You are on page 1of 29

International Journal of Neuropsychopharmacology (2014), 17, 1009–1037.

© CINP 2013 ARTICLE


doi:10.1017/S1461145712001496

On ‘polypharmacy’ and multi-target agents,


complementary strategies for improving
the treatment of depression:
a comparative appraisal

Mark J. Millan
Unité du Recherche et Découverte en Neurosciences, Institut de Recherches Servier, Croissy sur Seine, Paris, France

Abstract

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Major depression is a heterogeneous disorder, both in terms of symptoms, ranging from anhedonia to
cognitive impairment, and in terms of pathogenesis, with many interacting genetic, epigenetic,
developmental and environmental causes. Accordingly, it seems unlikely that depressive states could be
fully controlled by a drug possessing one discrete mechanism of action and, in the wake of disappointing
results with several classes of highly selective agent, multi-modal treatment concepts are attracting
attention. As concerns pharmacotherapy, there are essentially two core strategies. First, multi-target anti-
depressants that act via two or more complementary mechanisms and, second, polypharmacy, which refers
to co-administration of two distinct drugs, usually in separate pills. Both multi-target agents and poly-
pharmacy ideally couple a therapeutically unexploited action to a clinically established mechanism in
order to enhance efficacy, moderate side-effects, accelerate onset of action and treat a broader range of
symptoms. The melatonin MT1/MT2 agonist and 5-HT2C antagonist, agomelatine, which is effective in
the short- and long-term treatment of depression, exemplifies the former approach, while evidence-
based polypharmacy is illustrated by the adjunctive use of second-generation antipsychotics with serotonin
reuptake inhibitors for treatment of resistant depression. Histone acetylation and methylation, ghrelin
signalling, inflammatory modulators, metabotropic glutamate-7 receptors and trace amine-associated-1
receptors comprise attractive substrates for new multi-target and polypharmaceutical strategies. The
present article outlines the rationale underpinning multi-modal approaches for treating depression, and
critically compares and contrasts the pros and cons of established and potentially novel multi-target vs.
polypharmaceutical treatments. On balance, the former appear the most promising for the elaboration,
development and clinical implementation of innovative concepts for the more effective management of
depression.
Received 2 October 2012; Reviewed 8 November 2012; Revised 9 November 2012; Accepted 13 November 2012;
First published online 30 May 2013
Key words: Depression, epigenetic, histone, multi-functional, trace amine, treatment-resistance.

Introduction In light of their multi-factorial origins and complex


clinical profiles, it seems unlikely that disorders such
Psychiatric disorders are generally characterized by a
as major depression and schizophrenia could be fully
broad range of symptoms and high co-morbidity
controlled in all patients by a drug possessing one
with other conditions, and they can be attributed to a
single mechanism of action. This point is of consider-
diverse palette of neurochemical and structural dys-
able significance since the last decade has been domi-
functions provoked in turn by multifarious genetic,
nated by the search for highly selective agents acting
developmental, environmental and epigenetic factors.
at a single target hypothesized to play a key role in
the induction and/or control of specific disorders. Un-
fortunately, despite often compelling preclinical data,
Address for correspondence: Dr M. J. Millan, Unité du Recherche et
many selective agents have yielded mitigated results
Découverte en Neurosciences, Institut de recherches Servier,
125 chemin du ronde, 78290 Croissy sur Seine, Paris, France.
in clinical trials and not been progressed to authoriz-
Tel: 00 33 1 55 722425 Fax: 00 33 1 55 722082 ation. This is not to say, however, that the target in
Email: mark.millan@fr.netgrs.com question is necessarily irrelevant or inactive and,
1010 M. J. Millan

apart from the use of selective drugs in subpopulations dopamine (DA) D2/D3/5-HT2A antagonism plus
of patients and/or against certain subsets of symptoms, 5-HT1A partial agonism for antipsychotics – accounts
many targets might be more effectively exploited in for their beneficial effects in treating depression and
association with other mechanisms of therapeutic schizophrenia, respectively (Millan, 2006; Meltzer
activity. As outlined in this article, there are essentially et al., 2012; Quesseveur et al., 2012).
two ways of achieving this goal with pharmacothera- Currently, there is particular interest in novel multi-
peutics: polypharmacy (drug combinations); and multi- modal therapies that act at discrete subsets of targets
target drugs (several actions possessed by one drug). mediating desirable actions while side-stepping mech-
In fact, the term ‘polypharmacy’ has something of anisms evoking undesirable side-effects. In this case,
an unfortunate reputation since it is sometimes prac- both polypharmacy (drug combinations) and multi-
tised in an unstandardized and ad hoc basis without target agents are of potential interest and these strat-
formal supporting data, particularly in patients suffer- egies share a suite of therapeutic objectives, which,
ing from schizophrenia (Goodwin et al., 2009; Zink if fully realized, could markedly enhance the treatment
et al., 2010) and bipolar disorder (Lin et al., 2006; and lives of patients suffering from depression

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Goldberg et al., 2009). Counterproductively, the con- (Table 1). The present article discusses the basic prin-
comitant prescription of numerous drugs to certain ciples and the pros and cons of polypharmacy as com-
psychiatrically ill patients – in particular the elderly, pared to multi-target medication, evoking particular
who typically take around six to eight – may com- mechanisms that have been harnessed by both. It
promise efficacy and provoke serious adverse drug also considers a palette of innovative concepts under
reactions (Prudent et al., 2008). This type of poly- experimental exploration, which may eventually lead
pharmacy is often associated with co-morbidity and to the availability of more effective multi-modal anti-
generally encompasses a broad range of medication, depressants. The focus on major depression seems par-
including agents for controlling somatic complaints, ticularly appropriate in light of the disturbing current
as well as psychiatric and/or neurological problems trend to disengage from ‘R and D’ in this therapeutic
(Mojtabai and Olfson, 2010). Major depression is also domain despite well-recognized problems of treat-
characterized by an increasing tendency for multiple ment-resistance and sub-optimal remission (Miller,
drug use over the past decades, not only in geriatric 2010; Nutt and Goodwin, 2011).
populations but also in adults and the young (Glezer
et al., 2009; Goodwin et al., 2009; McIntyre and Jerrell,
Major depression, a complex and heterogeneous
2009; Mojtabai and Olfson, 2010; Serna et al., 2010).
clinical profile with multiple risk factors
Conversely, the present article is concerned with
the more positive dimension of evidence-based poly- Major depression is characterized by the cardinal
pharmacy. That is, the controlled, validated and symptoms of depressed mood and anhedonia, coupled
authorized use of drug combinations for improving to a state of despair and hopelessness, severe fatigue
antidepressant efficacy without any worsening of toler- and loss of concentration, suicidal ideation and, in
ance, in particular following guidelines in treatment- extreme cases, actual attempts at suicide (Millan,
resistant patients who fail to respond adequately to 2006; Sartorius et al., 2007; Uher, 2011). In addition,
monotherapy (Trivedi et al., 2006; Melander et al., it is accompanied by a broad spectrum of other symp-
2008; Shelton et al., 2010; Connolly and Thase, 2011; toms. Some are seen in a majority of depressed individ-
Al-Harbi, 2012; Schlaepfer et al., 2012). uals, such as cognitive impairment and anxiety,
As regards drugs possessing multiple mechanisms whereas others are seen in only a minority, such as
of action, there has been a tendency to disparagingly psychotic episodes and pain (Fig. 1). Moreover,
consider them as ‘dirty’ and to accentuate the side- depression is commonly co-morbid with: (1) psychia-
effects triggered by their ‘off-target’ actions. For tric conditions such as obsessive–compulsive disorder,
example, the antimuscarinic, α1-adrenoceptor (AR) generalized anxiety disorder and schizophrenia;
antagonist and histaminergic H1 antagonist properties (2) neurological disorders such as Parkinson’s disease,
of tricyclics (such as amitriptyline) and antipsychotics Alzheimer’s disease and chronic pain; (3) somatic dis-
(such as clozapine) compromise cognition and elicit eases such as diabetes, osteoporosis and cardiovascular
numerous autonomic and cardiovascular side-effects disease (Millan, 2006; Sartorius et al., 2007; McIntyre
(Millan, 2006; Gillman, 2007; Sartorius et al., 2007). On et al., 2012). While not every patient will display all
the other hand, other components of their multi-target symptoms, depression is clearly a heterogeneous dis-
profiles – monoamine reuptake inhibition plus 5-HT2C order with a broad range of symptoms requiring
antagonist properties for tricyclic antidepressants and treatment.
Complementary strategies for improving the treatment of depression 1011

Table 1. Major therapeutic aims of polypharmacy and multi-target pharmacotherapy in the treatment of major depression

Enhance treatment efficacy in terms both of: (1) the extent and breadth of remission in individual patients; (2) the number of
patients showing clinically significant improvement.
Improve quality of treatment not only in terms of rating scales but also as assessed by self-evaluation, and by concrete measures of
quality of life, real-world function and socio-professional integration.
Reduce the risk of relapse: (1) in the course of treatment (tachyphylaxis); (2) following termination of administration.
Accelerate therapeutic efficacy, both in terms of: (1) initial delay to onset; (2) attainment of full effect.
Broaden the therapeutic range of treatment to control not only depressed mood and anhedonia (cardinal symptoms) but also
symptoms like cognitive impairment and anxiety.
Reduce the incidence of side-effects (especially compared to SSRIs) and increase the ‘therapeutic window’ of doses eliciting
undesirable effects as compared to those eliciting beneficial actions.
Promote patient compliance and decrease the risk of premature discontinuation of treatment.

SSRIs, Selective serotonin reuptake inhibitors.

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


A number of related terms are applied (not always consistently) for the use of more than one drug to treat depression and other
central nervous system disorders, like association, combination, add-on, adjunctive, augmentation, etc. Polypharmacy in the pre-
sent context mainly refers to ‘augmentation’ strategies, whereby a second potentiating drug does not possess significant anti-
depressant properties itself. In contrast, a ‘combination’ strategy refers to the co-joint administration of two mechanistically
different antidepressants. However, this distinction is often ambivalent since, even if a drug class is not authorized as a clinically
effective treatment for depression, it may exert a favourable influence on depressed mood, anhedonia and other symptoms of
depression. Examples include antagonists at 5-HT2C and NK1 receptors.

It is possible that a single anomaly, such as a genetic 2012; Heim and Binder, 2012; Schloesser et al., 2012).
mutation, could ultimately trigger a broad range of Thus, the multifaceted symptomology of depression
symptoms, in particular if it occurred early in develop- is mirrored by a complex hierarchy of cellular (neuro-
ment – and perinatal trauma is indeed a risk factor nal and glial) anomalies manifested across overarch-
for depression (Uher, 2011; Heim and Binder, 2012). ing cerebral regions. It is also important to note that
However, although certain monogenic neurodevelop- certain pathological changes anticipate the appearance
mental conditions are associated with a broad range of unambiguous clinical symptoms and diagnosis
of dysfunction affecting both the brain and other (Millan, 2006; Heim and Binder, 2012; Duman and
organs (Kramer and van Bokhoven, 2009; Millan, Voleti 2012).
2013; Sanchez-Mut et al., 2012), depression is a poly- The above observations have several important
genic disorder (cumulative impact of many genes implications for treatment. First, since network shifts
with minor effects) that can be triggered or aggravated are not always reversible, treatment should be initiated
by adverse environmental events occurring at essen- as soon as possible – ideally preventatively, once
tially any time of life from conception and foetal devel- robust biomarkers become available (Millan 2008;
opment via infancy and puberty to adulthood and Schmidt et al., 2011; Uher, 2011). Second, as discussed
senescence (Fig. 2) (Millan, 2006; Lohoff, 2010; Uher, herein, it is likely that multiple mechanisms will need
2011). Interestingly, both genetic and environmental to be harnessed for the broad-based control of de-
factors converge onto epigenetic mechanisms control- pressed states across substantial populations of
ling gene expression, which include DNA methylation patients, questioning the wisdom of highly selective
and post-transcriptional histone marking as well as the agents. Third, multi-modal therapies concern both
control of mRNA processing and translation by non- agents designed to normalize a pathological process
coding RNAs (Covington et al., 2010; Lohoff, 2010; causing depression, as well as symptomatic inter-
Uher, 2011; Millan, 2013; Mouillet-Richard et al., ventions for relief of symptoms by the manipulation
2012; Sun et al., 2012). This interplay of mechanisms of ‘intact’, compensatory substrates not affected by
favouring (and countering) depressed states leads to depression per se.
a complex pattern of neuroplastic signalling and struc-
tural anomalies, both within and between neurones,
Genome-driven, rational drug discovery: a decade
and impacting a diversity of neural networks in the
of selective antidepressants in perspective
frontal hippocampus, amygdala and other corticolim-
bic structures (Fig. 1; Millan 2006; Covington et al., There is a multitude of (interrelated and non-exclusive)
2010; Autry and Monteggia, 2012; Dumas and Voleti, explanations for the limited progress seen over the last
1012 M. J. Millan

Agents with complementary mechanisms of action

Circadian
dysruption
Poor Sexual
sleep dysfunction

Somatic Depressed
Psychotic
symptoms, mood,
episodes
pain anhedonia

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Motor
retardation, Cognitive
excitation deficits
Anxiety,
nervousness

Improved efficacy, ↑ rapidity of action, control of other symptoms

Fig. 1. Symptoms other than depressed mood commonly associated with major depression. In addition to depressed mood
and anhedonia, which are cardinal symptoms of major depression, it is often accompanied by a broad palette of other
symptoms contributing to functional impairment. As outlined in the text, depression is also frequently co-morbid with a
wide range of other central nervous system and somatic diseases, such generalized anxiety disorder and schizophrenia,
Parkinson’s and Alzheimer’s disease, diabetes and cardiovascular disease. The complex pattern of symptoms and
co-morbidity implies the involvement of numerous pathological processes and supports arguments in favour of treatments
acting via several, complementary mechanisms of action.

few decades in the pharmacotherapy of depression. drugs from national institutions post-launch, in par-
They relate to all dimensions of the ‘R and D’ process ticular bearing in mind the comparatively low rate of
as well as to authorization of new medication, embrac- success, yet high cost of development for central ner-
ing preclinical, clinical, regulatory, intellectual prop- vous system (CNS) drugs vs. other therapeutic areas
erty and commercial issues. Several important points (Brady et al., 2009; Miller, 2010; Nutt and Goodwin,
may be highlighted: persistent question-marks over 2011).
the pertinence of animal models of depression; poor Three additional and interlinked issues that have
experimental validation of novel targets; the relevance dominated the agenda over the last decade should be
of tests for evaluating potential antidepressant activity; briefly discussed since they are highly relevant to
their translational dimension, whether comparable pro- the development and use (or non-use) of multi-target
cedures can be undertaken in humans; lack of rigorous drugs and polypharmacy. First, the impact of geno-
patient recruitment for clinical investigations, use of mics and ‘rational’ drug discovery (Fig. 3); second,
antiquated rating scales and a strikingly high response the above-mentioned preoccupation with highly-
to placebo (the most common cause of failed trials); the selective agents; third, the prioritization of novel, non-
need to show superiority of a new drug if not over pla- monoaminergic targets – with a particular focus on
cebo, then over another antidepressant; increasingly intracellular substrates of plasticity (Millan, 2006,
stringent safety concerns; limited patent life for new 2008; Covington et al., 2010; Duman and Voleti, 2012;
drugs, generic competition and the difficulty in rapidly Autry and Monteggia, 2012; Connolly and Thase,
negotiating a realistic level of reimbursement for novel 2012; Schloesser et al., 2012). It was originally thought
Complementary strategies for improving the treatment of depression 1013

Vast array of potential causes – diverse pathologies

Lifelong: Developmental:
Stress
environmental/internal childhood/adolescent
adverse events trauma

aNumerous
Diverse bComplex

depressed changes in DNA/


interacting
histone
risk genes, states and ‘marking’ by
CNVs, inherited
and de novo
symptoms methylation,
acetylation,
mutations

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


phosphorylation

Genetica risk factors: Epigeneticb control


Non -coding of gene expression
epistatic networks
RNAs

(Control of mRNA processing and translation)

No single trigger - no single solution


(Causal factors NOT synonymous with treatment targets)

Fig. 2. Multiple and interacting causes of major depression throughout life. Mirroring the heterogeneity of major
depression in terms of its clinical profile, no single factor has been identified as causal. Dependent on the individual
(and probably symptom) it reflects a deleterious interplay of both genetic and environmental (early and late life) factors,
which in many cases converge upon epigenetic mechanisms that modify gene expression without any change in gene
sequence. As regards genetic factors, multiple genes of small effect appear to be incriminated, as well as more extensive
copy number variants (CNVs) and both inherited and de novo mutations. As for epigenetic mechanism, anomalous
histone (and probably DNA) marking, a dysfunction of non-coding RNAs (such as microRNAs) and aberrant control
of mRNA translation are likely involved. While uncontrolled, severe and chronic stress is a threat throughout life,
stress during early childhood is an important risk factor, both immediately and for later life even when stress has
dissipated.

that cloning the human genome would yield a sig- high-throughput screening would ‘revolutionize’ the
nificant number of new genes with novel functions, detection of innovative drugs at established and
permitting a clarification of the causes of CNS dysfunc- novel targets (the ‘magic of high numbers’), this did
tion in psychiatric disorders and offering innovative not materialize, not least since the procedures used
targets for improved therapy. With hindsight, this were rather simplistic and mainly aimed at identifi-
proved naive. The number of protein-coding genes cation of highly selective ligands (Brady et al., 2009;
(about 21 000, or 3% of the genome) in humans scar- Campbell, 2010). In addition, despite impressive con-
cely differs from many ‘simpler species’ and complex- ceptual foundations and extensive experimental data,
ity is derived more from epigenetic and other modes of many selective antidepressants advanced into patients
transcriptional control, including regulatory and struc- failed to demonstrate efficacy sufficient for registration
tural roles of many classes of protein non-coding RNA, (Millan, 2006, 2009; Miller, 2010). Finally, reflecting an
and the operation of genes in networks (Millan, 2006; understandable desire to explore novel therapeutic
Cardon et al., 2009; Manolio et al., 2009; Penrod mechanisms, monoaminergic substrates were largely
et al., 2011). Moreover, studies of the human genome neglected (‘from monoamines to genomics’) despite
have not yet led to any radically new insights the fact that there remained (and still remains)
into the causes of depression nor unveiled a palette scope for their potentially more effective exploitation
of new sites for its treatment. Further, although it when coupled to other, non-monoaminergic sites
was anticipated that combinatorial chemistry and (see below).
1014 M. J. Millan

a Gene-rush: patents taken on many genes


REALITY
b Genetic,
developmental
(difficult, long, costly)
and environmental factors
b Many interacting genes implicated
c Low success rate
Functiond

Human ‘Druggable Depressionb Selective


Development
genome genea’ related protein Antidep.

SNP, man/KO, mice

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Combinatorial chemistry/HTSc:
Hit → Lead
d Many (pleiotropy)
d Beneficial and deleterious
d Challenging to establish
‘RATIONAL’
Fig. 3. The rational and reality of drug discovery for improved antidepressant (antidep) agents. Upon sequencing of the
human genome, it was hoped that its ‘mining’ would rapidly identify many new potential targets for treating depression and
other poorly controlled disorders. Unfortunately, as outlined in Figs. 1 and 2, depression is complex both as regards its causes
and its characteristics, so the simplistic idea of finding a dysfunctional gene [by, for example, single nucleotide polymorphism
(SNP) studies in man and gene knock-out (KO) techniques in mice] has proven elusive. Moreover, much of the genomic
signal can be overwritten by epigenetic processes, many genes reciprocally interact with others to modify their mutual effects
(epistasis), many have several functions – both favourable and detrimental – and gene functions are in general hard to
establish. In addition, many genes were locked up in patents at the time of cloning the genome so ironically lost to
research. Finally, ‘rational’ drug discovery based on high throughput screening (HTS) techniques coupled to combinatorial
chemistry did not prove very effective in the search for leads for clinically effective antideps. The ‘reality’ is that
genome-driven rational drug discovery remains very challenging, time-consuming and expensive, just like other modes of
drug discovery.

From highly selective to multi-modal strategies for conviction remains that NK1 receptors, CRF1 receptors
the improved treatment of depression and certain other targets unsuccessfully evaluated in
the clinic, as well as targets where clinical data are
Ironically, then, the disavowal of monoaminergic mech- awaited, such as metabotropic glutamate (mGluR) 2
anisms as obsolete in the search for improved treat- receptors (Bespalov et al., 2008; Campo et al., 2011),
ment was not followed by the anticipated emergence α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
of novel drugs aimed at hitherto unknown ‘genomic’ (AMPA) receptors (Farley et al., 2010; Nations et al.,
targets, but rather by a frustrating succession of incon- 2012) and 5-HT7 receptors (Mnie-Filali et al., 2011),
clusive clinical trials with selective ligands acting at may well be of therapeutic relevance. However, they
non-monoaminergic sites, such as neurokinin1 (NK1) will best be exploited in a more effective multi-modal
and corticotrophin releasing factor (CRF1) receptors manner for the treatment of depression; that is, either
(Quartara et al., 2009; Kehne and Cain, 2010). This in association with ‘alternative’ (non-medication) treat-
led to the inference that these mechanisms might actu- ments or integrated with complementary mechanisms
ally be red herrings generated by misleading pre- in polypharmacy and multi-target drugs (Fig. 4).
clinical observations. However, these therapeutic In addition to negative findings with highly selec-
trials were performed in far more challenging circum- tive agents in clinical trials, several arguments support
stances (not least, a high response to placebo) than pre- the assertion that multi-modal pharmacotherapy may
decessor substances such as selective inhibitors of 5-HT be more effective. First, despite their improved toler-
reuptake (SSRIs) and the assumption of a lack of ance, SSRIs are no more effective than tricyclic agents.
clinical relevance may be an over-interpretation. The While 5-HT/noradrenaline (NA) reuptake inhibitors
Complementary strategies for improving the treatment of depression 1015

Co-administration: ‘true’ polypharmacy

a e.g. Citalopram, SSRI a


+ NK 1 Ant b 2 drugs in 2
Fluoxetine separate pills
b e.g. GR205,171,
‘Hybrid’
MK869

NK 1 Ant 2 drugs in one and


SSRI NK 1 Ant the same pill
SSRI

SSRI: ↑Synaptic
levels of 5-HT Multi-target

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


1 drug possessing
NK 1 Ant/ both properties
NK1 antagonist: blockade
SSRI – in one pill
of Substance P overdrive

Selectively Multi- S41744 Dual- Designed Synonyms


Non-selective functional acting bipotent

Fig. 4. Basic principles of polypharmacy as compared to multi-target drugs as illustrated by inhibition of 5-HT reuptake and
blockade of neurokinin 1 (NK1) receptors. 5-HT reuptake inhibitors (SSRIs) are designed to elevate extracellular levels of
5-HT, while NK1 antagonists (Ant) are thought to counter the deleterious effects of Substance P and also to potentiate the
actions of SSRIs. These complementary and synergistic mechanisms of action can be united either by genuine ‘polypharmacy’
(co-administration of two selective drugs), multi-target pharmacotherapy, whereby the two pharmacological actions are
united in one chemical structure, or a ‘hybrid’ solution, whereby two separate drugs are mixed into one pill. The choice of
solution will be influenced by many factors, including drug availability for polypharmacy and whether structure–activity
relationships actually allow the two actions to be incorporated into one single, developable structure.

appear to globally be more effective than SSRIs (with Thase, 2011; Al-Harbi, 2012). Other mechanisms of
the possible exception of escitalopram – see below), SSRI potentiation likewise act at sites very different
tricyclics are still recognized as the most effective of from 5-HT transporters – see below. Third, both precli-
antidepressants, likely reflecting their interaction not nical and clinical evidence suggests that electroconvul-
only with 5-HT and/or NA transporters, but also sive therapy as well as deep-brain and transcranial
other sites such as 5-HT2A, 5-HT2C, 5-HT3, 5-HT7 and stimulation techniques for treatment of medication-
α2-ARs. Furthermore, they exert a broad pattern of refractory depression act via recruitment of broad-
effects across various cerebral loci (Millan, 2006; based neural circuits and multiple cellular and
Gillman, 2007; Sartorius et al., 2007; Cipriani et al., neurochemical substrates. Although fewer mechanistic
2009). Second, antidepressant augmentation strategies data are available, the same seems to hold for sleep
are not simply a case of more of the same, but rather deprivation, which is rapidly effective in the relief of
of something else on top; that is, the incorporation of depressed states (Eitan and Lerer, 2006; Millan, 2006;
additional, mechanistically distinct actions. For tri- Hemmeter et al., 2010; Bunney and Bunney, 2012;
cyclics themselves, efficacy can be reinforced by Del’Osso et al., 2012; Lee et al., 2012; Minichino
adjunctive treatment with lithium or (although seldom et al., 2012).
employed) the thyroid hormones, triiodothyronine and While multi-target strategies are primarily con-
thyroxine, while second generation antipsychotics such ceived for promoting efficacy and accelerating onset
as aripiprazole, quetiapine and olanzapine amplify of efficacy, they may even be better tolerated
the efficacy of SSRIs, reflecting their interaction with (Table 1). In this regard, older tricyclics display inter-
a variety of other cellular substrates (McIntyre and actions at sites other than those mediating positive
Moral, 2006; Crossley and Bauer, 2007; Connolly and actions (see above), but the goal is to build what has
1016 M. J. Millan

been termed ‘selectively non-selective’ agents directed Catena-Dell’Osso et al., 2012; Stein et al., 2012).
against a chosen palette of targets transducing ben- Inasmuch as melatonin is ineffective alone in the relief
eficial properties while eliminating interactions with of depression and selective 5-HT2C antagonists are
sites eliciting side-effects (Hopkins et al., 2006; Millan, unavailable for therapeutic exploration, this profile is
2006; Morphy and Rankovic, 2006, 2007; Zimmermann unique and cannot be mimicked by polypharmacy.
et al., 2007; Wong et al., 2010; Peters et al., 2012). Nonetheless, motivated by studies of enhanced neuro-
Thus, although the risk of additional side-effects genesis in rodents, a mixture of melatonin and buspir-
should not be ignored, judiciously designed multi- one (a 5-HT1A partial agonist) is in development and it
modal strategies may even possess improved toler- recently yielded positive findings in a controlled trial
ance. This is illustrated by antipsychotics possessing (Fava et al., 2012).
α2-AR and/or 5-HT2A antagonist properties to blunt In contrast to agomelatine, as depicted in Table 2,
induction of extrapyramidal actions and by the intro- the majority of multi-modal concepts for improving
duction of 5-HT2C antagonist properties into anti- treatment of depression have been articulated around
depressants to oppose side-effects of excessive 5-HT, inhibition of 5-HT reuptake. That is, in the case of poly-

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


such as sexual dysfunction and nervousness at the pharmacy, adjunctive administration with SSRIs of a
onset of treatment (Dekeyne et al., 2000, 2012; Millan, second agent with a contrasting mechanism of action
2006; Meltzer et al., 2012). and, as regards multi-target agents, the coupling to
Finally, as summarized in Table 1 and exhaustively 5-HT reuptake inhibition of a further and different
discussed elsewhere (Millan, 2006), multi-modal treat- pharmacological action. While the diversity of ideas
ments with complementary mechanisms of action under experimental and/or clinical exploration cannot
should afford additional scope for the control of a be comprehensively discussed herein (Millan, 2006,
greater range of symptoms accompanying depression 2009; Butler and Meegan, 2008; Carvalho et al., 2008;
(Fig. 1) and for the more effective alleviation of de- Shelton et al., 2010; Wong et al., 2010; Schlaepfer
pression co-morbid with other conditions, such as et al., 2012; Al-Harbi, 2012; Connolly and Thase,
anxiety disorders or Parkinson’s disease. 2012), the following observations are of particular
interest within the framework of comparisons between
multi-target and polypharmaceutical strategies.
Complementary mechanisms of antidepressant
Several monoaminergic (‘SSRI-plus’) strategies have
action: polypharmacy and multi-target exploitation
been based around suppressing reuptake of NA and/or
It remains unclear to what extent the combination DA as well as 5-HT, blocking the negative feedback of
of established antidepressants at effective doses monoamines at presynaptic sites inhibitory to release
will guarantee a better patient response and, in certain and/or blocking 5-HT2C receptors which, via recruit-
cases, a lack of further improvement may reflect redun- ment of GABAergic interneurons, restrain monoami-
dancy in the neuronal substrates mediating their nergic transmission (Millan et al., 2000a, b, 2009;
actions (Millan, 2006; Blier et al., 2010; Bobo et al., Millan, 2006; Sartorius et al., 2007). In principle, these
2011; Connolly and Thase, 2011; Rush et al., 2011; goals can be attained by either drug association or by
Al-Harbi, 2012; Rocha et al., 2012). Although there the use of equivalent multi-target drugs. Currently,
are examples where remission is indeed augmented, 5-HT/NA reuptake inhibitors (SNRIs) such as venlafax-
the most appealing feature of multi-modal strategies ine and duloxetine are generally considered somewhat
is the generation of novel antidepressant mechanisms more effective than SSRIs (Montgomery et al., 2007;
that cannot otherwise be reproduced by the use of Cipriani et al., 2009; Papakostas, 2010). One exception
existing agents. may be the ‘SSRI’, escitalopram, possibly reflecting
Melatonin MT1/MT2 receptors have attracted much its distinctive actions at allosteric sites on 5-HT trans-
interest in recent years in view of their role in control- porters as well as its binding to a high affinity site on
ling sleep and circadian rhythms, which are often per- NA transporters (Nguyen et al., 2013; Plenge et al.,
turbed in depression (Bunney and Potkin, 2008). The 2012; Zhong et al., 2012). Not surprisingly, SNRIs are
most compelling example of an innovative multi-target prescribed in preference to the association of a NA
agent is currently agomelatine, a melatonin MT1/MT2 reuptake inhibitor with a SSRI. The DA reuptake inhi-
agonist and 5-HT2C antagonist, which has been clini- bitor, buspirone, appears to be beneficial on top of
cally shown to relieve depressed states upon both SSRIs (Trivedi et al., 2006; Connolly and Thase, 2011)
short and long-term administration, and which also but, rather than dual-acting drugs recognizing 5-HT
possesses anxiolytic properties (De Bodinat et al., and DA transporters, there has been considerable inter-
2010; Kasper et al., 2010; Srinivasan et al., 2011; est in triple monoamine reuptake inhibitors – although
Table 2. Strategies for enhancement of SSRI efficacy based upon augmentation with a complementary mechanism of action, either as pharmacotherapy or as an
equivalent multi-target agent

Primary neuronal Clinically tested Multi-target


Mechanism of action substrates Potential advantages Potential disadvantages agents equivalent References

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


NA reuptake inhibition NA ext Delay CV side-effects Reboxetine VenlafaxineL Millan et al., 2001a, b;
DA ext (FCX) Motor retardation Desipramine DuloxetineL Lopez-Munoz et al., 2007;
(efficacy) DesvenlafaxineL Sartorius et al., 2007
MilnacipranL
DA/NA reuptake NA ext Delay CV tolerance Bupropion Amitifadine II Trivedi et al., 2006;
inhibition DA ext Motor retardation Abuse potential (efficacy) Tesofensine, II(D) Aluisio et al., 2008; Calderone
Sexual dysfunction Psychosis risk JNJ-7925476PC et al., 2010;
(Parkinson’s disease, Stimulation-sensitization JZAD-IV-22PC Tran et al., 2012
obesity)
5-HT1A antagonist 5-HT ext Delay Interference with (-)-Pindolol SB649,915PC Starr et al., 2007; Watson
ACh ext (FCX, hipp) Cognition (variable  WAY-211,612PC

Complementary strategies for improving the treatment of depression


(autoreceptor) post-synaptic and Dawson, 2007;
5-HT1A/1B antagonist Sleep antidepressant properties. efficacy/delay) Beyer et al., 2012
(autoreceptor) (Alzheimer’s disease)
5-HT1A partial agonist NA ext Anxiety Poor tolerance Buspirone VilazodoneL Dawson and Bromidge, 2007;
DA ext (FCX) Cognition Nausea (little  efficacy) OPC14523PC De Paulis, 2007; Frampton,
ACh (FCX, hipp) Sexual dysfunction Nervousness 2011
(GAD)
5-HT2C antagonist NA/DA ext (FCX, hipp). Anxiety Weight gain Ritanserin TrazodoneL Millan, 2005; Cremers et al.,
Control of stress responsive Sexual dysfunction Motor stimulation Mirtazapine Lu-AA24530 2007; Blier et al., 2010
limbic circuits Sleep (efficacy)
α2-AR antagonist NA ext Cognition, CV tolerance Yohimbine S35966PC Cordi et al., 2001; Sanacora et al.,
(autoreceptor) DA/ACh (FCX) Sexual dysfunction Endocrine side-effects (efficacy/delay) R226,161PC 2004; Andres et al., 2007
ACh (FCX, hipp) Parkinson’s disease.
Dopamine Recruitment of mesolimbic Anhedonia Nausea Pramipexole WS-50030 Rogoz and Skuza, 2006;
D2 agonist reward systems Reward Abuse potential OPC34712 Brennan et al., 2010
(Parkinson’s disease) CV side-effects (efficacy?)
Psychosis
Histamine Hist/ACh ext Cognition Wakefulness Not described JNJ-28583847PC Barbier et al., 2007;
H3 antagonist (FCX, hipp) Attention Stocking et al., 2010
NA/DA ext (FCX) (Parkinson’s,
Alzheimer’s disease,
EDSD, narcolepsy)
Acetylcholinesterase ACh ext Cognition Poor tolerance Not described RS-1259PC Abe et al., 2003
inhibitor (degradation) (Alzheimer’s disease)

1017
Table 2 (cont.)

1018 M. J. Millan
Primary neuronal Clinically tested Multi-target

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Mechanism of action substrates Potential advantages Potential disadvantages agents equivalent References

Nicotinic Limbic actions: precise Delay Unclear (S)-Mecamylamine Not described George et al., 2008; Bacher et al.,
(α4/β2 and α3β4>α7) neuronal substrates (TC-5224), IIID 2009; Philip et al., 2012
antagonist unclear (variable efficacy)
AMPA BDNF Cognition Overstimulation in Not described LY392,098PC Li et al., 2003; Farley et al., 2010;
positive allosteric Neurogenesis Attention presence of high LY404,187PC Nations et al., 2012
modulator Neural plasticity Circadian glutamate levels?
synchronization
(ADHD)
NMDA 5-HT/DA ext Neurotoxic damage Psychosis AZD6765 Not described Aan het Rot et al., 2012; Bunney
channel blocker BDNF and Delay Motor disruption (Efficacy study and Bunney, 2012; Ibrahim
mTOR/GSK-3β (Parkinson’s disease) Cognitive deficits underway) et al., 2012; Zarate et al., 2013
AMPA signalling
Neurokinin1 5-HT ext (FCX, hipp) Delay Unclear Vestipitant, IID GSK424,887PC Gobert et al., 2008; Millan et al.,
antagonist NA/DA (FCX) Anxiety Results not disclosed S41744PC 2010; Quartara et al. 2009; Ratti
BDNF Pain and nausea et al., 2011
(GAD, OCD)
CRF1 HPA overdrive and Anxiety Basal corticosterone (R121,919, IID, Several patents Millan, 2009; Kehne and Cain,
antagonist limbic over-activation Stress release GR561,679,IID) 2010
in response to stress Inflammation Interaction studies
(GAD, inflammatory probably not done?
bowel disorder)
Glucocorticoid Neurotoxic and ‘amnesic’ Neuroprotective Baseline HPA activity. Mifepristone, IID? Not described? Shatzberg and Lindley, 2006;
antagonist/synthesis actions of excess stress Psychosis Not desirable for atypical/ Metapyrone, IID? Gallagher et al., 2008
inhibitor corticosterone. Cognition seasonal depression (variable efficacy)
AMPA expression
COX 2 inhibition* NA/5-HT ext. Anti-inflammatory Gastrointestinal side-effects. Celecoxib Not described Müller et al., 2006, 2010; Abbasi
Pro-inflammatory Analgesic. Risk heart attacks and Naproxen et al., 2012; Johansson et al.,
mediators like IL6 Adjunctive treatment stroke (?). 2012
of schizophrenia?
Melanocortin Stress-induced activation Anxiety Metabolic, CV and Not evaluated MCL-0042PC Chaki et al., 2005; Chaki and
4 agonist of HPA, limbic actions HPA overdrive endocrine side-effects Okubo, 2007
Oestrogen receptor NA/5-HT ext. Cognition Endocrine and oncological 17 β-oestradiol Not described Hughes et al., 2008;
β agonist Modulation GABA Neuroprotection side-effects (efficacy) Molinda-Hernandez and
transmission. Tellez-Alcantara 2011
P21-kinase and ERK
recruitment.
Complementary strategies for improving the treatment of depression 1019

none has as yet reached the market and any decisive

are depicted, as well as use for treatment of co-morbid disorders such as generalized anxiety (GAD), attention-deficit hyperactivity (ADHD) and obsessive–compulsive (OCD)
inhibition alone. In certain cases, no comparable multi-target agent has been described. Further, in most cases – although experimentally characterized – clinical data for multi-target

example, essentially all antidepressant mechanisms favour limbic brain-derived neurotrophic factor (BDNF) expression and neurogenesis and induce structural changes in dendritic
depression. Rather, for polypharmacy, it focuses on association with selective serotonin reuptake inhibitors (SSRIs) since this accords with current guidelines, clinical trials and real-
This is not an exhaustive inventory of all drugs to date tested in association with antidepressants, nor a compendium of every multi-target evaluated in experimental models of

world practice. By analogy, the table depicts comparable SRI-based multi-target concepts where experimental data suggest that the mechanism presents benefits vs. 5-HT reuptake

agents are lacking. Certain augmenting agents have multiple mechanisms of action, such as second-generation antipsychotics but also, for example, mirtazapine. Neuronal substrates
indicated are not exhaustive, with a focus on primary drug actions and how they influence extracellular (ext) monoamines in frontal cortex (FCX) and hippocampus (hipp). For
DA, Dopamine; AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid; PC, preclinical; L, launched (approved); D, discontinued (note, actual drug status not always

plasticity, so this is not generally indicated (Duman and Voleti, 2011; Autry and Monteggia, 2012). Potential advantages in terms of control of symptoms other than depressed mood
* Celecoxib also tested in association with the noradrenaline (NA) reuptake inhibitor, reboxetine. Olanzapine has only been tested in association with fluoxetine (Symbax®).
Goodwin et al., 2007; Carvalho
et al., 2008; Connolly and gains in efficacy over SSRIs (or SNRIs) remains to be
demonstrated (Skolnick and Basile, 2006; Aluisio
et al., 2008; Millan, 2009; Tran et al., 2012).
In an effort to mimic the 5-HT1A (dendritic)
and 5-HT1B (terminal) autoreceptor desensitization
Thase, 2011

thought to underlie the progressive onset of anti-


depressant efficacy for SSRIs, both polypharmaceutical
obvious); CV, cardiovascular; EDSD, excessive daytime sleep disorder; HPA, hypothalamo-pituitary axis; COX 2, cyclooxygenase-2; PAG, partial agonist.

and multi-drug pistes have been pursued. However,


despite a plethora of preclinical findings and promis-

For further information see Carvalho et al., 2006; Millan, 2006, 2009; Shelton et al., 2010; Connolly and Thase, 2011, 2012; Schlaepfer et al., 2012).
ing clinical evidence for an accelerated onset of action,
Not described

gains in efficacy have proven inconsistent upon co-


administration of the 5-HT1A antagonist, (−)-pindolol,
with SSRIs (Artigas et al., 2006; Dawson and

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Bromidge, 2008; Connolly and Thase, 2012). This
may reflect its residual intrinsic efficacy at autorecep-
tors and, by analogy, it has proven difficult to link
pure antagonist properties at 5-HT1A and/or autorecep-
AripiprazoleL
OlanzapineL

tors to suppression of 5-HT reuptake, although several


QuetiapineL

(efficacy)

mixed ligands have now been documented (Millan,


2006, 2009; Starr et al., 2007; Watson and Dawson,
2007; Beyer et al., 2009). Rather paradoxically, then,
notwithstanding: (1) studies in rats indicating that
5-HT1A partial agonists blunt SSRI-induced increases
(akathesia) side-effects
CV, metabolic, motor

in 5-HT levels (Gobert et al., 1999); (2) less than scintil-


lating findings with adjunctive use of the 5-HT1A par-
tial agonist, buspirone, in depression (Millan, 2006;
Weight gain

Connolly and Thase, 2011), it is the mixed 5-HT1A par-


tial agonist/SSRI, vilazodone, that has been granted
approval for treatment of depression (De Paulis,
disorders. Some mechanisms are especially promising for accelerating onset.

2007; Dawson and Bromidge, 2008; Frampton, 2011;


Reed et al., 2012). Rather than 5-HT, alterations in
levels of other transmitters such as acetylcholine, NA
(schizophrenia,

and DA may account for any potential advantages of


OCD, GAD)

vilazodone over SSRIs (Millan et al., 1994; Millan,


Anxiety

2006; Watson and Dawson, 2007).


While partial agonism at 5-HT1A receptors favours
NA and DA release in the frontal cortex (Millan et al.,
possibly glutamate (FCX)

2000a, b), their elevation can be more robustly and


NA/DA/Ach ext and

reproducibly achieved by blockade of 5-HT2C receptors.


The induction of NA and DA release marries well with
Lu-AA24530 is also a 5-HT3 antagonist.

inhibition of 5-HT reuptake and 5-HT2C antagonism


affords additional anxiolytic and sleep-enhancing
GSK-3β

properties while opposing the sexual dysfunction pro-


voked by excessive levels of 5-HT (Dekeyne et al., 2000,
2008; Millan, 2005; Quesseveur et al., 2012). 5-HT2C
antagonism is inherent to many tricyclic agents as
(5-HT2A/2C antagonist,

well as to trazodone (of which a new long-release


5-HT1A PAG, etc)
Second generation

form is becoming available), but these drugs all have


antipsychotics

other undesirable actions like histamine H1 receptor


and α1-AR blockade (Millan, 2006; Gillman, 2007;
Stahl, 2009). Hence, dual 5-HT2C antagonists/SSRIs
shorn of detrimental actions still appear attractive;
1020 M. J. Millan

in particular, since selective 5-HT2C antagonists for inconsistent clinical efficacy remain to be elucidated,
association with SSRIs are not clinically available and but it is worth noting evidence that both blockade
since mirtazapine, which potentiates the actions of and/or stimulation of α4β2 nicotinic sites in rodents
SSRIs, shares the potent H1 and α1-AR antagonist positively influences depressed mood in rodents,
actions of tricyclics (Blier et al., 2010; Connolly and while the molecular substrates for putative anti-
Thase, 2011; Watanabe et al., 2011). More favourably, depressant actions of (S)-mecamylamine warrant fur-
mirtazapine antagonizes α2-ARs inhibitory to mono- ther investigation (Millan, 2006; George et al., 2008;
aminergic transmission and, mirroring preclinical Bacher et al., 2009; Andreasen et al., 2011; Philip
work, clinical data suggest that α2-AR blockade has- et al., 2012). Regrettably, although there is considerable
tens and intensifies antidepressant properties elicited cross-talk amongst nicotinic ligands (certain of which
by monoamine reuptake inhibition (Sanacora et al., interact with 5-HT transporters) and SSRIs (several of
2004). Correspondingly, mixed α2-AR/SSRIs and which recognize nicotinic receptor subtypes, Millan,
α2-AR/SNRIs, in which negative feedback at α2-ARs 2006), these structure-activity insights do not appear
autoreceptors is interrupted, display powerful anti- to been systematically exploited for generation of

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


depressant profiles in rodents (Cordi et al., 2001; multi-target nicotinic agents of potentially more robust
Andres et al., 2007; Millan, 2009; Serres et al., 2012). antidepressant efficacy than either class of agent
It is frustrating that these agents have not been author- alone – or, indeed, their combination.
ized for the control of treatment-resistant depression, Another mechanism worth highlighting is
primarily due to concerns of untoward cardiovascular N-methyl-D-aspartate (NMDA) receptor blockade in-
side-effects. Inasmuch as the above lines evoked the asmuch as a suite of recent studies has shown that
interest of associating 5-HT2C and α-AR antagonism the NMDA receptor channel blocker, ketamine, at sub-
with SSRI properties, it is of note that dual 5-HT2C/ anaesthetic doses, elicits robust and rapid antidepress-
α2-AR antagonists lacking the potential side-effects of ant properties (Aan Het Rot et al., 2012; Bunney and
mirtazapine were recently described, although their Bunney, 2012). NMDA receptor antagonists are
clinical effects await characterization (Dekeyne et al., known to recruit frontocortical and subcortical mono-
2012; Millan et al., 2012). aminergic pathways in rodents. In addition, AMPA
All the above-evoked monoaminergic strategies receptors and the modulator of mRNA translation
would be expected to reinforce dopaminergic trans- and neuroplasticity, mammalian target of rapamycin
mission in the frontal cortex and hence to counter (mTOR) – which is deficient in depression (Jernigan
depressed mood and impairment of cognitive domains et al., 2011) – are implicated in the actions of ketamine
such as executive function and working memory, but (Maeng et al., 2008; Li et al., 2010; Aan Het Rot et al.,
only triple reuptake inhibitors should elevate extra- 2012). To date, the combined administration of keta-
cellular levels of DA in limbic structures (Millan et al., mine with SSRIs has not been documented, but a clini-
2000a, b, 2012; Skolnick and Basile, 2006; Aluisio et al., cal evaluation of this question is currently underway
2008; Millan, 2009; Tran et al., 2012). Mesolimbic with a further, low affinity non-competitive antagonist
actions counter both motor retardation and defective at NMDA receptors, AZD6765 (Connolly and Thase,
mechanisms of reward, of particular relevance to mel- 2012). The integration of 5-HT transporter and NMDA
ancholic depression and depression co-morbid with channel blocking activity into a single drug seems unli-
Parkinson’s disease. An alternative approach would kely but an alternative approach may be to combine
be to directly recruit post-synaptic D2 receptors by SSRI activity with selective blockade of the NR2B
use of drugs possessing agonist properties and both receptor subunit in a single drug since: (1) NMDA re-
polypharmaceutical and multi-target approaches ceptors containing this subunit seem to be the major
have been described in this regard but, perhaps in class involved in the control of mood and depressed
view of numerous issues of tolerance, little real pro- states; (2) selective legends at NR2B subunits are
gress has been made and this line of research does under evaluation for potential antidepressant proper-
not appear particularly inspiring (Rogoz and Skuza, ties in animals and man (Aan Het Rot et al., 2012;
2006; Brennan et al., 2010; Al-Harbi, 2012). Ibrahim et al., 2012). The major concerns for the
Positive phase II efficacy studies associating the broader use of this concept, whether as drug combi-
nicotinic antagonist, (S)-mecamylamine, with SSRIs nations or multi-target agents, is the induction of psy-
led to much excitement, but more recent phase III chosis and blunting of efficacy upon prolonged
investigations were something of an anti-climax, lead- administration (Aan Het Rot et al., 2012; Mathew,
ing to termination of its development in depression. 2012). In any event, the actions of ketamine may pro-
The precise reasons underlying inadequate and/or vide insights into molecular substrates permitting
Complementary strategies for improving the treatment of depression 1021

more rapid control of depression by other, more tract- selective NK1 antagonists and SSRIs, although data
able strategies. Interestingly, mGluR2 antagonists, from humans are awaited (Brocco et al., 2008; Gobert
which likewise possess potential antidepressant prop- et al., 2009; Millan et al., 2010). Further, mirroring
erties, mimic ketamine in recruiting mTOR in the fron- findings with concomitant administration of selective
tal cortex (Dwyer et al., 2012). Note, however, the NK1 antagonists and SSRIs in rodents, vestipitant
caveat that: (1) NMDA receptor blockade is has been under development as polypharmacy with
pro-psychotic; (2) mGluR2 receptors are deficient in SSRIs for the treatment of major depression (Millan,
schizophrenia and antagonists possess potential pro- 2009). To date, clinical data have not been disclosed,
psychotic properties (Nicoletti et al., 2011); while (3) the status of this project is uncertain and it may have
over-expression of mTOR in the frontal cortex accounts been abandoned for strategic reasons related to factors
for certain cognitive deficits of both schizophrenia and other than clinical efficacy. The theoretical and exper-
neurodevelopmental disorders like Fragile X (Meffre imental arguments supporting co-joint blockade of
et al., 2012; Millan, 2013). NK1 receptors and 5-HT reuptake for treatment of
In view of the impressive pedigree of data sup- depression are compelling. Both classes of drug are

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


porting their roles in the response to stress, the control safe and well studied, so this still remains an ideal plat-
of mood and the induction of depressed states, two form for a structured comparison of multi-target drugs
of the most conspicuous failures to achieve authoriz- vs. polypharmacy for the ameliorated treatment of
ation (so far) are CRF1 and NK1 receptor antagonists depression.
(Quartara et al., 2009; Kehne and Cain, 2010; Ratti Contrasting with NK1 antagonists, there is abundant
et al., 2011). As pointed out above, the core problem support for the use of second-generation antipsycho-
may be that these mechanisms are not sufficient for tics as adjuncts to SSRIs in the control of treatment-
robust efficacy alone and require integration with resistant depression and data are particularly solid
other actions. While compounds inhibiting 5-HT trans- for aripiprazole and olanzapine (both authorized),
port and blocking CRF1 receptors are feasible and although the latter was evaluated only with fluoxetine
active in vivo (M.J. Millan, unpublished observations), (Connolly and Thase, 2011). The precise mechanisms
appropriate pharmacokinetic parameters remain elu- involved in the potentiation of SSRI efficacy remain
sive. Rather oddly, the effects of co-administration of to be defined, but a role for partial agonist properties
CRF1 antagonists and SSRIs in humans do not appear at 5-HT1A receptors, as well as blockade of 5-HT2A/2C
to have been investigated despite the inherent logic receptors and α2-ARs seems probable. These mechan-
underpinning such studies, especially in psychotic de- isms were evoked above and blockade of 5-HT7 sites
pression and mixed depression/anxiety. Today, CRF1 may likewise be implicated inasmuch as their inacti-
antagonists are mainly being pursued for anxiety dis- vation is associated with antidepressant properties in
orders and irritable bowel syndrome (Millan, 2009; rodents – although clinical data for 5-HT7 antagonists
Kehne and Cain, 2010). As for NK1 receptors, a high- alone and together with SSRIs are not yet available
profile report of antidepressant actions of the selective (Mnie-Filali et al., 2011). Partly in view of the complex
NK1 antagonist, aprepitant, had considerable re- pharmacological profiles of second-generation antipsy-
sonance but these actions could not be consistently chotics, but also in light of the robust results attained
confirmed by succeeding studies, even at doses upon their association with SSRIs, there have been no
shown to saturate NK1 receptors – possibly due to an real attempts to reproduce this profile with a multi-
intervening amplification of the response to placebo target strategy. Note that the major metabolite of que-
in the course of clinical trials, rather than any ostensi- tiapine possesses affinity for NA transporters and
ble loss of efficacy (Millan, 2009; Quartara et al., ziprasidone for 5-HT transporters, suggesting that
2009; Ratti et al., 2011). Although this remains to be such multi-target compounds would be feasible from
clarified, other selective NK1 antagonists have hardly a chemical perspective (McIntyre et al., 2007; Millan,
fared better so they are now employed mainly for con- 2009).
trol of chemotherapy-evoked nausea and vomiting Depressive states in humans and experimental
arising in the treatment of cancer – as part of a multi- models of depression in rodents are associated
drug treatment package designed to enhance efficacy with inflammation and elevations in levels of inter-
(Ratti et al., 2011; Dos Santos et al., 2012). Mixed leukins. These observations have prompted studies
NK1 antagonists/SSRIs have been documented in pre- of the potential adjunctive use of inhibitors of
clinical studies, where they manifest the positive attri- cyclooxygenase-2 like Celecoxib, both with SSRIs and
butes of each of these class of compounds, and their with the NA reuptake inhibitor, reboxetine (Müller
profiles resemble those of combined injection of et al., 2006; Guo et al., 2009, 2009; Muller, 2010;
1022 M. J. Millan

Capuron and Miller, 2011; Abbasi et al., 2012; Leonard Thase, 2011, 2012). Another prominent example is
and Maes, 2012). Despite promising data, and preclini- modafinil (and its active isomer, armodafanil) which
cal studies suggesting that Celecoxib acts both by anti- exerts a complex pattern of actions via monoaminergic,
inflammatory mechanisms and via an influence on glutamatergic and orexinergic mechanisms. Modafinil
monoaminergic transmission (Muller, 2010; revealed modest and possibly non-sustained im-
Johansson et al., 2012; Krause et al., 2012; Leonard provements in mood upon addition to SSRIs and, not
and Maes, 2012), clinical observations remain limited. surprisingly in view of its wake-promoting character-
Further, not all data support benefits of associating istics, reduced fatigue (Minzenberg and Carter, 2008;
anti-inflammatory agents with SSRIs (Warner- Shelton et al., 2010; Connolly and Thase, 2012;
Schmidt et al., 2011). Accordingly, there is a need for Millan, 2013).
further evaluation of this intriguing concept in Finally, the most striking feature of Table 2 is just
patients, yet studies have – not unexpectedly – been how few drug combinations and/or comparable multi-
restrained by questions concerning the cardiovascular target agents have been formally authorized and
safety of this class of drugs. become available to patients despite an abundance of

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


To date, no mixed SSRI/cyclooxygenase 2 inhibitors auspicious preclinical data, compelling hypotheses
are known and, despite the above indications that and at least small-scale clinical testing. Clearly, there
many other classes of multi-target drug incorporating is considerable scope for further work to clinically vali-
5-HT reuptake can be designed, a number of augmen- date these and other (see below) multi-modal concepts
tation mechanisms would be challenging to incor- for the improved treatment of depression.
porate into a single chemical structure. For example,
as regards the actions of thyroid hormones (tri-
Polypharmacy and multi-target agents: similarities
iodothyronine and thyroxine), inhibitors of corticoster-
and differences, advantages and disadvantages
one synthesis, glucocorticoid 2 receptor antagonists
and agonists at oestrogen β receptors (Millan, 2006; As outlined above, theoretical considerations, exper-
Carvalho et al., 2008; Dhir and Kulkarni, 2008; imental studies and, in several instances, clinical feed-
Gallagher et al., 2008; Hughes et al., 2008; Schatzberg back underscores the interest of multi-modal treatment
and Lindley, 2008; Molina-Hernandez and Tellez- strategies for the improved treatment of depression,
Alcantara, 2011; Connolly and Thase, 2012). Note, although much remains to be learned. While polyphar-
however, that thyroid hormones are little used today maceutical strategies and multi-target agents share
and that data for oestrogen agonists (mostly tested certain features, they also reveal differences and both
in peri-menopausal women) remain preliminary. have their merits and their disadvantages, as summar-
Moreover, despite considerable efforts and a strong ized in Table 3.
rationale, clinical studies with mefepristone (a gluco- The principal challenge with multi-target agents is
corticoid antagonist) and metyrapone (an inhibitor of the integration of two or more (preferably overlapping)
glucocorticoid synthesis) – which should protect cells pharmacophores into a small structure (molecular
from detrimental effects of excessive stress-induced weight not more than 500 if possible) in order to recon-
corticosterone release and favour cognitive perform- cile chosen promiscuity with favourable pharmaco-
ance – have not been brought to fruition, even in kinetic properties of CNS penetration and biostability
patients showing psychotic depression, where the (Hopkins et al., 2006; Millan, 2006; Morphy and
hypothalamic-pituitary axis activity is disinhibited Rankovic, 2006, 2007; Zimmermann et al., 2007;
(Millan, 2006; Gallagher et al., 2008; Schatzberg and Wong et al., 2010). Ironically, more than a decade of
Lindley, 2008). focus on highly selective ligands has enhanced the abil-
Other examples (not depicted in Table 2) of absent ity of chemists to eliminate (unwanted) activities rather
multi-target lookalikes for a clinically tested SSRI than favour their integration when desired. Nonethe-
augmentation strategy include instances where the less, while structure–activity relationships for multi-
drug added on: (1) has a poorly defined mechanism; target ligands can be challenging, it is not usually
(2) acts itself in a multi-target fashion; (3) the associ- necessary to generate compounds with high affinities
ation has yielded ambivalent experimental and clinical for two sites. As a rule, more modest affinities than
data providing only a modest incentive to launch a dis- for selective agents suffice and the real issue is acquir-
covery programme for an equivalent multi-target ing an appropriate balance as defined less in vitro than
agent. Anticonvulsants such as riluzole and lamotri- by use of in vivo experimental models of depression
gine illustrate this point, as does the ‘stimulant’ (see above citations). This can be arduous and it is
methylphenidate (Carvalho et al., 2008; Connolly and hard to be certain that the ratio of activities is optimal,
Complementary strategies for improving the treatment of depression 1023

Table 3. General features, advantages and drawbacks, of polypharmacy as compared to multi-target agents

Factor Polypharmacy (drug combinations) Multi-target agents

General acceptance Non-evidence-based polypharmacy is discouraged No inherent difference in attitude to


for CNS disorders and especially problematic in multi-target vs. selective agents for patients or
the elderly. However, when mechanisms are prescribers, although the potential advantages
supported by clinical data, administration is of multi-target over highly selective agents are
carefully monitored and drugs are prescribed in still insufficiently recognized.
accordance with guidelines to poorly responsive
patients, it is an important option.
Generation and design If individual drugs exist, they can be combined, In principle, any hypothetical association
of drugs although there may be issues of intellectual of two or more actions can be built
property and availability. If drugs with desired into one chemical structure. However,
pharmacological activities are inexistent, then structure–activity relationships are more

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


combination is impossible. A mixture of more challenging and onerous than for selective
than three compounds is rarely a realistic agents and some may be chemically
proposition. unrealizable.
Hypothesis validation, Characterization of drug combinations requires Once the hypothesis is validated, multi-target
experimental evaluation testing of components alone and in association, drugs can be characterized similarly to
time-consuming for determination of optimal selective agents. However, the ratio of
ratios, which may be test-symptom dependent. respective activities is invariant and cannot be
However, drug ratios can still be titrated in the adjusted in the clinic.
clinic.
Therapeutic range and Additional activities should expand therapeutic Same potential advantages and
properties range, although they may also introduce limitations as for polypharmacy.
constraints, e.g. dopamine D2 agonist properties
are fine for Parkinson’s disease but unacceptable
for psychotic depression.
Tolerance, therapeutic Additional activities may bring other side-effects, Same potential advantages and
window yet tolerance can be improved. Thus, a second limitations as for polypharmacy.
mechanism of additive or synergistic
antidepressant activity allows for reduced doses
of each at equivalent efficacy, hence enhancing
the therapeutic window. Certain actions may
directly counter side-effects, e.g. 5-HT2C
antagonism to counter sexual dysfunction and
nervousness.
Safety/toxicity testing Toxicological and safety pharmacology profiles Spared the drawback of polypharmacy, no
need to be available for both drugs separately as disadvantage vs. selective agents.
well as together.
Drug pharmacokinetics Pharmacokinetic profiles (time to max. effect, Spared the drawback of polypharmacy, no
half-life, etc.) of component drugs need to be disadvantage vs. selective agents.
comparable for co-administration to make sense.
Drug interactions Drug interactions among component ligands need Spared the drawback of polypharmacy, no
careful monitoring, such as those related to disadvantage vs. selective agents.
cytochrome 450 isoform inhibition. Interactions
with other drug classes especially complicated.
Clinical testing Designs complex, both vs. placebo and vs. Spared the drawback of polypharmacy, no
comparative agents. Large scale, long-term trials disadvantage vs. selective agents.
not easy, especially if other than add-on to SSRIs.
Industry primarily interested in testing novel
monotherapy*.
Compliance Major issue for antidepressants: may be even more Spared the drawback of polypharmacy, no
problematic for multiple pills although improved disadvantage vs. selective agents.
efficacy should favour adherence.
1024 M. J. Millan

Table 3 (cont.)

Factor Polypharmacy (drug combinations) Multi-target agents

Intellectual property, Association (and use) patents tend to be weak, Robust protection. Multiple components of
patent issues, financial especially if one or other of the drugs is generic pharmacological activity offer additional
considerations and/or the property of another firm. Use of opportunities for patent strategies compared to
additional drugs will increase health-care costs. selective agents. Costs of mechanistically novel
drugs superior to existing agents may be high
at launch.

CNS, Central nervous system; SSRIs, selective serotonin reuptake inhibitors.


*Fluoxetine plus olanzapine is an interesting exception, since both drugs belong to Lilly.
The pros and cons of polypharmacy and multi-target drugs are indicated for a broad range of characteristics from discovery and
experimental characterization to clinical development and therapeutic exploitation. Comparisons are made among these two

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


families of agents as well as to monotherapy with highly selective agents. Multi-target drugs share most of the benefits of poly-
pharmacy yet lack many of their disadvantages and they also display several distinctive benefits of their own. However, they
reveal a few drawbacks, notably in their design and discovery.

a core consideration since it is obviously invariant and drawbacks. On balance, although it would be unwise
cannot later be modified in the clinic. to neglect polypharmacological opportunities, assum-
Conversely, drug associations can be titrated in ing that the requisite drug can be made, multi-target
patients, with one component (like a SSRI) remaining strategies appear to be the most promising strategy
constant and the other being systematically varied, for future progress.
although, for pragmatic reasons and in view of time
and cost, a fairly well-defined notion of the desired
drug:dose ratio should be available before com- Perspectives for future development of multi-target
mencing clinical studies. Moreover, as outlined in drugs and polypharmacy
Table 3, in particular where the two drugs to be com-
A general strategy for exploiting multi-target
bined are not well characterized in humans, there are
concepts and polypharmacy
many developmental, pharmacokinetic and clinical
issues that are more complex to address for drug com- As articulated in detail elsewhere (Millan, 2006, 2009),
binations than for multi-target (or selective) agents. one pragmatic way of reconciling the competing de-
To date, most polypharmaceutical concepts have mands and merits of polypharmacy, multi-target
been limited to small-scale trials, where at least one drugs and selective agents would be to simultaneously
drug is commercially available, and the concomitant pursue all three of these complementary strategies. For
development from scratch of two novel ligands in all new targets, it is imperative to identify highly selec-
association has not, to the author’s knowledge, been tive ligands as exploratory pharmacological tools and
accomplished. Indeed, while polypharmacy for de- they can be evaluated both alone and in association
pression is incarnated by the approval of adjunctive with: (1) SSRIs; (2) other classes of clinically available
use of second-generation antipsychotics (such as aripi- antidepressant; (3) promising and novel modes of anti-
prazole) with SSRIs in treatment-resistant subjects, depressant activity. This would provide a framework
both of these drug classes are well known, of proven both for the development of drugs alone and in associ-
clinical utility and inherently ‘safe’. Further, olanza- ation. In parallel, and guided by such data, multi-target
pine has been specifically developed in association modes of target exploitation can likewise be generated
with fluoxetine, both drugs emanating from the same based on the integration of one or more complementary
company (Lilly). The development of drug mixtures pharmacological action into a single drug, either estab-
containing two or more new chemical entities from lished or new. Thus, where feasible and desirable,
the outset is a far more formidable challenge. multi-target drugs should systematically be designed
Polypharmacy and multi-target agents both have and progressed in parallel with selective ligands
their merits as compared to selective agents (some (required for polypharmacy) at innovative targets for
shared, some distinctive) and both present certain the potentially improved treatment of depression.
Complementary strategies for improving the treatment of depression 1025

Novel targets for multi-target and possesses anxiolytic properties and appears to promote
polypharmaceutical exploitation: sleep (De Bodinat et al., 2010; Kasper et al., 2010;
new vistas for monoaminergic mechanisms Racagni et al., 2011; Catena-Dell’Osso et al., 2012).
Clearly, there remains an urgent need to pursue
Underscoring the notion that novel monoaminergic
other multi-target strategies oriented around a non-
mechanisms may still have something to offer in the
monoaminergic target for further gains in effectiveness
multi-target treatment of depression, 5-HT4 agonists
and control of other symptom clusters. In this regard, it
have attracted interest as rapid onset antidepressants
is worth mentioning some novel avenues of research
(Lucas, 2009) and, as mentioned above, efforts are
(highlighted in Fig. 5), which could profitably be devel-
being made to develop 5-HT7 antagonists (Mnie-Falili
oped along lines evoked above.
et al., 2011). Although for reasons accentuated herein,
First, in light of the above-mentioned use of triio-
it is dubious whether such mechanisms will suffice
dothyronine for augmentation of tricyclics and, less
alone for adequate efficacy, both 5-HT4 agonists and
consistently, SSRIs (Cooper-Kazaz and Lerer, 2008;
5-HT7 receptor blockade may potentiate at least some
Connolly and Thase, 2012), it is intriguing that the

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


actions of SSRIs (Lucas, 2009; Shelton et al., 2009)
endogenous (decarboxylated) derivative, 3-iodothyro-
and several multi-target monoaminergic antidepress-
namine behaves like tyramine, phenylethylamine and
ants are currently under investigation. Notably, clinical
other trace amines as an agonist at trace amine associ-
data support the effectiveness of Lu-AA21004, which
ated receptor 1 receptors (Sotnikova et al., 2009;
inhibits 5-HT reuptake, acts as a partial agonist at
Di Cara et al., 2012). Their activation modulates mono-
5-HT1A and 5-HT1B sites and as an antagonist at
amine transport and exerts a variety of other cellular
5-HT3 and 5-HT7 receptors (Alvarez et al., 2012;
effects associated with potential antidepressant proper-
Katona et al., 2012; Mork et al., 2012). However, it is
ties and ligands at these sites may also be useful for
not yet clear which (if any) advantages it offers over
controlling psychosis associated with depression (Xie
currently available antidepressants.
and Miller, 2009; Revel et al., 2012a, b). Trace amine
Another innovative idea would be to couple DA
associated receptor 1 receptors are a novel and promis-
D3 antagonist properties to SSRIs and other antide-
ing site for new classes of multi-modal antidepressant.
pressant mechanisms in view of compelling evidence
Second, as depicted in Fig. 2, there is increasing
for improvements in a broad spectrum of depression-
interest in epigenetic mechanisms in the pathogenesis
compromised cognitive domains, such as social
of depressed states and the broad range of epigenetic
cognition, working memory, attention and executive
processes controlling gene expression independent of
function. Moreover D3 receptor blockade elevates
DNA sequence (from DNA methylation and histone
neurogenesis in the hippocampus (which is defective
marking to non-coding RNA control of mRNA splicing
in depression), counters stress-induced drug-seeking
and translation) provides a broad palette of potential
behaviour (for example, of cocaine) and promotes
targets for new classes of multi-modal antidepressant
motor function in models of Parkinson’s disease
(Millan, 2011; Uher, 2011; Mouillet-Richard et al.,
(Millan et al., 2007; Millan and Brocco, 2008;
2012; Sun et al., 2012). Of particular interest are histone
Heidbreder and Newman, 2010; Loiseau et al., 2009;
deacetylase 2 inhibitors, which possess antidepressant
Egeland et al., 2012; Watson et al., 2012). Selective D3
properties in rodents, as well as complementary
antagonists have not yet reached patients, yet multi-
pro-cognitive actions (Graff and Mansuy, 2009; Cov-
target antidepressants possessing D3 receptor antagon-
ington et al., 2010; Day and Sweatt, 2012; Lin et al.,
ist properties could be of particular use for treatment,
2012; Yamawaki et al., 2012). Interestingly, there is evi-
not only of major depression but also of depressed
dence that folate (folic acid) is lacking in depression
mood co-morbid with substance abuse, schizophrenia
and its deficiency may not only compromise active
and Parkinson’s disease (see above citations).
DNA formation but also methylation of both histones
and DNA (Gilbody et al., 2007). Antidepressant-like
Novel targets for multi-target and
actions of folate have been reported in rodents and
polypharmaceutical exploitation:
studies are being pursued of its administration alone
non-monoaminergic concepts
and together with SSRIs to patients (Taylor et al.,
Agomelatine still holds its position as a uniquely 2004; Stahl, 2007; Molina-Hernandez et al., 2011,
innovative and clinically proven, multi-target anti- 2012). On this basis, and in view of the significance
depressant reflecting its complementary (perhaps of DNA and histone methylation to the mood and cog-
synergistic) agonist and antagonist properties at mela- nitive deficits of neurodevelopmental disorders,
tonin and 5-HT2C receptors, respectively. It also histone methyltransferases and demethylases, as well
1026 M. J. Millan

> 30 Many potential


monoaminergic non-monoaminergic
targets 5-HT targets

1 Neurotransmitters
2 Neuropeptides
3 Hormones
4 Cellular modulators
DA NA

Modulator Activity Functional properties

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


1 Trace amines TAAR1 Ago ↓ Psychosis
Core, clinically 1 Acetylcholine α7Ago/PAM ↑ Cognition +
validated 1 Glutamate mGluR5 Ant, mGluR7 Ago ↓ Anxiety/Psychosis Complementary
monoaminergic 2 Neuropeptide Y mechanism of
Y1 Ago, Y2 or Y5Ant ↓ Anxiety/Obesity
mechanism activity
3 Ghrelin Ago ↑ Cognition
4 Histone deacetylase Inhibitor ↑ Cognition
Multi-target concepts/Polypharmacy combinations

Fig. 5. Novel mechanisms under exploration for multi-target and/or polypharmaceutical exploitation in the improved
treatment of depression. Including receptors, transporters and enzymes for degradation, there are about 30 ways of
manipulating monoaminergic transmission, of which several – but not all – are already clinically exploited. In addition, there
are innumerable, potential non-monoaminergic mechanisms for improving various dimensions of depressed states, although
none has yet become available to patients as selective drugs. It should be possible to harness clinically validated mechanisms
of antidepressant action [such as 5-HT and/or noradrenaline (NA) reuptake suppression or 5-HT2C receptor blockade] and
add on complementary mechanisms (either mono- or non-monoaminergic) to strengthen efficacy, improve tolerance, reduce
delay to action and control a broader range of symptoms. An example of a multi-target agent would be agomelatine
[melatonin agonist (Ago) plus 5-HT2C antagonist (Ant)] and an example of polypharmacy would be fluoxetine plus
olanzapine for treatment-resistant depression. Many more permutations remain to be experimentally and clinically explored
(see text). Dependent upon drug availability and other factors, both polypharmacy (drug combinations) and multi-target
agents are appropriate vehicles for uniting the mechanisms of action indicated. DA, Dopamine; TAAR, trace amine associated
receptor; PAM, positive allosteric modulator; mGluR, metabotropic glutamatergic.

as enzymes controlling DNA methylation, justify Fourth, neuropeptide Y has long been implicated
evaluation as potential targets for antidepressants in the response to stress and in the control of mood.
(Kramer and van Bokhoven, 2009; Gomes and Joca, Activation of post-synaptic Y1 sites, or blockade of
2011; Sanchez-Mut et al., 2012; Millan, 2013). their Y2 presynaptic counterparts, has been associated
Third, in addition to ionotropic AMPA and NMDA with antidepressant and anxiolytic properties in
receptors (vide supra), recent evidence supports a role rodents (Ishida et al., 2007; Tasan et al., 2010; Gelfo
for various classes of mGluR receptor in the control et al., 2012). Interestingly, subthreshold doses of
of mood and, specifically, in depressive states. Thus, neuropeptide Y and fluoxetine synergistically elicited
blockade of mGluR2 (Bespalov et al., 2008; Campo antidepressant effects (Molina-Hernandez and Tellez-
et al., 2011; Dwyer et al., 2012) and mGluR5 (Hughes Alcantara, 2011). Finally, suggesting a further dimen-
et al., 2012; Inta et al., 2012) receptor subtypes on the sion to the role of neuropeptide Y, a non-peptidergic
one hand and activation of the mGluR7 subtype antagonist at Y5 receptors was recently found to elicit
on the other (Palucha et al., 2007; Bradley et al., 2012) antidepressant effects (Packiarajan et al., 2011).
is associated with antidepressant (and anxiolytic) Fifth, another intriguing peptide for which data are
properties. Apart from multi-target agents, adjunctive less extensive is the appetite-stimulating hormone
use of mGluR ligands is an attractive possibility ghrelin, which is secreted by the gut yet enters the
(Matrisciano et al., 2008). brain where it facilitates cognition and counters the
Complementary strategies for improving the treatment of depression 1027

deleterious impact of stress on mood (Lutter et al., While mechanistic details of how the above drug
2008; Atcha et al., 2009; Chuang and Zigman, 2010). classes affect mood and other functions disrupted in
Although its precise role in the control of anxious depression are beyond the scope of this review (see
and depressed states remains somewhat unclear, ghre- above citations), they suggest scope for the exploration
lin may possess antidepressant properties and further of novel multi-modal strategies for the improved treat-
studies of therapeutic relevance to the control of ment of depressed states.
depression is warranted (Chuang and Zigman, 2010;
Carlini et al., 2012; Ishitobi et al., 2012). Integration of multi-modal pharmacotherapy with
Sixth, as mentioned above, there is considerable ‘alternative’ treatments
interest in the influence of nicotinic receptors upon
depressed mood, with most attention to date directed Not surprisingly, in view of persistent difficulties in
towards antagonists at α4β2 receptors – despite the identifying improved pharmacotherapy for depression,
recent disappointment of (S)-mecamylamine as adjunc- there is ever-increasing interest in ‘alternative’ (and,
tive therapy for SSRIs (Ledford, 2011; Philip et al., from a network perspective, ‘multi-target’) strategies

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


2012). α7 receptors are likewise of interest in view of as diverse as, for example, mindfulness (Marchand,
their positive influence upon cognition (Graef et al., 2012), cognitive behavioural therapy (DeRubeis et al.,
2011; Levin, 2012; Millan et al., 2012). Although 2008), light exposure (Crowley and Youngstedt, 2012)
affinities of α7 ligands at 5-HT transporters, and reci- and transcranial direct stimulation (Del’Osso et al.,
procally of SSRIs at nicotinic receptors, complicates 2012). Ultimately, the goal would arguably be to abol-
studies of interactions, there is evidence that α7 recep- ish artificial barriers and the knowledge gap between
tor blockade potentiates the actions of SSRIs in rodent medication and validated, non-pharmacotherapeutic
models of antidepressant properties (Millan, 2006; strategies and to unite them in a coherent and
Andreasen et al., 2012). Although rapid antidepressant evidence-based manner to the profit of individual
actions of the muscarinic antagonist, scopolamine, (subsets of) patients. Thus, a final issue concerns the
have been reported, it would appear more appropriate potential use of multi-target antidepressants and poly-
to identify and exploit the underlying cellular sub- pharmacy in association with alternative treatment
strates – perhaps analogous to those harnessed by strategies, where medication is not sufficient alone
ketamine or sleep deprivation (Bunney and Bunney, for satisfactory remission. Cognitive behavioural
2012) – rather than pursue multi-target drugs block- therapy acts at the ‘cerebral circuit’ level rather than
ing muscarinic receptors in view of gender differences homing in on any discrete molecular substrate and it
and their notorious side-effect profile, although recruits contrasting cerebral mechanisms vs. pharma-
allosteric sites may represent a hitherto unexplored cotherapy (Kennedy et al., 2007). This supports the
opportunity for therapeutics (Drevets and Furey, logic of its combination with pharmacotherapy for
2010). improving efficacy, reducing relapse and enhancing
Finally, although not illustrated in Fig. 5, a few quality of life (Guidi et al., 2011; Ishak et al., 2011;
words should be devoted to glial mechanisms, long Köhler et al., 2011). While such dual treatment
implicated in the pathogenesis of depression (and strategies would be difficult to integrate into early
other CNS disorders) but still not harnessed thera- development programmes, small-scale pilot trials and
peutically (Czeh and Di Benedetto, 2012). An interest- post-authorization studies should prove useful in
ing link has been made to a further potential substrate more fully exploring the clinical utility of novel classes
for multi-modal treatment of depression: p38-mitogen of multi-target drug and polypharmacy. Finally, stimu-
activated kinase, which phosphorylates and increases lation procedures for alleviating depression might be
the membrane expression of 5-HT leading to reduced combined with antidepressant agents in an attempt
levels of extracellular 5-HT (Bruchas et al., 2011; to improve efficacy (Eitan and Lerer, 2006).
Connolly and Thase, 2012). Glycogen synthase
kinase-β inhibition remains under consideration as
Concluding comments
well as a potential antidepressant strategy (Millan,
2009; Connolly and Thase, 2012; Duman and Voleti, As compared to highly selective agents, multi-modal
2012). Nonetheless, even for multi-target drugs, the therapeutic strategies appear to offer greater promise
above mechanisms all raise important questions of for the broad-based and improved treatment of
specificity and safety if they are to be therapeutically depressed states. The systematic pursuit of selective
exploited for treatment of depression or other CNS and multi-target agents together with the exploration
disorders. of novel drug combinations is a reasonable blueprint
1028 M. J. Millan

for future progress. There is nothing inherently radical on symptoms and serum IL-6 concentrations in patients
in all this inasmuch as multi-modal strategies are with major depressive disorder: randomized double-blind
already a focal point for the improved control of placebo-controlled study. J Affect Disord 141:308–314.
complex disorders as diverse as cancer, malaria, Abe Y, Aoyagi A, Hara T, Abe K, Yamazaki R, Kumagae Y,
Naruto S, Koyama K, Marumoto S, Tago K, Toda N,
Parkinson’s and Alzheimer’s disease (Toda et al.,
Takami K, Yamada N, Ori M, Kogen H, Kaneko T (2003)
2003; Millan, 2006; Van der Schyf et al., 2007; Petrelli
Pharmacological characterization of RS-1259, an orally
and Giordano, 2008; Piazzi et al., 2008). On the con- active dual inhibitor of acetylcholinesterase and serotonin
trary, it is surprising that multi-modal thinking is not transporter, in rodents: possible treatment of Alzheimer’s
more predominant in the field of depression ‘R and disease. J Pharmacol Sci 93:95–105.
D’ as a potential foundation for future progress. Al-Harbi KS (2012) Treatment-resistant depression: thera-
While combinations of clinically characterized com- peutic trends, challenges, and future directions. Patient
pounds remain feasible and potentially important (as Prefer Adhere 6:369–388.
illustrated by second-generation antipsychotics plus Aluisio L, Lord B, Bsarbier AJ, Fraser IC, Wilson SJ, Boggs J,
SSRIs), the development of mixtures of two new Dvorak LK, Letavic MA, Maryanoff BE, Carruthers NI,

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


drugs is more challenging and there are several limit- Bonaventure P, Lovenberg TWI (2008) In vitro and in vivo
characterization of JNJ-7925476, a novel triple monoamine
ations to polypharmaceutical approaches for treating
uptake inhibitor. Eur J Pharmacol 587:141–146.
depression. Conversely, while not ignoring the chemi-
Alvarez E, Perez V, Dragheim M, Loft H, Artigas F (2012)
cal difficulty of multi-target drug design and their A double-blind, rendomized, placebo-controlled, active
invariant ratio of component pharmacological activi- reference study of Lu AA21004 in patients with major
ties, they share many of the benefits of polypharmacy depressive disorder. Int J Neuropsychopharmacol
while lacking a number of important drawbacks. All 15:589–600.
things considered, multi-target agents may be the Andreasen JT, Nielsen EO, Christensen JK, Olsen GM,
most attractive route towards novel and improved Peters D, Mirza NR, Redrobe JP (2011) Subtype-selective
antidepressant agents. Currently, they are exemplified nicotinic acetylcholine receptor agonists enhance the
by the mechanistically innovative agent, agomelatine, responsiveness to citalopram and reboxetine in the mouse
but it is to be hoped that complementary multi-target forced swim test. J Psychopharmacol 25:1347–1356.
Andreasen JT, Redrobe JP, Nielsen EO (2012) Combined 7
agents with other advantages for treating additional
nicotinic acetylcholine receptor agonism and partial
dimensions of depression will eventually become
serotonin transporter inhibition produce antidepressant-
available. Together with improvements in translational like effects in the mouse forced swim and tail suspension
measures of drug actions between rodents and tests: a comparison of SSR180711 and PNU-282987.
humans, more objective biomarkers of incipient and Pharmacol Biochem Behav 100:624–629.
actual depressed states and innovations in clinical Andres JI, Alcazar J, Alonso JM et al. (2007) Tricyclic
evaluation of new drugs, a shift in mindset from highly isoxazolines identification of R226161 as a new
selective to multi-modal treatment strategies may help antidepressant that combines potent serotonin reuptake
revitalize a domain greatly in need of further progress inhibition and α2-adrenoceptor antagonism. Bioorg Med
in view of the insufficient current treatment of major Chem 15:3649–3660.
depression. Artigas F, Adell A, Celada P (2006) Pindolol augmentation of
antidepressant response. Curr Drug Targets 7:139–147.
Atcha Z, Chen WS, Ong AB, Wong FK, Neo A, Browne ER,
Witherington J, Pemberton DJ (2009) Cognitive enhancing
Acknowledgements
effects of ghrelin receptor agonists. Psychopharmacology
The author thanks Carmen Munoz, Elisabeth Mocaer 206:415–427.
and Philippe Delagrange for helpful comments on Autry AE, Monteggia LM (2012) Brain-derived neurotrophic
the paper, Marianne Soubeyran for preparing citations factor and neuropsychiatric disorders. Pharmacol Rev
64:238–258.
and tables and Alain Gobert for help with the figures.
Bacher I, Wu B, Shytle DR, George TP (2009) Mecamylamine –
a nicotinic acetylcholine receptor antagonist with potential
for the treatment of neuropsychiatric disorders. Expert
References
Opin Pharmacother 10:2709–2721.
Aan Het Rot M, Zarate CA, Charney DS, Mathew SJ (2012) Barbier AJ, Aluisio L, Lord B, Qu Y, Wilson SJ, Boggs JD,
Ketamine for depression: where do we go from here? Bonaventure P, Miller K, Fraser I, Dvorak L, Pudiak C,
Biol Psychiatry 72:537–547. Dugovic C, Shelton J, Mazur C, Letavic MA, Carruthers NI,
Abbasi SH, Hosseini F, Modabbernia A, Ashrafi M, Lovenberg TW (2007) Pharmacological characterization
Akhondzadeh S (2012) Effect of celecoxib add-on treatment of JNJ-28583867, a histamine H3 receptor antagonist
Complementary strategies for improving the treatment of depression 1029

and serotonin reuptake inhibitor. Eur J Pharmacol resilience in models of depression and addiction. Neuron
576:43–54. 71:498–511.
Bespalov AY, van Gaalen MM, Sukhotina IA, Wicke K, Bunney BG, Bunney WE (2012) Rapid-acting
Mezler M, Schoemaker H, Gross G (2008) Behavioral antidepressant strategies: mechanisms of action. Int J
characterization of the mGlu group II/III receptor Neuropsychopharmacol 15:695–713.
antagonist, LY-341495, in animal models of anxiety and Bunney JM, Potkin SG (2008) Circadian abnormalities,
depression. Eur J Pharmacol 592:96–102. molecular clock genes and chronobiological treatments in
Beyer CE, Lin Q, Platt B, Malberg J, Hornby G, Sullivan KM, depression. Br Med Bull 86:23–32.
Smith DL, Lock T, Mitchell PJ, Hatzenbuhler NT, Butler SG, Meegan J (2008) Recent developments in the
Evrard DA, Harrison BL, Magolda R, Pangalos MN, design of anti-depressive therapies: targeting the serotonin
Schechter LE, Rosenzweig-Lipson S, Andree TH (2009) transporter. Curr Med Chem 15:1737–1761.
Preclinical characterization of WAY-211612: a dual 5-HT Campbell JB (2010) Improving lead generation success
uptake inhibitor and 5-HT (1A) receptor antagonist and through integrated methods: transcending drug discovery
potential novel antidepressant. Br J Pharmacol 157:307–319. by numbers. IDrugs 13:874–877.
Blier P, Ward HE, Tremblay P, Laberge L, Hébert C, Campo B, Kalinichev M, Lambeng N, El Yacoubi M,

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Bergeron R (2010) Combination of antidepressant Royer-Urios I, Schneider M, Legrand C, Parron D, Girard F,
medications from treatment initiation for major depressive Bessif A, Poli S, Vaugeois JM, Le Poul E, Celanire S (2011)
disorder: a double-blind randomized study. Am J Characterization of an mGluR2/3 negative allosteric
Psychiatry 167:281–288. modulator in rodent models of depression. J Neurogenet
Bobo WV, Chen H, Trivedi MH, Stewart JW, Nierenberg AA, 25:152–166.
Fava M, Kurian BT, Warden D, Morris DW, Luther JF, Capuron L, Miller AH (2011) Immune system to brain
Husain MM, Cook IA, Lesser IM, Kornstein SG, signaling: neuropsychopharmacological implications.
Wisniewski SR, Rush AJ, Shelton RC (2011) Randomized Pharmacol Ther 130:226–238.
comparison of selective serotonin reuptake inhibitor Cardon LR, Chakravarti A, Cho JH, Guttmacher AE, Kong A,
(escitalopram) monotherapy and antidepressant Kruglyak L, Mardis E, Rotimi CN, Slatkin M, Valle D,
combination pharmacotherapy for major depressive Whittemore AS, Boehnke M (2009) Finding the missing
disorder with melancholic features: a CO-MED report. heritability of complex diseases. Nature 461:747–753.
J Affect Disord 133:467–476. Carlini VP, Machado DG, Buteler F, Ghersi M, Ponzio MF,
Bradley SR, Uslaner JM, Flick RB, Lee A, Groover KM, Martini AC, Schioth HB, de Cuneo MF, Rodrigues AL,
Hutson PH (2012) The mGluR7 allosteric agonist AMN082 de Barioglio SR (2012) Acute ghrelin administration
produces antidepressant-like effects by modulating reverses depressive-like behavior induced by bilateral
glutamatergic signaling. Pharmacol Biochem Behav olfactory bulbectomy in mice. Peptides 35:160–2165.
101:35–40. Carvalho AF, Machado JR, Cavalcante JL (2008)
Brady LS, Winsky L, Goodman W, Oliveri ME, Stover E Augmentation strategies for treatment-resistant depression.
(2009) NIMH initiatives to facilitate collaborations among Curr Opin Psychiatry 22:7–12.
industry, academia, and government for the discovery and Catena-Dell’Osso M, Marazziti D, Rotella F, Bellantuono C
clinical testing of novel models and drugs for psychiatric (2012) Emerging targets for the pharmacological treatment
disorders. Neuropsychopharmacology 34:229–243. of depression: focus on melatonergic system. Curr Med
Brennan JA, Graf R, Grauer SM, Navarra RL, Pulicicchio CM, Chem 19:428–437.
Hughes ZA, Lin Q, Wantuch C, Rosenzweig-Lipson S, Chaki S, Okubo T (2007) Melanocortin-4 receptor antagonists
Pruthi F, Lai M, Smith D, Goutier W, van de Neut M, for the treatment of depression and anxiety disorders.
Robichaud AJ, Rotella D, Feenstra RW, Kruse C, Broqua P, Curr Top Med Chem 7:1145–1151.
Beyer CE (2010) WS-50030 [7-{4-[3-(1H-inden-3-yl)propyl] Chaki S, Oshida Y, Ogawa S et al. (2005) MCL0042: a
piperazin-1-yl}−1,3-benzoxazol-2(3H)-one]: a novel nonpeptide MC4 receptor antagonist and serotonin
dopamine D2 receptor partial agonist/serotonin reuptake reuptake inhibitor with anxiolytic and antidepressant-like
inhibitor with preclinical antipsychotic-like and activity. Pharmacol Biochem Behav 82:621–626.
antidepressant-like activity. J Pharmacol Exp Ther 332:190–201. Chuang JC, Zigman JM (2010) Ghrelin’s roles in stress, mood,
Brocco M, Dekeyne A, Mannoury la Cour C, Touzard M, and anxiety regulation. Int J Peptides pii:4605549.
Girardon S, Veiga S, de Nanteuil G, deJong TR, Olivier B, Cipriani A, Santilli C, Furukawa TA, Signoretti A,
Millan MJ (2008) Cellular and behavioural profile of Nakagawa A, McGuire H, Churchill R, Barbui C (2009)
the novel, selective neurokinin1 receptor antagonist, Escitalopram vs. other antidepressive agents for depression.
vestipitant: a comparison to other agents. Eur Cochrane Database Syst Rev 15:CD006532.
Neuropsychopharmacol 10:729–750. Connolly KR, Thase ME (2011) If at first you don’t succeed.
Bruchas MR, Schindler AG, Shankar H, Messinger DI, Drugs 71:43–64.
Miyatake M, Land BB, Lemos JC, Hagan CE, Neumaier JF, Connolly KR, Thase ME (2012) Emerging drugs for major
Quintana A, Palmiter RD, Chavkin C (2011) Selective p38α depressive disorder. Expert Opin Emerging Drugs
MAPK deletion in serotonergic neurons produces stress 17:105–126.
1030 M. J. Millan

Cooper-Kazaz R, Lerer B (2008) Efficacy and safety of neurochemical, and electrophysiological characterization.
triiodothyronine supplementation in patients with major J Pharmacol Exp Ther 340:765–780.
depressive disorder treated with specific serotonin reuptake Dell’Osso B, Zanoni S, Ferrucci R, Vergari M, Castellano F,
inhibitors. Int J Neuropsychopharmacol 11:685–699. D’Urso N, Dobrea C, Benatti B, Arici C, Priori A,
Cordi AA, Berque-Bestel I, Persigand T, Lacoste M, Altamura AC (2012) Transcranial direct current stimulation
Newman-Tancredi A, Audinot V, Millan MJ (2001) for the outpatient treatment of poor-responder depressed
Potential antidepressants display combined patients. Eur Psychiatry 27:513–517.
α2-adrenoceptor antagonist and monoamine uptake De Maat SM, Dekker J, Schoevers RA, de Jonghe F (2007)
inhibitor properties. J Med Chem 44:787–805. Relative efficacy of psychotherapy and combined therapy in
Covington HE, Maze I, LaPlant QC, Vialon VE, Ohnishi YN, treatment of depression: a meta-analysis. Eur Psychiatry
Berton O, Fass DM, Renthal W, Rush AJ, Wu EY, Ghose S, 22:1–8.
Krishnan V, Russo SJ, Tamminga C, Haggarty SJ, Nestler EJ De Paulis T (2007) Drug evaluation: vilazodone – a combined
(2009) Antidepressant actions of histone deacetylase SSRI and 5-HT1A partial agonist for the treatment of
inhibitors. J Neurosci 29:11451–11460. depression. Idrugs 10:193–201.
Covington HE, Vialou V, Nestler EJ (2010) From synapse to DeRubeis RJ, Siegle GJ, Hollon SD (2008) Cognitive therapy

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


nucleus: novel targets for treating depression. vs. medication for depression: treatment outcomes and
Neuropharmacology 58:683. neural mechanisms. Nat Rev Neurosci 9:788–796.
Cremers TI, Rea K, Bosker FJ, Wikstrom HV, Hogg S, Mork A, Dhir A, Kulkarni SK (2008) Antidepressant-like effect of 17
Westerink BH (2007) Augmentation of SSRI effects on β-estradiol: involvement of dopaminergic, serotonergic,
serotonin by 5-HT2C antagonists: mechanistic studies. and (or) sigma-1 receptor systems. Can J Physiol Pharmacol
Neuropsychopharmacology 32:1550–1557. 86:726–735.
Crossley NA, Bauer M (2007) Acceleration and augmentation Di Cara B, Maggio R, Aloisi G, Rivet JM, Lundius EG,
of antidepressant with lithium for depressive disorders: Yoshitake T, Svenningsson P, Brocco M, Gobert A,
two meta-analyses of randomized, placebo-controlled trials. De Groote L, Cistarelli L, Veiga S, De Montrion C,
J Clin Psychiatry 68:935–940. Rodriguez M, Galizzi JP, Lockhart BP, Cogé F, Boutin JA,
Crowley SK, Youngstedt SD (2012) Efficacy of light therapy Vayer P, Verdouw PM, Groenink L, Millan MJ (2011)
for perinatal depression: a review. J Physiol Anthropol Genetic deletion of trace amine 1 receptors reveals their role
31:15. in auto-inhibiting the actions of ecstasy (MDMA). J
Czeh B, Di Benedetto B (2012) Antidepressants act directly on Neurosci 31:16928–16940.
astrocytes: evidence and functional consequences. Dos Santos LV, Souza FH, Brunetto AT, Sasse AD,
Eur Neuropsychopharmacol 23:171–185. da Silveira Nogueira Lima JP (2012) Neurokinin-1 receptor
Dawson LA, Bromidge SM (2008) 5-HT1 receptor antagonists for chemotherapy-induced nausea and
augmentation strategies as enhanced efficacy: therapeutics vomiting: a systematic review. J Natl Cancer Inst
for psychiatric disorders. Curr Top Med Chem 8:1008–1023. 104:1280–1292.
Day JJ, Sweatt JD (2012) Epigenetic treatments for cognitive Drevets WC, Furey ML (2010) Replication of scopolamine’s
impairments. Neuropsychopharmacology 37:247–260. antidepressant efficacy in major depressive disorder: a
De Bodinat C, Guardiola-Lemaitre B, Mocaër E, Renard P, randomized, placebo-controlled clinical trial.
Muñoz C, Millan MJ (2010) Agomelatine, the first Biol Psychiatry 67:432–438.
melatonergic antidepressant: discovery, characterization Duman RS, Voleti B (2012) Signaling pathways underlying the
and development. Nat Rev Drug Discov 9:628–642. pathophysiology and treatment of depression: novel
Dekeyne A, Denorme B, Monneyron S, Millan MJ (2000) mechanisms for rapid-acting agents. Trends Neurosci
Citalopram reduces social interaction in rats by activation 35:47–56.
of serotonin 5-HT2C receptors. Neuropharmacology Dwyer JM, Lepack AE, Duman RS (2012) mTOR activation
39:114–1117. is required for the antidepressant effects of mGluR2/3
Dekeyne A, Mannoury la Cour C, Gobert A, Brocco M, blockade. Int J Neuropsychopharmacol 15:429–434.
Lejeune F, Serres F, Sharp T, Daszuta A, Soumier A, Egeland M, Zhang X, Millan MJ, Mocaer E, Svenningsson P
Papp M, Rivet JM, Flik G, Cremers TI, Muller O, Lavielle G, (2012) Pharmacological or genetic blockade of the
Millan MJ (2008) S32006, a novel 5-HT2C receptor dopamine D3 receptor increases cell proliferation in the
antagonist displaying broad-base antidepressant and hippocampus of adult mice. J Neurochem 123:811–823.
anxiolytic properties in rodent models. Eitan R, Lerer B (2006) Neuropharmacological, somatic
Psychopharmacology 199:549–568. treatments of depression: electroconvulsive therapy and
Dekeyne A, Brocco M, Loiseau F, Gobert A, Rivet JM, Di novel brain stimulation modalities. Dialogues Clin Neurosci
Cara B, Cremers TI, Flik G, Fone KC, Watson DJ, Papp M, 8:241–258.
Sharp T, Serres F, Cespuglio R, Olivier B, Chan JS, Farley S, Apazoglou K, Witkin JM, Giros B, Tzavara ET (2010)
Lavielle G, Millan MJ (2012) S32212, a novel serotonin Antidepressant-like effects of an AMPA receptor potentia-
type 2C receptor inverse agonist/α2-adrenoceptor tor under a chronic mild stress paradigm. Int J
antagonist and potential antidepressant: II. A behavioral, Neuropsychopharmacol 13:1207–1218.
Complementary strategies for improving the treatment of depression 1031

Fava M, Targum SD, Nierenberg AA, Bleicher LS, Carter TA, combination treatment with antipsychotics. Eur
Wedel PC, Hen R, Gage FH, Barlow C (2012) An Neuropsychopharmacol 19:520–532.
exploratory study of combination buspirone and melatonin Graef S, Schönknecht P, Sabri O, Hegerl U (2011) Cholinergic
SR in major depressive disorder (MDD): a possible role receptor subtypes and their role in cognition, emotion, and
for neurogenesis in drug discovery. Psychiatr Res vigilance control: an overview of preclinical and clinical
3956:00246–00244. findings. Psychopharmacology 215:205–229.
Frampton JE (2011) Vilazodone in major depressive disorder. Graff J, Mansuy IM (2009) Epigenetic dysregulation in
CNS Drugs 25:615–627. cognitive disorders. Eur J Neurosci 30:1–8.
Gallagher P, Malik N, Newham J, Young AH, Ferrier IN, Guidi J, Fava GA, Fava M, Papakostas GI (2011) Efficacy
Mackin P (2008) Antiglucocorticoid treatments for mood of the sequential integration of psychotherapy and
disorders. Cochrane Database Syst Rev 1:CD005168. pharmacotherapy in major depressive disorder: a
Gelfo F, Tirassa P, De Bartolo P, Croce N, Bernardini S, preliminary meta-analysis. Psychol Med 41:321–331.
Caltagirone C, Petrosini L, Angelucci F (2012) NPY Guo JY, Li CY, Ruan YP, Sun M, Qi XL, Zhao BS, Luo F
intraperitoneal injections produce antidepressant-like (2009) Chronic treatment with celecoxib reverses chronic
effects and downregulate BDNF in the rat hypothalamus. unpredictable stress-induced depressive-like behavior

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


CNS Neurosci Ther 18:487–492. via reducing cyclooxygenase-2 expression in rat brain.
George TP, Sacco KA, Vassicchio JC, Weinberger AH, Eur J Pharmacol 612:54–60.
Shytle RD (2008) Nicotinic antagonist augmentation of Heidbreder CA, Newman AH (2010) Current perspectives
selective serotonin reuptake inhibitor-refractory major on selective dopamine D(3) receptor antagonists as
depressive disorder: a preliminary study. J Clin pharmacotherapeutics for addictions and related disorders.
Psychopharmacol 28:340–344. Ann N Y Acad Sci 1187:4–34.
Gilbody S, Lightfoot T, Sheldon T (2007) Is low folate a risk Heim C, Binder EB (2012) Current research trends in early life
factor for depression? A meta-analysis and exploration of stress and depression: review of human studies on sensitive
heterogeneity. J Epidemiol Community Health 61:631–637. periods, gene-environment interactions, and epigenetics.
Gillman PK (2007) Tricyclic antidepressant pharmacology and Exp Neurol 233:102–111.
therapeutic drug interactions updated. Br J Pharmacol Hemmeter UM, Hemmeter-Spernal J, Krieg JC (2010)
151:737–748. Sleep deprivation in depression. Expert Rev Neurother
Glezer A, Byatt N, Cook R, Rothschild AJ (2009) 10:1101–1015.
Polypharmacy prevalence rates in the treatment of Hopkins AL, Mason JS, Overington JP (2006) Can we
unipolar depression in an outpatient clinic. J Affect Disord rationally design promiscuous drugs? Curr Opin Struct Biol
117:18–23. 16:127–136.
Gobert A, Rivet JM, Cistarelli L, Melon C, Millan MJ (1999) Hughes ZA, Liu F, Platt BJ, Dwyer JM, Pulicicchio CM,
Buspirone modulates basal and fluoxetine-stimulated Zhang G, Schechter LE, Rosenzweig-Lipson S, Day M
dialysate levels of dopamine, noradrenaline and serotonin (2008) WAY-200070, a selective agonist of estrogen receptor
in the frontal cortex of freely moving rats: activation of β as a potential novel anxiolytic/antidepressant agent.
serotonin1A receptors and blockade of α2-adrenergic Neuropharmacology 54:1136–1142.
receptors underlie its actions. Neuroscience 93:1251–1262. Hughes ZA et al. (2012) Negative allosteric modulation of
Gobert A, Brocco M, Dekeyne A, Di Cara B, Bouchez G, metabolic glutamate receptor 5 results in broad spectrum
Lejeune F, Gannon RL, Millan MJ (2009). Neurokinin1 activity relevant to treatment resistant depression.
antagonists potentiate antidepressant properties of Neuropharmacology 66:202–214.
serotonin reuptake inhibitors, yet blunt their anxiogenic Ibrahim L, DiazGranados N, Jolkovsky L, Brutsche N,
actions: a neurochemical, electrophysiological, and Luckenbaugh DA, Herring J, Potter WZ, Zarate CA (2012)
behavioral characterization. Neuropsychopharmacology A randomized, placebo-controlled, crossover pilot trial of
34:1039–1056. the oral selective NR2B antagonist MK-0657 in patients
Goldberg JF, Brooks JO, Kurita K, Hoblyn JC, Ghaemi N, with treatment-resistant major depressive disorder. J Clin
Perlis RH, Miklowitz DJ, Ketter TA, Sachs GS, Thase ME Psychopharmacol 32:551–557.
(2009) Depressive illness burden associated with complex Inta D, Filipovic D, Lima-Ojeda JM, Dormann C, Pfeiffer N,
polypharmacy in patients with bipolar disorder: findings Gasparini F, Gass P (2012) The mGlu5 receptor antagonist
from the STEP-BD. J Clin Psychiatry 70:155–162. MPEP activates specific stress-related brain regions
Gomes MV, Joca SR (2011) Antidepressant-like effect and lacks neurotoxic effects of the NMDA receptor
induced by systemic and intra-hippocampal administration antagonist MK-801: significance for the use as anxiolytic/
of DNA methylation inhibitors. Br J Pharmacol antidepressant drug. Neuropharmacology 62:2034–2039.
164:1711–1721. Ishak WW, HA K, Kapitanski N, Bagot K, Fathy H,
Goodwin G, Fleischhacker W, Arango C, Baumann P, Swanson B, Vilhauer J, Balayan K, Bolotaulo NI,
Davidson M, de Hert M, Falkai P, Kapur S, Leucht S, Rapaport MH (2011) The impact of psychotherapy,
Licht R, Naber D, O’Keane V, Papakostas G, Vieta E, pharmacotherapy, and their combination on quality of
Zohar J (2009) Advantages and disadvantages of life in depression. Harv Rev Psychiatry 19:277–289.
1032 M. J. Millan

Ishida H, Shirayama Y, Iwata M, Katayama S, Yamamoto A, Ledford H (2011) Depression drug disappoints: failure of a
Kawahara R, Nakagome K (2007) Infusion of neuropeptide promising compound casts a shadow on others. Nature
Y into CA3 region of hippocampus produces 479:278–280.
antidepressant-like effect via Y1 receptor. Hippocampus Lee JC, Blumberger DM, Fitzgerald P, Daskalakis Z,
17:271–280. Levinson A (2012) The role of transcranial magnetic
Ishitobi Y, Kohno K, Kanehisa M, Inoue A, Imanaga J, stimulation in treatment-resistant depression: a review.
Maruyama Y, Ninomiya T, Higuma H, Okamoto S, Curr Pharm Des 18:5846–5852.
Tanaka Y, Tsuru J, Hanada H, Isogawa K, Akiyoshi J (2012) Leonard B, Maes M (2012) Mechanistic explanations
Serum ghrelin levels and the effects of antidepressants in how cell-mediated immune activation, inflammation
major depressive disorder and panic disorder. and oxidative stress pathways and their sequels
Neuropsychobiology 66:185–192. and concomitants play a role in the pathophysiology
Jernigan CS, Goswami DB, Austin MC, Iyo AH, Chandran A, of unipolar depression. Neurosci Biobehav Res 36,
Stockmeier CA, Karolewicz B (2011) The mTOR signaling 764–785.
pathway in the prefrontal cortex is compromised in major Levin ED (2012) α7-Nicotinic receptors and cognition.
depressive disorder. Prog Neuropsychopharmacol Biol Curr Drug Targets 13:602–606.

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Psychiatry 35:1774–1779. Li N, Lee B, Liu RJ, Banasr M, Dwyer JM, Iwata M, Li XY,
Johansson D, Falk A, Marcus MW, Svensson TH (2012) Aghajanian G, Duman RS (2010) mTOR-dependent synapse
Celecoxib enhances the effect of reboxetine and fluoxetine formation underlies the rapid antidepressant effects of
on cortical noradrenaline and serotonin output in NMDA antagonists. Science 329:959–964.
the rat. Prog Neuropsychopharmacol Biol Psychiatry Li X, Witkin JM, Need AB, Skolnick P (2003) Enhancement
39:143–148. of antidepressant potency by a potentiator of AMPA
Kasper S, Hajak G, Wulff K, Hoogendijk WJ, Montejo AL, receptors. Cell Mol Neurobiol 23:419–430.
Smeraldi E, Rybakowski JK, Quera-Salva MA, Lin D, Mok H, Yatham LN (2006) Polytherapy in bipolar
Wirz-Justice AM, Picarel-Blanchot F, Baylé FJ (2010) disorder. CNS Drugs 20:29–42.
Efficacy of the novel antidepressant agomelatine on the Lin H, Geng X, Dang W, Wu B, Dai Z, Li Y, Yang Y, Zhang H,
circadian rest-activity cycle and depressive and anxiety Shi J (2012) Molecular mechanisms associated with the
symptoms in patients with major depressive disorder: a antidepressant effects of the class I histone deacetylase
randomized, double-blind comparison with sertraline. inhibitor MS-275 in the rat ventrolateral orbital cortex.
J Clin Psychiatry 71:109–120. Brain Res 1447:119–125.
Katona C, Hansen T, Olsen CK (2012) A randomized, Lohoff FW (2010) Overview of the genetics of
double-blind, placebo-controlled, duloxetine-referenced, major depressive disorder. Curr Psychiatry Rep
fixed-dose study comparing the efficacy and 12:539–546.
safety of Lu AA21004 in elderly patients with Loiseau F, Millan MJ (2009) Blockade of dopamine
major depressive disorder. Int Clin Psychopharmacol D3 receptors in frontal cortex, but not in sub-cortical
27:215–223. structures, enhances social recognition in rats: similar
Kehne JH, Cain CK (2010) Therapeutic utility of non-peptidic actions of D1 receptor agonists, but not of D2 antagonists.
CRF1 receptor antagonists in anxiety, depression, and Eur Neuropsychopharmacol 19:23–33.
stress-related disorders: evidence from animal models. Lopez-Munoz F, Alamo C, Rubio G, Garcia-Garcia P, Pardo A
Pharmacol Ther 128:460–487. (2007) Reboxetine combination in treatment-resistant
Kennedy SH, Konarski JZ, Segal ZV, Lau MA, Bieling PJ, depression to selective serotonin reuptake inhibitors.
McIntyre RS, Mayberg HS (2007) Differences in brain Pharmacopsychiatry 40:14–19.
glucose metabolism between responders to CBT and Lucas G (2009) Serotonin receptors, type 4: a new hope?
venlafaxine in a 16-week randomized controlled trial. Curr Drug Targets 10:1085–1095.
Am J Psychiatry 164:778–788. Lutter M, Sakata I, Osborne-Lawrence S, Rovinsky SA,
Köhler S, Hoffmann S, Unger T, Steinacher B, Dierstein N, Anderson JG, Jung S, Birnbaum S, Yanagisawa M,
Fydrich T (2011) Effectiveness of cognitive-behavioural Elmquist JK, Nestler EJ, Zigman JM (2008) The orexigenic
therapy plus pharmacotherapy in inpatient treatment of hormone ghrelin defends against depressive symptoms of
depressive disorders. Clin Psychol Psychother. Advance chronic stress. Nat Neurosci 11:752–753.
online publication. doi: 10.1002/cpp.795. McIntyre M, Moral MA (2006) Augmentation in
Kramer JM, van Bokhoven H (2009) Genetic and treatment-resistant depression. Drugs Fut 31:1069–1081.
epigenetic defects in mental retardation. Int J Biochem McIntyre RS, Jerrell JM (2009) Polypharmacy in children and
Cell Biol 41:96–107. adolescents treated for major depressive disorder: a claims
Krause DL, Riedel M, Müller N, Weidinger E, Schwarz MJ, database study. J Clin Psychiatry 70:240–246.
Myint AM (2012) Effects of antidepressants and McIntyre RS, Soczynska JK, Woldeyohannes HO,
cyclooxygenase-2 inhibitor on cytokines and kynurenines in Alsuwaidan M, Konarski JZ (2007) A preclinical and clinical
stimulated in vitro blood culture from depressed patients. rationale for quetiapine in mood syndromes. Expert Opin
Inflammopharmacology 20:169–176. Pharmacother 8:1211–1219.
Complementary strategies for improving the treatment of depression 1033

McIntyre RS, Rosenbluth M, Ramasubbu R, Bond DJ, disorders (McArthur R, Borsini F, eds), pp32–89. New York:
Taylor VH, Beaulieu S, Schaffer A (2012) Canadian Academic Press.
network for mood and anxiety treatments (CANMAT) task Millan MJ (2009) Dual- and triple-acting agents for treating
force. Managing medical and psychiatric comorbidity in core and co-morbid symptoms of major depression: novel
individuals with major depressive disorder and bipolar concepts, new drugs. Neurotherapeutics 6:53–77.
disorder. Ann Clin Psychiatry 24:163–169. Millan MJ (2011) MicroRNA in the regulation and expression
Maeng S, Zarate CA, Du J, Schloesser RJ, McCammon J, of serotonergic transmission in the brain and other tissues.
Chen G, Manji HK (2008) Cellular mechanisms underlying Curr Opin Pharmacol 11:11–22.
the antidepressant effects of ketamine role of Millan MJ (2013) An epigenetic framework for
alpha-amino-3-hydroxy-5-methylosoxazole-4-propionic neurodevelopmental disorders: from pathogenesis to
acid receptors. Biol Psychiatry 63:349–352. therapy. Neuropharmacology 68:2–82.
Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Millan MJ, Brocco M (2008) Cognitive impairment in
Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, schizophrenia: a review of developmental and genetic
Chakravarti A, Cho JH, Guttmacher AE, Kong A, models, and pro-cognitive profile of the optimised
Kruglyak L, Mardis E, Rotimi CN, Slatkin M, Valle D, D(3)>D(2) antagonist, S33138. Therapie 63:187–229.

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Whittemore AS (2009) Finding the missing heritability of Millan MJ, Canton H, Gobert A, Lejeune F, Rivet JM,
complex diseases. Nature 461:747–753. Bervoets K, Brocco M, Widdowson P, Mennini T,
Marchand WR (2012) Mindfulness-based stress reduction, Audinot V, Honore P, Renouard A, Le
mindfulness-based cognitive therapy, and Zen meditation Marouille-Girardon S, Verreille L, Gressier H, Peglion JL
for depression, anxiety, pain, and psychological distress. (1994) Novel benzodioxopiperazines acting as antagonists
J Psychiatr Pract 18:233–252. at postsynaptic 5-HT1A receptors and as agonists at
Mathew SJ (2012) Ketamine for treatment-resistant unipolar 5-HT1A autoreceptors: a comparative pharmacological
depression: current evidence. CNS Drugs 26:189–204. characterization with proposed 5-HT1A antagonists.
Matrisciano F, Zusso M, Panaccione I, Turriziani B, Caruso A, J Pharmacol Exp Ther 268:337–352.
Iacovelli L, Noviello L, Togna G, Melchiorri D, Debetto P, Millan MJ, Lejeune F, Gobert A (2000a) Reciprocal
Tatarelli R, Battaglia G, Nicoletti F, Giusti P, Girardi P autoreceptor and heteroreceptor control of serotonergic,
(2008) Synergism between fluoxetine and the mGlu2/3 dopaminergic and noradrenergic transmission in the frontal
receptor agonist, LY379268, in an in vitro model for cortex: relevance to the actions of antidepressant agents.
antidepressant drug-induced neurogenesis. J Psychopharmacol 14:114–138.
Neuropharmacology 54:428–437. Millan MJ, Newman-Tancredi A, Audinot V, Cussac D,
Meffre J, Chaumont-Dubel S, Mannoury la Cour C, Loiseau F, Lejeune F, Cogé F, Galizzi JP, Boutin JA, Rivet JM,
Watson DJG, Dekeyne A, Seveno M, Rivet J-M, Gaven F, Dekeyne A, Gobert A (2000b) Agonist and antagonist
Deleris P, Herve D, Fone KCF, Bockaert J, Millan MJ, actions of yohimbine as compared to fluparoxan at a
Marin P (2012) 5-HT6 receptor recruitment of mTOR as a α2-adrenergic receptors (AR)s, serotonin (5-HT(1A), 5-HB
mechanism for perturbed cognition in schizophrenia. (1B), 5-HT(1D) and dopamine D(2) and D(3) receptors.
EMBO Mol Med 4:1043–1056. Significance for the modulation of frontocortical
Melander H, Salmonson T, Abadie E, Van Zwieten-Boot B monoamingeric transmission and depressive states.
(2008) A regulatory apologia – a review of Synapse 35:79–95.
placebo-controlled studies in regulatory submissions of Millan MJ, Dekeyne A, Papp M, La Rochell CD,
new generation antidepressants. Eur MacSweeny C, Peglion JL, Brocco M (2001a) S33005, a novel
Neuropsychopharmacol 18:623–627. ligand at both serotonin and norepinephrine transporters:
Meltzer HY, Massey BW, Horiguchi M (2012) Serotonin II. Behavioral profile in comparison with venlafaxine,
receptors as targets for drugs useful to treat psychosis reboxetine, citalopram, and clomipramine. J Pharmacol
and cognitive impairment in schizophrenia. Curr Pharm Exp Ther 298:581–591.
Biotechnol 13:1572–1586. Millan MJ, Lejeune F, De Nanteuil G, Gobert A (2001b)
Millan MJ (2005) Serotonin 5-HT2C receptors as a target for the Selective blockade of NK1 receptors facilitates the activity of
treatment of depressive and anxious states: focus on novel adrenergic pathways projecting to frontal cortex and dorsal
therapeutic strategies. Therapie 60:441–460. hippocampus in rats. J Neurochem 76:1949–1954.
Millan MJ (2006) Multi-target strategies for the improved Millan MJ, Di Cara B, Dekeyne A, Panayi F, De Groote L,
treatment of depressive states: conceptual foundation Sicard D, Cistarelli L, Billiras R, Gobert A (2007) Selective
and neuronal substrates, drug discovery and therapeutic blockade of dopamine D3 vs D2 receptors enhances
application. Pharmacol Ther 110:135–370. frontocortical cholinergic transmission and social memory
Millan MJ (2008) The discovery and development of in rats: a parallel neurochemical and behavioural analysis.
pharmacotherapy for psychiatric disorders: a critical J Neurochem 100:1047–1061.
survey of animal and translational models, and Millan MJ, Dekeyne A, Gobert A, Mannoury la Cour C,
perspectives for their improvement. In: Translational Brocco M, Rivet JM, Di Cara B, Lejeune F, Cremers TI,
models for CNS drug discovery. Vol. 1: Psychiatric Flik G, de Jong TR, Olivier B, de Nanteuil G (2010) S41744,
1034 M. J. Millan

a dual neurokinin (NK)1 receptor antagonist and Morphy R, Rankovic Z (2006) The physicochemical
serotonin (5-HT) reuptake inhibitor with potential challenges of designing multiple ligands. J Med Chem
antidepressant properties: a comparison to aprepitant 49:4961–4970.
(MK869) and paroxetine. Eur Neuropsychopharmacol Morphy R, Rankovic Z (2007) Fragments, network biology
20:599–621. and designing multiple ligands. Drug Discov Today
Millan MJ, Mannoury la Cour C, Chanrion B, Dupuis DS, Di 12:156–160.
Cara B, Audinot V, Cussac D, Newman-Tancredi A, Mouillet-Richard S, Baudry A, Launay JM, Kellermann O
Kamal M, Boutin JA, Jockers R, Marin P, Bockaert J, (2012) MicroRNAs and depression. Neurobiol Dis
Muller O, Dekeyne A, Lavielle G (2012) S32212, a novel 46:272–278.
serotonin type 2C receptor inverse agonist/α2-adrenoceptor Muller N (2010) COX-2 inhibitors as antidepressants and
antagonist and potential antidepressant: antipsychotics: clinical evidence. Curr Opin Invest Drugs
I. A mechanistic characterization. J Pharmacol Exp Ther 11:31–42.
340:750–764. Müller N, Schwarz MJ, Dehning S, Douhe A, Cerocecki A,
Miller G (2010) Is pharma running out of brainy ideas. Science Goldstein-Müller B, Spellmann I, Hetzel G, Maino K,
329:502–504. Kleindienst N, Möller HJ, Arolt V, Riedel M (2006) The

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Minichino A, Bersani FS, Capra E, Pannese R, Bonanno C, cyclooxygenase-2 inhibitor celecoxib has therapeutic effects
Salviati M, Chiaie RD, Biondi M (2012) ECT, rTMS, and in major depression: results of a double-blind, randomized,
deep TMS in pharmacoresistant drug-free patients with placebo controlled, add-on pilot study to reboxetine.
unipolar depression: a comparative review. Neuropsychiatr Mol Psychiatry 11:680–684.
Dis Treat 8:1–10. Müller N, Krause D, Dehning S, Musil R, Schennach-Wolff R,
Minzenberg MJ, Carter CS (2008) Modafinil: a review of Obermeier M, Möller HJ, Klauss V, Schwarz MJ,
neurochemical actions and effects on cognition. Riedel M (2010) Celecoxib treatment in an early stage
Neuropsychopharmcology 33:1477–1502. of schizophrenia: results of a randomized, double-blind,
Mnie-Filali O, Faure C, Lambas-Senas L, El Mansari M, placebo-controlled trial of celecoxib augmentation of
Belblidia H, Gondard E, Etiévant A, Scarna H, Didier A, amisulpride treatment. Schizophr Res 121:118–124.
Berod A, Blier P, Haddjeri N (2011) Pharmacological Nations KR, Dogterom P, Bursi R, Schipper J, Greenwald S,
blockade of 5-HT7 receptors as a putative fast acting Zraket D, Gertsik L, Johnstone J, Lee A, Pande Y, Ruigt G,
antidepressant strategy. Neuropsychopharmacology Ereshefsky L (2012) Examination of Org 26576, an AMPA
36:1275–1288. receptor positive allosteric modulator, in patients
Mojtabai R, Olfson M (2010) National trends in psychotropic diagnosed with major depressive disorder: an exploratory,
medication polypharmacy in office-base psychiatry. randomized, double-blind, placebo-controlled trial.
Arch Gen Psychiatry 67:26–36. J Psychopharmacol 26:1525–1539.
Molina-Hernandez M, Tellez-Alcantara NP (2011) Fluoxetine, Nguyen HT, Guiard BP, Bacq A, David DJ, David I,
17-β estradiol or folic acid combined with intralateral septal Quesseveur G, Gautron S, Sanchez C, Gardier AM (2013)
infusions of neuropeptide Y produced antidepressant-like Blockade of the high-affinity norepinephrine transporter
actions in ovariectomized rats forced to swim. Peptides (NET) by the selective serotonin reuptake inhibitor
32:2400–2406. escitalopram: an in vivo microdialysis study in mice.
Molina-Hernández M, Téllez-Alcántara NP, Olivera-López JI, Br J Pharmacol 168:103–116.
Jaramillo MT (2012) Intra-lateral septal infusions of folic Nicoletti F, Bockaert J, Collingridge GL, Conn PJ,
acid alone or combined with various antidepressant drugs Ferraguti F, Schoepp DD, Wroblewski JT, Pin JP
produce antidepressant-like actions in male Wistar rats (2011) Metabotropic glutamate receptors: from the
forced to swim Prog Neuropsychopharmacol Biol workbench to the bedside. Neuropharmacology
Psychiatry 36:78–84. 60:1017–10441.
Montgomery SA, Baldwin DS, Blier P, Blier P, Fineberg NA, Nutt D, Goodwin G (2011) ECNP Summit on the future of
Kasper S, Lader M, Lam RW, Lépine JP, Möller HJ, Nutt DJ, CNS drug research in Europe 2011: report prepared for
Rouillon F, Schatzberg AF, Thase ME (2007) Which ECNP by David Nutt and Guy Goodwin Eur
antidepressants have demonstrated superior efficacy? Neuropsychopharmacol 21:495–499.
A review of the evidence. Int Clin Psychopharmacol Packiarajan M, Marzabadi MR, Desai M, Lu Y, Noble SA,
22:323–329. Wong WC, Jubian V, Chandrasena G, Wolinsky TD,
Mork A, Pehrson A, Brennum LT, Nielsen SM, Zhong H, Zhong H, Walker MW, Wiborg O, Andersen K (2011)
Lassen AB, Miller S, Westrich L, Boyle NJ, Sanchez C, Discovery of Lu AA33810: a highly selective and potent
Fischer CW, Liebenberg N, Wegener G, Bundgaard C, NPY5 antagonist with in vivo efficacy in a model of mood
Hogg S, Bang-Anderson B, Stensbol TB (2012) disorder. Bioorg Med Chem Lett 21:5436–5441.
Pharmacological effects of Lu AA21004: a novel Palucha A, Klak K, Banski P, van der Putten H, Flor PJ, Pilc A
multimodal compound for the treatment of major (2007) Activation of the mGlu7 receptor elicits
depressive disorder. J Pharmacol Exp Ther antidepressant-like effects in mice. Psychopharmacology
340:666–675. 194:555–562.
Complementary strategies for improving the treatment of depression 1035

Papakostas GI (2010) The efficacy, tolerability, and safety Kilduff TS, Biemans B, Pouzet B, Caron MG, Canales JJ,
of contemporary antidepressants. J Clin Psychiatry 71 Wallace TL, Wettstein JG, Hoener MC (2012a) Trace
(Suppl. E1):e03. amine-associated receptor 1 partial agonism reveals novel
Penrod NM, Cowper-Sal-lari R, Moore JH (2011) Systems paradigm for neuropsychiatric therapeutics. Biol Psychiatry
genetics for drug target discovery. Trends Pharmacol Sci 72:934–942.
32:623–630. Revel FG, Moreau JL, Pouzet B, Mory R, Bradaia A, Buchy D,
Peters J-U, Hert J, Bissantz C, Hillebrecht A, Gerebtzoff G, Metzler V, Chaboz S, Groebke Zbinden K, Galley G,
Bendels S, Tillier F, Migeon J, Fischer H, Guba W, Kansy M Norcross RD, Tuerck D, Bruns A, Morairty SR, Kilduff TS,
(2012) Can we discover pharmacological promiscuity early Wallace TL, Risterucci C, Wettstein JG, Hoener MC (2012b)
in the drug discovery process? Drug Discovery Today A new perspective for schizophrenia: TAAR1 agonists
17:325–335. reveal antipsychotic – and antidepressant-like activity,
Philip NS, Carpenter LL, Tyrka AR, Price LH (2012) The nic- improve cognition and control body weight.
otinic acetylcholine receptor as a target for antidepressant Mol Psychiatry. Advance online publication.
drug development. Sci World J 104105:1–7. doi:10.1038/mp.2012.57.
Piazzi L, Cavalli A, Colizzi F, Belluti F, Bartolini M, Mancini F, Rocha FL, Fuzikawa C, Riera R, Hara C (2012) Combination

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


Recanatini M, Andrisano V, Rampa A (2008) of antidepressants in the treatment of major depressive
Multi-target-directed coumarin derivatives: hAChE and disorder: a systematic review and meta-analysis. J Clin
BACE1 inhibitors as potential anti-Alzheimer compounds. Psychopharmacol 32:278–281.
Bioorg Med Chem Lett 18:423–426. Rogoz Z, Skuza G (2006) Mechanism of synergistic action
Petrelli A, Giordano S (2008) From single- to multi-target following co-treatment with pramipexole and fluoxetine or
drugs in cancer therapy: when aspecificity becomes an sertraline in the forced swimming test in rats. Pharmacol
advantage. Curr Med Chem 15:422–432. Rep 58:493–500.
Plenge P, Shi L, Beuming T, Te J, Newman AH, Weinstein H, Rush AJ, Trivedi MH, Stewart JW, Nierenberg AA, Fava M,
Gether U, Loland CJ (2012) Steric hindrance mutagenesis in Kurian BT, Warden D, Morris DW, Luther JF, Husain MM,
the conserved extracellular vestibule impedes allosteric Cook IA, Shelton RC, Lesser IM, Kornstein SG,
binding of antidepressants to the serotonin transporter. Wisniewski SR (2011) Combining medications to enhance
J Biol Chem 287:39316–39326. depression outcomes (CO-MED): acute and long-term
Prudent M, Dramé M, Jolly D, Trenque T, Parjoie R, outcomes of a single-blind randomized study. Am J
Mahmoudi R, Lang PO, Somme D, Boyer F, Lanièce I, Psychiatry 168:689–701.
Gauvain JB, Blanchard F, Novella JL (2008) Potentially Sanacora G, Berman RM, Cappiello A, Oren DA, Kugaya A,
inappropriate use of psychotropic medications in Liu N, Gueorguieva R, Fasula D, Charney DS (2004)
hospitalized elderly patients in France: cross-sectional Addition of the α2-antagonist yohimbine to fluoxetine:
analysis of the prospective, multicenter SAFEs cohort. effects on rate of antidepressant response.
Drugs Aging 25:933–946. Neuropsychopharmacol 29:1166–1171.
Quartara L, Altamura M, Evangelista S, Maggi CA (2009) Sanchez-Mut JV, Huertas D, Esteller M (2012) Aberrant
Tachykinin receptor antagonists in clinical trials. epigenetic landscape in intellectual disability. Prog Brain
Expert Opin Investig Drugs 18:1843–1864. Res 197:53–71.
Quesseveur G, Nguyen HT, Gardier AM, Guiard BP (2012) Sartorius N et al. (2007) Antidepressant medications and other
5-HT2 ligands in the treatment of anxiety and depression. treatments of depressive disorders: a CINP Task Force
Expert Opin Investig Drugs 21:1701–1725. report based on a review of evidence. Int J
Racagni G, Riva MA, Molteni R, Musazzi L, Calabrese F, Neuropsychopharmacol 1:S1–S207.
Popoli M, Tardito D (2011) Mode of action of agomelatine: Schatzberg AF, Lindley S (2008) Glucocorticoid antagonists
synergy between melatonergic and 5-HT2C receptors. in neuropsychiatric disorders. Eur J Pharmacol
World J Biol Psychiatry 12:574–587. 583:358–364.
Ratti E, Bellew K, Bettica P, Bryson H, Zamuner S, Archer G, Schlaepfer TE, Agren H, Monteleone P, Gasto C, Pitchot W,
Squassante L, Bye A, Trist D, Krishnan KR, Fernandes S Rouillon F, Nutt DJ, Kasper S (2012) The hidden third:
(2011) Results from 2 randomized, double-blind, improving outcome in treatment-resistant depression.
placebo-controlled studies of the novel NK1 receptor J Psychopharmacol 26:587–602.
antagonist casopitant in patients with major depressive Schloesser RJ, Martinowich K, Manji HK (2012)
disorder. J Clin Psychopharmacol 31:727–733. Mood-stabilizing drugs: mechanisms of action. Trends
Reed CR, Kajdasz DK, Whalen H, Athanasiou MC, Neurosci 25:36–46.
Gallipoli S, Thase ME (2012) The efficacy profile of Schmidt HD, Shelton RC, Duman RS (2011) Functional
vilazodone, a novel antidepressant for the treatment of biomarkers of depression: diagnosis, treatment, and
major depressive disorder. Curr Med Res Opin 28:27–39. pathophysiology. Neuropsychopharmacology
Revel FG, Moreau JL, Gainetdinov RR, Ferragud A, 36:2375–2395.
Velazquez-Sanchez C, Sotnikova TD, Morairty SR, Serna MC, Cruz I, Real J, Gasco E, Galvan L (2010)
Harmeier A, Groebke Zbinden K, Norcross RD, Bradaia A, Duration and adherence of antidepressant treatment
1036 M. J. Millan

(2003 to 2007) based on prescription database. Eur Toda N, Tago K, Marumoto S, Takami K, Ori M, Yamada N,
Psychiatry 25:206–213. Koyama K, Naruto S, Abe K, Yamazaki R, Hara T,
Serres F, Millan MJ, Sharp T (2012) Molecular adaptation to Aoyagi A, Abe Y, Kaneko T, Kogen H (2003)
chronic antidepressant treatment: evidence for a more rapid A conformational restriction approach to the development
response to the novel α2-adrenoceptor antagonist/ of dual inhibitors of acetylcholinesterase and serotonin
5-HT-noradrenaline reuptake inhibitor (SNRI), S35966, transporter as potential agents for Alzheimer’s disease.
compared to the SNRI, venlafaxine. Int J Bioorg Med Chem 11:4389–4394.
Neuropsychopharmacol 15:617–629. Tran P, Skolnick P, Czobor P, Huang NY, Bradshaw M,
Shelton J, Bonaventure P, Li X, Yun S, Lovenberg T, McKinney A, Fava M (2012) Efficacy and tolerability of the
Dugovic C (2009) 5-HT7 receptor deletion enhances REM novel triple reuptake inhibitor amitifadine in the treatment
sleep suppression induced by selective serotonin reuptake of patients with major depressive disorder: a randomized,
inhibitors, but not by direct stimulation of 5-HT1A receptor. double-blind, placebo-controlled trial. J Psychiatr Res
Neuropharmacology 56:448–454. 46:64–71.
Shelton RC, Osuntokun O, Heinloth AN, Corya SA (2010) Trivedi MH, Fava M, Wisniewski SR, Thase ME, Quitkin F,
Therapeutic options for treatment-resistant depression. Warden D, Ritz L, Nierenberg AA, Lebowitz BD,

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022


CNS Drugs 24:131–161. Biggs MM, Luther JF, Shores-Wilson K, Rush AJ, STAR*D
Skolnick P, Basile AS (2006) Triple reuptake inhibitors as Study Team (2006) Medication augmentation after the
antidepressants. Drug Discov Today Ther Strat 3:489–494. failure of SSRIs for depression. N Engl J Med
Sotnikova TD, Caron MG, Gainetdinov RR (2009) Trace 354:1243–1252.
amine-associated receptors as emerging therapeutic targets. Uher R (2011) Genes, environment, and individual differences
Mol Pharmacol 76:229–235. in responding to treatment for depression. Harv Rev
Srinivasan V, De Berardis D, Shillcutt SD, Brzezinski A Psychiatry 19:109–124.
(2011) Role of melatonin in mood disorders and the Van der Schyf CJ, Geldenhuys WJ, Youdim MBH (2007)
antidepressant effects of agomelatine. Exp Opin Invest Multifunctional neuroprotective–neurorescue drugs for
Drugs 21:1503–1522. Parkinson’s disease. Future Neurol 2:411–423.
Stahl S (2007) Novel therapeutics for depression: Warner-Schmidt JL, Vanover KE, Chen EY, Marshall JJ,
L-methylfolate as a trimonoamine modulator and Greengard P (2011) Antidepressant effects of selective
antidepressant-augmenting agent. CNS Spectr 12:739–744. serotonin reuptake inhibitors (SSRIs) are attenuated by
Stahl SM (2009) Mechanism of action of trazodone: a anti-inflammatory drugs in mice and humans. Proc Natl
multi-functional drug. CNS Spectr 14:536–546. Acad Sci USA 108:9262–9267.
Starr KR, Price GW, Watson JM, Atkinson PJ, Arban R, Watanabe N, Omori IM, Nakagawa A, Cipriani A, Barbui C,
Melotto S, Dawson LA, Hagan JJ, Upton N, Duxon MSl Churchill R, Furukawa TA (2011) Mirtazapine vs. other
(2007) SB-649915-B, a novel 5-HT1A/B autoreceptor antidepressants for depression. Cochrane Database 12:
antagonist and serotonin reuptake inhibitor, is anxiolytic CD006528.
and displays fast onset activity in the rat social interaction Watson DJ, Loiseau F, Ingallinesi M, Millan MJ, Marsden CA,
test. Neuropsychopharmacology 32:2163–2172. Fone KC (2012) Selective blockade of dopamine
Stein DJ, Ahokas A, Albarran C, Olivier V, Allgulander C D3 receptors enhances while D2 receptor antagonism
(2012) Agomelatine prevents relapse in generalized impairs social novelty discrimination and novel object
anxiety disorder: a 6-month randomized, double-blind, recognition in rats: a key role for the prefrontal cortex.
placebo-controlled discontinuation study. J Clin Psychiatry Neuropsychopharmacology 37:770–786.
73:1002–1008. Watson JM, Dawson LA (2007) Characterization of the potent
Stocking EM, Letavic MA, Bonaventure P, Carruthers NL 5-HT1A/B receptor antagonist and serotonin reuptake
(2010) Exploration of structure-activity relationships for inhibitor SB-649915: preclinical evidence for hastened onset
dual serotonin reuptake inhibitors-histamine H3 receptor of antidepressant/anxiolytic efficacy. CNS Drug Rev
antagonists. Curr Top Med Chem 10:596–616. 13:206–223.
Sun H, Kennedy PJ, Nestler EJ (2012) Epigenetics of Wong EH, Tarazi FI, Shahid M (2010) The effectiveness of
the depressed brain: role of histone acetylation and multi-target agents in schizophrenia and mood disorders:
methylation. Neuropsychopharmacology 38:124–137. relevance of receptor signature to clinical action. Pharmacol
Tasan RO, Nguyen NK, Weger S, Sartori SB, Singewald N, Ther 126:173–185.
Heilbronn R, Herzog H, Sperk G (2010) The central and Xie Z, Miller GM (2009) Trace amine-associated receptor 1 as a
basolateral amygdala are critical sites of neuropeptide Y/Y2 monoaminergic modulator in brain. Biochem Pharmacol
receptor-mediated regulation of anxiety and depression. 78:1095–1104.
J Neurosci 30:6282–6290. Yamawaki Y, Fuchikami M, Morinobu S, Segawa M,
Taylor MJ, Carney SM, Goodwin GM, Geddes JR (2004) Matsumoto T, Yamawaki S (2012) Antidepressant-like effect
Folate for depressive disorders: systematic review and of sodium butyrate (HDAC inhibitor) and its molecular
meta-analysis of randomized controlled trials. mechanism of action in the rat hippocampus. World J
J Psychopharmacol 18:251–256. Psychiatry 13:458–467.
Complementary strategies for improving the treatment of depression 1037

Zarate CA Jr., Mathews D, Ibrahim L, Chaves JF, transporter – a review of current understanding of its
Marquardt C, Ukoh I, Jolkovsky L, Brutsche NE, Smith MA, mechanism of action. Psychopharmacology 219:1–13.
Luckenbaugh D (2013) A randomized trial of a low- Zimmermann GR, Lehar J, Keith CT (2007) Multi-target
trapping nonselective N-methyl-D-asparate channel therapeutics when the whole is greater than the sum of the
blocker in major depression. Biol Psychiatry. Advance parts. Drug Discov Today 12:34–42.
online publication. doi: 10.1016/j.biopsych. Zink M, Englisch S, Meyer-Lindenberg A (2010)
Zhong H, Haddjeri N, Sánchez C (2012) Escitalopram, an Polypharmacy in schizophrenia. Curr Opin Psychiatry
antidepressant with an allosteric effect at the serotonin 23:103–111.

Downloaded from https://academic.oup.com/ijnp/article/17/7/1009/720340 by guest on 25 June 2022

You might also like