You are on page 1of 9

REVIEW ARTICLE

Retinoblastoma for Pediatric Ophthalmologists


Alaa AlAli, MD, FRCSC,* Stephanie Kletke, MD,*
Brenda Gallie, MD, FRCSC,*†‡§ and Wai-Ching Lam, MD, FRCSC†¶

and later to visceral organs. Late diagnosis is therefore associated


Abstract: Retinoblastoma can present in 1 or both eyes and is the most with high morbidity and mortality. We predict that improved un-
common intraocular malignancy in childhood. It is typically initiated by derstanding of retinoblastoma among primary care providers, pe-
biallelic mutation of the RB1 tumor suppressor gene, leading to malignant diatric ophthalmologists, and parents will enable early diagnosis
transformation of primitive retinal cells. The most common presentation and timely intervention. With safe, evidence-based new therapeu-
is leukocoria, followed by strabismus. Heritable retinoblastoma accounts tic approaches to clearly define disease stage, collaborative care
for 45% of all cases, with 80% being bilateral. Treatment and prognosis with Internet communication specific to retinoblastoma care3,5
of retinoblastoma is dictated by the disease stage at initial presentation. (http://depictrb.technainstitute.com/), and location of nearby cen-
The 8th Edition American Joint Committee on Cancer (AJCC) TNMH ters for retinoblastoma care worldwide (http://map.1rbw.org/), we
Downloaded from http://journals.lww.com/apjoo by BhDMf5ePHKbH4TTImqenVD7nNEO3Od0IK7zXffF07KoCI626PX2hbNB5FUQQCFK/fUCIbOgmav8= on 12/08/2019

(tumor, node, metastasis, heritable trait) staging system defines evidence- would hope that no child should die from retinoblastoma.3 The
based clinical and pathological staging for overall prognosis for eye(s) yet unaddressed major cause of later death of these children is
and child. Multiple treatment options are available in 2018 for retino- second primary malignancy.
blastoma management with a multidisciplinary team, including pediatric
ocular oncology, medical oncology, radiation oncology, genetics, nurs-
ing, and social work. Survival exceeds 95% when disease is diagnosed EPIDEMIOLOGY
early and treated in centers specializing in retinoblastoma. However, Retinoblastoma occurs worldwide at an estimated rate of 1
survival rates are less than 50% with extraocular tumor dissemination. in 16,000–18,000 live births, with approximately 8000 new cases
We summarize the epidemiology, genetics, prenatal screening, diagnosis, predicted annually.3 Mean age at diagnosis is 27 months for uni-
classification, investigations, and current therapeutic options in the man- lateral and 15 months for bilateral retinoblastoma in Canada but
agement of retinoblastoma. 36 and 25 months, respectively, in Kenya, where many children
present with extraocular disease.4 There are no ethnic or sexual
Key Words: retinoblastoma, TNMH classification, RB1 mutation predilections and no geographic or environmental associations
with retinoblastoma.
(Asia-Pac J Ophthalmol 2018;7:160–168)

GENETICS

R etinoblastoma can present in 1 or both eyes and is considered


the most common intraocular malignancy in childhood. The
main therapeutic priority for retinoblastoma is to first save the
Retinoblastoma is the prototypic genetic cancer.1 The tumors
have mutation of both copies of the RB1 tumor suppressor gene
located on the long arm of chromosome 13 (13q14), which en-
child’s life through early tumor detection and prevention of meta- codes the retinoblastoma protein (pRB). The cellular origin of ret-
static spread. Secondary goals are globe salvage and maximizing inoblastoma is likely a uniquely susceptible cone photoreceptor
visual potential. With timely intervention while tumors remain precursor cell in the infant retina, which remains within the inner
small, the survival rate for affected children is nearly 100%.1 nuclear layer after losing both alleles of the RB1 tumor suppressor
Globally, mortality is considerably higher due to delayed diag- gene.3,6 This initiates a benign precursor, “retinoma,” which usu-
nosis, poor access to multidisciplinary retinoblastoma-specific ally progresses to retinoblastoma with accumulation of increasing
health care and pathology, lack of genetic testing and counsel- genomic changes and uncontrolled cellular proliferation.7,8
ing, and socioeconomic factors.2‒4 Advanced retinoblastoma can There are 2 major genetic forms of retinoblastoma: heritable
directly invade the orbit, spread via the optic nerve to the central (also known as germline) and nonheritable. Heritable retinoblas-
nervous system, or metastasize hematogenously to bone marrow, toma accounts for 45% of all cases; 80% are bilateral, 15% unilat-
eral, and 5% trilateral (bilateral retinoblastoma with pineal/mid-
line neuroectodermal tumor).1 One RB1 allele is mutated in all
From the *Department of Ophthalmology & Vision Sciences, University of Toronto,
Toronto; †Department of Ophthalmology & Vision Sciences, The Hospital
cells and a second somatic mutagenic event affects the remaining
for Sick Children, Toronto; ‡Techna Institute, University Health Network, allele in a primitive retinal cell (known as the “2-hit model”),
Toronto; §Departments of Molecular Genetics and Medical Biophysics,
University of Toronto, Toronto, Canada; and ¶Department of Ophthalmology,
thus initiating tumorigenesis.9 A mutated RB1 allele is inherited
The University of Hong Kong, Hong Kong. in only 5% of children with heritable retinoblastoma.1 Children of
Received for publication February 28, 2018; accepted April 23, 2018.
A.A. and S.K. contributed equally to this review as co-first authors.
a parent with heritable retinoblastoma have a 45% chance of be-
B.L.G. is unpaid Medical Director at Impact Genetics. ing affected (50% risk of inheritance and 90% penetrance). How-
The authors have no other funding or conflicts of interest to declare.
Reprints: Wai-Ching Lam, MD, FRCSC, Room 301, Level 3, Block B, Cyberport
ever, most heritable retinoblastoma arises de novo and the RB1
4, 100 Cyberport Road, Hong Kong. E‑mail: waichlam@hku.hk. mutation may be present in all cells (preconception mutation) or
Copyright © 2018 by Asia Pacific Academy of Ophthalmology
ISSN: 2162-0989
only a subset of cells, termed “mosaicism” (postconception muta-
DOI: 10.22608/APO.201870 tion) (Fig. 1).1,10,11 Most children with retinoblastoma are the first

160 | www.apjo.org © 2018 Asia-Pacific Academy of Ophthalmology

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 Retinoblastoma for Pediatric Ophthalmologists

A B

C D

FIGURE 1. A, Pedigree of a family with a unilaterally affected father discovered to be mosaic (H1) for a high penetrance RB1 mutation when his second
child was found to have bilateral retinoblastoma (H1).11 The child had late diagnosis requiring intensive therapy, with suboptimal visual prognosis.
B, DePICTRB visualization3,5 demonstrating the child’s follow-up and treatment timeline. C, At presentation, the right eye was staged TNMH29 cT2b and
IIRC25 Group D and the left eye TNMH29 cT1b and IIRC25 Group B. D, After extensive treatments, there was tumor regression with inactive scars bilaterally.
Visual acuity was fix and follow (F&F) in the right eye and 0.2 logarithm of the minimum angle of resolution (logMAR) by Cardiff in the left eye.

family member (proband) with the disease. Children with herita- villus sampling (between 11‒14 weeks’ gestation) or amniocen-
ble retinoblastoma are usually diagnosed earlier and present with tesis (after 16 weeks’ gestation) may be considered so parents
bilateral, multifocal tumors. Frequent examinations under anes- have choices of how to manage the pregnancy if the fetus has the
thesia (EUA) for retinal examination with scleral indentation are RB1 mutation. Amniocentesis can also be offered at 33 weeks’
recommended up to at least 3 years of age to treat tumors when gestation when the risks of miscarriage are lower and manage-
they are as small as possible with minimally invasive therapies.12 able.1 Examination for tumors as soon as possible after birth, fol-
Persons with RB1 germline predisposition also have a high life- lowed by repeated EUA for the first few years of life, facilitates
time risk of developing second primary malignancies, especially early detection of tumors that can be managed with less invasive
osteosarcomas, soft-tissue sarcomas, and melanomas.13‒15
Approximately 55% of cases have nonheritable retinoblasto-
ma that is always unilateral. Of nonheritable retinoblastoma, 98%
result from somatic loss of both RB1 alleles in a susceptible reti-
nal precursor cell, but 2% have normal RB1 alleles (RB1+/+) and
are initiated by somatic amplification of the MYCN oncogene.16
These tumors are always unilateral, nonheritable, and diagnosed
at median 4.5 months.
Genetic testing and counseling are extremely important in
the management of retinoblastoma, involving a laboratory with
high sensitivity to detect RB1 mutations.12 RB1 genetic testing of
the proband may identify heritability, and at-risk family members
may then undergo testing for the specific mutation. Children of a
proband with bilateral retinoblastoma have a 50% risk of carrying
the RB1 mutation, whereas children of an untested unilateral pro-
band have a 7.5% risk (50% × 15%), which can be accurately up-
dated to either 100% or 0% by genetic testing of the proband and FIGURE 2. The Internet and public media now help with awareness
then the fetus if the proband is found to carry an RB1 mutation.11 of retinoblastoma. This mother noticed photoleukorcoria when
The frequency of follow-up for children at risk of retinoblastoma she downloaded pictures from her camera and took her son to the
is informed by the results of genetic testing and counseling. emergency department the next day when Internet searches revealed
that it could be something sinister, like retinoblastoma. Once their son
was diagnosed, the family wanted to spread the word by contacting
PRENATAL SCREENING media outlets. This Finnish newspaper featured the photoleukocoria
Children with a family history of retinoblastoma who car- photographs of the child in July, which directly resulted in another child
ry the RB1 mutation are at risk of tumors at birth. Chorionic being diagnosed with retinoblastoma in September.

Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 www.apjo.org | 161

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
AlAli et al Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018

interventions.17 Infants who had prenatal RB1 mutation detection Retinoblastoma may be diagnosed during an office eye ex-
followed by early full-term delivery (36‒38 weeks’ gestation) and amination by a general doctor or pediatrician. All children with
coordinated retinal examination had smaller tumors at birth and high suspicion for retinoblastoma should immediately be re-
improved visual outcomes. ferred to a retinoblastoma center for diagnostic confirmation and
a staging EUA.24 This also includes a detailed fundus diagram
during dilated indirect ophthalmoscopy with scleral indentation
DIAGNOSIS 360 degrees to view the entire retina, documenting the location
Retinoblastoma most commonly presents with leukocoria and size of all tumors relative to the fovea and optic nerve, pres-
(white pupillary reflex), resulting from light reflecting off the tu- ence of calcification, subretinal fluid, and vitreous and/or subreti-
mor causing a “glint,” “cat’s eye reflex,” or other similar descrip- nal seeding.
tions by parents and other people caring for the baby (Fig. 2).4,18‒20
Retinoblastoma must be the key differential diagnosis suspected
by the primary care physician or pediatric ophthalmologist when CLASSIFICATION
parents report an abnormal “glow” or “squint.” Too commonly, Treatment and prognosis of retinoblastoma depend on the
the primary physician has forgotten about retinoblastoma and staging at initial presentation. Tumor size, location, presence of
does not react with immediate concern to the observation of the subretinal fluid and/or seeding, and histopathological features are
parent, leading to significant delay in diagnosis, despite the strong all predictors of overall outcome. A multitude of different classifi-
guidelines for “red reflex” testing as part of routine care.21 cation systems for retinoblastoma has led to significant confusion
Leukocoria is now commonly detected as a white reflection in the literature, preventing proper comparison among studies and
by flash photography, termed “photoleukocoria” (Fig. 2).22,23 Stra- confounding patient care. The International Intraocular Retino-
bismus is the second most common presentation, which occurs blastoma Classification (IIRC) staged eyes as Group A (very low
with vision loss secondary to central macular involvement.4,18,19 risk) through E (very high risk).25 A subsequent modification of

B C D

E F G

FIGURE 3. Presentation and staging of eyes with retinoblastoma. A, Fundus photograph of a clinically invisible solitary tumor (TNMH29 cT1a, IIRC25 Group
A) detected by OCT at 38 weeks’ gestation, 15 days of age; the child was diagnosed in utero to be H1, her mother’s RB1 mutation. B, As the tumor grows,
it gains blood supply and becomes globular. This fundus photo demonstrates a large central perifoveal tumor and multiple smaller tumors; yellow luteal
pigment at the inferior edge of the central tumor indicates the foveal location. There was less than 5 mm associated subretinal fluid and no evidence of
seeding (TNMH29 cT1b, IIRC25 Group B). C, D, Tumor extends beyond the inner limiting membrane giving rise to vitreous seeding and into the subretinal
space with seeding (TNMH29 cT2b, IIRC25 Group D). E, Fundus photograph of a total exudative retinal detachment overlying a large inferior tumor
(TNMH29 cT2a, IIRC25 Group D). F, Ultrasound biomicroscopy demonstrating an anteriorly situated tumor adjacent to the ciliary body (TNMH29 cT3, IIRC25
Group E); histopathology after enucleation confirmed invasion into the ciliary muscle. G, Unilateral retinoblastoma can mimic advanced Coats disease,
particularly in the presence of retinal detachment. This child was initially diagnosed with Coats disease based on total exudative retinal detachment
without calcification, but vitreous and subretinal seeding were evident during EUA and retinoblastoma diagnosis was confirmed after enucleation.43

162 | www.apjo.org © 2018 Asia-Pacific Academy of Ophthalmology

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 Retinoblastoma for Pediatric Ophthalmologists

this classification26 resulted in severely advanced eyes being clas- calcification (Fig. 3B). More advanced tumors may have asso-
sified with less-affected eyes, so that “Group E” no longer was ciated vitreous seeding (tumor cells extending beyond the inner
understood to designate eyes in danger of extraocular disease that limiting membrane into the vitreous cavity) (Fig. 3C) and subreti-
should be promptly enucleated.3,27,28 nal seeding (tumor extension into the subretinal space) (Fig. 3D),
The 2017 American Joint Committee on Cancer (AJCC) 8th along with exudative retinal detachment (Fig. 3E).
edition TNMH (tumor, node, metastasis, heritable trait) clinical Advanced retinoblastoma may present with anterior chamber
and pathological staging system is the first evidence-based sys- invasion, elevated intraocular pressure with neovascularization,
tem for predicting overall prognosis of both eye(s) and patients hyphema, vitreous hemorrhage, cataract, or aseptic orbital celluli-
(Fig. 3, Table 1).29 It is informed by an international survey of tis. The most dangerous presentation is aseptic orbital cellulitis,30
1728 eyes with retinoblastoma comparing all the previous clas- often due to central retinal arterial occlusion at the optic nerve
sifications and best separates severity of eye disease and the like- head, causing massive acute tumor and ophthalmic necrosis.31
lihood to save the eye without use of external beam irradiation; Retinoblastoma is importantly the first cancer to recognize
the TNMH has become the gold standard to define the extent of the role of germline predisposition by incorporating stage cat-
retinoblastoma at diagnosis in 2018. egory “H” into the AJCC classification. H1 is supported by the
Small retinoblastoma tumors initially present as round, presence of bilateral disease (Fig. 1, Table 1), a family history
gray intraretinal lesions (Fig. 3A).25 With further growth and ex- of retinoblastoma, a concomitant central nervous system midline
pansion of blood supply, the tumor assumes an irregular lobu- embryonic tumor (trilateral disease), or the presence of high-qual-
lated shape, opaque white or pink color, and may have areas of ity molecular evidence of an RB1 mutation.29 Those at unknown

TABLE 1. TNMH Clinical Staging in Comparison With IIRC Group Classification of Eyes With Intraocular Retinoblastoma

AJCC 8th Edition TNMH Staging29 IIRC Group25

cTX Unknown evidence of intraocular tumor


cT0 No evidence of intraocular tumor
cT1 Intraretinal tumor(s) with subretinal fluid <5 mm from the tumor base. No vitreous or subretinal seeding
cT1a Tumors ≤3 mm in size and >1.5 mm away from the optic disc and fovea A
cT1b Tumors >3 mm in size and <1.5 mm away from the optic disc and fovea B
cT2 Tumors with retinal detachment/subretinal seeding/vitreous seeding
cT2a Subretinal fluid >5 mm from the tumor base C/D
cT2b Tumors with vitreous seeding and/or subretinal seeding C/D/E
cT3 Advanced intraocular tumor(s) E
cT3a Phthisis or prephthisis bulbi
cT3b Tumor invasion of choroid, pars plana, ciliary body, lens, zonules, iris, or anterior chamber
cT3c Raised intraocular pressure with neovascularization and/or buphthalmos
cT3d Hyphema and/or massive vitreous hemorrhage
cT3e Aseptic orbital cellulitis
cT4 Extraocular tumor(s) involving orbit, including optic nerve
HX Unknown or insufficient evidence of RB1 constitutional mutation
H0 Normal RB1 alleles in blood
H0* Normal RB1 in blood with <1% residual risk mosaicism
H1 Bilateral retinoblastoma, trilateral retinoblastoma, family history of retinoblastoma, or molecular definition of constitutional
RB1 gene mutation
pTX Unknown evidence of intraocular tumor
pT0 No evidence of intraocular tumor
pT1 Intraocular tumor(s) without any local invasion, focal choroidal invasion, or pre- or intralaminar involvement of the optic
nerve head
pT2 Intraocular tumor(s) with local invasion
pT2a Concomitant focal choroidal invasion and pre- or intralaminar involvement of the optic nerve head
pT2b Tumor invasion of stroma of iris and/or trabecular meshwork and/or Schlemm canal
pT3 Intraocular tumor(s) with significant local invasion
pT3a Massive choroidal invasion (>3 mm in largest diameter, or multiple foci of focal choroidal involvement totaling >3 mm, or
any full-thickness choroidal involvement)
pT3b Retrolaminar invasion of the optic nerve head, not involving the transected end of the optic nerve
pT3c Any partial-thickness involvement of the sclera within the inner two thirds
pT3d Full-thickness invasion into the outer third of the sclera and/or invasion into or around emissary channels
pT4 Evidence of extraocular tumor: tumor at the transected end of the optic nerve, tumor in the meningeal spaces around the
optic nerve, full-thickness invasion of the sclera with invasion of the episclera, adjacent adipose tissue, extraocular
muscle, bone, conjunctiva, or eyelids

Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 www.apjo.org | 163

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
AlAli et al Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018

risk or who have variants of unknown significance are catego- changing (17%) clinical decisions.35 Additionally, OCT is ex-
rized HX. A family member who tests negative for the known tremely helpful in the detection of small invisible tumors that
RB1 mutation of the proband is considered at population risk cannot be identified by indirect ophthalmoscopy alone (Fig.
(0.007%) and may be assigned H0. If no mutation is found in a 3A).3,35‒38
proband or an unaffected parent of a proband in a lab with sensi- For advanced retinoblastoma, ultrasound biomicroscopy
tivity exceeding 97% for bilaterally affected patients, the residual (UBM) is valuable in the precise evaluation of anterior segment
risk for undetectable mosaicism is less than 1% and the designa- and ciliary body extension (Fig. 3F). In these cases, UBM may
tion H0* is suggested.1 provide useful clinical information for management, such as indi-
Pathological staging (pTNM) can only be performed after cation for enucleation.39
enucleation of the affected eye. Features such as massive cho- Magnetic resonance imaging (MRI) of the brain and orbit
roidal invasion (more than 3 mm in largest diameter, or multiple is preferred over computed tomography (CT) to evaluate optic
foci of focal choroidal involvement totaling more than 3 mm, or nerve and orbital extension and intracranial involvement and to
any full-thickness choroidal involvement) (Fig. 4), retrolaminar rule out trilateral disease at initial presentation. The use of MRI
invasion of the optic nerve head, scleral invasion, or evidence offers better soft-tissue resolution and avoids the radiation of CT,
of extraocular tumor confer high risk for metastases.29 The pres- which increases the risk of second primary malignancy in patients
ence of such high-risk features (pT3, pT4) warrants metastatic with germline predisposition to cancer because of their RB1 mu-
surveillance with cerebrospinal fluid and bone marrow analyses tation. The diagnostic sensitivity of MRI to detect high-risk fea-
and prophylactic adjuvant systemic chemotherapy.32,33 tures, including postlaminar optic nerve, choroidal, and scleral
invasion, is reportedly 59%, 74%, and 88%, respectively.40

INVESTIGATIONS
Diagnosis of retinoblastoma is made clinically and does not DIFFERENTIAL DIAGNOSIS
rely on histopathology; diagnostic fine needle aspiration biopsy Multiple ocular conditions that present with leukocoria or
would impose risk of systemic tumor dissemination.7 B-scan ul- strabismus may mimic retinoblastoma and may be differenti-
trasonography can assess tumor size and calcification, help to ated by a comprehensive history, clinical exam, and appropriate
distinguish retinoblastoma from other pathologies such as Coats testing.
disease and persistent fetal vasculature (PFV), and can be per- Generally, PFV and Coats disease are considered the most
formed with the child awake in the office or at the staging EUA. common differential diagnoses for unilateral retinoblastoma.19
Wide-field fundus photography with scleral depression to view Of these, PFV is a sporadic developmental ocular disorder re-
the entire retina during the staging EUA will document the ex- sulting from failure of normal hyaloid vasculature regression.41
tent and location of the tumor(s); accurate fundus drawings aid in It often presents with leukocoria in the first days to weeks of life.
monitoring response of tumors to treatment that may alter future Two thirds of cases are unilateral and may be associated with mi-
management. crophthalmos, retrolenticular fibrovascular mass, elongated cili-
Handheld optical coherence tomography (OCT) performed ary processes, cataract, and a funnel-shaped retinal detachment
on the supine child is valuable in the diagnosis and management in advanced cases. A characteristic vascular stalk extending from
of retinoblastoma for localization of the tumor, detection of optic the optic nerve to the posterior lens capsule is usually noted on
nerve infiltration, evaluation of scars and retinal structure after clinical exam and/or B-scan ultrasonography.
treatment, and assessment of foveal integrity, which is impor- Coats disease is a rare, unilateral, idiopathic nonhereditary
tant to assess visual potential.34 In a study of 339 OCT sessions, exudative retinopathy presenting most commonly in males dur-
OCT was found to play an important role in confirming (83%) or ing the first or second decade of life.42 It presents with a “yellow”

A B C

FIGURE 4. High-risk histopathological features predictive of increased metastatic risk. A, Low magnification histopathological section of a TNMH29 cT2b
(IIRC25 Group D) eye demonstrating massive choroidal invasion, transscleral and extrascleral invasion, and tumor extending beyond the lamina cribrosa.
Clinically, the tumor was multilobulated with overlying exudative retinal detachment and extensive subretinal seeding; optic nerve and fovea were
not visualized at presentation. B, Histopathological section under high magnification demonstrating full thickness tumor invasion of sclera, adjacent
episclera, and adipose tissue (TNMH29 pT4). C, Histopathological section under high magnification showing post‒lamina cribrosa invasion; the tumor did
not extend to the resection margin of the optic nerve.

164 | www.apjo.org © 2018 Asia-Pacific Academy of Ophthalmology

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 Retinoblastoma for Pediatric Ophthalmologists

pupillary reflex due to intraretinal and/or subretinal exudation visual function. Many treatment options are available and depend
secondary to retinal telangiectasia, often associated with serous on the laterality and extent of disease (Fig. 5). A multidisciplinary
retinal detachment (Fig. 3G). Subretinal lipid deposits can be team approach, including 1 or more ophthalmologists focused on
seen on ultrasonography, and fluorescein angiography shows the care for children with retinoblastoma, pediatric medical oncolo-
classic telangiectatic vessels. Clinically distinguishing Coats dis- gist, pediatric radiation oncologist, geneticist, social worker, and
ease from retinoblastoma can be challenging in advanced cases others, facilitates optimized care. Retinoblastoma only leads to
with little potential for useful vision. Enucleation is a reasonable death when the disease escapes the confines of the eye; delayed
diagnostic/treatment option given the life-threatening potential of diagnosis and treatment and excessive focus on saving a blind eye
undiagnosed retinoblastoma.43 with advanced disease put the child at risk for extraocular exten-
Congenital cataracts may present with leukocoria but the re- sion, metastases, and death.
mainder of the ocular exam is usually normal, and B-scan ultra- Primary enucleation remains the definitive treatment for ad-
sonography shows no posterior abnormalities. Severe retinopathy vanced unilateral retinoblastoma (TNMH29 cT3 and IIRC25 Group
of prematurity, incontinentia pigmenti, and Norrie disease may E eyes), allowing the child to return to normal life and enabling
progress to retinal detachment and leukocoria. Infectious causes histopathological analysis.44 For unilateral retinoblastoma, enu-
of leukocoria that can mimic retinoblastoma include ocular toxo- cleation is the definitive treatment when the optic nerve is not
cariasis and toxoplasmosis, typically in older children with histo- visible and in the presence of retinal detachment (TNMH29 cT2a
ry of soil or contaminated food ingestion or exposure to animals. and IIRC25 Group D eyes) or extensive vitreous seeds (TNMH29
They typically present with uveitis and posterior or peripheral cT2b and IIRC25 Group D eyes) that otherwise require invasive
subretinal granulomas.17 treatments over several years that are costly to the child and fam-
Other benign tumors such as astrocytic hamartoma and me- ily to save an eye with poor vision. Enucleation is strongly rec-
dulloepithelioma are included in the differential diagnosis of ommended when orbital or optic nerve involvement is suspected,
retinoblastoma. Myelinated nerve fiber layer, morning glory syn- when there is anterior segment invasion, neovascular glaucoma,
drome, traumatic vitreous hemorrhage, coloboma of the choroid intraocular hemorrhage, orbital cellulitis, and no potential for
or optic disc, along with photographic artifact from light hitting useful vision.
the optic nerve may also present with leukocoria. However, as The enucleation technique for retinoblastoma minimizes
retinoblastoma is a potentially life-threatening diagnosis, leuko- the potential for globe penetration while obtaining 8–12 mm of
coria requires urgent referral for full expert fundus examination the optic nerve. Postoperative movement of the prosthesis is bet-
and diagnosis.19 ter with the myoconjunctival technique of a simple polymethyl
Retinoma is not a differential diagnosis for retinoblastoma, methacrylate implant and rectus muscles sutured to the conjunc-
as it is on a continuous spectrum of malignancy from benign pre- tival fornices than with integrated implants with muscles imbri-
malignant to malignant retinoblastoma, and most retinomas in- cated to the implant.45
deed progress to malignancy.7,8 Attempts to salvage an eye require consideration of stag-
ing, visual potential, and considerable discussion with the child’s
family. Although small tumors may be managed with focal thera-
2018 MANAGEMENT OPTIONS pies alone, chemotherapy with focal consolidation is required for
FOR RETINOBLASTOMA eyes with tumors too large for primary focal therapy or too close
The primary goal of retinoblastoma management is to save to the optic nerve and macula (TNMH29 cT1b, cT2 and IIRC25
the child’s life, followed by salvage of the eye and optimization of Group B, C, D eyes). Chemotherapy can be systemic or, recently,

FIGURE 5. The multimodal therapeutic options for intraocular retinoblastoma in 2018 include chemotherapy combined with focal therapies and surgical
approaches. A relative “weighting” of value of each therapy for the eye and patient is suggested by font size and box width. Therapies highlighted in
gray are today reserved for salvage of tumors refractory to other modalities.

Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 www.apjo.org | 165

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
AlAli et al Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018

intra-arterial in specific situations. Systemic chemotherapy (most the outcomes of IAC to eradicate the cancer efficiently are im-
commonly intravenous carboplatin, etoposide, and vincristine) pressive.47 Systemic and ocular complications are uncommon.
is given over 4 to 6 cycles (every 3‒4 weeks) depending on the Systemic complications include unsuccessful catheterization,
extent of disease. Chemotherapy reduces tumor size, and focal stroke or a neurological complication (rare), groin hematoma,
therapy on repeated follow-up EUAs destroys remnants of tumor fever, neutropenia, bronchospasm, nausea, and vomiting. Ocular
(Fig. 6A). Handheld OCT is excellent to accurately assess tumor complications include central retinal artery occlusion, optic atro-
activity related to focal therapy scars.35 phy, extraocular muscle dysfunction, retinal detachment, vitreous
Intra-arterial chemotherapy (IAC) has an important role in hemorrhage, ptosis, enophthalmos, and phthisis bulbi. Transient
unilateral cT1b and cT2 (IIRC C/D) eyes, but important long-term periorbital edema and loss of eyelashes may also be observed.
outcomes, such as visual acuity, extraocular disease, and death, Incompletely reported are extraocular tumor and metastasis.
have not yet been scientifically compared.46 Characteristically the Focal therapies include laser photocoagulation (Fig. 6) and
choroid shows changes after IAC (Fig. 6C), so the lifelong vision cryotherapy.48 Laser photocoagulation involves argon laser in-
outcome will not be known until there is long-term follow-up and direct (532 nm) and transpupillary indirect or transcleral diode
will best be determined by prospective studies. Intra-arterial che- (810 nm) laser to first destroy the tumor blood supply, then di-
motherapy is excellent for children with unilateral retinoblastoma rect treatment on the regressing tumor. Laser treatment is best
[cT1b/cT2 (IIRC Groups B/C/D) eyes (Fig. 6B)] when the optic given after chemotherapy is completed, as it cuts off the access
nerve and fovea are visualized at presentation so there is no risk of chemotherapy to the tumor. Optical coherence tomography has
of masking high-risk features for extraocular extension. The most greatly improved the accuracy of laser treatment and minimized
commonly used intra-arterial chemotherapeutic agent is melpha- local complications, by both discovering tiny tumors invisible to
lan, sometimes combined with topotecan and/or carboplatin. Al- direct viewing with indirect ophthalmoscopy or cameras (Fig.
though IAC reportedly has high rates of globe salvage (66% of 3A) and accurately identifying residual or recurrent tumor.34‒36
all eyes and 57% with advanced disease), the literature is limited Cryotherapy with a triple freeze-thaw technique for small and re-
by variable staging schemes, absence of defined drug protocols, current peripheral tumors directly kills tumor cells. A single cryo-
and lack of long-term follow-up data.46 For well-selected patients, therapy freeze in peripheral retina 24‒48 hours before systemic
chemotherapy (prechemo cryotherapy) disrupts the blood-retina
barrier and increases penetration of chemotherapy.49
A Vitreous seeds are generally resistant to systemic and intra-
arterial chemotherapy mainly due to the avascular nature of the
vitreous. Intravitreal chemotherapy (melphalan and/or topotecan)
is used to control vitreous seeds, previously the most difficult
form of intraocular retinoblastoma to cure.50,51 The technique
involves an anterior chamber paracentesis or ocular massage to
reduce intraocular pressure, followed by intravitreal injection
under microscopic visualization, with 3 cycles of freeze-thaw
B cryotherapy at the injection site after needle removal. Intravit-
real melphalan injection can be repeated monthly depending on
the clinical response. The main complications include vitreous
hemorrhage, retinal tears/detachment, damage to the lens leading
to cataract, and endophthalmitis. Focal retinal pigment epithelial
changes reflect direct retinal toxicity when the drug is not well
dispersed through the vitreous.52,53 A recent retrospective study of
intravitreal chemotherapy injections in 10 retinoblastoma treat-
C ment centers worldwide found no extraocular extension of tumor
when used with precautions, such as selection of eyes where the
source of seeds is controlled, preinjection lowering of intraocular
pressure, and postinjection cryotherapy to the needle and needle
track.50,51,54
External beam radiotherapy is now only considered as a last
resort salvage option for eyes with tumors refractory to all other
FIGURE 6. Chemotherapy reduces tumor volume so that focal therapy, therapies. Radiation increases the risk of second primary malig-
such as laser photocoagulation, can be used to physically eradicate nancy with standardized incidence ratio of 20.4 for radiotherapy,
any remaining live tumor cells. A, Tumor regression after systemic increasing to 26.4 when radiation was combined with alkylating
chemotherapy and laser treatment to obtain long-term regression and chemotherapy55 in H129 patients. Although proton beam radia-
inactive scar (TNMH29 cT1b and IIRC25 Group B). B, Tumor regression tion56 and stereotactic radiation57,58 intend to reduce radiation dos-
after IAC and laser treatment in unilateral retinoblastoma (TNMH29 es to normal tissues, they still may carry risk for second primary
cT1b and IIRC25 Group B) to obtain long-term regression and inactive malignancy.59 Other local long-term adverse effects of radiation
scar. C, Note the peripheral choroidal pigmentary disturbance include health of soft tissues and bone around the eye, cataract
characteristic of treatment with IAC, which is not seen in the untreated formation, and keratoconjunctivitis sicca.60,61
normal right eye. In contrast, this choroidal pigmentary change is not Brachytherapy is focal therapy using a radioactive source
evident after treatment with systemic chemotherapy. (iodine-125, ruthenium-106) applied externally to the eye to

166 | www.apjo.org © 2018 Asia-Pacific Academy of Ophthalmology

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 Retinoblastoma for Pediatric Ophthalmologists

treat specific localized intraocular tumors.58,62,63 The radioactive Natl Acad Sci U S A. 1971;68:820‒823.
plaques are shielded to minimize radiation exposure to bone, and 10. Rushlow D, Piovesan B, Zhang K, et al. Detection of mosaic RB1 mutations
there is no evidence of risk to increase second primary malignan- in families with retinoblastoma. Hum Mutat. 2009;30:842‒851.
cy.64 The adverse effects of plaque radiotherapy are local, includ- 11. Roelofs K, Shaikh F, Astle W, et al. Incidental neuroblastoma with bilateral
ing cataract, glaucoma, and radiation retinopathy. retinoblastoma: what are the chances? Ophthalmic Genet. January 16, 2018.
The most recent addition to the treatment choices through [Epub ahead of print].
the long course of therapies to save an eye from retinoblastoma 12. Skalet AH, Gombos DS, Gallie BL, et al. Screening children at risk for
is pars plana vitrectomy (PPV). Previously, opening an eye with retinoblastoma. Ophthalmology. October 18, 2017. [Epub ahead of print].
active tumor was considered heresy! Now, after vitreous seeds 13. Kleinerman RA, Tucker MA, Abramson DH, et al. Risk of soft tissue
are uncontrolled by main-line therapies, PPV with melphalan in sarcomas by individual subtype in survivors of hereditary retinoblastoma. J
the irrigation fluid is shown to be an effective option without evi- Natl Cancer Inst. 2007;99:24‒31.
dence of extraocular disease.65 However, PPV is reserved for eyes 14. Kleinerman RA, Yu CL, Little MP, et al. Variation of second cancer risk by
with no evidence of optic nerve involvement both clinically and family history of retinoblastoma among long-term survivors. J Clin Oncol.
on neuroimaging, in which case enucleation is the safest option. 2012;30:950‒957.
In patients who are candidates for PPV, a laser barrier around the 15. MacCarthy A, Bayne AM, Brownbill PA, et al. Second and subsequent
solid tumor source of seeds can be applied before the planned tumours among 1927 retinoblastoma patients diagnosed in Britain 1951-
PPV, with endolaser reinforcement and silicone oil tamponade at 2004. Br J Cancer. 2013;108:2455‒2463.
the time of PPV to stabilize the retina. 16. Rushlow DE, Mol BM, Kennett JY, et al. Characterisation of retinoblastomas
without RB1 mutations: genomic, gene expression, and clinical studies.
Lancet Oncol. 2013;14:327‒334.
CONCLUSIONS 17. Soliman SE, Dimaras H, Khetan V, et al. Prenatal versus postnatal screening
Our current effort should aim to achieve a near to zero- for familial retinoblastoma. Ophthalmology. 2016;123:2610‒2617.
death cancer in retinoblastoma within the next 10 years. This 18. Abramson DH, Beaverson K, Sangani P, et al. Screening for retinoblastoma:
can be potentially achieved through improved understanding of presenting signs as prognosticators of patient and ocular survival. Pediatrics.
retinoblastoma among primary care providers, pediatric ophthal- 2003;112(Pt 1):1248‒1255.
mologists, parents, and the public. Early diagnosis and timely 19. Balmer A, Munier F. Differential diagnosis of leukocoria and strabismus,
intervention is key to improved overall survival of patients with first presenting signs of retinoblastoma. Clin Ophthalmol. 2007;1:431‒439.
retinoblastoma. Optimal management of retinoblastoma is multi- 20. Zhao J, Li S, Shi J, et al. Clinical presentation and group classification of
disciplinary, including the ophthalmologist(s) focused on retino- newly diagnosed intraocular retinoblastoma in China. Br J Ophthalmol.
blastoma, medical and radiation oncologists, geneticists, nurses, 2011;95:1372‒1375.
social workers, psychologists, and so on. Initiatives such as na- 21. Muen W, Hindocha M, Reddy M. The role of education in the promotion of
tional tumor board rounds and DePICTRB visualization (demo at red reflex assessments. JRSM Short Rep. 2010;1:46.
http://depictrb.technainstitute.com/)3,5 to support communication 22. Murphy D, Bishop H, Edgar A. Leukocoria and retinoblastoma-pitfalls of
across the whole management team, including parents, aim to max- the digital age? Lancet. 2012;379:2465.
imize the understanding of retinoblastoma from both clinical and 23. Abdolvahabi A, Taylor BW, Holden RL, et al. Colorimetric and longitudinal
research perspectives and maximize opportunities for patients. analysis of leukocoria in recreational photographs of children with
retinoblastoma. PLoS One. 2013;8:e76677.
24. Canadian Retinoblastoma Soiety. National retinoblastoma strategy Canadian
REFERENCES guidelines for care / Stratégie thérapeutique du rétinoblastome guide clinique
1. Soliman SE, Racher H, Zhang C, et al. Genetics and molecular diagnostics in Canadien. Can J Ophthalmol. 2009;44(Supp 2):S1‒S88.
retinoblastoma--an update. Asia Pac J Ophthalmol (Phila). 2017;6:197‒207. 25. Murphree AL. Intraocular retinoblastoma: the case for a new group
2. Soliman SE, Dimaras H, Souka AA, et al. Socioeconomic and psychological classification. Ophthalmol Clin North Am. 2005;18:41‒53.
impact of treatment for unilateral intraocular retinoblastoma. J Fr Ophtalmol. 26. Shields CL, Mashayekhi A, Au AK, et al. The International Classification
2015;38:550‒558. of Retinoblastoma predicts chemoreduction success. Ophthalmology. 2006;
3. Dimaras H, Corson TW, Cobrinik D, et al. Retinoblastoma. Nat Rev Dis 113:2276‒2280.
Primers. 2015;1:15021. 27. Novetsky DE, Abramson DH, Kim JW, et al. Published international
4. Dimaras H, Kimani K, Dimba EA, et al. Retinoblastoma. Lancet. 2012;379: classification of retinoblastoma (ICRB) definitions contain inconsistencies--
1436‒1446. an analysis of impact. Ophthalmic Genet. 2009;30:40‒44.
5. Chiu HH, Dimaras H, Downie R, et al. Breaking down barriers to 28. Mallipatna A, Dimaras H, Héon E, et al. Published International
communicating complex retinoblastoma information: can graphics be the Classification of Retinoblastoma (Icrb) definitions contain inconsistencies:
solution? Can J Ophthalmol. 2015;50:230‒235. an analysis of impact. Evidence-Based Ophthalmol. 2009;10:183‒185.
6. Xu XL, Singh HP, Wang L, et al. Rb suppresses human cone-precursor- 29. Mallipatna A, Gallie BL, Chévez-Barrios P, et al. Retinoblastoma. In: Amin
derived retinoblastoma tumours. Nature. 2014;514:385‒388. MB, Edge SB, Greene FL, eds. AJCC Cancer Staging Manual. 8th ed. New
7. Dimaras H, Khetan V, Halliday W, et al. Loss of RB1 induces non- York, NY: Springer; 2017:819‒831.
proliferative retinoma: increasing genomic instability correlates with 30. Chong EM, Coffee RE, Chintagumpala M, et al. Extensively necrotic
progression to retinoblastoma. Hum Mol Genet. 2008;17:1363‒1372. retinoblastoma is associated with high-risk prognostic factors. Arch Path
8. Dimaras H, Khetan V, Halliday W, et al. Retinoma underlying retinoblastoma Lab Med. 2006;130:1669‒1672.
revealed after tumor response to 1 cycle of chemotherapy. Arch Ophthalmol. 31. Valverde K, Pandya J, Héon E, et al. Retinoblastoma with central retinal
2009;127:1066‒1068. artery thrombosis that mimics extraocular disease. Med Pediatr Oncol.
9. Knudson AG. Mutation and cancer: statistical study of retinoblastoma. Proc 2002;38:277‒279.

Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018 www.apjo.org | 167

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.
AlAli et al Asia-Pacific Journal of Ophthalmology • Volume 7, Number 3, May/June 2018

32. Honavar SG, Singh AD, Shields CL, et al. Postenucleation adjuvant therapy Ophthalmol. 1996;114:1390‒1395.
in high-risk retinoblastoma. Arch Ophthalmol. 2002;120:923‒931. 50. Munier FL, Soliman S, Moulin AP, et al. Profiling safety of intravitreal
33. Kim JW. Retinoblastoma: evidence for postenucleation adjuvant injections for retinoblastoma using an anti-reflux procedure and sterilisation
chemotherapy. Int Ophthalmol Clin. 2015;55:77‒96. of the needle track. Br J Ophthalmol. 2012;96:1084‒1087.
34. Yarovaya V, Sioufi K, Shields CL. Parafoveolar retinoblastoma regression 51. Munier FL, Gaillard MC, Balmer A, et al. Intravitreal chemotherapy for
with foveal preservation following intra-arterial chemotherapy documented vitreous disease in retinoblastoma revisited: from prohibition to conditional
on hand-held optical coherence tomography in a newborn. Int J Retina indications. Br J Ophthalmol. 2012;96:1078‒1083.
Vitreous. 2017;3:43. 52. Shields CL, Manjandavida FP, Arepalli S, et al. Intravitreal melphalan for
35. Soliman SE, VandenHoven C, MacKeen LD, et al. Optical coherence persistent or recurrent retinoblastoma vitreous seeds: preliminary results.
tomography-guided decisions in retinoblastoma management. JAMA Ophthalmol. 2014;132:319‒325.
Ophthalmology. March 18, 2017. [Epub ahead of print]. 53. Aziz HA, Kim JW, Munier FL, et al. Acute hemorrhagic retinopathy
36. Rootman DB, Gonzalez E, Mallipatna A, et al. Hand-held high-resolution following intravitreal melphalan injection for retinoblastoma: a report of
spectral domain optical coherence tomography in retinoblastoma: clinical two cases and technical modifications to enhance the prevention of retinal
and morphologic considerations. Br J Ophthalmol. 2013;97:59‒65. toxicity. Ocul Oncol Pathol. 2017;3:34‒40.
37. Saktanasate J, Vongkulsiri S, Khoo CT. Invisible retinoblastoma. JAMA 54. Francis JH, Abramson DH, Ji X, et al. Risk of extraocular extension in
Ophthalmol. 2015;133:e151123. eyes with retinoblastoma receiving intravitreous chemotherapy. JAMA
38. Berry JL, Cobrinik D, Kim JW. Detection and intraretinal localization of an Ophthalmol. 2017;135:1426‒1429.
‘invisible’ retinoblastoma using optical coherence tomography. Ocul Oncol 55. Wong JR, Morton LM, Tucker MA, et al. Risk of subsequent malignant
Pathol. 2016;2:148‒152. neoplasms in long-term hereditary retinoblastoma survivors after
39. Vasquez LM, Giuliari GP, Halliday W, et al. Ultrasound biomicroscopy in chemotherapy and radiotherapy. J Clin Oncol. 2014;32:3284‒3290.
the management of retinoblastoma. Eye (Lond). 2011;25:141‒147. 56. Mouw KW, Sethi RV, Yeap BY, et al. Proton radiation therapy for the
40. de Jong MC, de Graaf P, Noij DP, et al. Diagnostic performance of magnetic treatment of retinoblastoma. Int J Radiat Oncol Biol Phys. 2014;90:863‒869.
resonance imaging and computed tomography for advanced retinoblastoma: a 57. Sahgal A, Millar BA, Michaels H, et al. Focal stereotactic external beam
systematic review and meta-analysis. Ophthalmology. 2014;121:1109‒1118. radiotherapy as a vision-sparing method for the treatment of peripapillary
41. Rizvi SW, Siddiqui MA, Khan AA, et al. Bilateral persistent hyperplastic and perimacular retinoblastoma: preliminary results. Clin Oncol. 2006;18:
primary vitreous: a close mimic of retinoblastoma. Semin Ophthalmol. 628‒634.
2013;28:25‒27. 58. Eldebawy E, Patrocinio H, Evans M, et al. Stereotactic radiotherapy as an
42. Shields JA, Shields CL, Honavar SG, et al. Classification and management alternative to plaque brachytherapy in retinoblastoma. Pediatr Blood
of Coats disease: the 2000 Proctor Lecture. Am J Ophthalmol. 2001;131: Cancer. 2010;55:1210‒1212.
572‒583. 59. Sethi RV, Shih HA, Yeap BY, et al. Second nonocular tumors among
43. Soliman SE, Wan MJ, Héon E, et al. Retinoblastoma versus advanced Coats’ survivors of retinoblastoma treated with contemporary photon and proton
disease: is enucleation the answer? Ophthalmic Genet. July 19, 2016. [Epub radiotherapy. Cancer. 2014;120:126‒133.
ahead of print]. 60. Blach LE, McCormick B, Abramson DH. External beam radiation therapy
44. Mallipatna AC, Sutherland JE, Gallie BL, et al. Management and outcome and retinoblastoma: long-term results in the comparison of two techniques.
of unilateral retinoblastoma. J AAPOS. 2009;13:546‒550. Int J Radiat Oncol Biol Phys. 1996;35:45‒51.
45. Shome D, Honavar SG, Raizada K, et al. Implant and prosthesis movement 61. Chan MP, Hungerford JL, Kingston JE, et al. Salvage external beam
after enucleation: a randomized controlled trial. Ophthalmology. 2010;117: radiotherapy after failed primary chemotherapy for bilateral retinoblastoma:
1638‒1644. rate of eye and vision preservation. Br J Ophthalmol. 2009;93:891‒894.
46. Yousef YA, Soliman SE, Astudillo PP, et al. Intra-arterial chemotherapy for 62. Francis JH, Barker CA, Wolden SL, et al. Salvage/adjuvant brachytherapy
retinoblastoma: a systematic review. JAMA Ophthalmol. 2016;134:584‒591. after ophthalmic artery chemosurgery for intraocular retinoblastoma. Int J
47. Munier FL, Mosimann P, Puccinelli F, et al. First-line intra-arterial versus Radiat Oncol Biol Phys. 2013;87:517‒523.
intravenous chemotherapy in unilateral sporadic group D retinoblastoma: 63. Abouzeid H, Moeckli R, Gaillard MC, et al. (106)Ruthenium brachytherapy
evidence of better visual outcomes, ocular survival and shorter time to for retinoblastoma. Int J Radiat Oncol Biol Phys. 2008;71:821‒828.
success with intra-arterial delivery from retrospective review of 20 years of 64. Shields CL, Shields JA, Cater J, et al. Plaque radiotherapy for retinoblastoma:
treatment. Br J Ophthalmol. December 7, 2016. [Epub ahead of print]. long-term tumor control and treatment complications in 208 tumors.
48. Hamel P, Héon E, Gallie BL, et al. Focal therapy in the management of Ophthalmology. 2001;108:2116‒2121.
retinoblastoma: when to start and when to stop. J AAPOS. 2000;4:334‒337. 65. Zhao J, Li Q, Wu S, et al. Pars plana vitrectomy and endoresection of
49. Wilson TW, Chan HS, Moselhy GM, et al. Penetration of chemotherapy refractory intraocular retinoblastoma. Ophthalmology. Novemer 16, 2017.
into vitreous is increased by cryotherapy and cyclosporine in rabbits. Arch [Epub ahead of print].

168 | www.apjo.org © 2018 Asia-Pacific Academy of Ophthalmology

Copyright © 2018 Asia-Pacific Academy of Ophthalmology. Unauthorized reproduction of this article is prohibited.

You might also like