Sei sulla pagina 1di 14

human psychopharmacology

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. Published online in Wiley InterScience (www.interscience.wiley.com) DOI: 10.1002/hup.1074

REVIEW ARTICLE

Serotonin norepinephrine reuptake inhibitors (SNRIs) in anxiety disorders: a comprehensive review of their clinical efcacy
Bernardo DellOsso1*, Massimiliano Buoli1, David S. Baldwin2 and A. Carlo Altamura1
1

Department of Psychiatry, University of Milan, Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via F. Sforza 35, Milano, Italy 2 Clinical Neuroscience Division, School of Medicine, University of Southampton, UK

Anxiety disorders are common psychiatric conditions that typically require long-term treatment. This review summarizes current knowledge of the pharmacological treatment of anxiety disorders with serotonin norepinephrine reuptake inhibitors (SNRIs) with specic emphasis on the ndings of recent randomized clinical trials and relevant neurobiological investigations. It is now well established that gabaergic, noradrenergic and serotonergic systems play a critical role in the pathophysiology of anxiety disorders, abnormalities in these systems being related to structural and functional alterations in specic brain areas such as the amygdala, prefrontal cortex, locus coeruleus and hippocampus, as repeatedly shown by neuroimaging studies. SNRIs selectively inhibit norepinephrine and serotonin reuptake and have shown to be efcacious and generally well tolerated treatments in patients with anxiety disorders, with some potential clinical advantages over selective serotonin reuptake inhibitors (SSRIs), which are considered by many to represent rst-line pharmacological treatments in patients with anxiety disorders. Anxiety disorders are characterized by a typically chronic course, high rates of comorbidity and frequent partial response to standard treatments, and the increasing use of SNRIs reects currently unmet clinical need, in terms of overall response, remission rates and treatment tolerability. Copyright # 2009 John Wiley & Sons, Ltd. key words serotonin norepinephrine reuptake inhibitors (SNRIs); anxiety disorders (ADs); selective serotonin reuptake inhibitors (SSRIs)

INTRODUCTION Epidemiologic data and general clinical aspects of ADs Anxiety disorders are the most common mental disorders and are characterized by the presence of impairing anxiety symptoms that vary widely in terms of their nature, severity, frequency, persistence and consequences. According to the Diagnostic and Statistical Manual of Mental Disorders (DSM-IVTR, American Psychiatric Association, 2000), this group of disorders includes panic disorder (PD), generalized anxiety disorder (GAD), specic (simple) phobia, social anxiety disorder (SAD, also known as social phobia), post-traumatic stress disorder (PTSD) and obsessive compulsive disorder (OCD). The anxiety disorders are extremely prevalent conditions, with a lifetime prevalence in Europe of
* Correspondence to: B. DellOsso, Department of Psychiatry, University of Milan, Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Via F. Sforza 35, 20122, Milano, Italy. Fax: 39-0250320310. E-mail: bernardo.dellosso@mssm.edu

approximately 13.621% in the general population (Alonso et al., 2004; Wittchen and Jacobi, 2005). According to the Epidemiologic Catchment Area (ECA) study, 6% of men and 13% of women in the United States full criteria for an anxiety disorder in a 6-month observation period (Leon et al., 1995). The lifetime prevalence of individual disorders varies widelyPD, 1.53.5%; SAD, 2.413.3%; GAD, 4.16.6%; PTSD, 1.37% and OCD, 0.78.0% (Fontenelle et al., 2006; Keane et al., 2006; Kessler et al., 1994; Lepine, 2002; Lieb et al., 2005; Perkonigg et al., 2000; Wittchen and Hoyer, 2001). It is difcult to provide reliable prevalence rates for specic phobia, as the majority of affected patients do not present with these problems to health care services; however, in the National Comorbidity Study, almost 50% of respondents reported a lifetime occurrence of an unreasonably strong fear of one or more phobic stimuli (Kessler et al., 1994). Taken together, these prevalence data demonstrate that anxiety disorders represent one of the most prevalent and disabling groups of mental disorders, with a substantial social and economic
Received 22 December 2008 Accepted 2 September 2009

Copyright # 2009 John Wiley & Sons, Ltd.

18

b. dellosso

ET AL.

impact on affected people, their families and wider society (Kroenke et al., 2007). There is a considerable degree of overlap between anxiety disorders (Body et al., 1984; Kessler et al., 1994), the occurrence of one disorder increasing the probability of having another. Approximately 75% of patients with an anxiety disorder will also meet diagnostic criteria for at least another psychiatric condition (De Girolamo et al., 2006). In the large majority of patients affected by co-morbid disorders in which one condition is anxiety disorder, there is good evidence that the anxiety disorder usually appears rst (Kessler et al., 1996; Regier et al., 1998). Although there are important differences in the clinical course of the various anxiety disorders (Yonkers et al., 2003), most are long-term conditions with either an episodic course, with alternating phases of ill-health and relative well-being, or chronic conditions waxing and waning in severity but without full remission of symptoms. The aims of treatment, therefore, are both the resolution of acute symptoms and the prevention of relapse (Angst et al., 2009). However, despite the high prevalence, morbidity, comorbidity and chronic course, fewer than 30% of affected patients in Europe present with anxiety symptoms to healthcare services (Alonso et al., 2007), and furthermore even fewer patients receive evidence-based treatment. Unmet clinical needs in the pharmacological treatment of ADs Several characteristics are required for an optimal anxiolytic treatment, including its effectiveness across a wide range of anxiety symptoms, the capacity to achieve remission and prevent future recurrences, and a favourable tolerability prole in terms of few side effects, few interactions and no discontinuation symptoms (Baldwin, 2006). Although effective in reducing anxiety symptoms in some patients, tricyclic antidepressants are not recommended in long-term treatment, being associated with more side effects when compared to selective serotonin reuptake inhibitors (SSRIs) (Hirschfeld, 2000). In most recent evidence-based guidelines, SSRIs are regarded as the rst-line pharmacological treatment for anxiety disorders (Allgulander et al., 2003; Baldwin et al., 2005; Bandelow et al., 2002; Koran et al., 2007; Ursano et al., 2004). With over 20 years of clinical experience practice and clear evidence of efcacy from randomized placebo-controlled trials, SSRIs have clear advantages over tricyclics and monoamine oxidase inhibitors (MAOIs), at least in
Copyright # 2009 John Wiley & Sons, Ltd.

initial treatment. Nevertheless, many patients undergoing SSRI treatment develop troublesome side effects such as sexual dysfunction (Baldwin, 2004) and weight gain (Papakostas, 2008) and these may represent a major problem in terms of adherence, particularly over long-term treatment. Benzodiazepines are still often used in clinical practice to reduce acute anxiety symptoms but are not recommended for long-term treatment in patients other than those with the most severe and disabling disorders (Argyropoulos and Nutt, 1999; Baldwin et al., 2005). Benzodiazepines may be more effective in relieving somatic symptoms of anxiety than some antidepressant drugs, but less effective in reducing psychological symptoms, and are often associated with side effects such as sedation and cognitive impairment (Shader and Greenblatt, 1993). Another hazard associated with use of benzodiazepines is the development of tolerance, dependence and a withdrawal syndrome, which is more frequent after long times use at higher doses and when the drugs are abruptly discontinued (Shader and Greenblatt, 1993). There is also an increasing body of evidence suggesting that some anticonvulsants, in particular pregabalin, have anxiolytic potential and might therefore be an alternative treatment option in some anxiety syndromes (Baldwin and Ajel, 2007; Mula et al., 2007; Van Ameringen et al., 2004; Zwanzger et al., 2007). The serotonin norepinephrine reuptake inhibitors (SNRIs) are dual action antidepressants that inhibit the reuptake of both serotonin (5-hydroxytryptamine) and norepinephrine (noradrenaline). SNRIs are a useful alternative to SSRIs and are often used in patients with anxiety disorders, following a partial response or nonresponse to SSRI treatment (Allgulander et al., 2001) (Table 1.). This reviews summarizes the main neurochemical and neuroimaging data related to the pathophysiology of anxiety disorders and the rationale for the use of SNRIs with particular emphasis on their pharmacological properties. The ndings of randomized controlled trials assessing efcacy and tolerability of SNRI treatment are presented and discussed along with consideration of the potential advantages for SNRIs over SSRIs in the treatment of anxiety disorders. NEUROBIOLOGICAL BASES OF ANXIETY DISORDERS Neurochemical aspects of anxiety disorders Investigations have evaluated the role of disturbances of different neurotransmitter systems in the pathogenHum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders


Table 1. FDA approved drugs for the treatment of anxiety disorders Panic disorder Benzodiazepines Alprazolam SSRIs Fluoxetine Paroxetine Sertraline Generalized Anxiety Disorder SNRIs Duloxetine Venlafaxine SNRIs Venlafaxine

19

SSRIs Paroxetine Sertraline Escitalopram Social Anxiety Disorder

Anticonvulsants Pregabalin

SSRIs Paroxetine Sertraline Post-Traumatic Stress Disorder SSRIs Paroxetine Sertraline Obsessive-Compulsive Disorder Tryciclics Clomipramine SSRIs Fluoxetine Fluvoxamine Sertraline

esis of ADs, in particular in the limbic system, including the amygdala and other structures (Davis, 1997; Garakani et al., 2006; LeDoux, 1998). Research has focused principally on the role of GABAA/BZD complex, the norepinephrine and serotonin systems (Gorman et al., 2002). GABAA receptors are thought to modulate anxiety responses through projections to limbic areas with a resultant decrease in turnover of monoamines, ` and to the locus coeruleus and raphe nuclei with suppression of neuronal ring (Kardos, 1999). In particular, it has been hypothesized that downregulation of the GABAA-benzodiazepine receptor in anxiety disorders would result in a decreased function of endogeneous neurotransmitters and the expression of characteristic symptoms (Argyropoulos and Nutt, 2003). The norepinephric neurons of the locus coeruleus give rise to diffuse projections to the forebrain, and may play a critical role in mediating response to fear, stress and arousal (Bremner et al., 1996). The central effects of norephinephrine are mediated through postsynaptic a1 and b1 receptors and presynaptic a2 receptors which are important in mediating the presynaptic inhibition of norephinephrine release (autoreceptors) and the release of other neurotransmitters, when located on the terminals of nonnoradrenergic neurons. The a2-adrenergic receptor
Copyright # 2009 John Wiley & Sons, Ltd.

antagonist yohimbine increases the ring of noradrenergic cell bodies in the locus coeruleus and induces anxiety, whereas agents that reduce the ring of these neurons, such as the a2 receptor agonist clonidine reduce symptoms of anxiety (Grimsley, 1995). ` Serotonergic neurons located in the raphe nuclei project to large areas of the brain, including the limbic system and hypothalamus and are integrally involved in the mediation of anxiety responses (Ressler and Nemeroff, 2000). Presynaptic 5-HT1 receptors and postsynaptic 5-HT2 receptors are principally involved in the modulation of anxiety (Salzman et al., 1993). Stimulation of terminal 5-HT1 autoreceptors attenuates the release of serotonin at the nerve ending (Stahl, 1998). Studies of the plasma concentrations of serotonin and its metabolites and receptors have suggested a dysfunction of the serotonergic system; for example, levels of 5-HT in the cerebrospinal uid were reported to be low in patients with anxiety disorders (Johnson and Lydiard, 1995) and administration of m-chlorophenylpiperazine, a non-selective 5HT1 and 5HT2 agonist, leads to an increase of hostility and anxiety in patients with GAD (Germine et al., 1992). It has been hypothesized that SSRIs may act through reducing central 5-HT transmission, suggesting there may be a state of increased serotonergic function in anxiety disorders. However, two major serotonergic systemsone originating from ` the medial raphe nuclei, the other from the dorsal raphe nucleihave been implicated in anxiety, it being hypothesized that each system mediates different aspects of anxiety, and that dysfunction of one or both of these systems would result in differing forms of anxiety disorders (Deakin and Graeff, 1991; Bell and Nutt, 1998). While gabaergic, noradrenergic and serotonergic systems still represent the main focus of attention in relation to the pathophysiology of ADs and to the mechanism of action of different anxiolytic treatments, researchers are moving their efforts to assess the importance of the balance between these systems and of other transmitters and modulators. Structural bases of anxiety and neuroimaging data in ADs Functional imaging techniques have been used intensively to identify possible neurobiological correlates between core anxiety symptoms and anatomical and neurophysiological alterations in the central nervous system (Kilts, 2003). Robust evidence indicates that the amygdala mediates states of
Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

20

b. dellosso

ET AL.

increased arousal and fear responses (Ohman, 2005). The central nucleus of the amygdala receives information from the visual, auditory, olfactory, nocioceptive and visceral pathways, and mediates the integration of information and execution of autonomic and behavioural fear responses (Kalin et al., 2004). Two types of fear responses have been described: a rapid, less nely tuned mode, in response to immediate threats and activated by a direct pathway from the sensory thalamus to the amygdala, and a slower one that is activated by a thalamo-corticoamygdalo circuit and which allows valuable cortical assessments of threat-related information (Kalin et al., 2004). The prefrontal cortex and the hippocampus are two other key brain structures known to be important in the pathophysiology of anxiety disorders (Shin et al., 2006). The hippocampus is considered to have a role in the processing of contextual information, differentiating between safe versus potentially dangerous contexts; hippocampal dysfunction, consequently, may produce an anxiety response to innocuous stimuli with an overestimation of potentially threatening contexts (Shin et al., 2006). The medial prefrontal cortex may play a critical role in the process of extinction, dened as the reduction of conditioned fear responses when a tone is repeatedly presented without a shock previously associated with the tone (Milad and Quirk, 2002). In animal models, individuals with dysfunctions in the medial prefrontal cortex seem to have difculties in memorizing previous associations between potentially threatening stimuli and lack of shock (Tamminga, 2006). Structural and functional imaging studies have highlighted the role of different brain areas such as the temporal lobe, prefrontal cortex, insula and motor striatal regions in the neural circuitry of PD (Graeff and Del-Ben, 2008). The most consistent ndings suggest: (1) the presence of a left-to-right asymmetry in hippocampal metabolism; (2) hypometabolism in parieto-tempopral areas which may rectify on treatment and (3) changes in anterior cingulate or orbitofrontal metabolism (Malizia, 2003). Studies in patients with PTSD suggest a hyperresponsivity of the amygdala and decient activation of the ventral/medial prefrontal cortex and hippocampus (Liberzon and Sripada, 2008). Structural imaging studies have reported smaller volumes of ventral/ medial prefrontal cortex and hippocampus in patients with PTSD, when compared to healthy controls (Karl et al., 2006). In SAD, functional neuroimaging studies have shown that patients differ from normal controls in
Copyright # 2009 John Wiley & Sons, Ltd.

the processing of social threat-related stimuli and conditioned aversive stimuli (Malizia, 2003); in particular, fMRI studies have shown the involvement of both the amygdala and hippocampus (Etkin and Wager, 2007). In OCD, obsessions have been associated with overactivity of the frontal cortex, possibly as a consequence of impaired thalamic gating, attributable in turn to decient striatal function; by contrast, compulsions may be the result of aberrant striatal activity (Saxena and Rauch, 2000). Resting state PET and SPECT studies in patients with OCD showed increased activity in the orbifrontal cortex and striatum, when compared to healthy controls (Whiteside et al., 2004). An fMRI study has found a signicant correlation between anxiety and degree of activation of the amygdala (Mataix-Cols et al., 2003). In patients with GAD, a PET study found metabolic differences in occipital lobe, limbic regions and basal ganglia in GAD, when compared to healthy controls, after benzodiazepine treatment (Wu et al., 1991). More recently, an fMRI study found that individual differences in the degree of rostral anterior cingulate cortex and amygdala activation predicted better treatment outcomes to venlafaxine (Whalen et al., 2008). Taken together, these neuroimaging ndings have led some to hypothesize a potential neurobiological classication of the anxiety disorders, based on predominant involvement of the amygdala (SAD), the combination of amygdala and cortical involvement (PTSD and PD), or on predominant involvement of cortico-striatal systems; however, there is at present insufcient consistent evidence to categorize GAD and OCD according to this scheme (Cannistraro and Rauch, 2003). Nevertheless, scientic advances in neurobiology are progressively elucidating fundamental brain mechanisms and the areas underlying anxiety, and this should eventually provide a rational base for pharmacological treatment in anxiety disorders. Detailed description of the genetic bases of anxiety disorders is beyond the scope of this review, but a robust body of evidence from family, twin and adoptee studies indicates a complex genetic component is involved in the development of anxiety-related traits (Hettema et al., 2001). For example, allelic variation of 5-HT transporter expression and function, in particular, seems to play a crucial role in the vulnerability to anxiety disorders. In addition, a specic association of the 5-HT transporter polymorphism and amygdala activation is supported by the ndings of a recent meta` analysis (Munafo et al., 2008).
Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders


Table 2. Pharmacokinetics of SNRIs Venlafaxine IR/SR Therapeutic dose range 75225 (divided dose IR) (mg/day) (single dose SR) Bioavailability 45% Biotransformation pathways CYP2D6 and others Major metabolites O-desmethyl-venlafaxine (ODV) (active) Half-life Elimination routes Protein binding NE/5-HT afnity ratio 4 h (parent) Urine (87%) 27% (parent) 15.7 Desvenlafaxine (ODV) 50 Duloxetine Milnacipran

21

60120 (with a maximum 25200 of 60 mg each dose) 80.5% 50% (reduced of a third in smokers) 85% CYP3A4 (minimal) CYP2D6 and CYP1A2. CYP2D6 or CYP2C19 No active metabolites 4-hydroxy duloxetine No active metabolites glucuronide and 5-hydroxy, 6-methoxy duloxetine sulfate (no active) 10 h 12 h (parent) 12 h (parent) Urine unchanged, heces Urine unchanged (<1%), Urine unchanged metabolites urine metabolites (about 70%), (50%), urine conjugated (30%), heces metabolites (about 20%) urine oxidized (20%) 30% >90% (parent) 13% (parent) 13.8 9.3 2

MECHANISM OF ACTION OF SNRIs SNRIs bind to 5-HT and norepinephrine transporters to selectively inhibit the reuptake of these neurotransmitters from the synaptic clefts, and therefore have a dual mode of action to increase the availability of 5-HT and norepinephrine within the central nervous system. The rationale for use of SNRIs comes from the possibility to correct dysfunctions of central serotonergic and noradrenergic functions which are thought to play a critical role in the pathophysiology of anxiety disorders. In addition, SNRIs have the advantage to act simultaneously on different areas of functioning that some authors ascribe to particular monoamines (for example, aggression to 5-HT, and decits in motivation to norepinephrine) (Healy and McMonagle, 1997; Stahl, 1999). The class of SNRIs currently includes the following agents: venlafaxine and its active metabolite drug desvenlafaxine, duloxetine and milnacipran (Table 2).

Venlafaxine

reached within 4 days of treatment. Renal elimination is the primary route of excretion. Both venlafaxine and its active metabolite, O-desmethylvenlafaxine (ODV) have linear kinetics. Venlafaxine is available in both immediate-release (IR) and extended release (XR) formulations: the extended release formulation simplies administration, as once-daily dosing achieves similar bioavailability to that with the twice-daily dose. The starting dose is generally 75 mg/day and the recommended maximum dose is 375 mg/day. Reduced doses are recommended in patients with hepatic cirrhosis and severe renal disease. In healthy elderly patients, however, dose adjustments may not be necessary (Kent, 2000). Both venlafaxine and its metabolites have little afnity for muscarinic, cholinergic, H1-histaminic or a-adrenergic receptors, and have no monoamine oxidase A or B inhibitory activity (Kent, 2000). Venlafaxine is though to act primarily as an SSRI at low therapeutic dosages (<100 mg/day), only becoming a dual reuptake inhibitor as the dosage increases (Kelsey, 1996; Roseboom and Kalin, 2000). Desvenlafaxine

Venlafaxine was the rst SNRI to be introduced. It is a bicyclic phenylethylamine compound (Thase and Sloan, 2006). After oral administration Venlafaxine is well absorbed from the gastrointestinal tract and undergoes extensive hepatic rst-pass metabolism. Peak plasma concentrations are achieved within 2 h of oral intake and steady-state plasma concentrations are
Copyright # 2009 John Wiley & Sons, Ltd.

Desvenlafaxine is the only major active metabolite of venlafaxine and like the parent compound is well absorbed after oral administration (80% bioavailability) with a half-life of approximately 910 h. The time
Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

22

b. dellosso

ET AL.

to maximum concentration is 7.5 h, and steady-state concentrations of desvenlafaxine are reached within 3 4 days of treatment. The compound has low protein binding, independent of its concentration. The recommended dose of desvenlafaxine is 50 mg/day, without regard to meals. Elimination occurs in the form of a glucuronide conjugate metabolite, with a low possibility of drug interactions (Septien-Velez et al., 2007). In addition, the inhibition of CYP450 enzymes by the agent is minimal (Shilling et al., 2005). Desvenlafaxine has a NE/5-HT afnity ratio of 13.8 (Roseboom and Kalin, 2000), and when compared to VEN its norepinephrine-binding afnity is almost three times higher (Sopko et al., 2008). The main pharmacokinetic difference between desvenlafaxine and its parent drug is the route of metabolism: whereas venlafaxine is metabolized primarily by CYP2D6, desvenlafaxine is conjugated via uridine diphosphate glucuronyl transferase (UGT), and therefore there may be more potential drug interactions for venlafaxine than desvenlafaxine during concomitant administration with other medications that affect the CYP2D6 pathway (Sopko et al., 2008). 3.3 Duloxetine

NE with Kis of 0.8 nM and 7.5 nM, respectively (having a NE/5-HT afnity ratio of 9:3) (Barkin and Barkin, 2005). Milnacipran

Milnacipran shows linear pharmacokinetics over a dose range of 25200 mg/day. It is rapidly and extensively absorbed (>85%) and has a half-life of 12 h with steady-state reached within 48 h when administrated twice a day (Delini-Stula, 2000). Milnacipran is not metabolized by cytocrome P450 and is eliminated by renal excretion (Preskorn, 2004); as a consequence, clearance is signicantly prolonged in patients with renal failure, with a half-life three times longer than in normal volunteers (Puozzo et al., 1998b). By contrast its pharmacokinetic properties are essentially unchanged in patients with severe liver impairment (Puozzo et al., 1998a). Plasma protein binding is low and non-saturable. None of its metabolites have meaningful pharmacological activity at clinical levels. The NE/5-HT afnity ratio of milnacipran is 2:1 (Puozzo et al., 2002), this being the closest to a 1:1 ratio of all the SNRIs. RANDOMIZED CONTROLLED TRIALS AND OPEN STUDIES WITH SNRIs IN ANXIETY DISORDERS

Duloxetine shows linear kinetics at the therapeutic dosage (60120 mg/day). The mean elimination halflife is 12.5 h and steady concentration is reached within 3 days at each dose (Hunziker et al., 2005). Duloxetine undergoes phase I oxidation carried out by cytochrome P450 isozymes CYP2D6 and CYP1A2; oxidized duloxetine is then conjugated with glucuronic acid. The primary metabolite observed in plasma is the glucuronide conjugate of 4-hydroxy duloxetine. Intermediate unconjugated metabolites are not detectable in signicant levels in plasma so that conjugation of oxidized duloxetine must occur rapidly. The conjugated metabolites have low afnity for the 5HT, NE or dopamine transporter and are considered to be inactive (Kuo et al., 2004). Elimination is primarily renal, with 72% excreted in the urine and 19% in faeces, the latter resulting from biliary excretion (Lantz et al., 2003). Duloxetine inhibits the uptake of 5HT and
Copyright # 2009 John Wiley & Sons, Ltd.

Panic disorder Two early open-label case series provided preliminary indications of the efcacy of venlafaxine in the treatment of PD even at low dosages (37.5 to 75 mg/ day) (Geracioti, 1995; Papp et al., 1998). A subsequent double-blind placebo-controlled study of 8 weeks in a sample of 25 patients found more certain evidence of efcacy (Pollack et al., 1996). Although one placebocontrolled investigation found rather mixed evidence of efcacy (Bradwejn et al. 2005), a later study found that venlafaxine (225 mg/day) had statistically superior response and remission rates versus placebo, good overall tolerability and similar efcacy to the SSRI paroxetine (40 mg/day) in short-term treatment in a 12-week study involving 664 patients (Pollack et al., 2007). Venlafaxine also has long-term efcacy and a 26-week double-blind placebo-controlled relapse preHum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders

23

vention study in patients who had previously responded to open treatment found signicantly fewer relapses during continuation treatment with venlafaxine (Ferguson et al., 2007). There is only limited data for the efcacy of other SNRIs in PD. A single case report describes successful treatment with duloxetine 60 mg/day (Crippa and Zuardi, 2006), and a 10-week open-label study in 31 patients found milnacipran to be effective at doses of 50100 mg/day, 58% of the sample achieving symptomatic remission (Blaya et al., 2007).

signicant improvement in anxiety (Gama et al., 2006). Social Anxiety Disorder (SAD) An early 15-week open-label study suggested the efcacy of exible doses of venlafaxine (112.5 187.5 mg/day) in patients who had not responded to SSRI treatment (Altamura et al., 1999). A subsequent multicentre randomized, double-blind, placebo-controlled, parallel group study found that venlafaxine-XR (75225 mg/day) had signicant superiority over placebo in the short-term treatment, with efcacy and tolerability similar to that of paroxetine (2050 mg/day) (Liebowitz et al., 2005a). Further double-blind placebo controlled trials have conrmed the efcacy of venlafaxine-XR (75225 mg/ day) in both short- and long-term treatment (Allgulander et al., 2004; Davidson, 2003; Liebowitz et al., 2005b; Rickels et al., 2004; Stein et al., 2005). As in most other anxiety disorders, the data for other SNRIs in SAD is limited. A small case series described the successful acute treatment of two patients with duloxetine at the dosage of 120 mg/day (Crippa et al., 2007), and 12 weeks of treatment with milnacipran was benecial in six out of 11 patients with TaijinKyofusho (a form of social phobia common in Japan) (Nagata et al., 2003). Post-traumatic Stress Disorder (PTSD) An open case series of patients with PTSD and depressive symptoms, treated with venlafaxine for 6 weeks reported a signicant reduction in symptom severity and improvement in global functioning (Smajkic et al., 2001). More recently, a 6-month double-blind placebo-controlled study showed that exible doses of venlafaxine-XR (37.5300 mg/day) were effective and generally well-tolerated (Davidson et al., 2006a). A parallel 12-week, multicentre, doubleblind, placebo-controlled study suggested that venlafaxine (37.5300mg/day) was superior to placebo in improving PTSD symptoms, whereas sertraline (25 200 mg/day) was not (Davidson et al., 2006b). Only limited data are available regarding the use of duloxetine in patients with PTSD. Combination of electroconvulsive therapy with both duloxetine and olanzapine produced full remission in a patient with PTSD and comorbid major depressive disorder (Hanretta and Malek-Ahmadi, 2006). There are as yet no published studies with milnacipran in patients with PTSD.
Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

Generalized Anxiety Disorder (GAD) Many placebo-controlled studies have demonstrated that venlafaxine-XR is effective in reducing both the somatic and psychological symptoms of GAD at doses of 75225 mg/day (Davidson et al., 1999; Hackett, 2000; Rickels et al., 2000) and in preventing relapses during 6-month double-blind treatment (Allgulander et al., 2001; Gelenberg et al., 2000). Comparisons of the SSRIs paroxetine and escitalopram with venlafaxine did not nd any difference of efcacy in acute treatment of GAD although these trials may have had insufcient power to distinguish reliably between treatments (Bose et al., 2008; Kim et al., 2006). Duloxetine was approved by the FDA (February 2007) and the European Medicine Agency (EMEA) (August 2008) for the treatment of GAD. Double-blind short-term placebo-controlled trials nd consistent evidence of efcacy for duloxetine in acute treatment (Allgulander et al., 2007; Endicott et al., 2007; Hartford et al., 2007; Koponen et al., 2007; Rynn et al., 2008). With the exception of the multicentre trial of Koponen et al. (2007) in which a xed dose was administered, all studies lasted 910 weeks and employed exible doses (60120 mg/die) (Allgulander et al., 2007; Endicott et al., 2007; Rynn et al., 2008). Long-term efcacy was demonstrated through the ndings of a double-blind placebo-controlled relapse prevention study (Davidson et al., 2008). Duloxetine is efcacious in reducing signicant pain symptoms (Russell et al., 2007), and has similar efcacy to venlafaxine in reducing somatic symptoms (Nicolini et al., 2008). For milnacipran, data is only limited. A pre-clinical study suggested that chronic administration of milnacipran at doses of 50100 mg/day/kg had the potential to be benecial in reducing anxiety symptoms (Moojen et al., 2006); and a case series of schizophrenic patients with intense anxiety symptoms, treated with clozapine and milnacipran at a dose of 100 mg/day suggested a
Copyright # 2009 John Wiley & Sons, Ltd.

24

b. dellosso

ET AL.

Obsessive-Compulsive Disorder (OCD) Double-blind comparative studies support the efcacy of venlafaxine in patients with OCD and a previous poor response to SSRI treatment (DellOsso et al., 2006). An open naturalistic study of 39 patients with OCD (29 being regarded as treatment-resistant) found that venlafaxine was effective (Hollander et al., 2003). A 12-week randomized double-blind comparative study found that venlafaxine was equally effective to paroxetine in treating OCD (Denys et al., 2003). A previous study, however, did not support the efcacy of venlafaxine in OCD (Yariyura-Tobias and Neziroglu, 1996). No randomized placebo-controlled studies of duloxetine in patients with OCD have yet been published, although it may be helpful in treatment-resistant patients, as suggested in a recent case-series of OCD patients with comorbid mood/anxiety disorders who had not responded to previous treatment with serotonin reuptake inhibitors (SRIs) at adequate doses for at least 12 weeks, (DellOsso et al., 2008). A pre-clinical study with milnacipran, administered at dosages >10 mg/ day, showed inhibition of marble burying behaviour in mice (an animal model of OCD), suggesting potential efcacy in OCD treatment (Sugimoto et al., 2007). We are not aware of any studies evaluating the use of desvenlafaxine in patients with anxiety disorders. Preliminary data suggest some efcacy of this molecule in reducing anxiety symptoms in patients with major depressive disorder (Tourian et al., 2007). SAFETY AND TOLERABILITY DATA Most patients with anxiety disorders will require longterm treatment, particularly those with the most severe symptoms (Kjernisted and Bleau, 2004). However many will stop treatment prematurely due to the onset of side effects (Devane et al., 2005). SSRI treatment generally shows lower rates of treatment discontinuation than with tricyclic antidepressants (McManus et al., 2004), but many patients experience troublesome side effects; indeed, a recent primary care study found that SSRIs were the most frequent class of drug responsible for adverse effects (Gandhi et al., 2003). Two common stated reasons for discontinuation of SSRIs treatment are sexual dysfunction and weight gain; a naturalistic long-term comparison of SSRIs in the treatment of PD found rates of 6.38.2% for weight gain and 515% for sexual dysfunction (Dannon et al., 2007). Venlafaxine may have some advantage over SSRIs in terms of reduced propensity for weight gain (Kim
Copyright # 2009 John Wiley & Sons, Ltd.

et al., 2006), although not for sexual dysfunction (Gregorian et al., 2002). By contrast, elevation of blood pressure is more common in patients treated with venlafaxine when compared to those undergoing SSRI treatment; although a recent naturalistic study in 37 patients treated with high venlafaxine doses (225 525 mg) found a favourable cardiovascular prole (Mbaya et al., 2007). A review of the prevalence of sexual dysfunction with antidepressants suggested that duloxetine was the least likely to provide these side effects (Werneke et al., 2006). Meta-analysis of four double-blind studies found that patients receiving duloxetine (40 120 mg/day) had lower rates of sexual dysfunction during the rst 8 weeks of treatment, when compared to patients receiving paroxetine (20 mg/day) (Delgado et al., 2005). A recent placebo controlled study of duloxetine versus escitalopram in depressed patients found an incidence of treatment-emergent sexual dysfunction of 33.3% for duloxetine, 43.6% for escitalopram and 25.0% for placebo (Clayton et al., 2007). Duloxetine appears to have time-course dependent effect on body weight, with slight weight loss during short-term treatment and subsequent modest weight gain over the long-term (Wise et al., 2006). Data for the other SNRIs is limited. A recent metaanalysis of 16 randomized controlled trials conducted in depressed patients reported that milnacipran showed similar tolerability to other antidepressants in the shortterm, but fewer adverse events and lower premature withdrawal rates (Nakagawa et al., 2008). Milnacipran is associated with the development of sexual dysfunction, although in the majority of patients sexual function improves as depressive symptoms reduce (Baldwin et al., 2008). Considerations of desvenlafaxine are based on its safety prole in depressed patients; in general, the tolerability prole is nearly identical to that of venlafaxine, the most common side-effects being nausea, insomnia, somnolence and dizziness (Sopko et al., 2008). DISCUSSION Anxiety disorders are common conditions associated with a considerable functional impairment (Carpiniello et al., 2002; Demertzis and Craske, 2006). Early diagnosis and long-term treatment are important for improving quality of life of patients as longer duration of untreated illness may be associated with a less favourable course of illness (Altamura et al., 2005, 2008). SSRIs and cognitive-behaviour therapy are held to be the principal treatments for patients with anxiety
Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders

25

disorders, but cannot be regarded as ideal. A potential advantage for SNRIs is the selective double action on both norepinephrine and serotonin, as this has been found to offer some advantage over conventional SSRI treatment in depressed patients (Silverstone, 2004). Meta-analyses have indicated that treatment of major depressive disorder with venlafaxine achieves greater response and remission rates than is seen with conventional SSRIs (Papakostas et al., 2007; Rudolph, 2002; Smith et al., 2002; Stahl et al., 2002; Thase et al., 2001; Thase, 2008). It is premature to state whether this might also translate into superiority for SNRIs when compared to SSRIs in anxiety disorders (Machado et al., 2006). The relative onset of action of SNRIs and SSRIs in anxiety disorders deserves further research, as at least one placebo-controlled study with venlafaxine in GAD and a placebo-controlled study with duloxetine in depressed patients showed a signicant reduction in anxiety symptoms by the second week of treatment (Allgulander et al., 2001; Detke et al., 2002). Anxiety disorders are often complicated by the presence of comorbid conditions, in particular mood and substance use disorders (Belzer and Schneier, 2004; Brady et al., 2007). Venlafaxine has been found effective in patients with anxiety disorders complicated by other psychiatric conditions (Feighner et al., 1998; Khan et al., 1998; Rudolph et al., 1998), and duloxetine has been found benecial in patients with physical illhealth arising from diabetic neuropathic pain and bromyalgia (Sultan et al., 2008). Furthermore, SNRIs have been usefully combined with atypical antipsychotics in patients with co-morbid mood and anxiety disorders (McIntyre et al., 2007). Another potential advantage for SNRIs over other compounds in the treatment of anxiety disorders may reside in the specic efcacy of these compounds in relieving somatic symptoms which represent a core dimension of many disorders. This effect has been already reported in patients with major depressive disorder and somatic complaints and in patients with anxiety disorders but needs further investigation (Barkin and Barkin, 2005).

approvals for some disorders. In depression, SNRI treatment may combine a shorter latency of onset with higher rates of overall remission than is seen with conventional SSRIs, and may be of particular value in patients with comorbid physical symptoms. These potential advantages cannot yet be regarded as conrmed but even modest differences in efcacy and tolerability for one treatment approach over another may have important public health implications given the high prevalence, disability and chronic nature of these conditions. REFERENCES
Allgulander C, Bandelow B, Hollander E, et al. 2003. WCA recommendations for the long-term treatment of generalized anxiety disorder. CNS Spectr 8: 5361. Allgulander C, Hackett D, Salinas E. 2001. Venalfaxine extended release (ER) in the treatment of generalized anxiety disorder: twenty-four-week placebo controlled dose-ranging study. Br J Psychiatry 179: 1522. Allgulander C, Hartford J, Russell J, et al. 2007. Pharmacotherapy of generalized anxiety disorder: results of duloxetine treatment from a pooled analysis of three clinical trials. Curr Med Res Opin 23: 1245 1252. Allgulander C, Mangano R, Zhang J, et al. 2004. Efcacy of Venlafaxine ER in patients with social anxiety disorder: a double-blind, placebo-controlled, parallel-group comparison with paroxetine. Hum Psychopharmacol 19: 387396. Alonso J, Angermeyer MC, Bernert S, et al. European Study of the Epidemiology of Mental Disorders (ESEMeD). Project. Prevalence of mental disorders in Europe: results from the European Study of the Epidemiology of Mental Disorders (ESEMeD) project. 2004. Acta Psychiatr Scand Suppl 420: 2127. Alonso J, Lepine JP. ESEMeD/MHEDEA 2000 Scientic Committee. 2007. Overview of key data from the European Study of the Epidemiology of Mental Disorders (ESEMeD). J Clin Psychiatry 68: 39. Altamura AC, Pioli R, Vitto M, Mannu P. 1999. Venlafaxine in social phobia: a study in selective serotonin reuptake inhibitor non-responders. Int Clin Psychopharmacol 14: 239245. Altamura AC, Dellosso B, DUrso N, Russo M, Fumagalli S, Mundo E. 2008. Duration of untreated illness as a predictor of treatment response and clinical course in generalized anxiety disorder. CNS Spectr 13: 415422. Altamura AC, Santini A, Salvadori D, Mundo E. 2005. Duration of untreated illness in panic disorder: a poor outcome risk factor? Neuropsychiatr Dis Treat 1: 345347. American Psychiatric Association. 2000. Diagnostic and Statistical Manual of Mental Disorders, 4th edn, Text Revision. American Psychiatry Publishing: Washington DC. Angst J, Gamma A, Baldwin DS, Ajdacic-Gross V, Rossler W. 2009. The generalized anxiety spectrum: prevalence, onset, course and outcome. Eur Arch Psychiatry Clin Neurosci 259: 3745. Argyropoulos SV, Nutt DJ. 1999. The use of benzodiazepines in anxiety and other disorders. Eur Neuropsychopharmacol 9: 407412. Argyropoulos SV, Nutt DJ. Neurochemical aspects of anxiety. In Anxiety Disorders, Chap 11, Nutt DJ, Ballenger JC (eds). 2003. Blackwell Science Ltd: Malden, MA; 181199. Baldwin DS. 2004. Sexual dysfunction associated with antidepressant drugs. Expert Opin Drug Saf 3: 457470. Baldwin DS. 2006. Evidence-based guidelines for anxiety disorders: can they improve clinical outcomes? CNS Spectr 11: 3439. Baldwin DS, Ajel K. 2007. The role of pregabalin in the treatment of generalized anxiety disorder. Neuropsychiatric Disease Treat 3: 185191. Baldwin DS, Anderson IM, Nutt DJ, et al. 2005. Evidence-based guidelines for the pharmacological treatment of anxiety disorders: recommendations from the British Association for Psychopharmacology. J Psychopharmacol 19: 567596.

CONCLUSIONS Over the last two decades, there has been much progress in our understanding of the epidemiology, neurobiology and treatment of anxiety disorders. Whilst SSRIs are regarded as rst-line pharmacological treatments in most anxiety disorders, SNRIs are receiving increasing consideration, and venlafaxine and duloxetine have already received regulatory
Copyright # 2009 John Wiley & Sons, Ltd.

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

26

b. dellosso

ET AL.

Baldwin DS, Moreno R, Briley M. 2008. Resolution of sexual dysfunction during acute treatment of major depression with milnacipran. Hum Psychopharmacol 23: 527532. Bandelow B, Zohar J, Hollander E, Kasper S, Moller HJ. World Federation of Societies of Biological Psychiatry Task Force on Treatment Guidelines for Anxiety, Obsessive-Compulsive and Posttraumatic Stress Disorders. 2002. World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the pharmacological treatment of anxiety, obsessive-compulsive and posttraumatic stress disorders. World J Biol Psychiatry 3: 171199. Barkin RL, Barkin S. 2005. The role of venlafaxine and duloxetine in the treatment of depression with decremental changes in somatic symptoms of pain, chronic pain, and the pharmacokinetics and clinical considerations of duloxetine pharmacotherapy. Am J Ther 12: 431438. Bell CJ, Nutt DJ. 1998. Serotonin and panic. Br J Psychiatry 172: 465471. Belzer K, Schneier FR. 2004. Comorbidity of anxiety and depressive disorders: issues in conceptualization, assessment, and treatment. J Psychiatr Pract 10: 296306. Blaya C, Seganfredo AC, Dornelles M, et al. 2007. The efcacy of milnacipran in panic disorder: an open trial. Int Clin Psychopharmacol 22: 153158. Bose A, Korotzer A, Gommoll C, Li D. 2008. Randomized placebocontrolled trial of escitalopram and venlafaxine XR in the treatment of generalized anxiety disorder. Depress Anxiety 25: 854861. Bradwejn J, Ahokas A, Stein DJ, Salinas E, Emilien G, Whitaker T. 2005. Venlafaxine extended-release capsules in panic disorder: exible-dose, double-blind, placebo-controlled study. Br J Psychiatry 187: 352 359. Brady KT, Verduin ML, Tolliver BK. 2007. Treatment of patients comorbid for addiction and other psychiatric disorders. Curr Psychiatry Rep 9: 374380. Bremner JD, Kristal JH, Southwick SM, Charney DS. 1996. Noradrenergic mechanisms in stress and anxiety: I. Preclinical studies. Synapse 23: 28 38. Cannistraro PA, Rauch SL. 2003. Neural circuitry of anxiety: evidence from structural and functional neuroimaging studies. Psychopharmacol Bull 37: 825. Clayton A, Kornstein S, Prakash A, Mallinckrodt C, Wohlreich M. 2007. Changes in sexual functioning associated with duloxetine, escitalopram, and placebo in the treatment of patients with major depressive disorder. J Sex Med 4: 917929. Carpiniello B, Baita A, Carta MG, et al. 2002. Clinical and psychosocial outcome of patients affected by panic disorder with or without agoraphobia: results from a naturalistic follow-up study. Eur Psychiatry 17: 394398. Crippa JA, Zuardi AW. 2006. Duloxetine in the treatment of panic disorder. Int J Neuropsychopharmacol 9: 633634. Crippa JA, Filho AS, Freitas MC, Zuardi AW. 2007. Duloxetine in the treatment of social anxiety disorder. J Clin Psychopharmacol 27: 310. Dannon PN, Iancu I, Lowengrub K, et al. 2007. A naturalistic long-term comparison study of selective serotonin reuptake inhibitors in the treatment of panic disorder. Clin Neuropharmacol 30: 326334. Davidson JR. 2003. Pharmacotherapy of social phobia. Acta Psychiatr Scand Suppl 417: 6571. Davidson JR, DuPont RL, Hedges D, Haskins JT. 1999. Efcacy, safety and tolerability of venlafaxine extended release and buspirone in outpatients with generalized anxiety disorder. J Clin Psychiatry 60: 528535. Davidson J, Baldwin D, Stein DJ, et al. 2006a. Treatment of posttraumatic stress disorder with venlafaxine extended release: a 6-month randomized controlled trial. Arch Gen Psychiatry 63: 11581165. Davidson J, Rothbaum BO, Tucker P, Asnis G, Benattia I, Musgnung JJ. 2006b. Venlafaxine extended release in posttraumatic stress disorder: a sertraline- and placebo-controlled study. J Clin Psychopharmacol 26: 259267. Davidson JR, Wittchen HU, Llorca PM, et al. 2008. Duloxetine treatment for relapse prevention in adults with generalized anxiety disorder: a double-blind placebo-controlled trial. Eur Neuropsychopharmacol 18: 673681. Davis M. 1997. Neurobiology of fear responses: the role of amygdale. J Neuropsychiatry Clin Neurosci 9: 382402.

De Girolamo G, Polidori G, Morosini P, et al. 2006. Prevalence of common mental disorders in Italy: results from the European Study of the Epidemiology of Mental Disorders (ESEMeD). Soc Psychiatry Psychiatr Epidemiol 41: 853861. Deakin JFW, Graeff FG. 1991. 5-HT and mechanisms of defense. J Psychopharmacol 5: 305315. Delgado PL, Brannan SK, Mallinckrodt CH, et al. 2005. Sexual functioning assessed in 4 double-blind placebo- and paroxetine-controlled trials of duloxetine for major depressive disorder. J Clin Psychiatry 66: 686692. Delini-Stula A. 2000. Milnacipran: an antidepressant with dual selectivity of action on noradrenaline and serotonin uptake. Hum Psychopharmacol 15: 255260. DellOsso B, Mundo E, Marazziti D, Altamura AC. 2008. Switching from serotonin reuptake inhibitors to duloxetine in patients with resistant obsessive compulsive disorder: a case series. J Psychopharmacol 22: 210213. DellOsso B, Nestadt G, Allen A, Hollander E. 2006. Serotonin-norepinephrine inhibitors in the treatment of obsessive-compulsive disorder: a critical review. J Clin Psychiatry 67: 600610. Demertzis KH, Craske MG. 2006. Anxiety in primary care. Curr Psychiatry Rep 8: 291297. Denys D, van der Wee N, van Megen HJ, Westenberg HG. 2003. A double blind comparison of venlafaxine and paroxetine in obsessive-compulsive disorder. J Clin Psychopharmacol 23: 568575. Detke MJ, Lu Y, Goldstain DJ, McNamara RK, Demitrack MA. 2002. Duloxetine 60 mg once daily dosing versus placebo in the acute treatment of major depression. J Psych Res 36: 383390. Devane CL, Chiao E, Franklin M, Kruep EJ. 2005. Anxiety disorders in the 21st century: status, challenges, opportunities, and comorbidity with depression. Am J Manag Care 11: 344353. Endicott J, Russell JM, Raskin J, et al. 2007. Duloxetine treatment for role functioning improvement in generalized anxiety disorder: three independent study. J Clin Psychiatry 68: 518524. Etkin A, Wager TD. 2007. Functional neuroimaging of anxiety: a metaanalysis of emotional processing in PTSD, social anxiety disorder, and specic phobia. Am J Psychiatry 164: 14761488. Feighner JP, Entusuah AR, McPherson MK. 1998. Efcacy of once-daily venlafaxine extended release (XR) for symptoms of anxiety in depressed outpatients. JAD 47: 5562. Ferguson JM, Khan A, Mangano R, Entsuah R, Tzanis E. 2007. Relapse prevention of panic disorder in adult outpatient responders to treatment with venlafaxine extended release. J Clin Psychiatry 68: 5868. Fontenelle LF, Mendlowicz MV, Versiani M. 2006. The descriptive epidemiology of obsessive-compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry 30: 327337. Gama CS, Zanatto VC, Picon F, Lobato MI, Belmonte-de-Abreu PS. 2006. Efcacy of milnacipran in treating anxiety symptoms in schizophrenic patients receiving clozapine: a case series study. Rev Bras Psiquiatr 28: 339340. Gandhi TK, Weingart SN, Borus J, et al. 2003. Adverse drug events in ambulatory care. N Engl J Med 17: 15561564. Garakani A, Mathew SJ, Charney DS. 2006. Neurobiology of anxiety disorders and implications for treatment. Mt Sinai J Med 73: 941949. Gelenberg AJ, Lydiard RB, Rudolph RL, Aguiar L, Haskins JT, Salinas E. 2000. Efcacy of venlafaxine extended-release capsules in nondepressed outpatients with generalized anxiety disorders: a 6-month randomized controlled trial. JAMA 283: 30823088. Geracioti TD Jr. 1995. Venlafaxine treatment of panic disorder: a case series. J Clin Psychiatry 56: 408410. Germine M, Goddard AW, Woods SW, Charney DS, Heninger GR. 1992. Anger and anxiety responses to m-chlorophenylpiperazine in generalized anxiety disorder. Biol Psychiatry 32: 457461. Gorman JM, Hirschfeld RMA, Ninan PT. 2002. New developments in the neurobiological basis of anxiety disorders. Psychopharmacol Bull 36: 4967. Graeff FG, Del-Ben CM. Neurobiology of panic disorder: from animal models to brain neuroimaging. Neurosci Biobehav Rev 2008 32: 1326 1335. Gregorian RS, Golden KA, Bahce A, Goodman C, Kwong WJ, Khan ZM. 2002. Antidepressant-induced sexual dysfunction. Ann Pharmacother 36: 15771589.

Copyright # 2009 John Wiley & Sons, Ltd.

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders


Grimsley SR. 1995. Anxiety disorders. In Applied Therapeutics: The Clinical Use of Drugs, 6th edn, Vol. 76, Young LY, Koda-Kimble MA (eds). Applied Therapeutics Inc: Vancouver, Canada; 128. Hackett D. 2000. Venlafaxine XR in the treatment of anxiety. Acta Psychiatr Scand 406: 3035. Hanretta AT, Malek-Ahmadi P. 2006. Combined use of ECT with duloxetine and olanzapine: a case report. J ECT 22: 139141. Hartford J, Kornstein S, Liebowitz M, et al. 2007. Duloxetine as an SNRI treatment for generalized anxiety disorder: results from a placebo and active-controlled trial. Int Clin Psychopharmacol 22: 167174. Healy D, McMonagle T. 1997. The enhancement of social functioning as a therapeutic principle in the management of depression. J Psychopharmacol 11: 2531. Hettema JM, Neale MC, Kendler KS. 2001. A review and meta-analysis of the genetic epidemiology of anxiety disorders. Am J Psychiatry 158 (10): 15681578. Hirschfeld RM. 2000. Antidepressants in long-term therapy: a review of tricyclic antidepressants and selective serotonin reuptake inhibitors. Acta Psychiatr Scand Suppl 403: 3538. Hollander E, Friedberg J, Wasserman S, Allen A, Birnbaum M, Koran LM. 2003. Venlafaxine in treatment-resistant obsessive-compulsive disorder. J Clin Psychiatry 64: 546550. Hunziker ME, Suehs BT, Bettinger TL, Crismon ML. 2005. Duloxetine hydrocloride: a new dual-acting medication for the treatment of major depressive disorder. Clin Therapheutics 27: 11261143. Johnson MR, Lydiard RB. 1995. The neurobiology of anxiety disorders. Psychiatr Clin North Am 18: 681725. Kalin NH, Shelton SE, Davidson RJ. 2004. The role of the central nucleus of the amygdala in mediating fear and anxiety in the primate. J Neurosci 16: 55065515. Kardos J. 1999. Recent advances in GABA research. Neurochem Int 34: 353358. Karl A, Schaefer M, Malta LS, Dorfel D, Rohleder N, Werner A. 2006. A meta-analysis of structural brain abnormalities in PTSD. Neurosci Biobehav Rev 30: 10041031. Keane TM, Marshall AD, Taft CT. 2006. Posttraumatic stress disorder: etiology, epidemiology, and treatment outcome. Annu Rev Clin Psychol 2: 161197. Kelsey JE. 1996. Dose-response relationship with venlafaxine. J Clin Psychopharmacol 16: 2126. Kent JM. 2000. SNaRIs, NaSSAs and NaRIs: new agents for the treatment of depression. Lancet 355: 911918. Kessler RC, McGonagle KA, Zhao S, et al. 1994. Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbid Survey. Arch Gen Psychiatry 51: 819. Kessler RC, Nelson CB, McGonagle KA, Liu J, Swartz M, Blazer DG. 1996. Comorbidity of DSM-III-R major depressive disorder in the general population: results from the US National Comorbidity Survey. Br J Psychiatry Suppl 30: 1730. Khan A, Upton GV, Rudolp RL, Entsuash R, Leventer SM. 1998. The use of venlafaxine in the treatment of major depression and major depression associated with anxiety: a dose-response study. Venlafaxine Investigator Study Group. J Clin Psychopharmacol 18: 1925. Kilts C. 2003. In vivo neuroimaging correlates of the efcacy of paroxetine in the treatment of mood and anxiety disorders. Psychopharmacol Bul 37: 1928. Kim TS, Pae CU, Yoon SJ, et al. 2006. Comparison of venlafaxine extended release versus paroxetine for treatment of patients with generalized anxiety disorder. Psychiatry Clin Neurosci 60: 347351. Kjernisted KD, Bleau P. 2004. Long-term goals in the management of acute and chronic anxiety disorders. Can J Psychiatry 49: 51S63S. Koponen H, Allgulander C, Erickson J, et al. 2007. Efcacy of duloxetine fort he treatment of generalized anxiety disorder: implications for primary care physicians. Prim Care Companion J Clin Psychiatry 9: 100107. Koran LM, Hanna GL, Hollander E, Nestadt G, Simpson HB. American Psychiatric Association. 2007. Practice guideline for the treatment of patients with obsessive-compulsive disorder. Am J Psychiatry 164: 553.

27

Kroenke K, Spitzer RL, Williams JB, Monahan PO, Lowe B. 2007. Anxiety disorders in primary care: prevalence, impairment, comorbidity, and detection. Ann Intern Med 146: 317325. Kuo F, Gillespie TA, Kulanthaivel P, et al. 2004. Synthesis and biological activation of some known and putative duloxetine metabolites. Bioorg Med Chem Lett 14: 34813486. Lantz RJ, Gillespie TA, Rash TJ, et al. 2003. Metabolism, excretion, and pharmacokinetics of duloxetine in healthy human subjects. Drug Metab Dispos 31: 11421150. LeDoux J. 1998. Fear and the brain: where have we been, and where are we going? Biol Psychiatry 44: 12291238. Leon AC, Portera L, Weissman MM. 1995. The social costs of anxiety disorders. Br J Psychiatry 27: 1922. Lepine JP. 2002. The epidemiology of anxiety disorders: prevalence and societal costs. J Clin Psychiatry 63: 48. Liberzon I, Sripada CS. 2008. The functional neuroanatomy of PTSD: a critical review. Prog Brain Res 167: 151169. Lieb R, Becker E, Altamura C. 2005. The epidemiology of generalized anxiety disorder in Europe. Eur Neuropsychopharmacol 15 (4): 445452. Liebowitz MR, Gelenberg AJ, Munjack D. 2005a. Venlafaxine extended release vs placebo and paroxetine in social anxiety disorder. Arch Gen Psychiatry 62: 190198. Liebowitz MR, Mangano RM, Bradwejn J, Asnis G. SAD Study Group. 2005b. A randomized controlled trial of venlafaxine extended release in generalized social anxiety disorder. J Clin Psychiatry 66: 238247. Machado M, Iskedjian M, Ruiz I, Einarson TR. 2006. Remission, dropouts, and adverse drug reaction rates in major depressive disorder: a metaanalysis of head-to-head trials. Curr Med Res Opin 22: 18251837. Malizia AL. 2003. Brain imaging and anxiety disorders. In Anxiety Disorders, Chap 12, Nutt DJ, Ballenger JC (eds). Blackwell Science Ltd: Malden, MA; 201228. Mataix-Cols D, Cullen S, Lange K, et al. 2003. Neural correlates of anxiety associated with obsessive-compulsive symptom dimensions in normal volunteers. Biol Psychiatry 53: 482493. Mbaya P, Alam F, Ashim S, Bennett D. 2007. Cardiovascular effects of high dose venlafaxine XL in patients with major depressive disorder. Hum Psychopharmacol 22: 129133. McIntyre A, Gendron A, McIntyre A. 2007. Quetiapine adjunct to selective serotonin reuptake inhibitors or venlafaxine in patients with major depression, comorbid anxiety, and residual depressive symptoms: a randomized, placebo-controlled pilot study. Depress Anxiety 24: 487 494. McManus P, Mant A, Mitchell P, Dudley J. 2004. Length of therapy with selective serotonin reuptake inhibitors and tricyclic antidepressants in Australia. Aust NZJ Psychiatry 38: 450454. Milad MR, Quirk GJ. 2002. Neurons in medial prefrontal cortex signal memory for fear extinction. Nature 420: 7074. Moojen VKM, Martins MR, Reinke A, et al. 2006. Effects of milnacipran in animal models of anxiety and memory. Neurochem Res 31: 571577. Mula M, Pini S, Cassano GB. 2007. The role of anticonvulsant drugs in anxiety disorders: a critical review of the evidence. J Clin Psychopharmacol 27 (3): 263272. ` Munafo MR, Brown SM, Hariri AR. 2008. Serotonin transporter (5HTTLPR) genotype and amygdala activation: a meta-analysis. Biol Psychiatry 63: 852857. Nagata T, Oshima J, Wada A, Yamada H, Kiriike N, Iketani T. 2003. Open trial of milnacipran for Taijin-Kyofusho in Japanese patients with social anxiety disorder. Int J Psychiatr Clin Pract 7: 107112. Nakagawa A, Watanabe N, Omori IM, et al. 2008. Efcacy and tolerability of milnacipran in the treatment of major depression in comparison with other antidepressants: a systematic review and meta-analysis. CNS Drugs 22: 587602. Nicolini H, Bakish D, Duenas H, et al. 2008. Improvement of psychic and somatic symptoms in adult patients with generalized anxiety disorder: examination from a duloxetine, venlafaxine extended-release and placebo-controlled trial. Psychol Med 19: 110. Ohman A. 2005. The role of the amygdala in human fear: automatic detection of threat. Psychoneuroendocrinology 30: 953958. Papakostas GI. 2008. Tolerability of modern antidepressants. J Clin Psychiatry 69: 813.

Copyright # 2009 John Wiley & Sons, Ltd.

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

28

b. dellosso

ET AL.

Papakostas GI, Thase ME, Fava M, Nelson JC, Shelton RC. 2007. Are antidepressant drugs that combine serotonergic and noradrenergic mechanisms of action more effective than the selective serotonin reuptake inhibitors in treating major depressive disorder? A meta-analysis of studies of newer agents. Biol Psychiatry 62: 12171227. Papp LA, Sinha SS, Martinez JM, Coplan JD, Amchin J, Gorman JM. 1998. Low-dose venlafaxine treatment in panic disorder. Psychopharmacol Bull 34: 207209. Perkonigg A, Kessler RC, Storz S, Wittchen HU. 2000. Traumatic events and post-traumatic stress disorder in the community: prevalence, risk factors and comorbidity. Acta Psychiatr Scand 101: 4659. Pollack M, Mangano R, Entsuah R, Tzanis E, Simon NM, Zhang Y. 2007. A randomized controlled trial of venlafaxine ER and paroxetine in the treatment of outpatients with panic disorder. Psychopharmacology (Berl) 194: 233242. Pollack MH, Worthington JJ, Otto MW, et al. 1996. Venlafaxine for panic disorder: results from a double-blind, placebo controlled study. Psychopharmacol Bull 32: 667670. Preskorn SH. 2004. Milnacipran: a dual norepinephrine and serotonin reuptake pump inhibitor. J Psychiatr Pract 10: 119126. Puozzo C, Albin H, Vincon G, Deprez D, Raymond JM, Amouretti M. 1998a. Pharmacokinetics of milnacipran in liver impairment. Eur J Drug Metab Pharmacokinet 23: 273279. Puozzo C, Pozet N, Deprez D, Baille P, Ung HL, Zech P. 1998b. Pharmacokinetics of milnacipran in renal impairment. Eur J Drug Metab Pharmacokinet 23: 280286. Puozzo C, Panconi E, Deprez D. 2002. Pharmacology and pharmacokinetics of milnacipran. Int Clin Psychopharmacol 17: 2535. Regier DA, Rae DS, Narrow WE, Kaelber CT, Schatzberg AF. 1998. Prevalence of anxiety disorders and their comorbidity with mood and addictive disorders. Br J Psych Suppl 34: 2428. Ressler KJ, Nemeroff CB. 2000. Role of serotonergic and noradrenergic systems in the pathophysiology of depression and anxiety disorders. Depress Anxiety 12: 219. Rickels K, Pollack MH, Sheehan DV, Haskins JT. 2000. Efcacy of extended-release venlafaxine in nondepressed outpatients with generalized anxiety disorders. Am J Psychiatry 157: 968974. Rickels K, Mangano R, Khan A. 2004. A double-blind, placebo-controlled study of a exible dose of venlafaxine ER in adult outpatients with generalized social anxiety disorder. J Clin Psychopharmacol 24: 488 496. Roseboom PH, Kalin NH. 2000. Neuropsychopharmacology of venlafaxine. Depress Anxiety 12: 2029. Rudolph RL. 2002. Achieving remission from depression with venlafaxine and venlafaxine extended release: a literature review of comparative studies with selective serotonin reuptake inhibitors. Acta Psychiatr Scand Suppl 415: 2430. Rudolph RL, Entsuah R, Chitra R. 1998. A meta-analysis of the effects of venlafaxine on anxiety associated with depression. J Clin Psychopharmacol 18: 136144. Russell JM, Weisberg R, Fava M, Hartford JT, Erickson JS, DSouza DN. 2007. Efcacy of duloxetine in the treatment of generalized anxiety disorder in patients with clinically signicant pain symptoms. Depress Anxiety 25: 111. Rynn M, Russell J, Erickson J, et al. 2008. Efcacy and safety of duloxetine in the treatment of generalized anxiety disorder: a exible-dose, progressive-titration, placebo-controlled trial. Depress Anxiety 25: 182189. Salzman C, Miyawaki EK, le Bars P, Kernhard TN. 1993. Neurobiologic basis of anxiety and its treatment. Harv Rev Psychiatry 1: 197206. Saxena S, Rauch SL. 2000. Functional neuroimaging and the neuroanatomy of obsessive-compulsive disorder. Psychiatr Clin North Am 23: 563 586. Septien-Velez L, Pitrosky B, Padmanabhan SK, Germain J, Tourian KA. 2007. A randomized, double-blind, placebo-controlled trial of desvenalfaxine succinate in the treatment of major depressive disorder. Int Clin Psychopharmacol 22: 338347. Shader RI, Greenblatt DJ. 1993. Use of benzodiazepines in anxiety disorders. N Eng J Med 328: 13981405. Shilling A, Young-Sciame R, Leung L. 2005. Comparison of inhibitory effects of desvenlafaxine succinate, venlafaxine, SS duloxetine, parox-

etine, sertraline and bupropion on human cytochrome P450 activities. Poster presented at International Society for the Study of Xenobiotics, Maui, HI. Shin LM, Rauch SL, Pitman RK. 2006. Amygdala, medial prefrontal cortex, and hippocampal function in PTSD. Ann N Y Acad Sci 1071: 6779. Silverstone PH. 2004. Qualitative review of SNRIs in anxiety. J Clin Psychiatry 65: 1928. Smajkic A, Weine S, Duric-Bijedic Z, Boskailo E, Lewis J, Pavkovic I. 2001. Sertraline, paroxetine and venlafaxine in refugee post traumatic stress disorder with depression symptoms. Med Arh 55: 3538. Smith D, Dempster C, Glanville J, Freemantle N, Anderson I. 2002. Efcacy and tolerability of venlafaxine compared with selective serotonin reuptake inhibitors and other antidepressants: a meta-analysis. Br J Psychiatry 180: 396404. Sopko MA Jr, Ehret MJ, Grgas M. 2008. Desvenlafaxine: another Me Too Drug? (October) (CE). Ann Pharmacother 42: 14391446. Stahl SM. 1998. Mechanism of action of selective serotonin reuptake inhibitors. Serotonin receptors and pathways mediate mediate therapeutic effects and side effects. JAD 51: 215235. Stahl SM. 1999. Why settle for silver, when you can go for gold? Response vs. recovery as the goal of antidepressant therapy. J Clin Psychiatry 60: 213214. Stahl SM, Entsuah R, Rudolph RL. 2002. Comparative efcacy between venlafaxine and SSRIs: a pooled analysis of patients with depression. Biol Psychiatry 52: 11661174. Stein MB, Pollack MH, Bystritsky A, Kelsey JE, Mangano RM. 2005. Efcacy of low and higher dose extended-release venlafaxine in generalized social anxiety disorder: a 6-month randomized controlled trial. Psychopharmacology (Berl) 177: 280288. Sugimoto Y, Tagawa N, Kobayashi Y, Hotta Y, Yamada J. 2007. Effects of the serotonin and noradrenaline reuptake inhibitor (SNRI) milnacipran on marble burying behaviour in mice. Biol Pharm Bull 30: 2399 2401. Sultan A, Gaskell H, Derry S, Moore RA. 2008. Duloxetine for painful diabetic neuropathy and bromyalgia pain: systematic review of randomised trials. BMC Neurol 8: 29. Tamminga CA. 2006. The anatomy of fear extinction. Am J Psychiatry 63: 961. Thase ME. 2008. Are SNRIs more effective than SSRIs? A review of the current state of the controversy. Psychopharmacol Bull 41: 5885. Thase ME, Entsuah AR, Rudolph RL. 2001. Remission rates during treatment with venlafaxine or selective serotonin reuptake inhibitors. Br J. Psychiatry 179: 79. Thase ME, Sloan DME. 2006. Venlafaxine. In Essentials of Clinical Psychopharmacology, Chap. 11, Schatzberg AF, Nemeroff CB (eds). American Psychiatric Publishing, Inc.: Wahington, DC; 159170. Tourian KA, Ahmed S, Patroneva A, Zitek B, Graepel J, Pitrosky B. 2007. Improvement of anxiety symptoms in patients with major depressive disorder treated with desvenlafaxine succinate: a pooled analysis. Poster presented at APA Meeting, San Diego. Ursano RJ, Bell C, Eth S, et al. 2004. Practice guideline for the treatment of patients with acute stress disorder and posttraumatic stress disorder. Am J Psychiatry 161: 331. Van Ameringen M, Mancini C, Pipe B, Bennett M. 2004. Antiepileptic drugs in the treatment of anxiety disorders: role in therapy. Drugs. 64 (19): 21992220. Werneke U, Northey S, Bhugra D. 2006. Antidepressants and sexual dysfunction. Acta Psychiatr Scand 114: 384397. Whalen PJ, Johnstone T, Somerville LH, et al. 2008. A functional magnetic resonance imaging predictor of treatment response to venlafaxine in generalized anxiety disorder. Biol Psychiatry 63: 858863. Whiteside SP, Port JD, Abramowitz JS. 2004. A meta-analysis of functional neuroimaging in obsessive-compulsive disorder. Psychiatry Res 132: 69 79. Wise TN, Perahia DG, Pangallo BA, Losin WG, Wiltse CG. 2006. Effects of the antidepressant duloxetine on body weight: analyses of 10 clinical studies. Prim Care Companion J Clin Psychiatry 8: 269278. Wittchen HU, Hoyer J. 2001. Generalized anxiety disorder: nature and course. J Clin Psychiatry 62: 1519.

Copyright # 2009 John Wiley & Sons, Ltd.

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

snris in anxiety disorders


Wittchen HU, Jacobi F. 2005. Size and burden of mental disorder in Europe: a critical review and appraisal of 27 studies. Eur Neuropsychopharmacol 15: 357376. Wu JC, Buchsbaum MS, Hershey TG, Hazlett E, Sicotte N, Johnson JC. 1991. PET in generalized anxiety disorder. Biol Psychiatry 29: 11811199. Yariyura-Tobias JA, Neziroglu FA. 1996. Venlafaxine in obsessive-compulsive disorder. Arch Gen Psychiatry 53 (7): 653654.

29

Yonkers KA, Bruce SE, Dyck IR, Keller MB. 2003. Chronicity, relapse, and illnesscourse of panic disorder, social phobia, and generalized anxiety disorder: ndings in men and women from 8 years of follow-up. Depress Anxiety 17: 173179. Zwanzger P, Eser D, Rupprecht R. 2007. Anticonvulsants in the treatment of anxietyan alternative treatment option? Nervenarzt 78 (11): 1274 1282.

Copyright # 2009 John Wiley & Sons, Ltd.

Hum. Psychopharmacol Clin Exp 2010; 25: 1729. DOI: 10.1002/hup

Copyright of Human Psychopharmacology: Clinical & Experimental is the property of John Wiley & Sons Ltd. 1996 and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use.

Potrebbero piacerti anche