Canna~Fangled Abstracts

Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders.

By December 27, 2014No Comments

Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders

Under a Creative Commons license
  Open Access


Abstract

elsevierPathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example D-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.

Keywords

  • Fear extinction;
  • Exposure therapy;
  • Augmented relearning;
  • Reconsolidation;
  • Drug development;
  • Cognitive enhancer

Abbreviations

  • 5-HT5-hydroxytryptamine = serotonin;
  • ACadenylate cyclase;
  • AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid;
  • AMYamygdala;
  • BAbasal amygdala;
  • BDNFbrain-derived neurotrophic factor;
  • BLAbasolateral amygdaloid complex;
  • BZDbenzodiazepine;
  • CaMKIICa2+/calmodulin-dependent protein kinase II;
  • cAMP3′-5′-cyclic adenosine monophosphate;
  • Cav 1.2L-type calcium channel alpha 1C isoform;
  • Cav 1.3L-type calcium channel alpha 1D isoform;
  • CBTcognitive behavioral therapy;
  • CCKcholecystokinin;
  • CeAcentral amygdala;
  • CeMcentromedial amygdala;
  • CeLcentrolateral amygdala;
  • CNScentral nervous system;
  • CREBcAMP response element binding;
  • CSconditioned stimulus;
  • DAdopamine;
  • DCSD-cycloserine;
  • DNAdeoxyribonucleic acid;
  • eCBendogenous cannabinoiod;
  • ERKextracellular regulated kinase;
  • ERPexposure and prevention CBT;
  • FGF2fibroblast growth factor-2;
  • GABAγ-aminobutyric acid;
  • GABAAGABAA receptor;
  • GADgeneralized anxiety disorder;
  • GAD (65/67)glutamate decarboxylase 65/67;
  • GluNNMDA receptor subtype;
  • GRglucocorticoid receptor;
  • H3histone H3 protein;
  • H4histone H4 protein;
  • HAThistone acetyltransferase;
  • HDAChistone deacetylase;
  • HDAC2histone deacetylase 2;
  • HPAhypothalamic-pituitary-adrenal axis;
  • HPChippocampus;
  • icvintracerebroventricular;
  • ILinfralimbic cortex;
  • ITCintercalated cell masses;
  • Klysine;
  • KORkappa opioid receptor;
  • LAlateral amygdala;
  • L-DOPAlevodopa;
  • MAPKmitogen-activated protein kinase;
  • MDMA3,4-methylenedioxy-N-methylamphetamine;
  • Mg2+magnesium ion;
  • mGluRmetabotropic glutamate receptor;
  • miRNAmicro-RNA;
  • mPFCmedial prefrontal cortex;
  • mRNAmessenger ribonucleic acid;
  • MS-275entinostat (Pyridin-3-ylmethyl N-[[4-[(2-aminophenyl)carbamoyl] phenyl]methyl]carbamate);
  • NMDAN-methyl-D-aspartate;
  • NMDARNMDA receptor;
  • nsnot studied;
  • NOnitric oxide;
  • NPSneuropeptide S;
  • NPYneuropeptide Y;
  • OCDobsessive-compulsive disorder;
  • OXToxytocin;
  • PAGperiaqueductal gray;
  • PEPA4-[2-(Phenylsulphonylamino)ethylthio]-2,6-difluorophenoxyacetamide;
  • PKCas Ca2+ /phospholipid-dependent protein kinase C;
  • PTSDpost-traumatic stress disorder;
  • PLprelimbic cortex;
  • SADsocial anxiety disorder;
  • SAHAvorinostat (N-hydroxy-N’-phenyl-octanediamide);
  • SNPsingle nucleotide polymorphism;
  • SNRIselective noradrenaline reuptake inhibitor;
  • SSRIselective serotonin reuptake inhibitor;
  • TrkBtropomyosin-related kinase B;
  • USunconditioned stimulus;
  • VGCCvoltage-gated calcium channel;
  • VRETvirtual reality exposure;
  • Zn2+zinc ion

1. Introduction

Fear, anxiety and trauma-related disorders are associated with excessive fear reactions triggered by specific objects, situations or internal and external cues in the absence of any actual danger, and often include an inability to extinguish learned fear and to show adequate safety learning [(Michael et al., 2007; Wessa & Flor, 2007; Milad et al., 2009; Jovanovic et al., 2012; Milad et al., 2013) reviewed in (Holmes & Singewald, 2013) and (Kong et al., 2014)]. Pathological fear and anxiety occur in a range of psychiatric conditions, including various types of phobia (e.g. social phobia, agoraphobia or specific phobia), panic disorder with/without agoraphobia, obsessive-compulsive disorder (OCD), generalized anxiety (GAD) and post-traumatic stress disorder (PTSD) (ICD-10, 1994; DSM-5, 2013). These disorders comprise the most common mental disorders and are estimated to have a life-time prevalence of up to 28% amongst western populations (Kessler et al., 2005; Wittchen et al., 2011; Kessler et al., 2012). In addition to the personal suffering of patients, the economic burden caused by anxiety disorders is heavy (Gustavsson et al., 2011).

Available pharmacological and psychotherapeutic treatments (Bandelow et al., 2007) which aim to reduce fear and anxiety are associated with decreased symptom severity, but up to 40% of anxiety patients show only partial long-term benefit, and a majority of them fail to achieve complete remission (Hoffman & Mathew, 2008; Stein et al., 2009; Bandelow et al., 2012) clearly underlining the need for further improvement. Current pharmacological approaches either induce rapid anxiolytic effects (e.g. benzodiazepines, some antipsychotics) or require prolonged, chronic treatment (e.g. antidepressants) to attenuate symptoms of pathological fear and anxiety. Commonly employed psychotherapeutic interventions apply cognitive behavioral strategies and exposure techniques to help patients overcome the maladaptive beliefs and avoidance behaviors that reinforce the pathology related to fear-eliciting cues. Meta-analyses show that cognitive behavioral therapy (CBT) does have efficacy for several anxiety disorders, including PTSD, but patients have difficulty bearing the demanding and exhausting process of therapy and many who do manage to cope with it respond only partially and often relapse with time (Choy et al., 2007).

One strategy to improve CBT is to augment psychotherapy with adjunctive pharmacological treatments. Early attempts at combining ‘CBT’ with anxiolytic medications [e.g. benzodiazepines (BZD)] showed that the combination was no more effective [in some instances even counterproductive (Marks et al., 1993; Wilhelm & Roth, 1997)] than psycho- or pharmacotherapy alone [for details see (Otto et al., 2010a; Otto et al., 2010b; Rodrigues et al., 2011; Dunlop et al., 2012; Hofmann, 2012)]. However, at least in some cases, this failure may have reflected idiosyncratic effects of the drugs tested (especially BZDs) rather than utility of the strategy itself, and there has been an intense search to identify agents that serve as more effective adjuncts to CBT. The preclinical assay most frequently used in this search is fear extinction – the focus of this current review. Extinction of fear following Pavlovian fear learning (Pavlov, 1927) in animals is procedurally similar to exposure-based CBT (Milad & Quirk, 2012). We will briefly outline different aspects of Pavlovian fear learning [(which is thought to be involved in the etiology and maintenance of anxiety disorders, e.g. (Amstadter et al., 2009)] and extinction highlighting the key processes that could be targeted to augment fear extinction (see Figure 1 for an overview).

Full-size image (79 K)
Figure 1.

Different modes of fear alleviation and sites of possible pharmacological intervention. A, Fear alleviation is mediated via different mechanisms leading to acute or sustained fear relief. Sustained fear relief can be obtained either with exposures to the feared cues/situations or, in some instances, also without them. For a detailed description of the involved mechanisms, see (Riebe et al., 2012). Pharmacological interventions to boost fear relief can target different mechanisms at various levels. Examples are given with numbers 1-4, for detail see text. B, Fear conditioning represents a training phase in which a novel conditioned stimulus (CS) is paired with an unconditioned stimulus (US) (redline). Throughout fear training and testing, fear is measured as a conditioned response (y-axis), typically freezing, fear-potentiated startle, increased heart rate, and other quantifiable behavioural measures of fear. Following this training period, mice undergo a consolidation phase which transfers the labile newly formed fear memory into a stable long-term memory. Fear can be extinguished by repeated presentations of the CS (without the US; red line) resulting in fear extinction. Following the extinction training session, and akin to fear learning, consolidation processes are initiated to stabilize this labile fear extinction memory into a long-term memory. Poor extinction is evidenced by high fear responding (red bar), and successful extinction retrieval is shown by reduced fear (green) during a retrieval test. Extinguished fear can recover via three main mechanisms: spontaneous recovery (recovery of extinguished fear responses occurs with the passage of time in the absence of any further training.), fear renewal (when the conditioned CS is presented outside the extinction context, for example the conditioning or novel contexts), and fear reinstatement (when un-signalled presentations of the US are interposed between the completion of extinction training and a subsequent retention test). Drugs (green arrows) can be administered either immediately prior to (to induce extinction acquisition also called ‘within-session extinction’ and/or extinction consolidation) or immediately following (to rescue/boost extinction consolidation processes) the training session to modulate extinction mechanisms. A clinical aim of drug augmentation strategies is to promote good extinction retrieval and to protect against return-of-fear phenomena to provide good ‘longterm extinction’. C, Fear memory can be reactivated (red bar) by presentation of the CS, which transfers the stabilized memory into a labile phase which requires reconsolidation (a process by which previously consolidated memories are stabilized after fear retrieval). The fear memory can then be tested during another retrieval test (red bar) to assess reconsolidation. Drugs (green arrow) can be administered either immediately prior to or after the retrieval session to modulate reconsolidation mechanisms. This effect can then be tested during subsequent retrieval tests. Evidence for successful interference with reconsolidation of the original fear memory is revealed in reduced freezing during the test session.

Fear and fear extinction

Experimentally, fear conditioning occurs when a previously neutral stimulus [conditioned stimulus (CS) – such as a tone or light-] is paired with an aversive, unconditioned stimulus (US – e.g. electric shock to the forearm in humans, mild foot shock in rodents), resulting in a CS-US association whereby the CS alone elicits a conditioned fear response (e.g. freezing in rodents or increased skin conductance in humans). Following a successful CS-US association, fear memories require consolidation, a process involving a cascade of molecular and cellular events that alter synaptic efficacy, as well as a prolonged systems level interaction between brain regions, to stabilize the memory (McGaugh, 2000). Once consolidated, fear memories, reactivated by presenting the CS, are de-stabilized to render the original fear memory liable to pharmacological/behavioral interference (see overview in Figure 1) and this is then followed by a second phase of molecular and cellular events to re-stabilize (re-consolidate) the (adapted) memory (Nader et al., 2000).

Fear memories can be attenuated by various processes and interventions (including pharmacological and psychological approaches), some producing temporary blunting of fear behaviors and others causing more long-lasting relief. Interfering with the re-consolidation by inhibiting molecular and cellular events supporting fear memory re-stabilization [Figure 1C, (Nader et al., 2000; Lee et al., 2006)], for example with ß-adrenoceptor blockers (Debiec & Ledoux, 2004; Kindt et al., 2009), has been proposed as a clinical approach to alleviating fear memories. For discussion on this and other means of reducing fear (e.g. via US habituation) we refer the reader to some excellent prior reviews (Graham et al., 2011; Schwabe et al., 2014). Other potential ways to relieve fear include safety learning (Rogan et al., 2005; Kong et al., 2014) and erasure-like mechanisms such as destruction of erasure-preventing perineuronal nets (Gogolla et al., 2009).

Alternatively, fear memories can also be extinguished. Fear extinction, a process originally described by Pavlov (Pavlov, 1927), entails repeated exposure to anxiety-provoking cues to establish a new memory that counters the original fear memory. The process is highly relevant to fear, anxiety and trauma-related disorders which are associated with negative emotional reactions triggered by specific objects, situations or internal and external cues that are excessive to the actual danger posed. Moreover, extinction in animals is procedurally similar to forms of CBT that rely on exposure to anxiety-provoking cues (see Figure 1) (Milad & Quirk, 2012), and anxiety disorders are associated with an inability to extinguish learned fear and to respond adequately to safety signals (Michael et al., 2007; Wessa & Flor, 2007; Milad et al., 2009; Jovanovic et al., 2012; Milad et al., 2013) [reviewed in (Holmes & Singewald, 2013)]. Thus, fear extinction has considerable translational utility. The key processes that can be targeted to pharmacologically augment fear extinction are summarized in Figure 1.

Extinction is a learning process driven by violation of the original CS=US contingency (termed ‘prediction error’ for review see (Pearce & Bouton, 2001; McNally & Westbrook, 2006)], but it also contains other elements including habituation and desensitization some also say erasure/destabilization (Lin et al., 2011). Some authors therefore favor the term ‘relearning’ over extinction [for discussion see (Riebe et al., 2012)]. As with fear learning, extinction occurs in two phases: extinction acquisition and extinction consolidation retrieval. The decrement in the fear response during ’extinction training’ is also termed ‘within-session extinction’. Similar to fear memory, stabilization of the extinction memory requires both a cascade of overlapping, but dissociable, molecular and cellular events [for review see (Myers & Davis, 2007; Orsini & Maren, 2012)] that alter synaptic efficacy and brain systems-level interactions (Pape & Pare, 2010). The strength of extinction memory can be assessed at some interval (usually >1 day) after extinction training in “extinction retrieval” sessions (also termed ‘extinction retention’ or ‘extinction expression’, but note that we use the term “extinction retrieval” throughout this review).

That extinction memories are prone to re-emergence (due to insufficient ‘longterm extinction’) indicates that the original fear memory is still in place and the extinction memory is weaker/more labile than the fear memory. This may be particularly true of older (‘remote’) fear memories (Tsai & Graff, 2014). The re-emergence of extinguished fear occurs under multiple circumstances: (i) renewal, when the CS is presented in a different context to that in which extinction training occurred; (ii) reinstatement, when the original US or another stressor as is given unexpectedly; and (iii) spontaneous recovery, when a significant period of time has elapsed following successful extinction training (Myers & Davis, 2007; Herry et al., 2010). The likelihood of fear re-emergence is dependent on the strength of the extinction memory which is also determined by the type of extinction protocol used see e.g. (Li & Westbrook, 2008; Laborda & Miller, 2013). Spontaneous recovery (Rowe & Craske, 1998b, a; Schiller et al., 2008), reinstatement (Schiller et al., 2008) and renewal (Effting & Kindt, 2007) are all observable in clinical settings, and can be readily exploited in the laboratory to identify drug- and other interventions that can prevent or fear re-emergence in animals and relapse in humans (Vervliet et al., 2013).

Our goal in the current review is to offer a comprehensive overview of preclinical work on the possible pharmacological approaches (see Box 1 for an overview) to augment fear extinction and protect against the re-emergence of fear, and to discuss the potential translational value of these candidates as adjuncts to exposure-based CBT in anxiety patients.

Table 1.Serotonergic signaling in fear extinction (preclinical studies).
FACILITATING 5-HT SIGNALING


DRUG/
MANIPULATION
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
SERT KO6 ns ns no drug (Hartley et al., 2012)
no effect ns no drug (Wellman et al., 2007)
acute fluoxetine (SSRI) (-)## no effect ns ip (Lebron-Milad et al., 2013)
subchronic
citalopram (SSRI, 9d)
no effect ns ns ip (Burghardt et al., 2013)
chronic citalopram (SSRI, 22d) (-) ns ip (Burghardt et al., 2013)
chronic (14d)
fluoxetine (SSRI)
(+)# (+) ns po (Lebron-Milad et al., 2013)
chronic Fluoxetine (SSRI) no effect + +
(Ren-A,
SR,
Re-in)
ip (Karpova et al., 2011)
no effect ns +
(Re-in)
ip,po (Deschaux et al., 2011; Deschaux et al., 2013)
no effect* +*4 ns po (Camp et al., 2012)
no effect + ns po (Popova et al., 2014)
acute venlafaxine (SSRI) no effect + ns ip (Yang et al., 2012)
chronic venlafaxine (SSRI) ns ns +
(Re-in)
ip (Yang et al., 2012)
8-OH-DPAT (5HT1 ag) +*5 (-)*5 ns ip (Wang et al., 2013)
tandospirone (5HT1 ag) ns +* ns ip (Saito et al., 2013)
tcb2 (5HT2A ag) + ns ns ip (Zhang et al., 2013)
psilocybin (5HT2A ag) (+) ns ns ip (Catlow et al., 2013)
5-HT3 OE no effect ns ns no drug (Harrell & Allan, 2003)
INHIBITING 5-HT SIGNALING
5,7-dihydroxytryptamine (+)# + ns BLA (Izumi et al., 2012)
5,7-dihydroxytryptamine no effect no effect ns IL (Izumi et al., 2012)
5HT3 KO ns ns no drug (Kondo et al., 2014)
MDL11,939 (5HT2A ant) Ns ns ip (Zhang et al., 2013)
ketanserin (5HT2A/C ant) (+) Ns ns ip (Catlow et al., 2013)
granisetron (5HT3 ant) no effect (+) ns ip (Park & Williams, 2012)
1drug administration following extinction training; 2 SERT-KO results in increased synaptic 5-HT levels; 3 7 days; 4 protection from context over-generalization; 5 valproate-induced model of autism, 6 SERT-KO results in increased synaptic 5-HT levels* facilitates rescue of impaired fear extinction; # reduced fear expression at the beginning of extinction training; ## enhanced fear expression at the beginning of extinction training
+, improved; -, impaired; (+) or (-), only minor effects; po, peroral administration; ip, intraperitoneal injection; ns, not studied; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; Ren-A, Fear renewal in conditioning context; SR, spontaneous recovery; Re-in, reinstatement; ag, agonist; ant, antagonist, KO, knock-out; OE, over-expression
Table 1A.Human trials: Serotonergic drugs combined with CBT.
Disorder Study Design Outcome (compared to placebo control group) Reference
Healthy volunteers 14 days po escitalopram pretreatment; Fear conditioning paradigm Accelerated extinction learning (skin conductance responses) (Bui et al., 2013)
Panic disorder with/without agoraphobia Fluvoxamine or placebo followed by exposure therapy; psychological panic management followed by exposure therapy or exposure therapy alone Self-reported measures
All treatments effective, however fluvoxamine plus CBT superior to all other treatments
(de Beurs et al., 1995)
Panic disorder with/without agoraphobia 12 weeks paroxetine or placebo plus CBT Reduced number of panic attacks in paroxetine/CBT group (Oehrberg et al., 1995)
Panic disorder with/without agoraphobia 10 weeks of paroxetine or placebo plus CBT in week 5 and 7 No significant difference in primary CGI outcome.
Secondary outcome: Higher proportion of panic-free patients in paroxetine-CBT group.
(Stein et al., 2000)
Panic disorder with/without agoraphobia 12 weeks fluvoxamine or placebo with or without CBT All groups except placebo without CBT improved.
No difference within other groups.
(Sharp et al., 1997)
Panic disorder with/without agoraphobia 12 weeks of sertraline or placebo treatment plus self-administered CBT or no CBT Reduced anticipatory anxiety in
sertraline plus self-administered CBT
No significant improvements in CGI.
(Koszycki et al., 2011)
Social anxiety disorder 24 weeks of sertraline/ placebo with or without exposure therapy (8 sessions in the first 12 weeks of treatment) All groups improved (also placebo without CBT)
Sertraline treatment (without CBT) showed higher improvement than CBT groups (placebo or sertraline). Placebo without CBT showed lowest benefits.
(Blomhoff et al., 2001)
Social anxiety disorder Follow-up study of (Blomhoff et al., 2001)
Assessment of long-term effects 28 weeks after cessation of medical treatment
Exposure therapy alone (without placebo or sertraline) showed a further improvement in CAPS 28 weeks after treatment cessation, however only reached improvement levels comparable with those of the sertraline alone group after the initial 24 weeks
CAPS score in the other groups (Exposure + placebo or sertraline and placebo alone) stayed constant (no improvement compared to 24 week CAPS score)
(Haug et al., 2003)
Social anxiety disorder 14 weeks of fluoxetine or placebo plus weekly CBT or no CBT
CBT consisted of group treatment combining in vivo exposure, cognitive restructuring and social skills training
All treatments were superior to placebo (without CBT) but no differences between groups themselves (Davidson et al., 2004)
PTSD 10 exposure therapy sessions
(1x / week) plus paroxetine CR
Optional 12 weeks of maintenance treatment
Greater CAPS improvement in paroxetine group vs placebo after 10 weeks
Higher rate of remission (61.5% vs 23.1% in placebo group) after 10 weeks
No changes after additional 12 weeks. CAVE drop-out of remitters.
(Schneier et al., 2012)
CR…controlled release
Table 2.Dopaminergic signaling in fear extinction (preclinical studies).
FACILITATING DA SIGNALING


DRUG/
MANIPULATION
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
L-DOPA ns +1 +
(SR, Re-in2
, Ren-A
ip (Haaker et al., 2013)
amphetamine ns ns ip (Borowski & Kokkinidis, 1998)
no effect ns ns ip (Carmack et al., 2010)
(+)3 (-) ns ip (Mueller et al., 2009)
cocaine ns ns ip (Borowski & Kokkinidis, 1998)
Methyl-
phenidate
+ (+) ns ip (Abraham et al., 2012)
ns +1 ns ip (Abraham et al., 2012)
SKF38393
(pAg D1)
ns ns ip (Borowski & Kokkinidis, 1998)
+4 ns ns ip (Dubrovina & Zinov’eva, 2010)
ns +1 ns CA1 (Fiorenza et al., 2012)
ns no effect1 ns BLA (Fiorenza et al., 2012)
ns no effect1 ns IL (Fiorenza et al., 2012)
ns +6 ns ip (Rey et al., 2014)
ns 7 ns ip (Rey et al., 2014)
Quinpirole
(D2 ag)
ns ns ip (Ponnusamy et al., 2005)
ns ns ip (Nader & LeDoux, 1999)
4 ns ns ip (Dubrovina & Zinov’eva, 2010)
INHIBITING DA SIGNALING
6-OH DOPA ns ns mPFC (Morrow et al., 1999)
(-) ns mPFC (Fernandez Espejo, 2003)
D1 knock-out ns ns global KO (El-Ghundi et al., 2001)
SCH23390
(D1 ant)
ns no effect1 ns IL (Fiorenza et al., 2012)
no effect ns BLA (Hikind & Maroun, 2008)
no effect ns IL (Hikind & Maroun, 2008)
ns 1 ns CA1 (Fiorenza et al., 2012)
ns no effect1 ns BLA (Fiorenza et al., 2012)
Sulpiride
(D2 ant)
ns ns ip (Dubrovina & Zinov’eva, 2010)
ns + ns ip (Ponnusamy et al., 2005)
no effect no effect ns ip (Mueller et al., 2010)
Raclopride
(D2 ant)
(-) no effect ns ip (Mueller et al., 2010)
no effect ns IL (Mueller et al., 2010)
Haloperidol
(D2/D3 ant)
ns ns ip (Holtzman-Assif et al., 2010)
(-) ns ns icv (Holtzman-Assif et al., 2010)
(-) ns NAcb (Holtzman-Assif et al., 2010)
L-741,741
(D4 ant)
no effect ns mPFC (Pfeiffer & Fendt, 2006)
+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; ns, not studied; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; mPFC, medial prefrontal cortex; CA1, cornu ammonis 1; NAcb, nucleus accumbens; Ren-A, Fear renewal in conditioning context; SR, spontaneous recovery; Re-in, reinstatement; ag, agonist; ant, antagonist, KO, knock-out;
1
drug administration following extinction training, 2 37 days, 3 increased locomotion, 4 passive avoidance paradigm, 5 reduced locomotion with 0.3mg/kg, no effect on locomotion, extinction training and retrieval with 0.1mg/kg, 6 rescued impaired extinction retrieval in estrus/metestrus/diestrus, 7 impairs extinction retrieval in proestrus
facilitates rescue of impaired fear extinction; # reduced fear expression at the beginning of extinction training; ## enhanced fear expression at the beginning of extinction training
Table 2A.Human trials: Dopaminergic enhancers combined with CBT.
Disorder Study Design Outcome (compared to placebo control group) Reference
Healthy volunteers Fear conditioning paradigm, L-DOPA po following extinction training Protection from renewal (skin conductance responses) (Haaker et al., 2013)
PTSD (treatment-refractory ) 2 single oral MDMA prior
to 8h therapy session (3-5 weeks apart, patients also received therapy prior to first MDMA exposure as well as between and in follow-up)
MDMA augmented CBT in comparison to placebo (83% vs 25% response)
(2 month follow-up); open-label MDMA use offered to patients from the placebo group after last assessment
(Mithoefer et al., 2011)
PTSD (treatment-refractory ) Follow-up study from (Mithoefer et al., 2011) MDMA augmented CBT produced long-lasting effects (17-74 months) (Mithoefer et al., 2013)
Table 3.Noradrenergic (NE) signaling in fear extinction (preclinical studies).
ENHANCING NORADRENERGIC SIGNALING


DRUG/
MANIPULATION
EXTINCTION
LEARNING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
noradrenaline ns + ns icv (Merlo & Izquierdo, 1967)
methylphenidate + + ns ip (Abraham et al., 2012)
noradrenaline ns +1 ns BLA (Berlau & McGaugh, 2006)
ns no effect1 ns BLA (Fiorenza et al., 2012)
ns no effect1 ns CA1 (Fiorenza et al., 2012)
ns 1 ns IL (Fiorenza et al., 2012)
atomoxetine (NE reuptake inhibitor) ns no effect6 +
(SR)5
systemic (Janak & Corbit, 2011)
isoproterenol (β ag) no effect ns ns ip (subchronic) (Do-Monte et al., 2010b)
ns (+)1 ns ip (subchronic) (Do-Monte et al., 2010b)
ns (+)1 ns mPFC (Do-Monte et al., 2010b)
yohimbine (α2 ant)8 + ns ns sc (Cain et al., 2004)
ns no effect1 ns sc (Cain et al., 2004)
+#2 +2 ns sc (Cain et al., 2004)
+ + ip (Morris & Bouton, 2007)
no effect7 no effect ns ip (Mueller et al., 2009)
ns +* ns ip (Hefner et al., 2008)
ns no effect6 +
(SR)5
systemic (Janak & Corbit, 2011)
atipamezole (α2 ant)8 ns no effect4 ns sc (Davis et al., 2008)
INHIBITING NORADRENERGIC SIGNALING
prazosine (α1 ant) ns no effect1 ns ip (subchronic) (Do-Monte et al., 2010a)
ns ns ip (subchronic) (Do-Monte et al., 2010a)
ns ns intra-mPFC (Do-Monte et al., 2010a)
propanolol (β ant) no effect ns ns sc (Cain et al., 2004)
ns no effect1 ns sc (Cain et al., 2004)
no effect2 (-)3 ns sc (Cain et al., 2004)
(+)# no effect ns ip (Rodriguez-Romaguera et al., 2009)
ns ip (subchronic) (Do-Monte et al., 2010b)
ns no effect1 ns ip (subchronic) (Do-Monte et al., 2010b)
atenolol (β ant) ns ns mPFC (Do-Monte et al., 2010b)
ns no effect1 ns mPFC (Do-Monte et al., 2010b)
sotalol (β ant) no effect no effect ns ip (Rodriguez-Romaguera et al., 2009)
timolol (β ant) ns +1 ns BLA (Fiorenza et al., 2012)
propanolol (β ant) no effect ns IL (Mueller et al., 2008)
ns no effect1 ns IL (Mueller et al., 2008)
timolol (β ant) ns +1 ns IL (Fiorenza et al., 2012)
ns no effect1 ns CA1 (Fiorenza et al., 2012)
* facilitates rescue of impaired fear extinction; ** facilitates extinction of remote memories, *** only in older animals (7 months), younger ones (2 months) are not affected, # reduced fear expression at the beginning of extinction training+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; injection; sc, subcutanous injection; icv, intracerebroventricular injection; ns, not studied; HPC, intra-hippocampal administration; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; PL, prelimbic cortex; CA1, cornu ammonis 1; Ren, Fear renewal; SR, spontaneous recovery; Re-in, reinstatement; ag, agonist; ant, antagonist, KO, knock-out; #, reduced fear expression at start of extinction training
1
drug administration following extinction training; 2 spaced CS extinction, US=0.7mA; 3 spaced CS extinction when US=0.4mA; no effect when US=0.7mA, 4 cocaine-conditioned place preference, 5 4 weeks, 6 instrumental lever press response, 7 reduced fear expression at start of extinction training, if extinction is performed in the same context as conditioning, 8 note that the net effect of α2 blockade is enhancement of noradrenergic signaling (see text)
Table 3A.Yohimbine combined with CBT in small-scale clinical trials.
Disorder Study Design Outcome (compared to placebo control group) Reference
Social anxiety disorder (SAD) 4 sessions consisting of yohimbine being administered prior to a CBT asession Yohimbine augmented CBT-induced improvement in SAD (self-report measures; 21 day follow-up) (Smits et al., 2014)
Claustrophobic fear 1 single oral yohimbine dose prior to an exposure b session Yohimbine augment CBT-induced reduced fear of enclosed spaces (7 day follow-up) (Powers et al., 2009)
Fear of flying 2 single oral yohimbine prior to VRET session No fear augmenting effect of yohimbine c (Meyerbroeker et al., 2012)
VRET, virtual reality exposure.
a
CBT involved an oral presentation on challenging topics in front of the therapists, other group members, and confederates,
b
behavioral approach task,
c
lack of yohimbine effect might possibly have been due to the powerful influence of VRET exposure itself leading to a greater impact on the results from this than from the manipulation (drug condition) that ‘washed out’ or overrode any effect of manipulation (Meyerbroeker et al., 2012),
Table 4.Glutamatergic signaling in fear extinction (preclinical studies).
FACILITATING AMPA SIGNALING


DRUG/
MANIPULATION
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
PEPA (AMPA potentiator) no effect no effect ns ip (Whittle et al., 2013)
no effect + ns ip (Yamada et al., 2011)
+# + +
(Re-in)
ip (Yamada et al., 2009)
+ + ns ip (Zushida et al., 2007)
PEPA + NBQX (AMPA ant) no effect no effect ns ip (Zushida et al., 2007)
PEPA (AMPA potentiator) (+) (+) ns PL (Zushida et al., 2007)
+ + ns BLA/ceA (Zushida et al., 2007)
INHIBITING AMPA SIGNALING
CNQX
(AMPA ant)
ns no effect ns BLA (Falls et al., 1992; Lin et al., 2003b; Zimmerman & Maren, 2010)
FACILITATING NMDA SIGNALING
D-cycloserine ns + ns ip (Walker et al., 2002)
ns +1 ns systemic (Ledgerwood et al., 2003)
ns +1 +
(Re-in)
systemic (Ledgerwood et al., 2004)
ns +1 ns systemic (Ledgerwood et al., 2005)
ns +1 ns ip (Parnas et al., 2005)
ns + ns ip (Yang & Lu, 2005)
ns + ns ip (Lee et al., 2006)
ns + no effect sc (Woods & Bouton, 2006)
ns + ns sc (Werner-Seidler & Richardson, 2007)
ns + ns systemic (Weber et al., 2007)
ns + ns ip (Yang et al., 2007)
ns + ns ip (Yang et al., 2007)
ns + ns ip (Hefner et al., 2008)
ns + ns po (Yamamoto et al., 2008)
ns + ns ip (Matsumoto et al., 2008)
+ + ns ip (Silvestri & Root, 2008)
ns + ns sc (Langton & Richardson, 2008)
ns + no effect sc (Bouton et al., 2008)
ns + ns ip (Lin et al., 2010)
+# + +
(Re-in)
ip (Yamada et al., 2009)
+ ns ns ip (Lehner et al., 2010)
ns +1 ns systemic (Langton & Richardson, 2010)
no effect + ns ip (Yamada et al., 2011)
no effect + ns ip (Toth et al., 2012b)
ns +1 ns ip (Toth et al., 2012b)
ns + ns systemic (Gupta et al., 2013)
ns + ns ip (Matsuda et al., 2010)
+ + ns ip (Bai et al., 2014)
ns +1 ns ip (Whittle et al., 2013)
ns + ns BLA (Walker et al., 2002)
ns +1 ns BLA (Ledgerwood et al., 2003)
ns + ns BLA (Lee et al., 2006)
ns + ns BLA (Mao et al., 2008)
ns + ns BLA (Lee et al., 2006)
ns + ns BLA (Akirav et al., 2009)
ns no effect ns BLA (Bolkan & Lattal, 2013)
ns + ns HPC (Bolkan & Lattal, 2013)
ns + ns HPC (Ren et al., 2013)
spermidine ns +1 ns HPC (Gomes et al., 2010)
INHIBITING NMDA SIGNALING
MK-801 (non-competitive NMDA ant) ns ns systemic (Baker & Azorlosa, 1996; Storsve et al., 2010)
ns ns +
(Re-in)
sc (Johnson et al., 2000)
ns 1 ns ip (Lee et al., 2006; Liu et al., 2009)
ns ns systemic (Langton et al., 2007; Chan & McNally, 2009)
CPP (competitive NMDA ant) no effect 5 ns ip (Santini et al., 2001; Sotres-Bayon et al., 2007)
no effect ns mPFC (Burgos-Robles et al., 2007)
ns 1 ns mPFC (Burgos-Robles et al., 2007)
ns BLA (Parsons et al., 2010)
AP-5 (competitive NMDA ant) ns ns BLA (Falls et al., 1992)
## ns BLA (Lee & Kim, 1998)
ns 1 ns BLA (Lin et al., 2003a; Laurent et al., 2008) (Fiorenza et al., 2012)
no effect ns BLA (Lin et al., 2003a; Zimmerman & Maren, 2010)
ns ns CA1 (Szapiro et al., 2003)
ns 1 ns CA1 (Fiorenza et al., 2012)
ns 1 ns mPFC (Fiorenza et al., 2012)
ifenprodil (non-comp NR2B-NMDA ant) ns ip (Sotres-Bayon et al., 2007)
ns 1 ns systemic Sotres-Bayon et al., 2009
ns BLA (Sotres-Bayon et al., 2007; Laurent et al., 2008; Laurent & Westbrook, 2008)
ns no effect1 ns BLA (Laurent et al., 2008; Laurent & Westbrook, 2008; Sotres-Bayon et al., 2009)
no effect ns mPFC (Laurent & Westbrook, 2008)
no effect no effect ns mPFC (Sotres-Bayon et al., 2009)
no effect 1 ns mPFC (Laurent & Westbrook, 2008; Sotres-Bayon et al., 2009)
ns 1 ns HPC (Gomes et al., 2010)
Ro25-6981 (non-comp NR2B-NMDA ant) no effect ns ip (Dalton et al., 2008; Dalton et al., 2012)
FACILITATING mGluR SIGNALING
CDPPB (mGluR5 pos modulator) +3 ns ns sc (Gass & Olive, 2009)
AMN082 (mGluR7 ag) + + ns ip (Whittle et al., 2013)
ns + ns po (Fendt et al., 2008)
no effect ns ip (Toth et al., 2012b)
ns 1 ns ip (Toth et al., 2012b)
(+) no effect ns BLA (Dobi et al., 2013)
ns mPFC (Morawska & Fendt, 2012)
INHIBITING mGluR SIGNALING
CPCCOEt (non-comp mGluR1 ant) ns BLA (Kim et al., 2007)
no effect no effect no effect
(SR)
ip (Mao et al., 2013)
mGluR4 KO ns + ns no drug (Davis et al., 2013)
mGluR5 KO ns no drug (Xu et al., 2009)
MTEP (mGluR5 ant) no effect
(SR)
ip (Mao et al., 2013)
ns ns
(SR)
BLA (Mao et al., 2013)
MPEP (allosteric mGluR5 ant) no effect no effect ns ip (Toth et al., 2012b)
ns no effect1 ns ip (Toth et al., 2012b)
no effect ns ip (Fontanez-Nuin et al., 2011)
no effect ns IL (Fontanez-Nuin et al., 2011)
ns IL (Sepulveda-Orengo et al., 2013)
mGluR7 KO 4 ns ns no drug (Callaerts-Vegh et al., 2006; Goddyn et al., 2008)
+ + ns no drug (Fendt et al., 2013)
siRNA knock-down of mGluR7 2 ns ns no drug (Fendt et al., 2008)
mGluR8 KO no effect# no effect ns no drug (Fendt et al., 2013)
(S)-3,4-DCPG (mGluR8 ag) no effect ns BLA (Dobi et al., 2013)
+, improved; -, impaired; (+) or (-), only minor effects; po, peroral administration; ip, intraperitoneal injection; sc, subcutaneous injection; ns, not studied; BLA, intra-basolateral amygdala administration; HPC, hippocampus; IL, infralimbic cortex; SR, spontaneous recovery; Re-in, reinstatement; ag, agonist; ant, antagonist, KO, knock-out;
1
drug administration following extinction training; 2conditioned taste aversion; 3 cocaine conditioned place preference; 4 food-rewarded operant conditioning, 5 reduced locomotion
facilitates rescue of impaired fear extinction; # reduced fear expression at the beginning of extinction training; ## enhanced fear expression at the beginning of extinction training
Table 4A.Human trials: D-cycloserine (DCS) combined with CBT.
Disorder Study Design Outcome (compared to placebo control group) Reference
Healthy volunteers DCS or placebo 2-3h prior extinction training No effect on fear extinction or fear recovery in healthy volunteers (Guastella et al., 2007)
Healthy volunteers DCS, placebo, valproic acid or combination of DCS and valproic acid 1.5h prior extinction training Administration of DCS, valproic acid or DCS/valproic acid combination facilitates fear extinction and protects from reinstatement (Kuriyama et al., 2011)
Healthy volunteers DCS or placebo 2h prior extinction training No effect on extinction learning and retention in healthy volunteers (Klumpers et al., 2012)
Acrophobia DCS or placebo 1h prior 2 sessions of virtual reality exposure
3 month follow-up
reduced fear symptoms in DCS group at all timepoints (Ressler et al., 2004)
Acrophobia 2 sessions virtual reality exposure plus DCS or placebo after the sessions
1 month follow-up
Re-evaulation of Tart et al., 2013
no advantage of DCS over placebo augmented VRE
DCS effect depends on success of exposure sessions (within-session fear reduction)
(Tart et al., 2013)
(Smits et al., 2013a)
Snake phobia DCS or placebo 1h prior a single exposure session same level of improvement with DCS, but DCS group achieved fear reduction quicker (Nave et al., 2012)
Panic disorder DCS or placebo 1h prior CBT session 3-5
1 month follow-up
DCS group showed greater reduction in panic symptom severity at all timepoints (Otto et al., 2010c)
Panic disorder with agoraphobia DCS or placebo 1h prior 3 individual exposure sessions + 8 group CBT sessions
5 months follow-up
no benefits of DCS at all timepoints, but initial benefical effects in severely symptomatic patients? (Siegmund et al., 2011)
PTSD DCS or placebo 1h prior 10 weekly exposure sessions No overall enhancement of treatment effects.
Higher symptom reduction in severe cases.
(de Kleine et al., 2012)
PTSD
(combat-related)
DCS or placebo 30min prior exposure session 2-6 Weaker symptom reduction compared to placebo group (Litz et al., 2012)
PTSD DCS or placebo 1.5h prior 12 weekly VRE session
6 months follow-up
DCS group showed earlier and greater improvement as well as higher remission rates (Difede et al., 2014)
pediatric PTSD DCS or placebo 12 1h prior session 5-12 No difference in symptom reduction, DCS group showed trend for faster response and better retention in 3 month follow-up (Scheeringa & Weems, 2014)
SAD 1x psychoeducation; DCS or placebo 1h prior to 4h exposure session (5x, individual or group)
1 month follow-up
Decreased self-reported social anxiety symptoms in DCS group after 1 month (Hofmann et al., 2006)
SAD DCS or placebo 1h prior to 4h exposure therapy (5x) Fewer social fear and avoidance in DCS group. Significant differences in DCS vs placebo following 3rd exposure session. (Guastella et al., 2008)
SAD DCS or placebo with CBT Greater overall rates of improvement and lower post-treatment severity (Smits et al., 2013b)
SAD 12 weeks; DCS or placebo 1h prior 5 exposure sessions (12 sessions at all); 6-months follow-up similar response and remission rates, DCS group improved quicker (Hofmann et al., 2013b)
OCD D-cycloserine (DCS) or placebo 4h prior each exposure session, 12 weeks (one exposure session/week) No statistically significant difference.
High response rates in treatment and placebo group.
(Storch et al., 2007)
OCD 2x/week DCS or placebo 2h prior exposure and response prevention (ERP) sessions
(max 10)
3 months follow-up
Faster improvements in DCS group
No difference between DCS and placebo group (no further benefit).
(Kushner et al., 2007)
OCD 2x/week DCS or placebo 1h prior 10 ERP sessions
1 month follow-up
Re-evaluation of Wilhelm et al., 2008
Faster improvements in DCS group
No difference between DCS and placebo group (no further benefit).
specific improvements of DCS in the first 5 sessions, 6x faster than placebo group
(Wilhelm et al., 2008)
(Chasson et al., 2010)
Pediatric OCD DCS or placebo 1h prior weekly session 4-10 (psychoeduction, cognitive training and ERP) for 10 wks modest reduction of obsessive symptoms (Storch et al., 2010)
Pediatric OCD DCS or placebo 1h prior session 5-9 ERP-CBT Significant improvements in OCD severity from posttreatment to 1-month follow-up in severe and difficult-to-treat pediatric OCD (Farrell et al., 2013)
Pediatric OCD DCS or placebo immediately after each of 10 CBT (ERP) sessions
1 year follow-up
both groups improved;
no significant advantage of DCS at any timepoint
(Mataix-Cols et al., 2014)
Table 5.GABAergic signaling in fear extinction (preclinical studies).
FACILITATING GABA SIGNALING


DRUG/
MANIPULATION
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
GABAA Agonists GABA-binding site (αβ interface)
muscimol +# no effect ns BLA (Akirav et al., 2006)
ns +1,7 ns BLA (Akirav et al., 2006)
# ns BLA (Laurent et al., 2008; Laurent & Westbrook, 2008)
ns 1 ns BLA (Laurent & Westbrook, 2008)
(+)# ns BLA (Sierra-Mercado et al., 2011)
ns mPFC (Laurent & Westbrook, 2008)
ns 1 ns mPFC (Laurent & Westbrook, 2008)
ns no effect1, ns IL (Akirav et al., 2006)
+# (+) ns IL (Akirav et al., 2006)
ns IL (Sierra-Mercado et al., 2011)
no effect ns IL (Laurent & Westbrook, 2009)
(+)# no effect ns PL (Laurent & Westbrook, 2009; Sierra-Mercado et al., 2011)
ns ns +
(Ren-A)8
dHPC (Corcoran et al., 2005)
no effect
(Ren-A)
dHPC (Corcoran et al., 2005)
(+)# ns vHPC (Sierra-Mercado et al., 2011)
GABAA: Agonists BZD-binding site αγ interface)
chlordiazepoxide ns ns ip (Goldman, 1977)
ns ns ip (Bouton et al., 1990)
diazepam ns ns ip (Pereira et al., 1989)
ns ns ip (Bouton et al., 1990)
midazolam ns 1 ns ip (Bustos et al., 2009)
##6 ns ip (Hart et al., 2009, 2010; Hart et al., 2014)
#6 ns BLA (Hart et al., 2009, 2010; Hart et al., 2014)
GABAB receptor agonists
baclofen ns ns ip (Heaney et al., 2012)
GS39783 (positive modulator GABAB) ns no effect ns po (Sweeney et al., 2013)
INHIBITING GABA SIGNALING
DRUG EXTINCTION
LEARNING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
GAD67 KD in BLA ns ns no drug (Heldt et al., 2012)
GAD65 KO 2 2 ns no drug (Sangha et al., 2009)
α5 KD in HPC ns 3 ns no drug (Yee et al., 2004)
α1 KO in CRF+ cells ns no drug (Gafford et al., 2012)
picrotoxin ns + ns ip (McGaugh et al., 1990)
ns +4 ns IL (Thompson et al., 2010)
ns +4 ns PL (Thompson et al., 2010)
ns +* ns (Fitzgerald et al., 2014b)
bicuculline ns +1 ns BLA (Berlau & McGaugh, 2006)
bicuculline + propanolol ns no effect1 ns BLA (Berlau & McGaugh, 2006)
FG7142 ns ip (Harris & Westbrook, 1998)
ns ns ip (Kim & Richardson, 2007, 2009)
GABAB receptor antagonism
GABAB(1b) KO 5 ns ns no drug (Jacobson et al., 2006)
phaclofen ns no effect ns ip (Heaney et al., 2012)
CGP52432 ns no effect ns po (Sweeney et al., 2013)
1 drug administration following extinction training; 2 only cued memory, contextual intact, 3 trace conditioning, 4 reconsolidation (injection prior reactivation of memory), 5 conditioned taste aversion, 6 midazolam may not impair fear extinction if initial extinction occurs drug-free [see (Hart et al., 2014)], 7 short extinction training (5 CS presentations), 8 injection prior renewal-testing# reduced fear expression at start of extinction training; ## enhanced immobility at start of extinction training;
+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; po,peroral administration; ns, not studied; HPC, intra-hippocampal administration; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; PL, prelimbic cortex; Ren-A, Fear renewal in conditioning context; ag, agonist; ant, antagonist, KO, knock-out;
Table 6.Cannabinoid signaling in fear extinction (preclinical studies).
ENHANCED CANNABINOID SIGNALING


DRUG/
MANIPULATION
EXTNCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM EXTNCTION ROUTE REFERENCE
cannabidiol + + ns icv (Bitencourt et al., 2008)
+ + ns IL (Do Monte et al., 2013)
anandamide ns +1 ns dCA1 (de Oliveira Alvares et al., 2008)
AM404
(eCB uptake inhibitor)
ns + ns ip (Pamplona et al., 2008)
ns + +
(Re-in)
ip (Chhatwal et al., 2005b)
+ (+) ns icv (Bitencourt et al., 2008)
no training + ns IL (Lin et al., 2009)
AM3506
(FAAH
Inhibitor)
ns + ns ip (Gunduz-Cinar et al., 2013b)
WIN 55,212-2
(low-dose CB1 Ag)
+ no effect2 ns ip (Pamplona et al., 2006)
WIN 55,212-2
(high-dose CB1 Ag)
(-) no effect2 ns ip (Pamplona et al., 2006)
WIN 55,212-2
(CB1 Ag)
ns ip (Pamplona et al., 2006)
ns + nd ip (Pamplona et al., 2008)
WIN 55,212-2 (CB1 Ag) ns +4 ns BLA (Ganon-Elazar & Akirav, 2013)
ns +4 ns HPC (Ganon-Elazar & Akirav, 2013)
ns no effect4 ns mPFC (Ganon-Elazar & Akirav, 2013)
ns + no effect
(Re-in, SR3)
IL (Lin et al., 2009)
no training + ns IL (Lin et al., 2009)
ns no effect ns PL (Kuhnert et al., 2013)
HU210 no training + ns IL (Lin et al., 2009)
URB597 (AEA hydrolysis
inhibitor)
no training + ns IL (Lin et al., 2009)
AM3506 (FAAH
Inhibitor)
+ SR141716A (CB1 Ant)
ns no effect ns ip + BLA (Gunduz-Cinar et al., 2013b)
AM3506
(FAAH
Inhibitor)
ns + ns BLA (Gunduz-Cinar et al., 2013b)
no training no effect ns BLA (Gunduz-Cinar et al., 2013b)
cannabidiol
+ SR141716A (CB1 Ant)
no effect no effect ns IL + ip (Do Monte et al., 2013)
REDUCED CANNABINOID SIGNALING
DRUG/
MANIPULATION
EXTNCTION
LEARNING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ADMINISTRATIONROUTE REFERENCE
CB1 KO ns no drug (Cannich et al., 2004)
CB1 KO ns ns no drug (Marsicano et al., 2002; Kamprath et al., 2006; Dubreucq et al., 2010; Plendl & Wotjak, 2010)
D1-CB1 KO ns ns no drug (Terzian et al., 2011)
SR141716A
CB1 ant
ns no effect1 ns sc (Marsicano et al., 2002)
ns ns sc, ip (Kamprath et al., 2006; Niyuhire et al., 2007; Plendl & Wotjak, 2010)
ns 1 ns ip (Suzuki et al., 2004)
no effect ns ip (Pamplona et al., 2006; Pamplona et al., 2008)
ns ns ip (Chhatwal et al., 2005b)
## no effect ns ip (Bowers & Ressler, 2014)
AM251
(CB1 inverse
ag)
ns ns ip (Reich et al., 2008)
ns 1 ns dCA1 (de Oliveira Alvares et al., 2008)
ns ns IL (Lin et al., 2009)
no training no effect ns IL (Lin et al., 2009)
ns ns PL (Kuhnert et al., 2013)
+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; ip, sc, subcutanous injection; ns, not studied; HPC, intra-hippocampal administration; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; PL, prelimbic cortex; CA1, cornu ammonis 1; Re-in, reinstatement; SR, spontaneous recovery, ag, agonist; ant, antagonist, KO, knock-out;
1
drug administration following extinction training; 2 drug-free retrieval of contextual fear memory; 3 5 days, 4 single-prolonged-stress model: injection immediately after trauma
, Recent memory; °, remote memory; ##, enhanced immobility at start of extinction training;
Table 6A.Human trials: Cannabinoids combined with CBT.
Disorder Study Design Outcome (compared to placebo control group) Reference
Healthy volunteers Fear conditioning paradigm, Cannabidiol inhalation prior or following extinction Reduced fear in retrieval
Protection against reinstatement
(Das et al., 2013)
Healthy volunteers Fear conditioning paradigm, tetrahydro-cannabinol prior extinction No effect (Klumpers et al., 2012)
Healthy volunteers Fear conditioning paradigm,
Dronabinol prior extinction
Reduced fear in retrieval (Rabinak et al., 2013)
Table 7.Neuropeptide signaling in fear extinction (preclinical studies).
NEUROPEPTIDES


DRUG/
MANIPULATION
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
oxytocin ns ns icv (Toth et al., 2012a)
+# + ns DLS (Zoicas et al., 2014)
-*5 -*5 ns ip (Eskandarian et al., 2013)
chronic oxytocin (30d) ns no effect*6 ns in (Bales et al., 2014)
NPY + + ns icv (Gutman et al., 2008)
NPY KO 3⁎⁎⁎ ns no drug (Verma et al., 2012)
NPY Y1 KO no effect ns no drug (Verma et al., 2012)
NPY Y2 KO no effect no effect ns no drug (Verma et al., 2012)
NPY Y1/Y2 KO ns no drug (Verma et al., 2012)
Leu31Pro-NPY
(Y1 ag)
ns + ns BLA (Lach & de Lima, 2013)
BIBO 3304 (Y1 ant) ns ns BLA (Gutman et al., 2008)
NPS (+)4 no effect no effect BLA (Jungling et al., 2008)
+ ns ns icv (Slattery et al., in press)
SHA68 (NPS-R ant) 4,⁎⁎⁎
(Ren-A)
BLA (Jungling et al., 2008)
naloxone ns systemic (McNally & Westbrook, 2003)
ns no effect1 ns systemic (McNally & Westbrook, 2003)
ns vlPAG (McNally et al., 2004)
ns vlPAG (Parsons et al., 2010)
no effect no effect ns BLA (Parsons et al., 2010)
CTAP (MOR ant) ns vlPAG (McNally et al., 2005)
dynorphin KO – 3 ns no drug (Bilkei-Gorzo et al., 2012)
nor-BNI (KOR ant) ns 1 ns systemic (Bilkei-Gorzo et al., 2012)
+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; icv, intra-cerebroventricular injection; ns, not studied; HPC, intra-hippocampal administration; BLA, intra-basolateral amygdala administration; DLS, dorsolateral septum; vlPAG, ventrolateral periaqueductal gray; Ren-A, fear renewal in conditioning context; ag, agonist; ant, antagonist, KO, knock-out;
1
drug administration following extinction training; 2 social fear conditioning;3 animals show accelerated fear learning; 4 administration 2h prior extinction training; 5 single prolonged stress model; 6 BTBR mouse autism model and C57BL6J
⁎⁎⁎
enhanced fear expression at the beginning of extinction training, # reduced fear expression at the beginning of extinction training
Table 8.Glucocorticoid signaling in fear extinction (preclinical studies).
FACILITATING GLUCOCORTICOID SIGNALING


DRUG EXTINCTION TRAINING EXTINCTION RETRIEVAL LONG-TERM EXTINCTION ROUTE REFERENCE
corticosterone (GR/MR ag) ns +1 no effect
(Re-in)
ip (Blundell et al., 2011)
ns 2 ns ip (Den et al., 2014)
dexamethasone (GR ag) ns + ns ip (Yang et al., 2006; Yang et al., 2007; Ninomiya et al., 2010)
RU28362 (GR ag) ns + ns AMY (Yang et al., 2006)
ganoxolone (allopregnanolone analog) +*3 +*3 ns ip (Pinna & Rasmusson, 2014)
INHIBITING GLUCOCORTICOID SIGNALING
metyrapone (CORT/cortisol synthesis inhibitor) no effect -* sc (Barrett & Gonzalez-Lima, 2004; Clay et al., 2011)
ns ns sc (Yang et al., 2006; Yang et al., 2007; Blundell et al., 2011)
mifepristone (GR and progesterone ant) ns ns AMY (Yang et al., 2006)
+, improved; -, impaired; ip, intraperitoneal injection; sc, subcutaneous injection; ns, not studied; AMY, intra-amygdala administration; Re-in, reinstatement; ag, agonist; ant, antagonist; GR, glucocorticoid receptor; MR, mineralocorticoid receptor; CORT, corticosteroid
1
drug administration following extinction training; 2 in adolescent but not adult rats if exposed to CORT one week prior to (drugfree) testing; adults also show impaired extinction retrieval if they were exposed to one week of CORT in their adolescence; 3 socially isolated mice, CAVE ganoxolone was administered following a reactivation session 24h prior extinction training
Table 8A.Human trials: Glucocorticoids combined with CBT.
Disorder Study Design Outcome (compared to placebo control group) Reference
Social phobia Cortisone or placebo administered orally 1 h before a socio-evaluative stressor Reduced self-reported fear during anticipation and exposure (Soravia et al., 2006)
Spider phobia Cortisol or placebo administered orally 1 h before exposure to a spider photograph in 6 sessions distributed over 2 weeks Progressive reduction of stimulus-induced fear
Reduction of fear was maintained also 2 days following session ending
(OFF-drug)
(Soravia et al., 2006)
Spider phobia Cortisol or placebo administered orally 1 h before 2 exposure therapy sessions
Follow-up after 1 month
Significant decrease in phobic symptoms assessed with the Fear of Spiders Questionnaire
Cortisol-treated patients reported significantly less anxiety during exposure to living spiders at follow-up
(Soravia et al., 2014)
Acrophobia Cortisol or placebo was administered orally 1 h before virtual reality exposure to heights
Follow-up after 1 month
Greater reduction in fear of heights measured with the acrophobia questionnaire both at post-treatment and at follow-up (de Quervain et al., 2011)
PTSD (combat-related ) Memory reactivation task followed by intravenous administration of cortisol or saline
Follow-up after 1 month
Reduced PTSD symptomatic in cortisol-treated patients
No differences between cortisol and saline treated patients (initial improvement was lost after 1 month)
(Suris et al., 2010)
PTSD 10 weekly sessions of prolonged exposure therapy, cortisol or placebo 30min prior session
3-10
Accelerated and greater decline in PTSD symptoms
Caveat – includes only 2 patients
(1 cortisol / 1 placebo)
(Yehuda et al., 2010)
Table 9.Neurotrophins and miscellaneous targets in fear extinction (preclinical studies).
NEUROTROPHINS


DRUG/
MANIPULATION
EXTINCTION LEARNING EXTINCTION RETRIEVAL LONG-TERM EXTINCTION ROUTE REFERENCE
FGF2 (+)# + ns sc (Graham & Richardson, 2009)
ns +1 +
(Re-in)
(Ren-A)
sc (Graham & Richardson, 2009, 2010)
ns +1 +
(Ren-A)
BLA (Graham & Richardson, 2011b)
BDNF no training +**2 no effect (Re-in) IL (Peters et al., 2010; Rosas-Vidal et al., 2014a)
no training no effect2 ns PL (Rosas-Vidal et al., 2014a)
7.8-dihydroxyflavone (+)* no effect (+)
(Re-in)
ip (Andero et al., 2011)
no effect no effect* +
(Ren-A)
ip (Baker-Andresen et al., 2013)
Lentiviral transfected dominant negative form of TrkB no effect ns BLA (Chhatwal et al., 2006)
BDNF KD in HPC ns ns no drug (Heldt et al., 2007)
Val66Met BDNF SNP ns ns no drug (Soliman et al., 2010)
BDNF +/- KO -*** -*** ns no drug (Psotta et al., 2013)
BDNF antibody ns IL (Rosas-Vidal et al., 2014a)
no effect no effect ns PL (Rosas-Vidal et al., 2014a)
BDNF KO in forebrain ns no effect# ns PL (Choi et al., 2010)
1 drug administration following extinction training; 2 drug administration 24h prior to extinction retrieval
3 drug administration 30 min prior to extinction retrieval 4 ABA scheme, 40mg/kg
METHYLENE BLUE, NITRIC OXIDE, HISTAMINE, and LTCCs
DRUG/
MANIPULATION
EXTINCTION LEARNING EXTINCTION RETRIEVAL LONG-TERM EXTINCTION ROUTE REFERENCE
methylene blue ns +1 ns ip (Gonzalez-Lima & Bruchey, 2004)
ns ns +1*
(Ren-A)
ip (Wrubel et al., 2007)
Histamine ns +1 ns CA1 (Bonini et al., 2011)
dimaprit (H2 ag) ns +1 ns CA1 (Bonini et al., 2011)
ranitidine (H2 ant) ns 1 ns CA1 (Fiorenza et al., 2012)
ns 1 ns BLA (Fiorenza et al., 2012)
ns 1 ns PFC (Fiorenza et al., 2012)
SKF9188 (histamine methyl-transferase inhibitor) ns +1 ns CA1 (Fiorenza et al., 2012)
ns +1 ns BLA (Fiorenza et al., 2012)
ns +1 ns PFC (Fiorenza et al., 2012)
L-NAME (nNOS inhibitor)4 ns ip (Luo et al., 2014)
CamKII-Cre Cav1.2 KO no effect ns ns no drug (McKinney et al., 2008)
Cav1.3 KO no effect ns ns no drug (Busquet et al., 2008)
no effect ns ns no drug (McKinney & Murphy, 2006)
Nifedipine (LTCC ant) ns ns ip (Cain et al., 2002)
no effect ns ns icv (Busquet et al., 2008)
no effect ns BLA (Davis & Bauer, 2012)
ns 1 ns HPC (de Carvalho Myskiw et al., 2014)
Verapamil (VGCC ant) no effect ns BLA (Davis & Bauer, 2012)
* facilitates rescue of impaired fear extinction; ** facilitates extinction of remote memories, *** only in older animals (7 months), younger ones (2 months) are not affected, # reduced fear expression at the beginning of extinction training+, improved; -, impaired; (+) or (-), only minor effects; ip, intraperitoneal injection; sc, subcutanous injection; icv, intracerebroventricular injection; ns, not studied; HPC, intra-hippocampal administration; BLA, intra-basolateral amygdala administration; IL, infralimbic cortex; PL, prelimbic cortex; CA1, cornu ammonis 1; Ren-A, Fear renewal in conditioning context; SR, spontaneous recovery; Re-in, reinstatement; ag, agonist; ant, antagonist, KO, knock-out;
1
drug administration following extinction training; 2 drug administration 24h prior extinction retrieval
3
drug administration 30 min prior extinction retrieval 4 ABA scheme, 40mg/kg
Table 10.Studies showing that HDAC inhibitors augment exposure-based fear extinction and rescue extinction learning deficits.
EXTINCTION
TRAINING
EXTINCTION
RETRIEVAL
LONGTERM
EXTINCTION
ROUTE REFERENCE
HDAC1 KO (HPC) ns – # ns no drug (Bahari-Javan et al., 2012)
HDAC2 KO in forebrain CamKII neurons ns + *# ns no drug (Morris et al., 2013)
Vorinostat/SAHA ns + ns ip (Fujita et al., 2012)
ns + ** ns ip (Matsumoto et al., 2013)
ns + ** ns ip (Hait et al., 2014)
TSA (Trichostatin A) ns + * ns ip, HPC (Lattal et al., 2007)
Sodium Butyrate + +
(SR)
ip, HPC (Stafford et al., 2012)
ns + ns ip (Itzhak et al., 2012)
ns + ns ip, HPC (Lattal et al., 2007)
Valproic Acid + * ns ip (Bredy et al., 2007)
+ * +
(Ren-A)
ip (Bredy & Barad, 2008)
+ ** + ** +
(SR,
Ren-N)
ip (Whittle et al., 2013)
MS-275 (Entinostat) + ** +
(SR,
Ren-N)
ip (Whittle et al., 2013)
Cl-994 ns + *** +
(SR)
ip (Graff et al., 2014)
FTY720 (Fingolimod) + ** ns ip (Hait et al., 2014)
* partial extinction training: reduction of fear during the extinction training session was not to pre-conditioning levels, ** facilitates rescue of impaired fear extinction; *** facilitate remote memories combined with memory re-consolidation update paradigm; # extinction protocol based on a re-consolidation/update paradigm+, improved; -, impaired; ip, intraperitoneal injection; ns, not studied; HPC, intra-hippocampal administration; Ren-A, Fear renewal in the conditioning context; Ren-N, Fear renewal in a novel context; SR, spontaneous recovery; # reduced freezing levels at the beginning of extinction training
Box 1.Why pharmacological augmentation of extinction?
A limitation of extinction-based exposure therapy is that patients can often relapse with the passage of time, with changes in context (out of the therapy context), or under conditions of stress or other provocations, such as experiencing trauma reminders. In the parlance of learning theory, extinction memories are labile and fragile. A key goal for pharmacotherapy, therefore, is to identify compounds that overcome this fragility, by bolstering the formation, persistence and possibly context independence of extinction memories. One approach to achieving this goal is to use drugs as adjuncts to exposure therapy (cognitive enhancers) as a way to augment the extinction learning process. In this review, we discuss the various neurochemical and molecular signalling pathways that have been targeted to this end. On the one hand, there remain significant challenges to overcome, including the selective targeting of extinction-related processes without concurrent effects on original fear memories. On the other hand, there are clearly a plethora of potentially promising avenues to pursue, and we are optimistic that real advances can be made in treating trauma-related disorders.

2. Neuronal substrates of fear extinction

Using a number of complementary techniques (including electrophysiology, immediate-early gene mapping, tracing studies, lesioning/inactivation approaches and optogenetics) research is revealing the complex and interconnected brain circuitry mediating fear extinction. Several key brain areas including the amygdala (AMY), hippocampus (HPC), medial prefrontal cortex (mPFC), periaqueductal grey (PAG), bed nucleus of the stria terminalis and others have been implicated in extinction [for recent detailed reviews see (Myers & Davis, 2007; Ehrlich et al., 2009; Herry et al., 2010; Pape & Pare, 2010; Knapska et al., 2012; Orsini & Maren, 2012; Duvarci & Pare, 2014)]. Of these, we focus on the AMY, HPC and mPFC as major, well-defined components of the fear circuitry (see Figure 2).

Full-size image (51 K)
Figure 2.

Anatomy of fear extinction and expression.

Fear extinction and expression rely on neuronal processing in an anatomical circuitry centering on the AMY, PFC, and HPC. Glutamatergic and GABAergic neurons, among others, are important components of connectivity and regulation of fear. The AMY is critically involved in the expression of aversive (fear) memories. While fear neurons in the BA send excitatory projections directly to the centromedial AMY (CeM) driving expression of fear (right panel), during extinction (left panel), the infralimbic cortex (IL) inhibits CeM output by driving inhibitory ITC neurons. IL inputs might also synapse directly on “extinction” neurons within the BA. “Extinction” neurons can influence activity within the central AMY (CeA) through several routes, possibly by driving inhibitory ITC or CeL (off PKC+) neurons that limit CeM activity. There is also a BA-CeL pathway contributing to ultimate inhibition of the CeM. The hippocampus is involved in contextual aspects of extinction via its projections to both the IL and the BA, among other brain regions. Hence, inhibitory memories built following extinction are encoded by the AMY and the PFC and are modulated by the HPC. It is thought that extinction training and exposure therapy produce long- lasting changes in synaptic plasticity and interneuronal communication in this circuitry ultimately reducing fear responses via output stations including the CeM. For further details, the reader is refered to recent reviews of (Orsini & Maren, 2012; Duvarci & Pare, 2014).

While the AMY and the mPFC are crucial for the formation and maintenance of fear extinction memories, the HPC, linked with the mPFC and the AMY (Pape & Pare, 2010), processes contextual information linked with extinction (Orsini & Maren, 2012). Altering these hippocampal contributions to extinction memory or mimicking the hippocampal response within the extinction context (see below) may be mechanisms that render extinction context-independent. The AMY is a core hub in fear extinction processing. Data in rodents and humans suggest sustained AMY activity in extinction-impaired individuals, possibly resulting from a failure to engage pro-extinction circuits in cortical areas and subregions of the AMY [reviewed in (Holmes & Singewald, 2013)]. Different AMY subregions and neuronal populations have differential contributions to extinction. The centromedial AMY (CeM) is the major output station of the AMY that drives fear via its connections to the hypothalamus and brainstem regions (LeDoux et al., 1988; Fendt & Fanselow, 1999; Maren, 2001), and its responding is modulated following extinction via intra-amygdala and remote inputs. Extinction training has been shown to cause a rapid reduction of CS-evoked responses of lateral AMY (LA) neurons possibly via depotentiation of thalamic inputs (Duvarci & Pare, 2014). The basal AMY (BA) contains extinction-encoding neurons which drive GABAergic cells in the medial intercalated cell masses (ITC) and neurons in the centrolateral AMY (CeL) to inhibit the centromedial AMY (CeM) and the expression of fear (Herry et al., 2008; Duvarci & Pare, 2014). Following from these initial findings, additional studies involving optogenetic approaches, have shown that a subpopulation of the BA pyramidal neurons which express the Thy1 gene may mark the extinction neuron subpopulation (Jasnow et al., 2013).

It is also becoming apparent that different ITCs are part of a GABAergic feed-forward relay station interconnecting AMY nuclei with distinct networks within the ITCs that are engaged in particular fear stages exerting different influences on extinction (Whittle et al., 2010; Busti et al., 2011; Duvarci & Pare, 2014). Separate neuronal populations in the CeL have been found to have contrasting roles in fear and fear extinction. CeL-OFF neurons (PKCδ+ phenotype) inhibit fear-promoting CeM neurons, while CeL-ON (PKCδ- phenotype) cells stimulate fear-promoting CeM neurons (Ciocchi et al., 2010; Haubensak et al., 2010). Additionally, a very recent finding suggests that a subpopulation of neurons within the CeM –that of the Tac2 peptide-expressing cells -, is critically involved in fear learning and fear expression (Andero et al., 2014).

Another important node within the extinction circuitry is the mPFC, which shares strong interconnections with the HPC and the AMY. Fear extinction recruits the ventral segment of the mPFC, the infralimbic (IL) subdivision [the rodent correlate of the human ventromedial PFC (vmPFC)] which projects to BA extinction and ITC neurons and can thereby inhibit the activity of CeM fear output neurons [reviewed in (Amir et al., 2011; Senn et al., 2014)]. When a conditioned cue following extinction is presented in the extinction (therapy) context, the HPC activates the IL (an activation which is absent or less in a novel context), supporting subsequent CeM inhibition (Herry et al., 2010). The IL subdivision is involved in the consolidation of extinction memory and synaptic plasticity in this region and is crucial for successful extinction retrieval (Mueller & Cahill, 2010). Impaired extinction has been associated with relatively low activity in IL neurons, and successful extinction with relatively high IL neuronal activity [reviewed in (Holmes & Singewald, 2013)]. The region of the vmPFC neighboring the IL [the prelimbic cortex (PL) in rodents and the dorsal anterior cingulate in humans] plays an opposite role to the IL in regulating fear and extinction [reviewed in (Milad & Quirk, 2012)]. The PL interacts with BA fear neurons, augmenting output of the basolateral amygdaloid complex (BLA) and hence CeM activity (Senn et al., 2014).

There is now strong evidence from both animal and human studies that these various components of the neural circuit underlying extinction are functionally disturbed in individuals with impaired extinction (reviewed in (Holmes & Singewald, 2013) and importantly that successful extinction in animals (Whittle et al., 2010) and exposure therapy in humans (Hauner et al., 2012) can correct these abnormalities by triggering a lasting reorganization of this network.

Note to users: Accepted manuscripts are Articles in Press that have been peer reviewed and accepted for publication by the Editorial Board of this publication. They have not yet been copy edited and/or formatted in the publication house style, and may not yet have the full ScienceDirect functionality, e.g., supplementary files may still need to be added, links to references may not resolve yet etc. The text could still change before final publication.
Although accepted manuscripts do not have all bibliographic details available yet, they can already be cited using the year of online publication and the DOI, as follows: author(s), article title, Publication (year), DOI. Please consult the journal’s reference style for the exact appearance of these elements, abbreviation of journal names and use of punctuation.
When the final article is assigned to an volumes/issues of the Publication, the Article in Press version will be removed and the final version will appear in the associated published volumes/issues of the Publication. The date the article was first made available online will be carried over.
twin memes II