Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Nov 25.
Published in final edited form as: Expert Opin Drug Metab Toxicol. 2008 Sep;4(9):1223–1234. doi: 10.1517/17425255.4.9.1223

Riluzole in Psychiatry

A Systematic Review of the Literature

Carlos Zarate
PMCID: PMC2587133  NIHMSID: NIHMS58462  PMID: 18721116

Abstract

Background

The glutamate system appears to be an important contributor to the pathophysiology of mood and anxiety disorders. Thus, glutamatergic modulators are reasonable candidate drugs to test in patients with mood and anxiety disorders. Riluzole, a neuroprotective agent with anticonvulsant properties approved for the treatment of amyotrophic lateral sclerosis (ALS) is one such agent.

Objective

To assess the potential risks and benefits of riluzole treatment in psychiatric patients.

Methods

A PubMed search was performed using the keywords “riluzole”, “inhibitor of glutamate release”, and “glutamatergic modulator” in order to identify all clinical studies and case reports involving riluzole in psychiatric patients.

Results/Conclusion

Riluzole’s side effect profile is favorable, and preliminary results regarding riluzole for the treatment of severe mood, anxiety, and impulsive disorders are encouraging.

Keywords: antidepressant, bipolar disorder, depression, glutamate, mania, plasticity, psychiatry, riluzole, anxiety disorder, treatment

1. Introduction

Riluzole, a neuroprotective agent with anticonvulsant properties, is a member of the benzothiazole class. Chemically, riluzole is 2-amino-6-(trifluoromethoxy) benzothiazole, and is the only drug currently approved (by the Food and Drug Administration (FDA) in the United States, the Committee on Proprietary Medicinal Products (CPMP) in Europe, and the Ministry of Health, Labor, and Welfare (MHW) in Japan) for the treatment of amyotrophic lateral sclerosis (ALS). Its use in other psychiatric or neurologic disorders is considered off-label.

Riluzole possesses both glutamatergic modulating, antiepileptic, and neuroprotective properties, all of which make it a promising candidate for the treatment of mood and anxiety disorders. An increasing number of publications regarding its use in neurodegenerative and psychiatric disorders are emerging. In this review, we investigate the pharmacology, pharmacokinetics, side effect profile, and presumptive mechanism of action of riluzole, as well as its use as a treatment in psychiatric disorders.

2. Method

A PubMed literature search was performed using the keywords “riluzole”, “inhibitor of glutamate release”, and “glutamatergic modulator”. These papers were then reviewed to identify all clinical studies in psychiatric populations. In addition, in order to assess potential risks and benefits not identified in psychiatric studies, a review of all clinical studies and case reports involving riluzole was undertaken.

3. Mechanism of Action

Before discussing the mechanism of action of riluzole, it is important to frame this discussion based on current theories on the etiology of depression and presumptive mechanism of action of effective antidepressants and mood stabilizers. Contemporary hypotheses of depression postulate that impairments in cellular resilience and plasticity occur in patients with severe and recurring mood disorders (major depressive disorder (MDD) and bipolar disorder (BPD)) [1]. These impairments are believed to result in atrophy of neurons in critical circuits that mediate hedonic drive, motor behavior emotions, cognition, etc. Thus, severe mood disorders are increasingly being seen as disorders of synapses and circuits rather than simply as the consequence of neurotransmitter alterations [2]. In support of this view, postmortem studies have shown decreases in the number, size, and density of neurons in key areas presumed to be involved in depression (e.g., anterior cingulate cortex, dorsolateral prefrontal cortex, orbitofrontal cortex) (reviewed in [3]). These anatomical changes on a tissue level are consistent with neuroimaging studies showing regional reductions in brain volume [4].

Several mechanisms are believed to result in impairments of cellular resilience and neuroplasticity, including stress, cortisol increases, reduced neurotrophic factors, and excess levels of glutamate. With regards to glutamate, its extracellular concentrations are tightly regulated; excesses are believed to result in neurotoxicity associated with neurodegenerative diseases, trauma, and ischemia. Thus, there has been a recent focus on the development of glutamatergic modulators in the hopes that their neuroprotective properties would be beneficial for these conditions. Such neuroprotective agents could be effective by more tightly regulating glutamate concentrations to minimize glutamate’s deleterious effects on neurons and glia in depression, and by enhancing adequate trophic support so that the intracellular signaling changes required for antidepressant action take place.

Riluzole is a glutamatergic modulator with neuroprotective and plasticity-enhancing properties and for these reasons could potentially play an important role in the treatment of mood and anxiety disorders. Evidence supporting the favorable CNS therapeutic profile of riluzole includes a growing body of work showing that this agent has neuroprotective properties in animal models of brain ischemia [5], spinal cord injury [6, 7], traumatic brain injury [8], Parkinson’s disease [8], post-traumatic peripheral neuropathy [9], and acute noise-induced hearing loss [10]. It has been suggested that inhibition of tyrosine phosphorylation (which is observed in response to brain ischemia) may contribute to riluzole’s neuroprotective properties against excitotoxic injury [11]. In addition, riluzole 10 M was found to be neuroprotective in glutamate-induced toxicity in primary glial cell culture, as evidenced by decreases in lactate dehydrogenase activity and nitrite levels [12].

Although the evidence for riluzole’s neuroprotective effects in the preclinical models cited above is robust, evidence in humans is mixed. There are positive controlled trials for riluzole’s use in ALS [13, 14], and negative studies for its use in Huntington’s disease [15], multiple system atrophy [16], and Parkinson’s disease [17]. Trials to assess its effectiveness in other neurodegenerative conditions studies are underway. In one study involving 14 patients with multiple sclerosis, riluzole showed neuroprotective properties as evidenced by a reduced rate of cervical cord atrophy and development of hypointense T1 brain lesions on magnetic resonance imaging (MRI) [18].

Table 1 lists the different mechanisms of action that have been ascribed to riluzole, some of which might explain its antidepressant effects. However, it is unknown which of them is directly relevant to its potentially mood-enhancing effects. The strongest evidence for riluzole’s mood enhancing and anxiolytic effects come from its action on the glutamatergic system, presumptively involving its glutamate inhibitory release, alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA, a post-synaptic glutamate receptor) trafficking, and glutamate reuptake properties. Discerning which of these mechanisms within the glutamatergic system is important or relevant to the antidepressant and anxiolytic properties of riluzole is still unknown. Few preclinical studies have attempted to discern what mechanisms might be relevant to riluzole’s therapeutic effects in the context of depression and anxiety models; such studies are currently underway.

Table 1. Mechanisms of action for riluzole.

Mechanism of action References
Inhibited release of glutamate as evidenced by its ability to reduce K+ -evoked release from slices of hippocampal area CA1 [19]
Inhibited release of glutamate as evidenced by its ability to reduce K+ -evoked release from rat cortical synaptosomes [20]
Inactivation of voltage-dependent sodium channels [88]
Inactivation of P/Q-type calcium channels [89]
Direct PKC inhibition [90]
AMPA receptor potentiation [26]
Potentiation of postsynaptic GABA(A) receptor function [50]
Suppression of HPA axis [53]
Stimulation of NGF, BDNF, and GDNF in cultured mouse astrocytes [34]
Increased clearance of glutamate from synaptic space through enhancement of reuptake in rat astrocytes [25]
Enhanced high-affinity glutamate uptake in rat spinal cord synaptosomes in vitro and after treatment in vivo [23]
Inhibition of glutamate release from presynaptic terminals through a mechanism linked to G-protein signaling [89]
Activation of TREK, a member of the two-pore domain potassium (K2P) channel [91]
Increased GLT1 expression (also known as EAAT2, the physiologically dominant astroglial protein (G. Sanacora, unpublished findings) [21]
Enhanced activity of glutamate transporters GLAST, GLT1 and EAAC1 [22]

3.1 Regulation of Extracellular Glutamate

3.1.1. Inhibition of glutamate release

Riluzole inhibits the release of glutamate as evidenced by its ability to reduce K+-evoked release from slices of hippocampal area CA1 [19] and rat cortical synaptosomes [20].

3.1.2 Enhancing glutamate transporter reuptake

The excitatory amino acid transporters (EAATs), a family of sodium-dependent carrier proteins, are also capable of regulating glutamate synaptic levels. The mechanism whereby this uptake process occurs is uncertain, but a recent report identified riluzole’s ability to increase GLT1 expression (also known as EAAT2, the physiologically dominant astroglial protein (G. Sanacora, unpublished findings)) as a possibility [21]. More recently it was reported that riluzole enhances the activity of the glutamate transporters GLAST, GLT1, and EAAC1 [22]. In this study, concentrations of riluzole ranging from 0.01 to 100 μM increased specific glutamate uptake in HEKGLT1, HEKEAAC1, and HEKGLAST in a dose-dependent fashion; at the highest concentrations the increase was about 30% in the three cell lines. The authors speculated that riluzole acts by changing the relative affinity for glutamate rather than modifying the expression or the trafficking of transporters, as indicated by the lack of change or the small changes in the maximal uptake rate in the different cell lines [22]. Azbill and colleagues (2000) reported that 0.1 and 1uM riluzole significantly increased glutamate uptake in vitro and after in vivo treatment in animals [23]. Dunlop and colleagues (2003) found a 20-25% increase in glutamate uptake in rat spinal cord synaptosomes at higher riluzole concentrations (10 to 300 uM) [24]. Finally, Frizzo and colleagues (2004) reported that riluzole had a biphasic effect with significant increases in uptake at 1 and 10 μM (15%); higher concentrations were found to be ineffective or even toxic in astrocyte cultures [25].

3. 1. 3 Increasing AMPA trafficking

Of the glutamatergic drugs available for study in mood disorders, riluzole has particularly diverse effects within the glutamatergic system, including inhibition of glutamate release, AMPA trafficking (involved in synaptic plasticity), and enhancement of glutamate reuptake. Riluzole also affects AMPA receptors by increasing cell surface expression of the AMPA subunits GLUR1 and GLUR2, and inducing changes involved in AMPA trafficking; the latter is a component of synaptic plasticity [26]. It has no known direct effects on NMDA or kainate receptors [27].

As will be discussed below, studies indicate that riluzole might have antidepressant properties. Because all FDA-approved antidepressants affect serotonin and/or norepinephrine, it would be important to determine whether riluzole modulates these same neurotransmitters and if it does, whether this effect could possibly explain its antidepressant properties. Riluzole has been found to strongly attenuate electrically evoked release of dopamine and norepinephrine but not serotonin; such neurotransmitters have been widely implicated in the mechanism of antidepressant action [28].

3.2 Regulation of Neurotrophic Factors

3.2.1. Increasing vascular endothelial growth factor (VEGF)

VEGF has been implicated in neuronal survival, neuroprotection, regeneration, growth, differentiation, and axonal outgrowth, all of which are associated with the pathophysiology of MDD [29]. Recently, VEGF mRNA levels in the peripheral leukocytes of 32 drug-naïve patients with MDD were found to be significantly higher than those from a group of age- and sex-matched controls; furthermore, the magnitude of the decrease of VEGF mRNA after eight weeks of treatment with paroxetine (10-40 mg/day) significantly correlated with clinical improvement [30]. However, no association was found between two single nucleotide polymorphic markers of the VEGF gene and MDD or its clinical subtypes [30]. The results of this study are in contrast to the finding that stress decreases levels of VEGF in the adult hippocampus [31] and that VEGF signaling is necessary for the behavioral actions of antidepressants [32]. A recent study found that riluzole injection (10 mg/kg/d, IP) for five days significantly reversed insulin increases in VEGF levels in oxygen-induced retinopathy in rats; its consequences included retinal hemorrhage in rat pups [33].

3.2.2. Brain Derived Neurotrophic Factor (BDNF)

Riluzole stimulates nerve growth factor, brain derived growth factor, and glia cell line-derived neurotrophic factor synthesis in cultured mouse astrocytes [34]. Another report found that repeated, but not single, injections of riluzole resulted in prolonged elevation of hippocampal BDNF, and were associated with increased numbers of newly generated cells in the granule cell layer [35].

4. Clinical Studies in Psychiatry

Although multiple studies have been conducted with riluzole in a number of psychiatric conditions, it is currently only indicated for use in ALS

Riluzole easily crosses the blood brain barrier [36] and ample long-term safety data exist concerning its use in humans. Riluzole has been reported to be well-tolerated for long periods of up to almost seven years [37]. Evidence from a variety of studies with experimental animals and with humans indicates that it is devoid of the psychotomimetic or other behavioral side-effects commonly associated with excitatory amino acid antagonists. Clinical use and clinical investigations support riluzole’s antidepressant and anxiety properties (see Table 2). These reports comprise either case series or open-label studies in patients with treatment-resistant conditions, including unipolar and bipolar depression, generalized anxiety disorders, obsessive-compulsive spectrum disorders, and cocaine dependence. Controlled studies, which are needed to confirm these preliminary findings, are currently underway.

Table 2. Clinical studies of riluzole in treatment-resistant neuropsychiatric patients.

Authors Study design Study Population/n Dose (all given BID) Findings References
Mood Disorders
Singh et al 2004 Case report, add-on Bipolar depression, n=1 200 mg/day Successful augmentation therapy with riluzole in a patient with treatment-resistant bipolar depression who developed a severe rash from lamotrigine [39]
Zarate et al 2004 Open-label for 6-weeks, 2wk washout, monotherapy MDD, n=19 Mean dose 169 mg/day 6-week open label monotherapy study in 19 patients with treatment-resistant MDD. Riluzole had significant antidepressant effects [40]
Zarate et al 2005 Open-label for 8 wks, add-on to lithium, wk washout from other antidepressants Bipolar depression, n=14 Mean dose 171 mg/day In 14 patients with bipolar depression of moderate severity who had failed to respond to a prospective trial of therapeutic levels of lithium, augmentation of lithium with riluzole for 8 weeks significantly improved depressive symptoms. [41]
Sanacora et al 2007 Open-label, no washout, add-on MDD, n=10 Mean dose 95 mg/day When added to ongoing antidepressant therapy in 10 patients with treatment-resistant MDD, led to significant improvements in depressive symptoms after 6-12 weeks [42]
Obsessive-Compulsive Spectrum Disorders
Coric et al 2003 Case report, no washout, add-on MDD and OCD, n=1 100 mg/day In a patient diagnosed with OCD and MDD, significant anti-obsessional and antidepressant effects were noted with eight weeks of riluzole (50 mg bid) treatment when added to the ongoing medication regimen [55]
Coric et al. 2005 Open-label, 6-12 wks OCD, n=13 100 mg/day 7/13 (54%) demonstrated a >35% reduction in Y-BOCS scores, and 5/13 (39%) were responders. HAM-D and HAM-A scores for the group also significantly improved over time. Riluzole was well tolerated with no serious adverse effects noted. [56]
Grant et al. 2007 Open-label for 12 wks, no washout, add-on psychotropics 4/6, K-SADS OCD in children, n=6 10 mg increased every few days, and in 2 cases the dose exceeded 120 mg/day; all subjects except one took at least 50 mg BID 6 subjects, ages 8-16 years received 12-weeks of open-label riluzole 50 mg BID added to ongoing treatment. 4/6 subjects had clear benefit with reduction of more than 46% (39%) overall on the Children’s Yale-Brown Obsessive-Compulsive Scale and “Much Improved” or “Very Much Improved” on the Clinical Global Impressions-Improvement scale; there were no adverse effects leading to discontinuation or dose reduction [57]
Generalized Anxiety Disorder
Mathew et al. 2005 Open-label for 8 wks treatment in GAD patients GAD, n=18 100 mg/day 12/15 patients who completed the trial responded to riluzole. At 8 weeks, 8/15 achieved remission of their anxiety. The median time to response was 2.5 weeks [92]
Case report/series
Pittenger et al. 2005 Cases of patients with self-injurious behavior 25 yo woman with PTSD and BPD; 45 yo woman with MDD, OCD and GAD, n=2 50 mg BID 25 yo woman with PTSD and BPD had no more cutting after 6 wks on riluzole at 50 mg bid added to other medications; 45 yo woman with MDD, OCD and GAD. Hitting and cutting self ceased after 4 wks on riluzole 50 mg bid added to ongoing meds and remained improved 6 months later [61]
Sasso et al. 2006 Case report, add-on to fluoxetine OCD, MDD, and anorexia nervosa, n=1 200 mg/day Significant improvement in symptoms of eating disorder, pathological skin picking, depression, obsessions and compulsions by 4 wks [59]
Coric et al. 2007 Case report, riluzole added to citalopram Trichotillomania and depression, n=1 Up to 200 mg/day At 16 weeks HAM-D score was 7, down from 26 at baseline; at 72 weeks symptoms (depression and urge to pull hair) continued to be minimal [60]

4.1 Mood disorders

Preclinical studies support the antidepressant properties of riluzole as evidenced by its antidepressant-like effects in the unpredictable stress paradigm [38].

A case report indicated successful augmentation therapy with riluzole in a patient with treatment-resistant bipolar depression who had developed a severe rash from lamotrigine [39]. This case report was followed by a six-week open label monotherapy study with riluzole in 19 patients with treatment-resistant MDD; riluzole at a mean daily dose of 169 mg/day had significant antidepressant effects [40]. Subsequently, riluzole was studied in 14 patients with bipolar depression of moderate severity who had failed to respond to a prospective trial of lithium within therapeutic levels. Augmentation of lithium with riluzole (mean daily dose 171 mg) for eight weeks resulted in significantly improved depressive symptoms [41]. In this small study, there was no evidence of hypomania or mania. Finally, in a recent trial, riluzole (mean dose 95 mg/day) was added to ongoing antidepressant therapy in 10 patients with treatment-resistant MDD; these patients showed significant improvement in depressive symptoms after six to 12 weeks [42]. Notably, antidepressant response in these studies occurred within the first few weeks of starting riluzole. The most common side effects were fatigue, nausea, and weight loss.

In an attempt to better understand the trajectory of response to riluzole in depression and the factors predictive of response, data from the two studies discussed above were pooled [40, 41]. This group comprised 33 patients aged 18-70 years who were treated for six weeks with riluzole (50 to 200mg/day) in monotherapy (for MDD) or in combination with lithium for bipolar depression. Response to riluzole was apparent at week one and remained significant for six weeks (the endpoint for the study with MDD patients, and the timepoint at which the eight-week study of bipolar depression patients was thus stopped). The mean daily dose leading to antidepressant response was 170 mg for both studies.

Several demographic and clinical factors were examined as predictors of response for patients with unipolar (n=19) and bipolar depression (n=14). However, none were associated with change in Montgomery-Asberg Depression Rating Scale (MADRS) scores. Pearson’s correlation for these factors was male gender (0.22), age (0.014), unipolar and bipolar diagnosis, (0.156), duration of current major episode (-0.15), past substance abuse or dependence (0.107), melancholic features (-0.003), atypical depression (0.055), mean daily dose of riluzole (0.162), and MADRS score at baseline (0.33).

Riluzole has not yet been studied in mania. Riluzole’s antidepressant properties in bipolar depression and its ability to reduce amphetamine hyperactivity (an animal model of mania) suggest that it should be considered for further testing for this facet of BPD. Notably, in mice, pretreatment with 10 mg/kg, but not 3 mg/kg, of riluzole moderately decreased amphetamine-(but not MK-801-) induced hyperlocomotion [43].

Although studies of riluzole’s efficacy as a treatment for anxiety disorders will be reviewed in greater detail below, it is worth noting that the above-mentioned analysis of the combined dataset involving 33 patients also assessed changes in anxiety symptoms, as they are highly prevalent during depressive episodes. Furthermore, such anxiety changes have been associated with a worse outcome, as indicated by an increased risk for relapse, suicide, and functional disability; a significant proportion of patients presenting with depression and anxiety have poor outcome related to residual and/or refractory anxiety, increased risk of suicidality, and poor treatment response [44-49]. In the merged dataset, a significant improvement in anxiety symptoms was found from week two to study endpoint.

Interestingly, both riluzole and lamotrigine (the latter is approved for relapse prevention in BPD) are inhibitors of glutamate release and have anticonvulsant and neuroprotective properties. Although they share several preclinical findings, they are clinically dissimilar. Unlike lamotrigine, riluzole does not require dose titration, nor has it been associated with severe skin rash (see Table 3). Nevertheless, riluzole has not yet been submitted to the same rigorous mood disorders research as lamotrigine. Additional controlled studies with riluzole in mood and anxiety disorders are needed.

Table 3. Comparison of riluzole and lamotrigine.

Riluzole Lamotrigine
Neuroprotective Yes Yes
Anticonvulsant Yes Yes
Efficacy in acute MDD Yes (only open-label studies) ? Yes (the majority of controlled studies are not positive; clear evidence in maintenance treatment)
Inhibition of voltage-gated Na+ channels Yes Yes
Ca2+ channel blockade Yes (P/Q type) Yes (N- and P-type)
AMPA potentiator Yes Yes
Increased expression of BDNF Yes ??
Dose titration None or rapid Very slow
Steven-Johnson’s syndrome No Yes
Response to the other agent 3 out of 4 lamotrigine responders responded to riluzole ?

4.2 Generalized Anxiety Disorders (GAD)

The anti-anxiety effects of riluzole have been postulated to be the result of postsynaptic GABAA receptor function potentiation in hippocampal neurons [50]. In preclinical work, an earlier report described the anxiolytic effects of riluzole as evidenced by the antagonism of the anxiogenic properties of beta-carboline FG 7142 [51]. A more recent study found that riluzole 3-30 mg/kg p.o. had anxiolytic effects in the rat-conditioned emotional response model of anxiety [52].

The hypothalamic-pituitary-adrenal (HPA) axis hormones are often studied to determine the effects of a particular compound on stress as a model of anxiolytic effects. Riluzole had no significant effects on HPA system activity under baseline and cognitive-stress induced conditions in healthy elderly subjects. However, there was a trend for dampening of the endocrine responses (ACTH, cortisol) [53]. How this finding would translate to patients with anxiety disorders remains to be determined.

Recently, Mathew and colleagues (2008) found, in an eight-week open-label study, that riluzole 100 mg/day was effective in treating anxiety symptoms in patients with GAD. In this study, 80% of completers responded to riluzole, and 53% met remission criteria at eight weeks [54]. In the same study, it was found that hippocampal NAA concentration changes were greater in responders to riluzole than in non-responders.

4.3 Obsessive-Compulsive Spectrum Disorders

In a patient diagnosed with obsessive-compulsive disorder (OCD) and MDD, significant anti-obsessional and antidepressant effects were noted with eight weeks of riluzole treatment (50 mg BID) added to the ongoing medication regimen [55]. Coric and colleagues examined riluzole in an open-label study in 13 patients with treatment-resistant OCD. Riluzole was prescribed at 50 mg BID for six to 12 weeks and 54% of subjects demonstrated a >35% reduction in Y-BOCS scores [56]. More recently, Grant and colleagues [57] found that 12 weeks of open-label treatment with riluzole 50 mg BID was effective in the treatment of OCD in six children aged eight to 16 years. In contrast, in animal studies, riluzole 0.3-10 mg/kg i.p. in mice had no effect on marble-burying behavior, an animal model of OCD [58].

In a recent letter to the editor, it was reported that riluzole 100 mg BID added to fluoxetine ameliorated symptoms of disordered eating, pathological skin picking, depression, obsessions, and compulsions in a 52 year old women with a diagnosis of OCD, MDD, and anorexia nervosa [59]. In another case report, riluzole at doses up to 200 mg/day was found to improve symptoms of trichotillomania [60]. Improvements have also been reported with riluzole in compulsive skin picking [59] and compulsive self-injurious behavior [61]. For more information the reader is referred to a comprehensive review on the topic of OCD [62].

4.4 Cocaine Dependence

In a multi-arm, modified blinded, placebo-controlled screening trial of patients with cocaine dependence, riluzole 100 mg/day had no beneficial effects [63].

5. Side Effect Profile

Controlled studies in ill patients taking multiple concomitant medications indicate that riluzole is well-tolerated [64]. The most frequent dose-related adverse events include nausea, asthenia, and elevated liver enzyme levels. Nausea was reported by 12 to 21% of riluzole-treated patients, compared with 12% for placebo [14]. Less than 1% of patients with asthenia discontinued treatment. Dizziness, diarrhea, anorexia, and circumoral paresthesia occurred more frequently with 200-mg/day of riluzole. It should be noted that concomitant medications were permitted in the trials that examined riluzole, thus adverse events for riluzole may be over-attributed (approximately 72% of subjects took at least one concomitant medication; the mean number of concomitant medications per patient during this trial period was 6.6). Elevated alanine transaminase levels accounted for most treatment discontinuations based on laboratory adverse events. This represented less than four percent of any treatment group (50 mg, 100 mg, or 200 mg riluzole) [14]. In 169 patients with ALS, high inter-individual variability in riluzole serum concentrations was noted. For instance, patients with high serum levels and area under the curve per kilogram of body weight more often had diarrhea [65].

In clinical trials of riluzole for mood disorders, the most common adverse events were headache, fatigue, gastrointestinal distress (nausea or vomiting), decreased salivation, constipation, and tension/inner unrest [64]. In a few patients, a reversible increase in liver function tests was also seen; similar side effects have been observed in ALS trials [13]. No serious adverse events with riluzole have been reported in studies of adults or children with mood or anxiety disorders.

Riluzole has been associated with rare life-threatening events. One case consisted of a 50-year-old man diagnosed with ALS who, 14 days after starting riluzole 50 mg BID, developed myalgia, fever, and increased liver function tests. In this case, a hypersensitivity reaction (myalgia, fever, rash, increased liver function tests, pericarditis) to riluzole was diagnosed [66]. Another report mentions the onset of hallucinations one week after riluzole 50 mg BID was added to memantine and bupropion in a 43-year old woman with a long history of treatment-resistant MDD [67]. There have also been case reports of acute hepatitis [68], pancreatitis [69], and hypersensitivity pneumonitis [70]. Two cases of methemoglobinemia have also been reported [71, 72]. The literature review also found reports that riluzole increases blood pressure in patients with multiple system atrophy and ALS [73, 74], however, this risk does not appear to be significantly increased in individuals without this disease. Granulocytopenia was described in three patients out of 4000 taking riluzole [75]. More recently, another case of reversible granulocytopenia two weeks after starting riluzole was reported [76].

6. Dosing, Pharmacokinetics, and Drug Interactions

6.1 Dosing

The optimal dose of riluzole in ALS is 100 mg/day given BID. However, in studies involving adults with a psychiatric disorder, the dose of riluzole typically ranged from 100-200 mg/day (again, this dosage and use are off-label). Most published reports in patients with mood disorders used flexible dosing up to 200 mg/day. Fixed doses of 100 mg/day were generally used in OCD with a few exceptions. In children with OCD, the dose of riluzole was initiated at 10 mg/day and increased by 10 mg every few days with the mode dose being 100 mg/day [57]. Table 2 contains a summary of riluzole dosing in these studies. Because the majority of the trials conducted in psychiatry used doses higher than that considered optimal in ALS, future studies will need to determine whether a higher dose offers any advantage, particularly because there is a dose-dependent risk for increases in transaminases and gastrointestinal disturbances.

6.2 Pharmacokinetics

The time to peak concentration of riluzole is one to 1.5 hours, and a maximum serum concentration of 214 ng/ml has been reported after a single 50 mg oral dose [77]. The bioavailability of riluzole is 60% (range 30% to 100%) [77], and its total protein binding is 97.5%. Riluzole is extensively metabolized in the liver (Prod Info Rilutek(R), 2006) by direct glucuronidation and hydroxylation by the cytochrome P450IA2 enzyme (CYP1A2) (Prod Info Rilutek(R), 2006). The metabolism of riluzole results in N-hydroxyriluzole and glucuronide conjugates, both of which are inactive. Approximately 85% to 95% of a 50 mg oral tablet is eliminated in the urine in the first 24 hours, and almost all of the metabolites are excreted in the urine. Less than 10% is excreted in feces. It is unknown if riluzole is excreted into human breast milk. The elimination half-life is 12 to 14 hours.

Pharmacokinetic studies of riluzole show a large inter-individual variability in the drug’s clearance and serum concentrations, and food decreases absorption. The inter-individual variability of riluzole’s peak was reported to be as high as 74% [78]. An in vitro study of riluzole metabolism in liver and yeast microsomes revealed that riluzole is transformed to N-hydroxyriluzole by cytochrome P450 enzyme subform 1AS (CYP1A2) and by glucuronidation to an unidentified metabolite [79]. In vivo studies addressing glucuronidation of riluzole are lacking. In a previous study of ALS patients using caffeine as a metabolic probe, it was found that differential CYP1A2 activity was responsible for 37% of the observed variability in riluzole clearance [80]. A recent study did not find that genetic variations in CYP1A1 and CYP1A2 genes seemed to influence riluzole levels [81]. Therefore, alternative metabolic pathways, such as glucuronidation, may also play a role in the inter-individual variability of riluzole elimination.

Glucuronidation is catalyzed by a family of uridine-diphosphate-glucuronosyltransferases (UGTs). These enzymes are responsible for the conjugation of glucuronic acid with many endogenous (e.g., bilirubin, steroids) and xenobiotic compounds [82]. Several subforms of UGT have been identified with both hepatic and intestinal expression sites [83]. The UGT1A1 subform has a genetic polymorphism that results in decreased gene expression. The UGT1A1 wildtype has a six-fold TA repeat ((TA)6) in the promoter region of the UGT gene, while increasing numbers of tandem repeats ((TA)7, designated as UGT1A1*28), result in decreased expression. So far this is the only functional genetic polymorphism that has been elucidated for UGT [84].

Recent evidence suggests that glucuronidation is a major metabolic pathway. Limited data are available on the in vivo metabolic elimination of riluzole. In vitro experiments suggest that CYP1A2 seems to be mainly involved in riluzole clearance. However, in vitro studies also suggest that formation of riluzole-glucuronide plays a prominent role and may, to some extent, determine the drug’s pharmacokinetic variability in patients. It is yet unknown whether riluzole-glucuronide has any therapeutic effect, and how this relates to the effect of the parent compound [81]. However, genetic variability in UGT1A1 was reported not to be the reason for inter-individual variability in riluzole serum concentrations [81].

In terms of brain penetrance, Colovic and colleagues previously reported that riluzole is distributed to brain when administrated orally [85, 86]. More recently, it was reported that riluzole is transported by p-gp at the blood brain barrier level [87]. The comparison between brain/plasma concentration ratios in CF1 mdr l a (-/-) and mdr l a (+/+) mice was 1.4 for riluzole. At least in ALS, transport out of the brain through p-gp could, at least in part, explain why riluzole plasma level does not always correlate with therapeutic effects or side effects. The relevance between p-gp and clinical response or side effects in psychiatric disorders is currently unknown.

6.3 Drug interactions with riluzole

The following drugs are metabolized by cytochrome P450: amitriptyline, imipramine, chlordiazepoxide, ciprofloxacin, digoxin, norfloxacin, phenacetin, rifampin, sulfasalazine, and tacrine. There is thus a theoretical risk of drug toxicity when they are used in combination with riluzole.

7. Conclusion

Riluzole, a neuroprotective agent with anticonvulsant properties, appears to be a promising candidate drug for the treatment of neuropsychiatric disorders; however this remains to be proven. It is a glutamatergic modulator with diverse effects within the glutamatergic brain system, a system that is being increasingly linked with the pathophysiology of mood and anxiety disorders. Riluzole’s side effect profile is favorable, and preliminary studies in severe mood, anxiety, and impulsive disorders are encouraging. These initial trials will need to be followed by more rigorous controlled studies to confirm the nature and extent of riluzole’s therapeutic effects. Riluzole has already led to essential insights into the role of the glutamatergic system in the mechanism of action of antidepressants.

8. Expert opinion

Contemporary theories of mood disorders note that they are characterized by disruptions in cellular resilience and neuroplasticity. Disruptions in neuroplasticity are believed to be largely due to alterations in CNS glutamate neurotransmission. Riluzole is among a series of candidate glutamatergic modulators that is being actively explored for use in a variety of neuropsychiatric disorders; it appears to modulate many different aspects of glutamate’s regulatory system. Riluzole has proved ineffective in some trials of neurodegenerative conditions such as Huntington’s disease, multiple system atrophy, and Parkinson’s disease; although the reason for these failed trials is unknown, it is possible that glutamatergic modulation with an agent such as riluzole would be more useful in individuals earlier in the disease process rather than later.

Riluzole is currently afforded orphan drug status and is approved (by the Food and Drug Administration (FDA) in the United States, the Committee on Proprietary Medicinal Products (CPMP) in Europe, and the Ministry of Health, Labor, and Welfare (MHW) in Japan) only for the treatment of ALS. At present, it is quite expensive, and insurance coverage for this drug is mainly for patients with ALS. For that reason, its use in a clinical population for patients with psychiatric disorders is limited at this time and off-label. Controlled studies, though lacking, are currently underway. Positive results in patients with mood and anxiety disorders would certainly impact riluzole’s clinical use and further testing in psychiatric disorders. Finally, much is being learned from riluzole in preclinical and clinical studies. When it becomes generic, or when new, improved versions of it become available, riluzole will undoubtedly have both short- and long-term significance in the treatment of psychiatric disorders.

Acknowledgments

The authors would like to acknowledge the support of the Intramural Research Program of the National Institute of Mental Heath, the Stanley Medical Research Institute and NARSAD. Ioline Henter provided outstanding editorial assistance.

Conflict of Interest

The authors would like to acknowledge the support of the Intramural Research Program of the National Institute of Mental Health. The authors declare that, except for income received from our primary employer, no financial support or compensation has been received from any individual or corporate entity over the past three years for research or professional service and there are no personal financial holdings that could be perceived as constituting a potential conflict of interest.

References

  • 1.Zarate CA, Jr., Singh J, Manji HK. Cellular plasticity cascades: targets for the development of novel therapeutics for bipolar disorder. Biol Psychiatry. 2006 Jun 1;59(11):1006–20. doi: 10.1016/j.biopsych.2005.10.021. [DOI] [PubMed] [Google Scholar]
  • 2.Schloesser RJ, Huang J, Klein PS, et al. Cellular plasticity cascades in the pathophysiology and treatment of bipolar disorder. Neuropsychopharmacology. 2008 Jan;33(1):110–33. doi: 10.1038/sj.npp.1301575. [DOI] [PubMed] [Google Scholar]
  • 3.Newberg AR, Catapano LA, Zarate CA, et al. Neurobiology of bipolar disorder. Expert Rev Neurother. 2008 Jan;8(1):93–110. doi: 10.1586/14737175.8.1.93. [DOI] [PubMed] [Google Scholar]
  • 4.Drevets WC. Neuroimaging studies of mood disorders. Biol Psychiatry. 2000 Oct 15;48(8):813–29. doi: 10.1016/s0006-3223(00)01020-9. [DOI] [PubMed] [Google Scholar]
  • 5.Heurteaux C, Laigle C, Blondeau N, et al. Alpha-linolenic acid and riluzole treatment confer cerebral protection and improve survival after focal brain ischemia. Neuroscience. 2006;137(1):241–51. doi: 10.1016/j.neuroscience.2005.08.083. [DOI] [PubMed] [Google Scholar]
  • 6.Ates O, Cayli SR, Gurses I, et al. Comparative neuroprotective effect of sodium channel blockers after experimental spinal cord injury. J Clin Neurosci. 2007 Jul;14(7):658–65. doi: 10.1016/j.jocn.2006.03.023. [DOI] [PubMed] [Google Scholar]
  • 7.Schwartz G, Fehlings MG. Evaluation of the neuroprotective effects of sodium channel blockers after spinal cord injury: improved behavioral and neuroanatomical recovery with riluzole. J Neurosurg. 2001 Apr;94(2 Suppl):245–56. doi: 10.3171/spi.2001.94.2.0245. [DOI] [PubMed] [Google Scholar]
  • 8.Benazzouz A, Boraud T, Dubedat P, et al. Riluzole prevents MPTP-induced parkinsonism in the rhesus monkey: a pilot study. Eur J Pharmacol. 1995 Sep 25;284(3):299–307. doi: 10.1016/0014-2999(95)00362-o. [DOI] [PubMed] [Google Scholar]
  • 9.Medico M, Nicosia A, Grech M, et al. Riluzole restores motor activity in rats with post-traumatic peripheral neuropathy. Neurosci Lett. 2004 Mar 18;358(1):37–40. doi: 10.1016/j.neulet.2003.12.090. [DOI] [PubMed] [Google Scholar]
  • 10.Ruel J, Wang J, Pujol R, et al. Neuroprotective effect of riluzole in acute noise-induced hearing loss. Neuroreport. 2005 Jul 13;16(10):1087–90. doi: 10.1097/00001756-200507130-00011. [DOI] [PubMed] [Google Scholar]
  • 11.Peyclit A, Keita H, Juvin P, et al. Effects of riluzole on N-methyl-D-aspartate-induced tyrosine phosphorylation in the rat hippocampus. Brain Res. 2001 Jun 8;903(12):222–5. doi: 10.1016/s0006-8993(01)02429-5. [DOI] [PubMed] [Google Scholar]
  • 12.Dagci T, Yilmaz O, Taskiran D, et al. Neuroprotective agents: is effective on toxicity in glial cells? Cell Mol Neurobiol. 2007 Mar;27(2):171–7. doi: 10.1007/s10571-006-9082-4. [DOI] [PubMed] [Google Scholar]
  • 13.Bensimon G, Lacomblez L, Meininger V, ALS/Riluzole Study Group A controlled trial of riluzole in amyotrophic lateral sclerosis. N Engl J Med. 1994 Mar 3;330(9):585–91. doi: 10.1056/NEJM199403033300901. [DOI] [PubMed] [Google Scholar]
  • 14.Lacomblez L, Bensimon G, Leigh PN, et al. Amyotrophic Lateral Sclerosis/Riluzole Study Group II Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Lancet. 1996 May 25;347(9013):1425–31. doi: 10.1016/s0140-6736(96)91680-3. [DOI] [PubMed] [Google Scholar]
  • 15.Landwehrmeyer GB, Dubois B, de Yebenes JG, et al. Riluzole in Huntington’s disease: a 3-year, randomized controlled study. Ann Neurol. 2007 Sep;62(3):262–72. doi: 10.1002/ana.21181. [DOI] [PubMed] [Google Scholar]
  • 16.Seppi K, Peralta C, Diem-Zangerl A, et al. Placebo-controlled trial of riluzole in multiple system atrophy. Eur J Neurol. 2006 Oct;13(10):1146–8. doi: 10.1111/j.1468-1331.2006.01452.x. [DOI] [PubMed] [Google Scholar]
  • 17.Jankovic J, Hunter C. A double-blind, placebo-controlled and longitudinal study of riluzole in early Parkinson’s disease. Parkinsonism Relat Disord. 2002 Mar;8(4):271–6. doi: 10.1016/s1353-8020(01)00040-2. [DOI] [PubMed] [Google Scholar]
  • 18.Kalkers NF, Barkhof F, Bergers E, et al. The effect of the neuroprotective agent riluzole on MRI parameters in primary progressive multiple sclerosis: a pilot study. Mult Scler. 2002 Dec;8(6):532–3. doi: 10.1191/1352458502ms849xx. [DOI] [PubMed] [Google Scholar]
  • 19.Martin D, Thompson MA, Nadler JV. The neuroprotective agent riluzole inhibits release of glutamate and aspartate from slices of hippocampal area CA1. Eur J Pharmacol. 1993 Dec 21;250(3):473–6. doi: 10.1016/0014-2999(93)90037-i. [DOI] [PubMed] [Google Scholar]
  • 20.Lingamaneni R, Hemmings HC., Jr. Effects of anticonvulsants on veratridine- and KCl-evoked glutamate release from rat cortical synaptosomes. Neurosci Lett. 1999 Dec 3;276(2):127–30. doi: 10.1016/s0304-3940(99)00810-1. [DOI] [PubMed] [Google Scholar]
  • 21.Rothstein JD, Dykes-Hoberg M, Pardo CA, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996 Mar;16(3):675–86. doi: 10.1016/s0896-6273(00)80086-0. [DOI] [PubMed] [Google Scholar]
  • 22.Fumagalli E, Funicello M, Rauen T, et al. Riluzole enhances the activity of glutamate transporters GLAST, GLT1 and EAAC1. Eur J Pharmacol. 2008 Jan 14;578(23):171–6. doi: 10.1016/j.ejphar.2007.10.023. [DOI] [PubMed] [Google Scholar]
  • 23.Azbill RD, Mu X, Springer JE. Riluzole increases high-affinity glutamate uptake in rat spinal cord synaptosomes. Brain Res. 2000 Jul 21;871(2):175–80. doi: 10.1016/s0006-8993(00)02430-6. [DOI] [PubMed] [Google Scholar]
  • 24.Dunlop J, Beal McIlvain H, She Y, et al. Impaired spinal cord glutamate transport capacity and reduced sensitivity to riluzole in a transgenic superoxide dismutase mutant rat model of amyotrophic lateral sclerosis. J Neurosci. 2003 Mar 1;23(5):1688–96. doi: 10.1523/JNEUROSCI.23-05-01688.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Frizzo ME, Dall’Onder LP, Dalcin KB, et al. Riluzole enhances glutamate uptake in rat astrocyte cultures. Cell Mol Neurobiol. 2004 Feb;24(1):123–8. doi: 10.1023/b:cemn.0000012717.37839.07. [DOI] [PubMed] [Google Scholar]
  • 26.Du J, Suzuki K, Wei Y, et al. The anticonvulsants lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization: relationship to clinical effects in mood disorders. Neuropsychopharmacology. 2007 Apr;32(4):793–802. doi: 10.1038/sj.npp.1301178. [DOI] [PubMed] [Google Scholar]
  • 27.Debono MW, Le Guern J, Canton T, et al. Inhibition by riluzole of electrophysiological responses mediated by rat kainate and NMDA receptors expressed in Xenopus oocytes. Eur J Pharmacol. 1993 Apr 28;235(23):283–9. doi: 10.1016/0014-2999(93)90147-a. [DOI] [PubMed] [Google Scholar]
  • 28.Jehle T, Bauer J, Blauth E, et al. Effects of riluzole on electrically evoked neurotransmitter release. Br J Pharmacol. 2000 Jul;130(6):1227–34. doi: 10.1038/sj.bjp.0703424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Warner-Schmidt JL, Duman R. VEGF as a potential target for therapeutic intervention in depression. Curr Opin Pharmacol. 2008;8:14–9. doi: 10.1016/j.coph.2007.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Iga J, Ueno S, Yamauchi K, et al. Gene expression and association analysis of vascular endothelial growth factor in major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2007 Apr 13;31(3):658–63. doi: 10.1016/j.pnpbp.2006.12.011. [DOI] [PubMed] [Google Scholar]
  • 31.Heine VM, Zareno J, Maslam S, et al. Chronic stress in the adult dentate gyrus reduces cell proliferation near the vasculature and VEGF and Flk-1 protein expression. Eur J Neurol. 2005;21:1301–14. doi: 10.1111/j.1460-9568.2005.03951.x. [DOI] [PubMed] [Google Scholar]
  • 32.Warner-Schmidt JL, Duman R. VEGF is an essential mediator of the neurogenic and behavioral actions of antidepressants. Proc Natl Acad Sci USA. 2007;104:4647–52. doi: 10.1073/pnas.0610282104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yoo MH, Yoon YH, Chung H, et al. Insulin increases retinal hemorrhage in mild oxygen-induced retinopathy in the rat: inhibition by riluzole. Invest Ophthalmol Vis Sci. 2007 Dec;48(12):5671–6. doi: 10.1167/iovs.07-0395. [DOI] [PubMed] [Google Scholar]
  • 34.Mizuta I, Ohta M, Ohta K, et al. Riluzole stimulates nerve growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor synthesis in cultured mouse astrocytes. Neurosci Lett. 2001 Sep 14;310(23):117–20. doi: 10.1016/s0304-3940(01)02098-5. [DOI] [PubMed] [Google Scholar]
  • 35.Katoh-Semba R, Asano T, Ueda H, et al. Riluzole enhances expression of brain-derived neurotrophic factor with consequent proliferation of granule precursor cells in the rat hippocampus. Faseb J. 2002 Aug;16(10):1328–30. doi: 10.1096/fj.02-0143fje. [DOI] [PubMed] [Google Scholar]
  • 36.Benavides J, Camelin JC, Mitrani N, et al. 2-Amino-6-trifluoromethoxy benzothiazole, a possible antagonist of excitatory amino acid neurotransmission--II. Biochemical properties. Neuropharmacology. 1985 Nov;24(11):1085–92. doi: 10.1016/0028-3908(85)90196-0. [DOI] [PubMed] [Google Scholar]
  • 37.Lacomblez L, Bensimon G, Leigh PN, et al. Long-term safety of riluzole in amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002 Mar;3(1):23–9. doi: 10.1080/146608202317576507. [DOI] [PubMed] [Google Scholar]
  • 38.Banasr M, Chowdhury G, Duman R, et al. Riluzole blocks stress-related effects on behavior and enhances glutamate/glutamine cycle flux. Neuropsychopharmacology. 2006;2006:S159–S60. [Google Scholar]
  • 39.Singh J, Zarate CA, Jr., Krystal AD. Case report: Successful riluzole augmentation therapy in treatment-resistant bipolar depression following the development of rash with lamotrigine. Psychopharmacology (Berl) 2004 Apr;173(12):227–8. doi: 10.1007/s00213-003-1756-8. [DOI] [PubMed] [Google Scholar]
  • 40.Payne JL, Quiroz J, et al. Zarate CA Jr., editor. An open-label trial of riluzole in patients with treatment-resistant major depression. Am J Psychiatry. 2004 Jan;161(1):171–4. doi: 10.1176/appi.ajp.161.1.171. [DOI] [PubMed] [Google Scholar]
  • 41.Zarate CA, Jr., Quiroz JA, Singh JB, et al. An open-label trial of the glutamate-modulating agent riluzole in combination with lithium for the treatment of bipolar depression. Biol Psychiatry. 2005 Feb 15;57(4):430–2. doi: 10.1016/j.biopsych.2004.11.023. [DOI] [PubMed] [Google Scholar]
  • 42.Sanacora G, Kendell SF, Levin Y, et al. Preliminary evidence of riluzole efficacy in antidepressant-treated patients with residual depressive symptoms. Biol Psychiatry. 2007 Mar 15;61(6):822–5. doi: 10.1016/j.biopsych.2006.08.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lourenco Da Silva A, Hoffmann A, Dietrich MO, et al. Effect of riluzole on MK-801 and amphetamine-induced hyperlocomotion. Neuropsychobiology. 2003;48(1):27–30. doi: 10.1159/000071825. [DOI] [PubMed] [Google Scholar]
  • 44.Fawcett J, Kravitz HM. Anxiety syndromes and their relationship to depressive illness. J Clin Psychiatry. 1983 Aug;44(8 Pt 2):8–11. [PubMed] [Google Scholar]
  • 45.Fava M, Rush AJ, Alpert JE, et al. What clinical and symptom features and comorbid disorders characterize outpatients with anxious major depressive disorder: a replication and extension. Can J Psychiatry. 2006 Nov;51(13):823–35. doi: 10.1177/070674370605101304. [DOI] [PubMed] [Google Scholar]
  • 46.Joffe RT, Bagby RM, Levitt A. Anxious and nonanxious depression. Am J Psychiatry. 1993 Aug;150(8):1257–8. doi: 10.1176/ajp.150.8.1257. [DOI] [PubMed] [Google Scholar]
  • 47.Coryell W, Endicott J, Andreasen NC, et al. Depression and panic attacks: the significance of overlap as reflected in follow-up and family study data. Am J Psychiatry. 1988 Mar;145(3):293–300. doi: 10.1176/ajp.145.3.293. [DOI] [PubMed] [Google Scholar]
  • 48.Davidson JR, Meoni P, Haudiquet V, et al. Achieving remission with venlafaxine and fluoxetine in major depression: its relationship to anxiety symptoms. Depress Anxiety. 2002;16(1):4–13. doi: 10.1002/da.10045. [DOI] [PubMed] [Google Scholar]
  • 49.Fawcett J. Suicide risk factors in depressive disorders and in panic disorder. J Clin Psychiatry. 1992 Mar;53(Suppl):9–13. [PubMed] [Google Scholar]
  • 50.He Y, Benz A, Fu T, et al. Neuroprotective agent riluzole potentiates postsynaptic GABA(A) receptor function. Neuropharmacology. 2002 Feb;42(2):199–209. doi: 10.1016/s0028-3908(01)00175-7. [DOI] [PubMed] [Google Scholar]
  • 51.Stutzmann JM, Cintrat P, Laduron PM, et al. Riluzole antagonizes the anxiogenic properties of the beta-carboline FG 7142 in rats. Psychopharmacology (Berl) 1989;99(4):515–9. doi: 10.1007/BF00589901. [DOI] [PubMed] [Google Scholar]
  • 52.Munro G, Erichsen HK, Mirza NR. Pharmacological comparison of anticonvulsant drugs in animal models of persistent pain and anxiety. Neuropharmacology. 2007 Oct;53(5):609–18. doi: 10.1016/j.neuropharm.2007.07.002. [DOI] [PubMed] [Google Scholar]
  • 53.Kniest A, Wiesenberg C, Weber B, et al. The glutamate antagonist riluzole and its effects upon basal and stress-induced activity of the human hypothalamus-pituitary-adrenocortical system in elderly subjects. Neuropsychobiology. 2001;43(2):91–5. doi: 10.1159/000054873. [DOI] [PubMed] [Google Scholar]
  • 54.Mathew SJ, Price RB, Mao X, et al. Hippocampal N-acetylaspartate concentration and response to riluzole in generalized anxiety disorder. Biol Psychiatry. 2008 May 1;63(9):891–8. doi: 10.1016/j.biopsych.2007.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Coric V, Milanovic S, Wasylink S, et al. Beneficial effects of the antiglutamatergic agent riluzole in a patient diagnosed with obsessive-compulsive disorder and major depressive disorder. Psychopharmacology (Berl) 2003 May;167(2):219–20. doi: 10.1007/s00213-003-1396-z. [DOI] [PubMed] [Google Scholar]
  • 56.Coric V, Taskiran S, Pittenger C, et al. Riluzole augmentation in treatment-resistant obsessive-compulsive disorder: an open-label trial. Biol Psychiatry. 2005 Sep 1;58(5):424–8. doi: 10.1016/j.biopsych.2005.04.043. [DOI] [PubMed] [Google Scholar]
  • 57.Grant P, Lougee L, Hirschtritt M, et al. An open-label trial of riluzole, a glutamate antagonist, in children with treatment-resistant obsessive-compulsive disorder. J Child Adolesc Psychopharmacol. 2007 Dec;17(6):761–7. doi: 10.1089/cap.2007.0021. [DOI] [PubMed] [Google Scholar]
  • 58.Egashira N, Okuno R, Harada S, et al. Effects of glutamate-related drugs on marble-burying behavior in mice: Implications for obsessive-compulsive disorder. Eur J Pharmacol. 2008 Feb 5; doi: 10.1016/j.ejphar.2008.01.035. [DOI] [PubMed] [Google Scholar]
  • 59.Sasso DA, Kalanithi PS, Trueblood KV, et al. Beneficial effects of the glutamate-modulating agent riluzole on disordered eating and pathological skin-picking behaviors. J Clin Psychopharmacol. 2006 Dec;26(6):685–7. doi: 10.1097/01.jcp.0000245567.29531.d6. [DOI] [PubMed] [Google Scholar]
  • 60.Coric V, Kelmendi B, Pittenger C, et al. Beneficial effects of the antiglutamatergic agent riluzole in a patient diagnosed with trichotillomania. J Clin Psychiatry. 2007 Jan;68(1):170–1. doi: 10.4088/jcp.v68n0123f. [DOI] [PubMed] [Google Scholar]
  • 61.Pittenger C, Krystal JH, Coric V. Initial evidence of the beneficial effects of glutamate-modulating agents in the treatment of self-injurious behavior associated with borderline personality disorder. J Clin Psychiatry. 2005 Nov;66(11):1492–3. doi: 10.4088/jcp.v66n1121d. [DOI] [PubMed] [Google Scholar]
  • 62.Pittenger C, Krystal JH, Coric V. Glutamate-modulating drugs as novel pharmacotherapeutic agents in the treatment of obsessive-compulsive disorder. NeuroRx. 2006 Jan;3(1):69–81. doi: 10.1016/j.nurx.2005.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ciraulo DA, Sarid-Segal O, Knapp CM, et al. Efficacy screening trials of paroxetine, pentoxifylline, riluzole, pramipexole and venlafaxine in cocaine dependence. Addiction (Abingdon, England) 2005 Mar;100(Suppl 1):12–22. doi: 10.1111/j.1360-0443.2005.00985.x. [DOI] [PubMed] [Google Scholar]
  • 64.Miller RG, Mitchell JD, Lyon M, et al. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND) Cochrane database of systematic reviews (Online) 2007;(1):CD001447. doi: 10.1002/14651858.CD001447.pub2. [DOI] [PubMed] [Google Scholar]
  • 65.Groeneveld GJ, Van Kan HJ, Kalmijn S, et al. Riluzole serum concentrations in patients with ALS: associations with side effects and symptoms. Neurology. 2003 Oct 28;61(8):1141–3. doi: 10.1212/01.wnl.0000090459.76784.49. [DOI] [PubMed] [Google Scholar]
  • 66.Sorenson EJ. An acute, life-threatening, hypersensitivity reaction to riluzole. Neurology. 2006 Dec 26;67(12):2260–1. doi: 10.1212/01.wnl.0000249182.09143.20. [DOI] [PubMed] [Google Scholar]
  • 67.Pittenger C, Naungayan C, Kendell SF, et al. Visual hallucinations from the addition of riluzole to memantine and bupropion. J Clin Psychopharmacol. 2006 Apr;26(2):218–20. doi: 10.1097/01.jcp.0000203228.64117.9f. [DOI] [PubMed] [Google Scholar]
  • 68.Remy AJ, Camu W, Ramos J, et al. Acute hepatitis after riluzole administration. J Hepatol. 1999 Mar;30(3):527–30. doi: 10.1016/s0168-8278(99)80115-9. [DOI] [PubMed] [Google Scholar]
  • 69.Rodrigo L, Moreno M, Calleja S, et al. Riluzole-induced acute pancreatitis. Am J Gastroenterol. 2001 Jul;96(7):2268–9. [Google Scholar]
  • 70.Cassiman D, Thomeer M, Verbeken E, et al. Hypersensitivity pneumonitis possibly caused by riluzole therapy in ALS. Neurology. 2003 Oct 28;61(8):1150–1. doi: 10.1212/01.wnl.0000082385.33604.1d. [DOI] [PubMed] [Google Scholar]
  • 71.Woolf A, Carstairs SD, Tanen DA. Riluzole-induced methemoglobinemia. Ann Emerg Med. 2004 Feb;43(2):294. doi: 10.1016/j.annemergmed.2003.08.012. [DOI] [PubMed] [Google Scholar]
  • 72.Viallon A, Page Y, Bertrand JC. Methemoglobinemia due to riluzole. N Engl J Med. 2000 Aug 31;343(9):665–6. doi: 10.1056/NEJM200008313430918. [DOI] [PubMed] [Google Scholar]
  • 73.Lipp A, Tank J, Stoffels M, et al. Riluzole and blood pressure in multiple system atrophy. Clin Auton Res. 2003 Aug;13(4):271–4. doi: 10.1007/s10286-003-0100-z. [DOI] [PubMed] [Google Scholar]
  • 74.Scelsa SN, Khan I. Blood pressure elevations in riluzole-treated patients with amyotrophic lateral sclerosis. Eur Neurol. 2000;43(4):224–7. doi: 10.1159/000008180. [DOI] [PubMed] [Google Scholar]
  • 75.Wagner ML, Landis BE. Riluzole: a new agent for amyotrophic lateral sclerosis. Ann Pharmacother. 1997 Jun;31(6):738–44. doi: 10.1177/106002809703100614. [DOI] [PubMed] [Google Scholar]
  • 76.North WA, Khan AM, Yamase HT, et al. Reversible granulocytopenia in association with riluzole therapy. Ann Pharmacother. 2000 Mar;34(3):322–4. doi: 10.1345/aph.19153. [DOI] [PubMed] [Google Scholar]
  • 77.Wokke J. Riluzole. Lancet. 1996 Sep 21;348(9030):795–9. doi: 10.1016/S0140-6736(96)03181-9. [DOI] [PubMed] [Google Scholar]
  • 78.Groeneveld GJ, van Kan HJ, Torano JS, et al. Inter- and intraindividual variability of riluzole serum concentrations in patients with ALS. J Neurol Sci. 2001 Oct 15;191(12):121–5. doi: 10.1016/s0022-510x(01)00613-x. [DOI] [PubMed] [Google Scholar]
  • 79.Sanderink GJ, Bournique B, Stevens J, et al. Involvement of human CYP1A isoenzymes in the metabolism and drug interactions of riluzole in vitro. J Pharmacol Exp Ther. 1997 Sep;282(3):1465–72. [PubMed] [Google Scholar]
  • 80.van Kan HJ, Groeneveld GJ, Kalmijn S, et al. Association between CYP1A2 activity and riluzole clearance in patients with amyotrophic lateral sclerosis. Br J Clin Pharmacol. 2005 Mar;59(3):310–3. doi: 10.1111/j.1365-2125.2004.02233.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.van Kan HJ, van den Berg LH, Groeneveld GJ, et al. Pharmacokinetics of riluzole: evidence for glucuronidation as a major metabolic pathway not associated with UGT1A1 genotype. Biopharm Drug Dispos. 2008 Apr;29(3):139–44. doi: 10.1002/bdd.594. [DOI] [PubMed] [Google Scholar]
  • 82.de Wildt SN, Kearns GL, Leeder JS, et al. Glucuronidation in humans. Pharmacogenetic and developmental aspects. Clin Pharmacokinet. 1999 Jun;36(6):439–52. doi: 10.2165/00003088-199936060-00005. [DOI] [PubMed] [Google Scholar]
  • 83.Fisher MB, Paine MF, Strelevitz TJ, et al. The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev. 2001 Aug-Nov;33(34):273–97. doi: 10.1081/dmr-120000653. [DOI] [PubMed] [Google Scholar]
  • 84.Miners JO, McKinnon RA, Mackenzie PI. Genetic polymorphisms of UDP-glucuronosyltransferases and their functional significance. Toxicology. 2002 Dec 27;181-182:453–6. doi: 10.1016/s0300-483x(02)00449-3. [DOI] [PubMed] [Google Scholar]
  • 85.Colovic M, Caccia S. Liquid chromatographic determination of minocycline in brain-to-plasma distribution studies in the rat. J Chromatogr B Analyt Technol Biomed Life Sci. 2003 Jul 5;791(12):337–43. doi: 10.1016/s1570-0232(03)00247-2. [DOI] [PubMed] [Google Scholar]
  • 86.Colovic M, Zennaro E, Caccia S. Liquid chromatographic assay for riluzole in mouse plasma and central nervous system tissues. J Chromatogr B Analyt Technol Biomed Life Sci. 2004 Apr 25;803(2):305–9. doi: 10.1016/j.jchromb.2004.01.004. [DOI] [PubMed] [Google Scholar]
  • 87.Milane A, Fernandez C, Vautier S, et al. Minocycline and riluzole brain disposition: interactions with p-glycoprotein at the blood-brain barrier. J Neurochem. 2007 Oct;103(1):164–73. doi: 10.1111/j.1471-4159.2007.04772.x. [DOI] [PubMed] [Google Scholar]
  • 88.Urbani A, Belluzzi O. Riluzole inhibits the persistent sodium current in mammalian CNS neurons. Eur J Neurosci. 2000 Oct;12(10):3567–74. doi: 10.1046/j.1460-9568.2000.00242.x. [DOI] [PubMed] [Google Scholar]
  • 89.Wang SJ, Wang KY, Wang WC. Mechanisms underlying the riluzole inhibition of glutamate release from rat cerebral cortex nerve terminals (synaptosomes) Neuroscience. 2004;125(1):191–201. doi: 10.1016/j.neuroscience.2004.01.019. [DOI] [PubMed] [Google Scholar]
  • 90.Noh KM, Hwang JY, Shin HC, et al. A novel neuroprotective mechanism of riluzole: direct inhibition of protein kinase C. Neurobiol Dis. 2000 Aug;7(4):375–83. doi: 10.1006/nbdi.2000.0297. [DOI] [PubMed] [Google Scholar]
  • 91.Mathie A, Veale EL. Therapeutic potential of neuronal two-pore domain potassium-channel modulators. Curr Opin Investig Drugs. 2007 Jul;8(7):555–62. [PubMed] [Google Scholar]
  • 92.Mathew SJ, Amiel JM, Coplan JD, et al. Open-label trial of riluzole in generalized anxiety disorder. Am J Psychiatry. 2005 Dec;162(12):2379–81. doi: 10.1176/appi.ajp.162.12.2379. [DOI] [PubMed] [Google Scholar]

RESOURCES