The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Special ArticlesFull Access

Neuroprotective Benefits of Antidepressants in Multiple Sclerosis: Are We Missing the Mark?

Abstract

The potential of antidepressant medication to have a neuroprotective effect for people with multiple sclerosis (MS) has received increased interest in recent years. The possibility of antidepressants, particularly fluoxetine, for potential repurposing to treat primary progressive and secondary progressive MS is of interest as a result of the relative lack of disease-modifying medications for these subtypes. A number of animal studies have found positive results for a neuroprotective effect of antidepressant use in MS, with human studies showing mixed results. These human studies all have a significant limitation: they exclude people with moderate to severe depressive symptoms, a core symptom of MS beyond that of reactive depression. It is likely that reregulation of the common mechanisms in depression and MS, such as inflammation, serotonin, norepinephrine, glutamate and brain-derived neurotropic factor disruption, and hypothalamic-pituitary-thalamic axis dysregulation, are important to the neuroprotective value of antidepressant medication. Given that MS is known for its heterogeneity, the question might be less about whether antidepressant medication provides neuroprotective benefits to people with multiple sclerosis but for whom they provide benefits and whether we are designing studies that will detect a benefit. To answer these questions, studies must include people with MS and depressive symptoms as well as people with relapsing remitting and chronic subtypes.

Multiple sclerosis (MS) is the most common neurodegenerative disease to affect young adults (1, 2), with onset typically occurring between the ages of 20 and 40 years (3). Depression and anxiety disorders are frequently present in people with MS (PwMS), and depression is considered a core symptom of the disease (4, 5). The lifetime prevalence of depression is approximately 50% following a diagnosis of MS (6), in contrast to 11.6% for the general Australian population (7). The rate of depression in MS is higher than most other chronic or neurologic diseases (8) and is thought to be driven by a combination of psychosocial factors consequential to the disease and biological changes that result from neurological damage (5). Depression in PwMS has been shown to be more enduring than in people without MS, who usually recover within 6 to 12 months (60% and 80%, respectively) (9). Studies have shown that PwMS and depression at baseline continued to experience depression at 4 and 10 years follow-up (9, 10). There has been less research into anxiety disorders; however, prevalence of clinically significant anxiety has been reported as between 25% and 51% (1114).

Consistent with the higher rate of psychological disorder experienced by PwMS, compared with another chronic health condition and the general population, there is a greater prevalence of antidepressant medication use, which is reported to be between 21% and 35% in PwMS (1517), whereas population-wide usage is around 8%−10% (18, 19). However, a study that reported 51% of PwMS had clinically significant depressive symptoms (32% were in the moderate to severe range) found only 41% of PwMS identified as having depressive symptoms were taking antidepressant medication, suggesting underdetection and treatment of depression in PwMS (16).

Stress, Depression, and Progression In MS

Stress is an important biological function that in small amounts is adaptive and has the ability to help motivate us to achieve goals. Sustained or uncontrollable stress, however, may lead to depression, especially in individuals who are predisposed (20). Unsurprisingly, a review of studies that have assessed the relationship between stress and depression in PwMS has found the link to be robust, with a proposed moderate to large effect (5).

The relationship between stress and disease progression has gained considerable research attention because of the long-considered view among many PwMS that significant stressful life events are involved in the onset of disease and occurrence of relapses, as well as the transient worsening of existing symptoms. Animal studies have shown a relationship between induced stress and disease onset and progression in mice infected with experimental autoimmune encephalomyelitis, the mouse model of MS (21). However, the relationship is complex and dependent on factors including gender, timing, duration, and type of induced stress. Additionally, severe stress has been shown to improve disease, whereas milder stress progressed it (22). An association dependent on the severity of a stressful relationship has also been shown in a human study. Brown et al. (23) recruited 101 consecutive PwMS through two MS clinics and followed them over a 2-year period. They reported acute stress, measured as stressful events of less than 6 months, to be related to exacerbation, whereas chronic stress, measured as stressful events of more than 6 months duration, was not. A review of research investigating the relationship between stress and disease onset or progression deduced that there is evidence of stress influencing disease onset and clinical course, although “no secure conclusions could be drawn” (24). There have been contrasting results from studies about the relationship between stress and exacerbation; however, the majority have found a relationship (for a review, see Mitsonis et al. [25]), which was reported to be moderate in a meta-analysis that included results of 14 studies (effect size, Cohen’s d=0.53) (26). Clearly, the relationship between stress and disease onset, progression, or exacerbation is not straightforward and is likely mediated or moderated by a range of psychosocial factors, including depression and anxiety (25, 27).

Research suggests there is also a relationship between depression and relapse/progression in MS. A study of 132 PwMS reported a correlation between higher depressive symptoms and exacerbation, with depressive symptoms remaining 6 months after the MS relapse (28). Furthermore, a retrospective assessment of 2,312 PwMS found that women (but not men) who had experienced a psychiatric illness (depression, anxiety, or bipolar) within a 10-year period had significantly higher scores on the Expanded Disability Status Scale than those who had not (β=0.31, p=0.0004) (29), which is consistent with an earlier animal study that found a greater stress-progression response in female mice infected with experimental autoimmune encephalomyelitis (30). However, depression was not related to greater disability progression at 10-year follow-up in another smaller study (N=96), despite depression remaining stable in people assessed at baseline (10). It is possible this study was underpowered, although the difference in psychiatric measurements may also be responsible for the contrasting results. Koch et al. (10) recruited through an MS clinic and used the Center for Epidemiological Studies Depression scale (CES-D), with a score of 16 or greater determining depressive symptoms in PwMS. The CES-D ranges between 0 and 60. By comparison, McKay et al. (29) identified psychiatric disorder from physician and hospital claim files (≥ five physician claims or ≥ one hospital claim), suggesting PwMS in this sample had a greater severity of psychiatric difficulties, given that they were seeking health care management in relation to them.

Similarities Of Dysregulation Occur In MS, Stress, and Depression

The experience of stress activates the hypothalamo-pituitary-adrenal (HPA) axis through the secretion of corticotrophin releasing hormone from the hypothalamus, which activates the pituitary gland and secretes adrenal corticotrophin, stimulating cortisol production from the adrenal gland; among other things, it helps regulate stress and immune response. Cortisol receptors provide a negative feedback loop to regulate cortisol production and the cortisol-to-cortisone ratio (20, 31). Production of cortisol is regulated by circadian rhythms, with highest levels in the morning and a reduction of cortisol throughout the day. Regulation of cortisol is important for both physical and psychological health. However, the natural rhythm of cortisol production is altered with HPA-axis disruption (31). HPA-axis dysregulation and inflammation have been related to psychiatric pathology (32). Increased cortisol levels, modulated by the HPA axis, have been associated with acute and chronic stress and depression in people with and without MS (33, 34). Dysregulation of the HPA axis is well established in PwMS; hyperactivity correlates with the clinical course of MS, such that mild hyperactivity is seen in relapsing remitting MS (RRMS), whereas primary-progressive MS (PPMS) shows the most pronounced changes. It is proposed that HPA-axis dysregulation may be driven by inflammation in RRMS but by neurodegeneration in secondary progressive MS (SPMS) (3537). A study that assessed cortisol awakening response (CAR) in people with RRMS and SPMS in comparison to healthy controls found that CAR was significantly higher in people with RRMS than the control group, but not people with SPMS. Additionally, in people with RRMS, increased CAR at baseline was predictive of greater disability 9 months later (38). Prior studies have also found cortisol hyperresponsiveness and HPA-axis dysregulation predicted clinically measured disease progression in PwMS (39). It is possible that glucocorticoid-insensitive immune cells may be responsible for excessive inflammatory response seen in RRMS, whereas glucocorticoid-insensitivity is absent or reduced in people with chronic disease types SPMS and PPMS (39). Taken together, the reregulation of the HPA-axis may be time critical for people with RRMS, before irreversible neuronal damage that causes dysregulation in the absence of inflammation. Importantly, treatment with antidepressant medication has shown attenuation of HPA-axis function in depression (40), and HPA-axis hyperactivity in MS is attenuated by immunomodulatory medications (35).

There is a well-established association between immune system inflammation and severity of depression (41, 42). The MS disease-modifying medication fingolimod, which has anti-inflammatory effects, has recently displayed antidepressant effects, including hippocampal neurogenesis in mice exposed to either chronic unpredictable stress or depressive symptoms induced by chronically treated corticosterone (43). Fingolimod increases brain-derived neurotropic factors (BDNF). BDNF support neurons and glial cells within the central nervous system and are important to neuronal survival and plasticity. BDNF are expressed in immune system cells and have been linked to inflammation. Other immunomodulatory therapy used in MS, including glatiramer acetate and hematopoietic stem cell therapy, have been shown to modulate BDNF. BDNF are similarly reduced in people experiencing depression, and fluoxetine has been found to increase BDNF in cultured mouse astrocytes (4446).

A number of neurotransmitters have been shown to be altered in both MS and depression. For example, norepinephrine, known for its anti-inflammatory and neuroprotective effects, has been found to be reduced in PwMS (47, 48), as have serotonin transporters and platelet content (49, 50). Excessive glutamine, known to have neurotoxic effects, has been shown in PwMS (51). Serotonin or norepinephrine depletion has long been considered a factor in depression, and this is supported by antidepressant medications that act to increase the synaptic availability of these neurotransmitters to treat depression (52, 53). Glutamate has similarly been implicated in depression, and there is evidence that chronic antidepressant administration reduces the presynaptic release of glutamate (54). These commonalities between neurotransmitter disturbance in MS and depression may point to a potential overlap in disease mechanisms.

What We Know About Antidepressants and Neuroprotection In MS

Animal Studies

The importance of the serotonergic system in immune modulation has been established in animal models with autoimmune encephalomyelitis, where knocking out the serotonin transporters resulted in increased susceptibility to inflammatory response (55). Serotonergic transmission has been shown to be altered in PwMS, with fewer seretonergic transporters (SERT) in the limbic and paralimbic systems and more SERT in the frontal cortex (50). Studies have shown benefits of the antidepressant medications sertraline and fluoxetine in suppressing inflammatory responses and reducing disease onset and progression in mice with autoimmune encephalomyelitis (5659). In addition, the MAO inhibitor phenelzine reduced progression of autoimmune encephalomyelitis and showed increased levels of serotonin, norepinephrine, dopamine, and GABA (60), whereas the tricyclic clomipramine was found to reduce T-cell proliferation, oxidative stress, and B-lymphocytic activity in acute and chronic phases of autoimmune encephalomyelitis (61). Although it would be ideal to assess whether neuroprotective effects of antidepressants in mouse model autoimmune encephalomyelitis show a difference in effect size dependent on level of stress or depression-like symptoms, this has not been possible, because the motor impairment produced by autoimmune encephalomyelitis precludes assessment of animal depressive-like behavior (43).

Human Studies

A summary of human studies is provided in Table 1. Most human studies have been small. A double-blind placebo-controlled study (N=38, n=19 selective serotonin reuptake inhibitor [SSRI]/placebo) that assessed the effect of fluoxetine on new MRI lesion formation in people who had RRMS or SPMS with relapses found a reduced frequency of new lesions in the group administered fluoxetine compared with the control group (62). Additionally, a study assessing the effect of stress as a risk for relapse in people with RRMS taking an SSRI, escitalopram, compared with a control group, found that the control group had a 2.9 times greater chance of relapse; this was predicted by long-term but not short-term stress-related events (63). Another very small study (N=11) showed a neuroprotective effect from short-term use of fluoxetine through an increased N-acetylaspartate/creatine ratio of cerebral white matter, proposing a reversible effect of axonal damage (64). In contrast, a study undertaken by Mostert et al. (65) that assessed fluoxetine in people with SPMS (N=42, n=20 SSRI, n=22 placebo) found no significant difference in treatment groups on disability progression. However, the authors noted that disability progression in the placebo group was slower in their study than other studies assessing progressive MS using similar measures, and they proposed that the study was underpowered (65). An important difference between the studies that have shown an effect and the Mostert et al. (65) study is that the latter included only people with SPMS. The Mostert et al. (62) study required PwMS to have evidence of relapse or gadolinium-enhancing lesion on MRI within the prior 1–2 years, whereas the Mostert et al. (64) study included people with RRMS (N=7) and SPMS (N=4). Results from these small studies point to the possibility of inflammation as the important aspect in the disease-modulating ability of antidepressant medications in MS.

TABLE 1. Human studies assessing the neuroprotective value of antidepressant medication in people with multiple sclerosis (MS)a

AuthorObjectiveSampleDesignInterventionMeasuresOutcomesComments
Mostert et al. (65)Pilot study to assess whether fluoxetine (SSRI) has neuroprotective properties for people with SPMS or PPMSPwMS, N=42; IG/CG, N=20/22; PPMS or SPMS, IG PP/SP, N=6/14; CG PP/SP, N=7/15Double-blind randomized placebo-controlled trial; longitudinal prospective for 2 yearsFluoxetine, 20 mg twice dailyEvery 3 months: EDSS; 9-HPT; AI. baseline, 1 and 2 years: MRI scans; MSFC; FIS; GNDS; SF–36Thirty-five percent (7/20) in the intervention group and 32% (7/22) in the control group experienced sustained progression on the EDSS, 9-HPT or AI at 2 years, with no significant between-group effect on time to progression time to progression and no between-group differences identified for T2 lesion load, gray or white matter volume.Authors reported an unusually low rate of disability progression in the placebo group, which they suggested decreased statistical power. Participants were required to have an EDSS score between 3.5 and 6.5. Excluded participants were those with moderate to severe depressive symptoms.
Mostert et al. (62)Exploratory study to assess whether fluoxetine (SSRI) has neuroprotective properties for people with RRMS or relapsing SPMSN=38; IG/CG, N=19/19; RRMS or SPMS, IG RR/SP, N=18/1; CG RR/SP, N=16/3Double-blind randomized placebo-controlled trial; longitudinal prospective for 6 monthsFluoxetine, 20 mg/dayParticipants assessed for gadolinium-enhancing lesions on brain MRI at 4, 8, 16 and 24 weeksAt 16 weeks: mean new enhancing lesions was 1.21 (SD=2.6) in the fluoxetine group and 3.16 (SD=5.3) in the control group (p=0.05); number of new lesions on scans was 24% (nine lesions) compared with 47% (18 lesions) in the fluoxetine compared with placebo group, respectively, (p=0.03); 63% (N=12) versus 26% (N=5) of people did not show new enhancing lesions in the fluoxetine versus control group respectively, (p=0.02); at 24 weeks: mean new enhancing lesions was 1.84 (SD=2.9) in the fluoxetine group and 5.16 (8.6) in the control group (p=0.15); number of new lesions on scans was 25% (19 lesions) compared with 41% (31 lesions) in the fluoxetine compared with placebo group respectively, (p=0.04); 32% (N=6) versus 21% (N=4) of people did not show new enhancing lesions in the fluoxetine versus control group respectively, (p=0.71).Participants required an EDSS score ≤6 and at least one relapse in the prior year or two relapses in the prior 2 years or one gadolinium-enhancing lesion on MRI. Excluded participants were those with moderate to severe depressive symptoms.
Mostert et al. (64)Exploratory study to assess whether fluoxetine (SSRI) improves NAA/creatine ratio or choline/creatine ratio in cerebral white matter of people with MSPwMS, N=11; RRMS, N=7; SPMS, N=4Longitudinal prospective for 2 weeks; no control armFluoxetine, 20 mg/per for 7 days (week 1) then 20 mg twice daily for 7 days (week 2)H-MRS at baseline, week 1 and week 2. T25WT; AFQA significant difference was found between NAA/Cr ratio mean at baseline, 1.77 (SD=0.18) and 2 weeks, 1.84 (SD=0.20) (p=0.007), which the authors reports to represent evidence of reversible axonal dysfunction from the effects of fluoxetine; there was no significant difference for Cho/Cr ratio mean at baseline, 1.06 (SD=0.13) and 2 weeks, 1.06 (SD=0.13) (p=0.84); there was no significant difference between baseline and 2 weeks for the T25WT or AFQ (both p values >0.05).Did not report on the participants’ level of depressive symptoms or whether this was an exclusion criterion.
Mitsonis et al. (63)Assessment of the effects of escitalopram (SSRI) on stress-related MS relapses in women with MSPwMS, N=41; all women; all RRMS; IG/CG, N=21/20Open-label randomized-controlled trial; longitudinal prospective for 1 yearEscitalopram, 10 mg/payStressful life events diary confirmed by a psychiatrist at monthly appointment; categorized into short- or long-term; neurologist confirmed relapses and EDSS assessed at monthly appointmentsStressful life event risk for relapse was 2.9 times higher for the control group than the escitalopram group (p<0.001); this risk was only influenced by long-term but not short-term stressful life events; there was a cumulative risk of relapse with increased stressful life events in the control group (hazard ratio for stressful life event: 1=3.3, p=0.007; 2=13.2, p<0.001; and ≧ 3=20.9, p<0.001), whereas, the hazard ratio was only significant for three stressful life events in the escitalopram group (hazard ratio for SLE: 5.3 (p<0.001), suggesting that escitalopram provides a protective buffer.Participants were premenopausal, had at least one MS relapse within the year prior to study enrollment, and an EDSS score. Participants with clinically significant mental health issues, (per DSM-IV criteria) at baseline were excluded, including those with unipolar and bipolar depressive disorder and anxiety disorders. Stressful events directly related to MS were excluded.
Cambron et al. (70)FLUOX-PMS: assessment of whether fluoxetine (SSRI) has neuroprotective properties for people with SPMS or PPMSPwMS, N=139; PPMS, 40%; SPMS, 60%; IG/CG, N=69/68Double-blind randomized placebo-controlled trial; longitudinal prospective for 2 yearsFluoxetine, 40 mg/day; started at 20 mg/day and titrated to 40 mg/day by week 129-HPT; T25WT; AI; MRI; cognitive assessmentNo significant difference between groups for time to disease progression, which was measured as the proportion of patients without sustained 20% increase in T25WT or 9-HPT; 66% in the fluoxetine group and 58% in the control group did not experience disease progression (p=0.07); no significant difference was found between the groups on the AI (p=0.37).Results were obtained from media reports following the ECTRIMS 2016 meeting where results were announced. Excluded participants were those with moderate to severe depressive symptoms. MRI results have not yet been reported.
Chataway et al. (69)MS-SMART: assessment of 445 people with SPMS to identify whether there is a neuroprotective benefit from amiloride, fluoxetine (SSRI), or riluzole compared with placeboPwMS, N=445; all SPMS; randomized across four armsPhase II double-blind randomized placebo-controlled four-arm trial; longitudinal prospective for 96 weeks (2 years).Three intervention conditions: amiloride, 5 mg twice daily for 96 weeks (5 mg/day for first 4 weeks); fluoxetine, 50 mg twice daily for 96 weeks (50 mg/day for first 4 weeks); riluzole, 50 mg twice daily for 96 weeks (50 mg/day for first 4 weeks)9-HPT; T25WT; AI; MRI; cognitive assessmentAt the present time, only preliminary findings have been announced; the researchers stated that none of the three drugs assessed, including fluoxetine, showed a neuroprotective effect in SPMS.Information obtained from the MS Society. Excluded participants were those with moderate to severe depressive symptoms. Participant EDSS score requirement between 4 and 6.5.

aAFQ=Abbreviated Fatigue Questionnaire, AI=Ambulatory Index, CG=control group, Cho/Cr ratio=choline to creatine ratio, EDSS=Expanded Disability Status Scale, FIS=Fatigue Impact Scale, FLUOX-PMS=fluoxetine in progressive multiple sclerosis, GNDS=Guy’s Neurological Disability Scale, IG=intervention group, MSFC=Multiple Sclerosis Functional Composite, MS-SMART=Multiple Sclerosis-Secondary Progressive Multi-Arm Randomisation Trial, NAA/CR ratio=N-acetylaspartate to creatine ratio, 9-HPT=9 Hole Peg Test, PPMS=Primary Progressive Multiple Sclerosis, PwMS=People with Multiple Sclerosis, RRMS=relapsing remitting multiple sclerosis, SF–36=36-item Short-Form Health Survey; SRI=selective serotonin reuptake inhibitor, SPMS=secondary progressive multiple sclerosis, T25WT=Timed 25-Foot Walk Test.

TABLE 1. Human studies assessing the neuroprotective value of antidepressant medication in people with multiple sclerosis (MS)a

Enlarge table

This is further supported by studies that have found an effect of fluoxetine on recovery following ischemic stroke (66). Studies have also shown untreated depression following stroke is related to greater impairment at follow-up, including cognitive impairment and greater difficulty performing activities of daily living, as well as increased mortality. Similar to MS, there is a high prevalence of depression following stroke; this is thought to be a combination of reactive and acute neurological inflammation factors (67).

A recent evaluation of potential neuroprotective oral antidepressant medications suggested that fluoxetine should be assessed for possible repurposing in SPMS, highlighting the relevant mechanisms to SPMS (68), including stimulation of glycogenosis and enhancement of brain-derived neurotrophic factors found to be reduced in SPMS. A four-arm trial that assessed three potential medications, including fluoxetine, against placebo (N=445) found no support for a neuroprotective effect in people with SPMS (69). A previous study similarly assessing the neuroprotective potential of fluoxetine against placebo in people with SPMS or PPMS (N=108) reported preliminary results at ECTRIMS 2016 also reported no significant results between groups for clinical disease progression, although there was a trend toward significance, p=0.07 (70). Both of these studies excluded people experiencing symptoms of depression (70, 71). MRI results from these trials are yet to be published.

What Are We Missing?

An important consideration for interpreting human studies that assess the relationship between antidepressant administration and exacerbation or progression in PwMS is that they excluded people with moderate to severe depressive symptoms. Excluding people with depressive symptoms is problematic, given that depression is a symptom of MS with a biological basis beyond that of reactive depression (5). The inclusion of people with depressive symptoms in a double-blind randomized-controlled study provides a layer of complexity because, once identified, the randomization of people experiencing moderate to severe depressive symptoms to the placebo arm raises ethical concerns. However, there is precedent for this practice when assessing antidepressant efficacy (72). Alternatively, a single-blind randomized control trial would provide participants in the placebo arm with autonomy to discontinue the trial if they chose to seek treatment for depressive symptoms, while still blinding study outcome evaluation.

Furthermore, studies that are limited to PPMS or SPMS, both characterized with ongoing progression of disability and brain atrophy in the absence of, or with minimal, inflammation, may limit the ability to detect an effect or require a larger sample to detect a smaller effect. Inflammation is a common mechanism associated with both MS and depression (41, 42) and is therefore a potential common mechanism by which antidepressant medication may exert an effect to reduce progression. Similarly, a shared disruption to neurotransmitters, including serotonin, norepinephrine, and glutamate, as well as to BDNF and HPA-axis activity in PwMS and people with depression supports the inclusion of PwMS and comorbid depression (35, 40, 50, 53, 73), especially given the ability of antidepressant medication to reregulate BDNF levels and HPA-axis activity in people experiencing depression (40, 74). Thus, if there is a link between depression and disease progression in MS, exclusion of participants with comorbid depression may be considered a systematic sampling error, because it risks not finding a neuroprotective relationship from SSRI use (or any antidepressant in general) when there is one, if results are generalized to the broader MS population.

Although there is a high priority to find neuroprotective agents for SPMS and PPMS given the dearth of therapeutics currently available for these subtypes, it is remiss to exclude people with RRMS, who often still experience neurological damage with current therapeutics. These people may be granted a greater level of neuroprotection, and therefore quality of life, if the addition of antidepressant medication offers neuroprotective benefits.

Conclusions

Given the high prevalence of depression and other emotional disorders in PwMS, and the similarities of neurotransmitter, BDNF, and HPA-axis dysregulation in PwMS and people experiencing stress and depression, it is vital that studies assessing the neuroprotective benefits of antidepressant medication in PwMS include people who are also experiencing depressive symptoms, and people with RRMS subtype, where inflammation is a key disease component. Should current research assessing the neuroprotective benefits of antidepressants in MS miss a signal because the study design excludes the very PwMS who would have shown a neuroprotective benefit, it risks the MS research community losing enthusiasm for this line of research. Thus, a potential opportunity to improve the quality of life for people living with MS and to reduce health care expenditure resulting from greater physical and psychological disability progression will also be lost.

From the Department of Psychological Sciences, Swinburne University, Melbourne, Australia (Grech); the Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, Australia (Grech); the Melbourne School of Psychological Sciences, University of Melbourne, Australia (Grech, Hester); the Department of Neurology, Monash Health, Victoria, Australia (Butler); the Department of Imaging, Monash Health, Victoria, Australia (Stuckey); and the Department of Imaging, School of Clinical Sciences, Monash Health, Monash University, Victoria, Australia (Stuckey).
Send correspondence to Dr. Grech ().

The authors report no financial relationships with commercial interests.

References

1 Julian LJ, Mohr DC: Cognitive predictors of response to treatment for depression in multiple sclerosis. J Neuropsychiatry Clin Neurosci 2006; 18:356–363LinkGoogle Scholar

2 Wallin MT, Wilken JA, Turner AP, et al.: Depression and multiple sclerosis: review of a lethal combination. J Rehabil Res Dev 2006; 43:45–62Crossref, MedlineGoogle Scholar

3 Cohen JA, Rae-Grant A: Handbook of Multiple Sclerosis, 2nd ed. London, Springer Healthcare, 2012CrossrefGoogle Scholar

4 Minden SL, Feinstein A, Kalb RC, et al.: Evidence-based guideline: assessment and management of psychiatric disorders in individuals with MS: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology 2014; 82:174–181Crossref, MedlineGoogle Scholar

5 Arnett PA, Barwick FH, Beeney JE: Depression in multiple sclerosis: review and theoretical proposal. J Int Neuropsychol Soc 2008; 14:691–724Crossref, MedlineGoogle Scholar

6 Feinstein A: Multiple sclerosis and depression. Mult Scler 2011; 17:1276–1281Crossref, MedlineGoogle Scholar

7 National Survey of Mental Health and Wellbeing 2007. Canberra, Australia, Australian Bureau of Statistics, 2008. www.abs.gov.auGoogle Scholar

8 Feinstein A: The Clinical Neuropsychiatry of Multiple Sclerosis, 2nd ed. Cambridge, United Kingdom, Cambridge University Press, 2007CrossrefGoogle Scholar

9 Koch MW, Patten S, Berzins S, et al.: Depression in multiple sclerosis: a long-term longitudinal study. Mult Scler 2015; 21:76–82Crossref, MedlineGoogle Scholar

10 Koch M, Uyttenboogaart M, van Harten A, et al.: Fatigue, depression and progression in multiple sclerosis. Mult Scler 2008; 14:815–822Crossref, MedlineGoogle Scholar

11 Janssens ACJW, van Doorn PA, de Boer JB, et al.: Anxiety and depression influence the relation between disability status and quality of life in multiple sclerosis. Mult Scler 2003; 9:397–403Crossref, MedlineGoogle Scholar

12 Janssens ACJW, van Doorn PA, de Boer JB, et al.: Impact of recently diagnosed multiple sclerosis on quality of life, anxiety, depression and distress of patients and partners. Acta Neurol Scand 2003; 108:389–395Crossref, MedlineGoogle Scholar

13 Korostil M, Feinstein A: Anxiety disorders and their clinical correlates in multiple sclerosis patients. Mult Scler 2007; 13:67–72Crossref, MedlineGoogle Scholar

14 Moreau T, Schmidt N, Joyeux O, et al.: Coping strategy and anxiety evolution in multiple sclerosis patients initiating interferon-beta treatment. Eur Neurol 2009; 62:79–85Crossref, MedlineGoogle Scholar

15 Kessing LV, Harhoff M, Andersen PK: Increased rate of treatment with antidepressants in patients with multiple sclerosis. Int Clin Psychopharmacol 2008; 23:54–59Crossref, MedlineGoogle Scholar

16 Cetin K, Johnson KL, Ehde DM, et al. Antidepressant use in multiple sclerosis: epidemiologic study of a large community sample. Multiple Sclerosis (13524585). 2007;13(8):1046-53.Crossref, MedlineGoogle Scholar

17 Windt R, Glaeske G, Hoffmann F: Treatment of multiple sclerosis in Germany: an analysis based on claims data of more than 30,000 patients. Int J Clin Pharm 2013; 35:1229–1235Crossref, MedlineGoogle Scholar

18 Simon GE, Stewart C, Beck A, et al.: National prevalence of receipt of antidepressant prescriptions by persons without a psychiatric diagnosis. Psychiatr Serv 2014; 65:944–946LinkGoogle Scholar

19 Patterns of Use of Mental Health Services and Prescription Medications, 2011. Canberra, Australia, Australian Bureau of Statistics, 2016Google Scholar

20 Pariante CM: Depression, stress and the adrenal axis. J Neuroendocrinol 2003; 15:811–812Crossref, MedlineGoogle Scholar

21 Gold SM, Heesen C: Stress and disease progression in multiple sclerosis and its animal models. Neuroimmunomodulation 2006; 13:318–326Crossref, MedlineGoogle Scholar

22 Lovera J, Reza T: Stress in multiple sclerosis: review of new developments and future directions. Curr Neurol Neurosci Rep 2013; 13:398Crossref, MedlineGoogle Scholar

23 Brown RF, Tennant CC, Sharrock M, et al. Relationship between stress and relapse in multiple sclerosis: Part I. Important features. Mult Scler 2006; 12:453-464.Crossref, MedlineGoogle Scholar

24 Artemiadis AK, Anagnostouli MC, Alexopoulos EC: Stress as a risk factor for multiple sclerosis onset or relapse: a systematic review. Neuroepidemiology 2011; 36:109–120Crossref, MedlineGoogle Scholar

25 Mitsonis CI, Potagas C, Zervas I, et al.: The effects of stressful life events on the course of multiple sclerosis: a review. Int J Neurosci 2009; 119:315–335Crossref, MedlineGoogle Scholar

26 Mohr DC, Hart SL, Julian L, et al. Association between stressful life events and exacerbation in multiple sclerosis: a meta-analysis. BMJ 2004; 328(7442):731Crossref, MedlineGoogle Scholar

27 Mohr DC: Stress and multiple sclerosis. J Neurol 2007; 254(Suppl 2):II65–II68Crossref, MedlineGoogle Scholar

28 Moore P, Hirst C, Harding KE, et al.: Multiple sclerosis relapses and depression. J Psychosom Res 2012; 73:272–276Crossref, MedlineGoogle Scholar

29 McKay KA, Tremlett H, Fisk JD, et al.: Psychiatric comorbidity is associated with disability progression in multiple sclerosis. Neurology 2018; 90:e1316–e1323Crossref, MedlineGoogle Scholar

30 Harpaz I, Abutbul S, Nemirovsky A, et al.: Chronic exposure to stress predisposes to higher autoimmune susceptibility in C57BL/6 mice: glucocorticoids as a double-edged sword. Eur J Immunol 2013; 43:758–769Crossref, MedlineGoogle Scholar

31 Ray JA, Meikle AW: Cortisol-cortisone shuttle: A functional indicator of 11B-HSD Activity, in Cortisol. Edited by Esposito A, Bianchi V. Hauppauge, NY, Nova Science Publishers, 2012, pp 91–105Google Scholar

32 Renoir T, Hasebe K, Gray L: Mind and body: how the health of the body impacts on neuropsychiatry. Front Pharmacol 2013; 4:158Crossref, MedlineGoogle Scholar

33 Spencer RL, Hutchison KE: Alcohol, aging, and the stress response. Alcohol Res Health 1999; 23:272–283MedlineGoogle Scholar

34 Munafò MR: The serotonin transporter gene and depression. Depress Anxiety 2012; 29:915–917Crossref, MedlineGoogle Scholar

35 Kümpfel T, Schwan M, Weber F, et al.: Hypothalamo-pituitary-adrenal axis activity evolves differentially in untreated versus treated multiple sclerosis. Psychoneuroendocrinology 2014; 45:87–95Crossref, MedlineGoogle Scholar

36 Then Bergh F, Kümpfel T, Trenkwalder C, et al.: Dysregulation of the hypothalamo-pituitary-adrenal axis is related to the clinical course of MS. Neurology 1999; 53:772–777Crossref, MedlineGoogle Scholar

37 Heesen C, Gold SM, Raji A, et al.: Cognitive impairment correlates with hypothalamo-pituitary-adrenal axis dysregulation in multiple sclerosis. Psychoneuroendocrinology 2002; 27:505–517Crossref, MedlineGoogle Scholar

38 Kern S, Krause I, Horntrich A, et al.: Cortisol awakening response is linked to disease course and progression in multiple sclerosis. PLoS One 2013; 8:e60647Crossref, MedlineGoogle Scholar

39 Gold SM, Mohr DC, Huitinga I, et al.: The role of stress-response systems for the pathogenesis and progression of MS. Trends Immunol 2005; 26:644–652Crossref, MedlineGoogle Scholar

40 Reul JM, Stec I, Söder M, et al.: Chronic treatment of rats with the antidepressant amitriptyline attenuates the activity of the hypothalamic-pituitary-adrenocortical system. Endocrinology 1993; 133:312–320Crossref, MedlineGoogle Scholar

41 Howren MB, Lamkin DM, Suls J: Associations of depression with C-reactive protein, IL-1, and IL-6: a meta-analysis. Psychosom Med 2009; 71:171–186Crossref, MedlineGoogle Scholar

42 Lu Y, Feng L, Feng L, et al.: Systemic inflammation, depression and obstructive pulmonary function: a population-based study. Respir Res 2013; 14:53Crossref, MedlineGoogle Scholar

43 di Nuzzo L, Orlando R, Tognoli C, et al.: Antidepressant activity of fingolimod in mice. Pharmacol Res Perspect 2015; 3:e00135Crossref, MedlineGoogle Scholar

44 Kong EKC, Peng L, Chen Y, et al.: Up-regulation of 5-HT2B receptor density and receptor-mediated glycogenolysis in mouse astrocytes by long-term fluoxetine administration. Neurochem Res 2002; 27:113–120Crossref, MedlineGoogle Scholar

45 Lühder F, Gold R, Flügel A, et al.: Brain-derived neurotrophic factor in neuroimmunology: lessons learned from multiple sclerosis patients and experimental autoimmune encephalomyelitis models. Arch Immunol Ther Exp (Warsz) 2013; 61:95–105Crossref, MedlineGoogle Scholar

46 Allaman I, Fiumelli H, Magistretti PJ, et al.: Fluoxetine regulates the expression of neurotrophic/growth factors and glucose metabolism in astrocytes. Psychopharmacology (Berl) 2011; 216:75–84Crossref, MedlineGoogle Scholar

47 Polak PE, Kalinin S, Feinstein DL: Locus coeruleus damage and noradrenaline reductions in multiple sclerosis and experimental autoimmune encephalomyelitis. Brain 2011; 134:665–677Crossref, MedlineGoogle Scholar

48 Feinstein DL, Kalinin S, Braun D: Causes, consequences, and cures for neuroinflammation mediated via the locus coeruleus: noradrenergic signaling system. J Neurochem 2016; 139(Suppl 2):154–178Crossref, MedlineGoogle Scholar

49 Baidina TV, Akintseva YV, Trushnikova TN: Platelet serotonin in multiple sclerosis and its relationship with fatigue syndrome. Neurochem J 2013; 7:226–229CrossrefGoogle Scholar

50 Hesse S, Moeller F, Petroff D, et al.: Altered serotonin transporter availability in patients with multiple sclerosis. Eur J Nucl Med Mol Imaging 2014; 41:827–835Crossref, MedlineGoogle Scholar

51 Macrez R, Stys PK, Vivien D, et al.: Mechanisms of glutamate toxicity in multiple sclerosis: biomarker and therapeutic opportunities. Lancet Neurol 2016; 15:1089–1102Crossref, MedlineGoogle Scholar

52 Moret C, Briley M: The importance of norepinephrine in depression. Neuropsychiatr Dis Treat 2011; 7(Suppl 1):9–13MedlineGoogle Scholar

53 Nutt DJ. Relationship of neurotransmitters to the symptoms of major depressive disorder. J Clin Psychiatry. 2008; 69 Suppl E1:4-7.MedlineGoogle Scholar

54 Sanacora G, Treccani G, Popoli M: Towards a glutamate hypothesis of depression: an emerging frontier of neuropsychopharmacology for mood disorders. Neuropharmacology 2012; 62:63–77Crossref, MedlineGoogle Scholar

55 Hofstetter HH, Mössner R, Lesch KP, et al.: Absence of reuptake of serotonin influences susceptibility to clinical autoimmune disease and neuroantigen-specific interferon-gamma production in mouse EAE. Clin Exp Immunol 2005; 142:39–44Crossref, MedlineGoogle Scholar

56 Taler M, Gil-Ad I, Korob I, et al.: The immunomodulatory effect of the antidepressant sertraline in an experimental autoimmune encephalomyelitis mouse model of multiple sclerosis. Neuroimmunomodulation 2011; 18:117–122Crossref, MedlineGoogle Scholar

57 Yuan XQ, Qiu G, Liu XJ, et al.: Fluoxetine promotes remission in acute experimental autoimmune encephalomyelitis in rats. Neuroimmunomodulation 2012; 19:201–208Crossref, MedlineGoogle Scholar

58 Bhat R, Mahapatra S, Axtell RC, et al.: Amelioration of ongoing experimental autoimmune encephalomyelitis with fluoxetine. J Neuroimmunol 2017; 313:77–81Crossref, MedlineGoogle Scholar

59 Ghareghani M, Zibara K, Sadeghi H, et al.: Fluvoxamine stimulates oligodendrogenesis of cultured neural stem cells and attenuates inflammation and demyelination in an animal model of multiple sclerosis. Sci Rep 2017; 7:4923Crossref, MedlineGoogle Scholar

60 Benson CA, Wong G, Tenorio G, et al.: The MAO inhibitor phenelzine can improve functional outcomes in mice with established clinical signs in experimental autoimmune encephalomyelitis (EAE). Behav Brain Res 2013; 252:302–311Crossref, MedlineGoogle Scholar

61 Faissner S, Mishra M, Kaushik DK, et al.: Systematic screening of generic drugs for progressive multiple sclerosis identifies clomipramine as a promising therapeutic. Nat Commun 2017; 8:1990Crossref, MedlineGoogle Scholar

62 Mostert JP, Admiraal-Behloul F, Hoogduin JM, et al.: Effects of fluoxetine on disease activity in relapsing multiple sclerosis: a double-blind, placebo-controlled, exploratory study. J Neurol Neurosurg Psychiatry 2008; 79:1027–1031Crossref, MedlineGoogle Scholar

63 Mitsonis CI, Zervas IM, Potagas CM, et al.: Effects of escitalopram on stress-related relapses in women with multiple sclerosis: an open-label, randomized, controlled, one-year follow-up study. Eur Neuropsychopharmacol 2010; 20:123–131Crossref, MedlineGoogle Scholar

64 Mostert JP, Sijens PE, Oudkerk M, et al.: Fluoxetine increases cerebral white matter NAA/Cr ratio in patients with multiple sclerosis. Neurosci Lett 2006; 402:22–24Crossref, MedlineGoogle Scholar

65 Mostert J, Heersema T, Mahajan M, et al.: The effect of fluoxetine on progression in progressive multiple sclerosis: a double-blind, randomized, placebo-controlled trial. ISRN Neurol 2013; 2013:370943Crossref, MedlineGoogle Scholar

66 Chollet F, Tardy J, Albucher J-F, et al.: Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol 2011; 10:123–130Crossref, MedlineGoogle Scholar

67 Robinson RG, Spalletta G: Poststroke depression: a review. Can J Psychiatry 2010; 55:341–349Crossref, MedlineGoogle Scholar

68 Vesterinen HM, Connick P, Irvine CMJ, et al.: Drug repurposing: a systematic approach to evaluate candidate oral neuroprotective interventions for secondary progressive multiple sclerosis. PLoS One 2015; 10:e0117705Crossref, MedlineGoogle Scholar

69 Chataway J: A message for MS-Smart trial participants from Professor Jeremy Chataway. London, MS Society, 2018. www.mssociety.org.uk/research/explore-our-research/research-we-fund/search-our-research-projects/can-three-existing-drugs-protect-nerves-from-damage-in-ms/ms-smart-resultsGoogle Scholar

70 Cambron M, Mostert J, Haentjens P, et al.: Fluoxetine in progressivemultiple sclerosis (FLUOX-PMS): study protocol for a randomized controlled trial. Trials 2014; 15:37Crossref, MedlineGoogle Scholar

71 MS-SMART: Multiple Sclerosis-Secondary Progressive Multi-Arm Randomisation Trial (MS-SMART). Bethesda, Md, National Library of Medicine, 2016. www.clinicaltrials.gov/ct2/show/NCT01910259?term=%22fluoxetine%22+AND+%22multiple+sclerosis%22&rank=2Google Scholar

72 Irving K, Brett JD, Tania BH-M, et al. Initial severity and antidepressant benefits: a meta-analysis of data submitted to the Food and Drug Administration. PLoS Med 2008; 5(2):e45MedlineGoogle Scholar

73 Barkhatova VP, Zavalishin IA, Askarova LSh, et al.: Changes in neurotransmitters in multiple sclerosis. Neurosci Behav Physiol 1998; 28:341–344Crossref, MedlineGoogle Scholar

74 Lee B-H, Kim Y-K: The roles of BDNF in the pathophysiology of major depression and in antidepressant treatment. Psychiatry Investig 2010; 7:231–235Crossref, MedlineGoogle Scholar