Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-05T00:45:51.453Z Has data issue: false hasContentIssue false

Antidepressant medications in dementia: evidence and potential mechanisms of treatment-resistance

Published online by Cambridge University Press:  09 January 2023

Harry Costello*
Affiliation:
Institute of Cognitive Neuroscience, University College London, London, UK
Jonathan P. Roiser
Affiliation:
Institute of Cognitive Neuroscience, University College London, London, UK
Robert Howard
Affiliation:
Division of Psychiatry, University College London, London, UK
*
Author for correspondence: Harry Costello, E-mail: harry.costello@ucl.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Depression in dementia is common, disabling and causes significant distress to patients and carers. Despite widespread use of antidepressants for depression in dementia, there is no evidence of therapeutic efficacy, and their use is potentially harmful in this patient group. Depression in dementia has poor outcomes and effective treatments are urgently needed. Understanding why antidepressants are ineffective in depression in dementia could provide insight into their mechanism of action and aid identification of new therapeutic targets. In this review we discuss why depression in dementia may be a distinct entity, current theories of how antidepressants work and how these mechanisms of action may be affected by disease processes in dementia. We also consider why clinicians continue to prescribe antidepressants in dementia, and novel approaches to understand and identify effective treatments for patients living with depression and dementia.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Depression in dementia is common. Individuals with dementia are twice as likely as age-matched controls to develop major depressive disorder and up to one-third of all-cause dementia patients develop depression (Asmer et al., Reference Asmer, Kirkham, Newton, Ismail, Elbayoumi, Leung and Seitz2018).

Despite the widespread use of antidepressants in patients with dementia (Breining et al., Reference Breining, Bonnet-Zamponi, Zerah, Micheneau, Riolacci-Dhoyen, Chan-Chee and Leperre-Desplanques2017; Jester, Molinari, Zgibor, & Volicer, Reference Jester, Molinari, Zgibor and Volicer2021) there is little evidence of therapeutic efficacy (see Fig. 1) (Dudas, Malouf, McCleery, & Dening, Reference Dudas, Malouf, McCleery and Dening2018). Though relatively few trials of antidepressants for depression in dementia have been conducted to date and most have studied depression in Alzheimer's dementia (AD), the largest and best quality studies have shown limited signal of any clinically meaningful difference (Banerjee et al., Reference Banerjee, Hellier, Dewey, Romeo, Ballard, Baldwin and Burns2011). The lack of therapeutic effect cannot be explained by age effects as depressed older adults without dementia respond as well to antidepressants as other age groups (Gerson, Belin, Kaufman, Mintz, & Jarvik, Reference Gerson, Belin, Kaufman, Mintz and Jarvik1999; Gildengers et al., Reference Gildengers, Houck, Mulsant, Pollock, Mazumdar, Miller and Reynolds2002), though the effective dose range differs (Furukawa et al., Reference Furukawa, Cipriani, Cowen, Leucht, Egger and Salanti2019).

Fig. 1. Reproduced forest plot of meta-analysis of included Cochrane review randomised controlled trials of antidepressants for depression in dementia. (Dudas et al. Reference Dudas, Malouf, McCleery and Dening2018) (AD - Alzheimer's dementia, VD - Vascular dementia, M - mixed dementia, PDD - Parkinson's disease dementia, SSRI - selective serotonin reuptake inhibitor, TCA - tricyclic antidepressant, SNRI - serotonin-noradrenaline reuptake inhibitor, NASSA - noradrenaline and specific serotonergic antidepressant).

In fact, antidepressant use in dementia is not only ineffective but potentially harmful, as it is associated with increased risk of falls, hospitalisation and higher risk of mortality (Banerjee et al., Reference Banerjee, High, Stirling, Shepstone, Swart, Telling and Tabet2021; Johnell et al., Reference Johnell, Jonasdottir Bergman, Fastbom, Danielsson, Borg and Salmi2017).

Clearly further high-quality trials of antidepressants in dementia types other than AD are needed. To date only one randomised controlled trial (RCT) of a selective serotonin reuptake inhibitor (SSRI) in 14 patients has been conducted in Lewy body dementia (LBD) which did not show efficacy and found high overall burden of side-effects (Culo et al., Reference Culo, Mulsant, Rosen, Mazumdar, Blakesley, Houck and Pollock2010). Vascular dementia has similarly been neglected. Antidepressants are effective for post-stroke depression in patients without dementia (Li & Zhang, Reference Li and Zhang2020). However, the overlap of vascular dementia with AD pathology (Attems & Jellinger, Reference Attems and Jellinger2014) and evidence that higher vascular risk factor scores predict reduced response to antidepressants in late life depression (Sheline et al., Reference Sheline, Pieper, Barch, Welsh-Boehmer, McKinstry, MacFall and Doraiswamy2010) does not suggest this patient group will buck the trend of a disappointing lack of efficacy of antidepressants in dementia. Even in frontotemporal dementia (FTD) where clinical use of SSRIs is widely advocated and seen as an effective therapy for depression and disinhibition in this patient group (Trieu et al., Reference Trieu, Gossink, Stek, Scheltens, Pijnenburg and Dols2020), the evidence supporting its use is mixed (Huey, Putnam, & Grafman, Reference Huey, Putnam and Grafman2006; Kaye, Petrovic-Poljak, Verhoeff, & Freedman, Reference Kaye, Petrovic-Poljak, Verhoeff and Freedman2010). Most studies in FTD are open-label or case series designs, and the only RCTs of SSRIs (RCTs) found opposing results in terms of efficacy (Deakin, Rahman, Nestor, Hodges, & Sahakian, Reference Deakin, Rahman, Nestor, Hodges and Sahakian2004; Moretti, Torre, Antonello, Cazzato, & Bava, Reference Moretti, Torre, Antonello, Cazzato and Bava2003), though one trial of trazodone in 26 patients reported reduced depressive symptoms (Lebert, Stekke, Hasenbroekx, & Pasquier, Reference Lebert, Stekke, Hasenbroekx and Pasquier2004).

In the UK over a quarter of all people living with dementia take an antidepressant and the rates of prescription are increasing (Martinez, Jones, & Rietbrock, Reference Martinez, Jones and Rietbrock2013). There is little evidence to support this rate of prescription and the lack of replication of the efficacy seen in depression without dementia could point to important mechanistic differences underlying depression in dementia.

Though there is evidence for the efficacy of some non-drug interventions in people with dementia (Orgeta, Qazi, Spector, & Orrell, Reference Orgeta, Qazi, Spector and Orrell2015; Watt et al., Reference Watt, Goodarzi, Veroniki, Nincic, Khan, Ghassemi and Straus2021), therapeutic outcomes for patients with depression and dementia are poor (Fritze, Ehrt, Hortobagyi, Ballard, & Aarsland, Reference Fritze, Ehrt, Hortobagyi, Ballard and Aarsland2011). Depression in dementia hastens cognitive decline, increases mortality, causes significant carer stress and accelerates admission to long-term care facilities (Asmer et al., Reference Asmer, Kirkham, Newton, Ismail, Elbayoumi, Leung and Seitz2018; Feng et al., Reference Feng, Scherer, Tan, Chan, Fong and Ng2010; Lavretsky et al., Reference Lavretsky, Zheng, Weiner, Mungas, Reed, Kramer and Mack2010; Rapp et al., Reference Rapp, Schnaider-Beeri, Wysocki, Guerrero-Berroa, Grossman, Heinz and Haroutunian2011).

Substantial evidence supports the hypothesis that depression is an early prodromal symptom of dementia, commonly occurring up to 10 years prior to diagnosis, and a consequence of the distinct pathophysiology of individual dementia syndromes (Singh-Manoux et al., Reference Singh-Manoux, Dugravot, Fournier, Abell, Ebmeier, Kivimäki and Sabia2017). In cognitively normal older adults, biomarkers of Alzheimer's disease pathology are associated with subsequent depression and presence of depression accelerates cognitive decline (Babulal et al., Reference Babulal, Ghoshal, Head, Vernon, Holtzman, Benzinger and Roe2016; Snowden et al., Reference Snowden, Atkins, Steinman, Bell, Bryant, Copeland and Fitzpatrick2015). The underlying aetiology of depression as a prodrome of dementia is therefore likely to differ from depression in the general population.

Effective treatments for depression in dementia are urgently required. Understanding why standard antidepressants are unsuccessful in dementia has the potential to provide insight into both their mechanisms of action in typical depressed patients and the mechanisms driving depression in dementia, as well as aiding identification of new therapeutic targets.

In this review we summarise the reasons why depression in dementia may be a distinct entity, current understanding of how antidepressants work and how these mechanisms of action are affected by disease processes in dementia. We also discuss why antidepressants will continue to be used in dementia and the attendant clinical dilemmas faced by clinicians, patients and carers.

A different syndrome of depression in dementia?

Depression is a heterogeneous and aetiologically complex syndrome. There are at least 256 possible unique symptom profiles that meet DSM-V criteria for a diagnosis of major depressive disorder (Buch & Liston, Reference Buch and Liston2021). This degree of clinical heterogeneity has led to efforts to define subtypes based on symptom profiles (Buch & Liston, Reference Buch and Liston2021). The reliability of different depression subtypes has not been established in the general population, largely due to transition between symptom profiles across and within depressive episodes (Lamers et al., Reference Lamers, Rhebergen, Merikangas, de Jonge, Beekman and Penninx2012). However, this has led to studies highlighting how specific symptoms of depression respond differently to interventions and may predict overall response to treatment (Buch & Liston, Reference Buch and Liston2021). For example, network analysis of individual depressive symptoms in antidepressant RCTs has identified improvement in low mood and insomnia as having greatest impact on subsequent remission and recovery (Komulainen et al., Reference Komulainen, Airaksinen, Savelieva, Gluschkoff, García Velázquez, Elovainio and Jokela2021).

Few studies have investigated how depressive symptoms differ across dementia types or in comparison with the non-dementia depression (Ehrt, Brønnick, Leentjens, Larsen, & Aarsland, Reference Ehrt, Brønnick, Leentjens, Larsen and Aarsland2006; Riedel, Heuser, Klotsche, Dodel, & Wittchen, Reference Riedel, Heuser, Klotsche, Dodel and Wittchen2010; Verdelho, Hénon, Lebert, Pasquier, & Leys, Reference Verdelho, Hénon, Lebert, Pasquier and Leys2004). However, the clinical complexity of making a distinction between a diagnosis of depression from some of the other behavioural and psychological symptoms of dementia (BPSD) can be challenging as shown by the overlap between depression rating scales with other symptoms captured by BPSD measures (See Table 1). This raises the question of whether antidepressants are ineffective because they have been used to treat different syndromes.

Table 1. Depressive symptoms across seven depression rating scales that are also captured by measures of other BPSD symptoms (not depression items) on four BPSD measures

NPI, Neuropsychiatric Inventory (Cummings et al., Reference Cummings, Mega, Gray, Rosenberg-Thompson, Carusi and Gornbein1994), Behave-AD, Behavioural Pathology in Alzheimer's Disease (Reisberg, Auer, & Monteiro, Reference Reisberg, Auer and Monteiro1997), CUSPAD, Columbia University Scale for Psychopathology in Alzheimer's disease (Devanand et al., Reference Devanand, Miller, Richards, Marder, Bell, Mayeux and Stern1992), MOUSEPAD, Manchester & Oxford University Scale for the Psychopathological Assessment of Dementia (Allen, Gordon, Hope, & Burns, Reference Allen, Gordon, Hope and Burns1996).

Coloured circles for a symptom indicate that a scale directly assesses that symptom, whereas empty circles indicate that a scale indirectly measures a symptom. The seven depression rating scales are: Beck Depression Inventory (Beck, Steer, Ball, & Ranieri, Reference Beck, Steer, Ball and Ranieri1996); Centre of Epidemiological Studies Depression Scale; Hamilton Rating Scale for Depression (Radloff, Reference Radloff1977); Inventory of Depressive Symptoms (Rush, Gullion, Basco, Jarrett, & Trivedi, Reference Rush, Gullion, Basco, Jarrett and Trivedi1996); Montgomery–Åsberg Depression Rating Scale (Montgomery & Åsberg, Reference Montgomery and Åsberg1979); Quick Inventory of Depressive Symptoms (Rush et al., Reference Rush, Trivedi, Ibrahim, Carmody, Arnow, Klein and Keller2003); Zung Self-Rating Depression Scale (Zung, Reference Zung1965). List of depressive symptoms adapted from ref – (Fried, Flake, & Robinaugh, Reference Fried, Flake and Robinaugh2022).

Studies of depressive symptom profile in dementia in long-term care facilities highlight how motivational symptoms such as apathy are common (Janzing, Hooijer, van ‘t Hof, & Zitman, Reference Janzing, Hooijer, van ‘t Hof and Zitman2002), and clinician-rated irritability is the most prevalent depressive symptom (Borza et al., Reference Borza, Engedal, Bergh, Barca, Benth and Selbæk2015). However, both apathy and irritability are common in dementia in the absence of depression, and highly influenced by environmental factors (Jao, Liu, Williams, Chaudhury, & Parajuli, Reference Jao, Liu, Williams, Chaudhury and Parajuli2019).

Apathy affects 49% (Zhao et al., Reference Zhao, Tan, Wang, Jiang, Tan, Tan and Yu2016) and 72% (Chow et al., Reference Chow, Binns, Cummings, Lam, Black, Miller and van Reekum2009) of Alzheimer's disease (AD) and FTD patients respectively, markedly higher than that reported in older adults with depression without dementia (38%) (Yuen et al., Reference Yuen, Bhutani, Lucas, Gunning, AbdelMalak, Seirup and Alexopoulos2015). Lesion studies have identified that the functional anatomy of apathy is associated with damage to fronto-striatal circuits (Levy & Dubois, Reference Levy and Dubois2006). Apathy also shares underlying neural and cognitive mechanisms with anhedonia, a core symptom of depression primarily characterised by a loss of interest in engaging in pleasurable activities. Motivational symptoms including apathy and anhedonia contribute to an ‘interest-activity’ cluster of depressive symptoms that predicts worse response to antidepressants in depression in the general population (Uher et al., Reference Uher, Perlis, Henigsberg, Zobel, Rietschel, Mors and McGuffin2012), but respond better than core mood symptoms to behavioural activation (Leontjevas et al., Reference Leontjevas, Teerenstra, Smalbrugge, Vernooij-Dassen, Bohlmeijer, Gerritsen and Koopmans2013). The presence of apathy in late-onset depression in mild cognitive impairment (MCI) also predicts subsequent development of AD (Ruthirakuhan, Herrmann, Vieira, Gallagher, & Lanctôt, Reference Ruthirakuhan, Herrmann, Vieira, Gallagher and Lanctôt2019). This indicates that apathy is not only a common symptom in depression in dementia, but is central to the presentation of depression as a prodromal stage of dementia and poor response to antidepressant treatment.

Agitation is common in dementia affecting half of patients (Livingston et al., Reference Livingston, Barber, Marston, Rapaport, Livingston, Cousins and Cooper2017), and is defined as inappropriate verbal, vocal, or motor activity that is not thought to be caused by an unmet need (Cummings et al., Reference Cummings, Mintzer, Brodaty, Sano, Banerjee, Devanand and Zhong2015). Irritability is a key component of agitation in dementia and strongly associated with depressive symptoms, both of which worsen with dementia progression (Volicer, Frijters, & Van der Steen, Reference Volicer, Frijters and Van der Steen2012). Though agitation is seen across psychiatric disorders, there is a clinical distinction between a patient with dementia and agitation consisting of wandering due to disorientation and a patient with agitated depression (Sampogna, Vecchio, Giallonardo, Luciano, & Fiorillo, Reference Sampogna, Vecchio, Giallonardo, Luciano and Fiorillo2020). It is likely that the prevalence of irritability as one of the most common symptoms in depression in dementia is due to this overlap, with agitation secondary to cognitive decline, and is a different entity to the irritability that characterises depression in the general population.

Few studies have assessed whether the phenomenology of depression in dementia is qualitatively different from depression in other populations (Ehrt et al., Reference Ehrt, Brønnick, Leentjens, Larsen and Aarsland2006; Riedel et al., Reference Riedel, Heuser, Klotsche, Dodel and Wittchen2010). However, apathy and irritability are examples of how key depressive symptoms in dementia, which are captured by most symptom scales, are also common in the context of BPSD without apparent depressed mood. This highlights just how challenging it is for clinicians to reliably diagnose depression in dementia and raises the question as to whether antidepressants are ineffective due to their being used to treat a different syndrome that isn't ‘depression’.

Due to the differing patterns of neurodegeneration across dementia subtypes it would be anticipated that the underlying mechanisms of depression, evolution of symptoms and response to treatment would be different in each disorder. However, few studies have investigated whether depressive symptom profiles or response to treatment differs across neurodegenerative disorders or stages of disease. Future research investigating the network dynamics of depressive symptoms over time could reveal how changes in specific symptoms are related, and which symptoms have the greatest impact on clinical improvement and treatment response.

Further trials of medications that treat depression as homogenous across dementia subtypes and stage of disease are likely to be ineffective. An alternative approach to understanding depression in dementia is to view it as a complex system of interacting depressive symptoms, which differ across dementia subtypes and stages of disease. Utilising this approach would promote tailored interventions for specific symptoms with greater importance at different stages of disease, development of depressive-symptom-network markers for each condition to guide interventions and diagnosis, and provide some mechanistic insights into the causes of depression.

Neurodegeneration of monoamine pathways

The monoamine hypothesis of depression is over 50 years old (Coppen, Reference Coppen1967). It proposes that diminished activity in monoamine pathways (predominantly serotonin, noradrenaline and dopamine) plays a causal role in the development of depression. In the age of complex systems neurobiology and computational neuroscience, there is a consensus that such reductionist biochemical theories are over-simplifications (Cowen & Browning, Reference Cowen and Browning2015; Harmer, Duman, & Cowen, Reference Harmer, Duman and Cowen2017). However, given that almost all effective antidepressants directly target monoamine system receptors or transporters, predominantly serotonin, monoamine function likely plays a central role in the therapeutic effect of antidepressants.

The monoamine systems project from small brainstem nuclei to widespread brain regions (Sasaki et al., Reference Sasaki, Shibata, Tohyama, Kudo, Endoh, Otsuka and Sakai2008). Combined with an array of post-synaptic receptors, this anatomical distribution enables a wide variety of functions but is also susceptible to disruption from neurodegeneration (see Table 2). A potential reason for the lack of therapeutic effect of antidepressants in dementia would be disruption of the monoamine pathways needed to mediate their effect.

Table 2. Neurodegeneration of monoaminergic nuclei in dementia subtypes

Serotonin is the monoamine that has primarily been implicated in the treatment of depression and is a target for most antidepressants, in particular SSRIs, the most widely used class of medications. The most direct evidence for serotonin's role in the pathophysiology of depression is from tryptophan depletion studies, in which transient reduction in serotonin synthesis is achieved via dietary depletion of its precursor amino acid, tryptophan. In recovered patients who are taking antidepressant medication, tryptophan depletion elicits a rapid worsening of depressive symptoms. However, tryptophan depletion has relatively little effect in healthy controls, currently depressed patients or even recovered patients who are unmedicated (Ruhé, Mason, & Schene, Reference Ruhé, Mason and Schene2007).

The dorsal raphe nucleus (DRN) is one of two brain stem nuclei which are the primary sites of serotonergic innervation in the brain. The human DRN consists of ~ 250 000 neurons, 70% of which contain serotonin (Steinbusch, Dolatkhah, & Hopkins, Reference Steinbusch, Dolatkhah, Hopkins, Di Giovanni and De Deurwaerdere2021). Projections from the DRN are topographically organised rostrocaudally and innervate regions involved in mood regulation, motivation and memory, including the striatum, entorhinal cortex, hippocampus, pre-frontal cortex and amygdala (Steinbusch et al., Reference Steinbusch, Dolatkhah, Hopkins, Di Giovanni and De Deurwaerdere2021).

During the early stages of AD pathology (Braak stage I-II), the DRN is heavily affected by tau tangles (Rüb et al., Reference Rüb, Del Tredici, Schultz, Thal, Braak and Braak2000) and exhibits marked neurodegeneration, with estimates of up to 77% neuronal cells lost depending on disease stage (Lyness, Zarow, & Chui, Reference Lyness, Zarow and Chui2003). As a consequence of the degeneration in the DRN, lower cortical serotonin, serotonin metabolites (5-hydroxyindoleacetic acid) and reduced serotonin transporter levels (Smith et al., Reference Smith, Barrett, Joo, Nassery, Savonenko, Sodums and Workman2017) have been reported in AD and other dementia syndromes (van der Zande et al., Reference van der Zande, Joling, Happach, Vriend, Scheltens, Booij and Lemstra2019). Degeneration in serotonergic pathways has also been implicated in the development of BPSD, including depression (Lanctôt, Herrmann, & Mazzotta, Reference Lanctôt, Herrmann and Mazzotta2001; Vermeiren et al., Reference Vermeiren, Van Dam, Aerts, Engelborghs, Martin and De Deyn2015).

The noradrenergic system is a further target of antidepressants, including the serotonin–noradrenaline reuptake inhibitors venlafaxine and duloxetine. The locus coeruleus (LC), a small, pigmented nucleus located in the pons is the primary site for the synthesis of noradrenaline in the brain (Holland, Robbins, & Rowe, Reference Holland, Robbins and Rowe2021). The LC projects widely and is implicated in modulating attention, memory, cognitive flexibility and sleep (Holland et al., Reference Holland, Robbins and Rowe2021; Swift et al., Reference Swift, Gross, Frazer, Bauer, Clark, Vazey and Poe2018). Degeneration of the LC occurs early in AD and Parkinson's disease (PD) (Betts et al., Reference Betts, Kirilina, Otaduy, Ivanov, Acosta-Cabronero, Callaghan and Hämmerer2019). Neuronal death of up to 30% of LC neurons has been reported in MCI, increasing to 55% in diagnosed Alzheimer's dementia (Kelly et al., Reference Kelly, He, Perez, Ginsberg, Mufson and Counts2017). Depletion of LC neurons in AD is more closely correlated with disease severity and staging than cholinergic degeneration in the nucleus basalis of Meynert (Holland et al., Reference Holland, Robbins and Rowe2021; Kelly et al., Reference Kelly, He, Perez, Ginsberg, Mufson and Counts2017). LC degeneration has been implicated in agitation and irritability in AD (Liu, Stringer, Reeves, & Howard, Reference Liu, Stringer, Reeves and Howard2018) and depression in PD (Wang et al., Reference Wang, Li, Huang, Wan, Zhang, Wang and Jin2018). However, no study to date has examined whether LC integrity predicts antidepressant response in depression in dementia.

Human and animal studies have shown that dopaminergic transmission is crucial in motivated behaviour and reward processing (Le Heron, Apps, & Husain, Reference Le Heron, Apps and Husain2018). Dopaminergic neurons project from the ventral tegmental area (VTA) and substantia nigra (SN) to the striatum (Sasaki et al., Reference Sasaki, Shibata, Tohyama, Kudo, Endoh, Otsuka and Sakai2008). Dopamine function is closely related to other neuromodulators such as serotonin which it is co-released within the striatum (Zhou et al., Reference Zhou, Liang, Salas, Zhang, De Biasi and Dani2005). This interplay with other neurotransmitters is believed to have an important role in cognitive functions and decision-making processes (Wert-Carvajal, Reneaux, Tchumatchenko, & Clopath, Reference Wert-Carvajal, Reneaux, Tchumatchenko and Clopath2022) implicated in depression (see Table 3). Though not first-line and less commonly used, dopaminergic agents such as buproprion (a noradrenaline-dopamine reuptake inhibitor), and pramipexole (a dopamine partial agonist) are effective treatments in the management of depression (Papakostas, Reference Papakostas2006).

Table 3. Examples of neurocognitive affective bias reported in depression and dementia

Dopaminergic dysfunction is associated with motivational symptoms of depression including apathy and anhedonia which are poorly responsive to antidepressant treatment (Husain & Roiser, Reference Husain and Roiser2018; Le Heron et al., Reference Le Heron, Apps and Husain2018). Greater atrophy and loss of pre-synaptic dopaminergic projections to the striatum is associated with increased apathy and anhedonia across neurodegenerative disorders (Drui et al., Reference Drui, Carnicella, Carcenac, Favier, Bertrand, Boulet and Savasta2014; Le Heron et al., Reference Le Heron, Apps and Husain2018). VTA and SN neurodegeneration is most prominent in PD where up to 80% of dopaminergic projections to the striatum are lost by the time of diagnosis (Cheng, Ulane, & Burke, Reference Cheng, Ulane and Burke2010; Kish, Shannak, & Hornykiewicz, Reference Kish, Shannak and Hornykiewicz1988). This striking loss of dopaminergic neurons may explain why apathy and anhedonia are particularly common in PD, estimated to affect 40% (den Brok et al., Reference den Brok, van Dalen, van Gool, Moll van Charante, de Bie and Richard2015) and 46% (Lemke, Brecht, Koester, Kraus, & Reichmann, Reference Lemke, Brecht, Koester, Kraus and Reichmann2005) of patients, respectively. It may also explain why depressive symptoms in PD are poorly responsive to serotonergic and noradrenergic antidepressants (Troeung, Egan, & Gasson, Reference Troeung, Egan and Gasson2013) but can respond to dopaminergic agonists used to treat motor symptoms (van der Velden, Broen, Kuijf, & Leentjens, Reference van der Velden, Broen, Kuijf and Leentjens2018).

Whether involving serotonin, noradrenaline or dopamine, antidepressant mechanisms of action involve increasing monoamine levels in the synapse by preventing either metabolism or reuptake into the pre-synaptic neuron. However, as described above, there is significant degeneration of monoamine neurons in early dementia, leading to a loss of pre-synaptic innervation (see Table 2). Following the death of these neurons, monoamine release is markedly reduced, resulting in a loss of function in these systems which presumably reduces any impact of antidepressants.

In addition to loss of innervation, degeneration of pre-synaptic monoamine nuclei would also lead to loss of feedback inhibition. For example, the serotonin-1A (5-HT1A) autoreceptor controls serotonergic tone through feedback inhibition and has been hypothesised to lead to a reduction in synaptic serotonin levels early on in SSRI treatment (Fritze, Spanagel, & Noori, Reference Fritze, Spanagel and Noori2017). The time taken for 5-HT1A autoreceptors to desensitise has been theorised to be an explanation of the delayed clinical effects of antidepressant treatment, and differences in 5-HT1A receptor levels or have been associated with depression, and the response to antidepressants (Richardson-Jones et al., Reference Richardson-Jones, Craige, Guiard, Stephen, Metzger, Kung and Leonardo2010). Though it is widely accepted that 5-HT1A receptors participate in autoinhibition, their action is far more complex and the function of feedback is regionally organised and dependent on behavioural states (Altieri, Garcia-Garcia, Leonardo, & Andrews, Reference Altieri, Garcia-Garcia, Leonardo and Andrews2012). Changes in 5-HT1A distribution and binding have been reported in dementia but vary depending on brain region and dementia subtype (Elliott et al., Reference Elliott, Ballard, Kalaria, Perry, Hortobágyi and Francis2009; Lai et al., Reference Lai, Tsang, Francis, Esiri, Keene, Hope and Chen2003). For example, increased 5-HT1A binding in the temporal cortex has been reported in vascular dementia (Elliott et al., Reference Elliott, Ballard, Kalaria, Perry, Hortobágyi and Francis2009) but reduced binding in the same region has been found in AD which also correlated with higher agitation levels (Lai et al., Reference Lai, Tsang, Francis, Esiri, Keene, Hope and Chen2003).

Understanding the relative deficits in monoamine function across dementia subtypes and stages of disease could guide which class of antidepressant may be most effective. However, as described above the dynamics and function of monoamine pathways are diverse, complex, and interdependent. There is considerable synergy between monoamine systems, for example co-release of dopamine and serotonin in the striatum, and innervation of the cholinergic system by the LC (Kelly et al., Reference Kelly, He, Perez, Ginsberg, Mufson and Counts2017; Zhou et al., Reference Zhou, Liang, Salas, Zhang, De Biasi and Dani2005). Further research into the relationship between these systems and association with depressive symptoms would be needed to guide best use of existing therapies or development of novel interventions.

Changes in neurocognitive processing

The cognitive neuropsychological model of depression suggests a causal role for negative affective biases in the development and treatment of depression (Roiser, Elliott, & Sahakian, Reference Roiser, Elliott and Sahakian2012). Depression is associated with several types of negative affective biases (see Table 3) including changes in reward processing, negative perception of social cues, greater attention to aversive information and increased recall of negative relative to positive information (Harmer et al., Reference Harmer, Duman and Cowen2017).

There is substantial evidence that antidepressants alter emotion and reward processing (Harmer et al., Reference Harmer, Duman and Cowen2017; Roiser et al., Reference Roiser, Elliott and Sahakian2012), thereby normalising negative affective biases. The cognitive neuropsychological model proposes that this modulation of core psychological processes in depression drives antidepressant therapeutic effect, explains why antidepressants take 4–6 weeks to improve depressive symptoms, and why they are most effective in conjunction with psychological therapy (Harmer et al., Reference Harmer, Duman and Cowen2017).

Dementia is characterised by progressive cognitive decline and it is probable that the cognitive adaptations induced by antidepressants to normalise negative biases are not sustained due to these cognitive deficits. Neurodegeneration of core neural circuits involved in affective biases (in addition to the monoamine systems innervating them) may lead to further loss of antidepressant effects (see Table 2). For example, the reward processing circuit includes the ventral striatum, anterior cingulate cortex, amygdala and prefrontal cortex (Perry & Kramer, Reference Perry and Kramer2015). Serotonin co-release with dopamine in the ventral striatum is believed to be crucial in mediating the temporal dynamics of dopamine mediated reward prediction errors which drive reinforcement learning (Zhou et al., Reference Zhou, Liang, Salas, Zhang, De Biasi and Dani2005). Activation in the ventral striatum is lower in both apathy and anhedonia (Husain & Roiser, Reference Husain and Roiser2018), and is directly altered by both serotonergic and dopaminergic medications (Admon et al., Reference Admon, Kaiser, Dillon, Beltzer, Goer, Olson and Pizzagalli2017; Stoy et al., Reference Stoy, Schlagenhauf, Sterzer, Bermpohl, Hägele, Suchotzki and Ströhle2012).

Degeneration of dopaminergic projections to the striatum is a core pathological feature of PD and LBD, with up to 80% of dopaminergic projections to the dorsal striatum lost by the time of diagnosis in PD (see Table 2) (Cheng et al., Reference Cheng, Ulane and Burke2010; Kish et al., Reference Kish, Shannak and Hornykiewicz1988). SSRIs such as fluoxetine bind at therapeutic doses in the ventral striatum and modify emotional valence and reward processing via changes in tonic and phasic activities of striatal neurons (Pasquereau et al., Reference Pasquereau, Drui, Saga, Richard, Millot, Météreau and Tremblay2021). Loss of these neurons is therefore likely to impact the pharmacodynamics and therapeutic effects of antidepressants.

Other regions such as the amygdala, involved in processing emotional aspects of information and generating emotional reactions, project to the hippocampus which participates in the creation and maintenance of emotional associations in memory (DeRubeis, Siegle, & Hollon, Reference DeRubeis, Siegle and Hollon2008). More negative recall of information is a core negative affective bias found in depression, and antidepressant therapy has been consistently shown to be associated with activation of the amygdala (Nord et al., Reference Nord, Barrett, Lindquist, Ma, Marwood, Satpute and Dalgleish2021). Prominent hippocampal and amygdala atrophy is a signature pattern of AD and FTD respectively (Barnes et al., Reference Barnes, Whitwell, Frost, Josephs, Rossor and Fox2006) and is associated with loss of emotional and behavioural regulation (Rosen et al., Reference Rosen, Perry, Murphy, Kramer, Mychack, Schuff and Miller2002). Neurodegeneration of these regions and other parts of the neural circuitry mediating affective biases is a likely key mechanism driving loss of therapeutic efficacy of antidepressants in dementia.

An alternative neurocognitive explanation for loss of antidepressant effect is that negative affective biases in depression in dementia are driven by different cognitive processes (see Table 3). For example, reward-related decision-making changes in AD have been found to be mediated by impairment in memory, inhibitory control and set-shifting rather than reward processing deficits (Perry & Kramer, Reference Perry and Kramer2015). Executive dysfunction, rather than reward valuation, has similarly been found to drive reward-based decision making in FTD (Perry & Kramer, Reference Perry and Kramer2015). In PD, reward processing impairment is dependent on dopaminergic medication state and subcategory of reward processing (Costello et al., Reference Costello, Berry, Reeves, Weil, Joyce, Howard and Roiser2021). These examples highlight how negative affective biases driving depression may have variable mechanisms which would not be targeted by existing antidepressant therapies.

Loss of neuroplasticity

Almost all antidepressants increase the expression of brain-derived neutrophic factor (BDNF) via neutrophic tyrosine kinase receptor 2 (TRKB) (Casarotto et al., Reference Casarotto, Girych, Fred, Kovaleva, Moliner, Enkavi and Castrén2021). BDNF promotes neuronal maturation, plasticity and synaptic transmission by increasing production of brain cholesterol which facilitates cell membrane phospholipid fluidity of lipid rafts where many antidepressants interact (Casarotto et al., Reference Casarotto, Girych, Fred, Kovaleva, Moliner, Enkavi and Castrén2021). Antidepressants, including SSRIs and even rapid-acting agents such as ketamine, bind to TRKB, facilitating synaptic localisation and activation by BDNF. This induces synaptic plasticity that is believed to be a key underlying mechanism of antidepressants effects on mood (Casarotto et al., Reference Casarotto, Girych, Fred, Kovaleva, Moliner, Enkavi and Castrén2021). Mutation of the TRKB antidepressant binding site leads to loss of cellular, behavioural and neuroplasticity induced changes by antidepressants in vitro and in animal models (Casarotto et al., Reference Casarotto, Girych, Fred, Kovaleva, Moliner, Enkavi and Castrén2021).

Across different dementia disorders, BDNF is markedly reduced and has been implicated as a key pathological mechanism of neurodegeneration (Ng, Ho, Tam, Kua, & Ho, Reference Ng, Ho, Tam, Kua and Ho2019; Zuccato & Cattaneo, Reference Zuccato and Cattaneo2009). Lower BDNF compared to healthy controls is seen in PD and AD, and worsens with disease progression (Zuccato & Cattaneo, Reference Zuccato and Cattaneo2009). Although, it remains unclear whether BDNF changes in PD and AD are a cause or consequence of disease, the first phase-I clinical trial of BDNF gene therapy for cognitive decline in AD commenced last year (Tuszynski, Reference Tuszynski2021). Substantial evidence supports a causal role of BDNF dysfunction in the pathogenesis of Huntington's dementia as the huntingtin protein plays a crucial role in the expression and vesicular transport of BDNF (Zuccato & Cattaneo, Reference Zuccato and Cattaneo2009).

Post-mortem studies have not supported a similar BDNF reduction in late-life depression compared to dementia (Nunes et al., Reference Nunes, Nascimento, Kim, Andreazza, Brentani, Suemoto and Lafer2018), and no study has yet investigated whether basal BDNF level predicts response to antidepressants. Further investigation of the effects of BDNF modulation on antidepressant response and mood symptoms in dementia is needed. It is likely that any BDNF-mediated therapeutic effect of antidepressants would be reduced in dementia due to marked reduction in BDNF levels. However, it could also be argued that if antidepressant efficacy is partly mediated by augmenting BDNF then strategies to increase BDNF levels could be an effective approach in dementia where it is lower.

Evidence suggests that post-synaptic modulation of calcium signalling is crucial for functional plasticity and regulation of the strength of synaptic transmission in response to neuronal activity via long-term potentiation or depression. Animal studies have shown SSRIs to protect against stress-induced synaptic neuroplasticity via direct blockade of voltage-activated L-type Ca2+ channels (Normann et al., Reference Normann, Frase, Haug, von Wolff, Clark, Münzer and Bischofberger2018). Epidemiological evidence has found an association between L-type calcium channel antagonist use (e.g. verapamil hydrochloride) and reduced rates of psychiatric hospitalisation and self-harm in bipolar disorder (Hayes et al., Reference Hayes, Lundin, Wicks, Lewis, Wong, Osborn and Dalman2019). This highlights how antidepressant function could be independent of their monoaminergic effects. Further investigation of the role calcium channel signalling has in mediating antidepressant effects is needed. However, this mechanism is also likely to be impacted by dementia pathology. Marked dysregulation of calcium homoeostasis is believed to play an important role in the development of AD neuropathology and hyperphosphorylated tau has been shown to directly impair L-type calcium channels (Stan et al., Reference Stan, Church, Randall, Harvey, Brown, Wilkinson and Marrion2022; Wang, Shi, & Wei, Reference Wang, Shi and Wei2017).

Why antidepressants are still used in dementia: The clinical dilemma

Given the different aetiology and potential risks of treatment, it may appear straightforward to conclude that antidepressants have no role in depression in dementia and should not be used; however, this neglects the dilemma that patients, carers and clinicians face.

First, although trials have shown no superiority of antidepressants compared to placebo, the choice to prescribe antidepressants in clinical practice is not between antidepressant and placebo, but antidepressant and nothing. Though non-pharmacological interventions are effective and should be considered first-line treatments, many patients do not respond or are unable to engage with these interventions, which often require a good level of cognitive function. Even if only acting as placebo, we know placebo is effective at improving symptoms in both depression and in dementia (Hyde, May, Xue, & Zhang, Reference Hyde, May, Xue and Zhang2017). Though clearly the use of antidepressants purely for their placebo benefit is unwise given the potential risks of harm and side effects, undoubtedly, this consideration will contribute to clinical decision making and represents an important reason why prescription of antidepressants in depressed people with dementia is continued.

Second, the lifetime risk of depression is high and it is estimated that up to half the population will experience one or more episodes of depression during their lifetime (Andrews, Poulton, & Skoog, Reference Andrews, Poulton and Skoog2005). When treating older adults with depression and dementia who have previously had episodes of depression successfully treated by antidepressants prior to developing dementia, the clinical question of whether to re-commence an antidepressant is difficult. Is this depression in the context of dementia or a relapse of existing major depressive disorder? Do antidepressants lose their efficacy in long-term depression users who develop dementia? Discontinuation of antidepressants in older adults with dementia who have remained on treatment for an extended period is similarly challenging, with evidence from one RCT showing worsening in depressive symptoms following discontinuation (Bergh, Selbæk, & Engedal, Reference Bergh, Selbæk and Engedal2012).

Third, accurate diagnosis of depression in the general population is reliant on recall of depressive symptoms over the previous two weeks and detailed description of subjective symptoms by patients. In patients with dementia who have marked cognitive impairment this is often not possible, and clinicians will be reliant on collateral information from relatives and carers. However, the accuracy of such information varies enormously and is dependent on the nature of the relationship; it can also place considerable demands on family members who may be experiencing significant carer stress and low mood themselves. High turnover of staff working shift patterns in long term-care facilities (Costello, Cooper, Marston, & Livingston, Reference Costello, Cooper, Marston and Livingston2020) can also complicate the process of making an accurate diagnosis and subsequent monitoring of depressive symptoms.

Finally, while antidepressants may not be effective in depression in dementia they are widely used for alternative indications which may overlap with depression. FTD guidelines support SSRI use to treat behavioural disinhibition and agitation despite no RCT having been conducted to date (O'Brien et al., Reference O'Brien, Holmes, Jones, Jones, Livingston, McKeith and Burns2017). Antidepressants are also used for treatment of neuropathic pain, which is highly prevalent in depression and dementia (Achterberg, Lautenbacher, Husebo, Erdal, & Herr, Reference Achterberg, Lautenbacher, Husebo, Erdal and Herr2019), and they are frequently (mis)used to treat agitation as a substitution strategy to avoid antipsychotic prescription (Maust, Kim, Chiang, & Kales, Reference Maust, Kim, Chiang and Kales2018).

The clinical complexity of diagnosis and management of depression in dementia, and use of antidepressants for other BPSD, clouds how clinicians decide whether to prescribe antidepressants. We must be honest with patients and carers about the lack of efficacy and potentially harmful consequences of antidepressant use in dementia, but also acknowledge how challenging clinical decision-making regarding prescription in this patient group can be.

Discussion

In this article we argue that antidepressants do not work in depression in dementia as it is likely a distinct entity that emerges as a consequence of a diverse array of pathological mechanisms, specific to each dementia syndrome, and which negates the therapeutic mechanisms of antidepressants. Antidepressants in depression without dementia have clinically meaningful small to moderate effects on improving clinical symptoms and preventing relapse (Cipriani et al., Reference Cipriani, Furukawa, Salanti, Chaimani, Atkinson, Ogawa and Geddes2018). However, expecting the same therapeutic effects on a heterogenous syndrome in a patient population with varying degrees of neurodegeneration, pathophysiology and cognitive function may simply be wishful thinking.

In recent years considerable progress has been made in our understanding of antidepressant mechanisms of action which has shed light on the reasons why they may be ineffective in dementia. Though currently no effective pharmacological interventions for depression in dementia exist, there is reason to be optimistic.

It is unlikely that a single well-specified neurobiological cause of depression in dementia will be discovered. However, if viewed as a core component of dementia pathology, it offers a potentially more homogenous entity to treat with a clearer understanding of the temporal onset and markers of disease than depression in the general population. For example, depression in PD, which often progresses to PD dementia, is characterised by early degeneration of dopamine neurons in the SN and loss of dopaminergic projections to the striatum. Apathy and anhedonia, both disorders of motivation which share cognitive mechanisms, are prominent in depression in PD, correlate with loss of dopaminergic projections to the striatum and are responsive to dopaminergic medication. Utilising this understanding of the progression of pathology, phenomenology, and cognitive deficits within each disease could open the door for novel interventions, such as dopaminergic agents for depression in PD or deep brain stimulation targeting the striatum. This clear involvement of a distinct pathological process with a consistent trajectory is not present in depression in patients without dementia and demonstrates how understanding depression in dementia can give mechanistic insights into depressive symptoms.

Effective development of treatments for depression in dementia will depend on refining phenotypes and integrating our understanding of the evolution of depression over time with the progression in dementia pathology. A personalised medicine approach using clinical and biological markers to identify different phenotypes and effective treatments is an important area of future research in this patient group. Recent work using existing large cohorts has identified different trajectories of depressive symptoms in older adults depending on AD biomarker status (Banning, Ramakers, Rosenberg, Lyketsos, & Leoutsakos, Reference Banning, Ramakers, Rosenberg, Lyketsos and Leoutsakos2021).

The processes discussed above of why antidepressants don't work in dementia offer insights and perspective for the future development of treatments for patients living with depression and dementia. Methods such as theory-driven computational modelling and approaching depression in dementia as a complex system of interacting behavioural, cognitive and neurobiological parts will be needed. However, these tools now exist and if integrated into future trials and drug development in depression and dementia could offer hope for identifying effective antidepressants for this patient group. The alternative of using the same approaches but expecting a different answer is unlikely to lead to the desperately needed treatments patients living with depression and dementia deserve.

Financial support

HC is supported by a Wellcome Trust Clinical Training Fellowship (175479) and JR is supported by Wellcome (101798/Z/13/Z).

Conflict of interest

There are no conflicts of interest for authors to disclose.

References

Achterberg, W., Lautenbacher, S., Husebo, B., Erdal, A., & Herr, K. (2019). Pain in dementia. Pain Reports, 5(1), e803. https://doi.org/10.1097/PR9.0000000000000803.CrossRefGoogle ScholarPubMed
Admon, R., Kaiser, R. H., Dillon, D. G., Beltzer, M., Goer, F., Olson, D. P., … Pizzagalli, D. A. (2017). Dopaminergic enhancement of striatal response to reward in major depression. American Journal of Psychiatry, 174(4), 378386. https://doi.org/10.1176/appi.ajp.2016.16010111.CrossRefGoogle ScholarPubMed
Allen, N. H. P., Gordon, S., Hope, T., & Burns, A. (1996). Manchester and Oxford Universities scale for the psychopathological assessment of dementia (MOUSEPAD). The British Journal of Psychiatry, 169(3), 293307. https://doi.org/10.1192/bjp.169.3.293.CrossRefGoogle ScholarPubMed
Altieri, S. C., Garcia-Garcia, A. L., Leonardo, E. D., & Andrews, A. M. (2012). Rethinking 5-HT1A receptors: Emerging modes of inhibitory feedback of relevance to emotion-related behavior. ACS Chemical Neuroscience, 4(1), 7283. https://doi.org/10.1021/cn3002174.CrossRefGoogle ScholarPubMed
Andrews, G., Poulton, R., & Skoog, I. (2005). Lifetime risk of depression: Restricted to a minority or waiting for most? The British Journal of Psychiatry, 187(6), 495496. https://doi.org/10.1192/bjp.187.6.495.CrossRefGoogle ScholarPubMed
Asmer, M. S., Kirkham, J., Newton, H., Ismail, Z., Elbayoumi, H., Leung, R. H., & Seitz, D. P. (2018). Meta-analysis of the prevalence of major depressive disorder among older adults with dementia. The Journal of Clinical Psychiatry, 79(5), 15460. https://doi.org/10.4088/JCP.17r11772.CrossRefGoogle ScholarPubMed
Attems, J., & Jellinger, K. A. (2014). The overlap between vascular disease and Alzheimer's disease – lessons from pathology. BMC Medicine, 12(1), 206. https://doi.org/10.1186/s12916-014-0206-2.CrossRefGoogle ScholarPubMed
Babulal, G. M., Ghoshal, N., Head, D., Vernon, E. K., Holtzman, D. M., Benzinger, T. L. S., … Roe, C. M. (2016). Mood changes in cognitively normal older adults are linked to Alzheimer's disease biomarker levels. The American Journal of Geriatric Psychiatry: Official Journal of the American Association for Geriatric Psychiatry, 24(11), 10951104. https://doi.org/10.1016/j.jagp.2016.04.004.CrossRefGoogle Scholar
Banerjee, S., Hellier, J., Dewey, M., Romeo, R., Ballard, C., Baldwin, R., … Burns, A. (2011). Sertraline or mirtazapine for depression in dementia (HTA-SADD): A randomised, multicentre, double-blind, placebo-controlled trial. The Lancet, 378(9789), 403411. https://doi.org/10.1016/S0140-6736(11)60830-1.CrossRefGoogle ScholarPubMed
Banerjee, S., High, J., Stirling, S., Shepstone, L., Swart, A. M., Telling, T., … Tabet, N. (2021). Study of mirtazapine for agitated behaviours in dementia (SYMBAD): A randomised, double-blind, placebo-controlled trial. The Lancet, 398(10310), 14871497. https://doi.org/10.1016/S0140-6736(21)01210-1.CrossRefGoogle ScholarPubMed
Banning, L. C. P., Ramakers, I. H. G. B., Rosenberg, P. B., Lyketsos, C. G., & Leoutsakos, J.-M. S. (2021). Alzheimer's disease biomarkers as predictors of trajectories of depression and apathy in cognitively normal individuals, mild cognitive impairment, and Alzheimer's disease dementia. International Journal of Geriatric Psychiatry, 36(1), 224234. https://doi.org/10.1002/gps.5418.CrossRefGoogle ScholarPubMed
Barnes, J., Whitwell, J. L., Frost, C., Josephs, K. A., Rossor, M., & Fox, N. C. (2006). Measurements of the amygdala and hippocampus in pathologically confirmed Alzheimer disease and frontotemporal lobar degeneration. Archives of Neurology, 63(10), 14341439. https://doi.org/10.1001/archneur.63.10.1434.CrossRefGoogle ScholarPubMed
Beck, A. T., Steer, R. A., Ball, R., & Ranieri, W. F. (1996). Comparison of beck depression inventories-IA and-II in psychiatric outpatients. Journal of Personality Assessment, 67(3), 588597. https://doi.org/10.1207/s15327752jpa6703_13.CrossRefGoogle ScholarPubMed
Bergh, S., Selbæk, G., & Engedal, K. (2012). Discontinuation of antidepressants in people with dementia and neuropsychiatric symptoms (DESEP study): Double blind, randomised, parallel group, placebo controlled trial. BMJ, 344, e1566. https://doi.org/10.1136/bmj.e1566.CrossRefGoogle ScholarPubMed
Betts, M. J., Kirilina, E., Otaduy, M. C. G., Ivanov, D., Acosta-Cabronero, J., Callaghan, M. F., … Hämmerer, D. (2019). Locus coeruleus imaging as a biomarker for noradrenergic dysfunction in neurodegenerative diseases. Brain, 142(9), 25582571. https://doi.org/10.1093/brain/awz193.CrossRefGoogle ScholarPubMed
Borza, T., Engedal, K., Bergh, S., Barca, M. L., Benth, J. Š., & Selbæk, G. (2015). The course of depressive symptoms as measured by the Cornell scale for depression in dementia over 74 months in 1158 nursing home residents. Journal of Affective Disorders, 175, 209216. https://doi.org/10.1016/j.jad.2014.12.053.CrossRefGoogle ScholarPubMed
Bradley, B. P., Mogg, K., & Millar, N. (1996). Implicit memory bias in clinical and non-clinical depression. Behaviour Research and Therapy, 34(11), 865879. https://doi.org/10.1016/S0005-7967(96)00074-5.CrossRefGoogle ScholarPubMed
Breining, A., Bonnet-Zamponi, D., Zerah, L., Micheneau, C., Riolacci-Dhoyen, N., Chan-Chee, C., … Leperre-Desplanques, A. (2017). Exposure to psychotropics in the French older population living with dementia: A nationwide population-based study. International Journal of Geriatric Psychiatry, 32(7), 750760. https://doi.org/10.1002/gps.4517.CrossRefGoogle ScholarPubMed
Broomfield, N. M., Davies, R., MacMahon, K., Ali, F., & Cross, S. M. B. (2007). Further evidence of attention bias for negative information in late life depression. International Journal of Geriatric Psychiatry, 22(3), 175180. https://doi.org/10.1002/gps.1655.CrossRefGoogle ScholarPubMed
Buch, A. M., & Liston, C. (2021). Dissecting diagnostic heterogeneity in depression by integrating neuroimaging and genetics. Neuropsychopharmacology, 46(1), 156175. https://doi.org/10.1038/s41386-020-00789-3.CrossRefGoogle ScholarPubMed
Casarotto, P. C., Girych, M., Fred, S. M., Kovaleva, V., Moliner, R., Enkavi, G., … Castrén, E. (2021). Antidepressant drugs act by directly binding to TRKB neurotrophin receptors. Cell, 184(5), 12991313.e19. https://doi.org/10.1016/j.cell.2021.01.034.CrossRefGoogle ScholarPubMed
Cheng, H.-C., Ulane, C. M., & Burke, R. E. (2010). Clinical progression in Parkinson's disease and the neurobiology of axons. Annals of Neurology, 67(6), 715725. https://doi.org/10.1002/ana.21995.CrossRefGoogle ScholarPubMed
Chow, T. W., Binns, M. A., Cummings, J. L., Lam, I., Black, S. E., Miller, B. L., … van Reekum, R. (2009). Apathy symptom profile and behavioral associations in frontotemporal dementia vs dementia of Alzheimer type. Archives of Neurology, 66(7), 888893. https://doi.org/10.1001/archneurol.2009.92.CrossRefGoogle ScholarPubMed
Cipriani, A., Furukawa, T. A., Salanti, G., Chaimani, A., Atkinson, L. Z., Ogawa, Y., … Geddes, J. R. (2018). Comparative efficacy and acceptability of 21 antidepressant drugs for the acute treatment of adults with major depressive disorder: A systematic review and network meta-analysis. Lancet (London, England), 391(10128), 13571366. https://doi.org/10.1016/S0140-6736(17)32802-7.CrossRefGoogle ScholarPubMed
Coppen, A. (1967). The biochemistry of affective disorders. The British Journal of Psychiatry, 113(504), 12371264. https://doi.org/10.1192/bjp.113.504.1237.CrossRefGoogle ScholarPubMed
Costello, H., Berry, A. J., Reeves, S., Weil, R. S., Joyce, E. M., Howard, R., … Roiser, J. P. (2021). Disrupted reward processing in Parkinson's disease and its relationship with dopamine state and neuropsychiatric syndromes: A systematic review and meta-analysis. Journal of Neurology, Neurosurgery & Psychiatry, 93(5), 555562. https://doi.org/10.1136/jnnp-2021-327762.CrossRefGoogle ScholarPubMed
Costello, H., Cooper, C., Marston, L., & Livingston, G. (2020). Burnout in UK care home staff and its effect on staff turnover: MARQUE English national care home longitudinal survey. Age and Ageing, 49(1), 7481. https://doi.org/10.1093/ageing/afz118.CrossRefGoogle Scholar
Cowen, P. J., & Browning, M. (2015). What has serotonin to do with depression? World Psychiatry, 14(2), 158160. https://doi.org/10.1002/wps.20229.CrossRefGoogle Scholar
Culo, S., Mulsant, B. H., Rosen, J., Mazumdar, S., Blakesley, R. E., Houck, P. R., & Pollock, B. G. (2010). Treating neuropsychiatric symptoms in dementia with Lewy bodies: A randomized controlled-trial. Alzheimer Disease & Associated Disorders, 24(4), 360364. https://doi.org/10.1097/WAD.0b013e3181e6a4d7.CrossRefGoogle ScholarPubMed
Cummings, J., Mintzer, J., Brodaty, H., Sano, M., Banerjee, S., Devanand, D. P., … Zhong, K. (2015). Agitation in cognitive disorders: International psychogeriatric association provisional consensus clinical and research definition. International Psychogeriatrics/Ipa, 27(1), 717. https://doi.org/10.1017/S1041610214001963.CrossRefGoogle ScholarPubMed
Cummings, J. L., Mega, M., Gray, K., Rosenberg-Thompson, S., Carusi, D. A., & Gornbein, J. (1994). The neuropsychiatric inventory: Comprehensive assessment of psychopathology in dementia. Neurology, 44(12), 23082308. https://doi.org/10.1212/WNL.44.12.2308.CrossRefGoogle ScholarPubMed
Deakin, J. B., Rahman, S., Nestor, P. J., Hodges, J. R., & Sahakian, B. J. (2004). Paroxetine does not improve symptoms and impairs cognition in frontotemporal dementia: A double-blind randomized controlled trial. Psychopharmacology, 172(4), 400408. https://doi.org/10.1007/s00213-003-1686-5.Google Scholar
Delazer, M., Sinz, H., Zamarian, L., & Benke, T. (2007). Decision-making with explicit and stable rules in mild Alzheimer's disease. Neuropsychologia, 45(8), 16321641. https://doi.org/10.1016/j.neuropsychologia.2007.01.006.CrossRefGoogle ScholarPubMed
den Brok, M. G. H. E., van Dalen, J. W., van Gool, W. A., Moll van Charante, E. P., de Bie, R. M. A., & Richard, E. (2015). Apathy in Parkinson's disease: A systematic review and meta-analysis. Movement Disorders, 30(6), 759769. https://doi.org/10.1002/mds.26208.CrossRefGoogle ScholarPubMed
DeRubeis, R. J., Siegle, G. J., & Hollon, S. D. (2008). Cognitive therapy versus medication for depression: Treatment outcomes and neural mechanisms. Nature Reviews Neuroscience, 9(10), 788796. https://doi.org/10.1038/nrn2345.CrossRefGoogle ScholarPubMed
Devanand, D. P., Miller, L., Richards, M., Marder, K., Bell, K., Mayeux, R., & Stern, Y. (1992). The Columbia University scale for psychopathology in Alzheimer's disease. Archives of Neurology, 49(4), 371376. https://doi.org/10.1001/archneur.1992.00530280051022.CrossRefGoogle ScholarPubMed
Doninger, N. A., & Bylsma, F. W. (2007). Inhibitory control and affective valence processing in dementia of the Alzheimer type. Journal of Neuropsychology, 1(1), 6583. https://doi.org/10.1348/174866407X180828.CrossRefGoogle ScholarPubMed
Drui, G., Carnicella, S., Carcenac, C., Favier, M., Bertrand, A., Boulet, S., & Savasta, M. (2014). Loss of dopaminergic nigrostriatal neurons accounts for the motivational and affective deficits in Parkinson's disease. Molecular Psychiatry, 19(3), 358367. https://doi.org/10.1038/mp.2013.3.CrossRefGoogle ScholarPubMed
Dudas, R., Malouf, R., McCleery, J., & Dening, T. (2018). Antidepressants for treating depression in dementia. Cochrane Database of Systematic Reviews, 8, 14651858. https://doi.org/10.1002/14651858.CD003944.pub2.Google ScholarPubMed
Ehrt, U., Brønnick, K., Leentjens, A. F. G., Larsen, J. P., & Aarsland, D. (2006). Depressive symptom profile in Parkinson's disease: A comparison with depression in elderly patients without Parkinson's disease. International Journal of Geriatric Psychiatry, 21(3), 252258. https://doi.org/10.1002/gps.1456.CrossRefGoogle ScholarPubMed
Elamin, M., Pender, N., Hardiman, O., & Abrahams, S. (2012). Social cognition in neurodegenerative disorders: A systematic review. Journal of Neurology, Neurosurgery & Psychiatry, 83(11), 10711079. https://doi.org/10.1136/jnnp-2012-302817.CrossRefGoogle ScholarPubMed
Elferink, M. W.-O., van Tilborg, I., & Kessels, R. P. C. (2015). Perception of emotions in mild cognitive impairment and Alzheimer's dementia: Does intensity matter? Translational Neuroscience, 6(1), 139149. https://doi.org/10.1515/tnsci-2015-0013.CrossRefGoogle Scholar
Elliott, M. S. J., Ballard, C. G., Kalaria, R. N., Perry, R., Hortobágyi, T., & Francis, P. T. (2009). Increased binding to 5-HT1A and 5-HT2A receptors is associated with large vessel infarction and relative preservation of cognition. Brain, 132(7), 18581865. https://doi.org/10.1093/brain/awp069.CrossRefGoogle ScholarPubMed
Erickson, K., Drevets, W. C., Clark, L., Cannon, D. M., Bain, E. E., Zarate, C. A., … Sahakian, B. J. (2005). Mood-Congruent bias in affective Go/No-Go performance of unmedicated patients with major depressive disorder. American Journal of Psychiatry, 162(11), 21712173. https://doi.org/10.1176/appi.ajp.162.11.2171.CrossRefGoogle ScholarPubMed
Fairfield, B., Colangelo, M., Mammarella, N., Di Domenico, A., & Cornoldi, C. (2017). Affective false memories in Dementia of Alzheimer's type. Psychiatry Research, 249, 915. https://doi.org/10.1016/j.psychres.2016.12.036.CrossRefGoogle ScholarPubMed
Feng, L., Scherer, S. C., Tan, B. Y., Chan, G., Fong, N. P., & Ng, T. P. (2010). Comorbid cognitive impairment and depression is a significant predictor of poor outcomes in hip fracture rehabilitation. International Psychogeriatrics, 22(2), 246253. https://doi.org/10.1017/S1041610209991487.CrossRefGoogle ScholarPubMed
Francis, P. T., & Perry, E. K. (2007). Cholinergic and other neurotransmitter mechanisms in Parkinson's disease, Parkinson's disease dementia, and dementia with Lewy bodies. Movement Disorders, 22(Suppl. 17), S351S357. https://doi.org/10.1002/mds.21683.CrossRefGoogle ScholarPubMed
Fried, E. I., Flake, J. K., & Robinaugh, D. J. (2022). Revisiting the theoretical and methodological foundations of depression measurement. Nature Reviews Psychology, 1(6), Art. 6. https://doi.org/10.1038/s44159-022-00050-2.CrossRefGoogle Scholar
Fritze, F., Ehrt, U., Hortobagyi, T., Ballard, C., & Aarsland, D. (2011). Depressive symptoms in Alzheimer's disease and Lewy body dementia: A one-year follow-up study. Dementia and Geriatric Cognitive Disorders, 32(2), 143149. https://doi.org/10.1159/000332016.CrossRefGoogle ScholarPubMed
Fritze, S., Spanagel, R., & Noori, H. R. (2017). Adaptive dynamics of the 5-HT systems following chronic administration of selective serotonin reuptake inhibitors: A meta-analysis. Journal of Neurochemistry, 142(5), 747755. https://doi.org/10.1111/jnc.14114.CrossRefGoogle ScholarPubMed
Furukawa, T. A., Cipriani, A., Cowen, P. J., Leucht, S., Egger, M., & Salanti, G. (2019). Optimal dose of selective serotonin reuptake inhibitors, venlafaxine, and mirtazapine in major depression: A systematic review and dose-response meta-analysis. The Lancet Psychiatry, 6(7), 601609. https://doi.org/10.1016/S2215-0366(19)30217-2.CrossRefGoogle ScholarPubMed
Gerson, S., Belin, T. R., Kaufman, A., Mintz, J., & Jarvik, L. (1999). Pharmacological and psychological treatments for depressed older patients: A meta-analysis and overview of recent findings. Harvard Review of Psychiatry, 7(1), 128.CrossRefGoogle ScholarPubMed
Gibb, W. R., Mountjoy, C. Q., Mann, D. M., & Lees, A. J. (1989). The substantia nigra and ventral tegmental area in Alzheimer's disease and Down's syndrome. Journal of Neurology, Neurosurgery & Psychiatry, 52(2), 193200. https://doi.org/10.1136/jnnp.52.2.193.CrossRefGoogle ScholarPubMed
Gildengers, A. G., Houck, P. R., Mulsant, B. H., Pollock, B. G., Mazumdar, S., Miller, M. D., … Reynolds, C. F. III (2002). Course and rate of antidepressant response in the very old. Journal of Affective Disorders, 69(1), 177184. https://doi.org/10.1016/S0165-0327(01)00334-2.CrossRefGoogle ScholarPubMed
Graham, N. L., Emery, T., & Hodges, J. R. (2004). Distinctive cognitive profiles in Alzheimer's disease and subcortical vascular dementia. Journal of Neurology, Neurosurgery & Psychiatry, 75(1), 6171.Google ScholarPubMed
Grinberg, L. T., Rüb, U., Ferretti, R. E. L., Nitrini, R., Farfel, J. M., Polichiso, L., … Group, B. B. B. S. (2009). The dorsal raphe nucleus shows phospho-tau neurofibrillary changes before the transentorhinal region in Alzheimer's disease. A precocious onset? Neuropathology and Applied Neurobiology, 35(4), 406416. https://doi.org/10.1111/j.1365-2990.2008.00997.x.CrossRefGoogle ScholarPubMed
Grudzien, A., Shaw, P., Weintraub, S., Bigio, E., Mash, D. C., & Mesulam, M. M. (2007). Locus coeruleus neurofibrillary degeneration in aging, mild cognitive impairment and early Alzheimer's disease. Neurobiology of Aging, 28(3), 327335. https://doi.org/10.1016/j.neurobiolaging.2006.02.007.CrossRefGoogle ScholarPubMed
Halahakoon, D. C., Kieslich, K., O'Driscoll, C., Nair, A., Lewis, G., & Roiser, J. P. (2020). Reward-processing behavior in depressed participants relative to healthy volunteers: A systematic review and meta-analysis. JAMA Psychiatry, 77(12), 12861295. https://doi.org/10.1001/jamapsychiatry.2020.2139.CrossRefGoogle ScholarPubMed
Halliday, G. M., Li, Y. W., Blumbergs, P. C., Joh, T. H., Cotton, R. G. H., Howe, P. R. C., … Geffen, L. B. (1990). Neuropathology of immunohistochemically identified brainstem neurons in Parkinson's disease. Annals of Neurology, 27(4), 373385. https://doi.org/10.1002/ana.410270405.CrossRefGoogle ScholarPubMed
Harmer, C. J., Duman, R. S., & Cowen, P. J. (2017). How do antidepressants work? New perspectives for refining future treatment approaches. The Lancet Psychiatry, 4(5), 409418. https://doi.org/10.1016/S2215-0366(17)30015-9.CrossRefGoogle ScholarPubMed
Hayes, J. F., Lundin, A., Wicks, S., Lewis, G., Wong, I. C. K., Osborn, D. P. J., & Dalman, C. (2019). Association of hydroxylmethyl glutaryl coenzyme A reductase inhibitors, L-type calcium channel antagonists, and biguanides with rates of psychiatric hospitalization and self-harm in individuals with serious mental illness. JAMA Psychiatry, 76(4), 382390. https://doi.org/10.1001/jamapsychiatry.2018.3907.CrossRefGoogle ScholarPubMed
Heitz, C., Noblet, V., Phillipps, C., Cretin, B., Vogt, N., Philippi, N., … Blanc, F. (2016). Cognitive and affective theory of mind in dementia with Lewy bodies and Alzheimer's disease. Alzheimer's Research & Therapy, 8(1), 10. https://doi.org/10.1186/s13195-016-0179-9.CrossRefGoogle ScholarPubMed
Holland, N., Robbins, T. W., & Rowe, J. B. (2021). The role of noradrenaline in cognition and cognitive disorders. Brain, 144(8), 22432256. https://doi.org/10.1093/brain/awab111.CrossRefGoogle ScholarPubMed
Huey, E. D., Putnam, K. T., & Grafman, J. (2006). A systematic review of neurotransmitter deficits and treatments in frontotemporal dementia. Neurology, 66(1), 1722. https://doi.org/10.1212/01.wnl.0000191304.55196.4d.CrossRefGoogle ScholarPubMed
Husain, M., & Roiser, J. P. (2018). Neuroscience of apathy and anhedonia: A transdiagnostic approach. Nature Reviews Neuroscience, 19(8), 470484. https://doi.org/10.1038/s41583-018-0029-9.CrossRefGoogle ScholarPubMed
Hyde, A. J., May, B. H., Xue, C. C., & Zhang, A. L. (2017). Variation in placebo effect sizes in clinical trials of oral interventions for management of the behavioral and psychological symptoms of dementia (BPSD): A systematic review and meta-analysis. The American Journal of Geriatric Psychiatry, 25(9), 9941008. https://doi.org/10.1016/j.jagp.2017.02.022.CrossRefGoogle ScholarPubMed
Janzing, J. G. E., Hooijer, C., van ‘t Hof, M. A., & Zitman, F. G. (2002). Depression in subjects with and without dementia: A comparison using GMS-AGECAT. International Journal of Geriatric Psychiatry, 17(1), 15. https://doi.org/10.1002/gps.526.CrossRefGoogle ScholarPubMed
Jao, Y.-L., Liu, W., Williams, K., Chaudhury, H., & Parajuli, J. (2019). Association between environmental stimulation and apathy in nursing home residents with dementia. International Psychogeriatrics, 31(8), 11091120. https://doi.org/10.1017/S1041610219000589.CrossRefGoogle ScholarPubMed
Jester, D. J., Molinari, V., Zgibor, J. C., & Volicer, L. (2021). Prevalence of psychotropic polypharmacy in nursing home residents with dementia: A meta-analysis. International Psychogeriatrics, 33(10), 10831098. https://doi.org/10.1017/S1041610220004032.CrossRefGoogle ScholarPubMed
Johnell, K., Jonasdottir Bergman, G., Fastbom, J., Danielsson, B., Borg, N., & Salmi, P. (2017). Psychotropic drugs and the risk of fall injuries, hospitalisations and mortality among older adults. International Journal of Geriatric Psychiatry, 32(4), 414420. https://doi.org/10.1002/gps.4483.CrossRefGoogle ScholarPubMed
Karantzoulis, S., & Galvin, J. E. (2011). Distinguishing Alzheimer's disease from other major forms of dementia. Expert Review of Neurotherapeutics, 11(11), 15791591. https://doi.org/10.1586/ern.11.155.CrossRefGoogle ScholarPubMed
Kaye, E. D., Petrovic-Poljak, A., Verhoeff, N. P. L. G., & Freedman, M. (2010). Frontotemporal dementia and pharmacologic interventions. The Journal of Neuropsychiatry and Clinical Neurosciences, 22(1), 1929. https://doi.org/10.1176/jnp.2010.22.1.19.CrossRefGoogle ScholarPubMed
Kelly, S. C., He, B., Perez, S. E., Ginsberg, S. D., Mufson, E. J., & Counts, S. E. (2017). Locus coeruleus cellular and molecular pathology during the progression of Alzheimer's disease. Acta Neuropathologica Communications, 5, 8. https://doi.org/10.1186/s40478-017-0411-2.CrossRefGoogle ScholarPubMed
Kish, S. J., Shannak, K., & Hornykiewicz, O. (1988). Uneven pattern of dopamine loss in the striatum of patients with idiopathic Parkinson's disease. New England Journal of Medicine, 318(14), 876880. https://doi.org/10.1056/NEJM198804073181402.CrossRefGoogle ScholarPubMed
Komulainen, K., Airaksinen, J., Savelieva, K., Gluschkoff, K., García Velázquez, R., Elovainio, M., … Jokela, M. (2021). Network dynamics of depressive symptoms in antidepressant medication treatment: Secondary analysis of eight clinical trials. Molecular Psychiatry, 26(7), 33283335. https://doi.org/10.1038/s41380-020-00884-3.CrossRefGoogle ScholarPubMed
Lai, M. K. P., Tsang, S. W. Y., Francis, P. T., Esiri, M. M., Keene, J., Hope, T., & Chen, C. P. L.-H. (2003). Reduced serotonin 5-HT1A receptor binding in the temporal cortex correlates with aggressive behavior in Alzheimer disease. Brain Research, 974(1), 8287. https://doi.org/10.1016/S0006-8993(03)02554-X.CrossRefGoogle ScholarPubMed
Lamers, F., Rhebergen, D., Merikangas, K. R., de Jonge, P., Beekman, A. T. F., & Penninx, B. W. J. H. (2012). Stability and transitions of depressive subtypes over a 2-year follow-up. Psychological Medicine, 42(10), 20832093. https://doi.org/10.1017/S0033291712000141.CrossRefGoogle ScholarPubMed
Lanctôt, K. L., Herrmann, N., & Mazzotta, P. (2001). Role of serotonin in the behavioral and psychological symptoms of dementia. The Journal of Neuropsychiatry and Clinical Neurosciences, 13(1), 521. https://doi.org/10.1176/jnp.13.1.5.CrossRefGoogle ScholarPubMed
Lavretsky, H., Zheng, L., Weiner, M. W., Mungas, D., Reed, B., Kramer, J. H., … Mack, W. J. (2010). Association of depressed mood and mortality in older adults with and without cognitive impairment in a prospective naturalistic study. The American Journal of Psychiatry, 167(5), 589597. https://doi.org/10.1176/appi.ajp.2009.09020280.CrossRefGoogle Scholar
Lebert, F., Stekke, W., Hasenbroekx, C., & Pasquier, F. (2004). Frontotemporal dementia: A randomised, controlled trial with trazodone. Dementia and Geriatric Cognitive Disorders, 17(4), 355359. https://doi.org/10.1159/000077171.CrossRefGoogle ScholarPubMed
Le Heron, C., Apps, M. A. J., & Husain, M. (2018). The anatomy of apathy: A neurocognitive framework for amotivated behaviour. Neuropsychologia, 118, 5467. https://doi.org/10.1016/j.neuropsychologia.2017.07.003.CrossRefGoogle Scholar
Lemke, M. R., Brecht, H. M., Koester, J., Kraus, P. H., & Reichmann, H. (2005). Anhedonia, depression, and motor functioning in Parkinson's disease during treatment with pramipexole. The Journal of Neuropsychiatry and Clinical Neurosciences, 17(2), 214220. https://doi.org/10.1176/jnp.17.2.214.CrossRefGoogle ScholarPubMed
Leontjevas, R., Teerenstra, S., Smalbrugge, M., Vernooij-Dassen, M. J. F. J., Bohlmeijer, E. T., Gerritsen, D. L., & Koopmans, R. T. C. M. (2013). More insight into the concept of apathy: A multidisciplinary depression management program has different effects on depressive symptoms and apathy in nursing homes. International Psychogeriatrics, 25(12), 19411952. https://doi.org/10.1017/S1041610213001440.CrossRefGoogle Scholar
Levy, R., & Dubois, B. (2006). Apathy and the functional anatomy of the prefrontal cortex-basal ganglia circuits. Cerebral Cortex (New York, N.Y.: 1991), 16(7), 916928. https://doi.org/10.1093/cercor/bhj043.CrossRefGoogle ScholarPubMed
Li, X., & Zhang, C. (2020). Comparative efficacy of nine antidepressants in treating Chinese patients with post-stroke depression: A network meta-analysis. Journal of Affective Disorders, 266, 540548. https://doi.org/10.1016/j.jad.2020.02.005.CrossRefGoogle ScholarPubMed
Liu, K. Y., Stringer, A. E., Reeves, S. J., & Howard, R. J. (2018). The neurochemistry of agitation in Alzheimer's disease: A systematic review. Ageing Research Reviews, 43, 99107. https://doi.org/10.1016/j.arr.2018.03.003.CrossRefGoogle ScholarPubMed
Livingston, G., Barber, J., Marston, L., Rapaport, P., Livingston, D., Cousins, S., … Cooper, C. (2017). Prevalence of and associations with agitation in residents with dementia living in care homes: MARQUE cross-sectional study. BJPsych Open, 3(4), 171178. https://doi.org/10.1192/bjpo.bp.117.005181.CrossRefGoogle ScholarPubMed
Lyness, S. A., Zarow, C., & Chui, H. C. (2003). Neuron loss in key cholinergic and aminergic nuclei in Alzheimer disease: A meta-analysis. Neurobiology of Aging, 24(1), 123. https://doi.org/10.1016/S0197-4580(02)00057-X.CrossRefGoogle ScholarPubMed
Mann, D. M., South, P. W., Snowden, J. S., & Neary, D. (1993). Dementia of frontal lobe type: Neuropathology and immunohistochemistry. Journal of Neurology, Neurosurgery, and Psychiatry, 56(6), 605614.CrossRefGoogle ScholarPubMed
Maria, G.-G., & Juan, G.-G. (2017). Negative bias in the perception and memory of emotional information in Alzheimer disease. Journal of Geriatric Psychiatry and Neurology, 30(3), 131139. https://doi.org/10.1177/0891988716686833.CrossRefGoogle ScholarPubMed
Martinez, C., Jones, R. W., & Rietbrock, S. (2013). Trends in the prevalence of antipsychotic drug use among patients with Alzheimer's disease and other dementias including those treated with antidementia drugs in the community in the UK: A cohort study. BMJ Open, 3(1), e002080. https://doi.org/10.1136/bmjopen-2012-002080.CrossRefGoogle ScholarPubMed
Maust, D. T., Kim, H. M., Chiang, C., & Kales, H. C. (2018). Association of the centers for Medicare & Medicaid Services’ national partnership to improve dementia care with the use of antipsychotics and other psychotropics in long-term care in the United States from 2009 to 2014. JAMA Internal Medicine, 178(5), 640647. https://doi.org/10.1001/jamainternmed.2018.0379.CrossRefGoogle ScholarPubMed
McGuinness, B., Barrett, S. L., Craig, D., Lawson, J., & Passmore, A. P. (2010). Attention deficits in Alzheimer's disease and vascular dementia. Journal of Neurology, Neurosurgery & Psychiatry, 81(2), 157159. https://doi.org/10.1136/jnnp.2008.164483.CrossRefGoogle ScholarPubMed
Mogg, K., Bradley, B. P., & Williams, R. (1995). Attentional bias in anxiety and depression: The role of awareness. British Journal of Clinical Psychology, 34(1), 1736. https://doi.org/10.1111/j.2044-8260.1995.tb01434.x.CrossRefGoogle ScholarPubMed
Montgomery, S. A., & Åsberg, M. (1979). A new depression scale designed to be sensitive to change. The British Journal of Psychiatry, 134(4), 382389. https://doi.org/10.1192/bjp.134.4.382.CrossRefGoogle ScholarPubMed
Moretti, R., Torre, P., Antonello, R. M., Cazzato, G., & Bava, A. (2003). Frontotemporal dementia: Paroxetine as a possible treatment of behavior symptoms. European Neurology, 49(1), 1319. https://doi.org/10.1159/000067021.CrossRefGoogle ScholarPubMed
Ng, T. K. S., Ho, C. S. H., Tam, W. W. S., Kua, E. H., & Ho, R. C.-M. (2019). Decreased serum brain-derived neurotrophic factor (BDNF) levels in patients with Alzheimer's disease (AD): A systematic review and meta-analysis. International Journal of Molecular Sciences, 20(2), Art. 2. https://doi.org/10.3390/ijms20020257.CrossRefGoogle Scholar
Nord, C. L., Barrett, L. F., Lindquist, K. A., Ma, Y., Marwood, L., Satpute, A. B., & Dalgleish, T. (2021). Neural effects of antidepressant medication and psychological treatments: A quantitative synthesis across three meta-analyses. The British Journal of Psychiatry, 219(4), 546550. https://doi.org/10.1192/bjp.2021.16.CrossRefGoogle ScholarPubMed
Normann, C., Frase, S., Haug, V., von Wolff, G., Clark, K., Münzer, P., … Bischofberger, J. (2018). Antidepressants rescue stress-induced disruption of synaptic plasticity via serotonin transporter–independent inhibition of L-type calcium channels. Biological Psychiatry, 84(1), 5564. https://doi.org/10.1016/j.biopsych.2017.10.008.CrossRefGoogle ScholarPubMed
Nunes, P. V., Nascimento, C. F., Kim, H. K., Andreazza, A. C., Brentani, H. P., Suemoto, C. K., … Lafer, B. (2018). Low brain-derived neurotrophic factor levels in post-mortem brains of older adults with depression and dementia in a large clinicopathological sample. Journal of Affective Disorders, 241, 176181. https://doi.org/10.1016/j.jad.2018.08.025.CrossRefGoogle Scholar
O'Brien, J. T., Holmes, C., Jones, M., Jones, R., Livingston, G., McKeith, I., … Burns, A. (2017). Clinical practice with anti-dementia drugs: A revised (third) consensus statement from the British Association for Psychopharmacology. Journal of Psychopharmacology (Oxford, England), 31(2), 147168. https://doi.org/10.1177/0269881116680924.CrossRefGoogle ScholarPubMed
Ohm, D. T., Peterson, C., Lobrovich, R., Cousins, K. A. Q., Gibbons, G. S., McMillan, C. T., … Irwin, D. J. (2020). Degeneration of the locus coeruleus is a common feature of tauopathies and distinct from TDP-43 proteinopathies in the frontotemporal lobar degeneration spectrum. Acta Neuropathologica, 140(5), 675693. https://doi.org/10.1007/s00401-020-02210-1.CrossRefGoogle ScholarPubMed
Orgeta, V., Qazi, A., Spector, A., & Orrell, M. (2015). Psychological treatments for depression and anxiety in dementia and mild cognitive impairment: Systematic review and meta-analysis. The British Journal of Psychiatry, 207(4), 293298. https://doi.org/10.1192/bjp.bp.114.148130.CrossRefGoogle ScholarPubMed
Papakostas, G. I. (2006). Dopaminergic-based pharmacotherapies for depression. European Neuropsychopharmacology, 16(6), 391402. https://doi.org/10.1016/j.euroneuro.2005.12.002.CrossRefGoogle ScholarPubMed
Park, H. K., & Chung, S. J. (2013). New perspective on Parkinsonism in frontotemporal lobar degeneration. Journal of Movement Disorders, 6(1), 18. https://doi.org/10.14802/jmd.13001.CrossRefGoogle ScholarPubMed
Pasquereau, B., Drui, G., Saga, Y., Richard, A., Millot, M., Météreau, E., … Tremblay, L. (2021). Selective serotonin reuptake inhibitor treatment retunes emotional valence in primate ventral striatum. Neuropsychopharmacology, 46(12), 20732082. https://doi.org/10.1038/s41386-021-00991-x.CrossRefGoogle ScholarPubMed
Perry, D. C., & Kramer, J. H. (2015). Reward processing in neurodegenerative disease. Neurocase, 21(1), 120133. https://doi.org/10.1080/13554794.2013.873063.CrossRefGoogle ScholarPubMed
Radloff, L. S. (1977). The CES-D scale: A self-report depression scale for research in the general population. Applied Psychological Measurement, 1, 385401. https://doi.org/10.1177/014662167700100306.CrossRefGoogle Scholar
Ramanan, S., Bertoux, M., Flanagan, E., Irish, M., Piguet, O., Hodges, J. R., & Hornberger, M. (2017). Longitudinal executive function and episodic memory profiles in behavioral-variant frontotemporal dementia and Alzheimer's disease. Journal of the International Neuropsychological Society, 23(1), 3443. https://doi.org/10.1017/S1355617716000837.CrossRefGoogle ScholarPubMed
Rapp, M. A., Schnaider-Beeri, M., Wysocki, M., Guerrero-Berroa, E., Grossman, H. T., Heinz, A., & Haroutunian, V. (2011). Cognitive decline in patients with dementia as a function of depression. The American Journal of Geriatric Psychiatry: Official Journal of the American Association for Geriatric Psychiatry, 19(4), 357363. https://doi.org/10.1097/JGP.0b013e3181e898d0.CrossRefGoogle ScholarPubMed
Reisberg, B., Auer, S. R., & Monteiro, I. M. (1997). Behavioral pathology in Alzheimer's disease (BEHAVE-AD) rating scale. International Psychogeriatrics, 8(Suppl. 3), 301308. https://doi.org/10.1017/S1041610297003529.CrossRefGoogle Scholar
Richardson-Jones, J. W., Craige, C. P., Guiard, B. P., Stephen, A., Metzger, K. L., Kung, H. F., … Leonardo, E. D. (2010). 5-HT1A autoreceptor levels determine vulnerability to stress and response to antidepressants. Neuron, 65(1), 4052. https://doi.org/10.1016/j.neuron.2009.12.003.CrossRefGoogle ScholarPubMed
Riedel, O., Heuser, I., Klotsche, J., Dodel, R., & Wittchen, H.-U. (2010). Occurrence risk and structure of depression in Parkinson disease with and without dementia: Results from the GEPAD study. Journal of Geriatric Psychiatry and Neurology, 23(1), 2734. https://doi.org/10.1177/0891988709351833.CrossRefGoogle ScholarPubMed
Rinne, J. O., Laine, M., Kaasinen, V., Norvasuo-Heilä, M. K., Någren, K., & Helenius, H. (2002). Striatal dopamine transporter and extrapyramidal symptoms in frontotemporal dementia. Neurology, 58(10), 14891493. https://doi.org/10.1212/WNL.58.10.1489.CrossRefGoogle ScholarPubMed
Rock, P. L., Roiser, J. P., Riedel, W. J., & Blackwell, A. D. (2014). Cognitive impairment in depression: A systematic review and meta-analysis. Psychological Medicine, 44(10), 20292040. https://doi.org/10.1017/S0033291713002535.CrossRefGoogle ScholarPubMed
Roiser, J. P., Elliott, R., & Sahakian, B. J. (2012). Cognitive mechanisms of treatment in depression. Neuropsychopharmacology, 37(1), 117136. https://doi.org/10.1038/npp.2011.183.CrossRefGoogle ScholarPubMed
Rosen, H. J., Perry, R. J., Murphy, J., Kramer, J. H., Mychack, P., Schuff, N., … Miller, B. L. (2002). Emotion comprehension in the temporal variant of frontotemporal dementia. Brain, 125(10), 22862295. https://doi.org/10.1093/brain/awf225.CrossRefGoogle ScholarPubMed
Rüb, U., Del Tredici, K., Schultz, C., Thal, D. R., Braak, E., & Braak, H. (2000). The evolution of Alzheimer's disease-related cytoskeletal pathology in the human raphe nuclei. Neuropathology and Applied Neurobiology, 26(6), 553567. https://doi.org/10.1046/j.0305-1846.2000.00291.x.CrossRefGoogle ScholarPubMed
Ruhé, H. G., Mason, N. S., & Schene, A. H. (2007). Mood is indirectly related to serotonin, norepinephrine and dopamine levels in humans: A meta-analysis of monoamine depletion studies. Molecular Psychiatry, 12(4), 331359. https://doi.org/10.1038/sj.mp.4001949.CrossRefGoogle ScholarPubMed
Rush, A. J., Gullion, C. M., Basco, M. R., Jarrett, R. B., & Trivedi, M. H. (1996). The inventory of depressive symptomatology (IDS): Psychometric properties. Psychological Medicine, 26(3), 477486. https://doi.org/10.1017/S0033291700035558.CrossRefGoogle ScholarPubMed
Rush, A. J., Trivedi, M. H., Ibrahim, H. M., Carmody, T. J., Arnow, B., Klein, D. N., … Keller, M. B. (2003). The 16-item quick inventory of depressive symptomatology (QIDS), clinician rating (QIDS-C), and self-report (QIDS-SR): A psychometric evaluation in patients with chronic major depression. Biological Psychiatry, 54(5), 573583. https://doi.org/10.1016/S0006-3223(02)01866-8.CrossRefGoogle ScholarPubMed
Ruthirakuhan, M., Herrmann, N., Vieira, D., Gallagher, D., & Lanctôt, K. L. (2019). The roles of apathy and depression in predicting Alzheimer disease: A longitudinal analysis in older adults with mild cognitive impairment. The American Journal of Geriatric Psychiatry, 27(8), 873882. https://doi.org/10.1016/j.jagp.2019.02.003.CrossRefGoogle ScholarPubMed
Sampogna, G., Vecchio, V. D., Giallonardo, V., Luciano, M., & Fiorillo, A. (2020). Diagnosis, clinical features, and therapeutic implications of agitated depression. Psychiatric Clinics, 43(1), 4757. https://doi.org/10.1016/j.psc.2019.10.011.Google ScholarPubMed
Sasaki, M., Shibata, E., Tohyama, K., Kudo, K., Endoh, J., Otsuka, K., & Sakai, A. (2008). Monoamine neurons in the human brain stem: Anatomy, magnetic resonance imaging findings, and clinical implications. NeuroReport, 19(17), 16491654. https://doi.org/10.1097/WNR.0b013e328315a637.CrossRefGoogle ScholarPubMed
Sheline, Y. I., Pieper, C. F., Barch, D. M., Welsh-Boehmer, K., McKinstry, R. C., MacFall, J. R., … Doraiswamy, P. M. (2010). Support for the vascular depression hypothesis in late-life depression: Results of a 2-site, prospective, antidepressant treatment trial. Archives of General Psychiatry, 67(3), 277285. https://doi.org/10.1001/archgenpsychiatry.2009.204.CrossRefGoogle ScholarPubMed
Singh-Manoux, A., Dugravot, A., Fournier, A., Abell, J., Ebmeier, K., Kivimäki, M., & Sabia, S. (2017). Trajectories of depressive symptoms before diagnosis of dementia: A 28-year follow-up study. JAMA Psychiatry, 74(7), 712718. https://doi.org/10.1001/jamapsychiatry.2017.0660.CrossRefGoogle ScholarPubMed
Sinz, H., Zamarian, L., Benke, T., Wenning, G. K., & Delazer, M. (2008). Impact of ambiguity and risk on decision making in mild Alzheimer's disease. Neuropsychologia, 46(7), 20432055. https://doi.org/10.1016/j.neuropsychologia.2008.02.002.CrossRefGoogle ScholarPubMed
Smith, G. S., Barrett, F. S., Joo, J. H., Nassery, N., Savonenko, A., Sodums, D. J., … Workman, C. I. (2017). Molecular imaging of serotonin degeneration in mild cognitive impairment. Neurobiology of Disease, 105, 3341. https://doi.org/10.1016/j.nbd.2017.05.007.CrossRefGoogle ScholarPubMed
Snowden, M. B., Atkins, D. C., Steinman, L. E., Bell, J. F., Bryant, L. L., Copeland, C., & Fitzpatrick, A. L. (2015). Longitudinal association of dementia and depression. The American Journal of Geriatric Psychiatry: Official Journal of the American Association for Geriatric Psychiatry, 23(9), 897905. https://doi.org/10.1016/j.jagp.2014.09.002.CrossRefGoogle ScholarPubMed
Stan, G. F., Church, T. W., Randall, E., Harvey, J. R. M., Brown, J. T., Wilkinson, K. A., … Marrion, N. V. (2022). Tau isoform-specific enhancement of L-type calcium current and augmentation of after hyperpolarization in rat hippocampal neurons. Scientific Reports, 12(1), 15231. https://doi.org/10.1038/s41598-022-18648-0.CrossRefGoogle Scholar
Steinbusch, H. W. M., Dolatkhah, M. A., & Hopkins, D. A. (2021). Chapter 2 – anatomical and neurochemical organization of the serotonergic system in the mammalian brain and in particular the involvement of the dorsal raphe nucleus in relation to neurological diseases. In Di Giovanni, G. & De Deurwaerdere, P. (Eds.), Progress in brain research (Vol. 261, pp. 4181). Elsevier. https://doi.org/10.1016/bs.pbr.2021.02.003.Google Scholar
Stoy, M., Schlagenhauf, F., Sterzer, P., Bermpohl, F., Hägele, C., Suchotzki, K., … Ströhle, A. (2012). Hyporeactivity of ventral striatum towards incentive stimuli in unmedicated depressed patients normalizes after treatment with escitalopram. Journal of Psychopharmacology, 26(5), 677688. https://doi.org/10.1177/0269881111416686.CrossRefGoogle ScholarPubMed
Strenziok, M., Pulaski, S., Krueger, F., Zamboni, G., Clawson, D., & Grafman, J. (2011). Regional brain atrophy and impaired decision making on the balloon analog risk task in behavioral variant frontotemporal dementia. Cognitive and Behavioral Neurology, 24(2), 5967. https://doi.org/10.1097/WNN.0b013e3182255a7c.CrossRefGoogle ScholarPubMed
Swift, K. M., Gross, B. A., Frazer, M. A., Bauer, D. S., Clark, K. J. D., Vazey, E. M., … Poe, G. R. (2018). Abnormal locus coeruleus sleep activity alters sleep signatures of memory consolidation and impairs place cell stability and spatial memory. Current Biology, 28(22), 35993609.e4. https://doi.org/10.1016/j.cub.2018.09.054.CrossRefGoogle ScholarPubMed
Taylor Tavares, J. V., Clark, L., Cannon, D. M., Erickson, K., Drevets, W. C., & Sahakian, B. J. (2007). Distinct profiles of neurocognitive function in unmedicated unipolar depression and bipolar II depression. Biological Psychiatry, 62(8), 917924. https://doi.org/10.1016/j.biopsych.2007.05.034.CrossRefGoogle ScholarPubMed
Torralva, T., Dorrego, F., Sabe, L., Chemerinski, E., & Starkstein, S. E. (2000). Impairments of social cognition and decision making in Alzheimer's disease. International Psychogeriatrics, 12(3), 359368. https://doi.org/10.1017/S1041610200006463.CrossRefGoogle ScholarPubMed
Trieu, C., Gossink, F., Stek, M. L., Scheltens, P., Pijnenburg, Y. A. L., & Dols, A. (2020). Effectiveness of pharmacological interventions for symptoms of behavioral variant frontotemporal dementia: A systematic review. Cognitive and Behavioral Neurology, 33(1), 115. https://doi.org/10.1097/WNN.0000000000000217.CrossRefGoogle ScholarPubMed
Troeung, L., Egan, S. J., & Gasson, N. (2013). A meta-analysis of randomised placebo-controlled treatment trials for depression and anxiety in Parkinson's disease. PLoS ONE, 8(11), e79510. https://doi.org/10.1371/journal.pone.0079510.CrossRefGoogle ScholarPubMed
Tuszynski, M. (2021). A Phase I Study to Assess the Safety, Tolerability and Preliminary Efficacy of AAV2–BDNF [Adeno-Associated Virus (AAV)-Based, Vector-Mediated Delivery of Human Brain Derived Neurotrophic Factor] in Subjects With Early Alzheimer's Disease and Mild Cognitive Impairment (Clinical Trial Registration No. NCT05040217). clinicaltrials.gov. https://clinicaltrials.gov/ct2/show/NCT05040217.Google Scholar
Uher, R., Perlis, R. H., Henigsberg, N., Zobel, A., Rietschel, M., Mors, O., … McGuffin, P. (2012). Depression symptom dimensions as predictors of antidepressant treatment outcome: Replicable evidence for interest-activity symptoms. Psychological Medicine, 42(5), 967980. https://doi.org/10.1017/S0033291711001905.CrossRefGoogle ScholarPubMed
van der Velden, R. M. J., Broen, M. P. G., Kuijf, M. L., & Leentjens, A. F. G. (2018). Frequency of mood and anxiety fluctuations in Parkinson's disease patients with motor fluctuations: A systematic review. Movement Disorders, 33(10), 15211527. https://doi.org/10.1002/mds.27465.CrossRefGoogle ScholarPubMed
van der Zande, J. J., Joling, M., Happach, I. G., Vriend, C., Scheltens, Ph., Booij, J., & Lemstra, A. W. (2019). Serotonergic deficits in dementia with Lewy bodies with concomitant Alzheimer's disease pathology: An 123I-FP-CIT SPECT study. NeuroImage: Clinical, 25, 102062. https://doi.org/10.1016/j.nicl.2019.102062.CrossRefGoogle Scholar
Verdelho, A., Hénon, H., Lebert, F., Pasquier, F., & Leys, D. (2004). Depressive symptoms after stroke and relationship with dementia: A three-year follow-up study. Neurology, 62(6), 905911. https://doi.org/10.1212/01.WNL.0000115107.66957.8C.CrossRefGoogle ScholarPubMed
Vermeiren, Y., Van Dam, D., Aerts, T., Engelborghs, S., Martin, J.-J., & De Deyn, P. P. (2015). The monoaminergic footprint of depression and psychosis in dementia with Lewy bodies compared to Alzheimer's disease. Alzheimer's Research & Therapy, 7(1), 7. https://doi.org/10.1186/s13195-014-0090-1.CrossRefGoogle ScholarPubMed
Volicer, L., Frijters, D. H. M., & Van der Steen, J. T. (2012). Relationship between symptoms of depression and agitation in nursing home residents with dementia. International Journal of Geriatric Psychiatry, 27(7), 749754. https://doi.org/10.1002/gps.2800.CrossRefGoogle ScholarPubMed
Wang, H., & Jia, J. (2007). [Different patterns of brain activation between patients of Alzheimer's disease with and without depression: A functional MRI study during emotion Stroop task]. Zhonghua Yi Xue Za Zhi, 87(13), 885888.Google ScholarPubMed
Wang, J., Li, Y., Huang, Z., Wan, W., Zhang, Y., Wang, C., … Jin, L. (2018). Neuromelanin-sensitive magnetic resonance imaging features of the substantia nigra and locus coeruleus in de novo Parkinson's disease and its phenotypes. European Journal of Neurology, 25(7), 949–e73. https://doi.org/10.1111/ene.13628.CrossRefGoogle ScholarPubMed
Wang, Y., Shi, Y., & Wei, H. (2017). Calcium dysregulation in Alzheimer's disease: A target for new drug development. Journal of Alzheimer's Disease & Parkinsonism, 7(5), 374. https://doi.org/10.4172/2161-0460.1000374.Google ScholarPubMed
Watt, J. A., Goodarzi, Z., Veroniki, A. A., Nincic, V., Khan, P. A., Ghassemi, M., … Straus, S. E. (2021). Comparative efficacy of interventions for reducing symptoms of depression in people with dementia: Systematic review and network meta-analysis. BMJ, 372, n532. https://doi.org/10.1136/bmj.n532.CrossRefGoogle ScholarPubMed
Wert-Carvajal, C., Reneaux, M., Tchumatchenko, T., & Clopath, C. (2022). Dopamine and serotonin interplay for valence-based spatial learning. Cell Reports, 39(2), 110645. https://doi.org/10.1016/j.celrep.2022.110645.CrossRefGoogle ScholarPubMed
Wong, S., Irish, M., Savage, G., Hodges, J. R., Piguet, O., & Hornberger, M. (2019). Strategic value-directed learning and memory in Alzheimer's disease and behavioural-variant frontotemporal dementia. Journal of Neuropsychology, 13(2), 328353. https://doi.org/10.1111/jnp.12152.CrossRefGoogle ScholarPubMed
Yang, Y., & Schmitt, H. P. (2001). Frontotemporal dementia: Evidence for impairment of ascending serotoninergic but not noradrenergic innervation. Acta Neuropathologica, 101(3), 256270. https://doi.org/10.1007/s004010000293.CrossRefGoogle Scholar
Yuen, G. S., Bhutani, S., Lucas, B. J., Gunning, F. M., AbdelMalak, B., Seirup, J. K., … Alexopoulos, G. S. (2015). Apathy in late-life depression: Common, persistent, and disabling. The American Journal of Geriatric Psychiatry, 23(5), 488494. https://doi.org/10.1016/j.jagp.2014.06.005.CrossRefGoogle ScholarPubMed
Zarow, C., Lyness, S. A., Mortimer, J. A., & Chui, H. C. (2003). Neuronal loss Is greater in the locus coeruleus than nucleus Basalis and substantia nigra in Alzheimer and Parkinson diseases. Archives of Neurology, 60(3), 337341. https://doi.org/10.1001/archneur.60.3.337.CrossRefGoogle ScholarPubMed
Zhao, Q.-F., Tan, L., Wang, H.-F., Jiang, T., Tan, M.-S., Tan, L., … Yu, J.-T. (2016). The prevalence of neuropsychiatric symptoms in Alzheimer's disease: Systematic review and meta-analysis. Journal of Affective Disorders, 190, 264271. https://doi.org/10.1016/j.jad.2015.09.069.CrossRefGoogle ScholarPubMed
Zhou, F.-M., Liang, Y., Salas, R., Zhang, L., De Biasi, M., & Dani, J. A. (2005). Corelease of dopamine and serotonin from striatal dopamine terminals. Neuron, 46(1), 6574. https://doi.org/10.1016/j.neuron.2005.02.010.CrossRefGoogle ScholarPubMed
Zubenko, G. S., Moossy, J., & Kopp, U. (1990). Neurochemical correlates of major depression in primary dementia. Archives of Neurology, 47(2), 209214. https://doi.org/10.1001/archneur.1990.00530020117023.CrossRefGoogle ScholarPubMed
Zuccato, C., & Cattaneo, E. (2009). Brain-derived neurotrophic factor in neurodegenerative diseases. Nature Reviews Neurology, 5(6), 311322. https://doi.org/10.1038/nrneurol.2009.54.CrossRefGoogle ScholarPubMed
Zung, W. W. K. (1965). A self-rating depression scale. Archives of General Psychiatry, 12(1), 6370. https://doi.org/10.1001/archpsyc.1965.01720310065008.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Reproduced forest plot of meta-analysis of included Cochrane review randomised controlled trials of antidepressants for depression in dementia. (Dudas et al. 2018) (AD - Alzheimer's dementia, VD - Vascular dementia, M - mixed dementia, PDD - Parkinson's disease dementia, SSRI - selective serotonin reuptake inhibitor, TCA - tricyclic antidepressant, SNRI - serotonin-noradrenaline reuptake inhibitor, NASSA - noradrenaline and specific serotonergic antidepressant).

Figure 1

Table 1. Depressive symptoms across seven depression rating scales that are also captured by measures of other BPSD symptoms (not depression items) on four BPSD measures

Figure 2

Table 2. Neurodegeneration of monoaminergic nuclei in dementia subtypes

Figure 3

Table 3. Examples of neurocognitive affective bias reported in depression and dementia