ArticlePDF AvailableLiterature Review

Abstract and Figures

It is established that neurodevelopmental disability (NDD) is common in neonates undergoing complex surgery for congenital heart disease (CHD); however, the trajectory of disability over the lifetime of individuals with CHD is unknown. Several ‘big issues’ remain undetermined and further research is needed in order to optimise patient care and service delivery, to assess the efficacy of intervention strategies and to promote best outcomes in individuals of all ages with CHD. This review article discusses ‘gaps’ in our knowledge of NDD in CHD and proposes future directions.
Content may be subject to copyright.
Open access
1
Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
To cite: Verrall CE, Blue GM,
Loughran-Fowlds A, etal. ‘Big
issues’ in neurodevelopment for
children and adults with
congenital heart disease. Open
Heart 2019;6:e000998.
doi:10.1136/
openhrt-2018-000998
Received 18 December 2018
Revised 18 March 2019
Accepted 26 April 2019
For numbered afliations see
end of article.
Correspondence to
Dr David Winlaw; david.
winlaw@ health. nsw. gov. au
‘Big issues’ in neurodevelopment for
children and adults with congenital
heart disease
Charlotte E Verrall,1,2 Gillian M Blue,1,2 Alison Loughran-Fowlds,2,3
Nadine Kasparian,1,4 Jozef Gecz,5 Karen Walker,3 Sally L Dunwoodie,6,7
Rachael Cordina,8,9 Gary Sholler,1,2 Nadia Badawi,2,3 David Winlaw 1,2
Congenital heart disease
© Author(s) (or their
employer(s)) 2019. Re-use
permitted under CC BY-NC. No
commercial re-use. See rights
and permissions. Published
by BMJ.
AbstrAct
It is established that neurodevelopmental disability (NDD)
is common in neonates undergoing complex surgery for
congenital heart disease (CHD); however, the trajectory
of disability over the lifetime of individuals with CHD is
unknown. Several ‘big issues’ remain undetermined and
further research is needed in order to optimise patient care
and service delivery, to assess the efcacy of intervention
strategies and to promote best outcomes in individuals
of all ages with CHD. This review article discusses ‘gaps’
in our knowledge of NDD in CHD and proposes future
directions.
INTRODUCTION
Care of children with congenital heart
disease (CHD) is associated with a low risk of
periprocedural mortality and morbidity in all
but the most complex of conditions, which
constitute fewer than 10% of presentations.
A broad suite of outcomes is now evaluated,
including cardiovascular functional status
(how well the circulation works), educational
attainment and employment, social engage-
ment and psychological well-being, which,
when combined, are assessed as ‘quality
of life’ (QOL). An understanding of these
issues is important not only for those looking
forward after CHD is ‘fixed’ but also for those
contemplating long-term expectations after
a fetal diagnosis. This is a major issue in the
field as it is recognised that QOL is reduced
in children with CHD,1 which, in turn, may
have a detrimental effect on the QOL of
the parents and support networks of those
affected.
In this review, we articulate the undeter-
mined ‘big issues’ in neurodevelopment
affecting those who require treatment for
CHD. We bring together a description of the
problem, an overview of the aetiology, eval-
uation of the effectiveness of current inter-
ventions and considerations for the future.
For the purpose of the review, CHD patients
those require early cardiac intervention are
of primary interest, which make up a minority
of CHD as a whole. Findings may not be
relevant to individuals with minor lesions,
including minor pulmonary valve abnormal-
ities and small ventricular septal defects, that
do not require surgery.
Incidence and manifestations of
neurodevelopmental disability
Central to normal growth and learning is
brain development. Abnormal brain develop-
ment is a greater issue for children with CHD
(~20% of those requiring cardiac surgical
intervention in infancy) compared with those
without major illness (~5%). There is a spec-
trum of manifestations in what is termed
neurodevelopmental disability (NDD)
(figure 1). At one end, there are typical phys-
ical manifestations of developmental ‘delay’
which may resolve or be remediated over
time, through to persisting behavioural and
psychosocial syndromes, and major disability
at the severe end of the phenotype. The
pathophysiology of the subentities of NDD
may well differ but are grouped for clinical
convenience.
Depending on the definition, NDD may
be the most common adverse outcome in
children with CHD. Up to 50% of children
requiring cardiac intervention exhibit NDD,
including mild cognitive impairments, diffi-
culties with attention and hyperactivity, defi-
cits in motor functioning, social interaction,
language and communication skills, and
delayed executive function,2–7 which may
persist into school age and beyond.8–10 As
expected, NDD has a detrimental effect on
educational achievement and attainment,
which consequently affects later employ-
ability, independence and relationships, and
may heighten the burden of psychological
disturbance and reduce overall QOL11–13
(figure 1).
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
Open Heart
2Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
Figure 1 Potential manifestations of NDD in people with
CHD and associated long-term outcomes. ADHD, attention-
decit/hyperactivity disorder; CHD, congenital heart disease;
NDD, neurodevelopmental disability.
The extent to which these manifestations continue into
adolescence and adulthood remains unclear and the subject
of ongoing research. While many older CHD patients are
doing much better physically than was ever expected at the
time of their surgery, understanding the impact of NDD
in later life is essential. The adult health system is not
well equipped to support these issues, particularly given
widespread stigma and negative societal attitudes towards
disability, and a lack of adequate resourcing for dedicated
neurocognitive services in CHD; a particularly vulnerable
time is during the transition to adult health services.14
Advancing our understanding of the onset, causes and
long-term trajectory of NDD is important to advance effec-
tive intervention and management.
NEURODEVELOPMENTAL OUTCOMES IN CHD AT DIFFERENT
AGES
Children
It is well established that NDD in CHD is an issue, with its
origins in early gestation. Infants and children with CHD
requiring intervention display NDD across a multitude of
domains with outcomes comparable to those observed in
premature infants and other sick neonates.3 4 15 In our clin-
ical experience, up to 29% of children requiring cardiac
surgery during infancy displayed moderate-to-severe
impairment in at least one area of neurodevelopment at
age 12 months. While many individuals scored between
‘normative means’, up to 28% had scores 2 SD below
mean performance in at least one neurodevelopmental
scale indicative of ‘mildly’ reduced assessment scores. This
suggests that many infants requiring cardiac surgery will
have reduced abilities compared with the typical popula-
tion.3
Abnormalities noted in infancy may persist into early
childhood. Utilising the Australian ‘National Assessment
Program—Literacy and Numeracy’ data, we demon-
strated that 13.1% of children that underwent a cardiac
procedure within the first year of life were classified as
having ‘special needs’ at school age compared with 4.4%
of children that had not had a cardiac procedure, as well
as displayed a higher proportion of learning disabilities
and speech impairments.16
Standardised assessments, such as the Bayley Scales
of Infant and Toddler Development (BSITD)17 and the
Wechsler Scales of Intelligence,18 are routinely used at key
time points throughout childhood, starting with assessments
as early as 1 month of age (table 1). The BSITD assessments
have been found to underestimate developmental delay19 20
and newer assessments with a high sensitivity and specificity
for early detection of cerebral palsy are being introduced to
assess CHD infants, such as the Prechtl Qualitative Assess-
ment of General Movements and the Hammersmith Infant
Neurological Examination (HINE).21
Assessment frequency is often determined by risk
profile, those deemed ‘at-risk’ are routinely re-assessed
at specific intervals, with the acknowledgement that
risk profile can change over time.22 Scores are typi-
cally interpreted compared with normative data, and
scores determined to be ‘below average’ usually acti-
vate recommendations for intervention involving health
professionals from a range of disciplines, including devel-
opmental paediatrics, neurology, psychology and allied
healthcare providers, such as physiotherapists, occupa-
tional therapists, speech and language pathologists, and
child life therapy.
Extending existing guidelines published by the Amer-
ican Academy of Paediatrics (AAP), the American Heart
Association recommends universal screening and long-
term surveillance for NDD in all children with CHD.22
Clinical practice and current research is based on the
2006 AAP algorithm for developmental surveillance
and screening that emphasises the importance of early
identification and management of developmental disor-
ders, which suggests that earlier intervention improves
outcomes23; however, evidence to support this in the CHD
cohort is limited and much-needed to support necessary
expenditure in health systems.
Adolescents
NDD is reported in nearly twice as many adolescents with
CHD compared with population norms,24 with poorer
neurodevelopmental outcomes observed across various
domains, including full-scale IQ, perceptual reasoning,
working memory, visual perception, visuomotor integra-
tion, and executive and motor functioning.25 Attendance
at special schools and lack of final school examination
occurs in as many as 12% of adolescents with CHD,26 with
up to 65% receiving remedial academic or behavioural
services and up to 50% requiring therapeutic services,
including physiotherapy, speech therapy, occupational
therapy and psychotherapy or counselling.25 27 When
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
3
Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
Congenital heart disease
Table 1 Example neurodevelopmental assessment tools commonly used in CHD
Assessment Age range Scores
Bayley Scales of Development, Version III*
Cognition
Expressive language
Receptive language
Fine motor
Gross motor
1–42 months ≥8 normal
6–7 mild
2–6 moderate
1–2 severe
Wechsler Scales of Intelligence, Version IV*
Full-scale IQ
Verbal IQ
Performance IQ
Preschool and primary
2.5 years–7 years 7 months
Children
6–16 years
Adults
>16 years
≥130 superior or ‘gifted’
120–129 very high
110–119 bright normal
>90 average–low average
69–70 borderline mental functionality
<69 mental retardation
*These assessments are commonly used assessment tools in the CHD population but various other assessments have been used for
evaluation in children and adolescents with CHD including: Woodcock Johnson; Wide Range Achievement Test; Clinical Evaluation of
Language Fundamentals; Expressive Vocabulary Test; Neuropsychological Assessment; Peabody Picture Vocabulary Test; Rey-Osterrieth
Complex Figure Test; Visual-Motor Integration; Conners’ Continuous Performance Test; Children’s Memory Scale; Wide Range Assessment
of Memory and Learning; Behavior Rating Inventory of Executive Function; Delis-Kaplan Executive Function System; Bruininks-Oseretsky
Test of Motor Prociency; Peabody Developmental Motor Scales; Scales of Independent Behavior-Revised; Attention Decit/Hyperactivity
Disorder Rating Scale; Child Behavior Checklist; Youth Self-Report; Conners’ Rating Scale-Revised; Diagnostic Interview Schedule for
Children; Basic Assessment System for Children and Diagnostic Adaptive Behavior Scale.22
CHD, congenital heart disease.
compared with sibling controls, outcomes appear worse
than when compared with population norms, particu-
larly for full-scale IQ and processing speed28 and may be
a better overall assessment.
Understanding the onset and trajectory of NDD
throughout life is important for effective manage-
ment and optimal intervention; however, the extent
to which early childhood outcomes predict later
disability is uncertain. The Boston Circulatory Arrest
Study (BCAS) and the Aachen Study are the only
prospective longitudinal studies in this field. Both
explored neurodevelopment at multiple time points
throughout childhood and adolescence in individuals
with surgically corrected transposition of the great
arteries (TGA). The BCAS study found that outcomes
in those with TGA were below population norms across
various neurodevelopmental domains at different time
points throughout childhood,9 10 29 30 and below mean
average scores continued to be observed at age 16 years
when compared with ‘healthy’ controls, particularly in
academic achievement and social cognition. A substan-
tial proportion of TGA patients scored 1 SD below
the expected population mean across various neuro-
developmental domains, including memory (35%),
academic achievement (26%–27%) and visual-spatial
skills (54%), and frequencies of scores greater than the
cut-off for clinical concern were significantly higher in
executive functioning (13% self-reports, 23% parent
reports and 38% teacher reports) and attention (19%).
This was accompanied by a higher incidence of
brain abnormalities detected by MRI; however, these
tended to be acquired rather than developmental and
no significant association was identified between MRI
abnormality and neurodevelopmental test scores.27
Similar findings were also demonstrated in the Aachen
study, where significant motor dysfunction, poorer
acquired abilities (learning knowledge) and speech
impairment were found at age 5 years compared with
population norms,31 32 and assessment at age 10 years
showed that neurological and speech impairments were
more frequent and motor function, acquired abilities
and language were reduced compared with norms.8 33
When re-assessed during adolescence, IQ scores were
within the normal range; however, the frequency of
scores 2 SD below the expected mean for performance
IQ was 11%, suggesting a greater incidence of specific
cognitive deficits compared with the normal popula-
tion through adolescence.26
Evidence-based predictors of later NDD will aid in iden-
tifying those ‘at-risk’ of adverse outcomes and provide
opportunities for intervention. As expected, CHD
complexity is predictive of worse neurodevelopmental
outcomes in adolescence.34 35 Children with ‘simple’
CHD, such as atrial septal defects, have demonstrated
some impairment compared with population norms,36
while others have found no differences in outcomes based
on CHD complexity.25 Most neonatal indices, including
birth weight and length, and 1 and 5 min Apgar score,
are not predictive of adolescent NDD, with the exception
of head circumference.35 Head circumference continues
to be smaller than average in adolescents with CHD, and
neuroimaging studies have demonstrated smaller total
brain volumes, total white matter (WM), and cortical and
deep grey matter volumes compared with controls, with
those with cyanotic CHD being more notably affected
compared with acyanotic CHD.37 Smaller brain volumes
have been found to correlate with functional outcomes
in adolescents with CHD but not control subjects,
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
Open Heart
4Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
highlighting the importance of quantitative imaging
measurements in this population.37
In time and with more research, these findings may
provide a diagnostic tool in the identification and inter-
vention of NDD in CHD; however, the generalisability
of findings is currently limited due to the studies being
small sample, single time point observations. To fully
understand the reliability and clinical significance of
these findings, longitudinal studies are needed.
Adults
Adults living with CHD now outnumber children,
accounting for up to 66% of the CHD population.38
The limited research to date shows that adults with
CHD (ACHD) display NDD across various domains,
including reduced abilities in executive functioning,39
information processing speed, psychomotor speed and
reaction time,40 overall and divided attention,39 40 fine
motor function,39 working memory41 and visuospatial
skills,41 with those with more complex CHD being more
notably affected compared with adults with simpler
CHD.39 40 42 A high frequency of MRI abnormalities and
reduced brain volumes in adults with cyanotic CHD has
also been observed.43 Adults with more complex CHD
have been found to have a higher frequency of neuro-
logical comorbidities, such as stroke and seizures, and
are more likely to be unemployed and receiving disa-
bility benefits despite educational attainment being no
different to those with simpler CHD.41
The rate of unemployment across all ACHD is esti-
mated at 18%–50%,11 44 and the incidence of comorbid
psychiatric disorders (eg, anxiety and depression),
pragmatic language impairment and delayed transi-
tion to independent living is increased.45 Understand-
ably, the overall QOL in ACHD is reduced compared
with population norms,46 and the accumulative effects
of neurological disability pose great demands on the
person affected, their support networks and societal
resources. Furthermore, the burden of neurocognitive
disability is likely to impact the rate of loss of follow-up
in this population, which has implications for the risk
of complications and consequentially, a higher cost
burden on resources. ACHD patients often require
additional nursing/allied health support to maintain
engagement, which also impacts the costs of care.
As the growing ACHD population ages, new concerns
are emerging regarding an increased risk of neurocog-
nitive decline and dementia, particularly early-onset
dementia, compared with population norms.38 47 This
may be evident earlier in life than typically expected and
associated with tachycardia, atrial fibrillation, hyperten-
sion, stroke, disordered glucose metabolism, coronary
artery disease and heart failure.47 48 Other risk factors for
dementia are also more common in the CHD population,
including genetic disorders and the impact of reduced
exercise capacity. Adults with severe CHD are considered
to have a greater risk of dementia, particularly those with
single ventricle morphology.47
AETIOLOGY OF THE CHD+NDD PHENOTYPE—WHAT CAN WE
MODIFY?
While we are starting to build a clearer picture of the inci-
dence of NDD over the course of a lifetime, the underlying
causes of the CHD+NDD paradigm are not fully under-
stood and the extent to which NDD can be prevented or
modified is unknown. Early research focused on intraop-
erative factors as the cause of adverse neurodevelopmental
outcomes in CHD.29 30 49–51 Use of prolonged deep hypo-
thermic circulatory arrest and extracorporeal membrane
oxygenation are still considered to be contributory risk
factors for adverse outcomes,2 51 and surgical techniques
have been adapted, where possible, to minimise potential
detrimental burden. Somewhat surprisingly, perioperative
factors have been found to explain only 5%–8% of varia-
bility in NDD outcomes following cardiac surgery.2 52 53
The fact that modification and improvements in surgical
techniques have not been accompanied by improvement in
neurodevelopmental outcomes supports this view.27 54 The
current understanding is that patient-specific and many
preoperative and antenatal factors are likely to contribute
to the majority of neurodevelopmental impairment in
people with CHD.
Development of the heart and brain are intimately
related and fetal neuroimaging demonstrates abnormal-
ities that are present from early gestation that are likely
to impact brain structure and growth in utero as a result
of altered perfusion and substrate delivery.55 56 Some
changes are dependent on the CHD anatomy, such as
retrograde perfusion of the aortic arch via the ductus in
fetuses with aortic atresia, and perfusion of the brain with
relatively hypoxic blood in fetuses with TGA.57 In support
of this concept, dysregulation of some angiogenic genes in
the brain of human fetuses with CHD has been observed,
possibly as a result of chronic hypoxia contributing to
abnormal brain development.58 59 Measurement of fetal
cerebral oxygenation by MRI demonstrates a reduction
in oxygen levels with increasing gestational age in fetuses
with CHD, which is considered to be significantly below
typical at as early as 32 weeks gestation; impaired perfu-
sion also correlates with smaller brain size.60 Sun et al61
demonstrated that a 15% reduction in cerebral oxygen
delivery and 32% reduction in cerebral VO2 in CHD
fetuses were associated with a 13% reduction in fetal
brain volume, supporting a direct link between reduced
cerebral oxygenation and impaired brain growth.
Disruption in fetal brain perfusion is thought to
contribute to greater susceptibility to brain injury in the
term neonate with CHD.62 63 Most commonly reported
is WM injury (WMI; up to 50% preoperatively and 62%
postoperatively64 65), which is comparable to the incidence
of periventricular leukomalacia identified in preterm
infants.63 66 The extent to which WMI worsens with surgery
is unclear, but WMI detected both preoperatively and
postoperatively is associated with NDD reported at various
points throughout childhood,65 67 and adolescents with
TGA have demonstrated reduced WM microstructure and
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
5
Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
Congenital heart disease
globally altered WM topology which correlates with worse
neurocognitive functioning across multiple domains.68 69
These data support the notion that the CHD brain may be
smaller and relatively underdeveloped at birth. Catch-up
growth may be possible as has been demonstrated in
infants after the arterial switch procedure for simple trans-
position,70 noting that persisting WM abnormalities have
been reported in other studies.69 71
Several strategies have been suggested to improve fetal
brain development. Maternal oxygen therapy is consid-
ered as one possible method and has also been used to
promote growth of small left ventricle morphology by
increasing fetal pulmonary blood flow and left atrial
return.59 While this is considered a diagnostic and ther-
apeutic tool in fetuses with some CHD subtypes, this
strategy is rarely used in clinical practice.59 72 Transla-
tional research in lambs offering an ‘artificial womb’,
where ‘normal’ substrate and oxygenation are provided
by extracorporeal support have allowed testing of the
concept that correction of flow and oxygenation abnor-
malities can improve brain development73 74 and may
eventually form the basis of care for preterm infants
with CHD, delaying the time to definitive cardiac care
to allow for brain maturation. The fetal brain in CHD
may also be affected by more generalised placental insuf-
ficiency, which may be difficult to detect using conven-
tional means.57 75 The placenta and fetal heart develop
in parallel and share a common vulnerability to genetic
defects, suggesting that deleterious defects in these
gene pathways could likely result in abnormality in the
morphology of both, with placental insufficiency further
exacerbating the development of key organs, including
both the heart and the brain.57 76–78 Chronic placental
insufficiency has been recognised to have serious conse-
quences on fetal growth, known as intrauterine growth
restriction (IUGR), which has been associated with
CHD79; however, the exact cause and effect mechanisms
of this relationship are unknown. Other factors, such as
folate metabolism, are also believed to compound the
issue of placental insufficiency and IUGR by impacting
the underlying mechanisms of cell growth and func-
tion.80 To add further ‘insult to injury’, fetal development
and growth may also be impacted by external environ-
mental factors, including developmental neurotoxicity
due to toxins, such as alcohol, drugs and environmental
organophosphates. Certain environmental chemicals
have been associated with cognitive and neurological
impairment, including diminished intellectual func-
tioning, learning disabilities, attention problems, hyper-
activity, attention-deficit/hyperactivity disorder (ADHD)
and autism, and are thought to disrupt the development
of the vulnerable fetal brain.81–83
Some very fundamental and readily accessible strategies
are known to optimise neurodevelopmental outcome.
Fetal cardiac diagnosis is important as it allows for coordi-
nation of perinatal care, and also provides an opportunity
for detailed antenatal counselling. Antenatal diagnosis
is associated with reduced risk of preoperative brain
injury, improved postnatal brain development and better
neurodevelopmental outcomes in infants with complex
CHD.73 74 Fetal diagnosis also allows planning of delivery
with current guidelines suggesting delivery between 39
and 40 weeks gestation.84 Early-premature and late-pre-
mature birth in the CHD population have been associ-
ated with greater neurodevelopmental impairment85 and
fetuses with CHD should not be delivered early for the
convenience of the treating teams.
Information provided during antenatal counsel-
ling should include discussion of neurodevelopmental
outcomes. For those requiring neonatal cardiac surgery,
our own practice is to outline the broad scope of neuro-
developmental impairment at a risk of 2–3 times the rate
observed in the population without CHD. We describe
a spectrum from motor delay to behavioural issues
(including autism and ADHD) and major disability.
Anticipated surgical complexity, as well as the risk of
acquired brain injury (eg, stroke), must also be consid-
ered.2 Thresholds for decisions regarding continuation
of pregnancy are individual and not necessarily based
on estimates of lesion severity or anticipated neuro-
developmental outcomes. Multidisciplinary support
and input into these decisions are required. Genomic
sequencing and fetal brain MRI have an emerging but, as
yet, unproven role in understanding mechanisms86 and
quantification of risk.61
There is considerable but, as yet, unwarranted opti-
mism that genetic variants explain much of the NDD
in CHD, possibly as a result of difficulty in identifying
other causes of the CHD+NDD phenotype. Nevertheless,
there is a growing body of evidence that genetic factors
are indeed contributing to altered fetal brain develop-
ment and often represent the key determinants of NDD
outcomes in patients with CHD.45 87 88 Of course, there are
several genetic syndromes in which both CHD and NDD
co-occur, such as Williams syndrome, Alagille syndrome,
Noonan syndrome or 22q11 deletion syndrome, among
others89; however, our principal interest remains the
much larger number of non-syndromic patients with a
largely sporadic mode of presentation.
Homsy et al studied 1213 CHD parent–offspring trios
and identified supporting evidence for the long-hy-
pothesised shared genetic origin of at least a propor-
tion of CHD with NDD.88 They found that genes highly
expressed in the heart were enriched for high expres-
sion in the developing brain and overlapped with
genes found to contain damaging de novo variants in
a number of NDD cohorts, comprising individuals with
isolated NDD. Jin et al90 extended this analysis to 2871
CHD probands, including 2645 parent–offspring trios,
and confirmed these findings and additionally identi-
fied an overlap between CHD and autism genes with
the suggestion that chromatin modifier genes have a
specific role. As CHD/NDD gene lists start to emerge,
genotype–phenotype correlations require large and
well-characterised patient cohorts necessitating multi-
centre collaboration.
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
Open Heart
6Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
While these studies mark a significant milestone in our
understanding of the genetic underpinning of NDD in
CHD, the approach is not ready for clinical application.
The ability to predict the development of NDD at an early
time point on the basis of DNA sequencing is an attractive
prospect. Early work in this field using known CHD and
NDD genes86 demonstrates substantial genetic variation in
CHD+NDD patients in both the ‘heart’ and ‘brain’ genes.
More than 1000 genes may be implicated in CHD+NDD
patients, with the likelihood that computational assess-
ment of variant burden may be more useful than anal-
ysis of specific variants and genes. This is in contrast to
the identification of causal variants in CHD (without
NDD), which can nowadays be effectively achieved using
massively parallel sequencing approaches.91–93 A unifying
developmental model for NDD in CHD is yet to be estab-
lished. Whole-genome sequencing approaches in larger
well-defined patient cohorts, including prenatal and post-
natal clinical data and outcomes, will be required.
While contemporary research is predominantly focused
on perinatal and genetic contributions to altered brain
maturation and neurodevelopment, we should again
consider whether we are underestimating the role of
perioperative factors and chronic circulatory abnormal-
ities, including hypoxia. The key study by Gaynor et al in
200753 introduced the now common understanding that
innate patient factors have a greater part to play in deter-
mining neurodevelopmental outcomes compared with
the perioperative factors previously understood. This
study was based on the neurodevelopmental outcomes
of 188 neonates and infants who underwent cardiac
surgery utilising cardiopulmonary bypass. Perioperative
risk factors still have important associations with reduced
neurodevelopmental ability.2 94 Acquired perioperative
brain injury remains common in CHD, and is correlated
with NDD,2 64 and postoperative factors are believed to
still have a significant role in outcomes.95
EFFECTIVENESS OF NEW ASSESSMENTS AND INTERVENTION
The multitude of complex and often non-modifiable
mechanisms contributing to CHD+NDD make it hard
to determine optimal methods of intervention. The
extent to which we can minimise or even prevent adverse
outcomes is uncertain. Optimal management for those
at-risk should include a multidisciplinary approach
with early identification, vigorous intervention and
routine assessments at various time points throughout
life,22 96 97 and many hospital-based Cardiac Neurodevel-
opment Programs now exist to provide this service.98 99
However, research focusing on the efficacy of neurode-
velopmental intervention and treatment strategies in the
CHD population is scarce.
Advances in the early detection and treatment of cere-
bral palsy, unrelated to CHD, are likely to be relevant
to CHD patients with milder forms of NDD. Evaluation
of newer assessments, such as the General Movements
(GMs) assessment, is occurring and has shown that this
test is highly sensitive and specific to detect cerebral
palsy in cardiac infants at 3 months of age and should
be incorporated into routine standardised follow-up for
these infants; however, further research is needed into
the precise prediction of long-term outcomes using this
test.100 Combinations of the GMs and the HINE together
with standard assessments, such as MRI and BSITD, can
provide accurate and early diagnosis of infants at high
risk of cerebral palsy as early as 3 months of age, and can,
thus, provide strong impetus for linkage into early inter-
vention programmes to take advantage of the stage of
neuroplasticity.21 100 101
The Congenital Heart disease Intervention Program102
is one of the limited intervention trials in CHD which
examined the influence of family, particularly maternal,
factors on infant neurodevelopment. Parents assigned
to the intervention received tailored psychoeducation,
narrative therapy, problem-solving techniques and
parenting skills training, delivered in six sessions (each of
1–2 hours duration) by a clinical psychologist and paedi-
atric cardiac nurse specialist.
The intervention was initiated when infants were 3 months
of age and at 6-month follow-up, infants in the intervention
group had significantly higher mental development scores
compared with infants in the control group, as well as
higher rates of breastfeeding, lower maternal worry scores
and greater positive appraisals. Psychomotor scores did not
differ between intervention and control infants at 6-month
follow-up, with mean scores for both groups indicative of
psychomotor delay. While the results of this study provide
preliminary evidence that early parental psychological
intervention may bolster mental developmental outcomes
for infants with CHD, evidence of the longer-term efficacy
of this intervention strategy is much-needed, and integrated
mental health interventions tailored to support neurobio-
logical health in children with CHD are currently being
trialled.103 Psychological and socioeconomic factors and
a nurturing family environment are no doubt important
for maximising neurodevelopmental potential; however,
adequately powered randomised controlled trials which
provide data on the long-term effects of structured psycho-
logical interventions are needed to determine efficacy in
reducing the neurodevelopmental burden associated with
CHD.
Calderon and Bellinger104 have described avenues for
intervention and treatment of NDD found to be successful
in other cohorts, such as the use of psychostimulant
medications commonly used in the treatment of atten-
tion-deficit disorder to improve working memory and
attention performance, intensive computerised training,
such as the use of the widely successful Cogmed training
programme,105 as well as other non-pharmacological tech-
niques, such as specialised assistance and support within
the school classroom. While it is theorised that these tech-
niques could improve neurodevelopmental functioning in
affected individuals, controlled trials implementing these
strategies in a CHD cohort are lacking, and thus their
viability is unclear.
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
7
Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
Congenital heart disease
If, as yet undetermined, genetic and epigenetic influ-
ences are considered key to the development of NDD, is it
reasonable to expect that we can actually modify outcomes?
Early evidence would suggest that despite potential genetic
causes, early and intensive intervention can improve
outcomes in those affected. There is a dynamic interplay
between genes and environment that is understood to form
the basis of typical neurobehavioural maturation, and it is
believed that these principles can be applied to neurodevel-
opmental disorders, even in the context of a strong genetic
component.38 Synaptic development and neural plasticity
in the newborn period are highly sensitive to modification
and continue to be influenced by environment-dependent
factors throughout the first months and years of life. After
birth, the brain increases over 100% in volume in the first
year and another 15% by the end of the second year of
life39 and these key developmental windows are considered
crucial in laying the foundations for neurodevelopmental
outcomes.40
Cost-effectiveness of intervention is another key concern.
Neurodevelopmental interventions are complex, may be
life-long, and require a multidisciplinary approach with
input from an extensive list of primary health services not
necessarily directly linked to CHD, such as psychologists,
developmental paediatricians, behavioural neurologists,
physiotherapists and occupational therapists, as well as
non-health resources relating to education performance,
employability and social participation.106 It is estimated
that interventions to improve neurodevelopment have
a high economic return if implemented during preg-
nancy and early childhood107; however tangible evidence
to support this is limited due to the infancy of evidence-
based intervention trials in CHD. As our understanding
of NDD outcomes in response to new intervention strate-
gies progresses, the cost-benefit of these services should,
in turn, be estimated in order to guide the direction of
future clinical programmes and care.
CONCLUSIONS
CHD+NDD remains a cause for concern across the lifespan
of the individual with CHD and the adverse outcomes
observed in childhood extend into adolescence and adult-
hood, potentially increasing the risk of early neurocogni-
tive decline. The aetiology is complex, multifactorial and
often speculative, involving many currently non-modifi-
able factors. Contemporary research efforts are focused
on improving intervention strategies to minimise burden
and maximise healthy outcomes; however, these strategies
are still in their infancy. Controlled intervention trials and
extended periods of follow-up are needed to assess the effi-
cacy and cost-effectiveness of these techniques and to opti-
mise patient care, resource planning and service delivery
for people of all ages with CHD.
Author afliations
1Heart Centre for Children, The Children’s Hospital at Westmead, Sydney, New
South Wales, Australia
2Discipline of Child and Adolescent Health, Sydney Medical School, Faculty of
Health and Medicine, University of Sydney, Sydney, NSW, Australia
3Grace Centre for Newborn Care, The Children’s Hospital at Westmead, Sydney,
New South Wales, Australia
4Discipline of Paediatrics, School of Women’s and Children’s Health, UNSW
Medicine, University of New South Wales, Sydney, NSW, Australia
5Faculty of Health and Medical Sciences, University of Adelaide School of Medicine,
Adelaide, South Australia, Australia
6Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research
Institute, Darlinghurst, New South Wales, Australia
7Faculties of Medicine and Science, University of New South Wales, Sydney, NSW,
Australia
8Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
9Discipline of Medicine, Sydney Medical School, Faculty of Health and Medicine,
University of Sydney, Sydney, NSW, Australia
Contributors All the authors have contributed signicantly to the content of the
article.
Funding NK is the recipient of a National Heart Foundation of Australia Future
Leader Fellowship (101229), and a 2018–2019 Harkness Fellowship in Health Care
Policy and Practice from the Commonwealth Fund.
Competing interests None declared.
Patient consent for publication Not required.
Provenance and peer review Not commissioned; externally peer reviewed.
Data sharing statement There are no data in this work.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which
permits others to distribute, remix, adapt, build upon this work non-commercially,
and license their derivative works on different terms, provided the original work is
properly cited, appropriate credit is given, any changes made indicated, and the use
is non-commercial. See:http:// creativecommons. org/ licenses/ by- nc/ 4. 0/.
RefeRences
1. Denniss DL, Sholler GF, Costa DSJ, etal. Need for Routine
Screening of Health-Related Quality of Life in Families of Young
Children with Complex Congenital Heart Disease. J Pediatr
2019;205:21-28.e2.
2. Verrall CE, Walker K, Loughran-Fowlds A, etal. Contemporary
incidence of stroke (focal infarct and/or haemorrhage) determined
by neuroimaging and neurodevelopmental disability at 12
months of age in neonates undergoing cardiac surgery utilizing
cardiopulmonary bypass†. Interact Cardiovasc Thorac Surg
2018;26:644–50.
3. Walker K, Badawi N, Halliday R, etal. Early Developmental
Outcomes following Major Noncardiac and Cardiac Surgery in Term
Infants: A Population-Based Study. J Pediatr 2012;161:748–52.
4. Walker K, Loughran-Fowlds A, Halliday R, etal. Developmental
outcomes at 3 years of age following major non-cardiac and
cardiac surgery in term infants: A population-based study. J
Paediatr Child Health 2015;51:1221–5.
5. Gaynor JW, Stopp C, Wypij D, etal. Neurodevelopmental
Outcomes After Cardiac Surgery in Infancy. Pediatrics
2015;135:816–25.
6. Gaynor JW, Ittenbach RF, Gerdes M, etal. Neurodevelopmental
outcomes in preschool survivors of the Fontan procedure. J Thorac
Cardiovasc Surg 2014;147:1276–83.
7. Hansen E, Poole TA, Nguyen V, etal. Prevalence of ADHD
symptoms in patients with congenital heart disease. Pediatr Int
2012;54:838–43.
8. Hövels-Gürich HH, Seghaye M-C, Schnitker R, etal. Long-term
neurodevelopmental outcomes in school-aged children after
neonatal arterial switch operation. J Thorac Cardiovasc Surg
2002;124:448–58.
9. Bellinger DC, Wypij D, duPlessis AJ, etal. Neurodevelopmental
status at eight years in children with dextro-transposition of
the great arteries: The Boston Circulatory Arrest Trial. J Thorac
Cardiovasc Surg 2003;126:1385–96.
10. Bellinger DC, Wypij D, Kuban KCK, etal. Developmental and
Neurological Status of Children at 4 Years of Age After Heart
Surgery With Hypothermic Circulatory Arrest or Low-Flow
Cardiopulmonary Bypass. Circulation 1999;100:526–32.
11. Ptzer C, Helm PC, Rosenthal L-M, etal. Educational level and
employment status in adults with congenital heart disease. Cardiol
Young 2018;28:32–8.
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
Open Heart
8Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
12. Kasmi L, Bonnet D, Montreuil M, etal. Neuropsychological and
Psychiatric Outcomes in Dextro-Transposition of the Great Arteries
across the Lifespan: A State-of-the-Art Review. Front. Pediatr.
2017;5.
13. Wilson WM, Smith-Parrish M, Marino BS, etal.
Neurodevelopmental and psychosocial outcomes across
the congenital heart disease lifespan. Prog Pediatr Cardiol
2015;39:113–8.
14. Everitt IK, Gerardin JF, Rodriguez FH, etal. Improving the quality of
transition and transfer of care in young adults with congenital heart
disease. Congenit Heart Dis 2017;12:242–50.
15. Morton PD, Ishibashi N, Jonas RA. Neurodevelopmental
Abnormalities and Congenital Heart Disease: Insights Into Altered
Brain Maturation. Circ Res 2017;120:960–77.
16. Lawley CM, Winlaw DS, Sholler GF, etal. School-Age
Developmental and Educational Outcomes Following Cardiac
Procedures in the First Year of Life: A Population-Based Record
Linkage Study. Pediatr Cardiol 2018.
17. Aylward GP. Bayley Scales of Infant and Toddler Development. In:
Kreutzer JS, DeLuca J, Caplan B, eds. Encyclopedia of Clinical
Neuropsychology. New York, NY: Springer New York, 2011: 357–8.
18. Flanagan DP, McGrew KS, Ortiz SO. The Wechsler Intelligence
Scales and Gf-Gc theory: A contemporary approach to
interpretation. Needham Heights, MA, US: Allyn & Bacon, 2000.
19. Acton BV, Biggs WSG, Creighton DE, etal. Overestimating
Neurodevelopment Using the Bayley-III After Early Complex
Cardiac Surgery. Pediatrics 2011;128:e794–800.
20. Chinta S, Walker K, Halliday R, etal. A comparison of the
performance of healthy Australian 3-year-olds with the standardised
norms of the Bayley Scales of Infant and Toddler Development
(version-III). Arch Dis Child 2014;99:621–4.
21. Novak I, Morgan C, Adde L, etal. Early, Accurate Diagnosis and
Early Intervention in Cerebral Palsy: Advances in Diagnosis and
Treatment. JAMA Pediatr 2017;171:897–907.
22. Marino BS, Lipkin PH, Newburger JW, etal. Neurodevelopmental
Outcomes in Children With Congenital Heart Disease: Evaluation
and Management. Circulation 2012;126:1143–72.
23. Medical Home Initiatives for Children With Special Needs Project
Advisory Committee. Identifying Infants and Young Children With
Developmental Disorders in the Medical Home: An Algorithm
for Developmental Surveillance and Screening. Pediatrics
2006;118:405–20.
24. Cassidy AR, White MT, DeMaso DR, etal. Executive Function in
Children and Adolescents with Critical Cyanotic Congenital Heart
Disease. J Int Neuropsychol Soc 2015;21:34–49.
25. Schaefer C, von Rhein M, Knirsch W, etal. Neurodevelopmental
outcome, psychological adjustment, and quality of life in
adolescents with congenital heart disease. Dev Med Child Neurol
2013;55:1143–9.
26. Heinrichs AKM, Holschen A, Krings T, etal. Neurologic and
psycho-intellectual outcome related to structural brain imaging
in adolescents and young adults after neonatal arterial switch
operation for transposition of the great arteries. J Thorac
Cardiovasc Surg 2014;148:2190–9.
27. Bellinger DC, Wypij D, Rivkin MJ, etal. Adolescents with
d-Transposition of the Great Arteries Corrected with the Arterial
Switch Procedure: Neuropsychological Assessment and Structural
Brain Imaging. Circulation 2011;124:1361–9.
28. Murphy LK, Compas BE, Reeslund KL, etal. Cognitive and
attentional functioning in adolescents and young adults with
Tetralogy of Fallot and d-transposition of the great arteries. Child
Neuropsychology 2017;23:99–110.
29. Bellinger DC, Jonas RA, Rappaport LA, etal. Developmental and
Neurologic Status of Children after Heart Surgery with Hypothermic
Circulatory Arrest or Low-Flow Cardiopulmonary Bypass. N Engl J
Med 1995;332:549–55.
30. Bellinger DC, Rappaport LA, Wypij D, etal. Patterns of
Developmental Dysfunction After Surgery During Infancy to Correct
Transposition of the Great Arteries. Journal of Developmental &
Behavioral Pediatrics 1997;18:75–83.
31. Hövels-Gürich HH, Seghaye M-C, Däbritz S, etal. Cognitive and
motor development in preschool and school-aged children after
neonatal arterial switch operation. J Thorac Cardiovasc Surg
1997;114:578–85.
32. Hövels-Gürich HH, Seghaye MC, Däbritz S, etal. Cardiological
and general health status in preschool- and school-age children
after neonatal arterial switch operation. Eur J Cardiothorac Surg
1997;12:593–601.
33. Hovels-Gurich HHetal. Long term behavioural outcome after
neonatal arterial switch operation for transposition of the great
arteries. Arch Dis Child 2002;87:506–10.
34. Karsdorp PA, Everaerd W, Kindt M, etal. Psychological and
Cognitive Functioning in Children and Adolescents with
Congenital Heart Disease: A Meta-Analysis. J Pediatr Psychol
2007;32:527–41.
35. Matos SM, Sarmento S, Moreira S, etal. Impact of Fetal
Development on Neurocognitive Performance of Adolescents with
Cyanotic and Acyanotic Congenital Heart Disease. Congenit Heart
Dis 2014;9:373–81.
36. Sarrechia I, Miatton M, De Wolf D, etal. Neurocognitive
development and behaviour in school-aged children after surgery
for univentricular or biventricular congenital heart disease. Eur J
Cardiothorac Surg 2016;49:167–74.
37. von Rhein M, Buchmann A, Hagmann C, etal. Brain volumes
predict neurodevelopment in adolescents after surgery for
congenital heart disease. Brain 2014;137:268–76.
38. Marelli AJ, Ionescu-Ittu R, Mackie AS, etal. Lifetime prevalence
of congenital heart disease in the general population from 2000 to
2010. Circulation 2014;130:749–56.
39. Tyagi M, Fteropoulli T, Hurt CS, etal. Cognitive dysfunction in
adult CHD with different structural complexity. Cardiol Young
2017;27:851–9.
40. Klouda L, Franklin WJ, Saraf A, etal. Neurocognitive and executive
functioning in adult survivors of congenital heart disease. Congenit
Heart Dis 2017;12:91–8.
41. Ilardi D, Ono KE, McCartney R, etal. Neurocognitive functioning
in adults with congenital heart disease. Congenit Heart Dis
2017;12:166–73.
42. Daliento L, Mapelli D, Volpe B. Measurement of cognitive
outcome and quality of life in congenital heart disease. Heart
2006;92:569–74.
43. Cordina R, Grieve S, Barnett M, etal. Brain Volumetrics, Regional
Cortical Thickness and Radiographic Findings in Adults with
Cyanotic Congenital Heart Disease. Neuroimage 2014;4:319–25.
44. Zomer AC, Vaartjes I, Uiterwaal CSP, etal. Social burden and
lifestyle in adults with congenital heart disease. Am J Cardiol
2012;109:1657–63.
45. Marelli A, Miller SP, Marino BS, etal. Brain in Congenital Heart
Disease Across the Lifespan. Circulation 2016;133:1951–62.
46. Hunter AL, Swan L. Quality of life in adults living with congenital
heart disease: beyond morbidity and mortality. J Thorac Dis
2016;8:E1632–E1636.
47. Bagge CN, Henderson VW, Laursen HB, etal. Risk of Dementia in
Adults With Congenital Heart Disease: Population-Based Cohort
Study. Circulation 2018;137:1912–20.
48. de la Torre JC. Cardiovascular risk factors promote brain
hypoperfusion leading to cognitive decline and dementia.
Cardiovasc Psychiatry Neurol 2012;2012:1–15.
49. Hoffman GM, Mussatto KA, Brosig CL, etal. Systemic venous
oxygen saturation after the Norwood procedure and childhood
neurodevelopmental outcome. J Thorac Cardiovasc Surg
2005;130:1094–100.
50. Massaro AN, El-dib M, Glass P, etal. Factors associated with
adverse neurodevelopmental outcomes in infants with congenital
heart disease. Brain and Development 2008;30:437–46.
51. Hamrick SEG, Gremmels DB, Keet CA, etal. Neurodevelopmental
Outcome of Infants Supported With Extracorporeal Membrane
Oxygenation After Cardiac Surgery. Pediatrics 2003;111:e671–5.
52. Gaynor JW, Stopp C, Wypij D, etal. Impact of Operative and
Postoperative Factors on Neurodevelopmental Outcomes After
Cardiac Operations. Ann Thorac Surg 2016;102:843–9.
53. Gaynor JW, Wernovsky G, Jarvik GP, etal. Patient characteristics
are important determinants of neurodevelopmental outcome at
one year of age after neonatal and infant cardiac surgery. J Thorac
Cardiovasc Surg 2007;133:1344–53.
54. Hirsch JC, Jacobs ML, Andropoulos D, etal. Protecting the infant
brain during cardiac surgery: a systematic review. Ann Thorac Surg
2012;94:1365–73.
55. Khalil A, Bennet S, Thilaganathan B, etal. Prevalence of prenatal
brain abnormalities in fetuses with congenital heart disease: a
systematic review. Ultrasound Obstet Gynecol 2016;48:296–307.
56. Brossard-Racine M, du Plessis AJ, Vezina G, etal. Prevalence and
Spectrum of In Utero Structural Brain Abnormalities in Fetuses
with Complex Congenital Heart Disease. American Journal of
Neuroradiology 2014;35:1593–9.
57. McQuillen PS, Goff DA, Licht DJ. Effects of congenital heart
disease on brain development. Prog Pediatr Cardiol 2010;29:79–85.
58. Sánchez O, Ruiz‐Romero A, Domínguez C, etal. Brain angiogenic
gene expression in fetuses with congenital heart disease.
Ultrasound Obstet Gynecol 2018;52:734–8.
59. Co-Vu J, Lopez-Colon D, Vyas HV, etal. Maternal
hyperoxygenation: A potential therapy for congenital heart disease
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
9
Verrall CE, etal. Open Heart 2019;6:e000998. doi:10.1136/openhrt-2018-000998
Congenital heart disease
in the fetuses? A systematic review of the current literature.
Echocardiography 2017;34:1822–33.
60. Lauridsen MH, Uldbjerg N, Henriksen TB, etal. Cerebral
Oxygenation Measurements by Magnetic Resonance Imaging in
Fetuses With and Without Heart Defects. Circ Cardiovasc Imaging
2017;10:e006459.
61. Sun L, Macgowan CK, Sled JG, etal. Reduced Fetal Cerebral
Oxygen Consumption Is Associated With Smaller Brain
Size in Fetuses With Congenital Heart Disease. Circulation
2015;131:1313–23.
62. Hövels-Gürich HH. Factors Inuencing Neurodevelopment after
Cardiac Surgery during Infancy. Front. Pediatr. 2016;4.
63. Guo T, Chau V, Peyvandi S, etal. White matter injury in term
neonates with congenital heart diseases: Topology & comparison
with preterm newborns. Neuroimage 2019;185:742–9.
64. Beca J, Gunn JK, Coleman L, etal. New white matter brain injury
after infant heart surgery is associated with diagnostic group and
the use of circulatory arrest. Circulation 2013;127:971–9.
65. Claessens NHP, Algra SO, Ouwehand TL, etal. Perioperative
neonatal brain injury is associated with worse school‐age
neurodevelopment in children with critical congenital heart disease.
Dev Med Child Neurol 2018;60:1052–8.
66. Hinton RB, Andelnger G, Sekar P, etal. Prenatal head growth and
white matter injury in hypoplastic left heart syndrome. Pediatr Res
2008;64:364–9.
67. Andropoulos DB, Ahmad HB, Haq T, etal. The association between
brain injury, perioperative anesthetic exposure, and 12-month
neurodevelopmental outcomes after neonatal cardiac surgery: a
retrospective cohort study. Paediatr Anaesth 2014;24:266–74.
68. Rollins CK, Newburger JW, Roberts AE. Genetic contribution to
neurodevelopmental outcomes in congenital heart disease: are
some patients predetermined to have developmental delay? Curr
Opin Pediatr 2017;29:529–33.
69. Panigrahy A, Schmithorst VJ, Wisnowski JL, etal. Relationship
of white matter network topology and cognitive outcome in
adolescents with d-transposition of the great arteries. Neuroimage
2015;7:438–48.
70. Ibuki K, Watanabe K, Yoshimura N, etal. The improvement
of hypoxia correlates with neuroanatomic and developmental
outcomes: Comparison of midterm outcomes in infants with
transposition of the great arteries or single-ventricle physiology. J
Thorac Cardiovasc Surg 2012;143:1077–85.
71. Bellinger DC, Rivkin MJ, DeMaso D, etal. Adolescents with
tetralogy of Fallot: neuropsychological assessment and structural
brain imaging. Cardiol Young 2015;25:338–47.
72. Kelly CJ, Makropoulos A, Cordero-Grande L, etal. Impaired
development of the cerebral cortex in infants with congenital heart
disease is correlated to reduced cerebral oxygen delivery. Sci Rep
2017;7:15088.
73. Partridge EA, Davey MG, Hornick MA, etal. An extra-uterine
system to physiologically support the extreme premature lamb. Nat
Commun 2017;8:15112.
74. Lawrence KM, McGovern PE, Mejaddam A, etal. Chronic
intrauterine hypoxia alters neurodevelopment in fetal sheep. J
Thorac Cardiovasc Surg 2019.
75. Rychik J, Goff D, McKay E, etal. Characterization of the Placenta in
the Newborn with Congenital Heart Disease: Distinctions Based on
Type of Cardiac Malformation. Pediatr Cardiol 2018;39:1165–71.
76. Maslen CL. Recent Advances in Placenta–Heart Interactions. Front
Physiol 2018;9:735.
77. Perez-Garcia V, Fineberg E, Wilson R, etal. Placentation defects
are highly prevalent in embryonic lethal mouse mutants. Nature
2018;555:463–8.
78. Sparrow DB, Boyle SC, Sams RS, etal. Placental Insufciency
Associated with Loss of Cited1 Causes Renal Medullary Dysplasia.
JASN 2009;20:777–86.
79. Wallenstein MB, Harper LM, Odibo AO, etal. Fetal congenital
heart disease and intrauterine growth restriction: a retrospective
cohort study. The Journal of Maternal-Fetal & Neonatal Medicine
2012;25:662–5.
80. Rosario FJ, Nathanielsz PW, Powell TL, etal. Maternal folate
deciency causes inhibition of mTOR signaling, down-regulation
of placental amino acid transporters and fetal growth restriction in
mice. Sci Rep 2017;7:3982.
81. De Felice A, Ricceri L, Venerosi A, etal. Multifactorial Origin of
Neurodevelopmental Disorders: Approaches to Understanding
Complex Etiologies. Toxics 2015;3:89–129.
82. Lyall K, Schmidt RJ, Hertz-Picciotto I. Maternal lifestyle and
environmental risk factors for autism spectrum disorders. Int J
Epidemiol 2014;43:443–64.
83. Slotkin TA. Cholinergic systems in brain development
and disruption by neurotoxicants: nicotine, environmental
tobacco smoke, organophosphates. Toxicol Appl Pharmacol
2004;198:132–51.
84. Donofrio MT, Moon-Grady AJ, Hornberger LK, etal. Diagnosis and
treatment of fetal cardiac disease: a scientic statement from the
American Heart Association. Circulation 2014;129:2183–242.
85. Goff DA, Luan X, Gerdes M, etal. Younger gestational age is
associated with worse neurodevelopmental outcomes after cardiac
surgery in infancy. J Thorac Cardiovasc Surg 2012;143:535–42.
86. Blue GM, Ip E, Walker K, etal. Genetic burden and associations
with adverse neurodevelopment in neonates with congenital heart
disease. Am Heart J 2018;201:33–9.
87. Blue GM, Kirk EP, Giannoulatou E, etal. Advances in the Genetics
of Congenital Heart Disease: A Clinician’s Guide. J Am Coll Cardiol
2017;69:859–70.
88. Homsy J, Zaidi S, Shen Y, etal. De novo mutations in congenital
heart disease with neurodevelopmental and other congenital
anomalies. Science 2015;350:1262–6.
89. A. Richards A, Garg V. Genetics of Congenital Heart Disease. Curr
Cardiol Rev 2010;6:91–7.
90. Jin SC, Homsy J, Zaidi S, etal. Contribution of rare inherited and
de novo variants in 2,871 congenital heart disease probands. Nat
Genet 2017;49:1593–601.
91. Alankarage D, Ip E, Szot JO, etal. Identication of clinically
actionable variants from genome sequencing of families with
congenital heart disease. Genet Med 2018.
92. Blue GM, Kirk EP, Giannoulatou E, etal. Targeted next-generation
sequencing identies pathogenic variants in familial congenital
heart disease. J Am Coll Cardiol 2014;64:2498–506.
93. Szot JO, Cuny H, Blue GM, etal. A Screening Approach to Identify
Clinically Actionable Variants Causing Congenital Heart Disease in
Exome Data. Circulation 2018;11:e001978.
94. Wernovsky G, Licht DJ. Neurodevelopmental Outcomes in Children
With Congenital Heart Disease—What Can We Impact? Pediatric
Critical Care Medicine 2016;17(8 Suppl 1):S232–S242.
95. Nattel SN, Adrianzen L, Kessler EC, etal. Congenital Heart
Disease and Neurodevelopment: Clinical Manifestations, Genetics,
Mechanisms, and Implications. Canadian Journal of Cardiology
2017;33:1543–55.
96. Knutson S, Kelleman MS, Kochilas L. Implementation of
Developmental Screening Guidelines for Children with Congenital
Heart Disease. J Pediatr 2016;176:135–41.
97. Guralnick MJ. Early Intervention for Children with Intellectual
Disabilities: An Update. Journal of Applied Research in Intellectual
Disabilities 2017;30:211–29.
98. Soto CB, Olude O, Hoffmann RG, etal. Implementation of a
Routine Developmental Follow-up Program for Children with
Congenital Heart Disease: Early Results. Congenit Heart Dis
2011;6:451–60.
99. Brosig C, Butcher J, Butler S, etal. Monitoring developmental risk
and promoting success for children with congenital heart disease:
Recommendations for cardiac neurodevelopmental follow-up
programs. Clinical Practice in Pediatric Psychology 2014;2:153–65.
100. Crowle C, Galea C, Walker K, etal. Prediction of neurodevelopment
at one year of age using the General Movements assessment in the
neonatal surgical population. Early Hum Dev 2018;118:42–7.
101. Crowle C, Badawi N, Walker K, etal. General Movements
Assessment of infants in the neonatal intensive care unit following
surgery. J Paediatr Child Health 2015;51:1007–11.
102. McCusker CG, Doherty NN, Molloy B, etal. A controlled trial
of early interventions to promote maternal adjustment and
development in infants born with severe congenital heart disease.
Child Care, Health Dev 2010;36:110–7.
103. Kasparian NA, Winlaw DS, Sholler GF. “Congenital heart health”:
how psychological care can make a difference. Med J Aust
2016;205:104–7.
104. Calderon J, Bellinger DC. Executive function decits in congenital
heart disease: why is intervention important? Cardiol Young
2015;25:1238–46.
105. Roording-Ragetlie S, Klip H, Buitelaar J, etal. Working Memory
Training in Children with Neurodevelopmental Disorders.
Psychology 2016;07:310–25.
106. Lamsal R, Zwicker JD. Economic Evaluation of Interventions for
Children with Neurodevelopmental Disorders: Opportunities and
Challenges. Appl Health Econ Health Policy 2017;15:763–72.
107. Edmond KM, Strobel NA, McAuley K, etal. Economic evaluation
of interventions delivered by primary care providers to improve
neurodevelopment in children aged under 5 years: protocol for a
scoping review. Syst Rev 2017;6:59.
on 4 July 2019 by guest. Protected by copyright.http://openheart.bmj.com/Open Heart: first published as 10.1136/openhrt-2018-000998 on 3 July 2019. Downloaded from
... A significant proportion of children with complex congenital heart disease require specialised habilitative services, supportive therapies, and educational supports into adulthood. Early identification of, and intervention for developmental concerns is critical to the wellbeing of children with CoHD and the responsibility of health professionals as an integral function of health care [92,93]. ...
... It must be equitable, inclusive and emphasise early initiation of evidence-based therapies to support infants, children, adolescents, and adults with CoHD in reaching their full potential. There must also be recognition that a person's level of neurodevelopmental risk or neurocognitive functioning can change over time, therefore continued monitoring is critically important [93,94]. ...
... From the time of diagnosis, and throughout the whole of life, patients with CoHD and their carers must have access to high-quality, up-to-date information and education on neurodevelopment and neurocognitive functioning. This information must be tailored to their heart condition and provided in formats that are culturally, linguistically and developmentally appropriate, and in line with trauma-informed care [93,95]. In the context of fetal cardiac diagnosis, provision of information and education to caregivers may begin as early as during pregnancy. ...
Article
Full-text available
Australian National Standards of Care fir Childhood-onset Heart Disease
... This was delineated further in the landmark Boston Circulatory Arrest Study, in which they longitudinally studied CHD patients and found that patients with transposition of the great arteries (TGA) were below the population norms and below mean average scores when compared to healthy controls in areas such as academic achievement and social cognition [25]. These issues affect the child and can manifest in areas such as repeated school absences, lower IQs, perceptual reasoning, working memory, visual perception, and executive and motor functioning skills [26]. ...
... The intervention was conducted in the UK and there was no mention of race/ethnicity or any other social determinants of health factors, thereby limiting the ability to translate these results. This, like other interventional programs, is a starting point, but it should be taken into consideration that often continued care may be hampered by parental ability to access these resources, rather it may be with difficulty with in-person or virtual visits, adequate means of transportation, or access to technology can be limited [26]. Even with the view that early and longitudinal intervention may assist neurodevelopment in children, a limited number of centers offer both cardiac and neurodevelopmental rehab, or other follow-up services that may not be easily accessible locally, such as in rural communities [20]. ...
Article
Full-text available
Congenital heart defects (CHDs) are complex conditions affecting the heart and/or great vessels that are present at birth. These defects occur in approximately 9 in every 1000 live births. From diagnosis to intervention, care has dramatically improved over the last several decades. Patients with CHDs are now living well into adulthood. However, there are factors that have been associated with poor outcomes across the lifespan of these patients. These factors include sociodemographic and socioeconomic positions. This commentary examined the disparities and solutions within the evolution of CHD care in the United States.
... In addition to the inherent morbidity of CHD, newborns with CHD are at an increased risk of extra-cardiac developmental abnormalities [3]. Up to 50% of patients with severe CHD will be diagnosed with a form of neurodevelopmental disability (NDD) [4], with 29% displaying moderate to severe developmental impairment [5]. Furthermore, 11% of adolescents meet the criteria for intellectual disability and 65% receive remedial academic or behavioral services [5]. ...
... Up to 50% of patients with severe CHD will be diagnosed with a form of neurodevelopmental disability (NDD) [4], with 29% displaying moderate to severe developmental impairment [5]. Furthermore, 11% of adolescents meet the criteria for intellectual disability and 65% receive remedial academic or behavioral services [5]. ...
Article
Full-text available
Up to 50% of patients with severe congenital heart disease (CHD) develop life-altering neurodevelopmental disability (NDD). It has been presumed that NDD arises in CHD cases because of hypoxia before, during, or after cardiac surgery. Recent studies detected an enrichment in de novo mutations in CHD and NDD, as well as significant overlap between CHD and NDD candidate genes. However, there is limited evidence demonstrating that genes causing CHD can produce NDD independent of hypoxia. A patient with hypoplastic left heart syndrome and gross motor delay presented with a de novo mutation in SMC5. Modeling mutation of smc5 in Xenopus tropicalis embryos resulted in reduced heart size, decreased brain length, and disrupted pax6 patterning. To evaluate the cardiac development, we induced the conditional knockout (cKO) of Smc5 in mouse cardiomyocytes, which led to the depletion of mature cardiomyocytes and abnormal contractility. To test a role for Smc5 specifically in the brain, we induced cKO in the mouse central nervous system, which resulted in decreased brain volume, and diminished connectivity between areas related to motor function but did not affect vascular or brain ventricular volume. We propose that genetic factors, rather than hypoxia alone, can contribute when NDD and CHD cases occur concurrently.
... The risk factors for mental health conditions in this population are many and multifactorial, including but likely not limited to those associated with chronic illness, including stress, learned helplessness, overprotection by parents in addition to underlying genetic syndromes and parental mental health conditions. [8,11,18,19]. Furthermore, as a reflection of the bidirectional relationship between medical and psychological well-being, emerging evidence suggests that sustained, elevated psychological stress or distress may affect CHD morbidity and mortality [20]. ...
Article
Full-text available
Mental health conditions are a common comorbidity among children living with heart disease. Children with congenital heart disease are more likely to have a mental health condition than their unaffected peers or peers with other chronic illnesses, and mental health risk persists across their lifetime. While poorer mental health in adults with congenital heart disease is associated with worse overall health outcomes, the association between mental health and cardiac outcomes for children with heart disease remains unknown. Despite this, it is suspected that mental health conditions go undiagnosed in children with heart disease and that many affected children and adolescents do not receive optimal mental health care. In this article, we review mental health in congenital heart disease across the lifespan, across domains of care, and across diagnoses. Further directions to support mental health care for children and adolescents with heart disease include practical screening and access to timely referral and mental health resources.
... Long-term survival of individuals with complex CHD has dramatically improved over recent decades due to advances in surgical strategies and clinical care [2,3]. Good long-term clinical outcomes are predominantly defined in terms of cardiac function and physiological health status; however, there is growing recognition of the cognitive [17][18][19][20] and psychological sequelae [4] experienced by this cohort which can impact physical and psychosocial health [4,21,46]. Associations between cognitive functioning and psychosocial factors in adults with complex CHD are poorly characterized. The current findings add to a growing body of literature that demonstrates a high prevalence of cognitive dysfunction in adults with complex CHD [18][19][20]. ...
Article
Full-text available
Adults with complex congenital heart disease (CHD) are at risk for cognitive dysfunction. However, associations between cognitive dysfunction and psychosocial outcomes are poorly defined. Between June and November 2022, we prospectively recruited 39 adults with complex CHD who completed a computerized cognitive assessment (Cogstate) and validated psychosocial scales measuring psychological distress, health-related quality of life (HRQOL), and resilience. Participants had a mean age of 36.4 ± 11.2 years. Over half (62%) were women, most (79%) had complex biventricular CHD, and 21% had Fontan physiology. Prevalence of cognitive dysfunction was greatest in the domains of attention (29%), working memory (25%), and psychomotor speed (21%). Adjusting for age and sex, Pearson partial correlations between Cogstate z-scores and self-reported cognitive problems were small. Participants who lived in the most disadvantaged areas and those with a below-average annual household income had lower global cognitive z-scores (p = 0.02 and p = 0.03, respectively). Two-thirds (64%) reported elevated symptoms of depression, anxiety, and/or stress. Small correlations were observed between psychological distress and cognitive performance. Greater resilience was associated with lower psychological distress (r ≥ −0.5, p < 0.001) and higher HRQOL (r = 0.33, p = 0.02). Our findings demonstrate that adults with complex CHD have a high risk of cognitive dysfunction, though may not recognize or report their cognitive challenges. Lower socioeconomic status may be an indicator for those at risk of poorer cognitive functioning. Psychological distress is common though may not be a strong correlate of performance-based cognitive functioning. Formal cognitive evaluation in this patient population is essential. Optimizing resilience may be a protective strategy to minimize psychological distress and bolster HRQOL.
... Given the high prevalence of neurodevelopmental impairments in children born with complex CHD, there is an increasing focus to determine potentially modifiable practices within the cardiac ICU to improve neurodevelopmental trajectories in this population. [1][2][3] The inherently unnatural and stressful environment of the cardiac ICU for the newborn infant with CHD has significant potential negative impacts on brain development. Developmentally appropriate practices aiming to mitigate the infant stress experience are promising modifiable factors to improve neurodevelopmental outcomes. ...
Article
Full-text available
We performed a single-centre, retrospective study to assess physiologic changes of infants in the cardiac ICU while being held by their parent. Continuous data streaming of vital signs were collected for infants included in the study from January 2021 to March 2022. Demographic and clinical characteristics were collected from the electronic medical record. The physiologic streaming data were analysed using mixed-effects models to account for repeated measures and quantify the effect of parental holding. Comparison analysis was also performed controlling for intubation, pre-operative versus post-operative status, and whether the holding was skin-to-skin or not. Ninety-five patients with complete physiologic data were included in the study. There were no immediate adverse events associated with holding. Heart rate decreased during the response time compared to its baseline value (p = 0.01), and this decrease was more pronounced for the non-intubated and pre-operative patients. The near-infrared spectroscopy-based venous saturation increased overall (p = 0.02) in patients while being held. We conclude that parental holding of infants in the cardiac ICU can be safely accomplished, and the haemodynamic and oximetric profile during the holding is favourable compared to the infants’ baseline prior to holding.
... To ensure that children identified as having a 'favourable' cardiovascular outcome did not leave NSW following their initial surgery and have a 'unfavourable' cardiovascular outcome recorded in another jurisdiction, children over the age of 10 years old were excluded if they did not link to NSW school attendance data, identified by lack of a NAPLAN school test record. Children who were classified as 'exempt' from NAPLAN were also excluded from having a 'favourable' outcome as poor neurocognitive outcomes in CHD patients are associated with poor cardiovascular function and insufficient cerebral oxygenation [10]. ...
Article
Full-text available
IntroductionContemporary care of congenital heart disease (CHD) is largely standardised, however there is heterogeneity in post-surgical outcomes that may be explained by genetic variation. Data linkage between a CHD biobank and routinely collected administrative datasets is a novel method to identify outcomes to explore the impact of genetic variation. Objective Use data linkage to identify and validate patient outcomes following surgical treatment for CHD. Methods Data linkage between clinical and biobank data of children born from 2001-2014 that had a procedure for CHD in New South Wales, Australia, with hospital discharge data, education and death data. The children were grouped according to CHD lesion type and age at first cardiac surgery. Children in each `lesion/age at surgery group' were classified into 'favourable' and 'unfavourable' cardiovascular outcome groups based on variables identified in linked administrative data including; total time in intensive care, total length of stay in hospital, and mechanical ventilation time up to 5 years following the date of the first cardiac surgery. A blind medical record audit of 200 randomly chosen children from 'favourable' and 'unfavourable' outcome groups was performed to validate the outcome groups. ResultsOf the 1872 children in the dataset that linked to hospital or death data, 483 were identified with a `favourable' cardiovascular outcome and 484 were identified as having a 'unfavourable' cardiovascular outcome. The medical record audit found concordant outcome groups for 182/192 records (95%) compared to the outcome groups categorized using the linked data. Conclusions The linkage of a curated biobank dataset with routinely collected administrative data is a reliable method to identify outcomes to facilitate a large-scale study to examine genetic variance. These genetic hallmarks could be used to identify patients who are at risk of unfavourable cardiovascular outcomes, to inform strategies for prevention and changes in clinical care.
Preprint
Full-text available
Background Brain magnetic resonance imaging (MRI) of infants with congenital heart disease (CHD) shows brain immaturity assessed via a cortical-based semi-quantitative score. Our primary aim was to develop an infant paralimbic-related subcortical-based semi-quantitative dysmaturation score, a brain dysplasia score (BDS), to detect abnormalities in CHD infants and predict clinical outcomes. Our secondary aim was to validate our BDS in a preclinical mouse model of hypoplastic left heart syndrome. Methods A paralimbic-related subcortical BDS, derived from structural MRIs of infants with CHD, was correlated with clinical risk factors, regional cerebral volumes, feeding and 18-month neurodevelopmental outcomes. The BDS was validated in a known CHD mouse model named Ohia with two disease-causing genes, Sap130 and Pchda9. To relate clinical findings, RNA-Seq was completed on Ohia animals. Findings BDS showed high incidence of paralimbic-related subcortical abnormalities (including olfactory, cerebellar, and hippocampal abnormalities) in CHD infants (n=215) compared to healthy controls (n=92). BDS correlated with reduced cortical maturation, developmental delay, poor language and feeding outcomes, and increased length of stay. Ohia animals (n=63) showed similar BDS findings, and RNA-Seq analysis showed altered neurodevelopmental and feeding pathways. Sap130 mutants correlated with a more severe BDS whereas Pcdha9 correlated with a milder phenotype. Interpretation Our BDS is sensitive to dysmaturational differences between CHD and healthy controls, and predictive of poor outcomes. A similar spectrum of paralimbic-related subcortical abnormalities exists between human and Ohia mutants suggesting a common genetic mechanistic etiology.
Chapter
The Fontan operation is a palliative procedure associated with a high risk of cardiac and extracardiac complications. Extracardiac complications, Fontan-associated liver disease and lymphatic dysfunction in particular, may be severe and the major determinant of clinical outcome. This chapter provides an overview of extracardiac complications of the Fontan operation and their management.
Article
Full-text available
Objective: We tested the hypothesis that chronic fetal hypoxia, at a severity present in many types of congenital heart disease, would lead to abnormal neurodevelopment. Methods: Eight mid-gestation fetal sheep were cannulated onto a pumpless extracorporeal oxygenator via the umbilical vessels and supported in a fluid-filled environment for 22 ± 2 days under normoxic or hypoxic conditions. Total parenteral nutrition was provided. Control fetuses (n = 7) were harvested at gestational age 133 ± 4 days. At necropsy, brains were fixed for histopathology. Neurons were quantified in white matter tracts, and the thickness of the external granular layer of the cerebellum was measured to assess neuronal migration. Capillary density and myelination were quantified in white matter. Data were analyzed with unpaired Student t tests or 1-way analysis of variance, as appropriate. Results: Oxygen delivery was reduced in hypoxic fetuses (15.6 ± 1.8 mL/kg/min vs 24.3 ± 2.3 mL/kg/min, P < .01), but umbilical blood flow and caloric delivery were not different between the 2 groups. Compared with normoxic and control animals, hypoxic fetuses had reduced neuronal density and increased external granular layer thickness. Compared with normoxic and control animals, hypoxic fetuses had increased capillary density in white matter. Cortical myelin integrity score was lower in the hypoxic group compared with normoxic and control animals. There was a significant negative correlation between myelin integrity and capillary density. Conclusions: Chronic fetal hypoxia leads to white matter hyper-vascularity, decreased neuronal density, and impaired myelination, similar to the neuropathologic findings observed in children with congenital heart disease. These findings support the hypothesis that fetal hypoxia, even in the setting of normal caloric delivery, impairs neurodevelopment.
Article
Full-text available
The purpose of the study was to evaluate school-age developmental and educational outcomes for children with and without a cardiac procedure in the first year of life to improve understanding of longer-term neurodevelopmental outcomes in children who have had a cardiac procedure for congenital heart disease, the most common serious congenital anomaly. A population-based cohort study using record linkage of state-wide data was undertaken, evaluating children born in New South Wales, Australia, 2001–2007. Those with and without a cardiac procedure in the first year of life with a linked developmental (Australian version Early Development Instrument testing result, age 4–6 years) and/or educational outcome (Australian National Assessment Program result, age 7–9 years) were included. Perinatal, perioperative and sociodemographic factors were examined using multivariable logistic regression models. Of 468,329 eligible children, 768 had a cardiac procedure in the first year of life and 582 were included. For those with a cardiac procedure and developmental outcome (n = 260), 13.1% were classified as having ‘special needs’ compared to 4.4% without a cardiac procedure. Of those with an educational outcome, after adjusting for perinatal, perioperative and demographic variables, children with a cardiac procedure (n = 396) were twice as likely to score below National Minimum Standard in school literacy and numeracy tests compared to their peers. Significant predictors included low birthweight, parent not completing school and having > 4 re-hospitalisations in their first six years. The developmental and educational trajectory of children who have had a cardiac procedure in their first year remains altered into primary school years. While perioperative factors did not impact outcomes, ongoing health and sociodemographic factors were important in identifying those children at greatest risk.
Article
Full-text available
Congenital heart disease (CHD) affects up to 1% of live births. However, a genetic diagnosis is not made in most cases. The purpose of this study was to assess the outcomes of genome sequencing (GS) of a heterogeneous cohort of CHD patients. Ninety-seven families with probands born with CHD requiring surgical correction were recruited for genome sequencing. At minimum, a proband–parents trio was sequenced per family. GS data were analyzed via a two-tiered method: application of a high-confidence gene screen (hcCHD), and comprehensive analysis. Identified variants were assessed for pathogenicity using the American College of Medical Genetics and Genomics–Association for Molecular Pathology (ACMG-AMP) guidelines. Clinically relevant genetic variants in known and emerging CHD genes were identified. The hcCHD screen identified a clinically actionable variant in 22% of families. Subsequent comprehensive analysis identified a clinically actionable variant in an additional 9% of families in genes with recent disease associations. Overall, this two-tiered approach provided a clinically relevant variant for 31% of families. Interrogating GS data using our two-tiered method allowed identification of variants with high clinical utility in a third of our heterogeneous cohort. However, association of emerging genes with CHD etiology, and development of novel technologies for variant assessment and interpretation, will increase diagnostic yield during future reassessment of our GS data.
Article
Full-text available
Large-scale phenotyping efforts have demonstrated that approximately 25–30% of mouse gene knockouts cause intrauterine lethality. Analysis of these mutants has largely focused on the embryo and not the placenta, despite the crucial role of this extraembryonic organ for developmental progression. Here we screened 103 embryonic lethal and sub-viable mouse knockout lines from the Deciphering the Mechanisms of Developmental Disorders program for placental phenotypes. We found that 68% of knockout lines that are lethal at or after mid-gestation exhibited placental dysmorphologies. Early lethality (embryonic days 9.5–14.5) is almost always associated with severe placental malformations. Placental defects correlate strongly with abnormal brain, heart and vascular development. Analysis of mutant trophoblast stem cells and conditional knockouts suggests that a considerable number of factors that cause embryonic lethality when ablated have primary gene function in trophoblast cells. Our data highlight the hugely under-appreciated importance of placental defects in contributing to abnormal embryo development and suggest key molecular nodes that govern placenta formation.
Article
Full-text available
Congenital heart defects (CHD) occur in ∼1 in every 100 live births. In addition, an estimated 10% of fetal loss is due to severe forms of CHD. This makes heart defects the most frequently occurring birth defect and single cause of in utero fatality in humans. There is considerable evidence that CHD is heritable, indicating a strong contribution from genetic risk factors. There are also known external environmental exposures that are significantly associated with risk for CHD. Hence, the majority of CHD cases have long been considered to be multifactorial, or generally caused by the confluence of several risk factors potentially from genetic, epigenetic, and environmental sources. Consequently, a specific cause can be very difficult to ascertain, although patterns of associations are very important to prevention. While highly protective of the fetus, the in utero environment is not immune to insult. As the conduit between the mother and fetus, the placenta plays an essential role in maintaining fetal health. Since it is not a fully-formed organ at the onset of pregnancy, the development of the placenta must keep pace with the growth of the fetus in order to fulfill its critical role during pregnancy. In fact, the placenta and the fetal heart actually develop in parallel, a phenomenon known as the placenta–heart axis. This leaves the developing heart particularly vulnerable to early placental insufficiency. Both organs share several developmental pathways, so they also share a common vulnerability to genetic defects. In this article we explore the coordinated development of the placenta and fetal heart and the implications for placental involvement in the etiology and pathogenesis of CHD.
Article
Full-text available
The placenta is a complex organ that influences prenatal growth and development, and through fetal programming impacts postnatal health and well-being lifelong. Little information exists on placental pathology in the presence of congenital heart disease (CHD). Our objective is to characterize the placenta in CHD and investigate for distinctions based on type of malformation present. Placental pathology from singleton neonates prenatally diagnosed and delivered at > 37 weeks gestation was analyzed. Placental findings of absolute weight, placental weight-to-newborn birth weight ratio, chorangiosis, villus maturity, thrombosis, and infarction were recorded and analyzed based on four physiological categories of CHD: (1) single ventricle-aortic obstruction, (2) single ventricle-pulmonic obstruction, (3) two-ventricle anomalies, and (4) transposition of the great arteries (TGA). Associations between fetal Doppler assessments of middle cerebral/umbilical arterial flow and placental findings were investigated. A total of 120 cases of complex CHD were analyzed. Overall placental-to-birth weight ratios were < 10th percentile for 77% and < 3rd percentile for 49% with abnormalities of chorangiosis (18%), hypomature villi (15%), thrombosis (41%), and infarction (17%) common. There was no association between fetal Doppler flow measures and placental abnormalities. Newborns with TGA had the greatest degree of placental abnormality. Placentas of newborns with CHD are smaller than expected and manifest a number of vascular abnormalities, with TGA most prominent. Fetal Doppler does not correlate with these abnormalities. Studies investigating the relationship between placental abnormalities and postnatal outcomes may offer insight into the fetal origins of outcome variability in CHD.
Article
Objectives: To assess health-related quality of life (HRQOL) in families of young children with complex congenital heart disease (CHD), and identify the demographic, clinical, and psychosocial factors that place these children and their mothers at greater risk of vulnerability. Study design: This cross-sectional study took place from June 2015 to October 2016 at The Sydney Children's Hospitals Network Cardiac Service, Australia. Mothers of a child aged 1-5 years with either single ventricle CHD or CHD requiring neonatal biventricular repair were invited to participate. Eighty-seven mothers completed a suite of validated measures, including the Pediatric Quality of Life Inventory, which assessed the outcomes of child and maternal HRQOL. Results: Sixty percent of children with single ventricle CHD and 25% of children with biventricular repair had total Pediatric Quality of Life Inventory scores within the at-risk range. Lower child HRQOL was strongly associated with single ventricle CHD (β = -0.38; P < .001), physical comorbidity (β = -0.32; P = .001), feeding difficulties (β = -0.26; P = .008), and greater maternal psychological stress (β = -0.18; P = .045), accounting for 52% of the variance in child HRQOL. Lower maternal HRQOL was strongly associated with poorer family functioning (β = 0.61; P < .001), greater maternal psychological stress (β = -0.23; P = .004), child physical comorbidity (β = -0.17; P = .01), and a 'difficult' child temperament (β = -0.14; P = .01), accounting for 73% of the variance in maternal HRQOL. Conclusions: Lower HRQOL is common in young children with complex CHD, particularly single ventricle CHD. Several predictors of HRQOL are potentially modifiable, offering possible pathways for prevention and early intervention. Routine screening is a necessary first step toward developing models of care to improve HRQOL in this population.
Article
Background: Neonates with congenital heart disease (CHD) are at high risk of punctate white matter injury (WMI) and impaired brain development. We hypothesized that WMI in CHD neonates occurs in a characteristic distribution that shares topology with preterm WMI and that lower birth gestational age (GA) is associated with larger WMI volume. Objective: (1) To quantitatively assess the volume and location of WMI in CHD neonates across three centres. (2) To compare the volume and spatial distribution of WMI between term CHD neonates and preterm neonates using lesion mapping. Methods: In 216 term born CHD neonates from three prospective cohorts (mean birth GA: 39 weeks), WMI was identified in 86 neonates (UBC: 29; UCSF: 43; UCZ: 14) on pre- and/or post-operative T1 weighted MRI. WMI was manually segmented and volumes were calculated. A standard brain template was generated. Probabilistic WMI maps (total, pre- and post-operative) were developed in this common space. Using these maps, WMI in the term CHD neonates was compared with that in preterm neonates: 58 at early-in-life (mean postmenstrual age at scan 32.2 weeks); 41 at term-equivalent age (mean postmenstrual age at scan 40.1 weeks). Results: The total WMI volumes of CHD neonates across centres did not differ (p = 0.068): UBC (median = 84.6 mm3, IQR = 26-174.7 mm3); UCSF (median = 104 mm3, IQR = 44-243 mm3); UCZ (median = 121 mm3, IQR = 68-200.8 mm3). The spatial distribution of WMI in CHD neonates showed strong concordance across centres with predilection for anterior and posterior rather than central lesions. Predominance of anterior lesions was apparent on the post-operative WMI map relative to the pre-operative map. Lower GA at birth predicted an increasing volume of WMI across the full cohort (41.1 mm3 increase of WMI per week decrease in gestational age; 95% CI 11.5-70.8; p = 0.007), when accounting for centre and heart lesion. While WMI in term CHD and preterm neonates occurs most commonly in the intermediate zone/outer subventricular zone there is a paucity of central lesions in the CHD neonates relative to preterms. Conclusions: WMI in term neonates with CHD occurs in a characteristic topology. The spatial distribution of WMI in term neonates with CHD reflects the expected maturation of pre-oligodendrocytes such that the central regions are less vulnerable than in the preterm neonates.
Article
Background Up to 20% of children with congenital heart disease (CHD) undergoing cardiac surgery develop neurodevelopmental disabilities (NDD), with some studies reporting persistent impairment. Recent large-scale studies have demonstrated shared genetic mechanisms contributing to CHD and NDD. In this study, a targeted approach was applied to assess direct clinical applicability of this information. Methods A gene panel comprising 148 known CHD and/or NDD genes was used to sequence 15 patients with CHD + NDD, 15 patients with CHD, and 15 healthy controls. The number and types of variants between the 3 groups were compared using Poisson log-linear regression, and the SNP-set (Sequence) Kernel Association Test-Optimized was used to conduct single-gene and gene-pathway burden analyses. Results A significant increase in rare (minor allele frequency < 0.01) and novel variants was identified between the CHD + NDD cohort and controls, P < .001 and P = .001, respectively. There was also a significant increase in rare variants in the CHD cohort compared with controls (P = .04). Rare variant burden analyses implicated pathways associated with “neurotransmitters,” “axon guidance,” and those incorporating “RASopathy” genes in the development of NDD in CHD patients. Conclusions These findings suggest that an increase in novel and rare variants in known CHD and/or NDD genes is associated with the development of NDD in patients with CHD. Furthermore, burden analyses point toward rare variant burden specifically in pathways related to brain development and function as contributors to NDD. Although promising variants and pathways were identified, further research, utilizing whole-genome approaches, is required prior to demonstrating clinical utility in this patient group.
Article
Aim To assess the impact of perioperative neonatal brain injury and brain volumes on neurodevelopment throughout school‐age children with critical congenital heart disease (CHD). Method Thirty‐four survivors of neonatal cardiac surgery (seven females, 27 males) were included. Neonatal preoperative and postoperative cerebral magnetic resonance imaging was performed and neurodevelopment was assessed at 24 months (SD 0.7, n=32, using Bayley Score of Infant and Toddler Development, Child Behavior Checklist) and 6 years (mean age 5y 11mo; SD 0.3, n=30, using Movement Assessment Battery for Children, Wechsler Preschool and Primary Scale of Intelligence, Child Behavior Checklist, Teacher Report Form). Brain injury, brain volumes, and cortical measures were related to outcome with adjustment for maternal educational level. Results Two‐year cognitive score and 6‐year Full‐scale IQ were poorer in children with neonatal white matter injury (n=21, all p<0.05), with higher teacher‐reported attention problems (p=0.03). Five of six children with involvement of the posterior limb of the internal capsule showed motor problems (p=0.03). Children with a below‐average Fulll‐scale IQ (<85, n=9) showed smaller volumes of basal ganglia thalami (−8%, p=0.03) and brain stem (−7%, p=0.03). Interpretation Our findings provide evidence of unfavourable outcome in school‐age children with critical CHD who acquire perioperative neonatal brain injury. What this paper adds • This paper extends knowledge about neonatal brain injury and long‐term outcome in congenital heart disease. • Children with white matter injury show lower IQ and more attention problems at school age. • Injury of the posterior limb of the internal capsule increases the risk of motor problems. • This study provides evidence for worse outcomes in neonates acquiring brain injury around cardiac surgery.