You are on page 1of 10

The

Oncologist ®

Pediatric Oncology
Retinoblastoma: Review of Current Management

MURALI CHINTAGUMPALA,a,c,d PATRICIA CHEVEZ-BARRIOS,b,h EVELYN A. PAYSSE,b,c


SHARON E. PLON,c–e RICHARD HURWITZa–c,f,g
a
Texas Children’s Cancer Center, bDepartment of Ophthalmology, cDepartment of Pediatrics, dSection of
Hematology/Oncology, eDepartment of Molecular and Human Genetics, fCenter for Cell & Gene Therapy,
and gDepartment of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA;
h
Department of Pathology, Weill Medical College of Cornell University at The Methodist Hospital,
Houston, Texas, USA

Key Words. Retinoblastoma • Chemotherapy • Genetics of retinoblastoma • Second malignancies • Animal models

Disclosure: No potential conflicts of interest were reported by the authors, planners, reviewers, or staff managers of this article.

LEARNING OBJECTIVES
After completing this course, the reader will be able to:
1. Discuss the need for a multidisciplinary approach to the management of children with retinoblastoma.
2. Identify the patient factors that need to be considered when choosing the most appropriate initial and subsequent
treatment for a child with retinoblastoma.
3. Describe the role of genetics in the follow-up of retinoblastoma patients.
CME Access and take the CME test online and receive 1 AMA PRA Category 1 Credit™ at CME.TheOncologist.com

ABSTRACT
The most common ocular cancer in children is retino- therapy along with local ophthalmic therapies can be
blastoma. It affects approximately 300 children in the used in the treatment of retinoblastoma. Cure rates are
U.S. every year. It can affect one or both eyes and the high in children when the tumor is confined to the eye
disease can be inherited. Altered discoloration of the pu- and has not spread systemically or into the orbit or
pil and strabismus are the usual symptoms that lead to brain. Children with the heritable form of retinoblas-
medical attention. Subsequent appropriate diagnostic toma are at high risk for developing subsequent malig-
studies and care provided by a multidisciplinary team, nancies, most commonly sarcomas. This risk is greater
including an ophthalmologist, a pediatric oncologist, a for those children with the heritable form of the disease
radiation oncologist, and a geneticist, among others, of- who were exposed to ionizing radiation at age <1 year.
ten result in optimal short-term and long-term care. Exciting discoveries using animal models are provid-
The best initial and subsequent treatments are based on ing new insights into the development of this disease
whether the child has unilateral or bilateral disease, the and opening new avenues for targeted therapies that
stage of the disease, and the age of the child. Enucle- may lead to high cure rates with minimal toxicities.
ation, chemotherapy, and various forms of radiation The Oncologist 2007;12:1237–1246

Correspondence: Murali Chintagumpala, M.D., 6701 Fannin Street, Clinical Care Center, 14th Floor, MC CC1410, Houston, Texas
77030, USA. Telephone: 832-822-1482; Fax: 832-825-1503; e-mail: mxchinta@txccc.org Received February 28, 2006; accepted for
publication July 17, 2007. ©AlphaMed Press 1083-7159/2007/$30.00/0 doi: 10.1634/theoncologist.12-10-1237

The Oncologist 2007;12:1237–1246 www.TheOncologist.com


1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1238 Current Management of Retinoblastoma

EPIDEMIOLOGY reditary form with constitutional mutations in the RB1 gene


Retinoblastoma is the most common malignant ocular tu- [11]. The RB1 gene was localized and then cloned based on
mor in childhood and affects approximately 1 in 18,000 rare children with retinoblastoma who carry a cytogeneti-
children ⬍5 years of age in the U.S. [1]. The incidence is cally visible constitutional deletion at chromosome
higher in developing countries, and in some countries in 13q14.2 [12]. Even in cases of bilateral retinoblastoma, the
Central and South America retinoblastoma is one of the majority of children do not have a family history of the dis-
most common solid tumor malignancies in children [2]. ease, and disease is a result of de novo mutations in the RB1
The reason for this higher incidence is not clear. Lower so- gene that primarily occur during spermatogenesis [13, 14].
cioeconomic status and the presence of human papilloma This suggests that mutations in the retinoblastoma gene lo-
virus sequences in the retinoblastoma tissue have been im- cus (RB1) occur more commonly during spermatogenesis
plicated [3]. A higher risk for diseases like retinoblastoma or that the paternal chromosome in the early embryo is at a
in children born through in vitro fertilization was described higher risk for mutation. Studies of retinoblastoma tumors
previously, but a recent large study does not support this as- in both the hereditary and nonhereditary forms reveal that
sociation [4]. Approximately 80% of children with retino- the second hit or mutation frequently results in loss of het-
blastoma are diagnosed before 3 years of age. The diagnosis erozygosity of polymorphic markers that flank the RB1
of retinoblastoma in children 6 years or older is extremely gene in the tumor. Loss of heterozygosity can arise from al-
rare. Children with bilateral retinoblastoma constitute lelic loss, mitotic recombination, or loss of whole chromo-
about 20%–30%. Patients with bilateral disease usually some 13.
present at a younger age (14 –16 months) than patients with Genetic counseling in families with retinoblastoma is an
unilateral disease (29 –30 months) [5, 6]. In approximately important aspect in their care and should be coordinated
20% of children diagnosed with bilateral retinoblastoma, with a medical geneticist or genetic counselor who is part of
there is a family history of the disease [7]. Histologically, the retinoblastoma team. The identification of genetically
retinoblastoma develops from immature retinal cells and re- susceptible family members can lead to early diagnosis that
places the retina and other intraocular tissues. It displays a cannot only be life saving but may avoid the need for enu-
high mitotic and apoptotic rate, and because of this elevated cleation of the affected eye. Genetic analysis also provides
turnover of tumor cells, there are many areas of necrosis important information with regard to the risk for parents
and dystrophic calcification. (and long-term survivors of retinoblastoma) to have addi-
tional children with retinoblastoma. For example, healthy
GENETICS parents of a child with bilateral retinoblastoma have a 7%
The study of retinoblastoma has provided valuable insights risk with each pregnancy of having a child with retinoblas-
into the genetic basis of cancer. A “two-hit” model, as pro- toma because of the possibility of germline mosaicism (i.e.,
posed by Knudson, was developed based on the finding that more than one sperm or egg carrying an RB1 mutation). Ge-
children with bilateral retinoblastoma developed multifo- netic testing for RB1 mutations is clinically available in
cal, bilateral tumors at an earlier age than children with uni- certified DNA diagnostic laboratories (http://www.
lateral, unifocal tumors [8, 9]. According to the two-hit genetests.org) and can detect constitutional mutations in
model, two events are necessary for the retinal cell or cells approximately 90% of patients with bilateral disease [15].
to develop into tumors. The first mutational event can be Patients with bilateral disease can be assumed to have a
inherited (germline or constitutional) and would then be constitutional mutation and DNA obtained from a blood
present in all cells in the body. The second event or “hit” sample is examined directly for mutations. Children with
results in the loss of the remaining normal allele and occurs unilateral tumors may or may not have a germline constitu-
within a particular retinal cell or cells with dysregulation of tional mutation. Therefore, for these cases, a frozen tumor
the cell cycle and inappropriate entry into S phase [10]. specimen and a blood sample are sent for analysis. There is
In the sporadic, nonheritable form of retinoblastoma, a wide variety of different RB1 mutations, including whole
both mutational events occur within a single retinal cell af- gene deletions, and small frameshift, nonsense, splice site,
ter fertilization (somatic events), resulting in unilateral ret- and missense mutations. Somatic changes can also include
inoblastoma. Unilateral multifocal retinoblastoma can silencing of the RB1 promoter by methylation. Therefore,
develop if the first mutation occurs during development genetic analysis often includes complete sequencing of
such that more than one retinal cell contains this hit. Con- the coding region, analysis for deletions and rearrange-
versely, not all children with unilateral disease represent so- ments, methylation analysis, and RNA analysis [16].
matic events. Overall, 85% of children with unilateral Mutation(s) identified in the tumor are then studied in a
disease represent somatic events, but 15% represent the he- DNA sample obtained from blood. Presence of the muta-
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Chintagumpala, Chevez-Barrios, Paysse et al. 1239

tion in blood is presumptive evidence of a germline or con- “seeds.” A secondary serous retinal detachment is often as-
stitutional mutation. Once the germline mutation is sociated with large retinal tumors and subretinal seeds. To
identified then all siblings or offspring of the patient should confirm the presence of the tumor, a detailed examination
be tested for that specific mutation in order to determine the under anesthesia through dilated pupils is performed.
need for surveillance for retinoblastoma [17]. Identification Ultrasonography of the eyes is often performed to iden-
of the same mutation in another young family member tify and analyze the intraocular mass. Heterogeneity and
should prompt frequent evaluations by an ophthalmologist calcifications provide strong evidence for the diagnosis of
using appropriate examination techniques. The frequency retinoblastoma. Ultrasonography is not as sensitive as com-
of such evaluations may be every 1–2 months for the first puted tomography (CT), which is the ideal imaging format
year of life, followed by every 2–3 months for the next year, to detect intraocular calcifications. CT, however, raises the
and then every 3 months until 3 years of age, and finally concern of exposure to radiation in children ⬍1 year of age
every 4 – 6 months until the age of 6. This schedule can be with germline mutations [22]. However, it is still frequently
modified by the examiner based on his own clinical expe- used to confirm the diagnosis. Magnetic resonance imaging
rience. (MRI) of the brain and orbits is the most sensitive means of
Knowledge of the specific mutation identified in the evaluating for extraocular extension. It gives better delin-
family has also been used to perform preimplantation ge- eation of the optic nerve and also the pineal area [23]. Pine-
netic diagnosis in order to implant in vitro fertilized em- oblastoma is a malignancy that is associated with
bryos that are unlikely to carry the mutation [18]. Even with retinoblastoma in rare cases and when present it is referred
extensive molecular analysis, the causative mutation is not to as trilateral retinoblastoma.
always identified. In these cases, if there are multiple family MRI of the brain and spinal cord and cytologic exami-
members with retinoblastoma, then predictive genetic test- nation of cerebral spinal fluid are also indicated when there
ing can be performed by linkage analysis using polymor- is gross evidence of involvement of the optic nerve by im-
phic markers that flank the RB1 gene, a technique also aging studies or microscopic involvement beyond the lam-
referred to as indirect genetic testing. ina cribrosa on histopathologic examination of the
enucleated eye. A bone marrow examination and a bone
CLINICAL PRESENTATION AND DIAGNOSIS scan are indicated only when the clinical examination is
The most common presentation of retinoblastoma in the de- suggestive of metastases or a blood count abnormality is
veloped world is leukocoria (an abnormal white discolora- present.
tion in one or both pupils). This is usually first noticed by a The diagnosis of retinoblastoma is based on examina-
parent or relative [19]. Leukocoria is the result of an altered tion by an ophthalmologist and imaging studies. A biopsy is
pupillary red reflex in the eye and usually occurs when there rarely indicated because the procedure carries a theoretical
is a large tumor present; however, it can occur also with risk for extraocular dissemination that would convert an in-
smaller tumors associated with retinal detachment. Because traocular, curable tumor into extraocular, metastatic disease
retinoblastoma has a white appearance, the normal red re- with an extremely poor prognosis. Therefore, in the absence
flex is replaced by a white or creamy pink discoloration of of a tissue diagnosis, benign conditions that can mimic the
the pupil and can be better visualized with dilation of the more ominous retinoblastoma must be carefully excluded.
pupil and the aid of an ophthalmoscope. The second most These diseases usually can be differentiated from retino-
common sign of retinoblastoma is strabismus [20]. Strabis- blastoma by an ophthalmologist skilled in ocular oncology.
mus (misalignment of the eye) in this case results from a These conditions include Toxocara canis endophthalmitis,
loss of central vision in one or both eyes causing the ocular persistent hyperplastic primary vitreous, and Coats’ disease
misalignment. Heterochromia (different color of pupils), [24 –27]. These conditions usually produce total loss of vi-
hyphema (blood in the anterior chamber), glaucoma (in- sion in the affected eye and therefore, to establish the cor-
creased intraocular pressure), and orbital cellulitis/inflam- rect diagnosis, enucleation is an accepted procedure in
matory presentation are less common presentations [21]. In cases where the diagnosis is still in question from clinical
patients with more advanced disease, the presenting signs examination.
and symptoms correlate with the degree of extraocular in- Retinoblastoma can invade the optic nerve into the chi-
vasion, which can result in orbital swelling and proptosis. asm or disseminate through the subarachnoid space. From
Funduscopy typically reveals a large white to creamy the subarachnoid space the tumor cells can spread to the
colored main tumor, frequently with satellite lesions in the brain and spinal cord. Tumors can also invade the choroid
retina, subretinal space, and/or vitreous. The satellite tu- and the vascular layer, and spread hematogenously to the
mors of the subretinal space and vitreous are referred to as bone and bone marrow [28 –32]. Tumors can spread ante-

www.TheOncologist.com
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1240 Current Management of Retinoblastoma

riorly and involve the aqueous venous channels, conjunc- Enucleation


tiva, and lymphatics or invade the sclera into the orbit with Most children with unilateral retinoblastoma present with
eventual spread to regional lymph nodes. advanced disease, and most of them require enucleation.
Other indications for enucleation are for children with bi-
TREATMENT lateral disease where enucleation may be indicated for the
Management of a child with retinoblastoma requires a mul- eye with the most advanced disease that does not respond to
tidisciplinary approach. Ophthalmologists, pediatric on- chemotherapy (rarely, enucleation is indicated for both
cologists, pediatric radiation oncologists, pathologists, eyes), for the eye that has failed all known effective thera-
genetic counselors, social workers, nurses, and others play pies, when active tumor is present in an eye with no vision,
important roles in the cure of the disease, salvage of vision, when glaucoma is present as a result of neovascularization
and support of the child with vision loss and potential long- of the iris or tumor invasion into the anterior chamber, and
term sequelae. Many therapeutic options are available, and when direct visualization of an active tumor is obstructed
the indications for a specific modality or a combination of by conditions including hemorrhage, corneal opacity, or
modalities vary with each patient. Furthermore, manage- cataract [37]. Enucleation is curative in ⬎95% of patients
ment varies for children with intraocular disease and ex- with unilateral disease. Care should be taken to avoid per-
traocular spread of the tumor. Most patients with unilateral foration of the globe during surgery and to obtain a long
disease present with advanced intraocular disease and segment of the optic nerve in order to minimize the chance
therefore usually undergo enucleation, which results in a of leaving tumor at the surgical margin [38]. Orbital
cure rate ⬎95%. Children with involvement of both eyes at implants made of silicone, plastic, hydroxyapatite, and
diagnosis usually require multimodality therapy (chemo- MedPore are used at major treatment centers. By con-
therapy, local therapies). Failure to control disease in chil- necting the implants to the orbital muscles, excellent cos-
dren with bilateral disease may lead to external beam metic appearance can be achieved. Complications such
radiation (EBR) therapy. Enucleation is usually reserved as wound dehiscence and conjunctival erosion, although
for eyes with recurrent disease and no useful vision. rare, may be seen with all types of implants.

Management of Intraocular Disease EBR Therapy


Staging of the disease has facilitated the assessment of EBR therapy is an effective means of curing retinoblas-
treatments and measurement of outcomes in oncology. The toma. The most common indication for EBR is for the eye
Reese-Ellsworth (R-E) classification for intraocular retino- in a young child with bilateral retinoblastoma who has ac-
blastoma, developed in the 1960s, was used during the last tive or recurrent disease after completion of chemotherapy
40 years in assessing outcomes of therapy, and this classi- and local therapies. Children with small tumors within the
fication facilitated the comparison of results from various macula that do not respond to chemotherapy or have recur-
studies [33]. The R-E classification was devised to predict rent disease following chemotherapy can benefit from
prognosis in eyes that were treated with EBR therapy. The EBR. With EBR therapy, the entire tumor-bearing area of
classification scheme has five groups. Eyes with disease the globe is included along with at least 1 cm of the optic
consistent with the lower groups have a lower risk for enu- nerve. The prescribed dose to the tumor ranges from 42 Gy
cleation following EBR and group V eyes have the highest to 46 Gy, with the radiosensitive lens receiving signifi-
risk for enucleation. With the advent of other therapies, in- cantly less. Preservation of the eye with control of the dis-
cluding chemotherapy, which has increasingly replaced ease using EBR therapy is in the range of 58%– 88%.
EBR therapy in the treatment of intraocular disease, the Radiation therapy has only a 50% local control rate in R-E
usefulness of the R-E scheme is less apparent. Several al- groups IV and V disease, with a 95% rate of preservation of
ternative schemes have been proposed recently by Shields the eye in R-E groups I–III. In a report of 63 R-E group Vb
et al. [34] and by Murphree [35], among others [34 –35]. eyes (tumors with vitreous seeds) that were irradiated at ini-
The classification proposed by Murphree is the basis for tial diagnosis, the ocular survival rate was 53.4% at 10 years
several protocols for the treatment of intraocular retinoblas- [39]. However, the probability of developing a second can-
toma within the Children’s Oncology Group (COG). While cer following initial EBR therapy for group Vb disease in
the R-E classification and the classification proposed by patients with bilateral disease was 29.7% by 10 years after
Murphree and others address intraocular disease, another diagnosis.
classification system has been proposed by Chantada et al. Patients with hereditary disease who received EBR ther-
[36] to address extraocular disease and microscopic disease apy are reported to have a cumulative incidence of second
following enucleation. cancers of 35%, compared with 6% for those who did not
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Chintagumpala, Chevez-Barrios, Paysse et al. 1241

receive EBR [40]. Furthermore, patients who are ⬍1 year Chemothermotherapy


of age and who receive radiation therapy are several times Larger tumors or tumors with subretinal seeds are usually
more likely to develop second and subsequent malignancies treated with a combination of thermotherapy and chemo-
than those who are ⬎12 months of age and receive radiation therapy. Tractional and vaso-occlusive complications that
therapy [41]. In a cohort of patients with hereditary retino- can be seen with thermotherapy alone appear to be less fre-
blastoma, a strong relationship between radiation dose and quent when thermotherapy is used in combination with che-
the development of soft tissue sarcomas was reported [42]. motherapy (chemothermotherapy). Chemotherapy and
In subsequent follow-up of this cohort, significantly thermotherapy are delivered within hours of each other. In
higher risks for melanoma, cancers of the bone, nasal cav- one study of 188 retinoblastomas, tumor control was
ities, and brain, and soft tissue sarcomas (with an excess of achieved in 86% of cases [57]. The complications of che-
leiomyosarcoma) were documented [43, 44]. Cataracts, op- mothermotherapy included focal iris atrophy, paraxial lens
tic nerve damage, total retinal vascular occlusion, vitreous opacity, sector optic disk atrophy, retinal traction, optic
hemorrhage, and facial and temporal bone hypoplasia are disk edema, retinal vascular occlusion, retinal detachment,
other complications associated with EBR therapy [45]. Pro- and corneal edema. Chemothermotherapy may be espe-
ton beam radiation therapy offers promise in reducing the cially useful for patients with small tumors adjacent to the
significant long-term side effects associated with conven- fovea and optic nerve, where radiation therapy or laser pho-
tional EBR therapy [46]. tocoagulation may result in significant visual loss.

Brachytherapy Laser Photocoagulation


Brachytherapy involves the placement of a radioactive im- Laser photocoagulation is recommended only for small
plant (plaque), usually on the sclera adjacent to the base of posterior tumors [58 – 60]. The treatment is delivered with
a tumor. Iodine-125 (125I), gold, and more recently ruthe- an argon or diode laser or a xenon arc. The purpose of this
nium have been used [47]. The intention is to deliver a dose treatment is to coagulate all the blood supply to the tumor.
of 4,000 – 4,500 cGy transclerally to the apex of the tumor Indirect ophthalmoscope laser photocoagulation has signif-
over a period of 2– 4 days. This treatment is limited to tu- icantly improved the delivery of photocoagulation. Effec-
mors that are ⬍16 mm in base and 8 mm in thickness, and tive therapy usually requires 2–3 sessions at monthly
can be used as the primary treatment or, more frequently, in intervals. Complications of this treatment include retinal
patients who had failed initial therapy including previous detachment, retinal vascular occlusion, retinal traction, and
EBR therapy [48 –51]. This modality can also be used when preretinal fibrosis.
there is a peripheral tumor with focal vitreous seeding
around it. Relative contraindications include larger tumors Cryotherapy
and those that involve the macula. Good tumor control has Cryotherapy induces tumor tissue to freeze rapidly, re-
been reported with this modality [52]. Side effects are less sulting in damage to the vascular endothelium with sec-
common than with EBR and include optic neuropathy, ra- ondary thrombosis and infarction of the tumor tissue.
diation retinopathy, and cataract formation. Second malig- Cryotherapy may be used as primary therapy for small
nancies do not appear to be associated with this type of local peripheral tumors or for small recurrent tumors previ-
therapy. An orbital implant with 125I seeds was shown to be ously treated with other modalities. Tumors are typically
effective in treating patients who were at high risk for or- treated three times per session, with one or two sessions
bital recurrence after enucleation [53]. at monthly intervals. Ninety percent of tumors ⬍3 mm in
diameter are cured permanently, and complications are
Thermotherapy few and rarely serious [61, 62]. Transient conjunctival
Thermotherapy involves the application of heat directly to edema and transient localized serous retinal detachments
the tumor, usually in the form of infrared radiation. A tem- can occur. Vitreous hemorrhage can be observed in large
perature between 45°C and 60°C is the goal of this thera- or previously irradiated tumors.
peutic approach and is below the coagulative threshold and
therefore spares the retinal vessels from coagulation [54, Chemotherapy
55]. Thermotherapy alone can be used for small retinoblas- Chemotherapy has been used to treat intraocular retinoblas-
tomas that are ⱕ3 mm in diameter without vitreous or sub- toma since the early 1990s. Chemotherapy is used to reduce
retinal seeds. In a study of 91 tumors, 92% of the tumors the size of the tumor to allow local ophthalmological ther-
that were ⬍1.5 mm in diameter were controlled with ther- apies, including cryotherapy and laser photocoagulation, or
motherapy alone [56]. thermotherapy, to eradicate the remaining disease. This

www.TheOncologist.com
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1242 Current Management of Retinoblastoma

combination of therapies has been promoted to avoid EBR boplatin, vincristine, and etoposide along with subtenon
therapy and/or enucleation and thereby decrease the poten- carboplatin for group C and D eyes [76, 77]. Eyes with
tial for long-term side effects while salvaging some useful diffuse vitreous seeding present a particularly difficult
vision. The common indications for chemotherapy for in- management problem and, as mentioned above, rarely
traocular retinoblastoma include tumors that are large and respond to chemotherapy alone. Although EBR therapy
that cannot be treated with local therapies alone in children is modestly successful in patients with vitreous seeds,
with bilateral tumors. Chemotherapy can also be used in pa- new approaches are needed. A recent phase I study using
tients with unilateral disease when the tumors are small but adenoviral vectors to deliver the herpes simplex thymi-
cannot be controlled with local therapies alone. Such pa- dine kinase gene followed by ganciclovir demonstrated
tients constitute only 10%–15% of all patients with unilat- durable clinical and histopathologic responses in heavily
eral disease. Most patients with unilateral disease are pretreated patients with vitreous seeds [78].
diagnosed with advanced intraocular disease and undergo
enucleation. Numerous studies have been published that
show that chemotherapy is very effective in eliminating the Management of Extraocular Disease
need for EBR/enucleation in R-E group I–III eyes, while Patients with extraocular disease have a very poor progno-
proving to be significantly less successful in eyes with sis with respect to survival. Recently, there have been en-
group IV or V disease [34, 63–70]. Selected patients with couraging data to suggest that patients with regional
group IV disease have also had a very good response to che- extraocular disease may benefit from a combination of con-
motherapy, and EBR and enucleation were avoided. The ventional chemotherapy and EBR and those with distant
chemotherapy regimen commonly used consists of carbo- metastatic disease may benefit from high-dose chemother-
platin, vincristine, and etoposide. Cyclosporine has been apy and EBR in conjunction with bone marrow stem cell
used in addition to these three agents in some institutions in transplantation. Regional extraocular disease includes pa-
order to try to overcome drug resistance [71]. Others have tients with orbital, preauricular disease and patients with tu-
used carboplatin and vincristine without etoposide and mor found at the optic nerve surgical margin. Chantada et
older studies used cyclophosphamide and doxorubicin. The al. [79] reported a 5-year EFS rate of 84% in 15 patients
chemotherapy regimens from the different groups of inves- with orbital or preauricular disease treated with chemother-
tigators vary in the number and frequency of chemotherapy apy that included vincristine, doxorubicin, and cyclophos-
cycles. All these regimens are well tolerated, with the ex- phamide or vincristine, idarubicin, cyclophosphamide,
pected side effects of myelosuppression and its conse- carboplatin, and etoposide. These patients also received
quences, which include invasive bacterial infections. EBR of 4,500 cGy administered to the optic nerve chiasm
Ototoxicity and renal toxicities are rare. There is also the for patients with orbital disease and to the involved nodes
potential risk for second malignancies, especially when us-
for those with preauricular lymphadenopathy. A subse-
ing etoposide (an epipodophyllotoxin) [72, 73]. The RE
quent study showed that 12 patients with positive optic
groups I–III and part of group IV correspond to group B dis-
nerve surgical margins were all event-free survivors fol-
ease of the new international classification scheme that is a
lowing chemotherapy as above and orbital radiation ther-
modification of the one proposed by Murphree. Patients
apy of 4,000 – 4,500 cGy. A similar successful study was
who have group B disease will be treated with carboplatin
reported from Brazil [80].
and vincristine in the proposed COG trial, and their event-
Patients with metastatic extraocular disease have a poor
free survival (EFS) at 2 years will be estimated, where an
event is described as the need for nonprotocol chemother- prognosis when treated with regimens of conventional
apy or EBR/enucleation. doses of chemotherapy. There are several reports now sug-
Chemotherapy alone is not very effective in avoiding gesting that high-dose chemotherapy with stem cell rescue
EBR/enucleation in patients with R-E group V eyes, es- combined with EBR for areas of bulky disease at diagnosis
pecially those with vitreous seeds. Friedman et al. [70] is beneficial, with some long-term survivors among pa-
showed that only 53% of 30 group V (C, D, or E in the tients with metastatic disease not involving the central ner-
proposed classification) eyes could be controlled with vous system (CNS) [81– 85]. It is rare for a patient with
chemotherapy alone. Data from Chan et al. [74] and Vil- metastatic CNS involvement to survive using the therapies
lablanca et al. [75] suggested that approximately 40% of described above. The proposed trial by the COG for pa-
group C and 70% of group D eyes failed systemic che- tients with metastatic disease involves conventional che-
motherapy alone. Based on these data, the trial proposed motherapy, stem cell harvest, high-dose chemotherapy with
by the COG involves systemic chemotherapy with car- stem cell rescue, and EBR of involved sites.
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Chintagumpala, Chevez-Barrios, Paysse et al. 1243

HIGH-RISK HISTOPATHOLOGIC FEATURES IN PRECLINICAL MODELS


ENUCLEATED EYES OF PATIENTS WITH Current therapies contribute significantly to vision salvage
UNILATERAL RETINOBLASTOMA in patients with intraocular disease and overall cure rates in
There is no consensus on the histopathologic prognostic patients with extraocular disease. However, these therapies
factors present in enucleated eyes that could predict dissem- are not without significant morbidities. A better under-
ination of the disease. Postlamina optic nerve spread of tu- standing of the pathogenesis of retinoblastoma may lead to
mor, massive choroidal invasion, and extrascleral extension therapies with fewer long-term morbidities. To this end,
have been recognized as risk factors for dissemination of emphasis is placed on the development of preclinical mod-
disease. Other factors like focal choroidal, anterior cham- els [106]. Retinoblastoma is uniquely a human disease; no
ber, ciliary body, and iris involvement by tumor have not natural occurring animal model exists. Xenograft models of
been clearly established as risk factors for spread of the dis- retinoblastoma using cell lines derived from patients’ tu-
ease. Extensively necrotic retinoblastoma was also recently mors and injected into the vitreous have been developed in
described as a high-risk factor [86]. immunocompromised mice and have been found to be use-
When there is evidence of active tumor at the surgical ful in studying therapeutic modalities [107–109]. These
margin of the optic nerve at the time of enucleation, the as- models resemble patients with vitreous seeds and closely
sociated mortality rate has been described to be as high as mimic the progression of nonmetastatic as well as meta-
50%– 81% [32, 87–94]. When tumor cells are present in the static human disease; however, the response of primary tu-
optic nerve posterior to the lamina cribrosa, the mortality mors and the immune component of the therapy cannot be
rate is in the range of 13%– 69% [32, 38, 87–98]. It is gen- studied. Transgenic mouse models of retinoblastoma have
erally accepted that tumor involvement anterior to the lam- also been developed [110, 111]. Because RB1 gene muta-
ina cribrosa is not associated with greater mortality. tions do not result in retinoblastoma in mice, multiple mem-
The significance of choroid involvement and its effect bers of the RB gene family have been genetically deleted or
on the prognosis are less clear. There are reports that claim sequestered using the targeted expression of simian virus 40
that choroidal invasion by tumor does not contribute to a T-antigen [112]. These animals are immunologically com-
poor outcome, while others claim that choroidal invasion petent but exhibit multifocal disease that is genetically and
can result in mortality in the range of 11%– 81% [32, 87– phenotypically distinct from human disease. Although nei-
102]. The combination of choroidal invasion along with ther model is identical to human disease, both models have
any degree of optic nerve invasion has also been recognized been useful in preclinical testing of novel approaches to
as a risk factor with an adverse influence on outcome by treating retinoblastoma [113, 114]. A recent study with pre-
some groups [48, 103]. clinical models suggests a potential target for chemother-
Recent studies suggest that adjuvant chemotherapy ef- apy in retinoblastoma [115, 116]. This potential target is the
fectively reduced the incidence of metastases in patients MDMX–p53 interaction, which is antagonized by nutlin-3
with retinoblastoma with massive choroidal invasion or and efficiently kills retinoblastoma cells. Local delivery
postlaminar optic nerve invasion [104, 105]. These statisti- of nutlin-3 is considered a potential option to treat
cal figures have been drawn from several retrospective retinoblastoma.
studies performed in the past and are difficult to interpret
because of the significant variations in the definition of SUMMARY
high-risk features and the different treatment regimens used Imaging techniques and a variety of treatment modalities
for each series. Therefore, to study this issue prospectively have resulted in vast improvements in the management of
the COG has opened a clinical trial to study the pathology of children with retinoblastoma. There is at present great co-
the enucleated eyes in all unilateral patients from participat- operation among various investigators internationally to
ing institutions in North America and assign high-risk sta- share experiences and identify common areas of clinical
tus based on well-defined histopathologic features. These and biologic research in retinoblastoma. There is wide-
patients are then eligible to receive adjuvant chemotherapy spread acknowledgment that awareness of the symptoms of
consisting of six cycles of carboplatin, vincristine, and eto- the disease and early detection accompanied by early inter-
poside. Patients who do not have high-risk features as de- vention will significantly reduce visual and systemic mor-
fined by the protocol do not require further therapy and will bidity and mortality. Molecular genetic studies with
be observed. This will represent the first study in North identification of germline mutations have made a tremen-
America to prospectively evaluate histopathologic features dous impact on the management of the siblings and off-
and outcome in patients with unilateral disease. Similar spring of affected individuals and have obviated the need
studies have been initiated in Europe. for prolonged clinical screening under anesthesia for many

www.TheOncologist.com
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1244 Current Management of Retinoblastoma

unaffected children. They have also enabled the possibility importance. Animal models to better understand the patho-
of preimplantation genetic diagnosis, an option that is likely genesis and treatment of retinoblastoma are being pursued
to be considered by many affected individuals. Epidemio- vigorously. Initiation of cooperative group trials will fur-
logic and biologic studies to understand the factors that pro- ther define the effectiveness of therapeutic options for this
mote dissemination of the disease are increasingly gaining rare disease.

REFERENCES 19 Shields JA, Shields CL, Parsons HM. Differential diagnosis of retinoblas-
toma. Retina 1991;11:232–243.
1 Devesa SS. The incidence of retinoblastoma. Am J Ophthalmol 1975;80:
263–265. 20 Shields JA, Augsburger JJ. Current approaches to the diagnosis and man-
agement of retinoblastoma. Surv Ophthalmol 1981;25:347–372.
2 Leal-Leal C, Flores-Rojo M, Medina-Sanson A et al. A multicentre report
from the Mexican Retinoblastoma Group. Br J Ophthalmol 2004;88: 21 Yoshizumi MO, Thomas JV, Smith TR. Glaucoma-inducing mechanisms
1074 –1077. in eyes with retinoblastoma. Arch Ophthalmol 1978;96:105–110.

3 Orjuela M, Castaneda VP, Ridaura C et al. Presence of human papilloma 22 Smith EV, Gragoudas ES, Kolodny NH et al. Magnetic resonance imag-
virus in tumor tissue from children with retinoblastoma: An alternative ing: An emerging technique for the diagnosis of ocular disorders. Int Oph-
mechanism for tumor development. Clin Cancer Res 2000;6:4010 – 4016. thalmol 1990;14:119 –124.
4 Lidegaard O, Pinborg A, Andersen AN. Imprinting diseases and IVF: 23 Brenner D, Elliston C, Hall E et al. Estimated risks of radiation-induced
Danish National IVF cohort study. Hum Reprod 2005;20:950 –954. fatal cancer from pediatric CT. AJR Am J Roentgenol 2001;176:289 –296.
5 Draper GJ, Sanders BM, Brownbill PA et al. Patterns of risk of hereditary 24 Kuker W, Ramaekers V. Persistent hyperplastic primary vitreous: MRI.
retinoblastoma and applications to genetic counselling. Br J Cancer 1992; Neuroradiology 1999;41:520 –522.
66:211–219.
25 Kaste SC, Jenkins JJ 3rd, Meyer D et al. Persistent hyperplastic primary
6 Abramson DH, Frank CM, Susman M et al. Presenting signs of retinoblas- vitreous of the eye: Imaging findings with pathologic correlation. AJR
toma. J Pediatr 1998;132:505–508. Am J Roentgenol 1994;162:437– 440.
7 Newsham IF, Hadjistilianou T, Cavenee WK. Retinoblastoma. In: Vo- 26 Chang MM, McLean IW, Merritt JC. Coats’ disease: A study of 62 histo-
gelstein B, Kinzler KW, eds. The Genetic Basis of Human Cancer. New logically confirmed cases. J Pediatr Ophthalmol Strabismus 1984;21:
York: McGraw-Hill, 1998:363–392. 163–168.
8 Knudson AG Jr. Mutation and cancer: Statistical study of retinoblastoma.
27 Morgan KS, McLean IW. Retinoblastoma and persistent hyperplastic vit-
Proc Natl Acad Sci U S A 1971;68:820 – 823.
reous occurring in the same patient. Ophthalmology 1981;88:1087–1089.
9 Hethcote HW, Knudson AG Jr. Model for the incidence of embryonal can-
28 Spencer WH. Optic nerve extension of intraocular neoplasms. Am J Oph-
cers: Application to retinoblastoma. Proc Natl Acad Sci U S A 1978;75:
thalmol 1975;80:465– 471.
2453–2457.
29 Karcioglu ZA, al-Mesfer SA, Abboud E et al. Workup for metastatic ret-
10 Harbour JW, Dean DC. Rb function in cell-cycle regulation and apoptosis.
inoblastoma. A review of 261 patients. Ophthalmology 1997;104:307–
Nat Cell Biol 2000;94:E65–E67.
312.
11 Van Quill KR, O’Brien JM. The Role of the Retinoblastoma Protein in
30 Tosi P, Cintorino M, Toti P et al. Histopathological evaluation for the
Health, Malignancy, and the Pathogenesis of Retinoblastoma. In: Albert
prognosis of retinoblastoma. Ophthalmic Paediatr Genet 1989;10:173–
DM, Polans A, eds. Ocular Oncology. New York: Marcel Dekker, Inc.,
177.
2003:103–128.
12 Friend SH, Bernards R, Rogelj S et al. A human DNA segment with prop- 31 Croxatto JO, Fernandez Meijide R, Malbran ES. Retinoblastoma mas-
erties of the gene that predisposes to retinoblastoma and osteosarcoma. querading as ocular inflammation. Ophthalmologica 1983;186:48 –53.
Nature 1986;323:643– 646. 32 Kopelman JE, McLean IW, Rosenberg SH. Multivariate analysis of risk
13 Dryja TP, Mukai S, Petersen R et al. Parental origin of mutations of the factors for metastasis in retinoblastoma treated by enucleation. Ophthal-
retinoblastoma gene. Nature 1989;339:556 –558. mology 1987;94:371–377.

14 Zhu XP, Dunn JM, Phillips RA et al. Preferential germline mutation of the 33 Reese AB, Ellsworth RM. Management of retinoblastoma. Ann N Y Acad
paternal allele in retinoblastoma. Nature 1989;340:312–313. Sci. 1964;114:958 –962.
15 Houdayer C, Gauthier-Villars M, Lauge A et al. Comprehensive screening 34 Shields CL, Mashayekhi A, Demirci H et al. Practical approach to man-
for constitutional RB1 mutations by DHPLC and QMPSF. Hum Mutat agement of retinoblastoma. Arch Ophthalmol 2004;122:729 –735.
2004;23:193–202.
35 Linn Murphree A. Intraocular retinoblastoma: The case for a new group
16 Lohmann DR, Gallie BL. Retinoblastoma: Revisiting the model proto- classification. Ophthalmol Clin North Am 2005;18:41–53.
type of inherited cancer. Am J Med Genet C Semin Med Genet 2004;
36 Chantada G, Doz F, Antoneli CB et al. A proposal for an international ret-
129:23–28.
inoblastoma staging system. Pediatr Blood Cancer 2006;47:801– 805.
17 Richter S, Vandezande K, Chen N et al. Sensitive and efficient detection
37 Abramson DH. Treatment of retinoblastoma. In: Blodi FC, ed. Retino-
of RB1 gene mutations enhances care for families with retinoblastoma.
blastoma. New York: Churchill Livingstone, 1985:3–93.
Am J Hum Genet 2003;72:253–269.
38 Abramson DH, Ellsworth RM. The surgical management of retinoblas-
18 Simpson JL, Carson SA, Cisneros P. Preimplantation genetic diagnosis
toma. Ophthalmic Surg 1980;11:596 –598.
(PGD) for heritable neoplasia. J Natl Cancer Inst Monogr 2005;(34):
87–90. 39 Abramson DH, Beaverson KL, Chang ST et al. Outcome following initial
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Chintagumpala, Chevez-Barrios, Paysse et al. 1245

external beam radiotherapy in patients with Reese-Ellsworth Group Vb indirect ophthalmoscope laser photocoagulation. J Pediatr Ophthalmol
retinoblastoma. Arch Ophthalmol 2004;122:1316 –1323. Strabismus 1995;32:317–322.
40 Roarty JD, McLean IW, Zimmerman LE. Incidence of second neoplasms 60 Shields JA. The expanding role of laser photocoagulation for intraocular
in patients with bilateral retinoblastoma. Ophthalmology 1988;95:1583– tumors. The 1993 H. Christian Zweng Memorial Lecture. Retina 1994;14:
1587. 310 –322.
41 Abramson DH, Frank CM. Second nonocular tumors in survivors of bi- 61 Abramson DH, Ellsworth RM, Rozakis GW. Cryotherapy for retinoblas-
lateral retinoblastoma: A possible age effect on radiation-related risk. toma. Arch Ophthalmol 1982;100:1253–1256.
Ophthalmology 1998;105:573–579; discussion 579 –580.
62 Shields JA, Parsons H, Shields CL et al. The role of cryotherapy in the
42 Wong FL, Boice JD Jr, Abramson DH et al. Cancer incidence after reti- management of retinoblastoma. Am J Ophthalmol 1989;108:260 –264.
noblastoma: Radiation dose and sarcoma risk. JAMA 1997;278:1262–
1267. 63 Gallie BL, Budning A, DeBoer G et al. Chemotherapy with focal therapy
can cure intraocular retinoblastoma without radiotherapy. Arch Ophthal-
43 Kleinerman RA, Tucker MA, Tarone RE et al. Risk of new cancers after mol 1996;114:1321–1328.
radiotherapy in long-term survivors of retinoblastoma: An extended fol-
low-up. J Clin Oncol 2005;23:2272–2279. 64 Shields CL, De Potter P, Himelstein BP et al. Chemoreduction in the ini-
tial management of intraocular retinoblastoma. Arch Ophthalmol 1996;
44 Kleinerman RA, Tucker MA, Abramson DH et al. Risk of soft tissue sar-
114:1330 –1338.
comas by individual subtype in survivors of hereditary retinoblastoma.
J Natl Cancer Inst 2007;99:24 –31. 65 Shields CL, Honavar SG, Shields JA et al. Factors predictive of recurrence
of retinal tumors, vitreous seeds, and subretinal seeds following chemore-
45 Imhof SM, Mourits MP, Hofman P et al. Quantification of orbital and mid-
duction for retinoblastoma. Arch Ophthalmol 2002;120:460 – 464.
facial growth retardation after megavoltage external beam irradiation in
children with retinoblastoma. Ophthalmology 1996;103:263–268. 66 Shields CL, Shelil A, Cater J et al. Development of new retinoblastomas
after 6 cycles of chemoreduction for retinoblastoma in 162 eyes of 106
46 Krengli M, Hug EB, Adams JA et al. Proton radiation therapy for retino-
consecutive patients. Arch Ophthalmol 2003;121:1571–1576.
blastoma: Comparison of various intraocular tumor locations and beam
arrangements. Int J Radiat Oncol Biol Phys 2005;61:583–593. 67 Kingston JE, Hungerford JL, Madreperla SA et al. Results of combined
47 Schueler AO, Fluhs D, Anastassiou G et al. Beta-ray brachytherapy of ret- chemotherapy and radiotherapy for advanced intraocular retinoblastoma.
inoblastoma: Feasibility of a new small-sized ruthenium-106 plaque. Arch Ophthalmol 1996;114:1339 –1343.
Ophthalmic Res 2006;38:8 –12. 68 Greenwald MJ, Strauss LC. Treatment of intraocular retinoblastoma with
48 Shields CL, Shields JA, De Potter P et al. Plaque radiotherapy in the man- carboplatin and etoposide chemotherapy. Ophthalmology 1996;103:
agement of retinoblastoma. Use as a primary and secondary treatment. 1989 –1997.
Ophthalmology 1993;100:216 –224. 69 Gunduz K, Shields CL, Shields JA et al. The outcome of chemoreduction
49 Hernandez JC, Brady LW, Shields CL et al. Conservative treatment of ret- treatment in patients with Reese-Ellsworth group V retinoblastoma. Arch
inoblastoma. The use of plaque brachytherapy. Am J Clin Oncol 1993;16: Ophthalmol 1998;116:1613–1617.
397– 401. 70 Friedman DL, Himelstein B, Shields CL et al. Chemoreduction and local
50 Desjardins L, Levy C, Labib A et al. An experience of the use of radioac- ophthalmic therapy for intraocular retinoblastoma. J Clin Oncol 2000;18:
tive plaques after failure of external beam radiation in the treatment of ret- 12–17.
inoblastoma. Ophthalmic Paediatr Genet 1993;14:39 – 42.
71 Chan HS, DeBoer G, Thiessen JJ et al. Combining cyclosporin with che-
51 Shields JA, Shields CL, De Potter P et al. Plaque radiotherapy for residual motherapy controls intraocular retinoblastoma without requiring radia-
or recurrent retinoblastoma in 91 cases. J Pediatr Ophthalmol Strabismus tion. Clin Cancer Res 1996;2:1499 –1508.
1994;31:242–245.
72 Le Deley MC, Vassal G, Taibi A et al. High cumulative rate of secondary
52 Shields CL, Shields JA, Cater J et al. Plaque radiotherapy for retinoblas- leukemia after continuous etoposide treatment for solid tumors in children
toma: Long-term tumor control and treatment complications in 208 tu- and young adults. Pediatr Blood Cancer 2005;45:25–31.
mors. Ophthalmology 2001;108:2116 –2121.
73 Bayar E, Robinson MG, Kurczynski TW. Unilateral retinoblastoma with
53 Sealy R, Stannard C, Shackleton D. Improved cosmesis in retinoblastoma acquired monosomy 7 and secondary acute myelomonocytic leukemia.
patients treated with iodine-125 orbital irradiation. Ophthalmic Paediatr Cancer Genet Cytogenet 1998;105:79 – 82.
Genet 1987;8:95–99.
74 Chan HSL, Heon E, Budning A et al. Improvement of the cure rate of in-
54 Murphree AL, Villablanca JG, Deegan WF 3rd et al. Chemotherapy plus traocular retinoblastoma without significantly increasing toxicity with
local treatment in the management of intraocular retinoblastoma. Arch higher dose carboplatin teniposide in a cyclosporine multidrug reversal
Ophthalmol 1996;114:1348 –1356. regimen. Presented at the 10th International Symposium on Retinoblas-
55 Murphree AL, Munier FL. Retinoblastoma. In: Ryan SJ, ed. Retina, 2nd toma, Ft. Lauderdale, FL, May 4, 2001.
ed. St Louis: Mosby-Year Book Inc., 1994:605– 606.
75 Villablanca JG, et al. Clinical outcome of group V eyes treated with cy-
56 Abramson DH, Schefler AC. Transpupillary thermotherapy as initial closporin A/carboplatin/etoposide/vincristine. Presented at the 10th Inter-
treatment for small intraocular retinoblastoma: Technique and predictors national Symposium on Retinoblastoma, Ft. Lauderdale, FL, May 4,
of success. Ophthalmology 2004;111:984 –991. 2001.
57 Shields CL, Santos MC, Diniz W et al. Thermotherapy for retinoblastoma. 76 Abramson DH, Frank CM, Dunkel IJ. A phase I/II study of subconjunc-
Arch Ophthalmol 1999;117:885– 893. tival carboplatin for intraocular retinoblastoma. Ophthalmology 1999;
58 Shields JA, Shields CL, Parsons H et al. The role of photocoagulation in 106:1947–1950.
the management of retinoblastoma. Arch Ophthalmol 1990;108:205–208. 77 Villablanca JG, Jubran RF, Murphree LA. Phase I study of subtenon car-
59 Shields CL, Shields JA, Kiratli H et al. Treatment of retinoblastoma with boplatin with systemic high dose carboplatin/etoposide/vincristine for

www.TheOncologist.com
1549490x, 2007, 10, Downloaded from https://theoncologist.onlinelibrary.wiley.com/doi/10.1634/theoncologist.12-10-1237 by Nat Prov Indonesia, Wiley Online Library on [29/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1246 Current Management of Retinoblastoma

eyes with disseminated intraocular. Presented at the 10th International 98 Rubin CM, Robison LL, Cameron JD et al. Intraocular retinoblastoma
Symposium on Retinoblastoma, Ft. Lauderdale, FL, May 4, 2001. group V: An analysis of prognostic factors. J Clin Oncol 1985;3:680 – 685.
78 Chevez-Barrios P, Chintagumpala M, Mieler W et al. Response of retino- 99 Schvartzman E, Chantada G, Fandino A et al. Results of a stage-based pro-
blastoma with vitreous tumor seeding to adenovirus-mediated delivery of tocol for the treatment of retinoblastoma. J Clin Oncol 1996;14:1532–
thymidine kinase followed by ganciclovir. J Clin Oncol 2005;23:7927– 1536.
7935.
100 Chantada G, Fandino A, Davila MT et al. Results of a prospective study
79 Chantada G, Fandino A, Casak S et al. Treatment of overt extraocular ret- for the treatment of retinoblastoma. Cancer 2004;100:834 – 842.
inoblastoma. Med Pediatr Oncol 2003;40:158 –161.
101 Khelfaoui F, Validire P, Auperin A et al. Histopathologic risk factors in
80 Antoneli CB, Steinhorst F, de Cassia Braga Ribeiro K et al. Extraocular retinoblastoma: A retrospective study of 172 patients treated in a single
retinoblastoma: A 13-year experience. Cancer 2003;98:1292–1298. institution. Cancer 1996;77:1206 –1213.
81 Namouni F, Doz F, Tanguy ML et al. High-dose chemotherapy with car- 102 Herm RJ, Heath P. A study of retinoblastoma. Am J Ophthalmol 1956;41:
boplatin, etoposide and cyclophosphamide followed by a haematopoietic 22–30.
stem cell rescue in patients with high-risk retinoblastoma: A SFOP and
SFGM study. Eur J Cancer 1997;33:2368 –2375. 103 Kobrin JL, Blodi FC. Prognosis in retinoblastoma: Influence of his-
topathologic characteristics. J Pediatr Ophthalmol Strabismus 1978;15:
82 Dunkel IJ, Aledo A, Kernan NA et al. Successful treatment of metastatic
278 –281.
retinoblastoma. Cancer 2000;89:2117–2121.
104 Uusitalo MS, Van Quill KR, Scott IU et al. Evaluation of chemoprophy-
83 Dunkel IJ, Aledo A, Finlay JL et al. Intensive multimodality therapy for
laxis in patients with unilateral retinoblastoma with high-risk features on
metastatic retinoblastoma. Pediatr Blood Cancer 2004;43:378.
histopathologic examination. Arch Ophthalmol 2001;119:41– 48.
84 Kremens B, Wieland R, Reinhard H et al. High-dose chemotherapy with
105 Honavar SG, Singh AD, Shields CL et al. Postenucleation adjuvant ther-
autologous stem cell rescue in children with retinoblastoma. Bone Mar-
apy in high-risk retinoblastoma. Arch Ophthalmol 2002;120:923–931.
row Transplant 2003;31:281–284.
85 Rodriguez-Galindo C, Wilson MW, Haik BG et al. Treatment of meta- 106 Zhang J, Schweers B, Dyer MA. The first knockout mouse model of ret-
static retinoblastoma. Ophthalmology 2003;110:1237–1240. inoblastoma. Cell Cycle 2004;3:952–959.

86 Chong EM, Coffee RE, Chintagumpala M et al. Extensively necrotic ret- 107 Gallie BL, Albert DM, Wong JJ et al. Heterotransplantation of retinoblas-
inoblastoma is associated with high-risk prognostic factors. Arch Pathol toma into the athymic “nude” mouse. Invest Ophthalmol Vis Sci 1977;16:
Lab Med 2006;130:1669 –1672. 256 –259.

87 Zimmerman LE. The Registry of Ophthalmic Pathology: Past, present and 108 Rowe SG, Lee WH, Madreperla S. Subretinal and vitreal growth of human
future. Trans Am Acad Ophthalmol Otolaryngol 1961;65:51–113. retinoblastoma cells in the mouse eye. Invest Ophthalmol Vis Sci 1992;
33:875.
88 Brown DH. The clinicopathology of retinoblastoma. Am J Ophthalmol
1966;61:508 –514. 109 del Cerro M, Seigel GM, Lazar E et al. Transplantation of Y79 cells into
89 Ellsworth RM. Orbital retinoblastoma. Trans Am Ophthalmol Soc 1974; rat eyes: An in vivo model of human retinoblastomas. Invest Ophthalmol
72:79 – 88. Vis Sci 1993;34:3336 –3346.

90 de Sutter E, Havers W, Hopping W et al. The prognosis of retinoblastoma 110 Albert DM, Griep AE, Lambert PF et al. Transgenic models of retinoblas-
in terms of survival. A computer assisted study. Part II. Ophthalmic Pae- toma: What they tell us about its cause and treatment. Trans Am Acad
diatr Genet 1987;8:85– 88. Ophthalmol Soc 1994;92:385– 400; discussion 400 – 401.

91 Magramm I, Abramson DH, Ellsworth RM. Optic nerve involvement in 111 Howes KA, Lasudry JGH, Albert DM et al. Photoreceptor cell tumors in
retinoblastoma. Ophthalmology 1989;96:217–222. transgenic mice. Invest Ophthalmol Vis Sci 1994;35:342–351.

92 Messmer EP, Heinrich T, Hopping W et al. Risk factors for metastases in 112 al-Ubaidi MR, Font RL, Quiambao AB et al. Bilateral retinal and brain
patients with retinoblastoma. Ophthalmology 1991;98:136 –141. tumors in transgenic mice expressing simian virus 40 large T antigen un-
93 Scott MH, Richard JM. Retinoblastoma in the state of Oklahoma: A clin- der control of the human interphotoreceptor retinoid-binding protein pro-
icopathologic review. J Okla State Med Assoc 1993;86:111–118. moter. J Cell Biol 1992;119:1681–1687.

94 Rootman J, Hofbauer J, Ellsworth RM et al. Invasion of the optic nerve by 113 Chévez-Barrios P, Hurwitz MY, Louie K et al. Metastatic and nonmeta-
retinoblastoma: A clinicopathological study. Can J Ophthalmol 1976;11: static models of retinoblastoma. Am J Pathol 2000;157:1405–1412.
106 –114. 114 Hurwitz MY, Marcus KT, Chévez-Barrios P et al. Suicide gene therapy
95 Carbajal UM. Observations on retinoblastoma. Am J Ophthalmol 1958; for treatment of retinoblastoma in a murine model. Hum Gene Ther 1999;
45:391– 402. 10:441– 448.

96 Haye C, Desjardins L, Elmaleh C et al. Prognosis and treatment of retino- 115 Elison JR, Cobrinik D, Claros N et al. Small molecule inhibition of HDM2
blastoma. 105 cases treated at Institut Curie. Ophthalmic Paediatr Genet leads to p53-mediated cell death in retinoblastoma cells. Arch Ophthalmol
1989;10:151–155. 2006;124:1269 –1275.
97 MacKay CJ, Abramson DH, Ellsworth RM. Metastatic patterns of retino- 116 Laurie NA, Donovan SL, Shih CS et al. Inactivation of the p53 pathway in
blastoma. Arch Ophthalmol 1984;102:391–396. retinoblastoma. Nature 2006;444:61– 66.

You might also like