Sunteți pe pagina 1din 17

Pediatric Neurology 63 (2016) 6e22

Contents lists available at ScienceDirect

Pediatric Neurology
journal homepage: www.elsevier.com/locate/pnu

Topical Review

Applying the Lessons of Tuberous Sclerosis: The 2015 Hower


Award Lecture
E. Steve Roach MD *
Division of Child Neurology, Ohio State University College of Medicine, Nationwide Childrens Hospital, Columbus, Ohio

abstract

Tuberous sclerosis complex is a dominantly inherited disorder that variably affects the brain, skin, kidneys, heart,
and other organs. Its neurological manifestations include epilepsy, autism, cognitive and behavioral dysfunction,
and giant cell tumors. A mutation of either TSC1 or TSC2 can cause tuberous sclerosis complex. Their two gene
products, hamartin and tuberin, form a physical complex which normally inhibits protein synthesis mediated
through the mechanistic target of rapamycin, so a TSC1 or TSC2 mutation results in overactivation of the mech-
anistic target of rapamycin cascade. In addition to their tumor suppressor roles, TSC1 and TSC2 help to regulate cell
size, neuronal migration, axon formation, and synaptic plasticity. Clinical trials of two different the mechanistic
target of rapamycin inhibitors have demonstrated substantial improvement of tuberous sclerosis complexerelated
tumors, and a recent trial also showed a benet from the mechanistic target of rapamycin inhibitor everolimus in
the treatment of refractory epilepsy due to tuberous sclerosis complex. Effective mechanism-based therapy is now
available for some manifestations of tuberous sclerosis complex.

Keywords: tuberous sclerosis complex, mTOR, rapamycin, everolimus, history, subependymal giant cell astrocytoma, epilepsy,
history
Pediatr Neurol 2016; 63: 6-22
2016 The Author. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://
creativecommons.org/licenses/by-nc-nd/4.0/).

Promise me youll always remember: youre braver than the individuals with TSC have a spontaneous mutation.
you believe, and stronger than you seem, and smarter than Because of its striking variability of clinical expression, the
you think. diagnosis of TSC is occasionally difcult, especially in
younger individuals or in those with subtle ndings.1 Two
A. A. Milne
genes are responsible for tuberous sclerosis: TSC1 on
Winnie-the-Pooh
chromosome 9q34 (which encodes the protein hamartin)
and TSC2 on chromosome 16p13.3 (which encodes the
Introduction protein tuberin). Effective treatment is now available for
some of the manifestations of TSC, therapy that specically
Tuberous sclerosis complex (TSC) is a dominantly targets the molecular dysfunction caused by TSC mutations.
inherited disorder of cellular differentiation and prolifera- My own involvement with TSC began with a presenta-
tion which variably affects the brain, skin, kidneys, heart, tion at the 1986 annual Child Neurology Society meeting in
and other organs. The neurological manifestations of TSC Boston. By the time of this presentation, TSC had already
include epilepsy, autism, cognitive and behavioral been known to physicians for 124 years. Yet much of what
dysfunction, and giant cell tumors. Up to three fourths of we now know about this often devastating disease has been
discovered in the three decades since 1986. That such
Article History: stunning progress can be made in the course of one gen-
Received June 9, 2016; Accepted in nal form July 7, 2016 eration of physicians provides hope that TSC can serve as a
* Communications should be addressed to: Dr. Roach; Division of prototype for the development of mechanism-based ther-
Child Neurology; Nationwide Childrens Hospital; 700 Childrens Drive;
Columbus, Ohio 43245.
apy for other genetic diseases. Here I provide a historical
E-mail addresses: esroach@earthlink.net, roache@nationwidechildrens.org perspective on some of the things we have learned about

0887-8994/ 2016 The Author. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
http://dx.doi.org/10.1016/j.pediatrneurol.2016.07.003
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 7

TSC in the last three decades. There are lessons from the angiobromas (Fig 3) with TSC. During the next few years,
progress we have made with TSC that can be applied to Kothe described the ungual bromas of TSC, and Campbell7
other serious genetic diseases. described the retinal TSC lesions (Fig 4).
In 1908, Heinrich Vogt9 proposed quasi-diagnostic
criteria for TSC, a triad consisting of epilepsy, idiocy, and
The early history of TSC
adenoma sebaceum. Because facial angiobromas (ade-
noma sebaceum) have a high level of specicity for TSC,
The false sense of security afforded by misinformation or
most individuals who manifest all three features of Vogts
misinterpretation of facts is far worse than simple
triad will in fact have TSC, but a clinician using Vogts triad
ignorance.
would likely fail to diagnose half the TSC patients that we
Tuberous sclerosis complex was rst described by von now recognize.
Recklinghausen (Fig 1) at a March 25, 1862, meeting of the Hypomelanotic macules (ash leaf spots) occur in over
Berlin Obstetrical Society.2 He presented a newborn baby 90% of the individuals with TSC, so it is surprising that the
who died after a few breaths because of multiple cardiac association of these lesions with TSC went unrecognized
myomata. The babys brain contained numerous sclerotic until a 1932 report by Critchley and Earl.10 Over three de-
areas, although von Recklinghausen did not elaborate. cades later, Gold and Freeman11 along with Fitzpatrick
Although von Recklinghausens patient almost certainly et al.12 nally established hypomelanotic macules (Fig 3) as
had TSC, his appreciation of its clinical manifestations was an important early sign of TSC.
clearly rudimentary.
Eighteen years later, in 1880, Bourneville3 (Fig 2) added
important clinical neurological details. He characterized the Chasing the tuberous sclerosis phenotypes
brain pathology as sclrose tubreuse des circonvolution
crbrales, thus coining the name we still use for the dis- No matter how well-accepted an idea it may be, without
order. Nevertheless, Bournevilles understanding of TSC was evidence, dogma is still just dogma.
initially limited. He noted his patients facial angiobromas The clinical features of TSC are highly variable, and even
but concluded that they were coincidental. Bourneville and affected members of the same family often manifest widely
Brissaud4 later would tie renal tumors to TSC. differing clinical ndings. The resulting clinical manifesta-
The genetic nature of TSC was quickly recognized. In tions often represent a substantial nancial and psychoso-
1885, only ve years after the report by Bourneville, Balzer cial burden to the patient and family.13 Not unexpectedly for
and Menetrier5 described TSC in a mother and daughter. a complicated disorder such as TSC, over the years, there
They were also the rst to connect the characteristic facial have been points of controversy.

Cutaneous lesions

Skin ndings of TSC include hypomelanotic macules,


ungual or periungual bromas, a shagreen patch, and facial
angiobromas (Fig 3). Hypomelanotic macules occur in over
90% of individuals with TSC. They are often difcult to see in
light-skinned newborns without the aid of an ultraviolet
light. Facial angiobromas occur in about three fourths of
patients and typically start to appear during the preschool
years. A shagreen patch occurs in 20% to 30% of TSC patients,
typically on the lower back or ank area. It is an irregularly
shaped, slightly raised or textured skin lesion. Ungual -
bromas are eshy lesions that arise adjacent to or from
underneath the nails. Single bromas occasionally develop
after trauma. Sometimes the broma itself is not visible but
creates a prominent longitudinal groove in the ngernail, a
nding which also has diagnostic signicance (Fig 3).

Retinal lesions

Retinal lesions occur in up to 87% of people with TSC.14


Varied retinal abnormalities include the classic mulberry
lesion adjacent to the optic disc, abnormalities of the retinal
pigment layer, plaque-like hamartomas, and depigmented
areas in the retina (Fig 4). Most retinal lesions are clinically
inconsequential, but occasional patients have visual
FIGURE 1. impairment due to the occurrence of a large macular lesion,
Friedrich Daniel von Recklinghausen, 1833 to 1910. a retinal detachment, or a vitreous hemorrhage.
8 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

FIGURE 2.
(A) Dsir-Magloire Bourneville, 1840 to 1909. (B) Bournevilles illustration of his patients brain tubers.3

Cardiac lesions of blood ow by an intraluminal tumor or to inadequate


normal myocardium to maintain perfusion. A few children
Two thirds to three fourths of newborns with TSC have later develop cardiac arrhythmias.
one or more cardiac rhabdomyomas on echocardiography,
but most of these lesions remain asymptomatic.15-17 Rhab- Renal lesions
domyomas are often multiple (Fig 5) and are commonly
evident during prenatal ultrasounds.18 These lesions do not Renal angiomyolipomas (AMLs) are demonstrable in
grow and typically shrink during the rst few months after 75% to 80% of TSC patients by 10 years of age.19 Multiple
birth. Most individuals with cardiac dysfunction present tumors are typical (Fig 6A), and lesions larger than 4 cm
soon after birth with heart failure due either to obstruction are more likely to become symptomatic than smaller

FIGURE 3.
The cutaneous lesions of tuberous sclerosis complex include (A) facial angiobromas (adenoma sebaceum), (B) ungual broma, (C) hypomelanotic
macules (ash leaf spots), and (D) a shagreen patch. Parts C and D are reproduced with permission from Roach.6
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 9

FIGURE 4.
(A) Retinal astrocytic hamartoma in an individual with tuberous sclerosis complex (TSC). (B) Retinal pigmentary lesions in an adult with TSC. Part A is
reproduced with permission from Roach.8

tumors. Most of these lesions represent histologically Neurological dysfunction


benign tumors with varying amounts of vascular tissue,
fat, and smooth muscle (Fig 6B). Renal cell carcinoma or The principal neurological manifestations of TSC are
other malignancies occur in a small number of TSC pa- intellectual disability, epilepsy, autism, and various
tients, and it can be difcult to noninvasively distinguish a behavioral abnormalities. Various types of seizures occur
malignant kidney tumor from an AML that contains little in over 90% of patients, and infantile spasms are particu-
fat. Single or multiple renal cysts are also a feature of TSC, larly common in younger individuals.21 Vigabatrin may be
and about 1% of the individuals with TSC also exhibit the more effective for infantile spasms due to TSC than those
severe phenotype of polycystic kidney disease (PKD). Iso- arising from other causes, so it is typically administered
lated renal cysts may disappear on repeated imaging. instead of corticosteroids or adrenocorticotrophic hor-
mone in these children.22-24 Although the seizures may be
refractory to therapy, seizure control with medication is
Pulmonary dysfunction often possible, and medication can even be halted in some
individuals without the return of seizures.25 Surgical
The classic pulmonary lesion of TSC is lymphangioleio- removal of the epileptogenic cortical lesion is an option for
myomatosis (LAM). Other patients exhibit multifocal some individuals.26,27 Hemispherectomy is usually neces-
micronodular pneumocytic hyperplasia. Manifestations sary for the children with hemimegalencephaly related to
include spontaneous pneumothorax, dyspnea on exertion, TSC,28 and corpus callosotomy can be helpful in children
and frank respiratory failure. Pulmonary disease is about with atonic seizures or frequent rapid secondary general-
ve times more common in females than in males, and it is ization of the seizures.
uncommon to develop pulmonary symptoms before Almost all individuals with marked cognitive impair-
adulthood. ment have epilepsy, but many patients who have epilepsy
are normally intelligent. Children with infantile spasms are
more likely to exhibit long-term cognitive impairment.29
Cognition ranges from normal to severe impairment, but
close to half of the patients are cognitively intact. Autism
spectrum disorder occurs in 25% to 50% TSC patients, and
attention decit disorder, aggressiveness, obsessive-
compulsive behavior, and even frank psychosis are com-
mon, often in conjunction with epilepsy or intellectual
decit.
The subependymal nodules which characterize TSC
typically arise adjacent to the lateral ventricular wall and
protrude into the ventricles (Fig 7).1 When calcied, these
lesions are easily conrmed with computed tomography
(Fig 8). Even uncalcied subependymal nodules are readily
apparent on magnetic resonance imaging (MRI), and the
cortical tubers are far more obvious with MRI than with
computed tomography. Evidence of abnormal neuronal
FIGURE 5. migration can be seen in some patients as a high signal
Prenatal ultrasound showing multiple echogenic cardiac rhabdomyomas in linear lesions running perpendicular to the cortex on T2-
a baby who later proved to have tuberous sclerosis complex. weighted MRI (Fig 9).
10 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

FIGURE 6.
(A) Longitudinally sectioned kidney reveals a large fat-laden angiomyolipoma (AML) in the lower pole. Several small AMLs are also evident in the renal
cortex. (B) Renal AMLs contain vascular, smooth muscle, and fat moieties (hematoxylin and eosin at 110). Part A was reproduced with permission from
Weiner et al.20

Depending on whether the diagnosis is rst made clini- lesions typically arise in the anterior portion of the lateral
cally or radiographically, subependymal giant cell tumors ventricles (Fig 9) and typically develop in children rather
(SEGAs) occur in 6% to 14% of individuals with TSC. These than in adults.31 Unlike the more common cortical tubers,

FIGURE 7.
(A) Brain section shows a cortical tuber in the left frontal region. (B) Subependymal nodules protruding into the ventricles (arrow). Reproduced with
permission from Roach.8
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 11

easily imaged, in effect ltering out all but the larger cortical
lesions.34 It is unlikely that a study using modern imaging
equipment that focused solely on the number of lesions
without regard to their size would show such a robust
correlation. Finally, recent studies suggest that the brain
lesions and the brains white matter connections may
evolve over time.36,37

Is surveillance testing worthwhile?

Another area of early debate centered on the need for


surveillance testing in an effort to identify TSC complications
at an earlier stage. Some physicians advocated an annual
complete assessment, whereas others preferred to deal with
any complications as they arose. Given the variety of
possible clinical manifestations, a complete annual evalua-
tion seemed burdensome to the families and often wasted
health care resources, but waiting on the complications to
occur could make them more difcult to address. At the
1998 consensus conference in Annapolis, Maryland, we
developed a systematic approach to periodic screening for
complications, which was based loosely on then existing
public health screening recommendations.38 To justify
screening, a complication should pose a signicant risk,
there should be a noninvasive means of evaluating it, and
earlier diagnosis should convey a substantial management
FIGURE 8. benet. We concluded that the lesions that met these three
Computed tomography reveals typical calcied subependymal nodules requirements were renal AMLs, especially in postadolescent
protruding into the lateral ventricles. Note also a large calcied cortical patients, and cerebral giant cell tumors before adulthood.
lesion in the left posterior hemisphere (black arrow) and scattered cortical
Each of these lesions is treatable and is easier to manage if
hypodense cortical tubers (white arrows). Reproduced with permission
from Roach et al.30
identied when smaller and asymptomatic, so the surveil-
lance guidelines focused on periodic imaging of the brain
SEGAs often enlarge and cause symptoms such as increased and kidneys.38 The latest revision of these guidelines
intracranial pressure, new focal neurological decits, or retained a similar philosophy but added periodic screening
deterioration of seizure control.31,32 Surgical resection of an for neuropsychiatric dysfunction,39,40 shifted the renal im-
expanding SEGA can be effective especially when the tumor aging modality from ultrasound to MRI, and added pulmo-
is not extensive, but increasingly, these lesions are managed nary screening of adult women.41
medically.31,33

Developing family partnerships


The lesion burden conundrum

Recognize the power of an effective partnership between


We suggested in 1987 that individuals with numerous
families and clinicians and researchers.
brain lesions on MRI were more likely to experience
intractable epilepsy and severe cognitive impairment.34 Real heroes are ordinary people who do extraordinary
This seemingly common sense observation was unexpect- things. Adrianne Cohen, Verna Morris, Linda Hamm, and
edly controversial, because until then, severe intellectual Susan Diaz, each a mother of a child affected by TSC, foun-
impairment in individuals with TSC had been attributed ded the National Tuberous Sclerosis Association (NTSA) in
solely to their uncontrolled seizures. A meta-analysis of this 1974. Dissatised by the lack of understanding of their
and other studies conrmed that the cortical tuber count childrens disease and the paucity of ongoing research,
constituted a biomarker that predicted an increased likeli- these four women gathered for coffee one morning in one of
hood of severe neurological impairment.35 their kitchens, and from this meeting grew NTSA.
The effect of the cortical lesion number on the severity of NTSA was renamed the Tuberous Sclerosis Alliance in
neurological dysfunction is likely to be far more nuanced 2000. The Tuberous Sclerosis Alliance has grown into a
than these early results suggest. The presence of numerous particularly effective support organization, providing in-
brain lesions (Fig 10), for example, does not mean that formation for professionals and families, supporting clinical
frequent seizures do not also contribute to a poor outcome. and basic research, and supporting patients with TSC and
Although the early analyses focused on the number of their families. The organization has provided start-up
cortical lesions, the size and location of the brain lesions are funding for basic and clinical research and training grants
likely to play a greater role than the number. Our 1987 study that launch new investigators. It supports research and
used a prototype .15T MRI unit; it had limited image reso- consensus conferences and press government ofcials for
lution and did not identify the smaller lesions that are now more research funding for TSC.
12 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

FIGURE 9.
(A) T1-weighted magnetic resonance imaging (MRI) shows subependymal nodules in a child with tuberous sclerosis complex. (B) Cranial MRI reveals radial
migration lines positioned between the subependymal area and the cortex. (C) MRI with gadolinium contrast shows the typical appearance and location
of a subependymal giant cell astrocytoma.

The Gomez textbook The Gomez book documented the phenotypic variability
of TSC and demonstrated the existence of milder TSC phe-
A well-researched book or seminal paper can dene a eld notypes. The recognition of TSCs diverse and milder phe-
of study, introduce new lines of thought, and ignite notypes heralded a several-fold increase in the prevalence
progress. of TSC, now estimated to be about 1 in 6000 to 1 in 10,000
A turning point in our understanding of TSC occurred in individuals.42,43
1979 with the publication of the rst edition of Tuberous
Diagnostic criteria for tuberous sclerosis
Sclerosis, edited by Manuel R. Manny Gomez (Fig 11).2 His
book was the rst comprehensive source of information
Diagnostic criteria represent only an operational denition
about TSC, with contributions from neurologists, cardiolo-
of a condition pending more information and insight.
gists, nephrologists, radiologists, dermatologists, ophthal-
mologists, and pathologists. For the rst time, there was one Aside from the 1908 Vogt triad, the rst TSC diagnostic
convenient source of information about all aspects of the criteria appeared in the Gomezs 1988 second edition of
disease and a broadening recognition of TSC manifestations Tuberous Sclerosis.44 The Gomez diagnostic criteria assumed
beyond a single discipline. that a number of its more suggestive clinical manifestations
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 13

FIGURE 10.
(A) T2-weighted magnetic resonance imaging (MRI) from a child with severe cognitive impairment shows numerous cortical and subcortical lesions. (B)
Note the paucity of hemispheric lesions on this MRI from another child with TSC who has normal cognitive function.

were pathognomonic of TSC, then tabulated the ndings to The criteria were revised at the 1998 consensus confer-
arrive at a denitive or a presumptive diagnosis. ence in Annapolis, Maryland.47 By this time, our under-
The Gomez diagnostic criteria provided a framework for standing of the clinical features of TSC had improved, and
a clinician to consider less-specic ndings when making a mutations could be identied at least in research settings,
diagnosis, and for typical patients with multiple classic making it easier to validate the diagnostic signicance of
features of TSC, the criteria were quite accurate. However, some TSC manifestations.47 The most recent iteration of TSC
there was no way to conrm the assumed pathognomonic diagnostic criteria resulted from the 2012 consensus con-
nature of the lesions, and indeed, some of the abnormalities ference in Washington, D.C.48 By then, commercially avail-
once considered specic for TSC are now known to occur in able mutational analysis was available, and the presence of
other settings. In addition, it was possible to reach a pre- a disease-causing mutation of TSC1 or TSC2 was accepted as
sumptive diagnosis of TSC via combination relatively the basis of a TSC diagnosis without regard to the clinical
nonspecic manifestations, such as the occurrence of a manifestations. It was also possible to further simplify the
gingival broma or dental pitting in an individual with an criteria. Members of this productive consensus group also
immediate relative with TSC or the co-occurrence of in- updated the recommended surveillance guidelines for TSC-
fantile spasms and myotonic, clonic, or atonic seizures.44 related complications and devised a standardized way to
The 1988 Gomez diagnostic criteria appeared the year assess individuals with TSC for the presence of neuropsy-
after TSC was linked to chromosome 9 by Fryer et al.45 and chological dysfunction.39-41
just before the effort to identify the second TSC gene. The
mistaken characterization of an unaffected individual as
On the trail of the TSC genes
affected can have a devastating effect on linkage analysis,
and, because of this concern, several of us were asked in
Clinical observations can expedite research and suggest
1992 to devise diagnostic criteria with a higher level of
new lines of inquiry. Pay attention!
specicity.46 Like Gomez, we had no way to ascertain the
true frequency or specicity of each TSC feature. To increase In 1987, Fryer et al.45 linked TSC to the distal long arm of
the level of specicity, we eliminated the notion of patho- chromosome 9 by analyzing ABO blood groups in 19 fam-
gnomonic TSC features altogether and structured the ilies affected by TSC. This report was among the rst to link
criteria to require at least two major features of TSC or one a specic disease to a specic noneX chromosome. It soon
major and two minor manifestations to characterize the became apparent that other multigenerational families
diagnosis as denitive. Somewhat controversially, we with TSC did not exhibit linkage to chromosome 9, and the
excluded epilepsy and cognitive impairment from the search for a second gene locus began.49,50
criteria altogether because both would almost always be At this time, it was not easy to establish gene linkage to a
associated with brain imaging abnormalities and these particular chromosome or to pinpoint the gene itself.
were already being tabulated.46 These criteria served their Identifying multigenerational families with TSC who were
initial purpose, but they were cumbersome for routine eager to participate in research was not easy. There were a
clinical use and likely left some individuals with TSC with limited number of established markers for each chromo-
only a probable diagnosis. some, and it could take months of painstaking work just to
14 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

testing probably represent somatic mosaicism,57 so it is


not surprising that individuals without an identied mu-
tation often exhibit a milder phenotype than those with a
dened mutation.58 Finally, germline mosaicism will not
be detected with standard mutational analysis. These in-
dividuals may have multiple offspring with TSC although
they have no clinical ndings to suggest TSC and despite
their lack of a disease-causing mutation on blood-based
testing.59

What do the TSC genes do?

The preservation of TSC1 and TSC2 across a broad range of


species was an early indication of their importance.
Plank et al. and others demonstrated in 1998 that tuberin
can be traced to the Golgi apparatus and that hamartin and
tuberin interact.60,61 Tuberin and hamartin form a protein
complex and work, probably explaining how a mutation on
either of the two genes could cause such a similar
phenotype.62,63
Rapamycin had been used clinically for several years
because of its broad antifungal and immunosuppressive
effects by the time it intersected with TSC research. Rapa-
mycin was named for Rapa Nui, the Easter Island whose soil
yielded rapamycins parent bacterium, Streptomyces hygro-
scopicus. Cafferkey et al.64 in 1993 identied two yeast
genes which, when mutated, eliminated rapamycin toxicity.
Subsequently, the proteins that arose from these two genes
were puried and characterized as the physical target of
rapamycin.65 Mammalian homologs of these proteins were
thus designated the mammalian targets of rapamycin
FIGURE 11.
(mTOR1 and mTOR2).66-68 In recognition of mTORs pres-
Manuel Rodriguez Gomez, 1928 to 2006.
ervation across a broad range of species and the entrenched
status of its abbreviation, mTOR was belatedly renamed the
mechanistic target of rapamycin.69,70
eliminate one chromosome. Raymond Kandt noted that An early indication of the importance of the TSC genes
occasional individuals with TSC experience polycystic kid- came in 2001 with the demonstration by Gao and Pan that
neys and that PKD had recently been linked to chromosome TSC1 mutations antagonize insulin signaling in
16. This bit of serendipity paid off handsomely, for Kandt Drosophila.71 It was subsequently demonstrated that the
et al.51 quickly found that TSC2 was not only linked to tuberinehamartin complex inhibits mTOR-mediated
chromosome 16, it was immediately adjacent to the PKD signaling. The mTOR proteins are serine/threonine protein
gene on chromosome 16. kinases that combine with several other proteins to form
Subsequent progress was rapid. In 1993 the TSC2 gene two large complexes (mTOR complex 1 and mTOR complex
was identied at 16p13.3,52 and, two years later, tuberin 2 or mTORC1 and mTORC2). Rapamycin inhibits mTOR by
was identied as its gene product.53 In 1997, ten years after interfering with the phosphorylation of the mTOR protein
its initial linkage to chromosome 9, TSC1 was nally iden- complexes.72 The hamartinetuberin complex normally
tied at 9q34, and hamartin was promptly identied as its dampens the distal mTOR pathway (Fig 12), so a TSC1 or
gene product.54 TSC2 mutation results in overactivation of the mTOR
Clinical gene testing for TSC1 and TSC2 became available cascade. mTORC1 phosphorylates and regulates several
in 2002. Conrmatory mutational analysis can help to downstream components of the cascade, including two
establish the TSC diagnosis in individuals with milder or well-studied proteins, namely ribosomal protein S6 kinase
atypical manifestations, and mutational analysis also aids 1 (S6K1) and eukaryotic initiation factor 4E-binding protein
genetic counseling. However, several factors limit the 1 (4E-BP1).73 These proteins, in turn, control several aspects
usefulness of mutational analysis for TSC.55 Most in- of translation and protein synthesis in response to changes
dividuals acquire TSC via a spontaneous mutation,56 and in nutrients, hormonal effects, and energy sufciency.74
mutational analysis is ordinarily not considered until after Given the frequency and the variety of tumors that occur
the diagnosis of the rst individual in the family. in individuals with TSC, the tumor suppressor role of TSC1
Commercially available mutational analysis fails to identify and TSC2 is fairly obvious. However, overactivation of the
the disease-causing mutation in about 10% to 15% of the mTOR pathway can also exert a dramatic effect on cell size,
individuals who meet the clinical diagnostic criteria for neuronal migration, axon formation, and synaptic plasticity.
TSC.57 Many of these individuals with unremarkable gene TSC-related neurological lesions result from impaired
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 15

FIGURE 12.
Schematic representation of the relationship of the tuberinehamartin complex to mTORC1 and mTORC2. Reproduced with permission from Tran and
Zupanc.21

neuronal migration along radial glial bers, abnormal the brain size remained normal, and the myelination
myelination, and from abnormal cell proliferation. Way remained close to normal (Fig 13).79
et al.75 created a mouse model with selective deletion of TSC1 and TSC2 are preserved across a broad range of
TSC2 from radial glial progenitor cells in the cerebral cortex. species and, via the mTOR pathway, inuence several
Reminiscent of human TSC, these mice developed postnatal important cellular processes.70 The pathway may play a role
megalencephaly, abnormal cerebral cortical and hippo- in the pathophysiology of such seemingly diverse condi-
campal lamination, heterotopias, abnormal myelination, tions as obesity, cancer, type 2 diabetes mellitus, aging, and
and enlarged dysplastic neurons and glia. There is also ev- neurodegenerative disorders. These topics are well beyond
idence that mTOR function inuences long-term synaptic the scope of this presentation, but detailed reviews of the
plasticity, which in turn could play a role in cognition and protean effects of the mTOR pathway are available.70
behavior that is distinct from these migrational and pro-
liferational aberrations.72,76 Impaired local protein synthe-
The rise of mTOR inhibitors
sis in axonal growth cones and synapses could represent a
unifying mechanism for these seemingly different
Repurposing an approved drug can save years of work and
processes.77
help patients much sooner than making a completely new
There are now several animal models of TSC,78 but mice
drug.
with prenatal loss of neuronal TSC1 by Meikle et al.79 was a
notable early contribution because the animals replicated The TSC1 and TSC2 mutations result in mTOR activa-
several of the clinical and neuropathological features of TSC tion, and rapamycin suppresses mTOR signaling. After TSC
in humans. These mice failed to thrive and experienced was shown to affect the mTOR pathway, clinicians soon
sometimes fatal seizures, typically dying between three began to study the already available mTOR inhibitor
weeks and two months of age. They exhibited enlarged rapamycin (sirolimus) in individuals with TSC. Clinical
dysplastic neurons, a larger than normal overall brain size, trials were also begun with everolimus, a rapamycin de-
and a striking lack of myelination. Remarkably, all these rivative that is said to act more selectively on mTOR
manifestations were mitigated by the early administration complex 1.21 For the rst time, targeted therapy based on
of an mTOR inhibitor. In the treated animals, seizures did an understanding of the molecular pathophysiology of
not occur, the median survival and weight gain normalized, TSC became a reality.80
16 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

FIGURE 13.
Improved myelination in TSC1 null-neuron mice in response to rapamycin. The top row depicts myelin stained samples from the retrosplenial granular
region of the cerebral cortex, whereas the bottom images are myelin-stained samples from the CA3 region of anterior hippocampus. Note the normal myelin
staining in a control animal (A, F) and in a control animal treated with rapamycin (E, J). Myelination is severely reduced in the untreated TSC1-decient
animal (B, G). Administration of rapamycin to the mutant mice dramatically improved myelination (C, H), and halting rapamycin after 30 postnatal days
resulted in improved but patchy myelination (D, I). Images 60; both scale bars equal 100 mm. Reproduced with permission from Meikle et al.79

Franz et al.81 initially treated ve TSC patients with oral hydrocephalus, or increased intracranial pressure.87 Over
rapamycin. The SEGA regressed in all ve of these in- half of the individuals who continued on therapy eventually
dividuals, exhibited transient regrowth after cessation of experienced a reduction in tumor size of more than 50%.88
therapy, and then shrank again upon reinstitution of rapa- Franz et al.89 conducted a randomized, placebo-
mycin (Fig 14). Interestingly, reduction of the tumor size in controlled trial of everolimus in 117 individuals with TSC
response to rapamycin may not be associated with an and at least one SEGA of more than one centimeter in
obvious histopathological change.82 diameter. Twenty-seven of the 78 individuals (35%) who
Bissler et al.83 conducted a 24-month, nonrandomized, received everolimus experienced a 50% or greater reduction
open-label trial of rapamycin (sirolimus) in individuals with in SEGA size versus none of the control group.89 With
TSC-related renal AMLs or LAM. Similar to the results of the extended follow-up, about half of the patients achieved a
earlier SEGA study by Franz et al., the renal AMLs regressed 50% reduction in tumor size.90 Most patients tolerate the
during rapamycin therapy (Fig 15A) but often grew again extended use of mTOR inhibitors well, but measuring
after therapy was halted.83 The spirometry measurements trough drug concentrations may be useful in some
also improved in the LAM patients, and the improvement individuals.91
persisted in some individuals (Fig 15B).83 Double-blind clinical trials of mTOR inhibitors are dif-
Subsequently, Bissler et al.84 conducted a placebo- cult to perform in individuals with TSC because of the drugs
controlled trial of the rapamycin derivative everolimus often striking effect on the facial angiobromas. Not sur-
(Fig 16) in 118 patients with either TSC or sporadic LAM who prisingly, a recently concluded multi-center clinical trial
had one or more renal AMLs that were more than 3 cm in demonstrated (Fig 17) that topically applied rapamycin
largest diameter. Thirty-three of the 79 (42%) patients who renders the facial angiobromas less prominent (Hope
received everolimus experienced a 50% or more reduction Northrup, personal communication September, 2015).
of the target AML volume.84 None of the lesions regressed in Another report suggests that topical everolimus is also
the 39 individuals who received placebo, and nine people in effective for these facial lesions.92
the control group discontinued study participation because Goyer et al.93 also successfully treated three neonates
of tumor growth.84 There was a similar robust response (i.e., with TSC-related tumors (two with a cardiac rhabdomyoma
more than 50% reduction of AML size in 80% of the patients) and one with a SEGA) with everolimus. All three neonates
among the younger patients receiving sirolimus in the SEGA seemed to benet from the medication and all tolerated it
clinical trial.86 well.
Krueger et al.87 conducted an open-label trial of ever-
olimus in 28 patients with TSC who exhibited increasing What about pre-emptive therapy of TSC?
SEGA size. All 28 individuals experienced a reduction in
SEGA size, and the tumor shrank by at least 30% in 21 pa- Most genetic disease therapies are aimed at secondary
tients (75%) and by 50% in nine individuals (32%). None of complications. Mechanism-based therapy may change that
the patients experienced new SEGA formation, worsening paradigm.
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 17

FIGURE 14.
These magnetic resonance images illustrate one patients subependymal giant cell tumor (SEGA) response to rapamycin. (A) Before starting therapy. (B)
After 2.5 months of therapy. (C) Follow-up imaging after stopping therapy for 4 months. (D) Eight months after resuming therapy, the SEGA is again smaller.
Images reproduced with permission from Franz, et al.81

Most children with TSC initially appear neurologically identifying individuals at high risk for seizures before the
normal, offering a potential opportunity to minimize or seizures actually begin.95
prevent some of the disorders complications. Would early Even more intriguing is the possibility of mitigating ep-
therapy prevent TSC neurological manifestations from ilepsy or cognitive dysfunction before their onset or at least
developing? The evidence is intriguing but far from soon after their appearance.94 The administration of rapa-
complete.80 mycin to TSC-decient mice appears to diminish seizures
Jozwiak et al.94 rst suggested that prophylactic admin- and behavioral abnormalities.80 Anecdotal reports from TSC
istration of an antiepileptic agent to infants at high risk for patient families suggest improvement of epilepsy or
epilepsy due to TSC before the onset of seizures greatly behavior after the administration of an mTOR inhibitor, and
improves their developmental outcome and reduces their some individuals in the sirolimus clinical trials reported a
likelihood of drug-resistant seizures. More recently, Wu substantial reduction in their seizure frequency.88,96
et al.95 assessed serial electroencephalography in 40 We had long assumed that the intractable epilepsy,
seizure-free children with TSC before seven months of age. cognitive impairment, and behavioral changes in in-
Two thirds of the children in this cohort subsequently dividuals with TSC resulted from long-standing multifocal
developed seizures, and just over half of them developed cortical dysplasia, aberrations of neuronal migration, and
infantile spasms. Seventy-four percent of the children who abnormal neuronal connections. This conjecture was
experienced seizures developed epileptiform discharges on bolstered by the apparent correlation between the brains
electroencephalography at an average of 1.9 months before lesion burden and the severity of the individuals epilepsy
the onset of seizures, demonstrating the feasibility of and cognitive impairment.34,35 However, substantial
18 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

Following these observations, clinical trials began to


determine if the mTOR inhibitor everolimus could improve
epilepsy or autistic symptoms related to TSC.
French and colleagues97 recently reported the initial re-
sults of the EXIST-3 trial, a randomized, multicenter,
placebo-controlled phase 3 of everolimus in 366 individuals
with refractory epilepsy due to TSC. The patients were
randomly assigned to receive placebo, lower-dose ever-
olimus (3 to 7 ng/mL), or higher-dose everolimus (9 to
15 ng/mL) after an initial eight-week observation period.
They determined that 15.1% of the placebo group, 28.2% of
the lower-dose everolimus group (P < 0.008 versus pla-
cebo), and 40% of the higher-dose everolimus group
(P < 0.001 versus placebo) reached the primary endpoint of
a 50% seizure reduction. They also analyzed the percentage
reduction in seizure frequency: 14.9% for placebo, 29.3% for
lower-dose everolimus (P < 0.003 versus placebo), and
39.6% for higher-dose everolimus (P < 0.001 versus pla-
cebo). These are remarkable results, especially given the
severity of the epilepsy in these individuals, but other
questions remain. Will earlier therapy minimize the en-
cephalopathy that often accompanies years of poorly
controlled epilepsy? Have we discovered the optimal
medication dose? Would an mTOR inhibitor affect seizures
due to other conditions that affect the mTOR pathway or
some forms of focal cortical dysplasia? Would there be a
benet to starting therapy after the TSC diagnosis but before
the onset of seizures?
Also left unanswered is whether mTOR inhibitors will
improve cognition and behavior in some individuals with
TSC. Clinical trials now in progress should provide answers
to these questions. The availability of mechanism-based
therapy might make it feasible to treat the disease rather
than waiting on its symptoms to develop.98

What causes TSC phenotypic variation?

Not everything is easily explained by simple Mendelian


genetics.
Phenotypic variability is striking in individuals with TSC.
FIGURE 15. Affected individuals within the same family, who typically
(A) All 18 individuals with tuberous sclerosis complexerelated renal
share the same mutation of the same TSC gene, a large
angiomyolipomas experienced a substantial reduction in tumor size during
12 months of rapamycin (sirolimus) therapy, but only ve of them main- number of other common genes, and similar environmental
tained a tumor reduction of 30% or more during the following year after circumstances, often exhibit strikingly different TSC mani-
discontinuation of therapy. (B) The individuals with lymphangioleiomyo- festations. Even identical twins often differ dramatically in
matosis exhibited improved spirometry measurements during sirolimus regard to their TSC manifestations.99 Our deepening un-
therapy. Note the improved performance in the forced vital capacity and derstanding of the genetic and molecular basis of TSC has
forced expiratory volume during the 12-month treatment period and the explained some parts of the phenotypic variability of TSC,
subsequent deterioration of the readings once therapy was discontinued.
but other aspects of the variability remain mysterious.
FEV1 forced expiratory volume 1; FVC forced vital capacity. Repro-
duced with permission from Bissler, et al.83 Clearly, some manifestations of TSC are age or gender
related, a factor that must be considered when assessing
phenotypic variation.1 Infants are unlikely to exhibit facial
improvement of long-established neurological dysfunction angiobromas or ungual bromas, and cardiac rhabdo-
in response to an mTOR inhibitor would suggest that at least myomas are unlikely to become symptomatic after the
some aspect of the dysfunction represents an ongoing neonatal period. Renal AMLs are evident in 80% of patients
cellular effect of the mutation, not just the effects of an by age ten years on imaging but seldom become symp-
already abnormal brain structure. tomatic until adolescence or adulthood.19 Pulmonary TSC
During the clinical trials of mTOR inhibitors for TSC- seldom becomes symptomatic until adulthood and is far
related tumors, families often documented a reduction in more likely to occur among women than among men. Ce-
the number of their childs seizures or described improved rebral giant cell tumors, in contrast, are relatively unlikely
cognition or behavior after starting the medication. to develop de novo in adults.32
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 19

FIGURE 16.
The chemical structure of rapamycin and everolimus. Reproduced with permission from Curatolo, et al.85

That up to three fourths of affected individuals develop The variable phenotype of TSC is not fully explained by
TSC via a spontaneous mutation has long been appreciated, the affected gene or the specic mutation. Not surprisingly,
but an estimated 15% or more of the individuals with TSC individuals with a TSC2 mutation are more likely to exhibit
exhibit somatic mosaicism and another 1% or so have multiple renal cysts. Genotypeephenotype analyses
germline mosaicism.59 Somatic mosaicism explains at least consistently show that TSC2 mutations tend to cause more
some of the cross-generational variability (a mildly affected severe manifestations than TSC1 mutations.58,100 In familial
mosaic parent may have severely affected children), cases, TSC1 and TSC2 mutations occur with roughly equal
whereas germline mosaicism explains instances in which frequency, whereas TSC2 mutations are identied more
seemingly unaffected couples have more than one child frequently among individuals with a new mutation.101 On
with identical TSC mutations. the whole, individuals with familial TSC tend to have milder
The variability of tumor formation from individual to disease manifestations than individuals with a new muta-
individual can be explained at least in part by Knudsons tion, possibly reecting the increased likelihood of TSC2
second hit phenomenon. Tumors arise after the tumor- mutations among people with spontaneous mutations. In-
forming tissue sustains one or more additional mutations dividuals who meet the clinical diagnostic criteria but have
that affect the function of the previously normal second no mutation identied on standard testing tend to have a
allele. This phenomenon may explain both the occur- milder phenotype.58 Both their mild phenotype and their
rence of a tumor and the variability in the rate of tumor unremarkable test results may be explained by somatic
growth. mosaicism.

FIGURE 17.
Facial angiobromas shown before (A) and after (B) administration of topical rapamycin. Photographs courtesy of Drs. Mary Beth Koenig and Hope
Northrup.
20 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

None of these factors, however, fully explains the 13. Rentz AM, Skalicky AM, Liu Z, et al. Tuberous sclerosis complex: a
phenotypic variability among affected family members, survey of health care resource use and health burden. Pediatr
Neurol. 2015;52:435-441.
who should have the same mutation of the same TSC gene. 14. Kiribuchi K, Uchida Y, Fukuyama Y, Maruyama H. High incidence of
Family members are susceptible to the effects of modier fundus hamartomas and clinical signicance of a fundus score in
genes and the inuences of other metabolic pathways, but tuberous sclerosis. Brain Dev. 1986;8:509-517.
even these factors cannot explain the occasional reports of 15. Gibbs JL. The heart and tuberous sclerosis. An echocardiographic
and electrocardiographic study. Br Heart J. 1985;54:596-599.
identical twins with widely differing phenotypes.99
16. DiMario FJ, Diana D, Leopold H, Chameides L. Evolution of cardiac
rhabdomyoma in tuberous sclerosis complex. Clin Pediatr. 1996;
12:615-619.
Conclusion 17. Jozwiak S, Kawalec W, Dluzewska J, Daszkowska J, Mirko-
wicz-Malek M, Michalowicz R. Cardiac tumors in tuberous
TSC is a dominantly inherited disorder of cellular differ- sclerosis: their incidence and course. Eur J Pediatr. 1994;153:
entiation and proliferation which variably affects the brain, 155-157.
18. Crawford DC, Garrett C, Tynan M. Cardiac rhabdomyomata as a
skin, kidneys, heart, and other organs. The predominant marker for the antenatal detection of tuberous sclerosis. J Med
neurological manifestations are epilepsy, autism, cognitive Genet. 1983;20:303-304.
and behavioral dysfunction, and SEGAs. TSC1 and TSC2 19. Ewalt DH, Shefeld E, Sparagana SP, Delgado MR, Roach ES. Renal
mutations cause an overlapping phenotype because their lesion growth in children with tuberous sclerosis complex. J Urol.
1998;160:141-145.
protein products tuberin and hamartin act together as a
20. Weiner DM, Ewalt DE, Roach ES, Hensle TW. The tuberous sclerosis
functional complex. Although the TSC2 phenotype is often complex: a comprehensive review. J Am Coll Surg. 1998;187:
more severe than that of TSC1, the variable phenotype of 548-561.
TSC is not fully explained by the gene that is affected nor by 21. Tran LH, Zupanc ML. Long-term everolimus treatment in in-
the specic mutation. Mechanism-based therapy with dividuals with tuberous sclerosis complex: a review of the current
literature. Pediatr Neurol. 2015;53:23-30.
mTOR inhibitors is now available for some TSC complica- 22. Jones K, Go C, Boyd J, et al. Vigabatrin as rst-line treatment for
tions, and pre-symptomatic therapy may prove to be infantile spasms not related to tuberous sclerosis complex. Pediatr
feasible in some situations. Although our understanding of Neurol. 2015;53:141-145.
TSC and our ability to prevent or treat its complications 23. Wanigasinghe J, Arambepola C, Sri RS, Sumanasena S,
Attanapola G. Randomized, single-blind, parallel clinical trial on
have grown tremendously in the last generation, ongoing
efcacy of oral prednisolone versus intramuscular corticotropin on
research is necessary if we are to solve the remaining rid- immediate and continued spasm control in West syndrome.
dles. Nevertheless, the progress so far suggests that TSC can Pediatr Neurol. 2015;53:193-199.
serve as a prototype for the development of mechanism- 24. Wanigasinghe J, Arambepola C, Sri RS, Sumanasena S,
based therapy for other genetic diseases. Muhandiram EC. The efcacy of moderate-to-high dose oral
prednisolone versus low-to-moderate dose intramuscular corti-
cotropin for improvement of hypsarrhythmia in West syndrome: a
This review article is derived from the Hower Award Lecture at the annual Child randomized, single-blind, parallel clinical trial. Pediatr Neurol.
Neurology Society meeting in Washington, D.C., on October 10, 2015. 2014;51:24-30.
25. Sparagana SP, Delgado MR, Batchelor LL, Roach ES. Seizure
remission and antiepileptic drug discontinuation in children with
tuberous sclerosis complex. Arch Neurol. 2003;60:1286-1289.
26. Bebin EM, Kelly PJ, Gomez MR. Surgical treatment for epilepsy in
References cerebral tuberous sclerosis. Epilepsia. 1993;34:651-657.
27. Fallah A, Rodgers SD, Weil AG, et al. Resective epilepsy surgery for
1. Roach ES, Sparagana SP. Diagnosis of tuberous sclerosis complex. tuberous sclerosis in children: determining predictors of seizure
J Child Neurol. 2004;19:643-649. outcomes in a multicenter retrospective cohort study. Neurosur-
2. Gomez MR. Tuberous Sclerosis. 1st ed. New York: Raven Press; gery. 2015;77:517-524.
1979. 28. Cuddapah VA, Thompson M, Blount J, Li R, Guleria S, Goyal M.
3. Bourneville DM. Sclrose tubreuse des circonvolution crbrales: Hemispherectomy for hemimegalencephaly due to tuberous scle-
Idiotie et pilepsie hemiplgique. Arch de Neurologie Paris. 1880;1: rosis and a review of the literature. Pediatr Neurol. 2015;53:
81-91. 452-455.
4. Bourneville DM, Brissaud E. Idiotie et epilepsie symptomatiques de 29. Shepherd CW, Houser OW, Gomez MR. MR ndings in tuberous
sclroses tubreuse ou hypertrophique. Arch Neurol (Paris). 1890; sclerosis complex and correlation with seizure development and
10:29-39. mental impairment. AJNR Am J Neuroradiol. 1995;16:149-155.
5. Balzer F, Menetrier P. Etude sur un cas dadnomes sbacs de la 30. Roach ES, Kerr J, Mendelsohn D, Laster DW, Raeside C. Diagnosis of
face et du cuir chevelu. Arch Physiol Norm Pathol (srie III). 1885;6: symptomatic and asymptomatic gene carriers of tuberous sclerosis
564-576. by CCT and MRI. Ann N Y Acad Sci. 1991;615:112-122.
6. Roach ES. Diagnosis and management of neurocutaneous syn- 31. Roth J, Roach ES, Bartels U, et al. Subependymal giant cell astro-
dromes. Semin Neurol. 1988;8:83-96. cytoma: diagnosis, screening, and treatment. Recommendations
7. Campbell AW. Cerebral sclerosis. Brain. 1906;28:382-396. from the International Tuberous Sclerosis Complex Consensus
8. Roach ES. Neurocutaneous syndromes. Pediatr Clin North Am. 1992; Conference 2012. Pediatr Neurol. 2013;49:439-444.
39:591-620. 32. Torres OA, Roach ES, Delgado MR, et al. Early diagnosis of sub-
9. Vogt H. Diagnostik der tubersen Sklerose. Z fr die Erforschung ependymal giant cell astrocytoma in patients with tuberous scle-
Behandlung des jugendlichen Schwachsinns auf Wissenschaftlicher rosis. J Child Neurol. 1998;13:173-177.
Grundlage. 1908;2:1-16. 33. Kotulska K, Borkowska J, Roszkowski M, et al. Surgical treatment of
10. Critchley M, Earl CJC. Tuberose sclerosis and allied conditions. subependymal giant cell astrocytoma in tuberous sclerosis com-
Brain. 1932;55:311-346. plex patients. Pediatr Neurol. 2014;50:307-312.
11. Gold AP, Freeman JM. Depigmented nevi: the earliest sign of tu- 34. Roach ES, Williams DP, Laster DW. Magnetic resonance imaging in
berous sclerosis. Pediatrics. 1965;35:1003-1005. tuberous sclerosis. Arch Neurol. 1987;44:301-303.
12. Fitzpatrick TB, Szabo G, Hori Y, Simone AA, Reed WB, 35. Goodman M, Lamm SH, Engel A, Shepherd CW, Gomez MR. Cortical
Greenberg MH. White leaf-shaped macules. Earliest visible sign of tuber count: a biomarker indicating cerebral severity of tuberous
tuberous sclerosis. Arch Dermatol. 1968;98:1-6. sclerosis complex. J Child Neurol. 1997;11:85-90.
E.S. Roach / Pediatric Neurology 63 (2016) 6e22 21

36. Vaughn J, Hagiwara M, Katz J, et al. MRI characterization and to be localized to cytoplasmic vesicles. Cancer Res. 1998;58:
longitudinal study of focal cerebellar lesions in a young tu- 4766-4770.
berous sclerosis cohort. AJNR Am J Neuroradiol. 2013;34: 61. van Slegtenhorst M, Nellist M, Nagelkerken B, et al. Interaction
655-659. between hamartin and tuberin, the TSC1 and TSC2 gene products.
37. Baumer FM, Song JW, Mitchell PD, et al. Longitudinal changes in Hum Mol Genet. 1998;7:1053-1057.
diffusion properties in white matter pathways of children with 62. Johnson MW, Emelin JK, Park S-H, Vinters HV. Co-localization of
tuberous sclerosis complex. Pediatr Neurol. 2015;52:615-623. TSC1 and TSC2 gene products in tubers of patients with tuberous
38. Roach ES, DiMario FJ, Kandt RS, Northrup H. Tuberous sclerosis sclerosis. Brain Pathol. 1999;9:45-54.
consensus conference: recommendations for diagnostic evalua- 63. Nellist M, Verhaaf B, Goedbloed MA, Reuser AJ, van den
tion. J Child Neurol. 1999;14:401-407. Ouweland AM, Halley DJ. TSC2 missense mutations inhibit tuberin
39. de Vries PJ, Whittemore VH, Leclezio L, et al. Tuberous sclerosis phosphorylation and prevent formation of the tuberin-hamartin
associated neuropsychiatric disorders (TAND) and the TAND complex. Hum Mol Genet. 2001;10:2889-2898.
Checklist. Pediatr Neurol. 2015;52:25-35. 64. Cafferkey R, Young PR, McLaughlin MM, et al. Dominant missense
40. Leclezio L, Jansen A, Whittemore VH, de Vries PJ. Pilot validation of mutations in a novel yeast protein related to mammalian phos-
the tuberous sclerosis-associated neuropsychiatric disorders phatidylinositol 3-kinase and VPS34 abrogate rapamycin cytotox-
(TAND) checklist. Pediatr Neurol. 2015;52:16-24. icity. Mol Cell Biol. 1993;13:6012-6023.
41. Krueger DA, Northrup H. Tuberous sclerosis complex surveillance 65. Kunz J, Henriquez R, Schneider U, Deuter-Reinhard M, Movva NR,
and management: recommendations of the 2012 International Hall MN. Target of rapamycin in yeast, TOR2, is an essential
Tuberous Sclerosis Complex Consensus Conference. Pediatr Neurol. phosphatidylinositol kinase homolog required for G1 progression.
2013;49:255-265. Cell. 1993;73:585-596.
42. Osborne JP, Fryer A, Webb D. Epidemiology of tuberous sclerosis. 66. Brown EJ, Albers MW, Shin TB, et al. A mammalian protein targeted
Ann N Y Acad Sci. 1991;615:125-127. by G1-arresting rapamycin-receptor complex. Nature. 1994;369:
43. OCallaghan FJK, Shiell AW, Osborne JP, Martyn CN. Prevalence of 756-758.
tuberous sclerosis estimated by capture-recapture analysis. Lancet. 67. Sabers CJ, Martin MM, Brunn GJ, et al. Isolation of a protein target
1998;351:1490. of the FKBP12-rapamycin complex in mammalian cells. J Biol Chem.
44. Gomez MR. Tuberous Sclerosis. 2nd ed. New York: Raven Press; 1995;270:815-822.
1988. 68. Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH.
45. Fryer AE, Chalmers A, Connor JM, et al. Evidence that the gene for RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-
tuberous sclerosis is on chromosome 9. Lancet. 1987;1:659-661. dependent fashion and is homologous to yeast TORs. Cell. 1994;78:
46. Roach ES, Smith M, Huttenlocher P, Bhat M, Alcorn D, Hawley L. 35-43.
Diagnostic criteria: tuberous sclerosis complex. Report of the 69. Curatolo P. Mechanistic target of rapamycin (mTOR) in tuberous
Diagnostic Criteria Committee of the National Tuberous Sclerosis sclerosis complex-associated epilepsy. Pediatr Neurol. 2015;52:
Association. J Child Neurol. 1992;7:221-224. 281-289.
47. Roach ES, Gomez MR, Northrup H. Tuberous sclerosis complex 70. Laplante M, Sabatini DM. mTOR signaling in growth control and
consensus conference: revised clinical diagnostic criteria. J Child disease. Cell. 2012;149:274-293.
Neurol. 1998;13:624-628. 71. Gao X, Pan D. TSC1 and TSC2 tumor suppressors antagonize
48. Northrup H, Krueger DA. Tuberous sclerosis complex diagnostic insulin signaling in cell growth. Genes Dev. 2001;15:
criteria update: recommendations of the 2012 International Tu- 1383-1392.
berous Sclerosis Complex Consensus Conference. Pediatr Neurol. 72. Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity,
2013;49:243-254. memory and disease. Trends Neurosci. 2010;33:67-75.
49. Kandt RS, Pericak-Vance MA, Hung WY, et al. Absence of linkage of 73. Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC,
ABO blood group locus to familial tuberous sclerosis. Exp Neurol. Blenis J. Tuberous sclerosis complex-1 and -2 gene products
1989;104:223-228. function together to inhibit mammalian target of rapamycin
50. Northrup H, Kwiatkowski DJ, Roach ES, et al. Evidence for genetic (mTOR)-mediated downstream signaling. Proc Natl Acad Sci U S A.
heterogeneity in tuberous sclerosis: one locus on chromosome 9 2002;99:13571-13576.
and at least one locus elsewhere. Am J Hum Genet. 1992;51: 74. Holz MK, Ballif BA, Gygi SP, Blenis J. mTOR and S6K1 mediate as-
709-720. sembly of the translation preinitiation complex through dynamic
51. Kandt RS, Haines JL, Smith M, et al. Linkage of an important gene protein interchange and ordered phosphorylation events. Cell.
locus for tuberous sclerosis to a chromosome 16 marker for 2005;123:569-580.
polycystic kidney disease. Nat Genet. 1992;2:37-41. 75. Way SW, McKenna III J, Mietzsch U, Reith RM, Wu HC,
52. The European Chromosome 16 Tuberous Sclerosis Consortium. Gambello MJ. Loss of Tsc2 in radial glia models the brain pathology
Identication and characterization of the tuberous sclerosis gene of tuberous sclerosis complex in the mouse. Hum Mol Genet. 2009;
on chromosome 16. Cell. 1993;75:1305-1315. 18:1252-1265.
53. Wienecke R, Konig A, DeClue JE. Identication of tuberin, the tu- 76. von der BC, Waltereit R, Zhang L, Beck H, Kirschstein T. Impaired
berous sclerosis-2 product. J Biol Chem. 1995;270:16409-16414. synaptic plasticity in a rat model of tuberous sclerosis. Eur J Neu-
54. van Slegtnenhorst M, de Hoogt R, Hermans C, Nellist M, Janssen B, rosci. 2006;23:686-692.
Verhoef S. Identication of the tuberous sclerosis gene TSC1 on 77. Martin KC. Local protein synthesis during axon guidance and
chromosome 9q34. Science. 1997;277:805-808. synaptic plasticity. Curr Opin Neurobiol. 2004;14:305-310.
55. Roach ES. Are diagnostic criteria for tuberous sclerosis still rele- 78. Sahin M, Henske E, Manning B, et al. Advances and future di-
vant? Pediatr Neurol. 2013;49:223-224. rections for tuberous sclerosis complex research: recommenda-
56. Roach ES, Kerr J, Mendelsohn D, Laster DW, Raeside C. Detection of tions from the 2015 strategic planning conference. Pediatr Neurol.
tuberous sclerosis in parents by magnetic resonance imaging. 2016;60:1-12.
Neurology. 1991;41:262-265. 79. Meikle L, Pollizzi K, Egnor A, et al. Response of a neuronal
57. Tyburczy ME, Dies KA, Glass J, et al. Mosaic and intronic mutations model of tuberous sclerosis to mammalian target of rapamycin
in TSC1/TSC2 explain the majority of TSC patients with no muta- (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead
tion identied by conventional testing. PLoS Genet. 2015;11: to improved survival and function. J Neurosci. 2008;28:
e1005637. 5422-5432.
58. Au KS, Williams AT, Roach ES, et al. Genotype/phenotype corre- 80. Julich K, Sahin M. Mechanism-based treatment in tuberous scle-
lation in 325 individuals referred for a diagnosis of tuberous rosis complex. Pediatr Neurol. 2014;50:290-296.
sclerosis complex in the United States. Genet Med. 2007;9:88-100. 81. Franz DN, Leonard J, Tudor C, et al. Rapamycin causes regression of
59. Rose VM, Au K-S, Pollom G, Roach ES, Prashner HR, Northrup H. astrocytomas in tuberous sclerosis complex. Ann Neurol. 2006;59:
Germ-line mosaicism in tuberous sclerosis: how common? Am J 490-498.
Hum Genet. 1999;64:986-992. 82. Cheng S, Hawkins C, Taylor MD, Bartels U. Pathological ndings of
60. Plank TL, Yeung RS, Henske EP. Hamartin, the product of the tu- a subependymal giant cell astrocytoma following treatment with
berous sclerosis 1 (TSC1) gene, interacts with tuberin and appears rapamycin. Pediatr Neurol. 2015;53:238-242.
22 E.S. Roach / Pediatric Neurology 63 (2016) 6e22

83. Bissler JJ, McCormack FX, Young LR, et al. Sirolimus for angio- 92. Dill PE, DeBernardis G, Weber P, Losch U. Topical everolimus for
myolipoma in tuberous sclerosis complex or lymphangioleio- facial angiobromas in the tuberous sclerosis complex. A rst case
myomatosis. N Engl J Med. 2008;358:140-151. report. Pediatr Neurol. 2014;51:109-113.
84. Bissler JJ, Kingswood JC, Radzikowska E, et al. Everolimus for 93. Goyer I, Dahdah N, Major P. Use of mTOR inhibitor everolimus in
angiomyolipoma associated with tuberous sclerosis complex or three neonates for treatment of tumors associated with tuberous
sporadic lymphangioleiomyomatosis (EXIST-2): a multicentre, sclerosis complex. Pediatr Neurol. 2015;52:450-453.
randomised, double-blind, placebo-controlled trial. Lancet. 2013; 94. Jozwiak S, Kotulska K, Domanska-Pakiela D, et al. Antiepileptic
381:817-824. treatment before the onset of seizures reduces epilepsy severity
85. Curatolo P, Moavero R. mTOR inhibitors in tuberous sclerosis and risk of mental retardation in infants with tuberous sclerosis
complex. Curr Neuropharmacol. 2012;10:404-415. complex. Eur J Paediatr Neurol. 2011;15:424-431.
86. Kingswood JC, Jozwiak S, Belousova ED, et al. The effect of ever- 95. Wu JY, Peters JM, Goyal M, et al. Clinical electroencephalographic
olimus on renal angiomyolipoma in patients with tuberous scle- biomarker for impending epilepsy in asymptomatic tuberous
rosis complex being treated for subependymal giant cell sclerosis complex infants. Pediatr Neurol. 2016;54:29-34.
astrocytoma: subgroup results from the randomized, placebo- 96. Wiegand G, May TW, Ostertag P, Boor R, Stephani U, Franz DN.
controlled, Phase 3 trial EXIST-1. Nephrol Dial Transplant. 2014; Everolimus in tuberous sclerosis patients with intractable epi-
29:1203-1210. lepsy: a treatment option? Eur J Paediatr Neurol. 2013;17:631-638.
87. Krueger DA, Care MM, Holland K, et al. Everolimus for sub- 97. French JA, Lawson JA, Yapici Z, et al. Adjunctive everolimus
ependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J therapy for treatment-resistant focal-onset seizures associated with
Med. 2010;363:1801-1811. tuberous sclerosis (EXIST-3): A phase 3, randomised, double-blind,
88. Franz DN, Agricola K, Mays M, et al. Everolimus for subependymal placebo-controlled study. Lancet. Published online September 6,
giant cell astrocytoma: 5-year nal analysis. Ann Neurol. 2015;78: 2016 http://dx.doi.org/10.1016/S0140-6736(16)31419-2.
929-938. 98. Roach ES. Mechanism-based therapy of genetic neurological dis-
89. Franz DN, Belousova E, Sparagana S, et al. Efcacy and safety of ease. Pediatr Neurol. 2014;50:285-286.
everolimus for subependymal giant cell astrocytomas associated 99. Gomez MR, Kuntz NL, Westmoreland BF. Tuberous sclerosis, early
with tuberous sclerosis complex (EXIST-1): a multicentre, rando- onset of seizures, and mental subnormality: study of discordant
mised, placebo-controlled phase 3 trial. Lancet. 2013;381:125-132. homozygous twins. Neurology. 1982;32:604-611.
90. Franz DN, Belousova E, Sparagana S, et al. Everolimus for sub- 100. Dabora SL, Jozwiak S, Franz DN, et al. Mutational analysis in a
ependymal giant cell astrocytoma in patients with tuberous scle- cohort of 224 tuberous sclerosis patients indicates increased
rosis complex: 2-year open-label extension of the randomised severity of TSC2, compared with TSC1, disease in multiple organs.
EXIST-1 study. Lancet Oncol. 2014;15:1513-1520. Am J Hum Genet. 2001;68:64-80.
91. Landecho MF, Alegre F, Lucena JF, Huerta A, Fernandez-Ros N, 101. Jones AC, Shyamsundar MM, Thomas MW, et al. Comprehensive
Aldaz A. Monitoring of everolimus trough concentrations for mutation analysis of TSC1 and TSC2- and phenotypic correlations
achieving optimal tolerance in the management of tuberous scle- in 150 families with tuberous sclerosis. Am J Hum Genet. 1999;64:
rosis complex. Pediatr Neurol. 2014;51:e9-e10. 1305-1315.

S-ar putea să vă placă și