31
1521-0081/66/4/10021032$25.00 http://dx.doi.org/10.1124/pr.114.009126 PHARMACOLOGICAL REVIEWS Pharmacol Rev 66:10021032, October 2014 Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics ASSOCIATE EDITOR: LESLIE A. MORROW Targeting the Modulation of Neural Circuitry for the Treatment of Anxiety Disorders David H. Farb and Marcia H. Ratner Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts Abstract.................................................................................... 1002 I. Introduction ................................................................................ 1003 II. Types of Anxiety Disorders ................................................................. 1004 III. Brain-Imaging Studies: Assessment of Brain Activation in Patients with Anxiety Disorders . . 1005 A. Functional Magnetic Resonance Imaging as a Method for Biomarker Identification....... 1006 B. Positron Emission Tomography: Advances and Caveats .................................. 1007 C. Single Photon Emission Computed Tomography ......................................... 1008 D. Magnetic Resonance Spectroscopy ....................................................... 1009 E. Overview ............................................................................... 1010 IV. Neural Systems and the Neurochemical Basis of Anxiety .................................... 1011 A. Neural Systems Underlying Anxiety .................................................... 1011 1. A Connectome Hypothesis. .......................................................... 1011 2. Optogenetics and Electrophysiology.................................................. 1012 B. Type A GABA Receptor and Serotonin Receptor Structure/Roles in Anxiety .............. 1013 1. GABA............................................................................... 1014 2. Serotonin............................................................................ 1016 V. Neuropsychopharmacology of Anxiety Disorders ............................................. 1018 A. Positive Allosteric Modulation of GABAergic Transmission .............................. 1018 1. Benzodiazepines and Related Modulators............................................. 1018 2. Natural Product Modulators......................................................... 1019 3. Increasing GABA Levels............................................................. 1020 B. Modulators of Glutamatergic Transmission .............................................. 1020 1. Inhibitors of Glutamatergic Activity.................................................. 1020 2. Potentiators of Glutamatergic Activity................................................ 1021 3. Neurosteroids as Modulators of Glutamatergic Transmission. ........................ 1021 Abstract——Anxiety disorders are a major public health concern. Here, we examine the familiar area of anxiolysis in the context of a systems-level understanding that will hopefully lead to revealing an underlying pharmacological connectome. The introduction of benzodiazepines nearly half a century ago markedly improved the treatment of anxiety disorders. These agents reduce anxiety rapidly by allosterically enhancing the postsynaptic actions of GABA at inhibitory type A GABA receptors but side effects limit their use in chronic anxiety disorders. Selective serotonin reuptake inhibitors and serotonin/norepinephrine reuptake inhibitors have emerged as an effective first-line alternative treatment of such anxiety disorders. However, many individuals are not responsive and side effects can be limiting. Research into a relatively new class of agents known as neurosteroids has revealed novel modulatory sites and mechanisms of action that are providing insights into the pathophysiology of certain anxiety disorders, potentially bridging the gap between the GABAergic and serotonergic circuits underlying anxiety. However, translating the pharmacological activity of compounds targeted to specific receptor subtypes in rodent models of anxiety to effective therapeutics in human anxiety has not been entirely successful. Since modulating any one of several broad classes of receptor targets can produce anxiolysis, we posit that a systems-level discovery platform combined with an individualized medicine approach based on noninvasive brain imaging would substantially advance the development of more effective therapeutics. Address correspondence to: Dr. David H. Farb, Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118. E-mail: [email protected] dx.doi.org/10.1124/pr.114.009126. 1002 by guest on July 7, 2020 Downloaded from

Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

1521-0081/66/4/1002–1032$25.00 http://dx.doi.org/10.1124/pr.114.009126PHARMACOLOGICAL REVIEWS Pharmacol Rev 66:1002–1032, October 2014Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics

ASSOCIATE EDITOR: LESLIE A. MORROW

Targeting the Modulation of Neural Circuitry for theTreatment of Anxiety Disorders

David H. Farb and Marcia H. Ratner

Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine,Boston, Massachusetts

Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1002I. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1003II. Types of Anxiety Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1004III. Brain-Imaging Studies: Assessment of Brain Activation in Patients with Anxiety Disorders . . 1005

A. Functional Magnetic Resonance Imaging as a Method for Biomarker Identification. . . . . . . 1006B. Positron Emission Tomography: Advances and Caveats. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1007C. Single Photon Emission Computed Tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1008D. Magnetic Resonance Spectroscopy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1009E. Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1010

IV. Neural Systems and the Neurochemical Basis of Anxiety. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011A. Neural Systems Underlying Anxiety . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011

1. A Connectome Hypothesis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10112. Optogenetics and Electrophysiology. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012

B. Type A GABA Receptor and Serotonin Receptor Structure/Roles in Anxiety . . . . . . . . . . . . . . 10131. GABA. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10142. Serotonin. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1016

V. Neuropsychopharmacology of Anxiety Disorders. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1018A. Positive Allosteric Modulation of GABAergic Transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1018

1. Benzodiazepines and Related Modulators.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10182. Natural Product Modulators. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10193. Increasing GABA Levels. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1020

B. Modulators of Glutamatergic Transmission. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10201. Inhibitors of Glutamatergic Activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10202. Potentiators of Glutamatergic Activity.. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10213. Neurosteroids as Modulators of Glutamatergic Transmission. . . . . . . . . . . . . . . . . . . . . . . . . 1021

Abstract——Anxiety disorders are a major publichealth concern. Here, we examine the familiar area ofanxiolysis in the context of a systems-level understandingthat will hopefully lead to revealing an underlyingpharmacological connectome. The introduction ofbenzodiazepines nearly half a century ago markedlyimproved the treatment of anxiety disorders. Theseagents reduce anxiety rapidly by allosterically enhancingthe postsynaptic actions of GABA at inhibitory type AGABA receptors but side effects limit their use in chronicanxiety disorders. Selective serotonin reuptake inhibitorsand serotonin/norepinephrine reuptake inhibitors haveemerged as an effective first-line alternative treatment ofsuch anxiety disorders. However, many individuals arenot responsive and side effects can be limiting. Research

into a relatively newclass of agents knownasneurosteroidshas revealed novel modulatory sites and mechanisms ofaction that are providing insights into the pathophysiologyof certain anxiety disorders, potentially bridging the gapbetween the GABAergic and serotonergic circuitsunderlyinganxiety.However, translating thepharmacologicalactivity of compounds targeted to specific receptorsubtypes in rodent models of anxiety to effectivetherapeutics in human anxiety has not been entirelysuccessful. Since modulating any one of several broadclasses of receptor targets can produce anxiolysis, weposit that a systems-level discovery platform combinedwith an individualized medicine approach based onnoninvasive brain imaging would substantially advancethe development of more effective therapeutics.

Address correspondence to: Dr. David H. Farb, Laboratory of Molecular Neurobiology, Department of Pharmacology and ExperimentalTherapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118. E-mail: [email protected]

dx.doi.org/10.1124/pr.114.009126.

1002

by guest on July 7, 2020D

ownloaded from

Page 2: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

C. Dopaminergic Transmission. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1021D. Epigenetics of Anxiolysis: Histone Deacetylase Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1021E. Modulators of Serotonergic and Noradrenergic Transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1022F. Neuroactive Steroids as Neuromodulators of Transmission . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1022

VI. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1025Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1026References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1026

I. Introduction

Anxiety is a recognized symptom of many psychiatricdisorders, including generalized anxiety disorder (GAD),social anxiety disorder (SAD), obsessive-compulsivedisorder (OCD), and post-traumatic stress disorder(PTSD). Although anxiety is a commonly reported clinicalmanifestation, it can be severely debilitating if leftuntreated. According to the Epidemiologic CatchmentArea Study, the prevalence of anxiety disorders is about7% in the adult population of the United States (Regieret al., 1990, 1998). Studies looking at the incidence ofanxiety disorders indicate that about 18% of adults in theUnited States may seek treatment for anxiety in anygiven year (Kessler et al., 2005b). Women are more likelythan men to be diagnosed with an anxiety disorder buthow much of this sex difference is due to socioeconomicfactors has not been established. That said, the need fora unified theory that will aid the development of targetedanxiolytics could not be more urgent with the emergenceof PTSD as a major global public health challenge facedafter more than a decade of war.The importance of an improved approach to treating

anxiety is highlighted by the inconsistent results seenwith the class of drugs considered to be the contempo-rary first-line treatment: selective serotonin reuptakeinhibitors (SSRIs) and serotonin and norepinephrinereuptake inhibitors (SNRIs). Although these agentshave been shown to be beneficial for the treatment ofcertain anxiety disorders, not all patients achieve anadequate clinical therapeutic response. The existenceof “nonresponders” indicates that SSRIs and SNRIs arenot the fail-safe solution to treating anxiety clinicianshave been looking for. In addition, SSRIs and SNRIsare associated with complications that can limit their

use in some patients, including delays in producingthe desired clinical reduction in anxiety or even po-tential worsening of the anxiety (Ravindran and Stein,2010; Sertraline, 2012; Venlafaxine, 2012; Fluoxetine,2013), not to mention the risk for the SSRI discontin-uation syndrome in noncompliant patient populations.Furthermore, some SSRIs (e.g., citalopram) are notsuitable for patients with heart problems (Wang andPae, 2013).

Another class of drugs, benzodiazepine (BZD) anxi-olytics, has played a central role in the pharmacologicmanagement of anxiety disorders for about 50 years.Although not as widely prescribed as in the past, thesecompounds nevertheless remain an effective alterna-tive to SSRIs. That said, the nonselective BZDs are notdevoid of side effects, which can include drowsiness,ataxia, impairment of cognition, as well as the risk ofdependence and withdrawal symptoms, including in-creased anxiety, tremors, and seizures (Noyes et al.,1988; Bennett et al., 1998; Yamawaki, 1999; Allgulanderet al., 2003). The identification of subtype-selectiveGABA receptor–positive allosteric modulators, such aszolpidem, which have proven to be effective in thetreatment of patients with insomnia while producingfewer untoward side effects than nonselective BZDs,suggested that similar drugs could be developed for anx-iety disorders. Unfortunately, although subunit-preferringcompounds are effective for insomnia, they are not yetproven to be any more beneficial for the managementof anxiety disorders than the classic BZDs, such asdiazepam (Farkas et al., 2013).

Another alternative to enhancing inhibitory neuro-transmission mediated by GABA is modulating excit-atory glutamatergic neurotransmission mediated by

ABBREVIATIONS: 5-HT, serotonin; BLA, basolateral amygdala; BNST, bed nucleus of the stria terminalis; BOLD, blood oxygen level–dependent; BZD,benzodiazepine; BZDR, benzodiazepine receptor; CBT, cognitive behavioral therapy; CL 218872, 3-methyl-6-[-3-(trifluoromethyl)phenyl]-1,2,4-triazolo[4,3-b]pyridazine; CNS, central nervous system; Co 3-0593, 3b-ethenyl-3a-hydroxy-5a-pregnan-20-one; DSM,Diagnostic and StatisticalManual ofMental Disorders;fMRI, functional magnetic resonance imaging; GABAAR, type AGABA receptor; GAD, generalized anxiety disorder; GPCR, G protein–coupled receptor; HPA,hypothalamic-pituitary-adrenocortical; K36, 5,7,29-trihydroxy-6,8-dimethoxyflavone; L655,708, ethyl(13aS)-7-methoxy-9-oxo-11,12,13,13a-tetrahydro-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylate; MRK-409, 7-cyclobutyl-6-(2-methyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2,6-difluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine; MRS, magnetic resonance spectroscopy; NMDA,N-methyl-D-aspartate; OCD, obsessive-compulsive disorder; PET, positron emissiontomography; PKA, protein kinase A; PTSD, post-traumatic stress disorder; rCBF, regional cerebral blood flow; Ro 15-4513, ethyl-8-azido-5,6-dihydro-5-methyl-6-oxo-4H-imidazo-1,4-benzodiazepine-3-carboxylate; Ro 16-6028, (13aS)-8-bromo-11,12,13,13a-tetrahydro-9-oxo-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylic acid 1,1-dimethylethyl ester; Ro 19-8022, (R)-1-[(10-chloro-4-oxo-3-phenyl-4H-benzo[a]quinolizin-1-yl)carbonyl]-2-pyrrolidine-methanol; RU 28362, 11b,17b-dihydroxy-6,21-dimethyl-17a-pregna-4,6-trien-20yn-3-one; RY80, ethyl-8-acetylene-5,6-dihydro-5-methyl-6-oxo-4H-imidazo[1,5a][1, 4]benzodiazepine-3-carboxylate,[ethyl-3H]; SAD, social anxiety disorder; SERT, serotonin transporter; SL651498, 6-fluoro-9-methyl-2-phenyl-4-(pyrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyrido[3,4-b]indol-1-one; SNRI, serotonin/norepinephrine reuptake inhibitor; SPECT, single photonemission tomography; SSRI, selective serotonin reuptake inhibitor; TPA023B, 7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine; VPA, valproic acid; VRE, virtual reality exposure.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1003

Page 3: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

N-methyl-D-aspartate (NMDA) receptors. AlthoughNMDAreceptors offer a promising therapeutic target fortreating anxiety disorders, the efficacy of NMDA re-ceptor modulators developed to date also remains limited(Swanson et al., 2005; Barkus et al., 2010).Collectively, these findings reveal a clear need for an

improved understanding of the neural systems underly-ing anxiety as well as a more effective approach to de-veloping anxiolytic agents. We believe that three recentdevelopments—the use of noninvasive imaging in humansubjects to identify biomarkers implicated in neurologicdisorders, and a convergence of pharmacologic effects inhuman populations of drugs that augment GABAergictone or serotonergic tone, combined with the develop-ment of systems-based approaches to drug discovery—offer a promising path forward. Systems-based drugdiscovery differs from traditional target-based discoveryin that it utilizes an understanding of complex systems tosearch for potential therapeutic agents, as opposed toidentifying agents based solely on interactions witha single target (Desbiens and Farb, 2012).Research into neuroactive steroids recently revealed

an important role for these agents in the pathophys-iology of anxiety, both influencing and being influencedby the GABAergic, serotonergic, and other neural sys-tems associated with anxiety and demonstrates boththe complexity of the problem and why focusing ona specific target without regard for the entire neuralnetwork may actually be counterproductive. This re-view aims to consider the basic molecular mechanismsby which neurotransmission can be modulated acrossvarious neural systems implicated in anxiety disor-ders, as well as to parse out how best to approach thetreatment of specific anxiety disorders based on theneural networks involved in each, with the ultimategoal of developing a systems-based approach to thediscovery of novel anxiolytics.Our understanding of anxiolytics starts with the

recognition that at least in some cases, therapeuticsappear to effectively interact with specific receptorsto modulate apparently simple and conserved neuralsystems implicated in hypervigilance and fear in bothhumans and animals. A closer look at the problem ofanxiety in humans reveals that complex cognitiveprocesses such as executive function mediated by thefrontal lobes also play a role in how subjects respond toenvironmental stimuli and that a single drug targetmay not be the most effective approach in the treat-ment of all types of anxiety. A growing body of researchis now also shedding light on the multiple genetic,environmental, and social factors that interact in thedevelopment of anxiety disorders (Lépine, 2002; Steinet al., 2002, 2014; Pigott, 2003; Gelernter et al., 2004;Hettema et al., 2005; Porcelli et al., 2012). In addition,it is increasingly evident that a diverse array ofneurotransmitters and their respective receptors andtransporters, including those for GABA, serotonin

(5-HT), and NMDA, play critical roles in the develop-ment of anxiety disorders (Swanson et al., 2005; Barkuset al., 2010; Graeff and Zangrossi, 2010). An increasedunderstanding of the neural systems underlying a path-ologic response to environmental stimuli that do notinduce anxiety in healthy subjects therefore begins withthe development of “connectivity maps” that in turn maylead to more individualized approaches to the pharma-cological management of various anxiety disorders.

II. Types of Anxiety Disorders

A comprehensive review of anxiety disorders must beginwith a definition of the problem. Anxiety per se is generallyconsidered to be synonymous with acute nervousness,fretfulness, and apprehension. Although the words fear,anxiety, and worry can sometimes be used interchange-ably, these terms generally represent distinct emotionalresponses that can be distinguished clinically by intensityand duration. Among healthy individuals, the magnitudeof this type of emotional response is proportional to thespecificity and intensity of the stimulus. The specificity ofthe stimulus is determined by its relevance to a particularindividual based on his or her current circumstancecombined with past life experience with that particularstimulus (i.e., individual sensitivity). For example, expo-sure to a stimulus that has previously been paired with anadverse life event will produce a different response thanwill exposure to a novel stimulus. Likewise, fear, fright,flight, and panic are more intense and acute reactions thantrepidation, apprehension, and worry and normally occurin close chronological and physical relation to an exposureto the appropriate stimuli. For example, an individual mayfeel slightly nervous and fretful while waiting to meeta blind date, or may be apprehensive about his or herability to perform a novel task in public, but fearful ofencountering an armed robber while walking down a darkalley alone at night. All of these responses are typicallybrief and resolve spontaneously with termination of ex-posure to the respective stimuli.

Fear, an intense fight-or-flight response that occursin reaction to a real or perceived threat, is a survivalresponse that is in contrast with panic disorder, whichis a fight-or-flight response in the absence of any identifiabledanger, therefore appearing to be a pathologic condition.Because these symptoms occur spontaneously and withoutwarning, persons with panic disorder will typically avoidsituations that their past experience suggests can triggerthe onset of an attack. Patients who have difficultyassigning a specific trigger for their attacks may becomesocially withdrawn.

Worry is synonymous with a more persistent concernor preoccupation with the unknown outcome of futurelife events. Worrying can be controlled so as to permitnormal functioning in social and occupational settings.By contrast, worrying such as that which occurs amongpatients with GAD is associated with persistent concern

1004 Farb and Ratner

Page 4: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

about routine life experiences, such as job performance orpersonal relationships, and is severe enough to interferewith the afflicted individual’s ability to concentrate andperform activities of daily living.The clinical anxiety disorders recognized in the fifth

edition of the Diagnostic and Statistical Manual ofMental Disorders (DSM-V), include the following: GAD,OCD, panic disorder, acute and chronic PTSD, and thevarious phobias, including agoraphobia, social phobia,and specific phobia (e.g., fear of flying) (American Psychi-atric Association, 2013). Each of these recognized clinicalanxiety disorders is unique with respect to the intensityand duration of the anxiety experienced as well as the typeof stimuli that can induce it.A systems-level approach to the development of

anxiolytic agents starts with a recognition of the rangeof anxiety disorders from which patients can suffer,including GAD, SAD, panic disorder, various phobias(e.g., fear of flying), and medication-induced anxietydisorder (DSM-V; American Psychiatric Association,2013). In addition, disorders that fall under their owncategory in the DSM-V, such as acute and chronic PTSD,include clinical manifestations, such as hypervigilance,exaggerated startle response, and fear as a component oftheir diagnostic criteria. Likewise, OCD includes fear ofsome dreaded event or situation among its diagnosticcriteria. Each of these disorders is unique with respect tothe intensity and duration of the symptoms experiencedby the patient as well as the type of stimuli that caninduce these. For example, persons with panic disorderexperience recurrent, discrete, and typically brief periodsof intense anxiety referred to as “uncued panic attacks”in the absence of any identifiable source of danger. Thisanxiety is associated with somatic symptoms, such asshortness of breath, chest pain, lightheadedness, andsweating. Persons with panic disorder often have per-sistent concerns about the causes or consequences ofhaving a panic attack. By contrast, “cued” anxiety attacksand phobias are defined as those in which the suffererexperiences debilitating symptoms in response to aclearly defined object or situation. In the cases ofphobias, exposure to the phobic stimulus produces animmediate onset of increased anxiety that is severeenough to interfere with the individual’s ability to functionnormally in social or occupational settings even when theperson recognizes that his or her own anxiety is excessiveor unreasonable.The onset of an anxiety disorder may be insidious or

may follow an identifiable life event. For example,acute stress disorder is characterized by symptomsthat occur during the first month after exposure to anextremely stressful event that evokes intense fear and/ora sense of helplessness, such as being in an automobileaccident or witnessing a murder. Symptoms of acutestress disorder include increased arousal, avoidance ofplaces and events that remind one of the traumaticevent, and recurrent intrusive memories or dreams

about the event. Although severe enough to interferewith activities of daily living, these symptoms typicallyresolve spontaneously within about 4 weeks after theevent. By contrast, chronic PTSD is characterized bysimilar symptoms that persist for more than 1 monthand, in many cases, can last for years after exposure toa traumatic event. These types of anxiety disordersrespond well to treatments that combine virtual realityexposure (VRE) therapy with pharmacologic interven-tions (Difede et al., 2014).

Unlike incident-evoked anxiety disorders, GAD ischaracterized by at least 6 months of persistent andexcessive apprehension and worry in response to aplethora of different stimuli, including such routine lifeevents as job performance, health of family members,or simply being on time for appointments. The onset ofGAD is typically insidious and is often not associatedwith a readily identifiable precipitating event. Individ-uals with GAD can be differentiated from those withnonpathological anxiety by two main criteria: 1) theyfind it difficult to control their worrying, and 2) thisperseverative thought process interferes with theirability to function in social and occupational settings.

The clinical manifestations of OCD are unique amongthe disorders that include fear and anxiety as a clinicalmanifestation in that the fear is about what will happenif the individual does not perform recurrent thoughts,impulses, or compulsions (e.g., repetitive hand washing)that the individual recognizes to be unreasonable andunrelated to real-life problems but nevertheless findsextremely difficult to stop. These symptoms cause distress,and thus, the affected individual may avoid situationsthat promote the obsession. For example, a person whoobsesses about personal contamination may not leavethe house to prevent placing himself in a situationwhere he might have to shake hands.

III. Brain-Imaging Studies: Assessment of BrainActivation in Patients with Anxiety Disorders

Although a range of clinical diagnostic criteria andassessment tools (e.g., Hamilton Anxiety Rating Scalesand the Minnesota Multiphasic Personality Inventory)have historically been used in the differential diagnosisof anxiety disorders, recent research has also shown thatvarious neuroimaging technologies can be employed toidentify common and unique brain regions associatedwith the different disorders. Neuroimaging studies canbe used to measure differences in regional cerebral bloodflow (rCBF) and for identifying significant changes inbinding to specific receptor subtypes and changes inuptake by transporters implicated in these disorders.Although it is premature to say that “silver bullets”targeting specific type A GABA receptor (GABAAR)subtypes or serotonin transporters (SERTs) withinparticular brain regions are on the horizon, it seemslikely that critical advances in brain imaging and

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1005

Page 5: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

subunit subtype analysis are improving the chancesfor optimizing therapeutic approaches to patientswith anxiety disorders.

A. Functional Magnetic Resonance Imaging asa Method for Biomarker Identification

It is natural to wonder whether serotonergic neuronsin the dorsal and median raphe nuclei that project tocorticolimbic circuits may be controlled by GABAergic,serotonergic, and neurosteroid modulators. A noninva-sive approach for assessing the elements of a potentialanxiety connectome is to evaluate the pharmacologic ef-fects of anxiolytics on specific brain regions by determiningchanges in rCBF associated with anxiety-inducingstimuli—these change in response to different pharmaco-logic interventions. Blood oxygen level–dependent (BOLD)functional magnetic resonance imaging (fMRI) can alsobe used to differentiate between types of anxiety, suchas between anxious apprehension (which involves verbalrumination and worry) and anxious arousal (which in-volves intense fear or panic or both) (Engels et al., 2007).An fMRI study of healthy subjects with no history

of psychiatric illness or treatment revealed that thosewho also tested positive for a regulatory variant(5-HTTLPR) in the SERT gene (SLC6A4), which waspreviously associated with reduced transcription of thetransporter, had greater amygdaloid reactivity in re-sponse to presentation of fearful and angry facesindependent of patient sex. These findings suggestthat the amygdala can be biased for reactivity basedon genetics and that this may represent a susceptibilityfactor for a pathologic response to stressful life ex-periences (Hariri et al., 2005). The observation thatSSRIs promote downregulation of SERTs (Mirza et al.,2007) also implicates dysregulation of serotoninergicneurotransmission in etiology of certain anxiety disor-ders. This polymorphism has also been associated withreduced responsiveness to SSRI treatment (Zanardiet al., 2001).The potential for an anxiolytic effect of 5-HT via

attenuation of amygdala activation was shown in astudy of healthy subjects treated with the SSRI citalopramfor 7 days. Compared with control subjects, the fMRIs ofindividuals treated with citalopram showed a bilateralreduction in activation of the amygdala, hippocampus,andmedial prefrontal cortex in response to presentation ofthreatening stimuli (Harmer et al., 2006).Evaluation of fMRI responses to the presentation of

pleasant or unpleasant words while monitoring leftversus right hemisphere activity indicates that the leftside (inferior frontal gyrus) is more active in patientswith anxious apprehension, consistent with the factthat this type of anxiety is more verbally mediated.These findings may seem to be discordant with datasupporting a left-sided bias in response to pleasantemotion, but positive emotional content preferentiallyactivates different regions of the left hemisphere: the

left frontal region (dorsolateral prefrontal cortex) (Engelset al., 2007).

Results from fMRI imaging studies support the clinicalimpression that GAD patients overreact to both pathology-specific and nonspecific cues and that treatment-inducedreduction of anxiety attenuates the response to both typesof cues (Hoehn-Saric et al., 2004). In a small cohort of sixpatients, subjects were initially studied using fMRIwhile each listened to verbal descriptions of a personalworry or a neutral statement before treatment withcitalopram and again after 7 weeks of treatment. ReducedBOLD responses to worry statements were observedin prefrontal regions, the striatum, and insula andparalimbic regions. In addition, contrasts before and aftertreatment revealed reductions in the differential responsethat existed between worry and neutral statements.Overall reduction of the BOLD response was mostprominent during neutral statements, particularly inthe left hemisphere. These findings are consistent withthe suggestion that frontolimbic structural connectivityunderlies the preservative thoughts as well as theemotional disturbances seen in patients with GAD(Tromp et al., 2012).

An fMRI study of 21 patients (8 males and 13 females)with generalized social phobia, characterized by anexaggerated and pervasive fear and avoidance ofscrutiny by others, suggested that activation of theamygdala in response to presentation of angry andfearful faces was attenuated relative to baseline after12 weeks of treatment with the SSRI sertraline (Phanet al., 2013). All patients received a stable dose (100–150mg/day) at the time of testing and had been titratedbased on their clinical response to treatment. At testing,14 of 21 patients were deemed to be responsive tosertraline treatment, whereas 7 were considered non-responders. All treated patients (clinical responders andnonresponders) were included in the study but bloodlevels of sertraline were not measured. A post hocanalysis looking at differences between responders andnonresponders indicated that the attenuation frompretreatment to post-treatment in amygdala reactivityto fearful faces was significant in responders (P = 0.01)but not in nonresponders (P = 0.22), as was attenuationin ventromedial prefrontal cortex reactivity to angryfaces (responders, P = 0.02; nonresponders, P = 0.29).Activation of the amygdala in all of the patients beforeand after treatment was also compared with that of19 healthy controls (10 males and 9 females). Consistentwith reports from other studies, activation of the amygdalain response to fearful faces before treatment was found tobe greater in the anxiety patients than in control patients;importantly, this difference was abolished by treatmentwith sertraline. By contrast, activation of the ventromedialprefrontal cortex in response to angry faces beforetreatment was found to be significantly lower in anxietypatients than in control patients, and this differencewas also abolished by sertraline treatment.

1006 Farb and Ratner

Page 6: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

The effects of chronic paroxetine treatment (20 mg/day)versus placebo have also been measured in patients withSAD using fMRI, along with performance of several tasksincluding a public-speaking task, public exposition of arecorded performance task, and an emotional face-processing task (Giménez et al., 2014). Paroxetinetreatment was associated with reduced activation inthe insula, thalamus, and anterior cingulate cortex atrest and during performance of the public expositionof a recorded performance task. By contrast, duringthe emotional face-processing task, paroxetine treat-ment was associated with an increase in activation ofthe right amygdala and the insula bilaterally.Collectively, these findings point to a role for corticolimbic

circuits, including the amygdala and medial prefrontalcortex in SAD, and suggest that reductions in the level ofactivation in these brain regions may prove to be a usefulbiologic marker for the efficacy of novel anxiolytics.

B. Positron Emission Tomography: Advancesand Caveats

rCBF can also be measured using positron emissiontomography (PET) scans. Studies of rCBF using PETsuggest that paralimbic structures together with theright inferior frontal cortex and subcortical nuclei mediatesymptoms across three different anxiety disorders: OCD,simple phobia, and PTSD (Rauch et al., 1997). Oxygen15–labeled carbon dioxide was used as a tracer to measurerCBF in 23 right-handed adult subjects (8 men and 15women) meeting criteria for one of these three disordersand relative rCBF measured in the context of subjects’specific symptom provocation paradigms (e.g., exposure ofa patient with OCD to a “contaminated” object). Overall,analysis of pooled imaging data revealed activation in theright inferior frontal cortex and right posterior medialorbitofrontal cortex, and bilateral activation in the insula,lentiform nuclei, and brainstem during symptomaticversus control conditions.The limitations of measuring changes in rCBF as a

measure of SSRI efficacy in the treatment of anxietydisorders is hampered by a deficiency of knowledgeabout the underlying neural circuitry, or functionalconnectome, that gives rise to feelings of anxiety andpharmacologically induced anxiolysis. For example, whenPET techniques were used to assess the effects of SSRItreatment in patients with SAD, the SSRI-treated subjectsshowed a selective deactivation of the left lateralamygdala not associated with a reduction in anxietystatus. By contrast, the left basomedial/basolateraland right ventrolateral regions of the amygdala weredeactivated in both SSRI-treated and placebo patientswho showed improvement in anxiety symptoms (Fariaet al., 2012) (Fig. 1). This latter deactivation was notobserved in nonresponders (i.e., subjects who showed noreduction in anxiety scores) irrespective of treatmentgroup, suggesting that this change was mediated by areduction in anxiety unrelated to SSRI treatment. Thus,

SSRI treatment was no more effective than placebo fortreating SAD, perhaps due to the difficulty of separatinganxiolysis from a placebo effect in conjunction witha small sample size. Unfortunately, the subjects werenot tested for the 5-HTTLPR polymorphism in the 5-HTtransporter gene (associated with reduced responsive-ness to SSRI treatment; Zanardi et al., 2001), furtherconfounding interpretation of the results.

An advantage of PET versus fMRI as a clinical re-search tool can be exemplified by a study that observeddecreased flumazenil binding in specific brain regions inmedication-free persons diagnosed with panic disorder(Malizia et al., 1998). Global reduction in BZD site bindingthroughout the brain were found in patients with panicdisorder (n = 7) compared with controls (n = 8) (Fig. 2A).In addition, several loci thought to be essential in theregulation of anxiety (right orbitofrontal cortex and rightinsula) were found to have the largest regional decreasein binding. These results are consistent with previousclinical psychopharmacologic evidence of involvementof the GABAAR (Hasler et al., 2008) using PET with[11C]flumazenil in patients with panic disorder comparedwith healthy controls, which showed reduced bindingpotential across regions of the frontal, temporal, andparietal cortices, particularly the dorsal anterolateralprefrontal cortex.

Dynamic three-dimensional PET scans show that theBZD-GABAAR may play a role in the pathophysiologyof PTSD, a finding that is consistent with previousanimal and clinical pharmacological studies (Geuze et al.,2008). In this study, radiolabeled [11C]flumazenil was

Fig. 1. PET scans of subjects with SAD who were treated with either anSSRI or placebo for a minimum of 6 weeks does not identify a brain regionmodulated by drug and associated with anxiolysis. Scans were madewhile subjects performed a public-speaking task. Subjects were dividedinto two groups based on clinical response to treatment (respondersversus nonresponders). An attenuation of rCBF that was independent oftreatment was observed in the left basomedial/basolateral and rightventrolateral amygdala of responders. Red indicates the site of reducedactivation irrespective of treatment (placebo versus SSRI) seen in responders;blue indicates a purely pharmacodynamic nonaxiolytic effect on rCBF thatwas seen in both responders and nonresponders treated with an SSRI;and yellow indicates an effect related to repeated testing. Reprinted withpermission from Faria et al. (2012). L, left; R, right.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1007

Page 7: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

used as a tracer in nine drug-naïve male veterans withdeployment-related PTSD and sevenmale veterans withoutPTSD. After a scan of 60 minutes, reduced [11C]flumazenilbinding was observed throughout the cortex, hippocampus,and thalamus of the PTSD patients (Fig. 2B).PET techniques have also proven valuable in evaluating

variations in the distribution of specific subtypes of theGABAAR within the human brain and the roles of thesesubunits in mediating different functions. Competitionstudies in vivo in the rat revealed that [11C]Ro 15-4513(ethyl-8-azido-5,6-dihydro-5-methyl-6-oxo-4H-imidazo-1,4-benzodiazepine-3-carboxylate) uptake was reduced tononspecific levels only by drugs that have affinity forthe a5 subtype, such as flunitrazepam, RY80 (ethyl-8-acetylene-5,6-dihydro-5-methyl-6-oxo-4H-imidazo[1,5a][1, 4]benzodiazepine-3-carboxylate,[ethyl-3H]), Ro 15-4513,and L655,708 [ethyl(13aS)-7-methoxy-9-oxo-11,12,13,13a-tetrahydro-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylate], but not by the a1-selective agonist zolpidem.[11C]Ro 15-4513 uptake was relatively greater in limbicareas compared with [11C]flumazenil, but lower in theoccipital cortex and cerebellum (Lingford-Hughes et al.,2002). The authors concluded that [11C]Ro 15-4513 PETlabels the GABAAR containing the a5 subunit in vivo inlimbic structures and can be used to further explore thefunctional role of this subunit in humans. This techniquewas used to characterize BZD recognition site occupancyin the human brain associated with administrationof several a2-selective compounds, such as TPA023B[7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine] andMRK-409 [7-cyclobutyl-6-(2-methyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2,6-difluorophenyl)-1,2,4-triazolo[4,

3-b]pyridazine] (Fig. 3) (Atack et al., 2006b, 2011a,b; VanLaere et al., 2008).

C. Single Photon Emission Computed Tomography

A single photon emission computed tomography (SPECT)study using 123I-labeled 2b-carbomethoxy-3b-(4-iodophenyl)-N-(3-fluoropropyl)-nortropane revealed an increase indopamine transporter binding in the striatum of SADsubjects treated with escitalopram for 12 weeks(Warwick et al., 2012). These subjects also showed adecrease in anxiety scale scores associated with treat-ment, but the scores were not correlated with changesin dopamine transporter binding. The scores remainedrelatively high after therapy, suggesting that treatmentmay not have been completely successful for all par-ticipants. These subjects were also not stratified asresponders versus nonresponders, thus limiting theinterpretation of the data.

As revealed by SPECT scans using [123I]iomazenil,patients with panic disorder exhibit decreased benzo-diazepine receptor (BZDR) binding in the left hippo-campus and precuneus relative to controls (Bremneret al., 2000). Further comparisons of patients withpanic disorder who either did or did not have a panicattack at the time of the scan revealed a decrease inBZDR binding in the prefrontal cortex among patientswho had a panic attack. These findings suggest thatBZDR function in the prefrontal cortex appears to beinvolved in changes in state-related panic anxiety.

These findings also suggest a role for brain regionsimplicated in episodic memory function, such as theprecuneus. This in turn may be amenable to cognitivebehavioral therapy (CBT) and targeting by pharmacologic

Fig. 2. The BZD binding site on the GABAAR plays a role in the pathophysiology of PTSD. (A) Results of PET studies using radiolabeled[11C]flumazenil to study BZDR kinetics in vivo. Axial images of middle brain (12 mm above the anterior commissure–posterior commissure plane)showing median volume of distribution map for controls (left) and patients with panic disorder (right). Note the generalized decrease in volume ofdistribution affecting the thalami and the heads of the caudate nucleus among patients with panic disorder. Reprinted with permission from Maliziaet al. (1998). (B) PET scan images highlighting those areas that showed decreased binding of [11C]flumazenil in drug-naïve subjects with PTSD (n = 9)compared with controls (n = 7). Reduced binding was observed in the cortex, hippocampus, and thalamus. Reprinted with permission from Geuze et al.(2008). CN, caudate nucleus; FC, frontal cortex; Ins, insula; L, left; OC, occipital cortex; R, right; TC, temporal cortex; Th, thalamus.

1008 Farb and Ratner

Page 8: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

interventions that can enhance extinction of salientmemories that trigger panic attacks. The results indicatethat the neuronal circuits implicated in panic attacks(i.e., the precuneus) are distinctly different from thoseimplicated in GAD (i.e., more involvement of the frontallobe).In another study of depressed patients with or without

panic disorder (N = 18), radiolabeled iomazenil SPECTscans demonstrated that those patients with panic disorderhad a significant decrease (P, 0.05) in the regional activityindex in the lateral inferior temporal lobes (right and left),medial inferior temporal lobes (left), and inferior frontallobes (right and left) after 2 hours (Kaschka et al., 1995).The findings may be due to either regional blood flowdifferences or BZDR effects, but the former hypothesis isconfirmed to some extent by similar findings in the scansacquired after 10 minutes. Only hypoactivity in the leftlateral temporal region seemed to be independent of thereduced blood flow seen in panic disorders.A SPECT study in patients with social phobia found

that citalopram therapy decreased activity in the leftcingulum, the anterior and lateral part of the left temporalcortex, and the anterior, lateral, and posterior part of theleft midfrontal cortex (Van der Linden et al., 2000).This broad engagement of cortical areas in anxiety

disorders and anxiolysis may be the consequence of aprimary abnormality or may be a cause of the disorders.

In some ways, similar to the phenomenon of “kineticambiguity” common to the study of enzyme mechanismsand bio-organic chemistry, “systems ambiguity” presentsa conundrum for the pharmacologist seeking to translatetarget-based pharmacology into pharmacotherapeuticeffects based on a mechanistic understanding of ananxiolysis connectome.

D. Magnetic Resonance Spectroscopy

Magnetic resonance spectroscopy (MRS) allows forthe noninvasive monitoring of levels of GABA as wellas other neurochemicals in the human brain (Goddardet al., 2001; Strawn et al., 2013; Zwanzger et al., 2013).MRS has been used to measure GABA levels in pa-tients with PTSD, social anxiety, and panic disorder(Ham et al., 2007; Pollack et al., 2008; Rosso et al.,2014). A study of 14 unmedicated patients with panicdisorder and 14 healthy control subjects matched forage and sex revealed a 22% mean reduction of GABA inthe occipital lobe of 12 of 14 subjects compared withcontrols. There were, however, no significant correla-tions between GABA levels in the occipital lobe andany measures of illness or state anxiety (Goddard et al.,2004). In a study of subjects with PTSD, MRS mea-surement of GABA levels in the insula revealed thatpatients had 30% less GABA than healthy controls. Thisfinding could not be accounted for by age or sex. This

Fig. 3. PET studies comparing TPA023B identify BZD binding sites in the human brain. (A) Comparison of the structures of TPA023B (Atack et al.,2011a), TPA023 (Atack et al., 2006b), and MRK-409 (Atack et al., 2011b). (B) Pseudocolor images on the left show the inhibition of [11C]flumazenilbinding produced by a single 1.5-mg oral dose of TPA023B in PET scans performed 1 hour before and 5 and 24 hours after dosing. TPA023B hasprolonged occupancy at the BZD binding site of GABAARs in the human brain. The right graph shows occupancy plotted as a function of thecorresponding plasma TPA023B concentrations. Modified with permission from Van Laere et al. (2008).

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1009

Page 9: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

difference could also not be accounted for by the differencein the percent gray matter versus white matter propor-tions within the voxel of interest. In addition there was asignificant negative correlation between State-TraitAnxiety Inventory scores and GABA levels (Rosso et al.,2014) (Fig. 4).MRS can also be used to study acute drug-induced

changes in brain glutamate levels. Experimental pharma-cologic induction of panic has been used to elucidate theneurobiological correlates of panic attacks. Cholecystokinin-tetrapeptide-4–induced panic attack was studied usingMRS and was found to be associated with an increase inglutamate plus glutamine/creatine ratios in the anteriorcingulate cortex (Zwanzger et al., 2013). This suggests thata chemically induced (i.e., by cholecystokinin-tetrapeptide-4)panic attack alters the glutamatergic system in the anteriorcingulate cortex and may be involved in the pathogenesisof panic attack. Given the activation of precuneus in salientepisodic memory–induced panic attack, it is tempting tospeculate that a targetable downstream network involvingthe anterior cingulate cortex susceptible to glutamatergicpharmacotherapy could be an effective approach towardtreating panic attack.By contrast, in patients with PTSD, positive modulators

of glutamatergic transmission (e.g., D-cycloserine and

acamprosate; see section V.B) have been used in combi-nation with behavioral therapy to extinguish salientmemories associated with PTSD.

E. Overview

Taken as a whole, brain-imaging studies implicatethe amygdala, prefrontal, insular, limbic, paralimbic,and occipital cortices as well as the basal ganglia moreoften than other regions in pathologic anxiety. Giventhe role that the insula plays in integrating sensory,emotional, and cognitive information, it is certainly notsurprising to find changes in the level of activation inthis brain region. Given that the insula spans from theprefrontal cortex to the posterior parietotemporal lobesand thereby creates extensive connections between thelateral prefrontal cortex, ventromedial prefrontal cortex,orbitofrontal cortex, cingulate, amygdala, bed nucleusof the stria terminalis (BNST), and ventral striatum, itseems reasonable to think of the insula as a node in theanxiety connectome.

These neuroimaging findings also reveal some evi-dence of lateralization and localization based on how theanxiety-inducing stimulus is presented (e.g., verballyversus visually), suggesting a path forward for differen-tiating among types of anxiety and tailoring treatment

Fig. 4. MRS studies showing that brain GABA levels in the right insula are lower in patients with PTSD. (A) T1-weighted images depicting voxelplacement as well as difference edited and 68-millisecond spectra from the ACC (A and B) and insula (C and D) of a representative subject. (B)Associations of GABA/Cr in right anterior insula with state anxiety (A) and trait anxiety (B) in the whole sample. Circles correspond to PTSD patients(asterisk indicates the single medicated patient) and triangles are control subjects. All fitted metabolite areas were normalized to total creatine(creatine and phosphocreatine combined). ACC, anterior cingulate cortex; Cho, choline; Cr, creatine; NAA, N-acetylaspartate. Reprinted withpermission from Rosso et al. (2014).

1010 Farb and Ratner

Page 10: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

appropriately based on systems-level analysis of data.For example, a meta-analysis of neuroimaging studiesrelated to emotional processing in SAD, PTSD, andspecific phobia suggests that common brain regions(hyperactivation of the amygdala and insula) play a rolein these anxiety disorders that can be triggered byspecific stimuli. In addition, the results revealed thathypoactivation in the dorsal and rostral anterior cingulatecortices and the ventromedial prefrontal cortex may play arole in the dysregulation of emotions associated withPTSD (Etkin and Wager, 2007).Human imaging studies in which both PET and

fMRI are used to study biomarkers in the same cohortmay be the most suitable for identifying relationshipsbetween individual variability in receptor binding andbrain region activation in response to treatments (althoughstudies employing one technology are also useful). Thisdual-technology approach was used by Fisher et al. (2006)to study 5-HT1A autoreceptor binding in relation toamygdala activation and by Rhodes et al. (2007) tostudy the association between 5-HT transporter bindingand amygdala activation. Kobiella et al. (2011) took thisapproach one step further and studied 5-HT transporterpolymorphisms in relation to amygdala region activation,further demonstrating the unique power of multimodalneuroimaging studies.Nonhuman animal imaging using experimental mod-

els of anxiety are also helping to parse out inferencesconcerning the possible differences and similaritiesunderlying various anxiety disorders. One MRS studyusing a chronic stress–induced depression model revealeda reduction in brain levels of N-acetyl-aspartate, totalcreatine, and choline-containing compounds in tree shrewsexposed to psychosocial stress. In this study, the voxelof interest was the forebrain including parasagittal neo-cortex, subjacent white matter, and portions of subcorticalforebrain structures, including striatum (Czéh et al.,2001). Postmortem analysis showed reduced cell pro-liferation in the dentate gyrus and a decreased GluR2expression in the prefrontal cortex. Concomitant admin-istration of a novel compound combining partial dopamineD2 receptor agonism with SSRI activity prevented thesechanges, suggesting that combining dopamine D2 receptoragonism with SSRI activity may serve as a novel treat-ment of those anxiety disorders presenting with comorbiddepression (Michael-Titus et al., 2008). In another studyusing fluorodeoxyglucose-PET, three specific character-istics of the anxious phenotype identified in rhesusmonkey (hypothalamic-pituitary-adrenal activity, freezingbehavior, and expressive vocalizations) were associatedwith a common elevation of activity in the central nucleusof the amygdala and the anterior hippocampus (Shackmanet al., 2013).However, can these types of imaging data produce

useful information for real-life therapy in humanpatients with a range of anxiety disorders? In patientswith SAD, fMRI was used to measure brain activation

prior to intervention with CBT and showed that changesin regional activation in response to angry versus neutralfaces or emotional versus neutral scenes are an effectivepredictor of response to CBT (Doehrmann et al., 2013).

IV. Neural Systems and the Neurochemical Basisof Anxiety

In the following sections, we explore the neurochemicalcomponents of the brain, placing these findings in thecontext of a systems-level approach to therapeutics basedon the real-time differences in rCBF demonstrated byfunctional neuroimaging studies and changes in brainlevels of GABA and glutamate revealed by MRS.

A. Neural Systems Underlying Anxiety

1. A Connectome Hypothesis. Anxiety is a complexemotional response that involves multiple neurotrans-mitters acting at various receptors within the limbicsystem (i.e., hippocampus, parahippocampal gyrus, cin-gulate gyrus, hypothalamus, and amygdala) and withinthe paralimbic regions of the cerebral cortex such as theorbital and medial prefrontal cortex (Rauch et al., 1997;Simpson et al., 2000; Pape and Stork, 2003; Phillipset al., 2003; Sah et al., 2003; Yilmazer-Hanke et al., 2003;Zwanzger et al., 2003; Kang-Park et al., 2004; Phan et al.,2004). From a neural systems perspective, fear andanxiety seem to be controlled by two major circuits: thoseinvolved in our most primitive innate responses to simpleand overt threatening stimuli, and those involved inmoderating responses to more complex situations. Theprefrontal cortex appears to be the brain region mostoften implicated in the latter.

The prefrontal cortex in particular is implicated inattenuation of responses to anxiety-inducing stimuli.Lesions to the ventrolateral prefrontal cortex havebeen associated with increased anxiety in nonhumanprimates (Agustín-Pavón et al., 2012). It has beenproposed that the amygdala is hyper-responsive tothreatening stimuli, whereas the response of theventromedial prefrontal cortex is a blunted in patientswith anxiety disorders, resulting in inadequate regu-lation of their responses to anxiety-inducing stimuli(Kent and Rauch, 2003; Rauch et al., 2003). Thishypothesis is supported in part by functional neuro-imaging studies showing that individuals with GADhave an attenuated discriminatory response pattern inthe ventromedial prefrontal cortex when presented withthreatening versus nonthreatening stimuli (Greenberget al., 2013).

The amygdala, which receives afferent inputs fromthe sensory cortex and sends its efferent projections tothe hypothalamus, periaqueductal gray matter, locusceruleus, raphe nuclei, and ventral tegmental area(Sah et al., 2003), appears to be primarily involved inthe encoding and consolidation of emotionally chargedmemories (Isenberg et al., 1999; Roozendaal et al.,

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1011

Page 11: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

2001; Sah et al., 2003; Pelletier and Paré, 2004). Thebasolateral amygdala (BLA), which is composed of thelateral, basomedial, and basolateral nuclei, plays acentral role in this process and sends projections to themedial temporal lobe and nucleus accumbens andseptal nuclei via the stria terminalis (Roozendaalet al., 2001; Pelletier and Paré, 2004). Lesions of theBLA or its major efferent pathway, the stria terminalis,interfere with the encoding and consolidation of mem-ories induced by emotional arousal (McGaugh, 2000;Roozendaal et al., 2001). In addition, the effects ofglucocorticoids such as RU 28362 (11b,17b-dihydroxy-6,21-dimethyl-17a-pregna-4,6-trien-20yn-3-one) on memoryconsolidation are dependent on an intact BLA nucleus andits efferent fibers that run through the stria terminalis(Roozendaal et al., 2001).The subunit composition and pharmacological prop-

erties of GABAARs vary widely between and withinbrain regions. For example, receptors containing the a2

and a5 subunits, which show pharmacologic profilessimilar to the classic BZD II or type 2 receptors, arefound in higher concentrations in the amygdala andhippocampus, whereas receptors containing a6 subunits,which are insensitive to BZD anxiolytics, are localizedto the cerebellum (Wieland et al., 1992; McKernan andWhiting, 1996; Whiting et al., 1999).The neuroanatomic distribution of 5-HT receptors is

complex, as would be expected based on the pharma-cological effects of receptor agonists, antagonists, andSSRIs. The limbic system is critical for the generationof motivational and affective states, with the limbicforebrain containing high levels of 5-HT (an evolutionarilyancient neurotransmitter that has functional orthologreceptors even in the Drosophila fruit fly) (Nichols et al.,2002). The 5-HT fibers that project into the forebrainoriginate for the most part from the median raphe nucleusand the dorsal raphe nucleus. These projections areheterogeneous in terms of morphology and electro-physiology, representing yet another level of intricacythat could influence treatment outcomes (Hensler, 2006).The hippocampus is well known to be involved in

the formation of new memories. Neurogenesis in thehippocampal dentate gyrus has been implicated in theformation of both spatial memory and anxiety disor-ders, as well as in the action of SSRIs. The temporalbehavior of dentate gyrus granule cells with respect totheta rhythm activity is different between rats withnormal and impaired levels of neurogenesis, suggest-ing that immature neurons could distinctly affect thetemporal dynamics of hippocampal encoding (Rangelet al., 2013). Serotonergic antidepressants can reversethe established state of neuronal maturation in adulthippocampal cells, and upregulation of 5-HT4 receptor–mediated signaling may play a role in this process(Kobayashi et al., 2010). The role of dentate granulecell hypoactivity in anxiety and in the mechanism ofaction of SSRIs was recently suggested based on the

observation that challenge-induced hypoactivation ofdentate gyrus neurons in mice with high trait anxietyis normalized by fluoxetine treatment (Sah et al., 2012).

Differences in the intrinsic physiology and synapticinhibition between the mature and immature cell pop-ulations could separate the activity of differently agedneurons in a temporal coding regimen, influencingdownstream CA3 neuron integration of informationconveyed by young and mature granule cells. In linewith this hypothesis, fluoxetine treatment induces activesomatic membrane properties in a manner reflective ofimmature granule cells (Kobayashi et al., 2010). Thesechanges do not seem to be explained by an increase innewly minted immature neurons and may be character-ized as “dematuration” of mature granule cells. Theseobservations suggest that normalization of hippocampalhypoactivity may serve as a neurobiological marker ofanxiolytic effectiveness and successful remission.

2. Optogenetics andElectrophysiology. Optogenetic tech-niques have been used to further elucidate the role of theamygdala in anxiety. Tye et al. (2011) used viral trans-fections to study the role of BLA–centrolateral nucleuspathway in the expression of anxiety. Illumination ofchannel rhodopsins expressed in BLA terminals located atthe central nucleus of the amygdala was shown to induce anacute reversible anxiolytic effect. Selective illumination ofspecific BLA terminals produces a behavioral responseopposite that induced by activation of all glutamatergicBLA terminals suggesting that multiple subpopulationsor projections of BLA neurons can act in opposition (e.g.,direct excitation of centromedial nucleus along with feed-forward inhibition of centromedial nucleus) (Fig. 5).

Subsequent studies demonstrated that bilateraloptogenetic inhibition of BLA axon terminals in theventral hippocampus reduces anxiety-related behaviors inmice, suggesting that BLA inputs to the ventral hippo-campus may play a role in modulating basal levels ofanxiety (Felix-Ortiz et al., 2013). A reduction of hippo-campal theta activity has also been implicated in theanxiolysis induced by various classes of anxiolytics, in-cluding buspirone (McNaughton et al., 2007; Chee et al.,2014). A reduction of hippocampal theta activity has alsobeen implicated in the anxiolysis induced by variousclasses of anxiolytics, including buspirone (McNaughtonet al., 2007; Chee et al., 2014). McNaughton et al. (2013)proposed that conflict-specific, right frontal activationrecorded noninvasively with surface electrodes is a read-ily obtainable noninvasive human functional homologof rodent hippocampal-cortical activity that may alsobe useful as an objective biologic marker of anxiolyticefficacy.

An optogenetics approach has also been used toelucidate the role of the dentate gyrus in the processesof learning and memory, as well as the pathophysiol-ogy of anxiety (Kheirbek et al., 2013). Specifically,granule cells in the ventral dentate gyrus suppressinnate anxiety but do not affect contextual learning,

1012 Farb and Ratner

Page 12: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

whereas those in the dorsal region control the encodingof contextual fear memories but not their retrieval. Asnoted, findings from this study suggest that distur-bances in pattern separation may underlie anxietydisorders in some patients, and therapies targeting theexcitability of the ventral dentate gyrus may offera beneficial approach in this population. These kinds oftechniques have also born fruit in animal models aswell, potentially allowing for translation of data fromanimal models to studies in humans. The connectionbetween hippocampal-mediated learning and anxiety

was further elucidated using optogenetic techniques,showing that hippocampal dentate gyrus neuronsactivated during fear learning in mice can be reac-tivated later using light stimulation, producing anincreased freezing response outside of the originalcontext (Liu et al., 2012).

Optogenetics combined with in vivo electrophysio-logical recordings may further elucidate activity inthe neural networks implicated in anxiety disorders.Optogenetic inhibition of the BNST in mice increasedexploration of open spaces in the elevated plus maze testand open field test, whereas stimulation of the BNSTwith the excitatory channelrhodopsin-2 was associatedwith increased anxiety in both tests. To elucidate theneural circuitry between the BLA and the anterodorsalBNST, microdrive electrode arrays containing stereotrodessurrounding a fiber-optic were implanted in the anterodorsalBNST of mice expressing channelrhodopsin-2 in theBLA. Excitation of the glutamatergic BLA terminalswas associated with increased activity of anterodorsalBNST neurons. Conversely, optically stimulating in-puts to the oval nucleus of the BNST induced a netinhibition of neural activity in the BLA, suggestingthat the oval nucleus and anterodorsal BNST playantagonistic roles in modulating anxiety (Kim et al.,2013). These findings are consistent with observationsthat the extended amygdala plays an important role inthe neural circuitry implicated in anxiety.

The BNST occupies a central position in the neuralcircuitry regulating the HPA axis response to stressand thus plays an important role in regulating theneuroendocrine and behavioral responses to stress (Henke,1984; Dunn, 1987; Fernandes et al., 2002; Walker et al.,2009). The release of neuroactive steroids in this brainregion may represent an endogenous homeostatic mech-anism for modulating anxiety and restoring normalGABAergic neurotransmission and HPA axis activityafter exposure to stress-inducing stimuli (Patchev et al.,1994; Barbaccia et al., 1996; Fernandes et al., 2002).Stressors that activate the BNST also activate seroto-nergic systems, and changes in 5-HT receptor expressionin this region were suggested to play a role in pathologicresponses to stress (Guo et al., 2009; Hazra et al., 2012).Thus, modulation of the activity of specific 5-HT receptorsubtypes in this region may be a novel approach totreating anxiety disorders.

B. Type A GABA Receptor and Serotonin ReceptorStructure/Roles in Anxiety

A disturbance of the nuanced integration of excit-atory and inhibitory transmission can occur on a localnetwork level that ultimately affects interconnecteddomains of larger-order structures linking to the outsideworld. Unraveling this will undoubtedly involve theaction of networks serving as low-pass as well as high-passfilters, all of which are based on the integrated action ofmultiple transmitters at their respective receptors.

Fig. 5. Projection-specific excitation induced by illumination of BLA terminalsin the CeA induces acute reversible anxiolysis in mice. (A) Illustrations showingwhere illumination was directed in the channelrhodopsin-2 (ChR2):BLA-CeA,eYFP:BLA-CeA, and ChR2:BLA (somata) mice. The mice received 5-millisecondlight pulses at 20 Hz for all light-on conditions. (B) Representative pathsin the elevated plus maze during light-on and light-off conditions. (C)Histograms showing increased probability of open arm entries during lighton phase in ChR2:BLA-CeAmice. (D and E) Open field test data demonstratingthat ChR2:BLA-CeA mice spend more time in center with light on than doeYFP:BLA-CeA and ChR2:BLA (somata) controls (P , 0.00001). CeA, centralamygdala; CeL, centrolateral nucleus; CeM, centromedial nucleus; eYFP,enhanced yellow fluorescent protein. Reprinted from Tye et al. (2011).

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1013

Page 13: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

1. GABA. There is general agreement that GABA-mediated inhibitory neurotransmission plays a partic-ularly important role in the modulation of emotionalresponses to fear-inducing stimuli (Davis and Myers,2002; Stork et al., 2002, 2003; Walker et al., 2003), andthe concept of positive and negative allosteric modulationof fast postsynaptic GABAergic inhibitory neurotrans-mission by BZDs was central to anxiolytic pharmacologyfor decades (Choi et al., 1977; Macdonald and Barker,1978; Chan and Farb, 1985). GABAARs are locatedprimarily in the postsynaptic membrane at synaptic andextrasynaptic sites and mediate the majority of fastpostsynaptic inhibitory neurotransmission in the adultcentral nervous system (CNS) (Rabow et al., 1995;Barnard et al., 1998; Ben-Ari, 2002). The action ofGABA on GABAARs can modulate both the frequencyand duration of chloride channel openings to maintainthe membrane near its resting voltage (Macdonald et al.,1989; Twyman et al., 1989).The GABAAR is a member of the cys-loop superfamily

of receptors, which includes the nicotinic acetylcholinereceptors, the glycine receptors, and the 5-HT3 receptors.These receptors all have a characteristic loop formed bya disulfide bond between two cysteine residues (Sieghartand Sperk, 2002). The GABAAR is a pentameric complex,which can be composed from the members of eightrecognized families of glycoprotein subunits (a, b, g, d, «,p, u, and r) (Fig. 6). Several of these subunit families areknown to include multiple isoforms (e.g., a1–a6 andg1–g3), giving rise to at least 19 distinct subunits that,when combined, permit the assembly of a large numberof receptor subtypes with variable affinities for GABAARmodulators (Barnard et al., 1998; Smith et al., 2001).Each of these receptor subunits has an extracellulardomain, which serves as a ligand-binding site, and fourtransmembrane domains, which are involved in chlorideion channel structure (Fig. 7).The majority of GABAARs are composed of two a

subunits, two b subunits, and a single g subunit (Pritchettand Seeburg, 1991; Chang et al., 1996; Graham et al.,1996; Tretter et al., 1997; Wingrove et al., 2002), with theGABA binding site located on the a/b subunit (Casalotti

et al., 1986). Residues that contribute to the GABAbinding site are F64 on the a subunit and Y157, T160, andY205 located on the b subunit (Amin and Weiss, 1993;Amin et al., 1997; Baur and Sigel, 2003). Substitution ofany of these four amino acids decreases the sensitivityof the GABAAR to activation by GABA and muscimol.However, these same amino acid substitutions do not alterGABAAR activation by pentobarbital, indicating thatthese three compounds act at distinct binding sites (Aminand Weiss, 1993). A large fraction of GABAARs have asubunit composition of a1b2g2, and possess many of thepharmacologic properties associated with the classic BZDI or type 1 receptors.

Compounds that affect GABAAR-mediated responsesfall into three categories: agonists, antagonists, andmodulators. Agonists, such as muscimol, directly andspecifically interact with the GABA recognition site onthe GABAAR to induce a response. Competitive antag-onists, such as bicuculline, or noncompetitive antago-nists, such as picrotoxin, bind to the receptor and blockGABA from activating a response via its recognitionsite. Allosteric modulators bind at modulatory sites andincrease, decrease, or null modulate the binding ofGABA to the GABAAR, thereby enhancing, inhibiting,or null modulating the ensuing response to GABA (Farbet al., 1984; Chan and Farb, 1985). Chlordiazepoxideand diazepam were the first demonstrated therapeuticsto act as positive allosteric modulators at the GABAAR(Braestrup and Squires, 1977; Choi et al., 1977; Möhlerand Okada, 1977; Macdonald and Barker, 1978; Gallagerand Tallman, 1983; Ferrero et al., 1984). These com-pounds appear to impart a conformational change in thetransmembrane receptor that alters its affinity for GABAand GABA agonists (Czajkowski and Farb, 1986; Lavoieand Twyman, 1996; Boileau and Czajkowski, 1999).Activation of GABAARs with the agonist muscimol alsoincreases the affinity for the receptor of positive allostericmodulators [e.g., diazepam and the triazolopyridine CL218872 (3-methyl-6-[-3-(trifluoromethyl)phenyl]-1,2,4-triazolo[4,3-b]pyridazine)], indicating that this rela-tionship is also reciprocal (Chiu and Rosenberg, 1979;Lippa et al., 1979; Villiger, 1984). Sensitivity to BZDs

Fig. 6. Schematic representation of GABAAR and the GABAergic synapse. (A) Top view of GABA and BZD binding sites. (B) Side view. Adapted fromBerezhnoy et al. (2007).

1014 Farb and Ratner

Page 14: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

appears to be mediated primarily by the g2 and a1, a2,a3, and a5 subunits (Brooks-Kayal and Pritchett, 1993;Boileau and Czajkowski, 1999).Dissimilar compounds influence GABAARs differently.

For example, although BZDs and barbiturates both acton GABAARs, electrophysiological studies suggest thatthe actual mechanism of action is different. These studiesindicate that diazepam increases the duration of GABA-induced channel openings or burst by only 9.3%, butincreases the number of bursts by 102.5%. In contrast,phenobarbital increases burst duration by 81.9%, butincreases the number of bursts by only 6.3% (Twymanet al., 1989). Although the mechanism of receptormodulation has not been fully elucidated, BZDs appearto enhance single-channel burst frequencies by increas-ing the affinity of GABA for its binding site, whereasbarbiturates appear to slow receptor deactivation bystabilizing the interaction between GABA and theGABAAR (Mathers, 1985; Twyman et al., 1989; Rabowet al., 1995; Steinbach and Akk, 2001). The binding ofBZDs to the allosteric BZD site increases the affinity ofthe receptor for GABA and enhances GABAergic neuro-transmission by increasing the number of chloridechannels opened at a given synaptic concentration ofGABA, which in turn hyperpolarizes the cell membrane.

The a subunit appears to be intimately related toGABAAR sensitivity to the prototypical BZDs andparticularly to diazepam, playing an important rolein drug binding and concentration response relation-ships (Pritchett et al., 1989; Pritchett and Seeburg,1990; Wafford et al., 1993; Smith et al., 2001) viainteracting with the a/g interface (Buhr and Sigel,1997). The g subunit contributes to receptor affinity forspecific ligands as well as the single-channel character-istics (Herb et al., 1992; Günther et al., 1995; Benkeet al., 1996).

GABAAR subtypes that are modulated by BZDs canbe divided into those that are diazepam sensitive andthose that are not (Lo et al., 1982). BZDRs that recog-nize the classic 5-phenyl-1,4-BZDs (e.g., diazepam andflunitrazepam) are referred to as “diazepam-sensitive”receptors, whereas those that do not recognize theseligands are referred to as “diazepam-insensitive” recep-tors (Malminiemi and Korpi, 1989; Hadingham et al.,1996; Knoflach et al., 1996). It was originally thought thatthere were just two subtypes of BZD-sensitive GABAARs.The first group, type 1 receptors that are enriched in thecerebellum and globus pallidus, display a higher affinityfor the triazolopyridine CL 218872 and the b-carboline[3H]propyl-b-carboline-3-carboxylate. By contrast, type II

Fig. 7. Molecular docking and transmembrane structure of the GABAAR. (A and B) The GABAAR interface (A) and structures (B) of BZD binding siteligands used in the study. Panel A shows the homology model of a1 and g2 subunits of the GABAAR with BZD binding site loops highlighted in yellow;loop F is green. Dashed lines indicate limits of cell membrane. Figure courtesy of Dr. Cynthia Czajkowski. Also see Hanson and Czajkowski (2008).

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1015

Page 15: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

receptors, which are found in the superficial layer of thesuperior colliculus, striatum, and the dentate gyrus ofthe hippocampus, show low affinity for CL 218872 and[3H]propyl-b-carboline-3-carboxylate (Braestrup andSquires, 1977, 1978; Braestrup and Nielsen, 1981;Young et al., 1981). It has since been shown that thereare multiple classes of BZDRs, reflecting differencesin GABAAR subunit compositions such as substitutionof the g3 and g2 subunits.Overall, it appears that sedation and amnesia associated

with classic BZD actions are mediated primarily via a1

subunit–containing GABAARs in rodents, whereas re-duction of vigilance is mediated primarily via the a2 anda3 subunits (Löw et al., 2000; McKernan et al., 2000; Diaset al., 2005), with activation of the g subunit also playing arole in BZD efficacy (Rudolph et al., 1999). The a2 subunitis largely expressed in limbic structures implicated inanxiety and fear, including the amygdala and hippocam-pus. Staining intensities of the a2 subunit are highest inthe accessory olfactory bulb, molecular layer of the dentate,hippocampal area CA3, central and lateral amygdaloidnuclei, septum, striatum, accumbens, and hypothalamus.The lateral septum has a high concentration of a2

subunits and has been shown to participate in theregulation various visceral and somatic functions aswell as in modulation of anxiety, aggression, and fearresponses in rodent models (Pesold and Treit, 1996;Gavioli et al., 1999). Neurons in this region are alsonot sensitive to zolpidem-induced enhancement ofGABAergic inhibition, which has been associated withactivation of receptors containing the a1 subunit (Duncanet al., 1995). Cat odor exposure induces Fos expres-sion in the posterior accessory olfactory bulb, whichhas also been shown to contain a high concentrationof the a2 subunit (McGregor et al., 2004). Cat odorinduction of Fos expression is also seen in the mainolfactory bulb and midazolam inhibited Fos expres-sion in this region. Midazolam also inhibited Fosexpression in key limbic regions involved in pheromonetransduction (medial amygdala and BNST) and de-fensive behavior (prelimbic cortex, lateral septum, lateraland medial preoptic areas, and dorsal premammillarynucleus).H101R mice in which the a2 subunit was rendered

insensitive to diazepam by a knock-in point mutation(His101→Arg) show reduced sensitivity to the anxio-lytic effects of diazepam. This mutation was notassociated with any overt phenotypic changes and theanimals expressed all subunits tested (a1, a2, a3, b2/3,and g2) at normal levels and distribution. After ad-ministration of diazepam, the a2 knock-in mice did notshow the increase in exploratory behavior (as mea-sured by the amount of time spent and the number ofentries into the open arms) on the elevated plus mazethat is typically seen in wild-type mice. This differencein behavior could not be attributed to motor impair-ment because the motor activity in the enclosed arms

was similar in a2 H101R and wild-type mice irrespectiveof the treatment (Löw et al., 2000).

A subsequent study looked at the behavior of H101Rmice in a conditioned emotional response task (Morriset al., 2006). In these experiments, lever pressing forfood on a variable interval schedule of reinforcementwas suppressed by delivery of a foot shock pairedwith a conditioned stimulus tone or light. During theconditioned emotional response test session, wild-typeand H101R mice were pretreated with diazepam(0, 0.5, 1, and 2 mg/kg) 30 minutes prior to testing.The wild-type mice showed an increase in lever pressingduring treatment with diazepam, whereas H101R micewere resistant to the anxiolytic effects of diazepam.

These observations naturally suggest that develop-ment of compounds that are selective for those receptorsubtypes implicated in anxiety may prove to be extremelyfruitful in the management of anxiety disorders. To date,efforts to fine-tune the binding affinity or functionalefficacy of compounds that display selectivity or prefer-ence for specific receptor subtypes remain promisingin rodent experimental animal systems, yet elusivein human clinical trials (Ballenger et al., 1991, 1992;Rudolph et al., 1999; Löw et al., 2000; Sandford et al.,2001; Atack, 2003; Lippa et al., 2005; Rudolph andMöhler, 2006; Atack et al., 2011a,b; Olivier et al., 2013).Additional consideration for targeting therapeutics,which take advantage of the rapid rates of GABAARturnover and the nonlysosomal pathway for degradation(Borden et al., 1984; Borden and Farb, 1988), may alsoyield greater selectivity and reduced adverse events.Further research directed along a neural-systems levelmay well resolve the drug discovery puzzle at hand ifthe receptor subtype and selective modulatory thera-peutic can be effectively aligned for the treatment ofspecific anxiety disorder subtypes.

2. Serotonin. The role of 5-HT in the pathophysiol-ogy of anxiety disorders has become increasingly wellestablished, and agents that inhibit the reuptake ofthis neurotransmitter now represent important com-ponents of current approaches to anxiolysis (Graeff andZangrossi, 2010; Sertraline, 2012; Venlafaxine, 2012;Fluoxetine, 2013). 5-HT is amonoamine whose postsynapticaction is terminated via 5-HT reuptake into the presynapticnerve terminal via a 5-HT transporter. The efficacy of SSRIsis thus based on augmenting the concentration of 5-HT inthe synaptic cleft by inhibiting its clearance (Ressler andNemeroff, 2000; Nichols and Nichols, 2008).

Effects of 5-HT are mediated by as many as 14 receptorsubtypes, including 13 G protein–coupled receptors(GPCRs) and a single ligand-gated ion channel. Thesereceptors are divided into seven distinct classes (5-HT1

to 5-HT7) (Fig. 8); however, postgenomic modificationscan produce at least 20 additional GPCRs, leaving morethan 30 5-HT receptors that signal through G proteins.In addition, activation may result in variable effects,depending on the brain region in which the receptors

1016 Farb and Ratner

Page 16: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

are expressed and on individual differences in respon-siveness mediated by genetic polymorphisms (Raymondet al., 2001; Hoyer et al., 2002; Gray and Roth, 2007).Although it is tempting to conceptualize the actions

of 5-HT at its receptor sites as being either inhibitoryor excitatory, the complexity of the signal transductionpathways activated ultimately may confound suchan appealing target- and hypothesis-driven approach.That said, 5-HT receptors generally have the followingfunctions (Polter and Li, 2011): 1) 5-HT1 receptors inhibitadenyl cyclase, reduce cyclic AMP, and inhibit proteinkinase A (PKA); 5-HT1A receptors are the prototypical5-HT1 receptors; 2) 5-HT2 receptors increase inositoltrisphosphate, increase intracellular calcium levels, increase

diacylglycerol, and activate PKA; 5-HT2A receptors are theprototypical type 5-HT2 receptors; 3) 5-HT3 receptors areligand-gated cation channels; 4) 5-HT4 receptors activateadenyl cyclase, increase cyclic AMP, and activate PKA; 5)5-HT5 receptors inhibit adenyl cyclase, reduce cyclic AMP,and inhibit PKA; and 6) 5-HT6 and 5-HT7 receptors activateadenyl cyclase, increase cyclic AMP, and activate PKA.

The complex distribution and functionality of thesereceptor subtypes contributes to their unique ability tomodulate emotional responses and other bodily func-tions via the systems into which they feed forward.The 5-HT3 receptor is a ligand-gated ion channel. Theremaining subtypes are GPCRs with the latter alsobeing defined as a type A family, rhodopsin-like receptors.Evidence suggests that most, if not all, of the 5-HT GPCRsundergo dimerization, and that dimerization may benecessary for proper functioning of the receptor (Nicholsand Nichols, 2008). For example, when inactive 5-HT2C

receptors are coexpressed with a wild-type 5-HT2C re-ceptor, the inactive receptor is able to inhibit theactivity of the wild-type receptor through the formationof a nonfunctional heterodimer, suggesting that di-merization is essential for receptor function (Herrick-Davis et al., 2005). Studies to date suggest that the5-HT receptors primarily related to anxiety and moodare 5-HT1A, 5-HT2A, and 5-HT2C (Toth, 2003; Van Oekelenet al., 2003; Mato et al., 2010).

Interestingly, the roles of these receptor subtypesmay vary depending on the form of anxiety involved, asoutlined in a review by Graeff and Zangrossi (2010).The authors suggest that SSRIs improve symptomsin patients with panic disorder by enhancing theresponsiveness of 5-HT1A and 5-HT2A receptors in thedorsal periaqueductal gray matter in the midbrain, butimprove symptoms in patients with generalized anxi-ety through the desensitization of 5-HT2C receptors and,perhaps, through stimulation of 5-HT1A receptors in theforebrain.

Understanding the effects of 5-HT in anxiety disordersrequires attention to variations in transporter reuptakesubtypes that may potentially influence the clinicalresponse to SSRIs. Genetic polymorphisms of the 5-HTtransporter gene promoter (5-HTTLPR) have beenassociated with efficacy of antidepressants (Porcelliet al., 2012), as well as with anxiety. For example,a functional polymorphism of the 59-flanking regionof the 5-HT transporter gene (SLC6A4) is associatedwith differences in anxiety-related personality traits(Heils et al., 1996; Lesch et al., 1996). A 43-bp deletion/insertion in 5-HTTLPR is associated with long “L”(16 repeats) and short “S” (14 repeats) alleles (Heilset al., 1996). There appear to be ethnic differences in theprevalence of these polymorphisms as well, with Cau-casians having about 22% S/LG alleles and Asians havingabout 60% S/LG alleles (Hu et al., 2006). Gene/environmentinteractions may therefore account for susceptibilityto anxiety disorders and for patient response to

Fig. 8. Model of a serotonergic neuron and synapse. 5-HT1A receptors canfunction both as autoreceptors on the serotonergic cell body and as postsynapticreceptors. Most 5-HT receptors are GPCRs and are located postsynaptically toserotonergic neurons. The 5-HT3 receptor is the exception because it is a ligand-gated ion channel. AC, adenylyl cyclase; DAG, diacylglycerol; GIRK, G protein–coupled inwardly rectifying potassium channel; IP3, inositol trisphosphate;PLC, phospholipase C; TPH, tryptophan hydroxylase. Adapted from Gray andRoth (2007).

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1017

Page 17: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

pharmacological treatment with SSRIs and SNRIs (Leschet al., 1996; Nakamura et al., 2000; Hu et al., 2006;Klumpers et al., 2012). The L(A) allele is overtransmittedby 2-fold to patients with OCD (Hu et al., 2006), whereassubjects with at least one short allele exhibit greaterfear-potentiated startle than do subjects with the longallele (Klumpers et al., 2012).Another genetic factor implicated in the clinical re-

sponse to SSRIs is a polymorphism in the gene that codesfor RGS2, a protein involved in neuronal GPCR responsesto neurotransmitters including 5-HT. Stein et al. (2014)found that a mutation that reduces expression of RGS2is associated with a poor clinical response to sertralinetreatment.

V. Neuropsychopharmacology ofAnxiety Disorders

Different combinations of subunits comprise theGABAA, NMDA, and 5-HT receptors, providing tre-mendous variability in affinity for endogenous andexogenous ligands and thus for the ability of the receptorto modulate neuronal function (Smith et al., 2001;Nichols and Nichols, 2008). Below, we review a rangeof established and investigational anxiolytic agents,outlining the impact of selective modulation of receptorsubtypes where data are available. The pharmacology ofdrug action is inherently the result of the net outcome ofsystems-level actions of the neuroactive agent. Therefore,it must be considered that, although subtype-preferringcompounds exist, highly selective therapeutics have notyet been developed.

A. Positive Allosteric Modulation ofGABAergic Transmission

1. Benzodiazepines and Related Modulators.Although the actions of BZDs on GABAARs can reduceanxiety, this therapeutic effect is not without severaladverse side effects, including drowsiness and ataxiaas well as the risk of dependence and withdrawal reactions(Noyes et al., 1988; Bennett et al., 1998; Yamawaki, 1999;Allgulander et al., 2003). Because of these untoward effects,the use of BZDs for long-term treatment of chronic anxietydisorders such as GAD is undesirable (Allgulander et al.,2003). As indicated previously, side effects and efficacy ofanxiolytic effects may be mediated through differenta subunits (Löw et al., 2000; McKernan et al., 2000;Dias et al., 2005), providing one impetus for researchinto the development of more selective compounds.Partial positive allosteric modulators have a lower

intrinsic ability to induce a modulatory effect at givenfractional occupancy as compared with full allostericpositive modulators and represent an approach todeveloping anxiolytics with fewer side effects and toxicity(Atack, 2003). The relationship between fractional re-ceptor occupancy and fractional effect can be assessed bydetermining the percentage of inhibition of flumazenil

binding to GABAARs and potentiation of GABA-evokedchloride currents. Potentiation of GABA-gated currentsby 25% occurs at about 35% receptor occupancy fordiazepam and about 45% for triazolam (Facklamet al., 1992b). The benzoquinolizinone Ro 19-8022[(R)-1-[(10-chloro-4-oxo-3-phenyl-4H-benzo[a]quinolizin-1-yl)carbonyl]-2-pyrrolidine-methanol], a BZDR partialallosteric modulator, produced 25% potentiation whenreceptor occupancy reached about 95%. However, theimidazobenzodiazepinone bretazenil, or Ro 16-6028[(13aS)-8-bromo-11,12,13,13a-tetrahydro-9-oxo-9H-imidazo[1,5-a]pyrrolo[2,1-c][1,4]benzodiazepine-1-carboxylicacid 1,1-dimethylethyl ester], which also acts as a partialallosteric positive modulator, did not produce 25%potentiation even at 100% receptor occupancy. Thesein vitro observations can be related to in vivo responsesin animal models, which indicate that bretazenil doesnot protect against seizures as well as the full allostericpositive modulator diazepam (Brouillet et al., 1991;Haefely et al., 1992). By contrast, most studies in humansand animals indicate that bretazenil can provide anxio-lytic effects with less sedation and motor impairmentsthan diazepam (Facklam et al., 1992a; Cole and Rodgers,1993; Busto et al., 1994). However, there is an increase insymptoms of sedation and deficits in certain measures ofattention and executive function (digit symbol) amongsubjects receiving 0.5 mg bretazenil (van Steveninck et al.,1996).

Although most compounds possessing the b-carbolinestructure are anxiogenic, the b-carboline abecarnil(isopropyl-6-benzyloxy-4-methoxymethyl-b-carboline-3-carboxylate) produces anxiolytic effects without sub-stantial sedation (Stephens et al., 1990; Ballengeret al., 1992). Abecarnil exhibits high affinity for the BZDrecognition site, although no differences in affinitieswere observed for recombinant receptors expressed inhuman embryonic kidney cells containing a1, a2, or a3

subunits (Hadingham et al., 1993). Another study showsthat abecarnil only partially inhibits diazepam-insensitivebinding of [3H]Ro 15-4513 in the cerebellum, suggestingthat this site can be differentiated into abecarnil-sensitiveand abecarnil-insensitive components (Mehta and Shank,1995).

Electrophysiological studies indicate that abecarnilpotentiates GABA-induced chloride currents less thanfull allosteric modulators. Interestingly, abecarnil ismost effective with the fewest side effects at the lowestdose examined in a double-blind, placebo-controlledclinical trial for treatment of GAD (Ballenger et al.,1991, 1992). However, higher concentrations (.7.5 mg/day)of abecarnil were associated with reports of symptoms andsigns of CNS depression including somnolence, difficultyconcentrating, and dizziness. These observations suggestthat efficacy alone may not account for all of the adverseside effects of BZDs.

The cyclopyrrolone pagoclone, another compoundbelieved to act as a partial positive allosteric modulator

1018 Farb and Ratner

Page 18: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

of the GABAAR, has also been investigated for itsanxiolytic effects. A randomized double-blind study ofpatients suffering from panic attacks found that thiscompound provides anxiolytic effects without the typicalside effects associated with BZDs (Sandford et al., 2001),but it has not been approved for use in the treatment ofanxiety disorders (Bateson, 2003; Atack et al., 2006a; deWit et al., 2006).Agents such as imidazenil and SL651498 [6-fluoro-9-

methyl-2-phenyl-4-(pyrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyrido[3,4-b]indol-1-one], which have a relativelyhigher affinity for specific GABAAR subunits, such asthe a2 subunit, also appear to produce reduced mea-sures of vigilance (a model for anxiolytic effects inhuman) with fewer side effects than the classic BZDs(Sieghart, 1994; Griebel et al., 2001; Williams and Akabas,2001; Collins et al., 2002; Licata et al., 2005). This isconsistent with the hypothesis that the a2 subunit–containing receptor subtype, which is largely expressedin the limbic system, may be an important contributorto the subtle positive modulation of GABAergic in-hibition necessary to induce a reduction in vigilancein rodents and possibly induce anxiolysis in humanswithout undesirable side effects (Pritchett et al., 1989;Pritchett and Seeburg, 1990; Löw et al., 2000; Kaufmannet al., 2003; Licata et al., 2005). Yet bridging the gapbetween convincing results of a therapeutic candidatein animal models for anxiety and its beneficial clinicalutility in humans remains an elusive if not dauntingobjective.Research into the allosteric modulatory effects of com-

pounds such as zolpidem, which act on the BZD site butdo not possess the 7-member ring associated with thestructure of the classic BZDs, has yielded compounds witha preference for specific GABAAR subunits (Ghiani et al.,1994; Serra et al., 1994; Drover, 2004). For example,zaleplon, zolpidem, and zopiclone exhibit a modestpreference of up to 6-fold for GABAARs containing a1

subunits compared with a2 or a3 subunits using modelsystems (Drover, 2004), and are effective sedative/hypnotic agents reported to produce less toleranceand dependence than diazepam (Follesa et al., 2002).Compounds that do not act at GABAARs containing

a1 subunits may be useful as nonsedating anxiolytics.Whereas zolpidem and zaleplon appear to lack signif-icant anxiolytic effects and seem to also not have anyactive metabolites, zopiclone is metabolized in vivoto the active metabolite (S)-desmethylzopiclone, whichhas been shown to produce anxiolysis with fewer neg-ative effects on locomotor activity than diazepam andalprazolam, suggesting that structurally related com-pounds may be useful in the treatment of anxietydisorders (Carlson et al., 2001; Fleck 2002).Ocinaplon (pyrazolo[1,5-a]-pyrimidine) is a novel

pyrazolopyrimidine that potentiates GABAergic neuro-transmission, increases the response of rats and primatesin measures of anxiety (conflict test), and attenuates

symptoms among human subjects with GAD as demon-strated by scores on the Hamilton Anxiety Rating Scales(Lippa et al., 2005). Electrophysiological studies indicatethat ocinaplon has lower potency and efficacy thandiazepam. However, the potentiation of GABA-gatedcurrents in recombinant receptors expressed in Xenopusoocytes observed in this study indicates that ocinaplonhas greater efficacy at a1 subunits than at a2 subunits,a finding that on the surface appears to be in conflictwith the hypothesis that anxiolysis is mediated by a2

subunit–containing GABAARs.A compound with comparable binding affinity but

different efficacies at the various receptor subtypescan exert its effects predominantly at those receptorsubtypes implicated in anxiolysis, rather than havingnonspecific effects and/or producing untoward sideeffects such as sedation (Atack, 2003). Interestingly,although ocinaplon shows selectivity for modulating a1

subunit–containing GABAARs, competitive bindingassays using native GABAAR membrane preparationsderived from rat cerebellum (wherein a1 subunits arehighly expressed) indicate that this compound is also muchless potent at inhibiting the binding of flunitrazepamthan is diazepam (Lippa et al., 2005). Since the clinicalpotency of a drug depends on its binding affinity for thetarget and its efficacy of signal transduction, it is plausiblethat lower receptor occupancy may contribute to theability of ocinaplon to produce anxiolysis without inducingsedation.

Alternatively, some positive modulation of GABAARscontaining a1 subunits may be necessary to effectivelyinduce anxiolysis in the human nervous system com-pared with mouse models, wherein even mild dysreg-ulation of higher-order cognitive processes mediated bythe frontal lobes has been implicated in controllingthreat assessment and anxiety (Cha et al., 2014). Ir-respective of which of these hypotheses or others turnsout to be themost valid, the development of this promisingcompound was halted due to reports of elevated liverenzymes in just a few subjects, underscoring the inherentdifficulties associated with bringing a novel anxiolytic tomarket even using a well defined target.

SL651498 is a pyridoindole that preclinical studiessuggest acts at the BZD binding site to produce anxiolyticeffects qualitatively similar to those of BZDs with re-duced ataxia or sedation. This agent is a full positivemodulator at recombinant rat GABAARs containing a2 ora3 subunits (approximately 95% enhancement) and apartial positive modulator at receptors containing a1 ora5 subunits (approximately 45% enhancement). Binding-affinity assays indicate that SL651498 does not differen-tiate between a1 (17 nM), a2 (73 nM), and a3 (83 nM)subtype–containing GABAARs (Griebel et al., 2001),indicating that binding affinity does not contribute tothe anxiolytic actions of this drug.

2. Natural Product Modulators. Some natural productsmay also provide new platforms for discovery. For example,

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1019

Page 19: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

chrysin, one of the first flavonoids shown to interactwith the BZD binding site, initiated the search fornatural anxiolytics among traditional medicinal herbs(Medina et al., 1989, 1990, 1997; Wolfman et al., 1994,1996). A number of other naturally occurring flavonoidsand their derivatives, such as K36 (5,7,29-trihydroxy-6,8-dimethoxyflavone) (Huen et al., 2003), hispidulin (Kavvadiaset al., 2004), wogonin (Hui et al., 2000, 2002), and relatedcompounds (Nielsen et al., 1988; Häberlein et al., 1994;Wolfman et al., 1996; Goutman et al., 2003), areallosteric modulators at GABAARs. In a mouse study,wogonin (7.5–30 mg/kg p.o.) was found to produce ananxiolytic response similar to diazepam in the elevatedplus maze without accompanying sedation (Hui et al.,2002). K36, a naturally occurring flavonoid, increasesentries onto the open arms of the elevated plus mazewithout changing locomotor activity, sedation, myorelax-ation, or motor incoordination in mice (Huen et al., 2003).3. Increasing GABA Levels. On the basis of all of

these results, it would be expected that increasingsynaptic concentrations of GABA, either by inhibitingits reuptake or degradation, could be another approachto enhancing GABAergic neurotransmission and re-ducing anxiety disorders. GABA-transaminase inhib-itors, such as vigabatrin (g-vinyl-GABA) and valproicacid (VPA) (Rando et al., 1981; Gram, 1988), increasesynaptic concentrations of GABA by preventing catab-olism of GABA (Zwanzger et al., 2001a,b; Nemeroff,2003). Vigabatrin raises levels of GABA in nerve terminalsand inhibits striatal dopamine release (Kushner et al.,1997). However, reports of psychosis and depressionassociated with vigabatrin suggest that this particularcompound may have significant clinical limitations(Levinson and Devinsky, 1999; Ashton and Young, 2003).In retrospect, elevating GABA in a way that is pro-portionate to its endogenous levels might be expected toactivate the natural distribution of relevant receptorsubtypes nonselectively, thus increasing the probabilityof side effects. A more nuanced, neural systems–levelapproach might be capable of modulating anxiety ina similar fashion to the effective SSRIs.

B. Modulators of Glutamatergic Transmission

A delicate balance between excitatory and inhibitoryneurotransmission is essential to modulation of anxi-ety and fear responses. Not surprisingly, disturbancesof glutamatergic as well as GABAergic neurotransmis-sion have been implicated in anxiety disorders (Jardimet al., 2005).1. Inhibitors of Glutamatergic Activity. Glutamatergic

neurotransmission in the lateral amygdala is essentialto the acquisition and extinction of fear-related memoriesand behavioral responses associated with re-exposure tofear-inducing stimuli (Sajdyk and Shekhar, 1997). Whena neutral stimulus is paired with an adverse stimulussuch as presentation of a tone paired with delivery ofa foot shock, the previously neutral stimulus is soon able

to elicit a fear response independently. NMDA receptorantagonists such as D,L-2-amino-5-phosphonopentanoicacid, which prevent induction of long-term potentiation,have also been shown to block the acquisition but notthe expression of conditioned fear responses in rodents(Miserendino et al., 1990; Falls et al., 1992). As pre-viously mentioned, the prefrontal cortex appears to bethe brain region most often implicated in attenuation ofanxiety.

Memantine is another NMDA receptor antagonistthat is currently approved for the treatment of Alzheimer’sdisease, but there is some evidence that it may also haveanxiolytic properties. A study in mice found that highstable doses of memantine (10, 30, and 100 mg/kgper day) produced some improvements in measures ofanxiety, including reductions in isolation-induced aggres-sion, escape latency (hidden platform), and wall swimmingtendency in the Morris water maze test (Minkevicieneet al., 2008). In addition, several case studies in humanshave shown some benefit for memantine in reducingsymptoms associated with PTSD, SAD, and GAD(Battista et al., 2007; Schwartz et al., 2012). For example,in a small study of four patients with combat exposure–induced PTSD, treatment with memantine led to consis-tent improvements in memory as well as depressive andhyperarousal symptoms (Battista et al., 2007).

Another study showed that acamprosate, an NMDAand metabotropic glutamate 5 receptor modulator, waseffective as a therapeutic for idiopathic and chemicalwithdrawal–induced anxiety disorders (Kotlinska andBochenski, 2008; Hertzman et al., 2009; Schwartz et al.,2010; Pałucha-Poniewiera and Pilc, 2012; Koltunowskaet al., 2013). Acamprosate seems to be a safe and ef-fective therapeutic option for patients presenting withcomorbid alcoholism. These findings were initiallynot entirely surprising given that enhancing inhibitoryneurotransmission is a well establishedmethod of treatinganxiety; thus, it logically followed that if acamprosateattenuated excitatory neurotransmission, it could yielda similar clinical outcome (Berton et al., 1998). However,subsequent research has revealed both enhancement aswell as inhibition of network synaptic elements underlyingglutamatergic and GABAergic transmission, further em-phasizing the need for a larger-scale understanding ofdrug action in the context of connectivity.

There is also evidence that riluzole, which reducessymptoms of GAD (Pittenger et al., 2008), may exertits effects by inhibiting voltage-gated sodium channelsand enhancing astrocytic uptake of extracellular gluta-mate (Urbani and Belluzzi, 2000; Wang et al., 2004). Inan open-label study of GAD patients, 8 of 15 patientstreated with riluzole had remission of anxiety over aperiod of 8 weeks, with a median time to response of2.5 weeks (Mathew et al., 2005). Thus, therapeutics thatinhibit glutamatergic neurotransmission (e.g., riluzole)may prove to be more useful in treating GAD and thehyperarousal seen in PTSD.

1020 Farb and Ratner

Page 20: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

2. Potentiators of Glutamatergic Activity. Admin-istration of D-cycloserine, a partial NMDA receptoragonist, has been shown to facilitate extinction of learnedfear responses in rodents and of phobias, SAD, and PTSDamong patients undergoing behavioral exposure therapy(Walker et al., 2002; Ressler et al., 2004; Woods andBouton, 2006; Mao et al., 2008; Aupperle et al., 2009;Difede et al., 2014).A randomized, double-blind, placebo-controlled clinical

trial examining the efficacy of a combination of D-cycloserineor placebo and exposure therapy for treatment of SADrevealed that short-term dosing of partial NMDA receptoragonists may also be effective in treating this disorder(Hofmann et al., 2006). This compound seems to be mosteffective for treating anxiety disorders that respond to“exposure therapy” in which learning not to fear thestimulus facilitates improvement in symptom measures.In a similar study, Difede and colleagues (2014) comparedthe effectiveness of VRE therapy alone versus thecombined treatment with D-cycloserine plus VRE, andfound that D-cycloserine augmented the therapeutic re-sponse to VRE, suggesting that negative memories maybe extinguished using cognitive enhancers. Overall, theseobservations suggest that compounds that increaseglutamatergic neurotransmission are useful in the treat-ment of certain types of anxiety disorders (e.g., specificphobias) and PTSD wherein re-exposure to a specificsalient stimuli can trigger a pathologic response thatinterferes with activities of daily living (Heresco-Levyet al., 2002; Difede et al., 2014).3. Neurosteroids as Modulators of Glutamatergic

Transmission. The observation that stress-inducedanxiety changes glutamatergic neurotransmission (Masneufet al., 2014) raises the possibility that endogenousmechanisms for modulating the balance of excitatoryto inhibitory neurotransmission could underlie thegenesis of anxiety. Neurosteroids such as pregnanolonesulfate, which enhances cognition, positively modulatesNMDAR function, and stimulates NMDAR trafficking tothe cell surface (Wu et al., 1991; Kostakis et al., 2013),may represent one such endogenous mechanism forlinking fearful stimuli and the emotional responsesubject to therapeutic manipulation (Plescia et al.,2013). Neuroactive steroids, such as pregnanolonesulfate and pregnanolone hemisuccinate, which in-hibit glutamatergic neurotransmission by negativelymodulating NMDAR function (Park-Chung et al.,1994) and may also exhibit mixed function by servingas both a negative modulator of the NMDAR anda prodrug for pregnanolone (a positive modulatorof GABAAR) delivery across the blood–brain barrier,may also represent a novel platform for discovery(Weaver et al., 1997, 2000).

C. Dopaminergic Transmission

In a recent study by Zweifel and colleagues (2011),cue conditioning was found to be significantly impaired

in knockout mice lacking functional NMDA receptorson dopaminergic neurons of the ventral tegmentalarea. This impairment was associated with developmentof a GAD-like phenotype, suggesting that dopaminergicneurotransmission plays a role in this anxiety disorder.Interestingly, increased anxiety is among the symptomsassociated with dopamine agonist withdrawal in humans(Nirenberg, 2013). The interplay between the glutamatergicand dopaminergic systems is well studied in drug ad-diction, wherein motivation for drug seeking is mediated,in part, by this relationship. Nucleus accumbens and pre-frontal cortex dopaminergic and glutamatergic systemshave been implicated in anxiety disorders and addiction(Ahmadi et al., 2013) and it has been suggested that theD2/D3 receptor agonist quinpirole may induce compulsivechecking behavior in rodents by enhancing dopaminergicactivity (Alkhatib et al., 2013). The role of motivation inOCD is well established and plays a key role in the abilityof an individual to override his or her pathologic responsesto anxiety-inducing stimuli; it is not uncommon for pa-tients with OCD and phobias to temporarily overcometheir fears to avoid life-threatening events.

In contrast with the D2/D3 receptor agonist quinpirole,the anxiolytic buspirone has been shown to block D3receptors in primates (Kim et al., 2014). However, therole that this mechanism plays in attenuating anxietyis not entirely clear since the compound also acts atserotonin and adrenergic receptors (Skolnick et al.,1984).

D. Epigenetics of Anxiolysis: HistoneDeacetylase Inhibitors

Combining histone deacetylase inhibitors with CBTcan also facilitate extinction of learned fear responses inhumans and animals (Heinrichs et al., 2013; Kuriyamaet al., 2013). Administration of VPA at a dose of 400 mg/kgto human subjects was shown to augment acquisitionand delayed offline consolidation of extinction training(Kuriyama et al., 2013). The unexpected effect of VPAon acquisition could be due to in part to an anxiolyticeffect associated with enhanced GABAergic neurotrans-mission mediated by inhibition of GABA transaminaseby VPA at this dose.

VPA seems to be rather nonspecific in terms of mech-anisms of action, at least at present. For example, inaddition to inhibition of histone deacetylases, VPA alsoincreases levels of brain-derived neurotrophic factorand cAMP response element-binding protein in thehippocampus and amygdala (Jeon et al., 2006; Casuet al., 2007; Jornada et al., 2010). A case of familialbipolar disorder with comorbid anxiety has beenstudied as part of the National Institute of MentalHealth Human Genetics Initiative and was found to beassociated with the genes that code for the extracellu-lar signal-regulated kinase/mitogen-activated proteinkinase and cAMP response element-binding protein–regulated intracellular signaling pathways (Kerner

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1021

Page 21: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

et al., 2013). Although further research in this area isneeded, these findings suggest that behavioral modifi-cation therapy could be combined with drugs thatmodulate gene expression to facilitate extinction of thefear-inducing stimuli associated with specific phobiasand PTSD.

E. Modulators of Serotonergic andNoradrenergic Transmission

Several studies reveal the potential benefits of SSRIsover BZD pharmacotherapy of certain types of anxiety.For example, it has been shown that patients whopresent with comorbid symptoms of depression andanxiety may respond better to SSRIs, whereas thosewho suffer from anxiety alone may respond better toBZDs. In addition, patients suffering from GAD re-spond well to BZDs, whereas those with PTSD andOCD have a relatively better clinical response to SSRIs(Rickels and Rynn, 2002; Varia and Rauscher, 2002;Blanco et al., 2003; Davidson, 2003).Paroxetine was the first SSRI to receive approval

from the US Food and Drug Administration for thetreatment of anxiety disorders and was superior toplacebo in short-term clinical trials among patientswith GAD (Allgulander et al., 2003). The SSRI fluoxetineis widely used for the treatment of patients who arecomorbid for depression and anxiety (Cassano et al.,2004). It seems unclear whether the anxiolytic effects offluoxetine may paradoxically be due entirely to enhance-ment of GABAergic tone at the neural-systems level.Electrophysiological results based on recombinantGABAARs transiently expressed in mammalian cellsreveal that fluoxetine increases the response of thesereceptors to submaximal GABA concentrations, althoughit does not alter the maximum current amplitude(Robinson et al., 2003), and this result is of uncertainrelevance to the physiologic effects of SSRIs.Although data are less abundant for SNRIs, several

clinical trials have established the efficacy of venlafaxinein patients with anxiety (Liebowitz et al., 2005; Steinet al., 2005). In one double-blind study of adult patientswith generalized SAD (N = 271, intent to treat), venlafaxineextended release produced significant improvements in theLiebowitz Social Anxiety Scale total score, Clinical GlobalImpressions-Severity of Illness scale, and the Social PhobiaInventory, compared with placebo over 12 weeks (Liebowitzet al., 2005). Similarly positive results were seen over6 months (Stein et al., 2005).A recent meta-analysis compared nine drugs for the

treatment of GAD in the United Kingdom (duloxetine,escitalopram, fluoxetine, lorazepam, paroxetine, pregabalin,sertraline, tiagabine, and venlafaxine). The investigatorsfound that fluoxetine was ranked first for response andremission (probability of 62.9 and 60.6%, respectively),whereas sertraline was ranked first for tolerability (49.3%)(Baldwin et al., 2011). These findings are generally in linewith current practice, in which SSRIs are often the

first-line treatment of a range of anxiety disorders. An-other review in the United States assessed data fromplacebo-controlled clinical trials of BZDs, azapirones,antidepressants, and anticonvulsant and antipsychoticdrugs, and concluded that patients with general anxietydisorder should receive either an SSRI or SNRI as first-line treatment (Reinhold et al., 2011).

F. Neuroactive Steroids as Neuromodulatorsof Transmission

Neuroactive steroid modulation of GABAergic neu-rotransmission in the central amygdala has beenimplicated in anxiety (Wang et al., 2007). There is someevidence that fluoxetine increases brain levels of theneurosteroid allopregnanolone, a positive allosteric mod-ulator of GABAARs (Matsumoto et al., 1999), and it hasbeen suggested that the anxiolytic effects of fluoxetineand possibly other SSRIs (e.g., paroxetine) are related tothis effect on allopregnanolone (Uzunova et al., 1998;Griffin and Mellon, 1999; Khisti and Chopde, 2000). Thishypothesis is supported by a recent study in a mousemodel of social isolation, which found that an SSRI-induced increase in allopregnanolone significantly con-tributes to the anxiolytic effects of SSRI treatment(Pinna, 2010). It is therefore of interest to take a closerlook at the mechanisms of action and modulatory sites ofneuroactive steroids, such as allopregnanolone.

The search for an endogenous system of new targetsfor modulation has been intensive, and neuroactivesteroid-based options offer some promise. Selye (1970)first demonstrated that endogenous steroids and theirmetabolites can produce rapid effects on CNS activity,including modulation of GABAergic neurotransmissionassociated with anxiety (Lambert et al., 2001, 2003;Yang et al., 2002).

The potential effects of neuroactive steroids on thedevelopment of anxiety disorders can be observed duringpuberty, a developmental stage associated with increasedlevels of reproductive hormones and onset of manypsychiatric disorders, including GAD, social anxiety,and panic attacks (Hayward and Sanborn, 2002; Kessleret al., 2005a; Merikangas et al., 2009, 2010; Reardonet al., 2009; Van Oort et al., 2009). Anxiety symptomshave been found to increase from middle to lateadolescence (Van Oort et al., 2009), with a particularlyhigh prevalence of all anxiety disorders reported amongadolescent girls (Merikangas et al., 2009; Leikangeret al., 2012; Legerstee et al., 2013).

Of particular interest in this setting are the neuroac-tive steroids allopregnanolone (in humans and rats) andpregnanolone (in humans only), metabolites of thereproductive hormone progesterone that are also pro-duced in the brain in response to stress (Purdy et al.,1991; Gïrdler et al., 2001). Allopregnanolone decreasesin the cerebral spinal fluid of women with PTSD(Rasmusson et al., 2006), but increases in depressedpatients receiving SSRIs (Uzunova et al., 1998). Moreover,

1022 Farb and Ratner

Page 22: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

administration of sodium lactate and cholecystokinin-tetrapeptide to persons with panic disorder–inducedattacks decreases plasma concentrations of bothpregnanolone and allopregnanolone, and increases theconcentration of the functional antagonistic isomer3b,5a-tetrahydroprogesterone (Ströhle et al., 2003).Together these findings suggest that changes in endoge-nous brain levels of neuroactive steroids associated withage, sex, stress, and administration of SSRIs may playa role in the onset and clinical response to treatment incertain anxiety disorders.Supporting this hypothesis are findings related to

the effects of allopregnanolone on gonadotrophin hormone-releasing hormone, the primary chemical messengerimplicated in the onset of puberty and sexual matura-tion (Moguilevsky and Wuttke, 2001). Allopregnanolonehas been found to suppress the release of hypothalamicgonadotrophin hormone-releasing hormone via alloste-ric modulation of GABAARs (Calogero et al., 1998; Simet al., 2001). Although this inhibition is increased byallopregnanolone administration before puberty and inadulthood, it is paradoxically reduced during puberty,leading to increased excitability of pyramidal cells inhippocampal region CA1. This effect appears to be duein part to inhibition of a4-containing GABAARs, whichare expressed at higher levels than normal in the CA1region of the hippocampus during puberty.It has also been shown that GABAARs of the a4b2d

subtype, which have a d subunit instead of a g subunit,play a role in tonic inhibition in areas such as the dentategyrus and cortex (Wisden et al., 1992). GABAAR-mediatedconductance is normally inhibitory; however, the reversalpotential of GABAAR-mediated postsynaptic current indentate gyrus granule cells is “positive” to the restingmembrane potential, making membrane hyperpolar-ization of GABAARs unlikely. Inhibition of shuntingappears to play a role in overcoming this process, such

that nonhyperpolarizing inhibitory conductance re-duces the depolarizing effect of postsynaptic poten-tials by decreasing proximal membrane resistance(Staley and Mody, 1992).

In the dentate gyrus and cortex, the GABAergiccurrent is inward (i.e., chloride flux is outward) (Staleyand Mody, 1992; Gulledge and Stuart, 2003); thus,inhibition in these areas is enhanced by allopregnanolone.However, in the CA1 region, the current is normallyoutward (Alger and Nicoll, 1982); thus, increasedexpression of a4b2d GABA receptors paradoxicallyresults in allopregnanolone attenuating rather thanenhancing inhibition. The reduction in current generatedby allopregnanolone at a4b2d GABAAR is dependentupon the presence of arginine 353 in the intracellularloop of a4, where it may serve as a chloride modulatorysite (Shen et al., 2007). This polarity-dependent decreasein inhibition mediated by allopregnanolone may haveimportant implications for how we approach the treat-ment of anxiety disorders in the future.

The modulation of GABAergic neurotransmission byneuroactive steroids is mediated by interactions withallosteric sites on GABAAR (Lambert et al., 2001,2003; Yang et al., 2002; Hosie et al., 2006). The in-teraction of certain neuroactive steroids with GABAARis stereoselective, suggesting that the binding sitesfor these compounds are of a specific dimension andshape (Harrison and Simmonds, 1984; Park-Chung et al.,1994; Covey et al., 2000). There is also evidence that thetransmembrane domains play a role in neuroactivesteroid–mediated modulation of GABAARs (Hosie et al.,2006) (Fig. 9), and that the effects of neuroactive steroids onGABAARs may be associated with its actions at d subunit–containing receptors (Mihalek et al., 1999, 2001; Spigelmanet al., 2003; Stell et al., 2003). Although changes in thesereceptors have not been associated with alterations inbehaviors considered indicative of vigilance or possibly

Fig. 9. Neurosteroid activity at the GABAAR is determined by a-subunit M1 domain residues. (A) The M1 through the end of M2 murine a1 and b2subunits were replaced with the corresponding sequence from the resistance to dieldrin (RDL) subunit, forming the chimeras aR and bR, respectively,which were previously shown to have very low sensitivity to potentiation and to lack direct activation by micromolar concentrations of neurosteroids.Polar residues present only in a1 (blue) are in bold, with Thr236 and Gln241 (red) highlighted. The transmembrane domains are boxed. Thepharmacology of receptors containing the modified a subunit was then compared with that of wild-type receptors using whole-cell recordings fromhuman embryonic kidney cells. (B) THDOC concentration–response curves for direct activation (red) and for potentiation (blue) of EC10 (concentrationcausing 10% of maximal response) GABA currents expressed as a percentage of the maximum GABA response, at wild-type a1b2g2 (squares) andchimeric aRb2g2 (circles) receptors showing that potentiation and direct activation by THDOC and allopregnanolone is abolished on receptorsincorporating aR. (C) Structures of ALLOP and THDOC. ALLOP, allopregnanolone; THDOC, tetrahydrodeoxycorticosterone. Reprinted withpermission from Hosie et al. (2006).

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1023

Page 23: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

anxiety in animals (Mihalek et al., 1999), evidencestrongly suggests that a change in subunit subtype froma1bxg2 to a4bxg2 increases receptor desensitization anddecreases GABAergic inhibition, a process that mayplay a role in epilepsy (Roberts et al., 2005, 2006; Lundet al., 2008; Grabenstatter et al., 2012). Could sucha change in subunit composition also underlie increasedanxiety and serve as a biomarker to identify patientsresistant to existing anxiolytic therapeutics? Couldswitching of receptor subunit subtypes—perhaps involvingthe a4 subunit, which is insensitive to BZD positivemodulators—be a control point in anxiolysis as in epilepsy?Both allopregnanolone and pregnenolone have been

implicated in the prevalence of anxiety disorders inmen and women (Le Mellédo and Baker, 2004), and arange of preclinical studies suggest potential mechanisticrationales for the anxiolytic effects of these neuroactivesteroids. It has been shown that allopregnanoloneexhibits anticonflict effects pharmacologically at 8 mg/kgin rats (Brot et al., 1997), and that it attenuates certaineffects of caffeine in rats indicative of vigilance or anxiety(Jain et al., 2005). Pregnanolone potentiates electro-physiological responses to and binding of muscimol toGABAARs (Majewska et al., 1988; Wu et al., 1993;Park-Chung et al., 1994), and exhibits sedative hypnoticproperties via its likely interaction with GABAARs(Wang et al., 2001). Overall, these observations suggestthat allopregnanolone and pregnanolone act as endoge-nous modulators of GABAergic neurotransmission invivo (Lambert et al., 2001, 2003; Yang et al., 2002;Ströhle et al., 2003). In support of the hypothesis thatallopregnanolone and pregnanolone have anxiolyticproperties, two small studies suggest that the plasma

concentrations of pregnenolone sulfate, a precursor ofthese steroids, is lower in males with generalized socialphobia or GAD (Semeniuk et al., 2001; Heydari and LeMellédo, 2002). Administration of pregnenolone is asso-ciated with an increase in allopreganolone and reducedactivity in the amygdala and insula, as well as a reductionin self-reported anxiety (Sripada et al., 2013) (Fig. 10).

Because they have poor bioavailability and canpotentially be metabolized to hormonally active ste-roids, endogenously occurring neuroactive steroidshave limited therapeutic use in patients. Syntheticanalogs of neuroactive steroids, which are more re-sistant to metabolism and better able to cross theblood–brain barrier, are now being investigated forpotential use as anxiolytics, anesthetics, and anticon-vulsants (Weaver et al., 1997; Gasior et al., 1999). Onesynthetic neurosteroid, pregnanolone hemisuccinate,produces sedation in mice and rats (Weaver et al.,1997), raising the possibility that it acts either byinhibiting the NMDA receptor or by crossing the blood–brain barrier and undergoing metabolism to pregnanolone.Synthetic neurosteroids bearing a hemisuccinate groupare more resistant to hydrolysis than the correspond-ing sulfate esters and are partly unionized at physiologicpH, allowing ready passage across the blood-brainbarrier.

Ganaxolone (3-hydroxy-3-methyl-5-pregnane-20-one),an orally active synthetic analog of allopregnanolone, isa positive allosteric modulator of GABAAR that hasshown promising basic and early stage clinical outcomesand is currently under investigation as a novel treat-ment of epilepsy and PTSD (Carter et al., 1997; Gasioret al., 1999; Reddy and Rogawski, 2012). Another

Fig. 10. Functional MRI studies showing that administration of PREG (400 mg) is associated with a reduction in activity in regions associated withgeneration of negative emotion. (A) Compared with placebo, pregnenolone administration decreased activation in right amygdala and right insula. (B)Compared with placebo, pregnenolone administration increased dorsal medial prefrontal cortex activation during appraisal. The percent signal changeis displayed next to each figure. PBO, placebo; PREG, pregnenolone administration group. Reprinted with permission from Sripada et al. (2013).

1024 Farb and Ratner

Page 24: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

synthetic analog of allopregnanolone, Co 3-0593 (3b-ethenyl-3a-hydroxy-5a-pregnan-20-one), was found to have anxi-olytic effects comparable to BZDs after both subcutane-ous and oral administration in rodents. An absence oftolerance to this drug is suggested by the fact that itseffects were maintained even after chronic administration(Wieland et al., 1997).Although these data suggest that anxiety disorders

may be amenable to treatment with synthetic neuro-steroids or through modulation of the synthesis ofendogenous neuroactive steroids, the more compellingvalue of these agents to date has been the insights theyhave provided into the pathophysiology of anxiety, bothinfluencing and being influenced by the GABAergic,serotonergic, and other neural systems in a range ofanxiety disorders.

VI. Conclusions

“Personalized medicine” is now a familiar concept, notonly in the scientific literature but also in the popularmedia. The concept is so familiar, in fact, that some arenow questioning whether the term has outlived itsusefulness, implying both too much and too little: toomuch in the sense that unique treatments are not beingdeveloped for every individual patient, and too little inthe sense that personalized medicine is what cliniciansalready aspire to every time they treat a patient(Katsnelson, 2013). Regardless of the term we use todescribe this aspiration, it is certainly true that the morewe are able to tailor treatments to a patient’s individualprofile, the greater the chance of clinical success. In thecases of mental disorders such as anxiety or depression,the idea of individualized approaches to therapy take oneven greater urgency, given the uniquely personal andcomplex mental processes that underlie the disorders.We believe that an adequate response to these

challenges will require a systems-based approach toresearch. Although traditional target-based discoveryhas shown success in identifying new drugs againstpreviously validated targets, the reductionist nature ofthis approach often leads to late-stage failure when thedrug is assessed in increasingly complex systems. Bycontrast, a systems-based approach may speed the drugdiscovery process by focusing less on drug interactionswith single targets and more on systems-level assaysrepresentative of the disease (Desbiens and Farb, 2012).A “pharmacologic connectome” is emerging based

upon which disorder-specific novel therapeutic agentsfor anxiety can be developed (Fig. 11). Functionalneuroimaging studies are revealing the brain regionsand nuclei implicated in specific anxiety disorders andare providing important clues about which pharmaco-logical interventions are the most appropriate for whichdisorders. The use of drugs that enhance extinction isgaining acceptance for the treatment of specific phobiasand the intrusive memories associated with PTSD

(Rothbaum et al., 2014). Compounds that inhibit excit-atory neurotransmission or enhance inhibitory neuro-transmission are proving to be more effective for treatingGAD and the hypervigilance associated with PTSD(Mathew et al., 2005). At the same time, we are beginningto understand that because of genetic polymorphisms,some patients may not respond to SSRIs and SNRIs(Zanardi et al., 2001). The opportunity to apply thisgrowing body of knowledge to the development of noveltherapeutics for anxiety disorders has not been greater.

Neuroactive steroids, which modulate various neuro-transmitter pathways and are a locus for the neuralcrosstalk involved in anxiety disorders, may prove to be animportant tool for advancing a systems-based approach todrug development. The unique place of neuroactivesteroids in this setting is given further support bya growing body of evidence showing that these agentsare important for both improving the survival of neuronsand increasing the number of newly formed neurons(Charalampopoulos et al., 2008).

Fig. 11. A pharmacologic connectome that depicts how specific brainregions and nuclei respond to therapeutic agents via connectivity mapsactivated in anxiety is beginning to be assembled as the human brain isdeconstructed. Episodic memories associated with anxiety-inducing eventsare stored in the parietal lobes (precuneus). The amygdala assigns an“appropriate” emotional-response tag to a stimulus, such as a smiling or anangry face. The frontal lobes in turn provide for the “top-down regulation”of emotional responses to these same stimuli. The cingulate and insulaform the major association pathways between the various regions involvedin anxiety, whereas the BNST connects the amygdala to the HPA axis,which in turn regulates how the endocrine system responds to the stimulus.In anxiety disorders that are triggered by a specific stimulus (e.g., phobiasand PTSD), the amygdaloid-mediated response to the stimuli is highlymemory-dependent. In disorders of increased apprehension and persever-ative cognition (e.g., GAD), the frontal lobe and top-down modulationof amygdaloid nucleus function would be altered.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1025

Page 25: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

The results reviewed in this article suggest thatcurrent research into brain activation and therapeutictargeting of GABA, 5-HT, and NMDA receptor subunitshas not produced a silver bullet for treating anxiety thatoptimizes efficacy and reduces risk of adverse events.Moreover, true personalized medicine in patients suffer-ing from various anxiety disorders would require acomprehensive and multifaceted approach beyond anal-ysis of neurologic activation. This could include assess-ment of patient demographics, clinical presentation,environmental factors, family history, relevant geneticpolymorphisms, and biomarkers, as well as treatmentoptions encompassing not only pharmacotherapy butalso psychotherapeutic approaches such as CBT andbehavioral activation therapy. Nevertheless, researchinto the brain regions and variations in receptorsubunits engaged in anxiety provides reason for opti-mism that the pathophysiology underlying this complexmosaic of perceptions will lead to treatments bettertailored to the individual needs of patients sufferingfrom a range of anxiety disorders based on results fromnoninvasive brain imaging and improved therapeutics.

Acknowledgements

The authors thank Dr. Cynthia Czajkowski for Fig. 7 and Dr. ConorC. Smith for artwork in Fig. 8.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Farb, Ratner.

ReferencesAhmadi H, Nasehi M, Rostami P, and Zarrindast MR (2013) Involvement of thenucleus accumbens shell dopaminergic system in prelimbic NMDA-inducedanxiolytic-like behaviors. Neuropharmacology 71:112–123.

Alger BE and Nicoll RA (1982) Pharmacological evidence for two kinds of GABAreceptor on rat hippocampal pyramidal cells studied in vitro. J Physiol 328:125–141.

Alkhatib AH, Dvorkin-Gheva A, and Szechtman H (2013) Quinpirole and 8-OH-DPATinduce compulsive checking behavior in male rats by acting on different functionalparts of an OCD neurocircuit. Behav Pharmacol 24:65–73.

Allgulander C, Bandelow B, Hollander E, Montgomery SA, Nutt DJ, Okasha A,Pollack MH, Stein DJ, and Swinson RP; World Council of Anxiety (2003) WCArecommendations for the long-term treatment of generalized anxiety disorder.CNS Spectr 8 (Suppl 1)53–61.

American Psychiatric Association (2013) Diagnostic and Statistical Manual of MentalDisorders, Fifth Edition (DSM-5), American Psychiatric Association, Washington,DC.

Amin J, Brooks-Kayal A, and Weiss DS (1997) Two tyrosine residues on the alphasubunit are crucial for benzodiazepine binding and allosteric modulation ofgamma-aminobutyric acidA receptors. Mol Pharmacol 51:833–841.

Amin J and Weiss DS (1993) GABAA receptor needs two homologous domains of thebeta-subunit for activation by GABA but not by pentobarbital. Nature 366:565–569.

Ashton H and Young AH (2003) GABA-ergic drugs: exit stage left, enter stage right.J Psychopharmacol 17:174–178.

Atack JR (2003) Anxioselective compounds acting at the GABA(A) receptor benzo-diazepine binding site. Curr Drug Targets CNS Neurol Disord 2:213–232.

Atack JR, Hallett DJ, Tye S, Wafford KA, Ryan C, Sanabria-Bohórquez SM, Eng WS,Gibson RE, Burns HD, and Dawson GR, et al. (2011a) Preclinical and clinicalpharmacology of TPA023B, a GABAA receptor α2/α3 subtype-selective partial agonist. J Psychopharmacol 25:329–344.

Atack JR, Pike A, Marshall G, Stanley J, Lincoln R, Cook SM, Lewis RT, BlackabyWP, Goodacre SC, and McKernan RM, et al. (2006a) The in vivo properties ofpagoclone in rat are most likely mediated by 59-hydroxy pagoclone. Neurophar-macology 50:677–689.

Atack JR, Wafford KA, Street LJ, Dawson GR, Tye S, Van Laere K, Bormans G,Sanabria-Bohórquez SM, De Lepeleire I, and de Hoon JN, et al. (2011b) MRK-409(MK-0343), a GABAA receptor subtype-selective partial agonist, is a non-sedatinganxiolytic in preclinical species but causes sedation in humans. J Psychopharmacol25:314–328.

Atack JR, Wafford KA, Tye SJ, Cook SM, Sohal B, Pike A, Sur C, Melillo D, Bristow L,and Bromidge F, et al. (2006b) TPA023 [7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine], an agonist selective

for alpha2- and alpha3-containing GABAA receptors, is a nonsedating anxiolytic inrodents and primates. J Pharmacol Exp Ther 316:410–422.

Agustín-Pavón C, Braesicke K, Shiba Y, Santangelo AM, Mikheenko Y, Cockroft G,Asma F, Clarke H, Man MS, and Roberts AC (2012) Lesions of ventrolateral pre-frontal or anterior orbitofrontal cortex in primates heighten negative emotion. BiolPsychiatry 72:266–272.

Aupperle RL, Hale LR, Chambers RJ, Cain SE, Barth FX, Sharp SC, Denney DR,and Savage CR (2009) An fMRI study examining effects of acute D-cycloserineduring symptom provocation in spider phobia. CNS Spectr 14:556–571.

Baldwin D, Woods R, Lawson R, and Taylor D (2011) Efficacy of drug treatments forgeneralised anxiety disorder: systematic review and meta-analysis. BMJ 342:d1199.

Ballenger JC, McDonald S, Noyes R, Rickels K, Sussman N, Woods S, Patin J,and Singer J (1991) The first double-blind, placebo-controlled trial of a partialbenzodiazepine agonist abecarnil (ZK 112-119) in generalized anxiety disorder.Psychopharmacol Bull 27:171–179.

Ballenger JC, McDonald S, Noyes R Jr, Rickels K, Woods SW, Patin J, Lydiard B,Garvey M, Cook B, and Goodard AW, et al. (1992) The first double-blind, placebo-controlled trial of a partial benzodiazepine agonist, abecarnil (ZK 112-119), ingeneralized anxiety disorder. Adv Biochem Psychopharmacol 47:431–447.

Barbaccia ML, Roscetti G, Trabucchi M, Mostallino MC, Concas A, Purdy RH,and Biggio G (1996) Time-dependent changes in rat brain neuroactive steroidconcentrations and GABAA receptor function after acute stress. Neuroendocrinol-ogy 63:166–172.

Barkus C, McHugh SB, Sprengel R, Seeburg PH, Rawlins JN, and Bannerman DM(2010) Hippocampal NMDA receptors and anxiety: at the interface between cog-nition and emotion. Eur J Pharmacol 626:49–56.

Barnard EA, Skolnick P, Olsen RW, Mohler H, Sieghart W, Biggio G, Braestrup C,Bateson AN, and Langer SZ (1998) International Union of Pharmacology. XV.Subtypes of gamma-aminobutyric acidA receptors: classification on the basis ofsubunit structure and receptor function. Pharmacol Rev 50:291–313.

Bateson A (2003) Pagoclone Indevus. Curr Opin Investig Drugs 4:91–95.Battista MA, Hierholzer R, Khouzam HR, Barlow A, and O’Toole S (2007) Pilot trialof memantine in the treatment of posttraumatic stress disorder. Psychiatry 70:167–174.

Baur R and Sigel E (2003) On high- and low-affinity agonist sites in GABAA recep-tors. J Neurochem 87:325–332.

Ben-Ari Y (2002) Excitatory actions of gaba during development: the nature of thenurture. Nat Rev Neurosci 3:728–739.

Benke D, Honer M, Michel C, and Mohler H (1996) GABAA receptor subtypes dif-ferentiated by their gamma-subunit variants: prevalence, pharmacology and sub-unit architecture. Neuropharmacology 35:1413–1423.

Bennett JA, Moioffer M, Stanton SP, Dwight M, and Keck PE Jr (1998) A risk-benefitassessment of pharmacological treatments for panic disorder. Drug Saf 18:419–430.

Berezhnoy D, Gravielle MC, and Farb DH (2007) Pharmacology of the GABA-Areceptor, in Handbook of Contemporary Neuropharmacology (Sibley D, KuharM, Hanin I, and Skolnick P eds), pp 465–568, John Wiley & Sons, Inc., Hoboken,NJ.

Berton F, Francesconi WG, Madamba SG, Zieglgänsberger W, and Siggins GR (1998)Acamprosate enhances N-methyl-D-apartate receptor-mediated neurotransmis-sion but inhibits presynaptic GABA(B) receptors in nucleus accumbens neurons.Alcohol Clin Exp Res 22:183–191.

Blanco C, Schneier FR, Schmidt A, Blanco-Jerez CR, Marshall RD, Sánchez-Lacay A,and Liebowitz MR (2003) Pharmacological treatment of social anxiety disorder:a meta-analysis. Depress Anxiety 18:29–40.

Boileau AJ and Czajkowski C (1999) Identification of transduction elements forbenzodiazepine modulation of the GABA(A) receptor: three residues are requiredfor allosteric coupling. J Neurosci 19:10213–10220.

Borden LA, Czajkowski C, Chan CY, and Farb DH (1984) Benzodiazepine receptorsynthesis and degradation by neurons in culture. Science 226:857–860.

Borden LA and Farb DH (1988) Mechanism of gamma-aminobutyric acid/benzodiazepine receptor turnover in neuronal cells: evidence for nonlysosomaldegradation. Mol Pharmacol 34:354–362.

Braestrup C and Nielsen M (1981) [3H]Propyl beta-carboline-3-carboxylate as a se-lective radioligand for the BZ1 benzodiazepine receptor subclass. J Neurochem 37:333–341.

Braestrup C and Squires RF (1977) Specific benzodiazepine receptors in rat braincharacterized by high-affinity (3H)diazepam binding. Proc Natl Acad Sci USA 74:3805–3809.

Braestrup C and Squires RF (1978) Pharmacological characterization of benzodiaz-epine receptors in the brain. Eur J Pharmacol 48:263–270.

Bremner JD, Innis RB, White T, Fujita M, Silbersweig D, Goddard AW, Staib L,Stern E, Cappiello A, and Woods S, et al. (2000) SPECT [I-123]iomazenil mea-surement of the benzodiazepine receptor in panic disorder. Biol Psychiatry 47:96–106.

Brooks-Kayal AR and Pritchett DB (1993) Developmental changes in human gamma-aminobutyric acidA receptor subunit composition. Ann Neurol 34:687–693.

Brot MD, Akwa Y, Purdy RH, Koob GF, and Britton KT (1997) The anxiolytic-likeeffects of the neurosteroid allopregnanolone: interactions with GABA(A) receptors.Eur J Pharmacol 325:1–7.

Brouillet E, Chavoix C, Bottlaender M, Khalili-Varasteh M, Hantraye P, Fournier D,Dodd RH, and Mazière M (1991) In vivo bidirectional modulatory effect of benzo-diazepine receptor ligands on GABAergic transmission evaluated by positronemission tomography in non-human primates. Brain Res 557:167–176.

Buhr A and Sigel E (1997) A point mutation in the gamma2 subunit of gamma-aminobutyric acid type A receptors results in altered benzodiazepine binding sitespecificity. Proc Natl Acad Sci USA 94:8824–8829.

Busto U, Kaplan HL, Zawertailo L, and Sellers EM (1994) Pharmacologic effects andabuse liability of bretazenil, diazepam, and alprazolam in humans. Clin PharmacolTher 55:451–463.

1026 Farb and Ratner

Page 26: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

Calogero AE, Palumbo MA, Bosboom AM, Burrello N, Ferrara E, Palumbo G, PetragliaF, and D’Agata R (1998) The neuroactive steroid allopregnanolone suppresses hy-pothalamic gonadotropin-releasing hormone release through a mechanism mediatedby the gamma-aminobutyric acidA receptor. J Endocrinol 158:121–125.

Carlson JN, Haskew R, Wacker J, Maisonneuve IM, Glick SD, and Jerussi TP (2001)Sedative and anxiolytic effects of zopiclone’s enantiomers and metabolite. Eur JPharmacol 415:181–189.

Carter RB, Wood PL, Wieland S, Hawkinson JE, Belelli D, Lambert JJ, White HS,Wolf HH, Mirsadeghi S, and Tahir SH, et al. (1997) Characterization of the anti-convulsant properties of ganaxolone (CCD 1042; 3a-hydroxy-3b-methyl-5a-pregnan-20-one), a selective, high-affinity, steroid modulator of the g-aminobutyricacid(A) receptor. J Pharmacol Exp Ther 280:1284–1295.

Casalotti SO, Stephenson FA, and Barnard EA (1986) Separate subunits for agonistand benzodiazepine binding in the gamma-aminobutyric acidA receptor oligomer. JBiol Chem 261:15013–15016.

Cassano P, Soares CN, Cohen LS, Lyster AK, and Fava M (2004) Sex- and age-related differences in major depressive disorder with comorbid anxiety treatedwith fluoxetine. Arch Women Ment Health 7:167–171.

Casu MA, Sanna A, Spada GP, Falzoi M, Mongeau R, and Pani L (2007) Effects ofacute and chronic valproate treatments on p-CREB levels in the rat amygdala andnucleus accumbens. Brain Res 1141:15–24.

Cha J, Greenberg T, Carlson JM, Dedora DJ, Hajcak G, and Mujica-Parodi LR (2014)Circuit-wide structural and functional measures predict ventromedial prefrontalcortex fear generalization: implications for generalized anxiety disorder. J Neurosci34:4043–4053.

Charalampopoulos I, Remboutsika E, Margioris AN, and Gravanis A (2008) Neurosteroidsas modulators of neurogenesis and neuronal survival. Trends Endocrinol Metab 19:300–307.

Chan CY and Farb DH (1985) Modulation of neurotransmitter action: control ofthe gamma-aminobutyric acid response through the benzodiazepine receptor.J Neurosci 5:2365–2373.

Chang Y, Wang R, Barot S, and Weiss DS (1996) Stoichiometry of a recombinantGABAA receptor. J Neurosci 16:5415–5424.

Chee SS, Menard JL, and Dringenberg HC (2014) Behavioral anxiolysis withoutreduction of hippocampal theta frequency after histamine application in the lateralseptum of rats. Hippocampus 24:615–627.

Chiu TH and Rosenberg HC (1979) GABA receptor-mediated modulation of3H-diazepam binding in rat cortex. Eur J Pharmacol 56:337–345.

Choi DW, Farb DH, and Fischbach GD (1977) Chlordiazepoxide selectively augmentsGABA action in spinal cord cell cultures. Nature 269:342–344.

Cole JC and Rodgers RJ (1993) An ethological analysis of the effects of chlordiaz-epoxide and bretazenil (Ro 16-6028) in the murine elevated plus-maze. BehavPharmacol 4:573–580.

Collins I, Moyes C, Davey WB, Rowley M, Bromidge FA, Quirk K, Atack JR,McKernan RM, Thompson SA, and Wafford K, et al. (2002) 3-Heteroaryl-2-pyridones: benzodiazepine site ligands with functional delectivity for alpha 2/alpha3-subtypes of human GABA(A) receptor-ion channels. J Med Chem 45:1887–1900.

Covey DF, Nathan D, Kalkbrenner M, Nilsson KR, Hu Y, Zorumski CF, and Evers AS(2000) Enantioselectivity of pregnanolone-induced gamma-aminobutyric acid(A)receptor modulation and anesthesia. J Pharmacol Exp Ther 293:1009–1016.

Czajkowski C and Farb DH (1986) Transmembrane topology and subcellular distri-bution of the benzodiazepine receptor. J Neurosci 6:2857–2863.

Czéh B, Michaelis T, Watanabe T, Frahm J, de Biurrun G, van Kampen M, BartolomucciA, and Fuchs E (2001) Stress-induced changes in cerebral metabolites, hippocampalvolume, and cell proliferation are prevented by antidepressant treatment withtianeptine. Proc Natl Acad Sci USA 98:12796–12801.

Davidson JR (2003) Pharmacotherapy of social phobia. Acta Psychiatr Scand Suppl417:65–71.

Davis M and Myers KM (2002) The role of glutamate and gamma-aminobutyric acidin fear extinction: clinical implications for exposure therapy. Biol Psychiatry 52:998–1007.

de Wit H, Vicini L, Haig GM, Hunt T, and Feltner D (2006) Evaluation of the abusepotential of pagoclone, a partial GABAA agonist. J Clin Psychopharmacol 26:268–273.

Desbiens S and Farb DH (2012) Current needs for new therapeutic agents and dis-covery strategies – a systems pharmacology approach, in Development of Thera-peutic Agents (Gad S ed) vol 416, pp 1–24, John Wiley & Sons, Inc., Hoboken, NJ.

Dias R, Sheppard WF, Fradley RL, Garrett EM, Stanley JL, Tye SJ, Goodacre S,Lincoln RJ, Cook SM, and Conley R, et al. (2005) Evidence for a significant role ofalpha 3-containing GABAA receptors in mediating the anxiolytic effects of benzo-diazepines. J Neurosci 25:10682–10688.

Difede J, Cukor J, Wyka K, Olden M, Hoffman H, Lee FS, and Altemus M (2014)D-cycloserine augmentation of exposure therapy for post-traumatic stress disorder:a pilot randomized clinical trial. Neuropsychopharmacology 39:1052–1058.

Doehrmann O, Ghosh SS, Polli FE, Reynolds GO, Horn F, Keshavan A, TriantafyllouC, Saygin ZM, Whitfield-Gabrieli S, and Hofmann SG, et al. (2013) Predictingtreatment response in social anxiety disorder from functional magnetic resonanceimaging. JAMA Psychiatry 70:87–97.

Drover DR (2004) Comparative pharmacokinetics and pharmacodynamics of short-acting hypnosedatives: zaleplon, zolpidem and zopiclone. Clin Pharmacokinet 43:227–238.

Duncan GE, Breese GR, Criswell HE, McCown TJ, Herbert JS, Devaud LL,and Morrow AL (1995) Distribution of [3H]zolpidem binding sites in relation tomessenger RNA encoding the alpha 1, beta 2 and gamma 2 subunits of GABAAreceptors in rat brain. Neuroscience 64:1113–1128.

Dunn JD (1987) Plasma corticosterone responses to electrical stimulation of the bednucleus of the stria terminalis. Brain Res 407:327–331.

Engels AS, Heller W, Mohanty A, Herrington JD, Banich MT, Webb AG, and MillerGA (2007) Specificity of regional brain activity in anxiety types during emotionprocessing. Psychophysiology 44:352–363.

Etkin A andWager TD (2007) Functional neuroimaging of anxiety: a meta-analysis ofemotional processing in PTSD, social anxiety disorder, and specific phobia. Am JPsychiatry 164:1476–1488.

Facklam M, Schoch P, Bonetti EP, Jenck F, Martin JR, Moreau JL, and Haefely WE(1992a) Relationship between benzodiazepine receptor occupancy and functionaleffects in vivo of four ligands of differing intrinsic efficacies. J Pharmacol Exp Ther261:1113–1121.

FacklamM, Schoch P, and Haefely WE (1992b) Relationship between benzodiazepinereceptor occupancy and potentiation of gamma-aminobutyric acid-stimulatedchloride flux in vitro of four ligands of differing intrinsic efficacies. J PharmacolExp Ther 261:1106–1112.

Falls WA, Miserendino MJ, and Davis M (1992) Extinction of fear-potentiated startle:blockade by infusion of an NMDA antagonist into the amygdala. J Neurosci 12:854–863.

Farb DH, Borden LA, Chan CY, Czajkowski CM, Gibbs TT, and Schiller GD (1984)Modulation of neuronal function through benzodiazepine receptors: biochemicaland electrophysiological studies of neurons in primary monolayer cell culture. AnnN Y Acad Sci 435:1–31.

Faria V, Appel L, Åhs F, Linnman C, Pissiota A, Frans Ö, Bani M, Bettica P, PichEM, and Jacobsson E, et al. (2012) Amygdala subregions tied to SSRI and placeboresponse in patients with social anxiety disorder. Neuropsychopharmacology 37:2222–2232.

Farkas RH, Unger EF, and Temple R (2013) Zolpidem and driving impairment—identifying persons at risk. N Engl J Med 369:689–691.

Felix-Ortiz AC, Beyeler A, Seo C, Leppla CA, Wildes CP, and Tye KM (2013) BLA tovHPC inputs modulate anxiety-related behaviors. Neuron 79:658–664.

Fernandes GA, Perks P, Cox NK, Lightman SL, Ingram CD, and Shanks N (2002)Habituation and cross-sensitization of stress-induced hypothalamic-pituitary-adrenal activity: effect of lesions in the paraventricular nucleus of the thalamusor bed nuclei of the stria terminalis. J Neuroendocrinol 14:593–602.

Ferrero P, Guidotti A, and Costa E (1984) Increase in the Bmax of gamma-aminobutyric acid-A recognition sites in brain regions of mice receiving diazepam.Proc Natl Acad Sci USA 81:2247–2251.

Fisher PM, Meltzer CC, Ziolko SK, Price JC, Moses-Kolko EL, Berga SL, and HaririAR (2006) Capacity for 5-HT1A-mediated autoregulation predicts amygdala re-activity. Nat Neurosci 9:1362–1363.

Fleck MW (2002) Molecular actions of (S)-desmethylzopiclone (SEP-174559), an an-xiolytic metabolite of zopiclone. J Pharmacol Exp Ther 302:612–618.

Fluoxetine (2013) Prescribing information. Eli Lilly and Company, Indianapolis, IN.Follesa P, Mancuso L, Biggio F, Cagetti E, Franco M, Trapani G, and Biggio G (2002)Changes in GABA(A) receptor gene expression induced by withdrawal of, but notby long-term exposure to, zaleplon or zolpidem. Neuropharmacology 42:191–198.

Gallager DW and Tallman JF (1983) Consequences of benzodiazepine receptor oc-cupancy. Neuropharmacology 22:1493–1498.

Gasior M, Carter RB, and Witkin JM (1999) Neuroactive steroids: potential therapeuticuse in neurological and psychiatric disorders. Trends Pharmacol Sci 20:107–112.

Gavioli EC, Canteras NS, and De Lima TC (1999) Anxiogenic-like effect induced bysubstance P injected into the lateral septal nucleus. Neuroreport 10:3399–3403.

Gelernter J, Page GP, Stein MB, and Woods SW (2004) Genome-wide linkage scan forloci predisposing to social phobia: evidence for a chromosome 16 risk locus. Am JPsychiatry 161:59–66.

Geuze E, van Berckel BN, Lammertsma AA, Boellaard R, de Kloet CS, Vermetten E,and Westenberg HG (2008) Reduced GABAA benzodiazepine receptor binding inveterans with post-traumatic stress disorder. Mol Psychiatry 13:74–83, 3.

Ghiani CA, Serra M, Motzo C, Giusti P, Cuccheddu T, Porceddu ML, and Biggio G(1994) Chronic administration of an anticonvulsant dose of imidazenil fails to in-duce tolerance of GABAA receptor function in mice. Eur J Pharmacol 254:299–302.

Giménez M, Ortiz H, Soriano-Mas C, López-Solà M, Farré M, Deus J, Martín-SantosR, Fernandes S, Fina P, and Bani M, et al. (2014) Functional effects of chronicparoxetine versus placebo on the fear, stress and anxiety brain circuit in SocialAnxiety Disorder: initial validation of an imaging protocol for drug discovery. EurNeuropsychopharmacol 24:105–116.

Gïrdler SS, Straneva PA, Light KC, Pedersen CA, and Morrow AL (2001)Allopregnanolone levels and reactivity to mental stress in premenstrual dysphoricdisorder. Biol Psychiatry 49:788–797.

Goddard AW, Mason GF, Almai A, Rothman DL, Behar KL, Petroff OA, Charney DS,and Krystal JH (2001) Reductions in occipital cortex GABA levels in panic disorderdetected with 1h-magnetic resonance spectroscopy. Arch Gen Psychiatry 58:556–561.

Goddard AW, Mason GF, Appel M, Rothman DL, Gueorguieva R, Behar KL,and Krystal JH (2004) Impaired GABA neuronal response to acute benzodiazepineadministration in panic disorder. Am J Psychiatry 161:2186–2193.

Goutman JD, Waxemberg MD, Doñate-Oliver F, Pomata PE, and Calvo DJ (2003)Flavonoid modulation of ionic currents mediated by GABA(A) and GABA(C)receptors. Eur J Pharmacol 461:79–87.

Grabenstatter HL, Russek SJ, and Brooks-Kayal AR (2012) Molecular pathwayscontrolling inhibitory receptor expression. Epilepsia 53 (Suppl 9):71–78.

Graeff FG and Zangrossi H Jr (2010) The dual role of serotonin in defense and themode of action of antidepressants on generalized anxiety and panic disorders. CentNerv Syst Agents Med Chem 10:207–217.

Graham D, Faure C, Besnard F, and Langer SZ (1996) Pharmacological profile ofbenzodiazepine site ligands with recombinant GABAA receptor subtypes. EurNeuropsychopharmacol 6:119–125.

Gram L (1988) Experimental studies and controlled clinical testing of valproate andvigabatrin. Acta Neurol Scand 78:241–270.

Gray JA and Roth BL (2007) Serotonin systems, in Handbook of ContemporaryNeuropharmacology (Sibley D, Kuhar M, Hanin I, and Skolnick P eds), pp 257–298,John Wiley & Sons, Inc., Hoboken, NJ.

Greenberg T, Carlson JM, Cha J, Hajcak G, and Mujica-Parodi LR (2013) Ventro-medial prefrontal cortex reactivity is altered in generalized anxiety disorder duringfear generalization. Depress Anxiety 30:242–250.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1027

Page 27: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

Griebel G, Perrault G, Simiand J, Cohen C, Granger P, Decobert M, Françon D,Avenet P, Depoortere H, and Tan S, et al. (2001) SL651498: an anxioselectivecompound with functional selectivity for alpha2- and alpha3-containing gamma-aminobutyric acid(A) (GABA(A)) receptors. J Pharmacol Exp Ther 298:753–768.

Griffin LD and Mellon SH (1999) Selective serotonin reuptake inhibitors directlyalter activity of neurosteroidogenic enzymes. Proc Natl Acad Sci USA 96:13512–13517.

Gulledge AT and Stuart GJ (2003) Excitatory actions of GABA in the cortex. Neuron37:299–309.

Günther U, Benson J, Benke D, Fritschy JM, Reyes G, Knoflach F, Crestani F, AguzziA, Arigoni M, and Lang Y, et al. (1995) Benzodiazepine-insensitive mice generatedby targeted disruption of the gamma 2 subunit gene of gamma-aminobutyric acidtype A receptors. Proc Natl Acad Sci USA 92:7749–7753.

Guo JD, Hammack SE, Hazra R, Levita L, and Rainnie DG (2009) Bi-directionalmodulation of bed nucleus of stria terminalis neurons by 5-HT: molecular ex-pression and functional properties of excitatory 5-HT receptor subtypes. Neuro-science 164:1776–1793.

Häberlein H, Tschiersch KP, and Schäfer HL (1994) Flavonoids from Leptospermumscoparium with affinity to the benzodiazepine receptor characterized by structureactivity relationships and in vivo studies of a plant extract. Pharmazie 49:912–922.

Hadingham KL, Garrett EM, Wafford KA, Bain C, Heavens RP, Sirinathsinghji DJ,and Whiting PJ (1996) Cloning of cDNAs encoding the human g-aminobutyric acidtype A receptor a 6 subunit and characterization of the pharmacology of a 6-containingreceptors. Mol Pharmacol 49:253–259.

Hadingham KL, Wingrove P, Le Bourdelles B, Palmer KJ, Ragan CI, and Whiting PJ(1993) Cloning of cDNA sequences encoding human alpha 2 and alpha 3 gamma-aminobutyric acidA receptor subunits and characterization of the benzodiazepinepharmacology of recombinant alpha 1-, alpha 2-, alpha 3-, and alpha 5-containinghuman gamma-aminobutyric acidA receptors. Mol Pharmacol 43:970–975.

Haefely W, Facklam M, Schoch P, Martin JR, Bonetti EP, Moreau JL, Jenck F,and Richards JG (1992) Partial agonists of benzodiazepine receptors for treatmentof epilepsy, sleep and anxiety disorders, in GABAergic Synaptic Transmission(Biggio G, Concas A, and Costa E eds) pp 379–394, Raven Press, NY.

Ham BJ, Sung Y, Kim N, Kim SJ, Kim JE, Kim DJ, Lee JY, Kim JH, Yoon SJ,and Lyoo IK (2007) Decreased GABA levels in anterior cingulate and basal gangliain medicated subjects with panic disorder: a proton magnetic resonance spectros-copy (1H-MRS) study. Prog Neuropsychopharmacol Biol Psychiatry 31:403–411.

Hanson SM and Czajkowski C (2008) Structural mechanisms underlying benzodi-azepine modulation of the GABA(A) receptor. J Neurosci 28:3490–3499.

Hariri AR, Drabant EM, Munoz KE, Kolachana BS, Mattay VS, Egan MF,and Weinberger DR (2005) A susceptibility gene for affective disorders and theresponse of the human amygdala. Arch Gen Psychiatry 62:146–152.

Harmer CJ, Mackay CE, Reid CB, Cowen PJ, and Goodwin GM (2006) Antidepres-sant drug treatment modifies the neural processing of nonconscious threat cues.Biol Psychiatry 59:816–820.

Harrison NL and Simmonds MA (1984) Modulation of the GABA receptor complex bya steroid anaesthetic. Brain Res 323:287–292.

Hasler G, Nugent AC, Carlson PJ, Carson RE, Geraci M, and Drevets WC (2008)Altered cerebral gamma-aminobutyric acid type A-benzodiazepine receptor bindingin panic disorder determined by [11C]flumazenil positron emission tomography.Arch Gen Psychiatry 65:1166–1175.

Hayward C and Sanborn K (2002) Puberty and the emergence of gender differencesin psychopathology. J Adolesc Health 30:49–58.

Hazra R, Guo JD, Dabrowska J, and Rainnie DG (2012) Differential distribution ofserotonin receptor subtypes in BNST(ALG) neurons: modulation by unpredictableshock stress. Neuroscience 225:9–21.

Heils A, Teufel A, Petri S, Stöber G, Riederer P, Bengel D, and Lesch KP (1996)Allelic variation of human serotonin transporter gene expression. J Neurochem 66:2621–2624.

Heinrichs SC, Leite-Morris KA, Rasmusson AM, and Kaplan GB (2013) Repeatedvalproate treatment facilitates fear extinction under specific stimulus conditions.Neurosci Lett 552:108–113.

Henke PG (1984) The bed nucleus of the stria terminalis and immobilization-stress:unit activity, escape behaviour, and gastric pathology in rats. Behav Brain Res 11:35–45.

Hensler JG (2006) Serotonergic modulation of the limbic system. Neurosci BiobehavRev 30:203–214.

Herb A, Wisden W, Lüddens H, Puia G, Vicini S, and Seeburg PH (1992) The thirdg subunit of the g-aminobutyric acid type A receptor family. Proc Natl Acad SciUSA 89:1433–1437.

Heresco-Levy U, Kremer I, Javitt DC, Goichman R, Reshef A, Blanaru M, and CohenT (2002) Pilot-controlled trial of D-cycloserine for the treatment of post-traumaticstress disorder. Int J Neuropsychopharmacol 5:301–307.

Herrick-Davis K, Grinde E, Harrigan TJ, and Mazurkiewicz JE (2005) Inhibition ofserotonin 5-hydroxytryptamine2c receptor function through heterodimerization:receptor dimers bind two molecules of ligand and one G-protein. J Biol Chem 280:40144–40151.

Hertzman M, Patt IS, and Spielman LA (2009) Open-label trial of acamprosate asa treatment for anxiety. Prim Care Companion J Clin Psychiatry 11:267.

Hettema JM, Prescott CA, Myers JM, Neale MC, and Kendler KS (2005) Thestructure of genetic and environmental risk factors for anxiety disorders in menand women. Arch Gen Psychiatry 62:182–189.

Heydari B and Le Mellédo JM (2002) Low pregnenolone sulphate plasma concen-trations in patients with generalized social phobia. Psychol Med 32:929–933.

Hoehn-Saric R, Schlund MW, andWong SH (2004) Effects of citalopram on worry andbrain activation in patients with generalized anxiety disorder. Psychiatry Res 131:11–21.

Hofmann SG, Meuret AE, Smits JA, Simon NM, Pollack MH, Eisenmenger K, ShiekhM, and Otto MW (2006) Augmentation of exposure therapy with D-cycloserine forsocial anxiety disorder. Arch Gen Psychiatry 63:298–304.

Hosie AM, Wilkins ME, da Silva HM, and Smart TG (2006) Endogenous neuro-steroids regulate GABAA receptors through two discrete transmembrane sites.Nature 444:486–489.

Hoyer D, Hannon JP, and Martin GR (2002) Molecular, pharmacological and func-tional diversity of 5-HT receptors. Pharmacol Biochem Behav 71:533–554.

Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, Xu K, Arnold PD,Richter MA, and Kennedy JL, et al. (2006) Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet78:815–826.

Huen MS, Hui KM, Leung JW, Sigel E, Baur R, Wong JT, and Xue H (2003) Natu-rally occurring 29-hydroxyl-substituted flavonoids as high-affinity benzodiazepinesite ligands. Biochem Pharmacol 66:2397–2407.

Hui KM, Huen MS, Wang HY, Zheng H, Sigel E, Baur R, Ren H, Li ZW, Wong JT,and Xue H (2002) Anxiolytic effect of wogonin, a benzodiazepine receptor ligandisolated from Scutellaria baicalensis Georgi. Biochem Pharmacol 64:1415–1424.

Hui KM, Wang XH, and Xue H (2000) Interaction of flavones from the roots ofScutellaria baicalensis with the benzodiazepine site. Planta Med 66:91–93.

Isenberg N, Silbersweig D, Engelien A, Emmerich S, Malavade K, Beattie B, LeonAC, and Stern E (1999) Linguistic threat activates the human amygdala. Proc NatlAcad Sci USA 96:10456–10459.

Jain NS, Hirani K, and Chopde CT (2005) Reversal of caffeine-induced anxiety byneurosteroid 3-alpha-hydroxy-5-alpha-pregnane-20-one in rats. Neuropharmacology48:627–638.

Jardim MC, Aguiar DC, Moreira FA, and Guimarães FS (2005) Role of glutamateionotropic and benzodiazepine receptors in the ventromedial hypothalamic nucleuson anxiety. Pharmacol Biochem Behav 82:182–189.

Jeon GS, Park SH, Lee KJ, Lee MS, Chun BG, and Shin KH (2006) Valproate pre-vents MK801-induced changes in brain-derived neurotrophic factor mRNA in therat brain. Eur J Pharmacol 545:142–146.

Jornada LK, Moretti M, Valvassori SS, Ferreira CL, Padilha PT, Arent CO, Fries GR,Kapczinski F, and Quevedo J (2010) Effects of mood stabilizers on hippocampusand amygdala BDNF levels in an animal model of mania induced by ouabain.J Psychiatr Res 44:506–510.

Kang-Park MH, Wilson WA, and Moore SD (2004) Differential actions of diazepamand zolpidem in basolateral and central amygdala nuclei. Neuropharmacology 46:1–9.

Kaschka W, Feistel H, and Ebert D (1995) Reduced benzodiazepine receptor bindingin panic disorders measured by iomazenil SPECT. J Psychiatr Res 29:427–434.

Katsnelson A (2013) Momentum grows to make ‘personalized’ medicine more‘precise’. Nat Med 19:249.

Kaufmann WA, Humpel C, Alheid GF, and Marksteiner J (2003) Compartmentationof alpha 1 and alpha 2 GABA(A) receptor subunits within rat extended amygdala:implications for benzodiazepine action. Brain Res 964:91–99.

Kavvadias D, Sand P, Youdim KA, Qaiser MZ, Rice-Evans C, Baur R, Sigel E, RauschWD, Riederer P, and Schreier P (2004) The flavone hispidulin, a benzodiazepinereceptor ligand with positive allosteric properties, traverses the blood-brain barrierand exhibits anticonvulsive effects. Br J Pharmacol 142:811–820.

Kent JM and Rauch SL (2003) Neurocircuitry of anxiety disorders. Curr PsychiatryRep 5:266–273.

Kerner B, Rao AR, Christensen B, Dandekar S, Yourshaw M, and Nelson SF (2013)Rare genomic variants link bipolar disorder with anxiety disorders to CREB-regulated intracellular signaling pathways. Front Psychiatry 4:154.

Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, and Walters EE (2005a)Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in theNational Comorbidity Survey Replication. Arch Gen Psychiatry 62:593–602.

Kessler RC, Chiu WT, Demler O, Merikangas KR, and Walters EE (2005b) Preva-lence, severity, and comorbidity of 12-month DSM-IV disorders in the NationalComorbidity Survey Replication. Arch Gen Psychiatry 62:617–627.

Kheirbek MA, Drew LJ, Burghardt NS, Costantini DO, Tannenholz L, Ahmari SE,Zeng H, Fenton AA, and Hen R (2013) Differential control of learning and anxietyalong the dorsoventral axis of the dentate gyrus. Neuron 77:955–968.

Khisti RT and Chopde CT (2000) Serotonergic agents modulate antidepressant-likeeffect of the neurosteroid 3alpha-hydroxy-5alpha-pregnan-20-one in mice. BrainRes 865:291–300.

Kim SW, Fowler JS, Skolnick P, Muench L, Kang Y, Shea C, Logan J, Kim D, CarterP, and King P, et al. (2014) Therapeutic doses of buspirone block D3 receptors inthe living primate brain. Int J Neuropsychopharmacol 17:1257–1267.

Kim SY, Adhikari A, Lee SY, Marshel JH, Kim CK, Mallory CS, Lo M, Pak S, MattisJ, and Lim BK, et al. (2013) Diverging neural pathways assemble a behaviouralstate from separable features in anxiety. Nature 496:219–223.

Klumpers F, Heitland I, Oosting RS, Kenemans JL, and Baas JM (2012) Geneticvariation in serotonin transporter function affects human fear expression indexedby fear-potentiated startle. Biol Psychol 89:277–282.

Knoflach F, Benke D, Wang Y, Scheurer L, Lüddens H, Hamilton BJ, Carter DB,Mohler H, and Benson JA (1996) Pharmacological modulation of the diazepam-insensitive recombinant gamma-aminobutyric acidA receptors alpha 4 beta 2gamma 2 and alpha 6 beta 2 gamma 2. Mol Pharmacol 50:1253–1261.

Kobayashi K, Ikeda Y, Sakai A, Yamasaki N, Haneda E, Miyakawa T, and Suzuki H(2010) Reversal of hippocampal neuronal maturation by serotonergic anti-depressants. Proc Natl Acad Sci USA 107:8434–8439.

Kobiella A, Reimold M, Ulshöfer DE, Ikonomidou VN, Vollmert C, Vollstädt-Klein S,Rietschel M, Reischl G, Heinz A, and Smolka MN (2011) How the serotonin trans-porter 5-HTTLPR polymorphism influences amygdala function: the roles of in vivoserotonin transporter expression and amygdala structure. Transl Psychiatr 1:e37.

Koltunowska D, Gibula-Bruzda E, and Kotlinska JH (2013) The influence ofionotropic and metabotropic glutamate receptor ligands on anxiety-like effect ofamphetamine withdrawal in rats. Prog Neuropsychopharmacol Biol Psychiatry 45:242–249.

Kostakis E, Smith C, Jang MK, Martin SC, Richards KG, Russek SJ, Gibbs TT,and Farb DH (2013) The neuroactive steroid pregnenolone sulfate stimulates

1028 Farb and Ratner

Page 28: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

trafficking of functional N-methyl D-aspartate receptors to the cell surface viaa noncanonical, G protein, and Ca21-dependent mechanism. Mol Pharmacol 84:261–274.

Kotlinska J and Bochenski M (2008) The influence of various glutamate receptorsantagonists on anxiety-like effect of ethanol withdrawal in a plus-maze test in rats.Eur J Pharmacol 598:57–63.

Kuriyama K, Honma M, Yoshiike T, and Kim Y (2013) Valproic acid but notD-cycloserine facilitates sleep-dependent offline learning of extinction and habit-uation of conditioned fear in humans. Neuropharmacology 64:424–431.

Kushner SA, Dewey SL, and Kornetsky C (1997) Gamma-vinyl GABA attenuatescocaine-induced lowering of brain stimulation reward thresholds. Psychopharma-cology (Berl) 133:383–388.

Lambert JJ, Belelli D, Harney SC, Peters JA, and Frenguelli BG (2001) Modulationof native and recombinant GABA(A) receptors by endogenous and synthetic neu-roactive steroids. Brain Res Brain Res Rev 37:68–80.

Lambert JJ, Belelli D, Peden DR, Vardy AW, and Peters JA (2003) Neurosteroidmodulation of GABAA receptors. Prog Neurobiol 71:67–80.

Lavoie AM and Twyman RE (1996) Direct evidence for diazepam modulation ofGABAA receptor microscopic affinity. Neuropharmacology 35:1383–1392.

Le Mellédo JM and Baker G (2004) Role of progesterone and other neuroactive ste-roids in anxiety disorders. Expert Rev Neurother 4:851–860.

Legerstee JS, Verhulst FC, Robbers SC, Ormel J, Oldehinkel AJ, and van Oort FV(2013) Gender-specific developmental trajectories of anxiety during adolescence:determinants and outcomes. The TRAILS study. J Can Acad Child AdolescPsychiatry 22:26–34.

Leikanger E, Ingul JM, and Larsson B (2012) Sex and age-related anxietyin a community sample of Norwegian adolescents. Scand J Psychol 53:150–157.

Lépine JP (2002) The epidemiology of anxiety disorders: prevalence and societalcosts. J Clin Psychiatry 63 (Suppl 14):4–8.

Lesch KP, Bengel D, Heils A, Sabol SZ, Greenberg BD, Petri S, Benjamin J, MüllerCR, Hamer DH, and Murphy DL (1996) Association of anxiety-related traits witha polymorphism in the serotonin transporter gene regulatory region. Science 274:1527–1531.

Levinson DF and Devinsky O (1999) Psychiatric adverse events during vigabatrintherapy. Neurology 53:1503–1511.

Licata SC, Platt DM, Cook JM, Sarma PV, Griebel G, and Rowlett JK (2005)Contribution of GABAA receptor subtypes to the anxiolytic-like, motor, anddiscriminative stimulus effects of benzodiazepines: studies with the functionallyselective ligand SL651498 [6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyridol[3,4-b]indol-1-one]. J Pharmacol Exp Ther313:1118–1125.

Liebowitz MR, Mangano RM, Bradwejn J, and Asnis G; SAD Study Group (2005) Arandomized controlled trial of venlafaxine extended release in generalized socialanxiety disorder. J Clin Psychiatry 66:238–247.

Lingford-Hughes A, Hume SP, Feeney A, Hirani E, Osman S, Cunningham VJ, PikeVW, Brooks DJ, and Nutt DJ (2002) Imaging the GABA-benzodiazepine receptorsubtype containing the alpha5-subunit in vivo with [11C]Ro15 4513 positronemission tomography. J Cereb Blood Flow Metab 22:878–889.

Lippa A, Czobor P, Stark J, Beer B, Kostakis E, Gravielle M, Bandyopadhyay S,Russek SJ, Gibbs TT, and Farb DH, et al. (2005) Selective anxiolysis producedby ocinaplon, a GABA(A) receptor modulator. Proc Natl Acad Sci USA 102:7380–7385.

Lippa AS, Coupet J, Greenblatt EN, Klepner CA, and Beer B (1979) A synthetic non-benzodiazepine ligand for benzodiazepine receptors: a probe for investigatingneuronal substrates of anxiety. Pharmacol Biochem Behav 11:99–106.

Liu X, Ramirez S, Pang PT, Puryear CB, Govindarajan A, Deisseroth K,and Tonegawa S (2012) Optogenetic stimulation of a hippocampal engram acti-vates fear memory recall. Nature 484:381–385.

Lo MM, Strittmatter SM, and Snyder SH (1982) Physical separation and charac-terization of two types of benzodiazepine receptors. Proc Natl Acad Sci USA 79:680–684.

Löw K, Crestani F, Keist R, Benke D, Brünig I, Benson JA, Fritschy JM, Rülicke T,Bluethmann H, and Möhler H, et al. (2000) Molecular and neuronal substrate forthe selective attenuation of anxiety. Science 290:131–134.

Lund IV, Hu Y, Raol YH, Benham RS, Faris R, Russek SJ, and Brooks-Kayal AR(2008) BDNF selectively regulates GABAA receptor transcription by activation ofthe JAK/STAT pathway. Sci Signal 1:ra9.

Macdonald RL and Barker JL (1978) Different actions of anticonvulsant and anes-thetic barbiturates revealed by use of cultured mammalian neurons. Science 200:775–777.

Macdonald RL, Rogers CJ, and Twyman RE (1989) Kinetic properties of the GABAAreceptor main conductance state of mouse spinal cord neurones in culture.J Physiol 410:479–499.

Majewska MD, Mienville JM, and Vicini S (1988) Neurosteroid pregnenolone sulfateantagonizes electrophysiological responses to GABA in neurons. Neurosci Lett 90:279–284.

Malizia AL, Cunningham VJ, Bell CJ, Liddle PF, Jones T, and Nutt DJ (1998)Decreased brain GABA(A)-benzodiazepine receptor binding in panic disorder:preliminary results from a quantitative PET study. Arch Gen Psychiatry 55:715–720.

Malminiemi O and Korpi ER (1989) Diazepam-insensitive [3H]Ro 15-4513 binding inintact cultured cerebellar granule cells. Eur J Pharmacol 169:53–60.

Mao SC, Lin HC, and Gean PW (2008) Augmentation of fear extinction byD-cycloserine is blocked by proteasome inhibitors. Neuropsychopharmacology 33:3085–3095.

Masneuf S, Lowery-Gionta E, Colacicco G, Pleil KE, Li C, Crowley N, Flynn S,Holmes A, and Kash T (2014) Glutamatergic mechanisms associated with stress-induced amygdala excitability and anxiety-related behavior.Neuropsychopharmacology85:190–197.

Mathers DA (1985) Spontaneous and GABA-induced single channel currents in cul-tured murine spinal cord neurons. Can J Physiol Pharmacol 63:1228–1233.

Mathew SJ, Amiel JM, Coplan JD, Fitterling HA, Sackeim HA, and Gorman JM(2005) Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry162:2379–2381.

Mato S, Vidal R, Castro E, Díaz A, Pazos A, and Valdizán EM (2010) Long-termfluoxetine treatment modulates cannabinoid type 1 receptor-mediated inhibition ofadenylyl cyclase in the rat prefrontal cortex through 5-hydroxytryptamine 1Areceptor-dependent mechanisms. Mol Pharmacol 77:424–434.

Matsumoto K, Uzunova V, Pinna G, Taki K, Uzunov DP, Watanabe H, Mienville JM,Guidotti A, and Costa E (1999) Permissive role of brain allopregnanolone contentin the regulation of pentobarbital-induced righting reflex loss. Neuropharmacology38:955–963.

McGaugh JL (2000) Memory—a century of consolidation. Science 287:248–251.McGregor IS, Hargreaves GA, Apfelbach R, and Hunt GE (2004) Neural correlates ofcat odor-induced anxiety in rats: region-specific effects of the benzodiazepinemidazolam. J Neurosci 24:4134–4144.

McKernan RM, Rosahl TW, Reynolds DS, Sur C, Wafford KA, Atack JR, Farrar S,Myers J, Cook G, and Ferris P, et al. (2000) Sedative but not anxiolytic propertiesof benzodiazepines are mediated by the GABA(A) receptor alpha1 subtype. NatNeurosci 3:587–592.

McKernan RM and Whiting PJ (1996) Which GABAA-receptor subtypes really occurin the brain? Trends Neurosci 19:139–143.

McNaughton N, Kocsis B, and Hajós M (2007) Elicited hippocampal theta rhythm:a screen for anxiolytic and procognitive drugs through changes in hippocampalfunction? Behav Pharmacol 18:329–346.

McNaughton N, Swart C, Neo P, Bates V, and Glue P (2013) Anti-anxiety drugsreduce conflict-specific “theta”—a possible human anxiety-specific biomarker.J Affect Disord 148:104–111.

Medina JH, Paladini AC, Wolfman C, Levi de Stein M, Calvo D, Diaz LE, and Peña C(1990) Chrysin (5,7-di-OH-flavone), a naturally-occurring ligand for benzodiaze-pine receptors, with anticonvulsant properties. Biochem Pharmacol 40:2227–2231.

Medina JH, Peña C, Levi de Stein M, Wolfman C, and Paladini AC (1989)Benzodiazepine-like molecules, as well as other ligands for the brain benzodiaze-pine receptors, are relatively common constituents of plants. Biochem Biophys ResCommun 165:547–553.

Medina JH, Viola H, Wolfman C, Marder M, Wasowski C, Calvo D, and Paladini AC(1997) Overview—flavonoids: a new family of benzodiazepine receptor ligands.Neurochem Res 22:419–425.

Mehta AK and Shank RP (1995) Interaction of abecarnil, bretazenil, and RO 19-8022with diazepam-sensitive and -insensitive benzodiazepine sites in the rat cerebel-lum and cerebral cortex. Life Sci 57:2215–2222.

Merikangas KR, He JP, Brody D, Fisher PW, Bourdon K, and Koretz DS (2010)Prevalence and treatment of mental disorders among US children in the 2001-2004NHANES. Pediatrics 125:75–81.

Merikangas KR, Nakamura EF, and Kessler RC (2009) Epidemiology of mentaldisorders in children and adolescents. Dialogues Clin Neurosci 11:7–20.

Michael-Titus AT, Albert M, Michael GJ, Michaelis T, Watanabe T, Frahm J,Pudovkina O, van der Hart MG, Hesselink MB, and Fuchs E, et al. (2008)SONU20176289, a compound combining partial dopamine D(2) receptor agonismwith specific serotonin reuptake inhibitor activity, affects neuroplasticity in ananimal model for depression. Eur J Pharmacol 598:43–50.

Mihalek RM, Banerjee PK, Korpi ER, Quinlan JJ, Firestone LL, Mi ZP, Lagenaur C,Tretter V, Sieghart W, and Anagnostaras SG, et al. (1999) Attenuated sensitivity toneuroactive steroids in gamma-aminobutyrate type A receptor delta subunitknockout mice. Proc Natl Acad Sci USA 96:12905–12910.

Mihalek RM, Bowers BJ, Wehner JM, Kralic JE, VanDoren MJ, Morrow AL,and Homanics GE (2001) GABA(A)-receptor delta subunit knockout mice have mul-tiple defects in behavioral responses to ethanol. Alcohol Clin Exp Res 25:1708–1718.

Minkeviciene R, Banerjee P, and Tanila H (2008) Cognition-enhancing and anxiolyticeffects of memantine. Neuropharmacology 54:1079–1085.

Mirza NR, Nielsen EØ, and Troelsen KB (2007) Serotonin transporter density andanxiolytic-like effects of antidepressants in mice. Prog Neuropsychopharmacol BiolPsychiatry 31:858–866.

Miserendino MJ, Sananes CB, Melia KR, and Davis M (1990) Blocking of acquisitionbut not expression of conditioned fear-potentiated startle by NMDA antagonists inthe amygdala. Nature 345:716–718.

Moguilevsky JA and Wuttke W (2001) Changes in the control of gonadotrophin secre-tion by neurotransmitters during sexual development in rats. Exp Clin EndocrinolDiabetes 109:188–195.

Möhler H and Okada T (1977) Benzodiazepine receptor: demonstration in the centralnervous system. Science 198:849–851.

Morris HV, Dawson GR, Reynolds DS, Atack JR, and Stephens DN (2006) Bothalpha2 and alpha3 GABAA receptor subtypes mediate the anxiolytic properties ofbenzodiazepine site ligands in the conditioned emotional response paradigm. Eur JNeurosci 23:2495–2504.

Nakamura M, Ueno S, Sano A, and Tanabe H (2000) The human serotonin trans-porter gene linked polymorphism (5-HTTLPR) shows ten novel allelic variants.MolPsychiatry 5:32–38.

Nemeroff CB (2003) Anxiolytics: past, present, and future agents. J Clin Psychiatry64 (Suppl 3):3–6.

Nichols CD, Ronesi J, Pratt W, and Sanders-Bush E (2002) Hallucinogens andDrosophila: linking serotonin receptor activation to behavior. Neuroscience 115:979–984.

Nichols DE and Nichols CD (2008) Serotonin receptors. Chem Rev 108:1614–1641.Nielsen M, Frøkjaer S, and Braestrup C (1988) High affinity of the naturally-occurring biflavonoid, amentoflavon, to brain benzodiazepine receptors in vitro.Biochem Pharmacol 37:3285–3287.

Nirenberg MJ (2013) Dopamine agonist withdrawal syndrome: implications for pa-tient care. Drugs Aging 30:587–592.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1029

Page 29: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

Noyes R Jr, DuPont RL Jr, Pecknold JC, Rifkin A, Rubin RT, Swinson RP, BallengerJC, and Burrows GD (1988) Alprazolam in panic disorder and agoraphobia: resultsfrom a multicenter trial. II. Patient acceptance, side effects, and safety. Arch GenPsychiatry 45:423–428.

Olivier JD, Vinkers CH, and Olivier B (2013) The role of the serotonergic and GABAsystem in translational approaches in drug discovery for anxiety disorders. FrontPharmacol 4:74.

Pałucha-Poniewiera A and Pilc A (2012) Involvement of mGlu5 and NMDA receptorsin the antidepressant-like effect of acamprosate in the tail suspension test. ProgNeuropsychopharmacol Biol Psychiatry 39:102–106.

Pape HC and Stork O (2003) Genes and mechanisms in the amygdala involved in theformation of fear memory. Ann N Y Acad Sci 985:92–105.

Park-Chung M, Wu FS, and Farb DH (1994) 3 alpha-Hydroxy-5 beta-pregnan-20-onesulfate: a negative modulator of the NMDA-induced current in cultured neurons.Mol Pharmacol 46:146–150.

Patchev VK, Shoaib M, Holsboer F, and Almeida OF (1994) The neurosteroid tet-rahydroprogesterone counteracts corticotropin-releasing hormone-induced anxietyand alters the release and gene expression of corticotropin-releasing hormone inthe rat hypothalamus. Neuroscience 62:265–271.

Pelletier JG and Paré D (2004) Role of amygdala oscillations in the consolidation ofemotional memories. Biol Psychiatry 55:559–562.

Pesold C and Treit D (1996) The neuroanatomical specificity of the anxiolytic effectsof intra-septal infusions of midazolam. Brain Res 710:161–168.

Phan KL, Coccaro EF, Angstadt M, Kreger KJ, Mayberg HS, Liberzon I, and SteinMB (2013) Corticolimbic brain reactivity to social signals of threat before and aftersertraline treatment in generalized social phobia. Biol Psychiatry 73:329–336.

Phan KL, Wager TD, Taylor SF, and Liberzon I (2004) Functional neuroimagingstudies of human emotions. CNS Spectr 9:258–266.

Phillips ML, Drevets WC, Rauch SL, and Lane R (2003) Neurobiology of emotionperception I: The neural basis of normal emotion perception. Biol Psychiatry 54:504–514.

Pigott TA (2003) Anxiety disorders in women. Psychiatr Clin North Am 26:621–672,vi–vii.

Pinna G (2010) In a mouse model relevant for post-traumatic stress disorder, selec-tive brain steroidogenic stimulants (SBSS) improve behavioral deficits by nor-malizing allopregnanolone biosynthesis. Behav Pharmacol 21:438–450.

Pittenger C, Coric V, Banasr M, Bloch M, Krystal JH, and Sanacora G (2008) Riluzolein the treatment of mood and anxiety disorders. CNS Drugs 22:761–786.

Plescia F, Marino RA, Cannizzaro E, Brancato A, and Cannizzaro C (2013) The role ofpregnenolone sulphate in spatial orientation-acquisition and retention: an in-terplay between cognitive potentiation and mood regulation. Behav Processes 99:130–137.

Pollack MH, Jensen JE, Simon NM, Kaufman RE, and Renshaw PF (2008) High-fieldMRS study of GABA, glutamate and glutamine in social anxiety disorder: responseto treatment with levetiracetam. Prog Neuropsychopharmacol Biol Psychiatry 32:739–743.

Polter AM and Li X (2011) Glycogen Synthase Kinase-3 is an Intermediate Modulatorof Serotonin Neurotransmission. Front Mol Neurosci 4:31.

Porcelli S, Fabbri C, and Serretti A (2012) Meta-analysis of serotonin transportergene promoter polymorphism (5-HTTLPR) association with antidepressant effi-cacy. Eur Neuropsychopharmacol 22:239–258.

Pritchett DB, Lüddens H, and Seeburg PH (1989) Type I and type II GABAA-benzodiazepine receptors produced in transfected cells. Science 245:1389–1392.

Pritchett DB and Seeburg PH (1990) Gamma-aminobutyric acidA receptor alpha 5-subunit creates novel type II benzodiazepine receptor pharmacology. J Neurochem54:1802–1804.

Pritchett DB and Seeburg PH (1991) gamma-Aminobutyric acid type A receptor pointmutation increases the affinity of compounds for the benzodiazepine site. Proc NatlAcad Sci USA 88:1421–1425.

Purdy RH, Morrow AL, Moore PH Jr, and Paul SM (1991) Stress-induced elevationsof gamma-aminobutyric acid type A receptor-active steroids in the rat brain. ProcNatl Acad Sci USA 88:4553–4557.

Rabow LE, Russek SJ, and Farb DH (1995) From ion currents to genomic analysis:recent advances in GABAA receptor research. Synapse 21:189–274.

Rando RR, Bangerter FW, and Farb DH (1981) The inactivation of gamma-aminobutyric acid transaminase in dissociated neuronal cultures from spinalcord. J Neurochem 36:985–990.

Rangel LM, Quinn LK, Chiba AA, Gage FH, and Aimone JB (2013) A hypothesis fortemporal coding of young and mature granule cells. Front Neurosci 7:75.

Rasmusson AM, Pinna G, Paliwal P, Weisman D, Gottschalk C, Charney D, KrystalJ, and Guidotti A (2006) Decreased cerebrospinal fluid allopregnanolone levels inwomen with posttraumatic stress disorder. Biol Psychiatry 60:704–713.

Rauch SL, Savage CR, Alpert NM, Fischman AJ, and Jenike MA (1997) The func-tional neuroanatomy of anxiety: a study of three disorders using positron emissiontomography and symptom provocation. Biol Psychiatry 42:446–452.

Rauch SL, Shin LM, and Wright CI (2003) Neuroimaging studies of amygdalafunction in anxiety disorders. Ann N Y Acad Sci 985:389–410.

Ravindran LN and Stein MB (2010) The pharmacologic treatment of anxiety dis-orders: a review of progress. J Clin Psychiatry 71:839–854.

Raymond JR, Mukhin YV, Gelasco A, Turner J, Collinsworth G, Gettys TW, GrewalJS, and Garnovskaya MN (2001) Multiplicity of mechanisms of serotonin receptorsignal transduction. Pharmacol Ther 92:179–212.

Reardon LE, Leen-Feldner EW, and Hayward C (2009) A critical review of the em-pirical literature on the relation between anxiety and puberty. Clin Psychol Rev 29:1–23.

Reddy DS and Rogawski MA (2012) Neurosteroids — endogenous regulators of sei-zure susceptibility and role in the treatment of epilepsy, in Jasper’s Basic Mech-anisms of the Epilepsies (Noebels JL, Avoli M, Rogawski MA, Olsen RW,and Delgado-Escueta AV eds) 4th ed, pp 984–1002, Little, Brown and Company,Boston, MA.

Regier DA, Narrow WE, and Rae DS (1990) The epidemiology of anxiety disorders:the Epidemiologic Catchment Area (ECA) experience. J Psychiatr Res 24 (Suppl 2):3–14.

Regier DA, Rae DS, Narrow WE, Kaelber CT, and Schatzberg AF (1998) Prevalenceof anxiety disorders and their comorbidity with mood and addictive disorders. Br JPsychiatry Suppl (34):24–28.

Reinhold JA, Mandos LA, Rickels K, and Lohoff FW (2011) Pharmacologicaltreatment of generalized anxiety disorder. Expert Opin Pharmacother 12:2457–2467.

Ressler KJ and Nemeroff CB (2000) Role of serotonergic and noradrenergic systemsin the pathophysiology of depression and anxiety disorders. Depress Anxiety 12(Suppl 1):2–19.

Ressler KJ, Rothbaum BO, Tannenbaum L, Anderson P, Graap K, Zimand E, HodgesL, and Davis M (2004) Cognitive enhancers as adjuncts to psychotherapy: use ofD-cycloserine in phobic individuals to facilitate extinction of fear. Arch GenPsychiatry 61:1136–1144.

Rhodes RA, Murthy NV, Dresner MA, Selvaraj S, Stavrakakis N, Babar S, Cowen PJ,and Grasby PM (2007) Human 5-HT transporter availability predicts amygdalareactivity in vivo. J Neurosci 27:9233–9237.

Rickels K and Rynn M (2002) Pharmacotherapy of generalized anxiety disorder.J Clin Psychiatry 63 (Suppl 14):9–16.

Roberts DS, Hu Y, Lund IV, Brooks-Kayal AR, and Russek SJ (2006) Brain-derivedneurotrophic factor (BDNF)-induced synthesis of early growth response factor 3(Egr3) controls the levels of type A GABA receptor alpha 4 subunits in hippocampalneurons. J Biol Chem 281:29431–29435.

Roberts DS, Raol YH, Bandyopadhyay S, Lund IV, Budreck EC, Passini MA,Wolfe JH, Brooks-Kayal AR, and Russek SJ (2005) Egr3 stimulation ofGABRA4 promoter activity as a mechanism for seizure-induced up-regulationof GABA(A) receptor alpha4 subunit expression. Proc Natl Acad Sci USA 102:11894–11899.

Robinson RT, Drafts BC, and Fisher JL (2003) Fluoxetine increases GABA(A) re-ceptor activity through a novel modulatory site. J Pharmacol Exp Ther 304:978–984.

Roozendaal B, de Quervain DJ, Ferry B, Setlow B, and McGaugh JL (2001) Baso-lateral amygdala-nucleus accumbens interactions in mediating glucocorticoid en-hancement of memory consolidation. J Neurosci 21:2518–2525.

Rosso IM, Weiner MR, Crowley DJ, Silveri MM, Rauch SL, and Jensen JE (2014)Insula and anterior cingulate GABA levels in posttraumatic stress disorder: pre-liminary findings using magnetic resonance spectroscopy. Depress Anxiety 31:115–123.

Rothbaum BO, Price M, Jovanovic T, Norrholm SD, Gerardi M, Dunlop B, Davis M,Bradley B, Duncan EJ, and Rizzo A, et al. (2014) A randomized, double-blindevaluation of D-cycloserine or alprazolam combined with virtual reality exposuretherapy for posttraumatic stress disorder in Iraq and Afghanistan War veterans.Am J Psychiatry 171:640–648.

Rudolph U, Crestani F, Benke D, Brünig I, Benson JA, Fritschy JM, Martin JR,Bluethmann H, and Möhler H (1999) Benzodiazepine actions mediated by specificgamma-aminobutyric acid(A) receptor subtypes. Nature 401:796–800.

Rudolph U and Möhler H (2006) GABA-based therapeutic approaches: GABAA re-ceptor subtype functions. Curr Opin Pharmacol 6:18–23.

Sah A, Schmuckermair C, Sartori SB, Gaburro S, Kandasamy M, Irschick R,Klimaschewski L, Landgraf R, Aigner L, and Singewald N (2012) Anxiety- ratherthan depression-like behavior is associated with adult neurogenesis in a femalemouse model of higher trait anxiety- and comorbid depression-like behavior.Transl Psychiatr 2:e171.

Sah P, Faber ES, Lopez De Armentia M, and Power J (2003) The amygdaloid com-plex: anatomy and physiology. Physiol Rev 83:803–834.

Sajdyk TJ and Shekhar A (1997) Excitatory amino acid receptors in the basolateralamygdala regulate anxiety responses in the social interaction test. Brain Res 764:262–264.

Sandford JJ, Forshall S, Bell C, Argyropoulos S, Rich A, D’Orlando KJ, GammansRE, and Nutt DJ (2001) Crossover trial of pagoclone and placebo in patients withDSM-IV panic disorder. J Psychopharmacol 15:205–208.

Schwartz TL, Siddiqui UA, and Raza S (2012) Memantine as an augmentationtherapy for anxiety disorders. Case Report Psychiatry 2012:749796.

Schwartz TL, Siddiqui UA, Raza S, and Costello A (2010) Acamprosate calcium asaugmentation therapy for anxiety disorders. Ann Pharmacother 44:1930–1932.

Selye H (1970) Adaptive steroids: retrospect and prospect. Perspect Biol Med 13:343–363.

Semeniuk T, Jhangri GS, and Le Mellédo JM (2001) Neuroactive steroid levels inpatients with generalized anxiety disorder. J Neuropsychiatry Clin Neurosci 13:396–398.

Serra M, Ghiani CA, Motzo C, Cuccheddu T, Floris S, Giusti P, and Biggio G (1994)Imidazenil, a new partial agonist of benzodiazepine receptors, reverses the in-hibitory action of isoniazid and stress on gamma-aminobutyric acidA receptorfunction. J Pharmacol Exp Ther 269:32–38.

Sertraline (2012) Prescribing information. Pfizer/Roehrig, New York.Shackman AJ, Fox AS, Oler JA, Shelton SE, Davidson RJ, and Kalin NH (2013)Neural mechanisms underlying heterogeneity in the presentation of anxioustemperament. Proc Natl Acad Sci USA 110:6145–6150.

Shen H, Gong QH, Aoki C, Yuan M, Ruderman Y, Dattilo M, Williams K, and SmithSS (2007) Reversal of neurosteroid effects at alpha4beta2delta GABAA receptorstriggers anxiety at puberty. Nat Neurosci 10:469–477.

Sieghart W (1994) Pharmacology of benzodiazepine receptors: an update. J Psychi-atry Neurosci 19:24–29.

Sieghart W and Sperk G (2002) Subunit composition, distribution and function ofGABA(A) receptor subtypes. Curr Top Med Chem 2:795–816.

Sim JA, Skynner MJ, and Herbison AE (2001) Direct regulation of postnatal GnRHneurons by the progesterone derivative allopregnanolone in the mouse. Endocri-nology 142:4448–4453.

1030 Farb and Ratner

Page 30: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

Simpson JR, Ongür D, Akbudak E, Conturo TE, Ollinger JM, Snyder AZ, GusnardDA, and Raichle ME (2000) The emotional modulation of cognitive processing: anfMRI study. J Cogn Neurosci 12 (Suppl 2):157–170.

Skolnick P, Paul SM, and Weissman BA (1984) Preclinical pharmacology of buspironehydrochloride. Pharmacotherapy 4:308–314.

Smith AJ, Alder L, Silk J, Adkins C, Fletcher AE, Scales T, Kerby J, Marshall G,Wafford KA, and McKernan RM, et al. (2001) Effect of alpha subunit on allostericmodulation of ion channel function in stably expressed human recombinantgamma-aminobutyric acid(A) receptors determined using (36)Cl ion flux. MolPharmacol 59:1108–1118.

Spigelman I, Li Z, Liang J, Cagetti E, Samzadeh S, Mihalek RM, Homanics GE,and Olsen RW (2003) Reduced inhibition and sensitivity to neurosteroids in hip-pocampus of mice lacking the GABA(A) receptor delta subunit. J Neurophysiol 90:903–910.

Sripada RK, Marx CE, King AP, Rampton JC, Ho SS, and Liberzon I (2013)Allopregnanolone elevations following pregnenolone administration are associatedwith enhanced activation of emotion regulation neurocircuits. Biol Psychiatry 73:1045–1053.

Staley KJ and Mody I (1992) Shunting of excitatory input to dentate gyrus granulecells by a depolarizing GABAA receptor-mediated postsynaptic conductance.J Neurophysiol 68:197–212.

Stein MB, Jang KL, Taylor S, Vernon PA, and Livesley WJ (2002) Genetic andenvironmental influences on trauma exposure and posttraumatic stress disordersymptoms: a twin study. Am J Psychiatry 159:1675–1681.

Stein MB, Keshaviah A, Haddad SA, Van Ameringen M, Simon NM, Pollack MH,and Smoller JW (2014) Influence of RGS2 on sertraline treatment for social anxietydisorder. Neuropsychopharmacology 39:1340–1346.

Stein MB, Pollack MH, Bystritsky A, Kelsey JE, and Mangano RM (2005) Efficacy oflow and higher dose extended-release venlafaxine in generalized social anxietydisorder: a 6-month randomized controlled trial. Psychopharmacology (Berl) 177:280–288.

Steinbach JH and Akk G (2001) Modulation of GABA(A) receptor channel gating bypentobarbital. J Physiol 537:715–733.

Stell BM, Brickley SG, Tang CY, Farrant M, and Mody I (2003) Neuroactive steroidsreduce neuronal excitability by selectively enhancing tonic inhibition mediated bydelta subunit-containing GABAA receptors. Proc Natl Acad Sci USA 100:14439–14444.

Stephens DN, Schneider HH, Kehr W, Andrews JS, Rettig KJ, Turski L, SchmiechenR, Turner JD, Jensen LH, and Petersen EN, et al. (1990) Abecarnil, a metabolicallystable, anxioselective beta-carboline acting at benzodiazepine receptors. J Phar-macol Exp Ther 253:334–343.

Stork O, Ji FY, and Obata K (2002) Reduction of extracellular GABA in the mouseamygdala during and following confrontation with a conditioned fear stimulus.Neurosci Lett 327:138–142.

Stork O, Yamanaka H, Stork S, Kume N, and Obata K (2003) Altered conditionedfear behavior in glutamate decarboxylase 65 null mutant mice. Genes Brain Behav2:65–70.

Strawn JR, Chu WJ, Whitsel RM, Weber WA, Norris MM, Adler CM, Eliassen JC,Phan KL, Strakowski SM, and DelBello MP (2013) A pilot study of anterior cin-gulate cortex neurochemistry in adolescents with generalized anxiety disorder.Neuropsychobiology 67:224–229.

Ströhle A, Romeo E, di Michele F, Pasini A, Hermann B, Gajewsky G, Holsboer F,and Rupprecht R (2003) Induced panic attacks shift gamma-aminobutyric acid typeA receptor modulatory neuroactive steroid composition in patients with panicdisorder: preliminary results. Arch Gen Psychiatry 60:161–168.

Swanson CJ, Bures M, Johnson MP, Linden AM, Monn JA, and Schoepp DD (2005)Metabotropic glutamate receptors as novel targets for anxiety and stress disorders.Nat Rev Drug Discov 4:131–144.

Toth M (2003) 5-HT1A receptor knockout mouse as a genetic model of anxiety. Eur JPharmacol 463:177–184.

Tretter V, Ehya N, Fuchs K, and Sieghart W (1997) Stoichiometry and assembly ofa recombinant GABAA receptor subtype. J Neurosci 17:2728–2737.

Tromp DP, Grupe DW, Oathes DJ, McFarlin DR, Hernandez PJ, Kral TR, Lee JE,Adams M, Alexander AL, and Nitschke JB (2012) Reduced structural connectivityof a major frontolimbic pathway in generalized anxiety disorder. Arch Gen Psy-chiatry 69:925–934.

Twyman RE, Rogers CJ, and Macdonald RL (1989) Differential regulation of gamma-aminobutyric acid receptor channels by diazepam and phenobarbital. Ann Neurol25:213–220.

Tye KM, Prakash R, Kim SY, Fenno LE, Grosenick L, Zarabi H, Thompson KR,Gradinaru V, Ramakrishnan C, and Deisseroth K (2011) Amygdala circuitry me-diating reversible and bidirectional control of anxiety. Nature 471:358–362.

Urbani A and Belluzzi O (2000) Riluzole inhibits the persistent sodium current inmammalian CNS neurons. Eur J Neurosci 12:3567–3574.

Uzunova V, Sheline Y, Davis JM, Rasmusson A, Uzunov DP, Costa E, and Guidotti A(1998) Increase in the cerebrospinal fluid content of neurosteroids in patients withunipolar major depression who are receiving fluoxetine or fluvoxamine. Proc NatlAcad Sci USA 95:3239–3244.

Van der Linden G, van Heerden B, Warwick J, Wessels C, van Kradenburg J, Zungu-Dirwayi N, and Stein DJ (2000) Functional brain imaging and pharmacotherapy insocial phobia: single photon emission computed tomography before and aftertreatment with the selective serotonin reuptake inhibitor citalopram. ProgNeuropsychopharmacol Biol Psychiatry 24:419–438.

Van Laere K, Bormans G, Sanabria-Bohórquez SM, de Groot T, Dupont P, DeLepeleire I, de Hoon J, Mortelmans L, Hargreaves RJ, and Atack JR, et al. (2008)In vivo characterization and dynamic receptor occupancy imaging of TPA023B, analpha 2/alpha 3/alpha 5 subtype selective gamma-aminobutyric acid-a partialagonist. Biol Psychiatry 64:153–161.

Van Oekelen D, Luyten WH, and Leysen JE (2003) 5-HT2A and 5-HT2C receptorsand their atypical regulation properties. Life Sci 72:2429–2449.

Van Oort FV, Greaves-Lord K, Verhulst FC, Ormel J, and Huizink AC (2009) Thedevelopmental course of anxiety symptoms during adolescence: the TRAILS study.J Child Psychol Psychiatry 50:1209–1217.

van Steveninck AL, Gieschke R, Schoemaker RC, Roncari G, Tuk B, Pieters MS,Breimer DD, and Cohen AF (1996) Pharmacokinetic and pharmacodynamicinteractions of bretazenil and diazepam with alcohol. Br J Clin Pharmacol 41:565–573.

Varia I and Rauscher F (2002) Treatment of generalized anxiety disorder with cit-alopram. Int Clin Psychopharmacol 17:103–107.

Venlafaxine (2012) Prescribing information. Pfizer/Wyeth Pharmaceuticals, Inc.,Philadelphia.

Villiger JW (1984) CL 218,872 binding to benzodiazepine receptors in rat spinal cord:modulation by gamma-aminobutyric acid and evidence for receptor heterogeneity.J Neurochem 43:903–905.

Wafford KA, Whiting PJ, and Kemp JA (1993) Differences in affinity and efficacy ofbenzodiazepine receptor ligands at recombinant gamma-aminobutyric acidA re-ceptor subtypes. Mol Pharmacol 43:240–244.

Walker DL, Miles LA, and Davis M (2009) Selective participation of the bed nucleusof the stria terminalis and CRF in sustained anxiety-like versus phasic fear-likeresponses. Prog Neuropsychopharmacol Biol Psychiatry 33:1291–1308.

Walker DL, Ressler KJ, Lu KT, and Davis M (2002) Facilitation of conditioned fearextinction by systemic administration or intra-amygdala infusions of D-cycloserineas assessed with fear-potentiated startle in rats. J Neurosci 22:2343–2351.

Walker DL, Toufexis DJ, and Davis M (2003) Role of the bed nucleus of the striaterminalis versus the amygdala in fear, stress, and anxiety. Eur J Pharmacol 463:199–216.

Wang C, Marx CE, Morrow AL, Wilson WA, and Moore SD (2007) Neurosteroidmodulation of GABAergic neurotransmission in the central amygdala: a role forNMDA receptors. Neurosci Lett 415:118–123.

Wang MD, Zhu D, Bäckström T, and Wahlström G (2001) The interaction betweenethanol and pregnanolone at induction of anaesthesia investigated with a thresh-old method in male rats. Br J Pharmacol 134:1393–1402.

Wang SJ, Wang KY, and Wang WC (2004) Mechanisms underlying the riluzole in-hibition of glutamate release from rat cerebral cortex nerve terminals (synapto-somes). Neuroscience 125:191–201.

Wang SM and Pae CU (2013) How much to worry about the FDA warning in the useof citalopram? Expert Rev Neurother 13:883–886.

Warwick JM, Carey PD, Cassimjee N, Lochner C, Hemmings S, Moolman-Smook H,Beetge E, Dupont P, and Stein DJ (2012) Dopamine transporter binding in socialanxiety disorder: the effect of treatment with escitalopram. Metab Brain Dis 27:151–158.

Weaver CE, Land MB, Purdy RH, Richards KG, Gibbs TT, and Farb DH (2000)Geometry and charge determine pharmacological effects of steroids on N-methyl-D-aspartate receptor-induced Ca(2+) accumulation and cell death. J PharmacolExp Ther 293:747–754.

Weaver CE Jr, Marek P, Park-Chung M, Tam SW, and Farb DH (1997) Neuro-protective activity of a new class of steroidal inhibitors of the N-methyl-D-aspartate receptor. Proc Natl Acad Sci USA 94:10450–10454.

Whiting PJ, Bonnert TP, McKernan RM, Farrar S, Le Bourdellès B, Heavens RP,Smith DW, Hewson L, Rigby MR, and Sirinathsinghji DJ, et al. (1999) Molecularand functional diversity of the expanding GABA-A receptor gene family. Ann N YAcad Sci 868:645–653.

Wieland HA, Lüddens H, and Seeburg PH (1992) A single histidine in GABAAreceptors is essential for benzodiazepine agonist binding. J Biol Chem 267:1426–1429.

Wieland S, Belluzzi J, Hawkinson JE, Hogenkamp D, Upasani R, Stein L, Wood PL,Gee KW, and Lan NC (1997) Anxiolytic and anticonvulsant activity of a syntheticneuroactive steroid Co 3-0593. Psychopharmacology (Berl) 134:46–54.

Williams DB and Akabas MH (2001) Evidence for distinct conformations of the twoalpha 1 subunits in diazepam-bound GABA(A) receptors. Neuropharmacology 41:539–545.

Wingrove PB, Safo P, Wheat L, Thompson SA, Wafford KA, and Whiting PJ (2002)Mechanism of alpha-subunit selectivity of benzodiazepine pharmacology atgamma-aminobutyric acid type A receptors. Eur J Pharmacol 437:31–39.

Wisden W, Laurie DJ, Monyer H, and Seeburg PH (1992) The distribution of 13GABAA receptor subunit mRNAs in the rat brain. I. Telencephalon, diencephalon,mesencephalon. J Neurosci 12:1040–1062.

Wolfman C, Viola H, Marder M, Wasowski C, Ardenghi P, Izquierdo I, Paladini AC,and Medina JH (1996) Anxioselective properties of 6,39-dinitroflavone, a high-affinity benzodiazepine receptor ligand. Eur J Pharmacol 318:23–30.

Wolfman C, Viola H, Paladini A, Dajas F, and Medina JH (1994) Possible anxiolyticeffects of chrysin, a central benzodiazepine receptor ligand isolated from Passifloracoerulea. Pharmacol Biochem Behav 47:1–4.

Woods AM and Bouton ME (2006) D-cycloserine facilitates extinction but does noteliminate renewal of the conditioned emotional response. Behav Neurosci 120:1159–1162.

Wu FS, Gibbs TT, and Farb DH (1991) Pregnenolone sulfate: a positive allostericmodulator at the N-methyl-D-aspartate receptor. Mol Pharmacol 40:333–336.

Wu FS, Gibbs TT, and Farb DH (1993) Dual activation of GABAA and glycinereceptors by beta-alanine: inverse modulation by progesterone and 5 alpha-pregnan-3 alpha-ol-20-one. Eur J Pharmacol 246:239–246.

Yamawaki S (1999) The use and development of anxiolytics in Japan. Eur Neuro-psychopharmacol 9 (Suppl 6):S413–S419.

Yang P, Jones BL, and Henderson LP (2002) Mechanisms of anabolic androgenicsteroid modulation of alpha(1)beta(3)gamma(2L) GABA(A) receptors. Neurophar-macology 43:619–633.

Yilmazer-Hanke DM, Roskoden T, Zilles K, and Schwegler H (2003) Anxiety-relatedbehavior and densities of glutamate, GABAA, acetylcholine and serotoninreceptors in the amygdala of seven inbred mouse strains. Behav Brain Res 145:145–159.

Targeted Modulation of Neural Circuits Implicated in Anxiolysis 1031

Page 31: Targeting the Modulation of Neural Circuitry for the Treatment of …pharmrev.aspetjournals.org › ... › pharmrev › 66 › 4 › 1002.full.pdf · N-methyl-D-aspartate (NMDA)

Young WS 3rd, Niehoff D, Kuhar MJ, Beer B, and Lippa AS (1981) Multiple benzo-diazepine receptor localization by light microscopic radiohistochemistry. J Phar-macol Exp Ther 216:425–430.

Zanardi R, Serretti A, Rossini D, Franchini L, Cusin C, Lattuada E, Dotoli D,and Smeraldi E (2001) Factors affecting fluvoxamine antidepressant activity: in-fluence of pindolol and 5-HTTLPR in delusional and nondelusional depression. BiolPsychiatry 50:323–330.

Zwanzger P, Baghai T, Boerner RJ, Möller HJ, and Rupprecht R (2001a) Anxiolyticeffects of vigabatrin in panic disorder. J Clin Psychopharmacol 21:539–540.

Zwanzger P, Baghai TC, Schuele C, Ströhle A, Padberg F, Kathmann N, Schwarz M,Möller HJ, and Rupprecht R (2001b) Vigabatrin decreases cholecystokinin-tetrapeptide(CCK-4) induced panic in healthy volunteers. Neuropsychopharmacology 25:699–703.

Zwanzger P, Eser D, Padberg F, Baghai TC, Schule C, Rötzer F, Ella R, Möller HJ,and Rupprecht R (2003) Effects of tiagabine on cholecystokinin-tetrapeptide (CCK-4)-induced anxiety in healthy volunteers. Depress Anxiety 18:140–143.

Zwanzger P, Zavorotnyy M, Gencheva E, Diemer J, Kugel H, Heindel W, Ruland T,Ohrmann P, Arolt V, and Domschke K, et al. (2013) Acute shift in glutamateconcentrations following experimentally induced panic with cholecystokinin tet-rapeptide—a 3T-MRS study in healthy subjects. Neuropsychopharmacology 38:1648–1654.

Zweifel LS, Fadok JP, Argilli E, Garelick MG, Jones GL, Dickerson TM, Allen JM,Mizumori SJ, Bonci A, and Palmiter RD (2011) Activation of dopamine neurons iscritical for aversive conditioning and prevention of generalized anxiety. Nat Neu-rosci 14:620–626.

1032 Farb and Ratner