11
Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease http://dx.doi.org/10.1016/B978-0-12-410529-4.00018-8 197 © 2015 Elsevier Inc. All rights reserved. CHAPTER 18 Autism Spectrum Disorders: Clinical Considerations Patricia Evans, Sailaja Golla, and Mary Ann Morris The University of Texas Southwestern Medical Center, Dallas, TX, USA INTRODUCTION Autism spectrum disorders (ASD) are a group of neurodevelopmental disabilities that typically first appear in childhood and are characterized by significant difficulties in social, communicative, and behavioral functioning. Although no one single cause has been found, it is suspected that many etiologies have a common final pathway within early neurologic development. Bearing a diagnosis of an ASD typically causes an individual and his/ her family difficult financial, emotional, and at times, physical burdens over the individual’s lifespan. New bur- dens are being experienced by state and federal agencies as communities seek ways to medically and financially support the increasing volume of adults with ASD. Consequently, research continues to better understand the complex process of ASD, find disease-specific interventions, and better ways to enable communities to provide resources for families. OVERVIEW Identifying individuals with an ASD ideally starts in infancy, and requires two levels of surveillance, each ad- dressing a distinct component of patient management. Routine developmental surveillance and screening specif- ically for ASD has been recommended to be performed on all children, with subsequent in-depth investigation specific for ASD as needed. Such an approach ensures that all children with risk for any type of atypical development are screened, and within that group, specifically identifying those children with a possible ASD, with appropriate referral. Specific information about the recommended developmental screening and diagnostic tools can be found at the American Academy of Neurology (AAN) website (http://www.aan.com). 1 The American Academy of Pediatrics as well as the AAN recommend developmental surveillance at all well-child visits from infancy through school age, and at any time afterwards in the context of concerns regarding social accep- tance, learning, or behavior. 2–4 A child has failed level one surveillance if he or she fails to meet any of the following specific milestones: no evidence of babbling by 12 months; no gesturing by 12 months; the lack of single words by 16 months; no two-word spontaneous phrases by 24 months; and finally, loss of any previously acquired language or social skills at any age. 3,4 Additionally, siblings of children with an ASD should be carefully monitored for acqui- sition of social, communication, and play skills, and the occurrence of maladaptive behaviors. Screening should be performed not only for ASD-related symptoms but also for language delays, learning difficulties, social problems, and anxiety or depressive symptoms. Screening specifically for an ASD should be performed on all children failing routine developmental surveillance procedures using one of the validated instruments. A release for all school records as well as any private, individ- ualized assessment is important to attain pertinent information. A child having failed initial surveillance typically requires assessment by practitioners with particular interest and training in ASD. CHAPTER 18

Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

Embed Size (px)

Citation preview

Page 1: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease http://dx.doi.org/10.1016/B978-0-12-410529-4.00018-8 197 © 2015 Elsevier Inc. All rights reserved.

C H A P T E R

18

Autism Spectrum Disorders: Clinical Considerations

Patricia Evans, Sailaja Golla, and Mary Ann Morris The University of Texas Southwestern Medical Center, Dallas, TX, USA

INTRODUCTION

Autism spectrum disorders (ASD) are a group of neurodevelopmental disabilities that typically first appear in childhood and are characterized by significant difficulties in social, communicative, and behavioral functioning. Although no one single cause has been found, it is suspected that many etiologies have a common final pathway within early neurologic development. Bearing a diagnosis of an ASD typically causes an individual and his/her family difficult financial, emotional, and at times, physical burdens over the individual’s lifespan. New bur-dens are being experienced by state and federal agencies as communities seek ways to medically and financially support the increasing volume of adults with ASD. Consequently, research continues to better understand the complex process of ASD, find disease-specific interventions, and better ways to enable communities to provide resources for families.

OVERVIEW

Identifying individuals with an ASD ideally starts in infancy, and requires two levels of surveillance, each ad-dressing a distinct component of patient management. Routine developmental surveillance and screening specif-ically for ASD has been recommended to be performed on all children, with subsequent in-depth investigation specific for ASD as needed. Such an approach ensures that all children with risk for any type of atypical development are screened, and within that group, specifically identifying those children with a possible ASD, with appropriate referral. Specific information about the recommended developmental screening and diagnostic tools can be found at the American Academy of Neurology (AAN) website (http://www.aan.com).1

The American Academy of Pediatrics as well as the AAN recommend developmental surveillance at all well-child visits from infancy through school age, and at any time afterwards in the context of concerns regarding social accep-tance, learning, or behavior.2–4 A child has failed level one surveillance if he or she fails to meet any of the following specific milestones: no evidence of babbling by 12 months; no gesturing by 12 months; the lack of single words by 16 months; no two-word spontaneous phrases by 24 months; and finally, loss of any previously acquired language or social skills at any age.3,4 Additionally, siblings of children with an ASD should be carefully monitored for acqui-sition of social, communication, and play skills, and the occurrence of maladaptive behaviors. Screening should be performed not only for ASD-related symptoms but also for language delays, learning difficulties, social problems, and anxiety or depressive symptoms.

Screening specifically for an ASD should be performed on all children failing routine developmental surveillance procedures using one of the validated instruments. A release for all school records as well as any private, individ-ualized assessment is important to attain pertinent information. A child having failed initial surveillance typically requires assessment by practitioners with particular interest and training in ASD.

C H A P T E R

18

Page 2: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

198 18. AuTISm SPECTRum DISoRDERS: ClInICAl ConSIDERATIonS

II. NEUROLOGIC DISEASES

CLINICAL FEATURES AND DIAGNOSTIC EVALUATION

The fifth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5)2 was released in May 2013. The changes from the previous Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, Text Revision (DSM-IV-TR)3 have been as dramatic as they have been controversial: DSM-5 no longer contains the subdiagnoses of Autistic Disorder, Asperger Syndrome, Pervasive Developmental – Not Otherwise Specified, Childhood Disintegrative Disorder, or Rett Disorder. The DSM-IV-TR’s autistic symptoms were divided into three areas, specifically, social reciprocity, communicative intent, restricted and repetitive behaviors; while the new DSM-5’s diagnostic criteria has been reduced into two areas, specifically, social communication/interaction and restricted/repetitive behaviors. If the diagnostic criteria for ASD are met, then the diagnosis must also specify if it is with or without accompanying intellectual impairment and/or language impairment, as well as noting if the diagnosis is associated with a known medical or genetic condition and/or environmental factor. Additionally, the ASD diagnosis must also be specified by severity levels (Level 1 – Requiring Support, Level 2 – Requiring Substantial Support, and Level 3 – Requiring Very Substantial Support).2,4 Children who have deficits only in social communication should be considered for the new DSM-5 diagnostic category of Social Communication Disorder (SCD).4,5

Clinical Diagnostic Procedures and Methods

Interview MethodThe Autism Diagnostic Interview – Revised (ADI-R)6 is a structured interview conducted with the parent or care-

taker of a child and adult with a mental age of at least 2 years old and who has been referred for the evaluation of a possible ASD. The ADI-R is useful for diagnosing ASD, planning treatment, and distinguishing ASD for other developmental disorders.6–8

The ADI-R typically takes 1–2 hours and focuses on the child’s current behavior or behavior at a certain point in the areas of reciprocal social interaction, communication and language, and patterns of behavior.7 The interview is divided into five sections: opening questions, communication questions, social development and play questions, repetitive and restricted behavior questions, and questions about general behavior problems. Due to the lengthy interview, the ADI-R is primarily used in clinical or research settings.8

Observational MethodsThe Autism Diagnostic Observation Schedule, Second Edition (ADOS-2) is a semi-structured set of observations

and series of activities involving the referred individual and a trained examiner.9 This revision improves an instru-ment already viewed as “the gold standard” for observational assessment of an ASD. With updated protocols, re-vised algorithms, a new Comparison Score, and a Toddler Module,10 the ADOS-2 provides a highly accurate picture of current symptoms, unaffected by language. It can be used to evaluate almost anyone suspected of having ASD, as young as a 12 months old with no language to verbally fluent adults.

Like its predecessor, the ADOS,11 the ADOS-2 is a semi-structured, standardized assessment of communication, social interaction, play, and restricted and repetitive behaviors. It presents various activities that elicit behaviors directly related to a diagnosis of ASD. By observing and coding these behaviors, information can be attained for diagnosis, treatment planning, and educational placement.

In Modules 1 through 4, algorithm scores are compared with cut-off scores to yield one of three classifications: Autism, Autism Spectrum, and Non-Spectrum. The difference between Autism and Autism Spectrum classifications is one of severity, with the former indicating more pronounced symptoms.9 In the Toddler Module, algorithms produce “ranges of concern” rather than classification scores.10

The Childhood Autism Rating Scale, Second Edition (CARS2) is a widely used autism assessment tool.12 The revised second edition expands its responsiveness to individuals that are high functioning on the autism spectrum with average and above cognitive abilities, as well as better verbal skills, and more subtle social and behavioral deficits.

The CARS2 includes the Standard Version Rating Booklet, for use with individuals younger than 6 years of age with communication challenges or below-average cognitive abilities, and the High-Functioning Version Rating Booklet, for the assessment of verbally fluent individuals, 6 years of age and older, with cognitive abilities above 80. Rating values for all items are summed to produce a total raw score that is converted to a standard score or percentile rank, as well as guidelines for score interpretation and suggestions for intervention.12

Page 3: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

ClInICAl fEATuRES AnD DIAgnoSTIC EvAluATIon 199

II. NEUROLOGIC DISEASES

Checklist and Rating Scale MethodsThe Social Responsiveness Scale, Second Edition (SRS-2) can be completed in approximately 15 minutes and iden-

tifies social impairment associated with ASD, as well as quantifying its severity.13 The SRS-2 has a Preschool Form (for ages 2.5–4.5 years) and the School-Age Form (for ages 4–18 years), which are completed by a parent/guardian or teacher. The Adult (Relative/Other Report) Form is for ages 19 years and up and is completed by a relative or friend and the Adult (Self-Report) Form is completed by the individual and begins at 19 years of age through adulthood.

The SRS-2 generates scores for Social Awareness, Social Cognition, Social Communication, Social Motivation, Restricted Interests, and Repetitive Behavior treatment scales that are useful in determining appropriate treatment and intervention.13

The Social Communication Questionnaire (SCQ) is a brief, 35-item, true/false questionnaire, completed by par-ents or guardians regarding the communication skills and social functioning of children who may be suspected of having ASD.14 It parallels the ADI-R in content and is used for brief screening to determine the need to conduct a full ADI-R interview and/or the ADOS-2.7,9 Because the SCQ is brief, quick, and easily completed by a parent or caregiver in less than 10 minutes, it allows clinicians and educators to routinely screen children for ASD, as well as determine the need for comprehensive diagnostic evaluation.

The Gilliam Autism Rating Scale, Third Edition (GARS-3) assists parents/guardians, teachers, and clinicians in identifying ASD in individuals, as well as estimating its severity.13 The GARS-3 items are based on the DSM-5 di-agnostic criteria for ASD.2 The GARS-3 yields standard scores, percentile ranks, severity levels, and the probability of an ASD. The GARS-3 is normed for ages 3-22 years, and takes approximately 10 minutes to rate. The instrument consists of 56 clearly stated items describing the characteristic behaviors of persons with ASD. The items are grouped into six subscales: Restrictive, Repetitive Behaviors, Social Interaction, Social Communication, Emotional Responses, Cognitive Style, and Maladaptive Speech.15

Medical Assessment

A detailed and careful history is imperative, and should include probes into family history with potential risk for consanguinity, postnatal deaths, and perinatal issues.16 Family prevalence is important: family studies note a 50–100-fold increase in the rate of ASD in first-degree relatives of autistic children.17 For all referrals of individuals with an ASD, a careful review of psychometric measures is critical, which has been detailed above. Children with ASD have an increased rate of larger head circumferences, often noted over the first few years of life rather than at birth, although only a few will have a true macrocephaly.1,18,19 There are well-known associations with a range of genetic disorders, including, but not limited to, tuberous sclerosis complex, fragile X syndrome, Angelman Syndrome, and a wide range of DNA genetic variations.20–25

The physical exam should include the graphing of height, weight, and head circumference, particularly in young children, as well as simply observing the child’s interaction with his family and caregivers during history gathering. In particular, attention should be given to possible dysmorphology, curvature of spine, the shape and size of the head, and the appearance of any birth marks.1,18,19 The degree to which a child can make and sustain eye contact with family members as well as with the examiners should be specifically described, and adjectives such as fleeting, brief, intermittent, and others, may all be appropriate to the exam. The mental status exam should include a description of the speech patterns, if present; the level of social maturity relative to age; and the capacity to engage with the exam-iner.26–28 Attention should be also given to the range and quality of speech; language deficits may range from mutism to verbal fluency, but with other pragmatic and receptive language difficulties.27,28 The neurologic examination may be quite limited depending upon the anxiety or timidity of the child; however, attempts should be made to assess optic discs, visual fields, strength, tone, at least a general sensory exam, gait, and coordination.1,19

Laboratory investigations that are recommended in the context of developmental delay, with or without elements of an ASD, typically include a formal audiology examination as well as lead screening for children who have had an extended oral–motor stage of play, even in the absence of true pica.4,18 Additional laboratory investigations will be guided by findings on exam; however, genetic testing yields abnormalities in more than 1% of children with an au-tistic phenotype, including abnormalities in the proximal long arm of chromosome 15q, such as Angelman Syndrome and Prader–Willi syndrome, disorders typically associated with moderate to severe intellectual impairment.22,23 Less than 5% of all children with an ASD phenotype have inborn errors of metabolism; however, assessing serologies for abnormalities in amino acid, carbohydrate, purine, peptide, and mitochondrial metabolism must be made based on each individual case.4,18,30 Electrophysiological testing is dictated by the history and physical. The prevalence of epilepsy in autistic children has been estimated at 7%18 to 14%,28 whereas the cumulative prevalence by adulthood is estimated at 20–35%.31,32 Seizure onset peaks in early childhood and again in adolescence. Intellectual disability, with

Page 4: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

200 18. AuTISm SPECTRum DISoRDERS: ClInICAl ConSIDERATIonS

II. NEUROLOGIC DISEASES

or without motor abnormalities or family history of epilepsy, is a significant risk factor for the development of sei-zures in individuals with an ASD.31–33 A higher incidence of epileptiform electroencephalogram (EEG) abnormalities in autistic children whose presentation includes a history of regression has been reported when compared to autistic children without clinical regression.34,35 Seizures or epileptiform discharges were more prevalent in children with re-gression who demonstrated cognitive deficits. Regression in cognition and language in adolescence associated with seizure onset has also been observed, but little is known about its cause or prevalence. There may be a causal rela-tionship between a subgroup of children with autistic regression and EEG-defined benign focal epilepsies, although there has not yet been consensus on how to treat.35 Radiographic images are rarely indicated in children with an autistic phenotype unless there is evidence for focal findings by exam or by EEG. Prevalence of lesions on magnetic resonance imaging (MRI) in children with an ASD is similar to normal control subjects.1,4,30,36 Computed tomography (CT) and MRI studies of autistic subjects screened to exclude those with disorders other than ASD have confirmed the absence of significant structural brain abnormalities.36,37 There is no evidence to support a role for functional neu-roimaging studies in the clinical diagnosis of an ASD at the present time.1,4,30

There is insufficient evidence to support the use of other tests such as hair analysis for trace elements, celiac an-tibodies, allergy testing for gluten, casein, candida, and other molds, immunologic or neurochemical abnormalities, micronutrients such as vitamin levels, intestinal permeability studies, stool analysis, urinary peptides, mitochondrial disorders, thyroid function tests, or erythrocyte glutathione peroxidase studies.4

THERAPEUTIC APPROACHES

Because of the complexity of the ASD condition, reducing the social and emotional discomfort for an individual with an ASD requires a multidisciplinary approach. Such approaches typically include providing supportive envi-ronments at home, school, and where applicable, work. Additionally, therapeutics that involve behavior modifica-tion and enhancement of effective communication are important. Finally, the use of psychopharmacologic agents can be helpful in reducing troublesome issues of disruptive, aggressive, or anxious behaviors.

Environmental Approaches

After a diagnosis of an ASD has been made, it is optimal to invite all caregivers to participate in a relaxed and sup-portive conference with the medical team to review specific challenges. Although predictability and consistent rou-tines are often difficult for many 21st century families, such qualities are key to an individual’s successful outcome.38 Evidence continues to reinforce that the earlier and more inclusive the intervention, the more successful individuals are. The individual’s environment must include consistency in schedules and physical space as well as supportive relationships between all caregivers.38–40

Nonpharmacologic Therapeutic Options

In addition to environmental supports and structure, the addition of formal therapies has been shown to be crit-ically important for many children and teenagers. Although speech and occupational therapies are particularly im-portant for infants and young children, the introduction of appropriate therapies at any point in development, from childhood to the adult years, can yield significant improvement in an individual’s independence and capacity for interaction with others. Of these, five are the most commonly used, and include speech and language therapy; occu-pational therapy; behavior therapy; social skills therapy; and virtual reality therapeutics.

Speech and language therapies include a wide range of techniques and methods, and may include didactic and naturalistic behavioral therapy: verbal behavior, natural language paradigm, pivotal response training, and milieu teaching have been studied.19,29,38,41 Augmentative and alternative communication modalities, such as American Sign Language, picture and communication boards, and even the child’s best gestural commands, can all be effective in building communication skills.41–44

Occupational therapy has been the cornerstone of addressing many of the sensory issues found in this population of children. While there are many approaches, the general goals seek to reduce sensory overstimulation and provide improved focus for learning tasks, both cognitively and for visual motor skills.45–47

Behavioral and social skills therapies are also important, and can range in goals from better navigating the prag-matics of conversation to simply not being as aggressive when feeling anxious, and include the use of discrete trial teaching (DTT) and applied behavioral analysis (ABA). Certainly the use of practicing scripted dialogue, particularly with improved eye contact, can provide individuals with tools to better understand their social environments.41,43,44,48

Page 5: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

THERAPEuTIC APPRoACHES 201

II. NEUROLOGIC DISEASES

Finally, individuals with an ASD can be greatly assisted by the emerging field of virtual reality therapeutics. Although often expensive, immersive virtual reality (VR) has the potential to be very helpful in coaching socializa-tion and self-confidence. Some studies have observed an increase in gaze duration, vocalizations, as well as signifi-cant reduction in social anxiety. Further research is needed in this very promising field.49,50

Pharmacologic Interventions

Patients with ASD typically present with impairments associated with social interaction, communication difficul-ties, and stereotyped behaviors. These difficulties most often manifest as relative degrees of aggression, self-injurious behaviors, irritability, sleep problems, anxiety, mood disorders, obsessive–compulsive behaviors, inattention, overactivity, and impulsiveness. Such behavioral problems may be responsive to the combination of environmental support, nontherapeutic interventions as reviewed, and adjunctive pharmacological intervention as well. Research into the pharmacotherapy of ASD and related disorders have been ongoing since the middle of the 20th century. But the utility of earlier research is limited by methodological flaws. Behavioral heterogeneity, over interpretation of case studies, and lack of appropriate outcome measures have been some of the major problems.51 Most current pharmaco-therapies target one or more of the several major biochemical pathways, most commonly through the dopaminergic and serotonergic pathways.52,53 A comprehensive review of psychopharmacology is beyond the scope of this chapter. Readers are referred to the ninth edition of Kaplan and Sadock’s Comprehensive Textbook of Psychiatry for a detailed discussion of psychopharmacology referenced below.54

Neuroleptics (Antipsychotics)Neuroleptics typically block D2 receptors in the dopaminergic system. In addition to D2 antagonistic effects,

neuroleptics also block serotonin receptors, with newer generations of atypical neuroleptics demonstrating greater selectivity of receptors.54 The use of atypical antipsychotics in children with ASD is based, in part, on their efficacy in the treatment of psychiatric disorders that have behavioral symptoms similar to the core symptoms seen with ASD, as well as their relatively safe adverse effect profile in comparison with older, conventional antipsychotics, and the possible role of dopamine in the etiology of ASD.55–57

Typical neuroleptics are less selective than atypical neuroleptics, and because the typical neuroleptics more often block dopamine receptors in the mesocortical pathway, tuberoinfundibular pathway, and the nigrostriatal pathway, typical neuroleptics such as haloperidol provoke frequent side effects, which include sedation, headaches, dizziness, hyperprolactinemia, sexual dysfunction, osteoporosis, weight gain, and orthostasis.54,58 Neuroleptics also produce an-ticholinergic effects, including angle-closure glaucoma, constipation, dry mouth, and reduced perspiration. Tardive dyskinesias occur more often for individuals taking high-potency first-generation neuroleptics such as haloperidol, and also appear after chronic, rather than acute, exposure to the drug. Tardive dyskinesias are manifest by repetitive, involuntary, and purposeless movements, most often of the face, lips, legs or torso; they tend to resist treatment and are frequently irreversible.54 Neuroleptic malignant syndrome is a potentially fatal condition characterized by au-tonomic instability, which can manifest with tachycardia, nausea, vomiting, diaphoresis, and hyperthermia; mental status alteration; and muscle rigidity, any of which may be associated with laboratory abnormalities, including ele-vated creatinine kinase, reduced iron plasma levels, and electrolyte derangements.54 Finally, the use of neuroleptics has also been associated with pancreatitis, pharyngitis, and changes on electrocardiogram, specifically a QT interval prolongation. Seizures have been observed, especially in the context of chlorpromazine and clozapine. Finally, other cardiac complications may include thromboembolism, myocardial infarction, stroke, and torsades de pointes.

Haloperidol, a typical neuroleptic, was one of the first medications studied systematically as a treatment for ASD-related symptoms. The effects of typical neuroleptics, such as haloperidol and chlorpromazine, are primarily me-diated through dopamine-receptor (D2) antagonism, and have been shown to be effective in targeting a range of maladaptive behavioral symptoms, withdrawal, and stereotypy, especially in school-age children.55,56 However, sig-nificant adverse effects including dose-related sedative effects and extrapyramidal symptoms such as dystonias and dyskinesias limit the use of haloperidol primarily to treatment-refractory patients.56 Hence, atypical neuroleptics, with significantly less severe extrapyramidal side effects, have emerged as the first-line drugs in the management of children with ASD.

Atypical neuroleptics, by contrast, are potent serotonin-receptor as well as dopamine-receptor antagonists, and these include aripiprazole, olanzapine, quetiapine, risperidone, and ziprasidone.54,59 The serotonin receptor antag-onism decreases the propensity for tardive dyskinesias and extrapyramidal symptoms. Risperidone is one of the most commonly used medications in this group. In 2003, the US Food and Drug Administration (FDA) approved risperidone for the short-term treatment of the mixed and manic states associated with bipolar disorder. In 2006,

Page 6: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

202 18. AuTISm SPECTRum DISoRDERS: ClInICAl ConSIDERATIonS

II. NEUROLOGIC DISEASES

the FDA approved risperidone for the treatment of irritability in children and adolescents with autism. The FDA’s decision was based, in part, on a study of autistic people with severe and enduring problems of violent meltdowns, aggression, and self-injury. Risperidone is not recommended for autistic people with mild aggression and explosive behavior without an enduring pattern. Weight gain is a major adverse effect of this drug. Hence, it is important to regularly monitor body mass index, lipid panel and blood glucose in patients receiving risperidone.58–60 Aripiprazole is another commonly used atypical neuroleptic. Weight gain is considered less severe with aripiprazole compared to risperidone, although some of the more recent studies have shown significant weight gain with it.59,61,62 Ziprasidone is found to be weight neutral, but can potentially cause QTc prolongation on the electrocardiogram.63

StimulantsHyperactivity, inattention, and impulsiveness are common traits among individuals with ASD. Stimulants have

been shown to decrease these symptoms in a significant proportion of such patients, although these agents can be problematic in this group of patients because of the propensity to exacerbate anxiety, a feature common in ASD.64,81 Stimulants exert their effects through a number of different pharmacological mechanisms, the most prominent of which include facilitation of norepinephrine, or dopamine, or both; adenosine receptor antagonism; and nicotinic acetylcholine receptor agonism.65 Commonly used stimulants in the ASD population include amphetamines; meth-ylphenidate; and dopamine and/or norepinephrine reuptake inhibitors.66

Amphetamines are a group of phenylethylamine stimulants such as amphetamine, methamphetamine, Dexedrine and levoamphetamine (Adderall). Amphetamines increase the levels of norepinephrine and dopamine in the brain via reuptake inhibition; however, the more important mechanism by which amphetamines cause stimulation is through the direct release of these catecholamines from storage vesicles in cells. Amphetamines are known to cause elevated mood and euphoria as well as rebound depression and anxiety, the latter a particular challenge for many individuals with an ASD.54,66

Methylphenidate (MPH; Ritalin, Concerta, Metadate, or Methylin) is a drug approved for treatment of attention- deficit hyperactivity disorder (ADHD), postural orthostatic tachycardia syndrome, and narcolepsy.65 It belongs to the piperidine class of compounds and increases the levels of dopamine and norepinephrine in the brain through reuptake inhibition of the monoamine transporters. It also increases the release of dopamine and norepinephrine. MPH possesses structural similarities to amphetamine, and, although it is less potent, its pharmacological effects are even more closely related to those of cocaine.65 A recent meta-analysis reviewed seven randomized-controlled trials looking at the effects of MPH on ADHD symptoms in children with pervasive developmental delay.64 This review found MPH to be an effective treatment for ADHD symptoms in these children, but noting that weight loss and car-diac toxicity are two of the major side effects associated with its use.

Norepinephrine and/or dopamine reuptake inhibitors (NRIs) also function as stimulants, and inhibit the reuptake of norepinephrine and/or dopamine, resulting in increased extracellular levels and, therefore, enhanced neurotrans-mission, ultimately producing a stimulant effect. Among individuals with an ASD, the NRI atomoxetine (Strattera) is commonly used in this population of patients. Limited studies have demonstrated its use in ASD as being both effective and safe.64–66

Also used is bupropirone (Wellbutrin), which blocks dopamine primarily, and to a much lesser extent, nor-epinephrine. Both drugs have a considerably lower abuse potential in comparison to other stimulants like the amphetamines.67,68

Alpha Adrenergic AgonistsClonidine and guanfacine are among the most commonly used alpha adrenergic agents in ASD in which inat-

tention and impulsivity prominently feature. The mechanism of action in the treatment of ADHD is to increase noradrenergic tone in the prefrontal cortex (PFC) directly by binding to postsynaptic α2A adrenergic receptors and indirectly by increasing norepinephrine input from the locus coeruleus.69,70 Alpha 2 agonists have been used as stimulant-treatment extenders in the context of immediate-release methylphenidate or amphetamine, when the goal is to limit both the number of stimulant doses given per day and extend the duration of therapeutic action in com-bination with a stimulant.71,72 Also, alpha 2 agonists, with their pronounced sedative side effects, are used to reduce the delay in sleep onset, a problem common in children with ADHD and impulse control problems.73 In contrast to clonidine, guanfacine is more selective for the alpha 2A receptor,74 with less alpha 2B activity, making it less sedating than clonidine. The use of these agents as adjunctive sleep aids should be short term at most.75,76

Alpha 2 adrenergic agonists are additionally used with a stimulant to enhance the stimulant efficacy. The use of combination of alpha 2 adrenergic agonists with a stimulant is often used in presence of ADHD with comorbid features, specifically oppositional defiant disorder, Giles de Tourette syndrome, and aggressive/impulsive behavior.

Page 7: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

THERAPEuTIC APPRoACHES 203

II. NEUROLOGIC DISEASES

The rationale for combination therapy in these cases has been that the primary effects of stimulants and alpha 2 agonists are mediated by different neurotransmitter systems. Functional neuroimaging studies78 demonstrate that guanfacine produces selective and circumscribed activation of frontal and frontal association areas while at the same time “turning down” or inhibiting striatal activity. Research suggests that stimulants such as methylphenidate im-prove working memory and regulate attention primarily through alpha 2A receptors to enhance norepinephrine tone in the prefrontal cortex.79 These effects of methylphenidate on prefrontal cortical function can be prevented by blocking either norepinephrine alpha 2 receptors or dopamine D1 receptors,79 establishing that both D1 and alpha 2A receptors are essential to the optimum functioning of the prefrontal cortex.

Anxiolytics and Selective Serotonin Reuptake Inhibitors (SSRIs)Selective serotonin reuptake inhibitors or serotonin-specific reuptake inhibitors (SSRIs) are a class of compounds

typically used as antidepressants in the treatment of depression, anxiety disorders, and some personality disorders. SSRIs are believed to increase the extracellular level of the neurotransmitter serotonin by inhibiting its reuptake into the presynaptic cell, increasing the level of serotonin in the synaptic cleft available to bind to the postsynaptic receptor.80

Buspirone is a serotonin agonist that is used primarily to treat anxiety disorders but has recently proven to be useful to treat ASD, as these children have alteration in brain serotonin synthesis during critical periods of brain develop-ment.82 Serotonin receptor inhibitors (SRIs) have been beneficial for obsessive–compulsive behaviors and repetitive behaviors with ASD. The most commonly used are fluoxetine, fluoxamine, and clomipramine.83–92 Further research is needed to better understand mechanisms affected by SSRIs and serotonin agonists during brain development in ASD.

AnticonvulsantsAnticonvulsants such as valproic acid,93 lamictal,94,95 carbamazepine and oxcarbamazapine96 have shown improve-

ment in a range of symptoms in ASD, particularly as mood stabilizers with mixed results. Overall very few studies and limited evidence limits the use of these medications. Some studies demonstrated some improvement in attention span and impulsivity.

Other Medications and Experimental TherapiesAMANTADINE HYDROCHLORIDE

The psychotropic effect of amantadine is related to its antagonism of the N-methyl-D-aspartate (NMDA) receptor. By decreasing the toxic effects of the glutamatergic neurotransmitter system, amantadine is thought to ameliorate issues particularly related to ASD. Two randomized controlled trials of amantadine were identified in children and adolescents. One reported beneficial effects in controlling the symptoms of irritability and hyperactivity in ASD and the other described a significant impact in ADHD. Studies in adults, with relevance to children and adolescents, re-ported effectiveness in resistant depression, obsessive–compulsive disorder and in counteracting side effects of some psychotropic medications. Available data for such use, although promising, require further confirmation.97

CHOLINESTERASE INHIBITORS

Cholinesterase inhibitors increase acetylcholine levels in the brain and have been used for a long time in the treatment of Alzheimer disease. Some of the commonly used agents are donepezil,98 galantamine,99 and rivastigmine tartrate.100 Very few studies have been attempted on ASD that have shown some improvement with hyperactivity and irritability.

SECRETIN

Evidence from seven randomized controlled trials supports a lack of effectiveness of secretin for the treatment of ASD symptoms. Secretin has been studied extensively in multiple randomized controlled trials, with clear evidence that it lacks any benefit. Given the high strength of evidence for a lack of effectiveness, secretin as a treatment ap-proach for ASDs warrants no further study.101 Other therapies like gluten-free diet102 and hyperbaric oxygen103 also have not shown any significant beneficial effect for children with ASD.

Treatment and Future Research

The goal of all current therapies in ASD is to relieve the core symptoms, most often anxiety, aggression, impulsiv-ity, and inattention. Many patients with ASD receive some form of pharmacotherapy, much of it off-label, with the primary therapeutic aim being to improve behaviors that interfere with behavioral and educational therapies.104–106 Because of the difficulties inherent in conducting randomized, placebo-controlled studies in pediatric populations, evidence of the benefits of this strategy is limited, and most such studies have been small and of short duration.1,30 There remains a great need for research into the long-term benefits and risks of pharmacotherapy in ASD.1,30

Page 8: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

204 18. AuTISm SPECTRum DISoRDERS: ClInICAl ConSIDERATIonS

II. NEUROLOGIC DISEASES

References 1. Filipek PA, Accardo PJ, Ashwal S, et al. Practice parameter: screening and diagnosis of autism: report of the Quality Standards

Subcommittee of the American Academy of Neurology and the Child Neurology Society. Neurology. 2000;55(4):468–479. doi:10.1212/wnl.55.4.468.

2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington, DC: American Psychiatric Association; 2013.

3. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders - Text Revision. 4th ed. Washington, DC: American Psychiatric Association; 2000.

4. Filipek PA, Accardo PJ, Ashwal S, et al. Practice parameter: screening and diagnosis of autism: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Child Neurology Society. Neurology. 2000;55(4):468–479, PMID:10953176.

5. New DSM-5 includes changes to autism criteria. American Academy of Pediatrics. AAP News. Retrieved from www.aapnews.org; June 4, 2013. Accessed 31.10.13.

6. Ozonoff S. Editorial perspective: Autism spectrum disorders in DSM-5 – an historical perspective and the need for change. J Child Psychol Psychiatry. 2012;53:1092–1094.

7. Rutter M, LeCouteur A, Lord C. Autism Diagnostic Interview-Revised (ADI-R). Los Angeles, CA: Western Psychological Services; 2003. 8. Kim SH, Lord C. New autism diagnostic interview-revised algorithms for toddlers and young preschoolers from 12 to 47 months of age.

J Autism Dev Disord. 2012;42(1):82–93. doi:10.1007/s10803-011-1213-1, PMID:21384244. 9. Lord C, Rutter M, DiLavore P, Risi S, Gotham K. Autism Diagnostic Observation Schedule – Second Edition (ADOS-2): Modules 1 through 4. Los

Angeles, CA: Western Psychological Services; 2012. 10. Lord C, Luyster R, Gotham K, Guthrie W. Autism Diagnostic Observation Schedule – Second Edition (ADOS-2): Toddler Module. Los Angeles,

CA: Western Psychological Services; 2012. 11. Lord C, Rutter M, DiLavore P, Risi S. Autism Diagnostic Observation Schedule (ADOS). Los Angeles, CA: Western Psychological Services; 1999. 12. Schopler E, Van Bourgondien ME, Wellman GJ, Love SR. Childhood Autism Rating Scale. 2nd ed. Los Angeles, CA: Western Psychological

Services; 2010. 13. Constantino JM, Gruber CP. Social Responsiveness Scale (SRS-2). 2nd ed. Los Angeles, CA: Western Psychological Services; 2012. 14. Rutter M, Bailey A, Lord C. Social Communication Questionnaire. Los Angeles, CA: Western Psychological Services; 2003. 15. Gilliam JE. Gilliam Autism Rating Scale. 3rd ed. Austin, TX: PRO-ED; 2013. 16. American Academy of Pediatrics. Management of children with autism spectrum disorder. Pediatrics. 2007;120(5):1183–1215. Available at

http://pediatrics.aappublications.org/content/120/5/1162.full, Accessed 01.03.14. 17. Center for Disease Control. Autism: Data and Statistics. Available at http://www.cdc.gov/ncbddd/autism/data.html, Accessed 01.03.14. 18. Dykens EM, Volkmar FR. Medical conditions associated with autism. In: Cohen DJ, Volkmar FR, eds. Handbook of Autism and Pervasive

Developmental Disorders. 2nd ed. New York, NY: John Wiley & Sons; 1997:388–410. 19. Evans PA, Morris MA. A Clinical Guide to Autistic Spectrum Disorders. New York, NY: Lippincott Williams & Wilkins; 2011. 20. Gillberg IC, Gillberg C, Ahlsen G. Autistic behaviour and attention deficits in tuberous sclerosis: a population-based study. Dev Med Child

Neurol. 1994;36:50–56, PMID:8132114. 21. Piven J, Gayle J, Landa R, Wzorek M, Folstein S. The prevalence of Fragile X in a sample of autistic individuals diagnosed using a

standardized interview. J Am Acad Child Adolesc Psychiatry. 1991;30:825–830, PMID:1938801. 22. Cook Jr. EH, Lindgren V, Leventhal BL, et al. Autism or atypical autism in maternally but not paternally derived proximal 15q duplication.

Am J Hum Genet. 1997;60:928–934, PMID:9106540. 23. Schroer RJ, Phelan MC, Michaelis RC, et al. Autism and maternally derived aberrations of chromosome 15q. Am J Med Genet. 1998;76:

327–336, PMID:9545097. 24. Weidmer–Mikhail E, Sheldon S, Ghaziuddin M. Chromosomes in autism and related pervasive developmental disorders: a cytogenetic

study. J Intellect Disabil Res. 1998;42:8–12, PMID:9534109. 25. Steffenburg S, Gillberg CL, Steffenburg U, Kyllerman M. Autism in Angelman syndrome: a population-based study. Pediatr Neurol.

1996;14:131–136 PMID:8703225. 26. Stone WL, Ousley OY, Yoder PJ, Hogan KL, Hepburn SL. Nonverbal communication in two-and three-year-old children with autism.

J Autism Dev Disord. 1997;27:677–696, PMID:9455728. 27. Rapin I. Preschool Children with Inadequate Communication: Developmental Language Disorder, ASD, Low IQ. London, UK: MacKeith Press;

1996. 28. Tuchman RF, Rapin I, Shinnar S. Autistic and dysphasic children. II Epilepsy. Pediatrics. 1991;88:1219–1225, PMID:1956740. 29. Greenspan SI, Wieger S. Engaging AutismASD: The Floortime Approach to Helping Children Relate, Communicate, and Think. Reading, MA:

Perseus Book; 2005. 30. Myers S, Johnson CPAmerican Academy of Pediatrics Council on Children with Disabilities. Management of children with autism

spectrum disorders. Pediatrics. 2007;120(5):1162–1182, PMID:17967921. 31. Rossi PG, Parmeggiani A, Bach V, Santucci M, Visconti P. EEG features and epilepsy in patients with autism. Brain Dev. 1995;17:169–174

PMID:7573755. 32. Volkmar FR, Nelson DS. Seizure disorders in autism. J Am Acad Child Adolesc Psychiatry. 1990;29:127–129, PMID:2295565. 33. Wong V. Epilepsy in children with autistic spectrum disorder. J Child Neurol. 1993;8:316–322, PMID:7693796. 34. Tuchman RF, Rapin I. Regression in pervasive developmental disorders: seizures and epileptiform electroencephalogram correlates.

Pediatrics. 1997;99:560–566, PMID:9093299. 35. Nass R, Gross A, Devinsky O. Autism and autistic epileptiform regression with occipital spikes. Dev Med Child Neurol. 1998;40:453–458,

PMID:9698058. 36. Cauda F, Costa T, Palermo S, et al. Concordance of white matter and gray matter abnormalities in autism spectrum disorders: a voxel-

based meta-analysis study. Hum Brain Mapp. 2013. doi:10.1002/hbm.22313 PMID:23894001.

Page 9: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

II. NEUROLOGIC DISEASES

REfEREnCES 205

37. Filipek PA. Neuroimaging in the developmental disorders: the state of the science. J Child Psychol Psychiatry. 1999;40:113–128, PMID:10102728.

38. National Research Council, committee on Education interventions for Children with Autism. In: Lord C, McGee JP, eds. Educating Children with Autism. Washington, DC: National Academies Press; 2001.

39. Goldstein H. Communication intervention for children with autism: a review of treatment efficacy. J Autism Dev Disord. 2002;32:373–396, PMID:12463516.

40. Ventola PE, Oosting D, Anderson LC, Pelphrey KA. Brain mechanisms of plasticity in response to treatments for core deficits in autism. Prog Brain Res. 2013;207:255–272. doi:10.1016/B978-0-444-63327-9.00007-2.

41. Paul R, Sutherland D. Enhancing early language in children with autism spectrum disorders. In: Volkmar FR, Paul R, Klin A, Cohen D, eds. Handbook of Autism and Pervasive Developmental Disorders; Vol II 3rd ed. Hoboken, NJ: John Wiley & Sons; 2005:946–976.

42. Solomon R, Necheles J, Ferch C, Ruckman D. Pilot study of a parent training program for young children with autism. Autism. 2007;11(3):205–224, PMID:17478575.

43. American Speech-Language-Hearing Association, Ad Hoc Committee on Autism Spectrum Disorders. Principles for Speech-Language Pathologists in Diagnosis, Assessment, and Treatment of Autism Spectrum Disorders Across the Life Span. Available at http://www.asha.org/public/speech/disorders/Autism.htm; 2005. Accessed 01.03.14.

44. DeThorn LS, Johnson CJ, Walder L, Mahurin-Smith J. When “Simon Says” doesn’t work: alternatives to imitation for facilitating early speech development. Am J Speech Lang Pathol. 2009;18(2):133–145, PMID:18930909.

45. Petrus C, Adamson SR, Block L, Einarson SJ, Sharifenjad M, Harris SR. Effects of exercise interventions on stereotypic behaviors in children with autism spectrum disorder. Physiother Can. 2008;60(2):134–145, PMID:20145777.

46. Chen YW, Cordier R, Brown NA. Preliminary study on the reliability and validity of using experience sampling method in children with autism spectrum disorders. Dev Neurorehabil. 2013 Dec 4, [Epub ahead of print] PMID:24304202.

47. Wauang Y, Wang C, Huang M, Su CY. The effectiveness of simulated developmental horse-riding. Adopt Phys Active Q. 2010;27(2):113–126, PMID:20440.

48. Weiss JA, Viecili MA, Sloman L, Lunsky Y. Direct and indirect psychosocial outcomes for children with autism spectrum disorder and their parents following a parent-involved social skills group intervention. J Can Acad Child Adolesc Psychiatry. 2013;22(4):303–309, PMID:24223050.

49. Wang M, Reid D. Using the virtual reality-cognitive rehabilitation approach to improve contextual processing in children with autism. Scientific World Journal. 2013;2013(Nov 13):716890. doi:10.1155/2013/716890.

50. Mineo BA, Ziegler W, Gill S, Salkin D. Engagement with electronic screen media among students with autism spectrum disorders. J Autism Dev Disord. 2009;39(1):172–187, PMID:18626761.

51. Volkmar FR. Pharmacological interventions in autism: theoretical and practical issues. J Clin Child Psychol. 2001;30(1):80–87, PMID:11294081.

52. Aman MG, Farmer CA, Hollway J, Arnold LE. Treatment of inattention, overactivity, and impulsiveness in autism spectrum disorders. Child Adolesc Psychiatr Clin N Am. 2008;17(4):713–738, vii. PMID: 18775366.

53. Stigler KA, McDougle CJ. Pharmacotherapy of irritability in pervasive developmental disorders. Child Adolesc Psychiatr Clin N Am. 2008;17(4):739–752, vii–viii. PMID:18775367.

54. Sadock BJ, Sadock VA, Ruiz P. Biological Therapies. Kaplan and Sadock’s Comprehensive Textbook of Psychiatry. 9th ed. New York, NY: Lippincott, Williams & Wilkins; 2009, SBN/ISSN: 9780781768993.

55. Campbell M, Anderson LT, Meier M. A comparison of haloperidol, behavior therapy, and their interaction in autistic children [proceedings]. Psychopharmacol Bull. 1979 Apr;15(2):84–86, PMID:373009.

56. Cohen IL, Campbell M, Posner D, Small AM, Triebel D, Anderson LT. Behavioral effects of haloperidol in young autistic children. An objective analysis using a within-subjects reversal design. J Am Acad Child Psychiatry. 1980;19(4):665–677, PMID:7204797.

57. Campbell M, Armenteros JL, Malone RP, et al. Neuroleptic-related dyskinesias in autistic children: a prospective, longitudinal study. J Am Acad Child Adolesc Psychiatry. 1997;36(6):835–843.

58. Pierre JM. Extrapyramidal symptoms with atypical antipsychotics: incidence, prevention and management. Drug Saf. 2005;28(3):191–208, PMID:15733025.

59. Ho J, Panagiotopoulos C, McCrindle B, Grisaru S, Pringsheim T. Management recommendations for metabolic complications associated with second-generation antipsychotic use in children and youth. Paediatr Child Health. 2011;16(9):575–580, PMCID: PMC3223901.

60. Scahill L. How do I decide whether or not to use medication for my child with autism? Should I try behavior therapy first? J Autism Dev Disord. 2008;38(6):1197–1198. doi:10.1007/s10803-008-0573-7, PMID 18463973.

61. Stigler KA, Posey DJ, McDougle CJ. Aripiprazole for maladaptive behavior in pervasive developmental disorders. J Child Adolesc Psychopharmacol. 2004;14(3):455–463, PMID:15650503.

62. Valicenti-McDermott MR, Demb H. Clinical effects and adverse reactions of off-label use of aripiprazole in children and adolescents with developmental disabilities. J Child Adolesc Psychopharmacol. 2006;16(5):549–560, PMID: 17069544.

63. Malone RP, Delaney MA, Hyman SB, Cater JR. Ziprasidone in adolescents with autism: an open-label pilot study. J Child Adolesc Psychopharmacol. 2007;17(6):779–790, PMID:18315450.

64. Reichow B, Volkmar FR, Bloch MH. Systematic review and meta-analysis of pharmacological treatment of the symptoms of attention-deficit/hyperactivity disorder in children with pervasive developmental disorders. J Autism Dev Disord. 2013;43(10):2435–2441, PMID:23468071.

65. Riddle EL, Fleckenstein AE, Hanson GR. Role of monoamine transporters in mediating psychostimulant effects. AAPS J. 2005;7(4):E847–E851. doi:10.1208/aapsj070481, PMC 2750953. PMID 16594636.

66. Gibbs TT. Pharmacological treatment of autism. In: Blatt GJ, Gibbs TT, eds. The Neurochemical Basis of Autism. 2010:245–267. doi:10.1007/978-1-4419-1272-5_15.

67. Arnold LE, Aman MG, Cook AM, et al. Atomoxetine for hyperactivity in autism spectrum disorders: placebo-controlled crossover pilot trial. J Am Acad Child Adolesc Psychiatry. 2006;45(10):1196–1205, PMID:17003665.

Page 10: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

206 18. AuTISm SPECTRum DISoRDERS: ClInICAl ConSIDERATIonS

II. NEUROLOGIC DISEASES

68. Lile JA, Nader MA. The abuse liability and therapeutic potential of drugs evaluated for cocaine addiction as predicted by animal models. Curr Neuropharmacol. 2003;1:21–46. doi:10.2174/1570159033360566.

69. Jaselskis CA, Cook Jr. EH, Fletcher KE, Leventhal BL. Clonidine treatment of hyperactive and impulsive children with autistic disorder. J Clin Psychopharmacol. 1992;12(5):322–327, PMID: 1479049.

70. Posey DJ, Posey DJ, Puntney JI, Sasher TM, Kem DL, McDougle CJ. Guanfacine treatment of hyperactivity and inattention in pervasive developmental disorders: a retrospective analysis of 80 cases. J Child Adolesc Psychopharmacol. 2004;14(2):233–241, PMID:15319020.

71. Pliszka SR, Crismon ML, Hughes CW, et al., Texas Consensus Conference Panel on Pharmacotherapy of Childhood Attention Deficit Hyperactivity Disorder. The Texas Children’s Medication Algorithm Project: revision of the algorithm for pharmacotherapy of attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2006;45:642–657, PMID:16721314.

72. Scahill L, Chappell PB, Kim YS, et al. A placebo-controlled study of guanfacine in the treatment of children with tic disorders and attention deficit hyperactivity disorder. Am J Psychiatry. 2001;158:1067–1074, PMID:11431228.

73. Wilens TE, Biederman J, Spencer T. Clonidine for sleep disturbances associated with attention-deficit hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 1994;33:424–426, PMID:8169189.

74. Uhlen S, Wikberg JE. Delineation of rat kidney alpha 2A- and alpha 2B-adrenoceptors with [3H] RX821002 radioligand binding: computer modeling reveals that guanfacine is an alpha 2A-selective compound. Eur J Pharmacol. 1991;202:235–243, PMID:1666366.

75. Kugler J, Seus R, Krauskopf R, Brecht HM, Raschig A. Differences in psychic performance with guanfacine and clonidine in normotensive subjects. Br J Clin Pharmacol. 1980;10(suppl 1):71S–80S, PMID:6994783.

76. Daviss WB, Patel NC, Robb AS, et al., the CAT STUDY TEAM. Clonidine for attention-deficit/hyperactivity disorder: II. ECG changes and adverse events analysis. J Am Acad Child Adolesc Psychiatry. 2008;47:189–198.

77. Biederman J, Melmed RD, Patel A, et al., for the SPD503 Study Group. A randomized, double-blind, placebo-controlled study of guanfacine extended release in children and adolescents with attention-deficit/hyperactivity disorder. Pediatrics. 2008;121:e73–e84.

78. Easton N, Shah YB, Marshall FH, Fone KC, Marsden CA. Guanfacine produces differential effects in frontal cortex compared with striatum: assessed by phMRI BOLD contrast. Psychopharmacology (Berl). 2006;189:369–385, PMID:17016709.

79. Arnsten AFT, Dudley AG. Methylphenidate improves prefrontal cortical cognitive function through alpha2 adrenoceptor and dopamine D1 receptor actions: relevance to therapeutic effects in attention deficit hyperactivity disorder. Behav Brain Funct. 2005;1:2, PMID: 15916700.

80. Ming X, Gordon E, Kang N, Wagner GC. Use of clonidine in children with autism spectrum disorders. Brain Dev. 2008;30:454–460, PMID:18280681.

81. Reichow B, Volkmar FR, Bloch MH. Systematic review and meta-analysis of pharmacological treatment of the symptoms of attention-deficit/hyperactivity disorder in children with pervasive developmental disorders. J Autism Dev Disord. 2013;43(10):2435–2441, PMID:23468071.

82. Edwards DJ, Chugani DC, Chugani HT, Chehab J, Malian M, Aranda JV. Pharmacokinetics of buspirone in autistic children. J Clin Pharmacol. 2006;46(5):508–514, PMID:16638734.

83. Soorya L, Kiarashi J, Hollander E. Psychopharmacologic interventions for repetitive behaviors in autism spectrum disorders. Child Adolesc Psychiatr Clin N Am. 2008;17(4):753–771. doi:10.1016/j.chc.2008.06.003 viii.

84. Oswald DP, Sonenklar NA. Medication use among children with autism spectrum disorders. J Child Adolesc Psychopharmacol. Jun 2007;17(3):348–355, PMID:17630868.

85. McPheeters ML, Warren Z, Sathe N, et al. A systematic review of medical treatments for children with autism spectrum disorders. Pediatrics. 2011;127(5):e1312–e1321, PMID:21464191.

86. Blier P. Pharmacology of rapid-onset antidepressant treatment strategies. J Clin Psychiatry. 2001;62(suppl 15):12–17. 87. Hollander E, Phillips A, Chaplin W, et al. A placebo controlled crossover trial of liquid fluoxetine on repetitive behaviors in childhood and

adolescent autism. Neuropsychopharmacology. 2005;30(3):582–589, PMID: 15602505. 88. Owley T, Walton L, Salt J, et al. An open-label trial of escitalopram in pervasive developmental disorders. J Am Acad Child Adolesc

Psychiatry. 2005;44(4):343–348, PMID:15782081. 89. Namerow LB, Thomas P, Bostic JQ, Prince J, Monuteaux MC. Use of citalopram in pervasive developmental disorders. J Dev Behav Pediatr.

2003;24(2):104–108, PMID:12692455. 90. Couturier JL, Nicolson R. A retrospective assessment of citalopram in children and adolescents with pervasive developmental disorders.

J Child Adolesc Psychopharmacol. Fall 2002;12(3):243–248, PMID:12427298. 91. Boyce RD, Handler SM, Karp JF, Hanlon JT. Age-related changes in antidepressant pharmacokinetics and potential drug-drug interactions:

a comparison of evidence-based literature and package insert information. Am J Geriatr Pharmacother. 2012;10(2):139–150, PMCID: PMC3384538.

92. US Food and Drug Administration. Celexa (citalopram hydrobromide): Drug Safety Communication – Abnormal Heart Rhythms Associated with High Doses. Available at http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm269481.htm, Accessed 24.10.12.

93. Hollander E, et al. An open trial of divalproex sodium in autism spectrum disorders. J Clin Psychiatry. 2001;62(7):530–534, PMID: 11488363. 94. Rugino TA, Samsock TC. Levetiracetam in autistic children: an open-label study. J Dev Behav Pediatr. 2002;23(4):225–230, PMID:12177568. 95. Douglas JF, Sanders KB, Benneyworth MH, et al. Brief report: retrospective case series of oxcarbazepine for irritability/agitation symptoms

in autism spectrum disorder. J Autism Dev Disord. 2013;43(5):1243–1247. doi:10.1007/s10803-012-1661-2. 96. Uvebrant P, Bauziene R. Intractable epilepsy in children. The efficacy of lamotrigine treatment, including non-seizure-related benefits.

Neuropediatrics. 1994;25(6):284–289. 97. Hosenbocus S, Chahal R. Amantadine: a review of use in child and adolescent psychiatry. J Can Acad Child Adolesc Psychiatry.

2013;22(1):55–60, PMID:23390434. 98. Hardan AY, Handen BL. A retrospective open trial of adjunctive donepezil in children and adolescents with autistic disorder. J Child Adolesc

Psychopharmacol. 2002;12(3):237–241, PMID:12427297. 99. Nicolson R, Craven-Thuss B, Smith J. A prospective, open-label trial of galantamine in autistic disorder. J Child Adolesc Psychopharmacol.

2006;16(5):621–629, PMID:17069550.

Page 11: Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease || Autism Spectrum Disorders

II. NEUROLOGIC DISEASES

REfEREnCES 207

100. Chez MG, et al. Treating autistic spectrum disorders in children: utility of the cholinesterase inhibitor rivastigmine tartrate. J Child Neurol. 2004;19(3):165–169, PMID: 15119476.

101. Krishnaswami S, McPheeters ML, Veenstra-Vanderweele J. A systematic review of secretin for children with autism spectrum disorders. Pediatrics. 2011;127(5):e1322–e1325. doi:10.1542/peds.2011-0428.

102. Dosman C, et al. Complementary, holistic, and integrative medicine: autism spectrum disorder and gluten- and casein-free diet. Pediatr Rev. 2013;34(10):e36–e41, PMID: 24085796.

103. Rossignol DA, Rossignol LW, James SJ, Melnyk S, Mumper E. The effects of hyperbaric oxygen therapy on oxidative stress, inflammation, and symptoms in children with autism: an open-label pilot study. BMC Pediatr. 2007;7(Nov 16):36, PMID:18005455.

104. Stahl SM. Stahl's Essential Psychopharmacology: Neuroscientific Basis and Practical Applications. Cambridge University Press; 2008. 105. Palermo M, Curatolo P. Pharmacologic treatment of autism. J Child Neurol. 2004;19:155–164, PMID:15119475. 106. Masi G. Pharmacotherapy of pervasive developmental disorders in children and adolescents. CNS Drugs. 2004;18:1031–1052, PMID:

15584771.