PD patients with depressive symptoms without major depression (minor depression, subclinical depression, dysthymia, etc.). On the other hand, this is the first prospective study specifically designed to assess the effect of vortioxetine on depressive symptoms and other NMSs in PD patients with depression. Despite these limitations, the results presented here are novel and are of great interest as there is a lack of knowledge on what effects vortioxetine can have over mood and many other symptoms in PD patients.
In conclusion, this study observed that PD patients improved in terms of their depressive symptoms and other related NMSs (cognition, apathy, and fatigue) 3 months after starting treatment with vortioxetine, with good tolerability. Based on these results and despite some limitations, these findings suggest that vortioxetine could be a good option for treating depression in patients with PD in clinical practice.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/brainsci12111466/s1, Table S1: Description of each of the scales used in this study; Table S2: Change in each item of the HAM-D17 from baseline to final visit.

Author Contributions

D.S.G.: conception, organization, and execution of the project; statistical analysis; writing of the first draft of the manuscript; recruitment and/or evaluation of participants. M.G.A.L.: review and critique; recruitment and/or evaluation of participants. I.C.H.: review and critique; recruitment and/or evaluation of participants. I.C.L.: review and critique; recruitment and/or evaluation of participants. R.Y.B.: review and critique; recruitment and/or evaluation of participants. R.A.R.: review and critique; recruitment and/or evaluation of participants. J.M.P.G.: review and critique; recruitment and/or evaluation of participants. C.C.B.: review and critique; recruitment and/or evaluation of participants. M.J.F.P.: review and critique; collaboration in the preparation of the first manuscript. M.C.Í.A.: review and critique; collaboration in the preparation of the first manuscript. I.G.D.: review and critique; collaboration in the preparation of the first manuscript. C.L.: review and critique; recruitment and evaluation of participants. All authors have read and agreed to the published version of the manuscript.

Funding

The present study is a study promoted by an independent researcher (promoter: Diego Santos García). Lundbeck Spain has financed its expenses. Diego Santos García has received honoraria for educational presentations and advice services from Abbvie, UCB Pharma, Lundbeck, KRKA, Zambon, Bial, Italfarmaco, Teva, Archímedes, Esteve, Stada, and grants from the Spanish Ministry of Economy and Competitiveness (PI16/01575), co-founded by ISCIII (Concesión de subvenciones de Proyectos de Investigación en Salud de la convocatoria 2020 de la Acción Estratégica en Salud 2017–2020 for the project “PROGRESIÓN NO MOTORA E IMPACTO EN LA CALIDAD DE VIDA EN LA ENFERMEDAD DE PARKINSON”). Maria Gema Alonso Losada has received honoraria for educational presentations and advice services from Zambon and Bial. Icíar Cimas Hernando has received honoraria for educational presentations and advice services from KRKA, Italfarmaco, Teva, Zambon, and Bial. Iria Cabo López has received honoraria for educational presentations and advice services from Abbvie and Zambon and Bial. Rosa Yáñez Baña has received honoraria for educational presentations from Teva, Bial, and Zambon. Jose Manuel Paz González has received honoraria for educational presentations and/or advice services from UCB Pharma, Lundbeck, KRKA, and Zambon.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Comité de Ética de la Investigación Clínica de Galicia from Spain (2020/129; 31/MAR/2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The protocol and the statistical analysis plan are available on request. Deidentified participant data are not available for legal and ethical reasons.

Acknowledgments

We would like to thank all the patients who collaborated in this study. Many thanks also to the Fundación Española de Ayuda a la Investigación en Enfermedades Neurodegenerativas of Origen Genético (https://fundaciondegen.org/, accessed on 13 September 2022), ADKNOMA (https://www.adknoma.com/es/, accessed on 13 September 2022), and Lundbeck España (https://www.lundbeck.com/es, accessed on 13 September 2022).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADLS: Schwab and England Activities of Daily Living Scale; AS, Apathy Scale; FSS, Fatigue Severity Scale; HAM-D17, 17-item Hamilton Depression Rating Scale score; H&Y: Hoenh and Yahr; NMS, non-motor symptoms; PD, Parkinson’s disease; PD-CRS, Parkinson’s Disease Cognitive Rating Scale; PDQ-39, 39-item Parkinson’s Disease Quality of Life Questionnaire; QoL, quality of life; UPDRS, Unified Parkinson’s Disease Rating Scale.

References

  1. Reijnders, J.S.A.M.; Ehrt, U.; Weber, W.E.J.; Aarsland, D.; Leentjens, A.F.G. A systematic review of prevalence studies of depression in Parkinson’s disease. Mov. Disord. 2007, 23, 183–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tan, L.C. Mood disorders in Parkinson’s disease. Park. Relat. Disord. 2012, 18 (Suppl. 1), S74–S76. [Google Scholar] [CrossRef]
  3. Schrag, A. Quality of life and depression in Parkinson’s disease. J. Neurol. Sci. 2006, 248, 151–157. [Google Scholar] [CrossRef] [PubMed]
  4. Holroyd, S.; Currie, L.J.; Wooten, G.F. Depression is associated with impairment of ADL, not motor function in Parkinson disease. Neurology 2005, 64, 2134–2135. [Google Scholar] [CrossRef] [PubMed]
  5. Bomasang-Layno, E.; Fadlon, I.; Murray, A.N.; Himelhoch, S. Antidepressive treatments for Parkinson’s disease: A systematic review and meta-analysis. Park. Relat. Disord. 2015, 21, 833–842. [Google Scholar] [CrossRef] [Green Version]
  6. Seppi, K.; Ray Chaudhuri, K.; Coelho, M.; Fox, S.H.; Katzenschlager, R.; Perez Lloret, S.; Weintraub, D.; Sampaio, C.; the collaborators of the Parkinson’s Disease Update on Non-Motor Symptoms Study Group on behalf of the Movement Disorders Society Evidence-Based Medicine Committee. Update on treatments for nonmotor symptoms of Parkinson’s disease-an evidence-based medicine review. Mov. Disord. 2019, 34, 180–198. [Google Scholar] [CrossRef] [Green Version]
  7. Bang-Andersen, B.; Ruhland, T.; Jørgensen, M.; Smith, G.; Frederiksen, K.; Jensen, K.G.; Zhong, H.; Nielsen, S.M.; Hogg, S.; Mørk, A.; et al. Discovery of 1-[2-(2,4-Dimethylphenylsulfanyl)phenyl]piperazine (Lu AA21004): A Novel Multimodal Compound for the Treatment of Major Depressive Disorder. J. Med. Chem. 2011, 54, 3206–3221. [Google Scholar] [CrossRef]
  8. Sanchez, C.; Asin, K.E.; Artigas, F. Vortioxetine, a novel antidepressant with multimodal activity: Review of preclinical and clinical data. Pharmacol. Ther. 2015, 145, 43–57. [Google Scholar] [CrossRef]
  9. Bobo, W.; Kelliny, M.; Croarkin, P.; Moore, K. Profile of vortioxetine in the treatment of major depressive disorder: An overview of the primary and secondary literature. Ther. Clin. Risk Manag. 2015, 11, 1193–1212. [Google Scholar] [CrossRef] [Green Version]
  10. Rosenblat, J.D.; Kakar, R.; McIntyre, R.S. The Cognitive Effects of Antidepressants in Major Depressive Disorder: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Int. J. Neuropsychopharmacol. 2015, 19, pyv082. [Google Scholar] [CrossRef]
  11. Cumbo, E.; Cumbo, S.; Torregrossa, S.; Migliore, D. Treatment Effects of Vortioxetine on Cognitive Functions in Mild Alzheimer’s Disease Patients with Depressive Symptoms: A 12 Month, Open-Label, Observational Study. J. Prev. Alzheimers Dis. 2019, 6, 192–197. [Google Scholar] [CrossRef]
  12. Baune, B.T.; Sluth, L.B.; Olsen, C.K. The effects of vortioxetine on cognitive performance in working patients with major depressive disorder: A short-term, randomized, double-blind, exploratory study. J. Affect. Disord. 2017, 229, 421–428. [Google Scholar] [CrossRef]
  13. McIntyre, R.S.; Lophaven, S.; Olsen, C.K. A randomized, double-blind, placebo-controlled study of vortioxetine on cognitive function in depressed adults. Int. J. Neuropsychopharmacol. 2014, 17, 1557–1567. [Google Scholar] [CrossRef]
  14. Mahableshwarkar, A.R.; Zajecka, J.; Jacobson, W.; Chen, Y.; E Keefe, R.S. A Randomized, Placebo-Controlled, Active-Reference, Double-Blind, Flexible-Dose Study of the Efficacy of Vortioxetine on Cognitive Function in Major Depressive Disorder. Neuropsychopharmacology 2015, 40, 2025–2037. [Google Scholar] [CrossRef] [Green Version]
  15. Yee, A.; Ng, C.G.; Seng, L.H. Vortioxetine Treatment for Anxiety Disorder: A Meta-Analysis Study. Curr. Drug Targets 2018, 19, 1412–1423. [Google Scholar] [CrossRef]
  16. Adamo, D.; Pecoraro, G.; Aria, M.; Favia, G.; Mignogna, M.D. Vortioxetine in the Treatment of Mood Disorders Associated with Burning Mouth Syndrome: Results of an Open-Label, Flexible-Dose Pilot Study. Pain Med. 2020, 21, 185–194. [Google Scholar] [CrossRef]
  17. Cao, B.; Park, C.; Rosenblat, J.D.; Chen, Y.; Iacobucci, M.; Subramaniapillai, M.; Mansur, R.B.; Zuckerman, H.; Lee, Y.; McIntyre, R.S. Changes in sleep predict changes in depressive symptoms in depressed subjects receiving vortioxetine: An open-label clinical trial. J. Psychopharmacol. 2019, 33, 1388–1394. [Google Scholar] [CrossRef]
  18. Chaudhuri, K.R.; Schapira, A.H. Non-motor symptoms of Parkinson’s disease: Dopaminergic pathophysiology and treatment. Lancet Neurol. 2009, 8, 464–474. [Google Scholar] [CrossRef]
  19. Russo, M.; Carrarini, C.; Dono, F.; Ferri, L.; Di Pietro, M.; Onofrj, M. Vortioxetine treatment of depression in Parkinson’s disease [abstract]. In Movement Disorders; 111 RIVER ST, HOBOKEN 07030-5774; Wiley: New York, NJ, USA, 2019; Volume 34, pp. S80–S81. [Google Scholar]
  20. Miliukhina, I.V. The use of vortioxetine for depression in patients with Parkinson’s disease in the early and advanced stages of the disease. Neurol. Neuropsychiatry Psychosom. 2020, 12, 40–45. [Google Scholar] [CrossRef]
  21. Barbato, G.; Marano, P.; Passarella, B.; Pilotto, A.; Riboldazzi, G.; Spagnolo, F.; Vanni, P. Vortioxetine for the treatment of depressive episodes associated with Parkinson’s disease: A case series of six patients. J. Psychopathol. 2020, 26, 248–255. [Google Scholar]
  22. Yoshimura, R.; Ikenouchi, A.; Okamoto, N.; Konishi, Y. Vortioxetine Improved Depressive State In Parkinson’s Disease. Cureus 2021, 13, e15750. [Google Scholar] [CrossRef] [PubMed]
  23. Daniel, S.E.; Lees, A.J. Parkinson’s Disease Society Brain Bank, London: Overview and research. J. Neural Transm. Suppl. 1993, 39, 165–172. [Google Scholar] [PubMed]
  24. DSM-IV-TR. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; American Psychiatric Association: Washington, DC, USA, 2000. [Google Scholar]
  25. Hamilton, M. Development of a rating scale for primary depressive illness. Br. J. Soc. Clin. Psychol. 1967, 6, 278–296. [Google Scholar] [CrossRef] [PubMed]
  26. Naarding, P.; Leentjens, A.F.G.; Van Kooten, F.; Verhey, F.R. Disease-specific properties of the Hamilton Rating Scale for depression in patients with stroke, Alzheimer’s dementia, and Parkinson’s disease. J. Neuropsychiatry Clin. Neurosci. 2002, 14, 329–334. [Google Scholar] [CrossRef] [PubMed]
  27. Dubois, B.; Burn, D.; Goetz, C.; Aarsland, D.; Brown, R.G.; Broe, G.; Dickson, D.; Duyckaerts, C.; Cummings, J.; Gauthier, S.; et al. Diagnostic procedures for Parkinson’s disease dementia: Recommendations from the movement disorder society task force. Mov. Disord. 2007, 22, 2314–2324. [Google Scholar] [CrossRef]
  28. Hoehn, M.M.; Yahr, M.D. Parkinsonism: Onset, progression and mortality. Neurology 1967, 17, 427–442. [Google Scholar] [CrossRef] [Green Version]
  29. Fanhn, S.; Elton, R.L. Members of the UPDRS Development Committee. Unified Parkinson’s Disease Rating Scale. In Recent Developments in Parkinson’s Disease; Fahn, S., Marsden, C.D., Calne, D.B., Goldstein, M., Eds.; Macmillan Health Care Information: Florham Park, NJ, USA, 1987; Volume 2, pp. 153–164. [Google Scholar]
  30. Starkstein, S.E.; Mayberg, H.S.; Preziosi, T.J.; Andrezejewski, P.; Leiguarda, R.; Robinson, R.G. Reliability, validity, and clinical correlates of apathy in Parkinson’s disease. J. Neuropsychiatry Clin. Neurosci. 1992, 4, 134–139. [Google Scholar]
  31. Pagonabarraga, J.; Kulisevsky, J.; Llebaria, G.; García-Sánchez, C.; Pascual-Sedano, B.; Gironell, A. Parkinson’s disease-cognitive rating scale: A new cognitive scale specific for Parkinson’s disease. Mov. Disord. 2008, 23, 998–1005. [Google Scholar] [CrossRef]
  32. Krupp, L.B.; Pollina, D.A. Mechanisms and management of fatigue in progressive neurological disorders. Curr. Opin. Neurol. 1996, 9, 456–460. [Google Scholar] [CrossRef]
  33. Jenkinson, C.; Fitzpatrick, R.; Peto, V.; Greenhall, R.; Hyman, N. The Parkinson’s Disease Questionnaire (PDQ-39): Development and validation of a Parkinson’s disease summary index score. Age Ageing 1997, 26, 353–357. [Google Scholar] [CrossRef] [Green Version]
  34. Da Rocha, N.S.; Power, M.J.; Bushnell, D.M.; Fleck, M.P. The EUROHIS-QOL 8-item index: Comparative psychometric properties to its parent WHOQOL-BREF. Value Health 2012, 15, 449–457. [Google Scholar] [CrossRef] [Green Version]
  35. Schwab, R.S.; England, A.C. Projection Technique for Evaluating Surgery in Parkinson’s Disease. In Proceedings of the Third Symposium on Parkinson’s Disease, Edingurgh, Scotland, 21–22 May 1968; E. And S. Livingstone: Edinburgh, UK, 1969; pp. 152–157. [Google Scholar]
  36. Guy, W. ECDEU Assessment Manual for Psychopharmacology; National Institute of Mental Health, Psychopharmacology Research Branch, Division of Extramural Research Programs, Eds.; U.S. Dept. of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, National Institute of Mental Health, Psychopharmacology Research Branch, Division of Extramural Research Programs: Rockville, MD, USA, 1976; pp. 218–222. [Google Scholar]
  37. Hamilton, M. A rating scale for depression. J. Neurol. Neurosurg. Psychiatry 1960, 23, 56–62. [Google Scholar] [CrossRef]
  38. Schade, S.; Mollenhauer, B.; Trenkwalder, C. Levodopa Equivalent Dose Conversion Factors: An Updated Proposal Including Opicapone and Safinamide. Mov. Disord. Clin. Pr. 2020, 7, 343–345. [Google Scholar] [CrossRef]
  39. Aarsland, D.; Påhlhagen, S.; Ballard, C.G.; Ehrt, U.; Svenningsson, P. Depression in Parkinson disease--epidemiology, mechanisms and management. Nat. Rev. Neurol. 2011, 8, 35–47. [Google Scholar] [CrossRef]
  40. Schrag, A.; Jahanshahi, M.; Quinn, N. What contributes to quality of life in patients with Parkinson’s disease? J. Neurol. Neurosurg. Psychiatry 2000, 69, 308–312. [Google Scholar] [CrossRef] [Green Version]
  41. Santos García, D.; de Deus Fonticoba, T.; Cores, C.; Muñoz, G.; Paz González, J.M.; Martínez Miró, C.; Suárez, E.; Jesús, S.; Aguilar, M.; Pastor, P.; et al. COPPADIS Study Group. Predictors of clinically significant quality of life impairment in Parkinson’s disease. NPJ Park. Dis. 2021, 7, 118. [Google Scholar] [CrossRef]
  42. Santos-García, D.; de Deus Fonticoba, T.; Cores Bartolomé, C.; Íñiguez Alvarado, M.C.; Feal Panceiras, M.J.; Suárez Castro, E.; Canfield, H.; Martínez Miró, C.; Jesús, S.; Aguilar, M.; et al. Predictors of the change in burden, strain, mood, and quality of life among caregivers of Parkinson’s disease patients. Int. J. Geriatr. Psychiatry 2022, 37. [Google Scholar] [CrossRef]
  43. Kano, O.; Ikeda, K.; Cridebring, D.; Takazawa, T.; Yoshii, Y.; Iwasaki, Y. Neurobiology of depression and anxiety in Parkinson’s disease. Parkinsons Dis. 2011, 2011, 143547. [Google Scholar] [CrossRef] [Green Version]
  44. Barone, P.; Poewe, W.; Albrecht, S.; Debieuvre, C.; Massey, D.; Rascol, O.; Tolosa, E.; Weintraub, D. Pramipexole for the treatment of depressive symptoms in patients with Parkinson’s disease: A randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010, 9, 573–580. [Google Scholar] [CrossRef]
  45. Menza, M.; Dobkin, R.D.; Marin, H.; Mark, M.H.; Gara, M.; Buyske, S.; Bienfait, K.; Dicke, A. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology 2009, 72, 886–892. [Google Scholar] [CrossRef] [Green Version]
  46. Tolosa, E.; Gaig, C.; Santamaría, J.; Compta, Y. Diagnosis and the premotor phase of Parkinson’s disease. Neurology 2009, 72 (Suppl. 7), 12–20. [Google Scholar] [CrossRef] [PubMed]
  47. D’Agostino, A.; English, C.D.; Rey, J.A. Vortioxetine (brintellix): A new serotonergic antidepressant. P T J. Formul. Manag. 2015, 40, 36–40. [Google Scholar]
  48. Pae, C.-U.; Wang, S.-M.; Han, C.; Lee, S.-J.; Patkar, A.A.; Masand, P.S.; Serretti, A. Vortioxetine: A meta-analysis of 12 short-term, randomized, placebo-controlled clinical trials for the treatment of major depressive disorder. J. Psychiatry Neurosci. 2015, 40, 174–186. [Google Scholar] [CrossRef] [PubMed]
  49. Thase, M.E.; Mahableshwarkar, A.R.; Dragheim, M.; Loft, H.; Vieta, E. A meta-analysis of randomized, placebo-controlled trials of vortioxetine for the treatment of major depressive disorder in adults. Eur. Neuropsychopharmacol. 2016, 26, 979–993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Koesters, M.; Ostuzzi, G.; Guaiana, G.; Breilmann, J.; Barbui, C. Vortioxetine for depression in adults. Cochrane Database Syst. Rev. 2017, 7, CD011520. [Google Scholar] [CrossRef] [PubMed]
  51. Katona, C.; Hansen, T.; Olsen, C.K. A randomized, doubleblind, placebo-controlled, duloxetine-referenced, fixed-dose study comparing the efficacy and safety of Lu AA21004 in elderly patients with major depressive disorder. Int. Clin. Psychopharmacol. 2012, 27, 215–223. [Google Scholar] [CrossRef]
  52. Henigsberg, N.; Mahableshwarkar, A.R.; Jacobsen, P.; Chen, Y.; Thase, M.E. A randomized, double-blind, placebo-controlled 8-week trial of the efficacy and tolerability of multiple doses of Lu AA21004 in adults with major depressive disorder. J. Clin. Psychiatry 2012, 73, 953–959. [Google Scholar] [CrossRef]
  53. Jain, R.; Mahableshwarkar, A.R.; Jacobsen, P.L.; Chen, Y.; Thase, M.E. A randomized, double-blind, placebo-controlled 6-wk trial of the efficacy and tolerability of 5 mg vortioxetine in adults with major depressive disorder. Int. J. Neuropsychopharmacol. 2012, 16, 313–321. [Google Scholar] [CrossRef] [Green Version]
  54. Mahableshwarkar, A.R.; Jacobsen, P.L.; Chen, Y. A randomized, double-blind trial of 2.5 mg and 5 mg vortioxetine (LuAA21004) versus placebo for 8 weeks in adults with major depressive disorder. Curr. Med. Res. Opin. 2013, 29, 217–226. [Google Scholar] [CrossRef]
  55. Baldwin, D.S.; Hansen, T.; Florea, I. Vortioxetine (Lu AA21004) in the long-term open-label treatment of major depressive disorder. Curr. Med. Res. Opin. 2012, 28, 1717–1724. [Google Scholar] [CrossRef]
  56. Frampton, J.E. Vortioxetine: A Review in Cognitive Dysfunction in Depression. Drugs 2016, 76, 1675–1682. [Google Scholar] [CrossRef]
  57. Harrison, J.E.; Lophaven, S.; Olsen, C.K. Which Cognitive Domains are Improved by Treatment with Vortioxetine? Int. J. Neuropsychopharmacol. 2016, 19, pyw054. [Google Scholar] [CrossRef] [Green Version]
  58. Kim, H.; Baik, S.Y.; Kim, Y.W.; Lee, S. Improved cognitive function in patients with major depressive disorder after treatment with vortioxetine: A EEG study. Neuropsychopharmacol. Rep. 2021, 42, 21–31. [Google Scholar] [CrossRef]
  59. Jeong, H.W.; Yoon, K.H.; Lee, C.H.; Moon, Y.S.; Kim, D.H. Vortioxetine Treatment for Depression in Alzheimer’s Disease: A Randomized, Double-blind, Placebo-controlled Study. Clin. Psychopharmacol. Neurosci. 2022, 20, 311–319. [Google Scholar] [CrossRef]
  60. Bennabi, D.; Haffen, E.; Van Waes, V. Vortioxetine for Cognitive Enhancement in Major Depression: From Animal Models to Clinical Research. Front. Psychiatry 2019, 10, 771. [Google Scholar] [CrossRef] [Green Version]
  61. Kehagia, A.A.; Barker, R.A.; Robbins, T.W. Cognitive impairment in Parkinson’s disease: The dual syndrome hypothesis. Neurodegener. Dis. 2012, 11, 79–92. [Google Scholar] [CrossRef] [Green Version]
  62. Skorvanek, M.; Gdovinova, Z.; Rosenberger, J.; Ghorbani Saeedian, R.; Nagyova, I.; Groothoff, J.W.; Van Dijk, J.P. The associations between fatigue, apathy, and depression in Parkinson’s disease. Acta Neurol. Scand. 2015, 131, 80–87. [Google Scholar] [CrossRef]
  63. Christensen, M.C.; Florea, I.; Lindsten, A.; Baldwin, D.S. Efficacy of vortioxetine on the physical symptoms of major depressive disorder. J. Psychopharmacol. 2018, 32, 1086–1097. [Google Scholar] [CrossRef] [Green Version]
  64. Florea, I.; Danchenko, N.; Brignone, M.; Loft, H.; Rive, B.; Abetz-Webb, L. The effect of vortioxetine on health-related quality of life in patients with major depressive disorder. Clin. Ther. 2015, 37, 2309–2323.e6. [Google Scholar] [CrossRef] [Green Version]
  65. Ekhart, C.; van Hunsel, F.; van Puijenbroek, E.; Chandler, R.; Meldau, E.-L.; Taavola, H.; Norén, G.N. Post-Marketing Safety Profile of Vortioxetine Using a Cluster Analysis and a Disproportionality Analysis of Global Adverse Event Reports. Drug Saf. 2022, 45, 145–153. [Google Scholar] [CrossRef]
  66. Baldwin, D.S.; Chrones, L.; Florea, I.; Nielsen, R.; Nomikos, G.G.; Palo, W.; Reines, E. The safety and tolerability of vortioxetine: Analysis of data from randomized placebo-controlled trials and open-label extension studies. J. Psychopharmacol. 2016, 30, 242–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Nemutlu Samur, D.; Akçay, G.; Yıldırım, S.; Özkan, A.; Çeker, T.; Derin, N.; Tanrıöver, G.; Aslan, M.; Ağar, A.; Özbey, G. Vortioxetine ameliorates motor and cognitive impairments in the rotenone-induced Parkinson’s disease via targeting TLR-2 mediated neuroinflammation. Neuropharmacology 2022, 208, 108977. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) HAM-D17 total score at VB (baseline) and V12w (12 weeks ± 14 days); p < 0.0001. (B) Number of cases with different type of depression at VB vs V12w: not depressed, HAM-D17 0–7; mild/minor depression, HAM-D17 8–13; moderate depression, HAM-D17 14–18; severe depression, HAM-D17 19–22; very severe depression, HAM-D17 > 23 (p < 0.0001). Data are presented as box plots, with the box representing the median and the two middle quartiles (25–75%). p values were computed using the Wilcoxon signed-rank test (A) and the marginal homogeneity test (B). Mild outliers (O) are data points that are more extreme than Q1–1.5. HAM-D17—17-item Hamilton Depression Rating Scale.
Figure 1. (A) HAM-D17 total score at VB (baseline) and V12w (12 weeks ± 14 days); p < 0.0001. (B) Number of cases with different type of depression at VB vs V12w: not depressed, HAM-D17 0–7; mild/minor depression, HAM-D17 8–13; moderate depression, HAM-D17 14–18; severe depression, HAM-D17 19–22; very severe depression, HAM-D17 > 23 (p < 0.0001). Data are presented as box plots, with the box representing the median and the two middle quartiles (25–75%). p values were computed using the Wilcoxon signed-rank test (A) and the marginal homogeneity test (B). Mild outliers (O) are data points that are more extreme than Q1–1.5. HAM-D17—17-item Hamilton Depression Rating Scale.
Brainsci 12 01466 g001
Figure 2. (A) PDQ-39 total score at VB (baseline) and V12w (12 weeks ± 14 days); p = 0.001. Mean score on each domain of the PDQ-39 at VB and V12 expressed as the Summary Index (range 0–100). The difference between both visits was significant for PDQ-39D3 (Emotional well-being; p < 0.0001) and PDQ-39D6 (Cognition; p = 0.033). (B) EUROSHIS-QOL8 total score at VB (baseline) and V12w (12 weeks ± 14 days); p < 0.0001. Mean score on each domain (range 0–5) of the EUROHIS-QOL8 at VB and V12w. The difference between both visits was significant (* p < 0.05; Table 2) for all domains except EUROSHIS-QOL8D7 (Economic capacity) and EUROSHIS-QOL8D8 (Habitat). Data are presented as box plots, with the box representing the median and the two middle quartiles (25–75%). p values were computed using the Wilcoxon signed-rank test. Mild outliers (O) are data points that are more extreme than Q1–1.5. PDQ-39—the 39-item Parkinson’s disease Questionnaire.
Figure 2. (A) PDQ-39 total score at VB (baseline) and V12w (12 weeks ± 14 days); p = 0.001. Mean score on each domain of the PDQ-39 at VB and V12 expressed as the Summary Index (range 0–100). The difference between both visits was significant for PDQ-39D3 (Emotional well-being; p < 0.0001) and PDQ-39D6 (Cognition; p = 0.033). (B) EUROSHIS-QOL8 total score at VB (baseline) and V12w (12 weeks ± 14 days); p < 0.0001. Mean score on each domain (range 0–5) of the EUROHIS-QOL8 at VB and V12w. The difference between both visits was significant (* p < 0.05; Table 2) for all domains except EUROSHIS-QOL8D7 (Economic capacity) and EUROSHIS-QOL8D8 (Habitat). Data are presented as box plots, with the box representing the median and the two middle quartiles (25–75%). p values were computed using the Wilcoxon signed-rank test. Mild outliers (O) are data points that are more extreme than Q1–1.5. PDQ-39—the 39-item Parkinson’s disease Questionnaire.
Brainsci 12 01466 g002
Figure 3. Clinical global impression of changes according to the patient’s (PGI-C) and clinician’s (CGI-C) opinion (N = 27). Total number of patients accorded to each category are shown (y-axis).
Figure 3. Clinical global impression of changes according to the patient’s (PGI-C) and clinician’s (CGI-C) opinion (N = 27). Total number of patients accorded to each category are shown (y-axis).
Brainsci 12 01466 g003
Table 1. Data about sociodemographic aspects, comorbidities, antiparkinsonian drugs and other therapies at baseline (N = 30).
Table 1. Data about sociodemographic aspects, comorbidities, antiparkinsonian drugs and other therapies at baseline (N = 30).
Age66.23 ± 10.27 (48–83)Family cases of depression (%) 33.3
Gender (males) (%)73.3Family cases of PD (%)26.7
Race (%)
   Caucasian100Time from symptoms onset4.16 ± 3.11 (0.33–11)
   Other0
Motor fluctuations (%)60
Civil status (%): Dyskinesia (%)23.3
   Married53.3
   Widowed23.3Treatment for PD (%):
   Single10   Levodopa96.7
   Divorced10   MAO-B inhibitor76.7
   Other3.4   COMT inhibitor23.3
   Dopamine agonist60
Living style (%)    Amantadine 6.7
   With the partner56.7
   Alone20L-dopa daily dose (mg)505.71 ± 392.56 (0–1910)
   With a son/daughter20LEDD (mg)765.25 ± 477.63 (100–2150)
   Other3.3
Other treatments (%):
Habitat (%):    Amitriptiline6.6
   Rural (<5000)10   Trazodone 10
   Semiurban (5000–20,000)26.7   Mirtazapine 3.3
   Urban (>20,000)63.3   Benzodiazepine43.3
   Antipsychotic 3.3
Comorbidities (%):    Analgesic20
   Arterial hypertension40
   Diabetes mellitus6.7Number of anti-PD drugs2.86 ± 1.3 (1–6)
   Dyslipemia36.7Number of non-PD drugs2.82 ± 2.8 (0–9)
   Hiperuricemia3.3Total number of drugs5.68 ± 2.96 (1–13)
   Cardiomyopathy3.3Number of pills for PD4.87 ± 2.26 (1–9.5)
   Cardiac arrhythmia3.3Number of pills for other cause2.62 ± 2.49 (0–8.5)
   Smoking6.7Total number of pills 7.5 ± 2.68 (3–13.75)
   Alcohol consumption0
The results represent % or mean ± SD (range). COMT, catechol-O-methyltransferase; LEDD, levodopa equivalent daily dose; MAO-B, Monoamine oxidase-B; PD, Parkinson’s disease.
Table 2. Change in the HAM-D17 total score and other scales of the study from VB (baseline; N = 30) to V12w (12 weeks ± 14 days; N = 27).
Table 2. Change in the HAM-D17 total score and other scales of the study from VB (baseline; N = 30) to V12w (12 weeks ± 14 days; N = 27).
VBV12wCohen’s d∆VB–V12wp
MOTOR ASSESSMENT
  H&Y-ON2 (1.75–2)N.A.N. AN.A.N.A.
  UPDRS-III-ON23.1 ± 9.85 (9–51)21.63 ± 8.28 (7–39)−0.21−6.90%0.483
  UPDRS−IV2.53 ± 2.04N.A.N.A.N.A.N.A.
NON MOTOR ASSESSMENT
  HAM-D17 21.5 ± 4.75 (16–33)10.44 ± 7.54 (1–30)−2.5−52.70%<0.0001
  AS17.6 ± 6.54 (1–31)11.29 ± 7.18 (1–26)−1.3−35.10%<0.0001
  PD-CRS 80.66 ± 19.14 (29–116)86.81 ± 20.45 (38–127)0.87.94%0.007
  PD-CRS FS sub-score54.17 ± 18.1959 ± 18.960.398.90%0.104
   Immediate verbal memory7.3 ± 2.03 (4–12)7.85 ± 2.14 (4–12)0.467.50%0.091
   Sustained attention7.37 ± 3.21 (0–10)8.33 ± 2.07 (2–10)0.4913%0.094
   Working memory5.9 ± 2.67 (0–9)6.19 ± 2.2 (0–19)0.084.90%0.946
   Clock drawing8.57 ± 2.3 (1–10)9 ± 1.54 (5–10)0.165%0.711
   Delayed verbal memory4.4 ± 2.67 (0–11)4.96 ± 2.54 (0–10)0.5412.70%0.047
   Alternating verbal fluency9.67 ± 4.22 (2–17)10.7 ± 4.71 (2–20)0.4810.60%0.114
   Action verbal fluency12.53 ± 4.68 (5–24)13.07 ± 5.61 (6–27)0.254.30%0.654
  PD-CRS PC sub-score26.5 ± 8.9427.81 ± 7.060.444.90%0.098
   Confrontation naming15.57 ± 4.98 (7–24)17.48 ± 3.78 (8–26)0.4612.30%0.067
   Clock copy9.2 ± 2.14 (1–10)9.37 ± 1.36 (4–10)0.221.80%0.566
  FSS38.7 ± 18.49 (9–76)29.04 ± 16.3 (9–60)−0.77−27.90%0.014
QOL AND AUTONOMY
  PDQ-3949.56 ± 19.39 (15–95)38.25 ± 22.6 (7–83)−0.78−23.80%0.001
   Mobility33.83 ± 22 (0–35)30.37 ± 24.27 (0–35)−0.26−10.20%0.109
   Activities of daily living22.64 ± 18.94 (0–18)20.22 ± 18.37 (0–16)−0.23−10.70%0.273
   Emotional well-being59.72 ± 24.05 (1–24)33.95 ± 24.23 (0–21)−1.28−43.20%<0.0001
   Stigmatization13.96 ± 18.18 (0–10)8.8 ± 13 (0–6)−0.46−36.90%0.092
   Social support11.39 ± 18.88 (0–8)7.41 ± 13.73 (0–6)−0.38−35.20%0.143
   Cognition34.17 ± 26.19 (0–15)27.31 ± 22.14 (0–13)−0.6−20.10%0.033
   Communication14.72 ± 16.47 (0–6)10.49 ± 15.77 (0–6)−0.48−28.70%0.069
   Pain and discomfort40.56 ± 21.52 (0–10)43.21 ± 26.85 (0–12)0.26.50%0.583
  EUROHIS-QOL825.1 ± 4.99 (12–38)28.19 ± 4.38 (20–36)1.3512.30%<0.0001
   Quality of life2.93 ± 0.94 (1–4)3.48 ± 0.7 (2–4)0.918.70%0.004
   Health status2.3 ± 0.87 (1–4)2.78 ± 0.93 (1–4)0.7120.80%0.02
   Energy2.73 ± 0.98 (1–5)3.33 ± 0.92 (1–5)0.9621.90%0.002
   Autonomy for ADL2.8 ± 1.03 (1–5)3.3 ± 0.95 (2–5)1.0117.80%0.002
   Self-esteem2.87 ± 1.04 (1–5)3.37 ± 1 (1–5)0.8617.40%0.004
   Social relationships3.7 ± 0.75 (1–5)4.04 ± 0.51 (3–5)0.619.10%0.025
   Economic capacity3.57 ± 0.72 (2–5)3.59 ± 0.84 (1–5)0.240.50%0.356
   Habitat4.2 ± 0.61 (3–5)4.3 ± 0.61 (3–5)0.512.30%0.059
  ADLS 82.66 ± 11.72 (50–100)84.81 ± 11.22 (50–100)0.322.60%0.227
   Functional dependency (%)23.314.8N.A.N.A.0.687
p values were computed using the Wilcoxon signed-rank or Mc Nemar test. The results represent mean ± SD (range), median [p25, p75] or %. Each domain of the PDQ-39 was expressed as a percentage: (score/total score) x 100. Cohen’s d formula was applied for measuring the effect size. It was considered: ignored, <0.2; small, 0.2 –<0.5; moderate, 0.5 –<0.8; large, 0.8–1.3; very large, ≥1.3. N.A., not applicable. ADLS, Schwab & England Activities of Daily Living Scale; AS, Apathy Scale; FSS, Fatigue Severity Scale; HAM-D17, 17-item Hamilton Depression Rating Scale score; H&Y: Hoenh & Yahr; PD-CRS, Parkinson’s Disease Cognitive Rating Scale; PDQ-39, 39-item Parkinson’s Disease Quality of Life Questionnaire; QoL, quality of life; UPDRS, Unified Parkinson’s Disease Rating Scale.
Table 3. Correlation between the change in the HAM-D17 total score from VB to V12w (∆V12w–VB) and the change in the score in other scales from VB to V12w.
Table 3. Correlation between the change in the HAM-D17 total score from VB to V12w (∆V12w–VB) and the change in the score in other scales from VB to V12w.
∆V12W–VBHAM-D17p
AS0.4650.015
PD-CRS −0.0870.667
PC-CRS FS sub-score−0.2210.268
PD-CRS PC sub-score0.010.961
FSS0.4970.008
PDQ390.4060.036
EUROHIS-QOL8−0.2350.238
ADLS0.1030.609
Pearson correlation coefficient was applied. ADLS, Schwab & England Activities of Daily Living Scale; AS, Apathy Scale; FSS, Fatigue Severity Scale; HAM-D17, 17-item Hamilton Depression Rating Scale score; H&Y: Hoenh & Yahr; PD-CRS, Parkinson’s Disease Cognitive Rating Scale; PDQ-39, 39-item Parkinson’s Disease Quality of Life Questionnaire; QoL, quality of life.
Table 4. Adverse events in patients from VB to V12w.
Table 4. Adverse events in patients from VB to V12w.
N
Total AEs, N11
   Nausea5
   Dizziness2
   Vomiting1
   Headache1
   Helicobacter pylori infection1
COVID-19 disease1
Patients with at least one AE, N (%)10 (33.3)
At least possibly related AEs, N8
   Definitely related2
   Probably related4
   Possibly related 2
   Unrelated3
Patients with at least possibly * related to vortioxetine AEs, N (%)7 (23.3)
Severity, N
   Mild9
   Moderate1
   Severe1
Total SAEs, N1
   Vomiting
Patients with al least one SAE, N (%)1 (3.3)
At least possibly * related to vortioxetine SAEs, N1
Patients with at least possibly related to vortioxetine SAEs, N (%)1 (3.3)
Patients with at least one AE leading to discontinuation, N (%)1 (3.3)
Patients with at least one possibly * related to vortioxetine AE leading to discontinuation N (%)1 (3.3)
Action taken with the AE, N
   Drug withdraw1
   Dose reduction1
   None9
Deaths, N (%)0 (0%)
Outcome of the EA, N
   Fully recovered9
   Improvement (not fully recovered)1
   Unknown1
* Considered “possibly”, “probably” or “definitely” related to treatment (vortioxetine). AE, adverse event; SAE, serious adverse event.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Santos García, D.; Alonso Losada, M.G.; Cimas Hernando, I.; Cabo López, I.; Yáñez Baña, R.; Alonso Redondo, R.; Paz González, J.M.; Cores Bartolomé, C.; Feal Painceiras, M.J.; Íñiguez Alvarado, M.C.; et al. Vortioxetine Improves Depressive Symptoms and Cognition in Parkinson’s Disease Patients with Major Depression: An Open-Label Prospective Study. Brain Sci. 2022, 12, 1466. https://doi.org/10.3390/brainsci12111466

AMA Style

Santos García D, Alonso Losada MG, Cimas Hernando I, Cabo López I, Yáñez Baña R, Alonso Redondo R, Paz González JM, Cores Bartolomé C, Feal Painceiras MJ, Íñiguez Alvarado MC, et al. Vortioxetine Improves Depressive Symptoms and Cognition in Parkinson’s Disease Patients with Major Depression: An Open-Label Prospective Study. Brain Sciences. 2022; 12(11):1466. https://doi.org/10.3390/brainsci12111466

Chicago/Turabian Style

Santos García, Diego, Maria Gema Alonso Losada, Icíar Cimas Hernando, Iria Cabo López, Rosa Yáñez Baña, Ruben Alonso Redondo, Jose Manuel Paz González, Carlos Cores Bartolomé, Maria José Feal Painceiras, Maria Cristina Íñiguez Alvarado, and et al. 2022. "Vortioxetine Improves Depressive Symptoms and Cognition in Parkinson’s Disease Patients with Major Depression: An Open-Label Prospective Study" Brain Sciences 12, no. 11: 1466. https://doi.org/10.3390/brainsci12111466

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop