Sie sind auf Seite 1von 46

COGNITION nitive dysfunction associated with the slow

progressive timeline of AD neurodegeneration


with the cerebral atrophy occurring due to
ROBERT L. HUDKINS,
opportunistic viral, fungal, protozoal, and bac-
MICHAEL J. MARINO,
terial infections in AIDS to a common mechan-
MICHAEL WILLIAMS
Cephalon, Inc., Worldwide istic pathway is, based on present knowledge,
Discovery Research, West an optimistic stretch.
Chester, PA
1.1. Cognition
1. INTRODUCTION Cognition is defined by the Merriam-Webster
Online Dictionary as “to become acquainted
The aging process and traumatic insults to the with, know.” Cognition can also be defined as
brain frequently result in a decline in cogni- the ability to process contemporary informa-
tive performance with an associated decrease tion in the context of existing knowledge to
in the individual quality of life. At the tissue appropriately respond—in terms of decision
level, this decline involves neuronal/glial dys- making—to a given situation. In either defini-
function and/or overt neurodegeneration with tion, cognition involves several distinct beha-
marked cell loss, disruption of discrete synap- vioral domains. These include attention, per-
tic pathways, and altered neural network ception, emotion, memory, action, and pro-
function [1]. blem solving [2].
Drugs to treat cognitive dysfunction are a Therapeutic approaches to ameliorating
high priority in biomedical research as the el- cognitive dysfunction can be simplistically
derly population grows, showing an increased viewed as involving two ends of a conti-
incidence of what has been termed “benign nuum—at one end the prophylatic enhance-
senescentforgetfulness”thatprogressestomild ment of cognitive function and at the other end
cognitive impairment (MCI) and Alzheimer’s the restoration of function and/or arrest of the
disease (AD). Aspects of cognitive dysfunction decline occurring to the aging or traumatized
also occur in association with Parkinson’s dis- brain. Cognition enhancers include psychos-
ease, AIDS-associated dementia, stroke, schi- timulants such as caffeine, the most widely
zophrenia, stress, sleep deprivation, depres- ingested drug in the world in the form of coffee
sion, anxiety, recreational, prescription drug and soft drinks [3], amphetamines [4], nootro-
usage, and surgical procedures. pics such as piracetam 1, and the antioxidant,
The degree of cognitive deficit and its un- idebenone 2 [5]. Drugs currently approved for
derlying causality differs both anatomically use in the treatment of neurodegenerative
and, to the extent known, mechanistically in disorders such as AD include the cholinester-
this broad spectrum of CNS disorders, making ase inhibitors, tacrine 3, donepezil 4, rivastig-
it somewhat na€ıve to expect that new chemical mine 5, and galanthamine 6 [6] and meman-
entities acting via a single mechanism will tine 7, a partial agonist at glutamate
have efficacy in all CNS disorders that involve receptors [7]. None of these drugs works espe-
some aspect of cognitive dysfunction. The mo- cially well in treating the symptoms of AD [8]
lecular and anatomical substrates of cognition probably reflecting the advanced stage of the
and associated behaviors that include atten- disease when it is diagnosed than the intrinsic
tion and memory are complex and diffuse and efficacy of these drugs. To restore function to a
suggest that drugs addressing this area will dead cell, a “Lazarus-like” effect, is a phenom-
necessarily be polypharmic [2]. As an exam- enon that has yet to be achieved outside the
ple, given the relative ease of diagnosis of realm of fiction.
AIDS and its relatively short progression, A variety of new chemical entities (NCEs)
AIDS dementia was for a period of time viewed are currently being explored to identify new
as a surrogate disease state for testing com- cognitive enhancers. Some have discrete me-
pounds that might have potential utility in the chanistic targets against that they are/
treatment of AD. However, equating the cog- were optimized. Others involve a heuristic

15

Burger’s Medicinal Chemistry, Drug Discovery, and Development, Seventh Edition,


edited by Donald J. Abraham and David P. Rotella
Copyright  2010 John Wiley & Sons, Inc.
16 COGNITION

O O NH2
NH2 MeO (CH2)10-OH

O N
MeO Me N
O

1 Piracetam 2 Idebenone 3 Tacrine


O
O Me
O N
Me
MeO
N Me
MeO
NMe2

4 Donepezil 5 Rivastigmine

Me
N NH2

HO
O Me
H OMe Me
6 Galantamine 7 Memantine

postrationalization of a target based on a phe- the more important of which have been Phases
notypic behavioral response while yet others II and III clinical trials where approaches to
have their origins in ayurvedic medicine or reducing brain amyloid have failed to show
folklore and currently lack a robust mechan- efficacy in altering disease progression [11].
ism of action (MoA), for example, Ginko biloba Approaches to altering the hyperphosphoryla-
and the nootropics [5]. tion state of tau as a target for AD treatment
In neurodegeneration-associated cognitive are similarly complex. There are some 80
deficit, significant efforts have been focused on serine and threonine residues on tau that are
the b-amyloid [9] and tau hyperphosophoryla- potential substrates for kinase activity [10].
tion [10] hypotheses of AD that reflect the With only 30 of these functioning as phosphor-
involvement of amyloid plaques and neurofi- ylation sites under normal physiological con-
brillary tangles (NFTs), respectively, in dis- ditions and 25 of these being identified as sites
ease pathophysiology. Both are found in post- of “abnormal phosphorylation” there is signif-
mortem AD brain although whether they are icant redundancy in the ability to alter tau
causative, or a result, of the disease remains to phosphorylation. Candidate kinases for tau
be determined [11]. Prevention of b-amyloid phosphorylation are the proline-directed ki-
(Ab) formation by altering cleavage products nases, GSK3b, CDK5, and ERK2 that can
via inhibition/alteration of secretase enzyme phosphorylate 13 residues in tau associated
processing [12], prevention of Ab deposition or with proline residues. Microtubule-affinity-
enhancing its removal by the use of metal regulating kinase (MARK) and cAMP-depen-
chelators or vaccination will eventually pro- dent protein kinase A (PKA) are nonproline-
vide information on the causative role of this directed kinases that may also affect tau hy-
peptide fragment in AD. However, the path perphosphorylation. Inhibitors of “tau
forward is far from certain as initial positive kinase,” dual GSK3/CDK5 inhibitors such as
results have, more often than not, been con- the indirubicins—while effective in altering
founded by results from subsequent studies, phosphorylation of key serine and threonine
INTRODUCTION 17

sites on tau in cellular and transgenic animal that an understanding of mental processing,
systems—have yet to demonstrate robust bio- the realm of cognitive psychology [16], was
chemical or phenotypic effects in native essential for interpreting the full spectrum of
systems. behavior, and of human behavior in
One inevitable outcome from research into particular.
cognition enhancers for the potential treat- A useful framework for understanding cog-
ment of disease-associated cognitive dysfunc- nitive processes is provided by the information
tion is that of nootropics or “smart drugs.” [14] processing models of cognitive psychology.
In the same way that caffeine and ampheta- These models range in complexity from the
mines are viewed as improving normal func- black-box model of the behaviorists (Fig. 1a)
tion via their stimulant actions, newer gen- to extremely complex connectionist mod-
erations of cognition enhancers are antici- els [17]. However, common themes are appar-
pated to improve performance [14]. ent that allow the derivation of a simplified
model (Fig. 1b). Importantly, this framework
1.1.1. Domains of Cognition and Memory allows for the deconstruction of the cognitive
Historically, the understanding of cognitive process into experimentally addressable do-
processes is based on the empirical school of mains that can be studied both preclinically
psychology known as behaviorism [15]. Beha- and clinically.
viorist theories, championed most notably by Perception A detailed description of the pro-
James Watson and B.F. Skinner, evolved from cesses underlying perception is outside of the
the early learning and memory studies of scope of cognition. However, deficits in pre-
Pavlov and Thorndike. These behaviorists attentive processing and attention play sig-
were primarily concerned with defining sti- nificant roles in defining the scope and con-
mulus–response relationships and focused on sequences of the cognitive deficits observed in
precise observation and mathematical form- a number of CNS disorders including schizo-
alism that viewed the intervening processing phrenia [18]. Since these early processing
of stimuli as irrelevant as only directly obser- events are critical for subsequent cognitive
vable behaviors were believed to be amenable processing, a brief treatment is necessary.
to scientific investigation. While this ap- Preattentive processing refers to the non-
proach was successful in rigorously character- conscious events that occur at the earliest
izing certain behaviors, it was soon apparent stage of information processing. Information

(a) Stimulus Response

(b) Perception
Preattentive Working
Stimulus Attention
Processing memory

Processing Response

Long-Term
memory

Figure 1. Schematic representation of information processing models of cognitive function. (a) The black-box
model employed in behaviorist theory. Stimulus and response are quantifiable while knowledge of the inner
workings of the cognitive process is considered intractable to experimental methods. (b) The more complex
model if information processing based on probing cognitive function in both animal and human using the tools
of psychology and neurobiology. See text for explanation of labels.
18 COGNITION

enters the CNS as a sensory stream (e.g., core of cognitive processing. These processes
visual or auditory input from the eyes or ears, are significantly impaired in the aged and/or
respectively) representing a massive amount traumatized brain. Memories from years past
of data that would quickly saturate down- appear facile to recall, while events that are
stream systems. A large number of distractors more recent cannot be recalled. Whether the
exist in the environment that must be quickly problem is in the acquisition of recent mem-
filtered out of the sensory stream to pass along ories or the ability to recall these is unknown
information that is behaviorally and contex- and has considerable impact on studying the
tually relevant. This is accomplished by a problem of cognitive dysfunction from a drug
system of filters that determine the saliency discovery perspective. If a memory is not ac-
of stimuli based on temporal–spatial fre- quired, searching for drugs to recall that mem-
quency and importance [19]. The preattentive ory at a point distal to its acquisition is an
processing and filtering of the sensory stream exercise in futility.
occurs rapidly, within 100 ms of the appear- Memory is divided into two main cate-
ance of a stimulus, and has been demonstrated gories: working, or short-term memory, and
by simple visual search experiments in which long-term memory.
a subject is asked to identify a unique target in Working Memory Working memory is classi-
a field of distractors [20]. For simple targets cally described as consisting of three subcom-
and dissimilar distractors, the time required ponents: a central executive, a verbal or pho-
for target identification is independent of the nological rehearsal loop, and a visuospatial
number of distractors, indicating that a con- sketchpad [22]. The central executive func-
scious search through the field is unnecessary tion, which maintains control over voluntary
and therefore the process is preattentive. activities, is described below. Both verbal re-
Once preattentive processing filters the hearsal and visuospatial sketchpad are meth-
sensory stream to a manageable number of ods of temporarily holding information arising
stimuli that then enter into consciousness, it from the perceptive process or which is re-
is necessary that the system attends and called from long-term memory. For example,
responds to the relevant stimulus. Attention keeping a phone number in mind by verbally
is the cognitive process of selectively focus- rehearsing until the number is dialed, or ima-
ing on a relevant stimulus while ignoring gining the landmarks on a street corner when
other stimuli in the environment. This focus- relaying directions. These working storage
ing of consciousness may be an overt process elements provide a dynamic representation of
in which attention is directed at a particular both the inner and the outer world that con-
stimulus by focusing the sensory apparatus stitutes the substrates for further cognitive
on that stimulus, or covert, where attention processing. The key characteristics of working
is mentally focused independently of sensa- memory are a limited duration of only a few
tion. In addition, attention may be focused seconds, and a relatively small capacity
on cognitive processing independent of known to range between 4 and 10
sensory input. This executive attention or items [23,24]. Because of its dynamic nature,
executive function is discussed in greater working memory is subject to rapid change
detail below. and can be easily disrupted. Therefore, infor-
Attention is far from a simple linear selec- mation can only be maintained through con-
tion process as it is subject to a number of solidation into long-term memory.
complex modulatory interactions [21]. Nota- The process of memory consolidation in
bly, significant top–down processing occurs which an item in working memory is trans-
during when information in working memory, ferred into long-term memory is essential to
conscious determination of the importance of all forms of learning. Memories are consoli-
stimuli, and motor control of the sensory dated over time, and with repetition or rehear-
stream, all play a role in directing and main- sal, in a process that is facilitated by sleep [25].
taining attention. Much is being uncovered regarding the mole-
Memory The processes of information acqui- cular substrates of memory consolidation dis-
sition (learning) and recall (memory) form the cussed further below.
INTRODUCTION 19

Long-Term Memory In contrast to working cesses that take the simple inputs and storage
memory, long-term memory is static, long devices described above to create conscious
lasting, and resistant to disruption and can cognition. These are the processes that beha-
be divided into declarative and nondeclarative viorists considered intractable in terms of the
memory [26]. Nondeclarative or nonconscious scientific method, and while they have begun
memory includes items learned by nonasso- to yield to well-designed experimentation,
ciative means such as habituation or sensiti- there is still much to be learnt.
zation, innate motor and cognitive skills, and As mentioned above, the concept of a cen-
dispositions. While it is debatable as to tral executive is a necessary component of
whether these items are learned without con- working memory and consolidation required
scious input, they all clearly involve uncon- to oversee verbal rehearsal and the visuospa-
scious processing. Declarative or conscious tial sketchpad. Furthermore, there is implicit
memory is the type of memory more obviously in the concept of attention the need for a
associated with cognitive processes and can be process that directs the focusing of conscious-
further divided into semantic and episodic ness. This observation led Broadbent [28] to
memories. distinguish between automatic and controlled
Semantic memories are factual recollec- processes ultimately leading to the concept of
tions. They involve facts about the world that the central executive as a type of orchestra
are independent of a particular place or time. conductor or CEO that oversees cognitive pro-
Semantic memories include facts about the cesses (for review, see Ref. [29]).
individual, others, or shared knowledge. For The central executive oversees a variety of
example, one’s height, a friend’s birthday, or functions often termed “higher order” pro-
the capital of a particular country all are items cesses. These include process such as plan-
of semantic memory. Semantic memories are ning, goal setting, and the initiation of actions.
distinguished from episodic memories in that Furthermore, the central executive is respon-
the former are things that are “known” rather sible for planning, motivation, problem sol-
than reexperienced or “remembered.” ving, language processing, and a variety of
Episodic memories hold specific relevance other higher order functions.
in time and space and are often autobiogra- From this brief overview of the processes
phical and involve life experience. As such, involved in cognition, it is obvious that drugs
these memories are self referential and fre- to potentially improve cognitive function will
quently context dependent involving relation- target particular domains of cognition. As the
ships to times, places, other individuals and disruption of cognitive domains in disease
other events. Episodic memories are typically states will be heterogeneous with respect to
organized around particular periods of time, the underlying pathologies, it is important to
and are recalled in a manner in which the understand the different indications for which
individual mentally recreates the events. cognitive enhancing drugs are being
It should be obvious that there is a close developed.
relationship between semantic and episodic
memories. Both types of memories may con- 1.1.2. Indications and Diagnostic Criteria The
tribute to a given recall event [27] and, over Diagnostic and Statistical Manual of Mental
time episodic memories are believed to be Disorders, Fourth Edition, Text Revised
transformed into semantic memories. For (DSM-IV-TR, 2004) includes cognitive dys-
example, a memory of an event that occurred function under the general heading
several years ago may not be recalled as a “Delirium, Dementia, and Amnestic Dis-
mental re-creation of the event but rather orders.” [30] This is a broad spectrum of CNS
a knowledge of attendance at the event and disorders that includes delirium (Diagnostic
a list of related occurrences. code 293), AD (294.1), vascular dementia
Processing The box labeled processing in (290), (Delirium) and amnestic disorders/
Fig. 1 is the cornerstone of cognitive function. Other Cognitive Disorders (294). Delirium,
It represents a generalization of a number of also known as acute confusional state or re-
complex, and, to date, poorly understood pro- versible madness is a behavioral response to
20 COGNITION

widespread disturbances in cerebral metabo- that lead to the AD patient being bedridden
lism that can be caused among other events and ultimately to death. AD is a major health-
by: substance intoxication or withdrawal, care challenge with current estimates of
head trauma, dehydration, congestive heart 25–34 MM individuals being currently
failure, sleep deprivation, endocrine dysfunc- affected worldwide [11] with projections of
tion, etc. Acetylcholine (ACh) is the primary triple this number by 2050. Since diagnosis
neurotransmitter involved in delirium with of AD is exclusionary, a major challenge in
the primary neuroanatomical site being the finding effective treatments for AD is the abil-
reticular formation. The clinical abnormal- ity to diagnose the disease at a stage early
ities associated with delirium occur in the enough when drugs may act to arrest and
domains of arousal, language and cognition, potentially reverse the disease process, for
perception, orientation;, mood, sleep and wa- example, before the cells are dead and the
kefulness, and neurological functioning. De- cellular substrates for drug action are no long-
mentia involves multiple cognitive deficits er present [34]. Current biomarkers, including
that are differentiated on the basis of etiol- plasma and CSF amyloid levels and brain
ogy [30] and include dementia due to AD, imaging [31] are insufficiently robust to be
Parkinson’s disease (PD), Pick’s disease, useful in disease diagnosis [11]. In October
Huntington’s disease, HIV disease, head trau- 2008, the FDA’s Peripheral and Central Ner-
ma, vascular disease, substance-induced per- vous System Drugs Advisory Committee in
sisting, other medical conditions, and medica- reviewing the use of radionucleotide imaging
tions. Quite clearly, the ability to diagnosis the for the detection of cerebral amyloid to assist
precise form of dementia, for example, cogni- in AD diagnosis “felt that too many questions
tive dysfunction, will dictate both defining the remain about the relationship between cere-
potential molecular lesion and treatment bral amyloid and AD to diagnose AD or predict
which has a major impact on identifying viable risk of the disease based on a positive test for
drug discovery approaches and effective pa- the marker.” [35] Given that the currently
tient treatments. Evaluation of medical his- approved treatments for AD have question-
tory, physical examination, routine and spe- able efficacy [8] and the major research focus
cialized laboratory tests and brain scans [30] on the amyloid hypothesis is also in ques-
are all pertinent to diagnosis underlying a tion [11], there is considerable effort ongoing
major need for definitive biomarkers for cog- to identify drug candidates that restore brain
nitive dysfunction [31–33]. function via effects in enhancing neurotrans-
Cognition in Alzheimer’s Disease AD and its mitter release, reducing brain inflammation
variants are neurodegenerative diseases and oxidative stress and in enhancing mito-
where cell death plays a major role in the chondrial function [11]. These approaches are
cognitive dysfunction phenotype. Memory im- discussed in detail below.
pairment, difficulty in solving problems and Schizophrenia Cognitive impairment in schi-
decreases in spontaneity, reaction speed and zophrenia occurs before the manifestation of
accuracy are early signs of AD. The inability to overt psychotic symptoms, remaining severe
remember names, misplacing items, forget- through the course of the disease. Schizophre-
ting what one was about to be done and diffi- nia-associated cognitive dysfunction involves
culty in word finding (anomia) are early signs multiple domains including executive func-
of AD that are usually dismissed as signs of tion, attention, processing, vigilance, verbal
“getting older” or “needing more sleep” that learning and memory, verbal and spatial
can lead to depression, aggression, confusion, working memory, semantic memory, and so-
and wandering that can further exacerbate cial cognition [30,36]. There is a major unmet
cognitive function. The memory loss in early medical need in finding effective treatments to
stage AD is most obvious with newly acquired treat the cognitive domain of schizophrenia
memories that also lead the individual to (CDS) since it is considered to be of equal or
avoiding new, unfamiliar situations. AD can greater importance than positive or negative
progress to overt memory loss, for example, symptoms in predicting the functional conse-
dementia with accompanying physical traits quences of schizophrenia, especially in regard
INTRODUCTION 21

to work status and quality of life [37]. The US cing dopamine formation and release, redu-
federal initiative, Measurement and Treat- cing brain inflammation and oxidative stress
ment Research to Improve Cognition in Schi- and enhancing mitochondrial function.
zophrenia (MATRICS) is discussed further Stroke/Vascular Dementia Vascular demen-
below. tia is the second most common form of demen-
Parkinson’s Disease While the major clinical tia after AD and is caused by a major or multi-
features of PD reflect tremor, rigidity and ple cerebrovascular accidents (CVAs), now
bradykinesia that result from the degenera- being more commonly known as stroke or
tion of dopaminergic neurons in the locus “brain attack.” A stroke is defined by the WHO
coeruleus and substantia nigra, dementia oc- as a “neurological deficit of cerebrovascular
curs in approximately 40% of PD patients cause that persists beyond 24 hours or is
older than 70 [38]. Like AD, the cerebral cortex interrupted by death within 24 hours” and
of PD patients contains amyloid plaques, occurs when blood flow to the brain is attenu-
NFTs, and also eosinophilic Lewy bodies. The ated by the ischemia resulting from thrombo-
latter also occur in Lewy body dementia sis or an embolism or from hemorrhage. Some
(LBD), a degenerative brain disease that is four million cases of stoke occur each year
a leading cause of dementia in the elderly highlighting the need for effective thera-
population and accounts for up to 20% of all pies [40]. Memory impairment following a
dementia cases. PD is a form of LBD. Treat- stroke is accompanied by aphasia, apraxia,
ment approaches to PD-associated dementia agnosis, and deficits in executive function,
and LBD, such as those for AD are currently spasticity, ataxia, hemiparesis, and white
focused on plaque removal with additional matter lesions [30]. While t-PA is effective as
initiatives in enhancing dopamine formation antithrombolytic therapy, the search for drugs
and release, reducing brain inflammation and to treat the consequences of the excitotoxic
oxidative stress and enhancing mitochondrial insult associated with stoke has been frustrat-
function. ing due to a lack of predictive animal models
HIV-Associated Dementia Dementia asso- and compounds advanced to clinical trials that
ciated with the HIV infection in AIDS results lacked efficacy or had side effects that limited
from the decline in immune competence asso- their use or made the consequences of the
ciated with viral load. In approximately 10% of stroke worse [41,42]. As with AIDS and PD,
HIV-positive individuals, the neurological there are expectations that medications effec-
symptoms that lead to dementia reflect the tive in treating the cognitive dysfunction as-
first sign of AIDS [30]. Cognitive disorders in sociated with AD will have utility in the treat-
AIDS dementia include memory impairment, ment of the cognitive decline following stroke.
lack of concentration and confusion. These are Life Style-Associated Cognitive Dysfunction In
accompanied by apathy, depression, anhedo- addition to stress, sleep deprivation, depres-
nia, delusions, and hallucinations as well as sion, and anxiety, life style choices associated
motor dysfunction that can lead to PD-like with impaired cognitive function include re-
symptoms. Neuropathological abnormalities creational and prescription drug usage and
occur in 90% of brains of individuals that severe alcohol dependence which is the third
reflect opportunistic viral, fungal, protozoal, leading cause of dementia. The latter occurs
and bacterial infections that lead to cerebral late in life following 15–20 years of heavy
atrophy and nonspecific white matter loss [39]. drinking [30]. Additionally, the use of seda-
Additionally, several of the antiviral agents tives, hypnotics, and/or anxiolytics can lead to
used to treat AIDS have their own effect on dementia.
CNS function leading to anxiety, depression,
and confusion. In addition to antiviral therapy 1.1.3. Disease Diagnosis ADAS-COG The
to reduce the cause of the neuropathological most commonly used rating instrument in
abnormalities in AIDS, treatment approaches trials of cognition enhancers is the cognitive
to PD-associated dementia and LBD, such as subscale of the AD Assessment Scale (ADAS-
those for AD are currently focused on plaque cog) [43,44]. This scale was designed to reliably
removal with additional initiatives in enhan- identify and rate the major characteristics of
22 COGNITION

AD across a range of dysfunction from mild to widespread, multifaceted impairments in cog-


severe dementia. The ADAS-cog is a relatively nitive function, including executive function,
brief test taking approximately 30 min to com- attention, processing, vigilance, verbal learn-
plete. The test consists of 11 parts that primar- ing and memory, verbal and spatial working
ily rate memory, language, and praxis (perfor- memory, semantic memory, and social cogni-
mance of an action). Notably, nearly half of the tion. Cognitive impairment in schizophrenia
items in the ADAS-cog relate to memory mak- may be of equal or greater importance than
ing this scale a very sensitive measure of positive or negative symptoms in predicting
memory dysfunction ideally suited for the as- functional outcomes and quality of life [37].
sessment of AD patients [44]. The recognition of the prevalence of cogni-
CANTAB While the ADAS-cog is a useful tive dysfunction in schizophrenia together
instrument for the study of AD, it is limited with the realization that there was a lack of
in the domains of cognitive function assessed, a consensus as to how cognition in schizophre-
and is subject to rater errors that can be nia should be measured, led the National In-
compounded during long disease progression stitute of Mental Health to sponsor the Mea-
trials where the raters may turnover several surement and Treatment Research to Im-
times during the course of the study [45]. The prove Cognition in Schizophrenia (MATRICS)
Cambridge Neurophysiological Test Auto- initiative that has produced the MATRICS
mated Battery (CANTAB) was developed as Consensus Cognitive Battery (MCCB) [47].
a computer-based tool that has high precision, The MCCB employs 10 tests that assess per-
speed, and reliability and can produce objec- formance in 7 cognitive domains: speed of
tive feedback [46]. The battery is administered processing, attention, working memory, ver-
at a computer terminal and relies entirely on bal learning, visual learning, reasoning and
nonverbal stimuli and responses. CANTAB problem solving, and social cognition. The
measures different aspects of cognitive func- battery is intended to be a comprehensive
tion including visual memory, attention, and assessment of cognitive dysfunction in schizo-
planning. As such, CANTAB can measure phrenia, and therefore requires more than 1 h
certain aspects of executive function that are for administration. The MCCB relies on neu-
not addressed in detail in the ADAS-cog. The ropsychological tests that are standardized
CANTAB instrument was designed as a bat- and widely used making it somewhat user
tery of tests that mimic behavioral paradigms friendly for the clinician. However, it lacks
used in preclinical studies that have helped to the translational power of CANTAB in that
establish the neuronal substrates of cognitive many of the tests that have no preclinical
function. Therefore, the battery has the un- analog. In an attempt to remedy this, efforts
ique potential to not only identify cognitive are ongoing to match domains of clinical effi-
dysfunction but also indicate possible sites of cacy with appropriately predictive animal
disease action in the brain. The CANTAB has models [48].
proven to be a sensitive measure effectively
detecting cognitive dysfunction in a number of
1.2. Substrates of Cognition
disorders including AD, PD, schizophrenia,
and mood disorders, as well as the cognitive 1.2.1. Neuroanatomical Our understanding
decline associated with normal aging. of the neuroanatomical substrates of cognition
MATRICS Schizophrenia is often thought of began in the 1920s with relatively crude lesion
in terms of the positive symptoms of the dis- studies pioneered by Karl Lashley during his
order including hallucinations and paranoia. search for the elusive memory trace termed
However, it is now understood that the cogni- the engram. While Lashely’s work was influ-
tive dysfunction associated with schizophre- ential and set the stage for all future research
nia is both severe and poorly treated by cur- on the neural basis of cognition, his focus on
rent therapies. Cognitive impairment in schi- the reductionist approach led him to famously
zophrenia begins before the onset of the psy- conclude after 30 years of research that he
chosis and can worsen throughout the course discovered “nothing directly of the real nature
of the illness. Schizophrenia is associated with of the engram” and that he sometimes felt in
INTRODUCTION 23

reviewing the evidence that “the necessary rate any new memories. Interestingly, HM
conclusion is that learning just is not possi- and other patients with damage to the medial
ble.” [49]. The development of functional ima- temporal lobes also experienced a temporally
ging methods along with the ability to produce graded retrograde amnesia with more re-
discrete anatomical, pharmacological, and cently encoded memories of events prior to
molecular lesions has greatly advanced the the damage being lost, but older memories
field now known as cognitive neuroscience. being maintained. This has led to the sugges-
Neuroanatomical Substrates of Memory As tion that the process of memory consolidation
first identified in the work of Penfield [50], is a slow multicomponent process with the
focal electrical stimulation delivered during initial encoding of long-term memories resid-
brain surgery to the cortex, the temporal lobes ing in the medial temporal complex followed
in particular, can evoke strong conscious by a slow transfer to the neocortex over
memories. These studies suggested that mem- time [52].
ories are somehow stored in the cortex, possi- Neuroanatomical Substrates of Executive Func-
bly in a distributed network formed by alter- tion As described above, the central execu-
ing synaptic connections and weights that can tive function is a necessary yet poorly
be selectively activated by electrical stimula- defined function that oversees the bulk of
tion. The medial temporal lobe that includes what is considered conscious processing.
the hippocampus, entorhinal, and perirhinal While the complexity of the overall roll of the
cortex is the key brain complex responsible for central executive makes it difficult to accu-
processing information from the neocortical rately define, it is clear that it resides in
and limbic regions and integrating the infor- prefrontal cortex [55]. The prefrontal cortex
mation into a memory that encodes for various is connected to all functional units within the
aspects of an event [51]. The medial temporal central nervous system [56] implying that it
lobe interconnects with a number of brain exerts a broad controlling influence on beha-
regions including sensory cortical regions, for vior. Of particular interest when considering
example, the superior temporal gyrus and central executive function is that the prefron-
insular cortex, regions that encode for fear tal cortex has extensive interconnections with
and emotion responses, including the amyg- the dorsomedial thalamic nucleus, a key in-
dale and cingulated cortex, and executive re- tegrator of information from the thalamus a
gions of the neocortex. The medial temporal region encoding emotional states that in-
lobe is therefore ideally situated to receive cludes the amygdale and cingulate cortex and
information regarding the multiple compo- the key memory areas in the medial temporal
nents of an experience (sensory, emotional, lobe.
and cognitive) and integrating these. The The prefrontal cortex, and the dorsolateral
“bound” memory is then consolidated into prefrontal cortex in particular, is involved in
long-term memory in the neocortex [52]. the processing of goals, actions, and planning,
Perhaps the most dramatic proof of the role executive functions that operate with working
of the medial temporal lobe in the formation of memory [57]. These functions include both the
new memories comes from the classic case of ability to guide behavior by internal represen-
the patient known as HM [53] who died in tations and the ability to adapt to changes that
2008. HM suffered from severe intractable require subsequent alterations in behavior.
epilepsy that was treated by bilateral removal This cognitive flexibility, or the ability to shift
of the medial temporal lobes [54]. This proce- cognitive set, is a key aspect of central execu-
dure was performed prior to an understanding tive function that is disrupted in number of
of the role of this brain structure, and pro- neuropsychiatric disorders involving impair-
duced a surprising and remarkable change in ments in prefrontal cortical function [58].
HM’s ability to encode memory in that he Damage to the prefrontal cortex induced
developed complete anterograde amnesia. by stroke or traumatic brain injury can pro-
While appearing otherwise normal, in terms duce a variety of syndromes that underscore
of problem solving ability, IQ, and language the importance of this structure in central
comprehension, HM was unable to incorpo- executive function. Resultant symptoms
24 COGNITION

include perseverative behaviors, flat affect or inactive at normal membrane potential, but
dramatic changes in personality, a loss of allows it to open at more depolarized poten-
ability to follow internal plans or maintain tials. Induction of LTP at the Schaffer-collat-
attention, and a loss of cognitive flexibility. eral-CA1 synapse requires activation of
These same behaviors are observed in neu- NMDA receptors [62]. Strong synaptic stimu-
rological and neuropsychiatric disorders as- lation, such as those produced by a high-fre-
sociated with prefrontal dysfunction as dis- quency (100 Hz) electrical stimulation of the
cussed below. Schaffer collateral fibers, leads to sufficient
AMPA receptor-mediated depolarization of
1.2.2. Molecular Substrates of Memory As the CA1 pyramidal neurons to relieve the
discussed above, memory is believed to exist in magnesium block of the NMDA receptor. This
a distributed network created by altering sy- allows an influx of calcium through the NMDA
naptic connections and weights. The concept receptor that triggers the biochemical cas-
that neuronal activity can lead to changes in cades that lead to a persistent change in sy-
synaptic coupling efficiency was first proposed naptic efficacy. While a large number of puta-
in Hebb’s postulate [59]. Hebb proposed that tive signaling molecules have been implicated
when a given neuron excites a second neuron in this process, the exact biochemical pathway
repeatedly and persistently, some change oc- underlying LTP remains unknown [63]. How-
curs such that the efficiency of coupling be- ever, there are several key events that have
tween the two cells is increased. This activity- been identified, which provide an understand-
dependent synaptic plasticity has been the fo- ing of molecular substrates. The calcium/cal-
cus of considerable research in an attempt to modulin-dependent kinase, CAMKII appears
identify the molecular substrates of memory. central to LTP induction. CAMKII is activated
Long-Term Potentiation The finding that re- by calcium influx and autophosphorylates
petitive activation of glutamatergic synapses with LTP induction [64]. Knockout of CAMKII
in the hippocampus led to a long-lasting in- or replacement with an autophosphorylation-
crease in synaptic strength provided evidence deficient variant prevents LTP induc-
for the first potential physiological substrate tion [65,66]. Other kinases have also been
for memory in a mammals [60]. This phenom- implicated in LTP induction including
enon, long-term potentiation (LTP) has since cAMP-dependent protein kinase [67], extra-
been observed at a number synapses and has cellular signal-related kinase [68], Src ki-
been the subject of intensive research efforts nase [69], and protein kinase C [70]. The
aimed at understanding the molecular events biochemical steps linking CAMKII activation
underlying synaptic plasticity. The best stu- and autophosphorylation to the expression of
died of these synapses remains the Schaffer LTP are not fully understood. Enhanced sy-
collateral-CA1 synapse of the hippocampal naptic efficacy appears to involve an increase
formation. in postsynaptic AMPA receptors produced by
There are two major types of ionotropic a modulation of receptor trafficking [71]. In-
glutamate receptors that contribute to fast tracellular AMPA receptors are trafficked
postsynaptic response at glutamatergic sy- from recycling endosomes to the plasma mem-
napses [61] named for their selective agonists, brane by a process that requires Rab11a, a
AMPA (a-amino-3-hydroxy-5-methyl-4-isoxa- small GTP binding protein [72]. The AMPA
zole propionic acid) and NMDA (N-methyl-D- receptors interact with a family of transmem-
aspartate). AMPA receptors families are brane AMPA receptor regulatory proteins
monovalent cation channels that are gated by (TARPs) that provide an interaction with post-
glutamate and produce rapid excitatory re- synaptic density proteins (PSDs) [73] direct-
sponses. NMDA receptors are permeant to ing the AMPA receptors to their appropriate
monovalent cations and to calcium. A critical location within the synaptic membrane. CAM-
feature of the NMDA receptor channel is that KII phosphorylation of TARPs appears to be
it is blocked by extracellular magnesium in a important for LTP expression [74,75] provid-
voltage-dependent fashion. This magnesium ing a plausible link between CAMKII
block essentially renders the NMDA receptor activation, an increase in postsynaptic AMPA
INTRODUCTION 25

receptor density, and resultant enhanced sy- sent a significant challenge for drug discovery
naptic efficacy. efforts to identify therapeutically relevant
Long-Term Depression The finding that the cognitive enhancing agents. While it is rela-
Schaffer collateral-CA1 synapse and other sy- tively simple to ask a human subject if they
napses can also express long-term depression recall a particular word or number, rodents
(LTD) suggests that bidirectional control of and nonhuman primates are typically less
synaptic strength is possible [76]. LTD can be cooperative. In addition, preclinical species do
induced by a prolonged low-frequency (1 Hz) not have the capability for certain aspects of
stimulation. Interestingly, this form of LTD, cognitive function, especially those involving
like LTP is NMDA receptor-dependent and language. The importance of assay and model
relies on calcium influx suggesting that the selection and careful interpretation of results
temporal and spatial dynamics of spike timing is underscored by a large increase in preclini-
and calcium influx into the postsynaptic den- cal studies reporting cognitive enhancement
dritic spine are critical in determining the in animal models that is not reflected in the
form of plasticity expressed [77]. NMDA re- paucity of new therapeutics emerging from
ceptor-dependent LTD appears to involve the clinical trials, a relationship that may be in
activation of the protein phosphatases, PP1 part due to an underappreciation of the com-
and PP2B [78]. Subsequent calcium-depen- plexity of measuring cognitive function in pre-
dent dephosphorylation of AMPA receptors clinical species [32]. Despite these hurdles, a
and associated proteins lead to a dissociation number of assays have been developed that
of the receptors from the postsynaptic complex allow a reliable and informative assessment of
and a triggering of clathrin and dynamin-de- cognition in both rodents and nonhuman
pendent endocytosis [79]. primates.

1.2.3. Other Forms of Synaptic Plasticity This 1.3.1. Disease Models The validity of an an-
brief overview of the molecular basis of plas- imal model for any disorder can be rated on
ticity has focused on NMDA-dependent plas- three scales; predictive, construct, and face
ticity in the hippocampal CA1 region. While validity. Predictive validity focuses on how
this represents the best-studied form of sy- well results produced in the animal model are
naptic modulation, plasticity occurs at many borne out in the clinic. More often, animal
other synapses within and outside of the hip- models are back-validated using clinical
pocampus. The events may be NMDA-inde- benchmarks to provide a basis for arguing
pendent [80], have a presynaptic locus of ac- for future predictive validity. This is particu-
tion [81], be mediated and expressed by larly problematic in the field of cognition
GPCRs [82], or involve retrograde synaptic because there are few approved drugs, and
transmission [83]. Furthermore, plasticity is those that are approved have modest effi-
not solely a property of excitatory synapses cacy [85]. Construct validity concerns the
also occurring at inhibitory GABAergic sy- theoretical rationale underlying the model.
napses [84]. Therefore, memory is very likely A model with a high degree of construct va-
encoded in a distributed fashion across both lidity would disrupt the same neurotransmit-
excitatory and inhibitory synapses expressing ter systems and engage the same neuronal
multiple forms of plasticity. This suggests that circuitry as the human disorder. Unfortu-
the identification of a single molecular sub- nately, understanding of the underlying
strate of memory, or even of a particular type pathophysiology of cognitive dysfunction is
of memory may be as elusive as Lashley’s far from complete, suggesting that construct
engram. validity is difficult to ascertain. Face validity
is a measure of how accurately the model
reproduces the symptoms of the human dis-
1.3. Preclinical Behavioral Assessment of
order. As noted, face validity can be a chal-
Cognition
lenge because there are certain aspects of
The facts that most, if not all, of the disorders cognitive function that are not expressed in
discussed above are uniquely human repre- preclinical species.
26 COGNITION

Because of the problems with model valid- drug discovery research are described. While
ity, and construct validity in particular, ani- the focus is on a basic overview of the tasks, it
mal models of cognitive dysfunction typically is import to keep in mind that these assays
model some aspect of a disorder rather than exist in multiple forms and often are con-
recapitulate the human syndrome. For exam- ducted with significant procedural differences
ple, attempts at generating an AD model by between laboratories. Most of the assays de-
genetically recreating alterations in the amy- scribed can be used with short or long inter-
loid system have recapitulated certain aspects trial intervals to assess both short- and long-
of the disease including cognitive dysfunction term memories, and can be employed to assess
and altered synaptic plasticity [86], the ap- function in genetically or pharmacologically
proach has failed to produce a mouse that impaired models. Furthermore, by varying
faithfully produces plaque deposition, tangle the timing of treatment, it is often possible to
formation, neurodegeneration, and cognitive tease out the aspect of memory formation
dysfunction and recent data have questioned impacted by the treatment such as acquisition
the amyloid hypothesis [11]. or consolidation.
A large number of genetic and pharmaco- The 5-Choice Serial Reaction Time Task as a
logical models of cognitive dysfunction exist. Measure of Attention and Impulsivity In the
Some commonly used models include scopola- clinic, attention, and impulsivity are typically
mine-induced impairments to mimic choliner- measured using the continuous performance
gic dysfunction in AD [87] and phencyclidine task (CPT) in which the subject is asked to
(PCP) 8-induced impairments thought to re- attend to a stream of stimuli and respond to an
plicate NMDA-hypofunction associated with infrequent target (e.g., the number 9) while
schizophrenia [88]. A major caveat to the use withholding response to irrelevant distractors
of these models in drug discovery is the parti- (e.g., the numbers 0–8). The 5-choice serial
cular pharmacological or genetic insult used to reaction time task (5-CSRTT) has been devel-
disrupt function may not be involved in the oped as a preclinical analog of the CPT that
clinical pathology, and even if it is, may not be allows the assessment of attention and impul-
the sole cause of cognitive dysfunction. There- sivity in rodents [89]. In its simplest form, the
fore, a risk exists that an NCE will be devel- task requires the animal to attend to five small
oped with utility restricted to treatment of the openings in the wall of a test chamber and wait
animal model. Perhaps the best path for de- for one of the openings to become illuminated.
veloping novel cognitive enhancers with broad The animal responds to the illumination with
therapeutic potential is lies in testing NCEs a nose poke into the appropriate opening and
for cognitive enhancing effects in normal ani- receives a food reward. The openings are illu-
mals, or in aged animals exhibiting age-asso- minated in a random fashion, and the animal
ciated cognitive impairment. The use of such is required to respond within approximately
models requires careful attention to verifying 1 s. Errors may be measured as incorrect re-
both the presence and the function of the sponses in which the animal pokes the wrong
target in the disease state. opening, misses in which the animal fails to
respond, or premature responses in which the
1.3.2. Preclinical Cognitive Assays As men- animal responds before the illumination. An
tioned above, there are aspects of human cog- error produces a short period of darkness
nition such as verbal memory and fluency that (time-out) that serves as a negative reinforce-
cannot be modeled in preclinical species. ment. Treatments that enhance attention
Furthermore, due to the complexity and varia- would be expected to decrease the number of
bility inherent in animal studies, the assess- errors, with premature response rate serving
ment of long-term memory is typically limited as an indication of impulsivity.
to a simple increase in intertrial time that The 5-CSRTT has been used to asses the
occurs on a time scale on the order of hours proattentive properties of a number of clin-
to days rather than weeks or years. Below ical and preclinical compounds including the
some of the assays more commonly used in psychostimulant methylphenidate 9, the
INTRODUCTION 27

selective norepinephrine uptake (NET) inhi- tigation time observed after a long intertrial
bitor atomoxetine 10 [90], the H3 histamine interval.
receptor inverse agonist ciproxifan 11 [91], In the novel object recognition paradigm,
and the a4b2 nicotinic agonist ABT-418 the animal is exposed to two objects and ex-
12 [92]. ploration is assessed as in the social recogni-
Social Recognition and Novel Object Recogni- tion assay. After an appropriate intertrial
tion as Measures of Working Memory Recog- interval, the animal is reexposed to one of the
nition memory has become an increasingly original objects plus a novel object. The ani-
popular way of assessing the effect of cognitive mal’s memory of the familiar object is re-
enhancing agents in rodents. These methods flected in an increased duration of exploration
rely on the animal’s natural curiosity regard- of the novel object [94]. As with social recogni-
ing a novel stimulus. The social recognition tion memory, longer intertrial intervals lead
assay is based on the finding that social re- to a loss of memory that can be rescued with
cognition memory of adult rats for juvenile the administration of cognitive enhancing
rats decreases as the time interval between agents.
presentations of the same juvenile rats to A large number of preclinical and clinical
adult rats is increased [93]. The assay is typi- compounds produce promnesic effects in re-
cally performed by exposing an adult rat to a cognition memory models including the hista-
juvenile and measuring the time the adult mine H3 receptor inverse agonist GSK189254
spends investigating the juvenile during a 13 [95], the cholinesterase inhibitor tacrine
short (3–5 min) trial. Reexposure to the same 3 [96], and the ampakine CX-546 14 [97].
juvenile after a short intertrial interval The Morris Water Maze as a Measure of Spatial
(5–15 min) will result in significantly de- Memory Working Memory One of the most
creased investigation time suggesting that the commonly used memory tasks in preclinical
adult rat remembers and recognizes the juve- drug discovery research is the Morris water
nile. Longer (>2 h) intertrial intervals produce maze (MWM) [98]. The assay employs a large
investigation times similar to those observed circular pool filled with opaque water in which
on the initial presentation suggesting a lack of a small escape platform is hidden. A rat or
memory for the juvenile. Administration of a mouse placed in the pool for the first time will
test compound that improves working mem- swim and randomly encounter the escape
ory would be expected to decrease the inves- platform. In the simplest form of the assay,

Me
CO2Me
O
N H
N Me
N
H

8 Phencyclidine 9 Methylphenidate 10 Atomoxetine

H
N Me
O
Me
O N
N N
O

11 Ciproxifan 12 ABT-418
28 COGNITION

the animal learns over successive trials to that is divided between an open illuminated
employ spatial cues to find the platform re- side and an enclosed dark side. Rodents have a
sulting in shorter escape latencies. Treat- natural preference for dark spaces, so the will
ments that enhance spatial memory are ex- quickly move into the dark side of the cham-
pected to accelerate the learning process such ber. Entry into the dark side of the chamber is
that the escape latency decreases significantly accompanied by a foot shock. On subsequent
with fewer trials. Importantly, swim speed trials, the latency to enter the dark chamber
should be measured and should not vary with increases as the animal learns to associate the
treatment to avoid assigning cognitive enhan- shock with the context of the dark compart-
cing properties to an agent that simply pro- ment. Contextual freezing is measured by
duces hyperlocomotion. Similar information placing the animal in a box with obvious con-
on spatial learning and memory can be ob- textual cues, and administering a foot shock.
tained using other maze-based assays, for ex- The animal is later placed in the same context
ample, the radial arm maze [99], however, and the amount of time the animal remains
these methods are more complex involving the immobile (freezes) is recorded. This freezing
use of food reward while the MWM relies on behavior is a natural response in expectation
the animals native escape response. A large of receiving a foot shock. Therefore, the time
literature exists describing the effects of phar- spent in the frozen posture is related to the
macological treatment on learning in the memory for the context in which the shock
MWM. A number of clinical and preclinical occurred. Conditioned fear assays are rela-
compounds are effective in this model includ- tively facile, and therefore in common use in
ing the cholinesterase inhibitors donepezil 4 drug discovery research. However, it must be
and rivastigmine 5 [100], the 5-HT6 receptor noted that these assays are primarily measur-
antagonist SB-271046 15 [101], and the a7 ing fear and emotional memory processed
nicotinic receptor positive allosteric modula- through the amygdala that may not represent
tor, NS1738 16 [102]. other forms of memory. Pharmacological ma-
Conditioned Fear as a Measure of Contextual nipulations improve conditioned fear memory
Memory A number of conditioned fear assays including the cholinesterase inhibitor physos-
have been employed in drug discovery re- tigmine 17 [105], the 5-HT1A antagonist,
search as a test for cognitive enhancement. WAY-100635 18 [106], and the PDE4 inhibi-
The most common variants of these methods tor, rolipram 19 [107].
are the passive avoidance task [103] and con- Set shifting as a Measure of Executive Function
textual freezing [104]. In the passive avoid- A commonly used clinical test of executive
ance task, the animal is placed in a chamber function is the Wisconsin card sorting test

O O
O O
N
Me N N N
H
O

13 GSK189254 14 CX-546

Me
Cl O H NH Cl CF3
S N N
S O O
OMe N N
OH H H Cl

15 SB-271046 16 NS1738
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 29

H N
Me
N O
Me N N
O OMe N
Me
N H N O
Me

17 Physostigmine 18 WAY-100635

H
N MeO
MeO O O
O S N
NH2 MeO
O
OMe

OMe
19 Rolipram 20 Modafinil 21 Papaverine

(WCST) [108]. The test employs a series of proving attentional set shifting include mod-
cards containing shapes (stars, circles, trian- afinil 20 [111], the 5-HT6 receptor antagonist,
gles, etc.) present in different number and in SB-271046 15 [112], and the nonselective
different colors. The subject is asked to sort the PDE10 inhibitor, papaverine 21 [113].
cards and is rewarded for discovering the
appropriate sorting rule (e.g., sort by shape).
2. SMALL-MOLECULE APPROACHES TO
The experimenter then changes the rule with-
COGNITIVE ENHANCEMENT
out telling the subject. The ability to shift set
to the new rule is considered a measure of
2.1. Historical
cognitive flexibility. Patients with deficits in
prefrontal executive function, for example, Amyloid plaques, neurofibrillary tangles, and
those with schizophrenia, will exhibit diffi- neuronal loss characterize AD. This loss of
culty shifting set, and will make numerous neurons produces insufficiencies in several
perseverative errors [109]. The rodent analog neurotransmitter systems; one of the more
of the WCST is termed attentional set shift- prominent is the loss of cholinergic neurons
ing [110]. In this assay, rodents are trained to in the basal forebrain, which project into the
dig in one of the two containers to obtain a hippocampus and cortex, brain regions that
hidden food reward. The reward may be paired play an important role in memory and cogni-
with a particular odor (e.g., lavender), or a tive function. Loss of cholinergic neurons re-
particular digging medium (e.g., sawdust). sults in up to 90% reduction in the activity of
Once an animal learns to correctly identify choline acetyltransferase (ChAT) needed for
the rewarded container, the rules are chan- synthesis of ACh. These findings led to the
ged, and the animal’s performance is mea- cholinergic hypothesis of AD, some 50 years
sured. The change may be an intradimen- old, that the dementia resulted from choliner-
sional shift (e.g., the correct container was gic dysfunction and/or loss [114,115]. This
associated with lavender and is now asso- hypothesis was supported with animal studies
ciated with a clove odor), or extradimensional of cholinergic dysfunction that impaired
(e.g., the correct container was associated with learning and memory tasks. Although the ex-
lavender and is now associated with shredded act relationship between the cholinergic neu-
paper). The experimenter can then measure ron loss and dementia is not well understood,
the number of trials needed for the animal to it is accepted that the cholinergic system is
learn the new rule. Compounds active in im- involved in cognitive (attention and memory)
30 COGNITION

and noncognitive (apathy, depression, psycho- levels of AChE decrease to approximately


sis, sleep disturbances, aggression) processes 10–15% with preferential loss of the G4 iso-
of AD. Patients with MCI have increased form, while BuChE increases to approxi-
ChAT in frontal cortex and hippocampus at mately 120%. With the preferential loss of the
autopsy, suggesting it may be a compensatory G4 isoform of AChE, an ideal AChEI for treat-
mechanism to slow disease progression [116]. ing AD may require high specificity for the G1
Early treatment strategies have therefore fo- form. Inhibition of AChE results in accumula-
cused on cholinergic replacement therapy tion of ACh in the synapse, thus producing
with the development of cholinesterase inhi- effects equivalent to enhanced stimulation of
bitors (ChEIs) and cholinomimetic agents. cholinergic receptors. The AChEIs currently
approved for treatment of mild to moderate
2.1.1. Cholinesterase Inhibitors Cholinester- AD include donepezil 4, rivastigmine 5, and
ase (ChE) enzymes function to hydrolyze and galantamine 6 (Table 1). The initially ap-
terminate the action of ACh at postsynaptic proved AChEI, tacrine 3, has been removed
sites. There are two forms of the enzyme, from the market and replaced by the newer
acetylcholinesterase (AChE) and butyrylcho- compounds due to modest efficacy, significant
linesterase (BuChE, also known as pseudo- side effects and hepatotoxicity.
cholinesterase), with different CNS and PNS Tacrine Tacrine 3 was the first generation
localization. Both forms are found in the brain, AChEI being approved in 1993 for treatment
although AChE represents approximately of mild to moderate AD. It is a reversible,
95% of the ChE activity in the normal human noncompetitive inhibitor with similar potency
brain and colocalizes with cholinergic sy- for AChE and BuChE and the G1 and G4
napses. BuChE is in lower abundance in brain, isoforms. Tacrine also displays multiple bio-
primarily in glial or satellite cells, and is chemical activities, including interaction with
virtually absent in neurons. BuChE is synthe- potassium channels, inhibition of histamine
sized in the liver and highly distributed in the N-methyltransferase, and weak blockade of
plasma, hematopoetic cells, intestine, liver, muscarinic receptors. The use of tacrine
heart, and lung. Although AChE selectively was limited by poor oral bioavailability, poor
hydrolyses ACh, BuChE hydrolyses ACh, in pharmacokinetics (qid dosing), and limiting
addition to multiple xenobiotic esters. Two adverse drug reactions (nausea, diarrhea,
forms of AChE exist in the brain, a tetrameric urinary incontinence, hepatotoxicity) such
extracellular membrane-anchored G4 iso- that few patients could tolerate therapeutic
form, and a monomeric, intracellular cytoplas- doses, which led its withdrawal from the
mic G1 isoform [117]. During AD progression, market [118].

Table 1. Summary of Marketed Cholinesterase Inhibitors


Donepezil Rivastigmine Galantamine
Chemotype Piperidine Carbamate Alkaloid
Inhibition Reversible, noncompe- Pseudo-irreversible, Reversible competitive;
titive; AChE > BuChe noncompetitive; AChE > BuChE; nAChR
AChE  BuChe
Human dose (mg) 5–10 6–12 16–32
Dosage interval qd bid bid
Plasma half-life (h) 50-70 1-1.5 7
Plasma protein >90 40 10–20
binding (%)
Metabolism CYP2D6, CYP3A4 Nonhepatic esterases CYP2D6, CYP3A4
Route of elimination Parent and metabolites Metabolites in urine Unchanged parent in
in urine urine
Improvement in ADAS- 2.9 4.9 3.9
cog scale
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 31

Donepezil Donezepil 4 is a benzyl piperi- up to 7 h postdosing [122]. Preclinical studies


dine-based reversible, noncompetitive inhibi- also showed that rivastigmine had selectivity
tor approved in 1996 for treatment of mild to for central versus peripheral AChE. In the
moderate AD and dementia. It had greater clinic, rivastigmine (6–12 mg/day) was effica-
than 570-fold selectivity for human AChE over cious as assessed by the ADAS-cog, CIBIC-
BuChE and greater potency for brain over plus and activities of daily living scale com-
peripheral AChE with no inhibition in cardiac pared to placebo. No blood pressure changes
and smooth muscle. It had a long elimination were observed, and the most serious adverse
half-life (70 h), excellent brain permeability, events reported were cholinergic side effects,
and oral bioavailability (%F ¼ 100) with no agitation, nausea, anorexia, and GI
hepatotoxicity. Donezepil is metabolized in disturbances.
the liver by CYP2D6 and CYP3A4 and thus Galantamine Galantamine 6 is a tertiary
has potential for drug–drug interactions with amine alkaloid from the bulbs of the Cauca-
drugs such as fluoxetine and paroxetine. In sian snowdrop, Galanthus woronowi, ap-
clinical efficacy and safety studies evaluating proved in 2001 for treatment of mild to mod-
both short- and long-term effects, donezepil erate AD. Galantamine has a unique dual
showed significant improvement in ADAS-cog mechanism of action combining reversible,
measures (2.49 at 5 mg; 2.88 at 10 mg) and the competitive inhibition of AChE, with positive,
CIBIC-plus rating compared to placebo [119]. albeit weak, allosteric modulation of the nico-
The double blind study was followed by a blind tinic acetylcholine receptor (nAChR) [123]. It
washout phase that found, on all measures, a is the only actively marketed drug approved
decline to values not different from placebo, for AD that has demonstrated both mechan-
indicating the treatment was symptomatic in isms of action. Galantamine has been
nature. The most serious issues noted were proposed to increase ACh levels and to facil-
insomnia, agitation, nausea, and leg cramps itate glutamate, 5-HT and norepinephrine
with GI disturbances. In addition to increas- release in key brain regions involved in cogni-
ing ACh levels, donezepil may also affect cel- tion [124]. In healthy volunteers, galantamine
lular and molecular processes involved in was rapidly absorbed after oral dosing and
early stage AD pathogenesis [120]. showed high oral bioavailability (%F ¼ 100)
Rivastigmine Rivastigmine 5 is a pseudo-ir- and a plasma half-life of 7 h. The plasma
reversible carbamate type AChEI approved in protein binding was low (10–20%) with no
2000 for the treatment of mildly to moderately accumulation of drug observed after 2–6
severe AD. It is also approved for the treat- months of dosing. Galantamine is metabolized
ment of Parkinson’s dementia. Rivastigmine by CYP2D6 and CYP3A4, and thus has
is a centrally and brain region selective inhi- the potential for drug–drug interactions.
bitor that facilitates cholinergic activity in the In double-blinded placebo-controlled clinical
cortex and hippocampus [121]. Increased le- studies, galantamine showed improvements
vels of ACh were seen with rivastigmine with- in ADAS-cog, the disability assessment for
out changes in levels of NE, 5-HT, DA, and dementia (DAD), and the AD cooperative
their metabolites. Rivastigmine showed equal study/activities of daily living (ADCS/
activity for AChE and BuChE, and displayed ADL) [124].
greater potency for the G1 verse the G4 iso- Miscellaneous AChEIs Only huperzine A 22,
form of the enzyme. Since rivastigmine is an alkaloid isolated from the Chinese moss
metabolized by esterases rather than liver Huperzia serrata that has been used for cen-
cytochrome P450 enzymes, there is minimal turies in Chinese folk medicine, is currently in
potential for drug–drug interactions. It had a clinical trials [125]. It is a reversible AChEI
short plasma half-life (1–1.5 h), although the with additional weak NMDA antagonist ac-
pharmacodynamic half-life (10 h) was much tivity. Phenserine 23, a dual AChE/b-amyloid
longer due to the pseudo-irreversible, slow (Ab) protein inhibitor, had been in phase III
dissociation from the enzyme. Pharmacoki- trials to evaluate its potential to lower levels of
netic/pharmacodynamic results showed a b-amyloid precursor protein (b-APP) and Ab
6 mg dose reduced AChE activity by 50% for levels in patients with mild to moderate
32 COGNITION

Alzheimer’s disease [126]. Clinical develop- M1 receptors are postsynaptic and abundant
ment of this compound was halted when re- in cortex and hippocampus and M2 receptors
sults from a phase III trial showed no benefit are presynaptic inhibiting ACh release. At
over placebo in the primary efficacy endpoints. least 12 putative M1 receptor selective ago-
As noted retrospective meta-analyses of nists have advanced to the clinic. However,
clinical trials has indicated that none of the due to the broad tissue distribution of M1
ACHEIs works especially well in treating the receptors and dubious selectivity and efficacy,
symptoms of AD [8]. a plethora of undesirable cholinergic side ef-

Me
H
Me
H N O
N N
O Me
NH2 O N H
Me
Me

22 Huperzine A 23 Phenserine

AChEIs in Schizophrenia Current marketed fects have limited the utility of these NCEs.
AChEIs have modest benefit on cognition and Xanomeline 24, the most widely studied M1
global functioning but do not alter the course agonist had good M1/M4 potency but modest
of the disease, raising concerns about their functional selectivity over M2 and M3 recep-
cost-effectiveness [127,128]. AChEIs have tors [133]. The M1 receptor was associated
been evaluated for cognitive enhancing activ- with the procognitive effects of xanomeline,
ities in schizophrenia patients in trials to while the M4 subtype may be responsible for
broaden clinical indications [129,130]. In dou- its antipsychotic-like actions [134]. Clinical
ble blind placebo-controlled clinical trials, do- data with xanomeline reported cognitive im-
nezepil 4 and rivastigmine 5, failed to demon- provements, but with a high incidence of cho-
strate a benefit in treating cognitive deficits, linergic side effects that led to considerable
while galantamine 6 did exert a benefit for patient dropout and discontinuation of the
processing speed and verbal memory, but clinical program [135]. Cevimeline (AF-
failed to show a significant difference in global 102B) 25 is an M1/M3 agonist originally stu-
composite score [131]. Galantamine 6 differs died for the treatment of AD. It later gained
from donezepil 4 and rivastigmine 5 in its dual approval for symptoms of dry mouth in pa-
mechanism; it acts also as a positive allosteric tients with Sj€ogren’s syndrome, an autoim-
modulator of both a7 and a4/b2 nAChRs [123]. mune rheumatic disease [136]. Alvameline
The allosteric nicotinic properties of galanta- (LU-25-109T) 26 had higher affinity and a
mine 6 could lead to increased release of ACh unique M1 agonist/M2 antagonist profile, but
or indirectly affect cognition through effects on was discontinued due to adverse events in-
glutamate and DA. Galantamine enhanced cluding salivation, dizziness, GI disturbances,
DA release in prefrontal cortex and hippocam- and cardiovascular side effects [137]. AF267B
pus via allosteric modulation of nAChRs [132]. (NGX267) 27 improved cognitive symptoms,
cholinergic markers, and tau hyperphosphor-
2.1.2. Muscarinics The muscarinic receptor ylation in vivo and reportedly advanced into
family is composed of five GPCRs, M1–M5. M1, phase I for cognitive impairment in schizo-
M3, and M5 receptors are coupled with Gq phrenia and for the treatment of AD dementia.
linked to phopholipase C and are associated It is currently in Phase II for treatment of
with an increase in intracellular Ca2 þ . M2 and xerostomia. In vitro, M1 agonists including
M4 subtypes are associated with Gi subunits AF267B increase secreted APP and decrease
coupled to adenylyl cyclase. The M1 and M2 Ab levels and tau hyperphosphorylation [138].
receptor subtypes are involved in cognition. Norclozapine (ACP-104) 28, the desmethyl
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 33

metabolite of clozapine 29, a partial agonist at cess believed to be the basis of learning and
M1 and M5 receptors failed in Phase II clinical memory [60]. In neurodegeneration of AD, an
trials for cognitive deficits in schizophre- increase in extracellular glutamate leads to
nia [139]. Due to the high sequence conserva- excessive activation of NMDA receptors with
tion in the M1–M5 orthosteric binding site, consequent deficits in cognitive function and
recent interest has focused on allosteric M1 ultimately, neuronal death. Memantine 7, an
agonist to achieve improved selectivity and side orally active, weak uncompetitive NMDA re-
effect profiles [140,141]. The positive allosteric ceptor antagonist, was approved in 2002 for
modulators, VU0090157 30 and VU0029767 31 treatment of moderate to severe AD [7]. Mem-
were identified using a functional screen and antine binds to the open channel state of the
function by potentiation of agonist activation at NMDA receptor and blocks tonic pathological
the M1 orthosteric site [142]. activation induced by excessive glutamate
concentrations. Preclinically, it was shown to
2.1.3. NMDA Modulators Memantine Glu- permit physiological activation of the NMDA
tamate is the main excitatory neurotransmit- receptor while protecting against cytotoxicity
ter in the human brain and exerts its effects under conditions of chronic glutaminergic sti-
via a number of receptors, including the mulation [143]. The clinical efficacy of mem-
NMDA subtype. Under normal conditions, antine has been questioned [8] and it is in-
NMDA receptor activation results in long- creasingly being used as an adjunct to ChEI
term potentiation of neuronal activity, a pro- therapy rather than replacement.

N
Me O N N
N N
O
S N Me
Me
N
S
N
N
Me
Me
24 Xanomeline 25 Cevimeline 26 Alvameline

Me H Me
O N N

S NH N N
N N
Cl Cl
N
Me N N
H H
27 AF267B 28 Norclozapine 29 Clozapine

O
O
HO
O2N O H O
EtO N N
HN O N
H
O N Me
Me

31 VU0029767
30 VU0090157
34 COGNITION

2.1.4. Nootropics The term nootropic was and hippocampal noradrenergic mechanisms
coined by Giurgea in 1972 from the Greek noos in its cognition enhancing profile [145]. It was
(mind) and tropos (turn) to describe the prop- abandoned in Phase III. Levetiracetam 35, the
erties of the first substance, piracetam 1, S-enantiomer of etiracetam, an antiepileptic
which had positive effects in the treatment of approved in the European Union, significantly
memory loss, age related memory decline and improved cognitive function and QoL in pa-
lack of concentration. Piracetam, which has no tients with refractory partial seizures based
known mechanism of action, stimulated the on performance time on the WCST and scores
design and synthesis of a large number of on cognitive and social function [146,147].
structural “acetam” analogs that had a simi-

OMe O
NH2 Me O
O
N NH2
N O N
O O N
O
OH
NH2

32 Aniracetam 33 Oxiracetam 34 Nebracetam 35 Levetiracetam

lar pharmacological profile with modest clin-


2.2. Emerging Targets
ical benefit. Nootropics have been proposed to
work through modulation of AMPA, NMDA, 2.2.1. Neuronal Nicotinic Agonists Neuronal
and cholinergic signaling and the preclinical nicotinic acetylcholine receptors (nAChRs)
in vivo effects are normally associated with are pentameric ligand-gated cationic chan-
conditions of impaired brain function such as nels, highly expressed in the CNS. Twelve
aging, hypoxia, glucose deprivation, injury, or subunit genes, designated a2–a10 and b2–b4
neurodegeneration, for example, normal have been identified, with five additional sub-
healthy animals show little benefit from treat- units expressed in the peripheral nervous
ment with nootropics such as piracetam [5]. system (a1, b1, g, d, and e). Various subunits
Studies have suggested effects on membrane can combine to provide a diversity of receptor
fluidity and mitochondrial dysfunction could subtypes with unique brain and neuron-spe-
explain the effects of piracetam [144]. Anira- cific distribution [148,149]. The a7 and a4/b2
cetam 32 was reported as more potent than subtypes are involved in cognition and atten-
piracetam, with essentially no side effects. In a tion, and are highly expressed in brain regions
10-patient clinical trial, it demonstrated en- involved in learning and memory, including
hanced vigilance based on EEG analysis simi- the hippocampus, thalamus, and cor-
lar to piracetam (2 g p.o.). Oxiracetam 33, an tex [150,151]. nAChRs are involved directly
analog of piracetam reversed scopolamine-in- and indirectly in release of neurotransmitters
duced deficits in the radial arm maze in rats, including ACh, dopamine (DA), glutamate,
but failed to show benefit in AD patients and and norepinephrine (NE), and in AD and schi-
produced insomnia, agitation, headaches, and zophrenia, levels of cortical nAChRs are de-
occasional GI upsets in the clinic [5]. Nebra- creased [152]. In schizophrenia, a link be-
cetam (WEB-1881 FU) 34 blocked scopola- tween loss of nAChRs and sensory gating
mine-induced disruption of spatial cognition deficits has been proposed as a self-medicating
at 10 mg/kg p.o. and enhanced oxotremorine- phenomenon for the higher incidence of cigar-
induced tremors, indicating a cholinergic en- ette smoking in schizophrenia patients. One
hancing mechanism. It also decreased the D9- neurophysiological abnormality in schizo-
tetrahydrocannabinol-induced disruption of phrenia patients, P50 auditory gating deficit,
spatial cognition, suggesting additional limbic indicates an impaired information processing
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 35

and a diminished ability to filter unimportant i.p.) improved cognitive performance in the
information or repetitive sensory information. social recognition test and reversed the scopo-
These deficits can be normalized by nicotine lamine-induced performance impairment in
36 and studies indicate that the a7 subtype is the MWM (MED ¼ 30 mg/kg i.p.).
involved in the cognition [153]. Nicotine 36 is Varenicline 42, a a4b2 partial agonist/a7
not selective and its clinical utility was limited full agonist launched for treatment of smoking
by side effects including seizures, irregular cessation. It reversed nicotine withdrawal in-
heartbeat, hypertension, and GI effects in- duced deficits in learning and memory precli-
cluding nausea. DMXB-A (GTS-21) 37, a weak nically, and improved mood and cognition
a7 partial agonist, had preclinical efficacy in during smoking abstinence in the
rodent models of auditory gating [154]. An clinic [161,162]. Varenicline is being evalu-
initial proof-of-concept trial in schizophrenia ated in Phase III for CDS. The full a4/b2
involving single-day administration showed agonist ABT-418 12, showed efficacy in acute
positive cognitive effects on attention [155]. studies in AD patients, but failed to show
A second clinical trial with 4-week adminis- efficacy in a double blind, placebo-controlled
tration (75 and 150 mg bid) showed significant AD trial. Ispronicline (TC-1734; AZD-3480)
improvement on the negative symptoms but 43 [163], showed preclinical activity in several
not in the cognitive battery assessment [156]. models of cognition (step through passive
Adverse events included mild tremor and nau- avoidance, object recognition, radial arm
sea. DMXB-A generates several active meta- maze) and was well tolerated up to 320 mg in
bolites in humans that may contribute to a Phase I, but similarly failed to show benefit in
mixed pharmacological profile. A-582941 38 is Phase IIb [164]. The pyridyl ether ABT-089 44
a pyridazine a7 partial agonist with good phy- and ABT-894 45 are a4b2 agonists that ad-
siochemical and pharmacokinetic properties, vanced to Phase II for cognitive impairment in
including CNS penetration. It had good effi- AD and ADHD, respectively. ABT-089 was
cacy in several rodent and primate cognition effective in preclinical models of impaired
models thought to reflect memory and learn- cognitive function, including aging, septal le-
ing [157]. SSR-180711 39, a partial a7 agonist sion, and scopolamine-induced deficits in the
that advanced to Phase II, displayed high MWM. Clinically, it showed positive signs of
affinity for rat and human a7 (22 and 14 nM, cognitive activity in a reaction time test [165].
respectively) with 330-fold selectivity over ABT-894 was efficacious in an adult ADHD
other nAChRs. In vivo it was active in object Phase II trial, comparable with atomoxetine
recognition memory with no tolerance, in the 10. The primary endpoint was the total score
MWM, and reversed MK-801 deficits on short- of the Connors Adult ADHD Rating Scale
term memory and novelty learning [158]. TC- (CAARS). Overall, progress in this field con-
5619 40, a moderately potent a7 agonist, is one tinues to be slow despite years of research, due
of the four additional NCEs where the struc- mainly with the lack of efficacy and selectivity-
ture is known reported to be in Phase I for related side effects including emesis, motor
treatment of CDS. The selective a7 partial dysfunction, and hallucinations. Current drug
agonist JN403 41, showed rapid CNS pene- discovery directions are focused on positive
tration after oral administration in mice and allosteric modulators with the goal of im-
rats and was active in the social recognition proved selectivity and avoiding agonist-in-
test over a broad dose range [159]. The urea duced receptor desensitization [166].
analog, NS-1738 16, a positive allosteric mod-
ulator of the a7 nAChR, has shown cognition- 2.2.2. Histamine The histamine GPCR fa-
enhancing activity [160]. NS-1738 was unable mily consists of four members: H1–H4. H3Rs
to activate a7 nAChR alone. It effectively en- are expressed predominately in the brain, lo-
hanced ACh-evoked currents in cells trans- calized to the cerebral cortex, amygdala, hip-
fected with human a7 nAChR as well as in rat pocampus, striatum, thalamus and hypothala-
hippocampal neurons. NS-1738 showed little mus, where they are expressed on presynaptic
effect on desensitization kinetics of a7 terminals and function as inhibitory auto- and
nAChRs. In rats, NS-1738 (10 and 30 mg/kg heteroreceptors [167]. H3 antagonists increase
36 COGNITION

the release of various neurotransmitters, in- ABT-239 46 is currently the most widely stu-
cluding histamine, ACh, NE, 5-HT, and DA, died H3R inhibitor [168]. It is a potent H3R
and have potential utility in treating cognitive inverse agonist and effective at low doses
deficits associated with various dementias and (0.1 mg/kg sc) in a repeat trial inhibitory
schizophrenia [168]. H3R knockout mice and avoidance task, the rat social recognition mod-
H3R inhibitors demonstrate enhanced learning el of short-term memory and in a water maze
and memory in various animal models of cog- model [170]. Although ABT-239 had an im-
nitive function. A nuance of the H3R and its pressive in vivo profile for cognition enhance-
ligands is the high degree of constitutive activ- ment, its development was halted due to car-
ity in vitro and in vivo [169]. diovascular liabilities [168]. GSK189254 13
The search for H3 antagonists with drug- had high affinity for recombinant human
like properties has focused exclusively on H3R (Ki ¼ 0.9 nM) and for rat H3R blockade in
amine-based compounds as NCEs with re- vivo (ID50 ¼ 0.05 mg/kg p.o.), with greater
duced side effect liabilities have been identi- than 10,000-fold selectivity for H3 versus
fied and advanced into clinical evaluation. other receptors [95]. GSK189254, currently in

N
H
OMe
N Me N N
H
Me N N N
N MeO H

36 Nicotine 37 DMXB-A 38 A-582941

O
O H

N O Br N N O
O
H
N O Me F
N N

N 41 JN403

39 SSR-80711 40 TC-5619

H
N iPrO O
N N
N H Me
H
N N Me N Me

42 Varenicline 43 TC-1734 (AZD-3480) 44 ABT-089 (Pozanicline)

H N
N Cl

N Cl

45 ABT-894 (Sofinicline)
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 37

Me
NC
Cl
N O N

47 BF2.649
46 ABT-239

Me
N
N O N

O
CF3
48 Merck

Phase II, was efficacious preclinically across a types are current targets of interest for drug
panel of models designed to test different cog- discovery and have resulted in several clinical
nitive domains in rodent at 0.3–3 mg/kg p.o., candidates for cognition [173,174].
reversing scopolamine-induced deficits in pas- 5-HT1A 5-HT1 receptors are grouped into five
sive avoidance tasks, improving performance major subtypes and are negatively coupled to
of aged rats in a water maze model and im- adenylyl cyclase via Go/i [172,176]. Evidence
proving memory in an object recognition task. suggests that 5-HT1A antagonists may reverse
GSK239512 (undisclosed structure) is in the cognitive deficits seen in AD [176]. 5-HT1A
Phase II for the treatment of AD. BF2.649 receptors are most highly concentrated in cor-
47, despite its unusual pharmacokinetics and tical and hippocampal pyramidal neurons and
safety profile is in late stage clinical trials for provides inhibitory tone to cholinergic and
various indications, including cognitive en- glutamatergic neurons. 5-HT1A antagonists
hancement [168]. Quinazolone 48 is a com- facilitate glutamate and cholinergic transmis-
pound of further interest from Merck [171]. H3 sion [172–175]. Serotonergic neurons also pro-
antagonists continue to be an area of intense vide inhibitory tone to the cortical pyramidal
interest for drug development as the field pathway via 5-HT1A receptors. Thus, 5-HT1A
awaits clinical data [168]. antagonists may reverse AD-associated cog-
nitive deficits both by enhancing excitatory
2.2.3. Serotonin The serotonin (5-hydroxy- cholinergic and glutamate neurotransmission
tryptamine; 5-HT) 49 system originates from and by blocking direct inhibitory 5-HT in-
the raphe nucleus in the mid- and hindbrain put [176]. 5-HT1A antagonists facilitate gluta-
regions and projects to virtually all brain re- matergic activation and signal transduction
gions, including the cortex, hippocampus, by blocking the hyperpolarization and Ca2 þ
amygdala, hypothalamus, and thalamic nu- flux induced by inhibitory 5-HT tone. Except
clei. There are presently 14 5-HT receptor for clozapine 29, antipsychotics have mixed
subtypes, some of which exist as multiple results on CDS [177,178]. The atypical anti-
splice variants that are classified into 7 fa- psychotics aripiprazole, clozapine, olanza-
milies according to their primary structures, pine, and quetiapine are partial 5-HT1A ago-
signal transduction coupling, and pharmacol- nists while risperidone 50 and sertindole 51
ogy [172,173]. Except for the 5-HT3 subtype, are full antagonists [174]. WAY-100635 18
which is a ligand-gated cation channel, 5-HT was the first well-characterized, selective 5-
receptors are GPCRs. While all subtypes have HT1A antagonist [179]. The first clinical com-
been linked to learning and memory, the 5- pound from this series, lecozotan 52, was
HT1A, 5-HT2A, 5-HT4, 5-HT6, and 5-HT7 sub- a potent full antagonist with greater than
38 COGNITION

100-fold selectivity against 50 other receptors in 54 showed improvement in the ANAM but
except the D4 DA receptor, and was active in not the WCST test of cognition in patients
several models of learning and mem- stabilized with antipsychotic therapy.
ory [180,181]. It completed a Phase II/III study M100907 (volinanserin) 55, active in preclini-
in combination with donepezil in patients with cal models of schizophrenia and cognition, and
mild to moderate AD. The ability to distin- showed fewer errors in the WCST, was dis-
guish full 5-HT1A antagonists from partial continued in Phase III.

NH2
Me
HO O N N
N
N
N
H O
F
49 Serotonin
50 Risperidone

F N CN
Me
O
N N
N O N
NH
N O N O

Cl
51 Sertindole 52 Lecozotan
Me
OPO3H2 NMe2 N HO
N
N F
N OMe
H
OMe

53 Psilocybin 54 Mianserin 55 Volinanserin

agonists in the assay systems has been pro- 5-HT4 5-HT4 receptors are enriched in the
blematic and remains a key issue in develop- nigrostriatal and mesolimbic regions. They
ing antagonists. are positively coupled to adenylyl cyclase and
5-HT2A Postsynaptic 5-HT2A receptors are modulate the release of ACh, DA, GABA, and
highly localized in cortical pyramidal neurons 5-HT. The partial agonist, RS17017 56, im-
and may play a role in integrating cognitive proved performance in rodent tests of social,
and perceptual information [174]. The 5-HT2A olfactory-associated learning, and spatial
receptor is colocalized with the NR1 subunit of memory, and also improved delayed match-
the NMDA receptor, suggesting that an an- ing-to-sample responses in young and old
tagonist may potentially be beneficial in treat- primates [175]. SL650155 (capeserod) 57, a
ing cognition in schizophrenia by normalizing partial agonist with high affinity for 5-HT4
NMDA-receptor function. Limited clinical (Ki ¼ 0.4 nM) had greater than 100-fold selec-
data on 5-HT2A antagonists are available. The tivity over other 5-HT receptor subtypes.
5-HT2A agonist, psilocybin 53 produced a def- In vivo, SL650155 improved performance in
icit in a continuous performance test in rodent models of learning and memory includ-
healthy volunteers. The antagonist, mianser- ing novel object recognition (0.001–0.1 mg/kg
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 39

i.p. or p.o.), the linear maze task in aged rats for AD binds with nanomolar potency to the 5-
(0.01 and 0.1 mg/kg i.p.), and the MWM in HT6 receptor [186]. Additionally, it also inter-
mice, where it reversed scopolamine-induced acts with 5-HT7 receptors, butyryl- and acet-
deficits at 0.1 and 0.3 mg/kg i.p. SL650155 had ylcholinesterase, L-type calcium channels, the
a greater than additive effect on cognitive mitochondrial permeability transition pore,
performance in the Y-maze in combination AMPA and NMDA receptors [187]. In a cohort
with rivastigmine 5 [182]. SL650155 of 183 patients with mild-to-moderate AD,
(0.1 mg/kg s.c.) improved performance in the dimebon demonstrated a significant improve-
5-CSRT task [183] and was advanced to Phase ment in ADAS-cog and CIBIC-plus
II before being terminated. Nonetheless, 5- scores [188]. If these initial findings are sub-
HT4 receptors remain an active area in drug stantiated in the second pivotal trial, this
discovery, largely due to actions on amyloid compound will represent a major milestone
deposition, although GI side effects of agonists in AD treatment.
may ultimately limit their use. 5-HT7 The 5-HT7 receptor is the newest
5-HT6 The 5-HT6 receptor is positively member of the 5-HT GPCR family. In addition
coupled to adenylyl cyclase via Gs with ex- to depression, schizophrenia, and migraine,
pression almost exclusively in the CNS in the antagonists of the 5-HT7 receptor may find
olfactory tubercles, cerebral cortex, nucleus utility in sleep disorders and cognitive dys-
accumbens, and hippocampus. Blockade of function, although the role of the 5-HT7 re-
5-HT6 receptor function increases cholinergic ceptor in learning and memory processes is
and glutaminergic transmission and in vivo still under investigation [189]. Interest in 5-
cognitive efficacy in rodent behavior mod- HT7 receptors for cognition is based on locali-
els [174,175]. Atypical antipsychotics, such as zation in the brain (thalamus, hypothalamus,
clozapine 29 and olanzapine 58, bind with and hippocampus, with lower levels in cortex
high affinity as inverse agonists at 5-HT6 and amygdale) and behavioral pharmacol-
receptors, which coupled with its distribution ogy [189,190]. A lack of selective 5-HT7 ligands
in key brain areas involved in learning and has slowed progress with this target. The
memory has enhanced interest in identifying sulfonamide, SB-258719 62 was one of the
clinical candidates for this target [174,175]. first 5-HT7 antagonists reported. Lead opti-
Early 5-HT6 receptor antagonists had high mization produced SB-269970A 63 (Ki ¼ 1
affinity and good selectivity, but were very nM), a compound with improved affinity and
hydrophilic and had poor brain penetration selectivity. SB-269970A enhanced working
that limited their utility. The preclinical in and reference memory in a radial arm maze
vivo data on SB-271046 15 led to its being the task but had poor pharmacokinetic properties
first NCE in clinical trials, but it was discon- with low oral bioavailability [191]. SB-
tinued after Phase I due to poor brain parti- 656104A 64, a potent and selective 5-HT7
tioning [184]. Further efforts led PRX-07034 receptor antagonist had a Ki value of 2 nM
59, SB-742457 60, and SAM-315 (undisclosed with low affinity for a1B adrenoceptors
structure) advancing into the clinic. SB- (Ki ¼ 220 nM) and greater than 100-fold selec-
742457 showed clinical proof-of-concept with tivity over other 5-HT receptor subtypes [192].
results from a Phase II trial demonstrating The 5-HT7 receptor remains a target of inter-
that treatment of patients with 35 mg of SB- est in drug discovery for cognition and also as
742457 for 24 weeks resulted in a significant an antipsychotic agent to address positive
improvement in global function compared to symptoms of schizophrenia. However, better
placebo. A second Phase II trial comparing SB- NCEs are needed for clinical proof of concept.
742457 and donepezil 4 to placebo demon-
strated that patients receiving SB-742457 had 2.2.4. Dopamine Interest in the role of do-
similar improvements in global function and pamine in cognitive function has focused on
cognitive function compared to donepezil- its ability to modulate executive function,
treated patients [185]. Dimebon 61, an orally including working memory, planning, and
active NCE approved in Russia as an antihis- attention [193]. In schizophrenia, positive
taminic that has emerged as a novel treatment symptoms are hypothesized to be due to
40 COGNITION

N
N Me
O N
O N N
O N N
MeO
O
N
Cl S Me
NH2 H
O Cl
NH2

56 RS17017 57 SL650155 58 Olanzapine

Me Me
Cl
N
O
S
MeO N
O
Me
MeO N
HN
O N
Me S N NH
N
O N
H Me

59 PRX-07034 60 SB-742457 61 Dimebon

HO
Me O
O Me
S N
S
N O
O N
Me
N Me
Me
62 SB-258719 63 SB-269970A

O
S N
O
N
H N O Cl

64 SB-656104A

subcortical hyperdopamineric function, while that D1 receptors in prefrontal cortex are


cognitive deficits result from hypodopaminer- upregulated in schizophrenia due to a loca-
gic activity in the prefrontal cortex [194]. lized decrease in DA activity, and that D1
Consistent with this view are clinical reports antagonists worsen psychotic symptoms
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 41

[195]. In monkeys, DA levels in prefrontal lective NE reuptake inhibitors (SNRIs) are an


cortex increase during working memory in additional approach to elevate extracellular
a delayed alternation task and inhibition of levels of NE in the brain. These include rebox-
prefrontal DA decreases working memory etine 73, approved for treatment of depression
performance [196]. Five distinct DA receptor in Europe and purportedly under evaluation for
subtypes are known: D1-like (D1 and D5) and treatment of CDS in the United States [204],
D2-like (D2, D3, and D4). The D1 receptors and atomoxetine 10, the first nonstimulant
interact with the Gs complex to activate ade- approved for use in ADHD. In a phase II study,
nylyl cyclase, while D2 receptors interact via adjunctive atomoxetine treatment to second-
Gi to inhibit cAMP production [197]. D1 ago- generation antipsychotics showed no improve-
nists are of interest as targets for cognition, ments on prefrontal cognitive ability and func-
while D4 agonists have cognitive efficacy in tion in schizophrenics [205]. Nicergoline 74,
animal models [195,196]. The isochroman, A- an ergot alkaloid, had been used to treat
68930 65, and the benzazepine, SKF-81297 symptoms of cognitive decline in elderly pa-
66, the first full D1 agonists reported had tients with cerebrovascular insufficiencies. It
procognitive effects in animal models [196]. has a broad spectrum of activity, including
Dihydrexidine 67, under evaluation for co- a1-adrenoceptor antagonism, vasodilation and
caine dependency, improved cognitive perfor- increased arterial blood flow, and enhancement
mance in rodents and primates and was un- of cholinergic and catecholaminergic neuro-
der development for treatment of CDS. It is a transmitter function [206].
potent full D1 agonist (Ki ¼ 5.5 nM) with mod-
est 11-fold selectivity over D2 receptors and 2.2.6. Glutamate The excitatory neuro-
23-fold over a2 receptors [198]. Although safe transmitter glutamate mediates its effects
and well tolerated in man, poor oral bioavail- via both ionotropic and metabotropic recep-
ability and short half-life hindered the ad- tors. ionotropic glutamate receptors (iGluRs)
vancement of this NCE. Adrogolide (ABT- include NMDA, AMPA, and kainate sub-
431/DAS-431) 68, a prodrug of the di-phenol types. The NMDA receptor is a ligand-gated
A-86929, reversed working memory deficits ion channel composed of a combination of two
associated with chronic antipsychotic drug NR1 and two NR2 subunits, and requires
therapy in primates but was inactive in the concomitant binding of glutamate at the NR2
MWM. Adrogolide failed in clinical trials for subunit and glycine or a glycine site coagonist
PD and is still being studies for cocaine de- for activation. AMPA receptors mediate fast
pendence. The DA D4 agonist, A-412997 69 excitatory transmission in the CNS and exist
was efficacious in a social recognition test of as hetero- and homotetrameric receptors
short-term memory and in a 5-trial repeated composed of GluA1–GluA4 subunits, with
acquisition inhibitory avoidance model, while each subunit comprised of one of the two
the nonselective agonist, PD168077 70 was splice variants. NMDA and AMPA receptors
active only in short term memory [199]. A- operate in an independent, complementary
412997 increased ACh and DA levels in the fashion in controlling excitatory neurotrans-
rat medial prefrontal cortex but not in the mission. The AMPA receptor conducts pri-
dorsal hippocampus. A major issue that has marily Na þ ions, while NMDA receptors are
plagued advancement of full D1 agonists is high conductance, slow activating nonselec-
receptor desensitization. tive cationic channels that are permeable to
calcium [61,207,208]. At normal membrane
2.2.5. Norepinephrine Noradrenergic neuro- potentials, the NMDA receptor channel is
transmission in the prefrontal cortex plays a subject to voltage-dependant Mg2 þ blockade
key role in attention and cognitive proces- and its opening requires membrane depolar-
sing [200–202]. ModerateincreasesinNElevels ization by AMPA receptors. Thus, increasing
can enhance cognitive function through activa- AMPA receptor activity can increase NMDA
tion of postsynaptic a2A receptors [203]. The a2A receptor function. As discussed above,
agonists clonidine 71 and guanfacine 72 im- NMDA receptor activation is involved in
proved cognitive function in humans [202]. Se- membrane trafficking of AMPA receptors, a
42 COGNITION

H
O HO HO H
N
HO N H H
NH2 H
HO HO
HO Cl

65 A-68930 66 SKF-81297 67 Dihydrexidine

O
nPr Me
S CN N N
N O
N H
N
H N Me
AcO N
H H
H
AcO

68 Adrogolide (ABT-431) 69 A-412997 70 PD-168077

Cl H H
Cl H
N N
N NH2
N
Cl O NH
Cl

71 Clonidine 72 Guanfacine
N

OEt Br
O
O O
O MeO
N
Me
H
N
H
N
73 Reboxetine Me
74 Nicergoline

process believed to underlie the basis of neu- tivity in the CNS and are targets of current
roplasticity (LTP and LTD) [61]. interest in treating schizophrenia and cogni-
Metabotropic Glutamate Receptors (mGluRs) tive dysfunction. Group I receptors are linked
Metabotropic glutamate receptors are GPCRs to Gq and increase phosphatidylinositol turn-
comprised of eight receptor subtypes grouped over via phospholipase C activation to elevate
into three families: Group I (mGluR1, intracellular Ca2 þ . Group II and Group III
mGluR5), Group II (mGluR2, mGluR3), and receptors are located presynaptic, inhibit ade-
Group III (mGluR4, mGluR6–mGluR8). nylyl cyclase activity via Gi and modulate
mGluRs have important roles in synaptic ac- glutamate release. The potential for group II
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 43

mGluR2/3 receptor agonists to treat positive tor ADX-63365 reportedly advanced to precli-
and negative symptoms of schizophrenia has nical development for treatment of mild cog-
been established with LY2140023 75, the or- nitive impairment.

O O O O O
H S H S
HO2C CO2H
HO2C CO2H H
H NH HO2C
H NH2
MeS O H NH2
CO2H
NH2
75 LY2140023 76 LY404039 77 LY341495

O O N
F
O N N
F
N CN
N N
H
79 ADX-47273
78 CDPPB

ally active prodrug of LY404039 76 [209]. AMPA Potentiators Positive modulation of


LY404039 also increased cortical DA turnover AMPA receptors may have therapeutic poten-
in rats, an event predictive of procognitive tial in the treatment of cognitive deficits and
activity. Conversely, mGluR2/3 antagonists, potentially avoid many of the issues of direct
such as LY341495 77 reduce memory perfor- AMPA receptor activation, for example, sei-
mance. mGluR5 receptors, in additional to zures, excitotoxicity, and loss of efficacy due to
being a potential antipsychotic target, potenti- desensitization [207,208]. Ampakines are a
ate NMDA receptor currents in a number of drug class that enhance attention, alertness,
brain regions, indicating that activation of and facilitate learning and memory by allos-
this target would result in cognitive enhance- teric activation of the AMPA receptor [212].
ment. mGluRs have subtype-specific allos- AMPA receptor potentiators include the pyr-
teric sites. Positive allosteric modulation of- rolidone nootropics, for example, piracetam 1
fers several advantages over classically and aniracetam 32, benzothiazides (cyclothia-
orthosteric competitive agonists, including zide 80), benzylpiperidines (CX-516 81, CX-
subtype selectivity and lower risk of toxicity 546 14 and CX-691 82), and biarylpropylsul-
by avoiding agonist overstimulation [210]. fonamides (LY404187 83 and LY503430 84).
The pyrazole, CDPPB 78 was the first suffi- These compounds enhance cognitive function
ciently selective mGluR5 positive allosteric in rodents, which appears to correlate with
modulator for in vivo testing. CDPPB shifted increased hippocampal activity. In addition to
the glutamate-induced Ca2 þ increase fourfold directly enhancing glutamatergic synaptic
with an EC50 of 20 nM, and reduced amphe- transmission, AMPA receptor activation can
tamine-induced locomotor activity and nor- increase neurotrophin expression in vitro and
malized amphetamine-induced disruption of in vivo, which may contribute to the functional
prepulse inhibition. The oxadiazole, ADX- and neuroprotective effects of LY404187 and
47273 79 increased novel object recognition LY503430 [207,208,212]. CX-516 had been in
and reduced impulsivity in the 5-CSRT test at numerous Phase II trials for the treatment of
1 and 10 mg/kg i.p., respectively [211]. The autism, schizophrenia and AD dementia but
selective mGluR5 positive allosteric modula- was discontinued for the treatment of MCI due
44 COGNITION

to lack of efficacy. The toxicity of CX-516 lim- toms, possibly due to the presence of the anti-
ited the ability to achieve dose levels compar- psychotic agent.

O O
H
N Cl N N N
N O
O
HN N
S S N
O O NH2
O
81 CX-516 82 CX-691/ORG-24448
80 Cyclothiazide

Me O O Me O
H H
NC N S iPr N S iPr
N F O
O
Me
83 LY404187 84 LY503430

able to the efficacious doses in animal studies. Blockade of the Type 1 glycine transporter
CX-691/ORG-24448 (faramptator) 82 is being (GlyT1), a member of the sodium/chloride-de-
evaluated as an adjunct therapy for CDS [213]. pendent transporter family is responsible for
It improved short-term memory, but appeared regulation of synaptic glycine levels. Its dis-
to impair episodic memory in a group of 16 tribution mirrors that of NMDA receptor ex-
healthy elderly volunteers. Side effects in- pression, suggesting colocalization with the
cluded headache, somnolence, and nausea. NMDA receptor. The GlyT2 transporter colo-
calizes with inhibitory strychnine-sensitive
GlyT1 Glycine is a major inhibitory neuro- glycine-A receptors. Inhibitors of GlyT1 are
transmitter in the cerebellum, brainstem, and either substrate-based (sarcosine series) or
spinal cord, acting via ligand-gated strych- nonsubstrate-based compounds [218]. Sarco-
nine-sensitive glycine-A receptors. It also acts sine 88 was efficacious as an add-on therapy
as a required positive allosteric modulator of against positive, negative and cognitive sys-
glutamate by binding to the glycine-B site on tems of schizophrenia in two trials, but had
the NMDA receptor, which facilitates gluta- weak GlyT1 inhibitory activity (IC50 ¼ 38 mM)
mate binding to the NR2 subunit of the NMDA with poor pharmacokinetic properties and
complex enhancing excitatory glutamateric brain penetration, thus limiting its clinical
transmission in cortex and hippocampus [214]. utility as a drug candidate. Newer sarcosine
The NMDA receptor antagonist PCP 8 mimics analogs include NFPS (ALX-5407) 89,
the positive, negative, and cognitive symp- ORG24461 90, and ORG-25935 91. Preclini-
toms of schizophrenia in man [215]. Glycine cally, ORG-25935 elevated glycine levels and
85 was efficacious in improving negative reversed PCP-induced deficits in novel object
symptoms and some aspects of cognitive dys- recognition. It is reportedly in Phase II for the
function as an add-on therapy in schizophre- treatment of psychosis. While these potent
nia [216,217]. Furthermore, NMDA and gly- and selective inhibitors have been instrumen-
cine agonists such as D-serine 86 and D-cyclo- tal in studying the role of GlyT1 in schizo-
serine 87 improved negative symptoms in phrenia and cognition, a variety of serious
schizophrenics undergoing conventional anti- side effects such as ataxia, hypoactivity, and
psychotic therapy, with apparent decreased decreased respiration have been observed
EPS/tardive dyskinesia [215]. These studies with sarcosine-based inhibitors in ro-
demonstrated improvements in negative or dents [219]. SSR504734 92, a selective and
cognitive symptoms, but not in positive symp- reversible nonsarcosine GlyT1 inhibitor had
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 45

O O NH2
H O
H2N H2N O
OH OH N
Me OH
O N
OH
H
85 Glycine 86 D-Serine 87 D-cycloserine 88 Sarcosine

F
Me O Me O
N N
OH OH
O O

CF3

89 NFPS 90 ORG-24461
O

MeO N
Me O H
N N Cl Cl
MeO OH H N O
H O S O
Cl

CF3

91 ORG-25935 92 SSR504734 93 DCCCyB

a human GlyT1 IC50 value of 18 nM [220]. comprising a-, b-, and g- subunits in a 2:2:1
SSR103800, an analog of SSR504734 (undi- stoichiometry. Binding of GABA to the GABAA
sclosed structure) is more potent (human receptor can modulate simultaneous binding
GlyT1 IC50 ¼ 1.9 nM) and reportedly has ad- of various modulators to allosteric sites on the
vanced into the clinic. SSR103800 and ion channel complex; the most studied is the
SSR504734 were active in the social recogni- benzodiazepine (BZ) site. Positive allosteric
tion model and potentiated MK-801- and am- modulators, for example, diazepam 94, of the
phetamine-induced disruption of latent inhi- BZ site enhance the action of GABA on GABAA
bition, models believed to be predictive of receptors. Negative allosteric modulators or
positive, negative, and cognitive aspects of inverse agonists reduce GABA effects on GA-
schizophrenia, respectively [221]. Two newer BAA receptors, whereas agents that block the
nonsarcosine-based NCEs are DCCCyB 93 actions of both positive and negative allosteric
and GSK1018921 (undisclosed structure) modulators are categorized as neutralizing
that have advanced into early clinical allosteric modulators, for example, the BZ
development. antagonist flumazenil (Ro-151788) 95. BZ site
agonists produce their anxiolytic, sedative,
2.2.7. GABAA Receptor g-Aminobutyric acid anticonvulsant, and cognitive-impairing ef-
(GABA) is the major inhibitory neurotrans- fects via GABAA receptors containing b- and
mitter in the CNS. GABAA receptors are pen- g- and a1–3 or a5 subunits. Diazepam 94,
tameric GABA-gated chloride channels, com- which has been used as an anxiolytic, hypno-
posed of four transmembrane subunits (a1–6, tic, and muscle relaxant for nearly 50 years
b1–3, g1–3, d, e, u, and p) [222]. Nineteen enhanced the inhibitory effects of GABA and
GABAA receptor subunits have been identi- impair learning and memory in man [223].
fied, with the majority of receptors in the brain DMCM 96, a full nonselective inverse agonist
46 COGNITION

Me
O Me
N N
CO2Et CO2Me
MeO
N
Cl N
N
MeO
F N N
Me
O H

94 Diazepam 95 Flumazenil 96 DMCM

N N N N N O
N
O
N N
CO2Et N
N Me Me
N H Me N
Me
O Me
MeO N O

O N Me
N N
N N
Me N

97 L-655708 98 α-5IA 99

not only enhanced cognitive performance in and the potential for proconvulsant activity
rats but also produced anxiogenic and pro- of a5-inverse agonists.
convulsant activity and altered attentional
processing [224]. Pharmacological and genet- 2.2.8. Other Approaches
ic research suggests that the a5-subunit-se- Adenosine The neuromodulator adenosine
lective inverse agonists may enhance cogni- plays a major role in the regulation of synaptic
tion. Mice lacking the a5 gene show improved transmission and neuronal excitability in the
performance in the MWM, whereas perfor- CNS. Four adenosine GPCRs (A1, A2A, A2B,
mance in nonhippocampal-dependent learn- and A3) are known [228]. A1 and A3 receptors
ing and anxiety tasks was unaltered com- are coupled to the inhibitory G-proteins Gi and
pared to wild type [225]. The a5 inverse ago- Go, and A2A and A2B receptors are coupled to
nist, L-655708 97 enhanced spatial learning stimulatory Gs proteins. A1 receptors are
in the MWM [226]. The clinical candidate a- highly expressed throughout the CNS, includ-
5IA 98 robustly enhanced LTP in mouse hip- ing the cortex, hippocampus, and cerebel-
pocampal slices and performed in a rat hip- lum—important areas for cognitive function.
pocampal-dependent test of learning and A2A receptors are localized in the striatum
memory. In humans, a-5IA was toxic due to where they are coexpressed with dopamine
the formation of the hydroxymethyl isoxazole D2 receptors in GABAergic striatopallidal
metabolite that precluded its use in long- neurons and play important roles in DA neu-
term studies. In healthy volunteers, a-5IA romodulation. In contrast, A2B and A3 recep-
reversed the memory-impairing effects of al- tors have low abundance in the brain.
cohol [227]. Pyrazolotriazine 99 had a better Preclinical pharmacological and genetic
preclinical efficacy and safety profile com- studies support the involvement of adenosine
pared to a-5IA, but questions remain regard- receptors in learning and memory [3,229].
ing its overall BZ subtype selectivity profile. Adenosine modulates cognition primarily
While the field awaits clinical efficacy data to through A1 receptors, but there is now emer-
validate the GABAA subtype selective in- ging evidence for a role of A2A receptors. Ad-
verse agonist approach to cognitive impair- ministration of selective A1 receptor agonists
ment, concerns remain regarding sedation, disrupt learning and memory in rodents,
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 47

while nonselective antagonists such as caf- the dorsal root ganglia [233,234]. A large body
feine 100 or theophylline 101, or selective of evidence indicates that NGF promotes sur-
A1 (DPCPX 102) or A2A antagonists vival of basal forebrain cholinergic neu-
(ZM241835 103) facilitate rodent learning and rons [235]. NGF reversed reductions in ChAT
memory in diverse behavioral tasks [230]. and AChE activity in nucleus basalis magno-
Clinical results on the cognitive effects of caf- cellularis (NBM) lesioned rats, promoted sur-
feine in nondemented humans were inconclu- vival of septal cholinergic neurons and im-
sive [229]. Apaxifylline 104, a selective and proved learning after fimbria–fornix transec-
potent A1 antagonist (Ki ¼ 5 nM) that had 100- tion, supporting its rationale for evaluation in
fold selectivity over A2A, reversed scopola- the clinic [235]. In the clinic NGF was infused
mine-induced behavioral deficits in rats, in- intraventricularly in one patient over 3
creased vigilance and enhanced ACh release months resulting in an increase in uptake and
in cats. Apaxifylline advanced to Phase II for binding of [11 C]nicotine in the frontal and
treatment of AD but was discontinued due to temporal cortex and improved verbal episodic
its short half-life and the extensive formation memory [236]. In an alternate strategy to
of CNS active metabolites. enhance delivery of NGF to the brain by ex
A2A receptor antagonists such as istrade- vivo gene transfer, a Phase 1 trial in six pa-
fylline (KW-6002) 105 have been assessed for tients with mild AD revealed the rate of cog-
the treatment of PD acting as indirect DA nitive decline slowed by 36–51% based on
agonists [230]. They have also shown neuro- ADAS-cog and MMSE assessments, with no
protective and cognitive enhancing activ- reported adverse effects; however, the study
ity [230]. A2A receptor knockout mice were did not include a placebo group [237].
resistant to motor impairment and MPTP- Although showing encouraging results, the
induced neurotoxicity and had improved spa- therapeutic potential of polypeptides remains
tial recognition memory, while overexpression limited due to their size and pharmacokinetic
of A2A receptors resulted in working memory characteristics, which prevent their systemic
deficits in rats [231]. While istradefylline was administration for treatment of CNS diseases.
not approved for the treatment of PD due to Brain-derived neurotrophic factor (BDNF)
inconclusive Phase III trials, the selective A2A and its receptor tyrosine kinase TrkB are
antagonists SCH-420814 106, BIIB014 107, highly expressed in the hippocampus, cortex,
and SYN-115 (undisclosed structure) are in and basal forebrain and are targets for the
clinical trials for PD and have shown benefi- treatment of AD [238]. BDNF supports survi-
cial effects on cognitive-related functions in- val and differentiated function of ACh and DA
cluding motivation, attention and reward-re- neurons, and improved learning and memory
lated behavior [230]. The dual A1/A2A antago- in animals via activation of TrkB and the low
nist ASP5854 108 is under investigation to affinity NGF receptor p75 [239]. Low plasma
treat both motor disabilities and cognitive levels of BDNF mRNA have been suggested as
deficits in PD and AD [232]. As a class, the a marker for therapeutic monitoring in
main issues with development of A1 antago- AD [240]. Insulin-like growth factor I (IGF-
nists for CNS diseases have been poor water I) deficiency has also been implicated in cog-
solubility, pharmaceutical properties, poor nitive deficits seen in AD. IGF-I levels were
brain penetration, and cardiovascular side investigated for associated cognitive perfor-
effects. mance and decline, and were related to infor-
Neurotrophic Agents Neurotrophic factors mation processing speed, memory, and MMSE
are polypeptides that support the growth, dif- score [241].
ferentiation, and survival of neurons in devel- Cannabinoids The endocannabinoid system
opment and sustain neurons in the mature consists of two GPCRs, CB-1 and CB-2. CB-
adult nervous system. Nerve growth factor 1 receptors are abundant in brain regions
(NGF) has selective, survival promoting prop- associated with memory and learning, while
erties for cholinergic neurons in the CNS as CB-2 receptors are confined to cells of the
well as neurite outgrowth promoting proper- immune system. CB-1 receptor antagonists
ties on sympathetic and sensory neurons of may have therapeutic utility for the treatment
48 COGNITION

O R O
Me nPr NH2
N N N NH

N N
HO N N N
O N O N
Me nPr N N N O
H

100 Caffeine R = methyl 102 DPCPX 103 ZM241835


101 Theophylline R = H

O O
nPr Me
NH O Et
N N N OMe

O N N N OMe
O N
nPr Et

104 Apaxifylline 105 KW-6002 (Istradefylline)

NH2
O N N N
N OMe
N N
N
N
O

106 SCH-420814

O
O
N N iPr
N N
N
N H2N N
H2N N
F
Me

NH2 108 ASP5854


107 BIIB-014

of cognitive deficits associated with AD or ceptor antagonists as drugs is confounded by


schizophrenia [242]. The CB-1 antagonist ri- the psychiatric side effects associated with
monabant (SR141716) 109 improved olfactory Rimonabant use that has led to its non-
short-term memory assessed by the social approval as an antiobesity agent in the United
recognition test and enhanced spatial memory States and its withdrawal from the market in
in the radial-arm maze task in rodents. In the European Union for the same
addition, rimonabant reversed amnesia in- indication [244].
duced by i.c.v injections of b-amyloid frag- Neuropeptides Neuropeptides and their re-
ments in mice. In vivo rimonabant selectively ceptors have represented novel targets for
increased NE, DA, and ACh efflux in the pre- CNS disorders for more than half a century,
frontal cortex, suggesting a potential role for with minimal success as evidenced by the
CB-1 antagonists in treatment of attention inability to find improvements on morphine
and ADHD [242,243]. The future of CB-1 re- as analgesic acting via the opioid receptor
SMALL-MOLECULE APPROACHES TO COGNITIVE ENHANCEMENT 49

Me O
N N
N N
Cl
N H
N O
Me N
Cl Me
Cl
O
Cl
Cl

109 Rimonabant 110 Osanetant

Me

Et

N O

OH SO2
O2S
N

111 Talnetant 112 RWJ-57408

family and the failure of neurokinin-1 (NK-1) pounds suffer from poor pharmacokinetics
antagonists as analgesics and antidepres- and were abandoned [247].
sants. Nonetheless, several neuropeptides or GAL and its receptors (GALR1–GALR3)
their inhibitors including neurokinin B, an- are distributed in basal forebrain, cortex, hip-
giotensin IV, galanin (GAL), adrenocortico- pocampus, and amygdala, where it modulates
trophic hormone (ACTH), oxytocin (OT), argi- ACh, NE, and 5-HT pathways [248]. GAL
nine vasopressin (AVP), and thyrotrophin-re- inhibits ACh release in vitro and in vivo, and
leasing hormone (TRH) have shown efficacy in impairs cognitive performance in models of
cognition models. spatial learning and memory [249]. Mice over
The neurokinins (NKs) are a family of three expressing GAL have selective search and
neuropeptides, substance P (SP), neurokinin spatial navigation deficits with impaired
A (NKA), and neurokinin B (NKB), that med- learning and memory. The GALR1 antago-
iate their biological effects via activation of nist, RWJ-57408 112, reversed the GAL-in-
NK1, NK2, and NK3 GPCRs, respec- hibited release of ACh in vitro [250] and the
tively [245]. NK3 receptors are expressed in selective peptide antagonists, M35 and M40,
brain regions involved in emotion and cogni- reversed GAL-induced deficits in various mod-
tion, and stimulation of NK3 receptors can els of learning and memory in rats [248]. Ta-
enhance DA, NE, 5-HT, GABA, and ACh re- ken together, the data suggest GALR1 as a
lease. Antagonists may have therapeutic va- promising target for cognitive deficits in AD.
lue in treating psychosis and CDS. NK3 However, to date, high-throughput screening
knockout mice displayed deficits compared to and drug discovery efforts have failed to iden-
wild-type mice in several cognition tests, in- tify potent, drug-like NCEs for this target.
cluding passive avoidance, acquisition of con- ACTH modulates cognition and attention
ditioned avoidance responding and in humans. ACTH4–10 is a potent modulator
MWM [246]. Two NK3 antagonists, osanetant of attention in humans. It may also have
110 and talnetant 111 displayed antipsycho- neurotrophic or neuroprotective properties,
tic activity the clinic; however, both com- and was studied for the treatment of memory
50 COGNITION

disturbances in AD [251]. ORG2766 (H-Met affect, with modest improvement in semantic


(O2)-Glu-His-Phe-D-Ly-Phe-OH) enhanced memory [258].
recovery in behavioral models of forebrain The successful transition of peptide hor-
lesioned animals and ebiratide, an analog of mones to small-molecule mimetics or antago-
ACTH4–9, increased ChAT and AChE activ- nists remains a major challenge in CNS drug
ities in aged rats, suggesting potential ther- discovery perhaps due to the fact that as labile
apy in AD [252]. peptides, these neuromodulators have very
Angiotensin IV (Ang IV), initially thought short half-lives whereas small molecules may
to be an inactive product of Ang II degrada- be too long lasting in their effects to be effica-
tion, enhanced learning and memory in nor- cious and side effect free. Similarly, antago-
mal rodents and reverse memory deficits in nists may provide prolonged blockade of auto-
models of amnesia. The CNS effects are crine signaling that is deleterious to tissue
mediated by the AT4 receptor, found in high function. As examples, NK1 receptor antago-
levels in brain regions involved in cognition. nists MK-869 (Aprepitant) 113 and CP-
The AT4 receptor was identified as the trans- 122721 114 despite good drug-like properties
membrane enzyme, insulin-regulated mem- and copious preclinical efficacy, data have
brane aminopeptidase (IRAP). Inhibition of singularly failed in the clinic. Likewise the
IRAP via the AT4 receptor may inhibit degra- considerable efforts to improve on morphine
dation of neuropeptides involved in cognitive as an analgesic have yet to yield viable NCEs
enhancement [253]. that provide convincing evidence that opiod
There is growing body of evidence that the receptor subtype selectivity will avoid the side
neuropeptides OT and AVP modulate complex effects of morphine and its congeners includ-
social behavior and social cognition [254,255]. ing addiction, respiratory depression, consti-
OT knockouts display social amnesia in the pation, and euphoria.

CF3
MeO
Me

O O CF3
NH OCF3

N
H N
N H
O F
N N
H
113 MK-869 114 CP-122721

social recognition test, despite a normal ability Phosphodiesterase Inhibitors Phosphodies-


to recognize familiar nonsocial scents. The def- terases (PDEs) function to hydrolyze the phos-
icit in social recognition can be completely re- phodiester bond and degrade the key second
versed with OT infusion [256]. AVT increased messengers, cyclic adenosine monophosphate
glutamate release and intracellular Ca2 þ con- (cAMP), and cyclic guanosine monophosphate
centration in hippocampal and cortical astro- (cGMP) to control their intracellular le-
cytes and blocked Ab-induced impairment of vels [259]. At least 11 distinct PDE families
LTP in rat hippocampus in vivo [257]. (PDE1–PDE11) are known that are classified
In addition to its role in regulating thyroid by their substrate specificity. PDE3, PDE4,
function, TRH also modulates ACh activity in PDE7, and PDE8 are specific for cAMP; PDE5,
the CNS. TRH administration (0.3 mg/kg i.v.) PDE6, and PDE9 are cGMP selective en-
in 10 AD patients showed statistically signifi- zymes; and PDE1, PDE2, PDE10, and PDE11
cant increases in arousal and improvement in function as dual-substrate PDEs [259].
SMART DRUGS 51

Essentially all PDEs are expressed in the consolidation in rodent novel object recogni-
CNS with evidence suggesting that PDE2, tion tasks and reversed scopolamine-induced
PDE4, PDE5, PDE9, and PDE10 may have deficits in performance in a T-maze task [270].
therapeutic potential for cognitive disorders. PDE9A is a cGMP-specific PDE widely dis-
A substantial body of genetic and pharma- tributed in the CNS. The selective PDE9 in-
cological evidence demonstrates that the hibitors, BAY 73-6691 119 enhanced long-
cAMP response element binding (CREB) term memory in social recognition and object
protein is a required process in formation recognition tasks and attenuated scopola-
of long-term memory [260,261]; lower levels mine-induced deficit in passive avoidance and
of CREB result in memory impairment, MK-801-induced deficit in a T-maze alteration
while higher levels facilitate memory forma- task [271]. BAY 73-6691 is reportedly under
tion [262,263]. cAMP signaling through development for AD [265]. PDE10A is widely
PDE4 inhibition has been associated with expressed in brain regions associated with DA
consolidation and retention of LTP. The and glutamate function. Increasing interest in
prototype PDE4 inhibitor rolipram 19 facili- the discovery of PDE10A inhibitors stems
tated LTP in the hippocampus and in- from reports of the antipsychotic activity of
creased phosphorylation of CREB and ex- papaverine 21 and the potential for PDE10A
pression of the cAMP-dependent, memory- inhibitors to treat CDS [265,272]. Papaverine
related protein, Arc [264]; changes linked to enhanced CREB phosphorylation, reversed
retention of long-term memory. In vivo, ro- PCP-induced deficits in the EDID-set shift
lipram reversed scopolamine deficits in ob- assay, a test of executive function, and was
ject recognition and radial arm maze efficacious in the novel object recognition as-
tasks [265]. PDE4 inhibition and activation say [273,274]. PF-02545920 120, a selective,
of cAMP/CREB signaling cascade appear to picomolar PDE10A inhibitor, reportedly en-
specifically facilitate long-term, but not tered Phase II clinical trials for
short-term memory formation [264]. HT- schizophrenia [275].
0712 115, MK-0952 116, and MEM1414
(undisclosed structure) are PDE4 inhibitors
that reportedly advanced to the clinic. The 3. SMART DRUGS
primary obstacle with the development of
PDE4 inhibitors has been dose-limiting side As noted in the introduction [14], a natural
effects of emesis and nausea. The four PDE4 extension of the use of drugs intended to treat
subtypes (PDE4A–PDE4D) are differentially cognitive impairment in disease states such as
expressed in the CNS [266]. PDE4 knockout AD, PD, schizophrenia, and ADHD and in
and behavioral studies hypothesized that situations following brain trauma and stroke,
the PDE4D isozyme was specifically in- is the use of such agents to improve cognitive
volved in emesis [267] and PDE4B impli- performance and quality of life in healthy
cated in the regulation of LTP in the individuals. Stimulant compounds such as
hippocampus [268]. caffeine 100 (in coffee and soft drinks), am-
The PDE2A expression pattern in CNS phetamine 121, methylphenidate 9, and mod-
stimulated interest in the study of PDE2 in- afinil 20 are frequently used by students to
hibitors for treatment of cognitive disorders. improve examination performance, by shift
BAY 60-7550 117 (PDE2A IC50 ¼ 4.7 nM) had workers (including those in the medical pro-
selectivity versus PDE3B, PDE7B, PDE8A, fessions), airline crews, and by active military
PDE9A, PDE11A, and PDE1, increased cGMP personnel [14]. Their potential use in airport-
levels in culture and enhanced LTP in hippo- security screeners has also been suggested.
campal slices [269]. BAY 60-7550 improved This has led to an ethical debate on the use
performance in social and object recognition of cognition enhancers to improve “brain en-
memory tasks, and reversed MK-801-induced ergy” and overcome the effects of sleep depri-
deficits in T-maze spatial alteration. Although vation in “an overworked 24/7 society pushed
PDE5 expression in the brain is low the PDE5 to the limits of human endurance.” [14] In the
inhibitor, sildenafil 118 facilitated memory United States, the nonprescription use of the
52 COGNITION

O O
H
N N
MeO O
N N
O
F
Me
CO2H
115 HT-0712

116 MK-0952
O Me
H
N O Me
N
N H N
N O N
N
Me S
N N
O
O N
MeO Me OEt
Me
OMe
117 BAY 60-7550 118 Sildenafil

O N
H
Me N Me
N
N N N NH2
CF3 O
Cl
N N
Me

119 BAY 73-6691 120 PF-02545920 121 Amphetamine

stimulants amphetamine and methylpheni- in the healthy with the argument that these
date is a crime [276] creating a black market modalities may in time be viewed in the same
in their sale. Yet questions have been raised as prophylatic manner as vitamins or vaccines.
to whether the responsible use of cognition
enhancers in the healthy is actually a “good
health habit” and how, in these circum- 4. SUMMARY AND FUTURE CHALLENGES
stances, taking a prescription psychostimu-
lant differs from consuming a double espresso. The need for drugs to treat cognitive dysfunc-
Indeed, the increased consumption of coffee tion in all its forms has been clearly estab-
may be related to the fact that an 8 oz cup of lished and represents a major challenge in
Starbucks coffee contains nearly three times drug discovery given the many failures of
the caffeine as a regular coffee [277]. Modern- compounds robustly active in animal models
day society is excessively sleep deprived, of attention and memory in the clinic. It may
stressed, and overloaded with information via be argued that nearly every class of NCE
the Internet such that quality of life is at a active on CNS targets will, at one point or
premium. Accordingly, there have been another, be examined for effects on cognitive
calls [276,278] for further research to generate function. Similarly, the lack of clinical success
an evidence base on the risk and benefits may be argued as reflecting either the inade-
associated with the use of cognition enhancers quacy of the animal models used to triage CNS
REFERENCES 53

compounds for advancement to the 17. Mai TV, Cleeremans A. Trends Cog Sci 2005;9:
clinic [32,279] or the inability to diagnose 397–404.
patients at a sufficiently early stage in their 18. Geyer MA. Neurotox Res 2006;10:211–220.
disease progression to show benefit [11]. 19. Koch C, Ullman S. Hum Neurobiol 1985;4:
With an understanding of these limitations, 219–227.
a better temporal and informational interface 20. Smilek D, Frischen A, Reynolds MG, Gerritsen
between preclinical and clinical research activ- C, Eastwood JD. Percept Psychophys 2007;69:
ities [280] and the use of appropriate biomar- 1105–1116.
kers, it may be anticipated that newer com- 21. Knudsen EI. Ann Rev Neurosci 2007;30:57–78.
pounds advanced to the clinic may show im- 22. Repovs G, Baddeley A. Neuroscience 2006;139:
proved efficacy and patient benefit and add 5–21.
significantly to the quality of life in patients 23. Miller GA. Psychol Rev 1956;63:81–97.
with currently untreatable neurodegenerative 24. Cowan N. Behav Brain Sci 2001;24:86–14.
and traumatic disorders of the brain. 25. Stickgold R. Nature 2005;437:1272–1278.
26. Tulving E.In: Tulving E, Donaldson W, Ower
REFERENCES GH, editors. Organization of Memory. New
York: Academic Press; 1972. p 381–403.
1. Uhlhass PJ, Singer W. Neuron 2006;52: 27. Westmacott R, Black SE, Freedman M, Mos-
155–168. covitch M. Neuropsychologia 2004;42:25–48.
2. Squires LR, Kandel ER. Memory: From Mind 28. Broadbent DE. Psychol Rev 1957;64:205–215.
to Molecules. W.H. Freeman; 2000. 29. Goldberg E, The Executive Brain. New York:
3. Fredholm BB, B€ attig K, Holmen J, Nehlig A, Oxford University Press; 2001.
Zvartau EE. Pharmacol Rev 1999;51:83–133. 30. First MB, Tasman AC. DSM-IV-TR Mental
4. Barch DM, Carter CS. Schizophr Res 2005;77: Disorders: Diagnosis, Etiology and Treatment.
43–58. Chichester, UK: Wiley; 2004. p 263–309.
5. Gouliaev AH, Senning A. Brain Res Rev 1994; 31. Shaw LM, Korecka M, Clar CM, Lee WM-Y,
19:180–222. Trojanowski JQ. Nat Rev Drug Discov 2007;
6:295–303.
6. Birks J. Cochrane Database Syst Rev 2006;1:
CD005593. 32. Day M, Balci F, Wan HI, Fox GB, Rutkowski
JL, Feuerstein G. Curr Opin Invest Drugs
7. Rogawski MA, Wenk GL. CNS Drug Rev 2003;
2008;9:696–707.
9:275–308.
33. Chin A-V, Robinson DJ, O’Connell H, Hamil-
8. Raina P, Santaguida P, Ismaila A, Patterson C,
ton F, Bruce I, Coen R, Walsh B, Coakley D,
Cowan D, Levine M, Booker L, Oremus M. Ann
Molloy A, Scott J, Lawlor BA, Cunningham CJ.
Intern Med 2008;148:379–397.
Age Ageing 2008;37:559–564.
9. Castellani RJ, Lee H-G, Zhu X, Nunomura A,
34. Vellas B, Andrieu S, Sampaio C, Coley N, Wil-
Perry G, Smith MA. Acta Neuropathol 2006;
cock G. Lancet Neurol 2008;7:436–450.
111:503–509.
35. News in brief. Nature Rev Drug Disc 2008;7:
10. Mazanetz MP, Fischer PM. Nat Rev Drug
964–965.
Discov 2007;6:464–479.
36. Green MF. Ann Rev Clin Psychol 2007;3:
11. Williams M. Curr Opin Invest Drugs 2009;10:
159–180.
19–30.
37. McGurk SR. J Psychiatr Pract 2006;6:
12. Olson RE, Marcin LR. Ann Rep Med Chem
190–196.
2007;42:27–47.
38. Kaplan H, Saddock B, Grebb J. Kaplan and
13. Schneider A, Mandelkow E. Neurotherapeu-
Saddock’s Synopsis of Psychiatry. 7th ed. Bal-
tics 2008;5:443–457.
timore MD: Williams & Wilkins; 2003.
14. Sahakian B, Morein-Zamir S. Nature 2007;
39. Gray F, Adle-Biassette H, Chretien F, Lorin de
450:1157–1159.
la Grandmaison G, Force G, Keohane C. Clin
15. Thompson RF. Psychology Rev 1994;101: Neuropathol 2001;20:146–155.
259–265.
40. Zaleska MM, Mercado ML, Chavez J, Feuer-
16. Albright TD, Kandel ER, Posner MI. Curr Opin stein GZ, Pangalos MN, Wood A. Neurophar-
Neurobiol 2000;10:612–624. macology 2009;56:329–341.
54 COGNITION

41. Hall ED. Comp Med Chem II 2007;6:253–277. 65. Silva J, Paylor R, Wehner JM, Tonegawa S.
42. O’Collins VE, Macleod MR, Donnan GA, Hor- Science 1992;257:206–211.
ky LL, van der Worp BH, Howells DW. Ann 66. Giese KP, Fedorov NB, Filipkowski RK, Silva
Neurol 2005;59:467–477. AJ. Science 1998;279:870–873.
43. Rosen WG, Mohs RC, Davis KL. Am J Psychia- 67. Greengard P, Jen J, Nairn AC, Stevens CF.
try 1984;141:1356–1364. Science 1991;253:1135–1138.
44. Mohs RC, Rosen WG, Davis KL. Psychophar- 68. English JD, Sweatt JD. J Biol Chem 1996;
macol Bull 1983;19:448–450. 271:24329–24332.
45. Connor DJ, Sabbagh MN. J Alzheimer Dis 69. Lu YM, Roder JC, Davidow J, Salter MW.
2008;15:461–464. Science 1998;279:1363–1367.
46. Sahakian BJ, Owen AM. J Royal Soc Med 70. Malenka RC, Madison DV, Nicoll RA. Nature
1992;85:399–402. 1986;321:175–177.
47. Nuechterlein KH, Green MF, Kern RS, Baade 71. Malenka RC. Ann NY Acad Sci 2003;
LE, Barch DM, Cohen JD, Essock S, Fenton 1003:1–11.
WS, Frese FJ, Gold JM, Goldberg T, Heaton 72. Park M, Penick EC, Edwards JG, Kauer JA,
RK, Keefe RS, Kraemer H, Mesholam-Gately Ehlers MD. Science 2004;305:1972–1975.
R, Seidman LJ, Stover E, Weinberger DR, 73. Nicoll RA, Tomita S, Bredt DS. Science 2006;
Young AS, Zalcman S, Marder SR. Am J Psy-
311:1253–1256.
chiatry 2008;165:203–213.
74. Tsui J, Malenka RC. J Biol Chem 2006;
48. Geyer MA. Neurotox Res 2008;14:71–78. 281:13794–13804.
49. Lashley KS. Symp Soc Exp Biol 1950;4: 75. Tomita S, Stein V, Stocker TJ, Nicoll RA, Bredt
454–482. DS. Neuron 2005;45:269–277.
50. Penfield W. AMA Arch Neurol Psychiatry 76. Dudek SM, Bear MF. Proc Natl Acad Sci USA
1952;67:178–198.
1992;89:4363–4367.
51. Lipton PA, Eichenbaum H. Neural Plasticity 77. Dan Y, Poo MM. Physiol Rev 2006;86:
2008;2008: 258467. 1033–1048.
52. Squire LR, Alvarez P. Curr Opin Neurobiol 78. Mulkey RM, Herron CE, Malenka RC. Science
1995;5:169–177. 1993;261:1051–1055.
53. Corkin S. Nature Rev Neurosci 2002;3:
79. Ashby MC, De La Rue SA, Ralph GS, Uney J,
153–160. Collingridge GL, Henley JM. J Neurosci 2004;
54. Scoville WB, Milner B. J Neurol Neurosurg 24:5172–5176.
Psychiatr 1957;20:11–21. 80. Harris EW, Cotman CW. Neurosci Lett
55. Goldman-Rakic PS. Philos Trans R Soc Lond 1986;70:132–137.
1996;351:1445–1453.
81. Nicoll RA, Malenka RC. Nature 1995;377:
56. Nauta WJ. Acta Neurobiol Exp 1972;32: 115–118.
125–140. 82. Jin Y, Kim SJ, Kim J, Worley PF, Linden DJ.
57. Petrides M. Exp Brain Res 2000;133:44–54. Neuron 2007;55:277–287.
58. Hanes KR, Andrewes DG, Pantelis C. J Int 83. Gerdeman GL, Ronesi J, Lovinger DM. Nature
Neuropsychol Soc 1995;1:545–553. Neurosci 2002;5:446–451.
59. Hebb DO. The Organization of Behavior: A 84. Nugent FS, Kauer JA. J Physiol 2008;586:
Neuropsychological Theory. New York: John 1487–1493.
Wiley & Sons; 1949. 85. Raina P, Santaguida P, Ismaila A, Patterson C,
60. Bliss TV, Lomo T. J Physiol 1973;232: Cowan D, Levine M, Booker L, Oremus M. Ann
331–356. Intern Med 2008;148:379–397.
61. Dingledine R, Borges K, Bowie D, Traynelis 86. Jacobsen JS, Wu CC, Redwine JM, Comery TA,
SF. Pharmacol Rev 1999;51:7–61. Arias R, Bowlby M, Martone R, Morrison JH,
62. Harris EW, Ganong AH, Cotman CW. Brain Pangalos MN, Reinhart PH, Bloom FE. Proc
Res 1984;323:132–137. Natl Acad Sci USA 2006;103:5161–5166.
63. Sanes JR, Lichtman JW. Nat Neurosci 1999;2: 87. Patel S, Tariot PN. Psychiatr Clin North Am
597–604. 1991;14:287–308.
64. Fukunaga K, Muller D, Miyamoto E. J Biol 88. Amitai N, Semenova S, Markou A. Psycho-
Chem 1995;270:6119–6124. pharmacology 2007;193:521–537.
REFERENCES 55

89. Robbins TW. Psychopharmacology 2002;163: 106. Misane I, Ogren SO. Neuropsychophamacol-
362–380. ogy 2003;28:253–264.
90. Navarra R, Graf R, Huang Y, Logue S, Comery 107. Egawa T, Mishima K, Matsumoto Y, Iwasaki
T, Hughes Z, Day M. Prog Neuropsychopharm K, Iwasaki K, Fujiwara M. Jpn J Pharmacol
Biol Psych 2008;32:34–41. 1997;75:275–281.
91. Day M, Pan JB, Buckley MJ, Cronin E, Hol- 108. Eling P, Derckx K, Maes R. Brain Cogn 2008;
lingsworth PR, Hirst WD, Navarra R, Sullivan 67:247–253.
JP, Decker MW, Fox GB. Biochem Pharmacol 109. Everett J, Lavoie K, Gagnon JF, Gosselin J
2007;73:1123–1134. Psychiatry Neurosci 2001;26:123–130.
92. Hahn B, Sharples CG, Wonnacott S, Shoaib M, 110. Birrell JM, Brown VJ. J Neurosci 2000;20:
Stolerman IP. Neuropharmacology 2003;44: 4320–4324.
1054–1067. 111. Goetghebeur P, Dias R. Psychopharmacology
93. Thor DH, Wainwright KL, Holloway WR. Dev 2008; PMID:18392753.
Psychobiol 1982;15:1–8.
112. Rodefer JS, Nguyen TN, Karlsson JJ, Arnt J.
94. Ennaceur A, Cavoy A, Costa JC, Delacour J. Neuropsychopharmacology
Behav Brain Res 1989;33:197–207. 2008;33:2657–2666.
95. Medhurst D, Atkins AR, Beresford IJ, Brack- 113. Rodefer JS, Murphy ER, Baxter MG. Eur J
enborough K, Briggs MA, Calver AR, Cilia Neurosci 2005;21:1070–1076.
J, Cluderay JE, Crook B, Davis JB, Davis
114. Bartus RT, Dean RL, Beer B, Lippa AS.
RK, Davis RP, Dawson LA, Foley AG, Gar- Science 1982;217:408–414.
tlon J, Gonzalez MI, Heslop T, Hirst WD,
Jennings C, Jones DN, Lacroix LP, Martyn 115. Bartus RT. Exp Neurol 2000;163:495–529.
A, Ociepka S, Ray A, Regan CM, Roberts JC, 116. DeKosky ST, Ikonomovic MD, Styren SD,
Schogger J, Southam E, Stean TO, Trail BK, Beckett L, Wisniewski S, Bennett DA, Cochran
Upton N, Wadsworth G, Wald JA, White T, EJ, Kordower JH, Mufson EJ. Ann Neurol
Witherington J, Woolley ML, Worby A, Wil- 2002;51:145–155.
son DM. J Pharmacol Exp Ther 2007;321: 117. Davis KL, Mohs RC, Marin D, Purohit DP, Perl
1032–1045. DP, Lantz M, Austin G, Haroutunian V. JAMA
96. Scali C, Giovannini MG, Prosperi C, Bartolini 1999;281:1401–1406.
L, Pepeu G. Pharmacol Res 1997;36:463–469. 118. Watkins PB, Zimmerman HJ, Knapp MJ, -
97. Shimazaki T, Kaku A, Chaki S. Eur J Phama- Gracon SI, Lewis KW. JAMA 1994;271:
col 2007;575:94–97. 992–998.
98. Morris R. J Neurosci Methods 1984;11:47–60. 119. Shigeta M, Homma A. CNS Drug Rev 2001;7:
353–368.
99. Olton DS. Physiol and Behav
1987;40:793–797. 120. Jacobson SA, Sabbagh MN. Exp Opin Drug
100. Van Dam D, Abramowski D, Staufenbiel M, De Metab Toxicol 2008;4:1363–1369.
Deyn PP. Psychopharmacology 2005;180: 121. Gottwald MD, Rozanski RI. Exp Opin Investig
177–190. Drugs 1999;8:1673–1682.
101. Marcos B, Chuang TT, Gil-Bea FJ, Ramirez 122. Cutler NR, Polinsky RJ, Sramek JJ, Enz A,
MJ. Br J Pharmacol 2008;155:434–440. Jhee SS, Mancione L, Hourani J, Zolnouni P.
102. Timmermann DB, Grønlien JH, Kohlhaas Acta Neurol Scand 1998;97:244–250.
KL, Nielsen EØ, Dam E, Jørgensen TD, Ahr- 123. Samochocki M, H€offle A, Fehrenbacher A, Jo-
ing PK, Peters D, Holst D, Chrsitensen JK, stock R, Ludwig J, Christner C, Radina M,
Malysz J, Briggs CA, Gopalakrishnan M, Ol- Zerlin M, Ullmer C, Pereira EF, L€ubbert H,
sen GM. J Pharmacol Exp Ther 2007;323: Albuquerque EX, Maelicke A. J Pharmacol
294–307. Exp Ther 2003;305:1024–1036.
103. Banfi S, Cornelli U, Fonio W, Dorigotti L. J 124. Lilienfeld S. CNS Drug Rev 2002;8:159–176.
Pharmacol Methods 1982;8:255–263. 125. Bai DL, Tang XC, He XC. Curr Med Chem
104. Fanselow MS. Anim Learn Behav 1990;18: 2000;7:355–374.
264–270. 126. Greig NH, Sambamurti K, Yu QS, Brossi A,
105. Sienkiewicz-Jarosz H, Maciejak P, Krzaścik P, Bruinsma GB, Lahiri DK. Curr Alzheimer Res
Członkowska AI, Szyndler J, Bidziński A, 2005;2:281–290.
Kostowski W, Płaznik A. Phamacol Biochem 127. Vellas B, Andrieu S, Sampaio C, Wilcock G.
Behav 2003;75:491–496. Lancet Neurol 2007;6:56–62.
56 COGNITION

128. Benzi G, Moretti A. Eur J Pharmacol 1988; 145. Iwasaki K, Matsumoto Y, Fujiwara M. Jpn J
346:1–16. Pharmacol 1992;58:117–126.
129. Ferreri F, Agbokou nad C, Gauthier S. J Psy- 146. Abou-Khalil B. Neuropsychiatr Dis Treat
chiatry Neurosci 2006;31:369–376. 2008;4:507–523.
130. Friedman JL. Psychopharmacology 2004; 147. Zhou B, Zhang Q, Tian L, Xiao J, Stefan H,
174:45–53. Zhou D. Epilepsy Behav 2008;12:305–310.
131. Buchanan RW, Freedman R, Javitt DC, Abi- 148. Gotti C, Moretti M, Gaimarri A, Zanardi A,
Dargham A, Lieberman JA. Schizophr Bull Clementi F, Zoli M. Biochem Pharmacol
2007;33:1120–1130. 2007;74:1102–1111.
132. Schilstr€
om B, Ivanov VB, Wiker C, Svensson 149. Lippiello PM, Bencherif M, Hauser TA, Jordan
TH. Neuropsychopharmacology 2007;32:43–53. KG, Letchworth SR, Mazurov AA. Exp Opin
133. Jeppesen L, Olesen PH, Hansen L, Sheardown Drug Discov 2007;2:1185–1203.
MJ, Thomsen C, Rasmussen T, Jensen AF, 150. Levin ED. J Neurobiol 2002;53:633–640.
Christensen MS, Rimvall K, Ward JS, White- 151. Lloyd GK, Williams M. J Pharmacol Exp Ther
sitt C, Calligaro DO, Bymaster FP, Delapp 2000;292:461–467.
NW, Felder CC, Shannon HE, Sauerberg P. 152. Cincotta SL, Yorek MS, Moschak TM, Lewis
J Med Chem 1999;42:1999–2006. SR, Rodefer JS. Curr Opin Investig Drugs
134. Stanhope KJ, Mirza NR, Bickerdike MJ, 2008;9:47–56.
Bright JL, Harrington NR, Hesselink MB,
153. Freedman R, Coon H, Myles-Worsley M, Orr-
Kennett GA, Lightowler S, Sheardown MJ, Urtreger A, Olincy A, Davis A, Polymeropoulos
Syed R, Upton RL, Wadsworth G, Weiss SM, M, Holik J, Hopkins J, Hoff M, Rosenthal J,
Wyatt A. J Pharmacol Exp Ther 2001;
Waldo MC, Reimherr F, Wender P, Yaw J,
299:782–792. Young DA, Breese CR, Adams C, Patterson
135. Graul A, Castaner J. Drugs Future D, Adler LE, Kruglyak L, Leonard S, Byerley
1996;21:911–916. W. Proc Natl Acad Sci USA 1997;94:587–592.
136. Mizobe F, Nakahara N, Ogane N, Saito Y, Ise 154. Broad LM, Sher E, Astles PC, Zwart R, O’Neill
M, Iga Y, Fukui K, Fujise N, Haga T, Kawa- MJ. Drugs Future 2007;32:161–170.
nishi G.In: Nagatsu T, Fisher A, Yoshida M, 155. Olincy A, Harris JG, Johnson LL, Pender V,
editors. Basic, Clinical, and Therapeutic As- Kongs S, Allensworth D, Ellis J, Zerbe GO,
pects of Alzheimer’s and Parkinson’s Disease. Leonard S, Stevens KE, Stevens JO, Martin L,
New York: Plenum Press; 1990. p. 351–358.
Adler LE, Soti F, Kem WR, Freedman R. Arch
137. Mucke HAM, Castaner J. Drugs Future 1998; Gen Psychiatry 2006;63:630–638.
3:843 156. Freedman R, Olincy A, Buchanan RW, Harris
138. Caccamo A, Oddo S, Billings LM, Green KN, JG, Gold JM, Johnson L, Allensworth D, Guz-
Martinez-Coria H, Fisher A, LaFerla FM. Neu- man-Bonilla A, Clement B, Ball MP, Kutnick
ron 2006;49:671–682. J, Pender V, Martin LF, Stevens KE, Wagner
139. DailyDrugNewscom (Daily Essentials) June BD, Zerbe GO, Soti F, Kem WR. Am J Psychia-
17, 2008. try 2008;165:1040–1047.
140. Conn PJ, Tamminga C, Schoepp DD, Lindsley 157. Tietje KR, Anderson DJ, Bitner RS, Blomme
C. Mol Interv 2008;8:99–107. EA, Brackemeyer PJ, Briggs CA, Browman KE,
141. Thomas RL, Mistry R, Langmead CJ, Wood Bury D, Curzon P, Drescher KU, Frost JM,

MD, Challiss RA. J Pharmacol Exp Ther 2008; Fryer RM, Fox GB, Gronlien JH, Hakerud M,
327:365–374. Gubbins EJ, Halm S, Harris R, Helfrich RJ,
Kohlhaas KL, Law D, Malysz J, Marsh KC,
142. Marlo JE, Niswender CM, Days EL, Bridges
TM, Xiang Y, Rodriguez AL, Shirey JK, Brady Martin RL, Meyer MD, Molesky AL, Nikkel AL,
AE, Nalywajko T, Luo Q, Austin CA, Williams Otte S, Pan L, Puttfarcken PS, Radek RJ, Robb
MB, Kim K, Williams R, Orton D, Brown HA, HM, Spies E, Thorin-Hagene K, Waring JF,
Ween H, Xu H, Gopalakrishnan M, Bunnelle
Lindsley CW, Weaver CD, Conn PJ. Mol Phar-
macol 2009;75:577–588. WH. CNS Neurosci and Ther 2008; 14:65–82.
143. Danysz W, Parsons CG. Int J Geriatr Psychia- 158. Pichat P, Bergis OE, Terranova JP, Urani A,
try 2003;18(Suppl 1): S23–S32. Duarte C, Santucci V, Gueudet C, Voltz C,
Steinberg R, Stemmelin J, Oury-Donat F, Ave-
144. M€uller WE, Eckert GP, Eckert A. Pharmacop- net P, Griebel G, Scatton B. Neuropsychophar-
sychiatry 1999;32(Suppl 1): 2–9.
macology 2007;32:17–34.
REFERENCES 57

159. Feuerbach D, Lingenhoehl K, Olpe HR, Vass- 175. King MV, Marsden CA, Fone KC. Trends Phar-
out A, Gentsch C, Chaperon F, Nozulak J, Enz macol Sci 2008;29:482–492.
A, Bilbe G, McAllister K, Hoyer D. Neurophar- 176. Ogren SO, Eriksson TM, Elvander-Tottie E,
macology 2009;56:254–263. D’Addario C, Ekstr€om JC, Svenningsson P,
160. Timmermann DB, Groenlien JH, Kohlhaas KL, Meister B, Kehr J, Stiedl O. Behav Brain Res
Nielsen EO, Dam E, Joergensen TD, Ahring 2008;195:54–77.
PK, Peters D, Holst D, Chrsitensen JK, Malysz 177. Galletly CA, Clark CR, McFarlane AC. Eur
J, Briggs CA, Gopalakrishnan M, Olsen GM. J Neuropsychopharmacol 2005;15:601–608.
Pharmacol Exp Ther 2007; 323:294–307. 178. Meltzer HY, McGurk SR. Schizophr Bull
161. Patterson F, Jepson C, Strasser AA, Loughead 1999;2:233–255.
J, Perkins KA, Gur RC, Frey JM, Siegel S, 179. Forster EA, Cliffe IA, Bill DJ, Dover GM, Jones
Lerman C. Biol Psychiatry 2009;65:144–149. D, Reilly Y, Fletcher A. Eur J Pharmacol 1995;
162. Raybuck JD, Portugal GS, Lerman C, Gould 281:81–88.
TJ. Behav Neurosci 2008;122:1166–1171. 180. Childers WE, Jr, Abou-Gharbia MA, Kelly MG,
163. Gatto GJ, Bohme GA, Caldwell WS, Letch- Andree TH, Harrison BL, Ho DM, Hornby G,
worth SR, Traina VM, Obinu MC, Laville M, Huryn DM, Potestio L, Rosenzweig-Lipson SJ,
Reibaud M, Pradier L, Dunbar G, Bencherif M. Schmid J, Smith DL, Sukoff SJ, Zhang G,
CNS Drug Rev 2004;10:147–166. Schechter LE. J Med Chem 2005;48:
164. Morrison T. BioWorld September 17 2008. 3467–3470.
165. Rueter LE, Anderson DJ, Briggs CA, Donnelly- 181. Childers WE, Jr, Harrison BL, Abou-Gharbia
Roberts DL, Gintant GA, Gopalakrishnan M, MA, Raje S, Parks V, Pangalos MN, Schechter
Lin NH, Osinski MA, Reinhart GA, Buckley LE. Drugs Future 2007;32:399–407.
MJ, Martin RL, McDermott JS, Preusser LC, 182. Moser PC, Bergis OE, Jegham S, Lochead A,
Seifert TR, Su Z, Cox BF, Decker MW, Sullivan Duconseille E, Terranova JP, Caille D, Berque-
JP. CNS Drug Rev 2004;10:167–168. Bestel I, Lezoualc’h F, Fischmeister R, Du-
166. Arneric SP, Holladay M, Williams M. Biochem muis A, Bockaert J, George P, Soubrie P, Scat-
Pharmacol 2007;74:1092–1101. ton B. J Pharmacol Exp Ther 2002;302:
167. Hill SJ, Ganellin C, Timmermans H, Schwartz 731–741.
JC, Shankley N, Young JM, Schunack W, Levi 183. Hille C, Bate S, Davis J, Gonzalez MI. Behav
R, Haas HL. Pharmacol Rev 1997;49:253–278. Brain Res 2008;195:180–186.
168. Hudkins RL, Raddatz R. Ann Rep Med Chem 184. Robichaud AJ. Curr Top Med Chem 2006;6:
2007;42:49–63. 553–568.
169. Wieland K, Bongers G, Yamamoto Y, Hashi- 185. Johnson NC. Drugs Future 2008;33 (Suppl A):
moto T, Yamatodani A, Menge WM, Timmer- Abst L05.
man H, Lovenberg TW, Leurs R. J Pharmacol 186. Wu J, Li Q, Bezprozvanny I. Mol Neurodege-
Exp Ther 2001;299:908–914. ner 2008;3:15.
170. Fox GB, Esbenshade TA, Pan JB, Radek RJ, 187. Bachurin S, Bukatina E, Lermontova N, Tka-
Krueger KM, Yao BB, Browman KE, Buckley chenko S, Afanasiev A, Grigoriev V, Grigorieva
MJ, Ballard ME, Komater VA, Miner H, Zhang I, Ivanov Y, Sablin S, Zefirov N. Ann NY Acad
M, Faghih R, Rueter LE, Bitner RS, Drescher Sci 2001;939:425–435.
KU, Wetter J, Marsh K, Lemaire M, Porsolt 188. Doody RS, Gavrilova SI, Sano M, Thomas RG,
RD, Bennani YL, Sullivan JP, Cowart MD, Aisen PS, Bachurin SO, Seely L, Hung D.
Decker MW, Hancock AA. J Pharmacol Exp
Lancet 2008;372:207–215.
Ther 2005;313:176–190.
189. Perez-Garcıa G, Gonzalez-Espinosa C, Men-
171. Nagase T, Mizutani T, Sekino E, Ishikawa S, eses A. Behav Brain Res 2006;169:83–92.
Ito S, Mitobe Y, Miyamoto Y, Yoshimoto R,
190. Cifariello A, Pompili A, Gasbarri A. Behav
Tanaka T, Ishihara A, Takenaga N, Tokita S,
Sato N. J Med Chem 2008;51:6889–6901. Brain Res 2008;195:171–179.
172. Barnes NM, Sharp T. Neuropharmacology 191. Gasbarri A, Cifariello A, Pompili A, Meneses A.
1999;38:1083–1152. Behav Brain Res 2008;195:164–170.
192. Forbes IT, Douglas S, Gribble AD, Ife RJ,
173. Hannon J, Hoyer D. Behav Brain Res 2008;
195:198–213. Lightfoot AP, Garner AE, Riley GJ, Jeffrey
P, Stevens AJ, Stean TO, Thomas DR. Bioorg
174. Roth BL, Hanizavareh SM, Blum AE. Psycho-
Med Chem Lett 2002;12:3341–3344.
pharmacology 2004;174:17–24.
58 COGNITION

193. El-Ghundi M, O’Dowd BF, George SR. Rev 213. Wezenberg E, Verkes RJ, Ruigt GS, Hulstijn
Neurosci 2007;18:37–66. W, Sabbe BG. Neuropsychopharmacology
194. Guillin O, Abi-Dargham A, Laruelle M. Int Rev 2007;32:1272–1283.
Neurobiol 2007;78:1–39. 214. Bergeron R, Meyer TM, Coyle JT, Greene RW.
195. Goldman-Rakic PS, Castner SA, Svensson TH, Proc Natl Acad Sci USA 1998;95:15730–15734.
Siever LJ, Williams GV. Psychopharmacology 215. Marino MJ, Knutsen LJ, Williams M. J Med
2004;174:3–16. Chem 2008;51:1077–1107.
196. Rogers BN, Schmidt CJ. Ann Rep Med Chem 216. Heresco-Levy U, Ermilov M, Lichtenberg P, Bar
2006;41:3–21. G, Javitt DC. Biol Psychiatry 2004;55: 165–171.
197. Alexander SPH, Mathie MA, Peters JA. Br J 217. Javitt DC, Silipo G, Cienfuegos A, Shelley AM,
Pharmacol. 2008;153(Suppl 2): S34–S35. Bark N, Park M, Lindenmayer JP, Suckow R,
198. Salmi P, Isacson R, Kull B. CNS Drug Rev Zukin SR. Int J Neuropsychopharmacol 2001;
2004;10:230–242. 4:385–391.
199. Moreland RB, Patel M, Hsieh GC, Wetter JM, 218. Sur C, Kinney GG. Expert Opin Investig Drugs
Marsh K, Brioni JD. Pharmacol Biochem Be- 2004;13:515–521.
hav 2005;82:140–147. 219. Perry KW, Falcone JF, Fell MJ, Ryder JW, Yu
200. Briand LA, Gritton H, Howe WM, Young DA, H, Love PL, Katner J, Gordon KD, Wade MR,
Sarter M. Prog Neurobiol 2007;83:69–91. Man T, Nomikos GG, Phebus LA, Cauvin AJ,
Johnson KW, Jones CK, Hoffmann BJ, San-
201. Ramos BP, Arnsten AF. Pharmacol Ther
2007;113:523–536. dusky GE, Walter MW, Porter WJ, Yang L,
Merchant KM, Shannon HE, Svensson KA.
202. Coull JT. Drugs Aging 1994;5:116–126. Neuropharmacology 2008;55:743–754.
203. Arnsten AF. Psychopharmacology 2004;174: 220. Depoortere R, Dargazanli G, Estenne-Bouhtou
25–31. G, Coste A, Lanneau C, Desvignes C, Poncelet
204. ClinicalTrials.gov Web site (NCT00409201). M, Heaulme M, Santucci V, Decobert M, Cuden-
205. Friedman JL, Carpenter D, Lu J, Fan J, Tang nec A, Voltz C, Boulay D, Terranova JP, Stem-
CY, White L, Parrella M, Bowler S, Elbaz Z, melin J, Roger P, Marabout B, Sevrin M, Vige X,
Flanagan L, Harvey PD. J Clin Psychophar- Biton B, Steinberg R, Françon D, Alonso R,
macol 2008;28:59–63. Avenet P, Oury-Donat F, Perrault G, Griebel
206. Winblad B, Fioravanti M, Dolezal T, Logina I, G, George P, Soubrie P, Scatton B. Neuropsy-
Milanov IG, Popescu DC, Solomon A. Clin chopharmacology 2005;30:1963–1985.
Drug Investig 2008;28:533–552. 221. Black MD, Varty GB, Arad M, Barak S, De
207. O’Neill MJ, Bleakman D, Zimmerman DM, Levie A, Boulay D, Pichat P, Griebel G, Weiner
Nisenbaum ES. Curr Drug Targets CNS Neu- I. Psychopharmacology 2009;202:385–396.
rol Disord 2004;3:181–194. 222. Barnard EA, Skolnick P, Olsen RW, Mohler H,
208. Black MD. Psychopharmacology 2005;179: Sieghart W, Biggio G, Braestrup C, Bateson
154–163. AN, Langer SZ. Pharmacol Rev 1998;50:
209. Patil ST, Zhang L, Martenyi F, Lowe SL, 291–313.
Jackson KA, Andreev BV, Avedisova AS, Bar- 223. Ghoneim MM, Mewaldt SP. Anesthesiology
denstein LM, Gurovich IY, Morozova MA, Mo- 1990;72:926–938.
solov SN, Neznanov NG, Reznik AM, Smule- 224. Maubach K. Curr Drug Targets CNS Neurol
vich AB, Tochilov VA, Johnson BG, Monn JA, Disord 2003;2:233–239.
Schoepp DD. Nat Med 2007;13:1102–1107. 225. Collinson N, Kuenzi FM, Jarolimek W, Mau-
210. Williams DL, Jr, Lindsley CW. Curr Top Med bach KA, Cothliff R, Sur C, Smith A, Otu FM,
Chem 2005;5:825–846. Howell O, Atack JR, McKernan RM, Seabrook
211. Liu F, Grauer S, Kelley C, Navarra R, Graf R, GR, Dawson GR, Whiting PJ, Rosahl TW. J
Zhang G, Atkinson PJ, Wantuch C, Popiolek M, Neurosci 2002;22:5572–5580.
Day M, Khawaja X, Smith D, Olsen M, Kour- 226. Atack JR, Bayley PJ, Seabrook GR, Wafford
anova E, Gilbert A, Lai M, Pausch MH, Pruthi F, KA, McKernan RM, Dawson GR. Neurophar-
Pulicicchio C, Brandon NJ, Comery TA, Beyer macology 2006;51:1023–1029.
CE, Logue S, Rosenzweig-Lipson S, Marquis 227. Atack JR. CNS Neurosci Ther 2008;14:25–35.
KL. J Pharmacol Exp Ther 2008; 327:827–839.
228. Fredholm BB, Jzerman IAP, Jacobson KA,
212. Arai AC, Kessler M. Curr Drug Targets 2007; Klotz KN, Linden J. Pharmacol Rev 2001;
8:583–602. 53:527–552.
REFERENCES 59

229. Takahashi RN, Pamplona FA, Prediger RD. 247. Spooren W, Riemer C, Meltzer H. Nat Rev
Front Biosci 2008;13:2614–2632. Drug Discov 2005;4:967–975.
230. Simola N, Morelli M, Pinna A. Curr Pharm Des 248. Counts SE, Perez SE, Kahl U, Bartfai T, Bow-
2008;14:1475–1489. ser RP, Deecher DC, Mash DC, Crawley JN,
231. Gimenez-Llort L, Schiffmann SN, Shmidt T, Mufson EJ. CNS Drug Rev 2001;7:445–470.
Canela L, Camón L, Wassholm M, Canals M, 249. Vrontakis ME. Curr Drug Targets CNS Neurol
Terasmaa A, Fern andez-Teruel A, Tobeña A, Disord 2002;1:531–541.
PopovaE,Ferre S,AgnatiL,CiruelaF,Martınez 250. Scott MK, Ross TM, Lee DH, Wang HY, Shank
E, Scheel-Kruger J, Lluis C, Franco R, Fuxe K, RP, Wild KD, Davis CB, Crooke JJ, Potocki AC,
Bader M. Neurobiol Learn Mem 2007;87:42–56. Reitz AB. Bioorg Med Chem 2000;8: 1383–1391.
232. Mihara T, Mihara K, Yarimizu J, Mitani Y, 251. Hol EM, Gispen WH, B€
ar PR. Peptides
Matsuda R, Yamamoto H, Aoki S, Akahane A, 1995;16:979–993.
Iwashita A, Matsuoka N. J Pharmacol Exp
252. Matsumoto T, Tsuda S, Nakamura S. J Neural
Ther 2007;323:708–719. Transm Gen Sect 1995;100:1–15.
233. Thoenen H, Barde YA. Physiol Rev 1980;60: 253. Albiston AL, Morton CJ, Ng HL, Pham V,
1284–1335. Yeatman HR, Ye S, Fernando RN, De Bundel
234. Cuello AC, Bruno MA, Bell KF. Curr Alzhei- D, Ascher DB, Mendelsohn FA, Parker MW,
mer Res 2007;4:351–835. Chai SY. FASEB J 2008;22:4209–4217.
235. Hefti F, Schneider LS. Clin Neuropharmacol 254. Donaldson ZR, Young LJ. Science 2008;322:
1991;14(Suppl 1): S62–S66. 900–904.
236. Seiger A, Nordberg A, vonHolst H, Backman L, 255. Marazziti D, Dell’osso C. Curr Med Chem
Ebendal T, Alafuzoff I, Amberla K, Hartvig P, 2008;15:698–704.
Herlitz A, Lilja A, Lundqvist H, Langstrom B,
256. Ferguson JN, Young LJ, Hearn EF, Insel TR,
Meyerson B, Persson A, Viitanen M, Winblad Winslow JT. Nature Genetics 2000;25:
B, Olson L. Behav Brain Res 1993;57:255–261. 284–288.
237. Tuszynski MH, Thal L, Pay M, Salmon DP, Hoi 257. Jing W, Guo F, Cheng L, Zhang JF, Qi JS.
SU, Bakay R, Patel P, Blesch A, Vahlsing HL,
Neurosci Lett 2009;450:306–310.
Ho G, Tong G, Potkin SG, Fallon J, Hansen L,
Mufson EJ, Kordower JH, Gall C, Conner J. 258. Mellow AM, Sunderland T, Cohen RM, Lawlor
BA, Hill JL, Newhouse PA, Cohen MR,
Nat Med 2005;11:551–555.
Murphy DL. Psychopharmacology 1989;98:
238. Pezet S, Malcangio M. Expert Opin Ther Tar-
403–437.
gets 2004;8:391–399.
259. Rotella DP.In: Moos WH, editor. Comprehen-
239. Tapia-Arancibia L, Aliaga E, Silhol M, Aranci- sive Medicinal Chemistry II. Amesterdam:
bia S. Brain Res Rev 2008;59:201–220. Elsevier Press; 2007. p 919–957.
240. Gunstad J, Benitez A, Smith J, Glickman E, 260. Silva AJ, Kogan JH, Frankland PW, Kida S.
Spitznagel MB, Alexander T, Juvancic-Heltzel
Annu Rev Neurosci 1998;21:127–148.
J, Murray L. J Geriatr Psychiatry Neurol
2008;21:166–170. 261. Dash PK, Hochner B, Kandel ER. Nature 1990;
345:718–721.
241. Dik MG, Pluijm SM, Jonker C, Deeg DJ, Lo-
mecky MZ, Lips P. Neurobiol Aging 2003;24: 262. Barco A, Alarcon JM, Kandel ER. Cell 2002;
573–581. 108:689–703.
242. Grotenhermen F. Curr Drug Targets CNS 263. Josselyn SA, Shi C, Carlezon WA, Jr, Neve RL,
Nestler EJ, Davis M. J Neurosci 2001;21:
Neurol Disord 2005;4:507–530.
2404–2412.
243. Wotjak CT. Mini Rev Med Chem 2005;5:
659–670. 264. Monti B, Berteotti C, Contestabile A. Neurop-
sychopharmacology 2006;31:278–286.
244. Jones D. Nat Rev Drug Discov 2008;7:961–962.
265. Halene TB, Siegel SJ. Drug Discov Today
245. Holmes A, Heilig M, Rupniak NM, Steckler T, 2007;12:870–878.
Griebel G. Trends Pharmacol Sci 2003;24:
266. Menniti FS, Faraci WS, Schmidt CJ. Nat Rev
580–588.
Drug Discov 2006;5:660–670.
246. Siuciak JA, McCarthy SA, Martin AN, Chapin
DS, Stock J, Nadeau DM, Kantesaria S, Bryce- 267. Robichaud A, Stamatiou PB, Jin SL, Lachance
Pritt D, McLean S. Psychopharmacology N, MacDonald D, Laliberte F, Liu S, Huang Z,
Conti M, Chan CC. J Clin Invest 2002;110:
2007;194:185–195.
1045–1052.
60 COGNITION

268. Ahmed T, Frey JU. Neuroscience 2003;117: 274. Siuciak JA, Chapin DS, Harms JF, Lebel LA,
627–638. McCarthy SA, Chambers L, Shrikhande A,
269. Boess FG, Hendrix M, van der Staay FJ, Erb C, Wong S, Menniti FS, Schmidt CJ. Neurophar-
Schreiber R, van Staveren W, de Vente J, macology 2006;51:386–396.
Prickaerts J, Blokland A, Koenig G. Neuro- 275. Verhoest RP. 236th ACS National Meeting,
pharmacology 2004;47:1081–1092. Philadelphia, PA, August 17–21, 2008. MEDI
270. Devan BD, Sierra-Mercado D, Jr, Jimenez M, 219.
Bowker JL, Duffy KB, Spangler EL, IngramDK. 276. Greely H, Sahakian B, Harris J, Kessler RC,
Pharmacol Biochem Behav 2004;79:691–699. Gazzaniga M, Campbell P, Farah MJ. Nature
271. van der Staay FJ, Rutten K, B€
arfacker L, 2008;456:702–705.
Devry J, Erb C, Heckroth H, Karthaus D, 277. Mitler MM, O’Malley MB.In: Kryger MH, Roth
Tersteegen A, van Kampen M, Blokland A, T, Diment WC, editors. Principles and Practice
Prickaerts J, Reymann KG, Schr€oder UH, of Sleep Medicine. 4th ed. Philadelphia, PA:
Hendrix M. Neuropharmacology 2008;55: Elsevier Saunders; 2005. p 485.
908–918. 278. Lanni C, Lenzken SC, Pascale A, Del Vecchio I,
272. Schmidt CJ, Chapin DS, Cianfrogna J, Cor- Racchi M, Pistoia F, Govoni S. Pharmacol Res
man ML, Hajos M, Harms JF, Hoffman WE, 2008;57:196–213.
Lebel LA, McCarthy SA, Nelson FR, Proulx- 279. Markou A, Chiamulera C, Geyer MA, Trickle-
LaFrance C, Majchrzak MJ, Ramirez AD, bank M, Steckler T. Neuropsychopharmacol-
Schmidt K, Seymour PA, Siuciak JA, Tingley ogy 2009;34:74–89.
FD 3rd, Williams RD, Verhoest PR, Menniti
280. Enna S, Williams M. J Pharmacol Exp Ther
FS. J Pharmacol Exp Ther 2008;325:681–690. 2009;329:404–411.
273. Roderfer JS, Murphy ER, Baxter MG. Eur J
Neurosci 2005;21:1070–1076.

Das könnte Ihnen auch gefallen