You are on page 1of 23

Neuropsychol Rev (2007) 17:275–297

DOI 10.1007/s11065-007-9031-0

Neurocognitive Effects of Methamphetamine: A Critical


Review and Meta-analysis
J. Cobb Scott & Steven Paul Woods & Georg E. Matt &
Rachel A. Meyer & Robert K. Heaton &
J. Hampton Atkinson & Igor Grant

Published online: 13 August 2007


# Springer Science + Business Media, LLC 2007

Abstract This review provides a critical analysis of the related disorders) and neuromedical (e.g., HIV infection)
central nervous system effects of acute and chronic comorbidities. Finally, these findings are discussed with
methamphetamine (MA) use, which is linked to numerous respect to their potential contribution to the clinical
adverse psychosocial, neuropsychiatric, and medical prob- management of persons with MA abuse/dependence.
lems. A meta-analysis of the neuropsychological effects of
MA abuse/dependence revealed broadly medium effect Keywords Central nervous system . Methamphetamine .
sizes, showing deficits in episodic memory, executive Abuse . Dependence . Neuropsychological assessment .
functions, information processing speed, motor skills, Cognition
language, and visuoconstructional abilities. The neuropsy-
chological deficits associated with MA abuse/dependence Methamphetamine (MA) is a potent, addictive psychostim-
are interpreted with regard to their possible neural mech- ulant that has dramatic effects on the central nervous
anisms, most notably MA-associated frontostriatal neuro- system (CNS). This review provides a critical analysis of
toxicity. In addition, potential explanatory factors are the CNS sequelae of MA abuse and dependence, focusing
considered, including demographics (e.g., gender), MA on studies from both the neuropsychological and neuro-
use characteristics (e.g., duration of abstinence), and the imaging literatures. First, we provide a brief introduction to
influence of common psychiatric (e.g., other substance- the prevalence, patterns, and history of MA use, a description
of the acute effects of MA, and an overview of its
J. C. Scott neurotoxicity, including affected neural systems. Next,
Joint Doctoral Program in Clinical Psychology, results from neuroimaging studies are critically reviewed,
San Diego State University and University of California, including those using structural (MR), functional (fMRI),
San Diego, CA 92120, USA
and metabolic (i.e., positron emission technology [PET] and
S. P. Woods : R. A. Meyer : R. K. Heaton : J. H. Atkinson : magnetic resonance spectroscopy [MRS]) techniques. Then
I. Grant we present a meta-analysis of the neuropsychological
Department of Psychiatry (0847), School of Medicine, literature on persons with long-term MA abuse or depen-
University of California,
San Diego, La Jolla,
dence. Extending prior qualitative reviews of this literature
CA 92093, USA (e.g., Meredith et al. 2005; Nordahl et al. 2003), we provide
novel information by arriving at quantitative estimates of
G. E. Matt the potential effects of long-term MA abuse on various
Department of Psychology, San Diego State University,
San Diego, CA 92182, USA
neurocognitive functions. To this end, we provide effect
size estimates for each of nine neurocognitive ability
S. P. Woods (*) domains. The review concludes with a discussion of the
HIV Neurobehavioral Research Center, Department of Psychiatry results from the meta-analysis, potential moderating factors
(0847), University of California, San Diego,
150 West Washington Street, 2nd floor,
(e.g., demographics) and comorbidities (e.g., psychiatric
San Diego, CA 92103, USA disorders), the limitations of previous studies, and areas in
e-mail: spwoods@ucsd.edu which further research is urgently needed.
276 Neuropsychol Rev (2007) 17:275–297

Introduction pseudoephedrine). While data suggest that such measures


have been somewhat effective in reducing MA production
Scope of Methamphetamine Use in the U.S., MA production in Mexico has concurrently
increased and threatens to offset the decline seen domesti-
Although MA was first synthesized in the late 1800s in cally (National Drug Intelligence Center 2006).
Japan, it did not become widely used until World War II, Currently, MA is primarily sold illicitly for recreational
when Japanese, United States, and German military use, although it has been available legally as a pharmaceu-
personnel used it to combat fatigue and increase alertness. tical tablet under the trade name Desoxyn® since the 1940s.
In the U.S., MA was first illegally produced on a large scale There are a few accepted medical reasons for its use, such
during the 1960s by underground labs on the West Coast, as treatment of Attention-Deficit/Hyperactivity Disorder
and abuse of the substance quickly spread throughout that (ADHD), narcolepsy, and obesity, although it is only
area. This burgeoning illicit market was first controlled by available as a prescription that cannot be refilled (Bray
motorcycle gangs in the California Bay Area, only later to 1993; Mitler et al. 1993; National Institute on Drug Abuse
be replaced by Mexican-based distributors, who, along with 2006). The increasing prevalence of illicit MA is in part due
a number of “super-labs” in California and the U.S. to its somewhat easy and cost-efficient synthesis in
Southwest capable of producing large quantities of MA in clandestine laboratories with inexpensive, over-the-counter
a single production cycle, are the primary manufacturers of ingredients. Prior to restrictions placed on phenyl-2-
the drug at the present time (see Anglin et al. 2000 for a propanone (P2P) with the Federal Chemical Diversion and
comprehensive review of the history of MA use and Trafficking Act of 1988, the most popular method for MA
distribution). synthesis was the “P2P method” of reduction. A number of
Methamphetamine abuse has recently been described as different methods currently exist for synthesizing MA from
an “epidemic” in the U.S. due to the rising prevalence rates legal precursor materials (with instructions widely available
throughout the 1990s and into the present (Hunt 1995). over the Internet), but the most popular, cheap, and simple
According to the U.S. 2005 National Survey on Drug Use method is the ephedrine/pseudoephedrine reduction. In the
and Health, approximately 10 million people (around 4.5% U.S., the drug is most commonly found as a colorless
of the U.S. population) have tried MA at some point in their crystalline solid, sold under various street names (e.g.,
lives, while 1.3 million persons reported using MA in the ‘crystal meth,’ ‘ice,’ ‘tina’). However, the drug comes in
past year (Substance Abuse and Mental Health Services many forms and can be administered in a variety of ways,
Administration 2006b). From 1995 to 2002, emergency such as injecting, snorting, smoking, or orally ingesting.
room visits due to MA use increased 88% for persons under The amount administered for a single use varies widely,
18 years old, and 54% across all age groups (Substance depending on route of administration, drug purity, and
Abuse and Mental Health Services Administration 2006a). individual tolerance, although chronic users often use MA
During this same period (1994–2004), there was also a in cycles of binges or “runs” that last for a few days,
dramatic increase in individuals seeking substance abuse followed by periods of abstinence.
treatment with MA as their primary substance of abuse
(Substance Abuse and Mental Health Services Administra- Acute Effects of Methamphetamine Use
tion 2006c). However, recent estimates also indicate that
the MA epidemic in the U.S. may have plateaued, as use The timing, intensity, and spectrum of effects after MA
among the civilian, non-institutionalized population de- administration depend on both the amount of MA con-
clined between 2002 and 2005 (Substance Abuse and sumed and the method of administration. Generally, acute
Mental Health Services Administration 2006b). In addition, MA use results in a number of effects on the sympathetic
the number of individuals using MA for the first time branch of the autonomic nervous system, including hyper-
decreased sharply from 318,000 in 2004 to 192,000 in tension, tachycardia, hyperthermia, increased breathing
2005. Nonetheless, the MA problem is still rampant in the rate, and constriction of blood vessels. The desired
Southwestern area of the U.S. (e.g., San Diego), and cognitive and emotional effects include euphoria, enhanced
indicators point to a spread into other areas of the country energy and alertness, feelings of increased physical and
(Community Epidemiology Work Group 2006; National mental capacity, and a surge in productivity (Cretzmeyer
Drug Intelligence Center 2006). Legal measures have been et al. 2003; Hart et al. 2001). Elevated self-esteem and an
enacted specifically to counter the growing problem of MA increase in libido are also fairly common. Effects from
use, including the Comprehensive Methamphetamine Con- smoking and injection tend to be almost instantaneous,
trol Act of 1996 and the Combat Methamphetamine while ingestion and snorting have more delayed and less
Epidemic Act of 2005, both of which attempt to control intense effects with slower absorption into the body. Given
the precursor materials used for MA production (e.g., that the elimination half-life of MA ranges from 8 to 13 h,
Neuropsychol Rev (2007) 17:275–297 277

its effects typically last for a similar amount of time, which MA-associated automobile accidents or violations, finding
is notably more than other stimulants such as cocaine that all of the accidents were caused by the MA-using
(Busto et al. 1989), and may partly explain the relatively drivers, whose blood concentrations were generally well
quicker progression from first use to addiction in MA users above the therapeutic dose. Moreover, emergency room
(Gonzalez Castro et al. 2000). visits associated with MA use are more likely to present
Binge use, in which the drug is repeatedly administered with various traumas (e.g., injury from assault) and to
over a few hours or days, is common among chronic MA require greater utilization of services than the visits of the
users, resulting in highly elevated blood concentrations of average emergency room patient (Richards et al. 1999).
the drug. During a binge, individuals often exhibit an Clinical studies present a complex picture regarding the
increase in sexual behaviors and repetitive, focused acute effects of MA on cognition. A number of studies have
activities, such as mechanical work or intense periods of reported an enhancement of cognitive performance with
cleaning, often accompanied by sleeplessness (Semple et al. acute administration of therapeutic doses, especially in
2003). The progressively higher doses required to maintain information processing speed, psychomotor functioning,
a high (i.e., tachyphylaxis) can also result in heightened and attention, in both non-drug using volunteers (Johnson
anxiety, paranoia, hallucinations, and even delirium (Harris et al. 2000; Mohs et al. 1978, 1980; Silber et al. 2006) and
and Batki 2000; Smith and Fischer 1970). As a binge stimulant-dependent individuals (Johnson et al. 2005).
progresses, the positive effects of the drug generally However, other studies have shown equivocal effects
become less pleasurable, while toxic effects become more (e.g., Comer et al. 2001), while still others suggest that
prominent, and individuals eventually experience a phase MA leads to deficits in inhibition and filtering out irrelevant
known as “tweaking,” characterized by increased anxiety, information upon acute administration (Mewaldt and
irritability, insomnia, and confusion. A withdrawal syn- Ghoneim 1979). Users who are sleep deprived show better
drome can occur at the termination of binge use and can performance on most tasks with MA administration (Hart
include dysphoria, depression, irritability, anxiety, poor et al. 2005), although the effects generally diminish as more
concentration, hypersomnia, fatigue, paranoia, akathisia, of the drug is metabolized (Wiegmann et al. 1996).
and drug craving (Barr et al. 2002; Zweben et al. 2004). Interpretation of this literature remains difficult, in part
These symptoms usually lessen in intensity within a week due to differing MA dosing regimens, the variety of
after cessation of use, although they are more likely to cognitive tests administered, and variable quality in
occur and are more pronounced in long-term and injection experimental design. In addition, it is unclear how relevant
MA users (Davidson et al. 2001; McGregor et al. 2005; these cognitive enhancements may be for abusers, who
Newton et al. 2004). While subjectively distressing, mostly use well above the therapeutic dose and often smoke
withdrawal from MA (and other stimulants) is not asso- or inject the drug (as opposed to oral administration, which
ciated with the life-threatening symptoms that can accom- is the norm in acute studies). Nonetheless, the existing
pany withdrawal from alcohol or sedative-hypnotic agents literature on acute cognitive effects of MA, combined with
(see Trevisan et al. 1998 for a review). qualitative reports regarding MA use and the literature from
Although a causal relationship has not been adequately other amphetamines (e.g., Soetens et al. 1995), tend to point
demonstrated, MA use is frequently associated with to an increase in attentiveness and speed of processing
increased impulsive, violent, and even homicidal behavior, accompanied by decreases in ability to filter information
as well as unsafe automobile driving. Japan instituted a with therapeutic doses of MA.
strict ban on MA in the 1950s when its use had increased
concurrently with MA-associated crimes and homicides Effects of Chronic Methamphetamine Use
(Anglin et al. 2000). In one study, a large majority of
patients entering treatment reported that their use of MA Long-term MA abuse or dependence can result in a host of
resulted in violent behavior (von Mayrhauser et al. 2002). medical, psychiatric, and psychosocial consequences. A
Similarly, Zweben et al. (2004) reported that 43% of number of cardiovascular effects from chronic MA use
individuals in treatment either reported a history of legal have been reported, including pulmonary hypertension
charges for violent crimes or stated that they had difficulty (Chin et al. 2006), acute aortic dissection (e.g., Davis and
controlling violent behavior. In a population of adult male Swalwell 1994), myocardial infarction (e.g., Hong et al.
California parolees, MA use was predictive of both 1991), and ischemic and hemorrhagic strokes (e.g., Olsen
recidivism (i.e., a return to custody for any reason) and 1977; Yen et al. 1994). In addition, a number of other
violent behavior (Cartier et al. 2006). Logan et al. (1998) detrimental physical and mental health consequences can
found that approximately 45% of deaths involving MA use occur, including nutritional deficiencies, sleep deprivation,
were associated with either suicidal or homicidal violence. and rapid tooth decay, known as “meth mouth” (ADA
In an earlier review, Logan (1996) examined 28 reports of Division of Communications 2005; National Institute on
278 Neuropsychol Rev (2007) 17:275–297

Drug Abuse 2006). A number of cases of MA-related inattention (e.g., difficulty focusing), hyperactivity (e.g.,
mortality have been reported, not only due to excessive use fidgety behavior), and impulsivity (e.g., often interrupts
or overdosing, but also because of hypersensitivity to MA others) (American Psychiatric Association 1994). ADHD can
or the drug’s prominent hypertensive effects (e.g., Delaney persist into adulthood, mostly with a symptom profile of
and Estes 1980; Hall et al. 1973). In addition, MA abusers inattention and distractibility (Faraone et al. 2000), and is a
have significantly elevated infection rates for diseases such risk factor for subsequent substance abuse (Wilens et al.
as human immunodeficiency virus (HIV) and hepatitis C 1995). Although its neuropathogenesis is not completely
virus (HCV) as a result of both injection drug use (i.e., understood, ADHD is currently conceptualized as a disorder
needle sharing) and risky sexual behavior (e.g., unprotected resulting from hypoactivity of dopaminergic systems and
sex), especially among the population of men who have sex imbalance in noradrenergic systems (Biederman 2005; di
with men (Bluthenthal et al. 2001; Gonzales et al. 2006; Michele et al. 2005), implicating cognitive and behavioral
Halkitis et al. 2001; Shoptaw 2006). dysfunction characteristic of an underlying frontal-striatal
Chronic MA abusers also have high rates of affective pathophysiology (Hervey et al. 2004). Moreover, current
distress and psychiatric disorders, which can be especially pharmacologic treatments for ADHD (e.g., methylphenidate)
severe during withdrawal (Newton et al. 2004). Psychosis, target dopaminergic systems, and, similar to MA, operate by
including both negative and positive symptoms, is more blocking reuptake of dopamine and norepinephrine (Pliszka
likely to occur in MA abusing individuals compared to non- 2007). This has led some researchers to propose that
abusing populations, even after adjusting for history of individuals with ADHD may experience more beneficial
psychotic disorders (Buffenstein et al. 1999; Harris and effects from initial MA use, leading to repeated use and
Batki 2000; Iwanami et al. 1994; McKetin et al. 2006; potentially to dependence (Jaffe et al. 2005). Most studies,
Srisurapanont et al. 2003). This psychosis is more likely to however, are cross-sectional in nature, and therefore are not
be seen in MA dependent injection users (Hall et al. 1996; able to establish cause and effect relationships. Thus,
McKetin et al. 2006), but individuals with predispositions whether MA use results in a similar symptom profile to
to psychotic symptoms, including schizotypal or schizoid ADHD, or whether individuals with these symptoms have a
traits and family histories of psychotic disorders, also have predilection for MA due to its initial effects on concentration
an increased risk of MA-associated psychosis (Chen et al. abilities remains debated.
2003, 2005). The presence of psychosis is usually transient, Considering its highly addictive potential, it is not
occurring during use or withdrawal and normally abating in surprising that chronic MA use leads to adverse psychoso-
a period of days. However, susceptibility to psychotic cial and behavioral outcomes. MA users are likely to be
episodes may persist for years after cessation of MA use unemployed and uninsured (Baberg et al. 1996). Given that
(e.g., Sato 1986; Ujike and Sato 2004). Moreover, this illicit MA use often results in irritability, agitation, and
increased susceptibility may be associated with a polymor- numerous other forms of psychiatric distress, chronic MA
phism in the hDAT1 gene (SLC6A3) encoding the abusers experience myriad interpersonal problems, includ-
dopamine transporter (Ujike et al. 2003) and/or noradren- ing poor coping skills, limited social support, and disorga-
ergic hyperactivity (or other metabolite alterations) in nized lifestyles (Cretzmeyer et al. 2003; Halkitis and Shrem
response to mild stressors (Iyo et al. 2004; Yui et al. 2006). Impulsive behavior on the part of MA abusers may
2002). Methamphetamine abusers are also at increased risk exacerbate their psychosocial difficulties and promote
of depression and suicidal ideation during active use, maintenance of drug-seeking behavior, and this impulsivity
withdrawal, and abstinence, and this risk is amplified in may be greater in depressed, younger users, those with low
women and injection users (Domier et al. 2000; Kalechstein educational levels, and those who use large amounts of MA
et al. 2000; Zweben et al. 2004). Abnormalities in the (Semple et al. 2004, 2005).
amygdala or anterior cingulate, areas largely modulated by
dopamine, may contribute to the increase in depressive Methamphetamine and the Brain
symptoms seen in MA abusers (London et al. 2004). MA
users may also have increased rates of antisocial personality Methamphetamine is a synthetic derivative of amphet-
disorder (Chen et al. 2003), as well as mania and bipolar amine, but due to the addition of a methyl group in its
disorder (Shoptaw et al. 2003), but the differential chemical structure, it has a relatively high lipid solubility,
diagnosis of MA-induced mood disorders and primary allowing more rapid transport of the drug across the blood–
mania can be problematic. brain barrier (Barr et al. 2006). Thus, compared with its
Recently, high rates of comorbidity have been observed parent drug, amphetamine, MA exerts more profound
between MA dependence and ADHD (Jaffe et al. 2005; effects on the CNS. Methamphetamine use principally
Matsumoto et al. 2005; Sim et al. 2002), which is a results in a cascading release of dopamine, but also of
neurodevelopmental disorder characterized by marked other monoamine neurotransmitters, including norepineph-
Neuropsychol Rev (2007) 17:275–297 279

rine and serotonin (Kokoshka et al. 1998). Animal models vesicular monoamine transporter type-2 [VMAT2]),
suggest that MA administration stimulates the release of changes that might be expected with damage to dopamine
dopamine via a number of different molecular mechanisms, nerve terminals. Interestingly, a later study from the same
including displacement of storage vesicles and inhibition of group found that dopamine levels were more severely reduced
monoamine oxidase (Zaczek et al. 1990), although the in the caudate than in the putamen at autopsy, an opposite
dopamine transporter (DAT) may be the primary site of pattern as is seen in Parkinson’s disease, perhaps explaining
action (Giros et al. 1996). In addition to increasing the the relatively low prevalence of parkinsonian motor symp-
efflux of dopamine, MA enhances DAT-mediated reverse toms observed in chronic MA abusers (Moszczynska et al.
transport of dopamine across the plasma membrane, 2004). One possible implication of these findings is that
thereby further increasing the dopamine concentration into cognitive functions may be affected more so than motoric
the synapse (Khoshbouei et al. 2003). abilities in MA abuse compared to Parkinson’s disease,
Methamphetamine-induced neurotoxicity is evident in because the caudate has more cognitive pathways (i.e.,
several neurotransmitter systems, but it is most notable in the three prefrontal-caudate-globus pallidus/substantia
nigrostriatal dopaminergic pathways, thus altering the nigra-thalamo-cortical loops) than the putamen (i.e., the
function of the dopamine rich fronto-striato-thalamo- supplementary motor-putamen-globus pallidus/substantia
cortical loops (Cass 1997). Although an acute, moderate nigra-thalamo-cortical loop; Alexander et al. 1986).
dose of MA is unlikely to reduce dopamine stores
permanently (e.g., Chan et al. 1994), a series of high-dose Neuroimaging of Methamphetamine Abuse
administrations in experimental animals results in a and Dependence
significant reduction in levels of dopamine and serotonin
and their metabolites, as well as tyrosine hydroxylase and Given the effects of MA on dopaminergic, as well as
tryptophan hydroxylase activity, enzymes involved in the perhaps serotonergic and GABAergic (e.g., Burrows and
synthesis of dopamine and other catecholamines (Axt and Meshul 1999) systems, a number of studies have investi-
Molliver 1991; Bakhit et al. 1981; Hotchkiss and Gibb gated the impact of MA abuse and dependence on the
1980; Morgan and Gibb 1980). Both high dose and chronic structure and function of frontostriatal and limbic systems.
administration of MA can result in depletion of dopamine, For example, studies using positron emission tomography
destruction of dopamine nerve terminals, and long-term (PET), which utilizes ligands that bind to transporters,
reduction in other markers of dopamine terminal integrity, provide in vivo insights into dopaminergic and serotonergic
although dopaminergic neuron cell bodies themselves do dysregulation in MA users. One study reported that MA
not seem to be destroyed (Harvey et al. 2000; Miller and abusers evidence reduced serotonin transporter density (a
O’Callaghan 2003; Ricaurte et al. 1982; Wagner et al. marker of the number or integrity of serotonin terminals),
1980). A number of pathways have been implicated in MA- which may be associated with increased aggressive behav-
induced neurotoxicity, including production of reactive ior (Sekine et al. 2006). Regarding the effect of MA on the
oxygen and nitrogen species, hyperthermia, or triggering dopaminergic system, McCann et al. (1998) first demon-
of an apoptotic cascade dependent upon mitochondria, but strated a reduction in striatal DAT levels in a small sample
the exact processes are still unclear (see Cadet et al. 2005; of abstinent MA abusers. Similar findings were reported by
Davidson et al. 2001 for more thorough reviews). More- Volkow et al. (2001c) and Sekine et al. (2001, 2003), who
over, evidence from experimental animal studies indicates observed low DAT availability in the striatum and prefron-
that MA neurotoxicity brought about by binge-like admin- tal cortex of MA users ranging from 20% to 33% relative to
istration might be partially reversible with time, although healthy comparison participants. Although these DAT
this recovery is likely dependent on the amount and dosing reductions are generally lower than those found in
of MA exposure (Friedman et al. 1998; Harvey et al. 2000) Parkinson’s disease (e.g., McCann et al. 1998), they are
and is likely incomplete (Woolverton et al. 1989). The nonetheless associated with impaired psychomotor and
mechanisms of recovery are unclear, but may be partly episodic memory functioning (Volkow et al. 2001c),
explained by the sprouting of remaining axons or a increased severity of psychiatric symptoms (Sekine et al.
compensatory increase in monoamine levels (Cass and 2001, 2003), and longer duration of MA use (Sekine et al.
Manning 1999; Friedman et al. 1998). 2001, 2003; Volkow et al. 2001c). Whether these DAT
Studies of MA-associated neurotoxicity in humans have changes reflect down-regulation in transporter expression,
been limited until recent years. Wilson et al. (1996) found a as suggested by reduced DAT with relative sparing of
reduction in levels of dopamine, DAT, and tyrosine VMAT2 (i.e., a more stable indicator of the structural
hydroxylase in postmortem striatum of chronic MA integrity of dopamine terminals; Wilson et al. 1996), or
abusers, although they did not find reductions in levels of actual dopamine terminal degeneration (e.g., Johanson et al.
other dopamine transporters and synthetic enzymes (e.g., 2006) remains unclear. Importantly, however, DAT avail-
280 Neuropsychol Rev (2007) 17:275–297

ability may recover in a time-dependent fashion with MA users consistently reveal grey matter volumetric
extended abstinence from MA (Volkow et al. 2001b) but abnormalities, but have differed in directionality (i.e.,
not necessarily to pre-MA levels (Johanson et al. 2006) or smaller versus larger volumes) and their associations with
with parallel cognitive normalization (Volkow et al. 2001b). MA use characteristics, demographics, and neuropsycho-
Several studies have also employed 1H magnetic logical impairment. Thompson et al. (2004) first published
resonance spectroscopy (MRS) methods to investigate the on structural brain alterations in MA-dependent persons in
possible metabolic alterations in chronic MA use. A a study that showed abnormally small limbic (e.g.,
majority of these studies indicate that individuals with cingulate) and hippocampal volumes, with the latter being
MA dependence show low levels of N-acetylaspartate associated with word recall impairment. Using a compre-
(NAA), a marker of neuronal integrity, relative to age- hensive morphometric approach, Jernigan et al. (2005)
and gender-matched comparison volunteers. Low NAA is reported that MA dependence was associated with abnor-
apparent in the basal ganglia, frontal white matter, and mally large volumes in the striatum, nucleus accumbens,
anterior cingulate (Ernst et al. 2000; Nordahl et al. 2002; and parietal lobes. Greater cortical volumes were not related
Sung et al. 2006; cf. Sekine et al. 2002) and is associated to psychiatric comorbidity or MA use characteristics, but
with greater lifetime MA use (Ernst et al. 2000; Sung et al. were associated with greater severity of global neuro-
2006). However, a recent study of 44 well-characterized cognitive impairment. Similarly, Chang et al. (2005a) found
MA dependent persons who were abstinent for an average larger basal ganglia (i.e., putamen and globus pallidus)
of 3 months found no differences in NAA (Taylor et al. volumes in chronic MA users; however, in contrast to
2007), raising questions about the persistence of the NAA Jernigan et al. (2005), smaller striatal volumes were
effects. MA users also show reduced basal ganglia levels of associated with greater severity of cognitive impairment
creatine (a marker of metabolic [e.g., mitochondrial] and greater cumulative MA use. Although somewhat
disturbance), which is correlated with duration of use surprising, the larger basal ganglia volumes apparent in
(Sekine et al. 2002) and psychiatric symptom severity MA users may represent a functional compensatory
(Iyo et al. 2004; Sekine et al. 2002). Elevated choline (a response, inflammatory mechanisms, neuritic growth, or
marker of cellular membrane synthesis and turnover) in the glial activation (Chang et al. 2005a; Jernigan et al. 2005).
anterior cingulate and frontal grey matter is also evident in In addition to grey matter abnormalities, several studies
MA dependent individuals (Ernst et al. 2000; Salo et al. have revealed evidence of white matter pathology in
2007) and may correlate with duration of MA use (Taylor et persons dependent on MA. Perhaps reflecting underlying
al. 2007). Finally, some studies showed abnormally high cerebrovascular disease (e.g., decreased perfusion of brain
levels of myo-inositol (a marker of glial activation) in the parenchyma), male MA abusers demonstrate greater sever-
frontal grey (Ernst et al. 2000) and left frontal white (Sung ity of white matter hyperintensities, which correlate with
et al. 2006) matter. Taken together, these MR spectroscopy lifetime quantity of MA use in the deep white matter (Bae
studies suggest that a variety of different neuropathological et al. 2006). Volumetric analyses have shown bilaterally
processes may be associated with MA dependence, includ- greater temporal, occipital, and cingulate white matter
ing neuronal injury or death, astrocytosis, cellular mem- volumes in MA dependent individuals, with the latter being
brane alterations, and dysregulation of energy metabolism. more pronounced in the right hemisphere (Thompson et al.
Not all studies have shown MA effects on these metabo- 2004). Chronic MA users have also been found to
lites, however (e.g., Taylor et al. 2007), perhaps due to demonstrate corpus callosum irregularities, including great-
differences in MA use severity and prevalence of comorbid er curvature in the genu and smaller width in posterior
conditions across study cohorts, which raises the possibility regions (e.g., isthmus; Oh et al. 2005). In the only diffusion
that brain metabolite changes occur when MA use rises tensor imaging study of MA to date, Chung et al. (2006)
over some threshold or that certain populations of MA found lower fractional anisotropy (i.e., less directional
users might be unusually resistant to MA-associated diffusivity of water, possibly indicating decreased parallel
neurotoxicity. Given the possible role of glutamergic ordering of axons) bilaterally in the frontal white matter of
alterations in MA-associated neurotoxicity (Brown et al. 32 MA abusers compared with 30 healthy comparison
2005), future studies of MA users might use 13C spectros- subjects. The reduced white matter anisotropy was more
copy to provide a dynamic evaluation of glutamate- prominent in men, particularly the right frontal region,
glutamine cycling (for further reading on this method, see which was associated with a greater number of errors on the
Mason and Rothman 2004; Ross et al. 2003). Wisconsin Card Sorting Test (WCST; Chung et al. 2006).
More recently, attention has turned to the possible effects Considering the marked cardiovascular and neurotoxic
of MA-associated neurotransmitter (e.g., dopaminergic) and effects of MA use, changes in regional cerebral blood flow
metabolic (e.g., NAA) disturbances on the structural (rCBF) may accompany MA dependence. Iyo et al. (1997)
integrity of brain parenchyma. Structural MRI studies of reported qualitative “focal perfusion deficits” in six out of
Neuropsychol Rev (2007) 17:275–297 281

nine chronic MA users. Using quantitative perfusion MR have yielded contradictory findings. Second, both the
imaging techniques, Chang et al. (2002) demonstrated demographic (e.g., age) and drug use characteristics of the
lower relative rCBF in the frontal cortex and basal ganglia participants studied have varied widely. For example, some
of 20 MA dependent individuals as compared with age- and studies assess treatment-seeking samples that have been
gender matched comparison volunteers. Commensurate abstinent for at least 30 days (with wide variability in actual
with previously reported metabolite (Nordahl et al. 2002) abstinence length), whereas other studies assess individuals
and gray matter density (Thompson et al. 2004) abnormal- who are currently using MA and are only abstinent on the
ities in the anterior cingulate, an area with a high density of day of testing. Such discrepancies create difficulty in
dopamine fibers, Hwang et al. (2006) observed lower estimating and interpreting the effects of chronic MA use
relative rCBF in the right anterior cingulate cortex in MA on neuropsychological functioning. Meta-analyses repre-
users. These changes were less pronounced in individuals sent one potentially powerful way to clarify contradictory
with prolonged abstinence from MA when compared to findings and possible explanatory variables across a
recently abstinent users, although rCBF was still lower than literature, and may more accurately reveal the effects of
healthy comparison participants. As measured by fMRI, chronic MA use on cognition. Thus, we conducted a meta-
chronic MA users show alterations in blood-oxygen- analysis to evaluate the hypothesis that chronic MA use is
dependent level (BOLD) signals in both prefrontal and associated with deficits in neuropsychological functioning.
parietal brain regions during decision-making when com- In addition, we proposed the following exploratory ques-
pared with healthy comparison volunteers (Monterosso et tions: (1) To what extent do demographic characteristics
al. 2006; Paulus et al. 2002, 2003). These BOLD signal and comorbid drug use influence differences between
changes are particularly evident in the dorsolateral and groups? and (2) What are the effects of MA use variables
orbitofrontal cortices (Paulus et al. 2002), which may be (e.g., abstinence from MA, duration of MA use) on the
mediated by low levels of dopamine D2 receptors (Volkow neuropsychological sequelae of chronic MA use?
et al. 2001a). Interestingly, prefrontal, parietal, and insular
hypoactivation during decision-making is also predictive of
incident MA relapse, thus demonstrating the potential Meta-analysis
relevance of MA-related CNS effects to treatment outcomes
(Paulus et al. 2005). Studies and Variables
It is unknown whether the changes in brain structure and
function evident in persons with MA dependence are Before initiating a literature search, we identified a set of
reversible. Large-scale outcome studies that have followed inclusion criteria that would allow us to best answer the
MA users over longer periods of time (i.e., years) do not principal research question: “What are the neuropsycholog-
exist, and therefore we do not know if any or all of the ical sequelae of long-term methamphetamine abuse and
brain changes will remit with prolonged abstinence. If dependence?” These study inclusion criteria were (1)
research in the CNS effects of alcoholism is a reasonable assessed human adults aged 18 years and older; (2)
guiding model (e.g., Grant 1987), then one might predict published in English; (3) members of the MA group were
that sustained abstinence from MA would be associated classified specifically as having lifetime histories of MA
with substantial recovery in brain structure and function. dependence or abuse (studies that only included amphet-
amine or stimulant dependent/abusing participants were
Neuropsychological Aspects of Methamphetamine Abuse eligible only if they reported separate statistics for a MA
and Dependence group) and identified MA as their primary substance of use;
(4) healthy comparison participants did not have a history
Extending the neurobiological and neuroimaging litera- of stimulant dependence or any severe neurologic or
tures, a number of studies have suggested that chronic MA psychiatric illnesses; (5) MA was not administered by the
use is often associated with mild-to-moderate neuropsycho- experimenters; (6) outcome measures included at least one
logical impairment. Current estimates suggest that approx- standardized neuropsychological test; and (7) sufficient data
imately 40% of persons with MA dependence demonstrate were provided to calculate effect sizes. These criteria were
global neuropsychological impairment (Rippeth et al. intentionally liberal in order to be more inclusive, which
2004). However, interpretation of the neuropsychological has the benefits of providing a more complete representa-
literature remains difficult due to a number of methodolog- tion of the existing research on the cognitive effects of MA
ical factors. First, only a handful of studies have used a and using an empirical approach to examine the relation-
comprehensive neuropsychological battery that assesses ships between various methodologies and study findings.
functioning in a number of cognitive domains, and such Preliminary literature searches using terms such as
studies have used divergent tests within their batteries and amphetamine, methamphetamine, cognition, neuropsycho-
282 Neuropsychol Rev (2007) 17:275–297

logical, and domain-specific keywords (e.g., memory, (1) simple reaction time; (2) attention/working memory
attention) were independently conducted by two of the (e.g., Wechsler Adult Intelligence Scale, 3rd Edition
investigators (JCS and RAM) through several online data- [WAIS-III] Digit Span, N-Back tasks); (3) executive
bases, including PubMed, PsychInfo, and ISI Web of functions (e.g., WCST, Stroop Color-Word interference
Science. Any article published prior to January 2007 was condition); (4) learning (e.g., California Verbal Learning
considered eligible. All articles identified as potentially Test [CVLT] trials 1–5); (4) memory (e.g., CVLT Delayed
eligible were manually reviewed to ensure that the criteria Recall); (5) motor (e.g., Grooved Pegboard); (6) language
for inclusion (specified above) were met. We also manually (e.g., Verbal Fluency, WAIS-III Vocabulary); (7) speed of
reviewed each article to examine for any potential omis- information processing (e.g., WAIS-III Digit Symbol); and
sions in our literature search. In addition, all possibly (8) visuoconstruction (i.e., Rey Complex Figure Copy). If
eligible studies published by the same group of authors multiple subtests assessing the same cognitive construct
were carefully reviewed to minimize the inclusion of were reported (e.g., CVLT Delayed Free Recall and Cued
overlapping data from a single participant cohort. After Recall), the subtest with the best evidence of construct
narrowing down the pool of possibly eligible studies, a total validity (based on consensus) was chosen for inclusion
of 18 studies with 951 participants, including 487 partic- (e.g., CVLT Delayed Free Recall). Table 1 lists the tests
ipants with MA abuse/dependence and 464 normal com- that were included in each cognitive domain along with
parison participants, were deemed eligible for inclusion. their frequency. Measures for which low scores indicate
The following information was extracted from each study: better performance were adjusted accordingly to assure that
(1) demographic variables (e.g., age and education); (2) a negative d indicated that the MA group performed worse
MA use characteristics (e.g., length of MA use and average than the normal comparison group.
length of abstinence from MA); (3) cohort factors (e.g.,
comorbid substance use); (4) sample size; and (5) summary Statistical Analyses
statistics for the calculation of effect sizes. Two authors
(JCS and SPW) independently coded each study, and any A mixed-effects multivariate model was used in our meta-
disagreements (e.g., due to a lack of clarity in studies analysis computations for a number of theoretical and
regarding participant characteristics) were resolved by practical reasons (for a more thorough review of these
conference consensus. models, see Arends et al. 2003; Kalaian and Raudenbush
1996). Multivariate meta-analysis allows more than one
Effect Size Calculation outcome per study (i.e., more than one test per neuro-
cognitive domain per study) by modeling the correlation
For each neuropsychological test that was administered in between multiple dependent variables in a study (i.e., by
these 18 studies, an effect size and its variance were modeling both fixed and random elements). In addition, the
calculated. The effect size used in this meta-analysis was model is flexible in allowing different numbers of effect
the standardized mean difference statistic (d). When sizes per study. Furthermore, a multivariate mixed-effects
possible, this statistic was calculated as d=(Me −Mc)/Sp, model for meta-analysis allows us to increase generaliz-
where M e and M c are the mean raw scores on a ability and make inferences about the population of studies
neuropsychological test for the MA using and normal on the neurocognitive effects of chronic MA use, including
comparison groups, respectively, and Sp is the pooled ones that differ from the included studies in such factors as
within-group standard deviation. Where these data were participants, MA use characteristics, and outcome mea-
not reported, standardized mean difference effect sizes were sures, instead of solely allowing inferences about this
derived from t-values based on independent t-tests or F- particular set of studies (a limitation of fixed effects
ratios from a two-group one-way analysis of variance models).
(Shadish et al. 1999). Hedges and Olkin’s (1985) correction A two-level mixed-effects model was used to examine
for small sample bias was applied to all effect sizes. The the variability of effect sizes between studies (random
variance for each d value was then calculated and used to factor) and the association between explanatory variables
determine a weighting factor for the unbiased effect size. (fixed factors) and effect sizes. We first tested the following
We coded 157 effect sizes from the 18 studies, with a simple model without explanatory variables to estimate an
range of 1 to 19 effect sizes and a mean of 8.72 (SD=7.38) overall mean effect size and the between-study variance:
per study. When studies offered results from multiple    
neurocognitive tests, the battery was independently yij ¼ α þ uj þ eij uj  N 0; σ2u eij  N 0; s2ij
reviewed by the raters who classified the tests into domains.
In the event of disagreement, the raters determined the where yij refers to effect sizes (i) within studies (j), α is a
domains for each test by consensus. These domains were: constant (i.e., the overall mean), uj are the study-level
Neuropsychol Rev (2007) 17:275–297 283

Table 1 Neuropsychological tests categorized by cognitive domain Table 1 (continued)

Domain Domain

Test k % Test k %

Attention/Working memory WAIS-III symbol search 1 5.26


CalCAP choice reaction time 2 11.11 WAIS-R digit symbol 2 10.53
RAVLT trial 1 1 5.56 Symbol digit 2 10.53
PASAT 1 5.56
WAIS-III letter-number sequencing 1 5.56 Verbal
Delayed match to sample 2 11.11 Letter fluency 6 54.55
Woodcock–Johnson backward digit span 2 11.11 Category fluency 2 18.18
WCST failure to maintain set 1 5.56 Shipley vocabulary 3 27.27
Continuous performance test RT 1 5.56
WMS-III letter-number sequencing 1 5.56 Motor skills
WMS-III visual memory span 1 5.56 Timed gait 1 16.67
N-Back (1 and 2) 2 11.11 Grooved pegboard 4 66.67
CVLT trial 1 1 5.56 Finger tapping 1 16.67
CANTAB spatial working memory 1 5.56
RVIP CANTAB 1 5.56 Reaction time
CalCAP simple reaction time 2 66.66
Abstraction/Executive functions Continuous performance test 1 33.33
CalCAP response reversal 1 3.23
Trail making test, part B 7 22.58 Visuoconstruction
Stroop interference 8 25.81 Rey complex figure copy 3 100.00
Category test 1 3.23
WCST 4 12.90 % = Percent of journal articles within each domain that included the
Shipley abstract thinking 2 6.45 neuropsychological test in the primary source, k = number of studies,
Response inhibition/switching task 1 3.23 CalCAP = California Computerized Assessment Package, RAVLT =
Rey Auditory Verbal Learning Test, PASAT = Paced Auditory Serial
Two-choice prediction task 1 3.23
Addition Test, WAIS = Wechsler Adult Intelligence Test, WCST =
Delay discounting task 2 6.45
Wisconsin Card Sorting Test, RT = Reaction Time, WMS = Wechsler
Stop signal reaction time 1 3.23 Memory Test, CVLT = California Verbal Learning Test, CANTAB =
Ruff figural fluency 1 3.23 Cambridge Automated Neuropsychological Assessment Battery, RVIP =
CANTAB extradimensional shift 1 3.23 Rapid Visual Information Processing, HVLT-R = Hopkins Verbal Learning
Stockings of Cambridge 1 3.23 Test-Revised, BVMT-R = Brief Visuospatial Memory Test-Revised, PAL =
Paired Associates Learning.
Memory
RAVLT delay 3 18.75
Rey complex figure recall 3 18.75
random effects, and eij is the effect-size level residual. s 2u is
HVLT-R delay 1 6.25
the variance parameter to be estimated for the between-
BVMT-R delay 1 6.25
Repeated memory word 2 12.50 study variance, and s2ij are the known conditional variances
Repeated memory picture 2 12.50 of the effect sizes. This analysis revealed that the overall
WMS-III logical memory 1 6.25 mean effect size was d=−0.558 (SE=0.034) and the
Babcock story 1 6.25 between-study variance estimate was 0.037 (SE=0.012,
CVLT delay 1 6.25 p<0.01), indicating that the variance between studies was
CANTAB PAL 1 6.25 significantly more than what could be explained solely by
sampling error alone.
Learning
HVLT-R total 1 14.29
The significance of the between-study variance prompt-
BVMT-R total 1 14.29 ed an exploration as to whether research group, neuro-
RAVLT total 3 42.86 cognitive test domain, average gender proportion, average
Babcock story immediate 1 14.29 education level, or comorbidity could account for some of
CVLT trial 5 1 14.29 the between-study variance. Because the relatively small
number of studies did not provide enough degrees of
Speed of information processing freedom, we could not jointly examine main or interaction
Trail making test, part A 7 36.84
effects. Therefore, the explanatory variables were examined
Stroop color/word 5 26.32
WAIS-III digit symbol 2 10.53 separately, and findings from these analyses should be
interpreted cautiously.
284 Neuropsychol Rev (2007) 17:275–297

To examine single explanatory variables, we fit the dependence and 445 normal comparison participants, with an
following model: average of 53.88 participants per study (range=19–130).
Table 2 presents the included participants’ demographic data
yij ¼ α þ βxij þ uj þeij  and MA use characteristics, and Table 3 presents a summary
 
uj  N 0; σ2u eij  N 0; s2ij of the studies used in the meta-analysis.

where β is the regression slope associated with the


explanatory variable. Results
All models were fit using program gllamm of Stata
version 9.2 (Grilli and Rampichini 2006; Rabe-Hesketh et Overall, significant differences in mean effect size estimates
al. 2004; StataCorp 2006). The level 1 variances of the were found across neurocognitive test domains (χ2 =23.6,
effect sizes were fixed to the estimates of the conditional p<0.01). Figure 1 displays the mean weighted effect sizes
effect-size variances. and 95% confidence intervals for each neurocognitive
domain across studies. The mean effect sizes for each
Research Groups domain across the 17 studies ranged from −0.34 to −0.66.
With the exception of simple reaction time, the 95%
Preliminary analyses indicated that effect size estimates confidence interval surrounding the mean effect size for
differed significantly between research groups (χ2(10)=30.1, each domain did not contain zero, and thus all effect sizes
p<0.001), which reduced the between study variance to in each domain were significantly different from zero. By
0.026 (SE=0.023). Thus, differences between research convention, d-values of 0.2, 0.5, and 0.8 correspond to
groups accounted for a significant proportion of between- small, medium, and large effect sizes, respectively (Cohen
study differences, and the remaining between-group variance 1988), although it should be noted that these categoriza-
was predominantly due to sampling error. Among the tions are broad and do not necessarily signify levels of
research groups, effect sizes reported by one research group practical significance. As illustrated in Fig. 1, the largest
(Gonzalez et al. 2007) were the largest overall (d=−1.37, effect sizes were seen in the domains of learning (d=−0.66),
SE=0.248), and significantly larger than all but those of two executive functions (d=−0.63), and memory (d=−0.59),
research groups (p<0.05). We cannot statistically address the which were all in the medium-to-large range. Effect sizes
reasons for these differences, as analyses to test these of a medium magnitude were observed in the domains of
hypotheses were not possible due to diminishing degrees of speed of information processing (d=−0.52) and motor
freedom. Several possibilities may account for this discrep- skills (d=−0.48), with slightly smaller effects in attention/
ancy, including significant demographic differences (i.e., a working memory (d=−0.39), visuoconstruction (d=−0.37),
higher proportion of men in the MA group) and the high and language (d=− 0.34). Visual inspection of a funnel
prevalence of polysubstance use disorders in the MA group. plot (Duval and Tweedie 2000) showed no significant
Due to these divergent study characteristics and the potential evidence of publication bias.
influence of the unusually large effect sizes, we chose to
exclude this study from subsequent analyses and limit our Demographic Variables
analyses to the remaining research groups. However, the
results outlined below did not change appreciably when this Analyses examining the associations between study char-
study was included in analyses. This exclusion left 17 studies acteristics and effect size estimates were performed with a
containing 916 participants: 471 participants with MA abuse/ number of explanatory variables, including participant

Table 2 Participants’ demographic data and methamphetamine use characteristics

k MA group NC group

N Mean (SD) Range N Mean (SD) Range

Age 17 471 34.62 (7.83) 31.1–41.4 425a 32.75 (8.44) 25.7–38.7


Education 15 443 12.47 (1.59) 10–13.7 390 13.87 (1.69) 12.8–15.3
% male 17 471 65.21 36.1–100 445 61.8 33.3–100
Length of MA use (months) 14 395 123.58 (60.62) 30.7–207
Length of MA abstinence (days) 12 395 294.89 (359.37) 0–1241

k=Number of studies, MA = methamphetamine, N=number of participants, NC = normal comparison.


a
k=16.
Neuropsychol Rev (2007) 17:275–297 285

Table 3 Summary of studies used in the meta-analysis

Authors Year MA group NC group NP domains assessed


(N) (N)

Chang, Ernst, Speck, et al. 2002 18 20 Attention/WM, simple RT, executive functions
Chang, Cloak, Patterson, 2005 43 28 Attention/WM, executive functions, learning, memory, motor, verbal, SIP,
et al. visuoconstruction
Gonzalez, Bechara, and 2007 16 19 Attention/WM, executive functions
Martina
Hoffman, Moore, Templin, 2006 41 41 Executive functions, learning, memory, motor, verbal, SIP,
et al. visuoconstruction
Johanson, Frey, Lundahl, 2006 16 18 Attention/WM, executive functions, learning, memory, motor, verbal, SIP
et al.
Kalechstein, Newton, and 2003 27 18 Attention/WM, executive functions, learning, memory, verbal, SIP,
Green visuoconstruction
Kim, Lyoo, Hwang, et al. 2005 33 21 Executive functions
London, Berman, Voytek, 2005 17 16 Attention/WM, simple RT
et al.
Monterosso, Ainslie, Xu, 2006 12 17 Executive functions
et al.
Monterosso, Aron, 2005 11 29 Executive functions
Cordova, et al.
Newton, Kalechstein, 2004 9 10 Attention/WM, simple RT
Hardy, et al.
Paulus, Hozack, Frank, 2003 14 14 Executive functions
et al.
Rippeth, Heaton, Carey, 2004 47 60 Attention/WM, executive functions, learning, memory, motor, verbal, SIP
et al.
Salo, Nordahl, Moore, et al. 2005 34 20 Executive functions
Salo, Nordahl, Natsuaki, In 36 16 Executive functions
et al. press
Salo, Nordahl, Possin, et al. 2002 8 12 Executive functions
Simon, Domier, Carnell, 2000 65 65 Attention/WM, executive functions, memory, verbal, SIP
et al.
Simon, Domier, Sim, et al. 2002 40 40 Attention/WM, executive functions, memory, verbal, SIP

MA = methamphetamine, N=number of participants, NC = normal comparison, NP = neuropsychological, RT = reaction time, SIP = speed of
information processing, WM = working memory.
a
Study excluded from final analyses.

demographics (e.g., mean age, mean years of education, more positive at a trend level, indicating less performance
gender proportion) and drug use characteristics (e.g., difference between the groups (bb =+0.05; p=0.09). Finally,
average length of abstinence from MA, exclusion of as a majority of studies (76%) did not match MA and
comorbid drug use). Including gender in a model with the normal comparison groups on education (see Table 2), a
neurocognitive test domains showed that for every 1% variable representing the raw difference in years of
increase of men in the MA group, the magnitude of the education between these groups was created for each study
effect size estimate (i.e., the difference between the groups) to examine the influence of this difference on the
increased by −0.01 (b b =−0.01; p<0.001), indicating greater magnitude of effect size. This analysis revealed that there
performance discrepancy between the groups. Similarly, was not a significant influence of educational discrepancy
with every 1% increase of men in the normal comparison on study effect sizes (b
b =0.06; p=0.13).
group, the magnitude of the effect size estimate became
slightly more positive (b b =+0.01; p<0.01). Results also Methamphetamine Use Variables
revealed that for each unit increase (i.e., year) in the
average age of the MA group in a study the magnitude Analyses of MA use variables revealed that neither average
of the effect size estimate increased by −0.04 (b b =−0.04; length of abstinence from MA nor average duration of
p<0.05). Similarly, with increasing age in the normal lifetime MA use had an appreciable influence on the
comparison group, the magnitude of the effect size became magnitude of the effect size (ps>0.10). We also created a
286 Neuropsychol Rev (2007) 17:275–297

Fig. 1 Mean effect sizes and


95% confidence intervals for
each neurocognitive test do-
main. *k=18, mean=grand
mean effect size, LB = lower
bound, UB = upper bound,
SIP = speed of information
processing, WM = working
memory, CI = confidence
interval

dichotomous variable to indicate whether studies had Cognitive and Motor Processes Affected
excluded individuals from the MA group with prior in Methamphetamine Users
substance-related disorders other than MA (excluded,
k=6; did not exclude, k=11). This variable also exhibited Of particular interest, MA use was associated with marked
no significant change on the magnitude of effect sizes detrimental effects on episodic memory, which is also
(p>0.10). among the most susceptible cognitive functions to MA
relapse (Simon et al. 2004). Interpretation of the medium-
to-large effects in MA users is complicated by the fact that
Discussion episodic memory is a multifactorial ability (e.g., encoding,
consolidation, and retrieval) subserved by multiple brain
A wealth of animal and human literature implicates chronic regions (e.g., frontal and temporal systems). Nevertheless,
MA use in metabolic and neurotransmitter abnormalities, we hypothesize that the MA effect on episodic memory is
structural brain alterations, and deficits in everyday func- driven by deficient executive aspects of encoding and
tioning, which are most often attributed to neurotoxicity in retrieval, likely related to MA-associated neurotoxicity in
dopaminergic and serotonergic fronto-striatal-thalamo- the frontostriatal circuits. In support of this hypothesis, a
cortical circuits. It has been hypothesized that these slightly larger effect was observed on learning as compared
alterations may also contribute to significant neuropsycho- with delayed recall, suggesting that impairment in learning
logical impairment. The results of this meta-analysis gener- (and perhaps retrieval) rather than consolidation (i.e.,
ally support this contention, revealing an overall medium retention) difficulties underlie the overall episodic memory
effect size, which was somewhat greater than the generally deficit. To this end, Woods et al. (2005b) recently
small effects reported in meta-analyses of the cognitive demonstrated impairment in overall learning, free recall,
sequelae of cocaine (d=−0.35; Jovanovski et al. 2005) and utilization of semantic clustering strategies, repetitions, and
marijuana (d=−0.15; Grant et al. 2003). Significant deficits non-semantically related intrusion errors in 87 persons with
of a medium magnitude were observed in several different MA dependence, who were nevertheless normal in reten-
cognitive processes dependent upon frontostriatal and limbic tion and recognition discrimination. Moreover, Volkow
circuits, including episodic memory, executive functions et al. (2001c) showed associations between reduced striatal
(e.g., response inhibition, novel problem solving), complex dopamine terminal function and deficits in list learning and
information processing speed, and psychomotor functions. recall in MA users. Taken together, these cognitive and
Slightly smaller effects were also evident in attention/ neurobiological data support the notion that MA use
working memory, language, and visuoconstruction. disrupts the organizational and tactical components of
Neuropsychol Rev (2007) 17:275–297 287

memory encoding and retrieval that are dependent upon associated with poorer medical, neuropsychological, and
frontostriatal networks, although the potential contribution psychosocial outcomes. Indeed, it has been hypothesized
of medial temporal abnormalities on MA-associated epi- that MA dependent individuals may evidence fundamental
sodic memory impairment cannot be entirely dismissed dysfunction in decision-making, including a predilection
(Thompson et al. 2004). Given the magnitude of the for stimulus-driven behavior (Paulus et al. 2002). This
learning and memory deficit in chronic MA users and its hypothesis is bolstered by findings that hypoactivation in
apparent relationship to frontostriatal circuits, exploration prefrontal, parietal, and insular cortex during a decision-
of other aspects of episodic memory (e.g., prospective making task, potentially signifying reduced processing
memory), perhaps as well as nondeclarative (e.g., proce- resources, predicted incident relapse to MA (Paulus et al.
dural) memory will be important areas for future research. 2005).
Also supporting the hypothesized strategic contribution A number of early preclinical studies reported pro-
to episodic memory deficits evident in MA abusers, the nounced motor effects with high doses of MA, which
current meta-analysis revealed evidence of significant initially raised fears about the widespread prevalence of
executive dysfunction in this population. Based on the tests gross motor abnormalities in MA abusers, including
that comprised this domain (e.g., Stroop Color-Word parkinsonism. However, the prevalence of severe move-
Interference, WCST perseverations, and Trail Making Test, ment abnormalities in both clinical practice and in the
Part B), one might argue that cognitive set shifting and, neuropsychological literature has been less than what might
perhaps to a greater extent (Salo et al. 2005), response be expected given the vulnerability of the striatum to MA-
inhibition may be particularly affected in MA users. associated neurotoxicity (Caligiuri and Buitenhuys 2005;
Although executive functions are subserved by distributed Moszczynska et al. 2004). The current meta-analysis
neural networks, research generally supports their strong revealed medium effect sizes for basic motor functioning
reliance upon the frontal cortex and frontostriatal systems (e.g., fine-motor speed and coordination) and information
(e.g., Stuss and Alexander 2007). In MA users specifically, processing speed, with the latter domain being of a slightly
WCST errors are associated with lower frontal grey matter greater magnitude. One possible explanation for the minor
density (Kim et al. 2006), as well as hypometabolism (Kim discrepancy between these domains is that subtle motor
et al. 2005) and lower fractional anisotropy (Chung et al. difficulties will be more prominent on psychomotor tasks
2006) in the frontal white matter. Furthermore, recovery in that require higher-level cognitive abilities (Caligiuri and
DAT binding after MA abstinence is related to reductions Buitenhuys 2005). In support of this contention, the Digit
in WCST errors (Chou et al. 2007). Interestingly, deficits in Symbol test was generally associated with considerably
selective inhibition in MA abusers may also be related to larger effect sizes than either TMT A or the noninterference
frontal systems dysregulation, as evidenced by its associa- trials of the Stroop Test within the processing speed
tion with low anterior cingulate levels of N-acetyl aspartate domain, although a direct statistical comparison was not
(NAA), a brain metabolite regarded as a marker of neuronal possible. Furthermore, this hypothesis is consistent with
integrity (Salo et al. 2007). reports of more pronounced dopaminergic depletion in
An important future direction for executive function cognitive (i.e., caudate) versus motor (i.e., putamen)
research in MA dependent individuals is examination of regions of the striatum of MA users (Moszczynska et al.
impulsivity and decision-making abilities. Similar to other 2004). Given the relatively smaller putamen effects, use of
stimulant using populations (e.g., Coffey et al. 2003), MA more sophisticated and sensitive neuromotor paradigms
dependent individuals evidence risky decision-making and (e.g., motor programming, velocity scaling, forced steadi-
impulsivity, as assessed by their sensitivity to delayed ness, and contraction time) may be useful in demarcating
versus immediate rewards (Hoffman et al. 2006; Monterosso motor abnormalities in MA users (Caligiuri and Buitenhuys
et al. 2006), selection of disadvantageous and/or impulsive 2005).
choices when compared to normal comparison participants Clinical descriptions of patients dependent on MA
(Gonzalez et al. 2007), and self-report of impulsive frequently highlight the apparent inattentiveness and dis-
behavior patterns (Semple et al. 2004, 2005). In fact, tractibility of this population. In support of such observa-
decision-making impairment may be linked to the execu- tions, this meta-analysis suggests that MA dependent
tive aspects of working memory deficits (Bechara and individuals demonstrate impairment on laboratory tasks of
Martin 2004), which were also evident in this meta- attention and working memory. It has been hypothesized
analysis of MA users. In combination, the decision-making that this effect may depend on the type of task employed,
and working memory deficits may predispose MA users such that basic attentional and processing abilities (e.g.,
toward “real world” risky behaviors, such as needle digit recognition) are unaffected (Chang et al. 2002), with
sharing or unprotected sex, that increase the risk of HIV more pronounced deficits emerging with increasingly
and HCV transmission, which (as discussed below) are complex processing demands, especially when working
288 Neuropsychol Rev (2007) 17:275–297

memory and decision-making abilities are taxed (e.g., N- significant, influence on the magnitude of the MA-
back task). Sustained attentional abilities may also be associated effect sizes. Specifically, a larger proportion of
particularly susceptible to MA, perhaps related to MA- men in the MA sample was associated with greater overall
associated neuronal damage in the anterior cingulate and levels of neuropsychological impairment. Although this
insular cortices (London et al. 2005). effect was generally small (b b =−0.01), it should be noted
The moderate language impairment demonstrated in MA that a 50% change in the gender composition of an MA
users may be partially explained by the information group would be associated with a difference in the effect
processing speed and executive deficits described above. size of 0.5 standard deviation units. Moreover, these data
The language domain was primarily comprised of measures raise a number of interesting hypotheses regarding gender
of verbal fluency, which require the rule-guided generation and MA-associated neurotoxicity. For example, this finding
of words under time constraints (typically 60 s). As such, may reflect differential CNS effects of MA use in men,
normal fluency performance taxes the integrity of the which is supported by prior studies showing including
lexicosemantic memory stores, as well as executive and reduced fractional anisotropy in the frontal cortex (Kim
speeded processes, such as rapid, constrained search, et al. 2006) and correlations between frontal hypometabo-
retrieval, switching, and production abilities. In light of lism and WCST errors (Kim et al. 2005) in male, but not
the prominent frontostriatal neurotoxicity of MA, one might female MA users. To this end, animal models suggest that
hypothesize that the apparent verbal fluency deficit reflects the gonadal steroid hormone, estrogen, may be protective
executive dyscontrol of search and retrieval strategies and/ against frontostriatal dopamine depletion associated with
or slowed information processing, rather than degraded MA use (see Dluzen and McDermott 2006 for a review).
lexicosemantic memory stores, as has been observed in Another possible explanation is that men may use MA
other clinical populations with frontostriatal involvement more frequently or in greater amounts (Brecht et al. 2004),
(e.g., Troyer et al. 1998; Woods et al. 2004). Furthermore, leading to more severe addiction and increased CNS
considering the motor and executive deficits apparent in MA effects. Men who use MA also have a higher prevalence
users, one possible direction for future research is examina- of comorbid substance-related disorders (e.g., alcohol),
tion of action (verb) fluency, which prior research shows to antisocial personality disorder, and closed head injuries,
be particularly sensitive to frontal systems pathology (Piatt which may also adversely impact cognition and should be
et al. 1999a, b), perhaps reflecting inefficiencies engaging considered in future studies of CNS functioning in MA
motor representations (Woods et al. 2005a). users.
Although visuospatial abilities have not been extensively Several other demographic factors deserve consideration
studied in chronic MA users, the current meta-analysis found in interpreting these data. Older age in the MA sample was
a small-to-medium effect size within the visuoconstruction associated with a modest increase in cognitive effect size;
domain. Unfortunately, this domain consisted exclusively of practically speaking, an increase of 12 years in the age of
effect sizes from studies that used the copy trial from the Rey an MA group would result in a half standard deviation
Complex Figure (e.g., Kalechstein et al. 2003), which limits change in the effect size. Normal aging is associated with
the conclusions that can be drawn from this finding. structural and functional changes in prefrontal systems
Nevertheless, the posterior parietal cortex and parieto- (e.g., Mielke et al. 1998), which are often accompanied by
striato-frontal circuits play an important role in spatial cognitive decline (e.g., Craik and Bialystok 2006), and
cognition (Lawrence et al. 2000), including visuoconstruc- therefore it may be that normal aging leads to additive
tional abilities. In this regard, MA users show reduced neural and cognitive effects in MA users. However, one
relative regional cerebral blood flow (Chang et al. 2002) and notable limitation is that participants in this meta-analysis
greater volumes (Jernigan et al. 2005) in the parietal cortex. were generally young to middle-aged, with the largest mean
Yet whether the visuoconstructional deficits observed herein age of 41.4 years. It is also possible that older MA
are related to perceptual organizational impairment, execu- dependent individuals had been using MA for a longer
tive dysfunction (e.g., planning), or constructional dyspraxia period of time, although length of use was not correlated
remains to be determined. Future research may examine with the magnitude of effect in our analyses. Despite the
these, along with other aspects of spatial cognition (e.g., relatively lower educational attainment of the MA sample
mental rotation) in MA users, particularly with respect to relative to the comparison group, neither education nor the
their cognitive and neurobiological mechanisms. between-group discrepancy in education had a significant
influence on the effect size estimates. The possibility that
Demographic Factors premorbid neurocognitive differences may exist between
MA using and healthy comparison individuals should also
Analyses of demographic explanatory variables revealed be considered. In particular, substance users may evidence
that gender had a relatively minor, but nonetheless poorer intellectual abilities, or certain cognitive factors may
Neuropsychol Rev (2007) 17:275–297 289

predispose individuals to seek out or abuse substances (Hoffman et al. 2006; Kalechstein et al. 2003; Simon et al.
(Block et al. 2002). However, data regarding these types of 2004; cf. Chang et al. 2005a, b). Volkow et al. (2001b)
factors (e.g., SES, premorbid IQ, and ethnicity) were not found that psychomotor and memory deficits persisted for
consistently reported across studies and therefore could not at least 9 months after last MA use, albeit in a less severe
be considered as possible explanatory variables. Accordingly, form, despite a significant degree of recovery in dopamine
future study groups (and relevant MA subgroups) should be terminal function. Longer periods of abstinence may lead to
demographically comparable and neuropsychological test further recovery of cognitive function, although some
selection might give special consideration to measures with deficits still remain, most notably in episodic memory and
demographically-adjusted normative standards. cognitive inhibition (Johanson et al. 2006). Nevertheless,
while this meta-analysis found that neither average length
Methamphetamine Use Factors of abstinence nor duration of MA use significantly changed
the effect sizes observed across studies, it did not allow for
It remains unresolved whether the extent of neuropsycho- more fine-grained analyses of abstinence or recovery (e.g.,
logical impairment is related to MA use characteristics subgroup analyses). Large-scale, longitudinal cohort studies
(e.g., abstinence, lifetime quantity of consumption, and are needed to examine whether sustained abstinence is
route of use). A few studies have reported that rates of associated with full or partial recovery of cognitive
cognitive impairment correlate with frequency (Simon et al. functioning in persons with MA dependence.
2000) or amount (Monterosso et al. 2005) of MA use, while
others have found no associations with measures assessing Clinical Relevance
the severity of MA dependence or MA use frequency,
amount, or duration (Chang et al. 2002; Hoffman et al. Regarding the possible clinical relevance of this study, MA-
2006; Johanson et al. 2006; Rippeth et al. 2004). This associated cognitive impairment, and particularly the
ambiguity may be due to the unreliability of self-report medium-to-large negative effect sizes in episodic memory
substance use data or may reflect the influence of other, and executive functions, may influence daily functioning
often unmeasured factors (e.g., psychiatric symptomatolo- outcomes, as has been shown in both normal and clinical
gy). It may also reflect the divergence in studies regarding populations (e.g., HIV infection; Heaton et al. 2004). To our
the actual MA use variables that are collected and/or knowledge, however, only one study to date has examined
reported; for example, some studies report average amount the possible impact of MA-associated cognitive impairment
of MA administered per day, while others report days per on everyday functioning (Sadek et al. in 2007), revealing
week of use. Unfortunately, the current meta-analysis cannot that MA dependence is associated with elevated cognitive
shed much light on this issue, due to inconsistent (and often complaints and self-reported dependence in IADL (e.g.,
incomplete) reporting of MA use characteristics across preparing meals, managing money). IADL impairments
studies. were strongly associated with depressive symptoms, where-
Although still limited, sufficient data were available on as cognitive complaints were related to both depressive
reported length of abstinence and duration of MA use, symptoms and objective neuropsychological impairment
which our analyses showed did not correspond to neuro- (Sadek et al. in 2007). Of additional interest is the possible
psychological impairment. This was somewhat surprising link between cognitive impairment and poor treatment
because, as mentioned above, a number of studies have outcomes in MA users, again, especially in terms of
shown relationships between duration of MA use and executive and memory deficits, which are theorized con-
extent of dopaminergic dysfunction (Sekine et al. 2001, tributors to the maintenance of drug-seeking behaviors
2003; Volkow et al. 2001c), as well as at least partial (Bechara and Damasio 2002). Neuropsychological func-
recovery of dopamine terminal function, brain metabolism, tioning plays an important role in state-of-the-art MA abuse
and rCBF with extended abstinence (e.g., Hwang et al. treatments, which commonly involve cognitively-based
2006; e.g., Volkow et al. 2001b; Wang et al. 2004). interventions (e.g., developing strategies to avoiding use
However, it is still unclear whether this recovery results in and prevent relapse; Schuckit 1994), as well as targeted
any substantive changes in neuropsychological function, pharmacotherapies (Meredith et al. 2005). In fact, substance
daily living skills, and other functional outcomes. Previous users with neuropsychological deficits are more likely to
studies suggest that cognitive deficits extend into absti- achieve poorer treatment outcomes, including high rates of
nence, are gradual to recover, and may even become worse program rule violations, poor cognitive skill acquisition,
with initial abstinence when compared to continuing users. and premature drop-out (Aharonovich et al. 2003; Fals-
In early stages of abstinence, deficits are, at the least, Stewart 1993). Clearly, additional research is needed to
equivalent to those seen in currently abusing individuals, better understand the effects of MA-associated neuropsy-
especially in the domains of episodic and working memory chological impairment on these important functional out-
290 Neuropsychol Rev (2007) 17:275–297

comes. In addition, given evidence that cognitive impairment injury and neurocognitive impairment in persons with MA
is an important feature of chronic MA use, future studies of dependence. This is an important question because individ-
MA treatment efficacy should consider including neuropsy- uals rarely use MA in isolation and many illicit (and licit)
chological functioning as an outcome variable of interest. drugs are associated with cognitive deficits. For example,
although between 20 and 35% of chronic MA users are also
Medical and Psychiatric Comorbidities dependent on alcohol (e.g., Simons et al. 2005), we are
unaware of any studies that have prospectively examined
Interpretation of the etiology of the neuropsychological the additive effects of these comorbid neurotoxic substances
deficits in MA users is confounded by the prevalence of on CNS functioning specifically in MA dependent indi-
numerous premorbid (e.g., ADHD) and incident comorbid viduals. Approximately 15% of MA users have comorbid
(e.g., cardiovascular disease) conditions, which make it marijuana abuse (Rippeth et al. 2004), which may not
difficult to ascribe the observed impairment exclusively to exacerbate MA-associated neuropsychological impairment
MA use. Comorbid neuromedical (e.g., cardiovascular (Gonzalez et al. 2004), but could further compromise
disease) conditions may exacerbate MA-associated cogni- cerebral glucose metabolism (Voytek et al. 2005). Another
tive impairment but were not sufficiently characterized commonly used drug, nicotine (e.g., Monterosso et al.
across studies to examine their statistical effects on the 2005), is hypothesized to be protective against the
meta-analytic results. For example, MA users are at neurotoxic effects of MA (Maggio et al. 1998) and may
increased risk of cardiovascular disease (e.g., hypertension), even lessen MA cravings (Hiranita et al. 2006). Yet no
including hemorrhagic and ischemic stroke (Perez et al. studies have thoroughly examined the effects of nicotine
1999). Furthermore, HIV infection is associated with on cognitive functioning or neurobiological markers of
additive neural injury (Chang et al. 2005b; Taylor et al. cerebral function in human MA abusers. Unfortunately, our
2007) and neuropsychological impairment (Carey et al. meta-analysis was unable to provide any insights into the
2006; Rippeth et al. 2004) in MA dependent individuals. effects of current substance comorbidities, since the studies
HCV infection is another important medical factor that is examined were not designed to answer such questions
common in MA users and is known to carry CNS effects (e.g., most excluded persons with current comorbid non-
(Forton et al. 2003; Hilsabeck et al. 2003). In fact, HCV is MA substance abuse). A post-hoc analysis demonstrated
an independent predictor of white matter injury (i.e., lower that the presence of prior non-MA substance use disorders
NAA; Taylor et al. 2004) and cognitive impairment (i.e., did not significantly change the effect size estimates, which
learning, executive functions, and motor skills) in MA users may be attributable to inconsistencies in the prevalence of
(Cherner et al. 2005; Letendre et al. 2005). Several past substance-related disorders across studies. Thus, well-
psychiatric factors (e.g., psychosis, major depressive disor- controlled, prospective studies are needed to adequately
der, bipolar disorder, and ADHD) that can affect cognitive examine the combined or interactive risk of comorbid use
functioning frequently co-occur in MA dependent popula- of substances other than MA.
tions. For example, ADHD is associated with greater
psychiatric symptom severity among MA users (e.g., Jaffe Summary and Conclusions
et al. 2005) and may worsen deficits in language, working
memory, and executive functions (Jaffe et al. 2005; Sim Illicit MA use has grown increasingly prevalent over the
et al. 2002). Moreover, both mood (London et al. 2004) and past 10 years and is now linked to a host of adverse
psychotic (Sekine et al. 2002) symptoms are associated psychosocial (e.g., unemployment), psychiatric (e.g., de-
with cerebral metabolism abnormalities in MA users, pression), and medical (e.g., cardiovascular) complications.
raising questions as to whether these psychiatric comorbid- Methamphetamine is also neurotoxic, and chronic use is
ities are associated with greater prevalence and severity of associated with detrimental effects on neurotransmission
cognitive impairment. Future neuropsychological studies of (e.g., dopamine depletion), as well as the neural structure
MA dependence are encouraged to characterize their and functional integrity of limbic and frontostriatal circuits.
cohorts on these psychiatric and neuromedical co-factors In addition, the results of the present meta-analysis indicate
and carefully examine their potential effects on cognitive that, at the group level, MA dependence is associated with
outcomes. Relatedly, future studies should consider the overall moderate neuropsychological impairment. Effect
contribution of various host genetic factors, such as size estimates of the MA literature revealed broadly
catechol-O-methyltransferase (COMT) to the expression medium group differences across several domains of
of MA-associated neuropsychological impairment (see functioning, with the largest impairments in episodic
Goldberg and Weinberger 2004). memory, executive functions, and information processing
It is also unknown whether the abuse of other substances speed, which were accompanied by slightly smaller impair-
(e.g., alcohol or cocaine) imparts additive risks of neural ments in motor skills, language, and visuoconstructional
Neuropsychol Rev (2007) 17:275–297 291

abilities. Considering the neural systems most susceptible Alexander, G. E., Delong, M. R., & Strick, P. L. (1986). Parallel
organization of functionally segregated circuits linking basal
to MA and their known cognitive sequelae, this pattern of
ganglia and cortex. Annual Review of Neuroscience, 9, 357–
neuropsychological impairment is most consistent with a 381.
frontostriatal neuropathogenesis, along with perhaps more American Psychiatric Association (1994). Diagnostic and Statistical
modest contributions from neurotoxicity in the posterior Manual of Mental Disorders—Fourth Edition: DSM-IV. Wash-
ington, D.C.: American Psychiatric Association.
parietal and medial temporal cortices. Candidate mecha-
Anglin, M. D., Burke, C., Perrochet, B., Stamper, E., & Dawud-
nisms for MA-associated neuropsychological deficits in- Noursi, S. (2000). History of the methamphetamine problem.
clude both necrotic and apoptotic etiologies resultant from Journal of Psychoactive Drugs, 32(2), 137–141.
such factors as cerebrovascular injury, oxidative stress, Arends, L. R., Voko, Z., & Stijnen, T. (2003). Combining multiple
outcome measures in a meta-analysis: An application. Statistics
gliosis, and neuritic growth. Analysis of possible moderat-
in Medicine, 22(8), 1335–1353.
ing factors revealed a potential interplay between MA Axt, K. J., & Molliver, M. E. (1991). Immunocytochemical evidence
dependence and demographic factors (i.e., male gender and for methamphetamine-induced serotonergic axon loss in the rat
older age) in the expression of cognitive deficits. Although brain. Synapse, 9(4), 302–313.
Baberg, H. T., Nelesen, R. A., & Dimsdale, J. E. (1996). Amphet-
there was no evidence supporting the moderating effects of
amine use: Return of an old scourge in a consultation psychiatry
MA use characteristics (i.e., length of abstinence and setting. American Journal of Psychiatry, 153(6), 789–793.
duration of use) or remote non-MA substance-related Bae, S. C., Lyoo, I. K., Sung, Y. H., Yoo, J., Chung, A., Yoon, S. J., et al.
disorders on the observed neuropsychological effect sizes, (2006). Increased white matter hyperintensities in male metham-
phetamine abusers. Drug and Alcohol Dependence, 81(1), 83–88.
interpretation of these analyses is tempered by methodo-
Bakhit, C., Morgan, M. E., Peat, M. A., & Gibb, J. W. (1981). Long-
logical limitations of the existing literature. Understanding term effects of methamphetamine on the synthesis and metabo-
the etiology of the cognitive deficits in chronic MA users is lism of 5-hydroxytryptamine in various regions of the rat brain.
also complicated by numerous common psychiatric (e.g., Neuropharmacology, 20(12A), 1135–1140.
Barr, A. M., Markou, A., & Phillips, A. G. (2002). A ‘crash’ course on
ADHD and depression) and medical (e.g., HIV and HCV
psychostimulant withdrawal as a model of depression. Trends in
infection) cofactors with known CNS effects, which will be Pharmacological Science, 23(10), 475–482.
critical targets for future neuropsychological and neurobi- Barr, A. M., Panenka, W. J., MacEwan, G. W., Thornton, A. E., Lang,
ological research. Despite these interpretive caveats, the D. J., Honer, W. G., et al. (2006). The need for speed: An update
on methamphetamine addiction. Journal of Psychiatry and
nature and magnitude of cognitive deficits associated with
Neuroscience, 31(5), 301–313.
chronic MA use ostensibly increase the risk of poorer Bechara, A., & Damasio, H. (2002). Decision-making and addiction
health outcomes, including dependence in daily functioning (part I): Impaired activation of somatic states in substance
(e.g., unemployment), high-risk behaviors, and treatment dependent individuals when pondering decisions with negative
future consequences. Neuropsychologia, 40(10), 1675–1689.
nonadherence and relapse. Accordingly, consideration of
Bechara, A., & Martin, E. M. (2004). Impaired decision making
neuropsychological functioning has clear implications for related to working memory deficits in individuals with substance
the clinical management of persons with MA dependence. addictions. Neuropsychology, 18(1), 152–162.
Biederman, J. (2005). Attention-deficit/hyperactivity disorder: A
selective overview. Biological Psychiatry, 57(11), 1215–1220.
Acknowledgement The research described was supported by Block, R. I., Erwin, W. J., & Ghoneim, M. M. (2002). Chronic drug
DA12065 and MH62512 from the National Institutes of Health. The use and cognitive impairments. Pharmacology Biochemistry and
views expressed in this article are those of the authors and do not Behavior, 73, 491–504.
reflect the official policy or position of the Department of the Navy, Bluthenthal, R. N., Kral, A. H., Gee, L., Lorvick, J., Moore, L., Seal,
Department of Defense, or the United States Government. K., et al. (2001). Trends in HIV seroprevalence and risk among
The authors thank Drs. Raul Gonzalez and John Monterosso for gay and bisexual men who inject drugs in San Francisco, 1988 to
kindly providing supplementary data for analyses. 2000. Journal of Acquired Immune Deficiency Syndromes, 28(3),
The following studies were included in the meta-analysis: Chang 264–269.
et al. 2002, 2005a; Gonzalez et al. 2007; Hoffman et al. 2006; Johanson Bray, G. A. (1993). Use and abuse of appetite-suppressant drugs in the
et al. 2006; Kalechstein et al. 2003; Kim et al. 2005; London et al. 2005; treatment of obesity. Annals of Internal Medicine, 119(7 Pt 2),
Monterosso et al. 2005, 2006; Newton et al. 2004; Paulus et al. 2003; 707–713.
Rippeth et al. 2004; Salo et al. 2002, 2005, 2007; Simon et al. 2000, Brecht, M. L., O’Brien, A., von Mayrhauser, C., & Anglin, M. D.
2004. (2004). Methamphetamine use behaviors and gender differences.
Addictive Behaviors, 29(1), 89–106.
Brown, J. M., Quinton, M. S., & Yamamoto, B. K. (2005).
References Methamphetamine-induced inhibition of mitochondrial complex
II: Roles of glutamate and peroxynitrite. Journal of Neurochem-
ADA Division of Communications (2005). For the dental patient. istry, 95, 429–436.
methamphetamine use and oral health. Journal of the American Buffenstein, A., Heaster, J., & Ko, P. (1999). Chronic psychotic illness
Dental Association, 136(10), 1491. from methamphetamine. American Journal of Psychiatry, 156(4),
Aharonovich, E., Nunes, E., & Hasin, D. (2003). Cognitive 662.
impairment, retention and abstinence among cocaine abusers in Burrows, K. B., & Meshul, C. K. (1999). High-dose methamphet-
cognitive-behavioral treatment. Drug and Alcohol Dependence, amine treatment alters presynaptic GABA and glutamate immu-
71(2), 207–211. noreactivity. Neuroscience, 90(3), 833–850.
292 Neuropsychol Rev (2007) 17:275–297

Busto, U., Bendayan, R., & Sellers, E. M. (1989). Clinical Coffey, S. F., Gudleski, G. D., Saladin, M. E., & Brady, K. T. (2003).
pharmacokinetics of non-opiate abused drugs. Clinical Pharma- Impulsivity and rapid discounting of delayed hypothetical
cokinetics, 16(1), 1–26. rewards in cocaine-dependent individuals. Experimental and
Cadet, J. L., Jayanthi, S., & Deng, X. (2005). Methamphetamine- Clinical Psychopharmacology, 11(1), 18–25.
induced neuronal apoptosis involves the activation of multiple Cohen, J. (1988). Statistical power analyses for the behavioral
death pathways. Neurotoxicity Research, 8(3–4), 199–206. sciences (2nd ed.). Mahwah, NJ: Lawrence Erlbaum Associates,
Caligiuri, M. P., & Buitenhuys, C. (2005). Do preclinical findings of Inc.
methamphetamine-induced motor abnormalities translate to an Comer, S. D., Hart, C. L., Ward, A. S., Haney, M., Foltin, R. W., &
observable clinical phenotype? Neuropsychopharmacology, 30 Fischman, M. W. (2001). Effects of repeated oral methamphet-
(12), 2125–2134. amine administration in humans. Psychopharmacology, 155(4),
Carey, C. L., Woods, S. P., Rippeth, J. D., Gonzalez, R., Heaton, R. K., 397–404.
& Grant, I. (2006). Additive deleterious effects of methamphet- Community Epidemiology Work Group (2006). Epidemiologic trends
amine dependence and immunosuppression on neuropsychological in drug abuse—advance report. Bethesda, MD: U.S. Department
functioning in HIV infection. AIDS and Behavior, 10(2), 185–190. of Health and Human Services, National Institutes of Health.
Cartier, J., Farabee, D., & Prendergast, M. L. (2006). Methamphet- Craik, F. I., & Bialystok, E. (2006). Cognition through the lifespan:
amine use, self-reported violent crime, and recidivism among Mechanisms of change. Trends in Cognitive Science, 10(3), 131–
offenders in California who abuse substances. Journal of 138.
Interpersonal Violence, 21(4), 435–445. Cretzmeyer, M., Sarrazin, M. V., Huber, D. L., Block, R. I., & Hall, J.
Cass, W. A. (1997). Decreases in evoked overflow of dopamine in rat A. (2003). Treatment of methamphetamine abuse: Research
striatum after neurotoxic doses of methamphetamine. Journal of findings and clinical directions. Journal of Substance Abuse
Pharmacology and Experimental Therapeutics, 280(1), 105–113. Treatment, 24(3), 267–277.
Cass, W. A., & Manning, M. W. (1999). Recovery of presynaptic Davidson, C., Gow, A. J., Lee, T. H., & Ellinwood, E. H. (2001).
dopaminergic functioning in rats treated with neurotoxic doses of Methamphetamine neurotoxicity: Necrotic and apoptotic mecha-
methamphetamine. Journal of Neuroscience, 19(17), 7653–7660. nisms and relevance to human abuse and treatment. Brain
Chan, P., Di Monte, D. A., Luo, J. J., DeLanney, L. E., Irwin, I., & Research. Brain Research Reviews, 36(1), 1–22.
Langston, J. W. (1994). Rapid ATP loss caused by methamphet- Davis, G. G., & Swalwell, C. I. (1994). Acute aortic dissections and
amine in the mouse striatum: Relationship between energy ruptured berry aneurysms associated with methamphetamine
impairment and dopaminergic neurotoxicity. Journal of Neuro- abuse. Journal of Forensic Sciences, 39(6), 1481–1485.
chemistry, 62(6), 2484–2487. Delaney, P., & Estes, M. (1980). Intracranial hemorrhage with
Chang, L., Cloak, C., Patterson, K., Grob, C., Miller, E. N., & Ernst, amphetamine abuse. Neurology, 30(10), 1125–1128.
T. (2005a). Enlarged striatum in abstinent methamphetamine di Michele, F., Prichep, L., John, E. R., & Chabot, R. J. (2005). The
abusers: A possible compensatory response. Biological Psychia- neurophysiology of attention-deficit/hyperactivity disorder. Inter-
try, 57(9), 967–974. national Journal of Psychophysiology, 58(1), 81–93.
Chang, L., Ernst, T., Speck, O., & Grob, C. S. (2005b). Additive Dluzen, D. E., & McDermott, J. L. (2006). Estrogen, testosterone, and
effects of HIV and chronic methamphetamine use on brain methamphetamine toxicity. Annals of the New York Academy of
metabolite abnormalities. American Journal of Psychiatry, 162 Sciences, 1074, 282–294.
(2), 361–369. Domier, C. P., Simon, S. L., Rawson, R. A., Huber, A., & Ling, W.
Chang, L., Ernst, T., Speck, O., Patel, H., DeSilva, M., Leonido-Yee, (2000). A comparison of injecting and noninjecting methamphet-
M., et al. (2002). Perfusion MRI and computerized cognitive test amine users. Journal of Psychoactive Drugs, 32(2), 229–232.
abnormalities in abstinent methamphetamine users. Psychiatry Duval, S., & Tweedie, R. L. (2000). Trim and fill: A simple funnel
Research, 114(2), 65–79. plot based method of testing and adjusting for publication bias in
Chen, C. K., Lin, S. K., Sham, P. C., Ball, D., el Loh, W., & Murray, meta-analysis. Biometrics, 56(2), 455–463.
R. M. (2005). Morbid risk for psychiatric disorder among the Ernst, T., Chang, L., Leonido-Yee, M., & Speck, O. (2000). Evidence
relatives of methamphetamine users with and without psychosis. for long-term neurotoxicity associated with methamphetamine
American Journal of Medical Genetics. Part B, Neuropsychiatric abuse: A 1H MRS study. Neurology, 54(6), 1344–1349.
Genetics, 136(1), 87–91. Fals-Stewart, W. (1993). Neurocognitive defects and their impact on
Chen, C. K., Lin, S. K., Sham, P. C., Ball, D., Loh, E. W., Hsiao, C. substance abuse treatment. Journal of Addictions and Offender
C., et al. (2003). Pre-morbid characteristics and co-morbidity of Counseling, 13, 46–57.
methamphetamine users with and without psychosis. Psycholog- Faraone, S. V., Biederman, J., Spencer, T., Wilens, T., Seidman, L. J.,
ical Medicine, 33(8), 1407–1414. Mick, E., et al. (2000). Attention-deficit/hyperactivity disorder in
Cherner, M., Letendre, S., Heaton, R. K., Durelle, J., Marquie-Beck, adults: An overview. Biological Psychiatry, 48(1), 9–20.
J., Gragg, B., et al. (2005). Hepatitis C augments cognitive Forton, D. M., Taylor-Robinson, S. D., & Thomas, H. C. (2003).
deficits associated with HIV infection and methamphetamine. Cerebral dysfunction in chronic hepatitis C infection. Journal of
Neurology, 64(8), 1343–1347. Viral Hepatitis, 10(2), 81–86.
Chin, K. M., Channick, R. N., & Rubin, L. J. (2006). Is Friedman, S. D., Castaneda, E., & Hodge, G. K. (1998). Long-
methamphetamine use associated with idiopathic pulmonary term monoamine depletion, differential recovery, and subtle
arterial hypertension? Chest, 130(6), 1657–1663. behavioral impairment following methamphetamine-induced
Chou, Y. H., Huang, W. S., Su, T. P., Lu, R. B., Wan, F. J., & Fu, Y. K. neurotoxicity. Pharmacology, Biochemistry and Behavior, 61
(2007). Dopamine transporters and cognitive function in meth- (1), 35–44.
amphetamine abuser after a short abstinence: A SPECT study. Giros, B., Jaber, M., Jones, S. R., Wightman, R. M., & Caron, M. G.
European Neuropsychopharmacology, 17(1), 46–52. (1996). Hyperlocomotion and indifference to cocaine and
Chung, A., Lyoo, I. K., Kim, S. J., Hwang, J., Bae, S. C., Sung, Y. H., amphetamine in mice lacking the dopamine transporter. Nature,
et al. (2006). Decreased frontal white-matter integrity in abstinent 379(6566), 606–612.
methamphetamine abusers. International Journal of Neuropsy- Goldberg, T. E., & Weinberger, D. R. (2004). Genes and the parsing of
chopharmacology, 1–11. cognitive processes. Trends in Cognitive Sciences, 8(7), 325–335.
Neuropsychol Rev (2007) 17:275–297 293

Gonzales, R., Marinelli-Casey, P., Shoptaw, S., Ang, A., & Rawson, tomatology in patients with chronic hepatitis C. Journal of the
R. A. (2006). Hepatitis C virus infection among methamphet- International Neuropsychological Society, 9(6), 847–854.
amine-dependent individuals in outpatient treatment. Journal of Hiranita, T., Nawata, Y., Sakimura, K., Anggadiredja, K., &
Substance Abuse Treatment, 31(2), 195–202. Yamamoto, T. (2006). Suppression of methamphetamine-seeking
Gonzalez, R., Bechara, A., & Martin, E. M. (2007). Executive behavior by nicotinic agonists. Proceedings of the National
functions among individuals with methamphetamine or alcohol Academy of Sciences of the United States of America, 103(22),
as drugs of choice: Preliminary observations. Journal of Clinical 8523–8527.
and Experimental Neuropsychology, 29(2), 155–159. Hoffman, W. F., Moore, M., Templin, R., McFarland, B., Hitzemann,
Gonzalez, R., Rippeth, J. D., Carey, C. L., Heaton, R. K., Moore, D. R. J., & Mitchell, S. H. (2006). Neuropsychological function and
J., Schweinsburg, B. C., et al. (2004). Neurocognitive perfor- delay discounting in methamphetamine-dependent individuals.
mance of methamphetamine users discordant for history of Psychopharmacology, 188(2), 162–170.
marijuana exposure. Drug and Alcohol Dependence, 76(2), Hong, R., Matsuyama, E., & Nur, K. (1991). Cardiomyopathy
181–190. associated with the smoking of crystal methamphetamine. JAMA,
Gonzalez Castro, F., Barrington, E. H., Walton, M. A., & Rawson, R. 265(9), 1152–1154.
A. (2000). Cocaine and methamphetamine: Differential addiction Hotchkiss, A. J., & Gibb, J. W. (1980). Long-term effects of multiple
rates. Psychology of Addictive Behaviors, 14(4), 390–396. doses of methamphetamine on tryptophan hydroxylase and
Grant, I. (1987). Alcohol and the brain: Neuropsychological corre- tyrosine hydroxylase activity in rat brain. Journal of Pharmacol-
lates. Journal of Consulting and Clinical Psychology, 55, 310– ogy and Experimental Therapeutics, 214(2), 257–262.
324. Hunt, D. (1995). Pulse check: National trends in drug abuse.
Grant, I., Gonzalez, R., Carey, C. L., Natarajan, L., Wolfson, T. Washington, DC: Office of National Drug Control Policy.
(2003). Non-acute (residual) neurocognitive effects of cannabis Hwang, J., Lyoo, I. K., Kim, S. J., Sung, Y. H., Bae, S., Cho, S. N., et
use: A meta-analytic study. Journal of the International Neuro- al. (2006). Decreased cerebral blood flow of the right anterior
psychological Society, 9, 679–689. cingulate cortex in long-term and short-term abstinent metham-
Grilli, L., & Rampichini, C. (2006). A review of tandom effects phetamine users. Drug and Alcohol Dependence, 82(2), 177–
modelling using gllamm in Stata. In Software reviews of 181.
multilevel analysis packages. University of Bristol, UK: Centre Iwanami, A., Sugiyama, A., Kuroki, N., Toda, S., Kato, N., Nakatani,
for Multilevel Modelling. Y., et al. (1994). Patients with methamphetamine psychosis
Halkitis, P. N., Parsons, J. T., & Stirratt, M. J. (2001). A double admitted to a psychiatric hospital in Japan. A preliminary report.
epidemic: Crystal methamphetamine drug use in relation to HIV Acta Psychiatrica Scandinavica, 89(6), 428–432.
transmission among gay men. Journal of Homosexuality, 41(2), Iyo, M., Namba, H., Yanagisawa, M., Hirai, S., Yui, N., & Fukui, S.
17–35. (1997). Abnormal cerebral perfusion in chronic methamphet-
Halkitis, P. N., & Shrem, M. T. (2006). Psychological differences amine abusers: A study using 99MTc-HMPAO and SPECT.
between binge and chronic methamphetamine using gay and Progress in Neuro-Psychopharmacology and Biological Psychi-
bisexual men. Addictive Behaviors, 31(3), 549–552. atry, 21(5), 789–796.
Hall, C. D., Blanton, D. E., Scatliff, J. H., & Morris, C. E. (1973). Iyo, M., Sekine, Y., & Mori, N. (2004). Neuromechanism of
Speed kills: Fatality from the self-administration of methamphet- developing methamphetamine psychosis: A neuroimaging study.
amine intravenously. Southern Medical Journal, 66(6), 650–652. Annals of the New York Academy of Sciences, 1025, 288–295.
Hall, W., Hando, J., Darke, S., & Ross, J. (1996). Psychological Jaffe, C., Bush, K. R., Straits-Tröster, K., Meredith, C., Romwall, L.,
morbidity and route of administration among amphetamine users Rosenbaum, G., et al. (2005). A comparison of methamphet-
in Sydney, Australia. Addiction, 91(1), 81–87. amine-dependent inpatients childhood attention deficit hyperac-
Harris, D., & Batki, S. L. (2000). Stimulant psychosis: Symptom tivity disorder symptomatology. Journal on Addictive Disorders,
profile and acute clinical course. American Journal on Addic- 24(3), 133–152.
tions, 9(1), 28–37. Jernigan, T. L., Gamst, A. C., Archibald, S. L., Fennema-Notestine,
Hart, C. L., Haney, M., Nasser, J., & Foltin, R. W. (2005). Combined C., Mindt, M. R., Marcotte, T. D., et al. (2005). Effects of
effects of methamphetamine and zolpidem on performance and methamphetamine dependence and HIV infection on cerebral
mood during simulated night shift work. Pharmacology, Bio- morphology. American Journal of Psychiatry, 162(8), 1461–
chemistry and Behavior, 81(3), 559–568. 1472.
Hart, C. L., Ward, A. S., Haney, M., Foltin, R. W., & Fischman, M. W. Johanson, C. E., Frey, K. A., Lundahl, L. H., Keenan, P., Lockhart, N.,
(2001). Methamphetamine self-administration by humans. Psy- Roll, J., et al. (2006). Cognitive function and nigrostriatal
chopharmacology, 157(1), 75–81. markers in abstinent methamphetamine abusers. Psychopharma-
Harvey, D. C., Lacan, G., Tanious, S. P., & Melega, W. P. (2000). cology, 185(3), 327–338.
Recovery from methamphetamine induced long-term nigrostriatal Johnson, B. A., Ait-Daoud, N., & Wells, L. T. (2000). Effects of
dopaminergic deficits without substantia nigra cell loss. Brain isradipine, a dihydropyridine-class calcium channel antagonist,
Research, 871(2), 259–270. on D-methamphetamine-induced cognitive and physiological
Heaton, R. K., Marcotte, T. D., Mindt, M. R., Sadek, J., Moore, D. J., changes in humans. Neuropsychopharmacology, 22(5), 504–512.
Bentley, H., et al. (2004). The impact of HIV-associated Johnson, B. A., Roache, J. D., Ait-Daoud, N., Wallace, C., Wells, L.
neuropsychological impairment on everyday functioning. Journal T., & Wang, Y. (2005). Effects of isradipine on methamphet-
of the International Neuropsychological Society, 10(3), 317–331. amine-induced changes in attentional and perceptual-motor skills
Hedges, L. V., & Olkin, L. I. (1985). Statistical methods for meta- of cognition. Psychopharmacology, 178(2–3), 296–302.
analysis. Orlando, FL: Academic Press. Jovanovski, D., Erb, S., & Zakzanis, K. K. (2005). Neurocognitive
Hervey, A. S., Epstein, J. N., & Curry, J. F. (2004). Neuropsychology deficits in cocaine users: A quantitative review of the evidence.
of adults with attention-deficit/hyperactivity disorder: A meta- Journal of Clinical and Experimental Neuropsychology, 27(2),
analytic review. Neuropsychology, 18(3), 485–503. 189–204.
Hilsabeck, R. C., Hassanein, T. I., Carlson, M. D., Ziegler, E. A., & Kalaian, H. A., & Raudenbush, S. W. (1996). A multivariate mixed
Perry, W. (2003). Cognitive functioning and psychiatric symp- linear model for meta-analysis. Psychological Methods, 1(3), 227.
294 Neuropsychol Rev (2007) 17:275–297

Kalechstein, A. D., Newton, T. F., & Green, M. (2003). Methamphet- McGregor, C., Srisurapanont, M., Jittiwutikarn, J., Laobhripatr, S.,
amine dependence is associated with neurocognitive impairment Wongtan, T., & White, J. M. (2005). The nature, time course and
in the initial phases of abstinence. Journal of Neuropsychiatry severity of methamphetamine withdrawal. Addiction, 100(9),
and Clinical Neurosciences, 15(2), 215–220. 1320–1329.
Kalechstein, A. D., Newton, T. F., Longshore, D., Anglin, M. D., van McKetin, R., McLaren, J., Lubman, D. I., & Hides, L. (2006). The
Gorp, W. G., & Gawin, F. H. (2000). Psychiatric comorbidity of prevalence of psychotic symptoms among methamphetamine
methamphetamine dependence in a forensic sample. Journal of users. Addiction, 101(10), 1473–1478.
Neuropsychiatry and Clinical Neurosciences, 12(4), 480–484. Meredith, C. W., Jaffe, C., Ang-Lee, K., & Saxon, A. J. (2005).
Khoshbouei, H., Wang, H., Lechleiter, J. D., Javitch, J. A., & Galli, A. Implications of chronic methamphetamine use: A literature
(2003). Amphetamine-induced dopamine efflux. A voltage- review. Harvard Review of Psychiatry, 13(3), 141–154.
sensitive and intracellular Na+-dependent mechanism. Journal Mewaldt, S. P., & Ghoneim, M. M. (1979). The effects and
of Biological Chemistry, 278(14), 12070–12077. interactions of scopolamine, physostigmine and methamphet-
Kim, S. J., Lyoo, I. K., Hwang, J., Chung, A., Hoon Sung, Y., Kim, J., amine on human memory. Pharmacology, Biochemistry and
et al. (2006). Prefrontal grey-matter changes in short-term and Behavior, 10(2), 205–210.
long-term abstinent methamphetamine abusers. International Mielke, R., Kessler, J., Szelies, B., Herholz, K., Wienhard, K., &
Journal of Neuropsychopharmacology, 9(2), 221–228. Heiss, W. D. (1998). Normal and pathological aging-findings of
Kim, S. J., Lyoo, I. K., Hwang, J., Sung, Y. H., Lee, H. Y., Lee, D. S., positron-emission-tomography. Journal of Neural Transmission,
et al. (2005). Frontal glucose hypometabolism in abstinent 105(8–9), 821–837.
methamphetamine users. Neuropsychopharmacology, 30(7), Miller, D. B., & O’Callaghan, J. P. (2003). Elevated environmental
1383–1391. temperature and methamphetamine neurotoxicity. Environmental
Kokoshka, J. M., Metzger, R. R., Wilkins, D. G., Gibb, J. W., Hanson, Research, 92(1), 48–53.
G. R., & Fleckenstein, A. E. (1998). Methamphetamine treatment Mitler, M. M., Hajdukovic, R., & Erman, M. K. (1993). Treatment of
rapidly inhibits serotonin, but not glutamate, transporters in rat narcolepsy with methamphetamine. Sleep, 16(4), 306–317.
brain. Brain Research, 799(1), 78–83. Mohs, R. C., Tinklenberg, J. R., Roth, W. T., & Kopell, B. S. (1978).
Lawrence, A. D., Watkins, L. H., Sahakian, B. J., Hodges, J. R., & Methamphetamine and diphenhydramine effects on the rate of
Robbins, T. W. (2000). Visual object and visuospatial cognition cognitive processing. Psychopharmacology, 59(1), 13–19.
in Huntington’s disease: Implications for information processing Mohs, R. C., Tinklenberg, J. R., Roth, W. T., & Kopell, B. S. (1980).
in corticostriatal circuits. Brain, 123(Pt 7), 1349–1364. Sensitivity of some human cognitive functions to effects of
Letendre, S. L., Cherner, M., Ellis, R. J., Marquie-Beck, J., Gragg, B., methamphetamine and secobarbital. Drug and Alcohol Depen-
Marcotte, T., et al. (2005). The effects of hepatitis C, HIV, and dence, 5(2), 145–150.
methamphetamine dependence on neuropsychological perfor- Monterosso, J. R., Ainslie, G., Xu, J., Cordova, X., Domier, C. P., &
mance: Biological correlates of disease. AIDS, 19(Suppl 3), London, E. D. (2006). Frontoparietal cortical activity of
S72–S78. methamphetamine-dependent and comparison subjects
Logan, B. K. (1996). Methamphetamine and driving impairment. performing a delay discounting task. Human Brain Mapping,
Journal of Forensic Sciences, 41(3), 457–464. 28(5), 383–393.
Logan, B. K., Fligner, C. L., & Haddix, T. (1998). Cause and manner Monterosso, J. R., Aron, A. R., Cordova, X., Xu, J., & London, E. D.
of death in fatalities involving methamphetamine. Journal of (2005). Deficits in response inhibition associated with chronic
Forensic Sciences, 43(1), 28–34. methamphetamine abuse. Drug and Alcohol Dependence, 79(2),
London, E. D., Berman, S. M., Voytek, B., Simon, S. L., 273–277.
Mandelkern, M. A., Monterosso, J., et al. (2005). Cerebral Morgan, M. E., & Gibb, J. W. (1980). Short-term and long-term
metabolic dysfunction and impaired vigilance in recently effects of methamphetamine on biogenic amine metabolism in
abstinent methamphetamine abusers. Biological Psychiatry, 58 extra-striatal dopaminergic nuclei. Neuropharmacology, 19(10),
(10), 770–778. 989–995.
London, E. D., Simon, S. L., Berman, S. M., Mandelkern, M. A., Moszczynska, A., Fitzmaurice, P., Ang, L., Kalasinsky, K. S.,
Lichtman, A. M., Bramen, J., et al. (2004). Mood disturbances Schmunk, G. A., Peretti, F. J., et al. (2004). Why is parkinsonism
and regional cerebral metabolic abnormalities in recently absti- not a feature of human methamphetamine users? Brain, 127(Pt
nent methamphetamine abusers. Archives of General Psychiatry, 2), 363–370.
61(1), 73–84. National Drug Intelligence Center (2006). 2006 National drug threat
Maggio, R., Riva, M., Vaglini, F., Fornai, F., Molteni, R., Armogida, assessment. McLean, VA: National Drug Intelligence Center.
M., et al. (1998). Nicotine prevents experimental parkinsonism in National Institute on Drug Abuse (2006). NIDA research report—
rodents and induces striatal increase of neurotrophic factors. methamphetamine abuse and addiction (NIH publication no. 06-
Journal of Neurochemistry, 71(6), 2439–2446. 4210). Rockville, MD: National Institutes of Health.
Mason, G. F., & Rothman, D. L. (2004). Basic principles of metabolic Newton, T. F., Kalechstein, A. D., Duran, S., Vansluis, N., & Ling, W.
modeling of NMR (13)C isotopic turnover to determine rates of (2004). Methamphetamine abstinence syndrome: Preliminary
brain metabolism in vivo. Metabolic Engineering, 6(1), 75–84. findings. American Journal on Addictions, 13(3), 248–255.
Matsumoto, T., Kamijo, A., Yamaguchi, A., Iseki, E., & Hirayasu, Y. Newton, T. F., Kalechstein, A. D., Hardy, D. J., Cook, I. A., Nestor,
(2005). Childhood histories of attention-deficit hyperactivity L., Ling, W., et al. (2004). Association between quantitative EEG
disorders in Japanese methamphetamine and inhalant abusers: and neurocognition in methamphetamine-dependent volunteers.
Preliminary report. Journal of Neuropsychiatry and Clinical Clinical Neurophysiology, 115, 194–198.
Neurosciences, 59(1), 102–105. Nordahl, T. E., Salo, R., & Leamon, M. (2003). Neuropsychological
McCann, U. D., Wong, D. F., Yokoi, F., Villemagne, V., Dannals, R. effects of chronic methamphetamine use on neurotransmitters
F., & Ricaurte, G. A. (1998). Reduced striatal dopamine and cognition: A review. Journal of Neuropsychiatry and
transporter density in abstinent methamphetamine and meth- Clinical Neurosciences, 15(3), 317–325.
cathinone users: Evidence from positron emission tomography Nordahl, T. E., Salo, R., Possin, K., Gibson, D. R., Flynn, N., Leamon,
studies with [11C]WIN-35,428. Journal of Neuroscience, 18(20), M., et al. (2002). Low N-acetyl-aspartate and high choline in the
8417–8422. anterior cingulum of recently abstinent methamphetamine-
Neuropsychol Rev (2007) 17:275–297 295

dependent subjects: A preliminary proton MRS study. Magnetic Salo, R., Nordahl, T. E., Natsuaki, Y., Leamon, M. H., Galloway, G.
resonance spectroscopy. Psychiatry Research, 116(1–2), 43–52. P., Waters, C., et al. (2007). Attentional control and brain
Oh, J. S., Lyoo, I. K., Sung, Y. H., Hwang, J., Kim, J., Chung, A., metabolite levels in methamphetamine abusers. Biological
et al. (2005). Shape changes of the corpus callosum in abstinent Psychiatry, 61(11), 1272–1280.
methamphetamine users. Neuroscience Letters, 384(1–2), 76–81. Salo, R., Nordahl, T. E., Possin, K., Leamon, M., Gibson, D. R.,
Olsen, E. R. (1977). Intracranial hemorrhage and amphetamine usage. Galloway, G. P., et al. (2002). Preliminary evidence of reduced
Review of the effects of amphetamines on the central nervous cognitive inhibition in methamphetamine-dependent individuals.
system. Angiology, 28(7), 464–471. Psychiatry Research, 111, 65–74.
Paulus, M. P., Hozack, N., Frank, L., Brown, G. G., & Schuckit, M. Sato, M. (1986). Acute exacerbation of methamphetamine psychosis
A. (2003). Decision making by methamphetamine-dependent and lasting dopaminergic supersensitivity—a clinical survey.
subjects is associated with error-rate-independent decrease in Psychopharmacology Bulletin, 22(3), 751–756.
prefrontal and parietal activation. Biological Psychiatry, 53(1), Schuckit, M. A. (1994). The treatment of stimulant dependence.
65–74. Addiction, 89(11), 1559–1563.
Paulus, M. P., Hozack, N. E., Zauscher, B. E., Frank, L., Brown, G. Sekine, Y., Iyo, M., Ouchi, Y., Matsunaga, T., Tsukada, H., Okada, H.,
G., Braff, D. L., et al. (2002). Behavioral and functional et al. (2001). Methamphetamine-related psychiatric symptoms
neuroimaging evidence for prefrontal dysfunction in metham- and reduced brain dopamine transporters studied with PET.
phetamine-dependent subjects. Neuropsychopharmacology, 26 American Journal of Psychiatry, 158(8), 1206–1214.
(1), 53–63. Sekine, Y., Minabe, Y., Kawai, M., Suzuki, K., Iyo, M., Isoda, H.,
Paulus, M. P., Tapert, S. F., & Schuckit, M. A. (2005). Neural et al. (2002). Metabolite alterations in basal ganglia associated
activation patterns of methamphetamine-dependent subjects with methamphetamine-related psychiatric symptoms. A proton
during decision making predict relapse. Archives of General MRS study. Neuropsychopharmacology, 27(3), 453–461.
Psychiatry, 62(7), 761–768. Sekine, Y., Minabe, Y., Ouchi, Y., Takei, N., Iyo, M., Nakamura, K., et
Perez, J. A., Jr., Arsura, E. L., & Strategos, S. (1999). Methamphet- al. (2003). Association of dopamine transporter loss in the
amine-related stroke: Four cases. Journal of Emergency Medi- orbitofrontal and dorsolateral prefrontal cortices with metham-
cine, 17(3), 469–471. phetamine-related psychiatric symptoms. American Journal of
Piatt, A. L., Fields, J. A., Paolo, A. M., Koller, W. C., & Tröster, A. I. Psychiatry, 160(9), 1699–1701.
(1999a). Lexical, semantic, and action verbal fluency in Sekine, Y., Ouchi, Y., Takei, N., Yoshikawa, E., Nakamura, K.,
Parkinson’s disease with and without dementia. Journal of Futatsubashi, M., et al. (2006). Brain serotonin transporter
Clinical and Experimental Neuropsychology, 21(4), 435–443. density and aggression in abstinent methamphetamine abusers.
Piatt, A. L., Fields, J. A., Paolo, A. M., & Tröster, A. I. (1999b). Archives of General Psychiatry, 63(1), 90–100.
Action (verb naming) fluency as an executive function measure: Semple, S. J., Patterson, T. L., & Grant, I. (2003). Binge use of
Convergent and divergent evidence of validity. Neuropsycholo- methamphetamine among HIV-positive men who have sex with
gia, 37(13), 1499–1503. men: Pilot data and HIV prevention implications. AIDS Educa-
Pliszka, S. R. (2007). Pharmacologic treatment of attention-deficit/ tion and Prevention, 15(2), 133–147.
hyperactivity disorder: Efficacy, safety and mechanisms of Semple, S. J., Patterson, T. L., & Grant, I. (2004). A comparison of
action. Neuropsychology Review, 17(1), 61–72. injection and non-injection methamphetamine-using HIV posi-
Rabe-Hesketh, S., Skrondal, A., & Pickles, A. (2004). GLLAMM tive men who have sex with men. Drug and Alcohol Depen-
manual. In Berkeley Division of Biostatistics working paper dence, 76(2), 203–212.
series, paper 160. Berkeley, CA: U.C. Berkeley Division of Semple, S. J., Zians, J., Grant, I., & Patterson, T. L. (2005).
Biostatistics. Retrieved from http://www.bepress.com/ucbbiostat/ Impulsivity and methamphetamine use. Journal of Substance
paper160. Abuse Treatment, 29(2), 85–93.
Ricaurte, G. A., Guillery, R. W., Seiden, L. S., Schuster, C. R., & Shadish, W. R., Robinson, L., & Lu, C. (1999). ES: A computer
Moore, R. Y. (1982). Dopamine nerve terminal degeneration program and manual for effect size calculation. Minneapolis,
produced by high doses of methylamphetamine in the rat brain. MN: Assessment Systems.
Brain Research, 235(1), 93–103. Shoptaw, S. (2006). Methamphetamine use in urban gay and bisexual
Richards, J. R., Bretz, S. W., Johnson, E. B., Turnipseed, S. D., populations. Topics in HIV Medicine, 14(2), 84–87.
Brofeldt, B. T., & Derlet, R. W. (1999). Methamphetamine abuse Shoptaw, S., Peck, J., Reback, C. J., & Rotheram-Fuller, E. (2003).
and emergency department utilization. Western Journal of Psychiatric and substance dependence comorbidities, sexually
Medicine, 170(4), 198–202. transmitted diseases, and risk behaviors among methamphet-
Rippeth, J. D., Heaton, R. K., Carey, C. L., Marcotte, T. D., Moore, D. amine-dependent gay and bisexual men seeking outpatient drug
J., Gonzalez, R., et al. (2004). Methamphetamine dependence abuse treatment. Journal of Psychoactive Drugs, 35(Suppl 1),
increases risk of neuropsychological impairment in HIV infected 161–168.
persons. Journal of the International Neuropsychological Society, Silber, B. Y., Croft, R. J., Papafotiou, K., & Stough, C. (2006). The
10(1), 1–14. acute effects of d-amphetamine and methamphetamine on
Ross, B., Lin, A., Harris, K., Bhattacharya, P., & Schweinsburg, B. attention and psychomotor performance. Psychopharmacology,
(2003). Clinical experience with 13C MRS in vivo. NMR in 187(2), 154–169.
Biomedicine, 16(6–7), 358–369. Sim, T., Simon, S. L., Domier, C. P., Richardson, K., Rawson, R. A.,
Sadek, J. R., Vigil, O., Grant, I., & Heaton, R. K. (2007). The impact & Ling, W. (2002). Cognitive deficits among methamphetamine
of neuropsychological functioning and depressed mood on users with attention deficit hyperactivity disorder symptomatol-
functional complaints in HIV-1 infection and methamphetamine ogy. Journal of Addictive Diseases, 21(1), 75–89.
dependence. Journal of Clinical and Experimental Neuropsy- Simon, S. L., Dacey, J., Glynn, S., Rawson, R., & Ling, W. (2004).
chology, 29(3), 266–276. The effect of relapse on cognition in abstinent methamphetamine
Salo, R., Nordahl, T. E., Moore, C., Waters, C., Natsuaki, Y., abusers. Journal of Substance Abuse Treatment, 27(1), 59–66.
Galloway, G. P., et al. (2005). A dissociation in attentional Simon, S. L., Domier, C., Carnell, J., Brethen, P., Rawson, R., & Ling, W.
control: Evidence from methamphetamine dependence. Biologi- (2000). Cognitive impairment in individuals currently using meth-
cal Psychiatry, 57(3), 310–313. amphetamine. American Journal on Addictions, 9(3), 222–231.
296 Neuropsychol Rev (2007) 17:275–297

Simons, J. S., Oliver, M. N., Gaher, R. M., Ebel, G., & Brummels, P. Ujike, H., & Sato, M. (2004). Clinical features of sensitization to
(2005). Methamphetamine and alcohol abuse and dependence methamphetamine observed in patients with methamphetamine
symptoms: Associations with affect lability and impulsivity in a dependence and psychosis. Annals of the New York Academy of
rural treatment population. Addictive Behaviors, 30(7), 1370– Sciences, 1025, 279–287.
1381. Volkow, N. D., Chang, L., Wang, G. J., Fowler, J. S., Ding, Y. S.,
Smith, D. E., & Fischer, C. M. (1970). An analysis of 310 cases of Sedler, M., et al. (2001a). Low level of brain dopamine D2
acute high-dose methamphetamine toxicity in Haight-Ashbury. receptors in methamphetamine abusers: Association with metab-
Clinical Toxicology, 3(1), 117–124. olism in the orbitofrontal cortex. American Journal of Psychiatry,
Soetens, E., Casaer, S., D’Hooge, R., & Hueting, J. E. (1995). Effect 158(12), 2015–2021.
of amphetamine on long-term retention of verbal material. Volkow, N. D., Chang, L., Wang, G. J., Fowler, J. S., Franceschi, D.,
Psychopharmacology, 119(2), 155–162. Sedler, M., et al. (2001b). Loss of dopamine transporters in
Srisurapanont, M., Ali, R., Marsden, J., Sunga, A., Wada, K., & methamphetamine abusers recovers with protracted abstinence.
Monteiro, M. (2003). Psychotic symptoms in methamphetamine Journal of Neuroscience, 21(23), 9414–9418.
psychotic in-patients. International Journal of Neuropsychophar- Volkow, N. D., Chang, L., Wang, G. J., Fowler, J. S., Leonido-Yee,
macology, 6(4), 347–352. M., Franceschi, D., et al. (2001c). Association of dopamine
StataCorp. (2006). Stata Statistical Software: Release 9.2. College transporter reduction with psychomotor impairment in metham-
Station, TX. phetamine abusers. American Journal of Psychiatry, 158(3),
Stuss, D. T., & Alexander, M. P. (2007). Is there a dysexecutive 377–382.
syndrome? Philosophical Transactions of the Royal Society of von Mayrhauser, C., Brecht, M. L., & Anglin, M. D. (2002). Use
London, Series B: Biological Sciences, 362(1481), 901–915. ecology and drug use motivations of methamphetamine users
Substance Abuse and Mental Health Services Administration (2006a). admitted to substance abuse treatment facilities in Los Angeles: An
Drug abuse warning network, 2004: National estimates of drug- emerging profile. Journal of Addictive Diseases, 21(1), 45–60.
related emergency department visits (DAWN series D-28, DHHS Voytek, B., Berman, S. M., Hassid, B. D., Simon, S. L., Mandelkern,
publication no. SMA 06-4143). Rockville, MD: Office of M. A., Brody, A. L., et al. (2005). Differences in regional brain
Applied Studies. metabolism associated with marijuana abuse in methamphet-
Substance Abuse and Mental Health Services Administration. amine abusers. Synapse, 57(2), 113–115.
(2006b). Results from the 2005 national survey on drug use Wagner, G. C., Ricaurte, G. A., Seiden, L. S., Schuster, C. R., Miller,
and health: National findings (NSDUH Series H-30, DHHS R. J., & Westley, J. (1980). Long-lasting depletions of striatal
publication no. SMA 06-4194). Rockville, MD: Office of dopamine and loss of dopamine uptake sites following repeated
Applied Studies. administration of methamphetamine. Brain Research, 181(1),
Substance Abuse and Mental Health Services Administration. (2006c). 151–160.
Treatment episode data set (TEDS): 1994–2004 (DASIS series: Wang, G. J., Volkow, N. D., Chang, L., Miller, E., Sedler, M.,
S-33, DHHS publication no. SMA 064180). Rockville, MD: Hitzemann, R., et al. (2004). Partial recovery of brain metabolism
Office of Applied Studies. in methamphetamine abusers after protracted abstinence. Amer-
Sung, Y. H., Cho, S. C., Hwang, J., Kim, S. J., Kim, H., Bae, S., et al. ican Journal of Psychiatry, 161(2), 242–248.
(2006). Relationship between N-acetyl-aspartate in gray and Wiegmann, D. A., Stanny, R. R., McKay, D. L., Neri, D. F., &
white matter of abstinent methamphetamine abusers and their McCardie, A. H. (1996). Methamphetamine effects on cognitive
history of drug abuse: A proton magnetic resonance spectroscopy processing during extended wakefulness. International Journal
study. Drug and Alcohol Dependence, 88(1), 28–35. of Aviation Psychology, 6(4), 379–397.
Taylor, M. J., Letendre, S. L., Schweinsburg, B. C., Alhassoon, O. M., Wilens, T. E., Prince, J. B., Biederman, J., Spencer, T. J., & Frances,
Brown, G. G., Gongvatana, A., et al. (2004). Hepatitis C virus R. J. (1995). Attention-deficit hyperactivity disorder and comor-
infection is associated with reduced white matter N-acetylaspartate bid substance use disorders in adults. Psychiatric Services, 46(8),
in abstinent methamphetamine users. Journal of the International 761–763, 765.
Neuropsychological Society, 10(1), 110–113. Wilson, J. M., Kalasinsky, K. S., Levey, A. I., Bergeron, C., Reiber,
Taylor, M., Schweinsburg, B., Alhassoon, O., Gongvatana, A., Brown, G., Anthony, R. M., et al. (1996). Striatal dopamine nerve
G., Young-Casey, C., et al. (2007). Effects of human immuno- terminal markers in human, chronic methamphetamine users.
deficiency virus and methamphetamine on cerebral metabolites Nature Medicine, 2(6), 699–703.
measured with magnetic resonance spectroscopy. Journal of Woods, S. P., Carey, C. L., Troster, A. I., & Grant, I. (2005a). Action
Neurovirology, 13(2), 150–159. (verb) generation in HIV-1 infection. Neuropsychologia, 43(8),
Thompson, P. M., Hayashi, K. M., Simon, S. L., Geaga, J. A., Hong, 1144–1151.
M. S., Sui, Y., et al. (2004). Structural abnormalities in the brains Woods, S. P., Conover, E., Rippeth, J. D., Carey, C. L., Gonzalez, R.,
of human subjects who use methamphetamine. Journal of Marcotte, T. D., et al. (2004). Qualitative aspects of verbal
Neuroscience, 24(26), 6028–6036. fluency in HIV-associated dementia: A deficit in rule-guided
Trevisan, L. A., Boutros, N., Petrakis, I. L., & Krystal, J. H. (1998). lexical-semantic search processes? Neuropsychologia, 42(6),
Complications of alcohol withdrawal: Pathophysiological 801–809.
insights. Alcohol Health and Research World, 22(1), 61–66. Woods, S. P., Rippeth, J. D., Conover, E., Gongvatana, A., Gonzalez,
Troyer, A. K., Moscovitch, M., Winocur, G., Alexander, M. P., & R., Carey, C. L., et al. (2005b). Deficient strategic control of
Stuss, D. (1998). Clustering and switching on verbal fluency: verbal encoding and retrieval in individuals with methamphet-
The effects of focal frontal- and temporal-lobe lesions. Neuro- amine dependence. Neuropsychology, 19(1), 35–43.
psychologia, 36(6), 499–504. Woolverton, W. L., Ricaurte, G. A., Forno, L. S., & Seiden, L. S.
Ujike, H., Harano, M., Inada, T., Yamada, M., Komiyama, T., Sekine, (1989). Long-term effects of chronic methamphetamine admin-
Y., et al. (2003). Nine- or fewer repeat alleles in VNTR istration in rhesus monkeys. Brain Research, 486(1), 73–78.
polymorphism of the dopamine transporter gene is a strong risk Yen, D. J., Wang, S. J., Ju, T. H., Chen, C. C., Liao, K. K., Fuh, J. L.,
factor for prolonged methamphetamine psychosis. Pharmacoge- et al. (1994). Stroke associated with methamphetamine inhala-
nomics Journal, 3(4), 242–247. tion. European Neurology, 34(1), 16–22.
Neuropsychol Rev (2007) 17:275–297 297

Yui, K., Ikemoto, S., & Goto, K. (2002). Factors for susceptibility to phetamine: Characterization suggests the presence of a factor
episode recurrence in spontaneous recurrence of methamphet- responsible for maintaining sequestration. Journal of Neuro-
amine psychosis. Annals of the New York Academy of Sciences, chemistry, 54(1), 195–204.
965, 292–304. Zweben, J. E., Cohen, J. B., Christian, D., Galloway, G. P., Salinardi,
Zaczek, R., Culp, S., & De Souza, E. B. (1990). Intrasynaptosomal M., Parent, D., et al. (2004). Psychiatric symptoms in metham-
sequestration of [3H]amphetamine and [3H]methylenedioxyam- phetamine users. American Journal on Addictions, 13(2), 181–190.

You might also like