ArticlePDF Available

[EXPRESS] The multimodal antidepressant vortioxetine causes analgesia in a mouse model of chronic neuropathic pain

Authors:

Abstract

Vortioxetine is a multimodal antidepressant that potently antagonizes 5-HT3 serotonin receptors, inhibits the high affinity serotonin transporter, activates 5-HT1A and 5-HT1B receptors, and antagonizes 5-HT1D and 5-HT7 receptors. 5-HT3 receptors largely mediate the hyperalgesic activity of serotonin that occurs in response to nerve injury. Activation of 5-HT3 receptors contributes to explain why selective serotonin reuptake inhibitors, such as fluoxetine, are not indicated in the treatment of neuropathic pain. Here, we studied the analgesic action of vortioxetine in the chronic constriction injury (CCI) model of neuropathic pain in mice. Vortioxetine was injected once a day for 27 days at doses (10 mg/kg, i.p.) that determine >90% 5-HT3 receptor occupancy in the CNS. The action of vortioxetine was compared to the action of equal doses of the serotonin-noradrenaline reuptake inhibitor, venlafaxine (one of the gold standard drugs in the treatment of neuropathic pain), and fluoxetine. Vortioxetine caused a robust analgesia in CCI mice, and its effect was identical to that produced by venlafaxine. In contrast, fluoxetine was inactive in CCI mice. Vortioxetine enhanced mechanical pain thresholds in CCI mice without changing motor activity, as assessed by the open field and horizontal bar tests. None of the three antidepressants caused analgesia in the complete Freund’s adjuvant (CFA) model of chronic inflammatory pain. These findings raise the attractive possibility that vortioxetine can be effective in the treatment of neuropathic pain, particularly in patients with comorbid depression and cognitive dysfunction.
1
The multimodal antidepressant vortioxetine causes analgesia in a
mouse model of chronic neuropathic pain
Anna Rita Zuena1*, Daniela Maftei1*, Giovanni Sebastiano Alemà1, Francesca Dal
Moro1, Roberta Lattanzi1, Paola Casolini1, Ferdinando Nicoletti1,2
1Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
2IRCCS Neuromed, Pozzilli, Italy
* Co-first author
Corresponding author:
Ferdinando Nicoletti, Department of Physiology and Pharmacology, Sapienza University
of Rome; Piazzale A. Moro, 5, 00185, Rome, Italy.
Email: ferdinandonicoletti@hotmail.com
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
2
Abstract
Vortioxetine is a multimodal antidepressant that potently antagonizes 5-HT3 serotonin
receptors, inhibits the high affinity serotonin transporter, activates 5-HT1A and 5-HT1B
receptors, and antagonizes 5-HT1D and 5-HT7 receptors. 5-HT3 receptors largely
mediate the hyperalgesic activity of serotonin that occurs in response to nerve injury.
Activation of 5-HT3 receptors contributes to explain why selective serotonin reuptake
inhibitors, such as fluoxetine, are not indicated in the treatment of neuropathic pain. Here,
we studied the analgesic action of vortioxetine in the chronic constriction injury (CCI)
model of neuropathic pain in mice. Vortioxetine was injected once a day for 27 days at
doses (10 mg/kg, i.p.) that determine >90% 5-HT3 receptor occupancy in the CNS. The
action of vortioxetine was compared to the action of equal doses of the serotonin-
noradrenaline reuptake inhibitor, venlafaxine (one of the gold standard drugs in the
treatment of neuropathic pain), and fluoxetine. Vortioxetine caused a robust analgesia in
CCI mice, and its effect was identical to that produced by venlafaxine. In contrast,
fluoxetine was inactive in CCI mice. Vortioxetine enhanced mechanical pain thresholds
in CCI mice without changing motor activity, as assessed by the open field and horizontal
bar tests. None of the three antidepressants caused analgesia in the complete Freund’s
adjuvant (CFA) model of chronic inflammatory pain. These findings raise the attractive
possibility that vortioxetine can be effective in the treatment of neuropathic pain,
particularly in patients with comorbid depression and cognitive dysfunction.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
3
Key words: vortioxetine - neuropathic pain - inflammatory pain fluoxetine
venlafaxine.
Introduction
A large body of evidence suggests that serotoninergic pathways descending from the
rostral ventromedial medulla (RVM) to the spinal cord are involved in the top-down
inhibitory control of pain (reviewed in Ref. 1,2). However, the effect of serotonin on
pain modulation in the spinal cord can be either inhibitory or facilitatory depending on
the receptor subtypes which are preferentially activated. Pharmacological studies have
shown that serotonin-induced analgesia is mediated by 5HT7 receptors whereas
hyperalgesia is mediated by 5HT3 receptors 3-6. While inhibition of nociceptive
transmission by serotonin may prevail in conditions of acute pain, serotonin-induced
hyperalgesia contributes to the development of chronic pain after tissue or nerve injury
6-9. Accordingly, shRNA interference-induced knock-down of the serotonin
synthesizing-enzyme, type-2 tryptophan hydroxylase, in the RVM causes analgesia in the
spinal nerve ligation (SNL) model of neuropathic pain in rats 10. An elegant work shed
light into the epigenetic mechanism underlying the hyperalgesic activity of serotonin in
response to nerve injury. SNL in rats caused a down-regulation of the GABA-
synthesizing enzyme, glutamate decarboxylase-65 (GAD-65), in the raphe magnus as a
result of a reduced H3-histone acetylation at the gad65 gene promoter 11. Disinhibition
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
4
of serotoninergic neurons might drive maladaptive changes in serotonin descending
pathways resulting into nociceptive sensitization and chronic pain. These findings
contribute to explain why pure serotoninergic drugs, e.g., selective serotonin reuptake
inhibitors (SSRIs), are ineffective in the treatment of neuropathic pain. In contrast, drugs
that also inhibit noradrenaline uptake, such as venlafaxine, duloxetine, and amitriptyline
show level A rating for efficacy in the treatment of neuropathic pain 12.
We reasoned that activation of 5-HT3 receptors could overcome a potential analgesic
activity of serotonin in neuropathic pain. This gave us the impetus to examine the action
of vortioxetine in a preclinical model of neuropathic pain. Vortioxetine is a new
multimodal antidepressant drug, which inhibits the high affinity serotonin transporter,
and also interacts with different types of serotonin receptors. In particular, vortioxetine
behaves as a full agonist of 5-HT1A receptors, a partial agonist of 5-HT1B receptors, and
an antagonist of 5-HT1B, 5-HT3, and 5-HT7 receptors. Vortioxetine displays the highest
affinity and CNS receptor occupancy for 5-HT3 receptors, and is nearly as potent as the
prototypical 5-HT3 receptor antagonist, ondansetron, in inhibiting the 5-HT3-dependent
Bezold-Jarisch reflex 13-15. Owing to these characteristics, vortioxetine is clinically
effective in improving cognitive dysfunction associated with unipolar depression (see
Discussion and References therein). We hypothesized that vortioxetine could be
beneficial in neuropathic pain by enhancing serotoninergic transmission and potently
inhibiting 5-HT3 receptors at the same time. We tested this hypothesis by comparatively
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
5
evaluating the effect of a chronic treatment with vortioxetine, the serotonin-noradrenaline
reuptake inhibitor (SNRI), venlafaxine, and the SSRI, fluoxetine, in mouse models of
chronic inflammatory and neuropathic pain.
Materials and Methods
Animals
The experiments were carried out in two months old male CD1 mice (Charles River,
Italy). Animals were housed 3-4 per cage in a controlled-temperature room (2123°C,
humidity 4050%) maintained on a 12-h light/dark cycle (light on 07:00 am); food
(Standard Diet Charles River 4RF21, Italy) and water were available ad libitum. All
efforts were made to minimize the number of animals used and to alleviate their
discomfort. All experimental procedures were performed in conformity with the
European Union Directive (2010/63/EU) on the protection of animals used for scientific
purpose and were approved by the Italian Ministry of Health (DDL 26/2014 and previous
legislation; protocol number n° 882/2017-PR).
Drugs
Vortioxetine was provided by H. Lundback A/S (Denmark). Fluoxetine and venlafaxine
were purchased from LKT Laboratories Inc (Minnesota, USA). All drugs were dissolved
in 5% of 2-hydroxypropyl-β-cyclodextrin and administered intraperitoneally (i.p.) at the
dose of 10 mg/kg (50 µl/10 gr b.w.). Control animals received the vehicle alone (50 µl/10
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
6
g, b.w.). The fixed dose of 10 mg/kg for the three drugs was selected on the basis of
previous studies using fluoxetine and/or venlafaxine in models of neuropathic pain [16-
20]. No studies with vortioxetine in pain models had been performed before. The dose
of 10 mg/kg of vortioxetine is used in most of the studies on cognitive function and
depressive-like behavior [21-24].
Induction of chronic inflammatory pain
Tissue inflammation was induced by a single sub-cutaneous (s.c.) injection of 20 µl of
Complete Freund’s Adjuvant (CFA, SigmaAldrich; 1 mg/ml) in the dorsal surface of the
right hind paw. Control mice (n = 7) were injected s.c. with saline in the right hind paw.
Three hours after CFA injection, mice were treated once a day (always at 2.00 p.m.) for
12 days with vortioxetine (n = 8), fluoxetine (n = 9), venlafaxine (n = 8) or their vehicle
(n = 7). Tactile allodynia was assessed before the injection of CFA (day 0) to determine
baseline thresholds and then 1, 3, 5, 7, 10 and 12 days after CFA (and drug) injections.
Induction of chronic neuropathic pain
Mononeuropathy was induced by the chronic constriction injury (CCI) of the sciatic nerve
in mice 25 anesthetized by i.p. injection of tiletamine-zolazepam + xylazine (30 mg/kg
+ 10 mg/kg). The sciatic nerve was exposed and three loose ligatures with 4–0 silk suture
thread were made around the nerve with a 1.0–1.5 mm interval between each of them. In
sham-operated mice, an identical dissection was performed on the same side, except that
the sciatic nerve was not tied. Fourteen days after CCI (day 0) mice were daily injected
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
7
with vortioxetine (n = 10), fluoxetine (n = 9), venlafaxine (n = 9) or vehicle (n = 8) for
28 days. All treatments were performed at 2.00 p.m. Tactile allodynia was measured prior
to CCI to determine baseline thresholds, in the morning (10.00 a.m.) of the starting day
of drug treatments, and then at days 1, 3, 5, 7, 12, 14, 16, 21 and 27 of antidepressant
treatments.
Assessment of tactile allodynia
Tactile allodynia was assessed in the hind paws using calibrated von Frey laments (2
Biological Instruments, Italy) and the up-down method previously described by Chaplan
26. Animals were placed in individual Plexiglas boxes on a raised metal mesh surface
and allowed to acclimatize for 30 min before the test. Testing was initiated with a
medium-sized filament, which was applied for 7 s to the plantar area (plantar territory of
the sural nerve) until the filament bent slightly. If the mouse withdrew or lifted the paw,
the response was considered positive and a one size smaller filament was tried.
Conversely, if no response was observed, a one size larger filament was tried. The
protocol was repeated until five changes in behavior had been observed. The 50% paw
withdrawal threshold (PWT) was determined according to the following equation: Xf +
kD, where Xf is the value of the last von Frey filament used, k is the Dixon value for the
positive/negative pattern, and D is the logarithmic difference between stimuli 27. Tests
were performed in the morning, before drug injections, thus reflecting the analgesic action
of the previous treatment day.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
8
Assessment of locomotor activity and catalepsy
Locomotor activity was assessed in an open field apparatus in all mice used for the study
of neuropathic pain (sham-operated mice and CCI mice treated with vehicle, fluoxetine,
vortioxetine, and venlafaxine) at day 18 after the onset of drug treatments (between day
16 and 21 of pain assessment). The open field apparatus was a Plexiglas squared arena
(40 x 40 cm) with grey walls (40 cm high) and an open roof, located in a sound-attenuated
and dimly-illuminated room. Animals were individually placed in the center of the arena
and allowed to explore for 10 minutes. The arena was cleaned with 50% ethanol solution
before each test. The frequency of line crossing was used to assess total general motor
activity. For this purpose, the floor was divided in 9 virtual quadrants of equal size. A line
crossing was considered when the animal entered another virtual quadrant with all four
paws. Behavioral data were acquired and analyzed using an automated video-tracking
system (Any-Maze, Stoelting, USA).
Catalepsy was assessed in the same mice at day 19 after the onset of treatments by the
horizontal bar test, in which the fore paws of mice were placed on a 15 cm long bar placed
at 4.5 cm above the surface level. The center of the bar was marked to assure identica l
placement of the mice during the test [28]. The time in which the fore paws remained in
the horizontal bar was recorded by an observer who was unaware of the treatments with
a cut-off time of 60 sec. If a mouse did not stay in the position after four attempts, the
cataleptic response time was registered as zero seconds.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
9
Statistical analysis
Statistical analysis of the paw withdrawal threshold (PWT) was performed by two-way
ANOVA for repeated measures followed by Bonferroni post-hoc comparisons, where
appropriate. Statistical significance was set at p<0.05.
Results
No analgesic effect of the selected antidepressants in the CFA model of chronic
inflammatory pain
A significant reduction of tactile withdrawal thresholds was observed in the ipsilateral
hind paw one day after unilateral CFA injection. In CFA mice chronically treated with
vehicle, tactile allodynia remained unaltered in the first 5 days and then gradually
decreased until the last day of pain assessment (12 days following CFA injection). None
of the selected antidepressants, including vortioxetine, caused changes on tactile
sensitivity in the hind paw ipsilateral to CFA injection (Fig. 1A). No changes in paw
withdrawal were found in the contralateral hind paw regardless of drug treatments (Fig.
1B).
Chronic treatment with vortioxetine caused analgesia in the CCI model of neuropathic
pain
Unilateral CCI of the sciatic nerve caused the expected reduction in mechanical pain
thresholds after 14 days with respect to basal thresholds and to thresholds detected in
sham-operated mice. Daily treatments with fluoxetine, venlafaxine, vortioxetine (all at 10
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
10
mg/kg, i.p.) or their vehicle started at day 14 after CCI immediately after pain assessment
(indicated as time 0 in Fig. 2A,B). In CCI mice treated with vehicle, the reduction of
tactile pain thresholds in the ipsilateral hind paw remained unchanged for the whole
duration of the treatment (27 days, corresponding to 41 days after CCI) (Fig. 2A).
Fluoxetine treatment in CCI mice had no effect on tactile allodynia, and paw withdrawal
threshold values were indistinguishable from those detected in mice treated with vehicle
(Fig. 2A). In contrast, treatments with either venlafaxine or vortioxetine caused a robust
analgesia that was initially observed at day 7 and became substantial at day 12 of the
treatment (Fig. 2A). There was no difference between the analgesic effect of vortioxetine
and the effect of the venlafaxine (Fig. 2A). No changes in mechanical thresholds were
found in the hind paw contralateral to CCI regardless of drug treatments (Fig. 2B).
Vortioxetine and the other antidepressants had no effect on spontaneous locomotor
activity and did not induce catalepsy in sham-operated and CCI mice.
To exclude that changes in motor activity could influence the evaluation of pain
thresholds, we measured spontaneous locomotor activity in sham-operated mice, and in
CCI mice treated with vehicle, fluoxetine, venlafaxine, or vortioxetine. Mice were
evaluated in an open field apparatus at day 18 after the onset of drug treatments (pain
thresholds were measured at days 16 and 21). Measurements of total distance travelled
and number of line crossing showed that CCI itself (mice treated with vehicle) and drug
treatments did not change spontaneous locomotor activity, as compared to sham-operated
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
11
mice (Fig. 3A,B). We also evaluated the ability of mice to remove their fore paws from a
horizontal bar (a test for the assessment of catalepsy) at day 19 after the onset of drug
treatments. All mice (sham-operated mice or CCI mice treated with vehicle, fluoxetine,
venlafaxine or vortioxetine) showed no cataleptic behavior, and immediately removed
both fore paws from the horizontal bar (not shown).
Discussion
Full functional recovery in patients affected by unipolar depression relies on the control
of residual symptoms and comorbid disorders. Cognitive dysfunction, which occurs in a
significant proportion of depressed patients, has a negative prognostic value and is
refractory to the majority of antidepressant medications. Depressed patients with
cognitive dysfunction, characterized by impairment in executive functions, speed of
processing, and working and episodic memory, often relapse in spite of continuous
medication, and this leads to a marked reduction in the quality of life 29-32. The
multimodal antidepressant, vortioxetine, has shown a remarkable efficacy in improving
cognitive dysfunction associated with unipolar depression 33-40. In a network
metanalysis of twelve clinical trials made uniform by the use of the digit-symbol
substitution test as a measure of cognitive function, vortioxetine has shown a large
superiority with respect to all other antidepressants drugs, such as SNRIs, SSRIs,
monoamine oxidase inhibitors, and tricyclic antidepressants 41. This unique property
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
12
of vortioxetine largely depends on its ability to potently antagonize 5-HT3 receptors in
the prefrontal cortex and hippocampus, a mechanism that restrains the inhibitory activity
of selected populations of GABAergic interneurons on pyramidal cells 42,43. In spite
of this remarkable feature, vortioxetine is not yet considered as a first-choice drug in
patients with depression associated with comorbid neuropathic pain because the action of
vortioxetine on pain transmission is still unknown. In contrast, antidepressants that inhibit
both serotonin and noradrenaline reuptake, such as duloxetine, venlafaxine, amitriptyline,
are gold standard drugs in the treatment of neuropathic pain 12. Using an established
mouse model of neuropathic pain, we have shown here for the first time that vortioxetine
displays a strong analgesic activity, which is indistinguishable from the activity exhibited
by venlafaxine. To our knowledge, this is the first example of a pure serotoninergic drug
showing analgesic activity in a model of neuropathic pain. In contrast, chronic treatment
with the SSRI fluoxetine did not cause analgesia in the CCI model of neuropathic pain
(see 44 for similar data obtained in rats).
As outlined above, the serotonergic pathway descending from the lower brainstem to the
dorsal horns of the spinal cord becomes hyperalgesic under conditions of neuropathic
pain because of epigenetic processes developing in the raphe magnus and other
mechanisms (see Introduction and References therein). Serotonin-induced hyperalgesia
is mediated by activation of 5-HT3 receptors, which counteract the analgesic activity of
5-HT7 receptors (reviewed in Ref. 1). Vortioxetine acts as a multimodal serotonergic
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
13
antidepressant, behaving as an inhibitor of the high affinity serotonin transporter, a full
agonist of 5-HT1A receptors, a partial agonist of 5-HT1B receptors, and an antagonist of
5-HT1D, -3, and -7 receptors (reviewed in Ref. 39). Measurements of target occupancy
in mice showed that 10 mg/kg of vortioxetine (the dose used in this study) nearly saturates
5-HT3 receptors and the serotonin transporter, but recruits only 20-30% of 5-HT7
receptors [45]. Thus, under conditions of serotonergic hyperactivity, as occurs in
neuropathic pain, vortioxetine will efficiently antagonize 5-HT3 receptors leaving the
vast majority of 5-HT7 receptors unoccupied and, therefore, available for activation by
serotonin. On the basis of these findings we could predict that SSRIs could be effective
as analgesic drugs in neuropathic pain if combined with a potent 5-HT3 receptor
antagonist. This interesting hypothesis warrants further investigation. We were surprised
to find that vortioxetine and venlafaxine were equally effective in causing analgesia in
the CCI model of neuropathic pain in spite of the different mechanism of action of the
two drugs. However, dose-response curves should be performed for a correct comparison
of the analgesic activities of vortioxetine and venlafaxine.
Because antidepressant drugs have intrinsic anti-inflammatory effects [46], we cannot
exclude that vortioxetine causes analgesia by restraining neuroinflammation associated
with neuropathic pain. Along this line, increasing evidence suggests that pain-associated
neuroinflammation is gender-dependent, with a greater involvement of microglia in
males, and T lymphocytes in females (reviewed by [47]. Sex steroids are known to affect
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
14
neuroinflammation by modulating microglial responses to injuries [48-50]. Here, we
examined the analgesic activity of vortioxetine only in males to avoid heterogeneity
caused by the ovarian cycle. It will be interesting to extend the study to non-synchronized,
synchronized, and ovariectomized female mice (w/wo hormonal replacement therapy) to
gain insights into the influence of gender and sex steroids on the analgesic activity of
vortioxetine in models of neuropathic pain.
None of the antidepressants used in the present study caused analgesia in the CFA model
of chronic inflammatory pain. In a previous study, fluoxetine at the dose of 10 mg/kg was
found to enhance mechanical pain thresholds in CFA-injected rats [51]. To our
knowledge, fluoxetine has never been tested in the CFA model in mice, except in one
study in which fluoxetine had no effect on CFA-induced oedema but pain thresholds were
not determined [52]. High doses of venlafaxine (50 and 100 mg/kg) were reported to
enhance mechanical pain thresholds after intraplantar injection of carrageenan in rats
[53], but the drug has never been tested before in CFA mice. The lack of effects of
venlafaxine and vortioxetine on pain thresholds in CFA mice supports the hypothesis that
the molecular and trans-synaptic mechanisms underlying nociceptive sensitization in
inflammatory and neuropathic pain are different. We have shown recently that a 7-day
treatment with the antidepressant, amitriptyline, is effective in causing analgesia in the
CFA model of inflammatory pain in mice 54. Although amitriptyline shares with
venlafaxine the ability to inhibit both serotonin and noradrenaline reuptake, other
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
15
mechanisms, such as inhibition of neurotransmitter receptors or voltage-sensitive sodium
channels, may contribute to the analgesic effect of amitriptyline and account for the
different actions of amitriptyline and venlafaxine in the CFA model of pain.
Our findings suggest that vortioxetine may enrich the therapeutic armamentarium in
patients affected by major depression associated with comorbid neuropathic pain. If
confirmed in clinical studies, the analgesic activity of vortioxetine will be particularly
helpful for patients with cognitive dysfunction, in which the drug shows a greater efficacy
with respect to all other classes of antidepressants. Another advantage is the good profile
of safety and tolerability of vortioxetine, which does not causes significant increases in
body weight and sexual dysfunction (as opposed to SSRIs) or cardiovascular adverse
effects (as opposed to SNRIs) 55-57. The analgesic effect of vortioxetine in the CCI
model paves the way for clinical studies in which vortioxetine should be compared to
gold standard antidepressants in the treatment of neuropathic pain.
Authors Note
The study was supported by an unrestricted grant from Lundbeck, Italy and by Lundbeck
Denmark for the pure ingredient.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
16
Authors contribution
A.R.Z. performed pain experiments and contributed to write the manuscript; D.M.,
F.D.M. and R.L. performed pain experiments; S.A. performed experiments on motor
behaviour; P.C. and F.N. planned research and wrote the manuscript.
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with respect to the research,
authorship, and/or publication of this article.
Funding
The author(s) received no financial support for the research, authorship, and/or
publication of this article.
References
1. Ossipov MH, Dussor GO and Porreca F. Central modulation of pain. J Clin Invest.
2010 Nov;120(11):3779-87. doi: 10.1172/JCI43766.
2. Zhuo M. Descending facilitation. Mol Pain. 2017 Jan;13:1744806917699212. doi:
10.1177/1744806917699212.
3. Bee LA and Dickenson AH. Descending facilitation from the brainstem determines
behavioural and neuronal hypersensitivity following nerve injury and efficacy of
pregabalin. Pain. 2008 Nov 15;140(1):209-23. doi: 10.1016/j.pain.2008.08.008.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
17
4. Chang EY, Chen X, Sandhu A, Li CY and Luo ZD. Spinal 5-HT3 receptors facilitate
behavioural hypersensitivity induced by elevated calcium channel alpha-2-delta-1
protein. Eur J Pain. 2013 Apr;17(4):505-13. doi:10.1002/j.1532-2149.2012.00221.x.
5. Dogrul A, Ossipov MH and Porreca F. Differential mediation of descending pain
facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors. Brain Res. 2009 Jul
14;1280:52-9. doi: 10.1016/j.brainres.2009.05.001.
6. Suzuki R, Rygh LJ and Dickenson AH. Bad news from the brain: descending 5-HT
pathways that control spinal pain processing. Trends Pharmacol Sci. 2004
Dec;25(12):613-7.
7. Porreca F, Ossipov MH and Gebhart GF. Chronic pain and medullary descending
facilitation. Trends Neurosci. 2002 Jun;25(6):319-25.
8. Ren K and Dubner R. Descending modulation in persistent pain: an update. Pain. 2002
Nov;100(1-2):1-6.
9. Ren K and Dubner R. Pain facilitation and activity-dependent plasticity in pain
modulatory circuitry: role of BDNF-TrkB signaling and NMDA receptors. Mol
Neurobiol. 2007 Jun;35(3):224-35.
10. Wei F, Dubner R, Zou S, Ren K, Bai G, Wei D and Guo W. Molecular depletion of
descending serotonin unmasks its novel facilitatory role in the development of persistent
pain. J Neurosci. 2010 Jun 23;30(25):8624-36. doi: 10.1523/JNEUROSCI.5389-09.2010.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
18
11. Zhang Z, Cai YQ, Zou F, Bie B and Pan ZZ. Epigenetic suppression of GAD65
expression mediates persistent pain. Nat Med. 2011 Oct 9;17(11):1448-55.
doi:10.1038/nm.2442.
12. Attal N, Cruccu G, Baron R, Haanpää M, Hansson P, Jensen TS and Nurmikko T.
European Federation of Neurological Societies. EFNS guidelines on the pharmacological
treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2010 Sep;17(9):1113-e88.
doi: 10.1111/j.1468-1331.2010.02999.x.
13. Leiser SC, Li Y, Pehrson AL, Dale E, Smagin G and Sanchez C. Serotonergic
Regulation of Prefrontal Cortical Circuitries Involved in Cognitive Processing: A Review
of Individual 5-HT Receptor Mechanisms and Concerted Effects of 5-HT Receptors
Exemplified by the Multimodal Antidepressant Vortioxetine. ACS Chem Neurosci. 2015
Jul 15;6(7):970-86. doi: 10.1021/cn500340j.
14. Mørk A, Pehrson A, Brennum LT, Nielsen SM, Zhong H, Lassen AB, Miller S,
Westrich L, Boyle NJ, Sánchez C, Fischer CW, Liebenberg N, Wegener G, Bundgaard
C, Hogg S, Bang-Andersen B and Stensbøl TB. Pharmacological effects of Lu AA21004:
a novel multimodal compound for the treatment of major depressive disorder. J
Pharmacol Exp Ther. 2012 Mar;340(3):666-75. doi: 10.1124/jpet.111.189068.
15. Riga MS, Sánchez C, Celada P and Artigas F. Involvement of 5-HT3 receptors in the
action of vortioxetine in rat brain: Focus on glutamatergic and GABAergic
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
19
neurotransmission. Neuropharmacology. 2016 Sep;108:73-81.
doi:10.1016/j.neuropharm.2016.04.023.
16. Jesse CR, Wilhelm EA, Nogueira CW. Depression-like behavior and mechanical
allodynia are reduced by bis selenide treatment in mice with chronic constriction injury:
a comparison with fluoxetine, amitriptyline, and bupropion. Psychopharmacology (Berl).
2010 Dec;212(4):513-22. doi:
10.1007/s00213-010-1977-6.
17. Yalcin I, Tessier LH, Petit-Demoulière N, Doridot S, Hein L, Freund-Mercier MJ,
Barrot M. Beta2-adrenoceptors are essential for desipramine, venlafaxine or reboxetine
action in neuropathic pain. Neurobiol Dis. 2009 Mar;33(3):386-94. doi:
10.1016/j.nbd.2008.11.003.
18. Murad H, Ayuob N. Co-Administration of Pioglitazone Improves Fluoxetine's
Antinociceptive, Neuroprotective, and Antidepressant Effects in Chronic Constriction
Injury in Rats. Pain Physician. 2015 Nov;18(6):609-20.
19. Mizoguchi H, Fukumoto K, Sakamoto G, Jin S, Toyama A, Wang T, Suzumura A,
Sato J. Maternal separation as a risk factor for aggravation of neuropathic pain in later
life in mice. Behav Brain Res. 2018 Jun 20. pii: S0166-4328(18)30294-8. doi:
10.1016/j.bbr.2018.06.015.
20. Zychowska M, Rojewska E, Makuch W, Przewlocka B, Mika J. The influence of
microglia activation on the efficacy of amitriptyline, doxepin, milnacipran, venlafaxine
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
20
and fluoxetine in a rat model of neuropathic pain. Eur J Pharmacol. 2015 Feb 15;749:115-
23. doi: 10.1016/j.ejphar.2014.11.022.
21. Felice D, Guilloux JP, Pehrson A, Li Y, Mendez-David I, Gardier AM, Sanchez C,
David DJ. Vortioxetine Improves Context Discrimination in Mice Through a
Neurogenesis Independent Mechanism. Front Pharmacol. 2018 Mar 12;9:204. doi:
10.3389/fphar.2018.00204.
22. Pehrson AL, Pedersen CS, Tølbøl KS, Sanchez C. Vortioxetine Treatment Reverses
Subchronic PCP Treatment-Induced Cognitive Impairments: A Potential Role for
Serotonin Receptor-Mediated Regulation of GABA Neurotransmission. Front
Pharmacol. 2018 Mar 6;9:162. doi: 10.3389/fphar.2018.00162.
23. Nackenoff AG, Simmler LD, Baganz NL, Pehrson AL, Sánchez C, Blakely RD.
Serotonin Transporter-Independent Actions of the Antidepressant Vortioxetine As
Revealed Using the SERT Met172 Mouse. ACS Chem Neurosci. 2017 May
17;8(5):1092-1100. doi: 10.1021/acschemneuro.7b00038.
24. Guilloux JP, Mendez-David I, Pehrson A, Guiard BP, Repérant C, Orvoën S, Gardier
AM, Hen R, Ebert B, Miller S, Sanchez C, David DJ. Antidepressant and anxiolytic
potential of the multimodal antidepressant vortioxetine (Lu AA21004) assessed by
behavioural and neurogenesis outcomes in mice. Neuropharmacology. 2013 Oct;73:147-
59. doi: 10.1016/j.neuropharm.2013.05.014.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
21
25. Bennett GJ and Xie YK. A peripheral mononeuropathy in rat that produces disorders
of pain sensation like those seen in man. Pain. 1988, 33: 87107.
26. Chaplan SR, Bach FW, Pogrel JW, Chung JM and Yaksh TL. Quantitative assessment
of tactile allodynia in the rat paw. J Neurosci Methods. 1994 Jul;53(1):55-63.
27. Dixon W J. Efficient analysis of experimental observations. Ann. Rev. Pharmacol.
Toxicol. 1980, 20, 441-62. https://doi.org/10.1146/annurev.pa.20.040180.002301
28. Fink-Jensen A, Schmidt LS, Dencker D, Sclein C, Wess J, Wörtwein G and
Woldbye DP. Antipsychotic-induced catalepsy is attenuated in mice lacking the M4
muscarinic acetylcholine receptor. Eur J Pharmacol. 2011 Apr 10;656(1-3):39-44. doi:
10.1016/j.ejphar.2011.01.018.
29. Goeldner C, Ballard TM, Knoflach F, Wichmann J, Gatti S and Umbricht D.
Cognitive impairment in major depression and the mGlu2 receptor as a therapeutic target.
Neuropharmacology. 2013 Jan;64:337-46. doi: 10.1016/j.neuropharm.2012.08.001.
30. Iverson GL, Brooks BL, Langenecker SA and Young AH. Identifying a cognitive
impairment subgroup in adults with mood disorders. J Affect Disord. 2011
Aug;132(3):360-7. doi: 10.1016/j.jad.2011.03.001.
31. McClintock SM, Husain MM, Greer TL and Cullum CM. Association between
depression severity and neurocognitive function in major depressive disorder: a review
and synthesis. Neuropsychology. 2010 Jan;24(1):9-34. doi: 10.1037/a0017336.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
22
32. McDermott LM and Ebmeier KP. A meta-analysis of depression severity and
cognitive function. J Affect Disord. 2009 Dec;119(1-3):1-8. doi:
10.1016/j.jad.2009.04.022.
33. Alvarez E, Perez V and Artigas F. Pharmacology and clinical potential of vortioxetine
in the treatment of major depressive disorder. Neuropsychiatr Dis Treat. 2014 Jul
15;10:1297-307. doi: 10.2147/NDT.S41387.
34. Baune BT, Sluth LB and Olsen CK. 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. 2018 (a) Mar 15;229:421-
428. doi: 10.1016/j.jad.2017.12.056.
35. Christensen MC, Loft H and McIntyre RS. Vortioxetine improves symptomatic and
functional outcomes in major depressive disorder: A novel dual outcome measure in
depressive disorders. J Affect Disord. 2018 Feb;227:787-794. doi:
10.1016/j.jad.2017.11.081.
36. Harrison JE, Lophaven S and Olsen CK. Which Cognitive Domains are Improved by
Treatment with Vortioxetine? Int J Neuropsychopharmacol. 2016 Jun 8. pii: pyw054. doi:
10.1093/ijnp/pyw054.
37. Mahableshwarkar AR, Zajecka J, Jacobson W, Chen Y and Keefe RS. A Randomized,
Placebo-Controlled, Active-Reference, Double-Blind, Flexible-Dose Study of the
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
23
Efficacy of Vortioxetine on Cognitive Function in Major Depressive Disorder.
Neuropsychopharmacology. 2015 Jul;40(8):2025-37. doi: 10.1038/npp.2015.52.
38. McIntyre RS, Harrison J, Loft H, Jacobson W and Olsen CK. The Effects of
Vortioxetine on Cognitive Function in Patients with Major Depressive Disorder: A Meta-
Analysis of Three Randomized Controlled Trials. Int J Neuropsychopharmacol. 2016
Aug 24. pii: pyw055. doi: 10.1093/ijnp/pyw055.
39. Sanchez C, Asin KE and Artigas F. Vortioxetine, a novel antidepressant with
multimodal activity: review of preclinical and clinical data. Pharmacol Ther. 2015
Jan;145:43-57. doi: 10.1016/j.pharmthera.2014.07.001.
40. Vieta E, Sluth LB and Olsen CK. The effects of vortioxetine on cognitive dysfunction
in patients with inadequate response to current antidepressants in major depressive
disorder: A short-term, randomized, double-blind, exploratory study versus escitalopram.
J Affect Disord. 2018 Feb;227:803-809. doi: 10.1016/j.jad.2017.11.053.
41. Baune BT, Brignone M and Larsen KG. A Network Meta-Analysis Comparing
Effects of Various Antidepressant Classes on the Digit Symbol Substitution Test (DSST)
as a Measure of Cognitive Dysfunction in Patients with Major Depressive Disorder. Int J
Neuropsychopharmacol. 2018 (b) Feb 1;21(2):97-107. doi: 10.1093/ijnp/pyx070.
42. Dale E, Zhang H, Leiser SC, Xiao Y, Lu D, Yang CR, Plath N and Sanchez C.
Vortioxetine disinhibits pyramidal cell function and enhances synaptic plasticity in the
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
24
rat hippocampus. J Psychopharmacol. 2014 Oct;28(10):891-902. doi:
10.1177/0269881114543719.
43. Riga MS, Teruel-Martí V, Sánchez C, Celada P and Artigas F. Subchronic
vortioxetine treatment -but not escitalopram- enhances pyramidal neuron activity in the
rat prefrontal cortex. Neuropharmacology. 2017 Feb;113(Pt A):148-155. doi:
10.1016/j.neuropharm.2016.09.024.
44. Hu B, Doods H, Treede RD and Ceci A. Duloxetine and 8-OH-DPAT, but not
fluoxetine, reduce depression-like behaviour in an animal model of chronic neuropathic
pain. Neurosci Lett. 2016 Apr 21;619:162-7. doi: 10.1016/j.neulet.2016.03.019.
45. Leiser SC, Pehrson AL, Robichaud PJ and Sanchez C. Multimodal antidepressant
vortioxetine increases frontal cortical oscillations unlike escitalopram and duloxetine--a
quantitative EEG study in rats. Br J Pharmacol. 2014 Sep;171(18):4255-72. doi:
10.1111/bph.12782.
46. ller N. Immunological aspects of the treatment of depression and schizophrenia.
Dialogues Clin Neurosci. 2017 Mar;19(1):55-63.
47. Coraggio V, Guida F, Boccella S, Scafuro M, Paino S, Romano D, Maione S, Luongo
L. Neuroimmune-Driven Neuropathic Pain Establishment: A Focus on Gender
Differences. Int J Mol Sci. 2018 Jan 17;19(1). pii: E281. doi: 10.3390/ijms19010281.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
25
48. Luongo L, Sajic M, Grist J, Clark AK, Maione S, Malcangio M. Spinal changes
associated with mechanical hypersensitivity in a model of Guillain-Barré syndrome.
Neurosci Lett. 2008 May 30;437(2):98-102. doi: 10.1016/j.neulet.2008.04.019.
49. Garcia-Segura LM, Melcangi RC. Steroids and glial cell function. Glia. 2006 Nov
1;54(6):485-98.
50. Habib P, Beyer C. Regulation of brain microglia by female gonadal steroids. J Steroid
Biochem Mol Biol. 2015 Feb;146:3-14. doi: 10.1016/j.jsbmb.2014.02.018.
51. Boyce-Rustay JM, Zhong C, Kohnken R, Baker SJ, Simler GH, Wensink EJ, Decker
MW, Honore P. Comparison of mechanical allodynia and the affective component of
inflammatory pain in rats. Neuropharmacology. 2010 Feb;58(2):537-43. doi:
10.1016/j.neuropharm.2009.08.008.
52. Maciel IS, Silva RB, Morrone FB, Calixto JB, Campos MM. Synergistic effects of
celecoxib and bupropion in a model of chronic inflammation-related depression in mice.
PLoS One. 2013 Sep 27;8(9):e77227. doi: 10.1371/journal.pone.0077227.
53. Aricioğlu F, Buldanlioğlu U, Salanturoğlu G, Ozyalçin NS. Evaluation of
antinociceptive and anti-inflammatory effects of venlafaxine in the rat. Agri. 2005
Oct;17(4):41-6.
54. Notartomaso S, Mascio G, Bernabucci M, Zappulla C, Scarselli P, Cannella M,
Imbriglio T, Gradini R, Battaglia G, Bruno V and Nicoletti F. Analgesia induced by the
epigenetic drug, L-acetylcarnitine, outlasts the end of treatment in mouse models of
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
26
chronic inflammatory and neuropathic pain. Mol Pain. 2017 Jan;13:1744806917697009.
doi: 10.1177/1744806917697009.
55. Carvalho AF, Sharma MS, Brunoni AR, Vieta E and Fava GA. The Safety,
Tolerability and Risks Associated with the Use of Newer Generation Antidepressant
Drugs: A Critical Review of the Literature. Psychother Psychosom. 2016;85(5):270-88.
doi: 10.1159/000447034.
56. Hughes S, Lacasse J, Fuller RR and Spaulding-Givens J. Adverse effects and
treatment satisfaction among online users of four antidepressants. Psychiatry Res. 2017
Sep;255:78-86. doi: 10.1016/j.psychres.2017.05.021.
57. Meeker AS, Herink MC, Haxby DG and Hartung DM. The safety and efficacy of
vortioxetine for acute treatment of major depressive disorder: a systematic review and
meta-analysis. Syst Rev. 2015 Mar 1;4:21. doi: 10.1186/s13643-015-0001-y.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
27
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
28
Fig. 1 - Chronic treatment with fluoxetine, venlafaxine, and vortioxetine did not
cause analgesia in the CFA mouse model of inflammatory pain.
Mice were injected with CFA in the right hind paw. Control mice received an equal
volume of saline in the hind paw. In CFA mice, systemic treatments with fluoxetine (10
mg/kg), venlafaxine (10 mg/kg), or vortioxetine (10 mg/kg) started three hours after CFA
injection. All drugs were injected i.p. once a day for 12 days. Tactile pain thresholds were
assessed at days 1, 3, 5, 7, 10 and 12 after CFA injection. Pain threshold measured in the
ipsilateral hind paw are shown in (A), where values are means + S.E.M. of 7-9 mice per
group. In order to assess the inflammatory effect of CFA injection we first compared each
of the CFA-injected groups with the group of sham-operated mice treated with vehicle.
Two-way ANOVA for repeated measures (treatment x days) indicated that all CFA-
injected groups were statistically different from sham (CFA/vehicle vs. sham/vehicle:
F1,98= 79.76, p<0.05; CFA/fluoxetine vs. sham/vehicle: F1,103= 16.36, p<0.05;
CFA/venlafaxine vs. sham/vehicle: F1,97= 12.39, p<0.05; CFA/vortioxetine vs
sham/vehicle F1,97= 11.99, p<0.05). Then we assessed the treatment effect comparing all
CFA-injected groups without the group of sham-operated mice treated with vehicle. Two-
way ANOVA for repeated measure indicated a significant effect of the day (F3,199= 98.29,
p<0.05) but no effect of antidepressant treatment (F3,199= 1.33, p= 0.3). Pain threshold in
the contralateral hind paw are shown in (B).
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
29
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
30
Fig. 2 Analgesic effects of vortioxetine and venlafaxine in the CCI model of
neuropathic pain.
Mice were subjected to CCI of the right sciatic nerve and treated, after 14 days, with
either fluoxetine (10 mg/kg), venlafaxine (10 mg/kg), vortioxetine (10 mg/kg), or their
vehicle. Treatments were performed i.p. once a day for 27 days (i.e., up to 41 days after
CCI). Tactile pain thresholds in the ipsilateral hind paw are shown in (A) where values
are means + S.E.M. of 8-10 mice per group. In order to assess the induction of neuropathic
pain after CCI we first compared all CCI groups with the group of sham-operated mice
treated with vehicle. Two-way ANOVA for repeated measures (treatment x days)
indicated that each of the CCI groups was statistically different from the group sham-
operated mice treated with vehicle (CFA/vehicle vs. sham/vehicle: F1,180= 255.25,
p<0.05; CFA/fluoxetine vs. sham/vehicle: F1,180= 264.59, p<0.05; CFA/venlafaxine vs.
sham/vehicle: F1,180= 98.16, p<0.05; CFA/vortioxetine vs. sham/vehicle: F1,190= 156.51,
p<0.05). Then, we assessed the treatment effect comparing all CCI groups without the
group of sham-operated mice. Two-way ANOVA for repeated measure indicated a
significant effect of the day (F3,370= 18.17, p<0.05), a significant effect of the treatment
(F3,370= 23.97, p<0.05) and an interaction between the two factors (F3,370= 10.61, p<0.05).
Values obtained in CCI mice treated with vortioxetine or venlafaxine were significantly
different with respect to values obtained in CCI mice treated with vehicle or fluoxetine
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
31
from day 12 of treatment (*p<0.05, Bonferroni’s post-hoc test). Mechanical pain
thresholds in the contralateral hind paw are shown in (B).
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
32
Fig. 3 Antidepressant treatment did not cause changes in spontaneous locomotor
activity in mice subjected to CCI of the sciatic nerve.
Spontaneous locomotor activity was assessed in mice subjected to CCI of the right sciatic
nerve treated with antidepressants or their vehicle (see legend of Fig. 2), and in sham-
operated mice at day 32 after surgery (day 18 of treatments in CCI mice). Distance
travelled (in meters) (A) and numbers of line crossing (B) were determined in an open
field apparatus (10 min of total observation). Values are means + S.E.M. of 8-10 mice
per group.
Molecular Pain
MPX Express
Accepted on 27 September, 2018
This article can be cited as 10.1177/1744806918808987
... Several hypotheses have been proposed in the literature regarding the basic mechanisms underlying the effect of Vo on pain. Accordingly, the blockade of serotonin transport by Vo, the increase of neurotransmitters such as noradrenaline and 5-HTin central and peripheral nervous system synapses, and its direct effect on receptor activity that can modulate pain transmission may explain the impact of Vo on pain (27,28). In addition to its immunomodulatory, antioxidant, and anti-inflammatory effects, Vo is considered potentially effective in chronic pain by increasing BDNF levels (29,30). ...
... In an experimental study, the effect of Vo on pain appeared after 7 days and gradually increased. Similarly, we observed a decrease in the pain complaints of many of our patients as of the end of the first month of treatment (28). Case series in the literature on Vo suggests that Vo may also have a positive effect on mood and restless legs syndrome symptoms through its activity on dopamine and gamma-aminobutyric acid (GABA) (38). ...
... In our study, no patient discontinued medication or was excluded from the study due to side effects, except for one patient who was voluntarily excluded from the study due to persistent dizziness. The findings of this study support the clinical findings reported in the literature regarding the efficacy of Vo on NP (28,42). In a study conducted on diabetic rats, Yucel et al. (43) showed that Vo, in addition to its neutral activity on glycemic control, significantly improved diabetes-induced hyperalgesia and allodynia responses without affecting motor coordination. ...
Article
Full-text available
Introduction and objective Several studies revealed the therapeutic potential of vortioxetine (Vo) for pain. In this context, we aimed to evaluate the efficacy of Vo as a safe and tolerable novel pharmacologic agent in treating neuropathic pain (NP) in patients with major depressive disorder (MDD). Materials and methods The population of this cross-sectional prospective study consisted of all consecutive patients who were newly diagnosed with MDD by a neurology doctor at a psychiatric clinic and had NP for at least 6 months. All patients included in the sample were started on Vo treatment at 10 mg/day. They were assessed with Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), Self-Reported Leeds Assessment of Neuropathic Symptoms and Signs (S-LANSS), Douleur Neuropathique 4 Questions (DN4), Montreal Cognitive Assessment (MoCA), and Neuropathic Pain Impact on Quality of Life (NePIQoL) at the beginning of treatment and during the follow visits conducted at the end of the first, second and third months of the treatment. During these follow-up visits, patients were also queried about any side effects of Vo. Results The mean age of 50 patients included in the sample, 76% of whom were female, was 45.8 ± 11.2 years. There was a significant reduction in patients’ NP complaints based on DN4 and S-LANNS, the subscales of NePIQoL, and significant improvement in MoCA. There was a significant reduction in patients’ NP complaints based on DN4 and S-LANNS scores and a significant improvement in scores of the subscales of NePIQoL and MoCA. Conclusion The study’s findings indicate that Vo, with its multiple mechanisms of action, can effectively treat NP independently of its mood-stabilizing effect. Future indication studies for Vo are needed to establish Vo’s efficacy in treating NP.
... [33]. Likewise, several preclinical and clinical studies have recently demonstrated the effectiveness of vortioxetine in managing CP [15,[34][35][36][37], although more evidence is needed. ...
... SSRIs are ineffective in treating neuropathic pain because they induce the disinhibition of serotoninergic neurons, which may result in maladaptive changes in the descending serotonin pathways, resulting in nociceptive sensitization and chronic pain [42][43][44]. Considering that 5-HT3 receptors are involved in 5HT-induced hyperalgesia, the ability of vortioxetine to inhibit 5-HT3 receptors and other 5-HT receptors, modulating rather than only promoting serotoninergic transmission [39], makes it an interesting candidate for CP treatment [3,34]. ...
Article
Full-text available
Chronic pain (CP) and depression/anxiety often coexist, worsening each other's symptoms. Treating this comorbidity is challenging. Tricyclic antidepressants and serotonin noradrenaline reuptake inhibitors are the first-line treatment options for this comorbidity, although sometimes they are not effective and/or well tolerated by patients, and there is little clinical evidence that selective serotonin reuptake inhibitors are useful for controlling CP. The antidepressant vortioxetine, with a multimodal mechanism that may help reduce pain, has proven clinical efficacy in patients with major depressive disorder (MDD). This study investigated vortioxetine's effectiveness for MDD and CP in clinical practice. This was a 3-month, multicenter, prospective, open-label, non-interventional pharmacoepidemiologic study. Patients (n = 64) with MDD (9-item Patient Health Questionnaire [PHQ-9] score ≥ 15) and CP (visual analogue scale [VAS] score ≥ 4) were treated with vortioxetine for 3 months (initiated with 10 mg/day, with flexible dosing thereafter [5–20 mg/day]). VAS, Clinical Global Impression (CGI), and Patient Global Impression (PGI) scales were used at baseline and at 1 and 3 months. Brief Pain Inventory (BPI), PHQ-9 scale, and Satisfaction with Medicines Questionnaire (SATMED-Q) were used at baseline and at 3 months. Adverse Events (AEs) were recorded. Descriptive statistics, chi-square tests, and Student's t-tests were used for paired data. MDD patients showed a statistically significant improvement in VAS from baseline (mean [standard deviation (SD)]: 7.42 [0.69]) to 1 month (mean [SD]: 6.1 [0.81], P < 0.001) and 3 months (mean [SD]: 5.09 [1.26], P < 0.0001). Similarly, BPI and PHQ-9 scores showed significant improvement from baseline (mean [SD]: 6.20 [0.80] and 16.63 [1.47], respectively) to 3 months (mean [SD]: 4.73 [0.98] and 7.30 [2.60], P < 0.0001, respectively). Patients showed clinical improvement with CGI and PGI scales and reported being satisfied with the treatment in the SATMED-Q. A few mild EAs were registered. Vortioxetine can relieve depressive and pain symptoms, with a good safety profile, in patients with MDD and CP.
... Basic research has shown that serotonin-induced hyperalgesia involved in chronic pain is strongly mediated by 5-HT 3 receptors [38]. Vortioxetine, as opposed to fluoxetine, caused a robust analgesic effect, similar to that produced by an SNRI, so it may be effective in patients with neuropathic pain, particularly with comorbid depression [39]. ...
... In summary, its differential action on several 5-HT targets and the ability to modulate a wide range of neurotransmitters confers vortioxetine not only antidepressant but also procognitive effects, as well as promising further analgesic and anti-inflammatory properties [39,40,45]. ...
Article
Full-text available
Introduction Vortioxetine is a multimodal-acting antidepressant that provides improvements on cognitive function aside from antidepressants and anxiolytic effects. Vortioxetine has been found to be one of the most effective and best tolerated options for major depressive disorder (MDD) in head-to-head trials. Areas covered The present review intends to gather the most relevant and pragmatic data of vortioxetine in MDD, specially focusing on new studies that emerged between 2015 and 2020. Expert opinion Vortioxetine is the first antidepressant that has shown improvements both in depression and cognitive symptoms, due to the unique multimodal mechanism of action that combine the 5-HT reuptake inhibition with modulations of other key pre- and post-synaptic 5-HT receptors (agonism of 5-HT1A receptor, partial agonism of 5-HT1B receptor, and antagonism of 5-HT3, 5-HT1D and 5-HT7 receptors). This new mechanism of action can explain the dose-dependent effect and can be responsible for its effects on cognitive functioning and improved tolerability profile. Potential analgesic and anti-inflammatory properties observed in preclinical studies as well as interesting efficacy and tolerability results of clinical studies with specific target groups render it a promising therapeutic option for patients with MDD and concomitant conditions (as menopause symptoms, pain, inflammation, apathy, sleep and/or metabolic abnormalities).
... Although this form addresses the problem of poor water solubility, long-term VTX hydrobromide therapy is frequently associated with persistent respiratory irritation and digestive dysfunction, both of which are attributed to hydrobromide. Furthermore, VTX was notorious for its poor oral absorption and fast metabolism (Zhang et al., 2019;Zuena et al., 2018). To solve these problems with a stable dosage form, we have developed two techniques for dissolution, swelling, adherence, and penetration enhancements for the VTX. ...
... Vortioxetine is a relatively new antidepressant with improved tolerability profile and cognition-enhancing effects (De Diego-Adeliño et al., 2022). Its potential to relieve chronic pain has recently been demonstrated in animals (constriction and chemotherapy-induced neuropathy) (Micov et al., 2020;Zuena et al., 2018) and humans (burning mouth syndrome) (Adamo et al., 2021). Vortioxetine's unique mechanism of action combines inhibition of the serotonin (5-hydroxytryptamine, 5-HT)-reuptake transporter (SERT) and direct action on serotonergic receptors that may provide pain relief (antagonism of 5-HT 3 and stimulation of 5-HT 1B and 5-HT 1A receptors) (Cortes-Altamirano et al., 2018;De Diego-Adeliño et al., 2022). ...
Article
Full-text available
Background and Purpose Vortioxetine, a multimodal‐acting antidepressant, has recently shown analgesic properties. We aimed to investigate its prophylactic effect in the osteoarthritis (OA) model and gain insights into the underlying molecular mechanisms. Duloxetine was studied as a reference. Experimental Approach In the monoiodoacetate (MIA)‐induced rat model of knee OA, pain‐related behaviour was assessed in weight‐bearing and Von Frey tests. Antidepressants were administered orally once daily for 28 days. Gene expression of pain‐related mediators (Ngf, Il‐1β, Tnf‐α, Bdnf, and Tac1 encoding substance P) and oxidative stress parameters were determined after completion of the treatment/behavioural testing protocol. Key Results Vortioxetine and duloxetine dose dependently reduced weight‐bearing asymmetry and mechanical hyperalgesia of the paw ipsilateral to the MIA‐injected knee. Vortioxetine reduced the increased Ngf mRNA expression in the MIA‐injected knees to the level in sham‐injected counterparts. It reduced oxidative stress parameters in the affected knees, more effectively in females than males. Duloxetine showed no effect on Ngf mRNA expression and oxidative stress. Both antidepressants decreased mRNA expression of pain‐related mediators in the lumbar L3–L5 ipsilateral DRGs and spinal cords, which were up‐regulated in MIA‐injected rats. This effect was male‐specific. Conclusion and Implications Vortioxetine may be effective against the development of chronic pain in OA. Its antihyperalgesic effect may be mediated, at least in part, by normalization of NGF expression in the affected joint. Decrease of localized oxidative stress and of expression of pain‐related mediators that contribute to central sensitization are also involved in vortioxetine's antihyperalgesic effect, in a sex‐specific pattern.
... In another study, Zuena et al. investigated the efficacy of vortioxetine against neuropathic pain in mice using a model of chronic constriction injury. The authors reported that the administration of this drug at a daily dose of 10 mg/kg (i.p.) for 27 days significantly increased the mechanical pain thresholds of the animals without changing their motor activity [16]. In a different study examining the effects of vortioxetine on pain hypersensitivity in a model of oxaliplatin-induced neuropathy in mice, it was suggested that both repeated prophylactic and acute therapeutic regimens of this drug (1-10 mg/kg, p.o.) dose-dependently reduce mechanical allodynia in the von Frey test and cold allodynia in the acetone test [18]. ...
Article
Full-text available
The therapeutic potential of vortioxetine on mechanical hyperalgesia/allodynia was investigated in rats with streptozotocin-induced diabetes, and its possible mechanism of action was elucidated in this study. The obtained findings demonstrated that subacute vortioxetine treatment (5 and 10 mg/kg for 2 weeks) increased the reduced paw-withdrawal thresholds of diabetic rats both in the Randall–Selitto and Dynamic plantar tests. Moreover, the falling latencies of animals did not change in the Rota-rod assessments. These results suggest that vortioxetine administration significantly improved diabetes-induced hyperalgesia and allodynia responses in the rats without affecting their motor coordination. The vortioxetine (5 mg/kg)-induced antihyperalgesic and antiallodynic effects were reversed by AMPT, yohimbine, ICI 118,551, sulpiride and atropine pre-treatments, suggesting the involvement of the catecholaminergic system, α2- and β2-adrenoceptors, D2/3 dopaminergic receptors and cholinergic muscarinic receptors in the exhibited pharmacological activity, respectively. Moreover, the data from the immunohistochemical studies indicated that the inhibition of c-Fos overexpression in dorsal horn neurons also mediates the beneficial effect of this drug. Vortioxetine induced no difference in plasma glucose levels in diabetic rats. If clinical studies confirm these findings, the concomitant beneficial effect of vortioxetine on mood disorders and its neutral activity profile on glycemic control may make it an alternative drug for the treatment of neuropathic pain.
Article
In multiple sclerosis (MS), alongside the physical symptoms, individuals often grapple with anxiety and depressive symptoms as prevalent comorbidity. Mood disturbances, frequently undertreated in clinical practice, significantly impact the quality of life of individuals with MS, exacerbating disability and hindering overall well-being. Furthermore, traditional antidepressant therapies are often associated with adverse events, such as sexual side effect, weight gain, which could limit their use in these patients. Vortioxetine is one of the most innovative antidepressant drugs in the current pharmacopeia. Its pharmacological profile includes serotonin reuptake inhibition, antagonism for hydroxytryptamine (HT) receptors 5-HT3, 5-HT1D and 5-HT7, partial agonism for 5-HT1B, and agonism for 5-HT1A. It has been shown to have a beneficial effect on depression-related cognitive dysfunction, as well as on anxiety, depression, anhedonia and emotional blunting. Recently a potential anti-inflammatory action was also described. Limited clinical studies have specifically explored the efficacy of vortioxetine in treating depressive symptoms in MS. However, extrapolating from existing research in major depressive disorder, it is plausible that vortioxetine's multimodal mechanism could provide a favorable therapeutic approach. This position paper, which summarizes the output of annual clinical meeting held by the DMSTs in MS Italian Study Group, is focused on the possible role that vortioxetine could play as symptomatic treatment (ST) of depressed patients with MS, hypothesizing a direct impact on the clinical course of the disease.
Article
Neuropathic pain is an important and disabling clinical problem, its management constitutes a challenge for healthcare professionals. Vortioxetine is a new antidepressant drug with multimodal action, which gives it a unique profile. Tricyclic antidepressants, in particular amitriptyline, and serotonin and norepinephrine reuptake inhibitors venlafaxine and duloxetine are first-line drugs in the treatment of neuropathic pain. The interaction between the pain and depression binomial is very frequent, being the most frequent psychological complication in patients with chronic pain. This comprehensive and descriptive review summarizes the most relevant pharmacological data on vortioxetine, as well as the specific literature on vortioxetine in neuropathic pain and chronic pain.
Article
Background Vortioxetine hydrobromide (VXT), a new therapeutic option in the treatment of major depressive disorder, is a poorly soluble drug, and instability under stress conditions has been reported. The aim of the present study was to prepare VXT liposomes (VXT-Ls) with an antidepressant-like effect, to improve drug stability and reduce toxicity of the free drug.Methods Liposomes were prepared using the thin lipid film hydration method and properly characterized. Forced degradation studies were conducted in photolytic and oxidative conditions. The cytotoxicity was evaluated in VERO cells through MTT assay and in vivo toxicity was assessed in mice. The antidepressant-like effect in mice was confirmed using the open-field test paradigm and tail suspension test.ResultsThe optimized VXT-Ls have multilamellar vesicles with an average size of 176.74 nm ± 2.43. The liposomal formulation increased the stability of VXT. VERO cell viability was maintained at around 40% when the VXT-Ls were tested at higher concentrations and no signs of acute toxicity were observed in mice. The antidepressant-like effect was effective, for VXT-Ls, at doses ranging from 2.5 mg/kg to 10 mg/kg, measured by the tail suspension test in mice. The non-liposomal formulation was effective at a dose of 10 mg/kg. The open field test was performed and any unspecific changes in locomotor activity were revealed.Conclusions Liposomes seem to be a promising alternative for an oral VXT formulation at lower doses (2.5 mg/kg).Graphical abstract
Article
The opinion article is devoted to the analysis of a large-scale comprehensive systematic review of relevant and practically oriented studies of vortioxetine use in depression for the period to 2020. The mechanisms of multimodal action, issues of efficacy and safety of the drug, including direct procognitive effects, effects on anhedonia, anxiety, sleep disorders, analgesic and anti-inflammatory effects are highlighted. An increase in the efficacy of vortioxetine when the dose is increased to 20 mg/day is emphasized. Prospective directions for antidepressant research are outlined.
Article
Full-text available
Background: Chronic pain may be associated with diabetes mellitus and/or depression. Use of therapies that target both comorbidities is encouraged. Objective: This study was designed to investigate the potential antinociceptive, neuroprotective, and antidepressant effects of combinations of pioglitazone or metformin with fluoxetine in chronic constriction injury (CCI) in rats. Study design: Experimental trial in rats. Setting: University lab in in Saudi Arabia. Methods: Two sets of experiments were performed. In each one, 9 groups of rats (n = 8) were used: sham, CCI, and 7 CCI-treated groups. Treatments were given orally starting on day 7 post-surgery as follows (mg/kg/day): fluoxetine (10, 20, and 40), pioglitazone (20), metformin (50), fluoxetine (20) +' pioglitazone, and fluoxetine (20) +' metformin. In the first set, on day 14 post-surgery mechanical allodynia, thermal hyperalgesia, and serum cytokines were measured. Moreover, immunoreactivity of glial fibrillary acidic protein (GFAP, a marker for astrocytic activation) in the spinal cord was assessed and histopathological changes in the ipsilateral sciatic nerve were examined. In the second set, on days 14 and 21 post-surgery the forced swimming test was done. Results: In the first set, all treatments significantly decreased mechanical allodynia while all treatments except F10 and F20 significantly decreased thermal hyperalgesia compared to the CCI group. The F20+'M group showed the highest decreases, however still significantly lower than those of the sham group. The treatments didn't impair motor function in the rotarod test. All treatments significantly decreased serum levels of tumor necrosis factor- α, interleukin-6, and monocyte chemoattractant protein-1 while increasing the level of interleukin-10. The CCI-induced marked increase of GFAP immunoexpression has been reduced to moderate with fluoxetine (40) and pioglitazone, and to mild with metformin and the combination groups. The CCI-induced changes in sciatic nerve were less in fluoxetine (40), pioglitazone, and metformin groups, and least in the combination groups. In the second set, the immobility duration was significantly reduced by F20, F40, P, F20+'P, and F20+'M compared to the CCI group. The F20+'P group showed the highest decrease, however still significantly lower than that of the sham group. The treatments didn't affect locomotor activity in the open field test. Limitations: Measuring the cytokines levels only in blood and not in the spinal cord and sciatic nerve and measuring the outcome measures in the first set of experiments at only one time-point. Conclusions: Co-administration of pioglitazone or metformin with low-dose fluoxetine improved mechanical allodynia, thermal hyperalgesia, and neurohistopathological changes while co-administration of pioglitazone, but not metformin, improved the depressive-like behavior in the peripheral nerve injury model of neuropathic pain in rats. Extrapolation of the current results to clinical reality could be beneficial for pain patients with diabetes and/or depression, however this needs further confirmatory studies.
Article
Full-text available
Major Depressive Disorders (MDD) patients may exhibit cognitive deficits and it is currently unclear to which degree treatment with antidepressants may affect cognitive function. Preclinical and clinical observations showed that vortioxetine (VORT, an antidepressant with multimodal activity), presents beneficial effects on aspects of cognitive function. In addition, VORT treatment increases adult hippocampal neurogenesis (AHN) in rodents, a candidate mechanism for antidepressant activity. Pattern separation (PS) is the ability to discriminate between two similar contexts/events generating two distinct and non-overlapping representations. Impaired PS may lead to overgeneralization and anxiety disorders. If PS impairments were described in depressed patients, the consequences of antidepressant treatment on context discrimination (CD) are still in its infancy. We hypothesized that VORT-increased AHN may improve CD. Thus, in an attempt to elucidate the molecular mechanism underpinning VORT treatment effects on CD, a rodent model of PS, the role of AHN and stress-induced c-Fos activation was evaluated in the adult mouse hippocampus. Chronic treatment with VORT (1.8 g/kg of food weight; corresponding to a daily dose of 10 mg/kg, 3 weeks) improved CD in mice. Interestingly, chronic treatment with VORT reversed ablation of AHN-induced delay in CD and freezing behavior. VORT treatment decreased stress-induced c-Fos activation in the dorsal but not ventral dentate gyrus. VORT treatment did not affect c-Fos activity in the hippocampus of mice with ablated neurogenesis. This study highlights a role of VORT in CD, which may be independent from AHN and hippocampal c-Fos activation. Further studies elucidating the mechanisms underlying VORT’s effects in CD could contribute to future strategies for alleviating the disease burden for individuals suffering from depression and/or anxiety disorders.
Article
Full-text available
Major depressive disorder (MDD) is associated with cognitive impairments that may contribute to poor functional outcomes. Clinical data suggests that the multimodal antidepressant vortioxetine attenuates some cognitive impairments in MDD patients, but the mechanistic basis for these improvements is unclear. One theory suggests that vortioxetine improves cognition by suppressing γ-amino butyric acid (GABA)ergic neurotransmission, thereby increasing glutamatergic activation. Vortioxetine’s effects on cognition, GABA and glutamate neurotransmission have been supported in separate experiments, but no empirical work has directly connected vortioxetine’s cognitive effects to those on GABA and glutamate neurotransmission. In this paper, we attempt to bridge this gap by evaluating vortioxetine’s effects in the subchronic PCP (subPCP) model, which induces impaired cognitive function and altered GABA and glutamate neurotransmission. We demonstrate that acute or subchronic vortioxetine treatment attenuated subPCP-induced deficits in attentional set shifting (AST) performance, and that the selective 5-HT3 receptor antagonist ondansetron or the 5-HT reuptake inhibitor escitalopram could mimic this effect. Furthermore, acute vortioxetine treatment reversed subPCP-induced object recognition (OR) deficits in rats, while subchronic vortioxetine reversed subPCP-induced Object Recognition and object placement impairments in mice. Finally, subPCP treatment reduced GABAB receptor expression in a manner that was insensitive to vortioxetine treatment, and subchronic vortioxetine treatment alone, but not in combination with subPCP, significantly increased GABA’s affinity for the GABAA receptor. These data suggest that vortioxetine reverses cognitive impairments in a model associated with altered GABA and glutamate neurotransmission, further supporting the hypothesis that vortioxetine’s GABAergic and glutamatergic effects are relevant for cognitive function.
Article
Full-text available
The role of neuroinflammatory cells in the establishment of neuropathic pain has been investigated in depth in the last few years. In particular, microglia have been shown to be key players in the induction of tactile allodynia, as they release proinflammatory molecules that, in turn, sensitize nociceptive neurons within the spinal cord. However, the role of peripheral immune cells such as macrophages, infiltrating monocytes, mast cells, and T-cells has been highlighted in the last few studies, even though the data are still conflicting and need to be clarified. Intriguingly, the central (microglia) and peripheral (T-cell)-adaptive immune cells that orchestrate maladaptive process-driven neuropathic pain seem to be involved in a gender-dependent manner. In this review, we highlight the role of the microglia and peripheral immune cells in chronic degenerative disease associated with neuro-immune-inflammatory processes.
Article
Full-text available
Background: Major Depressive Disorder (MDD) is a complex disease characterized by emotional, physical and cognitive symptoms. We explored the efficacy of vortioxetine versus placebo on outcomes of cognition, functioning and mood symptoms in working patients with depression, using paroxetine as an active reference. Methods: Gainfully employed patients (18-65 years, N = 152) with MDD were randomized 1:1:1 to 8 weeks' double-blind, parallel treatment either with vortioxetine (10mg/day) or paroxetine (20mg/day), or with placebo. The primary efficacy measure was the Digit Symbol Substitution Test (DSST), analyzed using a mixed model for repeated measurements, and the key secondary efficacy measure was the University of San Diego Performance-based Skills Assessment - Brief (UPSA-B), analyzed using analysis of covariance (last observation carried forward). Results: At week 8, DSST and UPSA-B performance had improved relative to baseline in all treatment groups, with no statistically significant differences between treatment groups. While improvements in mood were comparable for vortioxetine and paroxetine, numerical improvements in cognitive performance (DSST) were larger with vortioxetine. Vortioxetine significantly improved overall cognitive performance and clinician-rated functioning relative to placebo. The majority of adverse events were mild or moderate, with nausea being the most common adverse event for vortioxetine. Limitations: Small sample sizes implied limited statistical power. Conclusion: This explorative study showed no significant differences versus placebo in DSST or UPSA-B performance at week 8. However, secondary results support vortioxetine as an effective and well-tolerated antidepressant, supporting an added benefit for cognition and functioning, which could have particular therapeutic relevance for the working patient population.
Article
Full-text available
With symptomatic remission and functional recovery as the overarching therapeutic objectives of antidepressant therapy, composite endpoint measures that conjointly consider both aspects of treatment are needed. This analysis evaluated the combined effect of vortioxetine on depressive symptoms and functional capacity in adults with MDD.Methods: NCT01564862, a multinational, double-blind, placebo-controlled, duloxetine-referenced study, con-ducted between April 2012 and February 2014, in 602 adult outpatients (18–65 years) with moderate-tosevere MDD (Montgomery-Asberg Depression Rating Scale (MADRS) ≥ 26), a major depressive episode of ≥ 3 months’ duration, and self-reported cognitive symptoms were randomized to once-daily vortioxetine (10 or 20 mg), duloxetine (60 mg), or placebo for 8 weeks. Assessments included the University of California San Diego Performance-based Skills Assessment (UPSA) and the MADRS. Two versions of UPSA were utilized; UPSA ‒Validation of Intermediate Measures and UPSA Brief form. An aligned UPSA-B (communication and finance items) was examined for sensitivity analysis. Efficacy was analyzed versus placebo according to the dualresponse (change from baseline in UPSA ≥ 7 and ≥ 9 and reduction in MADRS total score from baseline ≥ 50%).Results: Significantly more vortioxetine-treated patients were classified as dual responders for change in MADRS total score and UPSA score of ≥ 7 (clinically important difference [CID]) (27.4% vs 14.5%; P = 0.004), and change above CID (≥ 9) (23.4% vs 13.9%; P = 0.025). Duloxetine did not differ significantly from placebo for these dual response criteria. Sensitivity analysis using the aligned UPSA-B confirmed these results for vortiox-etine.
Article
Full-text available
Background Major Depressive Disorder (MDD) is a heterogeneous disease characterized by emotional, physical and cognitive symptoms. This study explored the effects of vortioxetine versus escitalopram on outcomes of cognition, functioning and mood symptoms in depressed patients with inadequate response to current antidepressant treatment. Methods In this parallel-group, active-comparator study, adult patients (18–65 years, N = 101) with MDD with inadequate response to current antidepressant monotherapy were randomized 1:1 to 8 weeks’ double-blind treatment with flexible doses (10 to 20 mg/day) of either vortioxetine or escitalopram. Primary and key secondary efficacy measures were the Digit Symbol Substitution Test (DSST), analyzed using a mixed model for repeated measurements, and the University of San Diego Performance-based Skills Assessment – Brief (UPSA-B), analyzed using analysis of covariance (last observation carried forward method). Results At week 8, DSST and UPSA-B performance had improved in both treatment groups, with no statistically significant treatment differences. Numerical improvements across measures of cognition, functioning and mood symptoms generally favored vortioxetine. Most adverse events were mild or moderate, with nausea being the most common adverse event. Limitations This was an exploratory study with small sample sizes implying limited statistical power. Conclusion Although this explorative study did not meet primary endpoints, the results confirm vortioxetine in doses of 10 to 20 mg/day as an efficacious and well-tolerated antidepressant switch treatment. The overall direction of numerical effect sizes across cognition endpoints support previous findings that vortioxetine specifically benefits cognitive function in MDD.
Article
Full-text available
Background: Major depressive disorder (MDD) is a common condition that often includes cognitive dysfunction. A systematic literature review of studies and a network meta-analysis (NMA) were carried out to assess the relative effect of antidepressants on cognitive dysfunction in MDD. Methods: MEDLINE ®, Embase ®, Cochrane, CDSR, PsychINFO ® databases, clinical trial registries, and relevant conference abstracts were searched for randomised controlled trials (RCTs) assessing the effects of antidepressants/placebo on cognition. An NMA comparing antidepressants was conducted using a random effects model. Results: The database search retrieved 11,337 citations of which 72 RCTs from 103 publications met the inclusion criteria. The review identified 86 cognitive tests assessing the effect of antidepressants on cognitive functioning. However, the Digit Symbol Substitution Test (DSST), which targets multiple domains of cognition and is recognised as being sensitive to change, was the only test that was used across 12 of the included RCTs and that allowed the construction of a stable network suitable for the NMA. The interventions assessed included selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs) and other non-SSRI/SNRIs. The NMA using the DSST showed that vortioxetine was the only antidepressant that improved cognitive dysfunction on the DSST versus placebo (standardised mean difference: 0.325 (95% CI=[0.120; 0.529], p=0.009). When comparing to other antidepressants, Vortioxetine was statistically more efficacious on the DSST versus escitalopram, nortriptyline, and the SSRI and TCA classes. Conclusions: This study highlighted the large variability in measures used to assess cognitive functioning. The findings on the DSST indicate differential effects of various antidepressants on improving cognitive function in patients with MDD.
Article
Full-text available
Schizophrenia and major depression (MD) have been associated with immune system dysfunction. One example of this is the altered level of cytokines—important inflammatory mediators—in blood, and a proinflammatory immune state has been described in some subgroups of patients. A knock to the immune system in early life might trigger a life-long increased immune reactivity, and infections and autoimmune disorders are now known to be risk factors for development of schizophrenia and MD. Pro- and anti-inflammatory cytokines mediate indoleamine 2,3-dioxygenase activity; this enzyme drives metabolism of tryptophan and kynurenin in the central nervous system and degrades serotonin. Alterations of serotonergic, noradrenergic, and glutamatergic neurotransmission have been associated with low-level neuroinflammation, and anti-inflammatory compounds have a therapeutic benefit in MD and schizophrenia, as shown in meta-analyses. Moreover, antidepressants and antipsychotics have intrinsic immunomodulatory effects. With evidence pointing to the role inflammatory processes play in the pathogenesis of major psychiatric disorders, this review will look at various immunological aspects of treatment of such disorders.
Article
Psychological stresses such as social loss and separation during childhood induce hardship, referred to as emotional pain. These experiences are well-documented risk factors for the development of physical pain in adulthood. However, the underlying neuronal mechanisms of this exacerbation of pain are largely unknown, and consequently there is no effective pharmacotherapy. In this study, we sought to determine whether infant maternal separation (MS) contributes to aggravation of neuropathic pain in adult mice. MS increased anxiety- and depression-like behavioral responses to adult stress. In MS animals, chronic constriction injury (CCI) heightened the sensory dimension of chronic pain relative to that of control mice. However, MS mice treated with fluoxetine for 4 weeks after MS did not exhibit augmentation of allodynia, and their emotional response was attenuated. Microglia were more abundant in the spinal cord in MS/CCI mice than in control/CCI mice. These results suggest that emotional impairment is related to augmentation of neuropathic pain, and that dysfunction of microglial activation contributes to heightened pain sensitivity.