Review
Glutamatergic medications for the treatment of drug and behavioral addictions

https://doi.org/10.1016/j.pbb.2011.04.015Get rights and content

Abstract

Historically, most pharmacological approaches to the treatment of addictive disorders have utilized either substitution-based methods (i.e., nicotine replacement or opioid maintenance) or have targeted monoaminergic or endogenous opioidergic neurotransmitter systems. However, substantial evidence has accumulated indicating that ligands acting on glutamatergic transmission are also of potential utility in the treatment of drug addiction, as well as various behavioral addictions such as pathological gambling. The purpose of this review is to summarize the pharmacological mechanisms of action and general clinical efficacy of glutamatergic medications that are currently approved or are being investigated for approval for the treatment of addictive disorders. Medications with effects on glutamatergic transmission that will be discussed include acamprosate, N-acetylcysteine, d-cycloserine, gabapentin, lamotrigine, memantine, modafinil, and topiramate. We conclude that manipulation of glutamatergic neurotransmission is a relatively young but promising avenue for the development of improved therapeutic agents for the treatment of drug and behavioral addictions.

Research Highlights

► Glutamate plays a critical role in both drug and behavioral addictions. ► Medications with glutamatergic actions show promise for addiction treatment. ► Mechanisms of action and clinical efficacy of glutamatergic medications are reviewed.

Introduction

Drug addiction, defined by the American Psychiatric Association as substance dependence (American Psychiatric Association, 2002), has numerous maladaptive psychological and behavioral manifestations including: loss of control over drug intake, taking drugs in greater quantities than intended, repeated unsuccessful attempts at quitting or reducing drug use, continued drug use despite negative consequences, and the emergence of drug-specific symptoms of tolerance and/or withdrawal. In addition to numerous intangible humanistic factors such as the disruption of families and interpersonal relationships, social dysfunction, and loss of life, the socioeconomic burden that drug addiction places on society is enormous (Cartwright, 2008, Gilson and Kreis, 2009, Malliarakis and Lucey, 2007, Rehm et al., 2009, Spanagel, 2009, Thavorncharoensap et al., 2009). In recent years it has become evident that the neural substrates underlying addiction to drugs of abuse overlap considerably with those of non-drug “behavioral” addictions (i.e., pathological gambling, pornography/internet addiction, etc.) (Grant et al., 2010a).

To date, medications that have been developed to aid in the treatment of addictive disorders have shown only moderate success. Known barriers that compromise the efficacy of medication-based approaches to treatment to addiction disorders include poor medication compliance, adverse side effects, safety issues, variable medication responses within treatment groups, poor integration of medication management into psychosocial or cognitive-behavioral therapies, inaccessibility to medications or adequate health care, and relapse following discontinuation of the therapeutic medication (Koob et al., 2009, Montoya and Vocci, 2008, O'Brien, 2008, Ross and Peselow, 2009, Zahm, 2010). While numerous medications of various classes that have been approved for other medical conditions are currently being investigated as potential aids in the treatment of addictive disorders, the only medications approved specifically for the treatment thus far in the United States are varenicline, buproprion, and nicotine replacement therapies for smoking cessation, long-acting opioids (i.e., methadone or buprenorphine) for opiate dependence, and disulfiram, naltrexone, and acamprosate for alcohol dependence. No medications to aid in the treatment of addiction to cocaine, methamphetamine, or marijuana are currently approved, nor are any approved for the treatment of behavioral addictions.

The purpose of the present review is to provide a summary of the pharmacological mechanisms of action and general clinical efficacy of medications acting on glutamatergic transmission in the treatment of addictive disorders. These medications include acamprosate, N-acetylcysteine, d-cycloserine, gabapentin, lamotrigine, memantine, modafinil, and topiramate. It should be noted that many of these medications have mechanisms of action that include multiple neurotransmitter systems, and perhaps with the exception of d-cycloserine, none is known to selectively target glutamatergic transmission or specific glutamate receptors. However, there is a strong body of preclinical evidence arising from over two decades of animal studies suggesting a critical role for glutamate transmission and glutamate receptors in drug reward, reinforcement, and relapse (Bird and Lawrence, 2009, Bowers et al., 2010, Gass and Olive, 2008, Kalivas et al., 2009, Moussawi and Kalivas, 2010, Olive, 2009, Olive, 2010, Reissner and Kalivas, 2010, Tzschentke and Schmidt, 2003, Uys and LaLumiere, 2008). For an overview of glutamatergic transmission and glutamate receptors, the reader is referred to the review by Sanacora in the current issue (publisher – please insert correct page numbers here). In addition, the small but growing body of literature on the use of these medications to treat behavioral addictions such as compulsive gambling, and studies on this topic will also be reviewed.

Section snippets

Mechanism of action

Acamprosate (calcium acetylhomotaurine) is derived from homotaurine, a nonspecific γ-aminobutyric acid (GABA) agonist. The molecule is N-acetylated to facilitate penetration across the blood–brain barrier, and is formulated as a calcium salt to increase absorption of the compound from the gastrointestinal tract. Despite these chemical modifications, its overall bioavailability remains poor (i.e., < 20%) and requires doses in the range of 2–3 g per day to demonstrate efficacy. Many pharmacological

Summary and conclusions

With regards to the eight medications reviewed here that possess a glutamatergic mechanism of action (acamprosate, NAC, DCS, gabapentin, lamotrigine, memantine, modafinil, and topiramate), we conclude that NAC, modafinil, and topiramate have the most well-documented and greatest potential for use in the treatment of drug and behavioral addictions. Certainly any of the medications reviewed here will not be a panacea for all addictions, but more likely an effective pharmacological aid to standard

Acknowledgments

The authors would like to thank Katie Ris-Vicari for assistance with the generation of the artwork. This work was supported by NIH grants DA024355, DA025606, and AA013852 (MFO).

References (203)

  • C.A. Dackis et al.

    Modafinil and cocaine: a double-blind, placebo-controlled drug interaction study

    Drug Alcohol Depend

    (2003)
  • M. Davis et al.

    Effects of D-cycloserine on extinction: translation from preclinical to clinical work

    Biol Psychiatry

    (2006)
  • D.J. Dooley et al.

    Inhibition of K+-evoked glutamate release from rat neocortical and hippocampal slices by gabapentin

    Neurosci Lett

    (2000)
  • C. Eroglu et al.

    Gabapentin receptor α2δ-1 is a neuronal thrombospondin receptor responsible for excitatory CNS synaptogenesis

    Cell

    (2009)
  • L. Ferraro et al.

    The effects of modafinil on striatal, pallidal and nigral GABA and glutamate release in the conscious rat: evidence for a preferential inhibition of striato-pallidal GABA transmission

    Neurosci Lett

    (1998)
  • L. Ferraro et al.

    The vigilance promoting drug modafinil increases extracellular glutamate levels in the medial preoptic area and the posterior hypothalamus of the conscious rat: prevention by local GABAA receptor blockade

    Neuropsychopharmacology

    (1999)
  • J.T. Gass et al.

    Glutamatergic substrates of drug addiction and alcoholism

    Biochem Pharmacol

    (2008)
  • G. Gonzalez et al.

    Clinical efficacy of gabapentin versus tiagabine for reducing cocaine use among cocaine dependent methadone-treated patients

    Drug Alcohol Depend

    (2007)
  • J.E. Grant et al.

    N-acetyl cysteine, a glutamate-modulating agent, in the treatment of pathological gambling: a pilot study

    Biol Psychiatry

    (2007)
  • M. Haney et al.

    Smoked cocaine discrimination in humans: effects of gabapentin

    Drug Alcohol Depend

    (2005)
  • K.G. Heinzerling et al.

    Randomized, placebo-controlled trial of baclofen and gabapentin for the treatment of methamphetamine dependence

    Drug Alcohol Depend

    (2006)
  • P.W. Kalivas et al.

    Glutamate transmission in addiction

    Neuropharmacology

    (2009)
  • R.M. Kaminski et al.

    Topiramate selectively protects against seizures induced by ATPA, a GluR5 kainate receptor agonist

    Neuropharmacology

    (2004)
  • K.M. Kampman et al.

    A pilot trial of topiramate for the treatment of cocaine dependence

    Drug Alcohol Depend

    (2004)
  • K.M. Kampman et al.

    Initiating acamprosate within-detoxification versus post-detoxification in the treatment of alcohol dependence

    Addict Behav

    (2009)
  • K.M. Kampman et al.

    A double-blind, placebo-controlled pilot trial of acamprosate for the treatment of cocaine dependence

    Addict Behav

    (2011)
  • K.S. Kau et al.

    Blunted cystine-glutamate antiporter function in the nucleus accumbens promotes cocaine-induced drug seeking

    Neuroscience

    (2008)
  • L.A. Knackstedt et al.

    Glutamate and reinstatement

    Curr Opin Pharmacol

    (2009)
  • L.A. Knackstedt et al.

    The role of cystine-glutamate exchange in nicotine dependence in rats and humans

    Biol Psychiatry

    (2009)
  • L.A. Knackstedt et al.

    Ceftriaxone restores glutamate homeostasis and prevents relapse to cocaine seeking

    Biol Psychiatry

    (2010)
  • G. Lees et al.

    Studies on the mechanism of action of the novel anticonvulsant lamotrigine (Lamictal) using primary neurological cultures from rat cortex

    Brain Res

    (1993)
  • S. Akhondzadeh et al.

    Topiramate prevents ecstasy consumption: a case report

    Fundam Clin Pharmacol

    (2005)
  • M. al Qatari et al.

    Mechanism of action of acamprosate. Part II. Ethanol dependence modifies effects of acamprosate on NMDA receptor binding in membranes from rat cerebral cortex

    Alcohol Clin Exp Res

    (1998)
  • S.L. Amen et al.

    Repeated N-acetylcysteine reduces cocaine seeking in rodents and craving in cocaine-dependent humans

    Neuropsychopharmacology

    (2011)
  • American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 4th Edition, Text Revision....
  • M.L. Andersen et al.

    Dopamine transporter-related effects of modafinil in rhesus monkeys

    Psychopharmacology

    (2010)
  • N. Anderson et al.

    Oral topiramate effective for alcoholism

    J Fam Pract

    (2003)
  • R.M. Anthenelli et al.

    Preliminary evidence for gender-specific effects of topiramate as a potential aid to smoking cessation

    Addiction

    (2008)
  • R.F. Anton et al.

    Combined pharmacotherapies and behavioral interventions for alcohol dependence — the COMBINE study: a randomized controlled trial

    JAMA

    (2006)
  • B. Arbaizar et al.

    Decrease in tobacco consumption after treatment with topiramate and aripiprazole: a case report

    J Med Case Reports

    (2008)
  • A.J. Arias et al.

    Memantine for alcohol dependence: an open-label pilot study

    Addict Disord Their Treat

    (2007)
  • D.A. Baker et al.

    The origin and neuronal function of in vivo nonsynaptic glutamate

    J Neurosci

    (2002)
  • D.A. Baker et al.

    Neuroadaptations in cystine-glutamate exchange underlie cocaine relapse

    Nat Neurosci

    (2003)
  • J.S. Ballon et al.

    A systematic review of modafinil: potential clinical uses and mechanisms of action

    J Clin Psychiatry

    (2006)
  • D.A. Baltieri et al.

    Comparing topiramate with naltrexone in the treatment of alcohol dependence

    Addiction

    (2008)
  • S.P. Berger et al.

    A medication screening trial evaluation of reserpine, gabapentin and lamotrigine pharmacotherapy of cocaine dependence

    Addiction

    (2005)
  • M. Berk et al.

    Nail-biting stuff? The effect of N-acetyl cysteine on nail-biting

    CNS Spectr

    (2009)
  • F. Berton et al.

    Acamprosate enhances N-methyl-d-apartate receptor-mediated neurotransmission but inhibits presynaptic GABAB receptors in nucleus accumbens neurons

    Alcohol Clin Exp Res

    (1998)
  • M.K. Bird et al.

    Group I metabotropic glutamate receptors: involvement in drug-seeking and drug-induced plasticity

    Curr Mol Pharmacol

    (2009)
  • A. Bisaga et al.

    Acute effects of memantine in combination with alcohol in moderate drinkers

    Psychopharmacology

    (2004)
  • Cited by (136)

    • Mechanistic insights into the efficacy of memantine in treating certain drug addictions

      2021, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      This glutamatergic homeostasis is disrupted differently during different stages of drug addiction (Koob and Volkow, 2010). For instance, the use of cocaine, nicotine, or low doses of alcohol causes an increase in synaptic glutamate and an inhibition of NMDA function; conversely, the use of opiates or high doses of alcohol causes a reduction in glutamate and an upregulation in NMDA function in certain brain regions (reviewed in Olive et al., 2012a). In contrast, withdrawal from alcohol is associated with a marked increase in synaptic glutamate, possibly due to reduced glutamate uptake (Abulseoud et al., 2014).

    • Mitochondrial dysfunction and affective disorders: Focus on diet, exercise, and aging

      2021, Mitochondrial Dysfunction and Nanotherapeutics: Aging, Diseases, and Nanotechnology-Related Strategies in Mitochondrial Medicine
    View all citing articles on Scopus
    View full text