Autism Spectrum Disorders
Autism Spectrum Disorders
Autism Spectrum Disorders
Disorders
edited by
Eric Hollander
Mount Sinai School of Medicine
New York, New York, U.S.A.
MARCEL
ISBN: 0-8247-0715-X
Headquarters
Marcel Dekker, Inc., 270 Madison Avenue, New York, NY 10016, U.S.A.
tel: 212-696-9000; fax: 212-685-4540
The publisher offers discounts on this book when ordered in bulk quantities. For more
information, write to Special Sales/Professional Marketing at the headquarters address
above.
Neither this book nor any part may be reproduced or transmitted in any form or by any
means, electronic or mechanical, including photocopying, microfilming, and recording,
or by any information storage and retrieval system, without permission in writing from
the publisher.
10 9 8 7 6 5 4 3 2 1
Advisory Board
It is intriguing that conditions which clearly have always existed may nonetheless
only be recognized and accorded significance relatively late. This is the case with
autism. It was only in the 1940s that the first clinical descriptions were provided
by Kanner and Asperger (these were published almost simultaneously, although
both researchers seem to have been unaware of the other’s work). And yet it is
certain that severe, classical cases of autism occurred long before this. Uta Frith
and Rab Houston, in their recent book Autism in History, present the most detailed
evidence for such a diagnosis in the case of a young man living in the 18th
century, and in an earlier book, Frith hazarded the thought that autism might
explain the otherwise almost unintelligible behavior of the Franciscan monk
Brother Juniper, of the ‘‘blessed fools’’ of old Russia, and so on.
While Asperger regarded autism as organic, a biological defect of affective
contact, Kanner saw it as psychological, or reactive, a reaction to a supposed
lack of parental love or care, and, in particular, to a cold, “refrigerator mother.”
Asperger’s paper did not become widely known (it was only translated into En-
glish in the 1980s), and Kanner’s unfortunate formulation was rather widely ac-
cepted during the 1950s and 1960s, with sometimes dire consequences to both
autistic children and their parents. (Kanner’s psychological theory is still accepted
in many parts of the world.) But by the late 1960s it had become apparent that
autism was organic, although not a “disease” in the usual sense—not specifically
a disorder of childhood, but rather a disorder of development, the development
of certain parts of the mind and brain, and, as such, a lifelong condition.
Up to this point approaches had been purely clinical and epidemiological—
what was the incidence of autism? What were its causes? How did autism present
itself, evolve, change with time? What could be done with drugs, or behavioral
iii
iv A Historical Perspective
approaches, to mitigate or control the symptoms, and allow the autistic person
a more normal life?
There was little sense of the deep psychological structure of autism, much
less of its neurobiological basis. Attempts to delineate a deep, invariable core in
the syndrome started in the 1970s, and soon homed in on the social and communi-
cational problems of the autistic, and these were seen by some in the 1980s as
expressions of a very belated, slow, and at best very partial development of what
primate ethologists were now terming “theory of mind.” And yet, it was clear,
no such theory sufficed by itself: there were nearly always sensory symptoms,
strange sensitivities and insensitivities, never the same in any two individuals
(Temple Grandin writes of these, from her own experience, in the present vol-
ume). There were repetitive and compulsive behaviors; there could be impulsiv-
ity, explosiveness, aggressiveness; there were sometimes specific language disor-
ders. Sometimes there were seizures or EEG abnormalities.
And a large proportion of the autistic—at least, those with classical infan-
tile autism such as Kanner had described—were markedly retarded; language
and intelligence, by contrast, seemed normal in those with Asperger’s syndrome.
All this complicated matters. Was one dealing with a single core disorder, or
with a complex phenotype which could include much else? Could some autistic
traits appear in relatives of the autistic, even though one would never see them
as clinically autistic? What was the relation of Kanner-type autism to Asperger’s
syndrome? What of pervasive developmental disorders, so-called, or Rett’s syn-
drome, chilhood disintegrative disorder, etc.? Should one not speak of an entire
spectrum of such disorders?
(And the relationship of savant syndrome to autism, while strong—at least
10% of those with classical infantile autism exhibit savant traits—remains myste-
rious, so much so that it has become widely regarded as a syndrome unto itself,
and as such is not treated in this volume. While Langdon Down coined the term
“idiot savant” in the 19th century, and a most detailed analysis of such a patient
was provided by Kurt Goldstein a few years before the writings of Kanner and
Asperger, we still know very little about the neurobiology of savant talents, nor
why they should so often be associated with autism.)
The need for sorting out these varied but related conditions—all of which
can have different presentations and prognoses, different responses to drugs and
other treatments, and different neurobiological bases—became pressing by the
late 1980s, and it is especially such a sorting-out, such a dimensional approach,
which has so fruitfully occupied Eric Hollander and his colleagues, and which is
set out here, systematically and comprehensively, in Autism Spectrum Disorders.
The organization of this book begins with the initial clinical encounter, the
making of a diagnosis—not perhaps intellectually difficult, but a diagnosis which
is emotionally and morally charged, for what one is diagnosing is a lifelong
condition which will have to be lived with, negotiated with, for decades ahead.
A Historical Perspective v
vii
viii Preface
ACKNOWLEDGMENTS
I would like to thank:
Beth, Evan, and Zachary for their love and support
Hirschell and Deanna Levine and the Seaver Foundation for their support
and championing the needs of autism research
Kenneth L. Davis, M.D., Dean, Mount Sinai School of Medicine, for scien-
tific guidance
Eric Hollander
Contents
Assessment
Psychopharmacological Treatments
Index 423
Contributors
David J. Posey, M.D. Assistant Professor of Psychiatry, Riley Child and Ado-
lescent Psychiatry Clinic, Department of Psychiatry, Indiana University School
of Medicine, Indianapolis, Indiana, U.S.A.
Ronald R. Rawitt, M.D. Seaver Autism Research Center, Mount Sinai School
of Medicine, New York, New York, U.S.A.
Roberto Tuchman, M.D. Dan Marino Center, Miami Children’s Hospital, Mi-
ami, Florida, U.S.A.
validity of autism as a diagnosis (2). For example, in his original report Kanner
mentioned that the parents of the majority of his cases were remarkably successful
academically or professionally. This led some clinicians to believe that parental
psychopathology or some aspects of early experience might have a role in syn-
drome pathogenesis, a notion incompatible with the idea that children were born
with autism. However, the question of the role of experience in pathogenesis
plagued the field for many years and was resolved only as it became clear that
when one controls for ascertainment bias there is no particular social class pre-
dominance in autism (3). Second, it became clear that children with autism have
trouble interacting with all people—not just their parents—and that problems in
parent–child interaction may arise from the side of the child and not the par-
ents (4).
Kanner initially speculated that autism was not related to other medical
conditions, but subsequent research has shown that various conditions are associ-
ated with autism, including fragile X syndrome, tuberous sclerosis, and seizure
disorders (5). An additional source of confusion arose because of Kanner’s origi-
nal speculation that children with autism were not mentally retarded. This impres-
sion reflected his observation that these children looked intelligent and that on
some parts of IQ testing they did rather well. It took several decades to realize
that poor performance on other aspects of tests of intelligence was quite real and
did not simply reflect lack of motivation or child cooperation, and that a majority
of children with autism do exhibit some degree of mental handicap (6).
Probably the main source of dispute involved the confusion of autism with
childhood schizophrenia. The speculation that autism was the earliest form of
schizophrenia was quite common in the late 1940s and 1950s (2), and compli-
cated psychological phenomena such as hallucinations and delusions were attrib-
uted to mute, autistic children. Subsequent research has confirmed that autism is
not the earliest manifestation of schizophrenia (7).
features such as overanxiety and activity that may be important targets for treat-
ment?
In 1978 Rutter (8) synthesized Kanner’s original report and subsequent
research in what later became a highly influential definition of autism. This defi-
nition included four essential features: early onset; impaired social development
(not due to mental retardation); impaired communicative development (not due
to mental retardation); and unusual behaviors of the type Kanner had described,
including, for example, resistance to change, idiosyncratic responses to the envi-
ronment, and motor mannerisms. These criteria largely comprised the basis for
the definition of autism in the third edition of the Diagnostic and Statistical Man-
ual of Mental Disorders (DSM-III), in which it was included, for the first time,
as part of a new class of disorders—the pervasive developmental disorders (9).
Although the inclusion of autism in DSM-III was a major advance, the definition
proposed was not developmentally oriented and was overly stringent in some
respects. As a result, major changes were made in the revised third edition (DSM-
III-R) (10), in which the definition employed was much more developmentally
oriented but also somewhat broader than that used previously (11). Also, the
changes made increased the differences between the DSM definition and that in
the pending revision of the international diagnostic system (ICD-10). In addition
to the differences between the DSM definition and that in the definition of autism,
another major difference from ICD-10 was that the latter included a number of
new conditions within the PDD class, e.g., Asperger’s disorder, Rett’s disorder,
childhood disintegrative disorder, and atypical autism. These, and other consider-
ations, were encompassed in DSM-IV (12).
For DSM-IV, a series of literature reviews and data reanalyses were used
in an attempt to identify areas of consensus as well as controversy. A multisite,
international field trial (13) was then undertaken that included nearly 1000 cases
evaluated by over 100 raters around the world. The results suggested that a defi-
nition convergent with that used in ICD-10 had a good balance of sensitivity
and specificity. Furthermore, the data available did provide some support for the
inclusion of other conditions in the PDD class in DSM-IV. The DSM-IV defini-
tion requires the presence of at least six criteria, including at least two criteria
relating to social abnormalities (group 1) and one each relating to impaired com-
munication (group 2) and range of interests and activities (group 3) with onset
of the condition before age 3. The qualitative impairment in social interaction
can take the form of markedly impaired nonverbal behaviors, failure to develop
appropriate peer relationships, lack of shared enjoyment or pleasure, or lack of
social-emotional reciprocity. Impairments in communication can include delay
or lack of spoken language, impairment in conversational ability, stereotyped
language use, and deficits in imaginative play with no attempt to compensate for
the language problems through other means. The final group of features includes
4 Volkmar et al.
EPIDEMIOLOGY
Epidemiological studies provide information regarding the prevalence of autistic
disorder and related conditions. Such studies also provide insight into aspects of
syndrome pathogenesis and natural history and are important in clarifying needs
for intervention. Well-designed, large, epidemiological studies are crucial for an-
swering the most pressing question posed in the last two decades regarding these
disorders: is the incidence of autism and pervasive developmental disorders in-
creasing? Reports of increased prevalence of these disorders in some surveys and
reports of clusters of affected children within particular geographical regions have
alarmed parents and policymakers alike. These reports have had two specific
effects: they have heightened awareness of the disorders that may impact the
Diagnosis and Epidemiology 5
delivery of clinical services and research funding, and they have established the
need for carefully done studies concerning the prevalence of autism and related
conditions.
These issues underscore the need to use standardized, rigorous procedures.
The most accurate prevalence rates are obtained when a large area is sampled,
as opposed to a clinically referred population (20). The case-finding method
should be initially overly inclusive, to guard against cases being missed. Finer
discrimination of diagnostic categories can be accomplished after an initial
screening has identified all possible cases; at this stage a critical issue is the finer-
grained distinctions between autism and related disorders, particularly since it is
clear that such distinctions are, of necessity, somewhat arbitrarily drawn (21).
Kanner’s historical description of autism was that of a behavioral syn-
drome, and provided the most narrow syndrome definition (1). In contrast, Wing
and Gould’s “triad of impairments” encompassed a much larger group (3). The
three editions of the DSM (III, III-R, and IV) provided behavioral descriptions
with varying degrees of specificity. As noted previously, DSM-III-R included
the broadest definition of autism, yielding a 40% false-positive rate for case iden-
tification, which most certainly impacted epidemiological surveys (11). In fact,
prevalence rates are positively correlated with date of publication, suggesting an
influence of syndrome definition on the calculation of rates (20). Thus, historical
changes in the classification system have affected prevalence rates, and perhaps
confounded the estimation of the “true” incidence of autism.
True epidemiological samples offer several advantages over clinically re-
ferred samples. The latter are biased toward those who need services at a particu-
lar time and families aware of the availability of such services. In fact, the use
of clinically referred samples may have contributed to the early notion that autism
was associated with higher socioeconomic class (20). Sample size influences epi-
demiological outcomes as well—smaller sample sizes have yielded greater rates
of the disorder. Confidence intervals reflect the variability within a sample, and
indicate the precision of calculation of rates (22). Epidemiological studies with
extremely wide confidence intervals show predicted rates that are likely quite
imprecise. In addition, an understanding of the way in which age trends affect
rates is needed for interpretation of epidemiological studies (20). Because
changes in the absolute number of a certain age cohort over time will influence
numbers of affected children but not rates, absolute numbers cannot be reliably
compared. Finally, case finding affects the results obtained at different ages; for
example, typically, more school-aged children with autism are identified than
preschoolers, probably an effect of case finding and not reflecting overall changes
in rates.
Lotter (23) provided one of the earliest epidemiological studies of autism,
screening 78,000 children between the ages of 8 and 10 in a borough outside of
London. The large sample size, the use of a prescreening survey, parent inter-
6 Volkmar et al.
grants (29). Review of the methods used to ascertain these samples reveals that
a large proportion of immigrants moving in and out of the surveyed regions may
have skewed the estimation of rates (20). Additionally, the likelihood that fami-
lies with disabled children may emigrate to a more developed country to obtain
better services for their child should be considered.
The identification of geographical clusters of persons affected with autism
has raised suspicion regarding environmental causes of autism (30). However,
the approach taken in reviewing the incidence of autism in these clusters has
not been statistically sound (23). Rates within a geographical cluster cannot be
compared with population rates, simply because the boundaries of the cluster are
predefined in such a way as to select for autism cases. A more reliable method
is to track subsequent cases in a geographical region, or to use statistical tech-
niques that specifically evaluate the reality of the cluster phenomenon while elim-
inating bias in data collection (23). Finally, Fombonne (31) makes the point that
since autism is probably a disorder with strong genetic underpinnings, changes
in prevalence would not occur over as short a timespan as is indicated by some
studies.
COMORBID CONDITIONS
Interest in the issue of comorbidity in autism has increased in recent years. This
is a complicated issue for more general reasons as well as for reasons more spe-
cific to autism. For example, the hierarchical rules employed in official diagnostic
systems may artificially limit comorbid diagnoses, as in the current approach to
comorbid diagnosis of autism and attention-deficit disorder. On the other hand,
if all such rules are discarded, diagnoses tend to become more of a symptom
checklist. For many years it was assumed that if an individual had autism then all
his or her difficulties were a function of the autism, a notion known as “diagnostic
overshadowing” (32,33). It was as if having autism “immunized” the individual
against other disorders rather than, as would more reasonably be expected, serv-
ing as a factor that increased risk.
Autism has been reported to co-occur with various other developmental,
psychiatric, and medical conditions; some such associations are frequent while
others are quite rare. A critical statistical issue is whether such associations are
significantly more frequent than would be related by chance alone; e.g., it appears
that individuals with autism do develop schizophrenia but not at higher rates than
would be expected given the rate of schizophrenia in the general population (34).
When autism occurs at a greater-than-expected rate with another disorder, the
issue arises as to whether the two disorders are related to the same underlying
cause or whether one disorder causes the other (35).
Of the various conditions associated with autism, it is clear that mental
retardation is the most frequently associated one (6). Despite a substantial body
8 Volkmar et al.
of research into the nature of the cognitive deficit(s) involved in autism (36), the
fundamental mechanism remains unclear. Interestingly, genetic research suggests
that mental retardation per se is not one of the features commonly transmitted
in immediate family members (37). In terms of medical conditions, the most
commonly recognized associations include seizure disorders ( in perhaps 20–
25% of cases), fragile X syndrome (in about 1% of cases), and, much less fre-
quently, tuberous sclerosis (5).
Various other behavioral and psychiatric difficulties have been described
in individuals with autism. Unfortunately, much of the evidence in favor of such
associations is based on case reports; the significance of such reports is always
difficult to interpret given the bias for only positive reports to be published and
the lack of more controlled studies. Reported difficulties include hyperactivity,
obsessive-compulsive phenomena, self-injury and stereotypy, tics, and affective
symptoms (38–44). While it is clear that symptoms of these conditions can be
observed, the questions of syndromic interpretation and, more importantly, of
causal relationships are complex. For example, when is a diagnosis of Tourette’s
disorder justified? When do the perseverative and ritualistic behaviors often ob-
served in autism constitute evidence for obsessive-compulsive disorder (OCD)?
In some cases, e.g., stereotyped-movement disorder, a comorbid diagnosis of
autism is prohibited because stereotyped movements are included as a defining
feature of autism. Given the high rates of mutism and major communication
problems in autistic individuals, issues of assessment for related comorbid condi-
tions are often quite complex. Ideally what are needed are independent validators
of comorbid associations, for example, independently collected data on family
history. Family studies have suggested some important associations such as social
difficulties, language and learning problems, anxiety, and affective difficulties in
family members (37).
The issue of comorbidity might also be approached through response to
treatment. For example, it is clearly the case that phenomena reminiscent of at
least some aspects of obsessive-compulsive disorder are frequently observed in
adults with autism (45). These behaviors may respond to medications such as
selective serotonin-reuptake inhibitors (SSRIs) that have been used in more typi-
cal presentations of obsessive-compulsive disorder (44,46). However, response
to medication may be a function of many factors, and some investigators have
questioned the notion that autism and OCD are specifically related and suggested
that the seeming relationship may be a function of many factors. Various disor-
ders may be ameliorated with the same medication (e.g., tricyclic antidepressants
are helpful in the treatment of depression and enuresis, and both hyperactive and
normal children may respond to stimulant medication with improved attention).
In this context, investigators have noted that the ritualistic phenomena of autism
and typical obsessions and compulsions cannot be simply equated (47). Similarly,
there have been various reports of associations of autism with tic disorders, spe-
Diagnosis and Epidemiology 9
DIFFERENTIAL DIAGNOSIS
An alternative way of conceptualizing the PDDs is through a dimensional ap-
proach, which links symptom clusters with neurobiology and treatment (53). The
categorical approach of DSM provides for multiaxial assessment. The axes are
a useful tool for describing associated and underlying conditions, and tracking
changes in the developmental expression of the disorder.
Differential diagnosis of autism should begin with consideration of other
disorders within the PDD class. Other PDDs described in DSM-IV include Rett’s
disorder, childhood disintegrative disorder (Heller’s syndrome), Asperger’s dis-
order, and PDD-NOS.
Rett’s disorder and childhood disintegrative disorder present after a period
of apparently normal development. Fortunately, both conditions are quite rare.
Rett’s disorder, described by Andreas Rett in 1966 (54), is characterized by decel-
eration of head growth, loss of purposeful hand movements with substitution of
hand-washing stereotypies, motor and language impairment, mental retardation,
and loss of social engagement. This disorder has been described primarily in
girls. Childhood disintegrative disorder, originally described by Heller in 1906,
is distinct from autism and often associated with severe mental retardation. It
presents after a minimum of 2 years of normal development and affects children
in multiple areas of functioning, including deterioration in two of the following
five areas—language functioning, social skills, toileting, play, and motor skills—
as well as abnormal functioning in two of the three symptom clusters of autism.
Underlying neuropathological processes may be identified in some cases (55).
Asperger’s disorder was initially described in 1944 (56). It differs from
autism in that language and cognition are generally preserved, and unusual behav-
10 Volkmar et al.
iors and unusual environmental responsiveness are less likely to be present. Social
behavior is characterized by a desire for social relationships coupled with an
impaired ability to participate in a reciprocal fashion. Circumscribed interests,
about which the child may speak in a pedantic way, are frequently seen. Motor
deficits may be prominent, and, unlike the characteristic profile in autism, verbal
IQ may be significantly higher than performance IQ. Asperger’s disorder is gener-
ally diagnosed later than autism.
PDD-NOS is the diagnosis reserved for cases that do not meet the full
criteria for autism disorder. The majority of children diagnosed with a PDD carry
this diagnosis, which in DSM-IV was broadened so that instead of having deficits
in social interaction and deficits in communication or restricted interests, one
need only have difficulties in any one of the three spheres.
In addition to the various other PDDs, other conditions sometimes present
in ways suggestive of autism. For children with hearing deficits, disability in
spheres other than communication should be relatively preserved. Other disorders
in which presentation may mimic aspects of autism include untreated seizure
disorders, developmental language disorder, stereotyped movement disorder, and
psychotic disorders. Severe or profound mental retardation may involve stereoty-
pies and may resemble autism except that social and communicative functioning
is intact relative to the individual’s overall level of functioning. Other disorders
to be considered in the differential diagnosis of autism include selective mutism,
OCD, reactive attachment disorder, social phobia, and schizoid and avoidant per-
sonality disorders (57).
Associated medical conditions may, in an apparent minority of cases (esti-
mated between 10% and 25%) underlie autistic-like presentations (58,59). Spe-
cifically, fragile X syndrome and tuberous sclerosis are seen with greater-than-
chance frequency in the autistic population. Associations with congenital rubella,
cerebral palsy, phenylketonuria, neurofibromatosis, and Down’s syndrome ap-
pear to occur at chance rates in individuals with autism (58).
CONCLUSIONS
In summary, Leo Kanner’s description of autism has remained the fundamental
basis of current definitions. Categorical definitions, while useful, have impacted
epidemiological estimates and research strategies. Further work is required in
delineating the diagnoses of Asperger’s disorder, Rett’s disorder, and childhood
disintegrative disorder. Perhaps most critical for exploring neurobiological mech-
anisms, the diagnosis of PDD-NOS will benefit from further refinement: dimen-
sional definitions may prove to be the best strategy for elucidating this broad
diagnosis. Given that dimensional definitions will be most useful in linking the
neurobiology of the disorders to specific symptom clusters, greater refinement
of instruments measuring various dimensions of symptomatology is needed.
Diagnosis and Epidemiology 11
The available evidence does not support the view that the prevalence of
autism, or autism spectrum conditions, is increasing. Studies that report higher
prevalence rates consistently demonstrate methodological flaws in ascertainment
and sampling, and widely varying syndrome definitions. Most studies confirm a
sex ratio of 4:1, although some have suggested that girls may be underrepresented
for a variety of reasons. No conclusive evidence supports the contention that
children with autism are likely to come from the higher socioeconomic classes,
and, similarly, no evidence exists to support the view that immigrants have higher
rates of the disorder than nonimmigrants.
Investigation of comorbid conditions in PDDs is critical for clarifying neu-
robiological mechanisms and designing appropriate intervention. Family genetic
studies are a useful methodology for identifying the presence of comorbid condi-
tions. Data supporting whether associations arise by chance or at a greater-than-
chance rate are critical as well. The current nosology will be enhanced by greater
certainty regarding associated conditions combined with more precise conceptu-
alization of these conditions.
ACKNOWLEDGMENTS
This work was supported by National Institute of Child Health and Development
grant 1 P01 HD35482-01 and Yale Children’s Clinical Research Center grant
M01 RR06022, the General Clinical Research Centers Program, the National
Center for Research Resources and, the National Institutes of Health.
REFERENCES
1. Kanner L. Autistic disturbances of affective contact. Nerv Child 1943; 2:217–250.
2. Rutter M. Diagnostic validity in child psychiatry. Adv Biol Psychiatry 1978; 2:2–22.
3. Wing L, Gould J. Severe impairments of social interactions and associated abnor-
malities in children: epidemiology and classification. J Autism Dev Disord 1979;
9(1):11–29.
4. Volkmar FR, Carter A, Grossman J, Klin A. Social development in autism. In:
Cohen DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental
Disorders. 2nd ed. New York: Wiley, 1997:173–194.
5. Dykens EM, Volkmar FR. Medical conditions associated with autism. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:388–410.
6. Sparrow S. Developmentally based assessments. In: Cohen DJ, Volkmar FR, eds.
Handbook of Autism and Pervasive Developmental Disorders. 2nd ed. New York:
Wiley, 1997:411–447.
7. Volkmar FR, Klin A, Cohen DJ. Diagnosis and classification of autism and related
conditions: consensus and issues. In: Cohen DJ, Volkmar, FR, eds. Handbook of Au-
tism and Pervasive Developmental Disorders. 2nd ed. New York: Wiley, 1997:5–40.
12 Volkmar et al.
26. Treffert DA. Epidemiology of infantile autism. Arch Gen Psychiatry 1970; 22:431–
438.
27. Steinhausen H-C, Gobel D, Breinlinger M, Wohlloben B. A community survey of
infantile autism. J Am Acad Child Psychiatry 1986; 25:186–189.
28. Bryson SE, Clark BS, Smith IM. First report of a Canadian epidemiological study
of autistic syndromes. J Child Psychol Psychiatry 1988; 4:433–445.
29. Wing L. The definition and prevalence of autism: a review. Eur Child Adolesc
Psychiatry 1993; 2:61–74.
30. Baron-Cohen S, Saunders K, Chakrabarti S. Does autism cluster geographically?
A research note. Autism 1999; 3:39–43.
31. Fombonne E. Is the prevalence of autism increasing? J Autism Dev Disord 1996;
6:673–676.
32. Reiss S, Levitan GW, Szyszko J. Emotional disturbance and mental retardation:
Diagnostic overshadowing. Am J Ment Retard 1982; 86:567–574.
33. White MJ, Nichols CN, Cook RS, Spengler PM, Walker BS, Look KK. Diagnostic
overshadowing and mental retardation: a meta-analysis. Am J Ment Retard 1995;
100:293–298.
34. Volkmar FR, Cohen DJ. Comorbid association of autism and schizophrenia. Am J
Child Psychiatry 1991; 148(12):1705–1707.
35. Tsai LY. Brief report: comorbid psychiatric disorders of autistic disorder. J Autism
Dev Disord 1996; 26(2):159–163.
36. Prior M, Ozonoff S. Psychological factors in autism. In: Volkmar FR, ed. Autism
and Pervasive Developmental Disorders. Cambridge: Cambridge University Press,
1998:64–108.
37. Rutter M, Bailey A, Simonoff E, Pickles A. Genetic influences in autism. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:370–387.
38. Poustka F, Lisch S. Autistic behaviour domains and their relation to self-injurious
behaviour. Acta Paedopsychiatry 1993; 56(2):69–73.
39. Jaselskis CA, Cook EH, Fletcher KE, Leventhal BL. Clonidine treatment of hyper-
active and impulsive children with autistic disorder. J Clin Psychopharmacol 1992;
2(5):322–327.
40. Quintana H, Birmaher B, Stedge D, Lennon S, Freed J, Bridge J, Greenhill L. Use
of methylphenidate in the treatment of children with autistic disorder. J Autism Dev
Disord 1995; 25(3):283–294.
41. Realmuto GM, Main B. Coincidence of Tourette’s disorder and infantile autism. J
Autism Dev Disord 1982; 12(4):367–372.
42. Ghaziuddin M, Tsai L, Ghaziuddin N. Comorbidity of autistic disorder in children
and adolescents. Eur Child Adolesc Psychiatry 1992; 1(4):209–213.
43. Nelson EC, Pribor EF. A calendar savant with autism and Tourette syndrome: re-
sponse to treatment and thoughts on the interrelationships of these conditions. Ann
Clin Psychiatry 1993; 5(2):135–140.
44. McDougle CJ, Kresch LE, Goodman WK, Naylor ST, Volkmar FR, Cohen DJ,
Price LH. A case-controlled study of repetitive thoughts and behavior in adults with
autistic disorder and obsessive-compulsive disorder. Am J Psychiatry 1995; 152(5):
772–777.
14 Volkmar et al.
45. Rumsey JM, Rapoport JL, Sceery WR. Autistic children as adults: psychiatric, so-
cial, and behavioral. J Am Acad Child Psychiatry 1985; 24(4): 465–473.
46. McDougle, CJ, Naylor ST, Cohen DJ, Volkmar FR, Henninger GR, Price LH. A
double-blind, placebo controlled study of fluvoxamine in adults with autistic disor-
ders. Arch Gen Psychiatry 1996; 53(11):1001–1008.
47. Baron-Cohen S. Do autistic children have obsessions and compulsions? Br J Clin
Psychol 1989; 28(Pt 3):193–200.
48. Burd L, Fisher WW, Kerbeshian J, Arnold ME. Is development of Tourette’s disor-
der a marker for improvement in patients with autism and other pervasive develop-
mental disorders? J Am Acad Child Adolesc Psychiatry 1987; 26(2):162–165.
49. Lainhart JE, Folstein SE. Affective disorders in people with autism: a review of
published cases. J Autism Dev Disord 1994; 24(5):587–601.
50. Steingard R, Biederman J. Lithium responsive manic-like symptoms in two individ-
uals with autism and mental retardation. J Am Acad Child Adolesc Psychiatry 1987;
26(6):932–935.
51. Handen BL. Pharmacotherapy in mental retardation and autism. School Psychol
Rev 1993; 22(2):163–183.
52. Hellgren L, Gillberg IC. Children with deficits in attention, motor control and per-
ception (damp) almost grown up: at age 16 years. Eur Child Adolesc Psychiatry
1994; 3(1):11–15.
53. Hollander E, Cartwright C, Wong CM, DeCaria CM, DelGiudice-Asch G, Buchs-
baum MS, Aronowitz BR. A dimensional approach to the autism spectrum. Curr
Trends Autism 1999; 103–114.
54. Rett A. Uber ein eigenartiges hirntophisces syndroem bei hyperammonie im kinder-
salter. Wein Medizinische Wochenschrift 1966; 118:723–726.
55. Volkmar FR, Klin A, et al. Childhood disintegrative disorder. In: Cohen DJ, Vol-
kmar FR, eds. Handbook of Autism and Pervasive Developmental Disorders. 2nd
ed. New York: Wiley, 1997:47–59.
56. Asperger H. Die “autistichen psychopathen” im kindersalter. Archive für Psychia-
trie und Nervenkrankheiten 1944; 117:76–136.
57. Volkmar F, Cook E Jr, Pomeroy J, Realmuto G, Tanguay P. Summary of the practice
parameters for the assessment and treatment of children, adolescents and adults with
autism and other pervasive developmental disorders. J Am Acad Child Adolesc
Psychiatry 1999; 38(12):1611–1615.
58. Rutter M, Bailey A, Bolton P, Le Couter A. Autism and known medical conditions:
myth and substance. J Child Psychol Psychiatry 1994; 35:311–322.
59. Gilberg C, Coleman M. Autism and medical disorders: a review of the literature.
Dev Med Child Neurol 1996; 38:191–202.
2
Core Symptoms, Related Disorders,
and Course of Autism
INTRODUCTION
Autism, originally described by Kanner in 1943, is among the most severe of all
neurodevelopmental disorders. It is a pervasive disorder associated with substan-
tial deficits in reciprocal social interaction and communication, and the presence
of repetitive and stereotyped behaviors and unusual interests (1). These classic
features of autism typically appear in infancy, and the syndrome, by definition,
is always present by the age of 3 years. Its manifestations and course often change
throughout development, yet autism remains a chronic, lifelong, and disabling
condition.
Epidemiological studies indicate a lifetime prevalence of autistic disorder
of five to 17 per 10,000 individuals, with the rate of autism among siblings esti-
mated even higher, between 50 and 175 per 10,000 (2–4). Recent reports of
the rising incidence of autism have generated considerable support for increased
research into the causes and treatment of autism (5). Of note, the Child Health
Act of 2000 was the first U.S. governmental initiative to specifically address the
need for comprehensive research to elucidate the presumably complex causes and
nature of this disorder, thereby aiding diagnosis, detection, prevention, prognostic
accuracy, and treatment.
Autism is a developmental disorder and, as such, symptoms and behaviors
change over the course of development. It is heterogeneous with regard to clinical
15
16 Hollander and Nowinski
involves social deficits and repetitive, restricted interests but no delay in the de-
velopment of language, cognition or age-appropriate self-help skills. Thus, it is
difficult to clinically distinguish it from high-functioning autism, i.e., an individ-
ual with autism who also has average intellectual functioning and no language
delay.
Differences among autism spectrum disorders seem to be linked to intelli-
gence, level of adaptive functioning, and number of autistic symptoms rather
than to the presence of distinct symptoms. Yet, if one takes a dimensional view
of the pervasive developmental disorders, one can clearly see that the three core
areas of deficit that define autism manifest in various ways and to varying degrees.
Social impairments may range from inappropriate seeking of affection from
strangers to complete withdrawal. Communication deficits can range from good
language abilities with deviant language use within a social setting to mutism.
Impairments in behavior and activities may range from an ability to engage in
symbolic play but a preoccupation with one limited interest to a constant involve-
ment in nonfunctional repetitive actions. The child with autism may be mute but
able to perform certain sophisticated technical actions. These often contradictory
symptoms make sense within the idea of an autism continuum.
Autism does not appear to be a reflection of a single gene defect; rather,
several genes may play a role in shaping the core and associated symptoms of
this disorder. Moreover, autistic symptoms may be a final common pathway re-
sulting from a variety of genetic and/or environmental factors (e.g., birth compli-
cations, exposure to toxins, etc.) affecting various brain systems. The disorder
presents a broad phenotype, with multiple clinical symptoms of differing severity
from the three core symptom domains: social interaction, communication, and
repetitive behaviors. Current literature describes the “broad autism phenotype”
(BAP) to represent the wide range of symptoms observed in individuals with
autism spectrum disorders. Defining the BAP allows researchers to study autism
spectrum disorders by including individuals, specifically siblings, with autism-
related symptoms into population samples (8,9). By using larger samples in re-
search, scientists can increase the level of confidence that their findings are accu-
rate and significant. The more inclusive definition has been particularly useful
in examining the genetic influence on autism by studying groups of subjects with
similar phenotypes or symptom presentations (e.g., Ref. 10).
Our understanding of the numerous presentations of autism and how they
relate to variations within the normal population is still unclear. The three core
deficits can be viewed on a dimension, with the milder variants falling within
the BAP and the severe variants, when all are present concurrently, forming the
classic autistic phenotype. Since autism is a developmental disorder, its symp-
toms may change over the course of illness in line with important changes in brain
development, making it even more difficult to diagnose and treat this disorder.
Comorbid neurological conditions, e.g., mental retardation and seizure disorder,
18 Hollander and Nowinski
also complicate the picture. To some extent, better definition of clinical symptoms
through the development of valid and reliable diagnostic instruments, such as
the Autism Diagnostic Interview–Revised (ADI-R) (11), has allowed for the col-
lection of more homogeneous, or similar, study populations. Nevertheless, the
above issues of heterogeneity, developmental variation, neurological comorbid-
ity, and multiple-gene involvement contribute to difficulty in determining the
neurobiology, brain mechanisms, and selective treatment response of autism.
A significant limitation in research on the neurobiology and treatment of
autism has been the lack of attention to this heterogeneity and comorbidity. It
remains to be seen whether the disorders on the autism spectrum have multiple
etiologies and varied neural systems but result in the same core deficits that define
the spectrum—social, communication, and repetitive behaviors—or whether
there is a specific, common neural substrate that underlies autism, with different
cases showing different levels of severity of symptoms. Since multiple genes
appear to be involved in autism (e.g., Ref. 12), it may be hypothesized that spe-
cific genes may be linked to specific behavioral dimensions. It is the complex
interaction of these genes that gives rise to the overall clinical picture. Environ-
mental factors have been viewed as possible “second hits” that, in the context of
the BAP, lead to more severe deficits and produce the classic autistic phenotype.
Questions remain regarding the boundaries of this broader autistic phenotype.
For example, are features of one of the dimensions alone necessary or are deficits
from all three dimensions required in order to fall within the spectrum?
CATEGORIZING AUTISM
One potentially fruitful approach to increasing our understanding of autism in-
volves better characterizing specific core symptom domains and associated symp-
tom features of autistic phenomenology. The DSM-IV clearly identifies three
core dimensions of autism: 1) social interaction, 2) speech/communication, and
3) compulsive/repetitive behaviors (1). The categorical approach of the DSM-
IV specifies that patients must have at least one abnormality in each of the three
dimensions to meet criteria for autistic disorder (1). Despite prior views that the
social and language domains were inseparable, these deficits have been shown
to occur in isolation and neither appears to be a result of the other (13). Further-
more, the DSM-IV field trial for autism supports the independence of all three
dimensions and the necessity of symptoms in each domain to accurately diagnose
autism (14).
Since the late 1970s, researchers have attempted to categorize autistic pa-
tients to better define the population. A recent review of approaches to subtyping
autism (15) describes four methods: 1) social interaction/communication subtyp-
ing, 2) intellectual/developmental level and adaptive functioning subtyping, 3)
Symptoms, Related Disorders, and Course 19
Figure 1 The core dimensions of autism, plus associated features and related disorders.
Symptoms, Related Disorders, and Course 21
odd children were more spontaneous in their social approaches, but these were
odd and idiosyncratic. They were more aware of other people’s emotional reac-
tions. An example is an autistic child who approaches adults to feel their clothes
or hair.
The social dimension extends into the areas of personality disorders, e.g.,
schizoid and schizotypal personality, and social anxiety or phobia, although autis-
tic patients exhibit only selected symptoms of these disorders and do so in combi-
nation with a variety of behaviors from the other domains. There is overlap of
autistic social deficits with schizoid personality disorder, in which there is a de-
tachment from social relationships and a restricted range of emotional expression.
Wolff and Chick (32) described a group of children diagnosed with schizoid
personality disorder as resembling those with Asperger’s syndrome but differenti-
ating from children with autism spectrum disorders by the absence of delayed
language development, emotional unresponsiveness, gaze avoidance, and
ritualistic/compulsive behaviors. These children did, however, display features
of social isolation, reduced capacity for empathy, hypersensitivity, rigidity of
mental set, and an odd style of communication.
In schizotypal personality disorder, there is a pervasive pattern of social and
interpersonal deficits. Affected individuals experience considerable discomfort
in close intimate relationships, exhibit eccentric behavior, and display distinct
cognitive and perceptual distortions. They have significant social anxiety, which
is associated with suspiciousness of other people’s motives. Both schizoid and
schizoptypal personality disorders have a higher prevalence among first-degree
relatives of those diagnosed with schizophrenia.
The individual with social phobia fears social/performance situations, such
as exposure to unfamiliar people in which embarrassment may occur. Exposure to
the feared situation leads to the anxiety response, which in turn leads to significant
avoidance behavior, and, if chronic, the disorder overlaps significantly with
avoidant personality disorder. A significantly higher rate of social phobia has
been found in the family members of nonmentally retarded individuals with au-
tism (33).
Using a dimensional approach in autism research has been useful in charac-
terizing the social domain. For example, researchers have examined the role of
neuropeptide systems, specifically the oxytocin system, in relationship to this
symptom domain in determining possible etiological factors leading to autism.
Studies of highly social and asocial animal species have documented increased
oxytocin receptor density in limbic structures such as the anterior cingulate gyrus,
which has been implicated in autism (34). This finding suggested that peptide
systems, particularly the oxytocin system, may promote normal and appropriate
social functioning. Panksepp first hypothesized that a failure to shift oxytocin
receptor density from an infantile to a mature pattern may underlie the develop-
ment of autism (35). Decreased levels of plasma oxytocin have been found in
Symptoms, Related Disorders, and Course 23
autistic children compared with normal control subjects, and social impairments
were associated with changes in plasma oxytocin levels (20,36). While normal
children showed an increase in plasma oxytocin levels, children with autism
failed to show a similar developmental increase in levels. Furthermore, elevated
levels of oxytocin were associated with lower functioning in skills of daily living
and interpersonal relations in autistic subjects, unlike normal children who exhib-
ited a positive association (36).
To better clarify the role of oxytocin, Green and colleagues (37) investi-
gated precursor peptide forms of oxytocin. They found elevated oxytocin precur-
sor peptide levels in contrast with reduced plasma oxytocin levels, which may
indicate incomplete processing of oxytocin prohormone in autism (37). A pro-
cessing deficiency is likely due to other biochemical factors, e.g., prohormone
convertases, although future studies must address these patterns of alteration.
Green et al. propose a genetic explanation for oxytocin processing and regulation
and describes such an investigation as the next step in studying the relationship
among social impairment, the oxytocin system, and autism (37). This example
describes the utility of focusing on symptom domains in autism and linking these
with underlying neurobiological/genetic mechanisms that could potentially con-
tribute to the development of targeted treatment approaches for individuals with
autism.
speech and communication skills, they too frequently show some deviance in the
language dimension, particularly in their pragmatic language skills, such as the
use of idiosyncratic tone and volume, a formal, professorial style of speech, and
difficulty using speech to interact fully with the world.
As briefly described above, recent research advances have resulted from
looking at the BAP. This approach has been particularly useful in studying defi-
cits in the communication domain. By extending the definition of autism beyond
that of classic autism, researchers can include subjects with milder variants, such
as PDD-NOS and Asperger’s syndrome. Studies that examined milder language
phenotypes found higher rates of language impairment in relatives of autistic
patients. In particular, one twin study revealed a familial relationship in impair-
ment in nonverbal communication and verbal/nonverbal status as shown by the
ADI-R (38). Therefore, the use of language/communication as a means to subtype
autism appears to be a logical approach for genetics studies (39).
By specifically looking at the language phenotype, investigators may have
an increased ability to locate genes contributing to the BAP (40). Silverman et
al. (25) found reduced variation within families compared with between families
in the level of deficits in nonverbal communication, the presence of phrase
speech, and the age of onset of phrase speech. Although increased similarity
among biologically related siblings does not directly implicate genetic factors
(environmental factors might also explain evidence of “familiality”), the familial
factors identified led to obvious ways in which families might be stratified
for molecular genetic studies. In their sample, Silverman et al. found that the
variance within siblings was reduced for the repetitive behavior domain and
for delays in and the presence of useful phrase speech. These features and also
the nonverbal communication subdomain provided even somewhat stronger evi-
dence of familiality when the authors restricted the sibling groups to include only
those carrying the narrow diagnosis of autism to define multiply affected sibling
groups.
In accordance with these findings of familial patterns in the communication
domain, Buxbaum and colleagues (10) found that restricting autism-affected rela-
tive pairs to those with phrase speech delay was useful in locating an autism
susceptibility gene. Restricting the analysis to the subset of families with two or
more individuals having a narrow diagnosis of autism with phrase-speech delay
generated increased scores for linkage probability (10). An earlier linkage study
used a stricter definition of autism than that defined by the DSM-IV or the ADI-
R, specifically in language-impairment criteria, yet produced strong linkage that
implicated chromosome 7 as a susceptibility gene (41). A more recent study
showed that linkage signals on chromosomes 7 and 13 were due primarily to the
language phenotype (23). Such studies provide evidence for the usefulness of
subcategorizing families based on symptoms within the communication domain,
particularly in linkage analyses.
Symptoms, Related Disorders, and Course 25
followed by myoclonic and partial seizures, followed by atonic seizures and in-
fantile spasms (67). In addition to the high rate of seizures, approximately half
of all persons with autism have abnormal EEGs that demonstrate nonspecific
abnormalities including focal slowing, generalized slowing, focal or centropari-
etal spikes, bilateral or multifocal spikes, and generalized spikes (72). Studies
have failed to establish specific patterns of pathology (72,73).
Psychiatric symptoms of affective instability, aggression, impulsivity, and
self-injurious behavior are also common associated features of autism. Lainhart
and Folstein (74) reviewed 17 published cases and found 35% of the autistic
individuals to have an affective disorder. Among patients with Asperger’s syn-
drome, 24% were found to have an affective disorder. Recent data suggest that
depression is common among mentally retarded, emotionally disturbed patients
(75), and one study (76) estimates the prevalence rate at 9% for depressive mood
in that population. However, depression may be underdiagnosed in autism; symp-
toms of depression may be confused with autistic symptomatology and, because
of difficulties of communication, are rarely reported by the afflicted individual.
This may explain why most reported cases of affective disorder in autism have
described patients with higher levels of intelligence and better verbal skills (77).
Increased rates of affective disorders in first-degree relatives of autistic
probands would lend support to the diagnostic validity of affective disorders in
this population (74). Studies have reported increased rates of bipolar disorder
(78) and elevated rates of major depressive disorder and social difficulties (33,79)
in relatives of autistic probands compared to controls. Bolton et al. (80) found
that the rate of affective illness (minor/major/bipolar disorder) in first-degree
relatives of autistic probands was twice as high as in Down’s syndrome controls
(35% vs. 17.3%). In autistic probands, the proportion having a positive family
history of mood disorders increases significantly when the proband receives a
diagnosis of depression (81).
Aggression, both self- and other-directed, is another common associated
feature of autism (82), as is impulsivity (83). In a report on comorbid diagnoses
in autism, Tsai found a prevalence rate of 25–43% of self-injurious behavior,
considered to be one form of aggression, in autistic children, a finding supported
by other independent studies (76,84,85). The prevalence rate of self-injurious
behavior is 2–9% among noninstitutionalized mentally retarded individuals and
up to 40% among institutionalized mentally retarded individuals (86,87). Esti-
mates of aggressive behavior in mentally retarded individuals range from 8.9%
to 24% (88,89), with rates of up to 45% of institutionalized mentally retarded
individuals (90). Of child patients with autism entered into a placebo-controlled,
double-blind study of fluoxetine treatment, approximately 50% presented with
aggressive behavior as a significant comorbid symptom (Hollander et al., unpub-
lished data). There is also evidence of increased expression of an impulsivity trait
in relatives of autistic probands (91).
Symptoms, Related Disorders, and Course 29
CONCLUDING THOUGHTS
We have discussed a dimensional approach to the autism spectrum in which dif-
ferent but overlapping groups of children with PDD exist on a continuum, having
similar qualitative features but distinguished by the degree of impairment. The
fact that data from many disparate areas come together to implicate neurobiologi-
cal and genetic factors modulating core symptom domains in autism speaks to
the explanatory power of the conceptualization. It is clear that no single gene
Symptoms, Related Disorders, and Course 31
accounts for the transmission of autism, but rather that several genes may contrib-
ute, each contributing a small amount of the overall variance. It appears that core
symptom domains have greater variation between families than within families,
suggesting that the severity of these domains runs within families. Stratifying
the autistic population based on severity of the core symptom domains may pro-
vide more homogeneous populations with which to elucidate genes that mediate
the core domain phenotype. Various neurotransmitters, neuropeptide abnormali-
ties, and neurocircuitry abnormalities have been reported in autistic subjects, but
lack of replication has been the rule, rather than the exception, in autism. An
important contribution to this variance has been heterogeneity of autism. Further,
abnormalities in neurobiological mechanisms have not correlated with autistic
severity. However, specific neurobiological abnormalities, such as the 5-HT re-
ceptor subsystem, oxytocin function, autoimmune markers, and regional meta-
bolic activity, have been tightly mapped to the severity of specific core symptom
domains. Thus, elucidating the pathophysiology and etiology of autism may be
hampered by heterogeneity, but elucidating the pathophysiology and etiology of
core domains may be successful even in the face of heterogeneity. Finally, no
single treatment appears helpful in addressing all aspects of autism or all afflicted
individuals. Nevertheless, the development of targeted treatments for specific
core and associated features of autism may be successfully undertaken, may im-
prove global severity and quality of life in subjects, and may lead to determining
which patients and which symptom clusters respond to which targeted treatments.
Still, we must address the pitfalls of using the core domain approach to
both clinically and scientifically understand autism. First, the core symptom do-
mains may not be completely independent or orthogonal dimensions. For exam-
ple, patients with severe social deficits may also have severe speech/language
deficits and severe repetitive behaviors. Second, brain systems that mediate core
domains may also mediate other domains, such that oxytocin and serotonin abnor-
malities may mediate both repetitive behaviors and social-deficit domains. Third,
treatments that address one symptom domain may also modulate other symptom
domains either directly or indirectly. For instance, SSRIs, which improve repeti-
tive behaviors, may also improve social deficits. This could be directly due to
effects of 5-HT on social functioning, or indirectly in that patients less impaired
by severe compulsivity, craving for sameness, and rigid adherence to routines may
have greater opportunities for improving social functioning. Fourth, in addition
to the core symptom domains, autistic patients are often impaired by comorbid
conditions (e.g., seizures, mental retardation) and associated features (e.g.,
impulsivity/aggression, affective instability). Thus, genetic, neurobiological, and
treatment studies must also address these comorbid conditions and associated fea-
tures to have a more robust impact on understanding and treating autistic patients.
Future research must address the need to refine our categories of different
groups of individuals with autism. By identifying the diagnostic boundaries of
32 Hollander and Nowinski
autism, we may better understand the causes, features, and course of this disorder
and provide viable treatment options for individuals with autism. In the mean-
time, the core symptom domains of autism may allow clinicians and researchers
alike to best examine, understand, and treat the autism spectrum given our current
limitations.
ACKNOWLEDGMENT
This chapter was supported in part by the Seaver Foundation, Food and Drug
Administration (FDA) grants FDR001520 and FDR002026, and National Insti-
tute of Neurological Diseases and Stroke grant NS43979.
REFERENCES
1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders. 4th ed. Washington, DC: American Psychiatric Publishing, 1994:886.
2. Fombonne E. The epidemiology of autism: a review. Psychol Med 1999; 29:769–
786.
3. Chakrabarti S, Fombonne E. Pervasive developmental disorders in preschool chil-
dren. JAMA 2001; 285:3093–3099.
4. Gillberg C, Wing L. Autism: not an extremely rare disorder. Acta Psychiatr Scand
1999; 99:399–406.
5. Department of Developmental Services. Changes in the Population of Persons with
Autism and Pervasive Developmental Disorders in California’s Developmental
Services System: 1987 through 1998. Report to the Legislature, March 1, 1999.
Available at http:/ /www.dds.ca.gov.
6. World Health Organization. The ICD-10 Classification of Mental and Behavioural
Disorders: Clinical Descriptions and Diagnostic Guidelines. Geneva: World Health
Organization, 1992:xii, 362.
7. Myhr G. Autism and other pervasive developmental disorders: exploring the di-
mensional view. Can J Psychiatry 1998; 43:589–595.
8. Le Couteur A, Bailey A, Goode S, Pickles A, Robertson S, Gottesman I, Rutter
M. A broader phenotype of autism: the clinical spectrum in twins. J Child Psychol
Psychiatry 1996; 37:785–801.
9. Piven J, Palmer P, Jacobi D, Childress D, Arndt S. Broader autism phenotype:
evidence from a family history study of multiple-incidence autism families. Am
J Psychiatry 1997; 154:185–190.
10. Buxbaum JD, Silverman JM, Smith CJ, Kilifarski M, Reichert J, Hollander E,
Lawlor BA, Fitzgerald M, Greenberg DA, Davis KL. Evidence for a susceptibility
gene for autism on chromosome 2 and for genetic heterogeneity. Am J Hum Genet
2001; 68:1514–1520.
11. Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview–Revised: a revised
version of a diagnostic interview for caregivers of individuals with possible perva-
sive developmental disorders. J Autism Dev Disord 1994; 24:659–685.
Symptoms, Related Disorders, and Course 33
12. Cook EH Jr. Genetics of autism. Child Adolesc Psychiatr Clin N Am 2001; 10:
333–350.
13. Bailey A, Phillips W, Rutter M. Autism: towards an integration of clinical, genetic,
neuropsychological, and neurobiological perspectives. J Child Psychol Psychiatry
1996; 37:89–126.
14. Volkmar FR, Klin A, Siegel B, Szatmari P, Lord C, Campbell M, Freeman BJ,
Cicchetti DV, Rutter M, Kline W, et al. Field trial for autistic disorder in DSM-
IV. Am J Psychiatry 1994; 151:1361–1367.
15. Beglinger LJ, Smith TH. A review of subtyping in autism and proposed dimen-
sional classification model. J Autism Dev Disord 2001; 31:411–422.
16. Wing L, Gould J. Severe impairments of social interaction and associated abnor-
malities in children: epidemiology and classification. J Autism Dev Disord 1979;
9:11–29.
17. Tanguay PE, Robertson J, Derrick A. A dimensional classification of autism spec-
trum disorder by social communication domains. J Am Acad Child Adolesc Psy-
chiatry 1998; 37:271–277.
18. Stevens MC, Fein DA, Dunn M, Allen D, Waterhouse LH, Feinstein C, Rapin I.
Subgroups of children with autism by cluster analysis: a longitudinal examination.
J Am Acad Child Adolesc Psychiatry 2000; 39:346–352.
19. Rapin I. Practitioner review—developmental language disorders: a clinical update.
J Child Psychol Psychiatry 1996; 37:643–655.
20. Waterhouse L, Fein D, Modahl C. Neurofunctional mechanisms in autism. Psychol
Rev 1996; 103:457–489.
21. Rutter M, Bailey A, Bolton P, Le Couteur A. Autism and known medical condi-
tions: myth and substance. J Child Psychol Psychiatry 1994; 35:311–322.
22. Gillberg C. Subgroups in autism: are there behavioural phenotypes typical of un-
derlying medical conditions? J Intellect Disabil Res 1992; 36(Pt 3):201–214.
23. Bradford Y, Haines J, Hutcheson H, Gardiner M, Braun T, Sheffield V, Cassavant
T, Huang W, Wang K, Vieland V, Folstein S, Santangelo S, Piven J. Incorporating
language phenotypes strengthens evidence of linkage to autism. Am J Med Genet
2001; 105:539–547.
24. Hollander E, King A, Delaney K, Smith CJ, Silverman JM. Obsessive compulsive
behaviors in parents of multiplex autism families. Psychiatry Res. In Press.
25. Silverman JM, Smith CJ, Schmeidler J, Hollander E, Lawlor BA, Fitzgerald M,
Buxbaum JD, Delaney K, Galvin P. Symptom domains in autism and related con-
ditions: evidence for familiality. Am J Med Genet 2002; 114:64–73.
26. McDougle CJ, Naylor ST, Cohen DJ, Volkmar FR, Heninger GR, Price LH. A
double-blind, placebo-controlled study of fluvoxamine in adults with autistic disor-
der. Arch Gen Psychiatry 1996; 53:1001–1008.
27. McDougle CJ, Holmes JP, Carlson DC, Pelton GH, Cohen DJ, Price LH. A double-
blind, placebo-controlled study of risperidone in adults with autistic disorder and
other pervasive developmental disorders. Arch Gen Psychiatry 1998; 55:633–641.
28. Lord C, Risi S, Lambrecht L, Cook EH, Jr., Leventhal BL, DiLavore PC, Pickles
A, Rutter M. The autism diagnostic observation schedule-generic: a standard mea-
sure of social and communication deficits associated with the spectrum of autism.
J Autism Dev Disord 2000; 30:205–223.
34 Hollander and Nowinski
29. Baron-Cohen S. Do people with autism understand what causes emotion? Child
Dev 1991; 62:385–395.
30. Rutter M. The treatment of autistic children. J Child Psychol Psychiatry 1985; 26:
193–214.
31. Sigman M, Mundy P. Social attachments in autistic children. J Am Acad Child
Adolesc Psychiatry 1989; 28:74–81.
32. Wolff S, Chick J. Schizoid personality in childhood: a controlled follow-up study.
Psychol Med 1980; 10:85–100.
33. Smalley SL, McCracken J, Tanguay P. Autism, affective disorders, and social
phobia. Am J Med Genet 1995; 60:19–26.
34. Insel TR, Shapiro LE. Oxytocin receptor distribution reflects social organization
in monogamous and polygamous voles. Proc Natl Acad Sci USA 1992; 89:5981–
5985.
35. Panksepp J. Commentary on the possible role of oxytocin in autism. J Autism Dev
Disord 1993; 23:567–569.
36. Modahl C, Green L, Fein D, Morris M, Waterhouse L, Feinstein C, Levin H.
Plasma oxytocin levels in autistic children. Biol Psychiatry 1998; 43:270–277.
37. Green L, Fein D, Modahl C, Feinstein C, Waterhouse L, Morris M. Oxytocin and
autistic disorder: alterations in peptide forms. Biol Psychiatry 2001; 50:609–613.
38. MacLean JE, Szatmari P, Jones MB, Bryson SE, Mahoney WJ, Bartolucci G, Tuff
L. Familial factors influence level of functioning in pervasive developmental disor-
der. J Am Acad Child Adolesc Psychiatry 1999; 38:746–753.
39. Folstein SE, Santangelo SL, Gilman SE, Piven J, Landa R, Lainhart J, Hein J,
Wzorek M. Predictors of cognitive test patterns in autism families. J Child Psychol
Psychiatry 1999; 40:1117–1128.
40. Piven J. The broad autism phenotype: a complementary strategy for molecular
genetic studies of autism. Am J Med Genet 2001; 105:34–35.
41. International Molecular Genetic Study of Autism Consortium. A full genome
screen for autism with evidence for linkage to a region on chromosome 7q. Hum
Mol Genet 1998; 7:571–578.
42. Rumsey JM, Duara R, Grady C, Rapoport JL, Margolin RA, Rapoport SI, Cutler
NR. Brain metabolism in autism: resting cerebral glucose utilization rates as mea-
sured with positron emission tomography. Arch Gen Psychiatry 1985; 42:448–
455.
43. McDougle CJ, Kresch LE, Goodman WK, Naylor ST, Volkmar FR, Cohen DJ,
Price LH. A case-controlled study of repetitive thoughts and behavior in adults
with autistic disorder and obsessive-compulsive disorder. Am J Psychiatry 1995;
152:772–777.
44. Schain RJ, Freedman DX. Studies on 5-hydroxyindole metabolism in autistic and
other mentally retarded children. J Pediatr 1961; 58:315–320.
45. Cook EH, Leventhal BL. The serotonin system in autism. Curr Opin Pediatr 1996;
8:348–354.
46. McDougle CJ, Naylor ST, Cohen DJ, Aghajanian GK, Heninger GR, Price LH.
Effects of tryptophan depletion in drug-free adults with autistic disorder. Arch Gen
Psychiatry 1996b; 53:993–1000.
47. Hollander E, Novotny S, Allen A, Aronowitz B, Cartwright C, DeCaria C. The
Symptoms, Related Disorders, and Course 35
JS, Johnson CP, Kallen RJ, Levy SE, Minshew NJ, Ozonoff S, Prizant BM, Rapin
I, Rogers SJ, Stone WL, Teplin S, Tuchman RF, Volkmar FR. The screening and
diagnosis of autistic spectrum disorders. J Autism Dev Disord 1999; 29:439–484.
62. Nordin V, Gillberg C. The long-term course of autistic disorders: update on follow-
up studies. Acta Psychiatr Scand 1998; 97:99–108.
63. Gillberg C, Steffenburg S. Outcome and prognostic factors in infantile autism and
similar conditions: a population-based study of 46 cases followed through puberty.
J Autism Dev Disord 1987; 17:273–287.
64. Volkmar FR, Nelson DS. Seizure disorders in autism. J Am Acad Child Adolesc
Psychiatry 1990; 29:127–129.
65. Giovanardi Rossi P, Posar A, Parmeggiani A. Epilepsy in adolescents and young
adults with autistic disorder. Brain Dev 2000; 22:102–106.
66. Rossi PG, Parmeggiani A, Bach V, Santucci M, Visconti P. EEG features and
epilepsy in patients with autism. Brain Dev 1995; 17:169–174.
67. Tuchman RF, Rapin I, Shinnar S. Autistic and dysphasic children. II: Epilepsy.
Pediatrics 1991; 88:1219–1225.
68. Deykin EY, MacMahon B. The incidence of seizures among children with autistic
symptoms. Am J Psychiatry 1979; 136:1310–1312.
69. Olsson I, Steffenburg S, Gillberg C. Epilepsy in autism and autisticlike conditions:
a population-based study. Arch Neurol 1988; 45:666–668.
70. Rutter M. Autistic children growing up. Dev Med Child Neurol 1984; 26:122–
129.
71. Gillberg CL. The Emanuel Miller Memorial Lecture 1991. Autism and autistic-
like conditions: subclasses among disorders of empathy. J Child Psychol Psychia-
try 1992; 33:813–842.
72. Tuchman RF, Jayakar P, Yaylai I, Villalobos R. Seizures and EEG findings in
children with autism spectrum disorder. CNS Spectrums 1998; 3:61–70.
73. Hutt SJ, Hutt C, Lee D, Ounsted C. A behavioural and electroencephalographic
study of autistic children. J Psychiatr Res 1965; 3:181–197.
74. Lainhart JE, Folstein SE. Affective disorders in people with autism: a review of
published cases. J Autism Dev Disord 1994; 24:587–601.
75. Matson JL, Manikam R, Coe D, Raymond K, Taras M, Long N. Training social
skills to severely mentally retarded multiply handicapped adolescents. Res Dev
Disabil 1988; 9:195–208.
76. Chung SY, Luk SL, Lee PW. A follow-up study of infantile autism in Hong Kong.
J Autism Dev Disord 1990; 20:221–232.
77. Ghaziuddin M, Tsai L. Depression in autistic disorder. Br J Psychiatry 1991; 159:
721–723.
78. DeLong GR, Teague LA, McSwain Kamran M. Effects of fluoxetine treatment in
young children with idiopathic autism. Dev Med Child Neurol 1998; 40:551–562.
79. Piven J, Palmer P. Psychiatric disorder and the broad autism phenotype: evidence
from a family study of multiple-incidence autism families. Am J Psychiatry 1999;
156:557–563.
80. Bolton PF, Pickles A, Murphy M, Rutter M. Autism, affective and other psychiat-
ric disorders: patterns of familial aggregation. Psychol Med 1998; 28:385–395.
81. Ghaziuddin M, Greden J. Depression in children with autism/pervasive develop-
Symptoms, Related Disorders, and Course 37
mental disorders: a case-control family history study. J Autism Dev Disord 1998;
28:111–115.
82. Weller EB, Rowan A, Elia J, Weller RA. Aggressive behavior in patients with
attention-deficit/hyperactivity disorder, conduct disorder, and pervasive develop-
mental disorders. J Clin Psychiatry 1999; 60(suppl 15):5–11.
83. Jaselskis CA, Cook EH Jr, Fletcher KE, Leventhal BL. Clonidine treatment of
hyperactive and impulsive children with autistic disorder. J Clin Psychopharmacol
1992; 12:322–327.
84. Tsai LY. Brief report: comorbid psychiatric disorders of autistic disorder. J Autism
Dev Disord 1996; 26:159–163.
85. Simons JM. Observations on compulsive behavior in autism. J Autism Child
Schizophr 1974; 4:1–10.
86. Griffin JC, Ricketts RW, Williams DE, Locke BJ, Altmeyer BK, Stark MT. A
community survey of self-injurious behavior among developmentally disabled
children and adolescents. Hosp Commun Psychiatry 1987; 38:959–963.
87. Griffin JC, Williams DE, Stark MT, Altmeyer BK, Mason M. Self-injurious behav-
ior: a state-wide prevalence survey of the extent and circumstances. Appl Res Ment
Retard 1986; 7:105–116.
88. Gardner W, Cole C. Aggression and related conduct disorders: definition, assess-
ment and treatment. In: Matson JL, Barrett RP, eds. Psychopathology in the Men-
tally Retarded. Needham Heights, MA: Allyn and Bacon, 1993:213–245.
89. Jacobson J. Problem behavior and psychiatric impairment within a developmen-
tally disabled population: I. Behavior frequency. Appl Res Ment Retard 1982; 3:
121–139.
90. Eyman RK, Call T. Maladaptive behavior and community placement of mentally
retarded persons. Am J Ment Defic 1977; 82:137–144.
91. Murphy M, Bolton PF, Pickles A, Fombonne E, Piven J, Rutter M. Personality
traits of the relatives of autistic probands. Psychol Med 2000; 30:1411–1424.
92. Piven J, Harper J, Palmer P, Arndt S. Course of behavioral change in autism: a
retrospective study of high-IQ adolescents and adults. J Am Acad Child Adolesc
Psychiatry 1996; 35:523–529.
93. Howlin P, Mawhood L, Rutter M. Autism and developmental receptive language
disorder—a follow-up comparison in early adult life. II: Social, behavioural, and
psychiatric outcomes. J Child Psychol Psychiatry 2000; 41:561–578.
94. Mawhood L, Howlin P, Rutter M. Autism and developmental receptive language
disorder—a comparative follow-up in early adult life. I: Cognitive and language
outcomes. J Child Psychol Psychiatry 2000; 41:547–559.
95. Bagley C, McGeein V. The taxonomy and course of childhood autism. Percept
Mot Skills 1989; 69:1264–1266.
96. Goldstein G, Minshew NJ, Siegel DJ. Age differences in academic achievement
in high-functioning autistic individuals. J Clin Exp Neuropsychol 1994; 16:671–
680.
97. Minshew NJ, Goldstein G, Taylor HG, Siegel DJ. Academic achievement in high
functioning autistic individuals. J Clin Exp Neuropsychol 1994; 16:261–270.
98. Rutter M. Autistic children: infancy to adulthood. Semin Psychiatry 1970; 2:435–
450.
38 Hollander and Nowinski
INTRODUCTION
Autism and the related pervasive developmental disorders (PDDs) are neuropsy-
chiatric syndromes characterized by abnormalities in social relatedness, verbal
and nonverbal communication deficits, and the presence of restricted and stereo-
typed behaviors and interests. These disorders are unique in that the children are
often not diagnosed until years after the symptoms first emerge. The reasons for
delay are multiple. Between the wide spectrum of clinical presentations and the
changes in the definition and diagnostic criteria over the years—see the third,
third–revised, and fourth editions of the Diagnostic and Statistical Manual of
Mental Disorders (DSM-III, III-R, and IV)—clinicians who are not very familiar
with the spectrum may not recognize the disorder. Also, there is often the fear
of imposing an improper label on a child, as well as the hope that the child will
“catch up.” And finally, for years there were no uniform guidelines for the work-
up and evaluation of children with suspected autism spectrum disorders. Yet there
is increasing evidence that early intervention can improve outcomes (1–8), so
the speed and accuracy with which the diagnosis is made are crucial.
In recent years, three different sets of guidelines for the evaluation of chil-
dren with suspected autism have been published. In 1997, a meeting sponsored
by the Cure Autism Now (CAN) foundation resulted in the CAN consensus
guidelines for screening and diagnostic referral, which appeared in CNS Spec-
39
40 Spence and Geschwind
trums in 1998 (9). In 1998, selected members of the American Academy of Child
and Adolescent Psychiatry (AACAP) met to establish a set of practice parameters
for the Academy, which were published in the Journal of the American Academy
of Child and Adolescent Psychiatry in 1999 (10). Most recently, the American
Academy of Neurology (AAN) and the Child Neurology Society (CNS) formed
a committee with representatives from a wide range of organizations with exper-
tise in autism and related disorders to create their own practice parameters for
screening and diagnosis; these were published in Neurology in 2000 (11). A first,
more detailed paper based on this consensus-building approach had appeared in
the Journal of Autism and Developmental Disorders in 1999 (12). This chapter
reviews and summarizes these various guidelines as the state of the science in
screening and neurodevelopmental assessment for autism and autism spectrum
disorders.
INITIAL SCREENING
Many pediatricians are unclear about what to do with a child who presents with
language delay and/or social behavioral problems, especially in the setting of
normal motor development. Often, there is a significant reluctance to acknowl-
edge the problem and thus a delay in making the proper referrals for further
assessment. The most recent set of practice parameters published by the AAN/
CNS emphasizes that this is a major issue in the diagnosis of autism, stating that
part of the problem is that, in general, developmental concerns are not taken
seriously enough. They estimate that up to 25% of children in any primary-care
setting present with some type of developmental concern, yet fewer than 30%
of primary-care providers routinely use standardized tests to screen for develop-
mental issues (11). Thus, the AAN/CNS committee (12) made strong recommen-
dations about basic developmental screening at all well-child visits. Their ap-
proach is divided into two levels. The first is a general screen for all
developmental issues including autism, and the second is more specific to the
formal diagnosis of autism.
at all IQ levels (31). For older, more verbal children, there is the Australian Scale
for Asperger’s syndrome (32), a tool filled out by a parent or teacher to identify
high-functioning children of school age who have gone undetected. From birth
to age 3, there is the Pervasive Developmental Disorders Screening Test–Stage
I (PDDST), which was designed for use in the primary-care setting and is in the
process of being validated (33).
Gillberg and colleagues have developed a screening model for autism in
very young children based on the Symptoms of Autism in Babies (SAB) question-
naire used in both retrospective and prospective studies of children referred for
evaluation of autism (34,35) (see Ref. 36 for discussion). Stone and colleagues
have developed two tools: the Parental Interview for Autism (PIA) (37) and the
Screening Tool for Autism in Two-Year-Olds (STAT), the latter of which is
undergoing validation (38,39). Their group also recently reported that the Vine-
land Adaptive Behavior Scale could be reliably used to differentiate autism from
nonautistic developmental delay (40).
44 Spence and Geschwind
ANCILLARY SCREENING
Historically it was left up to the discretion of individual practitioners regarding
when and what to obtain for laboratory studies. This point was addressed in the
recent guidelines, and although no specific complete laboratory testing has yet
been delineated, there is agreement regarding the basic screening studies that
should be performed in any child failing the screening described above (9,10,12).
Audiological Evaluation
Any child with language and/or social delays requires formal audiological testing
(9,10,12). All groups agree that the assessment should be behaviorally based and
performed by an experienced pediatric audiologist. Electrophysiological test-
ing—e.g., brainstem auditory evoked response (BAER)—is probably necessary
only if the behavioral assessment is considered inconclusive or there is a specific
concern about a central nervous system abnormality. In this case, frequency-
specific testing or tone-evoked responses rather than clicks should be used be-
cause these have been shown to better estimate behavioral hearing thresholds
(41). Evoked otoacoustic emissions are another time- and cost-effective way of
testing cochlear function that can be used in children with autism (42).
Lead Screening
Children with developmental delay are at increased risk of lead toxicity because
of the prolonged period spent in oral-motor stages of play (with frequent mouth-
ing of objects) and the occurrence of pica. Substantially higher lead levels have
been reported in children with autism than in nonautistic controls (43). Another
study found that children with autism were older at time of diagnosis, had sub-
stantially prolonged lead-level elevations, and were at increased risk of re-expo-
sure despite close monitoring compared to a group of nonautistic children with
lead poisoning (44). Thus, the AACAP and CAN consensus committees suggest
lead screening in all children with suspected autism (9,10). The AAN/CNS guide-
lines recommend it only in the cases in which pica is reported (12), but they do
point out that the Centers for Disease Control and Prevention’s 1997 guidelines
for lead screening in the United States recommend that all children with any
developmental delay be screened for lead poisoning.
MEDICAL EVALUATION
History and Physical Examination
All the panels have put forth guidelines for further medical and neurological
work-up at the specialist level and uniformly recommend a detailed history and
physical examination in all children with autism or PDD. This should include
probes specific to birth history, medical history, developmental history, and fam-
ily history. The relationship between autism and other medical diseases is still
uncertain (9,10), and it is unclear exactly what percentage of cases of autism are
attributable to general medical conditions—some say it is low (45) and others
have found up to 25% (46). Thus, attention to these details may, in fact, aid in
diagnosis.
All published guidelines have made similar recommendations regarding the
further medical and/or neurological evaluation. The focus of these assessments
should be on finding any conditions that may be medically treatable or may have
genetic implications for the family (10). The CAN consensus group suggests at
least a thorough developmental and family history, a medical and neurological
examination, including a mental-status exam, and a comprehensive speech and
language evaluation. They also suggest an assessment of the child’s social and
emotional development, with special attention to the communication deficits
and behavioral problems often associated with autism (9).
The AAN/CNS guidelines go into more detail regarding several medical
issues that must be taken into account in evaluating children with autism. Infor-
mation can be obtained from the family history, medical history, and physical
exam. There are significant epidemiological data to support the increased preva-
lence (50- to 100-fold) in first-degree relatives of autistic children (47–49). Other
psychiatric disorders (especially affective disorder, obsessive compulsive disor-
der, and anxiety disorder) are also seen at a higher-than-expected frequency (50–
54). The association with fragile X syndrome necessitates that family histories
be examined for mental retardation and/or autism inherited in an X-linked pat-
46 Spence and Geschwind
tern. While early screening studies had reported rates of up to 25%, more recent
studies have reported much lower percentages (e.g., ⬍5%) of autistic patients
with the fragile X mutation (55–57), including the Autism Genetic Resource
Exchange (AGRE) sample (58). Further, it should be noted that several groups
have found no evidence of the expansion of the (CGG) trinucleotide repeat in
the FMR-1 gene in patients with autism spectrum disorders, leading them to
question whether there is any causal link at all (59–61). Finally, there is a strong
association with tuberous sclerosis complex (TSC), an inherited neurocutan-
eous disease with an autosomal dominant pattern of transmission. Estimates are
that from 20% (62) or 25% (63) to as much as 50 to 60% (64,65) of patients
with TSC have autism. Seizures (especially infantile spasms) and mental retarda-
tion appear to be risk factors for the development of autism in TCS patients
(66,67). However, the percentage of autistic patients who have TSC is much
lower: 0.4% to 3% overall and 8–14% in patients with epilepsy (67). Diagnosing
TSC is important not only for genetic counseling but also for health mainten-
ance because of the associated abnormalities in multiple organ systems requiring
monitoring.
A careful physical exam can reveal other features that may be associated
with autism. General observation can identify dysmorphic features such as those
consistent with the fragile X syndrome or other inherited syndromes. These might
lead to specific genetic testing or a referral for a thorough genetic evaluation.
Skin examination could demonstrate hypopigmented macules or ash-leaf spots
(best seen using an ultraviolet light source or a Wood’s lamp) or other cutaneous
findings associated with TSC. It has also been reported that many children with
autism have relatively large head circumferences that may not be present at birth
(68), but this does not appear indicative of any neuropathology. After a careful
review of the literature, Filipek concluded that macrocephaly alone in an autistic
child in the absence of any signs or symptoms of structural lesions is not an
indication for neuroimaging (69). Finally, there are frequently reported mild sen-
sorimotor deficits. The most common motor problems are hypotonia, limb
apraxia, and motor stereotypies (e.g., rocking, hand or finger mannerisms, un-
usual posturing) (70). The most common sensory difficulties are over- or under-
sensitivity or paradoxical reactions to environmental stimuli (71).
Genetic Testing
Currently the area that has drawn the most interest is genetic testing. As men-
tioned above, many practitioners are testing for fragile X, but most do not perform
routine chromosomal analyses. With the rapidly evolving tools of molecular ge-
netics this is an area of very active research, and multiple loci have been posited
to contribute to autism susceptibility. In the past few years there have been several
reports of full genomic screens looking for putative autism genes (72–75) and
many more reports testing individual candidate genes (see Refs. 49 and 76–78
for reviews). These studies have implicated loci on chromosomes 1, 4, 5, 6, 7,
10, 13, 15, 16, 18, 19, 22, and X. However, it should be noted that the studies
sometimes do not agree, and some of these loci have not been confirmed when
tested in another cohort (77). We are currently involved in ascertaining and study-
ing a large number of multiplex families in the United States, through the AGRE.
The project entails collection of DNA, family histories, and neurological, psychi-
atric, and behavioral data on affected siblings and first-degree relatives in over
400 multiplex families. Most importantly, these data and the biomaterials are
rapidly made available to the scientific community, providing an unprecedented
resource (www.agre.org) (58).
A thorough review is certainly beyond the scope of this chapter, but emerg-
ing genetic data are obviously something to which investigators and clinicians
alike need to pay close attention. So far, the AAN practice parameters are the
only ones to have set a standard of karyotyping and DNA analysis for fragile X
in any autistic child with mental retardation, or with a family history of fragile
X or undiagnosed mental retardation, or with dysmorphic features. However,
there has been at least one reported case of a chromosome 15 abnormality in the
absence of mental retardation (79). This raises the question of whether karyotyp-
ing should be done even in nonretarded autistic patients. Although the overall
incidence of chromosomal abnormalities is probably quite low, it does have major
implications for genetic counseling. Also, karyotyping is an expensive test and
the decision to do widespread screening may ultimately come down to cost.
EEG
Epidemiological studies have shown that there is a significant percentage of chil-
dren with autism who have epilepsy. Estimates range from 7 to 21% in children
(80,81) and up to 35% by adulthood (82). Practitioners should be aware that the
age of onset of the seizures is bimodal, with peaks in early childhood and adoles-
cence (82). Any type of seizure can be seen in autistic children, but complex
partial seizures are probably most prevalent (83). The diagnosis of seizure activity
in autistic individuals is also made more difficult because the behavioral ab-
normalities associated with complex partial seizures (e.g., staring, being unre-
sponsive to one’s name, repetitive motor behaviors) can all be attributed to the
48 Spence and Geschwind
autism (82). Obviously an EEG is recommended and is the standard of care for
all children with clinical or suspected subclinical seizures. The difficulty and
controversy arise when trying to decide whether all children with autism should
have an EEG.
One aspect of this controversy is the discordance of EEG abnormalities
and clinical seizures (e.g., seizures with normal EEG and abnormal EEG without
seizures). Most neurologists will treat clinical seizures regardless of EEG find-
ings, but most do not consider an abnormal EEG in the absence of clinical sei-
zures an indication for anticonvulsant medication. The true incidence of isolated
EEG abnormalities in autistic patients is difficult to estimate since routine EEGs
are not part of the work-up. Tuchman and Rapin (84) reported abnormal EEGs
in 8% of their cohort of over 500 autistic children without clinical epilepsy.
Numerous papers in the literature have explored the possible connection
between autism and Landau-Kleffner syndrome (LKS). LKS is an acquired epi-
leptic aphasia clinically described as the occurrence of auditory agnosia (the in-
ability to understand spoken language) in a typically developing child. This is
often characterized as a regression, and is usually accompanied by seizures
(85,86). It is this language regression, and the question of whether it could be
likened to that seen in the subset of children with autistic regression, that caught
the attention of those studying and treating autism (84).
LKS is also associated with a severe EEG abnormality present in the deep-
est stages of sleep, referred to as continuous spike and wave in slow-wave sleep
(CSWS) or electrical status epilepticus in sleep (ESES). LKS has been treated
with traditional anticonvulsant medications (87), corticosteroids (88), IVIg (89),
and even epilepsy surgery (90) (see Ref. 91 for review), and in many instances
both the seizures and the language impairment improve.
However, there is currently much debate about how much of a connection
exists between autism spectrum disorders and LKS. Does LKS, like autism, exist
on a spectrum and should it be considered in autistic children with abnormal
EEGs or even in all autistic children (91–93)? Anecdotal evidence indicates that
there is a subset of children with abnormalities in their EEG whose language
disturbance improves when treated with anticonvulsants even in the absence of
clinical seizures (91). A recent paper even suggests a causal relationship between
the EEG findings consistent with a benign form of childhood epilepsy, Rolandic
(e.g., centrotemporal spikes), and autistic regression (94). It is certainly tempting
to think that the EEG abnormalities play a role in the abnormal speech and or
behaviors in autistic children as they are presumed to do in LKS (84), because
if this is the case, then perhaps treatment might prove beneficial. But even then
the risks of treatments, which are so far of unproven value, need to be considered
very carefully (95). Thus, until there is more evidence, it seems unreasonable to
make recommendations regarding either routine monitoring or treatment of iso-
lated EEG abnormalities in children with autism spectrum disorders (91).
Screening and Neurodevelopment Assessment 49
CONCLUSION
It is encouraging that expert clinical panels are now addressing the difficult prob-
lems and controversies regarding screening and diagnosing autism and PDD. The
Screening and Neurodevelopment Assessment 51
benefits of the move toward uniform work-up and diagnosis extend beyond those
conveyed to the individual child and family; it will undoubtedly serve to further
research into this disorder. Proper and prompt diagnosis will aid in research re-
garding early intervention and outcomes. Comprehensive reporting of associa-
tions with other diseases, abnormal laboratory values, genetic evaluations, and
other ancillary testing will ultimately shed light on what is certain to be the multi-
factorial etiology of this disorder. It may be that tests now considered to be of
“unproven value” will turn out to be crucial in the evaluation, but this cannot be
determined until more rigorous data collection is completed.
REFERENCES
1. Harris SL, Handleman JS, Gordon R, Kristoff B, Fuentes F. Changes in cognitive
and language functioning of preschool children with autism. J Autism Dev Disord
1991; 21:281–290.
2. Harris SL, Handleman JS. Age and IQ at intake as predictors of placement for
young children with autism: a four- to six-year follow-up. J Autism Dev Disord
2000; 30:137–142.
3. McEachin JJ, Smith T, Lovaas OI. Long-term outcome for children with autism
who received early intensive behavioral treatment. Am J Ment Retard 1993; 97:
359–372; discussion 373–391.
4. Ozonoff S, Cathcart K. Effectiveness of a home program intervention for young
children with autism. J Autism Dev Disord 1998; 28:25–32.
5. Rapin I. Autism [comments]. N Engl J Med 1997; 337:97–104.
6. Rogers SJ. Brief report: early intervention in autism. J Autism Dev Disord 1996;
26:243–246.
7. Rogers SJ. Empirically supported comprehensive treatments for young children
with autism [comments]. J Clin Child Psychol 1998; 27:168–179.
8. Sheinkopf SJ, Siegel B. Home-based behavioral treatment of young children with
autism [comments]. J Autism Dev Disord 1998; 28:15–23.
9. Geschwind DH, Cummings JL, Hollander E, et al. Autism screening and diagnos-
tic evaluation: CAN consensus statement. CNS Spectrums 1998; 3:40–49.
10. Volkmar F, Cook EH Jr, Pomeroy J, Realmuto G, Tanguay P. American Academy
of Child and Adolescent Psychiatry Working Group on Quality Issues. Practice
parameters for the assessment and treatment of children, adolescents, and adults
with autism and other pervasive developmental disorders [erratum appears in J
Am Acad Child Adolesc Psychiatry 2000; 39(7):938]. J Am Acad Child Adolesc
Psychiatry 1999; 38:32S–54S.
11. Filipek PA, Accardo PJ, Baranek GT, et al. The screening and diagnosis of autistic
spectrum disorders [published erratum appears in J Autism Dev Disord 2000;
30(1):81]. J Autism Dev Disord 1999; 29(6):439–484.
12. Filipek PA, Accardo PJ, Ashwal S, et al. Practice parameter: screening and diagno-
sis of autism: Report of the Quality Standards Subcommittee of the American
Academy of Neurology and the Child Neurology Society. Neurology 2000; 55:
468–479.
52 Spence and Geschwind
13. Bricker D, Squires J. The ages and stages questionnaires. Baltimore: Paul H.
Brookes, 1999.
14. Brigance A. The BRIGANCE-R screens. North Billerica, MA: Curriculum Associ-
ates, 1986.
15. Ireton H. Child development inventories. Minneapolis: Behavior Science Systems,
1992.
16. Glascoe HP. Collaborating with parents: using parents’ evaluation of develop-
mental status to detect and address developmental and behavioral problems. Nash-
ville: Ellsworth & Vandermeer, 1998.
17. Glascoe FP. Using parents’ concerns to detect and address developmental and
behavioral problems. J Soc Pediatr Nurs 1999; 4:24–35.
18. Glascoe FP. Parents’ concerns about children’s development: prescreening tech-
nique or screening test? [comments]. Pediatrics 1997; 99:522–528.
19. Glascoe FP, Dworkin PH. The role of parents in the detection of developmental
and behavioral problems. Pediatrics 1995; 95:829–836.
20. Glascoe FP. It’s not what it seems: the relationship between parents’ concerns and
children with global delays. Clin Pediatr (Phila) 1994; 33:292–296.
21. Stone WL, Lee EB, Ashford L, et al. Can autism be diagnosed accurately in chil-
dren under 3 years? J Child Psychol Psychiatry 1999; 40:219–226.
22. Lord C. Follow-up of two-year-olds referred for possible autism. J Child Psychol
Psychiatry 1995; 36:1365–1382.
23. Cox A, Klein K, Charman T, et al. Autism spectrum disorders at 20 and 42 months
of age: stability of clinical and ADI-R diagnosis. J Child Psychol Psychiatry 1999;
40:719–732.
24. Charman T, Swettenham J, Baron-Cohen S, Cox A, Baird G, Drew A. Infants
with autism: an investigation of empathy, pretend play, joint attention, and imita-
tion. Dev Psychol 1997; 33:781–789.
25. Charman T, Swettenham J, Baron-Cohen S, Cox A, Baird G, Drew A. An experi-
mental investigation of social-cognitive abilities in infants with autism: clinical
implications. Infant Ment Health J 1998; 19:260–275.
26. Mars AE, Mauk JE, Dowrick PW. Symptoms of pervasive developmental disor-
ders as observed in prediagnostic home videos of infants and toddlers. J Pediatr
1998; 132:500–504.
27. Osterling J, Dawson G. Early recognition of children with autism: a study of first
birthday home videotapes. J Autism Dev Disord 1994; 24:247–257.
28. Baron-Cohen S, Allen J, Gillberg C. Can autism be detected at 18 months? The
needle, the haystack, and the CHAT. Br J Psychiatry 1992; 161:839–843.
29. Baron-Cohen S, Cox A, Baird G, et al. Psychological markers in the detection
of autism in infancy in a large population. Br J Psychiatry 1996; 168:158–
163.
30. Baird G, Charman T, Baron-Cohen S, et al. A screening instrument for autism at
18 months of age: a 6-year follow-up study. J Am Acad Child Adolesc Psychiatry
2000; 39:694–702.
31. Berument SK, Rutter M, Lord C, Pickles A, Bailey A. Autism screening question-
naire: diagnostic validity. Br J Psychiatry 1999; 175:444–451.
32. Garnett MS, Attwood AJ. The Australian Scale for Asperger’s Syndrome. In: Att-
Screening and Neurodevelopment Assessment 53
wood T, ed. Asperger’s Syndrome: A Guide for Parents and Professionals. Lon-
don: Kingsley, 1998:17–19.
33. Siegel B. Early screening and diagnosis in autistic spectrum disorders: the Perva-
sive Developmental Disorders Screening Test (PDDST). NIH State of the Science
in Autism: Screening and Diagnosis Working Conference, Bethesda, MD, June
15–17, 1998.
34. Gillberg C, Ehlers S, Schaumann H, et al. Autism under age 3 years: a clinical
study of 28 cases referred for autistic symptoms in infancy [comments]. J Child
Psychol Psychiatry 1990; 31:921–934.
35. Dahlgren SO, Gillberg C. Symptoms in the first two years of life: a preliminary
population study of infantile autism. Eur Arch Psychiatry Neurol Sci 1989; 238:
169–174.
36. Gillberg C, Coleman M. Diagnosis in Infancy: The Biology of the Autistic Syn-
dromes. Vol 153/4. London: MacKeith Press, 2000.
37. Stone WL, Hogan KL. A structured parent interview for identifying young children
with autism. J Autism Dev Disord 1993; 23:639–652.
38. Stone WL. STAT manual: Screening Tool for Autism in Two-Year-Olds. NIH
State of the Science in Autism: Screening and Diagnosis Working Conference,
Bethesda, MD, June 15–17, 1998.
39. Stone WL. Descriptive information about the Screening Tool for Autism in Two-
Year-Olds (STAT). NIH State of the Science in Autism: Screening and Diagnosis
Working Conference, Bethesda, MD, June 15–17, 1998.
40. Stone WL, Ousley OY, Hepburn SL, Hogan KL, Brown CS. Patterns of adaptive
behavior in very young children with autism. Am J Ment Retard 1999; 104:187–199.
41. Stapells DR, Oates P. Estimation of the pure-tone audiogram by the auditory brain-
stem response: a review. Audiol Neurootol 1997; 2:257–280.
42. Grewe TS, Danhauer JL, Danhauer KJ, Thornton AR. Clinical use of otoacoustic
emissions in children with autism. Int J Pediatr Otorhinolaryngol 1994; 30:123–
132.
43. Cohen DJ, Johnson WT, Caparulo BK. Pica and elevated blood lead level in autis-
tic and atypical children. Am J Dis Child 1976; 130:47–48.
44. Shannon M, Graef JW. Lead intoxication in children with pervasive developmental
disorders. J Toxicol Clin Toxicol 1996; 34:177–181.
45. Rutter M, Bailey A, Bolton P, Le Couteur A. Autism and known medical condi-
tions: myth and substance. J Child Psychol Psychiatry 1994; 35:311–322.
46. Gillberg C, Coleman M. Autism and medical disorders: a review of the literature.
Dev Med Child Neurol 1996; 38:191–202.
47. Rutter M, Bailey A, Simonoff E, Pickles A. Genetic influences and autism. In:
Cohen DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental
Disorders. 2nd ed. New York: Wiley, 1997:370–387.
48. Simonoff E. Genetic counseling in autism and pervasive developmental disorders.
J Autism Dev Disord 1998; 28:447–456.
49. Szatmari P, Jones MB, Zwaigenbaum L, MacLean JE. Genetics of autism: over-
view and new directions. J Autism Dev Disord 1998; 28:351–368.
50. Bolton PF, Pickles A, Murphy M, Rutter M. Autism, affective and other psychiat-
ric disorders: patterns of familial aggregation. Psychol Med 1998; 28:385–395.
54 Spence and Geschwind
51. DeLong R, Nohria C. Psychiatric family history and neurological disease in autis-
tic spectrum disorders. Dev Med Child Neurol 1994; 36:441–448.
52. Piven J, Chase GA, Landa R, et al. Psychiatric disorders in the parents of autistic
individuals [comments]. J Am Acad Child Adolesc Psychiatry 1991; 30:471–478.
53. Piven J, Palmer P. Psychiatric disorder and the broad autism phenotype: evidence
from a family study of multiple-incidence autism families. Am J Psychiatry 1999;
156:557–563.
54. Piven J, Gayle J, Chase GA, et al. A family history study of neuropsychiatric
disorders in the adult siblings of autistic individuals [comments]. J Am Acad Child
Adolesc Psychiatry 1990; 29:177–183.
55. 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.
56. Hashimoto O, Shimizu Y, Kawasaki Y. Brief report: low frequency of the fragile
X syndrome among Japanese autistic subjects. J Autism Dev Disord 1993; 23:
201–209.
57. Bailey A, Bolton P, Butler L, et al. Prevalence of the fragile X anomaly amongst
autistic twins and singletons. J Child Psychol Psychiatry 1993; 34:673–688.
58. Geschwind DH, Sowinski J, Jones P, et al. and the AGRE Scientific Steering Com-
mittee. The Autism Genetic Resource Exchange (AGRE): a resource for the study
of autism and related neuropsychiatric conditions. Am J Hum Genet 2001; 69:
463–466.
59. Hallmayer J, Pintado E, Lotspeich L, et al. Molecular analysis and test of linkage
between the FMR-1 gene and infantile autism in multiplex families. Am J Hum
Genet 1994; 55:951–959.
60. Klauck SM, Munstermann E, Bieber-Martig B, et al. Molecular genetic analysis
of the FMR-1 gene in a large collection of autistic patients. Hum Genet 1997;
100:224–229.
61. Meyer GA, Blum NJ, Hitchcock W, Fortina P. Absence of the fragile X CGG
trinucleotide repeat expansion in girls diagnosed with a pervasive developmental
disorder. J Pediatr 1998; 133:363–365.
62. Baker P, Piven J, Sato Y. Autism and tuberous sclerosis complex: prevalence and
clinical features. J Autism Dev Disord 1998; 28:279–285.
63. Hunt A, Shepherd C. A prevalence study of autism in tuberous sclerosis. J Autism
Dev Disord 1993; 23:323–339.
64. Gillberg IC, Gillberg C, Ahlsen G. Autistic behaviour and attention deficits in tuber-
ous sclerosis: a population-based study. Dev Med Child Neurol 1994; 36:50–56.
65. Smalley SL, Tanguay PE, Smith M, Gutierrez G. Autism and tuberous sclerosis.
J Autism Dev Disord 1992; 22:339–355.
66. Gutierrez GC, Smalley SL, Tanguay PE. Autism in tuberous sclerosis complex.
J Autism Dev Disord 1998; 28:97–103.
67. Smalley SL. Autism and tuberous sclerosis. J Autism Dev Disord 1998; 28:407–414.
68. Lainhart JE, Piven J, Wzorek M, et al. Macrocephaly in children and adults with
autism. J Am Acad Child Adolesc Psychiatry 1997; 36:282–290.
69. Filipek PA. The autistic spectrum disorders. In: Swaiman KF, Ashwal S, eds. Pedi-
atric Neurology: Principles and Practice. St. Louis: Mosby, 1999:606–628.
Screening and Neurodevelopment Assessment 55
70. Rapin I. Neurological examination. In: Rapin I, ed. Preschool Children with Inade-
quate Communication: Developmental Language Disorder, Autism, Low IQ. Lon-
don: MacKeith, 1996:98–122.
71. Kientz MA, Dunn W. A comparison of the performance of children with and with-
out autism on the Sensory Profile. Am J Occup Ther 1997; 51:530–537.
72. Barrett S, Beck JC, Bernier R, et al. An autosomal genomic screen for autism:
collaborative linkage study of autism. Am J Med Genet 1999; 88:609–615.
73. Risch N, Spiker D, Lotspeich L, et al. A genomic screen of autism: evidence for
a multilocus etiology. Am J Hum Genet 1999; 65:493–507.
74. Philippe A, Martinez M, Guilloud-Bataille M, et al. Genome-wide scan for autism
susceptibility genes: Paris Autism Research International Sibpair Study [erratum
appears in Hum Mol Genet 1999; 8(7):1353]. Hum Mol Genet 1999; 8(5):805–812.
75. IMGSAC. A full genome screen for autism with evidence for linkage to a region
on chromosome 7q. International Molecular Genetic Study of Autism Consortium.
Hum Mol Genet 1998; 7:571–578.
76. Rutter M. Genetic studies of autism: from the 1970s into the millennium. J Abnorm
Child Psychol 2000; 28:3–14.
77. Lamb JA, Moore J, Bailey A, Monaco AP. Autism: Recent molecular genetic
advances [erratum appears in Hum Mol Genet 2000; 9(9):1461]. Hum Mol Genet
2000; 9(6):861–868.
78. Gillberg C. Chromosomal disorders and autism. J Autism Dev Disord 1998; 28:
415–425.
79. Cook EH Jr, Lindgren V, Leventhal BL, et al. Autism or atypical autism in mater-
nally but not paternally derived proximal 15q duplication. Am J Hum Genet 1997;
60:928–934.
80. Volkmar FR, Nelson DS. Seizure disorders in autism. J Am Acad Child Adolesc
Psychiatry 1990; 29:127–129.
81. Tuchman RF, Rapin I, Shinnar S. Autistic and dysphasic children. II. Epilepsy
[erratum appears in Pediatrics 1992; 90(2 Pt 1):264]. Pediatrics 1991; 88(6):1219–
1225.
82. Minshew N, Sweeney JA, Bauman ML. Neurologic aspects of autism. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:344–369.
83. Olsson I, Steffenburg S, Gillberg C. Epilepsy in autism and autisticlike conditions:
a population-based study. Arch Neurol 1988; 45:666–668.
84. Tuchman RF, Rapin I. Regression in pervasive developmental disorders: seizures
and epileptiform electroencephalogram correlates. Pediatrics 1997; 99:560–566.
85. Landau WM, Kleffner F. Syndrome of acquired aphasia with convulsive disorder
in children. Neurology 1957; 7:523–530.
86. Landau WM, Kleffner FR. Syndrome of acquired aphasia with convulsive disorder
in children [1957—classical article]. Neurology 1998; 51:1241, 8 pp following
1241.
87. Deonna TW. Acquired epileptiform aphasia in children (Landau-Kleffner syn-
drome). J Clin Neurophysiol 1991; 8:288–298.
88. Lerman P, Kivity S. The benign partial nonrolandic epilepsies. J Clin Neurophysiol
1991; 8:275–287.
56 Spence and Geschwind
89. Mikati MA, Saab R. Successful use of intravenous immunoglobulin as initial mo-
notherapy in Landau-Kleffner syndrome. Epilepsia 2000; 41:880–886.
90. Morrell F, Whisler WW, Smith MC, et al. Landau-Kleffner syndrome: treatment
with subpial intracortical transection. Brain 1995; 118:1529–1546.
91. Tuchman R. Treatment of seizure disorders and EEG abnormalities in children
with autism spectrum disorders. J Autism Dev Disord 2000; 30:485–489.
92. Kanner AM. Commentary: the treatment of seizure disorders and EEG abnormali-
ties in children with autistic spectrum disorders—are we getting ahead of our-
selves? J Autism Dev Disord 2000; 30:491–495.
93. Mantovani JF. Autistic regression and Landau-Kleffner syndrome: progress or
confusion? Dev Med Child Neurol 2000; 42:349–353.
94. Nass R, Devinsky O. Autistic regression with rolandic spikes. Neuropsychiatry
Neuropsychol Behav Neurol 1999; 12:193–197.
95. Prasad AN, Stafstrom CF, Holmes GL. Alternative epilepsy therapies: the keto-
genic diet, immunoglobulins, and steroids. Epilepsia 1996; 37:S81–95.
96. Filipek PA. Brief report: neuroimaging in autism—the state of the science 1995.
J Autism Dev Disord 1996; 26:211–215.
97. Filipek PA. Neuroimaging in the developmental disorders: the state of the science.
J Child Psychol Psychiatry Allied Disc 1999; 40:113–128.
98. Rumsey JM, Ernst M. Functional neuroimaging of autistic disorders. Ment Retard
Dev Disabil Res Rev 2000; 6:171–179.
99. Page T. Metabolic approaches to the treatment of autism spectrum disorders. J
Autism Dev Disord 2000; 30:463–469.
100. Gillberg IC, Gillberg C, Kopp S. Hypothyroidism and autism spectrum disorders.
J Child Psychol Psychiatry 1992; 33:531–542.
101. Page T, Coleman M. Purine metabolism abnormalities in a hyperuricosuric sub-
class of autism. Biochim Biophys Acta 2000; 1500:291–296.
102. Konstantareas MM, Homatidis S. Ear infections in autistic and normal children.
J Autism Dev Disord 1987; 17:585–594.
103. Comi AM, Zimmerman AW, Frye VH, Law PA, Peeden JN. Familial clustering
of autoimmune disorders and evaluation of medical risk factors in autism. J Child
Neurol 1999; 14:388–394.
104. van Gent T, Heijnen CJ, Treffers PD. Autism and the immune system. J Child
Psychol Psychiatry 1997; 38:337–349.
105. Warren RP, Singh VK, Averett RE, et al. Immunogenetic studies in autism and
related disorders. Mol Chem Neuropathol 1996; 28:77–81.
106. D’Eufemia P, Celli M, Finocchiaro R, et al. Abnormal intestinal permeability in
children with autism. Acta Paediatr 1996; 85:1076–1079.
107. Horvath K, Papadimitriou JC, Rabsztyn A, Drachenberg C, Tildon JT. Gastrointes-
tinal abnormalities in children with autistic disorder [comments]. J Pediatr 1999;
135:559–563.
108. Quigley EM, Hurley D. Autism and the gastrointestinal tract [comment; editorial].
Am J Gastroenterol 2000; 95:2154–2156.
109. Wakefield AJ, Murch SH, Anthony A, et al. Ileal-lymphoid-nodular hyperplasia,
non-specific colitis, and pervasive developmental disorder in children [comments].
Lancet 1998; 351:637–641.
4
Assessment and Early Identification of
Autism Spectrum and Other Disorders
of Relating and Communicating
observable and known. What is observable and known are the functional develop-
mental limitations in critical areas, including emotional and social functioning,
language, motor planning and sequencing, and sensory processing and modula-
tion. In fact, as most clinicians recognize, there are huge differences among chil-
dren with autism: one may be relatively strong in visual-spatial processing and
another in auditory-verbal memory. In terms of motor planning or visual-spatial
processing, a child with autism may be more similar to one with Down’s syn-
drome than to another child with autism. A functional developmental approach
enables us to study all the relevant problems in their unique configurations and,
in this way, improve assessments and intervention for a range of developmental
disorders, including autism.
In the functional approach, assessments and interventions must include all
relevant areas of developmental functioning and deal with each child and family
in terms of their unique profiles. We have developed a model that identifies the
relevant areas of functioning, helps with the construction of each child’s func-
tional developmental profile, and provides a developmental framework for the
assessment and intervention process: the Developmental, Individual-Difference,
Relationship-Based (DIR) model (1,2). In this chapter, we describe the DIR
model and discuss its application to assessment, intervention, classification, and
prognosis.
The DIR model looks at all of the child’s developmental capacities in the
context of his unique, biologically based processing profile and his family rela-
tionships and interactive patterns. As a functional approach, it uses the complex
interactions between biology and experience to understand behavior. Using this
model requires the assessment of each infant’s or child’s relative strengths as
well as challenges as they simultaneously impact on functioning at each develop-
mental level. This contrasts with the typical deficit model, which describes the
deficits within each developmental domain—fine motor, gross motor, speech and
language, social, and other skills are evaluated independently by teams using
standardized instruments, often working in single-session arena style. In the defi-
cit model, results are typically presented from the point of view of deficits in
each domain with general recommendations. Crucial areas of interactive relation-
ships and emotional functioning are often omitted.
The model described below is distinguished in its emphasis on multiple
observations and in-depth interviews over time both in the natural environments
and in child-centered settings. In the developmental functional approach, the eval-
uations go beyond the assessment of skills to the assessment of functioning within
relationships. Standardized tools are used only for strategic purposes rather than
for the core assessment. The evaluation always includes multiple observations
of infant/child–parent (all significant caregivers) interactions and play, as well
as interaction with the evaluator whose relationship with the family effects the
evaluation, interpretation, and implementation of the intervention plan.
Assessment and Early Identification 59
The theoretical, clinical, and empirical rationale for these developmental levels
is discussed in The Development of the Ego (3) and Intelligence and Adapta-
tion (4).
To make a developmental assessment of the functional level, the clinician
should make a determination regarding each of the six developmental levels in
terms of whether they have been successfully negotiated, and whether there is a
deficit at any level that has not been successfully negotiated. Sometimes these
levels have been successfully negotiated but are not applied to the full range of
emotional themes. For example, a toddler may use two-way gestural communica-
tion to negotiate assertiveness and exploration by, for example, pointing at a
certain toy and vocalizing for his parent to play with him. The same child may
either withdraw or cry in a disorganized way when he wishes for increased close-
ness and dependency instead of, for example, reaching out to be picked up or
coming over and initiating a cuddle. This would indicate a constriction at that
level.
Sometimes children are able to negotiate a level with one parent and not
the other, with one sibling and not another, or with one substitute caregiver but
not another. If it should reasonably be expected that a particular relationship
is secure and stable enough to support a certain developmental level but that
level is not evident in that relationship, then there is a constriction at that level
as well.
It is useful to indicate which areas or relationships are not incorporated
into the developmental level. Consider the following areas of expected emotional
range: dependency (closeness, pleasure, assertiveness [exploration], curiosity,
anger, empathy [for children over 31/2 years]); stable forms of love, self-limit-
setting (for children over 18 months); interest and collaboration with peers (for
children over 2 years); participation in a peer group (for children over 21/2 years);
and the ability to deal with competition and rivalry (for children over 31/2 years).
If the child has reached a developmental level but the slightest stress, such
as being tired, having a mild illness (e.g., a cold), or playing with a new peer,
leads to a loss of that level, then there is an instability at that level.
A child may have a defect, constriction, or instability at more than one
level. Also, a child may have a defect at one level and a constriction or instability
at another. Therefore, the clinician should make a “developmental” judgment
based on the following developmental levels. The Functional Emotional Develop-
Assessment and Early Identification 61
mental Scale clinical and research applications have been developed and include
reliability and validity studies (5) (Table 1).
Individual Processing Differences: Constitutional-
Maturational Patterns
In addition to the functional developmental levels, the DIR model focuses on
constitutional-maturational characteristics. Constitutional patterns are the result
of genetic, prenatal, perinatal, and maturational variations and/or deficits. They
are expressed in and can be observed as part of the following individual pro-
cessing differences:
Sensory reactivity, including hypo- and hyperreactivity in each sensory mo-
dality (tactile, auditory, visual, vestibular, olfactory)
Sensory processing in each sensory modality (e.g., the capacity to decode
sequences, configurations, or abstract patterns), including auditory pro-
cessing and visual-spatial processing
Sensory affective processing in each modality (e.g., the ability to process
and modulate affects and connect affective “intent” to motor planning
and sequencing, symbol formation, and other sensory processing capaci-
ties) (6)
Muscle tone
Motor planning and sequencing
An instrument to clinically assess aspects of sensory functions in a reliable
manner has been developed and is available (7–9). The following section further
considers the constitutional and maturational patterns.
Sensory reactivity (hypo- or hyper-) and sensory processing can be ob-
served clinically. Is the child hyper- or hyposensitive to touch or sound? The
62 Greenspan and Wieder
same question must be asked in terms of vision and movement in space. In each
sensory modality, does the 4-month-old “process” a complicated pattern of infor-
mation input or only a simple one? Does the 41/2-year-old have a receptive lan-
guage (i.e., auditory processing) problem and is therefore unable to sequence
words he hears together or follow complex directions? Is the 3-year-old an early
comprehender and talker but slower in visual-spatial processing? If spatial pat-
terns are poorly comprehended, a child may be facile with words and sensitive
to every emotional nuance, but have no context, never see the big picture (the
“forest”); such children get lost in the “trees.” In the clinician’s office, they may
forget where the door is or have a hard time picturing that their mother is only
a few feet away in the waiting room. In addition to straightforward “pictures”
of spatial relationship (e.g., how to get to the playground), they may also have
difficulty with seeing the emotional big picture. If the mother is angry, the child
may think that the earth is opening up and he is falling in because he cannot
comprehend that she was nice before and will probably be nice again. Such a
child may be strong on the auditory processing side but weak on the visual-spatial
processing side. Our impression is that children with a lag in the visual-spatial
area can become overwhelmed by the affect of the moment. This is often intensi-
fied by precocious auditory-verbal skills. The child, in a sense, overloads himself
and does not have the ability to see how it all fits together.
Thus, at a minimum, it is necessary to have a sense of how the child reacts
in each sensory modality, how he or she processes information in each modality,
and particularly, as the child gets older, a sense of the auditory-verbal processing
skills in comparison to visual-spatial processing skills.
It is also necessary to look at the motor system, including muscle tone, motor
planning (fine and gross), and postural control. Observing how a child sits, crawls,
or runs; maintains posture; holds a crayon; hops, scribbles, or draws; and makes
rapid alternating movements and plans and executes complex actions (such as using
a toy in an innovative four-step manner) will provide a picture of the child’s motor
system. His security in regulating and controlling his body plays an important role
in how he uses gestures to communicate his ability to regulate dependency (being
close or far away), his confidence in regulating aggression (“Can I control my hand
that wants to hit?”), and his overall physical sense of self.
Other constitutional and maturational variables have to do with movement
in space, attention, and dealing with transitions.
Parent and Family Contributions
In addition to the functional developmental levels and individual processing dif-
ferences based on constitutional and maturational factors, it is important to de-
scribe family patterns. It is especially useful to describe family interactions with
regard to each functional developmental level. If a family system is aloof, it may
not negotiate engagement well; if a family system is intrusive, it may overwhelm
Assessment and Early Identification 63
Often, parents will bring a baby in, and we will watch the parent–infant
interaction in the first session, while we are hearing about their concerns. With
an older child—a 4-year-old, for example—we may have them wait to bring the
child in, because we want them to talk freely at the outset.
Usually, we let each parent interact and play with the child for 20 minutes
or more, and then we will interact with the child for about the same amount of
time. We look for a pattern of interaction in the context of the developmental
processes. We observe which levels are present and not present, and also look
for the range of emotional themes in each of the core processes and the stability
of these processes. For example, a 4-year-old may be self-absorbed and only
occasionally purposeful, pulling mom to the door, for example. We look at how
the child is relating with his caregiver (and later on with us), and the breadth of
emotion and the stability of the relatedness. We observe the degree to which the
child can interact with purposeful gestures, problem-solve with a continuous flow
of interactive gestures, use ideas, and think logically. We also observe the child’s
processing capacities (does he understand mom’s verbal comments?) and the
natural interactive patterns his parents employ.
For each of the developmental levels, or core processes, it is necessary to
look at the child’s constitutional and maturational status, the family and parent
and couple patterns, and the actual caregiver/parent–infant interaction (see Figure
1). For each level, one must look at what is influencing the successful or compro-
mised negotiation at that level. Therefore, a therapist wants to be able to reach
a conclusion on all levels about a child 3 years old and up, and for the child less
than 3 years, on the levels they should have attained (e.g., for a 21/2-year-old,
through the first three levels; for a 14-month-old, the first two levels: attention/
engagement and two-way communication).
It is also often necessary to conduct evaluation of speech and language
functions, motor and sensory capacities, and educational skills, and conduct a
thorough biomedical evaluation (see Table 2).
The evaluations listed in V–IX should be carried out only to answer specific questions that arise
from the history, review of challenges, and current functioning and observations of the child–care-
giver interaction patterns and family functioning.
66 Greenspan and Wieder
Developmental History
In the second session, we construct a developmental history for the child. (How-
ever, sometimes marital or other family problems erupt during the first session.
Assessment and Early Identification 67
The parents may be at each other’s throats; the mother and/or the father may be
extremely depressed. In such cases, in the second session we focus on the individ-
ual parent problems, as well as family functioning.)
We will usually start the session in an unstructured manner. We want to
hear how development unfolded and what the parents thought was important.
We encourage them to alternate between what the baby or child was like at differ-
ent stages and what they felt was going on as a family and as individuals in each
of those stages. We try to start with the planning for the child and progress
through the pregnancy and delivery. Next, we cover the six developmental stages,
outlined earlier, in order to organize the developmental history.
Family Patterns
The next session focuses in greater depth on the functioning of the caregiver and
family at each developmental phase. For example, the mother may say that she
was a little depressed or angry, or that there were marital problems at different
stages in the child’s development.
Sometimes clinicians who are only beginning to work with infants and
family feel reluctant to talk to the parents about any difficulties in the marriage.
However, an open and supportive approach can elicit relevant information. One
might ask, “What can you tell me about yourselves as people, as a married couple,
as a family?” We are also interested in concrete details of a history of mental
illness, learning disabilities, or special developmental patterns in either of the
parent’s families.
more challenging. Later, we will join them do some coaching and/or start to play
with the child.
During this time, we want to see the child interacting at his or her highest
developmental level, as well as how he relates to a new person whom he knows
only slightly. In addition, we want to determine how to bring out the highest
developmental level at which the child can function. For example, if a child is
withdrawn or self-absorbed and repetitively moving a truck, we will suggest join-
ing his play, trying to move the truck together or put up a fence (one’s hands),
or take another car and, with great joy and enthusiasm, announce “Here I come!”
to entice the child into interaction. Sometimes marching or jumping with an aim-
less child or lying next to a passive, withdrawn child and offering a backrub or
tickle will draw him in. If a child shows signs of symbolic functioning and the
parents do not support symbolic functioning (e.g., in their 3-year-old), we will
try, through coaching or directing, to get pretend play going.
We observe the way the child relates to us, that is, the quality of engage-
ment—overly familiar, overly cautious, or warm. We look for how intentional
he is in the use of gestures and how well he sizes up the situation and us (without
words). We try to determine his emotional range and his way of dealing with
anxiety (e.g., does he become aggressive or withdrawn?). During interaction with
a child, we also note his physical status, speech, receptive language, visuospatial
problem-solving skills (e.g., whether he can search for a toy), gross and fine
motor skills, and general state of health and mood.
In general, we want to systematically describe the child’s ability to attend,
engage, initiate and be purposeful with affects and motor gestures, open and close
many communication circles to solve problems, create ideas (pretend-play), and
converse and think logically.
The next step is to learn what is on the child’s mind. If the child is symbolic,
one looks at the content of the play and dialog, as well as the sequence of themes
that emerge from them. Often, observing how a child shifts from one activity or
theme to another (e.g., separation to perseveration and self-stimulation, aggres-
sion to protectiveness, or exploration to repetition) will provide some initial
hypotheses. The therapist’s job is to be reasonably warm, supportive, and skillful
in engaging the child and helping him evidence his capacities and elaborate.
Formulation
After learning about the child’s current functioning and history and observing
the child and family first-hand, there should be a convergence of impressions. If
a picture is not emerging, one may need to spend another session or two devel-
oping the history or observing further.
One then asks oneself a number of questions. How high up in the develop-
mental progression has the child gone, in terms of 1) attending, 2) regulating and
Assessment and Early Identification 69
EARLY IDENTIFICATION
The DIR model of assessment and intervention planning also lends itself to the
early identification of autism spectrum and other developmental disorders.
72 Greenspan and Wieder
these doors provides an important picture of his adaptive and maladaptive devel-
opment and the need for further evaluation and, possibly, intervention.
If observing and/or asking about a child’s functional developmental capaci-
ties raises questions about appropriate progress, a second level of monitoring
should be considered. This should involve screening questionnaires that have
been used with a large number of children and shown to identify different types
of developmental problems. Screening questionnaires that cover a broad range of
developmental competence include the Communication and Symbolic Behavior
Scales Developmental Profile (45) and the Ages and Stages Questionnaire (ASQ):
A Parent-Completed, Child-Monitoring System, Second Edition (46).
If a systematic screening questionnaire supports the impression from clini-
cal observations and questions, then a third level should be considered: a compre-
hensive developmental evaluation to determine the nature and extent of a sus-
pected problem.
In order to broaden our frame of reference and implement the first level
of monitoring of an infant or child’s progress, it may prove helpful to have a
functional developmental growth chart and questionnaire that identify the mile-
stones to be observed or asked about. The growth chart and questionnaire must
provide straightforward, clear descriptions that can readily be observed by par-
ents, health-care providers, and educators. It must cover all the areas of develop-
mental functioning—emotional, social, cognitive, language, motor, and sen-
sory—and all the stages of infancy and early childhood.
Figure 2 presents a functional developmental growth chart (followed by
the functional developmental questionnaire) similar to a physical growth chart.
The developmental growth chart enables clinicians to look at the pattern of a
child’s growth, rather than simply at a few items at a certain age. Patterns of
change over time often provide the most useful information about a child’s abili-
ties.
In Figure 2, the functional developmental capacities to be monitored are
listed on the horizontal axis. The child’s age is on the vertical axis. A 45-degree
line shows the expected age range at which a child is expected to master each
capacity. As can be observed, a child’s functional developmental accomplish-
ments can be charted in relation to the age at which the accomplishment is ex-
pected to emerge and the age at which it does emerge. When a child does not
evidence the next milestone during the expected time interval, the last functional
capacity mastered is recorded on the chart. The next milestone, if it occurs, is
then recorded at whatever later time it is manifested. The 45-degree line indicates
a typical developmental curve. A child who is precocious in a predictable manner
(e.g., 3 months ahead of expectations) will have a functional developmental curve
that parallels the typical one and is a little above it. A child who is a little behind
the expected curve (e.g., 3 months behind on the functional developmental mile-
stones) will have a curve that parallels the typical curve but may fall just below
Assessment and Early Identification 75
it. When a child’s curve is below the norm, the child should be evaluated to
identify the factors that may be contributing to the developmental lag and what
may be a helpful response.
Most worrisome, and a red flag, is a curve that arcs away from the line;
that is, the distance from the line keeps growing, indicating a delay that is increas-
ing as the child becomes older (shown as the lowest line on the chart). The point
at which the curve begins arcing is where immediate assessment and possible
intervention are indicated. It is also a red flag if the developmental curve is run-
ning parallel to the typical curve but is significantly below it.
This developmental chart can be used by parents, educators (including day-
care staff), and other child-care facilitators to monitor a child’s functional capaci-
ties. In general, a child will have mastered a milestone when she can, most of
the time, engage in the behavior associated with the milestone. Mastery is not
indicated in a child who is only occasionally able to mobilize the age-appropriate
milestone or requires extraordinary support to perform it. The Functional Devel-
opmental Growth Chart Questionnaire following the chart will help in determin-
ing the child’s functional developmental level.
tory, with little or no language, intermittent relating, and a very narrow range
of intermittent, purposeful, and problem-solving gestural sequences, and mixed
reactivity to sensations. Very gradually, a type II child expands purposeful and
problem-solving gestural interactions as well as imitative abilities. This type of
child very slowly acquires the use of words and pretend-play and, with a compre-
hensive program, makes very gradual but consistent progress, tending to remain
at a fragmented level of language capacity (i.e., using ideas but having a very
difficult time connecting ideas in a logical sequence). Receptive language is often
stronger than expressive language. Type II children are interested in peers but
have difficulty developing fully interactive communication.
Processing profile: type II is characterized by moderate motor planning
dysfunction (three- to four-step sequences) and more significant sensory pro-
cessing problems. This type has two subtypes.
Type IIA: Hypersensitive to sensation.
Type IIB: Underreactive to sensation.
Type III (Partially Purposeful and Engaged Type)
Very slow progress—severe processing dysfunction; self-absorbed and intermit-
tently engaged. Self-absorbed or avoidant, type III children evidence only inter-
mittent use of simple purposeful gestures (i.e., an “in-and-out” quality) with no
language initially. They can very slowly become more purposeful and engaged
and evidence intermittent problem-solving, gestural sequences. Eventually, iso-
lated words and intermittent use of phrases are possible.
Processing profile: type III is characterized by severe motor planning dys-
function (one- to two-step sequences), weaker auditory and visual-spatial pro-
cessing than type II, and more extreme sensory modulation difficulties (usually
underreactivity, but can be mixed).
Type IV (Aimless, Unpurposeful Type)
Very slow progress with no development of expressive language—very severe
processing dysfunction, aimless, and nonverbal. Very self-absorbed or avoidant,
type IV children are only intermittently and partially engaged, with only fleeting
purposeful interactions. They often evidence extreme motor planning difficulties,
especially oral-motor dysfunctions, making sound production very difficult. Over
time, they can become more purposeful and engaged and eventually use pictures
or other visual aids for intermittent communication, but do not develop consistent
motor planning, problem solving, vocal imitative, or expressive language.
Processing profile: type IV evidences very severe motor planning problems
(aimless to one-step sequences), with very severe oral motor capacity and more
severe auditory, visual-spatial, and sensory modulation problems (usually under-
or mixed reactivity) than in type III.
Assessment and Early Identification 79
Type IVA: May make limited progress and become more related, but
does not progress to full purposeful or problem-solving interactions.
Type IVB: No progress, or vacillations between small gains and losses
of functions. Often evidences more overt neurological symptoms.
Children characterized as type I can often do especially well and develop
levels of empathy and creativity not thought possible in children diagnosed with
autism spectrum disorder. In our review of 200 cases, we found that a group of
children with these characteristics progressed through the stages of relating with
greater and greater warmth and longer and longer sequences of gestural and af-
fective, problem-solving interactions. Over a period of years, they learned to use
language both creatively and logically, eventually enjoying peer relationships
and functioning well in regular educational settings. They are now bright, warm,
empathetic, creative youngsters with a range of interests. When we compared the
20 children from this group who had made the most progress with a matched group
of children with no history of developmental and/or learning problems, we found
that they were indistinguishable on assessments of their emotional capacities and
interactions with parents (as measured through analysis of videotaped interactions)
and in their adaptive behavior (as measured on the Vineland Scales) (30).
The type of progress this group of children can make raises an important
question: do these children have a more transient set of autistic symptoms that
is very responsive to an intensive, dynamic intervention? At present, many chil-
dren with autistic disorders are unable to progress to complex functional interac-
tions and pragmatic language. In our chart review, we observed that children
characterized as type II, while engaging and speaking, were still having signifi-
cant challenges in developing creative and abstract language use, as well as full
peer relationships. Type III and IV children tended to have continuing difficulties
in being fully engaged and purposeful, as well as in problem-solving, and had
limited or no verbal abilities.
CONCLUSION
In this chapter we have presented a developmentally based approach (DIR model)
to assessing children with autism spectrum disorder and other disorders of re-
lating and communicating. We have discussed its implications for intervention
planning, early identification, and the classification of clinically useful subtypes.
ber, the growth chart is simply a visual tool to draw attention to the develop-
mental areas in which a child is progressing as expected and those in which
he or she may be facing some challenges.
with him or her by using words or sequences of sounds that are clearly an
attempt to convey a word?
Is your toddler (by 24 months) able to imitate familiar pretend-like actions, such
as feeding or hugging a doll?
Is your toddler (by 24 months) able to meet some basic needs with one or a few
words, such as “juice,” “open,” or “kiss”? (A parent may have to say the
word first.)
Is your toddler (by 24 months) able to follow simple one-step directions from a
caregiver to meet some basic need, for example, “The toy is there” or
“Come give Mommy a kiss.”
Is your toddler (by 30 months) able to engage in interactive pretend-play with
an adult or another child (feeding dollies, tea parties, etc.)?
Is your toddler (by 30 months) able to use ideas—words or symbols—to share
his or her delight or interest (“See truck!”)?
Is your toddler able to use symbols—words, pictures, organized games—while
enjoying and interacting with one or more peers?
REFERENCES
1. Greenspan SI. Infancy and Early Childhood: The Practice of Clinical Assessment
and Intervention with Emotional and Developmental Challenges. Madison, CT: In-
ternational Universities Press, 1992.
2. Greenspan SI, Wieder S. The Child with Special Needs: Intellectual and Emotional
Growth. Reading, MA: Addison Wesley Longman, 1998.
3. Greenspan SI. The Development of the Ego: Implications for Personality Theory,
Psychopathology, and the Psychotherapeutic Process. Madison, CT: International
Universities Press, 1989.
4. Greenspan SI. Intelligence and Adaptation: An Integration of Psychoanalytic and
Piagetian Developmental Psychology. Psychological Issues, Monograph 47/68.
New York: International Universities Press, 1979.
5. Greenspan S, DeGangi G, Wieder S. The Functional Emotional Assessment Scale
(FEAS) for Infancy and Early Childhood: Clinical and Research Applications.
Bethesda, MD: Interdisciplinary Council on Developmental and Learning Disor-
ders, 2001.
6. Greenspan S. The affect diathesis hypothesis: the role of emotions in core deficit
in autism and the development of intelligence and social skills. J Dev Learning
Disord 2001; 5:1–46.
7. DeGangi G, Greenspan SI. The development of sensory functioning in infants. J
Phys Occup Ther Pediatr 1988; 8(4):21–33.
8. DeGangi G, Greenspan SI. The assessment of sensory functioning in infants. J Phys
Occup Ther Pediatr 1989; 9:21–33.
9. DeGangi G, Greenspan SI. Test of sensory functions in infants. Los Angeles: West-
ern Psychology Services, 1989.
10. Bristol M, Cohen D, Costello J, Denckla M, Eckberg T, Kallen R, Kraemer H, Lord
C, Maurer R, McIllvane W, Minshew N, Sigman M, Spence A. State of the science
in autism: Report to the National Institutes of Health. J Autism Dev Disord 1996;
26:121–154.
11. Rogers S. Brief report: early intervention in autism. J Autism Dev Disord 1996;
26:243–246.
12. Lovaas OI. Behavioral treatment and normal educational and intellectual function-
ing in young autistic children. J Consult Clin Psychol 1987; 55:3–9.
13. McEachin JJ, Smith T, Lovaas OI. Long-term outcome for children with autism
who received early intensive behavioral treatment. Am J Ment Retard 1993; 97:
359–372.
14. Gresham FM, MacMillan DL. Early intervention project: can its claims be substanti-
ated and its effects replicated? J Autism Dev Disord 1998; 28(1):5–12.
15. Greenspan SI. Commentary—guidance for constructing clinical practice guidelines
for developmental and learning disorders: knowledge vs. evidence-based ap-
proaches. J Dev Learn Disord 1998; 2(2):171–192.
Assessment and Early Identification 83
34. Dawson G, Galpert I. Mother’s use of imitative play for facilitating social respon-
siveness and toy play in young autistic children. Dev Psychopathol 1990; 2:151–
162.
35. Tanguay PE, Robertson J, Derrick A. A dimensional classification of autism spec-
trum disorder by social communication domains. J Am Acad Child Adolesc Psychi-
atry 1998; 37(3):271–277.
36. Tanguay PE. The diagnostic assessment of autism using social communication do-
mains. Presented at the Interdisciplinary Council on Developmental and Learning
Disorders’ Third Annual International Conference, Autism and Disorders of Re-
lating and Communicating, McLean, VA, Nov 12–14, 1999.
37. Osterling J, Dawson G. Early recognition of children with autism: a study of first
birthday home videotapes. J Autism Dev Disord 1994; 24(3): 247–257.
38. Baranek GT. Autism during infancy: a retrospective video analysis of sensory-motor
and social behaviors at 9–12 months of age. J Autism Dev Disord 1999; 29(3):
213–224.
39. Wetherby AM, Prizant BM. Profiling communication and symbolic abilities in
young children. J Child Commun Disord 1993; 15:23–32.
40. Teitelbaum P, Teitelbaum O. Motor indicators of autism in the first year. Presented
at the Interdisciplinary Council on Developmental and Learning Disorders’ Third
Annual International Conference on Autism and Disorders of Relating and Commu-
nicating, McLean, VA, Nov 12–14, 1999.
41. Williamson GG, Anzalone ME. Sensory integration: a key component of the evalua-
tion and treatment of young children with severe difficulties in relating and commu-
nicating. ZERO TO THREE 1997; 17:29–36.
42. Baron-Cohen S. The early detection of autism. Presented at the Interdisciplinary
Council on Developmental and Learning Disorders’ Third Annual International
Conference on Autism and Disorders of Relating and Communicating, McLean,
VA, Nov 12–14, 1999.
43. Greenspan SI, Lourie RS. Developmental structuralist approach to the classification
of adaptive and pathologic personality organizations: application to infancy and
early childhood. Am J Psychiatry 1981; 138 (6):725–735.
44. Fillipek P, Accardo P, Baranek G, Cook E, Dawson G, Gordon B, Gravel J, Johnson
C, Kallen R, Levy S, Minshew N, Prizant B, Rapin I, Rogers S, Stone W, Teplin
S, Tuchman R, Volkmar F. The screening and diagnosis of autistic spectrum disor-
ders. J Autism Dev Disord 1999; 29(6):439–484.
45. Wetherby A, Prizant B. Communication and Symbolic Behavior Scales Develop-
mental Profile–Research Edition. Chicago: Applied Symbolix, 1998.
46. Bricker D, Squires J. Ages and Stages Questionnaires: A Parent-Completed, Child-
Monitoring System. 2nd ed. Baltimore: Paul H. Brookes, 1999.
47. Volkmar FR, Schwab-Stone M. Annotation: childhood disorders in DSM-IV. J
Child Psychol Psychiatry 1996; 37(7):779–784.
48. Lord C, Rutter M, Goode S, Heemsbergen J, Jordan H, Mauhood. Autism Diagnos-
tic Observation Schedule: a standardized observation of communicative and social
behavior. J Autism Ment Disord 1989; 19(2):185–212.
49. Tordjman S, Anderson GM, McBride PA, Hertzig ME, Snow ME, Hall LM,
Assessment and Early Identification 85
INTRODUCTION
Three core features characterize autism spectrum disorders (ASDs): impairment
in socialization, impaired communication, and restrictive or repetitive behaviors,
interests, or activities (1). Since early and intensive behavioral intervention is
essential in the successful treatment of these disorders, early diagnosis is particu-
larly crucial. The diagnosis of ASD is a multistep process, beginning with screen-
ing and followed by more comprehensive assessments (2). This full evaluation
is necessary in order to rule out other disorders (such as verbal and nonverbal
learning disabilities or organic brain disorder), and to guide treatment and educa-
tional planning. Full evaluations include, in addition to diagnosis, cognitive as-
sessment, adaptive functioning assessment, and neuropsychological testing (e.g.,
memory, problem solving, planning, language, and visual-spatial reasoning).
Cognitive and neuropsychological evaluations of children with ASD are
useful to pediatricians and child psychiatrists in clarifying the diagnosis by ruling
out developmental learning disorders, disorders of speech and language, and be-
havioral disorders, and by ruling out/in mental retardation. Cognitive and neuro-
psychological evaluations are essential for school personnel in developing Indi-
vidual Education Plans (IEP), and are important in supporting educational
87
88
Table 1 Generally Used Cognitive, Adaptive, and Neuropsychological Tests
Scale Authors, year Assesses Description
executive function
Trail Making Test Reitan RM (1969) Speed for attention, se- Subject draws a line through 25 stimuli in numerical or-
quencing, flexibility, vi- der (part A) and of 25 encircled numbers and letters
sual search and motor in alternating order (part B)
function; child and
adult versions
VIGIL Continuous Per- For Thought, Ltd. Brain damage, in subjects Computerized flash task requiring the execution of one re-
formance Test (1996) aged 6 to adulthood sponse with simultaneous inhibition of another of stim-
uli presented visually, auditorally, or in both modalities.
Measures vigilance or the maintenance of concentration
on a simple or relatively complex task over time.
Matching Familiar Fig- Kagan J, et al. Reflection versus impul- Timed with two practice and 12 experimental items;
ures Test (MFFT) (1964) sivity in children, ado- measures delay of decision making in situations
lescents, and adults where a correct response is not obvious (reflection)
vs. the quick choice of an alternative without ade-
quate consideration of options (impulsivity).
Wisconsin Card Sorting Heaton RK, Chelune Cognitive set-shifting and Measure requires the ability to develop and maintain
Test (WCST) GJ, Talley JL, maintenance ability in re- an appropriate problem-solving strategy across chang-
Kay GG, Curtiss sponse to reinforcement ing stimulus conditions in order to achieve a future
G (1993) contingencies and non- goal
verbal abstract concept
89
Stroop Color and Word Golden CJ (1978) Cognitive flexibility, resis- Used in diagnosis of brain dysfunction in the evalua-
Test tance to interference tion of stress, personality, cognition, and psycho-
from outside stimuli, pathology
and reaction to cogni-
tive stress. Ages 17 to
adult.
Motor and graphomo-
tor function
Motor-Free Visual Per- Colarusso RP, Ham- Visual-perceptual abilities Assess visual perception in children; especially useful
ception Test mill DD (1996) without involving mo- for those who may have learning, cognitive, motor,
tor skills of children or physical disabilities. Measures five areas of per-
ages 4–12 ception: spatial relationship, visual discrimination,
figure-ground, visual closure, and visual memory.
Purdue Pegboard Test Tiffin J (1968) Finger dexterity and Tests individual’s ability to move right, left, and both
hand–eye coordination hands, fingers and arms (gross movement), and to con-
trol movements of small objects (fingertip dexterity)
Denckla Motor Perfor- Denckla MB (1985) Soft neurological signs in Looks for signs of adventitious movements as child fo-
mance Battery children with motor co- cuses on a delicate or difficult task (e.g., sticking out the
ordination problems in tongue while attempting to write, or grimacing, or mov-
the absence of diagnos- ing the mouth to the side while focusing on a delicate ac-
able neurological condi- tivity), simultaneous movements of another part of the
tions body like gesticulations and grimacing while moving a
another part of the body, or the mirror movements of
overflow indicating that difficulty exists in the specific
activation of a pathway without causing activation of
King et al.
COGNITIVE EVALUATION
A cognitive evaluation yields critical information concerning a child’s abilities,
disabilities, strengths, and weaknesses. Research has revealed specific cognitive
profiles of individuals with ASDs, including spared performance on tasks that
are rote, mechanical, or perceptual, and impaired performance on tasks that are
complex or abstract (2). This “saw tooth” profile—common in children with
ASDs—reveals significant inter-subtest peaks and valleys. This profile suggests
relative task-specific strengths in simple memory, visual-spatial problem solving,
and visual reasoning. Relative weaknesses include those in fine motor skills, com-
plex information processing, verbal comprehension, and planning (3). However,
this profile, although clinically common, has not been shown to be universally
diagnostic, reinforcing the belief that children with autism show great variability
in skills compared with one another as well as with other children (4).
The choice of test for a cognitive evaluation depends on the child’s verbal
ability and age. Children with verbal ability may be tested using the Wechsler
Preschool and Primary Scales of Intelligence–Revised (WPPSI-R) (5) or the
Wechsler Intelligence Scale for Children–Third Edition (WISC-III) (6). The in-
formation gathered from these tests reveals functioning in attention, memory,
visual-spatial problem solving, abstract thinking, processing speed, fund of infor-
mation, and comprehension. Another verbal test of intelligence is the Stanford-
Binet Intelligence Scale (SBIS–Fourth Edition) (7). The subtests that comprise
this battery cover four broad areas: verbal reasoning, abstract/visual reasoning,
quantitative reasoning, and short-term memory (8). Notably, the SBIS cannot
assess for mental retardation in 2-year-olds, and more severe MR cannot be as-
sessed in 3- to 4-year-olds due to the floor effect on this measure which truncates
results at the low end for children between 2 and 5 years of age. Although the
Assessment of Children 93
WPPSI-R, WISC-III, and SBIS have no normative data for children with develop-
mental delays, they are widely considered “gold standard” indices of cognitive
functioning and an integral component of neuropsychological evaluations.
Another cognitive test widely used with children is the Kaufman Assess-
ment Battery for Children (K-ABC) (9). This test battery assesses simultaneous
and sequential mental processing. Six of these 10 subtests are considered nonver-
bal and are well suited for children with communication deficits. However, al-
though the K-ABC battery is otherwise well standardized, normative data are
lacking for children with developmental disabilities.
Overall, verbal children who exhibit difficulty with attention, interpersonal
interaction (e.g., looking at the examiner, following the examiner’s instructions),
and negative behaviors will experience greater difficulty in completing these tests,
and the accuracy of the cognitive profiles should be judged with caution.
For youngsters who lack verbal communication, a nonverbal test of cogni-
tive ability is necessary to adequately assess the child’s nonverbal intelligence.
However, the same caveats apply as for children with verbal abilities: negative
behaviors (self- and other-directed aggression), repetitive behaviors, noncompli-
ance, attentional deficits, and over- or underactivity will compromise the validity
of the profile. Nonverbal measures of intelligence assess cognitive skills and apti-
tudes such as reasoning, spatial visualization, memory, attention, and speed of
processing. These concepts are evaluated through the use of pictures, figural illus-
trations, and coded symbols, with no reliance on perceiving or manipulating
words or numbers that are printed or verbally presented (10).
The Leiter International Performance Scale–Revised (Leiter-R) (10) is a
nonverbal cognitive battery designed for populations for whom verbally based
evaluations are not appropriate. The Leiter-R has been shown useful for testing
children who do not speak English, speak English as a second language, are
hearing-impaired, or have significant problems with overactivity (e.g., attention-
deficit/hyperactivity disorder), and may thus be especially useful for use in ASD
populations.
Other nonverbal measures of intelligence include Raven’s Progressive Ma-
trices (11), a series of tests of inductive reasoning. This series is appropriate for
nonverbal populations because it requires no verbal ability, skilled manipulative
ability, or subtle differentiation of visuospatial information on the part of the
examinee. It consists of three forms: the Standard Progressive Matrices (11,12),
the Colored Progressive Matrices (13,14), and the Advanced Progressive Matri-
ces (16). There are several shortcomings to this battery. First, because normative
data for these tests are outdated, percentiles obtained by an individual taking the
test today may not be comparable with those obtained by an individual 40 years
ago. Additionally, there are no normative data for use of Raven’s Progressive
Matrices with a developmentally delayed population.
94 King et al.
NEUROPSYCHOLOGICAL EVALUATION
Impairment in neuropsychological functioning is an important component in the
evaluation of children with autism. Children with autism show diverse neuropsy-
chological impairments in explicit and working memory, establishing rules, plan-
ning, and response inhibition (2,24,25). The evaluation of attention, executive
functions (e.g., memory), praxis, and visual processing can guide treatment and
educational strategies aimed specifically at the child’s individual strengths and
weaknesses, and, to a lesser extent, may predict outcome. Neuropsychological
evaluation of the child with autism increases in difficulty with the severity of the
child’s illness. Important factors to consider when testing children with autism
are that attentional difficulties can interfere with testing memory and visual pro-
cessing while the presence of negative behaviors (self- and other-directed aggres-
sion), repetitive behaviors, and noncompliance can significantly compromise the
validity of the testing results.
Memory
Many published tests are available for testing memory in children; however, few
have been used with an autistic population. A search of the literature (PsychInfo
and PubMed) revealed that the Wisconsin Card Sort Test (WCST) and the Cali-
fornia Verbal Learning Test–Children are two of the tests most often used for
memory research in an autistic population.
Assessment of Children 95
Executive Functions
Inability to establish rules and to plan are neuropsychological deficits often found
in children with autism. These skills fall under the broad category of executive
functioning, along with initiation, hypothesis generation, cognitive flexibility, de-
cision making, regulation, judgment, feedback utilization, and self-perception.
These functions are important to assess, formally or informally, especially when
determining whether an individual can live independently.
Planning and rule generation are important higher-order cognitive functions
that affect major areas of functioning, including decision making and daily living.
Of the neuropsychological tests developed to study planning and rule generation
96 King et al.
in children with autism, the WCST is among the most popular and has been used
as a measure of executive functions in several studies (27,32).
Attention
Attention and concentration impact children with autism in many ways. Deficits
in these areas impact learning, memory, daily living skills, and cognitive ability.
The abilities to inhibit responses, distinguish relevant from nonrelevant informa-
tion, develop a system of rules, and solve problems are dependent on a child’s
memory and concentration. Tests that have been used to study attention in chil-
dren with autism include the Continuous Performance Test (CPT) and the WCST
(34).
Conners’ CPT (35) is a widely used test of attention, primarily because
of the wide range of subject-response parameters evaluated and because of the
flexibility of the program (8). It is appropriate for ages 4 through 70, although
normative data for the test indicate a bias toward a younger sample (subjects
aged 4 to 34) (8). A computerized task, it requires the test subject to strike the
appropriate key for any letter except when an X appears on the computer screen.
There are six blocks, each with 20-trial sub-blocks. The interstimulus intervals
vary between blocks, providing a new stimulus at 1, 2, or 4 seconds. Conners’
CPT scores reflect several categories of response style, including number of Hits
(correct responses), Omissions (not responding to a target), Commissions (re-
sponding to a nontarget), Hit rate (mean response time), Hit Rate Standard Error
(consistency of response times), Attentiveness, and Risk Taking (response ten-
dency to be cautious or more impulsive).
Conners’ CPT shows low to moderate correlations with other measures of
attention, but good ability to distinguish clinical from nonclinical populations
(35). The advantage of Conners’ CPT is the considerable amount of data that
can be collected on a number of important aspects of attention. Also, the detailed
analysis allows for detailed characterization of an individual’s deficits.
CONCLUSION
Cognitive and neuropsychological evaluations of children with ASD serve many
purposes for clinicians. These evaluations can assist in clarifying an ASD diagno-
sis by ruling out other disorders, developing and supporting educational place-
ment decisions (e.g., special-education vs. full inclusion), and guide treatment
planning by providing valuable treatment targets for occupation, physical, and
behavioral therapies.
Although great strides have been made in understanding the diagnosis and
treatment of autism in the past two decades, controversy continues regarding
adequate cognitive and neuropsychological testing for low-functioning children
with autism. Currently, many measures used to assess autistic individuals are not
designed specifically for use with autism and do not provide normative data for
autistic populations. Measures that are used in general clinical populations are
typically useful in testing autistic children who are relatively high-functioning
because of their reliance on verbal comprehension and communication.
The measures discussed in this chapter have been used in research with
high-functioning autistic populations, providing a greater understanding of the
strengths, deficits, and neuropsychological components of autism that are crucial
for clarifying the initial diagnosis, ruling out disorders that better explain the
symptoms, and guiding education and treatment. However, a gap remains in de-
termining the specific cognitive and neuropsychological deficits in lower-func-
tioning autistic children because of the lack of instruments developed for use in
nonverbal and minimally communicative populations. This gap must be filled
with further research. Specifically, significant progress must be made in the abil-
ity to assess the cognitive ability of low-functioning children with autism so that
they can be better served by treatment and, especially, in educational placement.
REFERENCES
1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Dis-
orders. 4th ed. Washington, DC: American Psychiatric Publishing, 1994.
2. Filipek PA, Accardo PJ, Baranek GT, et al. The screening and diagnosing of autistic
spectrum disorders. J Autism Dev Disord 1999; 29(6):439–484.
98 King et al.
Cecelia McCarton
Albert Einstein College of Medicine
and McCarton Center for Developmental Pediatrics
New York, New York, U.S.A.
HISTORICAL PERSPECTIVE
Kanner (1) first described the behaviors of 11 children between the ages of 2
and 8 years who presented with common characteristics of a profound lack of
social engagement, a range of communication problems, and unusual responses
to inanimate objects and the environment. He called these behaviors “autistic
disturbances.”
Kanner believed that the 11 children he studied had one fundamental distur-
bance: “an inability to relate themselves in an ordinary way to people and situa-
tions from the beginning of life.” He reported that as infants these children failed
to make direct and sustained eye contact and often resisted being held. Lan-
guage—in terms of prosody, content, and usage—was impaired, with echolalia,
pronoun reversal, and a language-processing deficit. Play was repetitive and ste-
reotypic, often lacking imaginative themes. Finally, the desire for sameness often
overwhelmed the children behaviorally if any change was introduced into their
schedule or environment. Kanner’s lucid and clinically precise description of
these behaviors remains the hallmark of the diagnosis described in the fourth
edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV)
(2).
101
102 McCarton
Autism Disorder
For the purpose of this chapter, the term autism spectrum disorder (ASD) is used
synonymously with pervasive developmental disorder (PDD) as umbrella terms.
The term autism disorder, or autism, is used to refer to the specific diagnosis
defined by specific criteria in DSM-IV (2).
The presentation of autism can vary greatly in its severity. However, in
order to be diagnosed with autism disorder, a child must meet six of the 12 criteria
listed in DSM-IV (2) and these symptoms must appear prior to 3 years of age.
The criteria fall into three broad categories:
1. Impairment in communication and imaginative activity
2. Impairment in reciprocal social interaction
3. Markedly restricted repertoire of activities and interests
Additional behavioral and motoric abnormalities are also described as part
of this disorder but do not have to be present to make the diagnosis. The severity
of autism is a spectrum with presenting symptomatology ranging from mild to
severe. One can have a child with normal or above-average intelligence as well
as a child with mental retardation.
Communication Impairment
Children with autism often do not develop an index point or respond consistently
to their name. A portion of children often have a verbal apraxia, which is an
Pervasive Developmental Disorder 103
interruption in the neuromotor signals to the mouth, tongue, and lips necessary
to form sounds and words consistently. Other children have echolalia; they repeat,
parrot-like, what is said to them. Echolalia may be immediate: e.g., when a parent
asks, “Are you hungry?,” the child replies, “Are you hungry?” In delayed echola-
lia, the child recites dialog, scripts from videos, TV commercials, or stories read
to them days or weeks ago. Pronoun reversal is also common, particularly with
you and I. They may refer to themselves in the third person or by name. Jargoning
can also be present, as well as using words and phrases out of context. Compre-
hension of language is often poor; the child, at best, may understand simple com-
mands or directives that have been said many, many times before and that have
often been associated with a visual cue or prompt or said in a particular context.
Prosody—the tone of a child’s voice—can also be affected. It may have a mono-
tone or singsong quality or be loud and high-pitched.
Impaired Social Interactional Skills
One of the first behaviors reported by parents is lack of eye contact or variable
eye contact. Children may have better eye contact with one or both parents than
with any other individual. Parents describe children as “being in a world of their
own,” “zoning out,” “looking through people,” or “preferring to be alone.” Often,
children will not reach out to be held or cuddled, and in fact squirm or stiffen
to get away. Some children do not mind being held or cuddled but accept it in
a more passive way. Joint attention is often lacking, and they do not share in
doing or seeing something with another person.
In terms of their peers, children with autism will play by themselves, engage
in parallel play, or observe from the periphery. They tend to prefer children who
are younger or older than they are. They will often actively engage in chase games
or follow children in activities that occur in a park or outdoor setting. However,
the social cues of others (a smile, a frown, a wave) may be meaningless to them.
Abnormal Behaviors
Children with PDD/ASD may not play at all or have very unusual play patterns.
A child may demonstrate no pretend-play based on imaginative themes and in-
stead use toys in a functional cause-and-effect manner. Toys may be lined up or
carried about or used in unusual ways (e.g., dropping a toy continuously on a
surface or spinning the wheels of a car). Many children require a “sameness or
routine” in their play activities, and any intrusion or interruption is likely to result
in a tantrum. Transitions are often difficult throughout the day’s activities and
may result in “meltdowns.” Stereotypic movements such as hand and/or arm
flapping, finger flicking, rocking, spinning, and teeth grinding may also become
prominent whenever the child is excited or upset.
A preoccupation with certain objects may also be present. Some children
engage in repetitive actions such as opening and closing drawers, turning light
104 McCarton
switches on and off, and spinning objects. Strings, lights, water, and ceiling fans
are particularly attractive. Often children must smell or lick an object before they
engage with it. Lines and the edges of objects (e.g., a desk or a piece of paper)
can be especially appealing, sometimes with the child squinting or looking out
of the corner of his eye at the object.
Associated Impairments
Sensory: Besides the senses of sight, touch, taste, smell, and hearing, our
nervous system also senses pressure, movement, body position, and the force of
gravity. These senses are known as tactile (touch), vestibular (movement), and
proprioceptive (body position). These systems work closely to help a child make
appropriate responses to incoming sensations and to the environment. Children
with autism may greatly overreact to sensory stimuli or have almost no reaction
to it. They have difficulty “filtering out” sensations and stimuli. They may walk
around and cover their ears to block sounds we might not hear or consider moder-
ate (e.g., a hair dryer). On the other hand, they may delight in a blaring police
or fire siren. They may be fascinated with color patterns, lights, shapes, or the
configuration of letters and numbers. They may be preoccupied by certain tex-
tures and rub against a particular surface or avoid certain foods because of a
particular texture. After a child has engaged in spinning or rough-housing, parents
have reported greater alertness, verbalizations, or eye contact. Reactions to pain
may also be abnormal—many parents report that their child has a high tolerance
to pain. Finally, the senses of smell and taste seem to be used more often to
explore objects.
Asperger’s Syndrome
One year after Kanner (1) described autistic behaviors, a pediatrician named
Asperger (5) also described four children with “autistic psychopathy.” The main
features of Asperger’s syndrome are a severe and sustained impairment in social
interactions and the development of restricted, repetitive patterns of behavior
interests and activities (2). There is no evidence of a clinical delay in language
development, although children with Asperger’s often have a pedantic style of
speaking, with mechanical or formal phrasing. They are often described as “little
professors” and initially can fool adults who marvel at their speaking skills. How-
ever, they can be quite concrete, and often their responses or answers miss the
essence of what was asked. Overall, children with this disorder tend to be bright,
and many master reading (hyperlexia) at an early age.
Socially, individuals with Asperger’s syndrome are quite inept and are usu-
ally unable to form friendships. Tragically, they often desire to form these social
106 McCarton
bonds but do not know how to go about making a friend (6). Older children
with Asperger’s syndrome exhibit unusual interests and preoccupation with train
schedules, animal categorizations, maps, and sports statistics. They often have
both fine and gross motor deficits including clumsy, uncoordinated movements
and odd postures. Indeed, in formalized standardized testing, there is often a
marked discrepancy between their high scores on verbal tests and their poor
visual-spatial/visual-perceptual abilities (performance IQ). An excellent book on
Asperger’s syndrome edited by Klin et al. (7) covers the wide breadth of this
fascinating and tragic disorder.
Rett’s Syndrome
Rett’s syndrome is a progressive neurological disorder essentially limited to girls.
Children initially develop normally during the first year of life, but there is then
a rapid deterioration consisting of decelerating head growth, loss of purposeful
hand movements, development of ataxia, and severely impaired social, cognitive,
and language skills. Stereotypic hand movements consist of hand-washing move-
ments, licking, clapping, or wringing. Mental retardation becomes prominent, and
clinical seizures occur in about one-third of cases (8–12). Although the condition
stabilizes over a period of many years, global neurological cognitive, behavioral,
and language deficits remain prominent.
PREVALENCE OF PDD/ASD
In the past 10 years, the overall prevalence of PDD/ASD has reached alarming
proportions. Early epidemiological studies indicated a prevalence of 4–5 per
10,000, or one in every 2000 people (17–19). However, increased numbers of
children have now been identified with this disorder. Whether it is because of
broader clinical definitions of inclusion or greater clinical recognition on the part
of professionals is still open to debate. Regardless of the reason, current preva-
lence rates range from 10 to 20 per 10,000 or one in every 500 to 1000 children
Pervasive Developmental Disorder 107
(19–30). Rett’s syndrome and childhood disintegrative disorder are rarer, each
occurring in fewer than one per 10,000 live births. The male-to-female ratio for
PDD/ASD is 4:1, with the exception of Rett’s syndrome, which occurs only in
girls.
Studies in the United States have mirrored this same increase in prevalence
rates. In an April 1999 report, the California Department of Developmental Ser-
vices found a 273% increase between 1987 and 1998 in the number of new chil-
dren entering the California developmental services system with a professional
diagnosis of autism. The report further stated that “the number of persons with
autism grew markedly faster than the number of people with other developmental
disabilities (i.e., cerebral palsy, epilepsy, mental retardation” and “compared to
characteristics of 11 years ago, the present population of persons with autism is
younger and has a greater chance of exhibiting no or milder forms of mental
retardation” (31).
Other states are also reporting dramatic increases. Data from the 1998
Maryland Special Education Census revealed that the state experienced a 513%
increase in autism between 1993 and 1998, while the general population in Mary-
land from 1990 to 1998 increased only 7%. A comparative analysis of the 16th
and 20th annual reports to Congress on the implementation of the Individuals
with Disabilities Education Act (IDEA) showed increases of more than 300% in
autistic children served under IDEA between 1992 and 1997 in 25 states: Ala-
bama, Alaska, Arkansas, Colorado, Delaware, Illinois, Indiana, Iowa, Kentucky,
Maine, Maryland, Michigan, Montana, Nebraska, Nevada, New Mexico, North
Dakota, Ohio, Oklahoma, Oregon, Pennsylvania, Rhode Island, South Carolina,
Vermont, and Wisconsin (31).
The Centers for Disease Control and Prevention, in a report released in
April 2000, found that the incidence of autism in Brick Township, New Jersey,
in 1998 was one in 150 children. The Autism Society of America estimates that
“more than 500,000 people in the United States have autism or some form of
a pervasive developmental disorder,” making autism one of the most common
developmental disabilities (31).
Indeed, many clinicians consider this the “new epidemic.” PDD/ASD is
not a rare disorder; it is more prevalent in the pediatric population than cancer,
diabetes, spina bifida, and Down’s syndrome. Because this disorder is growing
at such an alarming rate, numerous national professional organizations and the
National Institutes of Health have attempted to combine resources to establish
centers of research and formulate practice parameters for the diagnosis and evalu-
ation of this entity (32,33).
ETIOLOGY OF PDD/ASD
The wide variation in the presenting symptoms of children with PDD/ASD points
to multiple etiologies that ultimately result in abnormal brain development. Origi-
108 McCarton
(MMR) vaccine may cause gastrointestinal problems that, in turn, lead to autistic
behaviors. Wakefield studied a group of children referred because of diarrhea
and abdominal pain, along with a history of normal behavior followed by a loss
of language and acquired skills within 2 weeks after receiving the MMR vaccina-
tions. Gastrointestinal abnormalities following MMR vaccination included
chronic inflammation of the colon, abnormal growth of small nodules of
lymphoid tissue, thrush-like ulcers, and swellings in the small bowel.
Wakefield’s research suggested that the immune system of certain children
with a genetic predisposition to autism may not be able to handle certain viruses
appropriately, possibly including attenuated strains used in vaccines. In these
individuals, the MMR vaccine may lead to impaired gastrointestinal function,
allowing food by-products called peptides, which can exhibit opium-like proper-
ties, to pass through the intestinal walls. These particles may disrupt normal brain
function and development. In addition, most subjects had elevated urinary con-
centrations of methylmalonic acid, which is indicative of a functional vitamin
B12 deficiency often seen in other gastrointestinal disorders. Vitamin B12 is essen-
tial for normal myelination of nerve cells, a process not completed until around
10 years of age. Wakefield has subsequently collected a sample of over 100
children who presented with the same profile. However, reanalysis of data from
a population study of autism performed in the late 1980s by Gillberg and Heijbel
(75) failed to support this connection between MMR and PDD/ASD.
The fact that mercury, in the form of thimerosal, is used as a preservative
in some vaccines has led to the theory that the vaccines have caused mercury
poisoning, which causes many traits and behaviors similar to those seen in autistic
children (31). Thimerosal is used in DPT, DPTH, tetanus, DT, Td, influenza,
meningococcal, and most DtaP, Hib, and hepatitis B vaccines. The total amount
of mercury that babies under 6 months old have been exposed to in childhood
vaccines has exceeded the Environmental Protection Agency (EPA) limit. Thi-
merosal-free vaccines are available, but parents must request them.
Abnormalities in the breakdown of peptides, especially those found in glu-
ten and casein, have also been implicated in a subset of children with PDD/ASD.
Removal of gluten and casein from the diet has been reported to reduce autistic
symptoms, increase attention, and improve language and socialization (76–79).
Gupta and coworkers (80) demonstrated a marked deregulated immune sys-
tem in children with autism, including abnormalities of T cells, B cells, and natu-
ral killer (NK) cells, as well as various immunoglobulin classes. This deregulation
may account for the autoimmune and allergic reactions common in autistic chil-
dren as well as a susceptibility to fungal, viral, and bacterial infections.
Finally, Singh and colleagues (81) found evidence suggesting that viruses,
or the immunizations against them, play a role in causing autism. The serum of
both autistic and nonautistic controls was analyzed for antibodies to measles and
human herpesvirus 6. Serum samples for two brain autoantibodies (anti-MBP
110 McCarton
and anti-NAFP), the presence of which indicates that the body is attacking its
own cells, were also analyzed.
The researchers reported that measles and herpes antibody titers were only
moderately higher in autistic subjects than in controls. However, all the normal
controls were negative for brain autoantibodies, while the majority of autistic
samples (90%) positive for virus antibodies were also positive for brain autoanti-
bodies. This finding supported the hypothesis that a virus-induced autoimmune
response may play a causal role in autism.
which may or may not have been diagnosed in the past. Although these atypical
behaviors are present, usually social skills and language development are normal.
Recently, a new diagnostic entity called sensory integration disorder (SID)
has been widely used to describe children who have difficulty organizing external
tactile, auditory, or visual stimuli (82). They are referred to as being “out-of-
sync” with the environment and often have difficulty entering a room in which
other children are present, going to birthday parties or circuses, and playing with
children, and become overwhelmed with smells and sounds. Many of these chil-
dren display prominent tactile defensiveness, often not wanting to touch certain
textures or objects and being particularly sensitive on their skin. Some of these
behaviors are also seen in children with PDD/ASD, but with pure SID communi-
cative and cognitive skills are usually preserved.
EVALUATION OF PDD/ASD
The diagnosis of PDD/ASD is behaviorally defined. It is based on clinical rather
than laboratory findings. There are no medical tests (chromosomal, radiological,
electrophysiological, or biochemical) that can be administered to establish a diag-
nosis of PDD/ASD, although tests can rule out or identify other underlying prob-
lems. In order to determine a diagnosis of PDD/ASD, professionals depend on
112 McCarton
hands over their ears) should be a reason for referral to an experienced pediatric
audiologist. Children with PDD/ASD are often thought to have a hearing loss
because of their lack of response to language. A hearing evaluation is usually
the first one ordered by a pediatrician. Moreover, a hearing loss (conductive,
sensorineural, or mixed) can co-occur in a child with autism (111,112). However,
accompanying behaviors of stereotypic activities, loss of language skills, and
social deficits often differentiate the child with autism from the child with a hear-
ing loss. In any event, a hearing evaluation consisting of a behavioral evaluation
(visual-reinforcement audiometry) or an electrophysiological assessment (brain-
stem auditory evoked responses) and tympanometry are the usual methods em-
ployed.
Measures of Fine and Gross Motor Skills/Sensory
Integration
Many children with PDD/ASD demonstrate problems with fine and gross motor
skills, hypotonia (3), motor planning or sequencing of movement patterns, organi-
zation of materials, and increased sensitivity to tactile and auditory environmental
stimuli (113,114). These behaviors are usually evaluated by an occupational ther-
apist and physical therapist. Assessment of these skills utilizes a variety of stan-
dardized tools appropriate to the developmental level of the child. Once again,
the most important aspect is that the evaluator be experienced in assessing chil-
dren with autistic behaviors because the testing may require adaptations.
Speech/Language Evaluation
Speech and language therapists who have experience in assessing children with
PDD/ASD are important in the comprehensive evaluation of a child suspected
of PDD/ASD. All aspects of language function—expressive, receptive, prag-
matic, and prosody (voice and speech production)—should be evaluated.
Wetherby and Prizant (115) and Crais (116) recommend that all evaluations do
the following:
1. Focus on functions of communication
2. Analyze preverbal communication (gaze, gestures, vocalizations)
3. Assess social-affective signaling
4. Profile social, communicative, and symbolic abilities
5. Directly assess the child
6. Make observations of spontaneous and initiated communication
7. Directly involve parents or caregivers during the assessment
No standardized test can provide an opportunity to assess all these areas, but the
most commonly used instruments include the Rossetti Infant-Toddler Language
Scale (117), the Preschool Language Scale–3 (PLS) (118), the Clinical Evalua-
116 McCarton
1. Fostering of development
2. Promotion of learning
3. Reduction of rigidity and stereotypy
4. Elimination of nonspecific maladaptive behaviors
5. Alleviation of family stress
Behavioral Therapy
In 1993, Dr. Ivan Lovaas (128) reported a 47% recovery rate for children with
autism given ABA therapy—a 40-hour-a-week comprehensive one-on-one teach-
ing program (1:1 discrete trials). Learning consisted of breaking down hundreds
of language, cognitive, and social tasks to their least common denominator and,
once the child mastered these tasks, using them as the foundation to raise the
task or activity to a more complex level. Goals of the therapy include increasing
expressive and receptive language; expanding play skills; learning cognitive con-
cepts; increasing eye contact, attention, and focus; promoting social interactional
skills; eliminating rigid or perseverative behaviors; and decreasing tantrums or
aggression.
Many parents feel that ABA therapy is the only one that “works” and attest
to the “cures” it has brought about in their PDD/ASD children. In fact, although
the original Lovaas study has been criticized on scientific grounds, it remains
the only technique for which long-term positive cognitive effects have been re-
ported in a group of PDD/ASD children.
Recently, Smith (129) compared two groups of children who began treat-
ment between the ages of 18 and 42 months. The children were divided into two
groups:
1. Seven children with autism and eight with PDD received 30 hours
per week, for 2–3 years, of intensive training based on the techniques
used in the UCLA Young Autism Project. Professional therapists con-
ducted most of the training, with parents assisting for several hours
per week.
118 McCarton
2. Seven children with autism and six with PDD received therapy pro-
vided by parents who were trained by professional therapists. In addi-
tion, the children were enrolled in special-education classes for 10 to
15 hours per week.
Medication
More and more children with PDD/ASD are now receiving medications at
younger and younger ages. It is important to remember that no medication can
cure autism. It can only help ameliorate behaviors that may interfere with the
child’s effective participation in his therapies or with his daily activities. Medica-
tion should be used only after educational and behavioral treatments have been
given an extended trial. Drugs should be used with caution, and only by a physi-
cian with extensive experience in treating children with PDD/ASD.
claims, both positive and negative, accompany each of them. To truly evaluate
the efficacy of these alternative therapies, it is imperative that each be subjected
to randomized clinical trials. This will, first of all, prove whether they work. If
they are efficacious, it will also determine which specific children would benefit
the most from these interventions. Until this happens, parents and children with
PDD/ASD often wander from therapy to therapy in hopes of finding help.
Family Coping
How does a family adjust to their child’s diagnosis of PDD/ASD? Do they ever
adjust? Initially, the diagnosis of PDD/ASD is a shock, even if the family has
suspected this might be the case, because to hear the diagnosis from a stranger
is overwhelming for most parents. Stereotypes of “autistic” children usually flood
a parent’s mind, along with a long list of why this could or could not apply to
their child. Many parents report that the blackest and worst day of their lives
was the day they were told their child had PDD/ASD. Waves of despair and
helplessness usually overwhelm parents, even though they may seem to be coping
well as they ask the doctor what they need to do next to help their child. Elements
of guilt, anger, resentment, and sadness often form a backdrop of emotions that
are ever-present but varying in intensity as the child’s developmental picture
unfolds. These emotions are all normal parental reactions to the diagnosis of
PDD/ASD. Parents will probably experience these emotions many times—not
only in the beginning but also later, after they think they have finally adjusted
to their child’s condition. These emotions are always very close to the surface
and never really scar over. Minor experiences, seemingly innocent and unex-
pected, often open the floodgates of tears and renewed pain.
The first step toward moving on is to acknowledge these feelings. They
are normal and natural. It takes a long time to adjust to the diagnosis of PDD/
ASD, and the healing process is a long journey. It is also important that parents
educate themselves about PDD/ASD. Numerous associations are ready to pro-
vide information to parents through books and other reading material and meet-
ings, as well as to be supportive. There are parent groups in almost every part
of the country; these are invaluable in showing parents that many other people
just like themselves have children with the same diagnosis. The Internet is also an
invaluable means of meeting other parents, getting information, and establishing a
support system. It is a source of instant knowledge to parents throughout the
United States and even the world. Some of the information is excellent, and some
of it is quite subjective. The most important aspect, though, is the power and
interconnectedness it offers parents, almost instantly.
Parents should also look for support within their own families—their parents,
siblings, and extended ring of relatives and friends. Often, parents desperately want
the support of family and friends but at the same time worry abut how they will
take the news. The parents may need to provide their family and friends with the
Pervasive Developmental Disorder 123
information they have gleaned and allow them to ask questions. It may take some
time for the extended family to enter into a circle of support. Some of them may
not be able, but the ones who can will prove to be invaluable assets to both the
parents and the child throughout the future years of development.
Finally, parents should try to form an ongoing relationship with a profes-
sional who is knowledgeable about all aspects of PDD/ASD. Such an invaluable
resource will assist the parents in the myriad of decisions that will need to be
made about the child’s therapies, education, and medical care. The person can
be a physician, social worker, or service coordinator. The most important factors
are that the person be knowledgeable, available, and willing to go through the
life experience with the parent and child.
with one or both parents who act as a lifeline and interpreter of the world. Often,
PDD/ASD children have a very difficult time if separated from that special parent
and will become very upset. Some PDD/ASD children completely ignore most
unfamiliar people, while others are indiscriminately “friendly” with strangers.
Most of these children relate better to adults than to other children except when
the other child is older or younger.
SUMMARY
PDD/ASD is a neurological disorder of unknown etiology that is characterized
by a triad of atypical behavioral manifestations: an impairment in socialization, an
Pervasive Developmental Disorder 125
REFERENCES
1. Kanner L. Autistic disturbances of affective contact. Nerv Child 1943; 2:217–250.
2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders. 4th ed. Washington, DC: American Psychiatric Publishing, 1994.
3. Rapin I. Neurological examination. In: Rapin I, ed. Preschool Children with Inade-
quate Communication: Developmental Language Disorder, Autism, Low IQ. Lon-
don: MacKeith, 1996:98–122.
4. Towbin KE. Pervasive developmental disorder not otherwise specified. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:123–147.
5. Asperger H. “Autistic psychopathy” in childhood. Translated and annotated by
Frith U. In: Frith U, ed. Autism and Asperger Syndrome. New York: Cambridge
University Press, 1991/1944:37–92.
6. Bonnet A, Gao XK. Asperger syndrome in neurologic perspective. J Child Neurol
1996; 11:483–489.
7. Klin A, Volkmar FR, Sparrow SS, eds. Asperger Syndrome. New York: Guilford
Press, 2000.
8. Armstrong DD. Review of Rett syndrome. J Neuropathol Exp Neurol 1997; 56:
843–849.
126 McCarton
9. Naidu S. Rett syndrome: a disorder affecting early brain growth. Ann Neurol 1997;
42:3–10.
10. Hagberg B, Aicardi J, Dias K, Ramos O. A progressive syndrome of autism, de-
mentia, ataxia and loss of purposeful hand use in girls: Rett’s syndrome: report
of cases. Ann Neurol 1983; 14:471–479.
11. Hagberg B. Rett syndrome: clinical peculiarities and biological mysteries. Acta
Paediatrica 1995; 84:971–976.
12. Percy A, Gillberg C, Hagberg B, Witt-Engerstrom I. Rett syndrome and the autistic
disorders. Neurol Clin N Am 1990; 8:659–676.
13. Volkmar FR, Rutter M. Childhood disintegrative disorder: results of the DSM-IV
autism field trial. J Am Acad Child Adolesc Psychiatry 1995; 34:1092–1095.
14. Volkmar FR, Kiln A, Marans W, Cohen DJ. Childhood disintegrative disorder. In:
Cohen DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental
Disorders. 2nd ed. New York: Wiley, 1997.
15. Malhotra S, Gupta N. Childhood disintegrative disorder. J Autism Dev Disord
1999; 29(6):491–498.
16. Tuchman R. Pervasive developmental disorder: neurologic perspective. Acta Neu-
ropaediatrica 1996; 2:82–93.
17. Lotter V. Epidemiology of autistic conditions in young children. I. Prevalence.
Soc Psychiatry 1966; 1:124–137.
18. Brask BH. A prevalence investigation of childhood psychoses. In: Nordic Sympo-
sium on the Care of Psychotic Children. Oslo: Barnepsychiatrist Forening, 1970.
19. Wing L, Gould J. Severe impairments of social interactions and associated abnormalities
in children: epidemiology and classification. J Autism Dev Disord 1979; 9:11–29.
20. Bryson SE. Brief report: epidemiology of autism. J Autism Dev Disord1996; 26:
165–167.
21. Bryson SE. Epidemiology of autism: overview and issues outstanding. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:41–46.
22. Bryson SE, Clark, BS, Smith IM. First report of a Canadian epidemiological study
of autistic syndromes. J Child Psychol Psychiatry 1988; 4:433–445.
23. Gillberg C, Steffenburg S, Schaumann H. Is autism more common now than ten
years ago? Br J Psychiatry 1991; 158:403–409.
24. Ishii T, Takahashi O. The epidemiology of autistic children in Toyota, Japan: prev-
alence. Jpn J Child Adolesc Psychiatry 1983; 24:311–321.
25. Sugiyama T, Abe T. The prevalence of autism in Nagoya, Japan: a total population
study. J Autism Dev Disord 1989; 19:87–96.
26. Wing L. The definition and prevalence of autism: a review of the literature. Ad-
olesc Psychiatry 1993; 2:61–74.
27. Tanoue Y, Oda S, Asano F, Kawashima K. Epidemiology of infantile autism in
Southern Ibaraki, Japan: differences in prevalence in birth cohorts. J Autism Dev
Disord 1988; 18:155–166.
28. Matsuishi T, Shiotsuki M, Yoshimura K, Shoji H, Imuta F, Yamashita F. High preva-
lence of infantile autism in Kurume City, Japan. J Child Neurol 1987; 2:268–271.
29. Fombonne E. Epidemiological surveys of autism. Autism Pervasive Devel Disord
1998; 2:32–63.
Pervasive Developmental Disorder 127
49. DeLong R. Children with autistic spectrum disorder and a family history of af-
fective disorder. Dev Med Child Neurol 1994; 36:674–687.
50. DeLong R, Nohria C. Psychiatric family history and neurological disease in autis-
tic spectrum disorders. Dev Med Child Neurol 1994; 36:441–448.
51. DeLong GR, Teague L, Kamran MM. Effects of fluoxetine treatment in young
children with idiopathic autism. Dev Med Child Neurol 1998; 40:551–562.
52. Bailey A, Palferman S, Heavey L, LeCouteur A. Autism: the phenotype in rela-
tives. J Autism Dev Disord 1998; 28:369–392.
53. Brown WT, Jenkins EC, Cohen IL, Fisch GS, Wolf-Schein EG, Gross A, Water-
house L, Fein D, Mason-Brothers A, Ritvo E. Fragile X and autism: a multicenter
survey. Am J Med Genet 1986; 23:341–352.
54. Wahlstrom J, Gillberg C, Gustavson KH, Holmgren G. Infantile autism and the
fragile X: a Swedish multicenter study. Am J Med Genet 1986; 23:403–408.
55. Bailey A, Bolton P, Butler L, LeCouteur A, Murphy M, Scott S, Webb T, Rutter
M. Prevalence of the fragile X anomaly amongst autistic twins and singletons. J
Child Psychol Psychiatry 1993; 34:673–688.
56. Feinstein C, Reiss AL. Autism: the point of view from fragile X studies. J Autism
Dev Disord 1998; 28:393–405.
57. Dykens EM, Volkmar FR. Medical conditions associated with autism. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2nd ed. New York: Wiley, 1997:388–410.
58. Harrison JE, Bolton PF. Annotation: tuberous sclerosis. J Child Psychol Psychiatry
1997; 38:603–614.
59. Smalley S, Smith M, Tanguay P. Autism and psychiatric disorders in tuberous
sclerosis. Ann NY Acad Sci 1991; 615:382–383.
60. Gillberg IC, Gillberg C, Kopp S. Hypothyroidism and autism spectrum disorders.
J Child Psychol Psychiatry Allied Disc 1992; 33:531–542.
61. Stromland K, Nordin V, Miller M. Autism in thalidomide embryopathy: a popula-
tion study. Dev Med Child Neurol 1994; 36:351–356.
62. Nordin V, Gillberg C. Autism spectrum disorders in children with physical or
mental disability or both. I. Clinical and epidemiological aspects. Dev Med Child
Neurol 1996; 38:297–313.
63. Courchesne E, Townsend J, Saitoh, O. The brain in infantile autism: posterior
fossa structures are abnormal. Neurology 1994; 44:214–223.
64. Filipek PA. Quantitative magnetic resonance in autism: the cerebellar vermis. Curr
Opin Neurol 1995; 8:134–138.
65. Piven J, Arndt S, Bailey J. An MRI study of brain size in autism. Am J Psychiatry
1995; 152(8):1145–1149.
66. Bachevalier J. Medial temporal lobe structures and autism: a review of clinical
and experimental findings. Neuropsychologia 1994; 32:627–648.
67. Haas RH, Townsend J, Courchesne E. Neurological abnormalities in infant autism.
J Child Neurol 1996; 11:84–92.
68. Volkmar FR, Cohen DJ. Neurobiologic aspects of autism. N Engl J Med 1988;
318:1390–1392.
69. Panksepp J, Sahley TL. Possible brain opioid involvement in disrupted social in-
Pervasive Developmental Disorder 129
tent and language development of autism. In: Schopler E, Mesibov GB, eds. Neu-
robiological Issues in Autism. New York: Plenum, 1987.
70. Bailey A, Phillips W, Rutter M. Autism: towards an integration of clinical, genetic,
neuropsychological, and neurobiological perspectives. J Child Psychol Psychiatry
1996; 37:89–126.
71. Lotspeich LJ, Ciaranello RD. The neurobiology and genetics of infantile autism.
Int Rev Neurobiol 1993; 35:87–129.
72. Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, Chugani HT.
Developmental changes in brain serotonin synthesis capacity in autistic and nonau-
tistic children. Ann Neurol 1999; 45(3):287–295.
73. Chugani DC, Muzik O, Rothermel R, Behen M, Chakraborty P, Mangmer T,
daSilva EA, Chugani HT. Altered serotonin synthesis in the denta to thalamo-
cortical pathway in autistic boys. Ann Neurol 1997; 42(4):666–669.
74. Wakefield AJ, Murch SH, Anthony A, Linnell J, Casson DM, Malek M, Berelowitz
M, Drillon AP, Thomson MA, Harvey P, Valentine A, Davies SE, Walker-Smith
JA. Ideal lymphoid-nodular dysplasia, non-specific colitis, and pervasive develop-
mental disorder in children. Lancet 1998; 351:637–641.
75. Gillberg C, Heijbel H. MMR and autism. Autism 1998; 2(4):423–424.
76. Shattock P, Lowdon G. Proteins, peptides and autism. Part 2. Brain Dysfunct 1991;
4:323–334.
77. Serussi K. Unraveling the Mysteries of Autism and PDD. NewYork: Simon &
Schuster, 2000.
78. Shaw W. Biological Treatments for Autism. 1998.
79. Lewis L. Special Diets for Special Children. Texas: Future Horizons, 1998.
80. Gupta S, Aggarwal S, Heads C. Brief report: dysregulated immune system in chil-
dren with autism: beneficial effects of intravenous immune globulin on autistic
characteristics. J Autism Dev Disord 1996; 26:439–452.
81. Singh VK, Lin, SX, Yang VC. Raid communication: serological association of
measles virus and human herpisvirus-6 with brain autoantibodies in autism. Clin
Immuol Immunopathol 1998; 89(1):105–108.
82. Kranowitz C. The Out-of-Sync Child. New York: Perigree Books, 1999.
83. Volkmar FR, Kiln A, Siegel B, Szatmari P, Lord C, Campbell M, Freeman BJ,
Cicchetti DV, Rutter M, Kline W. Field trial for autistic disorder in DSM-IV. Am
J Psychiatry 1994; 151:1361–1367.
84. Lord C, Storoschuk S, Rutter M, Pickles A. Using the ADI-R to diagnose autism
in preschool children. Infant Mental Health J 1993; 14:234–252.
85. Gillberg C. Infantile autism: diagnosis and treatment. Acta Psychiatrica Scandi-
navica 1990; 81:209–215.
86. Glascoe FP. It’s not what it seems: the relationship between parents’ concerns and
children with global delays. Clin Pediatr 1994; 33:292–296.
87. Glascoe FP. Parents’ concerns about children’s development: prescreening tech-
nique or screening test? Pediatrics 1997; 99:522–528.
88. Glascoe FP. Collaborating with parents: using parental evaluation of develop-
mental status to detect and address developmental and behavioral problems. Nash-
ville: Ellsworth & Vandermeer, 1998.
130 McCarton
89. Glascoe FP, Dworkin PH. The role of parents in the detection of development and
behavioral problems. Pediatrics 1995; 95:829–836.
90. Piven J, Gayle J, Chase GA, Fink B, Landa R, Wzorek MM, Folstein SE. A family
history study of neuropsychiatric disorders in the adult siblings of autistic individu-
als. J Am Acad Child Adolesc Psychiatry 1990; 29:177–183.
91. Piven J, Wzorek M, Landa R, Lainhart J, Bolton P, Chase GA, Folstein S. Personality
characteristics of the parents of autistic individuals. Psychol Med 1994; 24:783–795.
92. Bolton PF, Murphy M, Macdonald H, Whitlock B, Pickles A, Rutter M. Obstetric
complications in autism: consequences or causes of the condition? J Am Acad
Child Adolesc Psychiatry 1997; 36:272–281.
93. Fombonne E, Bolton P, Prior J, Jordan H, Rutter M. A family study of autism:
cognitive patterns and levels in parents and siblings. J Child Psychol Psychiatry
1997; 38:667–683.
94. Davidovitch M, Patterson B, Gartside P. Head circumference measurements in
children with autism. J Child Neurol 1996; 11:389–393.
95. Woodhouse W, Bailey A, Rutter M, Bolton P, Baird G, Couteur A. Head circum-
ference in autism and other invasive developmental disorders. J Child Psychol
Psychiatry 1996; 37:665–671.
96. Lainhart JE, Piven J, Wzorek M, Landa R, Santangelo SL, Coon H, Folstein SE.
Macrocephaly in children and adults with autism. J Am Acad Child Adolesc Psy-
chiatry 1997; 36:282–290.
97. Fiedler DJ, Bailey JN, Smalley SL. Macrocephaly in autism and other pervasive
developmental disorders. Dev Med Child Neurol 2000; 42:737–740.
98. Fombonne E, Roge B, Claverie J, Courty S, Fremolle J. Microcephaly and macro-
cephaly in Autism. J Autism Dev Disord 1999; 29(2):113–119.
99. Schopler E, Reichler R, Rochen-Renner B. The Childhood Autism Rating Scale
(CARS). Los Angeles: Western Psychological Services, 1988.
100. Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview–Revised: a revised
version of a diagnostic interview for caregivers of individuals with possible perva-
sive developmental disorders. J Autism Dev Disord 1994; 24:659–685.
101. Lord C, Pickles A, McLennan J, Rutter M, Bregman J, Folstein S, Fombonne E,
Leboyer M, Minshew N. Diagnosing autism: analyses of data from the Autism
Diagnostic Interview. J Autism Dev Disord 1997; 27:501–517.
102. DiLavore PC, Lord C, Rutter M. The pre-linguistic autism diagnostic observation
schedule. J Autism Dev Disord 1995; 25:355–379.
103. Lord C. The Autism Diagnostic Observation Schedule–Generic (ADOS-G). Pre-
sented at the NIH State of the Science in Autism: Screening and Diagnosis Work-
ing Conference, Bethesda, MD, 1998.
104. Minshew NJ, Goldstein G. Autism as a disorder of complex information pro-
cessing. Ment Retard Dev Disord Res Rev 1998; 4:129–136.
105. Bayley N. Bayley Scales of Infant Development. 2nd ed. San Antonio, TX: Psy-
chological Corp., 1993.
106. Thorndike RL, Hagen EP, Sattler JM. The Stanford-Binet Intelligence Scale. 4th
ed. Guide for Administering and Scoring. Chicago: Riverside, 1986.
107. Wechsler Preschool and Primary Scale of Intelligence–Revised (WPPSI). San An-
tonio, TX: Psychological Corp., 1989.
Pervasive Developmental Disorder 131
108. Wechsler D. Wechsler Intelligence Scale for Children (WISC-III). San Antonio,
TX: Psychological Corp., 1991.
109. Sparrow SS, Balla DA, Cicchetti DV. Adaptive Behavior Scales (Survey Edition).
Circle Pines, MN: American Guidance Service, 1984.
110. Klin A, Carter A, Volkmar FR, Cohen DJ, Marans WD, Sparrow S. Developmentally
based assessments. In: Cohen DJ, Volkmar FR, eds. Handbook of Autism and Perva-
sive Developmental Disorders. 2nd ed. New York: Wiley, 1997:411–447.
111. Jure R, Rapin I, Tuchman RF. Hearing-impaired autistic children. Dev Med Child
Neurol 1991; 33:1062–1072.
112. Klin A. Auditory brainstem responses in autism: brainstem dysfunction or periph-
eral hearing loss? J Autism Dev Disord 1993; 23:15–35.
113. Stone WL, Lemanek KL. Parental report of social behaviors in autistic preschool-
ers. J Autism Dev Disord 1990; 20:513–522.
114. Kientz MA., Dunn W. A comparison of the performance of children with and
without autism on the Sensory Profile. Am J Occup Ther 1997; 51:530–537.
115. Wetherby AM, Prizant BM. Profiling young children’s communicative compe-
tence. In: Warren S, Reichle J, eds. Causes and Affects in Communication and
Language Intervention. Baltimore: Paul H. Brookes, 1992.
116. Crais ER. Expanding the repertoire of tools and techniques for assessing the commu-
nication skills of infants and toddlers. Am J Speech-Lang Disord 1995; 4:47–59.
117. Rossetti L. Infant-Toddler Language Scale. East Moline, IL: Lingui Systems,
1990.
118. Zimmerman IL, Steiner VG, Pond RE. Preschool Language Scale–3. San Antonio,
TX: Psychological Corp., 1992.
119. Semel E, Wiig EH, Secord WA. Clinical Evaluation of Language Fundamentals–
3 Examiner’s Manual. New York: New York Psychological Corp., 1995.
120. Gardner MF. Expressive One-Word Picture Vocabulary Test–Revised. Novato,
CA: Academic Therapy Publications, 1990.
121. Gardner MF. Receptive One-Word Picture Vocabulary Test–Revised. Novato,
CA: Academic Therapy Publications, 1990.
122. Curry CJ, Stevenson RE, Aughton D, Byrne J, Carey JC, Cassidy S, Cunniff C,
Graham JM Jr, Jones MC, Kaback MM, Moeschler J, Schaefer GB, Schwartz S,
Tarleton J, Opitz J. Evaluation of mental retardation: recommendations of a Con-
sensus Conference: American College of Medical Genetics. Am J Med Genetics
1997; 72:468–477.
123. Minshew NJ, Sweeney, JA, Bauman ML. Neurologic aspects of autism. In: Cohen
DJ, Volkmar FR, eds. Handbook of Autism and Pervasive Developmental Disor-
ders. 2d ed. New York: Wiley, 1997:344–369.
124. Tuchman RF, Rapin I. Regression in pervasive developmental disorders: seizures
and epileptiform electroencephalogram correlates. Pediatrics 1997; 99:560–566.
125. Rapin I. Physicians’ testing of children with developmental disabilities. J Child
Neurol 1995; 10(suppl):S11–15.
126. Tuchman R, Jayakar P, Yaylali I, Villalobos R. Seizures and EEG findings in
children with autism spectrum disorders. CNS Spectrums 1997; 3:61–70.
127. Rutter M. The treatment of autistic children. J Child Psych Psychiatry Allied Disc
1985; 26:193–214.
132 McCarton
INTRODUCTION
Autism is a pervasive developmental disorder (PDD) characterized by communi-
cation and language impairments, social deficits, and stereotyped or repetitive
behaviors. These developmental abnormalities are apparent by early childhood,
or 36 months of age. Autism is a complex psychiatric disorder with oligogenic
inheritance (1). Twin studies and family studies show substantial evidence for
genetic predisposition in the majority of idiopathic cases (2–9). The population
prevalence of autism has been estimated at approximately 0.5–2 per 1000, but
the rate among siblings of affected probands is estimated from multiple studies
at 1–3%, which is profoundly higher (approximately 10–15 times greater) than
the general population prevalence (6–9). The concordance rate for dizygotic (DZ)
twins is similar to the rate for nontwin siblings, whereas the concordance rate
for monozygotic (MZ) twins is approximately 60–90% (2). This suggests that
the heritability for autism is greater than 90%, if you assume a multifactorial
threshold model (10), which exceeds that of other common psychiatric disorders
such as bipolar disorder, alcoholism, and schizophrenia. Other twin studies show
that this predisposition extends to a broader autism phenotype of milder, related
developmental disorders (2,3). This spectrum of nonpathological abnormalities
in behavior is present in parents and siblings of autistic individuals (11). Most
MZ co-twins who were nonautistic exhibited milder abnormalities similar to fea-
tures of autism. Concordance for the broader phenotype of autism for DZ twins
133
134 Reichert et al.
is considerably lower than the concordance for MZ twins: 10% and 92%, respec-
tively (2). Concordance and relative risk continue to drop off dramatically for
relatives outside the immediate family, such as cousins, although the concordance
rates for relatives are still greater than the population prevalence (6,11).
This, combined with the difference in concordance rates between MZ and
DZ twins, suggests the action of several genes acting together. Latent class mod-
eling suggests two to 10 genetic loci interacting, with three interactive loci being
the best model (12), while other studies point to the possibility of 10–100 loci,
each of weak effect (13).
GENETIC STUDIES
Three types of genetic studies are being conducted to identify susceptibility
loci for autism. The first is identifying chromosomal and cytogenetic abnormali-
ties in autistic individuals. The second includes candidate gene studies, while
genome-wide linkage studies constitute the third.
chromosomes with one or two extra copies of the area corresponding to the
Angelman/Prader-Willi critical region. These patients usually have mental retar-
dation (18,23,24).
Chromosome 15 has a high frequency of deletion events, and accounts for
approximately 50% of all supernumerary marker chromosomes in humans (25).
Smith et al. described a 1-Mb deletion in the region 15q22-23 in a patient with
autism, developmental delay, and a mild dysmorphism, and showed that DNA
segments are shared between this region and region 15q11-q13 (26). A 5-kb
deletion was recently found at D15S822, and occurred in families with autism
more frequently than in a control group (27).
A study of linkage analysis results on chromosomes 7, 8, 15, and 16 in
105 multiplex families revealed null alleles at the sites of four markers—D7S630,
D7S517, D8S264, and D8S272—which were the result of deletions ranging in
size from 5 to ⬎260 kb in 12 families with autism (28). When controls were
screened for deletions at these 4 sites, the deletion at D8S272 was found in all
populations. The remaining three deletion sites remained specific to multiplex
families with autism. These could be potential autism-susceptibility deletions, or
it is possible that autism-susceptibility alleles elsewhere induced errors during
meiosis, causing these deletions. The D7S630 deletion occurred close to regions
found to be positive for linkage (29,30). This marker is also very close to the
proximal breakpoint for the interstitial 7q inversion reported in a subject with
autism (31).
Numerous translocations have been found in patients with autism. Several
balanced and complex translocations involve chromosome 7q. Among them are
the reciprocal balanced translocations t(1;7)(p22:q21) (32) and t(5,7)(q14;q32)
(33) and a balanced chromosome rearrangement involving a breakpoint at 7q31.3
(34).
Ashley-Koch et al. studied an autistic disorder family with three affected
siblings: two autistic males, and a female with expressive language disorder (31).
All three had a paracentric inversion, inv(7)(q22–q31.2), inherited from the
mother, who did not have autism. Another group identified a novel gene, RAY1,
on chromosome 7q31 that was interrupted by a translocation breakpoint, t(7;13)
(q31.3;q21) in an individual with autism (35). The failure to identify phenotype-
specific variants in a mutation screening of 27 unrelated autistic individuals sug-
gested to them that the coding region mutations were not likely to be involved
in the etiology of autism. In a study of a kindred, KE, a three-generation pedigree
in which half the members are affected with a severe speech and language disor-
der, a gene (SPCH1) was localized to 7q31, which the authors proposed was
responsible for the disorder (36). The SPCH1 critical interval was narrowed from
a 5.6-cM region between D7S2459 and D7S643 to a 6.1 MB region of completed
sequence between new markers 013A and 330B. They also studied two unrelated
individuals with de novo translocations in 7q31, and a similar speech and lan-
136 Reichert et al.
that studies that use tests other than the MTDT, such as the TDT (51,52), do not
appear to represent evidence against association when converted to MTDT results.
The presence of hyperserotonemia in a subset of autistic subjects has sug-
gested that the serotonergic system may play a role in the etiology of autism.
The serotonin transporter gene, 5-HTT, has been considered a candidate gene
for autism. One report has suggested association, using the TDT, between the
short variant of HTTLPR and autism (55). Another report, attempting replication,
demonstrated preferential transmission of the long variant of HTTLPR in 65 sin-
gleton families (56). A third study by the International Molecular Genetic Study
of Autism Consortium (IMGSAC) found no significant evidence for linkage or
association for the HTTLPR locus or the HTT-VNTR locus, in a sample of 99
multiplex families (51). In another study of the serotonin transporter gene
SLC6A4, the TDT was conducted with 81 trios (57). The investigators found
transmission disequilibrium but not preferential transmission of 5-HTTPLR. In
this study, they sequenced SLC6A4 and its flanking regions in 10 probands, and
found 20 single-nucleotide polymorphisms (SNPs) and seven simple sequence
repeat (SSR) polymorphisms, which they typed in 115 autism trios. TDT analysis
of individual markers showed seven SNP markers and four SSR markers to have
nominally significant evidence of transmission disequilibrium.
138 Reichert et al.
expectation. Analysis of the HOXB1 locus did not reveal any statistically signifi-
cant results, although there was evidence of interaction between HOXB1,
HOXA1, and gender in ASD susceptibility. Another screening of the 24 exons
of HoxA1 and HoxB2 for DNA polymorphisms, in 24 autistic individuals, identi-
fied the same sequence variants (63). However, a TDT in 110 multiplex families
showed no association with autism.
GENOME-WIDE SCREENS
The cosegregation of polymorphic DNA markers with a disorder such as autism
can be tracked in families in order to map genes. By identifying DNA markers
with a known genetic location that significantly cosegregate with autism within
families, linkage mapping can infer the location and inheritance mode of nearby
susceptibility loci. Since multigenerational families with numerous affecteds are
3
4p 4.8 D4S412 1.55
4q 199 D4S1535 0.88
4q
5p 5 D5S417 0.84
6q 109 D6S283 2.23
7p 42 D7S2564 1.01
7q 140.5 D7S2533 3.63 147.2 D7S684 0.62
7q 144.7 D7S530 2.53 125 D7S486 0.83 138 D7S1804 0.93
8q
9
10p 51.9 D10S197 1.36
10q 167 D10S217 0.84
11q
13q 85.0 D13S193 0.59 55 D13S800 0.68
14q 32.2 D14S70 0.99
15q
15q 32 D15S118 1.10
16p 17.3 D16S407 1.51 21 D16S3075 0.74
16q
17p 11 D17S1876 1.21
18q 94 D18S68 0.62 100 D18S878 1.00
19q 48.2 D19S49 0.99 41 D19S226 1.37
22q 5.0 D22S264 1.39
Xq
140 Reichert et al.
Table 2 (Continued)
CLSA (75 families)d SARC (35 families)e IMGSAC 2001 (152 sib-pairs)f
Position Nearest Position Nearest Multipoint HLOD HLOD Position Nearest Multipoint
Chromosome (cM) marker MLS (cM) marker NPL (dom) (rec) (cM) marker MLS
1p
2q 186.2 D2S364 2.45 2.25 1.65
2q 204.5 D2S325 1.52 0.67 0.65 206.4 D2S2188 3.74
3
4p
4q 72.5 D4S3248 1.33
4q 167.6 D4S2368 1.52
5p
6q
7p
7q 104 D7S1813 2.20 119.6 D7S477 3.2
7q 150 D7S1824 0.80
8q 60.3 D8S1477 1.02
9
10p
10q
11q 147.8 D11S968 1.22
13q 55 D13S800 3.00
14q
15q 13.1 D15S975 0.50
15q 32 ACTC 0.54
16p 23.1 D16S3102 2.93
16q 100.4 D16S516 1.03
17p
18q
19q
22q
Xq
rare in autism, other methods of linkage mapping can assess linkage within many
nuclear families. In order to identify autism susceptibility loci, eight groups have
reported systematic scans of the entire genome of multiplex families (29,46,49,
50,64,65,67,71). The results are summarized in Table 2, showing overlapping
regions between studies and the most significant results for each. The results of
several focused linkage scans have also been reported (30,31,47,66), as well as
a genome-wide QTL analysis (Table 2) (71).
The IMGSAC group (50) performed the first genome-wide screen in two
stages, with 99 families, including 87 ASPs and 12 affected nonsib-pairs. The
first stage typed 354 markers in 39 families. A subset of 175 markers was used
in the second stage to genotype 60 additional families, focusing on regions of
interest from the first stage. Six chromosomes (4, 7, 10, 16, 19, and 22), with
regions generating a multipoint maximum LOD score (MLS) ⬎1, were identified.
Molecular Genetics 141
Table 2 (Continued)
Columbia (110 families)g Duke 2002 (99 families)h Alarcon et al. (152 families)I
1p
2q
198 D2S116 2.86
3 36 D3S3680 1.51
4p
4q
4q
5p 45 D5S2494 2.55 Broad
6q
7p
7q 123 D7S523 1.02 145 D7S495 1.66 152 D7S1824 2.85 (0.84)
7q 165 D7S483 2.13
8q 134 D8S1179 1.66 Broad
9
10p
10q
11q
13q
14q
15q
15q
16q 28 D16S2619 1.91 Narrow
16q
17p
18q
19q 52 D19S433 2.46 Narrow 60 D19S425 1.21
22q
Xq 82 DXS1047 2.67 Narrow 63 DXS6789 2.49
a
IMGSAC, 1998 (50).
b
Philippe et al., 1999 (46).
c
Risch et al., 1999 (49).
d
Barrett et al., 1999 (29).
e
Buxbaum et al., 2001 (67).
f
IMGSAC, 2001 (30).
g
Liu et al., 2001 (64).
h
Shao et al., 2002 (65,72).
i
Alarcon et al., 2002 (71).
The most significant region was between D7S530 and D7S684, and had a
multipoint MLS of 2.53. The region between D16S407 and D16S3114 was the
next most significant, with a multipoint MLS of 1.51. They found no evidence
for linkage in the Prader-Willi/Angelman critical region, 15q11–q13.
They performed a follow-up study (66) of 7q32–q35 by fine-mapping the
142 Reichert et al.
stage genome-wide screen. In the first stage, 416 microsatellite markers were
genotyped to 75 families. Their most significant results were for regions on chro-
mosomes 13 and 7, using a recessive model. Marker D13S800 had a maximum
multipoint heterogeneity LOD (HLOD) score of 3.0. A peak between D13S217
and D13S1229 scored a 2.3 HLOD, and marker D7S1813 had an HLOD score
of 2.2. These results overlap with the IMGSAC and Stanford findings for chromo-
somes 7 and 13. They also observed a 0.51 HLOD score at marker D15S975.
The Columbia Genome Center (64) performed a genome-wide screening
for autism susceptibility loci using 335 microsatellite markers and 110 multiplex
families. In addition to more modest evidence for linkage at 12 marker sites (2,
3, 4, 5, 8, 10, 11, 12, 15, 16, 18, 19, and 20), they found significant evidence
for linkage with regions on chromosomes 5, 19, and X. They analyzed their
results using both broader (autism, Asperger’s, or pervasive developmental disor-
der) and narrower (autism only) diagnosis schemes. Under the broad diagnosis,
the most significant region was at marker D5S2494 (MLS ⫽ 2.55). Other signifi-
cant results under this scheme were DXS10437 (MLS ⫽ 2.56), D5S2488 (MLS
⫽ 1.90), and D19S714 (MLS ⫽ 1.72). Under the narrower diagnosis, the most
significant region was on chromosome 19, near D19S714 (MLS ⫽ 2.53). Other
significant findings with the narrow scheme include DSX1047 (MLS ⫽ 2.67),
D16S2619 (MLS ⫽ 1.93), D5S2488 (MLS ⫽ 1.63), and D5S2494 (MLS ⫽ 1.41).
Initially they found no evidence for linkage on chromosome 7, but because
of overlapping findings of positive linkage for this chromosome in previous ge-
nome-wide screenings, a follow-up study was performed. Six microsatellite
markers from the original scan were included, as well as 28 additional markers
to densely cover the region on chromosome 7. These markers were genotyped
in 160 families, including the 110 from the original study. A peak LOD score
of 2.13 was found at D7S483, and another peak of 1.02 at D7S523.
A combination of the results from these studies implicates a locus on chro-
mosome 7q for autism etiology. However, the different studies report positive
peaks for loci on 7q that can be more than 20 cM apart.
A study from Duke University (31) focused the scope of their genome scan
based on the findings of the previously mentioned groups, and their own results
from a study of a family with a paracentric inversion on 7q. They genotyped 76
multiplex families, with nine markers, to this region. Their most significant results
were for the region between D7S2527 and D7S495. A multipoint HLOD score of
1.47 and an MLS of 1.03 were obtained for marker D7S495. They also observed a
peak NPL score of 2.01 for D7S640, and a peak MLS of 1.77 for D7S2527. This
group also noted an increased rate of recombination between autistic families vs.
CEPH controls. TDT analysis suggested transmission distortion at D7S495 ( p
⫽ 0.03), mostly with paternal contribution. Additionally, significant paternal con-
tribution to linkage disequilibrium was observed at D7S1824 ( p ⫽ 0.02), and to
IBD sharing at D7S640 ( p ⫽ 0.007). This increase in paternal contribution sup-
144 Reichert et al.
ports an imprinting effect. Overall, the results provide further supporting evidence
for an autism-susceptibility locus on 7q.
This group also conducted a follow-up study, genotyping 14 markers in
the chromosome 15q11–q13 region to 63 multiplex families (47). They found
evidence for linkage in this region, along with increased rates of recombination
near GABRB3 and D15S217, in autism families. For marker D15S217, they
found a maximum LOD score of 1.37 ( p ⫽ 0.03), under a recessive model, and
a Z-score of 1.78.
The Duke group’s recent genome-wide screening was performed in two
stages (65). In the first stage, 52 multiplex families were genotyped for 352 satel-
lite markers. Markers in eight regions on seven chromosomes (2, 3, 7, 15, 18,
19, and X) met a threshold MLS of ⱖ1.00, and were genotyped with their flanking
markers in the second stage, in 47 additional families. Other regions reported in
previous screenings (2, 7q, 13, and 15q11–q13) were also genotyped to these 99
families. Their peak linkage results were on chromosome X with a multipoint
MLS of 2.49. They also found that their peak at D3S3680, a unique finding,
remained strong with a pairwise MLS of 2.02. Two markers on chromosome
2, D2S2215 and D2S116, previously below the cutoff for inclusion in the sec-
ond stage, now both had MLOD ⱖ1.0, and overlapped with findings from other
studies (50,67). Their results for chromosome 7q (multipoint MLS ⫽ 1.66) also
overlapped with the IMGSAC findings (50), and their peaks on chromosome
15q11–q13 (MLOD ⱖ1.0) overlap with results from the PARISS group (46). The
regions on chromosome X and 19 also overlapped with the findings of IMGSAC,
PARISS, and Columbia (64).
Studies of both developmental language disorder, or specific language im-
pairment (SLI), and autism have had significant linkage results on chromosome
7q31. Although the two syndromes are diagnostically distinct, they overlap phe-
notypically (69). The prevalence of autism in families of children with SLI is
greater than expected, and the prevalence of SLI in families of children with
autism is also greater than expected. The two disorders appear to be genetically
related and may have some genes in common (70).
Alarcon et al. (71) performed nonparametric multipoint linkage analyses
of 152 nuclear families, with at least two children with an ASD, to search for
quantitative trait loci (QTLs). Nine ADI-R items were examined for familiality
and sibling correlations, and three had significant intersib correlations [age at
first word (WORD), age at onset of phrase speech (PHRASE), and repetitive
or stereotyped behavior (RSB)]. These three significant items were chosen as
endophenotypes. Previous family and linkage studies in autism have shown these
phenotypes to be relevant to genetic studies of autism. Two methods of linkage
analysis (nonparametric QTL and HE nonparametric) were used, and peaks with
Z-scores ⬎1.65 and LOD scores ⬎1.0, respectively, that were found in both
analyses, were reported. The highest Z-score was 2.98 on chromosome 7, between
Molecular Genetics 145
DISCUSSION
With several genome-wide linkage screens for autism having been completed,
the differing results are consistent with a disease model of genetic heterogeneity,
and multiple loci of weak effect. However, it is not altogether straightforward
to compare results, because of differences in methodology such as varying mark-
ers and marker maps, as well as different statistical analyses. Also, the criteria
for inclusion and diagnosis differ somewhat from study to study. It may be useful
to examine different subsets of affected probands, based on varying diagnostic
criteria, such as the recent study of language deficit and chromosome 2. Using
the three domains of autism disease (stereotyped or repetitive behaviors, language
deficits, and social deficits) to create subphenotypes may increase the power of
linkage and association findings for susceptibility loci for these subsets of af-
fected probands. These subphenotypes could be used to narrow down which loci
could relate to each domain of the disease. The subsets could also include subjects
who may not have autism but some diagnosis in the broader autism phenotype,
with high scores in one particular domain. Further study will help to distinguish
true susceptibility loci from false positives.
In summary, evidence from genome-wide scans, and analysis of dele-
tions and translocations, indicates a susceptibility gene for autism on chromo-
some 7q. Other loci are also concordant between screens, including chromosome
2. Using diagnostic criteria to restrict the families in further studies may decrease
heterogeneity, and thereby increase power to detect genes. In addition, broaden-
ing the definition of affected to include subthreshold cases may also increase
power.
REFERENCES
1. Rutter M. Routes from research to clinical practice in child psychiatry: retrospect
and prospect. J Child Psychol Psychiatry 1998; 39(6):805–816.
2. Bailey A, Le Couteur A, Gottesman I, Bolton P, Simonoff E, Yuzda E, Rutter M.
Autism as a strongly genetic disorder: evidence from a British twin study. Psychol
Med 1995; 25(1):63–77.
3. Folstein S, Rutter M. Infantile autism: a genetic study of 21 twin pairs. J Child
Psychol Psychiatry Allied Disc 1977; 18:297–321.
4. Ritvo ER, Freeman BJ, Mason-Brotheres A, Mo A, Ritvo AM. Concordance for
the syndrome of autism in 40 pairs of afflicted twins. Am J Psychiatry 1985; 142:
74–77.
5. Steffenburg S, Gillberg C, Hellgren L, Andersson L, Gillberg IC, Jakobsson G,
Bohman M. A twin study of autism in Denmark, Finland, Iceland, Norway and
Sweden. J Child Psychol Psychiatry 1989; 30:405–416.
6. Bolton P, Macdonald H, Pickles A, Rios P, Goode S, Crowson M, Bailey A, Rutter
M. A case-control family history study of autism. J Child Psychol Psychiatry Allied
Disc 1994; 35(5):877–900.
Molecular Genetics 147
36. Lai CSL, Fisher SE, Hurst JA, Levy ER, Hodgson S, Fox M, Jeremiah S, Povey
S, Jamison C, Green ED, Vargha-Khadem F, Monaco AP. The SPCH1 region on
human 7q31: genomic characterization of the critical interval and localization of
translocations associated with speech and language disorder. Am J Hum Genet
2000; 67:357–368.
37. Lai CS, Fisher SE, Hurst JA, Vargha-Khadem F, Monaco AP. A forkhead-domain
gene is mutated in a severe speech and language disorder. Nature 2001; 413(6855):
519–523.
38. Newbury DF, Bonora E, Lamb JA, Fisher SE, Lai CS, Baird G, Jannoun L, Slonims
V, Stott CM, Merricks MJ, Bolton PF, Bailey AJ, Monaco AP. FOXP2 is not a
major susceptibility gene for autism or specific language impairment. Am J Hum
Genet 2002; 70(5):1318–1327.
39. Goizet C, Excoffier E, Taine L, Taupiac E, El Moneim AA, Arveiler B, Bouvard
M, Lacombe D. Case with autistic syndrome and chromosome 22q13.3 deletion
detected by FISH. Am J Med Genet 2000; 96(6):839–844.
40. Sukumar S, Wang S, Hoang K, Vanchiere CM, England K, Fick R, Pagon B, Reddy
KS. Subtle overlapping deletions in the terminal region of chromosome 6q24.2-
q26: three cases studied using FISH. Am J Med Genet 1999; 87(1):17–22.
41. Thomas NS, Sharp AJ, Browne CE, Skuse D, Hardie C, Dennis NR. Xp deletions
associated with autism in three females. Hum Genet 1999; 104(1):43–48.
42. Monaghan KG, Van Dyke DL, Wiktor A, Feldman GL. Cytogenetic and clinical
findings in a patient with a deletion of 16q23.1: first report of bilateral cataracts
and a 16q deletion. Am J Med Genet 1997; 73(2):180–183.
43. Smith M, Escamilla JR, Filipek P, Bocian ME, Modahl C, Flodman P, Spence MA.
Molecular genetic delineation of 2q37.3 deletion in autism and osteodystrophy: re-
port of a case and of new markers for deletion screening by PCR. Cytogenet Cell
Genet 2001; 94(1–2):15–22.
44. Ghaziuddin M, Burmeister M. Deletion of chromosome 2q37 and autism: a distinct
subtype? J Autism Dev Disord. 1999; 29(3):259–263.
45. Cook EH Jr, Courchesne RY, Cox NJ, Lord C, Gonen D, Guter SJ, Lincoln A, Nix
K, Haas R, Leventhal BL, Courchesne E. Linkage-disequilibrium mapping of autis-
tic disorder, with 15q11-13 markers. Am J Hum Genet 1998; 62:1077–1083.
46. Philippe A, Martinez M, Guilloud-Bataille M, Gillberg C, Rastam M, Sponheim E,
Coleman M, Zappella M, Aschauer H, Van Maldergem L, Penet C, Feingold J,
Brice A, Leboyer M, van Malldergerme L. Genome-wide scan for autism suscepti-
bility genes. Paris Autism Research International Sibpair Study. Hum Mol Genet
1999; 8:805–812.
47. Bass MP, Menold MM, Wolpert CM, Donnelly SL, Ravan SA, Hauser ER, Maddox
LO, Vance JM, Abramson RK, Wright HH, Gilbert JR, Cuccaro ML, DeLong GR,
Pericak-Vance MA. Genetic studies in autistic disorder and chromosome 15. Neuro-
genetics 2000; 2(4):219–226.
48. Martin ER, Menold MM, Wolpert CM, Bass MP, Donnelly SL, Ravan SA, Zimmer-
man A, Gilbert JR, Vance JM, Maddox LO, Wright HH, Abramson RK, Delong GR,
Cuccaro ML, Pericak-Vance MA. Analysis of linkage disequilibrium in gamma-
aminobutyric acid receptor subunit genes in autistic disorder. Am J Med Genet
2000; 96:43–48.
150 Reichert et al.
Vance MA. No association between the WNT2 gene and autistic disorder. Am J
Med Genet 2002; 114(1):106–109.
62. Ingram JL, Stodgell CJ, Hyman SL, Figlewicz DA, Weitkamp LR, Rodier PM.
Discovery of allelic variants of HOXA1 and HOXB1: genetic susceptibility to au-
tism spectrum disorders. Teratology 2000; 62(6):393–405.
63. Li J, Tabor HK, Nguyen L, Gleason C, Lotspeich LJ, Spiker D, Risch N, Myers
RM. Lack of association between HoxA1 and HoxB1 gene variants and autism in
110 multiplex families. Am J Med Genet 2002; 114(1):24–30.
64. Liu J, Nyholt DR, Magnussen P, Parano E, Pavone P, Geschwind D, Lord C, Iversen
P, Hoh J, Ott J, Gilliam TC. A genomewide screen for autism susceptibility loci.
Am J Hum Genet 2001; 69(2):327–340.
65. Shao Y, Wolpert CM, Raiford KL, Menold MM, Donnelly SL, Ravan SA, Bass
MP, McClain C, von Wendt L, Vance JM, Abramson RH, Wright HH, Ashley-
Koch A, Gilbert JR, DeLong RG, Cuccaro ML, Pericak-Vance MA. Genomic screen
and follow-up analysis for autistic disorder. Am J Med Genet 2002; 114(1):99–
105.
66. IMGSAC. Search for autism susceptibility loci: genome screen follow-up and fine
mapping of a candidate region on chromosome 7q [abstr]. Am J Hum Genet 1999;
65(suppl):A106 (560).
67. Buxbaum JD, Silverman JM, Smith CJ, Kilifarski M, Reichert J, Hollander E,
Lawlor BA, Fitzgerald M, Greenberg DA, Davis KL. Evidence for a susceptibility
gene for autism on chromosome 2 and for genetic heterogeneity. Am J Hum Genet
2001; 68(6):1514–1520.
68. IMGSAC. Further characterization of the autism susceptibility locus AUTS1 on
chromosome 7q. Hum Mol Genet 2001; 10(9):973–982.
69. Folstein S, Mankoski R. Chromosome 7q: where autism meets language disorder?
Am J Hum Genet 2000; 67:278–281.
70. Folstein S. Autism. Int Rev Psychiatry 1999; 11:269–277.
71. Alarcon M, Cantor RM, Liu J, Gilliam TC, Geschwind DH. Evidence for a language
quantitative trait locus on chromosome 7q in multiplex autism families. Am J Hum
Genet 2002; 70(1):60–71.
72. Shao Y, Raiford KL, Wolpert CM, Cope HA, Ravan SA, Ashley-Koch AA, Abram-
son RK, Wright HH, DeLong RG, Gilbert JR, Cuccaro ML, Pericak-Vance MA.
Phenotypic homogeneity provides increased support for linkage on chromosome 2
in autistic disorder. Am J Hum Genet 2002; 70(4):1058–1061.
8
Immune Dysfunction in Autism
Gina DelGiudice
Mount Sinai School of Medicine
and Hospital for Special Surgery
New York, New York, U.S.A.
Eric Hollander
Mount Sinai School of Medicine
New York, New York, U.S.A.
INTRODUCTION
Cellular and humoral immune dysfunction, complement deficiencies, abnormal
antibody production, and abnormal cytokine levels have been documented in au-
tistic individuals. Complex interactions and links between the immune system,
the endocrine system, and the central nervous system have been well established,
and have relevance for the pathophysiology of autism. We review the literature
of the last 25 years pertaining to the relationship between autism and altered
immune response; propose three hypotheses of immunopathogenesis; and review
preliminary immunomodulatory treatment strategies. Additional research is
needed to determine the role of autoimmune and neuroendocrine factors in autism
since most findings are preliminary and the implications of the findings need to
be determined to see whether specific therapeutic agents targeting these systems
might ameliorate or cure symptoms of autism.
PATHOPHYSIOLOGY OF AUTISM
Autism is a severe neuropsychiatric and developmental disorder characterized by
social difficulties (e.g., lack of social reciprocity), language abnormalities (e.g.,
a delay in language development), and stereotyped patterns of behavior (e.g.,
153
154 DelGiudice and Hollander
eight autistic children compared with 21 healthy children. A study using high-
resolution MRI scanning to examine the basal ganglia of 35 autistic individuals
in comparison with control subjects found increased caudate nuclei volume that
was proportional to increased total brain volume in subjects with autism (17).
They reported an association between caudate volume and compulsions. Reviews
of SPECT scans in autistic children have found focal areas of decreased cerebral
blood perfusion (18,19). PET studies have demonstrated asymmetrical 5-HT syn-
thesis in autistic boys (20,21). Functional MRI studies of individuals with autism
have demonstrated abnormal activation of certain brain regions during neuropsy-
chological tasks (22,23).
Serotonin (5-HT) continues to be the focus of neurochemical studies in
autism. Double-blind studies of the serotonin-reuptake inhibitors clomipramine
(24), fluvoxamine (25), and fluoxetine (26), as well as open-label studies of flu-
oxetine (27) and sertraline (28) have documented efficacy in treating both global
autistic symptoms and symptoms of repetitive behaviors and restricted interests
in up to 60% of patients treated.
Several biological studies have also suggested 5-HT dysregulation in autis-
tic patients. McBride et al. (29) measured platelet 5-HT in 77 autistic subjects
compared with normal controls and prepubertal children with mental retardation.
Prepubertal autistic subjects had significant elevation in platelet 5-HT compared
with controls, but this was not statistically significant in postpubertal subjects.
Previous studies have shown that hyperserotonemia in autistic subjects is familial
(30). Leboyer et al. (31) replicated these results and demonstrated that whole-
blood serotonin levels in autistic subjects were age-independent, whereas the
whole-blood serotonin levels decreased with age in controls.
fluence the responses of other lymphocytes and “turn off ” the immune response.
A cytotoxic response requiring no induction with antigen is termed natural cyto-
toxicity. This spontaneous cytotoxicity arises from natural killer (NK) cells and
serves as an initial response before the induction of antigen-specific cytotoxic T
lymphocytes (32).
Other lymphocytes, B cells, are responsible for humoral immunity. Their
surface receptors for antigen are immunoglobulin (Ig) molecules. The five distinct
immunoglobulins are referred to as classes: M, D, E, G, and A. IgG is the main
immunoglobulin class produced. When a B cell encounters a specific antigenic
determinant it can recognize through its Ig surface receptor, it is stimulated to
divide and produces more Ig molecules. These secreted Ig molecules (also known
as antibodies) bind onto their specific Fc receptor and initiate phagocytosis, anti-
body-dependent cell-mediated cytotoxicity (ADCC), and the release of inflam-
matory mediators. IgM is the initial isotype elaborated in response to primary
antigen exposure and represents the first antibody class synthesized by the neo-
nate. IgG is the predominant antibody class in serum, accounting for approxi-
mately 75% of total serum immunoglobulin. IgG also accounts for the antibody
activity in serum and is the reservoir of immunological memory for previously
encountered antigens. Immunoglobulins can trigger secondary mechanisms in-
volving other plasma proteins, such as the complement components. The comple-
ment system consists of at least 30 proteins and following activation results in
cell lysis, increased vascular permeability, leukocyte chemotaxis, and viral neu-
tralization (33). The human immune response is composed of specific elements
that trigger the recruitment of cells and secretion of factors that nonspecifically
cause inflammation or tissue damage. This process should clear the host of patho-
genic organisms. However, dysregulation at any of the described steps can lead
to autoimmune disease.
IMMUNOLOGICAL TOLERANCE
The concept of immunological tolerance explains the numerous strategies that the
immune system has evolved to ensure that an immune response against self does
not occur (34). These mechanisms can be broadly classified as central and periph-
eral tolerance. One of their roles is to delete autoreactive lymphocytes. Apoptosis,
a form of programmed cell death, regulates tolerance induction of self-reactive T
cells in the thymus and B cells in the bone marrow (35). There are many regulators
of apoptosis. Bc1-2, a 25 kDa membrane-bound protein, is a potent antiapoptotic
regulatory protein (34). A recent report interestingly documents a significant in-
crease in Bc1-2 levels in the temporal cortex of patients with schizophrenia (36).
We present four hypotheses with regard to an autoimmune cause of autism:
an infectious hypothesis, an autoimmune hypothesis, an immunological intoler-
ance hypothesis, and a neuropeptide hypothesis. In addition, we examine prelimi-
nary efforts using immunomodulatory therapies to threat autism.
Infectious Hypothesis
In clinical reports, autism has been linked to numerous fetal infections: rubella,
cytomegalovirus, varicella zoster, herpes simplex, and toxoplasmosis (37–41).
Best documented is the relationship between autism and damage caused by fetal
rubella infections. In a longitudinal study of 243 children with congenital rubella,
a high rate of autism and “autism-like” disorders were found, suggesting that, at
least in a subset of patients, fetal viral infections produce an autism syndrome.
However, there was no differentiation to distinguish between autism and mental
retardation in the study subjects, and there was a lack of control subjects as well.
Deykin and MacMahon (42) found no significant association between childhood
autism and maternal viral infections during pregnancy. While some studies on
seasonal variation in the births of autistic children (arguing for a seasonal expo-
sure to an environmental pathogen) have not found any correlation, others have
found a significantly higher incidence among children born in March and August
(43). Geographical areas may also differ in their influence on seasonal exposure
to chemicals or climate, and this was not considered.
So far, there have been few studies of viral agents in the serum or cerebro-
spinal fluid in autistic patients. Investigators have postulated that immunopatho-
logical sequelae of infections may not be direct but rather due to cross-reacting
antibodies; this is known as molecular mimicry. A classic example is rheumatic
fever and Sydenham’s chorea following a group A beta-hemolytic streptococcal
infection (GABHS). Husby et al. (44) documented immunofluorescent antibodies
reacting with the cytoplasm of neurons in the caudate and subthalamic nuclei of
the brain (44).
The monoclonal antibody D8/17 reacts with an antigen on at least 20% of
the B cells of all rheumatic fever patients (45). Two independent laboratory
Immune Dysfunction 159
groups have identified more peripheral B cells expressing D8/17 in children with
obsessive-compulsive disorder (OCD) (46,47). The results suggest that mono-
clonal D8/17 may serve as a marker to susceptibility in subsets of children who
develop childhood-onset OCD. Hollander et al. (48) hypothesized that D8/17
might serve as a marker for susceptibility to autism, and data demonstrate sig-
nificantly greater expression of monoclonal antibody D8/17 in a subgroup of
autistic children than in matched medically ill children. Severity of repetitive
behaviors as assessed by the compulsion score on the Yale-Brown Obsessive
Compulsive Scale strongly positively correlated with D8/17 expression. This
suggests that D8/17 antigen expression may represent both a genetic vulnerability
and an environmental susceptibility to autism.
Autoimmune Hypothesis
Pathogenic autoimmunity occurs when the immune system becomes autoaggres-
sive. Studies have focused on cellular elements of the immune system:
1. Phagocytic cells
2. Cytokines and complement system
3. T cells
4. Humoral antibodies
Studies of Phagocytic Cells
Warren et al. (49) investigated the possibility that altered NK-cell activity may
play a role in the development of autism. In a study of autistic subjects and
healthy controls, 40% of the autistic subjects demonstrated reduced NK cytotox-
icity that was not correlated with a decrease in the number of NK cells (49). This
was subsequently replicated in a later study (50). Since the NK cell is part of
the basic defense mechanism against viral infection, the authors hypothesize that
a predisposition to a relative NK-cell deficiency could subject a developing cen-
tral nervous system to increased risk of damage. However, the authors do not
take into account the bidirectional chemical messengers connecting the immune
system and the central nervous system. In fact, an initial CNS defect could alter
immune system function.
Cytokine and Complement Studies
The importance of complement in immune defense is demonstrated by the sus-
ceptibility of individuals with a deficiency of complement components to infec-
tious and autoimmune illnesses because of defective clearance of antigen–anti-
body complexes. Patients with C4, C2, and C3 deficiencies have a high incidence
of systemic lupus erythematosus (SLE) or other vasculitic syndromes. Patients
with C5, C6, C7, or C8 deficiency more commonly suffer from recurrent infec-
tions (32).
160 DelGiudice and Hollander
A Neuropeptide Hypothesis
Nelson et al. (72) examined biological regulators of cerebral development in au-
tism from archived neonatal blood samples of autistic and MR children, children
with cerebral palsy (CP), and controls. Neuropeptides substance P, vasoactive
intestinal peptide (VIP), calcitonin gene-related peptide (CGRP), and neuro-
trophin nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF),
neurotrophic 3 (NT3), and neurotrophin 415 (NT415) were measured using im-
munoaffinity chromatography. Concentrations of VIP, CGRP, BDNF, and NT415
were significantly higher in autistic and MR (without autism) children than in
controls. These findings provide evidence of the complex interaction of biological
molecules and relate the neurobiological and cognitive disorders to key proteins
involved in brain development. Whether clinical symptoms of autism such as GI
disturbances or sleep disorders are related to these abnormal values warrants fur-
ther study.
attention span, and sleep duration. Systematic behavioral assessments were not
conducted, and no control group was studied.
Secretin, a gastrointestinal polypeptide, was reported to improve social re-
latedness and language skills in three children with autism (93). Three double-
blind placebo-controlled studies of single-dose intravenous secretin have been
conducted. Owley et al. (94) studied 20 children with autistic disorder in a cross-
over of secretin and placebo administered 4 weeks apart. There was no significant
difference between the treatment groups. Sandler et al. (95) found no significant
difference between secretin and placebo. Chez et al. (96) also found no difference
between secretin and placebo in a double-blind crossover study in autistic chil-
dren. These studies do not provide support for the use of secretin in treating
autism.
CONCLUSIONS
Only in the past decade has it become accepted that autism is a biological disor-
der. Recent research in the areas of genetics, neuroimaging, neurochemistry, and
Immune Dysfunction 167
ACKNOWLEDGMENTS
This work was supported by the Seaver Foundation, a National Alliance for Re-
search on Schizophrenia and Depression (NARSAD) Distinguished Investigator
Award (Eric Hollander), Cure Autism Now (CAN) pilot project funding (Gina
DelGiudice and Eric Hollander), and in part by a grant (5 MOI RR00071) from
the Mount Sinai General Clinical Research Center via the National Center for
Research Resources, National Institutes of Health.
We would like to acknowledge the intellectual contributions of Dr. John
B. Zabriskie.
REFERENCES
1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders. 4th ed. Washington, DC: American Psychiatric Press, 1994.
2. Dunn M. Neurophysiologic observations in autism and implications for neurologic
dysfunction. In: Bauman ML, Kemper TL, eds. The Neurobiology of Autism. Bal-
timore: Johns Hopkins University Press, 1994:45–65.
3. Spiker D, Lotspeich L, Kraemer HC, Hallmayer J, McMahon W, Petersen PB,
Nicholas P, Pingree C, Wiese-Slater S, Chiotti C, Wong DL, Dimicelli S, Ritvo
E, Cavalli-Sforza LL, Ciananello RD. Genetics of autism: characteristics of af-
fected and unaffected children from 37 multiplex families. Am J Med Genet 1994;
54:27–35.
4. Bailey A, LeCouteur A, Gottesman I, Bolton P, Simonoff E, Yuzda E, Rutter M.
Autism as a strongly genetic disorder: evidence from a British twin study. Psychol
Med 1995; 2S:63–78.
5. Jorde L, Hasstedt S, Ritvo E, Mason-Brothers A, Freeman B, Pongree C, McMa-
hon W, Peterson B, Jenson W, Moll A. Complex segregation analysis of autism.
Am J Hum Genet 1991; 49:932–938.
6. Ritvo E, Jorde L, Mason-Brothers A, Freeman B, Pingree C, Johes M, McMahon
W, Petersen P, Jenson W, Mo A. The UCLA–University of Utah epidemiologic
survey of autism: recurrence risk estimates and genetic counseling. Am J Psychia-
try 1989; 146:1032–1036.
168 DelGiudice and Hollander
39. Stubbs EG, Ash E, Williams CP. Autism and congenital cytomegalovirus. J Au-
tism Dev Disord 1984; 14:183–189.
40. Peterson MR, Torrey EF. Viruses and other infectious agents as behavioral teratogens.
In: Coleman M, ed. The Autistic Syndromes. New York: Elsevier, 1976:23–42.
41. Knobloch H, Pasamarrick B. Gesell and Amatruda’s Developmental Diagnosis.
3rd ed. New York: Harper & Row, 1974:320–339.
42. Deykin EY, MacMahon B. Viral exposure and autism. Am J Epidemiol 1979; 109:
628–638.
43. Barak Y, Ring A, Sulkes J, Gabbay U, Elizur A. Season of birth and autistic disor-
der in Israel. Am J Psychiatry 1995; 152:798–800.
44. Husby G, Van de Rijn E, Zabriskie JB, Abdin ZH, Williams RC. Antibodies re-
acting with cytoplasm of subthalamic and caudate nuclei neurons in chorea and
acute rheumatic fever. J Expt Med 1976; 144:1094–1110.
45. Zabriskie JB, Lavenchy D, Williams RC Jr, Fu SM, Yeadon CA, Fotino M, Braun
DG. Rheumatic fever-associated B cell alloantigens as identified by monoclonal
antibodies. Arthritis Rheum 1985; 28:1047–1051.
46. Murphy TK, Goodman WK, Fudge MW, et al. B lymphocyte antigen D8/17: a
peripheral marker for childhood-onset obsessive-compulsive disorder and Tou-
rette’s syndrome? Am J Psychiatry 1997; 154:402–407.
47. Swedo SE, Leonard HL, Mittleman BB, Allen AJ, Rapoport JL, Dow SP, Kanter
ME, Chapman F, Zabriskie J. Identification of children with pediatric autoimmune
neuropsychiatric disorders associated with streptococcal infections by a marker
associated with rheumatic fever. Am J Psychiatry 1997; 154:110–112.
48. Hollander E, DelGiudice-Asch G, Simon L, Schmeidler J, Cartwright C, DeCaria
CM, Kwon J, Cunningham-Rundles C, Chapman F, Zabriskie J. B lymphocyte
antigen D8/17 and repetitive behaviors in autism. Am J Psychiatry 1999; 156(2):
317–320.
49. Warren RP, Foster A, Margaretten NC. Reduced natural killer cell activity in au-
tism. J Am Acad Child Adolesc Psychiatry 1987; 26:333–335.
50. Warren RP, Cole P, Odell JB, Pingree CB, Warren W, White E, Yonk LJ, Singh
VK. Detection of maternal antibodies in infantile autism. J Am Acad Child Ad-
olesc Psychiatry 1990; 29:873–877.
51. Warren RP, Singh VK, Cole P, Odell JD, Pingree CB, Warren WL, White E.
Increased frequency of the null allele at the complement C4B locus in autism.
Clin Exp Immunol 1991; 83:438–440.
52. Denney DR, Frei BW, Gaffney GR. Lymphocyte subsets and interleukin-2 recep-
tors in autistic children. J Autism Dev Disord 1996; 26:87–97.
53. Warren RP, Foster A, Margaretterinc, Pace NC. Immune abnormalities in patients
with autism. J Autism Dev Disord 1986; 16:189–197.
54. Ferrari P, Mavescot M, Moulias R, et al. Immunitaire dans l’autisme infantile.
Encephale 1988; 14:339–344.
55. Yonk LJ, Warren RP, Burger RA. CD4 and helper T cell depression in autism.
Immunol Lett 1990; 25:341–346.
56. Plioplys AV. Autism: immunological investigations. Proceedings of the State of
the Art Conference on Autism: Diagnosis and Treatment, Goteborg, Sweden, May
8–10, 1989, pp. 133–138.
Immune Dysfunction 171
57. Young JG, Capanilo BK, Shaywitz BA, Johnson WT, Cohen DJ. Childhood au-
tism: cerebrospinal fluid examination and immunoglobulin levels. J Am Acad
Child Psychiatry 1977; 16:174–179.
58. Warren RP, Odell JD, Warren WL. Brief report: immunoglobulin A deficiency in
a subset of autistic subjects. J Autism Dev Disord 1997; 27:187–192.
59. Yuwiler A, Shih JC, Chen CH, Ritvo ER, Hauna G, Ellison GW, King BH. Hyper-
serotoninemia and antiserotonin antibodies in autism and other disorders. J Autism
Dev Disord 1992; 22:33–45.
60. Laszlo A, Horvath E, Eck E, Fekete M. Serum serotonin, lactate and pyruvate
levels in infantile autistic children. Clin Chim Acta 1994; 229(1–2):205–207.
61. Todd RD, Ciaranello RD. Demonstration of inter and intraspecies differences in
serotonin binding sites by antibodies from an autistic child. Neurobiology 1985;
82:612–616.
62. Todd RD. Pervasive developmental disorders and immunological tolerance.
Psychiatr Dev 1986; 2:147–165.
63. Cook EH Jr, Perry BD, Dawson G. Receptor inhibition by immunoglobulins: spe-
cific inhibition by autistic children, their relatives, and control subjects. J Autism
Dev Disord 1993; 23(1):67–78.
64. Singh VK, Fudenberg HH, Emerson D, Coleman M. Immunodiagnosis and immu-
notherapy in autistic children. Ann NY Acad Sci 1988; 540:602–604.
65. Courchesne E, Lueng-Courchesne R, Piess GA, Uesselink JR, Jennigan TL. Hypo-
plasia of cerebellar vermal lobules VI and VII in autism. N Engl J Med 1988;
318:1349–1354.
66. Toh BH, Gibbs CJ Jr, Gajdusek DC. The 200- and 150-Kda neurofilament proteins
react with IgG autoantibodies from patients with Kuru, Creutzfeldt-Jakob disease,
and other neurologic diseases. Proc Natl Acad Sci USA 1985; 82(10):3485–3489.
67. Rosengren LE, Ahlsen G, Belfrage M, Gillberg C, Haglid KG, Hamberger A. A
sensitive ELISA for glial fibrillary acidic protein: application in CSF of children.
J Neurosci Meth 1992; 44(2–3):113–119.
68. Connolly AM, Chez MG, Pestnonk A, et al. Serum antibodies to brain in Landau-
Kleffner variant, autism, and other neurologic disorders. J Pediatr 1999; 134:607–
613.
69. Stubbs EG, Ritvo ER, Mason-Brothers A. Autism and shared parental HLA anti-
gens. J Am Acad Child Psychiatry 1985; 24:182–185.
70. McConnachie PR, McIntryre JA. Maternal antipaternal immunity in couples pre-
disposed to repeated pregnancy losses. Am J Reprod Immunol 1984; 5(4):145–
150.
71. Warren RP, Yonk LJ, Burger RA, Cole P, Odell JD, Warren WL, White E, Singh
VK. Suppressor-inducer (CD4⫹ CD45RA⫹) T cells in autism. Immunol Invest
1990; 19:245–251.
72. Nelson KB, Grether JK, Croen LA, Dambrosia JM, Dickens BF, Jelliffe LL, Han-
son RL, Phillips TM. Neuropeptides and neurotrophins in neonatal blood of chil-
dren with Autism or mental retardation. Ann Neurol 2001; 49:597–606.
73. Rapin I. Autism. N Engl J Med 1997; 337:97–104.
74. Dwyer JM. Manipulating the immune system with immunoglobulin. N Engl J Med
1992; 326:4104–4109.
172 DelGiudice and Hollander
75. Gupta S, Agganual S, Heads C. Brief report: dysregulated immune system in chil-
dren with autism; beneficial effects of intravenous immune globulin on autistic
characteristics. J Autism Dev Disord 1996; 26:439–452.
76. DelGiudice-Asch G, Simon L, Schmeidler J, Cunningham-Rundles C, Hollander
E. A pilot open clinical trial of intravenous immunoglobulin in childhood autism.
J Autism Dev Disord 1998; 29(2): 157–160
77. Freeman BJ, Ritvo ER, Yokota A, Ritvo A. A scale for rating symptoms of patients
with the syndrome of autism in real life settings. J Am Acad Child Psychiatry
1986; 25:130-136.
78. Goodman W, Price L, Ramussen S, Mazure C. The Yale-Brown Obsessive Com-
pulsive Scale (Y-BOCS). Part I. Development, use, and reliability. Arch Gen Psy-
chiatry 1989; 46:1006–1011.
79. Goodman W, Price L, Ramusssen S, Mazure C. The Yale-Brown Obsessive Com-
pulsive Scale (Y-BOCS). Part II. Validity. Arch Gen Psychiatry 1989; 46:1012–
1016.
80. Guy W. E.C.D.E.U. Assessment Manual for Psychopharmacology. DHEW Publi-
cation 76-338. Washington, DC: U.S. Government Printing Office, 1976.
81. Rapoport J, Elkins R, Mikkelsen E. Clinical controlled trial of clomipramine in
adolescents with obsessive compulsive disorder. Psychopharm Bull 1980; 16:61–
63.
82. Selman K, Reine CS. Tumor necrosis factor mediates myelin and oligodendrocyte
damage in vitro. Ann Neurol 1988; 23:339–346.
83. Han J, Thompson P, Beutler B. Dexamethasone and pentoxifylline inhibit
endotoxin-induced cachectin: tumor necrosis factor synthesis at separate points in
the signaling pathway. J Exp Med 1990; 172:391–394.
84. Natal S, Louboutin J, Chabannes D. Pentoxifylline inhibits experimental allergic
encephalitis. Acta Neurol Scand 1993; 88:97–99.
85. Black RS, Barday LL, Nolan KA. Pentoxifylline in cerebrovascular dementia. J
Am Geriatr Soc 1992; 40:237–244.
86. Soganie S. On our experience in using pentoxifylline for abnormal behavior and
the autistic syndrome. Jap J Child Psychiatry 1978; 19:134–144.
87. Nakane A. Effect of pentoxifylline on autistic children. Tokepto Eisei Gakkashi
1980; 64:104–105.
88. Shimode M. Effect of pentoxifylline on infantile autism. Clin Exp Med 1981; 58:
285–288.
89. Turek S. Treatment of psychotic and autistic children with pentoxifylline. ASANA
1981; 1:51–60.
90. Fudenberg HH, Singh VK. Implications of immunomodulant therapy in Alzhei-
mer’s disease. Prog Drug Res 1988; 32:21–42.
91. Dawson CA, Lineham JH. Biogenic amines. In: Massaro E, ed. Lung Cell Biology.
New York: Marcel Dekker, 1989:1091.
92. Fudenberg HH. Dialysable lymphocyte extract (DLyE) in infantile onset autism:
a pilot study. Biotherapy 1996; 9:143–147.
93. Horvath K, Stefanatos G, Sokolski KN, Wachtel R, Nabors L, Tildon JT. Improved
social and language skills after secretin administration in patients with autistic
spectrum disorders. Assoc Acad Minor Phys 1998; 9:9–15.
Immune Dysfunction 173
94. Owley T, Steele E, Corsello C, Risi S, McKaig K, Lord C, Leventhal BL, Cook EH
Jr. A double-blind placebo-controlled trial of secretin for the treatment of autistic
disorder. Medscape Gen Med 1999; 1:10.
95. Sandler AD, Sutton KA, DeWeese J, Girardi MA, Sheppard V, Bodfish JW. Lack
of benefit of a single dose of synthetic human secretin in the treatment of autism
and pervasive developmental disorder. N Engl J Med 1999; 341:1801–1806.
96. Chez MG, Buchanan CP, Bagan BT, Hammer MS, McCarthy KS, Ovrutskaya I,
Nowinski CV, Cohen ZS. Secretin and autism: a two part clinical investigation.
J Autism Dev Disord 2000; 30:87–94.
97. Chez MG, Nowinski CV, Panchanan CP, Jones C. Donepezil (aricept) use in chil-
dren with autism spectrum disorders. Ann Neurol 2000; 48:541.
98. Rossi PG, Posar A, Parmeggiani A, Pipitone E, D’Agata M. Niaprazine in the
treatment of autistic disorder. J Child Neurol 1999; 14:547–550.
99. King BH, Cook EH, Sikich L, et al. A controlled trial of amantadine in the treat-
ment of autistic children. 46th Annual Meeting of the American Academy of Child
and Adolescent Psychiatry. Chicago, Oct 19–24, 2000.
100. Bolter ER. Autism and Clostridium tebani. Med Hypoth 1998, 51:133–144.
101. Sandler RH, Finegold SM, Bolte ER, Buchanan CP, Maxwell AP, Vaisanen ML,
Nelson MN, Wexler HM . Short-term benefit from oral vancomycin treatment of
regressive-onset autism. J Child Neurol 2000; 15:429–435.
9
Autism and Environmental Toxins
INTRODUCTION
The etiology of autism is not well understood, and most cases are of unknown
etiology. Autism is most likely a multifactorial disease in which genetics and
environmental factors combine to yield a wide range of phenotypes. As such,
heterogeneity is a hallmark of both the origins and clinical manifestations of
autism.
Numerous studies have implicated various environmental factors as etio-
logical agents for autism. In addition, the apparent rise in the global prevalence
of the disorder (if it is a real increase rather than a reflection of better ascertain-
ment) could indicate a significant role for nongenetic factors in disease pathogen-
esis (1). Reports of geographic clustering (1) and an observed seasonality of the
births of autistic children may similarly point to the importance of environmental
influences.
175
176 Evers et al.
However, the toxins and viruses under study may individually account for distinct
subsets of the disorder.
Much of the evidence concerning specific environmental factors is some-
what circumstantial. Although association is often established, firm causation is
more difficult to prove. Temporality, and therefore causality, are by nature diffi-
cult to deduce when examining prenatal and early postnatal influences in a syn-
drome typically diagnosed at 2 to 4 years of age. In addition, the detailed mecha-
nisms by which environmental factors alter brain development are largely
unknown. Despite such experimental difficulties, there is a compelling body of
research on the role of environmental pathogens in autism.
Specific proposed etiological factors are discussed later in this chapter.
Prevalence of Autism
The global prevalence of autism may be increasing. Nearly all epidemiological
studies done before the mid-1980s pointed to a prevalence of approximately 0.2–
.05 in 1000 (1). However, since 1985, 10 of 11 non-U.S. studies have indicated
rates at or in excess of 0.9 per 1000 (1). A prevalence of 1 or 2 per 1000 is
currently commonly cited (2). Some studies, however, have shown prevalence
in excess of 20 per 10,000 (1). Repeated surveys of Goteborg, Sweden, indicated
an autism prevalence of 4 per 10,000 in 1980, 7.6 per 10,000 in 1984, and 11.5
per 10,000 in 1988 (1)—nearly a threefold increase in less than a decade. It has
been estimated that the worldwide prevalence of autism is increasing by 3.8%
annually (1); this increase is not seen in U.S. data (2).
In the United States, the state of California reported an increase of 210%
in the autistic population accessing services of regional centers for the develop-
mentally disabled between 1987 and 1998 (2). This contrasts with an increase
of 60% in total population served by the centers over the same time period, and
an increase of 35-40% for disorders such as cerebral palsy and epilepsy.
Without comprehensive surveillance for the disorder, it is impossible to
state with a high degree of certainty the national or international prevalence of
autism. There appears to be a lower prevalence in the United States than the rest
of the world (3). However, we do not know whether the disorder’s occurrence
is uniform or varies markedly across regions or locales.
It is possible that the apparent rise in prevalence is actually a function of
better diagnosis and ascertainment rather than a true increase in cases of autism.
There is now greater recognition of a broader range of autism spectrum disorders;
current epidemiological studies may reflect only better physician diagnosis of a
complex heterogeneous syndrome. Such changes in diagnostic criteria hinder ef-
forts to track changes in prevalence over time. In addition, the consumption of
California mental health resources described above may be merely the increased
utilization of proffered services by a more aware and empowered public.
Environmental Toxins 177
One study assessed liver function in autistic subjects and controls using
three measures. Glucaric acid levels, a biomarker for liver stress brought on by
xenobiotic contamination or liver disease, were analyzed as an index of both liver
function and significance of xenobiotic exposure. Second, specific blood analyses
were conducted to detect toxins in the blood. Lastly, comprehensive liver detoxi-
fication evaluation was conducted to assess the ability of the liver to process
toxins without accumulation of free radical metabolites or a back-up of unpro-
cessed chemicals that would be stored in fatty tissue.
The results of this study were supportive of liver dysfunction in autism.
All autistic subjects displayed elevated glucaric acid levels and abnormal liver
detoxification profiles. In addition, all but two subjects showed blood levels of
a variety of toxic agents in excess of maximum adult tolerance levels. Interest-
ingly, the subjects all had different combinations of elevated toxins. The results
therefore implicated the inability of the liver to process a range of different xeno-
biotics, rather than the deleterious effect of a particular agent. However, most
subjects did exhibit elevated trimethylbenzenes (10).
A different study found that 90% of autistic subjects lacked phenosulfo-
transferase, an enzyme central to the detoxication process (11).
The developing CNS is most vulnerable to xenobiotic agents during the
fetal period, when it lacks a formed blood–brain barrier. Therefore, it is likely
that neurotoxicity to the forming brain would begin in utero. The autistic individ-
ual’s fetal liver is unable to detoxify ordinary environmental chemicals. Not prop-
erly processed, these lipophilic chemicals would then damage the individual in
two ways.
First, they could pass directly into the brain, causing direct injury to struc-
tures such as neurons, dendritic processes, and receptors. Mitochondrial DNA
could be damaged as well. Xenobiotics may also activate 2′5 A-synthetase and
protein kinase R (PKR); this would decrease the production of mRNA and, subse-
quently, proteins critical to neuronal function, such as axial fibrillary proteins
and dendrites. Dysfunction of neuronal proteins could lead to the brain pathology
and dysfunction seen in autism.
Xenobiotic agents could also trigger indirect brain injury via the provoca-
tion of an immune response. Toxic insult to the brain could lead to the release
of a variety of brain antigens into the peripheral circulation. These antigens would
then stimulate an autoimmune response, creating autoantibodies to a variety of
nervous system components. Different neuronal proteins, myelin, glial fibrillary
proteins, Purkinje cells, etc., could all be targeted. The autoimmune aspects of
autism are discussed elsewhere. It should be noted here that autoantibodies to
certain of the above nervous system components (e.g., myelin basic protein, neu-
ron-axon filament proteins) have been detected in autistic individuals (12).
This view of autistic pathogenesis is characterized by a great variety of
possible pathological outcomes. Pathology will vary based on numerous factors,
Environmental Toxins 181
including the severity of liver dysfunction; the specific chemicals involved; the
times, duration, and frequency of prenatal exposure to those factors; and the vigor
with which an autoimmune response is mounted to brain antigen. The variety of
pathological anomalies may relate to the variety of clinical manifestations of
autistic spectrum disorder.
cardiac and neuromuscular defect (20). Thus, autism associated with congenital
rubella is atypical in its prognosis, as well as the details of its attendant handicaps
and other clinical features.
The longitudinal studies following children with congenital rubella estab-
lished a strong association between maternal rubella infection and autism. Be-
yond proving association, studies on this population strongly suggested a patho-
genic role for rubella virus in autism via viral invasion of CNS and subsequent
brain damage. Rubella virus was recovered postmortem from the spinal fluid
and brain tissue of several of the rubella-infected children (18). The subsequent
recovery noted in many of the children, as well as the late onset of autism in
four other children, point to the disorder’s pathogenesis via chronic viral (CNS)
infection, in which recovery, worsening, and delayed effects may all take place.
Rubella may thus act as a “slow virus,” mediating chronic CNS damage, yielding
behavioral aberration as one of the ultimate results (21). Rubella has also been
implicated in chronic sclerosing panencephalitis, a chronic viral infection of the
CNS (21).
Prenatal rubella has also been associated with “partial autism,” or the broad
autistic phenotype. One study estimated that the virus was responsible for 13%
of partial autism cases, a very strong association, suggesting that partial autism
is a manifestation of congenital rubella (14). However, the 13% figure was
deemed an “unstable estimate” by the researchers, based on the small study size.
The same study indicated that the virus was also associated with “total” (classic)
autism. For the full phenotype the overall exposure rate of both the autism cases
and controls, and thus the percentage of autism cases attributable to the virus per
this study, was relatively low. Interestingly, rubella was the only virus in the
research in question to show an association with the partial autism phenotype;
the other virus looked at (pre- and postnatal mumps) was associated only with
the classic phenotype.
An epidemiological survey of autism in Utah revealed one autistic proband
with congenital rubella and one proband with possible congenital rubella (22).
The survey discovered 12 rare diseases (infectious and otherwise) with known
CNS pathology present in 233 autistic individuals (11% of those surveyed). The
presence of 12 different rare diseases in a relatively large proportion of those
with an additional rare disease (autism) caused the researchers to suggest or infer
that CNS pathology secondary to the rare conditions was an etiological factor
for autism in those individuals. Actual neuropathology was not assessed; the in-
ference was based on the high mathematical probability that such a convergence
of comorbidity would not occur in a random fashion.
Multiple studies have reported a wide range of CNS damage and dysfunc-
tion in conjunction with viral infection of CNS and panencephalitis due to con-
genital rubella (20). Observed damage has ranged from mild cognitive disability
to severe retardation and neuronal death, gliosis, and vasculitis (23). A study of
184 Evers et al.
Human Herpesvirus-6
Human herpesvirus-6 (HHV-6)–antibody-positive sera from autistic children has
been associated with the presence of two brain autoantibodies: anti–myelin basic
protein (anti-MBP) and anti–neuron-axon filament protein (anti-NAFP). These
are autoantibodies found by separate studies in autistic children (12). Nonautistic
HHV-6-antibody-positive controls showed no such association with anti-brain
antibodies. In addition, higher HHV-6-antibody titer levels correlated with a
greater likelihood of the presence of brain autoantibodies.
HHV-6 has been shown to have neurological sequelae (12). It has also
been linked to demyelination (in multiple sclerosis). Perhaps most importantly,
it displays “molecular mimicry” with MBP and NAFP, two common targets of
brain autoantibodies found in autistic individuals. Molecular mimicry occurs
when some aspect of a bacterial or viral pathogen resembles some part of the
host (such as a protein found in a particular tissue). Antibodies to the pathogen
made by the host may then mistakenly attack this aspect of self (as well as the
pathogen). This antibody-mediated assault on the self, caused by molecular mim-
icry, may be a pathogenic event in some autism cases. HHV-6 infection in certain
individuals could lead to the production of antibodies that are cross-reactive with
the aforementioned proteins (MBP and NAFP) in the brain. Subsequent antibody-
mediated insult to the brain would then produce autistic pathology and symptom-
atology. Alternatively, any pathogenesis could result from demyelination or some
yet unknown mechanism.
It should be noted that multiple studies have not shown a difference in
rates of herpes simplex virus or human herpesvirus infection between autism and
control populations (12,28).
Herpes Simplex
Several case studies have linked herpes simplex encephalitis to autism, albeit
with differing clinical features. DeLong et al. (29) reported on an 11-year old
girl who developed an autistic syndrome secondary to an acute encephalopathic
illness. The syndrome was characterized by such autistic features as stereotypies,
withdrawal, echolalia, diminution of spontaneous verbalization, perseveration,
global cognitive dysfunction and lack of imaginative play or gestures other than
pointing. The girl displayed elevated herpes simplex titers, supporting a diagnosis
186 Evers et al.
Mumps
A study of the relationship of viral exposures in prenatal and early postnatal life
found a positive association between both prenatal and postnatal mumps and
autism (14). The more novel finding was probably the association of postnatal
mumps and autism, as most viral associations with the disorder have been prena-
tal. However, the study estimated that the overall proportion of autism cases
attributable to such mumps exposures was likely very small. Mumps in animals
has been shown to block cerebrospinal fluid and lead to mild hydrocephalus (27).
sures than autistic children with lower oxytocin levels; this was the opposite of
findings for normal subjects. Oxytocin levels increased with age in normal con-
trols but not in autistic subjects. The oxytocin system is different in males and
females, a significant fact given that autism is a disorder seen predominantly in
males. Further, oxytocin and vasopressin receptors are expressed more in the
developing brain (46); this is consistent with the developmental nature of autism.
Pitocin is a synthetic analog of the hormone oxytocin. It is routinely used
to induce labor or support uterine contraction during deliveries. While oxytocin
has a short plasma half-life and does not cross the blood–brain barrier in adults,
its distribution in, and consequences for, the neonate are unknown (47). There
has been speculation that induction of labor with pitocin somehow disrupts the
oxytocin system of genetically susceptible neonates, ultimately leading to the
social deficits of autism. A firm association of autism with this use of pitocin
has not been established. While a high incidence of pitocin-induced labor has
been observed in a clinical population of autistic patients (48), another survey
found no such relationship (49).
Analysis of more data is necessary to resolve this question with any cer-
tainty. Numerous potential confounding factors surround such surveys, such as
other medication use, difficulties in delivery, and maternal history. Even if an
association is ultimately discerned, causality may be more difficult to establish.
It is possible, for instance, that pitocin induction is necessitated by abnormalities
already present in an infant destined to develop autism.
Measles, Mumps, and Rubella Vaccine
There has been speculation that some cases of autism have been caused by the
measles-mumps-rubella (MMR) vaccine. This speculation was triggered primar-
ily by a case series study in the United Kingdom in which 12 children with a
history of normal development developed chronic enterocolitis and regressive
developmental disorder (50). The parents of eight of the children identified the
MMR vaccine to be the immediate precursor of developmental regression. This
seeming temporal association, widely reported in the media, led to a fear that
MMR administration is a causal agent for autism.
Large-scale epidemiological studies have since contradicted any causal link
between the MMR vaccine and autism. A study of all children with autism born
in eight U.K. health districts since 1979 found no change in trend in incidence
or age at diagnosis related to the introduction of MMR vaccination to the United
Kingdom in 1988 (51). The study found no temporal association between onset
of autism within 1 or 2 years after vaccination. In addition, developmental regres-
sion was not clustered in the months after vaccination. The weight of epidemio-
logical data to date thus supports the safety of MMR vaccination.
The British medical journal Lancet has further pointed out problems with
the presentation and quality of evidence presented by vaccination opponents (52).
Environmental Toxins 193
Allergies
There is a high prevalence of allergic disorders among autistic individuals
(57,58). It has been suggested that food allergies and the toxicity of certain food
peptides may affect the CNS and be involved in the pathogenesis of autism.
Antibody reactivity to certain common foods is higher among autistic individuals
than among normal controls; the removal of these items from the diet has im-
proved behavioral symptoms.
SUMMARY
The etiology of autism is not well understood, and most cases are of unknown
etiology. Autism is most likely a multifactorial disease in which genetics and
environmental factors combine to yield a wide range of phenotypes.
There is a growing body of compelling evidence in support of an environ-
mental pathogenesis for some cases of autism. Numerous studies have implicated
various environmental factors as etiological agents for the disease. No single
agent has been put forth to explain all, or even most, of autism cases. However,
the toxins and viruses under study may individually account for distinct subsets
of the disorder.
An apparent rise in the global prevalence of autism, reports of geographic
clustering of the disease, and an observed seasonality of the births of autistic
children may similarly point to the importance of environmental influences.
FUTURE DIRECTIONS
Further research on the role of environmental agents in the pathogenesis of autism
is necessary.
Improving study quality—Some studies have not controlled for key vari-
ables: medical history, medication use, environment, demographics, diagnostic
criteria, control group composition, etc. Research has also been limited by small
Environmental Toxins 195
ACKNOWLEDGMENT
This chapter was supported in part by a grant from the Seaver Foundation.
REFERENCES
1. London E, Etzel R. The environment as an etiologic factor in autism: a new direction
for research. Environ Health Perspect 2000; 108(suppl 3):401–404.
2. Goldman L, Koduru S. Chemicals in the environment and developmental toxicity
to children: a public health and policy perspective. Environ Health Perspect 2000;
108(suppl 3):443–448.
3. Center for Children’s Health and the Environment. Autism and Environmental Ex-
posures. 2000.
4. Barak Y, Ring A, Sulkes J, Gabbay U, Elizur A. Season of birth and autistic disorder
in Israel. Am J Psychiatry 1995; 152(5):798–800.
5. Bolton P, Pickles A, Harrington R, Macdonald H, Rutter M. Season of birth: issues,
approaches and findings for autism. J Child Psychol Psychiatry 1992; 33(3):509–
530.
6. Gillberg C. Do children with autism have March birthdays? Acta Psychiatr Scand
1990; 82:152–156.
7. Rodier P, Ingram J, Tisdale B, Nelson S, Romano J. Embryological origin for au-
tism: developmental anomalies of the cranial nerve motor nuclei. J Compr Neurol
1996; 370:247–261.
8. Stromland K, Nordin V, Miller M, Akerstrom B, Gillberg C. Autism in thalidomide
embryopathy: a population study. Dev Med Child Neurol 1994; 36:351–356.
196 Evers et al.
46. Insel T, O’Brien J, Leckman J. Oxytocin, vasopressin, and autism: is there a connec-
tion? Biol Psychiatry 1999; 45(2):145–157.
47. Insel T. A neurobiological basis of social attachment. Am J Psychiatry 1997; 154(6):
726–735.
48. Hollander E, Cartwright C, Wong C, et al. A dimensional approach to the autism
spectrum. CNS Spectrums: Int J Neuropsychiatr Med 1998; 3:22–40.
49. Fein D, Allen D, Dunn D. Pitocin induction and autism. Am J Psychiatry 1997;
154(3):438–439.
50. Wakefield A, Murch S, Anthony A, Linnell J, Casson D, Malik M, Berelowitz M,
Dhillon A, Thomson M, Harvey P, Valentine A, Davies S, Walker-Smith J. Ileal-
lymphoid-nodular hyperplasia, non-specific colitis, and pervasive developmental
disorder in children. Lancet 1998; 351:637–641.
51. Taylor B, Miller E, Farrington C, Petropoulos M, Favot-Mayaud I, Li J, Waight P.
Autism and measles, mumps and rubella vaccine: no epidemiological evidence for
a causal association. Lancet 1999; 353:2026–2029.
52. Measles, MMR, and autism: the confusion continues [editorial]. Lancet 2000; 355:
1379.
53. Nanson J. Autism in fetal alcohol syndrome: a report of six cases. Alcohol Clin
Exp Res 1992; 16(3):558–565.
54. Davis E, Fennoy I, Laraque D. Autism and developmental abnormalities in children
with perinatal cocaine exposure. J Natl Med Assoc 1992; 84(4):315–319.
55. Ferriero D, Partridge J, Wong D. Congenital defects and stroke in cocaine exposed
neonates. Ann Neurol 1988; 24:348.
56. Ferriero D, Wong D, Townsend R, et al. Neurological complications in infants of
cocaine abusing mothers. Neurology 1988; 38(suppl 1):163.
57. Bidet B, Leboyer M, Descours B, Bouvard M, Benveniste J. Allergic sensitization
in infantile autism [letter]. J Autism Dev Disord 1993; 23(2):419–420.
58. Renzoni E, Beltrami V, Sestini P, Pompella A, Menchetti G, Zappella M. Brief
report: allergological evaluation of children with autism. J Autism Dev Disord 1995;
25(3):327–333.
10
Neurobiology of Serotonin Function
in Autism
INTRODUCTION
Serotonin [5-hydroxytryptamine (5-HT)] neurons are widely distributed through-
out the mammalian brain. This neuronal system is one of the earliest to develop,
and the turnover rate of 5-HT is higher in the immature mammalian brain than
at any other time in life. Serotonin plays a key role as a growth factor in the
immature brain, directing both proliferation and maturation (1). For these reasons
and others, 5-HT has been a target of investigation into the pathophysiology of
autistic disorder (autism) for nearly 50 years. This chapter reviews results from
studies of peripheral and central neurochemistry, behavioral/neuroendocrine
challenges, neuroimaging, and genetics related to 5-HT function in autism. The
reader is referred to other excellent reviews of this topic for additional informa-
tion (2–4). We first recount the historical developments that led, in part, to studies
of 5-HT function in autism.
199
200 McDougle et al.
HISTORICAL PERSPECTIVE
The landmark publication by Schain and Freedman in 1961 (5) is often acknowl-
edged as the first paper to describe an abnormality in 5-HT function in autism. Al-
though it was the first report of elevated whole-blood serotonin (WBS) levels in
subjects clearly defined as autistic, at least one prior study had been published regard-
ing dysregulation in other aspects of 5-HT function in this disorder (6) (see below).
In their classic paper (5), Schain and Freedman refer to four previously
published articles: three by Pare, Sandler, and Stacey (7–9), in which results
from studies of 5-HT function in “mental defectives” (in today’s terminology,
mental retardation) are described, and the other by Sutton and Read (6). It was
based on these data, in part, that Schain and Freedman pursued their studies of
5-hydroxyindole metabolism in children with autism. Because of the importance
of these early investigations to the subsequent work of Schain and Freedman,
we describe the results of these studies in some detail.
In their first study, Pare, Sandler, and Stacey (7) reported significantly lower
serum 5-HT levels (mean ⫾ SEM ⫽ 57 ⫾ 11 ng/ml) in subjects with phenylketo-
nuria (PKU) compared with children awaiting tonsillectomy (124 ⫾ 14 ng/ml)
and mentally defective controls (270 ⫾ 51 ng/ml). Similarly, mean values of
urinary 5-hydroxyindoleacetic acid (5-HIAA), the primary metabolite of 5-HT,
were significantly lower in the group with PKU (2.9 ⫾ 0.2 mg/g creatinine)
than in the mentally defective control group (4.9 ⫾ 0.7 mg/g creatinine), normal
children (6.6 ⫾ 0.9 mg/g creatinine), and children awaiting tonsillectomy (11.6
⫾ 1.3 mg/g creatinine). No correlation was found between the intelligence quo-
tient (IQ) of subjects with PKU and their serum 5-HT and urinary 5-HIAA excre-
tion. The investigators suggested that the decreased levels of serum 5-HT and
urinary 5-HIAA might be due to competition between phenylalanine and trypto-
phan, an essential amino acid and precursor of 5-HT, for the same hydroxylating
system. Furthermore, they hypothesized that, as a result, impaired 5-HT synthesis
may contribute to the causation of mental deficiency in subjects with PKU. This
hypothesis was based in part on the fact that 5-HT had recently been shown to
be normally present in the central nervous system (10). The investigators stated
that their studies were based on previous findings from Armstrong and Robinson
(11,12) that provided evidence of abnormal indole metabolism in PKU; Arm-
strong and Robinson had found indolelactic acid in the urine of affected subjects
and decreased urinary excretion of 5-HIAA in some of their subjects. Pare, San-
dler, and Stacey (7) thus hypothesized that the 5-HT pathway might be abnormal
in PKU and that such an anomaly might contribute to the nature of the mental
defect which remained unexplained and which had not been clearly linked to the
defective hydroxylation of phenylalanine to tyrosine.
In a subsequent study, Pare et al. (8) found that serum 5-HT levels increased
significantly, from 90 ng/ml to 142.3 ng/ml, in seven subjects with PKU adminis-
Neurobiology of Serotonin Function 201
tered a low-phenylalanine diet. The investigators stated that these results might
be explained by diminished competition between phenylalanine and tryptophan
for hydroxylation. In this same paper, the authors also described results from
a study designed to address the possibility that the decreased 5-hydroxyindole
production in PKU might be due to an inhibition of 5-hydroxytryptophan (5-
HTP) decarboxylase, which converts 5-HTP to 5-HT, by phenylacetic acid, phen-
ylpyruvic acid, and phenyl-lactic acid. This inhibition had been shown to occur
in vitro, and it had also been demonstrated that subjects with PKU excreted these
acids in excess. To test this hypothesis, the investigators administered 5-HTP 25
mg intravenously to four children with PKU and four age- and gender-matched
mentally defective children without PKU. The children with PKU gave apprecia-
bly lower peaks for the urinary excretion of 5-HT and 5-HIAA. Unchanged 5-
HTP was also found in postinjection samples. The investigators stated that these
results were in agreement with the postulated inhibition of 5-HTP decarboxylase
in PKU.
In the third paper from the series by Pare et al. (9) referenced by Schain
and Freedman (5), the group extended and replicated their original findings. In
49 subjects with PKU and 32 mentally defective controls, the investigators found
significantly lower values of urinary 5-HIAA in the subjects (2.2 mg/g creatinine)
compared with those of the controls (7.2 mg/g creatinine). Likewise, serum levels
of 5-HT were lower in the subjects (71.2 ng/ml) than the controls (283 ng/ml).
Contrary to their working hypothesis, there was no significant association be-
tween IQ and either 5-HT or 5-HIAA levels in the subjects with PKU. In this
report, it was pointed out that some of the mentally defective controls had values
for serum 5-HT that were as high as those found in patients with the carcinoid
syndrome (Figure 1). These data, in addition to observations of the potent percep-
tual effects of serotonergic hallucinogens, such as lysergic acid diethylamide
(LSD) (13), prompted Schain and Freedman’s early studies of WBS in autism.
Pare, Sandler, and Stacey summarized their studies by reporting that enzymes
from multiple other neurochemical systems, including catecholamine and
gamma-aminobutyric acid (GABA) systems, had recently been shown to be af-
fected by phenylalanine metabolites. Because of this multisystem involvement,
they stated that the failure to demonstrate any correlation between the degree of
5-hydroxyindole deficiency and IQ in the subjects with PKU was “hardly sur-
prising.”
her personality with an associated mental regression over the next 4 to 5 months.
As part of a metabolic assessment, urine samples were obtained from the subject,
her sibling, and each of their parents. In the children, but not the parents, a striking
departure from normal was observed on the amino acid chromatograms. The
pattern consisted of very low levels of glutamine and consistent excretion of
GABA along with a moderate increase in glutamic acid, but not in abnormal
amounts. The subject and a 20-month-old male control were given 0.25 g/kg of
L-tryptophan orally. In response to this challenge, the subject failed to excrete
detectable amounts of indoleacetic acid and idolelactic acid, and unchanged tryp-
tophan. The excretion of metabolites characteristic of the kynurenine pathway
(an alternative pathway of metabolism for tryptophan, rather than metabolism to
5-HT) were similar in the subject and control child. Subsequently, three more
control children were tested. The subject excreted the lowest amount of indolelac-
tic acid, indoleacetic acid, and 5-HIAA. The subject’s baseline levels of indole
metabolites were normal, but she was apparently limited in her ability to convert
large quantities of tryptophan to indolelactic acid, indoleacetic acid, and 5-HIAA.
The subject’s sibling continued to develop normally to the then present age of 16
months. It was not possible to test her ability to metabolize tryptophan. The authors
Neurobiology of Serotonin Function 203
concluded that the subject’s mental aberration was the result of an altered ability
to maintain normal brain 5-HT levels. To our knowledge, this study by Sutton and
Read (6) was the first published assessment of 5-HT function in autism.
The study by Schain and Freedman (5) involved 23 children diagnosed
with “infantile autism,” ages 6 to 18 years (average age of 10.8 years), who were
institutionalized at the Southbury Training School in Connecticut. Blood and
urine were obtained for determination of 5-HT and 5-HIAA, respectively. Other
“defective” children, roughly matched for age and sex, were also studied. Multi-
ple determinations of WBS were made in all the children.
The subjects studied were divided into three groups. One (group A) in-
cluded mildly retarded children with IQs of 60 to 80 described as “familial ‘sub-
cultural’ defective children, high-grade Mongols.” The second (group B) included
children with the diagnosis of “infantile autism” who functioned on a severely
retarded level. These children were described as being usually not trainable, and
it was mentioned that they were segregated with other low-grade retarded chil-
dren. The third (group C) consisted of severely retarded children without a diag-
nosis of autism (IQ less than 20). These children had diagnoses including “con-
genital cerebral defect” and “diffuse encephalopathy.”
The mildly retarded children (group A) (n ⫽ 12) had WBS levels averaging
0.072 gamma/cc, with a range of 0.042 to 0.156 gamma/cc, which was similar
to the normal values obtained in the investigators’ laboratory. The autistic chil-
dren (group B) (n ⫽ 23) had average WBS levels of 0.141 gamma/cc, with a
range of 0.033 to 0.540 gamma/cc. Six of the 23 autistic children had mean levels
over 0.200 gamma/cc, a value significantly above the range of normal for the
investigators’ laboratory (0.02 to 0.15 gamma/cc) (Figure 2). By comparison,
the investigators stated that values for adult chronic schizophrenic patients tested
in their laboratory were within the normal range. A small group of nonautistic
low-grade defective children (group C) (n ⫽ 7) had an average WBS concentra-
tion of 0.128 gamma/cc, which was not significantly different from that of the
autistic group as a whole. Some of the children in the study were receiving pheno-
barbital, Dilantin, or chlorpromazine, although none of these drugs seemed to be
associated with any changes in WBS levels.
Urine 5-HIAA levels were higher in the autistic group (5.9) (n ⫽ 12) than
in the mildly retarded group (1.6) (n ⫽ 6) when expressed as gamma/mg of
creatinine. The absolute amount of 5-HIAA/ml of urine was similar in both
groups. Creatinine values, however, were much lower in the autistic children,
indicating greater dilution of urine. Urine 5-HIAA levels were not reported for
the children in group C. The investigators stated that none of the 5-HIAA results
could be considered significantly abnormal.
Tryptophan loads consisting of 1 g of L-tryptophan daily for 3 days were
given to four autistic children. This resulted in no consistent change in WBS
levels.
204 McDougle et al.
Figure 2 Mean blood 5-HT levels for individuals of each group. (From Ref. 5.)
In the discussion section of their paper (5), Schain and Freedman stated
that the results of their study confirmed the impressions of Pare et al. (9) that
mentally defective children have elevated levels of 5-HT in blood. In particular,
Schain and Freedman emphasized that the elevations of 5-HT in the blood of
their subjects tended to be present only in some of the more severely defective
children. They went on to say that consistent unusual elevations of WBS were
found only in the autistic children, although the mean WBS level of the other
severely retarded group was higher than that of the mildly retarded group. The
investigators could not find any differences in the presenting symptomatology
between the six autistic children with the highest WBS levels and those who had
normal levels. The only feature they found noteworthy was the absence of seizure
disorders in the six autistic children with elevated WBS levels.
In 1970, Ritvo and colleagues (14) published a study whose results repli-
cated those of Schain and Freedman. WBS levels were determined in 24 autistic
children, ages 33 to 91 months, and 82 controls consisting of hospital staff, their
children, children hospitalized for neurotic and behavior disorders, and paid adult
and child volunteers. The controls ranged in age from less than 23 months to
greater than 360 months. All subjects had been drug-free for at least 3 months.
An inverse relationship between age and both WBS levels and platelet values
was identified in the control group. A comparison of WBS levels between the
24 autistic children and 36 age-matched controls demonstrated significantly
Neurobiology of Serotonin Function 205
higher values in the autistics (mean ⫾ SD ⫽ 0.263 ⫾ 0.063 µg/ml vs. 0.216 ⫾
0.061 µg/ml). Mean 5-HT per platelet values were not significantly different
between age-matched groups of autistics and controls.
In 1987, Anderson and other investigators from Yale published results
from their laboratory and reviewed and summarized data on WBS levels in au-
tism to that date (Table 1) (15). WBS and tryptophan were measured in 87 nor-
mal children and young adults and in 40 autistic subjects with a similar age
distribution. WBS concentrations were significantly higher in the drug-free autis-
tic subjects (mean ⫾ SE ⫽ 205 ⫾ 16 ng/ml) (n ⫽ 21) than in normal subjects
(136 ⫾ 5.4 ng/ml) (n ⫽ 87). When the 95th percentile of the normal group was
used to define “hyperserotonemia” (WBS greater than 220 ng/ml), 38% of the
autistic subjects were determined to be hyperserotonemic. The investigators did
not find that the elevation in WBS was due to a particular subgroup of autistic
subjects. The autistic subjects who were receiving anticonvulsants or typical
neuroleptics had significantly lower WBS levels than did the drug-free subjects.
An age effect was observed in young normal males only, as young boys had
higher WBS levels (181 ⫾ 15 ng/ml) (n ⫽ 9) than adult males (138 ⫾ 12 ng/
ml) (n ⫽ 17). Mean whole-blood tryptophan levels and platelet counts were no
different between the autistic and normal groups. The investigators concluded
that, while causal mechanisms for elevated WBS in autism remained elusive,
relative to other possibilities, it seemed more likely that an alteration in platelet
function accounted for the hyperserotonemia. They pointed out that the problem
of the link between central and peripheral regulation of 5-HT metabolism re-
mained to be clarified in order to deduce the functional “meaning” of deviant
peripheral measures.
In a subsequent study, McBride et al. (16) re-evaluated platelet 5-HT in
autism, measuring and controlling for effects of race and puberty. In addition,
the specificity of hyperserotonemia for autism vs. cognitive impairment was as-
sessed. Prepubertal autistic children (n ⫽ 58) had significantly higher 5-HT con-
centrations than prepubertal controls (n ⫽ 38), although the elevation (25%) was
less than typically reported. Twenty-two mentally retarded or otherwise cogni-
tively impaired prepubertal children without autistic features (n ⫽ 22) had levels
similar to those of the normal controls. White children had significantly lower
5-HT levels than black or Latino youngsters, regardless of diagnosis. Diagnosis
and race effects were nonsignificant in the postpubertal group. The investigators
emphasized, however, that the observed absence of a significant or substantial
elevation in platelet 5-HT in postpubertal autistic subjects was based on a rela-
tively small number of subjects. They stated that when 5-HT was expressed as
ng/ml blood, the postpubertal autistic group showed a modest (15%) elevation in
the mean level that might have proved statistically significant in a larger sample.
Postpubertal subjects had lower 5-HT concentrations than prepubertal subjects.
The investigators concluded that the prevalence of hyperserotonemia in autistic
206
Schain and Freedman 1961 Bioassay Whole blood 141 ⫾ 78 ng/ml, N ⫽ 23 65 ⫾ 17 ng/ml, N ⫽ 4 normals Mean autistic age 10.8.
72 ⫾ 33 ng/ml, N ⫽ 12 (mildly
retarded)
Ritvo, Yuwiler, Geller, 1970 Acid fluorescence Whole blood 263 ⫾ 63 ng/ml, N ⫽ 23 216 ⫾ ng/ml, N ⫽ 36 3–8-yr-old subjects; levels de-
Ornitz, Saeger, and clined with age; no group differ-
Plotkin ences when expressed as per
platelet.
Yuwiler, Ritvo, Bald, 1971 Acid fluorescence Whole blood 272 ⫾ 53 ng/ml, N ⫽ 7 183 ⫾ 28 ng/ml, N ⫽ 4 8 a.m. samples; similar group dif-
Kyper, and Kopen 760 ⫾ 113 ng/10 platelets, 494 ⫾ 42 ng/10 platelets, ferences seen at noon. No circa-
N⫽7 N⫽4 dian rhythm seen.
Campbell, Friedman, 1974 Acid fluorescence Platelet-rich 280 ng/ml, N ⫽ 11 170 ng/ml, N ⫽ 6 Mean age autistics 5; mean age
DeVito, Greenspan, plasma normals 6.
and Collins
Yuwiler, Ritvo, Geller, 1975 Acid fluorescence Whole blood 273 ⫾ 30 ng/ml, N ⫽ 12 205 ⫾ 17 ng/ml, N ⫽ 12 Platelet 5-HT uptake, efflux similar
Ornitz, and Saeger 911 ⫾ 106 ng/10 9 platelets, 650 ⫾ 48 ng/10 9 platelets, in both groups. Mean autistic
N ⫽ 12 N ⫽ 12 and normal ages 5 and 8 yrs, re-
spectively.
McDougle et al.
Goldstein and Coleman 1976 Acid fluorescence Whole blood 86 ⫾ 36 ng/ml, N ⫽ 72 73 ⫾ 23 ng/ml, N ⫽ 71 Age-matched children.
Hanley, Stahl, and 1976 Acid fluorescence Whole blood 135 ⫾ 57 ng/ml, N ⫽ 27 57 ⫾ 49 ng/ml, N ⫽ 6 normals 7–23-yr-old subjects; no age effect
Freedman 97 ⫾ 38 ng/ml, N ⫽ 23 (mildly seen; urinary 5-HIAA also higher
retarded) in autistics.
Takahashi, Kanai, and 1976 Ninhydrin fluo- Platelet pellet 980 ⫾ 357 ng/mg, N ⫽ 30 807 ⫾ 202 ng/mg, N ⫽ 30 Expressed as ng/mg platelet pro-
Miyamoto rescence tein; mean age 5 yrs; no age effect
from 2–12 yrs.
Hoshino, Kumashiro, 1979 Acid fluorescence Serum 218 ⫾ 79 ng/ml, N ⫽ 42 175 ⫾ 60 ng/ml, N ⫽ 320 Mean age autistics 5.7; mean age
and Kaneko normals 9.3.
Hoshino, Yamamoto, 1984 Acid fluorescence Whole blood 173 ⫾ 62 ng/ml, N ⫽ 37 124 ⫾ 44 ng/ml, N ⫽ 67 Tryptophan elevated in autistic sub-
Kaneko, Tachibana, Wa- jects.
tanabe, Ono, and Ku-
mashiro
Anderson et al. 1987 HPLC fluoro- Whole blood 205 ⫾ 73 ng/ml, N ⫽ 21 136 ⫾ 50 ng/ml, N ⫽ 87 Mean age autistics 14.5; mean age
metric 776 ⫾ 348 ng/10 9 platelets, 522 ⫾ 213 ng/10 9 platelets, normals 14.6; drugs reduced 5-HT
Neurobiology of Serotonin Function
Figure 3 Whole-blood serotonin (5-HT) levels among autistic patients aged less than
16 years (n ⫽ 55) and controls (n ⫽ 91) according to age. (From Ref. 18, by permission
of Elsevier Science. Copyright 1999 by the Society of Biological Psychiatry.)
subjects and controls (Figure 3). This was due to the fact that among controls,
WBS values diminished with age, whereas WBS levels among autistic subjects
appeared to be age-independent.
Several studies have identified positive correlations between either platelet
5-HT (19) or WBS (20–22) levels of autistic children and WBS levels of their
parents and siblings. Autistic children with siblings affected with autism have
also been shown to have higher WBS levels than autistic probands without af-
fected siblings (23). As part of the study described above, Leboyer et al. (18)
sought to determine whether elevated WBS levels may be associated with genetic
liability to the development of autism. The sample included the 62 subjects with
autism, 122 first-degree relatives (61 mothers, 42 fathers, 11 sisters, and 8 broth-
ers), and a group of healthy subjects (n ⫽ 118) age-matched for first-degree
210 McDougle et al.
relatives over 16 years. As compared with the 118 controls above 16 years of
age (0.42 ⫾ 0.14 µmol/L), mean WBS levels were higher in the 42 fathers (0.79
⫾ 0.32 µmol/L), the 61 mothers (0.85 ⫾ 0.21 µmol/L), and the 8 siblings greater
than age 16 (1.04 ⫾ 0.14 µmol/L). In fact, only 12 mothers (20%) and 4 fathers
(9.5%) exhibited WBS levels below 0.70 µmol/L vs. 94% of the 118 controls
older than 16 years. Considering the whole population of first-degree relatives,
31/61 (51%) mothers, 19/42 (45%) fathers, and 7/8 siblings older than 16 years
(87%) had hyperserotonemia (WBS levels greater than 0.90 µmol/L). The inves-
tigators concluded that such familial aggregation of quantitative variables within
first-degree relatives of individuals with autism might enhance the search for
genetic vulnerability factors in autism.
In summarizing results from studies of WBS in autism, many but not all
investigations have found elevated WBS levels in younger autistic subjects that
tend to remain higher than those of normal controls across the age range. In
contrast, most studies of normal subjects have demonstrated an age-related de-
cline in WBS levels with increasing age. Some investigators have suggested that
these results could be explained, in part, by abnormal maturational processes of
the 5-HT system in autistic subjects (15,18). Additional factors that may affect
WBS levels include race, pubertal status, and treatment with psychotropic medi-
cation. Whether WBS levels will prove to be a useful quantitative measure in
the search for genetic susceptibility to autism remains to be determined.
In general, studies of urinary excretion of 5-HIAA (24) and whole-blood
tryptophan concentrations (15) have not found significant differences between
autistic subjects and controls.
Figure 4 Clinician-rated global severity change scale scores at the ⫹300-minute period
(5 hours after the drink) after tryptophan depletion (11 of 17 patients significantly worse)
and sham depletion (none of 17 patients significantly worse) for 17 patients with autism
(p ⫽ 0.001, Fisher exact test). (From Ref. 33. Copyright 1996 by the American Medical
Association.)
lactin release was identified in the autistic subjects in response to a 60-mg oral
dose of the indirect 5-HT agonist fenfluramine (32).
As a follow-up to these studies, McDougle et al. (33) employed an acute
tryptophan-depletion paradigm in 17 drug-free adults with autism. Tryptophan
is the essential amino acid necessary for the production of 5-HT within the CNS.
Administration of tryptophan-free amino acid mixtures within this paradigm have
been shown to deplete plasma levels of tryptophan and CSF levels of tryptophan
and 5-HIAA within 5 hours in humans (34).
Administration of the tryptophan-free amino acid mixture led to a marked
and significant reduction in plasma levels of both free and total tryptophan at 5
hours postingestion. In contrast, administration of a similar amino acid mixture
containing tryptophan (sham depletion) led to a significant increase in plasma
free and total tryptophan, as one would expect following normal food intake.
Behavioral effects were observed and quantitated in a double-blind fashion using
standardized, validated rating scales. Eleven of the 17 subjects who completed
both test days showed a significant worsening in symptoms following short-term
tryptophan depletion. In contrast, none of the 17 subjects demonstrated a signifi-
cant change in clinical status from baseline following sham depletion (Figure 4).
212 McDougle et al.
NEUROIMAGING STUDIES
To our knowledge, results from only two studies that assessed central 5-HT func-
tion in autism with neuroimaging techniques have been published. In the first of
these investigations, using alpha-[11C]methyl-L-tryptophan (AMT) as a tracer
for 5-HT synthesis with positron emission tomography (PET), eight autistic chil-
dren (seven boys, one girl; ages 4.1–11.1 years; mean age 6.6 years) and five of
their siblings (four boys, one girl; ages 8.2–14.4 years; mean age 9.9 years) were
studied (37). The investigators reported “gross” asymmetries of 5-HT synthesis
in frontal cortex, thalamus, and cerebellum in all seven autistic boys but not in
the one autistic girl. Decreased [11C]AMT accumulation was seen in the left
frontal cortex and thalamus in five of the seven boys. This was accompanied by
elevated [11C]AMT accumulation in the right dentate nucleus of the cerebellum,
confirmed through PET/magnetic resonance imaging image coregistration. The
investigators noted that the dentate nucleus is not visualized with [11C]AMT in
normal adults. In the other two autistic boys, [11C]AMT accumulation was de-
creased in the right frontal cortex and thalamus and elevated in the left dentate
nucleus. No gross asymmetries were seen in the frontal cortex or thalamus of
the sibling group; however, one sibling showed increased [11C]AMT accumula-
tion in the right dentate nucleus. Interestingly, this boy had a history of calendar
calculation and he ritualistically lined up his toys. The overall difference for
Neurobiology of Serotonin Function 213
asymmetry scores between the autistic boys and siblings was statistically signifi-
cant. Differences for regional asymmetry scores in the frontal cortex and thalamus
were also statistically significant. The asymmetry scores for the dentate nucleus
approached but did not reach statistical significance. The investigators concluded
that these focal alterations in [11C]AMT accumulation may represent either aber-
rant innervation by 5-HT terminals or altered function in anatomically normal
pathways.
The second study on neuroimaging, also by Chugani et al. (38), was de-
signed to determine whether brain 5-HT synthesis capacity is higher in children
or adults and whether there are differences in 5-HT synthesis capacity between
autistic and nonautistic children. Serotonin-synthesis capacity was measured in
autistic and nonautistic children at different ages, using [11C]AMT and PET.
Global brain values for 5-HT synthesis capacity (K-complex) were obtained for
autistic children (24 boys and 6 girls; age range, 2.3–15.4 years; mean age, 6.41
⫾ 3.3 years), 8 of their nonautistic siblings (6 boys and 2 girls; age range, 2.1–
14.4 years; mean age, 9.18 ⫾ 3.4 years), and 16 children with epilepsy without
autism (9 boys and 7 girls; age range, 3 months to 13.4 years; mean age, 5.73
⫾ 3.6 years). K-complex values were plotted according to age and fitted to linear
and five-parameter functions, to determine developmental changes and differ-
ences in 5-HT synthesis between groups. For nonautistic children, 5-HT synthesis
capacity was more than 200% of adult values until the age of 5 years and then
declined toward adult values. Serotonin-synthesis capacity values declined at an
earlier age in girls than in boys. In autistic children, 5-HT synthesis capacity
increased gradually between the ages of 2 years and 15 years to values 1.5 times
adult normal values and showed no sex difference. Significant differences were
detected between the autistic and epileptic groups and between the autistic and
sibling groups for the change with age in 5-HT synthesis capacity (Figure 5).
The investigators concluded that humans undergo a period of high brain 5-HT
synthesis capacity during childhood, and that this developmental process is dis-
rupted in autistic children.
GENETIC STUDIES
Based on a number of abnormalities identified in various aspects of 5-HT function
in autism, as reviewed above, researchers have begun preliminary investigations
of genes involved in 5-HT neurotransmission in this disorder. The 5-HT trans-
porter (5-HTT), the site of action of serotonin-reuptake-inhibiting drugs, has been
considered a candidate gene for autism. Two polymorphisms have been described
for the 5-HTT gene: a functional insertion-deletion polymorphism in the pro-
moter region (HTTLPR) (39) and a variable-number tandem repeat in the sec-
ond intron (HTT-VNTR) (40). Cook et al. (41) were the first to describe results
from a study of the 5-HTT gene in autism. The results suggested the presence of
214 McDougle et al.
Figure 5 Serotonin synthesis capacity in children with autism (n ⫽ 30, 䊊), siblings of
children with autism (n ⫽ 8, 䉱), and children with epilepsy (n ⫽ 16, ■). Global brain
values for K complex (ml/g/min) were plotted as a function of age and linear fits were
obtained for each group. The slope parameter for the autism group was 0.000075 ⫾
0.000102 (⫾SD) (dashed line), for the sibling group was ⫺0.000565 ⫾ 0.000217 (thin
line), and for the epilepsy group was ⫺0.000315 ⫾ 0.000164 (thick line). The autistic
group was significantly different from both the sibling group (p ⫽ 0.007) and the epileptic
group (p ⫽ 0.044), whereas the sibling and epilepsy groups were not significantly different
( p ⫽ 0.358). (From Ref. 38, by permission of Wiley-Liss.)
association between the short variant of HTTLPR and autism using the transmis-
sion disequilibrium test (TDT) (42) in 86 singleton families from the United
States. In contrast, Klauck et al. (43) reported preferential transmission of the
long variant of HTTLPR in a sample of 65 singleton families from Germany
using the TDT test. These investigators suggested that these conflicting find-
ings concerning the preferentially transmitted alleles of 5-HTT between the Ger-
man and U.S. patient samples might reflect etiological heterogeneity, differences
in the selection of patients, or a low power of the tests due to a small sample
size.
A subsequent study, by Zhong et al. (44), was designed to determine the
distribution frequency of the HTTLPR long allele and short allele in 72 autistic
subjects, 11 fragile X syndrome subjects with autistic behavior, 43 normal sub-
jects, and 49 fragile X syndrome nonautistic subjects. The frequency of the long/
long genotype was somewhat higher in the autistics (44.4%) and autistic fragile X
individuals (54.5%) than in the controls (normals and fragile X syndrome without
autistic behaviors combined) (30.4%). The short/short genotype was lower in the
Neurobiology of Serotonin Function 215
autistics (12.5%) and autistic fragile X (9.1%) subjects than in the controls
(21.7%). These differences, however, were not statistically different. The distri-
bution of the HTTLPR variant among the groups also showed no statistically
significant differences. This study used a simple case-control approach rather
than the more conservative TDT analysis.
In a recent study from the International Molecular Genetic Study of Autism
(IMGSA) Consortium (45), a two-stage genome search for susceptibility loci in
autism was performed on 87 affected sib pairs plus 12 non-sib affected relatives
from a total of 99 families. Regions on six chromosomes (4, 7, 10, 16, 19, and 22)
were identified that generated a multipoint maximum lod score ⬎1. No significant
increased allele sharing was detected in the 17q11.1–q12 region which contains
the 5-HTT gene. Because the presence of a locus with a small effect could not
be excluded, however, the investigators used the TDT to examine the two 5-HTT
gene polymorphisms in the IMGSA Consortium family data set (46). No evidence
for linkage or association was found at the HTTLPR locus, the HTT-VNTR locus,
or their haplotypes. The investigators concluded that polymorphisms at the 5-
HTT locus do not have a major effect on susceptibility to autism in their family
data set. Even more recently, Persico et al. (47) assessed linkage by both the
TDT and haplotype relative risk method in two new ethnically distinct samples
yielding a total sample of 98 trios and found no association between the HTTLPR
locus and autism.
Studies into other genes involved in 5-HT neurotransmission are ongoing.
Recently, Lassig and colleagues (48), using the TDT, found no evidence for an
association between polymorphisms in the gene encoding the 5-HT7 receptor
and autism.
CONCLUSION
This chapter has reviewed results from investigations of peripheral and central
neurochemistry, behavioral/neuroendocrine challenges, neuroimaging, and ge-
netics related to 5-HT function in autism. From peripheral neurochemical studies,
we have learned that WBS levels generally are higher in prepubertal autistic
subjects than in age-matched normal controls. Results from a number of studies
suggest that an age-related decline in WBS levels occurs normally in humans
and that this process may be altered in autism. These findings have led some
investigators to propose that an abnormal maturational process of the 5-HT sys-
tem may contribute to the pathophysiology of autism. Investigators are currently
attempting to determine whether WBS levels can serve as an objective measure
to assist in identifying individuals and families with a genetic vulnerability to
autism. Studies of basal central neurochemistry have not found significant differ-
ences in measures of 5-HT function between autistic subjects and controls. How-
ever, results from behavioral/neuroendocrine challenge studies have suggested
216 McDougle et al.
5-HT–immune interactions) appears warranted. Such effort should bring the field
closer to a better understanding of the pathophysiology of autism, the develop-
ment of safer and more effective treatment interventions, and, ultimately, ad-
vances in identifying etiological aspects of the disorder.
ACKNOWLEDGMENTS
We thank Ms. Robbie Smith for preparing the manuscript. This work was sup-
ported in part by an Independent Investigator Award–Seaver Foundation Investi-
gator award from the National Alliance for Research in Schizophrenia and De-
pression (NARSAD) (Dr. McDougle), a NARSAD Young Investigator Award
(Dr. Posey), Research Unit on Pediatric Psychopharmacology (RUPP) Contract
NO1MH70001 from the National Institute of Mental Health to Indiana University
(Drs. McDougle and Posey), a Daniel X. Freedman Psychiatric Research Fellow-
ship Award (Dr. Posey), the State of Indiana Division of Mental Health, and
Department of Housing and Urban Development Grant B-01-SP-IN-0200 (Repre-
sentative Dan Burton).
REFERENCES
1. Whitaker-Azmitia PM. The role of serotonin and serotonin receptors in development
of the mammalian nervous system. In: Zagon IS, McLaughlin PJ, eds. Receptors
in the Developing Nervous System. Vol 2. Neurotransmitters. London: Chapman &
Hall, 1993:43–53.
2. Cook EH Jr, Leventhal BL. The serotonin system in autism. Curr Opin Pediatr 1996;
8:348–354.
3. Anderson GM, Hoshino Y. Neurochemical studies of autism. In: Cohen DJ, Vol-
kmar FR, eds. Handbook of Autism and Pervasive Developmental Disorders. To-
ronto: John Wiley & Sons, 1997:325–343.
4. Potenza MN, McDougle CJ. The role of serotonin in autism-spectrum disorders.
CNS Spectrums 1997; 2(5):25–42.
5. Schain RJ, Freedman DX. Studies on 5-hydroxyindole metabolism in autistic and
other mentally retarded children. J Pediatr 1961; 58:315–320.
6. Sutton HE, Read JH. Abnormal amino acid metabolism in a case suggesting autism.
Am J Dis Child 1958; 96:23–28.
7. Pare CM, Sandler M, Stacey RS. 5-Hydroxytryptamine deficiency in phenylketonu-
ria. Lancet 1957; i:551–553.
8. Pare CM, Sandler M, Stacey RS. Decreased 5-hydroxytryptophan decarboxylase
activity in phenylketonuria. Lancet 1958; ii:1099–1101.
9. Pare CM, Sandler M, Stacey RS. The relationship between decreased 5-hydroxyin-
dole metabolism and mental defect in phenylketonuria. Arch Dis Child 1959; 34:
422–425.
10. Woolley DW. Manipulation of cerebral serotonin and its relationship to mental dis-
orders. Science 1957; 125:752.
218 McDougle et al.
11. Armstrong MD, Robinson KS. Abnormal metabolites in phenylketonuria. Fed Proc
1954; 13:175.
12. Armstrong MD, Robinson KS. On the excretion of indole derivatives in phenylketo-
nuria. Arch Biochem 1954; 52:287.
13. Marrazzi AS. The action of psychotogens and a neurophysiological theory of hallu-
cination. Am J Psychiatry 1960; 116:911.
14. Ritvo ER, Yuwiler A, Geller E, Ornitz EM, Saeger K, Plotkin S. Increased blood
serotonin and platelets in early infantile autism. Arch Gen Psychiatry 1970; 23:
566–572.
15. Anderson GM, Freedman DX, Cohen DJ, Volkmar FR, Hoder EL, McPhedran P,
Minderaa RB, Hansen CR, Young JG. Whole blood serotonin in autistic and normal
subjects. J Child Psychol Psychiatry 1987; 28:885–900.
16. McBride PA, Anderson GM, Hertzig ME, Snow ME, Thompson SM, Khait VD,
Shapiro T, Cohen DJ. Effects of diagnosis, race, and puberty on platelet serotonin
levels in autism and mental retardation. J Am Acad Child Adolesc Psychiatry 1998;
37(7):767–776.
17. Croonenberghs J, Delmeire L, Verkerk R, Lin AH, Meskal A, Neels H, Van der
Planken M, Schcarpe S, Deboutte D, Maes M. Peripheral markers of serotonergic
and noradrenergic function in post-pubertal, caucasian males with autistic disorder.
Neuropsychopharmacology 2000; 22(3):275–283.
18. Leboyer M, Philippe A, Bouvard M, Guilloud-Bataille M, Dondoux D, Tabuteau
F, Feingold J, Mouren-Simeoni M-C, and Launay J-M. Whole blood serotonin and
plasma beta-endorphin in autistic probands and their first-degree relatives. Biol Psy-
chiatry 1999; 45:158–163.
19. Kuperman S, Beeghly JH, Burns TL, Tsai LY. Serotonin relationships of autistic
probands and their first-degree relatives. J Am Acad Child Adolesc Psychiatry 1985;
24(2):186–190.
20. Abramson RK, Wright HH, Carpenter R, Brennan W, Lumpuy O, Cole E, Young
SR. Elevated blood serotonin in autistic probands and their first-degree relatives. J
Autism Dev Disord 1989; 19(3):397–407.
21. Cook EH, Leventhal BL, Heller W, Metz J, Wainwright M, Freedman DX. Autistic
children and their first-degree relatives: relationships between serotonin and norepi-
nephrine levels and intelligence. J Neuropsychiatry Clin Neurosci 1990; 2(3):268–
274.
22. Leventhal BL, Cook EH, Morford M, Ravitz A. Relationships of whole blood sero-
tonin and plasma norepinephrine within families. J Autism Dev Disord 1990; 20:
499–511.
23. Piven J, Tsai GC, Nehme E, Coyle JT, Chase GA, Folstein SE. Platelet serotonin,
a possible marker for familial autism. J Autism Dev Disord 1991; 21:51–59.
24. Minderaa RB, Anderson GM, Volkmar FR, Akkerhuis GW, Cohen DJ. Urinary 5-
hydroxyindoleacetic acid and whole blood serotonin and tryptophan in autistic and
normal subjects. Biol Psychiatry 1987; 22(8):933–940.
25. Gillberg C, Svennerholm L, Hamilton-Hellberg C. Childhood psychosis and mono-
amine metabolites in spinal fluid. J Autism Dev Disord 1983; 13:383–396.
26. Gillberg C, Svennerholm L. CSF monoamines in autistic syndromes and other perva-
sive developmental disorders of early childhood. Br J Psychiatry 1987; 151:89–94.
Neurobiology of Serotonin Function 219
27. Narayan M, Srinath S, Anderson GM, Meundi DB. Cerebrospinal fluid levels of
homovanillic acid and 5-hydroxyindoleacetic acid in autism. Biol Psychiatry 1993;
33:630–635.
28. Cohen DJ, Shaywitz BA, Johnson WT, Bowers MB. Biogenic amines in autistic
and atypical children: Cerebrospinal fluid measures of homovanillic acid and 5-
hydroxyindoleacetic acid. Arch Gen Psychiatry 1974; 31:845–853.
29. Cohen DJ, Caparulo BK, Shaywitz BA, Bowers MB. Dopamine and serotonin me-
tabolism in neuropsychiatrically disturbed children: CSF homovanillic acid and 5-
hydroxyindoleacetic acid. Arch Gen Psychiatry 1977; 34:545–550.
30. Hoshino Y, Watanabe M, Tachibana R, Murata S, Kancko M, Yashima Y, Kumosh-
iro H. A study of the hypothalamus-pituitary function in autistic children by the
loading test of 5HTP, TRH and LH-RH. Jpn J Brain Res 1983; 9:94–95.
31. Hoshino Y, Tachibana JR, Watanabe M, Murata S, Yokoyama F, Kaneko M, Yas-
hima Y, Kumoshiro H. Serotonin metabolism and hypothalamic-pituitary function
in children with infantile autism and minimal brain dysfunction. Jpn J Psychiatry
Neurol 1984; 26:937–945.
32. McBride PA, Anderson GM, Hertzig ME, Sweeney JA, Kream J, Cohen DJ, Mann
JJ. Serotonergic responsivity in male young adults with autistic disorder: results of
a pilot study. Arch Gen Psychiatry 1989; 46:213–221.
33. McDougle CJ, Naylor ST, Cohen DJ, Aghajanian GK, Heninger GR, Price LH.
Effects of tryptophan depletion in drug-free adults with autistic disorder. Arch Gen
Psychiatry 1996; 53:993–1000.
34. Carpenter LL, Anderson GM, Pelton GH, Gudin JA, Kirwin PDS, Price LH, Hen-
inger GR, McDougle CJ. Tryptophan depletion during continuous CSF sampling
in healthy human subjects. Neuropsychopharmacology 1998; 19:26–35.
35. Novotny S, Hollander E, Allen A, Mosovich S, Aronowitz B, Cartwright C, DeCaria
C, Dolgoff-Kaspar R. Increased growth hormone response to sumatriptan challenge
in adult autistic disorders. Psychiatry Res 2000; 94:173–177.
36. Hollander E, Novotny S, Allen A, Aronowitz B, Cartwright C, DeCaria C. The rela-
tionship between repetitive behaviors and growth hormone response to sumatriptan
challenge in adult autistic disorder. Neuropsychopharmacology 2000; 22:163–167.
37. Chugani DC, Muzik O, Rothermel R, Behen M, Chakraborty P, Mangner T, da
Silva EA, Chugani HT. Altered serotonin synthesis in the dentatothalamocortical
pathway in autistic boys. Ann Neurol 1997; 42:666–669.
38. Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, Chugani HT.
Developmental changes in brain serotonin synthesis capacity in autistic and nonau-
tistic children. Ann Neurol 1999; 45:287–295.
39. Heils A, Teufel A, Petri S, Stober G, Riederer P, Bengel D, Lesch KP. Allelic
variation of human serotonin transporter gene expression. J Neurochem 1996; 66:
2621–2624.
40. Lesch KP, Balling U, Gross J, Strauss K, Wolozin BL, Murphy DL, Riederer P.
Organization of the human serotonin transporter gene. J Neural Transmission 1994;
95:157–162.
41. Cook EH, Courchesne R, Lord C, Cox NJ, Yan S, Lincoln A, Haas R, Courchesne
E. Evidence of linkage between the serotonin transporter and autistic disorder. Mol
Psychiatry 1997; 2:247–250.
220 McDougle et al.
42. Spielman RS, Ewens WJ. The TDT and other family-based tests for linkage disequi-
librium and association. Am J Hum Genet 1996; 59:983–989.
43. Klauck SM, Poustka F, Benner A, Lesch KP, Poustka A. Serotonin transporter (5-
HTT) gene variants associated with autism? Hum Mol Genet 1997; 6(13):2233–
2238.
44. Zhong N, Ye L, Ju W, Tsiouris J, Cohen I, Brown WT. 5-HTTLPR variants not
associated with autistic spectrum disorders. Neurogenetics 1999; 2:129–131.
45. International Molecular Genetic Study of Autism (IMGSA) Consortium. A full ge-
nome screen for autism with evidence for linkage to a region on chromosome 7q.
Hum Mol Genet 1998; 7(3):571–578.
46. Maestrini E, Lai C, Marlow A, Matthews N, Wallace S, Bailey A, Cook EH, Weeks
DE, Monaco AP, International Molecular Genetic Study of Autism (IMGSA) Con-
sortium. Serotonin transporter (5-HTT) and K-aminobutyric acid receptor subunit
3 (GABRB3) gene polymorphisms are not associated with autism in the IMGSA
families. Am J Med Genet (Neuropsychiatric Genetics) 1999; 88:492–496.
47. Persico AM, Militerni B, Bravaccio C, Schneider C, Melmed R, Conciatori M, Dam-
iani V, Baldi A, Keller F. Lack of association between serotonin transporter gene
promoter variants and autistic disorder in two ethnically distinct samples. Am J Med
Genet 2000; 96:123–127.
48. Lassig JP, Vachirasomtoon K, Hartzell K, Leventhal M, Courchesne E, Courchesne
R, Lord C, Leventhal BL, Cook EH Jr. Physical mapping of the serotonin 5-HT7
receptor gene (HTR7) to chromosome 10 and pseudogene (HTR7P) to chromosome
12, and testing of linkage disequilibrium between HTR7 and autistic disorder. Am
J Med Genet 1999; 88:472–475.
49. McDougle CJ, Naylor ST, Cohen DJ, Volkmar FR, Heninger GR, Price LH. A
double- blind, placebo-controlled study of fluvoxamine in adults with autistic disor-
der. Arch Gen Psychiatry 1996; 53:1001–1008.
50. McDougle CJ, Kresch L, Posey DJ. Repetitive thoughts and behavior in pervasive
developmental disorders: treatment with serotonin reuptake inhibitors. J Autism Dev
Disord 2000; 30(5):427–435.
11
Autism, Serotonin and the Cerebellum
A New, Comprehensive Hypothesis
Donatella Marazziti
University of Pisa
Pisa, Italy
INTRODUCTION
Autism is a neuropsychiatric disorder characterized by disturbances in the devel-
opment of social interactions, imaginative activities, communication, and speech.
Associated features include resistance to change, ritualistic or compulsive
behaviors, stereotypies, self- and outwardly directed aggressive behaviors, mood
lability, and persistent preoccupation with parts of objects (1). In adolescence
the ritualistic behaviors may develop into obessional symptoms. Although its
onset occurs during childhood, it is a persistent and disabling disorder, severely
impairing the whole lifespan. Its prevalence in school-aged children is estimated
to be about 4 in 10,000 and four times higher in boys than in girls.
are those involving serotonin (5-HT). Since the first observations of Schain and
Freedman (2), who reported increased whole-blood 5-HT levels, this finding has
been replicated in a high percentage of autistic patients ranging from 30 to 40%
and even in patients’ relatives (for review, see Ref. 3). Subsequently, other indices
of altered serotonergic function have been observed, such as a decreased prolactin
response to L-tryptophan (TRP) and fenfluramine (4), the presence of antibodies
against serotonergic neurons (5,6), and the worsening of symptoms after a TRP-
free diet (7). There is also indirect evidence, based on preliminary observations,
of the potential effectiveness of clomipramine and of selective 5-HT-reuptake
inhibitors (SSRIs), mainly on obsessive-compulsive symptoms (8). The pathoge-
netic role of 5-HT is supported by a recent PET study (9) showing altered 5-HT
synthesis, while using alpha-(11C)methyl-L-tryptophan, in the dentato-thalamo-
cortical pathway of a small sample of autistic subjects.
The intrasynaptic availability of serotonin (5-HT) is regulated mainly by
an active transport or reuptake mechanism that removes the neurotransmitter
and returns it to the presynaptic terminal after its release. The 5-HT transporter
has become the subject of intensive interest in recent years, since it is the main
target of tricyclic antidepressants and SSRIs (10,11). Research in this area has
been assisted by the identity between brain and platelet 5-HT transporters, as
demonstrated by cloning studies (12–14) and by the significant correlation be-
tween the rates of inhibition of the 5-HT transporter in brain and platelets by
SSRIs (15). Apart from the direct evaluation of the reuptake rate, the 5-HT
transporter has been investigated in the brain and in platelets by means of the
specific binding first of [3H]-imipramine ([3H]-IMI) and then of [3H]-paroxetine
([3H]-Par), which appears to bind to a single site corresponding to the trans-
porter itself (16,17).
Recently, the role of the 5-HT transporter in autism has received some
support from genetic studies, but the results are still controversial: in fact, a higher
frequency of both the long and short alleles of the 5-HT transporter gene–linked
polymorphic region (5-HTTLPR) has been observed (18,19). However, the direct
measurement of the platelet 5-HT transporter has shown no difference between
autistic patients and healthy controls in platelet 5-HT uptake or [3H]-IMI binding.
In particular, autistic patients were not reported to differ from healthy controls
in terms of 5-HT reuptake (20) or [3H]-IMI binding sites (21). However, the
samples in the two studies were small and the age range was wide. In addition,
[3H]-Par is more specific than [3H]-IMI and might reveal changes that would
otherwise be undetectable. Cook et al. (22) evaluated [3H]-Par binding sites in
relatives of autistic subjects and reported a trend toward a lower dissociation
constant in hyperserotonemic than in normoserotonemic subjects. Recently, we
have observed an increased density of [3H]-Par binding sites, as compared with
matched healthy controls (23). It is interesting to underline that the function of
the 5-HT transporter is no longer considered to be restricted to the removal of
Autism, Serotonin, and the Cerebellum 223
the neurotransmitter once released in the synaptic cleft. Rather, different data
suggest that it plays a fundamental role in brain development, plasticity, and
neurodegeneration (24,25). While in adult life 5-HTT expression appears to be
restricted to raphe neurons, in postnatal development it has been detected in the
cingulate cortex and thalamus. Furthermore, the dense transient innervation of
somatosensory, visual, and auditory cortices has been shown to originate in the
thalamus, thus demonstrating a transient expression of the 5-HT transporter regu-
lating its maturation (26).
I propose that, perhaps, a disturbance in the normal process of expression
of the 5-HT transporter, such as an exaggerated persistence in postnatal life, has
profound effects on normal brain development that might lead to some disorders,
such as autism. However, currently, we cannot rule out that the increased density
of the platelet 5-HT transporter might represent a compensatory rather than a
primary phenomenon. Future studies, by means of brain-imaging studies or in-
situ hybridization in lymphocytes in which the 5-HT transporter has been detected
by means of binding techniques (27), should aim to determine whether the in-
creased [3H]-Par binding is due to an overexpression of the transporter protein.
36). The observation that the cerebellum has more neurons than the whole brain
and that the development of the lateral hemispheres and dentate nuclei parallels
that of the neocortex in humans has led to the proposal that it might project to
associative regions of the brain (37). This hypothesis has recently been supported
by the demonstration of an anatomical network linking the cerebellum with the
nonmotor prefrontal cortex through a retrogradal transneural transport of a ret-
roviral tracer (38). Its general architecture is quite simple, consisting of the pri-
mary outputs, represented by inhibitory Purkinje cells, which receive inputs from
only one climbing fibre, while mossy fibers make excitatory synapses in the nu-
clei and branch to make excitatory synapses with a large number of granule and
Golgi cells in the cortex.
Therefore, it is currently believed that the cerebellum might play a role in
the control of some cognitive processes and, in this regard, nonmotor behavioral
deficits associated with cerebellar damage or abnormalities have been reported
by some investigators (39). Interestingly, Petersen (40) observed with PET an
increase in right lateral cerebellar blood flow during a language task.
Since the cerebellum receives diffuse serotonergic innervation by afferents
originating from the raphe nuclei, theoretically 5-HT might modulate such cere-
bellar functions through specific receptors that, to date, have never been described
in humans.
Figure 1 Distribution of 5-HT5A receptor mRNA in the cerebellum. (Top left and right)
dark-field photomicrograph of a coronal section of the cerellar cortex; the Purkinje cells
are heavily labeled. High-magnification bright-field (bottom left) and dark-field (bottom
right) photomicrographs showing a high level of hybridization on the Purkinje-cell peri-
karya.
CONCLUSIONS
In conclusion, I suggest that autism (and perhaps autism-related disorders) might
be underlined by a specific disturbance in 5-HT5A receptors (45). This hypothesis
can currently be tested by comparing the level of expression of the specific
mRNA for 5-HT5A receptors in autoptic samples of cerebellum of autistic subjects
with those of healthy controls. Further studies could be carried out exploring the
possibility of structural changes at the level of the gene for 5-HT5A receptors in
autistic patients, and knockout techniques might also be employed for generating
animals lacking 5-HT5A receptors, whose (possible) behavioral abnormalities
might be used as models for some autistic symptoms. Future studies with selec-
tive agonists and antagonists for this receptor subtype, currently lacking, would
also be useful for supporting or rejecting our hypothesis, since they might permit
the comparison of the pharmacological characteristics of 5-HT5A receptors in au-
tistic subjects with those in healthy controls in autoptic brain samples or in vivo
by means of PET. Furthermore, such compounds might be of potential therapeutic
value for autism and related-disorders.
REFERENCES
1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Dis-
orders. 4th ed. Washington, DC: American Psychiatric Publishing, 1994.
2. Schain RJ, Freedman DX. Studies on 5-hydroxyindole metabolism in autistic and
other mentally retarded children. J Pediatr 1961; 58:315–320.
3. Schmahmann JD. In: Bauman ML, Kemper TL, eds. The Neurobiology of Autism.
Baltimore: John Hopkins University Press, 1994:195–226.
4. Hoshino Y, Yamamoto T, Kaneko M. Blood serotonin and free tryptophan concen-
tration in autistic children. Neuropsychobiology 1984; 11:22–27.
5. Todd RD, Ciaranello RD. Demonstration of inter- and intraspecies differences in
serotonin binding sites by antibodies from an autistic child. Proc Natl Acad Sci
USA 1985; 82:612–616.
6. Singh VKK, Singh EA, Warren RP. Hyperserotonemia and serotonin receptor anti-
bodies in children with autism but not mental retardation. Biol Psychiatry 1997;
41:753–755.
Autism, Serotonin, and the Cerebellum 227
7. Mc Dougle CJ, Naylor ST, Cohen CJ. Effect of tryptophan depletion in drug-free
adults with autistic disorder. Arch Gen Psychiatry 1996; 53:993–1000.
8. Potenza MC, C.J. Dougle CJ. The role of serotonin in autism-spectrum disorders.
CNS Spectrum 1997; 2:25–42.
9. Chugani DC, Muzik O., Rothermel R. Altered serotonin synthesis in the dentato-
thalamo-cortical pathway in autistic boys. Ann Neurol 1997; 42:666–669.
10. Wennogle LP, Meyerson LR. Serotonin uptake inhibitors differentially modulate
high-affinity imipramine dissociation in human platelet membranes. Eur J Pharma-
col 1985; 86:303–307.
11. Marcusson JO, Ross SB. Binding of some antidepressants to the 5-hydroxytrypta-
mine transporter in brain and platelets. Psychopharmacology 1991; 102:145–155.
12. Hoffman BJ, Mezey E, Brownstein MJ. Cloning of a serotonin transporter affected
by antidepressants. Science 1991; 254:579–580.
13. Lesch KP, Wolozin BL, Murphy DL, Riederer A. Primary structure of the human
platelet serotonin uptake: identity with the brain serotonin transporter. J Neurochem
1993; 60:2319–2322.
14. Ramamoorthy S, Bauman AL, Moore KR, Han H, Yang-Feng T, Chang AS, Gana-
pathy V, Blakely RD. Antidepressant- and cocaine-sensitive human serotonin trans-
porter: molecular cloning, expression and chromosomal localization. Proc Natl
Acad Sci USA 1993; 90:2542–2546.
15. Rausch JJ, Hutchinson J, Li X. Correlations of drug action between human platelets
and human brain 5HT. Biol Psychiatry 1995; 137(suppl):600.
16. Mellerup ET, Plenge P, Engelstoft M. High-affinity binding of 3H-paroxetine and
3H-imipramine to human platelet membranes. Eur J Pharmacol 1983; 96:303–
309.
17. Laruelle M, Vanisberg MA, Maloteaux JM. Regional and subcellular localization
in human brain of 3H-paroxetine binding, a marker of serotonin uptake sites. Biol
Psychiatry 1988; 24:299–309.
18. Cook EH, Courchesne R, Lord C, Cox NJ, Yan S, Lincoln A. Evidence of linkage
between the serotonin transporter and autistic disorder. Mol Psychiatry 1997; 2:
247–250.
19. Klauch S, Poustka F, Benner A, Lesch KP, Poustka A. Serotonin transporter (5-
HTT) gene variants associated with autism? Hum Mol Genet 1997; 6:2233–2238.
20. Boullin D, Freeman BJ, Geller E, Ritvo E, M. Rutter, Yuwiler A. Toward the resolu-
tion of conflicting findings. J Autism Dev Disord 1982; 12:97–103.
21. Anderson GM, Minderaa RB, van Benthem PG, Volkmar FR, Cohen DJ. Platelet
imipramine binding in autistic subjects. Psychiatry Res 1984; 11:133–141.
22. Cook EH, Arora RC, Anderson GM, Berry-Kravis EM, Yan S, Yeoh HC. Platelet
serotonin studies in hyperserotonemic relatives of children with autistic disorder.
Life Sci 1993; 52:20006–20015.
23. Marazziti D, Muratori F, Cesari A, Masala I, Baroni S, Giannaccini G, Dell’Osso
L, Cosenza A, Pfanner P, Cassano GB. Autism and serotonin: new supportive find-
ings from a platelet study. Pharmacopsychiatry 2000; 33:165–168.
24. Lauder JM. Neurotransmitters as growth regulatory signals: role of receptors and
second messengers. Trends Neurosci 1993; 16:233–240.
25. Azmitia EC, Whitaker-Azmitia PM. Development and adult plasticity of serotoner-
228 Marazziti
gic neurons and their target cells. In: Baumgartner HG, Goethert M, eds. Serotoner-
gic Neurons and 5-HT Receptors in the CNS. New York: Springer, 1997:1–39.
26. Bruening G, Liangos O, Baumgartner HG. Prenatal development of the serotonin
transporter in mouse brain. Cell Tissue Res 1997; 289:211–221.
27. Marazziti D, Rossi A, Giannaccini G, Baroni S, Lucacchini A, Cassano GB. Pres-
ence and characterization of the serotonin transporter in human resting lymphocytes.
Neuropsychopharmacology 1998; 19:154–159.
28. Courchesne E, Yeung-Courchesne R, Press GA, Hesselink JR, Jemigan TL. Hypo-
plasia of cerebellar vermal lobules VI and VII in autism. N Engl J Med 1988; 318:
1349–1354.
29. Ritvo ER, Freeman BJ, Scheibel AB, Mason-Brothers A, Jenson WR, McMahon
WM, Petersen BD, Jorde LB, Mo A, Ritvo A. Lower Purkinje cell counts in the
cerebella of four autistic subjects: initial findings. Am J Psychiatry 1986; 143:862–
866.
30. Hashimoto T, Tayama M, Murakawa K, Myazaki M, Kuroda Y. Development of
the brain stem and cerebellum in autistic patients. J Autism Dev Disord 1995; 25:
1–18.
31. Courchesne E, Saitoh O, Yeung-Courchesne R, Press GA, Haas RH, Schreibman
L. Abnormality of cerebellar vermian lobules VI and VII in patients with infantile
autism: identification of hypoplastic and hyperplastic subgroups with MR imaging.
Am J Roentgenol 1994; 162:123–130.
32. Pivet J, Sabila K, Bailey J, Arnd S. An MRI study of autism: the cerebellum revis-
ited. Neurology 1997; 49:546–551.
33. Westman JC. Handbook of Learning Disabilities: A Multisystem Approach. Boston:
Allyn & Bacon, 1990.
34. Berntson GG, Torello MW. The paleocerebellum and the integration of behavioral
function. Physiol Psychol 1982; 10:2–12
35. Schmahmann JD. In: Bauman ML, Kemper TL, eds. The Neurobiology of Autism.
Baltimore: John Hopkins University Press, 1994:195–226.
36. Parsons LM, Fox PT, Downs J, Glass T, Hirsch TB, Martin CC, Jerabek PA, Lancas-
ter JL. Use of implicit motor imagery for visual shape discrimination as revealed
by PET. Nature 1995; 375:54–58.
37. Leiner HC, Leiner AL, Dow RS. Does the cerebellum contribute to mental skills?
Behav Neurosci 1986; 100:443–454.
38. Middleton FA, Strick PL. Anatomical evidence for the cerebellar and basal ganglia
involvement in higher cognitive function. Science 1994; 266:458–461.
39. Fiez JA. Cerebellar contributions to cognition. Neuron 1996; 16:13–15.
40. Petersen SE. The processing of simple words studies with positron emission tomog-
raphy. Ann Rev Neurosci 1993; 16:509–530.
41. Pasqualetti M, Ori M, Nardi I, Cassano GB, Castagna A, Marazziti D. Distribution
of serotonin 5A receptors in human brain. Mol Brain Res 1998; 56:1–8.
42. Plassat JL, Boschert U, Amlaiky N, Hen R. The mouse 5HT5 receptor reveals a
remarkable heterogeneity within the 5HT1D receptor family. EMBO J 1992; 11:
4779–4786.
43. Hartig PR, Brancheck TA., Weinshank RL. A sub-family of 5-HT1D receptor genes.
Trends Pharmacol 1992; 13:152–159.
Autism, Serotonin, and the Cerebellum 229
INTRODUCTION
Many medications have been used to ameliorate autistic symptoms and behav-
iors in individuals with autism. Currently, there are no pharmacological treat-
ments with established indications for autism (1). However, psychotropic medi-
cations have been used in autistic individuals to target core symptoms,
behavioral dyscontrol, treatment of concurrent psychiatric disorders, and man-
agement of associated medical conditions such as seizures. Antidepressant medi-
cations, particularly serotonin-reuptake inhibitors (SRIs), and anticonvulsant
medications are among the medications commonly used for autism spectrum
behaviors. The SRIs used include clomipramine (Ananfranil), fluoxetine (Pro-
zac), sertraline (Zoloft), paroxetine (Paxil), fluvoxamine (Luvox), and venlafax-
ine (Effexor). Some of these medications have been studied in an open-label as
well as double-blind manner. The results of these studies generally indicate that
these medications are efficacious in treating some of the symptoms of individuals
with autism spectrum disorder. Anticonvulsants, such as valproic acid and carba-
mazepine, are used particularly in individuals with comorbid seizure disorder,
as well as those with impulsive aggression and affective instability. However,
to date no placebo-controlled trials have been published examining the efficacy
of these medications. There have, however, been case reports and open-label
231
232 Novotny and Hollander
important and consistent data to support the use of SRIs, which increase synaptic
serotonin availability in adult autistic subjects.
Additionally, our pilot studies with the 5-HT agonists meta-chlorophenylpi-
perazine (m-CPP) and sumatriptan have demonstrated that altered neuroendo-
crine responses to challenges with these agents significantly correlate with sever-
ity of repetitive behavior (20; Hollander et al., in preparation). As such, there is
substantial evidence that supports the involvement of 5-HT dysfunction in autism
(primarily decreased 5-HT function) and provides the rationale for a treatment
with SRIs such as fluoxetine that increase the availability of synaptic 5-HT and
down-regulate inhibitory 5-HT autoreceptors in autism.
agitation and decreased appetite. Two patients discontinued treatment due to agi-
tation. DeLong et al. (37) studied 37 children aged 2–7 in an open-label study
of fluoxetine at doses of 0.2–1.4 mg/kg/day. Twenty-two of these children had
a beneficial treatment response sustained through several months of treatment
(13–33 months). Eleven of these children had an excellent response and were
able to be mainstreamed into regular classroom. The other 11 had a good response
but remained in special-education environments. Fifteen children had little or no
response to the medication. Side effects, which included hyperactivity, agitation,
and aggressiveness, were often the reason for discontinuation.
Peral et al. (38) reported on an open-label trial of fluoxetine in six children,
aged 4–7, with autistic disorder. Five of the six children improved markedly,
with decreased repetitive behavior and improved social interest. Side effects
noted included agitation, insomnia, and decreased appetite. In addition, pilot data
from a study by Hollander et al. (39) which examined the use of fluoxetine in
children, adolescents, and adults in a double-blind, placebo-controlled crossover
design, found fluoxetine to be superior to placebo in the acute treatment of global
autism severity. A recently published study examined metabloic activity in the
brain before and after fluoxetine treatment in six adult patients, three of whom
were responders. The study noted that metabolic activity increased in the right
frontal lobe, including the anterior cingulate gyrus, following treatment and that
individuals with increased activity in these areas prior to treatment had the most
response to the medication (40). In summary, fluoxetine has been a promising
treatment in several domains, including global autistic severity, repetitive/com-
pulsive and social behavior, problem behavior, stereotypies, and depression, but
has also been shown to have some side effects, such as gastrointestinal distur-
bances and agitation, particularly in younger patients.
Fluvoxamine, an SSRI, also appears promising. McDougle et al. (41) first
reported a single case of fluvoxamine treatment of coincident autism and OCD.
There was a marked improvement in the obsessive-compulsive symptoms and
in social interaction, and a decrease in temper tantrums. In a more recent double-
blind, placebo-controlled study of fluvoxamine treatment in adults with autism,
McDougle et al. (28) found that eight of 15 (53.33%) patients were responders
compared with none of 15 in the placebo group, with improvement in repetitive
thoughts and behavior, maladaptive behavior and aggression, and significant im-
provement in ratings of social relatedness and language usage. Side effects in-
cluded mild nausea and sedation in a minority of the patients, with no occurrence
of seizures, cardiovascular events, or dyskinesias. This study was 12 weeks in
duration in both inpatient (9) and outpatient (21) populations, and patient ages
ranged from 18 to 53 years. Only three of 30 patients were female. Dosage was
started at 50 mg/day and increased at a rate of 50 mg every 3–4 days to a maxi-
mum dose of 300 mg/day. Fluvoxamine blood levels were not obtained. In a
Antidepressants and Anticonvulsants 237
paroxetine has also been reported to decrease self-injurious behavior (48) and
improve irritability and temper tantrums (49) in case reports of children with
autism spectrum disorders.
Thus, SSRIs such as fluoxetine appear promising in treating global as well
as compulsive/repetitive and social symptoms of autism. Furthermore, they are
well tolerated in low doses, and do not have the seizure and cardiac risks associ-
ated with clomipramine. However, large-scale, controlled trials are needed to
establish their efficacy and safety.
Other antidepressants have also been used to treat autistic disorders. Trazo-
done has been shown to be effective in decreasing the aggressive and self-injuri-
ous behavior in one autistic patient; however, this medication has not yet been
studied in a group of individuals (50). Desipramine, a norepinephrine-reuptake
inhibitor, has also been studied but was found to be less effective than clomipra-
mine, an SRI (23). Venlafaxine treatment, examined in a small group of individu-
als with autism spectrum disorders ranging in age from 3 to 21, was found to be
effective in decreasing repetitive behaviors and hyperactivity, and improving so-
cial deficits, attention, communication, and language function. Side effects in-
cluded behavioral activation, nausea, and polyuria (51).
Landau-Kleffner Syndrome
Landau-Kleffner syndrome (LKS) is a rare syndrome in which normally devel-
oping children develop verbal auditory agnosia at the same time as seizure disor-
der. The sleep EEG is usually abnormal and is characterized by abnormalities
such as a continuous spike-and-slow-wave pattern, focal sharp waves with spikes,
Antidepressants and Anticonvulsants 239
be safe and effective in this population at doses that produced blood levels of 75–
100 Tg/ml. Five of the patients were on other psychotropic medication, including
clonidine, lithium, thioridazine, sertraline, and methylphenidate. Sovner (59) also
reported on five cases of mentally challenged individuals with comorbid bipolar
disorder, two of whom had autism, who responded to valproate with a decrease
of their mood symptoms. No side effects are reported in this study.
Other anticonvulsants have been used to improve behavior in individuals
with autism. There is one case report (60) of two adolescent patients with autism
who received carbamazepine for treatment of “bipolar-like” affective disorders.
Both individuals evidenced decreased mood lability and a discontinuation of their
episodic mood symptoms after achieving a steady blood level of the medication.
No side effects were reported. There have been no follow-up studies examining
the efficacy of carbamazepine in ameliorating the symptoms of autism, or of
affective disorders in individuals with autism. One study of treatment of refrac-
tory seizures in children with lamotrigine showed improvement in both seizure
control and autistic symptoms in eight of the 13 autistic children in the study
(61). However a blinded, placebo-controlled 12-week study of lamotrigine did
not show any difference between lamotrigine and placebo on measures of autism
and aberrant behavior (62).
SUMMARY
In conclusion, although no medication is currently approved for the treatment
of autistic disorders, many medications show promise in treating the concurrent
symptoms of this disorder. In particular, the SRIs, especially fluoxetine, fluvox-
amine, and venlafaxine, have been shown to decrease repetitive and compulsive
behavior and to improve social and language abilities. Divalproex sodium seems
to be particularly useful in decreasing impulsive aggression and affective instabil-
ity, as well as for treating clinical and subclinical seizure disorders. All these
medications, however, will need to be further examined using double-blind, pla-
cebo-controlled methods to validate their efficacy and safety in treating the co-
morbid symptoms of autistic disorders.
ACKNOWLEDGMENTS
Supported in part by grants from the Seaver Foundation, the Food and Drug
Administration, the Cure Autism Now Foundation, the National Alliance for Re-
search on Schizophrenia and Depression, Eli Lilly and Company, and Abbott
Laboratories.
REFERENCES
1. Aman M, Van Bourgondien ME, Wolford P, et al. Psychotropic and anticonvulsant
drugs in subjects with autism: prevalence and patterns of use. J Am Acad Child
Adolesc Psychiatry 1995; 34(12):1672–1680.
2. Ciarenello RD, Vanderberg SR, Anders TF. Intrinsic and extrinsic determinants of
neuronal development: relevance to infantile autism. J Autism Dev Disord 1982;
12:115–145.
3. Anderson GM, Freedman DX, Cohen DJ, Volkmar FR, Hoder EL, McPhedran P,
Minderaa RB, Hansen CR, Young JG. Whole blood serotonin in autistic and normal
subjects. J Child Psychol Psychiatry 1987; 28:885–900.
4. Hoshino Y, Yamamoto T, Kaneko M, Tachibana R, Wantabe M, Ono Y, Kumashiro
H. Blood serotonin and free tryptophan concentration in autistic children. Neuropsy-
chobiology 1984; 11:22–27.
242 Novotny and Hollander
5. Ritvo ER, Yuwiler A, Geller E, Ornitz EM, Saeger K, Plotkin S. Increased blood
serotonin and platelets in early infantile autism. Arch Gen Psychiatry 1970; 23(6):
566–572.
6. Campbell M, Plij M. Behavioral and cognitive measures used in psychopharmaco-
logical studies of infantile autism. Psychopharmacol Bull 1985; 21:1047–1053.
7. Ritvo ER, Freeman BJ, Yuwiler A, et al. Fenfluramine therapy for autism: promise
and precaution. Psychopharmacol Bull 1986; 22:133–140.
8. Young JG, Level LI, Newcorn JH, Knott PJ. Genetic and neurobiological ap-
proaches to the pathophysiology of autism and the pervasive developmental disor-
ders. In: Meltzer HY, ed. Psychopharmacology: The Third Generation of Progress.
New York: Raven Press, 1987:825–836.
9. Schain RJ, Freedman DX. Studies on 5-hydroxyindoleamine metabolism in autistic
and other mentally retarded children. J Pediatr 1961; 58:315–320.
10. Kuperman S, Beeghly JH, Burns TL, Tsai LY. Serotonin relationships of autistic
probands and their first-degree relatives. J Am Acad Child Adolesc Psychiatry 1985;
24:186–190.
11. Piven J, Chase GA, Landa R, Wzorek M, Gayle J, Cloud D, Folstein S. Psychiatric
disorders in the parents of autistic individuals. J Am Acad Child Adolesc Psychiatry
1991; 30:471–478.
12. Abrahamson RK, Wright HH, Carpenter R, et al. Elevated blood serotonin in autistic
probands and their first-degree relatives. J Autism Dev Disord 1989; 19:397–407.
13. Cook EH, Leventhal BL, Hellar W, et al. Relationships between serotonin and nor-
epinephrine levels and intelligence. J Neuropsychiatry Clin Neurosci 1990; 2:268–
274.
14. Marazziti D, Muratori F, Cesari A, Masala I, Baroni S, Giannaccini G, Dell’Osso
L, Cosenza A, Pfanner P, Cassano GB. Increased density of platelet serotonin trans-
porter in autism. Pharmacopsychiatry 2000; 33(5):165–168.
15. Croonenberghs J, Delmeire L, Verkerk R, Lin AH, Meskal A, Neels H, Van der
Planken M, Scharpe S, Deboutte D, Pison G, Maes M. Peripheral markers of seroto-
nin and noradrenergic function in post-pubertal, caucasian males with autistic disor-
der. Neuropsychopharmacology 2000; 22(3):275–283.
16. McDougle C, Naylor S, Cohen D, Aghajanian G, Heninger G, Price L. Effects of
tryptophan depletion in drug-free adults with autistic disorder. Arch Gen Psychiatry
1996; 53(11):993–1000.
17. McBride PA, Anderson GM, Hertzig ME, et al. Serotonergic response in male
young adults with autistic disorder. Arch Gen Psychiatry 1989; 46:205–212.
18. Todd RD, Ciaranello RD. Demonstration of inter- and intraspecies differences in
serotonin binding sites by antibodies from an autistic child. Proc Natl Acad Sci
USA 1985; 82:612–616.
19. Chugani DC, Muzik O, Rothermel R, Behen M, Chakraborty P, Mangner T, da
Silva EA, Chugani HT. Altered serotonin synthesis in the dentathalamocortical path-
way in autistic boys. Ann Neurol 1997; 42:666–669.
20. Hollander E, Novotny S, Allen A, Aronowitz B, DeCaria C. The relationship be-
tween repetitive behaviors and growth hormone response to sumatriptan challenge
in adult autistic disorder. Neuropsychopharmacology 2000; 22(2):163–167.
21. Novotny S, Hollander E, Allen A Mosovich S, Aronowitz B, Cartwright C, DeCaria
Antidepressants and Anticonvulsants 243
58. Hellings JA, Chibber S, Kilgore ER, Nickel J. Valproate response as a function of
blood levels in adolescents with autistic disorder and PDD NOS. Proceedings of
the 150th Annual American Psychiatric Association Meeting, San Diego, CA, 1997:
260.
59. Sovner R. The use of valproate in the treatment of mentally retarded persons with
typical and atypical bipolar disorders. J Clin Psychiatry 1989; 50(3):40–43.
60. Komoto J, Usui S. Infantile autism and affective disorder. J Autism Dev Disord
1984; 14(1):81–84.
61. Uvebrant P, Bauziene R. Intractable epilepsy in children: the efficacy of lamotrigine
treatment, including non-seizure related benefits. Neuropediatrics 1994; 25:284–
289.
62. Belsito KM, Law PA, Kirk KS, Landa RJ, Zimmerman AW. Lamotrigine therapy
for autistic disorder: a randomized, double-blind, placebo controlled trial. J Autism
Dev Disord 2001; 31(2):175–181.
63. Kerbeshian J, Burd L, Fisher W. Lithium carbonate in the treatment of two patients
with infantile autism and atypical bipolar symptomatology. J Clin Psychopharmacol
1987; 7(6):401–405.
64. Steingard R, Biederman J. Lithium responsive manic-like symptoms in two individ-
uals with autism and mental retardation. J Am Acad Child Adolesc Psychiatry 1987;
26(6):932–935.
65. Dostal T, Zvolsky P. Antiaggressive effect of lithium salts in the treatment of severe
mentally retarded adolescents. Int Pharmacopsychiatry 1970; 5:203–207.
66. Micev V, Lynch DM. Effects of lithium on disturbed severely mentally retarded
patients [letter]. Br J Psychiatry 1974; 125(0):110.
67. Craft M, Ismail IA, Krishnamurti D, Mathews J, Regan A, Seth RV, North PM.
Lithium in the treatment of aggression in mentally handicapped patients: a double
blind trial. Br J Psychiatry 1987; 150:685–689.
68. Tyrer SP, Walsh A, Edwards DE, Berney TP, Stephens DA. Factors associated
with a good response to lithium in aggressive mentally handicapped subjects. Prog
Neuropsychopharmacol Biol Psychiatry 1984; 8(4-6):751–755.
69. Campbell M, Fish B, Korein J, Shapiro T, Collins P, Koh C. Lithium and chlorprom-
azine: a controlled crossover study of hyperactive severely disturbed young chil-
dren. J Autism Child Schizophr 1972; 2(3):234–263.
13
Use of Atypical Antipsychotics
in Autism
INTRODUCTION
The atypical antipsychotics have been very useful adjunctive treatments for autis-
tic individuals, especially when treating severe symptoms such as physical ag-
gression and self-injury. Clinical treatment, however, is sometimes complicated
by their propensity to cause weight gain and other adverse effects.
This chapter briefly reviews the pharmacology of atypical antipsychotics
and describes a rationale for their use in the treatment of autistic disorder (autism)
and related pervasive developmental disorders (PDDs). Clinical drug studies of
atypical antipsychotics in this population are reviewed in depth. Potential adverse
effects associated with atypical antipsychotics are discussed, along with recom-
mendations for their practical use in the treatment of PDDs. Finally, future direc-
tions for research are proposed.
247
248 Posey and McDougle
idal symptoms (EPS), as well as the increased efficacy in treating the “negative”
symptoms of schizophrenia (1, p. 105).
Clozapine, risperidone, olanzapine, quetiapine, and ziprasidone are atypical
antipsychotics that are United States Food and Drug Administration (FDA)–
approved treatments for schizophrenia. They differ in their affinities for subtypes
of receptors (see Table 1) and side-effect profile (discussed below).
Serotonin
A dysregulation in the 5-HT system has been known since Schain and Freedman’s
classic 1961 article reporting elevations in whole-blood serotonin (WBS) levels
Atypical Antipsychotics 249
in a large minority of autistic children (2). This finding has been consistently
replicated (3), although not yet fully explained. Most recently, McBride and col-
leagues (4) reported results from a study of 77 individuals with autistic disorder,
65 normal controls, and 22 prepubertal children with mental retardation (without
autistic features). In the prepubertal autistic children, significantly higher levels
of WBS were found compared with both normals and the mentally retarded con-
trol group, although the 25% elevation was less than that typically reported. Afri-
can-American and Latino children had significantly lower levels of WBS than
Caucasian children, regardless of diagnosis. The postpubertal subjects had consis-
tently lower levels of WBS than the prepubertal subjects, and this was not affected
by diagnosis or race.
Blunted neuroendocrine responses to pharmacological probes of the 5-HT
system have also been reported in children, using 5-hydroxytryptophan (5), and
in adults, using fenfluramine (6). In a placebo-controlled study, McDougle and
colleagues (7) reported that acute tryptophan depletion caused a significant in-
crease in maladaptive behaviors in drug-free autistic adults. Hollander and associ-
ates (8) also recently reported that the severity of repetitive behaviors in adults
with PDDs positively correlated with the magnitude of growth hormone response
to sumatriptan (a 5-HT1D agonist) challenge.
Several medications affecting the 5-HT system have been studied. Fenflur-
amine, an indirect 5-HT agonist, was extensively studied following an influential
report suggesting that it improved symptoms in three boys with autism (9). How-
ever, double-blind, placebo-controlled studies with parallel groups design failed
to demonstrate any significant difference between fenfluramine and placebo. It
was subsequently removed from the market over concerns about its contributions
to cardiac valve abnormalities (10).
Serotonin-reuptake inhibitors (SRIs), including clomipramine (11), flu-
voxamine (12), fluoxetine (13), sertraline (14), and paroxetine (15), have been
reported to reduce several maladaptive behaviors associated with autism. In con-
trast to fenfluramine, double-blind, placebo-controlled studies have supported the
use of clomipramine in children, adolescents, and young adults with autistic dis-
order (11) and fluvoxamine in adults (12). For a comprehensive review of these
medications in autistic disorder and related PDDs, the reader is referred to a
recent summary by Posey and McDougle (16).
Dopamine
Following demonstrations that psychostimulants (medications that in part en-
hance DA transmission) often worsen clinical symptoms and stereotypies in autis-
tic children (17) and that typical antipsychotics (DA receptor antagonists) reduce
certain symptoms associated with autism (18), studies of DA metabolism in-
creased. Cerebrospinal fluid levels of homovanillic acid (HVA), the primary me-
250 Posey and McDougle
Advantages
Atypical antipsychotics are associated with a lower incidence of tardive dyskinesia
(TD). This is especially important when treating PDDs because tardive and with-
drawal dyskinesias (WDs) may be more frequent in this clinical population. In a
large, prospective study of 118 children with autistic disorder treated with haloperi-
dol, Campbell and colleagues (25) found a 33.9% incidence of dyskinesias. The
majority of these were WDs and reversible; however, nine developed TD.
Atypical antipsychotics have also been reported to be better than typical
antipsychotics for the treatment of negative symptoms of schizophrenia (26).
While schizophrenia is thought to be a distinct syndrome from autistic disorder
(27), there are similarities between the negative symptoms of schizophrenia (af-
fective flattening, alogia, and avolition) and the social withdrawal often seen in
autistic disorder. This similarity led others to hypothesize that atypical antipsy-
chotics may have benefits in treating the core social impairment of autism (28).
Clozapine
There has been only one report of the use of clozapine to treat symptoms of
autistic disorder. Zuddas and colleagues (30) described treatment of three chil-
dren (ages 8 to 12 years) with autistic disorder who had not responded to haloperi-
dol. All three subjects showed initial improvement improving in hyperactivity,
negativism, aggression, and communication at doses of clozapine ranging from
Atypical Antipsychotics 251
100 to 200 mg. One subject relapsed at 5 months. Adverse effects included tran-
sient sedation and enuresis.
The side-effect profile of clozapine raises two concerns in the treatment of
individuals with autism. First, clozapine is associated with seizures at high doses.
This side effect could be important in treating autism since seizure disorders
are diagnosed in a substantial minority of these patients. Second, because of the
possibility of agranulocytosis, frequent venipuncture is needed to monitor white
blood cell (WBC) counts. This is less than desirable both in children and in those
with comorbid mental retardation because these populations are frequently unable
to fully comprehend the necessity for blood testing and are often reluctant partici-
pants. Because of this, clozapine is rarely used as an initial treatment option in
this population.
Risperidone
There have been multiple positive reports of risperidone treatment of children,
adolescents, and adults with autistic disorder and other PDDs (Table 2). Aggres-
sion is one common target symptom that improves with treatment. Others include
irritability, self-injury, sleep disturbance, repetitive behavior, and hyperactivity.
Interestingly, several studies have also reported improvements in some aspects
of social relatedness. It is unclear whether this was a direct effect of risperidone
treatment or an indirect effect due to a reduction in other maladaptive behaviors.
The most commonly reported adverse effect in these studies has been weight
gain.
Risperidone’s efficacy for treating the associated symptoms of autism has
been described in children as young as 2 years old (49), as well as adolescents
and adults. Doses of risperidone have usually been in the range of 1 to 3 mg
daily. The majority of these reports have been either retrospective or of short
duration. More recently, there has been increased interest in the efficacy and
safety of risperidone over longer durations of treatment (50).
In contrast to the large number of open-label studies of risperidone in au-
tism, there has been only one controlled study. In this 12-week, double-blind,
placebo-controlled study of adults with autistic disorder and PDD not otherwise
specified (NOS), McDougle and colleagues (29) found risperidone treatment ef-
ficacious in 8 of 14 subjects compared with none of the 16 subjects treated with
placebo. Responders were defined by a rating of “much improved” or “very much
improved” on the Clinical Global Impressions (CGI) scale (57). The mean dose
of risperidone was 2.9 mg daily. Improvement was seen in repetitive behavior,
aggression, anxiety, depression, irritability, and the overall behavioral symptoms
of autism. Risperidone was well tolerated, with mild, transient sedation being
the most frequently reported adverse effect. In contrast to studies of risperidone
in children and adolescents, weight gain was reported in only two subjects.
252
Table 2 Studies of Atypical Antipsychotics in Autistic Disorder and Related Pervasive Developmental Disorders
Medication/first No. subjects/
author age range ( years) Methodology Results Areas of improvement Side effects
Clozapine
Zuddas, 1996 (30) 3 AD (8–12) Case series 2/3 sustained improvement Aggression, communication, hyperactiv- Enuresis, transient sedation
ity, negativism
Risperidone
Vanden Borre, 1993 37 MR ⫾ autistic fea- DBPC (crossover; Risperidone ⬎ placebo Not stated Sedation
(31) tures (15–58) on concomi-
tant drugs)
Purdon, 1994 (32) 2 PDD (29–30) Case series 2/2 improved Behavior, cognition, stereotypes
McDougle, 1995 (33) 2 AD, 1 PDDNOS Case series 3/3 improved Aggression, repetitive behavior, social Transient sedation
(20–44) relatedness
Simeon, 1995 (34) 1 PDDNOS (13) Case report Improved Apathy, defiance, irritability, social
withdrawal
Demb, 1996 (35) 3 AD (5–10) Case series 3/3 improved Aggression, hyperactivity, self-injury EPS, transient sedation,
weight gain
Fisman, 1996 (28) 4 AD, 9 AS, 1 Case series 14/14 improved Agitation, anxiety, repetitive behavior, Transient sedation
PDDNOS (9–17) social awareness
Hardan, 1996 (36) 5 AD, 6 PDDNOS Open-label 8/11 improved Aggression, hyperactivity, impulsivity, Galactorrhea, weight gain
(8–17) oppositionality, self-injury
Lott, 1996 (37) 13 PDDNOS (25–49) Case series 10/13 improved Aggression, self-injury EPS, sedation, weight gain
Findling, 1997 (38) 6 AD (5–9) Open-label 6/6 improved Aggression, fearfulness, irritability, rest- Weight gain, sedation
lessness, tantrums
Frischauf, 1997 (39) 1 AD (12) Case report Improved Aggression, concentration, depression,
language
Horrigan, 1997 (40) 11 AD (6–34) Open-label 11/11 improved Aggression, self-injury, sleep Weight gain
McDougle, 1997 (41) 11 AD, 3 AS, 1 CDD, Open-label 12/18 improved Aggression, impulsivity, repetitive be- Weight gain
3 PDDNOS (5–18) havior, social relatedness
Perry, 1997 (42) 6 PDD Open-label 5/6 improved Anger, mood lability, sociability Hepatotoxicity, weight gain,
withdrawal dyskinesia
Posey and McDougle
Rubin, 1997 (43) 1 AD, 1 PDDNOS Case series 2/2 improved Aggression, hyperactivity, self-injury None
(3–5)
Cohen, 1998 (44) 3 AD, 1 PDDNOS Case series 3/4 improved Aggression, disruptiveness, self-injury Akathisia, sedation
(31–48)
Doan, 1998 (45) 1 AD (3) Case report Improved Insomnia None
McDougle, 1998 (29) 17 AD, 14 PDD-NOS DBPC Risperidone (8/14 im- Aggression, anxiety, depression, irrita- Transient sedation
(18–43) proved) ⬎ placebo (0/ bility, repetitive behavior
16 improved)
Nicolson, 1998 (46) 10 AD (4–10) Open-label 8/10 improved Aggression, autistic symptoms Weight gain
Schwam, 1998 (47) 1 AD (3) Case report Improved Food refusal
Atypical Antipsychotics
Dartnall, 1999 (48) 2 AD (27–33) Case series Maintained improvement Aggression, noncompliance, self-injury Weight gain, gynecomastia
for 2 years
Posey, 1999 (49) 2 AD (2) Case series 2/2 improved Aggression, irritability, social relat- Tachycardia, QTc prolonga-
edness tion
Zuddas, 2000 (50) 9 AD, 2 PDD-NOS Open-label 10/11 improved; 7/7 main- Anger, autistic symptoms, hyperactiv- Weight gain, facial dysto-
(7–17) tained improvement for ity, uncooperativeness nia, amenorrhea
1 year
Olanzapine
Horrigan, 1997 (51) 1 AD (10) Case report Improved Aggression, hyperactivity, sleep None
Rubin, 1997 (43) 1 AD (17) Case report Improved Aggression, mood
Malek-Ahmadi, 1998 1 AD (8) Case report Improved Aggression, hyperactivity
(52)
Heimann, 1999 (53) 1 AD (14) Case report Improved at 40 mg/day Psychosis None
Potenza, 1999 (54) 5 AD, 3 PDD-NOS Open-label 6/8 improved Aggression, anxiety, depression, hyper- Weight gain, sedation
(5–42) activity, irritability, self-injury, social
behavior
Kemner, 2000 (55) 23 PDD (children) Open-label Improved Accessibility Weight gain
Quetiapine
Martin, 1999 (56) 6 AD Open-label 2/6 improved Irritability, self-injury Behavioral activation, seda-
tion, seizure, weight gain
AD ⫽ autistic disorder; MR ⫽ mental retardation; DBPC ⫽ double-blind, placebo-controlled; PDD ⫽ pervasive developmental disorder; PDD-NOS ⫽ pervasive developmental
disorder not otherwise specified; EPS ⫽ extrapyramidal symptoms; AS ⫽ Asperger’s syndrome; CDD ⫽ childhood disintegrative disorder.
253
254 Posey and McDougle
There have not yet been any published placebo-controlled studies of risperi-
done in children and adolescents with autistic disorder. In 1997, the National
Institute of Mental Health contracted with five university-affiliated medical cen-
ters (Indiana University, Ohio State University, the Kennedy-Krieger Institute/
Johns Hopkins University, Yale University, and the University of California at
Los Angeles) to create a Research Unit on Pediatric Psychopharmacology
(RUPP) network designed to investigate promising new drug treatments for the
maladaptive symptoms associated with autistic disorder (58). Risperidone was
chosen as the first drug to study through the RUPP network. In the first phase
of this double-blind study, 102 children and adolescents with autistic disorder
were randomized to 8 weeks of treatment with risperidone or placebo. Responders
to risperidone received an additional 4 months of treatment to examine the safety
and efficacy of risperidone over a longer duration. For a review of methodological
issues in the design of this study, the reader is referred to the article by Arnold and
colleagues (59). In this study, risperidone treatment led to significant response in
69.4% of subjects compared with the 11.5% response rate seen in placebo-treated
subjects (81).
Olanzapine
Several case reports have been published suggesting efficacy of olanzapine for
the treatment of aggression, hyperactivity, mood symptoms, sleep disturbance,
and psychosis occurring in autistic disorder (Table 2). In a 12-week, prospective,
open-label study of eight children, adolescents, and adults with PDDs (ages 5 to
42 years; five with autistic disorder, three with PDD-NOS), Potenza and col-
leagues (54) found that olanzapine (mean dose of 7.8 ⫹ 4.7 mg daily; range 5–
20 mg daily) led to significant improvement in six of the subjects based on a
rating of “much improved” or “very much improved” on the CGI global improve-
ment item. Improvement was seen in overall symptoms of autism, hyperactivity,
social relatedness, affectual reactions, sensory responses, language usage, self-
injury, aggression, irritability, anxiety, and depression. Olanzapine did not appear
to reduce interfering repetitive behavior. This is in contrast to studies showing
risperidone’s efficacy for this symptom cluster of autism (29,41). The authors
hypothesized that this may be due to risperidone’s greater affinity for the 5-HT1D
receptor (60). The 5-HT1D receptor is abundant in the basal ganglia (61), a brain
region strongly implicated in the pathophysiology of obsessive-compulsive disor-
der (62). Sedation and weight gain were the most frequent adverse effects in this
study. The weight gain was particularly concerning. The mean weight for the
group increased from 62.50 ⫹ 25.37 kg to 70.88 ⫹ 25.06 kg. Two of the six
responders (both children) discontinued olanzapine treatment following the study
due to weight gain.
In another study of 23 children with PDDs, 3 months of open-label olanza-
Atypical Antipsychotics 255
pine (dose range 1.25–20 mg daily) was efficacious on several measures includ-
ing the Aberrant Behavior Checklist (63) and the CGI (55). As in the above study,
weight gain was a significant adverse effect, with subjects gaining an average of
4.8 kg. To our knowledge, there have not yet been any placebo-controlled studies
of olanzapine in subjects with PDDs.
Quetiapine
Currently, there is only one report of the use of quetiapine in the treatment of
individuals with autism. Martin and colleagues (56) reported their results of a
16-week, open-label study of quetiapine (dose range 100 to 350 mg daily) in six
children and adolescents (ages 6 to 15 years) with autistic disorder. Only two
subjects were judged “responders” by the CGI global improvement item. The
other four subjects did not complete the trial of quetiapine; three dropped out
prematurely due to sedation and lack of response and another was removed be-
cause of a possible seizure. Other side effects included behavioral activation,
increased appetite, and weight gain. The investigators concluded that quetiapine
was poorly tolerated and associated with serious side effects in this clinical popu-
lation, although others have reported a different clinical impression (64). Further
studies are needed to better determine the safety and efficacy of quetiapine for
the treatment of individuals with PDDs.
Ziprasidone
Ziprasidone is the newest atypical antipsychotic to be marketed in the United
States. Studies of ziprasidone in Tourette’s syndrome are promising (65). In addi-
tion, ziprasidone may have a lower incidence of weight gain, possibly due to its
lower affinity for the Histamine1 receptor (see Table 1). Preliminary results from
a case series suggests that it may be efficacious in PDDs and not associated with
weight gain (66).
ADVERSE EFFECTS
Tardive Dyskinesia and Extrapyramidal Side Effects
Most antipsychotic medications have the potential to cause irreversible movement
disorders (e.g., tardive dyskinesia). These are especially important to recognize
in treating individuals with PDDs given the high incidence of TD and WD ob-
served with haloperidol treatment in children with autistic disorder (25). Fortu-
nately, the potent 5-HT2A antagonism of atypical antipsychotics may reduce the
risk of TD (67). However, there have been several reports of TD with risperidone
treatment (68,69), so it is important to remain vigilant in monitoring for this very
serious side effect.
256 Posey and McDougle
Weight Gain
As discussed above, weight gain has been a prominent and frequent adverse effect
associated with atypical antipsychotic treatment. The magnitude of this may be
greater in children and adolescents compared with adults. In a retrospective study
of 60 adolescent inpatients, those treated with risperidone (n ⫽ 18) gained a
mean of 8.64 kg over a 6-month observation period, compared with 3.03 kg for
those taking typical antipsychotics (n ⫽ 23) and a 1.04-kg weight loss for those
on no antipsychotic medication (n ⫽ 19) (71). Weight gain may also contribute
to the development of hyperglycemia and hepatotoxicity (see below). This is
especially concerning given that many autistic individuals treated with atypical
antipsychotics will require chronic treatment, thus predisposing the individual to
complications of obesity including hypertension, diabetes, and heart disease.
Hepatotoxicity
Concern about risperidone-induced hepatotoxicity followed a case report of two
adults with schizophrenia who developed this complication following risperidone
treatment (72). Kumra and colleagues (73) screened 13 schizophrenic children
and adolescents treated with risperidone and discovered two who had obesity,
elevated liver enzymes, and evidence of steatohepatitis. Both cases of hepatotox-
icity resolved after risperidone discontinuation and weight loss. Subsequent to
this, Szigethy and colleagues (74) reviewed the charts of 38 children and adoles-
cents treated with risperidone (mean dose 2.5 mg daily; mean duration of treat-
ment 15.2 months) and found only one subject who had an elevation in liver
enzymes (not clinically significant) in spite of significant weight gain occurring
in the group. The question as to whether to routinely monitor liver enzymes
during atypical antipsychotic treatment remains unanswered.
Prolactin Elevation
Risperidone has been reported to increase prolactin levels in some subjects and
has been associated with galactorrhea, amenorrhea, and gynecomastia. The inci-
dence of this may be lower with olanzapine. The significance of an elevated
prolactin level in the absence of clinically significant symptoms is unclear.
Atypical Antipsychotics 257
Cardiovascular Effects
Concerns about antipsychotics, such as thioridazine and pimozide, prolonging
the corrected QT interval and potentially leading to fatal cardiac arrhythmias have
been raised. Dose-related tachycardia and corrected QT-interval prolongation was
reported in a 2-year-old boy with autistic disorder treated with risperidone (49).
This resolved upon decreasing the dose. Finally, a case of cardiac-related sudden
death in a 34-year-old woman with schizophrenia treated with risperidone has
been reported (75). Electrocardiograms (EKGs) should be considered during the
course of treatment with risperidone, especially in the presence of tachycardia,
administration of concomitant medications with cardiac effects, or when treating
the very young.
tions with cardiac effects. Finally, clozapine treatment requires regular monitor-
ing of WBC counts throughout treatment.
FUTURE DIRECTIONS
In contrast to the large number of case reports and open-label studies of atypical
antipsychotics in autism, there is a paucity of controlled studies. There are two
placebo-controlled studies of risperidone in PDDs. Controlled studies of olanza-
pine, quetiapine, and ziprasidone would be welcome, especially to better deter-
mine whether there are significant differences in efficacy or side-effect profile
between the atypical antipsychotics in this population.
An intriguing result of some studies of atypical antipsychotics in subjects
with PDDs is an improvement in some aspects of social impairment. This im-
provement needs to be better characterized in a systematic manner to determine
whether this is a direct effect of the atypical antipsychotic or an indirect effect
mediated through the reduction of other maladaptive symptoms. The develop-
ment of rating scales that adequately track social impairment are needed.
One important limitation of the atypical antipsychotics is their potential to
cause weight gain, especially in children and adolescents. Ziprasidone, a new
atypical antispsychotic, may have a lower propensity to cause weight gain and
should be thoroughly studied in this population. Finally, more research is needed
to determine the mechanism underlying the weight gain associated with atypical
antipsychotics and to develop preventive strategies.
ACKNOWLEDGMENTS
We thank Ms. Teresa Sasher for assistance in preparing this chapter. This work
was supported in part by an Independent Investigator Award–Seaver Foundation
Investigator from the National Alliance for Research in Schizophrenia and De-
pression (NARSAD) (Dr. McDougle), the Theodore and Vada Stanley Research
Foundation (Dr. McDougle), Research Unit on Pediatric Psychopharmacology
(RUPP) Contract NO1MH70001 from the National Institute of Mental Health to
Indiana University (Drs. McDougle and Posey), a Daniel X. Freedman Psychiat-
ric Research Fellowship Award (Dr. Posey), a NARSAD Young Investigator
Award (Dr. Posey), the State of Indiana Division of Mental Health, and Depart-
ment of Housing and Urban Development Grant B-01-SP-IN-0200 (Representa-
tive Dan Burton).
REFERENCES
1. Janicak PG, Davis JM, Preskorn SH, Ayd FJ Jr. Principles and Practice of Psycho-
pharmacotherapy. Philadelphia: Lippincott Williams & Wilkins, 2001.
260 Posey and McDougle
17. Campbell M, Small AM, Collins PJ, Friedman E, David R, Genieser N. Levodopa
and levoamphetamine: a crossover study in young schizophrenic children. Curr Ther
Res 1976; 19:70–83.
18. Campbell M, Anderson L, Meier M, Cohen I, Small A, Samit C, Sachar E. A com-
parison of haloperidol and behavior therapy and their interaction in autistic children.
J Am Acad Child Adolesc Psychiatry 1978; 17(4):640–655.
19. Gillberg C, Svennerholm L, Hamilton-Hellberg C. Childhood psychosis and mono-
amine metabolites in spinal fluid. J Autism Dev Disord 1983; 13:383–396.
20. Narayan M, Srinath S, Anderson GM, Meundi DB. Cerebrospinal fluid levels of
homovanillic acid and 5-hydroxyindoleacetic acid in autism. Biol Psychiatry 1993;
33:630–635.
21. Minderaa RB, Anderson GM, Volkmar F, Harcherik D, Akkerhuis CW, Cohen DJ.
Neurochemical study of dopamine functioning in autistic and normal subjects. J
Am Acad Child Adolesc Psychiatry 1989; 28:200–206.
22. Anderson L, Campbell M, Grega D, Perry R, Small A, Green W. Haloperidol in
the treatment of infantile autism: effects on learning and behavioral symptoms. Am
J Psychiatry 1984; 141:1195–1202.
23. Anderson LT, Campbell M, Adams P, Small AM, Perry R, Shell J. The effects of
haloperidol on discrimination learning and behavioral symptoms in autistic children.
J Autism Dev Disord 1989; 19:227–239.
24. Perry R, Campbell M, Adams P, Lynch N, Spencer EK, Curren EL, Overall JE.
Long-term efficacy of haloperidol in autistic children: continuous versus discontinu-
ous drug administration. J Am Acad Child Adolesc Psychiatry 1989; 28:87–92.
25. Campbell M, Armenteros JL, Malone RP, Adams PB, Eisenberg ZW, Overall JE.
Neuroleptic-related dyskinesias in autistic children: a prospective, longitudinal
study. J Am Acad Child Adolesc Psychiatry 1997; 36:835–843.
26. Chouinard G, Jones B, Remington G, Bloom D, Addington D, MacEwan GW, La-
belle A, Beauclair L, Arnott W. A Canadian multicenter placebo-controlled study of
fixed doses of risperidone and haloperidol in the treatment of chronic schizophrenic
patients. J Clin Psychopharmacol 1993; 13:25–40.
27. Volkmar FR, Cohen DJ. Comorbid association of autism and schizophrenia. Am J
Psychiatry 1991; 148:1705–1707.
28. Fisman S, Steele M. Use of risperidone in pervasive developmental disorders: a
case series. J Child Adolesc Psychopharmacol 1996; 6:177–190.
29. McDougle CJ, Holmes JP, Carlson DC, Pelton GH, Cohen DJ, Price LH. A double-
blind placebo-controlled study of risperidone in adults with autistic disorder and
other pervasive developmental disorders. Arch Gen Psychiatry 1998; 55:633–641.
30. Zuddas A, Ledda MG, Fratta A, Muglia P, Cianchetti C. Clinical effects of clozapine
on autistic disorder. Am J Psychiatry 1996; 153:738.
31. Vanden Borre R, Vermote R, Buttiens M, Thiry P, Dierick G, Geutjens J, Sieben
G, Heylen S. Risperidone as add-on therapy in behavioural disturbances in mental
retardation: a double-blind placebo-controlled cross-over study. Acta Psychiatr
Scand 1993; 87:167–171.
32. Purdon SE, Lit W, Labelle A, Jones DW. Risperidone in the treatment of pervasive
developmental disorder. Can J Psychiatry 1994; 39:400–405.
33. McDougle CJ, Brodkin ES, Yeung PP, Naylor ST, Cohen DJ, Price LH. Risperidone
262 Posey and McDougle
66. McDougle CJ, Kem DL, Posey DJ. Use of ziprasidone for maladaptive symptoms
in youths with autism. J Am Acad Child Adolesc Psychiatry 2002; 41(8):921–927.
67. Moller HJ, Pelzer E, Kissling W, Riehl T, Wernicke T. Efficacy and tolerability of
a new antipsychotic compound (risperidone): results of a pilot study. Pharmaco-
psychiatry 1991; 24(6):185–189.
68. Feeney DJ, Klykylo W. Risperidone and tardive dyskinesia. J Am Acad Child Ad-
olesc Psychiatry 1996; 35(11):1421–1422.
69. Campbell M. Risperidone-induced tardive dyskinesia in first-episode psychotic pa-
tients. J Clin Psychopharmacol 1999; 19(3):276–277.
70. Mandoki MW. Risperidone treatment of children and adolescents: increased risk
of extrapyramidal side effects? J Child Adolesc Psychopharmacol 1995; 5(1):49–
67.
71. Kelly DL, Conley RR, Love RC, Horn DS, Ushchak CM. Weight gain in adoles-
cents treated with risperidone and convential antipsychotics over six months. J Child
Adolesc Psychopharmacol 1998; 8(3):151–159.
72. Fuller M, Simon M, Freedman L. Risperidone-associated hepatoxicity. J Clin Psy-
chopharmacol 1996; 16:84–85.
73. Kumra S, Herion D, Jacobsen LK, Briguglia C, Grothe D. Case study: risperidone-
induced hepatotoxicity in pediatric patients. J Am Acad Child Adolesc Psychiatry
1997; 36(5):701–705.
74. Szigethy E, Wiznitzer M, Branicky LA, Maxwell K, Findling RL. Risperidone-
induced hepatotoxicity in children and adolescents? A chart review study. J Child
Adolesc Psychopharmacol 1999; 9(2):93–98.
75. Ravin DS, Leverson JW. Fatal cardiac event following initiation of risperidone ther-
apy. Ann Pharmacother 1997; 31:867–869.
76. Edleman RJ. Risperidone side effects. J Am Acad Child Adolesc Psychiatry 1996;
35:4–5.
77. Devinsky O, Honigfeld G, Patin J. Clozapine-related seizures. Neurology 1991; 41:
396–371.
78. Lane H-L, Chang W-H, Chou JC-W. Seizure during risperidone treatment in an
elderly woman treated with concomitant medications. J Clin Psychiatry 1998; 59(2):
81–82.
79. Hudson RG, Cain MP. Risperidone associated hemorrhagic cystitis. J Urol 1998;
160(1):159.
80. Emborg C. Neuroleptic malignant syndrome after treatment with olanzapine.
Ugeskr Laeger 1999; 161(10):1424–1425.
81. Research Units on Pediatric Psychopharmacology Autism Network. Risperidone in
children with autism and serious behavioral problems. N Engl J Med 2002; 347:
314–321.
14
Treatment of Seizures in Children with
Autism Spectrum Disorders
Roberto Tuchman
Miami Children’s Hospital
Miami, Florida, U.S.A.
INTRODUCTION
Autism is a complex developmental disorder that is behaviorally defined. Autism
spectrum disorder (ASD) is an inclusive term that includes a heterogeneous group
of children with similar symptoms and multiple biological etiologies. As a group,
children with ASD are characterized by early onset of deficits in verbal and non-
verbal communication skills, sociocommunicative function, and repetitive behav-
iors. The behaviors characteristic of ASD are secondary to central nervous system
dysfunction. The well-documented increased frequency of seizures and abnormal
electroencephalographic (EEG) findings in ASD provided some of the initial his-
torical support that has led to the now accepted concept of ASD as a neurobiologi-
cal disorder.
265
266 Tuchman
that occurs in adolescence may be associated with the degree of cognitive dys-
function, but further research to answer this observation is needed (3).
The prevalence of epilepsy in ASD varies among studies, from a low of
7% to a high of 42% (4–6). The two major risk factors for epilepsy (defined as
more than one seizure) in ASD are level of cognitive functioning and the combi-
nation of cognitive and motor deficit. The combination of severe mental defi-
ciency and motor deficit is associated with epilepsy in 42% of individuals with
ASD (3). On the other hand, in children with ASD without severe mental defi-
ciency, motor deficit, associated perinatal or medical disorder, or a positive family
history of epilepsy, seizures occur in 6% of individuals, a figure analogous to
the 8% frequency of seizures in children with dysphasia without autism (3). It
is important to realize, then, that the same risk factors for epilepsy—that is, cogni-
tive and motor deficits—are also the risk factors for seizure occurrence in ASD.
In children with ASD and without severe cognitive or motor deficit, the
only other risk factor for seizures was the type of language dysfunction. In a
study that classified 197 autistic children without severe mental retardation or
hearing impairment into four language subtypes based on a modified clinical
classification scheme proposed by Rapin and Allen (7), the highest percentage
of epilepsy (41%) occurred in children with the most severe deficit in the compre-
hension of language: verbal auditory agnosia (VAA) (8). VAA is a severe re-
ceptive and expressive language disorder believed to arise from inadequate audi-
tory or phonological processing that engages activity in primary or secondary
auditory cortices (9,10). VAA can occur in a developmental or acquired form
(11). In the acquired form it is the language type associated with Landau-Kleffner
syndrome (LKS) and also known as acquired epileptic aphasia (12). LKS is a
rare epileptic syndrome, and clinical seizures are not a necessary part of the diag-
nosis. It is defined by having an acquired aphasia in association with an epilepti-
form EEG demonstrating spikes or spike-and-wave discharges over the temporal
and parietal head regions. It is important to point out that up to 25% of individuals
with this diagnosis do not have clinical seizures. This suggests that, at least in this
subgroup of individuals, it is the “subclinical seizures” as indexed by epileptiform
activity on the EEG and not the clinical seizures that are responsible for the
language and behavioral manifestations that occur in LKS (13). The importance
of LKS to epilepsy in ASD is that approximately 30% of individuals with autism
have a regression in language and an acquired aphasia (VAA) similar to that in
LKS. Some studies have shown no significant difference in the prevalence of
epilepsy between a group of youngsters with a history of regression and those
without regression (14). Other studies have reported that epilepsy is significantly
more frequent in those with a history of regression than in those without regres-
sion (31% in those with regression vs. 15% in those without) (15). VAA is an
important predictor of outcome in all children with language dysfunction and
Treatment of Seizures 267
seizures, and in a recent study the duration of language loss in a group of children
with developmental or acquired VAA was not influenced by the persistence of
clinical seizures. Premorbid language and behavior were more predictive of lan-
guage recovery in this group of children with VAA (16). Future studies will need
to look at children with language-epilepsy syndromes in terms of not only vari-
ables such as regression and seizures but also type and degree of language dys-
function and the behavioral profile.
To summarize, the variability in seizure frequency reported by different
investigators in ASD is likely due to three risk factors: 1) the age groups studied,
with the higher percent of seizures being found in studies that included adolescent
and young adults, 2) the level of cognitive function, with the higher percent of
seizures found in studies that include lower-functioning individuals, and 3) the
type and degree of language dysfunction, with the highest percent of seizures
occurring in individuals with VAA (3).
Epileptiform abnormalities on EEG (interictal spikes or spike-and-wave
complexes) are even more frequent in youngsters with ASD than clinical epi-
lepsy. In both their 1991 and 1997 studies, Tuchman and colleagues found epilep-
tiform abnormalities in 8% of autistic, nonepileptic children (3,14). In the 1997
study, when only those youngsters with EEG data were considered, the percent-
age of autistic, nonepileptic youngsters with epileptiform abnormalities rose to
15%. Furthermore, in that study there was an equal proportion of children with
epileptiform EEGs among those with a history of regression and those without.
However, when children with a history of epilepsy were excluded from the analy-
sis, there was a statistically significant increased risk of epileptiform abnormali-
ties in youngsters with a history of regression (19% in those with regression vs.
10% in those without). Kawasaki and colleagues, in a cohort of 158 individuals
with multiple sleep-recorded EEGs, found epileptiform EEG abnormalities in
60.8% (17). Lewine and colleagues found epileptiform EEG activity in 68% of
50 children with ASD and a history of regression. Utilizing the experimental,
relatively new technique of magnetoencephalography (MEG), they demonstrated
epileptiform activity in 82% of these 50 children (18). Although several studies
have documented a high frequency of epileptiform abnormalities in individuals
with ASD without seizures, especially with prolonged sleep studies, the role that
these interictal discharges have in accounting for the behavioral and language
dysfunction characteristic of ASD has not been elucidated.
Children with ASD, regression (an acquired VAA), and an abnormal epi-
leptiform EEG without clinical seizures have been described as having autistic
epileptiform regression (AER), and treatment strategies used in LKS have also
been tried in a limited number of studies in this subgroup of children. Although
there are clinical and electrophysiological similarities, there are important differ-
ences between AER and LKS (19). Some of these differences include:
268 Tuchman
TREATMENT ISSUES
The treatment of children with autism and seizures or with autism and epileptiform
abnormalities on EEG recordings without seizures needs to be discussed within the
context of the limited knowledge presently available on the role that epilepsy, both
clinical and subclinical, has on the behavioral and language profile of children with
ASD. In general terms, all types of seizures are reported in children with ASD,
and the treatment of these seizures per se is not usually difficult or different from
treatment of seizures in the general non-ASD population (21).
The observation that epileptiform discharges on the EEG without clinical
seizures can cause behavioral, language, and cognitive impairments (22) raises
the question of whether this is occurring to some extent in complex behavioral
disorders such as autism. For example, children can have language impairment
secondary to an active epileptic focus in an area subserving language even in
the absence of clinical seizures (23). There are also well-documented reports of
patients with benign partial epilepsy of childhood with centrotemporal spikes
who have fluctuating disturbances such as intermittent drooling, oromotor dys-
praxia, dysphagia, and transient isolated deterioration of speech production in
association with epileptic discharges without clinical seizures (24). Similar corre-
lations have recently been made with respect to electrical status epilepticus dur-
Treatment of Seizures 269
ing slow-wave sleep (ESES) (25–27). The concept that transitory changes in
higher cortical functions can be secondary to EEG discharges not accompanied
by seizures is not new; it was proposed over 50 years ago (28). The term transient
cognitive impairment (TCI) is used to describe individuals with epileptiform
EEG discharges in association with a momentary disruption of adaptive cerebral
function (29). It has also been shown that focal interictal spikes may transiently
disrupt aspects of cortical functioning corresponding to the neuroanatomical lo-
cation in which they occur (30,31). It is important to emphasize that at present
we do not have scientific evidence to either refute or confirm the premise that
interictal epileptiform discharges play a role in the symptoms and signs that
occur in ASD.
Treatment studies in children with AER are preliminary, limited, and contro-
versial. Published studies have included the use of valproic acid in a total of four
case reports of children with ASD without clinical seizures but with epileptiform
abnormalities on the EEG (32,33). There is one published report of the use of corti-
costeroids in a child with autism, VAA, and regression but with a normal EEG (34).
There are numerous abstracts and anecdotal reports on the use of valproic acid and
the use of steroids in children with AER, but without controlled clinical trials on
the use of these interventions no definite recommendations can be made.
There are also a few studies on children with autistic regression and epi-
lepsy (clinical seizures) that suggest that aggressive treatment such as epilepsy
surgery is associated with positive outcomes (35,36). It is important to re-empha-
size that these case reports were of children with intractable epilepsy and, as
such, the surgery was being done for treatment of the seizures and not for the
symptoms of autism. One study suggests that after surgical intervention in chil-
dren with ASD and intractable seizures, the seizures may improve but the ASD
symptoms do not (37). A recent publication reported improvement in language
and behavior in 12 of 18 children with ASD and a history of regression in lan-
guage, multifocal epileptiform EEGs, and symptoms of possible subclinical sei-
zures (staring episodes, rapid eye blinking) without clinical seizures who under-
went multiple subpial transections (18). This latter study not only is controversial
but highlights how the lack of controlled clinical trials using good assessment
tools may lead to inappropriate irreversible and potentially life-threatening inter-
ventions in children with ASD. The use of surgery in children with LKS to treat
the symptoms associated with this disorder is still not well validated and contro-
versial in its own right (38), and its use in children with ASD without intractable
epilepsy is presently unacceptable.
Many clinicians believe that it is reasonable to consider a trial of anticon-
vulsants or steroids in a child with ASD and an epileptiform EEG who has a
history of regression of language and VAA. In the absence of intractable clinical
seizures there is no scientific evidence to suggest that epilepsy surgery is a treat-
ment option in children with ASD. In addition, the evidence to suggest that treat-
270 Tuchman
REFERENCES
1. Gillberg C, Steffenburg S. Outcome and prognostic factors in infantile autism and
similar conditions: a population-based study of 46 cases followed through puberty.
J Autism Dev Disord 1987; 17(2):273–287.
2. Volkmar FR, Nelson DS. Seizure disorders in autism. J Am Acad Child Adolesc
Psychiatry 1990; 29(1):127–129.
3. Tuchman RF, Rapin I, Shinnar S. Autistic and dysphasic children. II. Epilepsy [pub-
lished erratum appears in Pediatrics 1992; 90(2 Pt 1):264]. Pediatrics 1991; 88(6):
1219–1225.
4. Schain R, Yannet H. Infantile autism. J Pediatr 1960; 57(4):560–567.
5. Deykin EY, MacMahon B. The incidence of seizures among children with autistic
symptoms. Am J Psychiatry 1979; 136(10):1310–1312.
6. Olson I, Steffenburg S, Gillberg C. Epilepsy in autism and austiclike conditions.
Arch Neurol 1988; 45:666–668.
7. Rapin I, Allen DA. Syndromes in developmental dysphasia and adult aphasia. In:
Plum F, ed. Language, Communication, and the Brain. New York: Raven Press,
1988:57–75.
8. Tuchman R, Rapin I, Shinnar S. Autistic and dysphasic children. I. Clinical charac-
teristics. Pediatrics 1991; 88(6):1211–1218.
9. Klein S, Kurtzberg D, Brattson A, Kreuzer J, Stapells D, Dunn M, et al. Electrophys-
iologic manifestations of impaired temporal lobe auditory processing in verbal audi-
tory agnosia. Brain Lang 1995; 51:383–405.
10. Mantovani JF, Landau WM. Acquired aphasia with convulsive disorder: course and
prognosis. Neurology 1980; 30:524–529.
11. Rapin I, Mattis S, Rowan JA, Golden G. Verbal auditory agnosia in children. Dev
Med Child Neurol 1977; 19(2):192–207.
12. Beaumanoir A. The Landau-Kleffner syndrome. In: Roger J, Dravet C, Bureau M,
Dreifuss FE, Wolf P, eds. Epileptic Syndromes in Infancy, Childhood and Adoles-
cence. London: John Libbey Eurotext, 1985:181–191.
13. Tuchman RF. Acquired epileptiform aphasia. Semin Pediatr Neurol 1997; 4(2):93–
101.
14. Tuchman RF, Rapin I. Regression in pervasive developmental disorders: seizures
and epileptiform electroencephalogram correlates. Pediatrics 1997; 99(4):560–566.
15. Kobayashi R, Murata T. Setback phenomenon in autism and long-term prognosis.
Acta Psychiatr Scand 1998; 98(4):296–303.
16. Klein SK, Tuchman RF, Rapin I. The influence of premorbid language skills and
behavior on language recovery in children with verbal auditory agnosia. J Child
Neurol 2000; 15(1):36–43.
17. Kawasaki Y, Yokota K, Shinomiya M, Shimizu Y, Niwa S. Brief report: electroen-
Treatment of Seizures 271
I. INTRODUCTION
Autism spectrum disorders are conditions usually presenting in early childhood
characterized by marked impairments in social interaction and communication
(1) and by extremely restricted and odd interests and activities (2). For at least
a subgroup of individuals, movement disorders are salient manifestations of the
condition. For example, anomalous motions are prominent presenting (3) and
persisting signs (4–7) in some persons with autism spectrum disorders (8). Move-
ment disorders occur in people with autism spectrum disorders both as manifesta-
tions of the underlying conditions and as adverse effects of therapeutic interven-
tions (9).
This chapter reviews the treatment of movement disorders in autism spec-
trum disorders. For information on the autism spectrum disorders, please refer
to chapters 1–6. This chapter concentrates on diagnostic and therapeutic aspects
of common movement disorders in autism spectrum disorders.
The goal of this chapter is to integrate the latest scientific findings on the
treatment of movement disorders in autism spectrum disorders in clinically mean-
273
274 Brǎsić
ingful ways. It is aimed at professionals who are treating individuals with autism
spectrum disorders, with an emphasis on effective strategies targeting specific
symptoms. The chapter provides clear empirical pharmacological, behavioral,
and language-based data from several related disciplines to support optimal clini-
cal care in the treatment of movement disorders in autism.
Section II discusses the characteristics of movement disorders commonly
seen in autism spectrum disorders. Section III addresses the diagnosis and treat-
ment of movement disorder commonly appearing as adverse effects of pharmaco-
logical therapy for autism spectrum disorders, namely, acute dystonia (acute dys-
tonic reactions), acute akathisia, and withdrawal and tardive dyskinesias. Section
IV begins with a discussion of the diagnosis and treatment of the movements
typically observed in Rett’s disorder and autistic disorder, and concludes with a
discussion of the diagnosis and treatment of self-injurious behavior. Although
only a minority of individuals with autism spectrum disorders experience self-
injury, self-injurious behaviors merit inclusion in this chapter because of the re-
sultant morbidity and mortality.
Please refer to Table 1 at the end of the chapter for abbreviations utilized
herein.
B. Classification of Hyperkinesias
Hyperkinesias can be subdivided broadly into seven common subgroups (defined
in Table 12). While there is considerable overlap among some of the subgroups,
they can be separated by component descriptive terms (11). The relationships of
the common hyperkinetic movement disorders are represented in Figure 1 (11).
Some movement disorders, such as tics (11,17,42), may be temporarily
suppressed by the individual. This may happen when the person is concentrating
intensely on intellectual or physical activities. Although tics may be temporarily
suppressed, the person usually experiences an urge to perform the suppressed
tic. When the tic is eventually expressed, the individual then feels a sense of
relief. Additionally, a volley of intense tics is likely to occur when the suppressed
tic is finally expressed. On the other hand, akathisia is a movement disorder
characterized by the subjective experience of inner restlessness and an urge to
move. While individuals with akathisia typically also exhibit objective evidence
of the disorder, such as marching in place, akathisia may be present in a mo-
tionless individual (34,35,37).
Movement disorders in autism spectrum disorders can be broadly classified
into two types: 1) movement disorders resulting from treatments for autism spec-
trum disorders and 2) movement-disorder manifestations of autism spectrum dis-
orders. Because movement disorders occur frequently as adverse effects of the
pharmacotherapy of autism, this review includes this serious subgroup.
Motion analysis of home videotapes of infants facilitates the diagnosis of
autism. For example, Teitelbaum and colleagues (3) have suggested that anoma-
276 Brǎsić
lies in the motions of babies who later exhibit autism may be diagnostic of autism
in infancy (43). Although this report holds great promise for the early identifica-
tion of children at risk to develop autism, the number of reported cases is small.
Also, the methods utilized by Teitelbaum and colleagues are highly specialized.
Because therapy for autism spectrum disorders, particularly pharmacother-
apy, can cause many motions, postures, sensations, and utterances, it may be
difficult to distinguish movements caused by autism from movements caused
by treatment for autism spectrum disorders. Although the movement disorders
resulting from autism spectrum disorders are poorly described, they may be pa-
thognomonic for autism (4). While the movements resulting from autism cause
individuals to stand out in crowds, the movements typically do not bother the
individuals themselves. The movements resulting from autism may upset family
members because these movements identify the individuals as psychiatric patients
to the general public. These movements may constitute a stigma as a sign to the
community of the deviance of the person (44). Movements resulting from autism
may be embarrassing to the families and caregivers without harming the individ-
Movement Disorders 277
ual. On the other hand, some movement disorders resulting from autism can cause
serious tissue damage to the individual (24) (see Section IV.B), to others (45)
(Section IV.C), and to property (Section IV.D). In particular, the movement disor-
ders classified as self-injurious behaviors may cause serious morbidity and mor-
tality and therefore demand immediate intervention to prevent permanent tissue
damage (24) (Section IV.B).
The second group of movement disorders in autism is caused by treatments
for autism. Medications, particularly typical neuroleptics such as chlorpromazine
(Thorazine), haloperidol (Haldol), and thioridazine (Mellaril), are responsible for
many movement disorders that are difficult to treat. Pharmacological agents that
block postsynaptic dopamine type 2 (D2) receptors in the brain, such as the typical
neuroleptics, often result in tardive dyskinesia, a movement disorder that usually
occurs within 3 months of the discontinuation of the agent. Additional informa-
tion about the diagnosis and treatment of tardive dyskinesia and associated condi-
tions can be obtained in a recent review (11). A related group of movement
disorders includes withdrawal dyskinesias, movement disorders occurring as the
dose of the pharmacological agent is reduced or discontinued. People with autism
spectrum disorders appear to be exquisitely sensitive to adverse effects from med-
ication. Individuals with autism appear prone to develop tardive dyskinesia—
abnormal movements occurring after the causative medication has been discon-
tinued—when treated with medication, especially dopamine antagonists. Since
tardive dyskinesias can be painful and extremely troubling to the individual, they
often lead people with autism and their families and caregivers to seek treatment
(11).
Clinicians are hampered in their quest for effective treatments for move-
ment disorders in people with autism spectrum disorders by the dearth of pub-
lished reports. Therefore, there exists a need for publication of clinical trials of
therapeutic interventions for movement disorders in autism spectrum disorders.
Publication of even single case reports of negative clinical trials of pharmacologi-
cal and other interventions is justified and needed to advance knowledge about
beneficial and adverse effects of interventions for autism spectrum disorders (16).
The desperation of parents of children with Rett’s disorder, autistic disorder, and
other autism spectrum disorders may lead them to employ novel interventions
to attempt to help their children. Publication of the results of these efforts is
crucial to distinguish beneficial therapies from harmful ones. Publication of case
reports of adverse effects of treatment on these children is valuable to alert clini-
cians and parents about harmful effects of new agents. Clinicians and researchers
can benefit from the regular use of established rating scales (37), including the
Abnormal Involuntary Movement Scale (AIMS) (Table 2) (10,11), the Children’s
Global Assessment Scale (CGAS) (Table 3) (12), the Family Compliance Check-
list (FCC) (Table 4) (13,14), the Hillside Akathisia Scale (HAS) (Table 6)
(11,15), the Movement Disorders Checklist (MDC) (Table 7) (11), the Myoclo-
278 Brǎsić
nus Versus Tic Checklist (MVTC) (Table 8) (17), the Psychoactive Medication
Quality Assurance Rating Survey (PQRS) (Table 9) (11,18), the Timed Self-
Injurious Behavior Scale (TSIBS) (Table 10) (24), and the Timed Stereotypies
Rating Scale (TSRS) (Table 11) (11,25) to substantiate clinical trials of therapeu-
tic interventions.
A. Acute Dystonia
Acute dystonia (acute dystonic reaction) is characterized by the sudden forceful
contractions of muscle groups (Table 12). This may present within hours of the
first dose of a dopamine antagonist or at any moment in the course of neuroleptic
treatment.
Although rare, a dystonic contraction blocking respiration may occur. This
constitutes a medical emergency. A tracheostomy (insertion of a tube into the
windpipe, below the larynx) may be required to permit ventilation to prevent a
fatality.
Several muscle groups are commonly affected by acute dystonia. For exam-
ple, the eye muscles may be affected, causing the eyes to deviate upward, down-
ward, or to the side. The neck muscles may also be affected. The head may extend
backward (retrocollis) or to either side (torticollis). In addition, the tongue and
mouth muscles may be affected, preventing the person from swallowing or speak-
ing. Lastly, arm and leg muscles may be affected. The occurrence of acute dysto-
280 Brǎsić
nia can be extremely upsetting. The person may believe that an outside force is
causing contraction of the muscles, and may be unable to verbalize the experi-
ence, especially if speech is affected. The experience of acute dystonia may cause
severe distress, escalating anxiety and further increasing muscular contraction.
To minimize the discomfort and upset associated with the occurrence of
acute dystonia, people who are being treated with dopamine antagonists are
warned before the institution of treatment that dystonic reactions are common
side effects that can be effectively treated. They are educated about the nature
of acute dystonia and advised that they may experience sudden muscle spasms.
They are warned to immediately notify staff if they experience symptoms of
acute dystonia in order to receive appropriate interventions. Furthermore, when
it appears that patients are suffering from acute dystonia, they are educated that
they are likely experiencing a temporary side effect of medication that can be
quickly relieved. The need for medication is re-evaluated and, if possible, therapy
with the causative agent is reduced or discontinued.
Patients may be unable to accurately articulate the possibility that they are
experiencing an acute dystonic reaction. Instead, they may indicate that they are
having trouble talking, swallowing, breathing, or walking, or that they are experi-
encing pain from muscular contraction. There may be no localizing neurological
signs. Therefore, experienced clinicians have a particularly high level of suspi-
cion that acute dystonia may be occurring in patients who are being treated with
dopamine antagonists for the first time. Intramuscular administration of 2 mg
benztropine mesylate (Cogentin) or 25 mg diphenhydramine hydrochloride (Ben-
adryl) for sudden apparent subtle deficits in speech or walking often immediately
confirms the diagnosis of acute dystonia by relieving the symptoms. Usually the
patient reports immediate relief. If symptoms are not helped, then another cause
must be sought.
Acute dystonia can be effectively treated by the administration of anticho-
linergic medications. Oral doses of anticholinerigic agents usually cause the dys-
tonia to subside in an hour or so. Patients often prefer parenteral administration
of the anticholinergic to obtain the faster beneficial effect. Since anticholinergic
medications produce unpleasant adverse effects, they are not routinely prescribed
until the individual develops at least one dystonic reaction. Therefore, unneces-
sary administration of anticholinergics, and the risk of their adverse effects, can
be avoided for many.
B. Acute Akathisia
Akathisia is a sense of inner restlessness and an urge to move resulting from
treatment with medications, commonly dopamine antagonists. Unlike the other
common movement disorders discussed in this chapter, which are characterized
Movement Disorders 281
mature neurons (65–67) and that MeCP2 deficiency results in instability of brain
functioning (68). Databases of MeCP2 mutations in Rett’s disorder (69–71) are
maintained at http://homepages.ed.ac.uk/skirmis and http://mecp2.chw.edu.au.
Dysfunction of excitatory neurotransmission is implicated in the pathogen-
esis of Rett’s disorder by the abnormalities in the densities of N-methyl-D-
aspartate (NMDA), alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
(AMPA), and gamma-aminobutyric acid (GABA) in the superior frontal gyrus
of people with the disorder (72). Impaired cerebral metabolism, particularly in
the frontal (73) and parietal lobes and the insular cortex, likely play a role in the
pathogenesis (74). Dysfunction in dendritogenesis is hypothesized to occur in
Rett’s disorder (75). Reduced cerebral bloodflow in both hemispheres probably
also contributes to the stereotyped hand movements (76).
Reductions in 1) AMPA and NMDA receptor densities in the putamen and
2) kainate-type glutamate receptor densities in the caudate nucleus are hypothe-
sized to contribute to the hand movements of Rett’s disorder (77). Altered dopa-
minergic neurotransmission in the striatum is hypothesized to contribute to the
movement disorders (78,79). These typically do not bother the individual, but
they may cause the person to stand out in a crowd. In this sense, the movements
may be disfiguring mannerisms that contribute to the stigma of mental illness
(44). The hand movements may additionally bother family, friends, and teachers.
Since there is no danger to the individual from the hand movements, no treatment
for the patient is indicated. Instead, the family, friends, teachers, and neighbors
may benefit from counseling about the benign nature of the movements. They
may be helped to learn to accept the movements.
People with Rett’s disorder typically can receive medical and surgical inter-
ventions when indicated. For example, general anesthesia with halothane, nitrous
oxide, and isoflurane has been successfully performed on a subject with Rett’s
disorder (80). Patients with Rett’s disorder require supervision around the clock
because they are unable to care for themselves.
Further information about Rett’s disorder can be obtained from the Rett
Syndrome Research Foundation (RSRF), 4600 Devitt Drive, Cincinnati, OH
45246, (513) 874-3020, at www.rsrf.org, and from the International Rett Syn-
drome Association (IRSA) at www.rettsyndrome.org.
2. Movements of Autistic Disorder
A wide variety of movements have been associated with autism spectrum disor-
ders. Some, such as tics (81–84), may represent other disorders, such as Tou-
rette’s syndrome (42,85), that may coexist with autism spectrum disorders (Table
12). Further information about tics and Tourette’s syndrome may be obtained
from the Tourette Syndrome Association, Inc., 42-40 Bell Blvd., Bayside, NY
11361, (718) 224-2999, www.tsa-usa.org.
286 Brǎsić
Additionally, repetitive movements often are carried out over and over by
persons with autistic disorder. The movements are often conducted in a ritualistic
manner consistent with compulsions. Orchestrated sequences of coordinated
movements—such as repeatedly dropping rocks from a particular height, repeat-
edly flushing the toilet while watching the water go down the drain, and repeat-
edly touching walls or spinning objects—have been classified as compulsions or
complex tics. Administration of diagnostic procedures designed for obsessions,
compulsions (86,87), tics (42,88,89), and other adventitious movements (11,37)
can be modified for the limitations of people with autism spectrum disorders to
obtain assessments with a modicum of reliability (28,37,90). Tics (42), Tourette’s
disorder, obsessive-compulsive disorder, and other comorbid disorders can ap-
propriately be treated with the usual therapies for those conditions.
The most common movements in autistic disorder, however, are an array
of motions categorized as stereotypies (40,41,91–93). Several specific motions
have been identified in autism spectrum disorders (4,94–96) and other neuropsy-
chiatric disorders (95). However, stereotypies occur spontaneously in humans
(97,98) and animals (99). Table 13 contains additional information about stereo-
typies. Some stereotypies, especially those of a complicated nature, may be also
classified as compulsions and complex tics (42,88,89).
Stereotypies in autism spectrum disorders typically do not bother the indi-
vidual. Although they may cause the person to stand out and to be identified as
a mental patient, they usually do not cause injury to the person or others. Parents,
family, teachers, neighbors, and friends may object to the presence of stereotypies
in part due to the associated stigma of mental illness (44). However, stereotypies
that do not harm self or others can usually be ignored. While treatments such as
dopamine agonists (100), dopamine antagonists (101), serotonergic agents (102),
an analog of adrenocorticotrophic hormone (103), naltrexone (104), and venlafax-
ine (105) may at least temporarily ameliorate stereotypies, they are associated
with adverse effects themselves (106). Although often effective in alleviating
stereotypies in children with autistic disorder (107), clomipramine has serious
side effects (106,108–112), including seizures and cardiac events. Therefore, use
of newer serotonergic agents, such as fluoxetine and fluvoxamine (113,114), may
be an appropriate initial approach (102,106). (Refer to Chapter 12 for further
information about the administration of serotonergic medications.) Since seroton-
ergic dysfunction characterizes a group of individuals with autism spectrum dis-
orders, caution must be exercised in the administration of selective serotonin-
reuptake inhibitors (SSRIs) because dyscontrolled behaviors may result from
their use. Utilization of rating scales for adverse effects of SSRIs before starting
treatment and regularly throughout the course of treatment (108,115) facilitates
the early identification of adverse effects so that appropriate interventions can
be instituted.
Movement Disorders 287
B. Self-Injurious Behaviors
While self-injurious behaviors—chronic movements causing external or internal
trauma on a mechanical basis—present challenges to classification (125), a subset
of these activities meet the criteria for stereotypies. Therefore, stereotypical be-
haviors resulting in injury to self are discussed in this chapter. Self-injurious
behaviors are also classified as self-mutilative and self-abusive behaviors
(126,127). Unlike most stereotypies in autism spectrum disorder, self-injurious
behaviors are serious problems requiring immediate intervention to prevent mor-
bidity and mortality. Fortunately, most individuals with autism spectrum disor-
ders never exhibit self-injurious behaviors. Those who do, however, are likely
to have additional deficits, including seizure disorders, language disorders, visual
impairments, and profound mental retardation (128–131). Self-injurious behav-
288 Brǎsić
iors are among the most challenging manifestations of autism spectrum disorders,
consuming large amounts of human and fiscal resources while causing great
suffering and disfigurement (132). In addition to afflicting people with autism
spectrum disorders, self-injurious behaviors are serious public-health problems
affecting patients with mental retardation (133), Tourette’s disorder (134,135),
Lesch-Nyhan syndrome (136), Cornelia de Lange syndrome (154), frontal-lobe epi-
lepsy (138), and other neuropsychiatric disorders (135). Self-injurious behaviors
also consume enormous amount of caregiver time. The direct and indirect costs
of treatment and management of self-injurious behaviors amount to billions of
dollars annually (132). These behaviors must be differentiated from the self-stim-
ulation occasionally seen in infants and toddlers, which is time-limited and never
inflicts permanent harm.
Prevalence rates of self-injurious behaviors range from 8% to 40% among
persons with mental retardation and other developmental disabilities. The preva-
lence roughly correlates with the degree of mental retardation, a possible cause
of the discrepant findings in different settings (132,137,139–145).
Self-injurious behaviors involve most bodily parts and functions. The com-
mon forms include head banging, hand biting, self-scratching, self-hitting, eye
gouging, and rectal digging. Less common forms of self-injury include pica (the
ingestion of nonnutritive substances) (146), coprophagia (the ingestion of feces),
and aerophagia (the swallowing of air) (147). Rumination (the ingestion of regur-
gitated food) is also occasionally classified as a self-injurious behavior. Some of
the more common sequelae of self-injurious behaviors are listed in Table 14.
These behaviors are a major roadblock for successful community placement of
affected individuals.
Animal models have been developed for self-injurious behavior. Head
banging (148) and other forms of self-injury have been observed in socially de-
prived monkeys (149). By analogy, humans may respond to a socially isolated
environment by self-injurious behavior as a form of self-stimulation (150).
Some individuals exhibit the desire to perform self-injuring behaviors de-
spite a myriad of attempted therapeutic interventions. Despite various hypotheses
(151) and clinical observations (152–154), the etiology of self-injurious behav-
iors in people with autism spectrum disorders and other neuropsychiatric disor-
ders is poorly understood.
Understanding the biological etiologies (155) of self-injury in people with
autism spectrum disorders can be fostered by examination of other genetically
distinct syndromes with self-injury (156), including the Lesch-Nyhan (136,152,
153), Cornelia de Lange (154), and Riley-Day (familial dysautonomia) syn-
dromes. A striking condition with self-injury is the Lesch-Nyhan syndrome, an
X-linked recessive disorder of males resulting from an absence or deficiency of
hypoxanthineguanine phosphoribosyltransferase (HGPRT), an enzyme to metab-
olize purine, a product of normal metabolism, to uric acid (136,152,153). Hall-
Movement Disorders 289
The GSIBS gives a longitudinal picture of the self-injury exhibited by the patient
in the past week. It is completed by the clinician, utilizing all sources of informa-
tion after a thorough interview and examination of the patient and an interview
with the caregivers, guardians, teachers, neighbors, and all others who have con-
tact with the patient. Clinicians assessing and treating people with self-injurious
behaviors benefit from the administration of the TSIBS and the GSIBS before
treatment begins and regularly during the course of treatment to assess its effects
(24,185).
The occurrence of self-injurious behaviors requires immediate intervention
to prevent morbidity and mortality. However, the nature of optimal treatment is
moot. Several treatment modalities have been suggested for self-injurious behav-
ior in people with autism spectrum disorders (132,186). However, poor methodol-
ogy hinders the objective assessment of many reports (187).
Psychiatric inpatient units may be reluctant to admit patients with self-
injurious behavior for several reasons. First, observation of the behavior is likely
to upset other patients and visitors. Second, self-injurious behaviors can disrupt
scheduled ward activities. Third, the need for immediate intervention requires
the active participation of multiple staff members at a moment’s notice. Fourth,
these patients typically require one-to-one observation by a staff member to pre-
vent injury. Fifth, many staff members lack training in the management of self-
injurious behaviors. These concerns on the part of administration and staff at
psychiatric facilities are understandable.
Therefore, because of the dearth of psychiatric inpatient facilities available
to treat self-injurious behaviors in people with autism spectrum disorders, there
exists a need for specialized inpatient programs for their evaluation and treatment.
For example, the Kennedy-Krieger Institute in Baltimore, Maryland, has a unit
where patients stay for days, weeks, or longer for the evaluation and treatment of
self-injurious and other challenging behaviors. However, because many patients
cannot readily travel to specialized units, particularly for an inpatient stay of
weeks or months, alternative strategies are needed. The shortage of such treatment
facilities is a problem that remains to be adequately addressed and resolved by
the policy-makers and planners of health care in many locations. Out of despera-
tion, some parents are forced to improvise physical restraints, including tying
together a child’s hands to prevent face slapping and eye poking or wrapping a
child in a blanket to prevent head-to-object or body-to-object banging. At-
tempting to handle a child with self-injurious behavior is a tremendous burden
on parents who are typically already exhausted by the burden of caring for a
severely disturbed child. Family situations may be complicated by physical abuse
when a frustrated parent loses control in attempting to handle a child with self-
injurious behavior who appears recalcitrant to all therapeutic interventions (6).
Adequate inpatient treatment facilities are needed for these patients. Referral to
a specialized facility such as the Kennedy-Krieger Institute may be the appro-
Movement Disorders 291
1. Behavioral Interventions
Behavior modification (188–193) employs the principle that the frequency of a
behavior will increase if it is rewarded and decrease if it is not rewarded. For
instance, a patient in a bleak environment may enjoy the staff attention that results
from self-injury. Alternatively, a patient may learn that onerous chores can be
avoided through self-injury. Program adjustment resulting in less desirable conse-
quences may then reduce or eliminate the occurrence of self-injury. Since behav-
ioral programs may require considerable staff intervention, a Staff Intensity Scale
has been developed (194).
Behavioral intervention applied consistently at home and at school by per-
sons experienced in the application of techniques for people with pervasive devel-
opmental disorders is probably the most generally effective therapeutic approach
for autism spectrum disorders (195). By consistently applying behavioral inter-
ventions on a regular basis at home, school, and work, and in other settings, the
onset of acute episodes of self-injurious behavior can frequently be aborted. Also,
the institution of behavioral techniques as soon as the diagnosis of autism spec-
trum disorder is made helps minimize the morbidity and mortality of the child
and the family. Since self-injury may represent an attempt to communicate basic
needs, an assessment of the possible intended communication may facilitate more
appropriate communication (196,197). Behavioral strategies may be more diffi-
cult to apply and less effective for older children and adults.
In higher-functioning persons with autism spectrum disorders, a token
economy is often helpful to lessen or abolish self-injurious behaviors. Patients
are given tangible rewards for on-task behaviors. That is, they are differentially
reinforced for behaviors other (DRO) than self-injury (198,199) and for behaviors
that are incompatible (DRI) with self-injury. The rewards take the form of items
that are intrinsically pleasurable, such as food and playing cards, or they may be
tokens for exchange for privileges or gifts. The form of the rewards varies ac-
cording to the developmental level of the subject. When self-injury occurs, the
subject is punished by forfeiting tokens or relinquishing rewards.
Refer to Chapter 17 for further information about this mainstay of the treat-
ment of autism spectrum disorders. Since self-injurious behaviors demand imme-
diate cessation, behavioral therapy may not be effective for acute intervention.
Behavioral methods have been classified as reduction or enhancement ap-
proaches (132).
D. Property Destruction
Rarely, stereotypies exhibited by people with autism spectrum disorders result
in potential property destruction. For example, repeated touching, hitting, or tap-
ping an object may cause it to break. Because such movements can quickly cause
severe damage, immediate intervention is needed. The discussions of self-injuri-
ous behavior (Section IV.B) and aggression toward others (Section IV.C) also
apply to stereotypies causing property damage.
Movement Disorders 297
V. SUMMARY
Movement disorders are a prominent feature of autism spectrum disorders. They
constitute a manifestation of the underlying autism spectrum disorder as well as
an adverse effect of many pharmacological interventions commonly utilized for
autism spectrum disorders. Unless a comprehensive movement-assessment bat-
tery is performed and recorded at baseline before the administration of any thera-
peutic intervention, clinicians may be unable to differentiate movement-disorder
adverse effects of treatment from movement-disorder manifestations of the au-
tism spectrum disorders. Acute dystonia (acute dystonic reaction), acute aka-
thisia, and tardive dyskinesia are movement disorders commonly observed in
people with autism spectrum disorders following pharmacological interventions,
particularly with dopamine antagonists.
People with autism spectrum disorders, particularly Rett’s disorder and au-
tistic disorder, have characteristic movements called stereotypies. Since most ste-
reotypies do not harm the individual or others, they can usually be adequately
managed by behavioral treatment. On the other hand, self-injurious behaviors
constitute challenging stereotypies exhibited by a minority of individuals with
autism spectrum disorders. Self-injurious behaviors demand immediate interven-
tion to prevent morbidity and mortality. Although many interventions are re-
ported to help treat movement disorders in specific individuals with autism spec-
trum disorders, no therapy is uniformly effective. All treatments for autism
spectrum disorders carry the risk of adverse events. Potential benefits of interven-
tions must be weighed against likely risks of unfavorable outcomes (304,305).
Published reports of treatments of movement disorders in autism are often anec-
dotal documents that are difficult to generalize to other individuals (306). Well-
designed research (307) is needed to assess diagnostic and therapeutic procedures
for movement disorders in autism spectrum disorders (308–310). Carefully
planned assessments of even single cases are needed to yield meaningful results
(311–313). The beneficial and adverse effects of treatments for these disorders
must be established through controlled clinical trials.
ACKNOWLEDGMENTS
This work is sponsored by the Department of Psychiatry of Bellevue Hospital
Center and the New York University School of Medicine, The Essel Foundation,
Family and Friends of Chelsea Coenraads, the National Alliance for Research
on Schizophrenia and Depression (NARSAD), the Rett Syndrome Research
Foundation (RSRF), and the Tourette Syndrome Association, Inc. The coopera-
tion of Bellevue Hospital Center and the Health and Hospitals Corporation of
the City of New York is gratefully acknowledged. This work was supported by
the Medical Fellows Program of the Consortium for Medical Education in Devel-
298 Brǎsić
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Examination Procedure
Either before or after completing the Examination Procedure, observe the subject unobtru-
sively at rest, e.g., in the waiting room. The chair to be used in this examination should
be a hard, firm one without arms.
Instructions
Complete the above Examination Procedure before making ratings. For movement ratings,
circle the highest severity observed.
300 Brǎsić
Table 2 Continued
Code:
0 None
1 Minimal, may be extreme normal
2 Mild
3 Moderate
4 Severe
Extremity Movements
5. Upper (arms, wrists, hands, fingers)—include choreic movements 0 1 2 3 4
(i.e., rapid, objectively purposeless, irregular, spontaneous),
athetoid movements (i.e., slow, irregular, complex, serpentine).
Do not include tremor (i.e., repetitive, regular, rhythmic).
6. Lower (legs, knees, ankles, toes)—e.g., lateral knee movement, 0 1 2 3 4
foot tapping, heel dropping, foot squirming, and inversion and
eversion of the foot
Trunk Movements
7. Neck, shoulders, hips—e.g., rocking, twisting, squirming, pelvic 0 1 2 3 4
gyrations
Global Judgments
8. Severity of abnormal movements 0 1 2 3 4
9. Incapacitation due to abnormal movements 0 1 2 3 4
10. Subject’s awareness of abnormal movements—rate only the 0 1 2 3 4
subject’s report.
Dental Status
11. Current problems with teeth and/or dentures 0 No
1 Yes
12. Does the subject usually wear dentures? 0 No
1 Yes
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions to rater: Please rate the subject’s most impaired level of general functioning
for the specified time period by selecting the lowest level that describes his or her function-
ing on a hypothetical continuum of health–illness. Use the intermediary levels (e.g., 35,
58, 62). Rate actual functioning regardless of treatment or prognosis. The examples of
behavior provided are only illustrative and are not required for a particular rating.
Table 3 Continued
50–41 Moderate degree of interference in functioning in most social areas or severe
impairment of functioning in one area, such as might result from suicidal preoc-
cupations and ruminations, school refusal and other forms of anxiety, obsessive
rituals, major conversion symptoms, frequent anxiety attacks, poor or inappro-
priate social skills, or frequent episodes of aggressive or other antisocial behav-
ior with some preservation of meaningful social relationships.
40–31 Major impairment in functioning in several areas and unable to function in one
of these areas, i.e., disturbed at home, at school, with peers, or in society at
large, e.g., persistent aggression without clear instigation; markedly withdrawn
and isolated behavior due to either mood or thought disturbance, suicidal at-
tempts with clear lethal intent; such children are likely to require special school-
ing and/or hospitalization or withdrawal from school (but this is not a sufficient
criterion for inclusion in this category).
30–21 Unable to function in almost all areas, e.g., stays at home, in ward, or in bed
all day without taking part in social activities or severe impairment in reality
testing or serious impairment in communications (e.g., sometimes incoherent
or inappropriate).
20–11 Needs considerable supervision to prevent hurting others or self (e.g., frequently
violent, repeated suicide attempts) or to maintain personal hygiene or gross
impairment in all forms of communication, e.g., severe abnormalities in verbal
and gestural communications, marked social aloofness, stupor.
10–1 Needs constant supervision (24-hour care) due to severely aggressive or self-
destructive behavior or gross impairment in reality testing, communication, cog-
nition, affect, or personal hygiene.
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions to rater: Please circle the appropriate response or write in the appropriate
number based on all available sources of information.
Family factors
4. The family has refused to permit the use of medication(s). Yes* No
5. Family members have refused to make appointments. Yes* No
6. Number of appointments missed over the last 6 months.
7. The family has refused to sign the treatment plan. Yes* No
Practitioner factors
8. Number of sessions with primary therapist over the past 6
months.
9. Team member or primary therapist has appropriate contact with Yes No*
other providers, e.g., school, group residence, foster care
agency.
* If this response is circled, then immediate review is needed to assess the need for and the effective-
ness of current psychopharmacotherapy and to consider alternative treatment plans.
Source: Adapted from Ref. 13. 1998 Psychological Reports.
304 Brǎsić
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions to rater: Utilizing all available sources of information, please circle the
number corresponding to the most appropriate response for the past week.
Number
None 0
Single self-injurious behavior 1
Two self-injurious behaviors 2
Three self-injurious behaviors 3
Four self-injurious behaviors 4
Five or more self-injurious behaviors 5
Frequency
None There is no evidence of self-injurious behaviors. 0
Rarely Self-injurious behaviors occur rarely. Bouts of self-injurious
behaviors are brief and uncommon. 1
Occasionally There are occasional durations without any self-injurious
behaviors. Brief bouts of self-injurious behaviors
occasionally occur. 2
Frequently Bouts of single self-injurious behaviors occur regularly. 3
Almost always Periods of sustained self-injurious behaviors occur
regularly. Bouts of two or more self-injurious behaviors
are common. 4
Always Self-injurious behaviors are present constantly. There
are no observed waking intervals free of self-injurious
behaviors. 5
Intensity
Absent Self-injurious behaviors are absent. 0
Minimal Self-injurious behaviors are less forceful than comparable
voluntary actions. They may be masked by voluntary actions,
and may be overlooked unless the subject is observed
closely. There is no tissue damage. 1
Movement Disorders 305
Table 5 Continued
Mild Self-injurious behaviors are no more forceful than
comparable voluntary actions. Although there is no acute
tissue damage, tissue damage could occur if specific self-
injurious behaviors persisted for more than a day. 2
Moderate Self-injurious behaviors are more forceful than comparable
voluntary actions. They call attention to the individual
because of the intensity of the force. There may be minimal
tissue damage, such as bruises. 3
Severe Self-injurious behaviors are more forceful than comparable
voluntary actions. They call attention to the individual
because of their forceful and exaggerated character. Major
tissue damage requiring medical and surgical intervention,
such as laceration and hemorrhage, may occur. 4
Extreme Self-injurious behaviors are extremely forceful and
exaggerated in expression. They call immediate attention to
the individual. There may be major tissue damage of life-
threatening proportions, including fractures, rupture of
internal organs, loss of teeth, and impairments of hearing and
vision. 5
Complexity
None If present, specific self-injurious behaviors are clearly
“simple” (sudden, brief, purposeless) in character. 0
Borderline Some self-injurious behaviors are not clearly simple in
character. 1
Mild Some self-injurious behaviors are clearly complex (purposive
in appearance) and mimic brief automatic behaviors that can
be readily camouflaged. 2
Moderate Some self-injurious behaviors are more complex (more
purposive and sustained in appearance), and may occur in
orchestrated bouts that are difficult to camouflage but
resemble normal behavior or speech. 3
Marked Some self-injurious behaviors are highly complex in
character and tend to occur in sustained orchestrated bouts
that are difficult to camouflage and cannot be easily
rationalized as normal behavior because of the duration and
the unusual, inappropriate, or bizarre character, and the
severity of tissue damage. 4
Severe Some self-injurious behaviors involve lengthy bouts of
orchestrated behavior that are impossible to camouflage or
successfully rationalize as normal because of the duration
and extremely unusual, inappropriate, or bizarre character
(lengthy displays), and the severity of the tissue damage. 5
306 Brǎsić
Table 5 Continued
Interference
None No self-injurious behaviors are present. 0
Minimal Specific self-injurious behaviors do not interrupt the flow of
behavior or speech. 1
Mild Specific self-injurious behaviors occasionally interrupt the
flow of behavior or speech. 2
Moderate Specific self-injurious behaviors frequently interrupt the flow
of behavior or speech. 3
Marked Specific self-injurious behaviors frequently interrupt the flow
of behavior or speech and occasionally disrupt the intended
action or communication. 4
Severe Specific self-injurious behaviors constantly disrupt intended
action or communication. 5
Impairment
None There is no impairment in social, occupational, or
educational functioning. 0
Minimal Self-injurious behaviors are associated with subtle difficulties
in self-esteem, family life, social acceptance, school and job
functioning, activities of daily living, daily chores, and
vocational training (infrequent upset or concern about self-
injurious behaviors regarding the future; periodic or slight
increase in family tensions because of self-injurious
behaviors; friends or acquaintances may occasionally notice
or comment about self-injurious behaviors in an upsetting way). 1
Mild Self-injurious behaviors are associated with minor difficulties
in self-esteem, family life, social acceptance, school and job
functioning, activities of daily living, daily chores, and
vocational planning. 2
Moderate Self-injurious behaviors are associated with definite
difficulties in self-esteem, family life, social acceptance,
school and job functioning, activities of daily living, daily
chores, and vocational training (episodes of dysphoria;
periodic distress and upheaval in the family; frequent
tensions in caretaking staff; frequent teasing by peers or
episodic social avoidance; periodic interference in school or
job performance because of self-injurious behaviors; periodic
interference in activities of daily living and daily chores by
self-injurious behaviors). 3
Severe Self-injurious behaviors are associated with major difficulties
in self-esteem, family life, social acceptance, school and job
functioning, activities of daily living, daily chores, and
vocational training. 4
Movement Disorders 307
Table 5 Continued
Interference
Extreme Self-injurious behaviors are associated with extreme
difficulties in self-esteem, family life, social acceptance,
school and job functioning, activities of daily living, daily
chores, and vocational training (severe depression with
suicidal ideation and attempts; disruption of family including
separation, divorce, and residential placement; interference
with the routine functioning of caretaking staff; disruption of
social ties and peer interactions including severely restricted
life because of social stigma and social avoidance; removal
from school or vocational activity; extreme interference with
activities of daily living and daily chores). 5
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Subjective score
Objective score
Total score
Table 6 Continued
Clinical Global Impression
Severity of akathisia Score
Considering your total experience with this particular population, how akathisic is the
subject at this time?
0 Not assessed
1 Normal, not akathisic
2 Borderline akathisia
3 Mildly akathisic
4 Moderately akathisic
5 Markedly akathisic
6 Severely akathisic
7 Among the most akathisic of subjects
Global improvement Score
Rate total improvement whether or not, in your judgment, it is entirely due to drug
treatment. Compared with subject’s condition at admission to the study, how much has
he or she changed.
0 Not assessed
1 Very much improved
2 Much improved
3 Minimally improved
4 No change
5 Minimally worse
6 Much worse
7 Very much worse
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions for rater: Fill out separate checklists for each different movement, posture,
or utterance observed. Do not rate two or more particular movements, postures, or
utterances on the same sheet. Complete the following items based on all available
sources of information concerning each movement, posture, or utterance on the date of
the rating.
1 Abnormal posture 0 1 9
2 Abrupt 0 1 9
3 Brief 0 1 9
4 Can be suppressed 0 1 9
5 Continuous 0 1 9
6 Coordinated 0 1 9
7 Feeling of restlessness 0 1 9
8 Intermittent 0 1 9
9 Movement flows randomly 0 1 9
10 Oscillatory 0 1 9
11 Patterned 0 1 9
12 Present at rest 0 1 9
13 Present when maintaining a posture 0 1 9
14 Purposeless 0 1 9
15 Regular 0 1 9
16 Repetitive 0 1 9
17 Ritualistic 0 1 9
18 Shock-like 0 1 9
19 Squeezing movement 0 1 9
20 Sudden 0 1 9
21 Sustained 0 1 9
22 Twisting movement 0 1 9
23 Urge to move 0 1 9
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions: Please complete the following items based on all available sources of
information for the past 7 days.
1. Simple jerks 0 1 9
2. Random, unpredictable body distribution 0 ⫺1 9
3. Wide range of amplitude 0 1 9
4. Wide range of forcefulness 0 1 9
5. Movements can be voluntarily suppressed 0 1 9
6. Movements increase with intentional acts 0 ⫺1 9
7. Sustained dystonic movements 0 1 9
8. Complex movements 0 1 9
To score, convert all 9s to 0s. If the total score is positive, tics are more likely. If the
total score is negative, myoclonus is more likely. If the total score is zero, myoclonus
and tics are equally likely.
Source: Adapted from Ref. 17. 2000 Psychological Reports.
312 Brǎsić
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions to rater: These guidelines apply to all items unless indicated otherwise.
After reviewing the subject’s chart for the 12 months before the rating date, circle Y if
the stated item is true. For example, if the response to the item is no, not applicable,
none, don’t know, other, or any response other than yes for an item, leave it blank.
You may write any additional information on the backs of the pages.
Table 9 Continued
20. Subject’s level of mental
retardation
1 ⫽ Profound (IQ below 20 or 25)
2 ⫽ Severe (IQ 20–25 to 35–40)
3 ⫽ Moderate (IQ 35–40 to 50–55)
4 ⫽ Mild (IQ 50–55 to approximately 70)
5 ⫽ Borderline (IQ 71 to 84)
6 ⫽ Not retarded
9 ⫽ Don’t know
21. Subject’s full-scale IQ, as
measured by standard individual
test
22. The subject is deceased. Y
Caregivers
23. Subject’s primary clinician
24. Clinician 1 managed medications for subject
25. Clinician 2 managed medications for subject
26. Clinician 3 managed medications for subject
Past history
27. Record of previous diagnostic evaluation requested* Y
28. Record of previous diagnostic evaluation obtained* Y
Current medical evaluation
29. Comprehensive nonpsychiatric medical evaluation is initiated* Y
30. Comprehensive nonpsychiatric medical evaluation is completed* Y
31. Relevant nonpsychiatric medical assessment components are completed* Y
Current psychiatric evaluation
32. Comprehensive psychiatric evaluation is initiated* Y
33. Comprehensive psychiatric evaluation is completed* Y
34. Relevant psychiatric assessment components are completed* Y
35. Psychiatric diagnoses other than mental retardation, if applicable, by
DSM-IV-TR diagnostic classification (2)* Y
36. Psychiatric diagnoses other than mental retardation, if applicable, by 1998
ICD-9-CM (22) classification* Y
Current intellectual evaluation
37. Level of mental retardation is documented by IQ derived from individual
formal testing* Y
38. Level of retardation is documented by adaptive functioning* Y
Current behavioral evaluation
39. Focused behavioral evaluation is initiated* Y
40. Focused behavioral evaluation is adequate for initiating treatment* Y
41. Behavioral symptoms related to psychiatric diagnoses are specified* Y
314 Brǎsić
Table 9 Continued
42. Behavioral symptoms related to medical diagnoses are specified* Y
43. Baseline behavioral symptoms similar to treatment side effects are specified* Y
Specific medical diagnoses
44. Pulmonary disease is diagnosed Y
45. Cardiovascular disease is diagnosed Y
46. Cataracts are diagnosed Y
47. The subject is hepatitis A antigen–positive Y
48. The subject is hepatitis B antigen–positive Y
49. Constipation is diagnosed Y
50. Toe infection is present Y
51. Ear/nose/throat disease is present Y
52. Respiratory infection is diagnosed Y
53. Central nervous system disease is diagnosed Y
54. Seizures are diagnosed Y
55. Endocrine disease is diagnosed Y
56. The subject ambulates with assistance Y
57. Mental retardation due to disorders of metabolism and nutrition is diagnosed Y
58. Mental retardation due to infection (e.g., rubella) or head trauma is diagnosed Y
59. Cerebral malformations are diagnosed Y
60. Down’s syndrome is diagnosed Y
61. Fragile X syndrome is diagnosed Y
62. Other chromosomal disorders are diagnosed Y
63. One or more seizures are recorded in the past 12 months Y
64. Stereotypies are diagnosed Y
65. Neuroleptic-related tardive or withdrawal dyskinesias are diagnosed Y
66. Additional medical diagnoses are given Y
67. It is specified if medical diagnoses contribute to target symptoms Y
68. Another nonpsychiatric medical condition, other than mental retardation,
is diagnosed Y
If yes, please list:
69. Other informal clinical symptom diagnoses, e.g., self-injurious behavior,
are given Y
70. Hierarchy of severity of medical diagnoses is evident Y
Specific psychiatric diagnoses
71. Autistic disorder is diagnosed Y
72. Other pervasive developmental disorder is diagnosed Y
73. Schizophrenia is diagnosed Y
74. Depression is diagnosed Y
75. Mania is diagnosed Y
76. Self-injurious behavior has been recorded Y
77. Aggression toward others has been recorded Y
78. Suicide has been attempted in the past year Y
79. Hyperactivity has been recorded in the past year Y
Movement Disorders 315
Table 9 Continued
80. Another nonmedical psychiatric diagnosis, other than mental retardation,
is diagnosed Y
If yes, please list:
81. Hierarchy of severity of psychiatric diagnoses is evident Y
Target symptoms
82. More than one staff member has recorded each significant symptom* Y
83. There is an obvious environmental cause for each symptom† Y
84. There is obvious bias by the observer; others do not agree† Y
85. More than one significant symptom is observed Y
86. A hierarchy of symptom priorities for treatment is listed* Y
87. Target symptom(s) for treatment are established* Y
88. Baseline ratings of symptoms are obtained by rating scales Y
89. Baseline ratings of symptoms are obtained by informal observation* Y
90. Other target behavior symptoms are identified Y
If yes, please list:
91. There is a hierarchy of severity of target behavioral symptoms* Y
Treatment selection
92. Only one available psychoactive treatment is considered and reviewed† Y
93. More than one available psychoactive treatment is considered and reviewed* Y
94. Beneficial and side effects of each psychoactive treatment are reviewed* Y
95. Sequence of psychoactive treatments is established* Y
96. Caution to do no harm to subject in treatment selection* Y
97. Informed consent is obtained prior to starting psychoactive medication* Y
98. Class of psychoactive medication selected in relation to psychiatric
diagnoses* Y
99. Class of psychoactive medication selected in relation to target behavioral
symptom(s)* Y
100. Class of psychoactive medication selected in relation to concurrent medical
illnesses* Y
101. Class of psychoactive medication selected in relation to drug interactions* Y
102. Contraindicated and ineffective psychoactive medication(s) were excluded Y
103. Alternative psychoactive medication(s) are recorded* Y
Treatment monitoring protocols
104. A behavioral treatment plan is specified* Y
105. A pharmacological treatment plan is specified* Y
106. The duration of psychoactive treatments is specified, other than monthly
renewals* Y
107. After the monthly review of the symptoms of the subject, medication
renewals are completed* Y
108. Outcome criteria are specified to determine continuation of therapy* Y
109. The time and the method to determine long-term side effects are recorded Y
110. Psychoactive medication review by preset schedule, other than monthly
medication renewals Y
316 Brǎsić
Table 9 Continued
111. Psychoactive medication review by original protocol schedule Y
112. Psychoactive medication review by number of drugs* Y
113. Psychoactive medication review by long-term side effects* Y
114. Nonmedication influences reviewed: baseline variation* Y
115. Nonmedication influences reviewed: environmental* Y
116. Nonmedication influences reviewed: concurrent treatments* Y
Medication dosage range
117. Psychoactive drug dosage in usual range according to Physicians’ Desk
Reference (23)* Y
118. Psychoactive drug dosage in usual range according to state manual* Y
119. Psychoactive drug dosage in usual range according to standard texts* Y
120. Psychoactive drug dosage in usual range according to scientific journals* Y
Monitoring medication dosage and treatment effects
121. Psychoactive drug dosage is monitored by formal protocol schedule Y
122. Psychoactive drug dosage is monitored by open drug trial Y
123. Psychoactive drug dosage is monitored by plasma drug levels Y
124. Psychoactive drug dosage is monitored by consideration of other drugs
currently taken* Y
125. Appropriate beneficial behavioral effects are monitored* Y
126. Appropriate medication side effects are monitored* Y
127. Monitoring by staff observation, with chart notes, at one site (day program
or living unit)* Y
128. Monitoring by staff, with chart notes, at two or more sites (including both
day program and living unit) Y
129. Monitoring by appropriate specific rating scales filled out by assigned raters Y
130. Monitoring by appropriate specific rating scales, with raters by convenience Y
131. Monitoring by open format Y
132. Monitoring by single-blind format Y
133. Monitoring by double-blind format Y
134. Monitoring includes use of placebo Y
135. Monitoring includes crossover of treatment components Y
Drug holidays
136. Drug holiday of at least 4 weeks each year planned Y
137. Drug holiday of at least 4 weeks this year attempted Y
138. Drug holiday of at least 4 weeks this year completed Y
139. No drug holiday, with documentation supporting this decision Y
140. Attempt at drug holiday was discontinued, with justification Y
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions: Please place a checkmark (⻫) on the appropriate line the first time the
indicated behavior occurs during each 10-second interval for each minute of the 10-
minute rating session.
Table 10 Continued
9. Eye poking
10. Anal poking
11. Other poking
Please describe:
12. Lip chewing
13. Hair removal
14. Nail removal
15. Teeth banging
16. Other self-injurious behavior
Please describe:
Table 10 Continued
12. Lip chewing
13. Hair removal
14. Nail removal
15. Teeth banging
16. Other self-injurious behavior
Please describe:
Table 10 Continued
15. Teeth banging
16. Other self-injurious behavior
Please describe:
Table 10 Continued
8:00 8:10 8:20 8:30 8:40 8:50
1. Skin picking
2. Self biting
3. Head punching
4. Head slapping
5. Head-to-object banging
6. Body-to-object banging
7. Body punching
8. Body slapping
9. Eye poking
10. Anal poking
11. Other poking
Please describe:
12. Lip chewing
13. Hair removal
14. Nail removal
15. Teeth banging
16. Other self-injurious behavior
Please describe:
Subject Name
Subject Number
Rater Name
Date of Rating
Time of Rating
Place of Rating
Instructions: Place a checkmark (⻫) in the appropriate box the first time the indicated stereotypy occurs during each 30-second interval.
Write a description of each “other” stereotypy, namely, items 19–24, 27–32, 46–51, 60–65, 68–73, and 77–89.
Brǎsić
Movement Disorders 323
324 Brǎsić
Movement Disorders
Chorea—an abrupt, quick, brief, continuous, irregular movement that appears to flow
randomly from one body part to another (11).
Choreic movements have dancelike qualities. The movements of chorea may be
camouflaged to resemble intentional movement. For example, a choreic hand
movement may be followed by a movement to touch the head, giving the appearance
that the individual deliberately groomed his or her hair.
Table 12 Continued
Tic—an abrupt, brief, discontinuous, and intermittent motion, sound, sensation, or
utterance that can be suppressed temporarily (11,42).
Tics commonly present in childhood, affecting boys much more frequently than
girls (2). An individual may experience an urge to perform a tic, which subsides with a
feeling of relief when the tic is completed. Tics can typically be suppressed, especially
when the individual is concentrating intensely on a physical or mental activity. When
the tics are finally expressed, there may be an explosion of them, as if to make up for
the time during which the expression of the tics was suppressed.
Motor and phonic, or vocal, tics are frequently observed. Common motor tics
include eye blinks, mouth twitches, and shoulder shrugs. Copropraxia is the
performance of obscene gestures. Common phonic tics include heavy breathing, throat
clearing, sniffing, bird sounds, and animal sounds, including barking. Coprolalia is the
utterance of obscene and profane expressions. Copropraxia and coprolalia occur in a
minority of individuals with Tourette’s disorder, which is characterized by the presence
of both motor and phonic tics for at least a year (2).
The Myoclonus Versus Tic Checklist (MVTC) (Table 8) (17) assists in the
differentiation of tics from myoclonus.
Further information about tics may be obtained from the Tourette Syndrome
Association, Inc. See page 285 for contact information.
Bouncing—jumping up and down repeatedly. Often occurs when the person is happy
and excited.
Bronx cheer—forcefully blowing air through tightly closed lips to produce a sound
similar to a flatulent report.
Ear covering—placing the palms over the external ears as if to block hearing.
Eye covering—placing the palms over the eyes as if to block vision.
Finger wiggling—movements resembling piano playing.
Forceful breathing—heavy expiration and inspiration through an open mouth.
Hand flapping—flinging both hands up and down repeatedly with a limp wrist.
Typically occurs when the individual is excited or happy; may be associated with
bouncing and rotating.
Hand rubbing—moving the palm repeatedly over an item. Appears to be associated
with an intense sensory interest in the feeling of the texture and configuration of an
item of interest.
Head tilting—leaning the head to one side. Often associated with fixation of visual
focus on a specific item in the environment. The individual appears to be attempting
to obtain a better view of the object.
Rotating—moving around a vertical axis in the head and body. May also be seen with
bouncing and hand flapping. Rotating may be a manifestation of excitement and
pleasure.
Toe wiggling—movements resembling playing piano with one’s toes.
REFERENCES
1. Dobbinson S, Perkins MR, Boucher J. Structural patterns in conversations with a
woman who has autism. J Commun Disord 1998; 31:113–134.
2. American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders. 4th ed., text revision. Washington, DC: American Psychiatric Publish-
ing, 2000.
3. Teitelbaum P, Teitelbaum O, Nye J, Fryman J, Maurer RG. Movement analysis
in infancy may be useful for early diagnosis of autism. Proc Natl Acad Sci USA
1998; 95:13982–13987.
4. Brašić JR. Movements in autistic disorder. Med Hypotheses 1999; 53(1):48–49.
5. Brašić JR. Pervasive developmental disorder: Asperger syndrome. eMedicine
Journal Jan 7, 2002; 3(1).
6. Brašić JR: Pervasive developmental disorder: autism. eMedicine Journal Feb 19,
2002; 3(2).
7. Brašić JR, Gianutsos JG. Neuromotor assessment and autistic disorder. Autism
Int J Res Pract 2000; 4(3):287–298.
8. Gillberg C, Coleman M. The Biology of the Autistic Syndromes. 3rd ed. London:
MacKeith Press, 2000.
9. Brašić JR, Barnett JY. Hyperkinesias in a prepubertal boy with autistic disorder
treated with haloperidol and valproic acid. Psychol Rep 1997; 80:163–170.
10. National Institute of Mental Health (NIMH), Alcohol, Drug Abuse, and Mental
Health Administration, Public Health Service, Department of Health, Education,
and Welfare. Abnormal Involuntary Movement Scale (AIMS). Psychopharmacol
Bull 1988; 24(4):781–783.
11. Brašić JR, Bronson B: Tardive dyskinesia. eMedicine Journal May 20, 2002; 3(5).
12. Shaffer D, Gould MS, Brasic J, Ambrosini P, Fisher P, Bird H, Aluwahlia S. A
Children’s Global Assessment Scale (CGAS) (for children 4 to 16 years of age).
Psychopharmacol Bull 1985; 21(4):747–748.
330 Brǎsić
13. Brašić JR, Will MV, Ahn SC, Nadrich RH, McNally G. A review of the literature
and a preliminary study of family compliance in a developmental disabilities clinic.
Psychol Rep 1998; 82:275–286.
14. Brašić JR, Nadrich RH, Kleinrock S, Brathwaite C. Do families comply with child
and adolescent psychopharmacology? Child Adolesc Psychopharmacol News
2001; 6(2):6, 7, 10.
15. Fleischhacker WW, Bergmann KJ, Perovich R, Pestreich LK, Borenstein M, Lie-
berman J, Kane J. The Hillside Akathisia Scale: a new rating instrument for neuro-
leptic-induced akathisia. Psychopharmacol Bull 1989; 25:222–226.
16. Medical Editors’ Trial Amnesty (META). J Neurol 1998; 245:563–564.
17. Brašić JR. Differentiating myoclonus from tics. Psychol Rep 2000; 86:155–156.
18. Brašić JR, Young JG, Furman J, Conte RM, Baisley WE, Jaslow RI. Psychoactive
Medication Quality Assurance Rating Survey (PQRS). J Dev Phys Disabil 1997;
9:311–336.
19. Brašić JR, Furman J, Conte RM, Baisley WE, Nadrich RH, Kaplan D, Ahmad R,
Rodriguez M, Mendonça MF, Will MV. Quality assurance of treatments for Tou-
rette syndrome and other tic disorders [abstr]. 3rd International Scientific Sympo-
sium on Tourette Syndrome, New York, June 4–6, 1999, p 20.
20. Brašić JR, Furman J, Conte RM, Baisley WE, Jaslow RI. Psychoactive Medication
Quality Assurance Rating Survey (PQRS) screening criteria. J Dev Phys Disabil.
In press.
21. Brašić JR, Furman J, Conte RM, Baisley WE, Jaslow RI. Improving the quality
of the utilization of psychoactive medication. J Dev Phys Disabil. In press.
22. 1998 International Classification of Diseases. Ninth Revision, Clinical Modifica-
tion, Fifth Edition. Salt Lake City: Medicode Publications, 1997.
23. Physicians’ Desk Reference. 56th ed. Montvale, NJ: Medical Economics, 2002.
www.PDR.net.
24. Brasic JR, Barnett JY, Ahn SC, Nadrich RH, Will MV, Clair A. Clinical assess-
ment of self-injurious behavior. Psychol Rep 1997; 80:155–160.
25. Campbell M. Timed Stereotypies Rating Scale. Psychopharmacol Bull 1985;
21(3):1082.
26. Steffenburg S, Gillberg C, Steffenburg U. Psychiatric disorders in children and
adolescents with mental retardation and active epilepsy. Arch Neurol 1996; 53:
904–912.
27. Brašić JR, Zagzag D, Kowalik S, Prichep L, John ER, Liang HG, Kutchko B,
Cancro R, Sheitman BB, Buchsbaum M, Brathwaite C. Progressive catatonia. Psy-
chol Rep 1999; 84:239–246.
28. Brašić JR, Zagzag D, Kowalik S, Prichep L, John ER, Barnett JY, Bronson B,
Nadrich RH, Cancro R, Buchsbaum M, Brathwaite C. Clinical manifestations of
progressive catatonia. Ger J Psychiatry 2000; 3:13–24.
29. Brašić JR. Catatonia. eMedicine Journal Sept 27, 2002; 3(9).
30. Brašić JR, Barnett JY, Kowalik S, Prichep L, John ER, Nadrich RH, Will MV,
Bronson B, Ahmad R, Kurz L, Brathwaite C, Cancro R. Catatonia in pervasive
developmental disorders [abstr]. Mov Disord 2001; 16(suppl 1):S46–S47.
31. Brašić JR, Zagzag D, Kowalik S, Prichep L, John ER, Buchsbaum M, Barnett JY,
Nadrich RH, Kaplan D, Ahmad R, Mendonça MF, Rodriguez M, Bronson B, Can-
Movement Disorders 331
50. Brašić JR, Furman JW, Conte RM, Baisley WE. Continuous quality improvement
in child and adolescent psychopharmacology. Child Adolesc Psychopharmacol
News 1999; 4(3):9–10.
51. Brašić JR, Furman J, Conte RM, Baisley WE, Jaslow RI. Assuring the quality of
the utilization of psychoactive medication by people with mental retardation and
developmental disabilities by assessing dosages. Ger J Psychiatry 2000; 3(3): 7–
12. www.gjpsy.uni-goettingen.de/gjp-article-brasic2.pdf.
52. Campbell M, Armenteros JL, Malone RP, Adams PB, Eisenberg ZW, Overall JE.
Neuroleptic-related dyskinesias in autistic children: a prospective, longitudinal
study. J Am Acad Child Adolesc Psychiatry 1997; 36:835–843.
53. Locascio JJ, Malone RP, Small AM, Kafantaris V, Ernst M, Lynch NS, Overall
JE, Campbell M. Factors related to haloperidol response and dyskinesias in autistic
children. Psychopharmacol Bull 1991; 27(2):119–126.
54. Armenteros JL, Adams PB, Campbell M, Eisenberg ZW. Haloperidol-related dys-
kinesias and prenatal and perinatal complications in autistic-children. Psychophar-
macol Bull 1995; 31:363–369.
55. Advokat CD, Mayville EA, Matson JL. Side effect profiles of atypical antipsychot-
ics, typical antipsychotics, or no psychotropic medications in persons with mental
retardation. Res Dev Disabil 2000; 21:75–84.
56. Simonic I, Gericke GS, Lippert M, Schoeman JF. Additional clinical and cytoge-
netic findings associated with Rett syndrome. Am J Med Genet 1997; 74:331–337.
57. Berger-Sweeney J, Hohmann CF. Behavioral consequences of abnormal cortical
development: insights into developmental disabilities. Behav Brain Res 1997; 86:
121–142.
58. Hohmann CF, Berger-Sweeney J. Cholinergic regulation of cortical development
and plasticity: new twists to an old story. Perspect Dev Neurobiol 1998; 5:401–
425.
59. Wan M, Zhao K, Lee SSJ, Francke U. MECP2 truncating mutations cause histone
H4 hyperacetylation in Rett syndrome. Hum Molec Genet 2001; 10:1085–1092.
60. Lee SSJ, Wan M, Francke U. Spectrum of MECP2 mutations in Rett syndrome.
Brain Dev 2001; 23(suppl 1):S138–S143.
61. Francke U. Understanding Rett Syndrome: where are we? [abstr]. 3rd Annual Rett
Syndrome Symposium, Baltimore, June 17–19, 2002. Cincinnati: Rett Syndrome
Research Foundation (RSRF), 2002:1.
62. Bailey MES, Mash V, Hever AM, Boxer M, Ørstavik KH, Reilly S, Kerr AM, Cass
H, Charman T. A detailed genotype-phenotype correlation study in Rett syndrome
[abstr]. 3rd Annual Rett Syndrome Symposium, Baltimore, June 17–19, 2002.
Cincinnati: Rett Syndrome Research Foundation (RSRF), 2002:35.
63. Vaudry D, Stork PJS, Lazarovici P, Eiden LE. Signaling pathways for PC12 cell
differentiation: making the right connections. Science 2002; 296:1648–1649.
www.sciencemag.org.
64. Stearns NA, Floerke-Nashner L, Berger-Sweeney J. Behavioral characterization of
a mouse model of Rett syndrome [abstr]. 3rd Annual Rett Syndrome Symposium,
Baltimore, June 17–19, 2002. Cincinnati: Rett Syndrome Research Foundation
(RSRF), 2002:33.
65. Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG binding
Movement Disorders 333
80. Maguire D, Bachman C. Anaesthesia and Rett syndrome: a case report. Can J
Anaesth 1989; 36:478–481.
81. Brašić JR, Barnett JY, Nadrich RH, Kaplan D, Ahmad R, Rodriguez M, Mendonça
MF. Evaluation of self-injurious tics [abstr]. 3rd International Scientific Sympo-
sium on Tourette Syndrome, New York, June 4–6, 1999, p 19.
82. Brašić JR, Zagzag D, Kowalik S, Prichep L, John ER, Buchsbaum M, Barnett JY,
Nadrich RH, Kaplan D, Ahmad R, Mendonça MF, Rodriguez M, Bronson B, Can-
cro R, Kurz L, Brathwaite C. Chronic motor tics are a manifestation of progressive
catatonia [abstr]. 3rd International Scientific Symposium on Tourette Syndrome,
New York, June 4–6, 1999, p 19.
83. Ringman JM, Jankovic J. Occurrence of tics in Asperger’s syndrome and autistic
disorder. J Child Neurol 2000; 15:394–400.
84. Kadesjo B, Gillberg C. Tourette’s disorder: epidemiology and comorbidity in pri-
mary school children. J Am Acad Child Adolesc Psychiatry 2000; 39:548–555.
85. Baron-Cohen S, Scahill VL, Izaguirre J, Hornsey H, Robertson MM. The preva-
lence of Gilles de la Tourette syndrome in children and adolescents with autism:
a large scale study. Psychol Med 1999; 29:1151–1159.
86. Goodman WK, Price LH, Rasmussen SA, Mazure C, Fleischmann RL, Hill CL,
Heninger GR, Charney DS. The Yale-Brown Obsessive Compulsive Scale. I. De-
velopment, use, and reliability. Arch Gen Psychiatry 1989; 46:1006–1011.
87. Goodman W, Price LH, Rasmussen SA, Mazure C, Delgado P, Heninger GR,
Charney DS. The Yale-Brown Obsessive Compulsive Scale. II. Validity. Arch
Gen Psychiatry 1989; 46:1012–1016.
88. Leckman JF, Riddle MA, Hardin MT, Ort SI, Swartz KL, Stevenson J, Cohen DJ.
The Yale Global Tic Severity Scale: initial testing of a clinician-rated scale of tic
severity. J Am Acad Child Adolesc Psychiatry 1989; 28:566–573.
89. Leckman JF, Towbin KE, Ort SI, Cohen DJ. Clinical assessment of tic disorder
severity. In: Cohen DJ, Bruun RD, Leckman JF, eds. Tourette’s Syndrome and
Tic Disorders. New York: John Wiley & Sons, 1988:55–78.
90. Arnold LE, Aman MG, Martin A, Collier-Crespin A, Vitiello B, Tierney E, Asar-
now R, Bell-Bradshaw F, Freeman BJ, Gates-Ulanet P, Klin A, McCracken JT,
McDougle CJ, McGough JJ, Posey DJ, Scahill L, Swiezy NB, Ritz L, Volkmar
F. Assessment in multisite randomized clinical trials of patients with autistic disor-
der: the Autism RUPP Network. J Autism Dev Disord 2000; 30:99–111.
91. Brasic JR, Barnett JY, Ahn SC, Roncoli M, Nadrich RH, Ahmad R. Characteriza-
tion of movement disorders in prepubertal boys with autistic disorder [abstr]. Mov
Disord 1996; 11(suppl 1):59.
92. Würbel H, Freire R, Nicol CJ. Prevention of stereotypic wire-gnawing in labora-
tory mice: effects on behaviour and implications for stereotypy as a coping re-
sponse. Behav Proc 1998; 42:61–72.
93. Brown DW. Autism, Asperger’s syndrome and the Crick-Mitchison theory of the
biological function of REM sleep. Med Hypotheses 1996; 47:399–403.
94. Ross LL, Yu D, Kropla WC. Stereotyped behavior in developmentally delayed or
autistic populations: rhythmic or nonrhythmic? Behav Modif 1998; 22:321–334.
95. Ridley RM. The psychology of perseverative and stereotyped behaviour. Prog
Neurobiol 1994; 44:221–231.
Movement Disorders 335
96. Lewis MH, Bodfish JW. Repetitive behavior disorders in autism. Ment Retard Dev
Disabil Res Rev 1998; 4(2):80–89.
97. Nijhof G, Joha D, Pekelharing H. Aspects of stereotypic behaviour among autistic
persons: a study of the literature. Br J Dev Disabil 1998; 44:3–13.
98. Magulac M, Landsverk J, Golshan S, Jeste DV. Abnormal involuntary movements
in neuroleptic-naı̈ve children and adolescents. Can J Psychiatry 1999; 44:368–
373.
99. Powell SB, Newman HA, Pendergast JF, Lewis MH. A rodent model of spontane-
ous stereotypy: initial characterization of developmental, environmental, and neu-
robiological factors. Physiol Behav 1999; 66:355–363.
100. Antoniou K, Kafetzopoulos E, Papadopoulou-Daifoti Z, Hyphantis T, Marselos
M. d-amphetamine, cocaine and caffeine: a comparative study of acute effects on
locomotor activity and behavioural patterns in rats. Neurosci Biobehav Rev 1998;
23:189–196.
101. Barik S, de Beaurepaire R. Evidence for a functional role of the dopamine D3
receptors in the cerebellum. Brain Res 1996; 737:347–350.
102. Chabrol H, Bonnet D, Roge B. Psychopharmacologie de l’autisme [the psycho-
pharmacology of autism]. Encéphale 1996; 22:197–203.
103. Buitelaar JK, Dekker MEM, van Ree JM, van Engeland H. A controlled trial with
ORG 2766, an ACTH-(4-9) analog, in 50 relatively able children with autism. Eur
Neuropharmacol 1996; 6:13–19.
104. Cazzullo AG, Musetti MC, Musetti L, Bajo S, Sacerdote P, Panerai A. β-Endophin
levels in peripheral blood mononuclear cells and long-term naltrexone treatment
in autistic children. Eur Neuropsychopharmacol 1999; 9:361–366.
105. Hollander E, Kaplan A, Cartwright C, Reichman D. Venlafaxine in children, ado-
lescents, and young adults with autism spectrum disorders: an open retrospective
clinical report. J Child Neurol 2000; 15:132–135.
106. Posey DJ, McDougle CJ. The pharmacotherapy of target symptoms associated
with autistic disorder and other pervasive developmental disorders. Harv Rev Psy-
chiatry 2000; 8:45–63.
107. Brasic JR, Barnett JY, Kaplan D, Sheitman BB, Aisemberg P, Lafargue RT, Ko-
walik S, Clark A, Tsaltas MO, Young JG. Clomipramine ameliorates adventitious
movements and compulsions in prepubertal boys with autistic disorder and severe
mental retardation. Neurology 1994; 44:1309–1312.
108. Brašić JR, Barnett JY, Sheitman BB, Lafargue RT, Kowalik S, Kaplan D, Tsaltas
MO, Ahmad R, Nadrich RH, Mendonça MF. Behavioral effects of clomipramine
on prepubertal boys with autistic disorder and severe mental retardation. CNS
Spectrums: Int J Neuropsychiatr Med 1998; 3(10):39–46.
109. Brašić JR, Barnett JY, Sheitman BB, Tsaltas MO. Adverse effects of clomipramine
[letter]. J Am Acad Child Adolesc Psychiatry 1997; 36:1165–1166.
110. Sanchez LE, Campbell M, Small AM, Cueva JE, Armenteros JL, Adams PB. A
pilot study of clomipramine in young autistic children. J Am Acad Child Adolesc
Psychiatry 1996; 35:537–544.
111. McDougle CJ, Kresch LE, Posey DJ. Repetitive thoughts and behavior in perva-
sive developmental disorders: treatment with serotonin reuptake inhibitors. J Au-
tism Dev Disord 2000; 30:427–435.
336 Brǎsić
148. Levison CA. The development of head banging in a young rhesus monkey. Am
J Ment Defic 1970; 75:323–328.
149. Allyn G, Deyme A, Bègue I. Self-fighting syndrome in macaques. I. A representa-
tive case study. Primates 1976; 17: 1–22.
150. Jones IH, Barraclough BM. Auto-mutilation in animals and its relevance to self-
injury in man. Acta Psychiatr Scand 1978; 58:40–47.
151. Cataldo MF, Harris J. The biological basis for self-injury in the mentally retarded.
Anal Interv Dev Disabil 1982; 2:21–38.
152. Kelley WM, Wyngaarden JB. Clinical syndromes associated with HGPRT defi-
ciency. In: Stanbury JB, Wyngaarden JB, et al., eds. The Metabolic Basis of Inher-
ited Disease. 5th ed. New York: McGraw-Hill, 1983:1115–1143.
153. Lesch M, Nyhan WL. A familial disorder of uric acid metabolism and central
nervous system function. Am J Med 1964; 36:561–570.
154. Shear CS, Nyhan WL, Kirman BH, Stern J. Self-mutilative behavior as a feature
of the de Lange syndrome. J Pediatr 1971; 78:506–509.
155. Gualtieri CT. The differential diagnosis of self-injurious behavior in mentally re-
tarded people. Psychopharmacol Bull 1989; 25(3):358–363.
156. Jones IH. Self-injury: toward a biological basis. Perspect Biol Med 1982; 26:137–
150.
157. Ciaranello RD, Anders TF, Barchas JD, Berger PA, Cann HM. The use of 5-
hydroxytryptophan in a child with Lesch-Nyhan syndrome. Child Psychiatry Hum
Dev 1976; 7:127–133.
158. Lloyd KG, Hornykiewicz O, Davidson L, Shannak K, Farley I, Goldstein M,
Shibuya M, Kelley WN, Fox IH. Biochemical evidence of dysfunction of brain
neurotransmitters in the Lesch-Nyhan syndrome. N Engl J Med 1981; 305:1106–
1111.
159. Lake CR, Ziegler MG. Lesch-Nyhan syndrome: low dopamine-beta-hydroxylase
activity and diminished sympathetic response to stress and posture. Science 1977;
196:905–906.
160. Mizuno TI, Yugari Y. Self mutilation in Lesch-Nyhan syndrome [letter]. Lancet
1974; i(7860):761.
161. Nyhan WL, Johnson HG, Kaufman IA, Jones KL. Serotonergic approaches to the
modification of behavior in the Lesch-Nyhan syndrome. Appl Res Ment Retard
1980; 1:25–40.
162. Mailis A. Compulsive targeted self-injurious behaviour in humans with neuro-
pathic pain: a counterpart of animal autotomy? Four case reports and literature
review. Pain 1996; 64:569–578.
163. Ritvo ER, Ornitz EM, La Franchi S. Frequency of repetitive behaviors in early
infantile autism and its variants. Arch Gen Psychiatry 1968; 19:341–347.
164. Delcomyn F. Neural basis of rhythmic behavior in animals. Science 1980; 210:
492–498.
165. Olson GA, Olson RD, Vaccarino AL, Kastin AJ. Endogenous opiates:1997. Pep-
tides 1998; 19:1791–1843.
166. Beckwith BE, Couk DI, Schumacher K. Failure of naloxone to reduce self-injuri-
ous behavior in two developmentally disabled females. Appl Res Ment Retard
1986; 7:183–188.
Movement Disorders 339
167. Davidson PW, Kleene BM, Carroll M, Rockowitz RJ. Effects of naloxone on self-
injurious behavior: a case study. Appl Res Ment Retard 1983; 4:1–4.
168. Sandman CA, Barron JL, Crinella FM, Donnelly JF. Influence of naloxone on
brain and behavior of a self-injurious woman. Biol Psychiatry 1987; 22:899–
906.
169. Sandman CA, Datta PC, Barron J, Hoehler FK, Williams C, Swanson JM. Nalox-
one attenuates self-abusive behavior in developmentally disabled clients. Appl Res
Ment Retard 1983; 4:5–11.
170. Ballinger BR, Armstrong J, Presley AS, Reid AH. Use of a standardized psychiat-
ric interview in mentally handicapped patients. Br J Psychiatry 1975; 127:540–
544.
171. Goldberg DP, Cooper B, Eastwood MR, Kedward HB, Shepherd M. A standard-
ized psychiatric interview for use in community surveys. Br J Prev Soc Med 1970;
24:18–23.
172. Read SG, Batchelor DH. Violent and self-injurious behaviour in mentally handi-
capped patients—psychopharmacological control. Int Clin Psychopharmacol
1986; 1(suppl 1):63–74.
173. Campbell M, Adams P, Small AM, Tesch LM, Curren EL. Naltrexone in infantile
autism. Psychopharmacol Bull 1988; 24(1):135–139.
174. Sandyk R. Naloxone abolishes self-injuring in a mentally retarded child. Ann Neu-
rol 1985; 17:520.
175. Spitzer RL, Williams JBW, Gibbon M, First MB. Structured Clinical Interview for
DSM-III-R–Patient Edition (SCID-P, Version 1.0). Washington, DC: American
Psychiatric Publishing, 1990.
176. Bernstein GA, Hughes JR, Mitchell JE, Thompson T. Effects of narcotic antago-
nists on self-injurious behavior: a single case study. J Am Acad Child Adolesc
Psychiatry 1987; 26:886–889.
177. Iwata BA, Pace GM, Kissel RC, Nau PA, Farber JM. The Self-Injury Trauma
(SIT) Scale: a method for quantifying surface tissue damage caused by self-injuri-
ous behavior. J Appl Behav Anal 1990; 23:99–110.
178. Baker SP, O’Neill B, Haddon W, Jr., Long WB. The Injury Severity Score: a
method for describing patients with multiple injuries and evaluating emergency
care. J Trauma 1974; 14(3):187–196.
179. Barancik JI, Chatterjee BF. Methodological considerations in the use of the Abbre-
viated Injury Scale in trauma epidemiology. J Trauma 1981; 21(8):627–631.
180. Champion HR, Sacco WJ, Lepper RL, Atzinger EM, Copes WS, Prall RH. An
anatomic index of injury severity. J Trauma 1980; 20(3):197–202.
181. Committee on Medical Aspects of Automotive Safety. Rating the severity of tissue
damage. I. The abbreviated scale. JAMA 1971; 215:277–280.
182. Committee on Medical Aspects of Automotive Safety. Rating the severity of tissue
damage. II. The comprehensive scale. JAMA 1972; 220:717–720.
183. Greenspan L, McLellan BA, Greig H. Abbreviated Injury Scale and Injury Severity
Score: a scoring chart. J Trauma 1985; 25:60–64.
184. Mayer T, Matlak ME, Johnson DG, Walker ML. The modified injury severity
scale in pediatric multiple trauma patients. J Pediatr Surg 1980; 15:719–726.
185. Brasic JR, Kisnad HV, Barnett JY, Kowalik S, Ahn SC, Nadrich RH, Mendonca
340 Brǎsić
203. Singh NN, Watson JE, Winton ASW. Treating self-injury: water spray versus fa-
cial screening or forced arm exercise. J Appl Behav Anal 1986; 19:403–410.
204. Peri T, Ben-Shakhar G, Orr, SP, Shalev AY. Psychophysiologic assessment of
aversive conditioning in posttraumatic stress disorder. Biol Psychiatry 2000; 47:
512–519.
205. Altmeyer BK, Williams DE, Sams V. Treatment of severe self-injurious and ag-
gressive biting. J Behav Ther Exp Psychiatry 1985; 16:169–172.
206. Brašić JR, Fogelman D. Clinician safety. Psychiatr Clin North Am 1999; 22:923–
940.
207. Azrin NH, Kaplan SJ, Foxx RM. Autism reversal: eliminating stereotyped self-
stimulation of retarded individuals. Am J Ment Defic 1973; 78:241–248.
208. Silverman K, Watanabe K, Marshall AM, Baer DM. Reducing self-injury and cor-
responding self-restraint through the strategic use of protective clothing. J Appl
Behav Anal 1984; 17:545–552.
209. Pace GM, Iwata BA, Edwards GL, McCosh KC. Stimulus fading and transfer in
the treatment of self-restraint and self-injurious behavior. J Appl Behav Anal1986;
19:381–389.
210. Luiselli JK. Modification of self-injurious behavior: an analysis of the use of con-
tingently applied protective equipment. Behav Modif 1986; 10:191–204.
211. Luiselli JK. Contingent glove wearing for the treatment of self-excoriating behav-
ior in a sensory-impaired adolescent. Behav Modif 1989; 13:65–73.
212. Lovaas I, Newson C, Hickman C. Self-stimulatory behavior and perceptual rein-
forcement. J Appl Behav Anal 1987; 20:45–68.
213. Clauser B, Gould K. Visual screening as a reductive procedure: an examination
of generalization and duration. Behav Residential Treat 1988; 3:51–61.
214. Watson J, Singh NN, Winton ASW. Suppressive effects of visual and facial screen-
ing on self-injurious finger-sucking. Am J Ment Defic 1986; 90:526–534.
215. Winton ASW, Singh NN, Dawson MJ. Effects of facial screening and blindfold
on self-injurious behavior. Appl Res Ment Retard 1984; 5:29–42.
216. Goodall E, Corbett J. Relationships between sensory stimulation and stereotyped
behaviour in severely mentally retarded and autistic children. J Ment Defic Res
1982; 26:163–175.
217. Stroh G, Buick D. The effect of relative sensory isolation on the behaviour of two
autistic children. In: Hutt SJ, Hutt C, eds. Behaviour Studies in Psychiatry. Oxford,
England: Pergamon Press, 1970:161–174.
218. Luiselli JK. Use of sensory extinction in treating self-injurious behavior: a caution-
ary note. The Behavior Therapist 1984; 7:142–160.
219. Norton RS. Side effects of sensory extinction and an alternative procedure for high
rates of self-injury. The Behavior Therapist 1985:8:86–87.
220. Rincover A. Sensory extinction: some answers and questions. The Behavior Thera-
pist 1985; 8:177–178.
221. Azrin NH, Besalel VA, Jamner JP, Caputo JN. Comparative study of behavioral
methods of treating severe self-injury. Behav Residential Treat 1988; 3:119–152.
222. Haring TG, Breen CG, Pitts-Conway V, Gaylord-Ross R. Use of differential rein-
forcemnt of other behavior during dyadic instruction to reduce stereotyped behav-
ior of autistic students. Am J Ment Defic 1986; 90:694–702.
342 Brǎsić
223. Matson JL, Keyes JB. A comparison of DRO to movement suppression time-out
and DRO with two self-injurious and aggressive mentally retarded adults. Res Dev
Disabil 1990; 11:111–120.
224. Parrish JM, Iwata BA, Dorsey MF, Bunck TJ, Slifer KJ. Behavior analysis, pro-
gram development, and transfer of control in the treatment of self-injury. J Behav
Ther Exp Psychiatry 1985; 16:159–167.
225. Repp AC, Deitz SM, Speir NC. Reducing stereotypic responding of retarded per-
sons by differential reinforcement of other behavior. Am J Ment Defic 1974; 79:
279–284.
226. Tarpley HD, Schroeder SR. Comparison of DRO and DRI on rate of suppression
of self-injurious behavior. Am J Ment Defic 1979; 84:188–194.
227. Hegel MT, Ferguson RJ. Differential reinforcement of other behavior (DRO) to
reduce aggressive behavior following traumatic brain injury. Behav Modif 2000;
24(1):94–101.
228. Luiselli JK. Functional assessment and treatment of aggressive and destructive
behaviors in a child victim of physical abuse. J Behav Ther Exp Psychiatry 1996;
27:41–49.
229. Paisey TJH, Whitney RB, Moore J. Person-treatment interactions across nonaver-
sive response-deceleration procedures for self-injury: a case study of effects and
side effects. Behav Residential Treat 1989; 4:69–88.
230. Lancioni GE, O’Reilly MF. A review of research on physical exercise with people
with severe and profound developmental disabilities. Res Dev Disabil 1998; 19:
477–492.
231. Repp AC, Singh NN, Olinger E, Olson DR. The use of functional analyses to test
causes of self-injurious behaviour: rationale, current status and future directions.
J Ment Defic Res 1990; 34:95–105.
232. Durand VM, Crimmins DB. Identifying the variables maintaining self-injurious
behavior. J Autism Dev Disord 1988; 18:99–117.
233. Iwata BA, Pace GM, Kalsher MJ, Cowdery GE, Cataldo MF. Experimental analy-
sis and extinction of self-injurious escape behavior. J Appl Behav Anal 1990; 23:
11–27.
234. Steege MW, Wacker DP, Cigrand KC, Berg WK, Novak CG, Reimers TM, Sasso
GM, DeRaad A. Use of negative reinforcement in the treatment of self-injurious
behavior. J Appl Behav Anal 1990; 23:459–467.
235. Bourland G, Jablonski EM, Lockhart DL. Multiple-behavior comparison of group
and individual instruction of persons with mental retardation. Ment Retard 1988;
26:39–46.
236. Durand VM. Self-injurious behavior as intentional communication. Adv Learn Be-
hav Disabil 1986; 5:141–155.
237. Bird F, Dores PA, Moniz D, Robinson J. Reducing severe aggressive and self-
injurious behaviors with functional communication training. Am J Ment Retard
1989; 94:37–48.
238. Carr EG, Durand VM. See me, help me. Psychol Today 1987; 21(11):62–64.
239. Day RM, Johnson WL, Schussler NG. Determining the communicative properties
of self-injury: research, assessment, and treatment implications. Adv Learn Behav
Disabil 1986; 5:117–139.
Movement Disorders 343
240. Day RM, Rea JA, Schussler NG, Larsen SE, Johnson WL. A functionally based ap-
proach to the treatment of self-injurious behavior. Behav Modif 1988; 12:565–589.
241. Durand VM, Carr EG. Functional communication training to reduce challenging
behavior: maintenance and application in new settings. J Appl Behav Anal 1991;
24:251–264.
242. Durand VM, Carr EG. Self-injurious behavior: motivating conditions and guide-
lines for treatment. School Psychol Rev 1985; 14(2):171–176.
243. Braithwaite KL, Richdale AL. Functional communication training to replace chal-
lenging behaviors across two behavioral outcomes. Behav Interv 2000; 15:21–36.
244. Schroeder SR, Rojahn J, Mulick JA. Ecobehavioral organization of developmental
day care for the chronically self-injurious. J Pediatr Psychol 1978; 3(2):81–88.
245. Arnaud M. Étude des stéréotypies dans l’autisme infantile [a study of stereotypies
in infantile autism]. Psychiatr Enfant 1986; 29:387–420.
246. Kennedy H, Moran GS. The developmental roots of self-injury and response to
pain in a 4-year-old boy. Psychoanal Study Child 1984; 39:195–212.
247. Kerbeshian J, Burd L, Avery K. Pharmacotherapy of autism: a review and clinical
approach. J Dev Phys Disabil 2001; 13:199–228.
248. Madrid AL, State MW, King BH. Pharmacologic management of psychiatric and
behavioral symptoms in mental retardation. Child Adolesc Psychiatr Clin North
Am 2000; 9:225–243.
249. Goldstein M, Kuga S. Dopamine (DA) agonist induced compulsive biting (CB)
behavior in monkeys: animal model for Lesch-Nyhan syndrome [abstr]. Soc Neu-
rosci Abstracts 1984; 10(part 2):804.
250. Gualtieri CT, Schroeder SR. Pharmacotherapy for self-injurious behavior: prelimi-
nary tests of the D1 hypothesis. Psychopharmacol Bull 1989; 25(3):364–371.
251. Allen SM, Freeman JN, Davis WM. Evaluation of risperidone in the neonatal 6-
hydroxydopamine model of Lesch-Nyhan syndrome. Pharmacol Biochem Behav
1998; 59:327–330.
252. Horrigan JP, Barnhill LJ. Risperidone and explosive aggressive autism. J Autism
Dev Disord 1997; 27:313–323.
253. Cohen SA, Ihrig K, Lott RS, Kerrick JM. Risperidone for aggression and self-
injurious behavior in adults with mental retardation. J Autism Dev Disord 1998;
28:229–233.
254. Mendonca MF, Brasic JR, Perry R, Krigsman S, Lucien M, Petty CB. Risperidone
ameliorates large amplitude complex stereotypies and high activity level in a boy
with autistic disorder [abstr]. Mov Disord 2001; 16(suppl 1):S36.
255. DeVane CL, Nemeroff CB. An evaluation of risperidone drug interactions. J Clin
Psychopharmacol 2001; 21:408–416.
256. Drug interactions of risperidone. Child Adolesc Psychopharmacol News 2002;
7(2):12.
257. Aman MG, Arnold LE, Armstrong SC. Review of serotonergic agents and persev-
erative behavior in patients with developmental disabilities. Ment Retard Dev Dis-
abil Res Rev 1999; 5:279–289.
258. Potenza MN, Holmes JP, Kanes SJ, McDougle CJ. Olanzapine treatment of chil-
dren, adolescents, and adults with pervasive developmental disorders: an open-
label pilot study. J Clin Psychopharmacol 1999; 19:37–44.
344 Brǎsić
259. Aman MG, Madrid A. Atypical antipsychotics in persons with developmental dis-
abilities. Ment Retard Dev Disabil Res Rev 1999; 5:253–263.
260. Divalproex sodium in autism spectrum disorders. Child Adolesc Psychopharmacol
News 2002; 7(2):10–11.
261. Hollander E, Dolgoff-Kaspar R, et al. An open trial of divalproex sodium in autism
spectrum disorders. J Clin Psychiatry 2001; 62:530–534.
262. Ereshefsky L, Riesenman C, Lam YWF. Serotonin selective reuptake inhibitor
drug interactions and the cytochrome P450 system. J Clin Psychiatry 1996;
57(suppl 8):17–25.
263. Oesterheld JR, Shader RI. Cytochromes: a primer for child and adolescent psychia-
trists. J Am Acad Child Adolesc Psychiatry 1998; 37:447–450.
264. Luchins DJ, Dojka D. Lithium and propranolol in aggression and self-injurious
behavior in the mentally retarded. Psychopharmacol Bull 1989; 25(3):372–375.
265. Reents SB, Beck CA Jr. Naloxone and naltrexone: application in COPD. Chest
1988; 93:217–219.
266. Cohen MR, Cohen RM, Pickar D, Murphy DL, Bunney WE Jr. Physiological
effects of high dose naloxone administration to normal adults. Life Sci 1982; 30:
2025–2031.
267. Grevert P, Albert LH, Inturrisi CE, Goldstein A. Effects of eight-hour naloxone
infusions on human subjects. Biol Psychiatry 1983; 18:1375–1392.
268. Newhouse PA, Tariot PN, Gross M, Murphy DL, Cohen RM. Failure of high dose
naloxone to relieve tardive dyskinesia [letter]. J Clin Psychopharmacol 1987; 7:
364–365.
269. Nasrallah HA, Varney N, Coffman JA, Bayless J, Chapman S. Effects of naloxone
on cognitive deficits following electroconvulsive therapy. Psychopharmacol Bull
1985; 21(1):89–90.
270. Pollock J, Kornetsky C. Naloxone prevents and blocks the emergence of neuroleptic-
mediated oral stereotypic behaviors. Neuropsychopharmacology 1991; 4:245–249.
271. Richardson JS, Zaleski WA. Naloxone and self-mutilation. Biol Psychiatry 1983;
18:99–101.
272. Barrett RP, Feinstein C, Hole WT. Effects of naloxone and naltrexone on self-
injury: a double-blind, placebo-controlled analysis. Am J Ment Retard 1989; 93:
644–651.
273. Malcolm R, Gabel T, Morton A. More on idiosyncratic reaction to naltrexone
[letter]. Am J Psychiatry 1989; 146:124–125.
274. Rascol O, Fabre N, Blin O, Poulik J, Sabatini U, Senard J-M, Ané M, Montastruc
J-L, Rascol A. Naltrexone, an opiate antagonist, fails to modify motor symptoms
in patients with Parkinson’s disease. Mov Disord 1994; 9:437–440.
275. Sandyk R, Iacono RP. Naltrexone attenuates the antiparkinsonian effects of
picoTesla range magnetic fields. Int J Neurosci 1994; 78:111–122.
276. Dennis K. Naltrexone. Treatment Issues: The GMHC Newsletter of Experimental
AIDS Therapies 1990; 4(1):1–2.
277. Wildt L, Leyendecker G. Induction of ovulation by the chronic administration of nal-
trexone in hypothalamic amenorrhea. J Clin Endocrinol Metab 1987; 64:1334–1335.
278. Igoin-Apfelbaum L, Apfelbaum M. Naltrexone and bulimic symptoms [letter].
Lancet 1987; ii(8567):1087–1088.
Movement Disorders 345
INTRODUCTION
The primary treatment for autism is educational, an intensive behavioral approach
that has become widespread despite some lingering controversy. Biological treat-
ments are strictly adjunctive at present. These consist of primarily psychotropic
medications that address certain aspects of the core symptoms such as stimulants
for hyperactivity and inattention, clonidine for insomnia, selective serotonin-
reuptake inhibitors (SSRIs) for repetitive obsessive-compulsive-like behaviors
and antipsychotics for aggression. When these medications are effective they
often also have a beneficial effect on language and social relatedness. In sum-
mary, they have a limited impact on some autistic symptoms in some patients.
However, parents of an autistic child for the most part seek more than just a
partial fix. They are desperate to find a solution to the puzzle of their strangely
disordered child. They commonly feel that the “perfect” child who was born to
them has been made captive to an unknown process and they are willing to make
any sacrifice to rescue their beloved son or daughter. Until more is known about
the causes of autism spectrum disorders and effective treatments are available, fami-
lies and patients will remain vulnerable to speculative theories and treatments.
General pediatricians should expect to have at least one autistic patient;
other practitioners, including neurologists, psychiatrists, and developmental pedi-
atricians, will see many more (1). They will all undoubtedly hear of many alterna-
347
348 Cartwright and Power
tive treatments. It is important for the physician to keep an open mind in working
with patients and families, who need above all an ally and professional sounding
board they can trust on the long road of raising an autistic child. In addition to
trying to find a “cure” after the child has been diagnosed, and the search for
treatments to improve the core symptoms, there will be many associated problems
that may become a focus of attention. These can include problems with feeding,
sleep, toileting, hyperactivity, and aggression.
Practitioners should evaluate alternative treatments using sound scientific
principles, carefully weighing the risks and benefits with their patients and care-
givers. At the same time it is important to understand that parents who are dealing
with troubling behaviors on a daily basis may not be willing to wait until defini-
tive studies have been performed, and that their threshold for testing a substance
that offers some hope for improvement is much lower than what objectivity would
dictate. Practitioners should ensure that parents feel comfortable sharing with
them all aspects of the child’s management. When unproven treatments are at-
tempted, as in any other type of pharmacotherapy it is paramount that parents
and practitioners work together to monitor efficacy, safety, and side effects. Par-
ents should also be educated on guidelines for evaluating treatments and encour-
aged to initiate only one change at a time, to target specific symptoms, and to
identify measurable objectives—ideally, together with the practitioner. It is also
preferable to use blinded observers such as teachers and therapists to assess the
efficacy of a given intervention.
This chapter discusses some of the commonly used alternative treatments:
vitamin and nutritional therapies, antifungals and antibiotics, detoxification of
heavy metals, and auditory integrative training. Secretin and immunoglobulin
therapies are discussed elsewhere.
SECRETIN
On October 7, 1998, the national news program “Dateline NBC” aired a story
in which Bernard Rimland, Ph.D., along with a parent of an autistic child, pro-
moted the neuropeptide secretin as being highly effective in treating, and in some
cases “curing,” autistic disorders. This set off an outpouring of intense interest
from the public, with secretin being viewed as a major medical breakthrough. Dr.
Rimland based his report on a study by Horvath and Stefanatos (2) that reported
significant improvements in three children with autism following treatment with
secretin as well as anecdotal reports he was receiving from parents about the
successful use of secretin. Following the broadcasting of the story, clinicians in
the field of autism and related disorders were inundated with calls from parents
making desperate pleas for treatment with this hormone for their children. There
were postings on the Internet by physicians who were administering this non-
FDA-approved treatment with claims of success. The excitement regarding secre-
Alternative Biological Treatments 349
the children showed better eye contact, a greater level of alertness, and improved
language skills.
In a study of gastrointestinal function in 36 children with autism and perva-
sive developmental disorder not otherwise specified (PDD-NOS) ranging in age
from 21/2 to 10 years (13), subjects were referred for a gastrointestinal workup due
to the presence of abdominal pain, chronic diarrhea, gaseousness/bloating, night-
time awakening, and unexplained irritability. Of these children, 47% were on a
gluten-free and/or casein-free diet. They underwent endoscopy under general anes-
thesia, including biopsies of esophagus, stomach, and duodenum for histology,
measurement of digestive enzymes of the pancreas (before and after secretin infu-
sion) and the small intestine, as well as bacterial and fungal cultures. Porcine secre-
tin, in a dose of 2 CU/kg body weight, was given as an intravenous infusion over
1 minute.
Comparisons were made with specimens collected from nonautistic chil-
dren who had undergone a similar procedure in the same setting. Test results
showed the presence of reflux esophagitis in 25 of the 36 (69%) children. Twenty-
two of these 25 children had nighttime awakening, irritability, and abdominal
upset. Fifteen of the children had chronic inflammation of the gastric mucosa
and 24 had chronic nonspecific duodenal inflammation. In addition there was
evidence of Paneth cell hyperplasia and hypertrophy in the majority of the chil-
dren scoped. The significance of this is not known. There was reduced activity
of one or more of the disaccharidases and glucoamylase in 21 of the 36 children.
All the children with reduced levels also reported the presence of loose stools
and gaseousness. All subjects had normal levels of pancreatic enzymes. The ma-
jority had a significantly increased pancreaticobiliary fluid output in response to
the secretin infusion. Nineteen of the 21 with chronic diarrhea had a significantly
increased pancreatic output, and most responded to secretin in the weeks after
the infusion with improved stool consistency. The authors hypothesized that the
increased pancreaticobiliary response to secretin was due to the up-regulation of
secretin receptors in the ductal cells of the pancreas and bile ducts. This up-
regulation is due to the reduced availability of secretin in these children.
In summary, the study found evidence to support the connections between
certain behavioral problems and the presence of gastrointestinal dysfunction in
autism. The authors felt that secretin may have opened a window into discovering
how common gastrointestinal disturbance is in autism, thereby linking brain–gut
dysfunction.
Treatment Studies
Open-Label Studies
In a small, open-label study by Perry and Bangaru (14) conducted in a private-
practice setting in New York, six children diagnosed with autism were treated
with single secretin infusions. Only one was found to respond with a clinically
Alternative Biological Treatments 353
Despite these negative findings, 69% of all study parents reported that they
remained interested in pursuing the use of secretin as a treatment for their chil-
dren’s difficulties (63% of parents of children who received secretin and 76% of
parents of children who received placebo). It is apparent that parents are likely
to continue to advocate for the use of secretin as they have done with other
alternative treatment for autism (dietary manipulations, vitamin therapies, and
sensory integration techniques) despite the lack of empirical evidence.
This study was notable for its use of standardized outcome measures to
assess change as well as the attempt to differentiate the diagnostic categories of
autism and PDD-NOS. Shortcomings include the short-term nature of the study
in a disorder that is unlikely to show change in such a short period, the use of
only a single dose of secretin whereas multiple doses may be more effective,
and the use of synthetic secretin—past reports of positive effects were following
treatment with porcine secretin. A critique of this study by Horvath (19) cautioned
that the majority of children who show improvement on secretin do so gradually
and after repeated injections. It was noted that subjects did not have serious gas-
trointestinal symptoms and the outcome assessments that were used were insensi-
tive to change. In addition, Horvath suggested that children be given the secretin
in a fasting state because of the possible antagonism from other gastrointestinal
hormones.
Chez and colleagues (4), following on their open-label study, decided to
further test the benefits of secretin and assess whether the above changes were
due to rater bias or the real effects of the drug. They chose to study the subgroup
of children who had shown apparent progress in the first study and included
additional subjects as well. In the second double-blind, placebo-controlled, cross-
over clinical study, 25 children (22 boys, 3 girls) with an average age of 6.0
years (SD 2.4) were included. Nine of the subjects had gastrointestinal symptoms.
Group A received secretin followed by placebo 4 weeks later and group B had
placebo followed by secretin. Subjects were evaluated at baseline and, weeks 4
and 8 on the CARS and detailed neurological and symptom questionnaires com-
pleted by physician or nurse (devised by the authors’ clinic). Parents also com-
pleted diaries and noted changes in behavior. The study found no statistically
significant overall differences in CARS scores between the groups at weeks 4
and 8. No significant adverse events were reported at the time of infusion with
secretin.
Within group A there were no statistically significant group differences in
CARS scores from baseline to week 4 (secretin arm) and weeks 4 to 8 (placebo
arm), i.e., no difference between placebo and secretin. In group B, there were no
significant differences after the placebo infusion; however, after secretin, parents
perceived their children as having improved, particularly in the areas of expres-
sive language, gastrointestinal function, eye contact, and receptive language. De-
spite this finding, the authors concluded that there were no significant differences
in CARS scores of children treated with secretin as compared with those treated
356 Cartwright and Power
with saline, and therefore that no obvious clinical benefits were evident in chil-
dren with varying degrees of severity of autism after receiving a single infusion
of secretin. No specific subgroup was identified who responded preferentially to
secretin. They suggested that the perceived improvements noted by parents (in
both the open-label and the controlled studies) may have been due to “expectancy
effects”—the trial led to an expectancy by parents of a positive response, which
was then reported to investigators. In a critique of the study, Dr. Bernard Rimland
(20) argued that the study design had a bias toward a negative outcome and that
the authors downplayed the positive findings in both their studies.
Chez et al. (4) acknowledged the limitations of their study. These included
the use of the CARS as an outcome measure, as this instrument was not designed
to be sufficiently sensitive to detect subtle changes in the course of a short-term
treatment trial. Future trials would need to use standardized assessment instru-
ments as well as a variety of standardized, well-validated parent, clinician, and
teacher outcome-assessment instruments. It would also be important to include
independent evaluators who could reliably assess behavioral change. Another
methodological weakness was that the children were not separated into those
who were receiving psychotropic medication (the majority, with 80% of the sam-
ple being on some form of medication) and those who were receiving secretin
alone. The study’s findings might therefore have been confounded by drug–secre-
tin interactions.
The authors suggested that future research should attempt to identify the
possible mechanisms of action of secretin as well as describe the neurobiology
of the effect of secretin on the brain, either directly (crossing the blood–brain
barrier) or indirectly through neural or humoral mechanisms.
In summary, as a single infusion, secretin has been found to be clinically
ineffective; however, cinical trials that investigate the efficacy of multiple doses
of secretin are in progress or nearing completion.
Conclusion
Researchers remain largely in the dark about the function of secretin and related
peptides and their respective receptors in the brain. It is not clear whether findings
from animal studies will be replicated among humans. It remains to be determined
whether peripheral neuropeptide dysfunction reflects central neuropeptide
changes. In cases where improvement has been noted, it is unclear whether the
therapeutic benefit is due to an improvement in gastrointestinal symptoms or
attributable to neurobiological changes in the brain.
Evidence from open-label and double-blind controlled treatment trials of
secretin in autism does not support any therapeutic benefit in the area of social
and communication deficits. Given the heterogeneity of autism, however, the
possibility remains that there is a small subgroup of children with autism who
may benefit from secretin treatment.
Alternative Biological Treatments 357
posed metabolic defect is unknown and currently there is no way to test for it.
Martineau et al. (26) suggested that it might decrease elevated levels of homova-
nillic acid in autistic subjects; the clinical significance of this is unknown. Others
have shown some evidence of changed in evoked potentials (27). Again, this is
an interesting finding whose significance is currently not understood.
In the case of metabolic, vitamin-responsive defects, high doses of a vita-
min will improve coenzyme binding of a mutant enzyme and partially improve
function. Whether this is the explanation for the apparent benefit of pyridoxine
in some autistic children has not been clarified.
VITAMIN A
At the 1999 conference of DAN! (Defeat Autism Now!, an organization of physi-
cians and scientists convened by the Autism Research Institute), pediatrician Dr.
Mary Megson hypothesized that autism may be due to a G-alpha protein defect
that is reversible with natural vitamin A (28). G-alpha protein diseases refer to
defects in the production or activity of G proteins, which are important signaling
molecules that are intermediaries for a host of chemical messengers. For the
most part these diseases have been limited to rare endocrine disorders. Recently,
however, they have been implicated in more common disease states, including
hypertension (29). According to Dr. Megson, the patients at risk for autism would
be those with a family history of at least one parent with a pre-existing G-alpha
protein defect, including night blindness, pseudohypoparathyroidism, or thyroid
or pituitary adenomas. She proposes that in these instances there may be a defect
in the hippocampal retinoid receptors that are essential for vision, sensory percep-
tion, language processing, and attention. Natural vitamin A would repair the de-
fective transmission. A review of the basic science literature certainly suggests
that retinoids are important in many brain structures, including the hippocampus,
where it promotes cell proliferation (30). It may also play an important role in
gene regulatory events postnatally (31).
This area of research is noteworthy, but it is not at all clear how it fits into
the autism puzzle. Dr. Megson reports encouraging results anecdotally in several
autistic patients, with disappearance of the common “sideways” glance, improved
eye contact being one of the first benefits, as well as immediate improvements
in language, attention, and social interaction, in addition to normalization of ab-
normal lipid profiles. She believes these changes are due to the stimulation of
blocked acetylcholine receptors by vitamin A in the form of cod liver oil in
association with urocholine, which purportedly acts as the “switch.”
An interesting study in mice was presented at the 2000 Society for Neuro-
science meeting in New Orleans, Louisiana. Entitled “A Required Role for Vita-
min A Signaling in Hippocampal Long-Term Synaptic Plasticity,” it was done
at the Salk Institute and funded in part by the National Institutes of Health. The
Alternative Biological Treatments 359
study suggested that withholding vitamin A from adult animals impairs learning
pathways, and that these changes are reversible with vitamin A administration.
Many questions remain as to the role of vitamin A in typically and atypi-
cally developing brains. It should be remembered, though, that vitamin A toxicity
is manifested by anorexia, hepatosplenomegaly, and increased intracranial pres-
sure. A clinical trial using cod liver oil in autistic children has been approved
by the American College for Advancement of Medicine, an organization dedi-
cated to alternative and complementary medicine based in Laguna Hills, Califor-
nia, and is now underway.
VITAMIN C
Vitamin C, found in high concentrations in the brain, fulfills vital functions
throughout the body. It has been proposed that high doses of vitamin C may
decrease stereotypic behaviors through a dopaminergic mechanism. In 1993
Dolske and collaborators (32) conducted a 30-week double-blind, controlled trial
with ascorbic acid as a supplemental pharmacological treatment in 18 autistic
children in a residential school. Behaviors were rated weekly using the Ritvo-
Freeman scale as well as sensory motor scores, and were found to be significantly
decreased in association with administration of ascorbic acid. The dose used was
8 g/70 kg/day. The only major concern in the possibility of kidney stones. How-
ever, further studies are necessary to delineate vitamin C’s efficacy and its place
in the pathogenesis and treatment of autistic individuals.
DIMETHYLGLYCINE
Dimethylglycine is classified as a nutritional supplement and has been proposed
by Bernard Rimland and others as a safe substance that should be used as a first-
line treatment of autistic symptoms. A recent double-blind, controlled pilot trial
was done in eight autistic males ranging from 41/2 to 30 years of age (33). Three
scales were used: the Campbell-NIMH rating scale, an experimental rating scale,
and an individualized rating scale for each subject. Analysis of the results re-
vealed no statistically significant differences. The major methodological weak-
ness was the low dose and small sample size. As with vitamin C, no conclusions
can be drawn based on such preliminary work.
ORG 2766
It has been demonstrated that ACTH (adrenocorticotropic hormone) plays a role
in recovery from brain damage, in part by modulating the activity of endogenous
opioids and the NMDA (N-methyl-D-aspartate) receptor (34). Other basic science
research has focused on the effect of ACTH (in the form of its analog ORG 2766)
360 Cartwright and Power
on learning (35) and social interest (36). ORG 2766 had been reported to improve
social and communicative behavior in autistic subjects. A controlled trial pub-
lished in 1996 failed to replicate earlier findings (37). Outcome was assessed
using the Aberrant Behavior Checklist by parents and teachers as well as by
detailed observation of 30 of the 50 study subjects. Interestingly, significant im-
provements were noted outside the defining variables, in a subgroup of patients
who were more hyperactive with more stereotypies and abnormal speech, less
initial eye contact, and a lower performance IQ. The authors of this study wonder
whether ORG 2766 would be useful for certain subtypes of autism. Basic science
research on ORG 2766 by Horvath’s group continues (38) and may prove to be
particularly germane if an excitotoxic mechanism of neuronal death is found to
be important in the pathogenesis of autism.
his book Biological Treatments for Autism and PDD, he believes that these com-
pounds are derived from intestinal bacteria and fungi. Laboratories such as his
own Great Plains Laboratory and the Great Smokies Laboratory test for these
metabolites in the urine of autistic children as well as for excessive amounts of
yeast and anaerobic bacteria in the stool. The typical scenario described is of a
child with frequent antibiotic treatments, usually for repeated ear infections, who
then develops chronic diarrhea and regresses into an autistic state. There are
anecdotal reports of improvement on antifungal treatments such as diflucan and
Nizoral, as well as with low-sugar, low-yeast diets.
Recently, in this same vein, Sandler et al. (47) reported unexpected behav-
ioral improvement on Vancomycin in eight of 10 autistic children. The subjects
all had a prior history of treatment with broad-spectrum antibiotics followed by
chronic diarrhea and a regressive type of onset in which prior developmental
gains prior to diagnosis had been lost. Behavior was evaluated by coded video-
tapes scored by a clinical psychologist blinded to treatment status. The improve-
ments did not persist at follow-up. However, the findings add to the hypothesis
of a “gut–brain connection” in autism. This idea is widely disseminated and is
cited in many alternative treatments, including diet for food allergies, immuno-
globulin therapy, antifungals, and secretin. It may have started with the clinical
observation of concomitant steatorrhea and autism, and continues with the MMR
controversy currently unfolding as its latest manifestation. Further scientific in-
quiry is necessary to understand whether the intestinal dysfunction leads to or
contributes to autistic symptoms, is coincidental, is a consequence of a primary
insult or is unrelated.
DIETARY INTERVENTION
Several metabolic defects have been described in association with autism (48).
These include phenylketonuria, histidinemia, adenylosuccinate lyase deficiency,
dihydropyrimidine dehydrogenase deficiency, 5″-nucleotidase superactivity, and
phosphoribosylpyrophosphate synthetase deficiency. It is clear that specific di-
etary interventions in these cases can often produce dramatic improvements. Most
of these defects have been described in a very small number of cases and have
associated abnormalities such as megaloblastic anemia, seizures, and other neuro-
logical abnormalities. In the case of hyperuricosuric, or “purine,” autism, as many
as 10–30% of the autistic populations are afflicted. Symptoms include cognitive
delays, poor muscle tone, hearing loss, seizures, poor social skills, and specific
dietary sensitivities. Plasma levels are usually within normal limits, but excretion
is increased. Diagnosis is done by the uricase method on a 24-hour urine collec-
tion. No effective treatment has been found, but a low-purine diet with or without
allopurinol has been helpful. It is unlikely that the elevated uric acid itself is
causing autism, as other disorders with much higher levels are not associated
with autistic symptoms (49).
Alternative Biological Treatments 363
discouraging results that are at best equivocal (52). In the latest study, done in
March of 2000, a crossover experimental design was provided for 16 autistic
children and again no differences were detected on teacher-rated measures, IQ,
or language comprehension. There was a decrease in adaptive/social behavior
scores and expressive language quotients. Fifty-six percent of the parents were
unable to identify in retrospect when their child received the treatment. Many
occupational therapists and schools for autistic children throughout the country
use techniques of sensory integration that are felt to often be helpful as part of
an overall educational program. Their benefits may be secondary to some other
mechanism. This is yet another area to be researched.
CONCLUSION
In evaluating unproven treatments for autism, it is important to be mindful of
the “Hawthorne phenomenon.” This is not simply a placebo effect; rather, it de-
scribes the effects of parental expectation. The extra attention given uncon-
sciously to the child has been shown to influence the results of a given treatment.
This awareness does not discount the possibility of a true effect, but it does high-
light the need to adhere to rigorous scientific standards in the research of this
still unfathomable and tragic disorder.
It is logical to presume that innovations in treatments for autism should
stem from breakthroughs in understanding the pathophysiological mechanisms
of the disorder. However, this is not always the path that scientific advancement
takes. Although some resources and hopes are diverted to alternative hypotheses
and treatments, there may be some benefit in having a free-for-all brainstorming
of ideas. In the literature—both mainstream and alternative—there are many
interesting hypotheses, especially in the area of a metabolic/immunological basis
for autism that is triggered by an environmental insult, whether it be an infection,
exposure to toxins, or vaccine-associated antigens. The increase in autism re-
search is encouraging; some definitive answers can be expected as well as many
more new questions.
SELECTED WEBSITES
Dr. Rimland, ARI, vitamin B6: www.autism.com/ari
Mercury and vaccines: www.safeminds.org
Dr. Megson, vitamin A: home.att.net/pediatricaac
Heavy metal detoxification: www.heall.com/healingnews/may/heavy
Purine autism: www2.dgsys.com/purine
Dr. William Shaw: www.greatplainslaboratory.com
Elimination diet: www.panix.com/donwiss/reichelt.html
Autism Society of America: www.autism-society.org/asa home.html
Alternative Biological Treatments 365
REFERENCES
1. Powell JE, et al. Changes in the incidence of childhood autism and other autistic
spectrum disorders in preschool children from two areas of the West Midlands, UK.
Dev Med Child Neurol 2000; 42(9):624–628.
2. Horvath K, Stefanatos G, et al. Improved social and language skills after secretin
administration in patients with autistic spectrum disorders. J Assoc Acad Minor
Phys 1998; 9(1):9–15.
3. Lightdale JR, Heyman MB. Secretin: cure or snake oil for autism in the new millen-
nium? J Pediatr Gastroenterol Nutr 1999; 29(2):114–115.
4. Chez MG, Buchanan CP, et al. Secretin and autism: a two-part clinical investigation.
J Autism Dev Disord 2000; 30(2):87–94.
5. Holtmann MH, Hadac EM. Molecular basis and species specificity of high affinity
binding of vasoactive intestinal polypeptide by the rat secretin receptor. J Pharmacol
Exp Ther 1996; 279(2):555–560.
6. Li P, Lee KY, et al. Mechanism of acid-induced release of secretin in rats: presence
of a secretin releasing peptide. J Clin Invest 1990; 86:1474–1479.
7. Holst JJ. Neuronal control of pancreatic exocrine secretion. Eur J Clin Invest 1990;
20(suppl 1):33–39.
8. Momany FA, Bowers CY. Speculations on the mechanism of hormone-receptor
interactions of the secretin/glucagon family of polypeptide hormones derived from
computational structural studies. Ann NY Acad Sci 1996; 26:172–181.
9. de Bono M, Bargmann CI. Natural variation in a neuropeptide Y receptor homolog
modifies social behavior and food response in C. elegans. Cell 1998; 94(5):679–
689.
10. Banks WA, Kastin AJ, et al. Passage of pituitary adenylate cyclase activating poly-
peptide1-27 and pituitary adenylate cyclase activating polypeptide1-38 across the
blood-brain barrier. J Pharmacol Exp Ther 1993; 267(2):690–696.
11. Conter RL, Hughes MT, et al. Intracerebroventricular secretin enhances pancreatic
volume and bicarbonate response in rats. Surgery 1996; 119(2):208–213.
12. Roskoski R Jr, White L, Knowlton R, Roskoski LM. Regulation of tyrosine hydrox-
ylase activity in rat PC12 cells by neuropeptides of the secretin family. Mol Pharma-
col 1989; 36(6):925–931.
13. Horvath K, Papadimitriou JC, et al. Gastrointestinal abnormalities in children with
autistic disorder. J Pediatr 1999; 135(5):559–563.
14. Perry R, Bangaru BS. Secretin in autism. J Child Adolesc Psychopharmacol 1998;
8(4):247–248.
15. Aman MG, Armstrong SA. Regarding secretin for treating autistic disorder. J Au-
tism Dev Disord 2000; 30(1):71–72.
16. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Dis-
orders. 4th ed. Washington, DC: American Psychiatric Publishing, 1994.
17. Owley T, Steele E, Corsello C, Risi S, McKaig K, Lord C, Leventhal BL, Cook EH
Jr. A double-blind, placebo-controlled trial of secretin for the treatment of autistic
disorder. MedGenMed Oct 6, 1999; E2.
18. Sandler AD, Sutton KA, et al. Lack of benefit of a single dose of synthetic human
secretin in the treatment of autism and pervasive developmental disorder. N Engl
J Med 1999; 341(24):1801–1806.
366 Cartwright and Power
19. Horvath K. Secretin treatment for autism. N Engl J Med 2000; 342(16):1216; dis-
cussion, 1218.
20. Rimland B. Comments on: Chez MG, et al. Secretin and autism: a two-part clinical
investigation. J Autism Dev Disord 2000; 30(2):95; discussion, 97–98.
21. Rimland B. In: Shaw W, Rimland B, Semon B, Lewis L. Biological Treatments
for Autism and PDD: What’s Going On? What Can You Do About It? Overland
Park, KS: Great Plains Laboratory, 1998.
22. Rimland B, Callaway E, Dreyfus P. The effect of high doses of vitamin B6 on
autistic children: a double blind crossover study. Am J Psychiatry 1978;135:472–
475.
23. Lelord G, Muh JP, Barthelemy C, Martineau J, Garreau B, Callaway E. Effects of
pyridoxine and magnesium on autistic symptoms: initial observations. J Autism Dev
Disord 1981; 11:219–230.
24. Findling RL, Maxwell K, Scotese-Wotjila L, Huang J, Yamashita T, Wiznitzer M.
High-dose pyridoxine and magnesium administration in children with autistic disor-
der: an absence of salutary effects in a double-blind, placebo-controlled study. J
Autism Dev Disord 1997; 27:467–478.
25. Pfeiffer SI, Norton J, Nelson L, Shott S. Efficacy of vitamin B6 and magnesium
in the treatment of autism: a methodology review and summary of outcomes. J
Autism Dev Disord 1995; 25:481–493.
26. Martineau J, Cheliakine C, Lelord G. Brief report: an open middle-term study of
combined vitamin B6-magnesium in a subgroup of autistic children selected for
their sensitivity to this treatment. J Autism Dev Discord 1988; 3:372–376.
27. Lelord G, et al. Cited in Coleman M, Gillberg C, eds. The Biology of Autistic
Syndromes. New York: Cambridge University Press, 2000:279.
28. Megson MN. Is autism a G-alpha protein defect reversible with natural vitamin A?
Med Hypotheses 2000; 54(6):979–983.
29. Farfel Z, Bourne HR, Iiri T. The expanding spectrum of G protein diseases. N Eng
J Med 1999; 340(13):1012–1020.
30. Chung JJ, et al. Activation of retinoic acid receptor gamma induces proliferation
of immortalized hippocampal progenitor cells. Brain Res Mol Brain Rev 2000;
83(1–2):52–62.
31. Zetterstrom R, et al. Role of retinoids in the CNS: differential expression of retinoid
binding proteins and receptors and evidence for presence of retinoic acid. Eur J
Neurosci 1999; 11(2):407–416.
32. Dolske MC, Spollen J, McKay S, Lancashire E, Tolbert L. A preliminary trial of
ascorbic acid as supplemental therapy for autism. Prog Neuropsychopharmacol Biol
Psychiatry 1993; 17(5):765–774.
33. Bolman WM, Richmond JA. A double-blind, placebo-controlled, crossover pilot
trial of low dose dimethylglycine in patients with autistic disorder. J Autism Dev
Disord 1999; 29(3):191–194.
34. van Rijzingen IM, Gispen WH, Spruijt BM. The ACTH(4-9) analog ORG 2766
and recovery after brain damage in animal models: a review. Behav Brain Res 1996;
74(1–2):1–15.
35. Horvath KM, Meerlo P, Felszeghy K, Nyakas C, Luiten PG. Early postnatal treat-
ment with ACTH4-9 analog ORG 2766 improves adult spatial learning but does
Alternative Biological Treatments 367
not affect behavioural stress reactivity. Behav Brain Res 1999; 106(1–2):181–
188.
36. Hol T, Ruven S, Van Ree JM, Spruijt BM. Chronic administration of Org2766 and
morphine counteracts isolation-induced increase in social interest: implication of
endogenous opioid systems. Neuropeptides 1996; 30(3):283–291.
37. Buitelaar JK, Dekker ME, van Ree JM, van Engeland H. A controlled trial with
ORG 2766, an ACTH-(4–9) analog in 50 relatively able children with autism. Eur
Neuropsychopharmacol 1996; 6:13–19.
38. Horvath KM, Abraham IM, Harkany T, Meerlo P, Bohus BG, Nyakas C, Luiten
PG. Postnatal treatment with ACTH-(4–9) analog ORG 2766 attenuates N-methyl-
D-aspartate-induced excitotoxicity in rat nucleus basalis in adulthood. Eur J Phar-
macol 2000; 405(1–3):33–42.
39. Shearer TR, Larson K, Neuschwander J, Gedney B. Minerals in the hair and nutrient
intake of autistic children. J Autism Dev Disord 1982; 12:25–34.
40. Jackson MJ, Garrod PJ. Plasma zinc, copper and amino acid levels in the blood of
autistic children. J Autism Child Schizophr 1978; 8:203–208.
41. Wiedel L, Coleman M. The autistic and control population of this study. In: Cole-
man M, ed. The Autistic Syndromes. Amsterdam: North-Holland, 1976:11–20.
42. Mahaffey KR. Recent advances in recognition of low-level methylmercury poison-
ing. Curr Opin Neurol 2000; 13(6):699–707.
43. Reich F, et al. Cytoxicity of dental composite components and mercury compounds
in lung cells. Dent Mater 2001; 17(2):95–101.
44. Eppright TD, Sanfacon JA, Horwitz EA. Attention deficit hyperactivity disorder,
infantile autism and elevated blood-lead: a possible relationship. Mol Med 1996;
93(3):136–138.
45. Alberti A, et al. Sulphation deficits in “low-functioning” autistic children: a pilot
study. Biol Psychiatry 1999; 46:420–424.
46. Shaw W, Kassen E, Chaves E. Increased urinary excretion of analogs of Krebs
cycle metabolites and arabinose in two brothers with autistic features. Clin Chem
1995; 41:1094–1104.
47. Sandler RH, Finegold SM, Bolte ER, et al. Short-term benefit from oral vancomycin
treatment of regressive-onset autism. J Child Neurol 2000; 15(7):429–435.
48. Page T. Metabolic approaches to the treatment of autism spectrum disorders. J Au-
tism Dev Disord 2000; 30(5):463–469.
49. Coleman M, Gillberg C. The Biology of the Autistic Syndromes. 3rd ed. New York:
Cambridge University Press, 2000.
50. Lucarelli S, Frediani T, Zingoni AM, Ferruzzi F, Giardini O, Quintieri F, Barbato
M, D’Eufemia P, Cardi E. Food allergy and infantile autism. Panminerva Med 1995;
37(3):137–141.
51. Wolraich M. Attention deficit hyperactivity disorder. Prof Care Mother Child 1998;
8(2):35–37.
52. Dawson G, Watling R. Interventions to facilitate auditory, visual and motor integra-
tion in autism: a review of the evidence. J Autism Dev Disord 2000; 30(5):415–
421.
17
Behavioral Assessment and Treatment
INTRODUCTION
Although autism is biological in origin, behavioral treatment is currently the best-
studied intervention for this disorder. Researchers have published more than 550
peer-refereed, data-based investigations on behavioral treatment (also called ap-
plied behavior analysis), and these investigations have shown that the treatment
confers a wide range of benefits (1). For example, it helps most individuals with
autism communicate with others, engage in play and leisure activities with peers
and caregivers, carry out self-care activities such as toileting and dressing, acquire
academic and vocational skills, and manage disruptive behaviors such as tantrums
or stereotypies (2).
From a behavioral perspective, individuals with autism have biological im-
pairments that reduce their ability and motivation to learn in ways that typically
developing children and adults do. In particular, individuals with autism have
little skill or interest in playing creatively, conversing, modeling other people’s
actions, exploring their environments, attending to teachers’ instructions, or read-
ing books on topics that are unfamiliar to them. As a result, a primary goal of
behavioral treatment is to provide learning situations that enable individuals with
autism to experience success and that motivate them to continue learning.
Because many interventions developed for individuals with autism have
proved ineffective or even harmful (3), behavioral practitioners believe that it is
essential to use interventions whose benefits have been documented in controlled
studies and that are derived from scientifically sound principles on how to pro-
369
370 Smith and Magyar
mote learning. Moreover, they believe that the effects of these interventions need
to be monitored carefully for each individual with autism who receives them.
Behavioral assessment and treatment are usually implemented by parapro-
fessionals who work under the close supervision of professional behavior ana-
lysts. Such professionals should have master’s, doctoral, or postdoctoral training
in behavior analysis from university departments of psychology, education, or
human development and a year or more of internship experience providing behav-
ioral assessment and treatment for individuals with autism (4).
ASSESSMENT
Prior to a behavioral assessment, individuals with autism should complete an
interdisciplinary evaluation to establish the diagnosis, rule out medical conditions
other than autism (e.g., hearing loss), and determine the individual’s current skill
level. The focus of behavioral assessment is on directly observing an individual
in order to obtain precise information on the activities that the individual per-
forms. The main purposes are: a) identifying behaviors to address in treatment
(target behaviors), b) examining how events influence these behaviors (functional
analysis), c) determining what skills an individual needs to acquire in order to
perform new behaviors (task analysis), and d) evaluating interventions (5).
Target behaviors are either behavioral excesses (activities that the individ-
ual with autism performs too often or too intensely, such as tantrums or stereoty-
pies) or behavioral deficits (activities that the individual with autism performs
too seldom or not at all, such as communicating or playing with toys). A func-
tional analysis focuses on three kinds of events: 1) antecedents, which are events
that occur immediately before the behavior and thus may trigger it, 2) conse-
quences, which are events that occur immediately after a behavior and thus may
increase or decrease the likelihood that the behavior recurs, and 3) establishing
operations, which are events that alter the effects of antecedents and conse-
quences.
Investigators have developed many methods for conducting a functional
analysis (6). Perhaps the simplest and most common approach is to set up an
antecedent–behavior–consequence (A-B-C) chart, on which each episode of the
behavior is recorded as it occurs, along with events that were observed immedi-
ately before and after the behavior. A more rigorous approach that is useful with
especially severe or complex behavior problems is to have trained raters observe
the individual with autism and score the antecedents, behavior, consequences,
and establishing operations on a standard rating form. Another approach is to set
up observation sessions during which antecedents and consequences are system-
atically presented and removed. Data are usually graphed and interpreted by vi-
sual inspection (e.g., looking for consequences that reliably follow the behavior),
but inferential statistics such as time series analyses may also be used.
Behavioral Assessment and Treatment 371
tings, teachers usually specify the settings in which the behavior is to occur. They
may do so by outlining a sequence of goals. For example, the first goal might
be to enable the individual to respond accurately to instructions in a one-to-one
teaching situation. After this goal has been achieved, the next goal might be for
the individual to respond accurately to the same instructions when given in a
small-group teaching situation, then to do so on community outings. Data are
collected on the number of opportunities to perform the skill being taught, the
number of times the individual correctly used the skill, and the level of assistance
that the individual needed to perform the skill (e.g., did he need manual guidance
to perform the skill, or did he perform the skill in response to an instruction
without extra help?). These data are graphed, and the rate of correct responding
and need for assistance are analyzed by visual inspection of the graphs to deter-
mine whether the rate of correct, unassisted responses is increasing. If so, the
intervention is continued until the individual attains the criterion for mastery. If
not, the instructional format is altered in an effort to increase the rate of progress.
In sum, behavioral assessment and treatment are intertwined. Initial assess-
ments serve as the basis for selecting interventions. Ongoing assessments of re-
sponse to interventions may lead to revisions of the initial assessments and modi-
fications of interventions.
BEHAVIORAL TREATMENT
Principles
Behavioral treatment of children with autism involves systematically applying
principles derived from research on learning in order to increase adaptive and
functional behavioral repertoires (2). The treatment is comprehensive, targeting
skills in all domains of development. Developmental sequences and educational
models for typically developing children are used to guide the treatment plan.
Intervention occurs across settings and often involves multiple instructors. Col-
laboration with caregivers and involvement of peers are also emphasized. Inter-
vention methods are designed to create multiple learning opportunities and enable
high rates of success for individuals with autism during both skill-acquisition
and skill-application phases. The four main teaching formats are as follows:
1. Discrete trial training (DTT): DTT is a highly structured, precise in-
structional procedure characterized by a) one-to-one interaction be-
tween the teacher and the individual with autism, b) short and clear
instructions from the teacher, c) carefully planned procedures for
prompting the individual to follow instructions and for fading these
prompts, and d) immediate reinforcement for each correct response.
2. Loosely structured training: for situations when individuals with au-
tism do not need the tightly controlled learning situations provided in
Behavioral Assessment and Treatment 373
Overview
Treatment progresses in stages, each of which has specific objectives. Stages
vary greatly in length depending on the individual’s skill level and response to
treatment. The beginning stage has two interdependent goals: teaching behaviors
that promote learning (e.g., sitting in a chair, attending to the teacher, and looking
at the teaching materials) and reducing behaviors that interfere with learning (e.g.,
aggression, noncompliance, and self-injury). Teachers usually employ DTT to
teach and reinforce learning-readiness skills. During DTT, they also use reductive
procedures, described in “Maladaptive Behaviors” below, to decrease interfering
behaviors. This phase of treatment is crucial to establishing basic rules of social
374 Smith and Magyar
Curriculum
Language and Communication
Behavioral training for establishing language and communication in individuals
with autism evolved out of the conceptual work of Skinner and the experimental
work of Salzinger and others who suggested that language, like many other
classes of behavior, could be conceptualized as operant behavior (i.e., behavior
influenced by contingencies of reinforcement) (8,9). Behavioral teachers have
developed curricula to enable individuals with autism to progress from being
preverbal to having extensive communication skills (10). For preverbal individu-
als, the teacher usually begins by using DTT to teach receptive language skills
such as following directions and selecting objects requested by the teacher. For
the latter, the teacher chooses objects that the individual prefers and that are
commonly found in the individual’s environment (e.g., favorite toys or foods).
In the first step of DTT, the teacher displays one object at a time, gives a verbal
cue (e.g., “touch book”), and may prompt the response (e.g., physically guiding
the individual’s hand to the object). The teacher immediately reinforces a correct
response (e.g., providing access to the object for a brief time) and corrects an
inaccurate response. At the end of this sequence, the teacher removes the object
briefly (1–3 seconds), then brings the object back and starts the sequence again.
Instruction continues until the individual reliably selects the object without a
prompt. Next, the teacher uses the same procedures to instruct the individual to
select a second object when that object is presented alone. Once the individual
reliably selects these two objects when either is presented alone, the teacher pre-
sents two objects simultaneously so that the individual learns to discriminate
between requests for each object (e.g., selecting a book in response to “touch
Behavioral Assessment and Treatment 375
book” and selecting a doll in response to “touch doll”). After the individual has
mastered this discrimination, the selection of additional objects is taught in the
same manner as with the first two. Also, the individual is taught to generalize
the skill by responding to varied instructions (e.g., “show me the book” or “point
to the book”), across different settings (e.g., home vs. school), with different
instructors.
Receptive language training expands quickly to include such skills as fol-
lowing multistep directions and comprehending verbs, prepositions, adjectives,
and functions and classification of objects. Teachers use a combination of DTT,
loosely structured approaches, and incidental teaching methods to establish skills
and generalize these skills to the individual’s everyday environment.
Once the individual receptively identifies a small set of items (usually about
10), expressive language training begins. For those with minimal expressive
speech, teachers use DTT to teach imitation of sounds. Later, they focus on teach-
ing how to combine sounds into syllables, words, and, eventually, phrases. Indi-
viduals who have difficulty imitating oral motor movements needed to make
sounds may receive intensive training in making such movements and while this
training is ongoing may be taught to use alternative or augmentative communica-
tion, such as a picture symbol system. Whether focusing on vocal language or
augmentative communication, the next goal of expressive language instruction
is to teach the individual to request and label a variety of objects (usually ones
previously mastered in receptive language training). Subsequently, individuals
are taught to label pictures and actions that they or others perform. Teachers
apply incidental teaching methods concurrently with DTT in order to help indi-
viduals use their new language skills to communicate in everyday settings (e.g.,
placing objects in sight but out of reach so that individuals are likely to request
them). Teachers may also implement loosely structured approaches to expand
these skills. For example, they may teach individuals with autism to play prere-
corded social phrases that prompt them to converse with peers. Once such skills
have been mastered, expressive language instruction may proceed to advanced
skills such as speaking in sentences that include parts of speech (e.g., prepositions
and pronouns) and morphemes (e.g., verb tenses and plurals), asking and answer-
ing “wh-“ questions, making conversational statements on a topic, and joining
ongoing conversations.
Play and Social Interaction
Through DTT and loosely structured approaches—especially BST—individuals
with autism learn simple imitation skills that they can use in play and social
interactions, such as blowing a kiss, clapping hands, waving, building with
blocks, rocking a doll, rolling a truck, and stirring a spoon in a pot. These skills
are then combined to form longer behavior sequences and pretend-play activities.
Subsequently, with BST, individuals with autism may master other social skills
376 Smith and Magyar
such as appropriate greetings and conversational statements (11). Peers often fa-
cilitate instruction, by serving either as models for new skills or as tutors who
direct individuals with autism to use their skills in everyday settings (12). To
help individuals play and engage in other leisure activities without supervision,
teachers can instruct them to follow picture activity schedules (13).
Cognitive and Academic Skills
Teachers employ DTT methods to teach preacademic skills such as matching,
counting, number and letter identification, and drawing (14). They then imple-
ment skills-training methods to teach more advanced academic skills such as
reading, mathematics, spelling, language arts, science, and social studies. Al-
though few studies have compared different BST approaches, clinical experience
suggests that the most effective ones include carefully planned sequences of skill
development and techniques for modifying instruction to suit individuals’ learn-
ing styles (e.g., relying on pictorial rather than verbal instruction and allowing
more time to complete tasks) (15).
Motor Skills and Independent Living Skills
Although fine and gross motor skills are often a strength for individuals with
autism, some exhibit delays in this area, particularly in their performance of
planned sequences of motor activities (16). Individuals with autism who are
skilled at imitation may learn a variety of activities when the teacher simply
models the activities and reinforces correct application. Others may also require
graduated guidance (i.e., providing physical assistance, which is systematically
reduced as the individual progresses) (13). In addition to these procedures, chain-
ing is useful for teaching activities that involve a sequence of behaviors. For
example, dressing is best taught using backward chaining: the last step in the
sequence is taught first, followed by the second-to-last step, and so on, until the
whole sequence is acquired. Thus, in teaching an individual with autism to put
on her pants, a parent might place the pants on the girl and zip up the front but
leave the top button undone. Using graduated guidance, the parent would assist
her to fasten the top button and then reinforce her. Once she had mastered this
step, the parent would leave the zipper down in addition to keeping the button
undone. This process would continue until the daughter learned to independently
step into her pants, pull them over her ankles, raise them up her legs, zip, and
button. In order to help individuals perform self-help activities without supervi-
sion, teachers often instruct them to follow picture schedules (13).
Maladaptive Behaviors
Individuals with autism often demonstrate behavior problems (e.g., aggression,
noncompliance, self-injury) that interfere with learning and overall functioning.
Behavioral Assessment and Treatment 377
visual skills tend to be more advanced than verbal skills, instructions may be
presented in pictures rather than words, and tasks may have visual prompts
(grooves to indicate where to place items, pictures of each step of a task, etc.).
Because classroom noise and intrusions from peers may be distracting or aver-
sive, individuals with autism often work at their own workstations rather than
with classmates, although some activities occur in small groups. Because transi-
tions from one activity to another may be difficult, individuals with autism may
have a highly structured schedule displayed at their workstations. A number of
uncontrolled and quasiexperimental studies have suggested that Project TEACCH
is effective, and some investigators have suggested that its benefits may be com-
parable to those of behavioral treatment. However, because no controlled studies
have evaluated TEACCH, additional research is needed (21).
In the Denver Model (23), teachers aim to establish “warm, affectionate,
playful relationships” with individuals with autism. They also encourage individ-
uals with autism to exercise control in these relationships by choosing activities,
asking to finish activities, and taking turns in songs and games. Such relationships
are intended to facilitate teaching skills in the following areas: communication,
sensory activities (e.g., messy art projects or play at a water table), independent
living skills, social interaction, motor activities, and participation in classroom
routines. Uncontrolled studies have indicated that individuals with autism may
make gains on developmental tests after participating in the Denver Model, but
this finding has yet to be confirmed in controlled investigations (21).
The main supplements to behavioral treatment are sensorimotor and rela-
tional therapies. In sensorimotor therapies, individuals with autism are viewed
as having difficulty processing sensory input from the environment and/or
translating such input into effective action. The most common sensorimotor ther-
apy for individuals with autism is sensory integration therapy (SIT), which is
based on the observation that many individuals with autism respond too much
or too little to sounds or tactile stimulation and may seem unaware of their bodies
(24). To improve their responsiveness and awareness, SIT involves activities de-
signed to stimulate sensory systems such as brushing or squeezing parts of the
body, spinning on specially constructed equipment, or engaging in activities that
require balance. Although individuals with autism probably do have sensory
deficits, SIT and other sensorimotor therapies have fared poorly in research. More
fundamentally, investigators have pointed out that there is no basis in biology
or learning theory for expecting that activities such as brushing or spinning would
alleviate sensory deficits (25). However, these activities may help desensitize
individuals to sensory stimuli that are aversive or overly arousing. Also, they
may be reinforcing and thus may be useful when offered as an incentive to com-
plete other activities (26).
In relational therapies, autism is viewed as a disorder that primarily affects
individuals’ ability to enter into meaningful interactions with others. Currently,
380 Smith and Magyar
FUTURE DIRECTIONS
In addition to the need to validate alternatives and supplements to behavioral
treatment, there are a number of directions for future development of this inter-
vention. First, recent research on learning may suggest ways to enhance this inter-
vention. For example, studies on stimulus equivalence may lead to improved
methods for promoting generalization of skills (30). Second, neuropsychological
investigations have identified areas that are particularly difficult for individuals
with autism yet have received little attention in behavioral treatment, such as
theory of mind (recognizing the mental states of others) and executive functioning
(planning, sequencing activities, and adjusting to changes in routines). Hence,
research on how to address these problems in behavioral treatment is likely to
be beneficial (21).
Finally—and perhaps most importantly—because autism is biological in
origin, behavioral interventions will need to be integrated with findings from
biomedical research. Two avenues for such integration may hold particular prom-
ise in the foreseeable future. First, nonresponders to treatment may constitute a
more homogeneous sample for studying associated biomedical problems than
would a general group of individuals with autism. For example, individuals who
remain nonverbal after treatment, who cannot give up routines for more appro-
priate leisure activities, or who do not learn to understand others’ mental states
may be especially informative about the neural basis of each of these deficits.
Second, the neural functioning of individuals who largely overcome such deficits
Behavioral Assessment and Treatment 381
with treatment may differ from untreated individuals, as has been observed in
other clinical populations (31). Thus, the exploration of such differences may
yield important information about brain–behavior relationships.
REFERENCES
1. Matson JL, Benavidez, DA, Compton LS, Paclawskyj T, Baglio C. Behavioral treat-
ment of autistic persons: a review of research from 180 to the present. Res Dev
Disabil 1996; 17:433–465.
2. Newsom CB. Autistic Disorder. In: Mash EJ, Barkley RA, eds. Treatment of Child-
hood Disorders. 2nd ed. New York: Guilford, 1998:416–467.
3. Jacobson JW, Mulick JA, Schwartz AA. A history of facilitated communication:
science, pseudoscience, and antiscience. Am Psych 1995; 50:750–765.
4. Autism Special Interest Group. Guidelines for Consumers of Applied Behavior Ana-
lytic Services to Individuals with Autism. Kalamazoo, MI: Association for Behavior
Analysis, 1998.
5. Haynes SN, O’Brien WH, Hayes W. Principles and Practice of Behavioral Assess-
ment. New York: Plenum, 2000.
6. Desrochers MN, Hile MG, Williams-Moseley TL. Survey of functional assessment
procedures used with individuals who have severe mental retardation. Am J Ment
Retard 1997; 101:535–546.
7. Anderson SR, Taras M, Cannon BO. Teaching new skills to young children with
autism. In: Maurice C, ed. Behavioral Intervention for Young Children with Autism.
Austin, TX: PRO-ED, 1996:181–194.
8. Skinner BF. Verbal Behavior. New York: Appleton-Century-Crofts, 1957.
9. Salzinger K. Experimental manipulation of verbal behavior: a review. J Gen Psych
1959; 61:65–94.
10. Taylor BA, McDonough KA. Selecting teaching programs. In: Maurice C, ed. Be-
havioral Intervention for Young Children with Autism. Austin, TX: PRO-ED, 1996:
63–180.
11. Krantz PJ, McClannahan LE. Teaching children with autism to initiate to peers:
effects of script fading procedure. J Appl Behav Anal 1993; 26:121–132.
12. Kohler F, Strain P. Peer-assisted interventions: early promises, notable achieve-
ments, and future aspirations. Clin Psych Rev 1990; 10:441–452.
13. Krantz PJ, McClannahan LE. Activity Schedules for Children with Autism: Teach-
ing Independent Behavior. Bethesda, MD: Woodbine House, 1998.
14. Lovaas OI, Ackerman AB, Alexander D, Firestone P, Perkins J, Young D. Teaching
developmentally disabled children: the ME book. Austin, TX: PRO-ED, 1981.
15. Etzel BC, Leblanc JM, Schillmoeller KJ, Stella ME. Behavior modification contri-
butions to education. In: Bijou SW, Ruiz R, eds. Stimulus Control Procedures in
the Education of Young Children. Hillsdale, NJ: Lawrence Erlbaum, 1981:3–37.
16. Rogers SJ, Bennetto L, McEvoy R, Pennington BF. Imitation and pantomime in
high-functioning adolescents with autism spectrum disorders. Child Dev 1996, 67:
2060–2073.
17. Reichle J, Wacker DP. Communicative Alternatives to Challenging Behavior: Inte-
382 Smith and Magyar
Audrey F. King
Mount Sinai School of Medicine
New York, New York
INTRODUCTION
The “normalization” movement has been integral in protecting civil rights, de-
institutionalizing and humanizing services for people with developmental handi-
caps (1). Section 504 of the Rehabilitation Act of 1973 and Public Law 94-142
(Education for All Handicapped Children Act of 1975) have had a tremendous
impact on providing quality services to individuals with developmental disabili-
ties. A key problem of service delivery, however, is that the needs of children
with mental retardation and children with autism are not well differentiated (2)
by the laws put in place. Additionally, issues pertaining to children with autism,
including medication, social-skills training, placement, and programming, are not
well addressed by this legislation (3), which is important because autism and
mental retardation are different in terms of developmental pattern and outcome
(4–6). In practice, the effort toward integration has sometimes led to a lack of
diversity of services, inappropriate practices, and a lack of responsiveness to
individuals’ needs (7). More recently, Public Law 106-310 (the Children’s Health
Act of 2000) has outlined plans to increase research and education on autism.
The passing of this law has been a substantial milestone in an effort to discover
the etiology of and effective treatments for autism.
Not only do the needs of children with autism differ from those of children
with mental retardation, but not all therapies for autism are appropriate for all
383
384 King
teaching model for all children with developmental disabilities. The idea of taking
into full account the specific needs and limitations of children with autism, along
with each child’s cognitive abilities, is an important consideration for education
(15).
Supportive interventions have also been studied as a way to increase the
academic potential of children with autism in full-inclusion or mainstream set-
tings. The practice of providing occupational therapy, speech therapy, and other
important services to children with autism in special-education, full-inclusion, or
mainstream educational settings is widely used and accepted as an important
aspect of service provision (16,17).
Koegel et al. (18,19) suggest that the identification by teachers and psychol-
ogists of “pivotal” target behaviors to be addressed in the classroom may impact
academic and social performance of these children. Such behaviors affect wide
areas of functioning and therefore have a global impact on the child’s functioning
(18).
Another environmental intervention used is the decrease of task size in
order to increase on-task behavior. Sweeney and LeBlanc (20) found that decreas-
ing the number of beads to string resulted in fewer negative or off-task behaviors
and increased the number of beads strung in five children diagnosed as severely
autistic and mentally retarded. This study was based on results using a well-
described but small group of subjects, and yielded some individual variability in
results. These results highlight the importance of decreasing sample heterogeneity
in autism studies by using a sample that not only is well defined but exhibits
homogeneous symptoms, or using a sample large enough to control for individual
differences.
Koegel et al. (21) assessed the effectiveness of teaching self-management
procedures to decrease disruptive behaviors of children with developmental disa-
bilities in full-inclusion classrooms. However, because of the small sample size
used, it is unclear whether this strategy could be generalized.
In summary, current research exploring classroom techniques helpful in
educating children with autism in mainstream, full-inclusion, or special-education
classes is plagued by several shortcomings. First, it may not be enough to classify
autistic children as “high-” or “low-” functioning. In order to determine which
children with autism will benefit from different educational environments, careful
description of their individual skills, deficits, and symptoms is crucial because
of the heterogeneity of the disorder. Therefore, the question is not only “will this
intervention work for autistic children?” but also “will this work for most, or
only a subgroup of, autistic children?” For example, Tjus et al. (22) found that
language age discriminated between the responses to treatment of a group of
autistic children with varying cognitive abilities. Second, the use of small sample
sizes makes generalizing results difficult. Especially because autism is such a
heterogeneous disorder, large-scale studies should be conducted in order to iden-
386 King
tify subgroups of children with autism who benefit from different educational
environments.
without specific and individualized curricula for children with autism may not
be the appropriate first-line placement for some, as these classrooms may lack
the intensity and flexibility to modify the behaviors of young or low-functioning
children with autism (30). Thus, children who do not receive or benefit from
intensive behavioral treatment provided before the age of 5 or 6 may not do well
in special-education classrooms. These children may benefit more from schools
with programs that directly address autism and offer specific curricula and envi-
ronments for children with autism (2).
The question remains: what responses to early behavioral intervention
would indicate that mainstreaming, full-inclusion, or special education would be
most appropriate for a given child? Intellectual ability at time of admission to
an intensive treatment program has been determined to be an important predictor
for outcome (24), as have age at first treatment and autism severity (32). Full
inclusion may be a good fit for children who achieve near-grade-level language
acquisition by the age of 6 or earlier.
One concern of parents who desire mainstreaming or full inclusion for their
autistic child is the opportunity for interaction with typical peers (33). The hope
is that through observation and modeling of typical peers, the child with autism
may begin to use more typical or adaptive behaviors, and may become better
socialized. However, autistic children rarely initiate social interactions with peers,
either autistic or nonautistic (11,19,25). In studies that have examined the effect
of academic and social peer tutoring on the social behaviors of autistic children,
there is no evidence for long-term gains in social peer tutoring, although some
short-term gains have been evident (34–36).
Also at issue is to what extent typically developing peers would engage
children with autism. While some typically developing peers [39], but not all
(38), say they would interact with nontypical peers, they may not interact with
them given the opportunity (39). Therefore, it is unclear whether children with
autism would gain a social benefit from mainstreaming or full inclusion.
In summary, while few studies have empirically examined which children
would benefit from full inclusion and which would benefit from mainstreamed
classes, factors such as IQ, age at initiation of treatment, successful early inten-
sive behavioral intervention, and presence of negative behaviors may be related
to academic performance and may determine which subgroups of children with
autism may benefit from full inclusion, mainstreaming, or special education. An
additional factor may be pre-existing functional abilities, because full-inclusion
classes typically do not address these types of “real-world” skills (40).
The extent to which mainstreaming or full inclusion leads to increased so-
cial behaviors and social integration is unclear since studies have yielded mixed
results. As with academic achievement, it is possible that children with autism
with less social impairment may benefit most from social inclusion. However,
studies have not rated children in social dimensions, so there is no evidence for
388 King
this. What is clear is the need for studies using very well-defined samples of
children who are carefully described, and rated on all their symptoms.
CONCLUSION
The goal of full inclusion for children with autism is a viable and important one.
While full-inclusion, mainstreaming, and special-education classrooms may not
be adequate or appropriate first-line interventions for children with autism, it may
be possible to identify subgroups of autistic children who will benefit from these
different environments, who will be fully integrated, and who will be placed in
special education. As yet, this question remains unanswered.
390 King
REFERENCES
1. Wolfensberger W. PASS: A Method of the Quantitative Evaluation of Human Ser-
vices. Toronto: National Institute on Mental Retardation, 1973.
2. Blau G. Autism—assessment and placement under the Education for All Handi-
capped Children Act: a case history. J Clin Psychol 1985; 41(3):440–447.
3. Katsiyannis A, Reid R. Autism and section 504: rights and responsibilities. Focus
Autism Other Dev Disabil 1999; 14(2):66–72.
4. Powell S, Jordan R. A psychological perspective on identifying and meeting the
needs of exceptional pupils. School Psychol Int 1991; 12(4):315–327.
5. Kamps D, Leonard B., Vernon S, Dugan E, Delquadri J, Gershon B, Wade L, Louise
F. Teaching social skills to students with autism to increase peer interactions in an
integrated first-grade classroom. J Appl Behav Anal 1992; 25:281–288.
6. Ando H, Yosimura I, Shinichiro W. Effects of age on adaptive behavior levels and
academic skill levels in autistic and mentally retarded children. J Autism Dev Disord
1980; 10(2):173–184.
7. Mesibov G. Normalization and its relevance today. J Autism Dev Disord 1990;
20(3):379–390.
8. Romanczyk R. Behavioral analysis and assessment: the cornerstone to effectiveness.
In: Maurice C, Green G, eds. Behavioral Intervention for Young Children with Au-
tism. Austin, TX: PRO-ED, 1996.
9. Autism Society of America. 2001. http:/ /www.autism-society.org/society/
options.html.
10. Silver A. Children with autistic behavior in a self-contained unit in the public
schools. Dev Behav Pediatr 1986; 7(2):84–92.
11. Mesibov G, Shea V. Full inclusion and students with autism. J Autism Dev Disord
1996; 26(3):337–346.
12. Kamps D, Dugan E, Leonard B, Daoust P. Enhanced small group instruction using
choral responding and student interaction for children with autism and develop-
mental disabilities. Am J Ment Retard 1994; 99(1):60–73.
Educational Intervention 391
13. Carr E, Darcy M. Setting generality of peer modeling in children with autism. J
Autism Dev Disord 1990; 20(1):45–49.
14. Kamps D, Walker D, Maher J, Rotholz D. Academic and environmental effects of
small group arrangements in classrooms for students with autism and other develop-
mental disabilities. J Autism Dev Disord 1992; 22(2).
15. Verheij F, Van Doorn E. Autism and mental retardation: the planning of a therapeu-
tic environment. Int J Rehabil Res 1990; 13(2):127–136.
16. Harrower J, Dunlap G. Including children with autism in general education class-
rooms: a review of effective strategies. Behav Modif 2001; 25(5):762–784.
17. Rapin I. An 8-year-old boy with autism. JAMA 2001; 285(13):1749–1757.
18. Koegel R, Koegel K, Carter C. Pivotal teaching interactions for children with au-
tism. School Psychol Rev 1999; 28(4):576–594.
19. Koegel L, Koegel R, Frea W, Fredeen R. Identifying early intervention targets for
children with autism in inclusive school settings. Behav Modif 2001; 25(5):745–
761.
20. Sweeney H, LeBlanc J. Effects of task size on work-related and aberrant behaviors
of youths with autism and mental retardation. Res Dev Disabil 1995; 16(2):97–
115.
21. Koegel L, Harrower J, Koegel R. Support for children with developmental disabili-
ties in full inclusion classrooms through self-management. J Posit Behav Interv
1999; 1(1):26–34.
22. Tjus T, Heimann M, Nelson K. Interaction patterns between children and their
teachers when using a specific multimedia and communication strategy: observa-
tions from children with autism and mixed intellectual disabilities. Autism 2001;
5(2):175–187.
23. Kasari C, Freeman S, Bauminger N, Alkin M. Parental perspectives in inclusion:
effects of autism and down syndrome. J Autism Dev Disord 1999; 29(4):297–305.
24. Harris S, Handleman J. Age and IQ at intake as predictors of placement for young
children with autism: a four- to six-year follow-up. J Autism Dev Disord 2000;
30(2):137–142.
25. Myles B, Simpson R, Ormsbee C, Erickson C. Integration preschool children with
autism with their normally developing peers: research findings and best practices
recommendations. Focus Autistic Behav 1993; 8:1–20.
26. Howlin P. Practitioner review: psychological and educational treatments for autism.
J Child Psychol Psychiatry 1998; 39(3):307–322.
27. Green G, Brennan L, Fein D. Intensive behavioral treatment for a toddler at high
risk for autism. Behav Modif 2002; 26(1):69–102.
28. AACAP. Practice parameters for the assessment and treatment of children, adoles-
cents, and adults with autism and other pervasive developmental disorders. J Am
Acad Child Adolesc Psychiatry 1999; 38:32S–54S.
29. Green G. Early behavioral intervention for autism. In: Maurince C, Green G, eds.
Behavioral Intervention for Young Children with Autism. Austin, TX: PRO-ED,
1996.
30. Lovaas I. Behavioral treatment and normal education and intellectual functioning
in young autistic children. J Consult Clin Psychol 1987; 55(1):3–9.
31. McClannahan L, Krantz P. The Princeton child development institute. In: Harris
392 King
SL, Handleman JS, eds. Preschool Education Programs for Children with Autism.
Austin, TX: PRO-ED, 1994:107–126.
32. Eaves L, Ho H. School placement and academic achievement in children with autis-
tic spectrum disorders. J Dev Phys Disabil 1997; 9(4):277–291.
33. McGregor E, Campbell E. The attitudes of teachers in Scotland to the integration
of children with autism into mainstream schools. Autism 2001; 5(2):189–207.
34. Dugan E, Kamps D, Leonard B. Effects of cooperative learning groups during social
studies of students with autism and fourth-grade peers. J Appl Behav Anal 1995;
28(2):175–188.
35. Roeyers H. The influence of nonhandicapped peers on the social interactions of
children with a pervasive developmental disorder. J Autism Dev Disord 1996; 26(3):
303–320.
36. Wolfberg P, Schuler A. Integrated play groups: a model for promoting social and
cognitive dimensions of play in children with autism. J Intellect Disabil Res 1999;
43(4):314–324.
37. Fisher D. According to their peers: inclusion as high school students see it. Ment
Retard 1999; 37(6):458–467.
38. Swaim K., Morgan S. Children’s attitudes and behavioral intentions toward a peer
with autistic behaviors: does a brief educational intervention have an effect? J Au-
tism Dev Disord 2001; 31(2):195–205.
39. Schleien S, Mustonen T, Rynders J. Participation of children with autism and non-
disabled peers in a cooperatively structured community art program. J Autism Dev
Disord 1995; 25(4):397–413.
40. Chesley G, Calaluce P. The deception of inclusion. Ment Retard 1997; 35(6):488–
490.
41. Trillingsgaard A, Sorensen E. School integration of high-functioning children with
autism: a qualitative clinical interview study. Eur Child Adolesc Psychiatry 1994;
3(3):187–196.
42. York A, von Fruanhofer J, Turk P, Sedgwick P. Fragile X syndrome, Down’s syn-
drome and autism: awareness and knowledge amongst special educators. J Intellect
Disabil Res 1999; 43(4):314–324.
43. Helps S, Newsom-Davis I, Callias M. Autism: the teacher’s view. Autism 1999;
3(3):287–298.
44. Meyer L, Fox A, Schermer A, Ketelson D, Montan N, Maley K, Cole D. The effects
of teacher intrusion on social play interactions between children with autism and
their nonhandicapped peers. J Autism Dev Disord 1987; 17(3).
19
Consumer Advocacy and Autism
John Maltby
Sleepy Hollow, New York, U.S.A.
INTRODUCTION
In recent years there has been remarkable growth in the amount of attention paid
to autism. Only a generation ago, the syndrome was thought to be too rare, and
its expression too diverse, to warrant much scientific attention. The condition
was blamed on poor parenting, treatment was primitive to nonexistent, and diag-
nosis was delayed or often simply not made at all. In the past 35 years we have
seen enormous gains, many of them arising from the efforts of motivated individ-
uals with autism and their families.
Autism is often depicted in terms of its impact on children; indeed, it was
formerly diagnosed as “childhood schizophrenia.” For a syndrome that has few
treatments and no cure, it is obvious that most people with autism are likely to
be adults. For most individuals and their families, advocacy is a lifelong commit-
ment. Similarly, the public image of the caregiver is generally that of a highly
motivated parent, vigorously arguing for a particular kind of treatment or style
of education. However, a significant majority of caregivers are in fact profes-
sional, mostly low-paid, workers in group homes or workshops, as well as educa-
tors, psychologists, social workers, and medical professionals. These profession-
als constitute the largest body of potential advocates for people with autism.
Autism was once considered a rare disability, with a prevalence of 2–5 per
10,000. Prevalence rates are now generally understood to be much higher. Studies
presented at the 1997 Centers for Disease Control/National Alliance for Autism
Research conference on prevalence put the rate as high as 25 per 10,000. The
CDC-funded study of Brick Township, New Jersey, in 1999 concluded that local
393
394 Maltby
vacuum this absence created, various treatment methods were promoted. Over
the years these have included the use of vitamins, hyperbaric chambers, holding
therapy, facilitated communication, electric shock, aversion treatment, auditory
integration, psychopharmacological “treatment,” and the use of high doses of
secretin. In many instances these were trumpeted as “cures,” sometimes on the
basis of very little evidence. None of them has been borne out when examined
in a rigorous scientific fashion. Some of them have lured families into spending
a great deal of money and time chasing rainbows. Some of them have posed
physical danger to the subjects. This problem is exacerbated by the public media,
which seize on touching stories and promising cures and ignore the slow and
steady process of developing real science. Despite the imperfections of the movie
“Rain Man,” it was fair to say that at that time any publicity was good publicity.
Colleagues would ask a parent whether their son could count cards, rather than
whether he was “artistic.” Ten years or so on, now that public awareness is higher,
the advocacy community needs to assert common sense and be more active in
refuting pseudo science.
Given the history of autism, it is not surprising that any research that points
to families with an autistic child as being in any way “different” treads a delicate
path. However, it is becoming clearer that in many instances there is evidence
of subclinical autistic-like symptoms in both the immediate and extended fami-
lies. With a disability with a potentially strong genetic component, this should
not be surprising. The advocacy community has tended to embody some of these
traits of obsessiveness and of difficulty in communication. Professionals are natu-
rally wary of alluding to these problems, but it is past time for all involved to
recognize this obstacle and find ways to work through it.
As a syndrome, autism covers a wide range of different levels of ability.
It is now hoped that eventually we will be able to better understand the different
disorders that the syndrome encompasses. When we hear a lecture by Temple
Grandin, Ph.D., inspiration for Oliver Sacks’ An Anthropologist on Mars (1995),
we are witness to a remarkably gifted person. Dr. Grandin eloquently describes
how she deals with life as a person with autism (see Chapter 20). Her insights
are an inspiration to other people who live and function with autism, and to the
families of those people with autism who have never spoken a word, appear
severely retarded, and demonstrate self-injurious behaviors, and whose lives are
an immense daily struggle. It is also clear, though, that what we are seeing across
the spectrum is a range of people with very different levels of functioning and
very different needs. Dr. Grandin’s insights are comforting but may not relate
to the situation of other, less able autistic individuals. Despite this diversity, the
diagnosis is likely to be the same. For example, a highly verbal yet profoundly
introverted individual lacking social skills might be given the same diagnostic
label as a nonverbal, self injurious person classified as severely retarded. On the
Consumer Advocacy 397
face of it, such a lumping together is nonsensical. The clinician making the diag-
nosis is typically not the professional who will be carrying out the treatment or
looking after the individual. Unfortunately, one label is taken to fit all.
The NIH treatment conference (1999) concluded that, while there is strong
support for applied behavior analysis as an education tool and indications that
some pharmacological treatments may reduce symptoms in some instances, we
are a long way from certainty for any particular regimen. Findings presented
there demonstrated that in many pharmacological studies the placebo effect was
much higher than that found in studies of other disabilities—often higher than
40%. This helps us understand the sincerity of the more vigorous proponents of
some of the fad treatments. It is hard to deny or oppose a “treatment” that may
seem to work.
Taken together, the lack of specificity of diagnosis, the fad treatments that
are promoted like wildfire, the natural fractiousness of the community, and the
wide range of ability and need that the spectrum presents make coordinated advo-
cacy very difficult. Over the years these factors have created periodic schisms
in the advocacy community. The worst of these arose in the mid-1980s. There
was violent disagreement over the use of “aversives,” ranging from the most
benign use of rewards to extremes such as the use of cattle prods, aggressive
bullying language, and severe physical restraints. In the absence of real treatment,
desperate measures were being attempted, and in some cases, as with almost all
autism treatments, were showing apparently positive results. This was also the
height of the “deinstitutionalization” movement, and some families were justifi-
ably afraid that in moving their sons or daughters “into the community” services
would be lost. Some of these fears have been shown to be unwarranted, especially
when there was a strong advocate for the individual. However, for many people
with autism, especially those with no family or advocate, there was a loss of
support, and they simply went from one expensive institutional setting to a
smaller, cheaper one–or worse.
These bitter ideological arguments of the mid-1980s wracked the autism
community. Despite the vocal extremists, most people found themselves some-
where in the middle, opposed to painful treatments but not opposed to rewards,
opposed to the worst of the institutional settings but not ready to throw their son
or daughter into an unfriendly society. One consequence of the arguments was
the creation of new advocacy groups, generally focused on a single issue. With
the progress being made in autism research these schisms are beginning to heal,
as the goals become clearer and, even more important, attainable.
In its history the ASA has been called on many times to endorse a treatment
that may have seemed to work for some people, or to come out against a particular
kind of aversive. It was advisedly reluctant to endorse one treatment over another,
or to endorse any at all, given the lack of hard science. It created the Options
398 Maltby
Policy, which states that individuals and families should be able to pursue treat-
ments that make sense to them, without an ASA endorsement. Naturally this
decision, in turn, disaffected many people.
RESEARCH ADVOCACY
The first major private funding initiative in basic autism research came with the
establishment of the Seaver Center in New York City in 1994. The Center is
primarily funded by the Seaver Foundation, which has strong links with local
advocacy groups in the New York area and has also reached out to the national
advocacy groups. Since its inception in 1994, the Foundation has invested over
$10 million in applied and basic autism research. In addition, the Center has
received funding from other voluntary groups and from the NIH.
After much lobbying by ASA and by the professional community, the NIH
held its first symposium on autism—“The State of the Science”—in 1995. For
the first time, this brought together leading researchers to summarize what is
known about causes and treatments for autism. The symposium highlighted the
need for increased research funding, for an organized effort to recruit consumer
research participants, This event and the growing body of substantive research
led to the creation of three national consumer-led research-funding organizations.
The National Alliance for Autism Research (NAAR), founded in 1995 in
Princeton, New Jersey, began funding researchers in 1997. NAAR’s focus has
been on funding basic science investigators, particularly those seeking to gather
initial data prior to submission for more substantial long-term grants to the NIH
and other funding sources. It assembled a stellar Scientific Advisory Board
(SAB), and its annual RFP process, which in 1997 attracted 27 proposals and
funded five grants, received 90 proposals and expected to fund 21 awards and
fellowships for a total of $1.5 million in 2000. In its first four years, NAAR has
funded 50 studies for a total of $3 million, in addition to its outreach work and its
Autism Tissue Program (ATP) joint venture with the ASA Foundation (ASAF).
In 1994, the ASA had provided significant funding to the Stanford autism
project when its program came under threat following the death of its leader, Dr.
Roland Ciaranello. This followed initial discussions at the Las Vegas Conference
of that year aimed at creating a biomedically based foundation. The Society de-
bated long and hard over whether to focus exclusively on biomedical research
or to add applied research to its mission. The ASAF was finally formed in 1996
and was tied very closely to the ASA governance process and to ASA’s member-
ship . In addition to funding basic research, the ASAF also funds applied research
to lead to better understanding and improved conditions in treatment, schooling,
work and housing, consumer rights, etc. In 2001, in addition to ASA’s funding
commitment to advocacy, services, and education, the ASAF funded five such
Consumer Advocacy 399
projects. These are projects of major interest and utility to its constituency that
focus on the lifelong need for support of the individual with autism. In a signifi-
cant change from the fractious history of autism advocacy the ASAF has created
close ties to other advocacy and research organizations. Rather than try to dupli-
cate the NAAR SAB, for example, ASAF directs the bulk of its basic science
funding through the NAAR SAB process, and has to date funded eight grants
this way. In addition, ASAF and NAAR created the ATP, which serves to recruit
potential brain-tissue donors from the ASA membership and to enable exchange
of existing tissue between researchers. This has been a very successful program,
doubling the limited amount of tissue available.
ASAF also created the Autism Research Registry (ARR), which enlists
ASA members and the broader community as potential research participants
through a clearinghouse designed to protect research subjects and researchers,
increase the number of research subjects, until now a very small pool, and reduce
the cost and time involved in recruitment. This registry currently includes 1500
people across the United States.
Cure Autism Now (CAN) was founded in 1995 in Los Angeles. CAN funds
basic and applied research, and has made commitments of $5.7 million in research
awards over the past five years. CAN created the Autism Genetic Research Ex-
change (AGRE), a collaborative gene bank for genetic research in autism, which
by the end of 2001 had created a bank of immortalized cell line DNA and frozen
serum samples from a total of 400 multiplex families.
None of this would be possible without the continuing sponsorship and
facilitation of the NIH. As recently as 10 years ago, some divisions of the NIH
felt that autism research would be futile because the syndrome was too diffuse
and too rare. Worse still, the autism advocacy community seemed to be bitterly
divided along numerous fault lines. They “couldn’t get their act together,” and
this turned the research community away from them. Thanks to the vigorous
activity of autism professionals in the NIH, these obstacles were gradually over-
come. In 1995, the year of the first State of the Science Conference, NIH funding
was $10 million. In 1999 NIH funded $26 million for autism research, and the
total for year 2000 was close to $47 million. Still modest in relation to the scope
of the disorder, but a vastly different profile and approach. Since the 1995 confer-
ence NIH has created an interdepartmental forum to exchange information from
all the different facets of NIH research, a forum that involves consumers.
The CDC and NAAR cosponsored a prevalence conference in 1997. Esti-
mates spanned the range from the historical estimate of 5 per 10,000 to as high as
30–40 per 10,000 in Japanese and Scandinavian studies. Recently after vigorous
consumer advocacy from families in Brick Township, New Jersey, a CDC study
concluded that the local prevalence was closer to 50 per 10,000. Whether in-
creased prevalence is based on improved or loosened diagnosis, driven by the
400 Maltby
fact that a diagnosis is the key to services, or whether there has been an actual
increase in incidence remains an open question. What is clear is that, as a result
of consumer activity, autism is recognized as a major public-health problem.
Applied behavior analysis (ABA) is sometimes identified with particular
practitioners but is a technique used to varying degrees in different educational
milieux. Although the number of large-scale controlled studies is limited, small-
scale studies demonstrate that individuals have had positive outcomes over an
extended period of time. Normally identified publicly as being most effective
with young children, it is also applicable to older individuals. The families of
young children with autism have been very vigorous in the past decade in pressur-
ing school districts to provide ABA-type learning environments for their children.
In some instances these have been incorporated into the public schools; in others,
families have been the driving force behind creating ABA-based classes and sepa-
rate schools. Obtaining these programs has often involved litigation, expensive
for both the families and the school district, in addition to the emotional toll that
the constant battle takes. To support this effort, many organizations have been
created at the local level, some with one purpose only—to create an ABA-based
program—and others that provide wider advocacy. The best known of these orga-
nizations is FEAT (Families for Early Autism Treatment). Founded in 1993 by
parents and professionals in Sacramento, California, FEAT sponsors a website
and workshops in conjunction with the UCLA Clinic for the Behavioral Treat-
ment of Children, promotes awareness of education issues, and provides support
on an ongoing basis to individuals with autism and their families.
they encounter and, while tact is always helpful, can be more in charge of their
own treatment and support.
The second step is to join a local voluntary group—an ARC or ASA chap-
ter, for example. Obviously some function better than others, but it is the best
way to learn how to deal with the local bureaucracy and what programs and
benefits the individual is entitled to, and to share experiences with others with
similar concerns. For most people with autism, day-to-day difficulties are very
local, and very time-consuming, for example, ensuring that receive their food
stamps, SSI, SSDI, Medicaid funding—processes that take many hours of work.
When they go in to an Individualized Education Program (IEP) meeting, they
need to be properly prepared, and they may need help getting transportation and
access to work and leisure. An advocate who knows the science, supported by
the local experience of other advocates, is likely to encounter far better treatment
by the bureaucracy than a passive consumer, with consequently more beneficial
outcomes. While it may sometimes seem otherwise, most social-services bureau-
crats did not go into the field because they were indifferent to people. They fre-
quently enjoy meeting someone who is highly motivated and who has done his
or her homework, and they are more responsive to such advocates.
Third, as the individual with autism grows older, advocates should find a
way to replace themselves. They should put in place the Guardianship structures
that might be needed—Special Needs Trusts, if possible. Information on these
legal structures is available from the ASA and also from most ARCs and the
UJA. These organizations can help to design a long-term financial and service
plan that will survive the advocate. One effective way to create such a support
system is to form a Circle of Support, comprising family and friends and commu-
nity members to share the workload and ensure ongoing support.
All the services an individual will receive in his or her lifetime are provided
by systems. In recent years we have seen a welcome reduction in the size of
some of these service-delivery mechanisms, but virtually by definition they are
likely to be highly structured and not well geared to the needs of the individual.
Systems tend to standardize services and be reimbursed in a uniform fashion,
resulting in generic services delivery. The advocate must therefore be the oil in
the gears. The more vigorous and informed the advocate, the more responsive
the system will perforce be.
LOCAL ADVOCACY
Throughout their lifetime, individuals with autism and advocates for individuals
with autism will find support from local associations. Most parents, regardless
of prognosis, will wonder why this has happened to their child and why it has
happened to them. They will ask this question of themselves for the rest of their
402 Maltby
lives, and it doesn’t have an answer. One of the many assumptions we make of
autism—that it cuts across all social classes and ethnicities—actually seems to
be very true. Most consumer advocates lead very different lives from one another
but have one profound difficulty in common, and it is only with other people
who share the same difficulty that they can ever truly feel comfortable dealing
with it. This common emotional bond should not be the raison d’être of an advo-
cacy group, but it is a necessary part of the foundation.
An effective local group does not have to be one that encompasses the
entire range of needs and abilities—it doesn’t have to be solely about autism,
although the stronger ASA chapters will afford the greatest level of support. The
spectrum is wide-ranging. Some individuals function almost independently and
are verbal and literate, but have excruciating social difficulties at school and work
and are constantly aware of having to “fit in.” Elsewhere on the spectrum, the
focus may be on safety, treatment of self-injurious behaviors, protection from
abuse by group-home housemates, or coping when communication is almost ab-
sent. At the local level, it is hard to form a group that will include people with
experience in different parts of the spectrum, and especially hard to reflect such
diversity if a group was originally formed by a small cadre of families with
particular shared needs. However, the key to effectiveness is to focus on those
things that everyone will have in common, e.g., dealing as a group with local
education authorities, finding a pediatrician or a dentist who works well with
developmentally disabled patients, or determining which local government office
is least obstructive in obtaining benefits. At the same time, bringing in speakers
on a range of topics is one way to address the diversity of need and to better
understand each other’s priorities.
At a very practical level, local organizations around the country have cre-
ated “ombudsman” programs. An ombudsman is a member of the community—
possibly a relative of an individual with a disability—who, after some training,
undertakes to visit a group home or a work or school program. Their job is not
to ensure compliance with regulations; that is a whole industry by itself. Rather,
their function is to visit the programs, act as a friend and support to the individuals
in them, and be alert to the potential for neglect, sterility of content, and quality-
of-life issues that are not covered by the regulatory framework. Twenty five years
after Willowbrook, gross neglect and severe abuse still occur in the group-home
industry; with an active ombudsman program there is an extra level of safeguard.
In many cases, the ombudsman may after a few years become the longest-serving
“staff” member of a program, given the frequency of staff turnover. The people
participating in the program, however, have control over the extent to which they
join with the ombudsman; their friendship is not forced. The programs have been
successful in raising the quality of life for disabled individuals, at a very modest
cost, and without additional regulatory hierarchy. The program is also extremely
rewarding for the ombudsman.
Consumer Advocacy 403
SUSTAINING ADVOCACY
People in the United States have formed countless community organizations over
the years, and it is not very hard to see why the successful ones work. In autism
it is important to have a “big tent” outlook. The spectrum is wide, the needs are
various, and, while not all of them can be met, they should all be considered.
Similarly, with treatment and living options, one size does not fit all. Successful
organizations stay mission-focused and have a structure that provides for succes-
sion and consensus. Fledgling organizations often benefit from a charismatic en-
trepreneur, but one doesn’t need to look far in the business news to see what
befalls organizations that do not learn how to move past that developmental stage.
Another aspect that some groups relegate to too minor a role is fund raising and
development. Organizations should be on a sound financial footing, and a board
should always consider this, along with changing the world, one of their primary
responsibilities. Often the best way to raise money at the local level is through
grants and government funding, a developmental process. The most successful
organizations work with their community, the local politicians, local and state
404 Maltby
NATIONAL ADVOCACY
The growth of public awareness coupled with real scientific advances in the 1990s
has created steady growth in the size and effectiveness of the national autism
advocacy movement. At the same time, it can appear on the surface to be ever
more diffuse as new organizations spring up almost every month. Perhaps the
real story is that national autism advocacy is finally growing up. We are moving
from a nationwide support group, united against an uncaring public and a mis-
guided medical establishment, to a national disability advocacy movement, lob-
bying for legislation to get serious about research funding, raising money for
services and research, and working with the medical and education community
to increase public awareness at the national level.
Advocacy at the national level falls into four main categories.
to $14 million and by 1999 to $26 million. The budget for the Centers for Disease
Control was increased from zero to $10 million in the same period. Legislation
already approved by the Senate and now in the House would increase annual
funding to $60 million. The three major research foundations—NAAR, ASAF,
and CAN—have developed a more unified approach to lobbying and to working
with the NIH and CDC. As a result, the legislature and the administration get a
common message and focus. There is still a great deal of work to do. The current
level of funding is a great improvement and has drawn many new scientists to
the field, but the research funding does not yet approach the level of the problem.
We continue to advocate for better funding for basic and applied research, fund-
ing for centers of excellence. In 1999 the ASAF was instrumental in lobbying
the National Bioethics commission to modify its position on human-subject re-
search on individuals with developmental disabilities. The original position
would have so constrained research as to be a denial of the right to direct research
to find treatments and cures for autism.
Educational Rights
The battle to provide for a free and appropriate public education for all children
was not ended by PL 94-142 in 1979. Every year there are attempts to dilute the
act, most recently on the grounds of the need to prevent unruly children from
disrupting classes (this against a background fear of disturbed children bringing
guns to school). Primarily funded by teachers’ unions, this unfortunately has the
effect of pitting the parents of children with disabilities against the parents of
normal children. Advocates need to do a better job of informing the public of
the cost benefit of improved education access, as well as its clinical benefit. In
some ways this is a harder struggle than finding money for research. Most people,
albeit naively, equate funding research with finding a cure. Maintaining the right
to remain integrated in the educational system is asking for a long-term social
change, one that has moved far in the generation since PL 94-142 passed, but
that will need several more generations to reach fulfillment. This advocacy relies
heavily on an organized nationwide grass-roots movement that can be called on
to write and phone its congressional representatives on short notice when faced
with a late session “end run.” We are improving with practice.
Poverty
In addition to the medical diagnosis, most individuals with autism have an addi-
tional handicap of poverty. People with developmental disabilities are the most
impoverished of all Americans. They might receive services that cost a great deal
of money, but remain personally impoverished and dependent. Allied with other
disability groups at the national level, autism organizations, principally the ASA,
have lobbied for the expansion and maintenance of SSI/SSDI, food stamps, heat-
ing and air conditioning grants, housing improvement grants, and transportation
subsidies. As we move further away from the long-term institutional model, these
programs will become a more important part of every individual’s support.
SERVICE STANDARDS
Nonprofit provider agencies still retain vestiges of the charitable institutions many
of them sprang from. However, most are small businesses to midsize corpora-
tions, distinguished only by tax treatment from their other private-sector brethren.
We still expect them to be caring and altruistic in purpose, and many are. How-
ever, the lingering aura of charity, and the low pay in the profession, allow for
deficiencies and variability in standards of care. The current euphemism for a
person with disabilities in this context is “consumer.” Most of our “consumers”
do not have much choice as to service provider, and are utterly in their power.
A long-term objective of the autism advocacy movement is to create national
standards for training and qualifications for people who work with individuals
with autism syndrome disorder (ASD). There are possibly as many as 500,000
people whose work is primarily involved with ASD individuals and who have
an interest in improving their professional qualifications, not only in the teaching
profession but, especially, in the human-services professions These professionals
should be the allies of the autism advocacy community.
dence of individuals with high-functioning ASD, and has spurred the creation of
self-advocacy and support groups around the world. The ASA (assuring anonym-
ity) provides details on how to participate in several such groups.
In 1999 the Shirley Foundation and the NSAC in the United Kingdom
created and hosted the first Virtual Conference on Autism. There were over
23,000 delegates from 112 countries. While the ASA has for some time been in
touch with organizations in other countries, this was the first truly international
convocation. Because the knowledge available in this kind of forum can reach
people all over the world, the impact on the millions of people with autism will
be extremely powerful.
significant need for more in-service training, for greater understanding of what
the different branches of the system do.
ADVOCACY NOW
It is finally dawning on the medical and political establishment that autism is a
worldwide health problem. The autism advocacy community is beginning to
come together far more effectively as research offers greater hope for the future,
and as all the work done for so many years on educating the public and advocating
for better services and recognition bears fruit. We are at the beginning of a new
phase in advocacy at the national level, one that presents huge opportunity. At
this writing there are two research funding bills in the House of Representatives
and one that has just been passed in the Senate. Since the first NIH-sponsored
State of the Science conference in 1995, there has been a series of others, includ-
ing Diagnosis, Treatment, and the Development of an Animal Model. Dozens of
studies in basic research have been funded by NAAR, ASAF, and CAN. ASAF
has funded a series of applied research studies. The ATP is a national resource
for obtaining and distributing brain tissue for research. The AGRE has facilitated
the use of genetic material for researchers across the country. The ARR is a
national clearinghouse for researchers and research participants.
While the future for national-based organizations looks promising, and the
level of community support has grown, at the end of the day advocacy is most
effective when it is most focused. Each advocate should reflect on where he or
she can do the most good. Advocacy movements can be waylaid by organiza-
tional politics, multiple agendas, and all the ills that corporations are prone to.
Sometimes it is best to think about actually achieving the next step, no matter
how small—make one good thing happen.
FURTHER INFORMATION
The Autism Society of America
800-3-AUTISM
www.autism-society.org
National Alliance for Autism Research, 888-777-NAAR
www.Naar.org
Cure Autism Now, 323-549-0500
www.Canfoundation.org
The Organization for Autism Research (OAR)
703-351-5031
OAR@autism.org
20
Autism: A Personal Perspective
Temple Grandin
Colorado State University
Fort Collins, Colorado, U.S.A.
409
410 Grandin
tracted to strong visual stimuli, yet high-pitched auditory stimuli hurt my ears.
I don’t think this is puzzling at all. Margaret Creedon (1992, unpublished) found
that I have a very slight jerkiness in my visual tracking although my vision is
otherwise normal. This slight defect may explain my attraction to things such as
flags, kites, and automatic doors.
My auditory processing problems are far greater than my visual processing
problems. When I was a small child I could understand what adults said when
they spoke directly to me, but when adults talked quickly to one another I could
not decipher what sounded to me like gibberish. As an adult, I still have difficulty
hearing hard consonant sounds. Often, I mix up similar sounding names such as
Crandell and Brandell. In such instances, I figure out words by the context cues.
For example, if one is talking about “fog” at the airport, I know from the context
that they are not saying “bog.” I was shocked at how badly I performed on a
series of central auditory processing tests although my pure tone hearing test was
normal. Nevertheless, a pure tone hearing test does not detect problems with
hearing hard consonants or other central auditory processing problems.
had to push just hard enough to get me out of my world. Children who have
more severe sensory problems than I did may not respond well to intensive pro-
grams such as that of Lovaas (9). I had touch and sound sensitivity problems,
but my senses provided a more or less accurate picture of the world. Children with
more severe sensory problems than mine may be driven into sensory overload if
a teacher grabs their chin.
Williams (10), a woman living with autism whose sensory processing prob-
lems are different from mine, explains that she is what can be referred to as
“mono channel.” She cannot attend to simultaneous visual and auditory input.
In other words, she has to either listen to or look at something but she cannot
do both. In her new book she describes visual shutdown, wherein her visual
system ceases to function when overloaded.
It is clear that sensory function in autism can vary a great deal. Recently,
I talked to a Lovaas therapist who was currently a witness to such variation. She
informed me that she was having great success with two children, but that a third
child was making almost no progress. The third child was moving into sensory
shutdown. Therefore, from a clinical standpoint, there appears to be two basic
types of 2- to 3-year-old children with autism.
The first type of autistic child has relatively mild sensory processing prob-
lems and will probably respond well to a Lovaas-style program. The therapist
can gently pull such children out of their world and reach them through their,
“front door” (11). I also believe strongly that good programs should include sen-
sory treatments. Sensory treatments such as Agren (11) sensory integration as
well as auditory and visual training can help a child’s nervous system become
more receptive to a structured educational program. Vigorous exercise will also
help reduce repetitive stereotypic and maladaptive behavior and Rimland and
Edelson (12) also found that auditory integration training was helpful in reducing
sound sensitivity and improving behavior (13–15). Both my work (16) and Gil-
lingham’s (17) stress the need for awareness of sensory problems in autism.
The second type of child has very severe sensory processing problems. In
severe cases, vision and audition may jumble together.
Williams (6) and Joliffe et al. (18) describe problems autism may cause
with an individual’s kinesthetic sense, the ability to discern body boundaries. For
children of this type, the world is a profoundly confusing place of sensory over-
load, something like living inside a rock and roll speaker. To such a child, fluo-
rescent lights would be like flashing strobe lights. Intrusive methods such as the
Lovaas method often work poorly with these children. If parents report that their
autistic child has tantrums and cannot tolerate large supermarkets, shopping
malls, or similar environments, this is often an indication of very severe sensory
processing problems. Since my sensory processing problems were fairly mild I
could enjoy certain demanding sensory environments such as the supermarket.
412 Grandin
One must be consistently aware that sensory problems are highly variable.
Some children may have visual problems and others may not. One child will be
able to tolerate fluorescent lights and another will not. Therapists working with
autistic children must be able to understand that fluorescent lights or fear of a
phone ringing, problems that arise from sensory processing difficulties, may make
it hard for a child to learn. Therapists and parents should make understanding
such sensory problems and fears a priority and modify the child’s environment
accordingly.
With the first type of child, the therapist should be gently intrusive in order
to enter his or her “front door.” However, with the second type of child, the
therapist must carefully tiptoe through the child’s “back door.” Children with
severe sensory problems will likely respond better if the therapist talks very qui-
etly to them in a darkened room, which cuts down on total sensory stimulation
and therefore reduces possible overstimulation. This allows the child to concen-
trate on one sensory channel at a time.
VISUAL THINKING
Many people with autism are visual thinkers, including myself. All of my
thoughts are like videotapes playing in my imagination. Even my abstract thought
is in pictures. For example, I have no generalized concept of beauty in my mind.
Instead, there are only specific examples of beautiful things that I have come
into contact with, such as a slide of a rainbow in Hawaii or the Rocky Mountains
near my house.
It was through questioning many people that I discovered the true extent
of my thinking differences. I can best explain the differences by specific exam-
ples. When most people are asked to access their memory of church steeples,
they usually report seeing a rather vague picture of a generic steeple (19). If they
are asked to think about specific church steeples they can do it, but their mind
tends to drift back to the generic picture.
I have discovered that most people think by mentally surveying a concept
and then moving to specific examples. My thinking works in the opposite manner.
I form concepts by looking at many different specific examples and comparing
them.
As a child, I figured out that cats and dogs were different. Even though
dachshunds and golden retrievers were very different, I still knew that both of
these animals were dogs. Although both breeds were dissimilar, they had com-
mon characteristics that cats did not have. By comparing the specific cat and dog
pictures in my imagination I was able to differentiate between dogs and cats at
a young age. I figured out that all cats have silky fur and retractable claws and
all dogs have the same type of nose. It was easier for me to form concepts when I
had many specific examples. The bottom line is that my thoughts go from specific
A Personal Perspective 413
examples to general principles and are devoid of language. They are just like a
video playing in my head.
Visual thinking is also associative and nonlinear. When I search my mem-
ory for information, it is something like surfing the Internet. My associative think-
ing goes from one video “web page” to the next. For example, if I think about
vacuum cleaners, the first picture that appears in my imagination is a giant vac-
uum cleaner used in my elementary school that terrified me. The next associative
picture is the electric broom in my closet, and the third picture is a video of me
walking through my house. The fourth associative picture is one of my first big
livestock design jobs. So how did I get from vacuum cleaners to livestock equip-
ment? After the image of the electric broom in my closet, my image changed to
me walking through my house and seeing a picture of a livestock project I have
on my wall. Associative thinking of this kind helps explain how an autistic child
can make connections that seem nonsensical.
MY MEDICATION VOYAGE
It was when I entered puberty that my anxiety attacks started (20). It felt like
being in a constant state of stage fright for absolutely no reason. My ner-
vous system was consistently activated and ready to fight in the absence of any
threat.
During this time, to help relieve my anxiety attacks, I built a device that
I could use to apply pressure to large areas of my body (21,22). Relief through
body pressure seems to be common in autism because many children and adults
with autism seek pressure by getting under mattresses and other objects. Agren
(11), McClure and Holz (21), and Zisserman (23) all report that pressure applied
to large areas of the body will reduce self-stimulatory behaviors and calm the
nervous system.
During my late teens and early 20s I was able to control my anxiety attacks
and calm my nerves by using my pressure machine and engaging in heavy physi-
cal exercise and work. When I reached my early 30s, however, the anxiety attacks
began to destroy me both physically and mentally. I would wake up abruptly at
3:00 a.m. with my heart pounding.
Approximately 20 years ago, when I was in my ealy 30s, fluoxetine was
not on the market. At this time I read a journal article by Sheean et al. (24) about
imipramine for anxiety and I asked my doctor to prescribe 50 mg/day. It worked
like magic and my anxiety attacks stopped. The 50 mg/day dose that controlled
my anxiety was much lower than the doses normally used to treat depression.
After taking imipramine for a few months I had a relapse. I resisted the
urge to increase the dose and, fortunately, after a few weeks my anxiety again
subsided. To help reduce my side effects I switched to desipramine and used it
for 10 years. A dosage of 50 mg/day successfully controlled my anxiety.
414 Grandin
Then I had a hysterectomy in which one ovary was removed along with
my uterus. Shortly after the hysterectomy I went on estrogen supplements to
relieve severe menopause symptoms. A dose of 0.66 mg/day of conjugated estro-
gen made me feel really good. I took the estrogen every day with no breaks. This
worked for about 2 years. After 2 years at 0.66 mg/day I started getting anxiety
attacks again. When I stopped taking the estrogen my anxiety diminished. I rea-
soned that I could control both, menopause symptoms and my anxiety by manipu-
lating the dose of estrogen. Depending on how I felt I switched back and forth
between 0.3 mg and 0.66 mg/day. This dosage strategy worked for about a year
and then I started having constant anxiety attacks and problems sleeping through
the night, so I reduced the dosage to 0.3 mg/day every day.
In the fall of 1996, during the book tour for Thinking in Pictures, my new
book, I had a severe anxiety attack. I really wanted to complete the book tour,
and I thought that my anxiety would probably lessen if I stopped taking estrogen.
I went off estrogen for 6 weeks, and my anxiety subsided during the first 4 weeks.
However, during the last 2 weeks without estrogen supplements, menopause
symptoms started and my anxiety increased. When I went back on the supple-
ments, I felt much better. Around Christmas of 1996 I started a new estrogen
strategy to mimic natural estrogen cycles. I took 0.3 mg every day from 10 days
to 2 weeks and then went off the medication from 10 days to 2 weeks. In other
words, I took daily doses of estrogen until I started to feel anxious and then
stopped until I felt aching joints or other menopause symptoms return.
One may wonder why I did not just take more trycyclics. The reasons are
fairly straightforward. I am already slightly overweight and I did not want to get
bigger—weight gain being a common side effect of tricyclics. Also, I have been
on tricyclics for over 20 years, and I was afraid that would have a bad reaction
if I tried to get off the tricyclic desipramine to switch to fluoxetine or one of the
other selective serotonin-reuptake inhibitors (SSRIs). My strategy of going on
and off estrogen is working, for the time being. At one point I tried buspirone,
and I had an awful experience. It felt like I had overdosed on allergy pills. My
mind was fuzzy and I could not think. It felt like I had taken too much cold
medicine!
for about 6 months and then the sleeping problems returned. I then thought to
myself “The female body is not designed for a steady-state dose of hormones”.
To more closely mimic the natural cycle, I started taking the combination of
estrogen and progesterone for 3 weeks and then going off all the hormones for
1 week. This 3-weeks-on-and-1-week-off cycle of both conjugated estrogen and
medroxyprogesterone worked well for about a year.
For the last 6 months of 1999 and up until the writing of this update I have
engaged in a program of vigorous exercise. I was very out of shape and my only
exercise had been walking through airports and walking around in meat plants
and feedlots. I now exercise for 15 to 20 minutes each day by jogging in place.
It took me 4 months to work up to jogging in place for 15 minutes without getting
winded. After I started to exercise it took about 2 weeks for my sleep to improve.
I had to find an exercise I could do easily in a hotel room or in front of the TV
at home. Going to a gym is too much trouble. At the present time, the combination
of exercise and the combination hormones of 3 weeks on and 1 week off is
working. My dose of desipramine is still 50 mg/day.
As I have gotten older, sleeping has become progressively more difficult.
I have more and more problems getting to sleep, and I would wake up in the
middle of the night and feel so “wired” that I could not get back to sleep. The
problem of lack of sleep was one of the main things that motivated me to do
further experiments with hormones. When I have a bad night I feel terrible, and
if I have too many bad nights I tend to get more colds and flu. When I sleep
well I function better.
CONCLUSION
Today, tricyclic antidepressants are not the first-choice medications for adults
with autism. Many of my autistic friends and associates are having good results
with fluoxetine or fluvoxamine. During my lectures at autism conferences I have
heard of many medication disasters. The most common is when too high a dose
of an SSRI is prescribed. People with autism often need much lower doses than
others (J. Ratey and E. Cook, personal communication). Too high a dose will
cause irritability, insomnia, and aggression. Discussions with hundreds of parents
and many people with autism indicate that low doses of fluoxetine, sertraline,
paroxetine, or fluvoxamine are often effective. Scientific studies also support the
use of fluoxetine or fluvoxamine for autistic adults (22,25). These are good first-
choice medications.
Epilepsy is common in people with autism [26]. During my travels I have
been told of three or four autistic cases in which grand mal seizures were triggered
by clomipramine. Clomipramine works well for autistic people with severe obses-
sive-compulsive disorder, but it is probably not the best first-choice medication
in autism due to the increased epilepsy hazard.
416 Grandin
REFERENCES
1. Grandin T, Scariano M. Emergence Labelled Autistic. New York: Warner Books,
1986.
2. Grandin T. Thinking in Pictures. New York: Vintage Books, 1995.
3. Bemporad ML. Adult recollections of a formerly autistic child. J Autism Dev Disord
1979; 9:179–197.
4. Stehli A. Sound of a Miracle. New York: Doubleday, 1991.
5. White DB, White MS. Autism from the inside. Medical Hypotheses 1987; 24:223–
229.
6. Williams D. Somebody Somewhere. New York: Time Books, 1994.
7. Coleman RS, Frankel F, Ritvoe E, Freeman BJ. The effects of fluorescent and incan-
descent illumination upon repetitive behaviors in autistic children. J Autism Dev
Disord 1976; 6:157–162.
8. Waterhouse L, Fein D, Modahl C. Neurofunctional mechanisms in autism. Psychol
Rev 1996; 103:457–489.
9. Lovaas I. Behavioral treatment and normal educational and intellectual functioning
in young autistic children. J Consult Clin Psychol 1987; 55:3–9:
10. Williams D. Autism: an Inside Outside Approach. Wiltshire, England: Cromwell
Press Melksham, 1996.
11. Agren JA. Sensory Integration and the Child. Los Angeles: Western Psychological
Services, 1979.
12. Rimland B, Edelson S. The effects of auditory integration training in autism. J
Speech Lang Pathol 1994; 5:16–24.
13. Elliot RO, Dobbin AR, Rose GD, Soper HV. Vigorous aerobic exercise versus gen-
eral motor training effects on maladaptive and stereotypic behavior of adults with
both autism and mental retardation. J Autism Dev Disord 1994; 24:565–576.
14. Walters RG, Walters WE. Decreasing self-stimulatory behavior with physical exer-
cise in a group of autistic boys. J Autism Dev Disord 1980; 10:379–387.
15. McGinsey JF, Favell JE. The effects or increased physical exercise on disruptive
behavior in retarded persons. J Autism Dev Disord 1988; 18:167–179.
16. Grandin T. Brief report: response to National Institutes of Health report. J Autism
Dev Disord 1996; 26:185–187.
17. Gillingham G. Autism—Fragile Handle with Care. Arlington, TX: Future Horizons,
1995.
18. Joliffe T. Lakesdown R, Robinson C. Autism, a personal account. Communication
1992: 26(3):12–19.
19. Grandin T. How people with autism think. In: Schopler E, Mesibov G, eds. Learning
and Cognition in Autism. New York: Plenum Press, 1995.
20. Grandin T. Calming effects of deep touch pressure on patients with autistic disor-
ders, college students and animals. J Child Adolesc Psychopharmacol 1992; 2:63–
70.
21. McClure MK, Holtz M. The effects of sensory stimulatory treatment on an autistic
child. Am J Occup Ther 1991; 45:1138–1142.
22. McDougal C, Sherman C. Fluvoxamine for obsessive-compulsive disorder. Med
Lett Drugs Ther 1995; 37:13–14.
A Personal Perspective 417
23. Zisserman L. The effects of deep pressure on self stimulating behavior in a child
with autism and other disabilities. Am J Occup Ther 1992; 46:547–551.
24. Sheean DV, Beh MB, Basllanger J, Jacobson G. Treatment of endogenous anxiety
with phobic, hysterical and hypochondriacal symptoms. Arch Gen Psychiatry, 1980;
37:51–59.
25. Cook EH, Rowlett R, Jasiskis C, Levanthal B. Fluoxetine treatment of children
and adults with autistic disorder and mental retardation. J Am Acad Child Adolesc
Psychiatry 1992: 31:739–745.
26. Gillberg C. The treatment of epilepsy in autism. J Autism Dev Disord 1991; 21:
61–77.
21
Future Trends
ASSESSMENTS
Additional research toward standardized screening and diagnosis of autism spe-
cifically, and the pervasive developmental delay spectrum disorders, will aid indi-
419
420 Hollander and Rawitt
vidual children and families, as well as studies of early intervention and out-
comes. Rigorous data collection will help reveal the multifactorial etiology of
autism and pervasive developmental delays, especially associated with other dis-
eases, by analyzing abnormal lab values, genetic testing, and other ancillary
testing.
Emphasis on a child’s ability to use a capacity toward an emotional goal,
or to satisfy a need, is described in a functional developmental approach, which
needs further systemization and exploration but may improve assessments and
intervention based on what is observable and known.
on the tolerability and response to SSRIs. These findings warrant further research
on 5-HT functioning in autism and its impact on pathophysiology and symptom
domains of autism, and may result in safer and more effective treatments for
specific symptom domains.
The autism phenotype may be a final common pathway of various patho-
physiological mechanisms, one of which involves autoimmune and neuroendo-
crine factors. Findings in this area are clearly preliminary, but perhaps specific
immunomodulatory therapeutic agents may result in amelioration of symptoms
in some patients.
Investigators have examined the relationships between neurochemical dys-
function of specific subtypes of the serotonergic system and neuroanatomical
abnormalities, especially in the cerebellum. Further studies are needed to test this
hypothesis.
TREATMENTS
Pharmacological approaches have been a key element in managing specific symp-
toms in autistic individuals. Mood stabilizers, antiepileptic drugs, antidepressants
(particularly the SSRIs), atypical antipsychotics, stimulants, and cholinergic
agents are available on a clinical basis, but need further double-blind, placebo-
controlled studies to validate safety and efficacy. In particular, studies in children
and adolescents are important and needed. There is also a need for new, better,
safer, and more efficacious compounds to be characterized and tested with appro-
priate outcome measures and study designs.
Studies of children with autism spectrum disorders and either clinical or
subclinical seizures need to be undertaken to determine the effects of seizures
on behavior and language. Epileptiforme discharges on EEG recordings, without
clinical seizures, can cause behavioral, language, and cognitive impairments, and
this needs to be studied in autism spectrum disorders as well. Additionally, treat-
ment modalities need to be systematized and studied in controlled clinical trials
that employ double-blind methods and placebo controls.
The treatment of autism spectrum disorder individuals with movement dis-
orders has been hampered by a lack of published reports and a need for clinical
trials, which are crucial in order to better characterize and treat these individuals.
A variety of alternative treatments for autism have gained a great degree
of popularity but do not have systematic data to support widespread use. These
include secretin, peridoxin and magnesium, vitamin A, vitamin C, dimethylgly-
cine, ORG2766, chelation of heavy metals, antifungals and antibiotics, various
dietary interventions, and auditory integration training.
In the future, behavioral assessment and treatment may be enhanced by
research on learning and neuropsychological correlates of specific areas of diffi-
culty. Research on how to address specific areas of difficulty with behavioral
422 Hollander and Rawitt
treatments, and how to integrate behavioral treatments with new biomedical find-
ings, is also needed. Individuals nonresponsive to a treatment may comprise a
specific subgroup, further clarifying information on the neural basis of specific
deficit areas. Alternatively, individuals who improve with the help of specific
treatments may again reveal a subclass of individuals, which may enhance our
understanding of autism spectrum disorders.
ADVOCACY
Autism research has been greatly facilitated by advocacy groups such as Cure
Autism Now, the National Alliance for Autism Research, and the Autism Society
of America that have developed a more unified approach to lobbying and working
with the National Institute of Health and the Centers for Disease Control and
Prevention. This has led to a common message to legislative and administrative
bodies, resulting in better funding for basic and applied research. Nevertheless,
more work is needed to improve funding. Advocacy in education to inform the
public of the cost benefits of improved educational access and the clinical benefits
is also needed. Efforts to find appropriate work and housing for people with
autism spectrum disorders are crucial in moving individuals from sheltered work-
shops to meaningfully compensated employment and out of perpetual poverty
and dependency. A long-term objective of advocacy groups is the creation of
national standards for the training and qualifications for people who work with
individuals with autism spectrum disorders. Also, there is a need for in-service
training for people working with individuals with autism spectrum disorders. As
part of this advocacy approach, reports from people with autism spectrum disor-
ders and their families are helpful in educating professionals and the general
public about these disorders, and in helping individuals, families, and our society
as a whole to integrate people with autism spectrum disorders and help them
become useful and productive members of society.
Index
423
424 Index