Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

REVIEW ARTICLE

Neurofibromatosis Type 1:
Review and Update on Emerging Therapies
Tanya Karaconji, FRANZCO,*† Eline Whist, BMed, MPHTM, FRANZCO,*‡
Robyn V. Jamieson, PhD, FRACP,§¶ǁ** Maree P. Flaherty, FRANZCO, FRACS, FRCOphth,*†† and
John R.B. Grigg, MD, FRANZCO*¶ǁ‡‡

ophthalmic features of this complex disorder are discussed in this


Abstract: Neurofibromatosis type 1 is an autosomal dominant review.
neurocutaneous disorder affecting 1 in 3000 births. This familial tumor
predisposition syndrome is diagnosed clinically and affects the skin,
bones, and nervous system. Malignant tumors can arise in childhood or EPIDEMIOLOGY
adulthood and are the most common cause of mortality in this population. Neurofibromatosis 1 is one of the most common genetic
Early diagnosis and management led by a multidisciplinary team remains disorders with no sex or racial predilection. It occurs in
the standard of care, particularly in the management of optic pathway approximately 1 in 3000 people.3 Estimates of the prevalence
glioma. Emerging concepts in the genetic patterns of this condition of NF1 range from 1:21904 to 1:7800.5 Some of the difficulties
have led to the introduction of new treatment modalities that target the with accurate prevalence data for this condition may be due to
mitogen-activated protein kinase and the mammalian target of rapamycin ascertainment bias6 partly because of the wide variability in the
pathways. In this review, the role of the ophthalmologist and approach expression of the condition, whereby mild cases may escape
to screening for optic pathway glioma are outlined based on previous study inclusion particularly in studies dependent on an affected
recommendations. Updates on choroidal involvement, as a diagnostic individual coming to medical attention.7 Reduced prevalence
criterion, will also be discussed, further highlighting the pivotal role of in later adulthood may be due to increased mortality seen in
the ophthalmologist in the diagnosis and management of this complex individuals with NF1 or to underascertainment of adult patients.8
condition. The physician Mark Akenside first recognized the disorder in
1768. Robert Smith, an Irish surgeon, published further details
Key Words: choroidal nodules, neurofibromatosis 1, optic pathway on the disease in 1849. However, it was the German pathologist
glioma, phakomatoses Friedrich von Recklinghausen who is credited with its recognition
in 1882.1
(Asia Pac J Ophthalmol (Phila) 2019;8:62–72)

GENETICS AND PATHOPHYSIOLOGY

N eurofibromatosis 1 (NF1) is an inherited neurocutaneous


disorder. It is characterized by the presence of multisystem
tumors throughout the skin and central nervous system (CNS),
The inheritance of NF1 is autosomal dominant with 100%
penetrance but highly variable expressivity.1,3,9 Importantly, a high
spontaneous mutation rate of up to 42% exists.3 The NF1 gene is
which carries a risk of malignant transformation.1 The hallmark located on chromosome 17q11.2 and was cloned in 1990. It was
clinical features of NF1 include multiple café au lait macules, originally cloned by the Wallace group10 along with the Cawthon
neurofibromas, intertriginous freckling, osseous lesions, Lisch and Viskochil group.11 It is an important tumor suppressor gene
nodules, and optic pathway gliomas (OPGs).2 An evolving found throughout the nervous system.12
understanding of both the genetic and newly recognized The NF1 gene encodes the protein neurofibromin, which
influences multiple signaling pathways affecting many cellular
processes throughout the body. Importantly, it acts as a negative
From the *Discipline of Ophthalmology, Save Sight Institute, Sydney Medical regulator of cell growth and proliferation.2 Several pathways are
School, University of Sydney, Australia; †Manchester Royal Eye Hospital,
Manchester, United Kingdom; ‡Royal Darwin Hospital, Darwin, Australia; thought to be involved in the development of tumors associated
§Disciplines of Paediatrics, Genomic Medicine and Ophthalmology, Sydney with NF1. Loss of neurofibromin increases rat sarcoma viral
Medical School, University of Sydney, Australia; ¶Eye Genetics Research
Unit, Children’s Medical Research Institute, University of Sydney, Australia; oncogene homolog (RAS) activity. This increased RAS activity
ǁSave Sight Institute and Eye Genetics Clinics, The Children’s Hospital at causes unopposed cell growth and activation of important
Westmead, Australia; **Westmead Hospital, Sydney, Australia; ††Department
of Ophthalmology, The Children’s Hospital at Westmead, Australia; and downstream intermediates such as the mitogen-activated protein
‡‡Sydney Eye Hospital, Sydney, Australia. kinase (MAPK) and the mammalian target of rapamycin (mTOR)
Submitted May 1, 2018; accepted August 23, 2018.
J.R.B.G. and R.V.J. are supported in part by NHMRC Centre for Research pathways.2,9
Excellence APP1116360. E.W. is supported in part by Sydney Eye Hospital Neurofibromin is also involved in the regulation of cyclic
Foundation.
The other authors have no funding or conflicts of interest to declare. adenosine monophosphate levels, which has been shown to affect
Reprints: Tanya Karaconji, Discipline of Ophthalmology, Save Sight Institute, the CNS in animal models, and importantly in OPG formation.12–14
Sydney Medical School, University of Sydney, NSW 2006, Australia. E-mail:
tkaraconji.@gmail.com. Although NF1 may result in a combination of benign and
Copyright © 2019 by Asia Pacific Academy of Ophthalmology malignant tumors of the central and peripheral nervous system,
ISSN: 2162-0989
DOI: 10.22608/APO.2018182 the initial event in tumor formation is the germline mutation in

62 | www.apjo.org Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019 Neurofibromatosis Type 1

the NF1 tumor-suppressor gene. The “second hit” in this “2- Lisch Nodules
hit hypothesis” pathway is a somatic mutation, causing loss of Austrian ophthalmologist Karl Lisch was the first to
heterozygosity of the NF1 gene, and consequently leading to emphasize what are now called Lisch nodules and their association
Schwann cell neurofibromas along with other tumors.9 with NF1 in 1937.20 Histologically Lisch nodules are melanocytic
hamartomas, consisting of a condensation of spindle cells on the
anterior iris surface. Stromal iris nevi underlie the pigmented
CLINICAL PRESENTATION nodules.21 They are present on the anterior iris surface or in the
Neurofibromatosis 1 is a neurocutaneous disorder with an anterior chamber angle. They are well-defined yellow-brown
age-dependent, variable clinical presentation. This often leads dome-shaped elevated lesions and can range in size from pinpoint
to difficulty in establishing an early diagnosis. Despite this, NF1 to large (Fig. 1).20,22 Although Lisch nodules do not cause ocular
has well-recognized diagnostic criteria as outlined in Table 1.15 morbidity or disability, they are important because they are one of
Although these “classical” criteria are widely used and agreed the diagnostic criteria for NF1. Their presence is age-dependent.
upon, additional new cutaneous and extracutaneous features Although their presence is unusual before the age of 2 years, they
have been described in recent years (Table 1). An example of are seen in half of 5-year-olds, 75% of 15-year-olds, and almost
this is the presence of unidentified bright objects on imaging. 100% of adults older than 30 years.2
These are seen as hyperintense lesions on T2-weighted brain
magnetic resonance imaging (MRI) scans, which may represent Skin
aberrant gliosis pathognomonic of NF1.16 Additionally, advanced Café au lait macules and skin fold freckling do not usually
molecular analysis is now available, refining our knowledge of cause complications and have no malignant potential.23 They are
the NF1 gene mutation.17 an important component of the diagnostic criteria of NF1. Café au
A small subset of patients present with a somatic mutation or lait patches are usually the first identified clinical manifestation in
“segmental” NF1 where the clinical features only affect a specific NF1.24 They are usually present within the first 2 years of life.18
area of the body and neither parent is affected.18 Other phenotypic They present as well-defined hyperpigmented macular lesions
presentations in patients with an NF1 gene mutation include that are generally absent from the scalp and eyebrows along with
neurofibromatosis-Noonan syndrome, which may occur in up to the palms and soles (Fig. 2).18 Histologically these lesions are due
12% of these patients. Clinical features are an overlap between to increased proliferation of epidermal melanocytes with basal
the 2 named syndromes and include the Noonan features of short hyperpigmentation of the epidermis.
stature, ocular hypertelorism, low-set ears, and downslanting Axillary and inguinal freckling usually presents after the
palpebral fissures.19 development of café au lait macules by the age of 5 to 8 years.24

TABLE 1. National Institutes of Health (NIH) Diagnostic Criteria for Neurofibromatosis 1 (NF1) and Other Common Findings2,6,9,14,15

The NIH Diagnostic Criteria are Met if 2 or More of the Following are Found
1. Six or more café au lait macules of over 5 mm in greatest diameter in prepubertal individuals and over 15 mm in greatest diameter in
postpubertal individuals
2. Two or more neurofibromas of any type or 1 plexiform neurofibroma
3. Freckling in the axillary or inguinal regions
4. Optic glioma
5. Two or more Lisch nodules (iris hamartomas)
6. A distinctive osseous lesion such as sphenoid dysplasia or thinning of long bone cortex
7. A first-degree relative with NF1 by the above criteria
Nondiagnostic Cutaneous and Extracutaneous Signs to Consider in Addition to the “Classical” Diagnostic Criteria When Evaluating
Patients with NF1
Genetic analysis Molecular analysis of the NF1 gene
Cutaneous signs Anemic nevus
Juvenile xanthogranuloma
Mixed vascular hamartomas and cherry angiomas
Hypochromic macules
“Soft-touch” skin
Hyperpigmentation
Extracutaneous Choroidal hamartomas
Large head circumference and hypertelorism
Unidentified bright objects on neuroimaging
Cerebrovascular dysplasia, moyamoya
Learning, speech and behavioral disabilities, headache and seizures
Neoplasms

© 2019 Asia-Pacific Academy of Ophthalmology www.apjo.org | 63

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Karaconji et al Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

Typically, they present as small freckles less than 5 mm in size in


the axillary or inguinal area.18,24,25 They may also present in areas
where skin folds are in apposition, including the neck and under
the breasts in women.7 These are given the eponymous name,
Crowe sign, after the physician Frank Crowe who first described
their association with café au lait macules in the diagnosis of NF1
in 1964.26

Neurofibromas
Neurofibromas are a hallmark feature of NF1, present in
almost all patients older than 30 years.25 They are benign soft tissue
tumors of Schwann cell origin that arise on peripheral nerves.27 In
addition to neoplastic Schwann cells, they also contain fibroblasts,
macrophages, and mast cells.28 Clinically, neurofibromas can be
FIGURE 1. Slit-lamp photograph showing multiple Lisch nodules. further classified as cutaneous, subcutaneous, or plexiform which
can be further divided into nodular or diffuse. The features of
each are outlined in Table 2.17–19,24 Although they typically present
symptomatically on the skin, they can also develop deep within
the body and may remain largely asymptomatic.24
Like most features in NF1, the presence of neurofibromas
is age-dependent. Although neurofibromas tend to increase with
age, the presence of plexiform neurofibromas may manifest at
birth and are most active during the first decade of life, developing
in 25% to 50% of individuals with NF1.29
The morbidity associated with plexiform neurofibromas is
2-fold. In addition to the disfigurement, bony destruction, and
pain associated with these lesions, they also carry an 8% to 13%
lifetime risk of malignant transformation [malignant peripheral
nerve sheath tumor (MPNSTs)].23,24 This rate of malignant
transformation is higher than previously thought.30
Unlike cutaneous neurofibromas, plexiform neurofibromas
can infiltrate surrounding soft tissue and arise from multiple
nerve fascicles, with multiple discrete tumors forming along
nerve trunks.23 Growth of these tumors is often unpredictable
and is usually rapid during the first decade of life, resulting in
significant disfigurement and functional impairment.25 Palpebral
plexiform neurofibromas typically affect the upper eyelid
unilaterally appearing in infancy or early childhood and may
result in significant functional impairment including amblyopia.25
This may be due to a mechanical ptosis causing astigmatism.
As always in pediatric patients, regular refraction, dilated
examination, and amblyopia treatment are of utmost importance,
particularly in NF1 patients presenting with eyelid plexiform
neurofibromas.
Unfortunately, MPNSTs have a poor prognosis with a 5-year
survival of up to 60%, warranting ongoing careful surveillance
FIGURE 2. Café au lait macules. with a low threshold for investigation.31 Poor prognosis is

TABLE 2. Clinical Features of Established Neurofibroma Subtypes17–19,24

Neurofibroma Types Location Clinical Signs Presentation


Cutaneous Epidermis and dermis Moves with skin, bluish tinge, local pruritus. Asymptomatic; most common type;
Size: ~2 mm to 3 cm presents in late teens
Subcutaneous Deep to dermis Skin moves over, firm and rounded feel, located Tenderness on palpation; tingling in
along peripheral nerves. Size: 3 to 4 cm distribution of affected nerve
Nodular plexiform Localized interdigitation “Bag of worms” feel May be present from birth, enlarges
with normal tissues during first decade of life
Diffuse plexiform Infiltrate widely and deeply Smooth slightly irregular skin thickening −

64 | www.apjo.org © 2019 Asia-Pacific Academy of Ophthalmology

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019 Neurofibromatosis Type 1

associated with early metastasis and poor response to systemic predicting response in humans.
chemotherapy. Delayed diagnosis remains an issue. Features Although these studies have shown promising results
including increased growth rate, irregular contour, and pain in in plexiform neurofibromas, unfortunately they have only
existing neurofibromas are symptoms and signs that may indicate demonstrated a transient benefit in the treatment of MPNST in
malignant transformation. These symptoms may also be present mice models.36,38 In contrast, mTOR pathway inhibitors such as
in benign lesions.31,32 rapamycin suppress the growth of these MPNST but have not
Although a histological diagnosis remains the gold standard demonstrated tumor regression.39 The mTOR pathway is critically
in differentiating benign from malignant lesions, imaging with deregulated in NF1-mutant MPNST, hence the role of mTOR
positron emission tomography and computed tomography inhibitors.39 However, when combined with heat shock protein 90
combined with a flurodeoxyglucose tracer provides a noninvasive inhibitors, potent tumor regression is observed.40
technique to identify malignant transformation based on in-vivo
glucose metabolism.31 Definitive diagnosis of a MPNST requires Anterior Segment and Uvea
pathological identification. The presence of choroidal abnormalities in neurofibromatosis
was reported in as early as the 1930s.41 These lesions were
Neurofibroma Treatment identified on enucleated specimens and attributed to proliferating
Plexiform neurofibromas are notoriously difficult to treat Schwann cells arranged as concentric rings around axons.42,43
surgically due to their infiltration of adjacent normal tissue and More recently, a number of case series have used confocal near-
the commonly reported risk of regrowth after excision.33 Targeted infrared reflectance (NIR) imaging as a means of identifying
chemotherapeutic agents such as imatinib (tyrosine kinase these choroidal lesions in NF1 patients.44–47 In each of these
inhibitor) have also been studied but have only demonstrated a studies, the choroidal abnormalities identified were not visualized
modest reduction in tumor volume size in a phase 2 trial in affected by simple ophthalmoscopic examination or fundus fluorescein
patients with clinically significant plexiform neurofibromas angiography.46 Further studies have found that these choroidal
due to NF1.34 Interferons exert antitumor activity via a number abnormalities tend to increase with age and are particularly
of mechanisms including antiproliferative, antiangiogenesis, localized around the posterior pole.45
and cytotoxic effects. A phase 1 trial of pegylated interferon- Near-infrared reflectance imaging at 815 nm and optical
alpha-1beta in young patients with progressive and unresectable coherence tomography have further elucidated the presence of
plexiform neurofibromas has demonstrated a promising reduction these choroidal bright patchy nodules in patients with NF1.46
and stabilization of plexiform neurofibroma size.35 Viola et al46 found choroidal bright patches in 82% of NF1
Clinical trials with biologic agents including mTOR and patients compared with only 7% of normal controls with a
MAPK (MEK) inhibitors are underway.36 Studies looking diagnostic sensitivity of 83% and specificity of 96%. These
at MEK inhibitors in mouse models have demonstrated that choroidal abnormalities have been further categorized as either
blocking MEK signaling was sufficient to profoundly shrink “domeshaped” or “placoid-like”.48 These lesions are also reported
existing neurofibromas.37 Clinical trials with MEK inhibitors at a high frequency in pediatric NF1 patients.47 The appearance of
for plexiform neurofibromas are ongoing or in development and these lesions on NIR imaging is demonstrated in Figure 3.
will allow further evaluation of the utility of the mouse model in These studies highlight the potential importance of choroidal

FIGURE 3. The near-infrared reflectance images of the right and left eyes of a patient with neurofibromatosis type 1 demonstrating bright patchy
areas localized to the posterior pole and vascular arcade. White arrows indicate 2 of multiple lesions in each eye (images courtesy of Francesco Viola
Università degli Studi di Milano).

© 2019 Asia-Pacific Academy of Ophthalmology www.apjo.org | 65

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Karaconji et al Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

involvement as a new diagnostic criterion for NF1. In particular, which may all predispose to glaucoma.51 Interestingly, Morales et
the use of NIR imaging to detect these choroidal abnormalities is al53 found ipsilateral glaucoma and increased axial length in 23%
gaining increasing interest particularly among ophthalmologists of 95 patients with orbitofacial involvement.
as a possible screening tool, as it is noninvasive and can be
applied to the pediatric population, at least in older children.46,47 Skeletal Anomalies
Additional choroidal changes that have also been described A number of skeletal manifestations are recognized in NF1
include a reduction in mean choroidal thickness, which may be patients. These may present as either generalized or focal bony
related to altered choroidal circulation in the presence of choroidal abnormalities. Generalized bony changes include osteopenia,
nodules (Fig. 4).48 Another case report has identified increased short stature, and macrocephaly.36 In NF1 patients older than 40
subfoveal choroidal thickness in a case of NF1.49 Pigmentary years, a 5-fold increase in fracture risk has been reported, with a
hamartomas (choroidal nevi) may also involve the posterior uveal 3-fold risk in children younger than 16 years.54
tract in up to 30% of affected patients.50 Focal bony abnormalities include tibial dysplasia/
Although the association of glaucoma and neurofibromatosis pseudarthrosis (2%), sphenoid wing dysplasia (1–6%), scoliosis
has been described in the literature as a rare association, the (10–15%), nonossifying fibrosis, pectus deformities of the chest,
mechanism is not well understood and is most likely related and rarely, defects in the occipital bone.24,36,55 Of particular
to angle closure.25,51,52 A retrospective histological study by significance to the ophthalmologists is the presence of sphenoid
Edward et al51 analyzed the eyes from 5 patients with NF1 wing dysplasia that may present with asymmetric orbits,
and glaucoma, ranging in age from birth to 13 years. They proptosis, or enophthalmos.24 More specifically, these patients
identified endothelialization of the anterior chamber angle, usually present with pulsatile proptosis due to herniation of the
which they hypothesized might be related to overexpression of temporal lobe into the orbit.23 Importantly, absence or thinning
the RAS-MAPK genes in these eyes.51 Clinically, infiltration of of the sphenoid wing may be due to the presence of an orbital
the iris and angle with neurofibromas may obstruct the angle plexiform neurofibroma.24
resulting in secondary angle closure.25 Other anterior segment Although the mechanisms underlying these osseous
features include congenital ectropion uvea, iris heterochromia, manifestations remain poorly understood, studies have
developmental anomaly of the angle, and posterior embryotoxon, demonstrated that neurofibromin plays a critical role in

FIGURE 4. Choroidal changes (arrows) are shown in a patient with neurofibromatosis type 1 as seen on near-infrared reflectance imaging, with
corresponding optical coherence tomography through the choroidal lesions in the right (A) and left (B) eye.

66 | www.apjo.org © 2019 Asia-Pacific Academy of Ophthalmology

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019 Neurofibromatosis Type 1

regulating mesenchymal stem/progenitor cell differentiation into symptoms or signs related to the OPG.60,67,68 Symptomatic
osteoblasts, affecting collagen synthesis and mineralization.36,56 OPG in children with NF1 generally results in decreased visual
A defect in this process is likely to contribute in part to some of acuity, diminished visual fields, abnormal pupillary function,
these clinical features. Ideally, specialized pediatric orthopedic or proptosis. It is important to note that young children rarely
surgeons should manage these children. complain of vision loss, necessitating the use of reliable,
reproducible measures to detect visual changes.58 Reduction in
Optic Pathway Gliomas visual acuity is the most common finding on screening.61 Other
Optic pathway gliomas account for 5% of pediatric less common ocular presentations include strabismus, proptosis,
intracranial tumors.57 Traditional figures suggest that 15% to 20% nystagmus, afferent pupil defect, and abnormal color vision.61
of children with NF1 will develop a low-grade glial neoplasm.58,59 Clinically, the optic discs usually appear pale. Low-grade glial
A large retrospective review of 861 patients seen at the tertiary tumors may also present with systemic manifestations such as
referral neurofibromatosis clinic at the Children’s Hospital precocious puberty or rarely, diencephalic syndrome.68 Although
at Westmead in Sydney found a prevalence of 6.6% in their it is extremely rare, the latter may occur in cases with tumor
population.60 involvement of the thalamus or hypothalamus and presents with
Optic pathway gliomas can occur anywhere in the brain or failure to thrive, vomiting, headaches, and vision abnormalities.69
spine in children with NF1. They are, however, most common Minimum requirements for cranial MRI at diagnosis include
in the optic pathway, hypothalamus, and (to a lesser extent) T2-weighted, T1-weighted, and gadolinium contrast-enhanced
brainstem.24,61 The traditional Dodge classification (DC) of OPGs T1-weighted sequences and fluid-attenuated inversion recovery.
proposed in 195862 defines tumors anatomically as involving the This is used to diagnose and monitor progression of these tumors.
optic nerves alone (stage 1), the chiasm with or without nerve Typical features include fusiform enlargement, tortuosity, and
involvement (stage 2), and the hypothalamus or other adjacent kinking of the optic nerves. There may also be involvement of the
structures (stage 3). The DC has long been used to select patients chiasm (best visualized on coronal sections), optic tract, or optic
for resection of optic nerve tumors and to provide an important radiation with diffuse enlargement.70 Optic pathway gliomas are
indicator of prognosis with tumors at the optic chiasm having a isointense or slightly hypointense to normal optic nerve on T1
poorer outcome. More recently, a modified DC has introduced imaging and hyperintense on T2 imaging71,72 (Fig. 5). Although
an imaging-based method using MRI scans and is referred to as they may demonstrate gadolinium enhancement, not all OPGs do.
the PLAN score.63 This updated score provides a more detailed Abnormal perineural tissue within the nerve sheath may also be
anatomical description that can help to categorize and select tumors present.71,72 The screening guidelines and management pathways
at a higher risk of visual morbidity to better guide treatment and of patients with NF1 are outlined in Figure 6.60,61
detect change. Taylor et al63 found that NF1-positive cases more
commonly involved both optic nerves and multiple locations Treatment
at other sites whereas NF1-negative tumors more commonly Current imaging is virtually always diagnostic, so biopsy of
involved the central chiasm and the hypothalamus. Most patients suspected tumors is no longer necessary.70 Surgery is recommended
will be diagnosed before 6 years of age,61 although other series for OPG cases with severe vision loss and disfiguring proptosis
have found that up to 30% of children presented with OPGs after only when assessed by a multidisciplinary team, as surgery in
the age of 6.60,64 Table 3 outlines the features of the most common NF1 OPG is rarely useful and will often sacrifice whatever vision
subtype, pilocytic astrocytoma. Although pilocytic astrocytomas remains.73
are common in NF1 patients and usually follow an indolent A combination of carboplatin-based chemotherapy, often
course, they may spontaneously resolve without intervention.36,65 in conjunction with vincristine, remains the gold standard for
Pilomyxoid astrocytoma, another subtype, is less common in NF1-associated low-grade gliomas requiring treatment.74 The
the context of NF1.2 It is found that NF1-associated OPGs have 5-year progression-free survival in these patients has been shown
active MAPK signaling and rarely transform into high-grade to be as high as 69%.74 This regimen also has lower toxicity
tumors with no malignant potential.2 and side-effect profile than other comparable agents.74 Other
These tumors may have a wide range of clinical presentations. traditional chemotherapeutic agents have also been associated
Many OPGs are asymptomatic. They may become evident either with a worrisome risk of secondary malignancies.36 In recurrent,
as a result of patient signs or symptoms or as an incidental finding refractory tumors, alternative agents such as vinblastine have
on “ baseline” neuroimaging studies.66 A study by Listernick et proven to be efficacious, although it is poorly tolerated due
al67 in 1989 demonstrated that, if all children with NF1 undergo to extensive bone marrow suppression in many patients.36
screening neuroimaging, 15% of them will have radiographic Pretreatment vision is an important prognostic indicator of final
evidence of OPG with only half of these children developing visual acuity after chemotherapy.36

TABLE 3. Characteristic Features of the Most Common Subtype of Optic Pathway Glioma (OPG) in Neurofibromatosis 1 (NF1): Pilocytic Astrocytoma13,54

Histological features GFAP-staining in cell cytoplasm and presence of Rosenthal fibers; contains piloid cells and astrocytes
Incidence (per 100,000 per year) 0.48
Association with NF1 Mainly OPG (50%)
Age 5–19 y
World Health Organization grade I
Location Cerebellum/brainstem/optic pathway

© 2019 Asia-Pacific Academy of Ophthalmology www.apjo.org | 67

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Karaconji et al Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

A B

FIGURE 5. Magnetic resonance imaging (MRI): A, Coronal T2 of the brain: bilateral optic nerve enlargement with tortuosity. B, axial T2 of the brain and
orbits demonstrating enlargement and tortuosity of both optic nerves, consistent with an optic pathway glioma.

Screening  guidelines   Management  Pathways  

Screening   3  Monthly  
Annual  ophthalmic   children  with   follow-­‐up  with  
assessment  (VA,  VF,   suspected  OPG   MRI  +   MRI  in  the  1st  
0  −  8  y   or  abnormal   MDT   year.  May  be  
pupils,  eyelids,  OMS,  iris,  
fundi,  refrac0onC   ophthalmic   extended  in  
examina0on   the  following  
year  if  clinically  
stable  

Second  yearly  
Screening  
ophthalmic  assessment  
asymptoma0c   >8  −  18  y  
(VA,  VF,  pupils,  eyelids,  
1F1  pa0ents  
OMS,  iris,  fundi,   During  chemotherapy:    
refrac0onC   3  monthly  ophthalmic  
Monitoring   assessment  +  OCT  +  MRI  
children  with  
confirmed  OPG  
Rou0ne  eye  care  only,   undergoing   Post  chemotherapy:    
>18  y  
no  screening   treatment   3-­‐6  monthly  ophthalmic  
assessment  +  OCT  +  MRI.  
Increase  intervals  if  
clinically  improved/stable  

FIGURE 6. Proposed screening guidelines and management pathways of patients with NF1. If unable to accurately assess a child clinically, there may be
a role for neuroimaging. Some centers do perform baseline neuroimaging in new diagnosis of NF1. MDT indicates multidisciplinary team (pediatrician,
neurologist, oncologist, neurosurgeons, geneticist); MRI, magnetic resonance imaging; NF1, neurofibromatosis 1; OCT, optical coherence tomography;
OMS, ocular motor score; OPG, optic pathway glioma; VA, visual acuity; VF, visual field (age-appropriate).

Newer therapies targeting MEK inhibitors and mTOR based therapy.76 Although further studies are required, this
pathways are currently under trial.70 Drugs targeting tumor presents a promising treatment in addition to standard therapies.
angiogenesis, such as bevacizumab, have shown objective The Pediatric Brain Tumour Consortium study is currently
responses radiologically and on vision testing in cases of evaluating the use of selumetinib, an MEK inhibitor, in children
refractory OPG.75 A study by Avery et al76 in 2014 looked at the with recurrent or refractory low-grade gliomas with promising
use of intravenous bevacizumab in 4 cases of pediatric OPGs who results.77
demonstrated progressive visual acuity or visual field loss despite
prior treatment with chemotherapy or proton-beam radiotherapy. Screening for Optic Pathway Tumors in NF1
All 4 subjects demonstrated a marked improvement in their One of the critical roles that ophthalmology plays in the
visual acuity, visual field, or both while receiving bevacizumab- management of children with NF1 is the regular monitoring

68 | www.apjo.org © 2019 Asia-Pacific Academy of Ophthalmology

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019 Neurofibromatosis Type 1

and screening for OPGs. Given the high frequency of learning average range.23 The incidence of true cognitive delay in NF1
and behavioral problems in NF1 children, this regular screening is, however, lower than expected.84 There are increased rates of
should be in the context of a multidisciplinary team.23 autism spectrum disorder, attention-deficit hyperactivity, and
Routine screening remains the commonest means to identify sleep disturbances in these patients.85 Language difficulties,
OPGs.61 The neuro-ophthalmic examination is a key component particularly receptive language and language structure, have
in the diagnosis and management of OPGs. In children with been identified as issues in children with NF1.86 In view of
NF1, examination guidelines have been developed by expert these findings, early neuropsychological screening assessment
consensus58 and are outlined in Figure 6. The Response Evaluation to help evaluate and support these children as they enter school
in Neurofibromatosis and Schwannomatosis Visual Outcomes should be considered.24 This also supports the need for ongoing
Committee recommends that visual acuity should be the primary routine screening, which is best performed in dedicated NF
outcome measure to assess for tumor progression, guide treatment multidisciplinary clinics.
decisions, and evaluate treatment effect.61
Pattern visual evoked potentials are performed in some Vascular Complications
centers at the same frequency as neuroimaging. These provide A number of cardiovascular abnormalities have also been
additional objective evidence for visual pathway dysfunction described in NF1 patients. Congenital heart disease, particularly
when performed in laboratories experienced with visual pulmonary stenosis, accounts for as many as 50% of these
electrophysiology in children. Multifocal visual evoked potentials abnormalities.87 Other NF1-related vascular abnormalities
offer a method for assessing the topographic optic nerve function, include renal and cerebral artery stenosis, moyamoya, aneurysmal
providing an objective measure of visual field function.78,79 Optical lesions, and arteriovenous fistula malformations.36
coherence tomography of the optic nerve has also proved to be an Interestingly, both adult and pediatric patients with NF1
effective, noninvasive tool to assess for progressive retinal nerve have a significantly increased risk of stroke when compared with
fiber layer thinning secondary to OPGs in NF1 patients; although the general population.88 Although the risk is most notable for
it is not considered standard surveillance, it has proved a potential hemorrhagic strokes, it is also increased for ischemic strokes
biomarker of vision in NF1 OPGs.59,80 in children due to a number of underlying vascular lesions.88,89
The decision on when to treat children with OPGs remains Various associations have included moyamoya arteriopathy,90
controversial and relies on input from a multidisciplinary team. cerebral aneurysms, and ectatic cerebral vessels.89
Treatment is reserved for progressive disease, which may
include either reduced vision or radiographic tumor growth, or a Prognosis
combination of the 2.59,70 In children with NF1-associated OPG after chemotherapy,
approximately a third will regain some vision with treatment.91
Central Nervous System and Other Systems Unlike vision, which will largely remain stable or may even
improve, NF1 reduces average life expectancy by 10 to 15 years,
Other CNS Tumors and malignant tumors are the most common cause of death in
Gliomas can present throughout the CNS in individuals with individuals with this syndrome.7 It is important to remember that
NF1.73 It is important to recognize that among patients with NF1 NF1 patients have a 5-fold risk for cancer and more than a 2000-
who are older than 10 years, the relative risk of a brain tumor is fold risk for neurogenic malignancies compared with the general
100 times higher than those without NF1.81 Most brain tumors population.92
in children with NF1 are low-grade gliomas [World Health
Organization (WHO) grade I–II], and the majority of those are
WHO grade I.9 These tumors do have malignant potential and can CONCLUSIONS
infiltrate the brain.9 As NF1 remains a multisystem disease with life-threatening
Brainstem gliomas associated with NF1 have been described complications, a multidisciplinary approach with close
and, in contrast to the general population, they tend to have a collaboration among NF1 clinicians will facilitate a uniform
more indolent course and may not require treatment.82 method for diagnosis and management of this condition.
Ophthalmologists have a crucial role in diagnosis and in
Non-CNS Tumors monitoring visual function. The recent characterization of the
A range of other non-CNS tumors has also been reported choroidal hamartomas emphasizes this role.
in patients with NF1 including pheochromocytomas, which may Despite the recent and exciting new advances in NF1
initially present with hypertension.9 Gastrointestinal fibromas research, including MEK and mTOR inhibitors, there still remain
have also been described.9 many questions about the disease. The continuing challenge
The lifetime breast cancer risk is 4 times higher in women for clinicians is the detection of progression of OPG as early as
with NF1 who are younger than 50 years compared with the possible so that effective treatment can be instigated to maximize
general population.83 Women with NF1 also have an increased the preservation of vision.
risk for invasive breast cancer across all age groups.83

Learning Difficulties; Speech, Language, and Cognitive ACKNOWLEDGMENT


Impairment The authors would like to thank Dr. Francesco Viola,
Learning difficulties and cognitive impairment remain Università degli Studi di Milano, UO Oculistica, Fondazione
the commonest neurological complication of NF1 in children. IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy,
The mean full-scale intelligence quotient is in the low-to- who contributed optical coherence tomography images.

© 2019 Asia-Pacific Academy of Ophthalmology www.apjo.org | 69

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Karaconji et al Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

REFERENCES 2007;44:81–88.
1. Tadini G, Milani D, Menni F, et al. Is it time to change the neurofibromatosis 24. Hirbe AC, Gutmann DH. Neurofibromatosis type 1: a multidisciplinary
1 diagnostic criteria? Eur J Intern Med. 2014;25:506–510. approach to care. Lancet Neurol. 2014;13:834–843.
2. Helfferich J, Nijmeijer R, Brouwer OF, et al. Neurofibromatosis type 25. Abdolrahimzadeh B, Piraino DC, Albanese G, et al. Neurofibromatosis: an
1 associated low grade gliomas: a comparison with sporadic low grade update of ophthalmic characteristics and applications of optical coherence
gliomas. Crit Rev Oncol Hematol. 2016;104:30–41. tomography. Clin Ophthalmol. 2016;10:851–860.
3. Kissil JL, Blakeley JO, Ferner RE, et al. What’s new in neurofibromatosis? 26. Crowe FW. Axillary freckling as a diagnostic aid in neurofibromatosis. Ann
Proceedings from the 2009 NF Conference: new frontiers. Am J Med Genet A. Intern Med. 1964;61:1142–1143.
2010;152A:269–283. 27. Hajdu SI. Peripheral nerve sheath tumors. Histogenesis, classification, and
4. Fuller LC, Cox B, Gardner RJ. Prevalence of von Recklinghausen prognosis. Cancer. 1993;72:3549–3552.
neurofibromatosis in Dunedin, New Zealand. Neurofibromatosis. 1989;2: 28. Munchhof AM, Li F, White HA, et al. Neurofibroma-associated growth
278–283. factors activate a distinct signaling network to alter the function of
5. Sergeyev AS. On the mutation rate of neurofibromatosis. Humangenetik. neurofibromin-deficient endothelial cells. Hum Mol Genet. 2006;15:
1975;28:129–138. 1858–1869.
6. Friedman JM. Epidemiology of neurofibromatosis type 1. Am J Med Genet. 29. Prada CE, Rangwala FA, Martin LJ, et al. Pediatric plexiform
1999;89:1–6. neurofibromas: impact on morbidity and mortality in neurofibromatosis type
7. Rasmussen SA, Friedman JM. NF1 gene and neurofibromatosis 1. Am J 1. J Pediatr. 2012;160:461–467.
Epidemiol. 2000;151:33–40. 30. Evans DG, Baser ME, McGaughran J, et al. Malignant peripheral nerve
8. Clementi M, Barbujani G, Turolla L, et al. Neurofibromatosis-1: a maximum sheath tumours in neurofibromatosis 1. J Med Genet. 2002;39:311–314.
likelihood estimation of mutation rate. Hum Genet. 1990;84:116–118. 31. Tovmassian D, Abdul Razak M, London K. The role of [18F]FDG-PET/
9. Brems H, Beert E, de Ravel T, et al. Mechanisms in the pathogenesis of CT in predicting malignant transformation of plexiform neurofibromas in
malignant tumours in neurofibromatosis type 1. Lancet Oncol. 2009;10: neurofibromatosis-1. Int J Surg Oncol. 2016;2016:6162182.
508–515. 32. Stucky CC, Johnson KN, Gray RJ, et al. Malignant peripheral nerve
10. Marchuk DA, Saulino AM, Tavakkol R, et al. cDNA cloning of the type sheath tumors (MPNST): the Mayo Clinic experience. Ann Surg Oncol.
1 neurofibromatosis gene: complete sequence of the NF1 gene product. 2011;19:878–885.
Genomics. 1991;11:931–940. 33. Needle MN, Cnaan A, Dattilo J, et al. Prognostic signs in the surgical
11. Cawthon RM, Weiss R, Xu GF, et al. A major segment of the management of plexiform neurofibroma: the Children’s Hospital of
neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and Philadelphia experience, 1974-1994. J Pediatr. 1997;131:678–682.
point mutations. Cell. 1990;62:193–201. 34. Robertson KA, Nalepa G, Yang FC, et al. Imatinib mesylate for plexiform
12. Rodriguez F, Stratakis CA, Evans DG. Genetic predisposition to peripheral neurofibromas in patients with neurofibromatosis type 1: a phase 2 trial.
nerve neoplasia: diagnostic criteria and pathogenesis of neurofibromatoses, Lancet Oncol. 2012;13:1218–1224.
Carney complex, and related syndromes. Acta Neuropathol. 2012;123: 35. Jakacki RI, Dombi E, Potter DM, et al. Phase I trial of pegylated interferon-
349–367. alpha-2b in young patients with plexiform neurofibromas. Neurology.
13. Buchanan ME, Davis RL. A distinct set of Drosophila brain neurons 2011;76:265–272.
required for neurofibromatosis type 1-dependant learning and memory. J 36. Widemann BC, Acosta MT, Ammoun S, et al. CTF meeting 2012:
Neurosci. 2010;30:10135–10143. translation of the basic understanding of the biology and genetics of NF1,
14. Warrington NM, Gianino SM, Jackson E, et al. Cyclic AMP suppression is NF2, and schwannomatosis toward the development of effective therapies.
sufficient to induce gliomagenesis in a mouse model of neurofibromatosis-1. Am J Med Genet A. 2014;164A:563–578.
Cancer Res. 2010;70:5717–5727. 37. Jessen WJ, Miller SJ, Jousma E, et al. MEK inhibition exhibits efficacy in
15. Neurofibromatosis. Conference statement. National Institutes of Health human and mouse neurofibromatosis tumors. J Clin Invest. 2013;123:
Consensus Development Conference. Arch Neurol. 1988;45:575–578. 340–347.
16. DiPaolo DP, Zimmerman RA, Rorke LB, et al. Neurofibromatosis type 1: 38. Patel AV, Eaves D, Jessen WJ, et al. Ras-driven transcriptome analysis
pathologic substrate of high-signal-intensity foci in the brain. Radiology. identifies aurora kinase A as a potential malignant peripheral nerve sheath
1995;195:721–724. tumor therapeutic target. Clin Cancer Res. 2012;18:5020–5030.
17. Sabbagh A, Pasmant E, Imbard A, et al. NF1 molecular characterization 39. Johannessen CM, Johnson BW, Williams SM, et al. TORC1 is essential for
and neurofibromatosis type 1 genotype-phenotype correlation: the French NF1-associated malignancies. Curr Biol. 2008;18:56–62.
experience. Hum Mutat. 2013;34:1510–1518. 40. De Raedt T, Walton Z, Yecies JL, et al. Exploiting cancer cell
18. Riccardi VM. Von Recklinghausen neurofibromatosis. N Engl J Med. vulnerabilities to develop a combination therapy for ras-driven tumors.
1981;305:1617–1627. Cancer Cell. 2011;20:400–413.
19. De Luca A, Bottillo I, Sarkozy A, et al. NF1 gene mutations represent the 41. Freeman D. Neurofibroma of the choroid. Arch Ophthalmol. 1934;11:
major molecular event underlying neurofibromatosis-Noonan syndrome. Am 641–645.
J Hum Genet. 2005;77:1092–1101. 42. Woog JJ, Albert DM, Craft J, et al. Choroidal ganglioneuroma in
20. Lubs ML, Bauer MS, Formas ME, et al. Lisch nodules in neurofibromatosis neurofibromatosis. Graefes Arch Clin Exp Ophthalmol. 1983;220:25–31.
type 1. N Engl J Med. 1991;324:1264–1266. 43. Kurosawa A, Kurosawa H. Ovoid bodies in choroidal neurofibromatosis.
21. Williamson TH, Garner A, Moore AT. Structure of Lisch nodules in Arch Ophthalmol. 1982;100:1939–1941.
neurofibromatosis type 1. Ophthalmic Paediatr Genet. 1991;12:11–17. 44. Yasunari T, Shiraki K, Hattori H, et al. Frequency of choroidal
22. Ragge NK, Falk RE, Cohen WE, et al. Images of Lisch nodules across the abnormalities in neurofibromatosis type 1. Lancet. 2000;356:988–992.
spectrum. Eye (Lond). 1993;7:95–101. 45. Nakakura S, Shiraki K, Yasunari T, et al. Quantification and anatomic
23. Ferner RE, Huson SM, Thomas N, et al. Guidelines for the diagnosis distribution of choroidal abnormalities in patients with type I
and management of individuals with neurofibromatosis 1. J Med Genet. neurofibromatosis. Graefes Arch Clin Exp Ophthalmol. 2005;243:

70 | www.apjo.org © 2019 Asia-Pacific Academy of Ophthalmology

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019 Neurofibromatosis Type 1

980–984. Am J Med Genet A. 2003;122A:95–99.


46. Viola F, Villani E, Natacci F, et al. Choroidal abnormalities detected 67. Listernick R, Charrow J, Greenwald MJ, et al. Optic gliomas in children
by near-infrared reflectance imaging as a new diagnostic criterion for with neurofibromatosis type 1. J Pediatr. 1989;114:788–792.
neurofibromatosis 1. Ophthalmology. 2012;119:369–375. 68. Listernick R, Darling C, Greenwald M, et al. Optic pathway tumors in
47. Vagge A, Camicione P, Capris C, et al. Choroidal abnormalities in children: the effect of neurofibromatosis type 1 on clinical manifestations
neurofibromatosis type 1 detected by near-infrared reflectance imaging in and natural history. J Pediatr. 1995;127:718–722.
paediatric population. Acta Ophthalmol. 2015;93:e667–e671. 69. Cavicchiolo ME, Opocher E, Daverio M, et al. Diencephalic syndrome as
48. Abdolrahimzadeh S, Felli L, Plateroti R, et al. Morphologic and vasculature sign of tumor progression in a child with neurofibromatosis type 1 and optic
features of the choroid and associated choroid-retinal thickness alterations pathway glioma: a case report. Childs Nerv Syst. 2013;29:1941–1945.
in neurofibromatosis type 1. Br J Ophthalmol. 2015;99:789–793. 70. Avery RA, Fisher MJ, Liu GT. Optic pathway gliomas. J Neuroophthalmol.
49. Rao RC, Choudhry N. Enhanced depth imaging spectral-domain optical 2011;31:269–278.
coherence tomography findings in choroidal neurofibromatosis. Ophthalmic 71. Imes RK, Hoyt WF. Magnetic resonance imaging signs of optic nerve
Surg Lasers Imaging Retina. 2014;45:466–468. gliomas in neurofibromatosis 1. Am J Ophthalmol. 1991;111:729–734.
50. Huson S, Jones D, Beck L. Ophthalmic manifestations of neurofibromatosis. 72. Brodsky MC. The “pseudo-CSF” signal of orbital optic glioma on magnetic
Br J Ophthalmol. 1987;71:235–238. resonance imaging: a signature of neurofibromatosis. Surv Ophthalmol.
51. Edward DP, Morales J, Bouhenni RA, et al. Congenital ectropion uvea 1993;38:213–218.
and mechanisms of glaucoma in neurofibromatosis type 1: new insights. 73. Korf BR. Malignancy in neurofibromatosis type 1. Oncologist. 2000;5:
Ophthalmology. 2012;119:1485–1494. 477–485.
52. Al Freihi SH, Edward DP, Nowilaty SR, et al. Iris neovascularization and 74. Dodgshun AJ, Maixner WJ, Heath JA, et al. Single agent carboplatin for
neovascular glaucoma in neurofibromatosis type 1: report of 3 cases in pediatric low-grade glioma: a retrospective analysis shows equivalent
children. J Glaucoma. 2013;22:336–341. efficacy to multiagent chemotherapy. Int J Cancer. 2016;138:481–488.
53. Morales J, Chaudhry IA, Bosley TM. Glaucoma and globe enlargement 75. Packer RJ, Jakacki R, Horn M, et al. Objective response of multiply
associated with neurofibromatosis type 1. Ophthalmology. 2009;116: recurrent low-grade gliomas to bevacizumab and irinotecan. Pediatr Blood
1725–1730. Cancer. 2009;52:791–795.
54. Heervä E, Koffert A, Jokinen E, et al. A controlled register-based study of 76. Avery RA, Hwang EI, Jakacki RI, et al. Marked recovery of vision in
460 neurofibromatosis 1 patients: increased fracture risk in children and children with optic pathway gliomas treated with bevacizumab. JAMA
adults over 41 years of age. J Bone Miner Res. 2012;27:2333–2337. Ophthalmol. 2014;132:111–114.
55. Raj A, Baidya K, Ghosh S, et al. Neurofibromatosis 1. Ophthalmology. 77. Banerjee A, Jakacki RI, Onar-Thomas A, et al. A phase I trial of the MEK
2009;116:598–598.e1. inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or
56. Wu X, Estwick SA, Chen S, et al. Neurofibromin plays a critical role in refractory low-grade glioma: a Pediatric Brain Tumor Consortium (PBTC)
modulating osteoblast differentiation of mesenchymal stem/progenitor cells. study. Neuro Oncol. 2016;19:1135–1144.
Hum Mol Genet. 2006;15:2837–2845. 78. Balachandran C, Klistorner AI, Billson F. Multifocal VEP in children: its
57. Rakotonjanahary J, De Carli E, Delion M, et al. Mortality in children with maturation and clinical application. Br J Ophthalmol. 2004;88:226–232.
optic pathway glioma treated with up-front BB-SFOP chemotherapy. PLoS 79. Klistorner AI, Graham SL, Grigg J, et al. Objective perimetry using the
One. 2015;10:e0127676. multifocal visual evoked potential in central visual pathway lesions. Br J
58. Listernick R, Ferner RE, Liu GT, et al. Optic pathway gliomas in Ophthalmol. 2005;89:739–744.
neurofibromatosis-1: controversies and recommendations. Ann Neurol. 80. Chang L, El-Dairi MA, Frempong TA, et al. Optical coherence tomography
2007;61:189–198. in the evaluation of neurofibromatosis type-1 subjects with optic pathway
59. de Blank PM, Fisher MJ, Liu GT, et al. Optic pathway gliomas in gliomas. J AAPOS. 2010;14:511–517.
neurofibromatosis type 1: an update: surveillance, treatment indications, and 81. Gutmann DH, Rasmussen SA, Wolkenstein P, et al. Gliomas presenting
biomarkers of vision. J Neuroophthalmol. 2017;37 Suppl 1:S23–S32. after age 10 in individuals with neurofibromatosis type 1 (NF1). Neurology.
60. Thiagalingam S, Flaherty M, Billson F, et al. Neurofibromatosis type 1 2002;59:759–761.
and optic pathway gliomas: follow-up of 54 patients. Ophthalmology. 82. Milstein JM, Geyer JR, Berger MS, et al. Favorable prognosis for brainstem
2004;111:568–577. gliomas in neurofibromatosis. J Neurooncol. 1989;7:367–371.
61. Fisher MJ, Avery RA, Allen JC, et al. Functional outcome measures for 83. Sharif S, Moran A, Huson SM, et al. Women with neurofibromatosis 1 are
NF1-associated optic pathway glioma clinical trials. Neurology. 2013;81(21 at a moderately increased risk of developing breast cancer and should be
Suppl 1):S15–S24. considered for early screening. J Med Genet. 2007;44:481–484.
62. Dodge HW Jr, Love JG, Craig WM, et al. Gliomas of the optic nerves. AMA 84. Huson SM, Hughes RA, eds. The Neurofibromatoses: A Pathogenic and
Arch Neurol Psychiatry. 1958;79:607–621. Clinical Overview. London: Chapman and Hall Medical; 1994.
63. Taylor T, Jaspan T, Milano G, et al. Radiological classification of optic 85. Johnson H, Wiggs L, Stores G, et al. Psychological disturbance and sleep
pathway gliomas: experience of a modified functional classification system. disorders in children with neurofibromatosis type 1. Dev Med Child Neurol.
Br J Radiol. 2008;81:761–766. 2005;47:237–242.
64. Segal L, Darvish-Zargar M, Dilenge ME, et al. Optic pathway gliomas in 86. Brei NG, Klein-Tasman BP, Schwarz GN, et al. Language in young children
patients with neurofibromatosis type 1: follow-up of 44 patients. J AAPOS. with neurofibromatosis-1: relations to functional communication, attention,
2010;14:155–158. and social functioning. Res Dev Disabil. 2014;35:2495–2504.
65. Parazzini C, Triulzi F, Bianchini E, et al. Spontaneous involution of optic 87. Lin AE, Birch PH, Korf BR, et al. Cardiovascular malformations and other
pathway lesions in neurofibromatosis type 1: serial contrast MR evaluation. cardiovascular abnormalities in neurofibromatosis 1. Am J Med Genet.
AJNR Am J Neuroradiol. 1995;16:1711–1718. 2000;95:108–117.
66. King A, Listernick R, Charrow J, et al. Optic pathway gliomas in 88. Terry AR, Jordan JT, Schwamm L, et al. Increased risk of cerebrovascular
neurofibromatosis type 1: the effect of presenting symptoms on outcome. disease among patients with neurofibromatosis type 1: population-based

© 2019 Asia-Pacific Academy of Ophthalmology www.apjo.org | 71

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Karaconji et al Asia-Pacific Journal of Ophthalmology • Volume 8, Number 1, January/February 2019

approach. Stroke. 2016;47:60–65. 91. Fisher MJ, Loguidice M, Gutmann DH, et al. Visual outcomes in children
89. Rosser TL, Vezina G, Packer RJ. Cerebrovascular abnormalities in with neurofibromatosis type 1-associated optic pathway glioma following
a population of children with neurofibromatosis type 1. Neurology. chemotherapy: a multicenter retrospective analysis. Neuro Oncol.
2005;64:553–555. 2012;14:790–797.
90. Koss M, Scott RM, Irons MB, et al. Moyamoya syndrome associated 92. Peltonen S, Kallionpää RA, Peltonen J. Neurofibromatosis type 1 (NF1)
with neurofibromatosis type 1: perioperative and long-term outcome after gene: beyond café au lait spots and dermal neurofibromas. Exp Dermatol.
surgical revascularization. J Neurosurg Pediatr. 2013;11:417–425. 2016;26:645–648.

72 | www.apjo.org © 2019 Asia-Pacific Academy of Ophthalmology

Copyright © 2019 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.

You might also like