The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Published Online:https://doi.org/10.1176/foc.6.4.foc486

Abstract

The Anxiety Disorders Association of America convened a conference of experts to address treatment-resistant anxiety disorders and review promising novel approaches to the treatment of refractory anxiety disorders. Workgroup leaders and other participants reviewed the literature and considered the presentations and discussions from the conference. Authors placed the emerging literature on new therapeutic approaches into clinical perspective and identified unmet needs and priority areas for future research. There is a relative paucity of efforts addressing inadequate response to anxiety disorder treatment. Systematic efforts to exhaust all therapeutic options and overcome barriers to effective treatment delivery are needed before patients can be considered treatment refractory. Cognitive behavioral therapy, especially in combination with pharmacotherapy, must be tailored to accommodate the effects of clinical context on treatment response. The literature on pharmacologic treatment of refractory anxiety disorders is small but growing and includes studies of augmentation strategies and non-traditional anxiolytics. Research efforts to discover new pharmacologic targets are focusing on neuronal systems that mediate responses to stress and fear. A number of clinical and basic science studies were proposed that would advance the research agenda and improve treatment of patients with anxiety disorders. Significant advances have been made in the development of psychotherapeutic and pharmacologic treatments for anxiety disorders. Unfortunately, many patients remain symptomatic and functionally impaired. Progress in the development of new treatments has great promise, but will only succeed through a concerted research effort that systematically evaluates potential areas of importance and properly uses scarce resources.

(Reprinted with permission from Depression and Anxiety 2008; 25:467–476)

INTRODUCTION

Remarkable progress has been made in the development of medication and psychosocial treatment strategies for anxiety disorders (Ballenger, 2001). However, many patients remain symptomatic despite improvements in therapy. Randomized, controlled trials (RCTs) of pharmacotherapy for anxiety disorders report response rates of 40–70% and remission rates of 20–47% (Ballenger, 1999). In addition, these reported improvement rates may be overestimates because of selection bias, failure to publish negative trials, and exclusion from RCTs of clinically relevant patients with comorbid psychiatric or medical disorders (Roy-Byrne and Cowley, 1998). Anxiety disorders are considered treatment resistant when there are residual symptoms or when symptoms do not improve at all after some form of therapeutic intervention. However, research on treatment-resistant anxiety disorders has lagged behind similar studies of schizophrenia and major depressive disorder (Roy-Byrne and Cowley, 1998). Clearly, much work is needed to advance therapeutic strategies and improve rates of clinically meaningful benefit for patients with treatment-resistant anxiety disorders.

The Anxiety Disorders Association of America convened a 2-day conference in Chantilly, Virginia on June 16–17, 2003 to address the issue of treatment-resistant anxiety disorders and review promising, novel therapeutic approaches. The conference consisted of presentations, open discussions, and workgroup sessions by clinicians and researchers in the fields of psychosocial therapies, psychopharmacology, and neurobiology (Appendix). The panel did not consider obsessive-compulsive disorder (OCD) because some believe that it may be better conceptualized as part of the obsessive-compulsive spectrum disorders and may be moved to a separate diagnostic category in the fifth edition of the diagnostic and statistical manual of mental disorders (DSM-V) (Hollander and Zohar, 2004). In this article, we consider issues associated with treatment resistance, cognitive behavioral therapy (CBT), pharmacologic approaches, and new drug discovery. The review of CBT approaches discusses factors associated with poor response to psychotherapy and focuses on extinction-based learning, which is a concept that can be applied to all anxiety disorders. The pharmacotherapy sections review reported novel pharmacological strategies for refractory anxiety and potential new targets for drug development. Finally, we outline research priorities for studies of treatment-resistant anxiety disorders.

THERAPEUTIC GOALS AND TREATMENT RESISTANCE

Many factors contribute to poor therapeutic response, including treatment-related factors (e.g., incorrect or incomplete diagnosis, inappropriate intervention, inadequate dosing, insufficient treatment duration), patient-related factors (e.g., comorbidity, poor adherence), and logistical factors (e.g., scarcity of trained treatment providers, inadequate insurance coverage). The dissemination of strategies to enhance appropriate diagnosis of anxiety disorders and relevant comorbidities, as well as the optimal use of pharmacologic and psychosocial strategies for their treatment is critical to improve therapeutic outcomes for affected individuals. Patient engagement, influenced by patients' beliefs about anxiety, treatment preferences, experience of stigma, and readiness to change, is also critical. Systematic efforts at optimizing treatment engagement must be addressed before patients truly can be considered to be treatment refractory. Such efforts include patient education about the natural course of the illness, the benefits of effective treatment, and expected time frame for symptom resolution. Guidance about the importance of exposure to feared situations and the use of effective coping strategies and skills development is needed to achieve maximum clinical gains. Finally, the contribution of life stressors, comorbidity, and other factors should be assessed and addressed with appropriate targeted interventions (e.g., individual, couples, or family psychotherapy for intrapsychic or interpersonal difficulties) in a patient with a suboptimal clinical outcome (Roy-Byrne and Cowley, 1998).

There is neither a universally accepted definition of treatment resistance nor consensus on the definitive number and characteristics of interventions needed to constitute an adequate treatment trial for persistently symptomatic anxiety disorders. Nonetheless, it appears intuitive that treatment goals should be to achieve well-defined therapeutic endpoints with sustained resolution of core anxiety symptoms, functional disability, and psychiatric comorbidities. Each anxiety disorder is unique in its natural course, presentation, and associated morbidity, and the metrics of response and remission must consider all affected domains, including vocational and social dysfunction and overall quality of life.

Consensus standards have been proposed for response and remission based on improvement in multiple dimensions of individual anxiety disorders (Ballenger, 2001), though there is little empiric validation for these criteria and a lack of universal adoption by leading professional societies. Nonetheless, it is encouraging that these guidelines focus on the resolution or significant improvement in core anxiety symptoms, functional impairment, and comorbid depression. Proposed general endpoints for remission across anxiety disorders include reduction in Hamilton Rating Scale for Anxiety scores to ≤7, Sheehan Disability Scale scores to ≤1 (i.e., mildly disabled), and Hamilton Rating Scale for Depression scores to ≤7. In addition, disorder-specific remission criteria include reduction in the Panic Disorder Severity Scale to <7 for panic disorder, Liebowitz Social Anxiety Scale score to ≤30 for social anxiety disorder, and the Treatment Outcome for Posttraumatic Stress Disorder (PTSD) Scale score to ≤5 or 6 for PTSD (Ballenger, 2001).

COGNITIVE-BEHAVIORAL APPROACHES

It is difficult to identify a standard trial of CBT because of the remarkable heterogeneity of interventions for anxiety disorders. For example, exposure interventions may include imaginal, in vivo or interoceptive exposure, or the use of virtual reality technology for hard-to-arrange exposures.

Attempts at defining treatment resistance in this context are fraught with difficulty given the wide range of CBT and the diversity of anxiety disorders. Nonetheless, certain key items should be addressed. An adequate course of CBT should include a full protocol of treatment, ideally defined by interventions found to be effective in at least two controlled trials. Treatment protocols often include 12–16 sessions of a combination of information, cognitive restructuring, and exposure interventions, with skill rehearsal and exposure in home-practice assignments (Barlow, 2001). The use of protocol-driven CBT for anxiety disorders is well tolerated, cost-effective, and efficacious acutely and in the long term (Foa et al., 2002; McHugh et al., in press; Otto et al., 2004), with evidence for further gains following acute CBT as patients consolidate and extend learning-based approaches (Liebowitz et al., 1999).

Supportive interventions, such as brief relaxation treatment or breathing retraining do not constitute an adequate CBT trial as these symptom management procedures alone do not, for example, add to the efficacy of CBT protocols for panic disorder (Schmidt et al., 2002). Failure to respond to an initial CBT protocol suggests the need for alternative interventions (e.g., greater attention to changing cognitions, refinement of the target or intensity of exposure interventions), other empirically supported treatments, or combined treatment modalities. Relevant recent findings on extinction learning and context that can be applied across the anxiety disorders are reviewed, and next-step strategies for non-response are considered.

EXTINCTION LEARNING AND LIMITING FACTORS

Anxiety disorders are characterized by exaggerated distress and disability in response to phobic cues. As long as an individual can learn and integrate new experiences with phobic stimuli, exaggerated responses to phobic cues should respond to extinction learning, and anxiety, thus diminish. However, this assumes that the correct fears are being targeted and appropriate learning conditions are identified and established by the therapist.

Informational and cognitive interventions often are used to facilitate and enhance initial exposure to fears, which are designed to become increasingly relevant to core fears as therapy progresses. Balance must be maintained between conditions that facilitate exposure to avoided phobic cues and conditions where exposure provides useful learning. Strategies that avert or attenuate anxiety in phobic situations (i.e., “safety behaviors”) may temporarily alleviate anxiety, but reduce long-term efficacy (Powers et al., 2004; Salkovskis et al., 1999).

What is learned during extinction in one context (e.g., when safety behaviors or medications are available) may not generalize to a different context (e.g., when no safety behavior or medications are available) (Bouton, 2002). Stronger findings have been reported for shifts in internal context, such as medication effects (Mystkowski et al., 2003). Therapeutic approaches for treatment-resistant anxiety disorders should include strategies that maximize fear extinction during exposure and generalize this learning across contexts, seeking to reverse the effects of distraction, safety cues, and context shifts on the longer-term acquisition of a sense of safety when confronted by phobic cues (Powers et al., 2006).

Attention to shifts in context, specifically internal context provided by pharmacotherapy, is relevant to combining medications with CBT. Context effects may explain the loss of efficacy seen in combination treatment trials when medications are later discontinued (e.g., Barlow et al., 2000). However, maintenance and extension of treatment gains are common when CBT is provided during and after medication taper (Otto et al., 2002).

Context effects introduce additional complexity into the selection of the next-step strategy following partial response to CBT. Should patients be switched to pharmacotherapy, risking the introduction of context effects that may require additional CBT to prevent relapse in the future? If there was no response to CBT, then there is little reason to be concerned about context effects introduced by medications. However, if patients achieved a partial response to CBT, further honing of the targets of CBT interventions and identification of context effects that may be preventing generalization of exposure-based learning should be considered before addition of pharmacologic alternatives.

There is a dearth of evidence addressing whether troubleshooting and persisting with CBT is better than switching treatment modalities (Kampman et al., 2002). In one small randomized trial, continuation of exposure-based treatment alone was at least as effective as its combination with imipramine or cognitive therapy (Fava et al., 1997). Further, dropout data from controlled trials of CBT and pharmacotherapy indicate that CBT tends to be more tolerable and acceptable than medication alternatives (Otto et al., 2005). Nonetheless, non-response to initial CBT does provide a call to therapists to re-evaluate whether core fears are being adequately targeted by cognitive and exposure interventions, with consideration of altering the timing, modality, and context (including the presence of safety cues that may undermine the efficacy of exposure treatments), to try to enhance response to additional CBT (e.g., Powers et al., 2004, 2006; Smits et al., 2006).

In addition, recent research has introduced a new approach to combined treatment, in which pharmacotherapy is used to enhance therapeutic learning rather than attenuate symptoms (Ressler et al., 2004). The N-methyl-D-aspartate (NMDA) agonist, D-cycloserine (DCS) augments extinction learning in animal models after a single dose administered immediately before extinction trials (Walker et al., 2002). In a proof of concept translational RCT, 28 patients with acrophobia were randomized to virtual-reality exposure (VRE) therapy plus one of two doses of DCS or placebo, administered before each of 2 weekly VRE sessions (Ressler et al., 2004). DCS resulted in significantly less fear of heights compared with placebo during the trial as well as following treatment. In an independent replication of the DCS enhancement effect, Hofmann et al. (2006) demonstrated better outcome in a sample of 27 outpatients with primarily generalized social anxiety disorder when treated with DCS versus placebo in the context of a five-session CBT protocol combining study pills with the latter four sessions of increasingly challenging public speech exposures. Furthermore, recent evidence also supports the efficacy of combined treatment with DCS for OCD, although there are indications that with ongoing exposure therapy CBT alone can catch up to the boost in efficacy provided by DCS (Kushner et al., in press). If these promising findings are further replicated, this strategy for combined treatment suggests a potentially expeditious way to improve the speed of onset and, perhaps, overall efficacy of CBT for anxiety.

CBT FOR PHARMACOTHERAPY NON-RESPONDERS

CBT has been successfully used to facilitate discontinuation of benzodiazepines and antidepressants (Otto et al., 2002; Schmidt et al., 2002) as well as to boost response in patients who have failed previous medication trials for panic disorder and PTSD (Otto et al., 2003, 1999; Heldt et al., 2006). Accordingly, CBT remains a central strategy for patients who do not respond fully to pharmacotherapy.

PHARMACOLOGIC APPROACHES

Though not intended as a study of treatment resistance, the results of a study in PTSD have relevance for the management of treatment-resistant anxiety disorders in general. Of 128 patients with PTSD who did not respond to an acute 12-week trial of sertraline, 54% converted to responders during a subsequent 24-week continuation phase (Londborg et al., 2001), suggesting that treatment may need to be continued for months beyond the typical acute 6–12-week course to consider a patient truly treatment refractory. However, as it is frequently difficult for patients to adhere to treatment for months with an inadequate response, initiation of alternative or additional treatment strategies is often necessary to maintain a sense of therapeutic optimism.

As is true for CBT, there is a relative dearth of data about pharmacotherapy for treatment-resistant anxiety disorders. Most available information comes from small, open-label, or retrospective reports. Nonetheless, the need for more effective and better-tolerated treatments has prompted exploration of pharmacological augmentation, novel uses of non-traditional anxiolytic drugs, and new drug development. No drug has yet received FDA approval for refractory anxiety so, as is typical in the management of refractory states, use of pharmacologic strategies in this context is off-label. In this section, we note evidence from randomized controlled trials (and in their absence, case series or reports) on pharmacotherapy for refractory anxiety disorders and novel uses of non-traditional antianxiety agents.

ATYPICAL ANTIPSYCHOTICS

Augmentation of existing antianxiety treatments with atypical antipsychotics having mixed effects at dopaminergic and serotonin receptors appears to be an effective strategy for refractory anxiety disorders as evidenced by double-blind randomized controlled trials with risperidone and olanzapine for generalized anxiety disorder (GAD) (Brawman-Mintzer et al., 2005; Pollack et al., 2006) and PTSD (Bartzokis et al., 2005; Hamner et al., 2003; Monnelly et al., 2003; Stein et al., 2002). Further there are case studies reporting the efficacy of olanzapine for refractory panic disorder (Etxebeste et al., 2000; Khaldi et al., 2003) and one small randomized controlled trial with olanzapine monotherapy suggesting its efficacy in social anxiety disorder (though not specifically refractory patients) (Barnett et al., 2002).

ANTICONVULSANTS

Open-label case reports and case series suggest the adjunctive efficacy of anticonvulsants for refractory GAD, including gabapentin, which acts as an α2 delta voltage-gated, calcium ion channel antagonist and may enhance γ-aminobutyric acid (GABA) synthesis (Pollack et al., 1998), the voltage-gated calcium channel modulator levetiracetam (Pollack, 2002) and the selective GABA reuptake inhibitor tiagabine (Crane, 2003). In addition, reports suggest efficacy for gabapentin (Pollack et al., 1998), tiagabine (Zwanzger et al., 2001), and valproic acid (Ontiveros and Fontaine, 1992) for refractory panic. Further, case and chart reviews describe the potential efficacy of the addition of gabapentin (Brannon et al., 2000), topiramate (Berlant and van Kammen, 2002), tiagabine (Berigan, 2002), phenytoin (Bremner et al., 2004), valproate (Szymanski and Olympia, 1991), and levetiracetam (Kinrys et al., 2006), in individuals with civilian or combat-related PTSD, providing signals for efficacy in the domains of sleep duration, nightmare frequency and severity, and intrusive recollections. Likewise, efficacy was suggested for topiramate, an anticonvulsant that blocks sodium channels, increases GABA levels, and potentiates GABA activity at non-benzodiazepine receptors (Shaywitz and Liebowitz, 2003; Van Ameringen et al., 2004), valproic acid (Kinrys et al., 2003), and levetiracetam (Simon et al., 2004) in open-label trials in social anxiety disorder.

Though not specifically tested in refractory patients, pregabalin, an α2 delta voltage-gated, calcium ion channel antagonist that has anticonvulsant and analgesic effects has been demonstrated to be effective for GAD and social anxiety disorder in controlled trials (Feltner et al., 2000; Rickels et al., 2005), with suggestions of an onset of anxiolytic response similar to the benzodiazepine comparator. Similarly, lamotrigine, an antagonist at sodium and calcium channels as well as a modulator of GABA and glutamatergic activity, was demonstrated effective in a small, randomized placebo-controlled study of individuals with PTSD (Hertzberg et al., 1999), as was gabapentin in an RCT for social anxiety disorder (Pande et al., 1999a).

NOVEL USES OF NON-TRADITIONAL ANXIOLYTICS AND OTHER PHARMACOLOGIC STRATEGIES

Pindolol, a β-blocker with antagonist effects on the 5HT1A autoreceptor (thus potentially facilitating serotonergic activity) was demonstrated effective in a double-blind randomized, placebo-controlled trial of selective serotonin reuptake inhibitor (SSRI)-resistant panic disorder (Hirschmann et al., 2000), though not social anxiety disorder (Stein et al., 2001), with significant improvement observed at 2 weeks. Prazosin, a centrally active α1-adrenergic antagonist, showed evidence of efficacy for the treatment of nightmares, sleep disturbance, and overall PTSD symptoms in a 20 week, double-blind, placebo-controlled crossover study of 10 combat veterans with chronic PTSD (Raskind et al., 2003). Similarly small case series suggest the potential efficacy of combined treatment with a tricyclic antidepressant and SSRI for panic disorder refractory to antidepressant monotherapy (Tiffon et al., 1994), buspirone augmentation of SSRI non-responders with social anxiety disorder (Van Ameringen et al., 1996) and the addition of triiodothyronine for PTSD (Agid et al., 2001).

NEW TARGETS FOR DRUG DEVELOPMENT

A tremendous research effort is underway to discover drugs that target specific neuronal systems involved in mediating the stress response. By focusing on discrete neurobiological pathways, it is hoped that anxiolytic drugs will be discovered that normalize pathological activity in brain circuits and produce fewer adverse effects, including tolerance, dependence, and withdrawal effects (Nemeroff, 2003). Identification of multiple serotonin receptors has allowed investigation of compounds that act selectively at individual receptors and may be better tolerated than currently available SSRIs. Drugs with GABA-potentiating properties that are distinct from those of the benzodiazepines also are being studied as anxiolytics, as in the studies of the anticonvulsants described earlier. The GABA-A α2 subtype has been identified as a potential target for anxiolytic effects without sedation or dependence (Mohler et al., 2001).

Some of the most promising avenues of new drug development involve non-monoamine, non-GABA neurotransmitters. For example, substance P (tachykinin NK1), corticotropin-releasing factor (CRF), L-glutamate, and other neurotransmitters that are pivotal to the regulation of anxiety and stress responses may be ideal targets for new drug development. Reports from preclinical studies have shown that manipulation of these neuronal systems produces evidence of anxiolytic and antidepressant effects.

NEXT-GENERATION SSRIS

The SSRIs increase available serotonin in the synapse and lead to indiscriminate activity at multiple different serotonin receptors, some of which are believed to be associated with SSRI-related side effects. The observations that mice lacking 5-HT1A receptors exhibit pronounced anxious and fearful behaviors and do not respond to SSRIs (Gross et al., 2002) has led to studies of specific postsynaptic 5-HT1A receptor agonists as possible treatments for anxiety disorders (Gorman, 2003). A recent neurobiological substrate has been identified for treatment resistance in anxiety disorders to SSRIs. Subjects with generalized social anxiety disorder possessing the “short” arm of the serotonin transporter gene were significantly less likely to respond (40% non-responder rate) to maximally titrated SSRIs in comparison to subjects with the “long” arm of the serotonin transporter gene (Stein et al., 2006). Similar results for SSRI treatment resistance have been noted in panic disorder (Perna et al., 2005). It is not clear what the functional consequences of possession of the short versus long arm of the serotonin transporter imply, but these important observations may provide clues to the pharmacogenomic underpinnings of SSRI treatment resistance.

NEUROPEPTIDES

Neuropeptides are short-chain amino acids that act as neurotransmitters in regions of the brain involved with regulation of mood and stress responses. Recognition of a large number of neuropeptide targets for anxiety disorders has provided the rationale for studies of small-molecule neuropeptide receptor ligands. Some of the most promising neuropeptide targets for anxiolytic drug discovery are substance P, CRF-1, CRF-2, neuropeptide Y, CCK-2, and galanin (Herranz, 2003; Holmes et al., 2003). A cholecystokinin (CCK) antagonist has been studied in patients with panic disorder (Pande et al., 1999b) and GAD (Adams et al., 1995), but was not more effective than a placebo. A possible explanation for the failure of the CCK antagonist to demonstrate its efficacy may be related to its relatively poor penetration.

The substance P antagonists are being investigated as treatments for depression and anxiety disorders. In late 2003, further development of MK-869, a substance P (NK1) antagonist that was studied through phase III clinical trials of depression (Kramer et al., 1998), was halted because of a failure to demonstrate efficacy. A second NK1 antagonist, L759274, is being studied in depression (Kramer et al., 2004). Efficacy of the CRF-1 receptor antagonist, R121919, was suggested by the findings of a small, open-label study of patients with major depression (Zobel et al., 2000). However, development of R121919 is no longer being pursued because of concerns about hepatotoxicity. Clinical trials of other CRF-1 receptor antagonists are currently underway (Gorman, 2003).

OTHER MOLECULAR TARGETS

Other molecular targets are being investigated as avenues toward anxiolytic drug development. The excitatory neurotransmitter, L-glutamate, and its NMDA and metabotropic glutamate (mGlu) receptors are potential drug development targets. The non-competitive NMDA receptor antagonists (MK-801) and mGlu receptor agonists (LY354740) possess antidepressant and anxiolytic properties preclinically (Papp and Moryl, 1994; Schoepp et al., 2003), and the NMDA receptor antagonist, riluzole (Mathew et al., 2005) has been observed to exert anxiolytic effects in patients with anxiety disorders. In contrast, the NMDA partial agonist, DCS, failed to show efficacy in a small pilot study in patients with PTSD (Heresco-Levy et al., 2002), although, as noted, appeared to enhance the effect of CBT in the treatment of acrophobia and social anxiety disorder (Hofmann et al., 2006; Ressler et al., 2004). Brain-derived neurotrophic factor (BDNF), which is critical to neuronal plasticity, is inhibited by stress and has therefore been recognized as a potential target for drug development in mood and anxiety disorders (Hall et al., 2003; Rasmusson et al., 2002). Neurosteroid modulators of the GABA system, such as allopregnanolone, are being studied for anxiolytic effects (Lydiard, 2003). Other molecular targets of interest are the opioid receptor partial agonists (Dautzenberg et al., 2001) and cytokine modulators (Koh and Lee, 2004).

FUTURE DIRECTIONS

The most pressing research priorities for advancing the treatment of refractory anxiety disorders were identified. There is a remarkable lack of empirical data in this area. When data are available, it is often obtained from small controlled trials, open-label pilot studies, case reports, or chart reviews. Therefore, the following list of research priorities are evidence-based when possible, but also include suggestions that are based on the opinions and clinical experiences of the experts who participated in this conference.

RESEARCH PRIORITIES FOR PSYCHOTHERAPY AND PHARMACOTHERAPY TRIALS

•. 

Conduct RCTs focused on strategies for achieving remission rather than partial response: aggressive psychotherapy regimens (i.e., intensive treatment, prolonged session times) and/or aggressive pharmacotherapy regimens (i.e., adequate dosing and duration [e.g., 6 months versus 6 weeks]), and outcome assessments that are more sensitive than measures of global improvement.

•. 

Conduct prospective studies and post hoc analyses of existing psychotherapy and pharmacotherapy databases to define early predictors of response/ non-response that would identify modifiers of outcome and patients who would benefit from additional interventions, more prolonged treatment, or an alternative therapeutic approach.

•. 

Include functional outcome assessments (e.g., measures of avoidance) in all studies and differentiate these measures from quality of life assessments.

•. 

Conduct additional studies of the factors (e.g., context effects, safety behaviors, attentional factors) that may diminish the benefits of exposure treatments.

•. 

Conduct randomized, controlled psychotherapy augmentation trials, particularly the role of memory enhancers, to examine whether pharmacotherapy can have a significant role in improving extinction learning and the salience of adaptive behaviors learned in therapy

•. 

Conduct RCTs of pharmacologic augmentation therapy to validate the clinical signals suggested by open-label studies and case reports.

•. 

Conduct effectiveness studies in patients with common comorbidities (e.g., depression, alcohol/substance abuse), women (including issues surrounding pregnancy/postpartum, menstrual cycle, menopause), underserved ethnic minorities, medically ill patients, and the elderly. Examine the impact of treatment on extant symptomatology and course of illness in at-risk and manifestly anxious children and adolescents.

•. 

Design and evaluate treatment algorithms for patients failing initial interventions, including testing approaches for combining different treatments (e.g., medication and CBT) with particular attention to context effects.

•. 

Conduct investigations of strategies to better disseminate efficacious psychosocial and pharmacologic treatments.

RESEARCH PRIORITIES FOR NEW DRUG DEVELOPMENT

•. 

Conduct gene expression studies in animal models to identify possible targets for drug development (“bottom-up” approach).

•. 

Identify neurodevelopment of anxiety-relevant circuits and evaluate the effect of drugs at different stages of development.

•. 

Combine neuroimaging with other modes of inquiry (e.g., genotyping, neuroendocrinology, psychophysiology, psychotherapy, pharmacotherapy).

•. 

Conduct neuroimaging studies of treatment response and non-response, including studies of placebo response.

•. 

Conduct genotyping studies to identify treatment responders and risk factors for adverse effects.

•. 

Conduct RCTs of new classes of medications to treat anxiety disorders.

•. 

Conduct translational research to build on promising animal models for the reduction of fear and test these models in humans.

CONCLUSIONS

Anxiety disorders are common and are associated with significant distress and dysfunction. Currently available interventions have demonstrable efficacy for reducing anxiety-related symptomatology. However, many patients remain symptomatic despite treatment, and a significant proportion remain impaired. Despite this, few controlled studies address inadequate treatment response in anxiety disorders. The need for refined CBT strategies pales in comparison to the importance of disseminating CBT for anxiety disorders more generally, as these interventions are rarely received by patients in clinical practice (Goisman et al., 1999). These data join a larger literature indicating that in clinical care across the United States, few patients receive an adequate “dose” of psychotherapy (Hansen et al., 2002), a finding that is in concordance with the high rates of inadequate pharmacotherapy observed in primary care and specialty care settings (Roy-Byrne et al., 1999; Wang et al., 2000; Weilburg et al., 2003). Optimal use of available medications and the development of novel pharmacological strategies and agents along with the further application of empirically derived psychosocial therapies offer promise in improving this current situation, but a concerted research effort is warranted to systematically evaluate potential areas of importance and prudently use scarce resources. This article summarizes the current state of knowledge and serves as a call to action to mobilize efforts aimed at improving the lives of the many individuals who are adversely impacted by anxiety disorders.

APPENDIX

The Anxiety Disorders Association of America Novel Approaches to Treatment of Refractory Anxiety Disorders conference Chair: Mark H. Pollack, MD.

WORKGROUP 1—PSYCHOTHERAPY

Michael W. Otto, PhD, Massachusetts General Hospital (currently at Boston University); Barbara O. Rothbaum, PhD, Emory University (Workgroup Leaders); Bruce Cuthbert, PhD, National Institutes of Mental Health; Edna Foa, PhD, University of Pennsylvania; Richard Heimberg, PhD, Temple University; Stefan Hoffman, PhD, Boston University; Terry Keane, PhD, National Center for PTSD; Jerilyn Ross, MA, LICSW, Ross Center for Anxiety and Related Disorders; Kerry Ressler, MD, PhD, Emory University; Sabine Wilhelm, MD Massachusetts General Hospital; Doug Mennin, PhD, Yale University

WORKGROUP 2— PSYCHOPHARMACOLOGY

Mark H. Pollack MD, Massachusetts General Hospital (Workgroup Leader); David Katzelnick, MD, Healthcare Technology Systems, Inc.; Barry Leibowitz, PhD, National Institutes of Mental Health; Bruce Lydiard, MD, Southeast Health Consultants; Phil Ninan, MD, Emory University; Michael Van Ameringen, MD, McMaster University; Peter RoyByrne, MD, University of Washington; Beth Salcedo, MD, Ross Center; Kathy Shear, MD, University of Pittsburgh; Michael Johnson, MD, University of Florida; Naomi Simon, MD, Massachusetts General Hospital; Amy Wagner, MD, University of Washington.

WORKGROUP 3— NOVEL BIOLOGICAL APPROACHES

Jack Gorman, MD, Mount Sinai School of Medicine; Jeremy Coplan, MD, Downstate Medical Center (Workgroup Leaders); Dennis Charney, MD, Mount Sinai School of Medicine; Joe LeDoux, PhD, New York University; Israel Liberzon, MD, University of Michigan; Paul Plotsky, PhD, Emory. Scott Rauch, MD, Massachusetts General Hospital; David Silbersweig, MD, Weill Cornell Medical College; Rachel Yehuda, PhD, Mt. Sinai School of Medicine; Michael Owens, PhD, Emory University; Justine Kent, MD, Columbia University; Elizabeth Hoge, MD, Massachusetts General Hospital; Sanjay Mathew, MD, Columbia University; Ruth Lanius, MD, PhD, Western Ontario.

REFERENCES

Adams JB, Pyke RE, Costa J, Cutler NR, Schweizer E, Wilcox CS, Wisselink PG, Greiner M, Pierce MW, Pande AC. 1995. A double-blind, placebo-controlled study of a CCK-B receptor antagonist, CI-988, in patients with generalized anxiety disorder. J Clin Psychopharmacol 15: 428– 434.CrossrefGoogle Scholar

Agid O, Shalev AY, Lerer B. 2001. Triiodothyronine augmentation of selective serotonin reuptake inhibitors in posttraumatic stress disorder. J Clin Psychiatry 62: 169– 173.CrossrefGoogle Scholar

Ballenger JC. 2001. Treatment of anxiety disorders to remission. J Clin Psychiatry 62: 5– 9.Google Scholar

Ballenger JC. 1999. Current treatment of the anxiety disorders in adults. Biol Psychiatry 46: 1579– 1594.CrossrefGoogle Scholar

Barlow DH. 2001. Clinical handbook of psychological disorders: a step-by-step treatment manual (3rd ed.). New York: Guilford Press.Google Scholar

Barlow DH, Gorman JM, Shear MK, Woods SW. 2000. Cognitive-behavioral therapy, imipramine, or their combination for panic disorder: a randomized controlled trial. JAMA 283: 2529– 2536.CrossrefGoogle Scholar

Barnett SD, Kramer ML, Casat CD, Connor KM, Davidson JRT. 2002. Efficacy of olanzapine in social anxiety disorder: a pilot study. J Psychopharmacol 16: 365– 368.CrossrefGoogle Scholar

Bartzokis G, Lu PH, Turner J, Mintz J, Saunders CS. 2005. Adjunctive risperidone in the treatment of chronic combat-related posttraumatic stress disorder. Biol Psychiatry 57: 474– 479.CrossrefGoogle Scholar

Berigan T. 2002. Treatment of posttraumatic stress disorder with tiagabine. Can J Psychiatry 47:788.Google Scholar

Berlant J, van Kammen DP. 2002. Open-label topiramate as primary or adjunctive therapy in chronic civilian posttraumatic stress disorder: a preliminary report. J Clin Psychiatry 63: 15– 20.CrossrefGoogle Scholar

Bouton ME. 2002. Context, ambiguity, and unlearning: sources of relapse after behavioral extinction. Biol Psychiatry 52: 976– 986.CrossrefGoogle Scholar

Brannon N, Labbate L, Huber M. 2000. Gabapentin treatment for posttraumatic stress disorder. Can J Psychiatry 45:84.CrossrefGoogle Scholar

Brawman-Mintzer O, Knapp R, Nietert P. 2005. Adjunctive risperidone in generalized anxiety disorder: a double-blind, placebo-controlled study. J Clin Psychiatry 66: 1321– 1325.CrossrefGoogle Scholar

Bremner D, Mletzko T, Welter S, Siddiq S, Reed L, Williams C, Heim CM, Nemeroff CB. 2004. Treatment of posttraumatic stress disorder with phenytoin: an open-label pilot study. J Clin Psychiatry 65: 1559– 1564.CrossrefGoogle Scholar

Crane D. 2003. Tiagabine for the treatment of anxiety. Depress Anxiety 18: 51– 52.CrossrefGoogle Scholar

Dautzenberg FM, Wichmann J, Higelin J, Py-Lang G, Kratzeisen C, Malherbe P, Kilpatrick GJ, Jenck F. 2001. Pharmacological characterization of the novel nonpeptide orphanin FQ/nociceptin receptor agonist Ro 64–6198: rapid and reversible desensitization of the ORL1 receptor in vitro and lack of tolerance in vivo. J Pharmacol Exp Ther 298: 812– 819.Google Scholar

Etxebeste M, Aragüés E, Malo P, Pacheco L. 2000. Olanzapine and panic attacks. Am J Psychiatry 157: 659– 670.CrossrefGoogle Scholar

Fava GA, Savron G, Zielezny M, Grandi S, Rafanelli C, Conti S. 1997. Overcoming resistance to exposure in panic disorder with agoraphobia. Acta Psychiatrica Scand 95: 306– 312.CrossrefGoogle Scholar

Feltner DE, Pollack MH, Davidson JRT, Stein MB, Futterer R, Jefferson JW, Lydiard RB, DuBoff E, Robinson P, Phelps M, Slomkowski M, Werth JL, Pande AC. 2000. A placebo-controlled, double-blind study of pregabalin treatment of social phobia: outcome and predictors of response. J Eur Coll Neuropsycho-pharmacol 10: S345.Google Scholar

Foa EB, Franklin ME, Moser J. 2002. Context in the clinic: how well do cognitive-behavioral therapies and medications work in combination? Biol Psychiatry 10: 987– 997.CrossrefGoogle Scholar

Goisman RM, Warshaw MG, Keller MB. 1999. Psychosocial treatment prescriptions for generalized anxiety disorder, panic disorder, and social phobia, 1991–1996. Am J Psychiatry 156: 1819– 1821.Google Scholar

Gorman JM. 2003. New molecular targets for antianxiety interventions. J Clin Psychiatry 64: 28– 35.Google Scholar

Gross C, Zhuang X, Stark K, Ramboz S, Oosting R, Kirby L, Santarelli L, Beck S, Hen R. 2002. Serotonin 1A receptor acts during development to establish normal anxiety-like behaviour in the adult. Nature 416: 396– 400.CrossrefGoogle Scholar

Hall D, Dhilla A, Charalambous A, Gogos JA, Karayiorgou M. 2003. Sequence variants of the brain-derived neurotrophic factor (BDNF) gene are strongly associated with obsessive-compulsive disorder. Am J Hum Genet 73: 370– 376.CrossrefGoogle Scholar

Hamner MB, Faldowski RA, Ulmer HG, Frueh BC, Huber MG, Arana GW. 2003. Adjunctive risperidone treatment in post-traumatic stress disorder: a preliminary controlled trial of effects on comorbid psychotic symptoms. Int Clin Psychopharmacol 18: 1– 8.CrossrefGoogle Scholar

Hansen NB, Lambert MJ, Forman EM. 2002. The psychotherapy dose-response effect and its implications for treatment delivery Services. Clin Psychol Sci Pract 9: 329– 343.CrossrefGoogle Scholar

Heldt E, Manfro GG, Kipper L, Blaya C, Isolan L, Otto MW. 2006. One-year follow-up of pharmacotherapy-resistant patients with panic disorder treated with cognitive-behavior therapy: outcome and predictors of remission. Behav Res Therapy 44: 657– 665.CrossrefGoogle Scholar

Heresco-Levy U, Kremer I, Javitt DC, Goichman R, Reshef A, Blanaru M, Cohen T. 2002. Pilot-controlled trial of D-cycloserine for the treatment of post-traumatic stress disorder. Int J Neuropsychopharmacol 5: 301– 307.CrossrefGoogle Scholar

Herranz R. 2003. Cholecystokinin antagonists: pharmacological and therapeutic potential. Med Res Rev 23: 559– 605.CrossrefGoogle Scholar

Hertzberg MA, Butterfield MI, Feldman ME, Beckham JC, Sutherland SM, Connor KM, Davidson JR. 1999. A preliminary study of lamotrigine for the treatment of posttraumatic stress disorder. Biol Psychiatry 45: 1226– 1229.CrossrefGoogle Scholar

Hirschmann S, Dannon PN, Iancu I, Dolberg OT, Zohar J, Grunhaus L. 2000. Pindolol augmentation in patients with treatment-resistant panic disorder: a double-blind, placebo-controlled trial. J Clin Psychopharmacol 20: 556– 559.CrossrefGoogle Scholar

Hofmann SG, Meuret AE, Smits JAJ, Simon NM, Pollack MH, Eisenmenger K, Shiekh M, Otto MW. 2006. Augmentation of exposure therapy for social anxiety disorder with D-Cycloserine. Arch Gen Psychiatry 63: 298– 304.CrossrefGoogle Scholar

Hollander E, Zohar J. 2004. Beyond refractory obsessions and anxiety states; toward remission. J Clin Psychiatry 65: 3– 5.Google Scholar

Holmes A, Heilig M, Rupniak NMJ, Steckler T, Griebel G. 2003. Neuropeptide systems as novel therapeutic targets for depression and anxiety disorders. Trends Pharmacol Sci 24: 580– 588.CrossrefGoogle Scholar

Kampman M, Keijsers GP, Hoogduin CA, Hendriks GJ. 2002. A randomized, double-blind, placebo-controlled study of the effects of adjunctive paroxetine in panic disorder patients unsuccessfully treated with cognitive-behavioral therapy alone. J Clin Psychiatry 63: 772– 777.CrossrefGoogle Scholar

Khaldi S, Kornreich C, Dan B, Pelc I. 2003. Usefulness of olanzapine in refractory panic attacks. J Clin Psychopharmacol 23: 100– 101.CrossrefGoogle Scholar

Kinrys G, Pollack MH, Simon NM, Worthington JJ, Nardi AE, Versiani M. 2003. Valproic acid for the treatment of social anxiety disorder. Int Clin Psychopharmacol 18: 169– 172.Google Scholar

Kinrys G, Wygant LE, Pardo TB, Melo M. 2006. Levetiracetam for treatment-refractory posttraumatic stress disorder. J Clin Psychiatry. 67: 211– 214.CrossrefGoogle Scholar

Koh KB, Lee Y. 2004. Reduced anxiety level by therapeutic interventions and cell-mediated immunity in panic disorder patients. Psychother Psychosom 73: 286– 292.CrossrefGoogle Scholar

Kramer MS, Winokur A, Kelsey J, Preskorn SH, Rothschild AJ, Snavely D, Ghosh K, Ball WA, Reines SA, Munjack D, Apter JT, Cunningham L, Kling M, Bari M, Getson A, Lee Y. 2004. Demonstration of the efficacy and safety of a novel substance P (NK1) receptor antagonist in major depression. Neuropsychopharmacol 29: 385– 392.CrossrefGoogle Scholar

Kramer MS, Cutler N, Feighner J, Shrivastava R, Carman J, Sramek JJ, Reines SA, Liu G, Snavely D, Wyatt-Knowles E, Hale JJ, Mills SG, MacCoss M, Swain CJ, Harrison T, Hill RG, Hefti F, Scolnick EM, Cascieri MA, Chicchi GG, Sadowski S, Williams AR, Hewson L, Smith D, Carlson EJ, Hargreaves RJ, Rupniak NM. 1998. Distinct mechanism for antidepressant activity by blockade of central substance P receptors. Science 281: 1640– 1645.CrossrefGoogle Scholar

Kushner MG, Kim SW, Donahue C, Thuras P, Adson D, Kotlyar M, McCabe J, Peterson J, Foa EB. in press. D-cycloserine augmented exposure therapy for obsessive compulsive disorder.Google Scholar

Liebowitz MR, Heimberg RG, Schneier FR, Hope DA, Davies S, Holt CS, Goetz D, Juster HR, Lin SH, Bruch MA, Marshall RD, Klein DF. 1999. Cognitive-behavioral group therapy versus phenelzine in social phobia: long-term outcome. Depress Anxiety 10: 89– 98.CrossrefGoogle Scholar

Londborg PD, Hegel MT, Goldstein S, Goldstein D, Himmelhoch JM, Maddock R, Patterson WM, Rausch J, Farfel GM. 2001. Sertraline treatment of posttraumatic stress disorder: results of 24 weeks of open-label continuation treatment. J Clin Psychiatry 62: 325– 331.CrossrefGoogle Scholar

Lydiard RB. 2003. The role of GABA in anxiety disorders. J Clin Psychiatry 64: 21– 27.Google Scholar

Mathew SJ, Amiel MJ, Coplan JD, Fitterling HA, Sackeim HA, Gorman JA. 2005. Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry 162: 2379– 2381.CrossrefGoogle Scholar

McHugh RK, Otto MW, Barlow DH, Gorman JM, Shear MK, Woods SW. in press. Cost-efficacy of individual and combined treatments of panic disorder. J Clin Psychiatry; anxiety disorders. Am J Psychiatry 155: 992– 993.CrossrefGoogle Scholar

Mohler H, Crestani F, Rudolph U. 2001. GABA(A)-receptor subtypes: a new pharmacology. Curr Opin Pharmacol 1: 22– 25.CrossrefGoogle Scholar

Monnelly EP, Ciraulo DA, Knapp C, Keane T. 2003. Low-dose risperidone as adjunctive therapy for irritable aggression in posttraumatic stress disorder. J Clin Psychopharmacol 23: 193– 196.CrossrefGoogle Scholar

Mystkowski JL, Mineka S, Vernon LL, Zinbarg RE. 2003. Changes in caffeine states enhance return of fear in spider phobia. J Consult Clin Psychol 71: 243– 250.CrossrefGoogle Scholar

Nemeroff CB. 2003. Anxiolytics: past, present, and future agents. J Clin Psychiatry 64: 3– 6.CrossrefGoogle Scholar

Ontiveros A, Fontaine R. 1992. Sodium valproate and clonazepam for treatment-resistant panic disorder. J Psychiatry Neurosci 17: 78– 80.Google Scholar

Otto MW, Smits JA, Reese HE. 2005. Combination psychotherapy and pharmacotherapy for mood and anxiety disorders in adults: review and analysis. Clin Psychol Sci Pract 12: 72– 86.CrossrefGoogle Scholar

Otto MW, Smits JA, Reese HE. 2004. Cognitive-behavioral therapy for the treatment of anxiety disorders. J Clin Psychiatry 65: 34– 41.Google Scholar

Otto MW, Hinton D, Korbly NB, Chea A, Phalnarith B, Gershuny BS, Pollack MH. 2003. Treatment of pharmacotherapy-refractory posttraumatic stress disorder among Cambodian refugees: a pilot study of combination treatment with cognitive-behavior therapy vs. sertraline alone. Behav Res Ther 41: 1271– 1276.CrossrefGoogle Scholar

Otto MW, Hong JJ, Safren SA. 2002. Benzodiazepine discontinuation difficulties in panic disorder: conceptual model and outcome for cognitive-behavior therapy. Curr Pharm Des 8: 75– 80.CrossrefGoogle Scholar

Otto MW, Pollack MH, Penava SJ, Zucker BG. 1999. Cognitivebehavior therapy for patients failing to respond to pharmacotherapy for panic disorder: a clinical case series. Behav Res Ther 37: 763– 770.CrossrefGoogle Scholar

Pande AC, Davidson JRT, Jefferson JW, Janney CA, Katzelnick DJ, Weisler RH, Greist JH, Sutherland SM. 1999a. Treatment of social phobia with gabapentin: a placebo-controlled study. J Clin Psychopharmacol 19: 341– 348.CrossrefGoogle Scholar

Pande AC, Greiner M, Adams JB, Lydiard RB, Pierce MW. 1999b. Placebo-controlled trial of the CCK-B antagonist, CI-988, in panic disorder. Biol Psychiatry 46: 860– 862.CrossrefGoogle Scholar

Papp M, Moryl E. 1994. Antidepressant activity of non-competitive and competitive NMDA receptor antagonists in a chronic mild stress model of depression. Eur J Pharmacol 263: 1– 7.CrossrefGoogle Scholar

Perna G, Favaron E, Di Bella D, Bussi R, Bellodi L. 2005. Antipanic efficacy of paroxetine polymorphism within the promoter of the serotonin transporter gene. Neuropsychopharmacology 30: 2230– 2235.CrossrefGoogle Scholar

Pollack M. 2002. Levetiracetam (Keppra) for anxiety. Curbside Consultant 1:4.Google Scholar

Pollack MH, Simon NM, Zalta AK, Worthington J, Hoge EA, Mick E, Kinrys G, Oppenheimer J. 2006. Olanzapine augmentation of fluoxetine for refractory generalized anxiety disorder: a placebo controlled study. Biol Psychiatry 59: 211– 215.CrossrefGoogle Scholar

Pollack MH, Matthews J, Scott EL. 1998. Gabapentin as a potential treatment for anxiety disorders. Am J Psychiatry 155: 992– 993.CrossrefGoogle Scholar

Powers MB, Smits JAJ, Leyro TM, Otto M. 2006. Translational research perspectives on maximizing the effectiveness of exposure therapy. In: Richard DCS, Lauterbach DL, editors. Comprehensive handbook of exposure therapies. Boston: Academic Press. p 109– 126.Google Scholar

Powers MB, Smits JA, Telch MJ. 2004. Disentangling the effects of safety-behavior utilization and safety-behavior availability during exposure-based treatment: a placebo-controlled trial. J Consult Clin Psychol. 72: 448– 454.CrossrefGoogle Scholar

Raskind MA, Peskind ER, Kanter ED, Petrie EC, Radant A, Thompson CE, Dobie DJ, Hoff D, Rein RJ, Straits-Troster K, Thomas RG, McFall MM. 2003. Reduction of nightmares and other PTSD symptoms in combat veterans by prazosin: a placebo-controlled study. Am J Psychiatry 160: 371– 373.CrossrefGoogle Scholar

Rasmusson AM, Shi L, Duman R. 2002. Downregulation of BDNF mRNA in the hippocampal dentate gyrus after re-exposure to cues previously associated with footshock. Neuropsychopharmacol 27: 133– 142.CrossrefGoogle Scholar

Ressler KJ, Rothbaum BO, Tannenbaum L, Anderson P, Graap K, Zimand E, Hodges L, Davis M. 2004. Cognitive enhancers as adjuncts to psychotherapy: use of D-cycloserine in phobic individuals to facilitate extinction of fear. Arch Gen Psychiatry 61: 1136– 1144.CrossrefGoogle Scholar

Rickels K, Pollack MH, Feltner DE, Lydiard RB, Zimbroff DL, Bielski RJ, Tobias K, Brock JD, Zornberg GL, Pande AC. 2005. Pregabalin for treatment of generalized anxiety disorder: a 4-week, multicenter, double-blind, placebo-controlled trial of pregabalin and alprazolam. Arch Gen Psychiatry 62: 1022– 1030.CrossrefGoogle Scholar

Roy-Byrne P, Cowley DS. 1998. Clinical approach to treatment-resistant panic disorder. In: Rosenbaum JF, Pollack MH, editors. Panic disorder and its treatment. New York: Marcel Dekker. p 205– 227.Google Scholar

Roy-Byrne PP, Stein MB, Russo J, Mercier E, Thomas R, McQuaid J, Katon WJ, Craske MG, Bystritsky A, Sherbourne CD. 1999. Panic disorder in the primary care setting: comorbidity, disability, service utilization, and treatment. J Clin Psychiatry 60: 492– 499.CrossrefGoogle Scholar

Salkovskis PM, Clark DM, Hackmann A, Wells A, Gelder MG. 1999. An experimental investigation of the role of safety-seeking behaviours in the maintenance of panic disorder with agoraphobia. Behav Res Ther 37: 559– 574.CrossrefGoogle Scholar

Schmidt NB, Wollaway-Bickel K, Trakowski JH, Santiago HT, Vasey M. 2002. Antidepressant discontinuation in the context of cognitive behavioral treatment for panic disorder. Behav Res Ther 40: 67– 73.CrossrefGoogle Scholar

Schoepp DD, Wright RA, Levine LR, Gaydos B, Potter WZ. 2003. LY354740, an mGlu2/3 receptor agonist as a novel approach to treat anxiety/stress. Stress 6: 189– 197.CrossrefGoogle Scholar

Shaywitz JE, Liebowitz MR. 2003. Antiepileptic treatment of anxiety disorders. Prim Psychiatry 51– 56.Google Scholar

Simon NM, Worthington JJ, Doyle AC, Hoge EA, Kinrys G, Fischmann D, Link N, Pollack MH. 2004. An open-label study of levetiracetam for the treatment of social anxiety disorder. J Clin Psychiatry 65: 1219– 1222.CrossrefGoogle Scholar

Smits JA, Powers MB, Buxkamper R, Telch MJ. 2006. The efficacy of videotape feedback for enhancing the effects of exposure-based treatment for social anxiety disorder: a controlled investigation. -Behav Res Ther. 44: 1773– 1785.CrossrefGoogle Scholar

Stein MB, Kline NA, Matloff JL. 2002. Adjunctive olanzapine for SSRI-resistant combat-related PTSD: a double-blind, placebo-controlled study. Am J Psychiatry 159: 1777– 1779.CrossrefGoogle Scholar

Stein MB, Sareen J, Hami S, Chao J. 2001. Pindolol potentiation of paroxetine for generalized social phobia: a double-blind, placebo-controlled, crossover study. Am J Psychiatry 158: 1725– 1727.CrossrefGoogle Scholar

Stein MB, Seedat S, Gelernter J. 2006. Serotonin transporter gene promoter polymorphism predicts SSRI response in generalized social anxiety disorder. Psychopharmacology (Berl) 187: 68– 72.CrossrefGoogle Scholar

Szymanski HV, Olympia J. 1991. Divalproex in posttraumatic stress disorder. Am J Psychiatry 148: 1086– 1087.Google Scholar

Tiffon L, Coplan JD, Papp LA, Gorman JM. 1994. Augmentation strategies with tricyclic or fluoxetine treatment in seven partially responsive panic disorder patients. J Clin Psychiatry 55: 66– 69.Google Scholar

Van Ameringen M, Mancini C, Pipe B, Oakman J, Bennett M. 2004. An open-label trial of topiramate in the treatment of generalized social phobia. J Clin Psychiatry 65: 1674– 1678.CrossrefGoogle Scholar

Van Ameringen M, Mancini C, Wilson C. 1996. Buspirone augmentation of selective serotonin reuptake inhibitors (SSRIs) in social phobia. J Affect Disord. 39: 115– 121.CrossrefGoogle Scholar

Walker DL, Ressler KJ, Lu KT, Davis M. 2002. Facilitation of conditioned fear extinction by systemic adminstration of intra-amygdala infusions of D-cycloserine as assessed with fear-potentiated startle in rats. J Neurosci 22: 2343– 2351.CrossrefGoogle Scholar

Wang PS, Berglund P, Kessler RC. 2000. Recent care of common mental disorders in the United States: prevalence and conformance with evidence-based recommendations. J Gen Intern Med 15: 284– 292.CrossrefGoogle Scholar

Weilburg JB, O'Leary KM, Meigs JB, Hennen J, Stafford RS. 2003. Evaluation of the adequacy of outpatient antidepressant treatment. Psychiatr Serv 54: 1233– 1239.CrossrefGoogle Scholar

Zobel AW, Nickel T, Kunzel HE, Ackl N, Sonntag A, Ising M, Holsboer F. 2000. Effects of the high-affinity corticotropin-releasing hormone receptor 1 antagonist R121919 in major depression: the first 20 patients treated. J Psychiatr Res 34: 171– 181.CrossrefGoogle Scholar

Zwanzger P, Baghai TC, Schüle C, Minov C, Padberg F, Möller H-J, Rupprecht R. 2001. Tiagabine improves panic and agoraphobia in panic disorder patients. J Clin Psychiatry 62: 656– 657.CrossrefGoogle Scholar