Sie sind auf Seite 1von 32

Retinoblastoma

They live and see...


Author

Author Andhra Pradesh State Ophthalmological


Society, 2001; Best of Show Award for the
Born in Pune, Dr Honavar studied in Banga- video film by the American Academy of
lore and had his basic medical education at Ophthalmology, 2002 and 2006; Achieve-
the Bangalore Medical College. He was the ment Award by the American Academy
Best Graduate of the Bangalore University of Ophthalmology, 2002; Dr Surya Prasad
in 1988. He received post-graduate training Rao Oration by the Andhra Pradesh State
in Ophthalmology, followed by a senior resi- Ophthalmological Association, 2006; and
dency in Ophthalmic Plastic Surgery, Pediat- Dr Siva Reddy International Award by the
ric Ophthalmology and Glaucoma at the Dr AIOS, 2007.
Rajendra Prasad Center for Ophthalmic Sci-
ences, All India Institute of Medical Scienc- Dr Honavar has been active in the organi-
es, New Delhi. During his residency training zation of scientific meetings. The Interna-
at the All India Institute of Medical Sciences, tional Congress of ocular Oncology that he
he was adjudged the Best Resident. conducted in 2004 brought together del-
egates from 66 countries. Sunayana 2007,
Dr Honavar further trained in Ocular Oncol- the annual meeting of the AIOS broke the
ogy and was mentored by Prof Jerry Shields records in terms of number of delegates
and Prof Carol Shields at the Wills Eye Hos- and set very high standards. Cutting Edge
pital, Thomas Jefferson University, Philadel- 2007 proved to be landmark event.
phia, PA, USA. He thereafter established
and now heads the comprehensive Ocular Dr Honavar’s overall research contributions
Oncology Service at the LV Prasad Eye In- that have had very significant impact on the
stitute, Hyderabad, the first such facility in diagnosis and management of retinoblast-
the country. His current research interests oma and its outcome. The comprehensive
comprise of tumors of the ocular surface, multispecialty Children’s Eye Cancer Center
orbit, retinoblastoma, and pediatric lacrimal that he has established at the LV Prasad Eye
disorders. Institute in collaboration with the SightSav-
ers international has done pioneering work
Dr Honavar has extensively published in peer- and is now recognized as one of the best
reviewed journals (over 125 manuscripts) in the World. Dr Honavar is currently the
and has written several book chapters. The Chairman of the Indian Retinoblastoma
worth of Dr Honavar’s scientific work can Group that plans to work in standardization
be gauged by his cumulative citation index of diagnosis and management of retinoblas-
of 617, H Index of 16 and G Index of 22. toma across the country.
He is the reviewer for American Journal of
Ophthalmology, Asian Journal of Ophthal- Despite the recent developments and im-
mology, British Journal of Ophthalmology, proved prognosis, delayed diagnosis and
Clinical and Experimental Ophthalmology, treatment has been a major detriment in
Eye, Indian Journal of Ophthalmology, Jour- the management of retinoblastoma. This
nal of Ophthalmic Plastic and Reconstruc- monograph is an attempt to propagate ba-
tive Surgery, Ophthalmology and Retina. sic information on the current diagnosis and
treatment of retinoblastoma to the peer
Some of the major awards and honors to group. This educational effort is supported
his credit include Gold Medals and the Best by the SightSavers International and the LV
Graduate Award by the Bangalore Universi- Prasad Eye Institute.
ty for excellence in MBBS, 1988; State Award
for Academic Excellence by the Government
of Karnataka, 1988; Pfizer Post-graduate
National Award & Medallion, 1990; Col
Rangachari Gold Medal by the AIOS, 1992;
Santosh G Honavar, MD, FACS
Most-Promising Young Ophthalmologist-in-
Research Citation, an International Award
Head of the Department of Ophthalmic by the ‘Physicians in Research’, USA, 1994,
Plastic Surgery, Orbit and Ocular 1996, 1997, and 1998; Best-Resident Award
Oncology; Associate Director, Patient by the Dr Rajendra Prasad Center for Oph-
Care Policies and Planning thalmic Sciences, All India Institute of Medi-
LV Prasad Eye Institute, Kallam Anji cal Sciences, New Delhi, 1995; ARVO-Sant-
Reddy Campus, LV Prasad Marg, en International Fellowship by the ARVO,
Banjara Hills, Hyderabad 500034, India 1996; Zeigler International Fellowship by
Voice +91-40-30612345 the Orbis International, New York, USA,
1999; Young Scientist Award by the Indian
Mobile +91-98483-04001
Society of Oncology, 2000; Dr Vengala Rao
Fax +91-40-23548271
Award for the best scientific paper by the
e-mail honavar@lvpei.org Andhra Pradesh State Ophthalmological
http://www.lvpei.org Society, 2001; Achievement Award by the

Foreword

Foreword others, including Wardrop, believed it


could be therapeutic. Others argued for
The story of retinoblastoma is a long wind- the use of poultices, home-made remedies,
ing tale that weaves through the dark for- and leeches for therapy. Later, in the early
ests of fatal treachery in the 1600s to the 1900s, Hilgartner observed the success of
steep mountains of new understanding in external beam radiotherapy for retinoblas-
the 1800s to the open plains of secure sur- toma. Globes with retinoblastoma were
vival in the late 1900s. It is a scary story of salvaged, often with poor vision and many
death from a frightful mass that filled the with ultimate phthisis bulbi. Later refine-
eye and enveloped the spirit of a young ments in radiotherapy technique using less
toddler. It is a suspense-ridden drama of toxic doses and avoidance of bone with
a blind child whose life was threatened multiple beam portals lead to fewer long-
with an eye cancer, but fortunately saved term complications. Further refinements
by enucleation. It is a fairy tale of scores with the use of focal radioactive applicators
of children cursed with a deadly, blinding surgically sewn onto the eye allowed for
eye malignancy that survived with chemo- improved therapy with minimal toxicity. Re-
therapy and regained their vision to live in ports on novel techniques and results of ra-
peaceful happiness. dioactive applicators from several countries
The story of retinoblastoma has been re- ensued including Stallard from England,
written many times since it was first recog- Ellsworth and Abramson from the USA,
nized by Pawius in 1657. The nightmare has Stannard from South Africa, Hungerford
gradually become a fairytale with a happy from England, Desjardins from France, and
ending. The prognosis for life and vision in Shields and Shields from the USA.
patients with retinoblastoma has greatly Until the 1990s, intraocular retinoblastoma
improved over the past centuries, due pri- showed little or inconsistent response to
marily to better understanding of this ma- systemic chemotherapy. Older studies in-
lignancy, earlier detection of the disease, vestigating chemotherapy indicated poor
and, most importantly, improved treatment control with various regimens. In 1996,
methods.1-4 One hundred years ago, retin- Kingston and associates reported success
oblastoma was nearly always fatal in the with vincristine, etoposide, and carbopla-
United States. Gradually, the prognosis im- tin for intraocular retinoblastoma.6 They
proved and approximately 30% of affected showed that the ocular salvage rate in
patients survived in the 1930’s, 80% in the advanced retinoblastoma treated with ex-
1960’s, and over 95% in the 1990’s. Simi- ternal beam radiotherapy alone was 30%
lar improvements have been found in India whereas the ocular salvage rate in those
with improved survival from 5% in 1896 to eyes treated with chemoreduction prior
81% in 1967. The trend for improved sur- to external beam radiotherapy was nearly
vival continues worldwide into the 21st cen- 70%.6 So, enter the new era of retinoblas-
tury. toma therapy, the chemoreduction era. 6-8
In today’s world, children with retinoblas-
We now have nearly 15 years experience
toma rarely die from this intraocular can-
with chemoreduction for retinoblastoma.
cer. However, these children are at risk for
Many observations on the successes and
death from related conditions such as trilat-
limitations of this technique have been pub-
eral retinoblastoma and second cancers. In
lished. An early study revealed that chem-
children under age 5 years, the most com-
oreduction caused regression of retinoblas-
mon cause of death is primary intracranial
toma by approximately 35% in tumor base
neuroblastic tumor (“trilateral retinoblasto-
and nearly 50% in tumor thickness.7 Shortly
ma”) and in patients over 5 years, death is
thereafter, it was noted that subretinal flu-
usually due to remote second malignancies,
id completely resolved in 76% of eyes that
particularly osteosarcoma. 5 Therefore, the
had initial total retinal detachment. 9 Subse-
tale of retinoblastoma does not end with
quently, several reports have elaborated on
enucleation or treatment of the eye and
the response of vitreous and subretinal seeds
cure of the intraocular tumor. The first
to chemoreduction.10-12 In spite of this suc-
chapter concludes but the second chapter Carol L Shields, MD
cess, vitreous and subretinal seeds pose the
opens as this is only the beginning of the Co-Director, Ocular Oncology Service,
greatest problem with potential for recur-
story. Long-term follow up for life is impor- Wills Eye Institute; Professor of
rence, often remote from the main tumor.
tant, particularly to screen for nonocular, Ophthalmology, Jefferson Medical
In a report on 158 eyes with retinoblastoma
potentially life-threatening cancers. School, Thomas Jefferson Hospital
treated using vincristine, etoposide, and
Therapy for retinoblastoma has evolved carboplatin for 6 cycles, all retinoblastomas, 840 Walnut Street
over the years. In the 1800s, arguments subretinal seeds, and vitreous seeds showed Philadelphia, PA,19107, USA
centered around the usefulness of enuclea- initial regression. However, approximately Telephone +1-215-928-3105
tion. Some clinicians felt enucleation was 50% of the eyes with vitreous seeds showed Fax +1-215-928-1140
not useful to the patient’s survival while at least one vitreous seed recurrence at 5 e-mail carolshields@gmail.com

Foreword

years and 62% of the eyes with subretinal of chemotherapy for children with high-risk
seeds showed at least one subretinal seed retinoblastoma, that is eyes with tumor in-
recurrence at 5 years. 11 Of the 158 eyes, vading the optic nerve or choroid.16 After
recurrence of at least one retinal tumor per establishing his practice of retinoblastoma in
eye was found in 51% eyes by 5 years. A Hyderabad, India, Honavar and team inves-
more recent analysis of 457 consecutive tigated very high-risk retinoblastoma with
retinoblastomas focused on individual tu- tumor invasion into the orbit and designed
mor control with chemoreduction and focal new therapeutic regimens with success.
tumor consolidation. 12 Tumors treated with Their approach to advanced retinoblastoma
chemoreduction alone showed recurrence has been adapted by experts worldwide.
in 45% by 7 years follow up, whereas those
treated with chemoreduction plus thermo- The nightmare of retinoblastoma has be-
therapy, cryotherapy, or both showed re- come a fairytale with early detection of can-
currence in 18% by 7 years. cer and better therapeutic regimens. Future
chapters will focus on detection of the tu-
Following chemoreduction, tumor consoli- mor by dilated pediatric screening protocols
dation with thermotherapy or cryotherapy at or before 6 months of age, documented
is critical. Macular retinoblastoma repre- fundus photography, and prevention of
sents a specifically difficult situation regard- retinoblastoma in germline mutation fami-
ing therapy. Controversy exists regarding lies using preimplantation genetic diagnosis
the need or benefit of adjuvant focal con- and in vitro fertilization. Better therapeutic
solidation with thermotherapy following alternatives with anti-vascular endothelial
chemoreduction. In an analysis of 68 macu- growth factors (anti-VEGF) and local deliv-
lar retinoblastoma, it was found that 35% ery of chemotherapy are under investiga-
of those treated with chemoreduction alone tion. Our joint goal for retinoblastoma is
showed recurrence by 4 years compared to provide intervention at a time when we
to 17% of those treated with chemoreduc- can save the child’s life and eye, and even
tion plus extrafoveal thermotherapy.13 Sur- provide reasonable vision for a lifetime.
prisingly, small retinoblastomas were most
likely to show tumor recurrence believed to References
be related to the reduced chemotherapy
dose from small feeder vessels or to the 1.Shields JA, Shields CL. Management and
more well differentiated features of small prognosis of retinoblastoma. In Intraocular
retinoblastomas with less responsiveness to Tumors. A Text and Atlas Philadelphia: WB
chemotherapy. Saunders 1992; 377-392.
2. Shields JA, Shields CL. Retinoblastoma. In
So it is evident that chemotherapy can
Intraocular Tumors. A Textbook and Atlas.
control retinoblastoma, but what about
Philadelphia: Lippincott Williams Wilkins
its toxicities. The current six cycle regimen
2008; 293-365.
causes transient bone marrow suppression
with pancytopenia and a risk for infection. 3. Epstein J, Shields CL, Shields JA. Trends in
The potential risk for induction of second the management of retinoblastoma; Evalua-
cancers is not known, but is predicted to tion of 1,196 consecutive eyes during 1974-
be minimal due to the low dose, short-term 2001. J Pediatr Ophthalmol Strabismus
treatment. There is a concern for etoposide- 2003;40:196-203.
induced leukemia in children treated with
4. Shields CL, Mashayekhi A, Demirci H,
higher total dose than our current proto-
Meadows AT, Shields JA. A practical ap-
col. Other concerns include ototoxicity and
proach to management of retinoblastoma.
nephrotoxicity. All of these complications
Arch Ophthalmol 2004; 122:729-735.
should be seriously considered before treat-
ing a young child with these potentially 5. Wong FL, Boice JD Jr, Abramson DH, Tar-
toxic agents. one RE, Kleinerman RA, Stovall M, Goldman
MB, Seddon JM, Tarbell N, Fraumeni JF Jr, Li
Honavar and coworkers have contributed FP. Cancer incidence after retinoblastoma.
substantially to our understanding of retin- Radiation dose and sarcoma risk. JAMA
oblastoma. While working with our team in 1997;278:1262-1267.
Philadelphia, we collaborated and published
several reports including the results of in- 6. Kingston JE, Hungerford JL, Madreperla
traocular surgery after treatment of retino- SA, Plowman PN. Results of combined chem-
blastoma, the trepidation of vitrectomy in otherapy and radiotherapy for advanced in-
eyes with unsuspected retinoblastoma, and traocular retinoblastoma. Arch Ophthalmol
numerous studies on the effectiveness of 1996;114:1339-1343.
chemoreduction for retinoblastoma. 10,11,14,15 7. Shields CL, De Potter P, Himelstein BP,
Perhaps one of his most important contri- Shields JA, Meadows AT, Maris JM. Chem-
butions was the publication on the benefits oreduction in the initial management of in-

Foreword

traocular retinoblastoma. Arch Ophthalmol 12. Shields CL, Mashayekhi A, Shelil A, Cater
1996;114:1330-1338. J, Meadows AT, Shields JA. Chemoreduc-
tion for retinoblastoma. Analysis of tumor
8. Ferris FL, Chew EY. A new era for the
control and risks for recurrence in 457 tu-
treatment of retinoblastoma. Arch Oph-
mors Am J Ophthalmol 2004;138:329-37.
thalmol 1996; 114: 1412.
13. Shields CL, Mashayekhi A, Shelil A, Ness
9. Shields CL, Shields JA, DePotter P, Himel- S, Cater J, Meadows AT, Shields JA. Macu-
stein BP, Meadows AT: The effect of chem- lar retinoblastoma managed with chemore-
oreduction on retinoblastoma-induced reti- duction. Analysis of tumor control with or
nal detachment. J Pediatric Ophthalmol & without adjuvant thermotherapy in 68 tu-
Strabismus1997,34:165-169. mors. Arch Ophthalmol 2005;123:765-73.
10. Shields CL, Honavar SG Meadows AT, 14. Honavar SG, Shields CL, Shields JA,
Shields JA, Demirci H, Singh AD, Friedman Demirci H, Naduvilath TJ. Intraocular sur-
D, Naduvilaths TJ. Chemoreduction plus fo- gery after treatment of retinoblastoma.
cal therapy for retinoblastoma: Factors pre- Arch Ophthalmol. 2001;119:1613-21.
dictive of need for treatment with external
15. Shields CL, Honavar S, Shields JA, Demir-
beam radiotherapy or enucleation. Am J
ci H, Meadows AT. Vitrectomy in eyes with
Ophthalmol 2002;133:657-664.
unsuspected retinoblastoma. Ophthalmol-
11. Shields CL, Honavar SG, Shields JA, ogy. 2000;107:2250-5.
Demirci H, Meadows AT, Naduvilath TJ. 16. Honavar SG, Singh AD, Shields CL,
Factors predictive of recurrence of retinal Meadows AM, Demirci H, Cater J, Shields
tumor, vitreous seeds, and subretinal seeds JA. Postenucleation adjuvant therapy in
following chemoreduction for retinoblasto- high-risk retinoblastoma. Arch Ophthalmol.
ma. Arch Ophthalmol 2002;120:460-464. 2002;120:923-931.


Retinoblastoma

Retinoblastoma
They live and see...


Introduction

Introduction stood at cross roads in the 1990s. The


outstanding issues related to identifi-
Retinoblastoma is the most common cation of a child at risk of developing
intraocular malignancy in children, with retinoblastoma by genetic testing, op-
a reported incidence ranging from 1 in timization of vision salvage by minimi-
15,000 to 1 in 18,000 live births.1 It is zation of the size of the tumor regres-
second only to uveal melanoma in the sion scar, reduction in the incidence of
frequency of occurrence of malignant second malignant neoplasm following
intraocular tumors. There is no racial external beam radiotherapy by explor-
or gender predisposition in the inci- ing for alternative therapeutic modali-
dence of retinoblastoma. Retinoblas- ties, reduction in the incidence of sys-
toma is bilateral in about 25 to 35% of temic metastasis following enucleation,
cases.2 The average age at diagnosis is and improvement in the prognosis of
18 months, unilateral cases being diag- orbital retinoblastoma and metastatic
nosed at around 24 months and bilat- retinoblastoma.
eral cases before 12 months. 2
The recent advances such as identifica-
Pawius described retinoblastoma as tion of genetic mutations, 5, 6 replace-
early as in 1597.3 In 1809, Wardrop re- ment of external beam radiotherapy by
ferred to the tumor as fungus haema- chemoreduction as the primary man-
todes and suggested enucleation as the agement modality, use of chemoreduc-
primary mode of management. 3 The tion to minimize the size of regression
discovery of ophthalmoloscope in 1851 scar with consequent optimization of
facilitated recognition of specific clini- visual potential, 7-11 identification of
cal features of retinoblastoma. Initially histopathologic high-risk factors follow-
thought to be derived from the glial ing enucleation 12 and provision of adju-
cells, it was called a glioma of the ret- vant therapy to reduce the incidence of
ina by Virchow (1864).3 Flexner (1891) systemic metastasis, 13 protocol-based
and Wintersteiner (1897) believed it to management of retinoblastoma with
be a neuroepithelioma because of the accidental perforation or intraocular
presence of rosettes.3 Later, there was surgery 14-16 and aggressive multimodal
a consensus that the tumor originated therapy in the management of orbital
from the retinoblasts and the Ameri- retinoblastoma 17,18 have contributed to
can Ophthalmological Society officially improved outcome in terms of better
accepted the term retinoblastoma in survival, improved eye salvage and po-
1926.4 tential for optimal visual recovery.

Retinoblastoma was associated with


near certain death just over a century
ago. Early tumor recognition aided by
Genetics of
indirect ophthalmoscopy and refined Retinoblastoma
enucleation technique contributed to
an improved survival from 5% in 1896 Out of the newly diagnosed cases of
to 81% in 1967.2 Advances in external retinoblastoma only 6% are familial
beam radiotherapy in the 1960s and while 94% are sporadic. 2,19 Bilateral
1970s and further progress in planning retinoblastomas involve germinal mu-
and delivery provided an excellent al- tations in all cases. Approximately 15%
ternative to enucleation and resulted of unilateral sporadic retinoblastoma is
in substantial eye salvage. 2 Focal thera- caused by germinal mutations affecting
peutic measures such as cryotherapy, only one eye while the 85% are spo-
photocoagulation and plaque brachy- radic.2
therapy allowed targeted treatment of
smaller tumors entailing vision salvage.2 In 1971, Knudson proposed the two hit
Parallel advancements in ophthalmic hypothesis.20 He stated that for retino-
diagnostics and introduction of ultra- blastoma to develop, two chromosom-
sonography, computed tomography, al mutations are needed. In hereditary
and magnetic resonance imaging con- retinoblastoma, the initial hit is a germi-
tributed to improved diagnostic accu- nal mutation, which is inherited and is
racy and early detection of extraocular found in all the cells. The second hit de-
retinoblastoma. velops in the somatic retinal cells lead-
ing to the development of retinoblas-
Despite all the advances that took toma. Therefore, hereditary cases are
place between 1960 and 1990, the predisposed to the development of no-
overall management of retinoblastoma nocular tumors such as osteosarcoma.

Genetics of Retinoblastoma

In unilateral sporadic retinoblastoma, Knowledge of the full range of muta-


both the hits occur during the devel- tions can aid in the design of screening
opment of the retina and are somatic tests for individuals at risk.6
mutations. Therefore there is no risk
of second nonocular tumors.
Mutational analysis of the RB1 gene in Indian
patients with retinoblastoma
Genetic counseling is an important as-
Ata-ur-Rasheed M, Vemuganti G, Honavar SG,
pect in the management of retinoblas- Ahmed N, Hasnain S, Kannabiran C.
toma. In patients with a positive family
Ophthalmic Genet. 2002;23:121-8
history, 40% of the siblings would be
at risk of developing retinoblastoma Twenty-one probands, twelve with bilateral
and nine with unilateral retinoblastoma, were
and 40% of the offspring of the af- screened for mutations in the RB1 gene using
fected patient may develop retinoblas- genomic DNA from peripheral blood leuko-
toma. In patients with no family his- cytes as well as tumors. Amplification of indi-
vidual exons and flanking regions of the RB1
tory of retinoblastoma, if the affected gene were carried out, followed by direct se-
child has unilateral retinoblastoma, 1% quencing of the amplified products. Sequenc-
of the siblings are at risk and 8% of es of affected individuals were compared with
those of controls. Mutations were identified in
the offspring may develop retinoblas- seven patients, five with bilateral and two with
toma. In cases of bilateral retinoblas- unilateral retinoblastoma. Six out of seven mu-
toma with no positive family history, tations involved the formation of premature
termination codons by means of single base
6% of the siblings and 40% of the off- substitutions (2), frameshifts due to splice-site
spring have a chance of developing mutations (2), or deletion and duplication (2).
retinoblastoma. 2 One missense mutation was identified. Of the
remaining fourteen patients, seven with bilat-
eral disease had no mutations in peripheral
Apart from empiric genetic counseling blood (7 cases) or tumors (3/7 cases). Analysis
as described above, the current trend is of the peripheral blood of seven patients with
to identify the mutation and compute unilateral disease also showed no mutations.
Mutations were detected in about one-third
specific antenatal risk. We screened of the cases, suggesting that hemizygous dele-
twenty-one probands, twelve with bi- tions at the RB1 locus or mutations outside the
lateral retinoblastoma and 9 with uni- coding regions of RB1 may be responsible for
the disease in the remaining patients.
lateral retinoblastoma, for mutations in
the RB1 gene using genomic DNA from
peripheral blood leukocytes as well as Mutational screening of the RB1 gene in Indi-
tumors. Amplification of individual ex- an patients with retinoblastoma reveals eight
ons and flanking regions of the RB1 novel and several recurrent mutations
gene were carried out, followed by di- Kiran VS, Kannabiran C, Chakravarthi K, Ve-
rect sequencing of the amplified prod- muganti GK, Honavar SG
ucts. Sequences of affected individuals Hum Mutat. 2003; 22:339
were compared with those of controls. Retinoblastoma is the most common primary
Mutations were identified in seven pa- intraocular malignancy in children, caused by
inactivation of the RB1 gene on chromosome
tients, five with bilateral and two with 13. We carried out a mutational screen of the
unilateral retinoblastoma. Analysis of exons and promoter of the RB1 gene in Indian
the peripheral blood of seven patients patients with retinoblastoma in order to de-
termine the range of mutations giving rise to
with unilateral disease showed no mu- disease. Forty-seven patients were screened
tations. 5 for mutations in all exons and promoter of
the RB1 gene by single strand conformation
polymorphism followed by sequencing. Tu-
Subsequently, we carried out mu- mors were available from 27 patients (12 bi-
tational screening of the exons and lateral and 15 unilateral retinoblastoma) while
promoter of the RB1 gene in Indian only peripheral blood was available from 20
patients, all with bilateral disease. Mutations
patients with retinoblastoma in order were found in 22 patients, 9 from the analysis
to determine the range of mutations of tumors and 13 from peripheral blood. Eight
giving rise to the disease. Eight novel novel mutations were identified, including 4
single base changes, 2 small deletions and 1 du-
mutations were identified, including plication. These are g.64365T>G (Tyr325Ter),
4 single base changes, 2 small dele- g.78131G>A (Trp515Ter), g.150061G>T (Glu-
tions and 1 duplication. These were 587Ter), g.170383C>G (S834X), g.41924A>C
(IVS3-2A>C), g.150064ins4, g.160792del22,
g.64365T>G (Tyr325Ter), g.78131G>A and g.76940del14 (IVS15 del +20-33). Almost
(Trp515Ter), g.150061G>T (Glu- all mutations produced nonsense codons or
587Ter), g.170383C>G (S834X), frameshifts. Recurrent mutations, especially
at CpG sites were seen predominantly. Detect-
g.41924A>C (IVS3-2A>C),g.150064ins4, able mutations in exons were found in 46%
g.160792del22, and g.76940del14 of patients tested. Large deletions, epigenetic
changes as well as mutations in non-coding
(IVS15 del +20-33). All mutations pro- regions may be the cause of disease in the
duced nonsense codons or frameshifts. remainder of patients. Knowledge of the full
Detectable mutations in exons were range of mutations can aid in the design of
screening tests for individuals at risk.
found in 46% of patients tested.

Histopathology of Retinoblastoma

Histopathology of Figure 1
Retinoblastoma
On low magnification, basophilic areas
of tumor are seen along with eosinophilic
areas of necrosis and more basophilic
areas of calcification within the tumor.
Poorly differentiated tumors consist of
small to medium sized round cells with
large hyperchromatic nuclei and scanty
cytoplasm with mitotic figures. Well-dif-
ferentiated tumors show the presence
of rosettes and fleurettes. These can be
of various types. Flexner-Wintersteiner Figure 2
rosettes consist of columnar cells ar-
ranged around a central lumen. This is
highly characteristic of retinoblastoma
and is also seen in medulloepithelioma.
Homer Wright rosettes consist of cells
arranged around a central neuromus-
cular tangle. This is also found in neu-
roblastomas, medulloblastomas and
medulloepitheliomas. Pseudorosette
refers to the arrangement of tumor
cells around blood vessels. They are not
signs of good differentiation. Fleurettes
are eosinophilic structures composed The clinical presentation of retinoblas-
of tumor cells with pear shaped eosi- toma depends on the stage of the
nophilic processes projecting through disease.10 Early lesions are likely to be
a fenestrated membrane. Rosettes and missed, unless an indirect ophthalmos-
fleurettes indicate that the tumor cells copy is performed. The tumor appears
show photoreceptor differentiation. In as a translucent or white fluffy retinal
addition basophilic deposits (precipitat- mass (Figure 1). The child may present
ed DNA released after tumor necrosis) with strabismus if the tumor involves
can be found in the walls of the lumen the macula or with reduced visual acu-
of blood vessels. 2 ity.

Clinical Manifestations of Moderately advanced lesions usually


present with leucocoria due to the re-
Retinoblastoma flection of light by the white mass in
the fundus (Figure 2). As the tumor
Leucocoria is the most common pre- grows further, three patterns are usu-
senting feature of retinoblastoma, fol- ally seen: 10
lowed by strabismus, painful blind eye
• Endophytic, in which the tumor grows
and loss of vision. Table 1 lists the com-
into the vitreous cavity (Figure 3). A
mon presenting signs and symptoms of
yellow white mass progressively fills
retinoblastoma. 21
the entire vitreous cavity and vitreous
seeds occur. The retinal vessels are
Table 1. Common Presenting Features of not seen on the tumor surface.
Retinoblastoma
• Exophytic, in which the tumor grows
1 Leucocoria 56% towards the subretinal space (Figure
2 Strabismus 20% 4). Retinal detachment usually occurs
and retinal vessels are seen over the
3 Red painful eye 7%
tumor.
4 Poor vision 5%
• Diffuse infiltrating tumor, in which
5 Asymptomatic 3%
the tumor diffusely involves the retina
6 Orbital Cellulitis 3% causing just a placoid thickness of the
7 Unilateral Mydriasis 2% retina and not a mass. This is gener- Figure 1. Early manifestation of retinoblastoma
8 Heterochromia Iridis 1%
ally seen in older children and usually with a localized tumor
there is a delay in the diagnosis (Fig- Figure 2. Lecocoria is the most common clinical
9 Hyphema 1% ure 5). presentation of retinoblastoma

Clinical Manifestations of Retinoblastoma

Advanced tumors manifest with prop-


Figure 7
tosis secondary to optic nerve extension
or orbital extension (Figure 6) and sys-
temic metastasis. 10 Retinoblastoma can
spread through the optic nerve with
relative ease especially once the lamina
cribrosa is breached. Orbital extension
may present with proptosis and is most
likely to occur at the site of the scleral
emissary veins. Systemic metastasis oc-
curs to the brain, skull, distant bones
and the lymph nodes.

Figure 3 Figure 8

Figure 4 Figure 9

Figure 5 Figure 10
Figure 3. Endophytic tumor with vitreous
seeds
Figure 4. Exophytic retinal tumor with exuda-
tive retinal detachment
Figure 5. Diffuse infiltrative retinoblastoma
with placoid retinal thickening seen on gross
examination of the enucleated eye in a 7-year-
old child
Figure 6. Retinoblastoma with orbital exten-
sion in a 3-year-old child
Figure 7. A 5-year-old child with retinoblasto-
ma with anterior segment seeding manifesting
with tumor hypopyon
Figure 6 Figure 11
Figure 8. A 4-year-old with spontaneous hy-
phema in the left eye. Ultrasonography con-
firmed the diagnosis of retinoblastoma.
Figure 9. Spontaneous vitreous hemorrhage as
the presenting feature of retinoblastoma in a
4-year-old child
Figure 10. An 18-month-old child with bilateral
retinoblastoma. The right eye has secondary
glaucoma and enlarged cornea while the left
eye is phthisical.
Figure 11. A 3-year-old child with retinoblas-
toma presenting with orbital cellulites
10
Diagnosis of Retinoblastoma

Some of the atypical manifestations of organ salvage while staging prognosti-


retinoblastoma include pseudohypopy- cates survival.
on (Figure 7), spontaneous hyphema
(Figure 8), vitreous hemorrhage (Figure The Reese Ellsworth classification was
9), phthisis bulbi (Figure 10) and presep- introduced to prognosticate patients
tal or orbital cellulites (Figure 11). 10 treated with methods other than enu-
cleation. 22 This classification was de-
vised prior to the widespread use of
Diagnosis of indirect ophthalmoscopy and focal
measures of management of retino-
Retinoblastoma blastoma and mainly pertained to eye
salvage with external beam radiother-
A thorough clinical evaluation with
apy. Although the Essen classification
careful attention to details, aided by
ultrasonography B-scan helps in the di-
Figure 12
agnosis.10 Computed tomography and
magnetic resonance imaging are gen-
erally reserved for cases with atypical
manifestations and diagnostic dilemma
and where extraocular or intracranial
tumor extension is suspected.10
A child with suspected retinoblastoma
necessarily needs complete ophthalmic
evaluation including a dilated fundus
examination under anaesthesia.10 The
intraocular pressure is measured and
the anterior segment is examined for
neovascularization, pseudohypopyon, Figure 13
hyphema, and signs of inflammation.10
Bilateral fundus examination with 360
degree scleral depression is mandatory.
Direct visualization of the tumor by an
indirect ophthalmoscope is diagnostic
of retinoblastoma in over 90% of cas-
es. 21 RetCam is a wide-angle fundus
camera, useful in accurately document-
ing retinoblastoma and monitoring re-
sponse to therapy (Figure 12).
Ultrasonography B-scan shows a round-
ed or irregular intraocular mass with Figure 14
high internal reflectivity representing
typical intralesional calcification (Fig-
ure 13). 10 Computed tomography de-
lineates extraocular extension and can
detect an associated pinealoblastoma
(Figure 14). 10 Magnetic resonance im-
aging is specifically indicated if optic
nerve invasion or intracranial extension
is suspected.10 On fluorescein angiogra-
phy, smaller retinoblastoma shows min-
imally dilated feeding vessels in the ar-
terial phase, blotchy hyperfluorescence
in the venous phase and late staining Figure 15
(Figure 15). 10 Figure 12. RetCam, a wide-angle digital fun-
dus camera and image archival system helps
in documentation and assessment of tumor
Classification of regression on follow-up
Figure 13. Ultrasonography B-scan showing
Retinoblastoma multifocal retinal tumors
Figure 14. Computed tomography scan shows
An ideal classification system for retino- pinealoblastoma
blastoma should include two compo- Figure 15. Fundus fluorescein angiography
nents: grouping and staging. Grouping in retinoblastoma in the early phase shows
is a clinical system of prognosticating blotchy hyperfluorescence
11
Classification of Retinoblastoma

Table 2. International Classification of Table 3. International Staging System for


Intraocular Retinoblastoma* Retinoblastoma
Group A Small tumors (< 3 mm) outside No enucleation (one or both eyes
macula Stage 0
may have intraocular disease)
Enucleation, tumor completely
Stage I
resected
Enucleation with microscopic
Stage II
residual tumor
Regional extension
Group B Bigger tumors (> 3 mm) or any tu- A Overt orbital disease
mor in macula or any tumor with Stage III
B Preauricular or cervical lymph
subretinal fluid
node extension
Stage IV Metastatic disease
A Hematogenous metastasis
1 Single lesion
2 Multiple lesions
B CNS Extension
Group C Localized seeds (subretinal or vitre-
ous) 1 Prechiasmatic lesion
2 CNS mass
3 Leptomeningeal disease

addressed some of the shortcomings of


Reese Ellsworth classification, it is con-
sidered too complex. Further, none of
the older systems of classification had
been designed to prognosticate chem-
oreduction, the current favored meth-
od of retinoblastoma management.
The new International Classification of
Intraocular Retinoblastoma is a logical
Group D Diffuse seeds (subretinal or vitre- flow of sequential tumor grading that
ous) linearly correlates with the outcome of
newer therapeutic modalities (Table 2).
23, 24

The new International Staging system


is the first such for retinoblastoma and
incorporates five distinct stages (Table
3). 25 Staging is based on collective in-
formation gathered by the clinical eval-
uation, imaging, systemic survey and
histopathology.

Group E Tumor touching the lens, Neovas- Management of


cular glaucoma, Tumor anterior Retinoblastoma
to anterior vitreous face involving
ciliary body or anterior segment, The primary goal of management of
Diffuse infiltrating retinoblastoma,
retinoblastoma is to save life. Salvage
Opaque media from hemorrhage,
Tumor necrosis with aseptic orbital
of the organ (eye) and function (vision)
cellulitis, and Phthisis bulbi are the secondary and tertiary goals
respectively. The management of retin-
oblastoma needs a multidisciplinary
team approach including an ocular on-
cologist, pediatric oncologist, radiation
oncologist, radiation physicist, genetist
Courtesy Carol L Shields, MD, Wills Eye Insti- and an ophthalmic oncopathologist.
tute, Philadelphia, PA, USA The management strategy depends on
12
Management of Retinoblastoma

Table 4. Current Suggested Protocol


A. Intraocular tumor, International Classification Group A to C, Unilateral or Bilateral
1. Focal therapy (cryotherapy or transpupillary thermotherapy) alone for smaller tumors (<
3mm in diameter and height) located in visually noncrucial areas
2. Standard 6 cycle chemoreduction and sequential aggressive focal therapy for larger tumors
and those located in visually crucial areas
3. Defer focal therapy until 6 cycles for tumors located in the macular and juxtapapillary
areas. Transpupillary thermotherapy or plaque brachytherapy for residual tumor in the
macular and juxtapapillary areas >6 cycles.
4. Focal therapy for small residual tumor, and plaque brachytherapy/external beam radiother-
apy (>12 months age) for large residual tumor if bilateral, and enucleation if unilateral.
B. Intraocular tumor, International Classification Group D, Unilateral or Bilateral
1. High dose chemotherapy and sequential aggressive focal therapy
2. Periocular carboplatin for vitreous seeds
3. Consider primary enucleation if unilateral, specially in eyes with no visual prognosis
C. Intraocular tumor, International Classification Group E, Unilateral or Bilateral
1. Primary enucleation
2. Evaluate histopathology for high risk factors
D. High risk factors on histopathology, International Staging, Stage 2
1. Baseline systemic evaluation for metastasis
2. Standard 6 cycle adjuvant chemotherapy
3. High dose adjuvant chemotherapy and orbital external beam radiotherapy in patients with
scleral infiltration, extraocular extension, and optic nerve extension to transection.
E. Extraocular tumor, International Staging, Stage 3A
1. Baseline systemic evaluation for metastasis
2. High dose chemotherapy for 3-6 cycles, followed by enucleation or extended enucleation,
external beam radiotherapy, and continued chemotherapy for 12 cycles
F. Regional Lymph Node Metastasis, International Staging, Stage 3B
1. Baseline evaluation for systemic metastasis
2. Neck dissection, high dose chemotherapy for 6 cycles, followed by external beam radio-
therapy, and continued chemotherapy for 12 cycles
F. Hematogenous or Central Nervous System Metastasis, International Staging, Stage 4
1. Intent-to-cure or Palliative therapy in discussion with the family

the stage of the disease – intraocular present with intraocular retinoblasto-


retinoblastoma, retinoblastoma with ma while 60-70% present at this stage
high-risk characteristics, orbital retino- in the developing world.10 Diagnosis of
blastoma and metastatic retinoblas- retinoblastoma at this stage and appro-
toma. priate management are crucial for life,
eye and possible vision salvage.
Management of retinoblastoma is high-
ly individualized and is based on several There are several methods to man-
considerations - age at presentation, age intraocular retinoblastoma - focal
laterality, tumor location, tumor stag- (cryotherapy, laser photocoagulation,
ing, visual prognosis, systemic condition, transpupillary thermotherapy, transcle-
family and societal perception, and, to ral thermotherapy, plaque brachythera-
a certain extent, the overall prognosis py), local (external beam radiotherapy,
and cost-effectiveness of treatment in a enucleation), and systemic (chemother-
given economic situation (Table 4). apy). While primary focal measures are
mainly reserved for small tumors, local
Management of Intraocular and systemic modalities are used to
treat advanced retinoblastoma.
Retinoblastoma
A majority of children with retinoblas- Cryotherapy
toma manifest at the stage when the
tumor is confined to the eye. About 90- Cryotherapy is performed for small
95% of children in developed countries equatorial and peripheral retinal tu-
13
Focal Therapy

mors measuring up to 4 mm in basal di- detachment, retinal vascular occlusion,


ameter and 2 mm in thickness.2, 10 Triple retinal hole, retinal traction, and pre-
freeze thaw cryotherapy is applied at retinal fibrosis. It is less often employed
4-6 week intervals until complete tumor now with the advent of thermothera-
regression. Cryotherapy produces a scar py. In fact, laser photocoagulation is
much larger than the tumor (Figure 16). contraindicated while the patient is on
Complications of cryotherapy include active chemoreduction protocol. 10
transient serous retinal detachment,
retinal tear and rhegmatogenous reti-
nal detachment. Cryotherapy adminis- Thermotherapy
tered 2-3 hours prior to chemotherapy In thermotherapy, focused heat gener-
can increase the delivery of chemo- ated by infrared radiation is applied to
therapeutic agents across the blood tissues at subphotocoagulation levels to
retinal barrier and thus has synergistic induce tumor necrosis.26 The goal is to
effect.10 achieve a slow and sustained tempera-
ture range of 40 to 60 degree C within
Laser Photocoagulation the tumor, thus sparing damage to the
retinal vessels (Figure 17). Transpupil-
Laser photcoagulation is used for small
lary thermotherapy using infrared ra-
posterior tumors 4 mm in basal diame-
diation from a semiconductor diode la-
ter and 2 mm in thickness. 2, 10 The treat-
ser delivered with a 1300-micron large
ment is directed to delimit the tumor
spot indirect ophthalmoscope delivery
and coagulate the blood supply to the
system has become a standard prac-
tumor by surrounding it with two rows
tice. It can also be applied transpupil-
of overlapping laser burns. Complica-
lary through an operating microscope
tions include transient serous retinal
or by the transscleral route with a di-
opexy probe. The tumor is heated until
Figure 16 A it turns a subtle gray. Thermotherapy
provides satisfactory control for small
tumors - 4 mm in basal diameter and
2 mm in thickness. Complete tumor re-
gression can be achieved in over 85%
of tumors using 3-4 sessions of thermo-
therapy. 26 The common complications
are focal iris atrophy, focal paraxial
lens opacity, retinal traction and serous
retinal detachment. The major applica-
tion of thermotherapy is as an adjunct
to chemoreduction. The application of
Figure 16 B heat amplifies the cytotoxic effect of
platinum analogues. This synergistic
combination with chemoreduction pro-
tocol is termed chemothermotherapy.

Plaque Brachytherapy

Plaque brachytherapy involves place-


ment of a radioactive implant on the
sclera corresponding to the base of the
tumor to transsclerally irradiate the tu-
mor. 27 Commonly used radioactive ma-
17 terials include Ruthenium 106 (Figure
Figure 17
18) and Iodine 125. The advantages of
plaque brachytherapy are focal deliv-
Figure 16. A peripheral retinal tumor that un- ery of radiation with minimal damage
derwent cryotherapy (16 A). The tumor has to the surrounding normal structures,
completely regressed but the scar is much larg- minimal periorbital tissue damage, ab-
er than the tumor itself (16 B). sence of cosmetic abnormality because
Figure 17. Two focal tumors treated with of retarded bone growth in the field
transpupillary thermotherapy: note flat scars of irradiation as occurs with external
with patent blood vessels coursing through the
scars. Transpupillary thermotherapy classically
beam radiotherapy, reduced risk of sec-
spares the blood vessels from occlusion and ond malignant neoplasm and shorter
produces a compact scar. duration of treatment.
14
External Beam Radiotherapy

Figure 18 Figure 19

Plaque brachytherapy is indicated in External beam radiotherapy can induce


tumors less than 16 mm in basal diam- second malignant neoplasm especially
eter and less than 8 mm thickness. It in patients with the hereditary form of
could be the primary or secondary mo- retinoblastoma (Figure 19). There is a
dality of management. Primary plaque high 30% chance of developing anoth-
brachytherapy is currently performed er malignancy by the age of 30 years in
only in situations where chemotherapy such patients if they are given external
is contraindicated. It is most useful as beam radiotherapy compared to a less
secondary treatment in eyes that fail than 6% chance in those who do not
to respond to chemoreduction and ex- receive external beam radiotherapy. 30
ternal beam radiotherapy or for tumor The risk of second malignant neoplasm
recurrences. is greater in children under 12 months
of age. 30
Plaque brachytherapy requires precise
tumor localization and measurement Enucleation
of its basal dimensions. The tumor
thickness is measured by ultrasonog- Enucleation is a common method of
raphy. The data is used for dosimetry managing advanced retinoblastoma.
on a three-dimensional computerized Just about 3 decades ago, a majority of
tumor modeling system. The plaque patients with unilateral retinoblastoma
design is chosen depending on the ba- and the worse eye in bilateral retino-
sal tumor dimensions, its location, and blastoma underwent primary enuclea-
19 A substantial reduction in the
tion.
configuration. The dose to the tumor
apex ranges from 4000-5000 cGy. The frequency of enucleation has occurred
plaque is sutured to the sclera after in the late last century. 31 Concurrently,
confirming tumor centration and is left there has been an increase in the use of
in situ for the duration of exposure, alternative eye- and vision-conserving
generally ranging from 36 to 72 hours. methods of treatment. 9, 32
The results of plaque brachytherapy Primary enucleation continues to be
are gratifying with about 90% tumor the treatment of choice for advanced
control. The common complications are intraocular retinoblastoma with neo-
radiation papillopathy and radiation vascularization of iris, secondary glau-
retinopathy. coma, anterior chamber tumor invasion,
tumors occupying >75% of the vitreous
External Beam Radiotherapy volume, necrotic tumors with second-
ary orbital inflammation, and tumors
External beam radiotherapy was the associated with hyphema or vitreous
preferred form of management of mod- hemorrhage where the tumor charac-
erately advanced retinoblastoma in late teristics can not be visualized, especially
1900s.28, 29 However with the advent of when only one eye is involved. 10
newer chemotherapy protocols, exter-
nal beam radiotherapy is being used There are specific considerations while
less often. Presently it is indicated in enucleating an eye with retinoblasto-
eyes where primary chemotherapy and ma. (Table 5) Minimum-manipulation
local therapy has failed, or rarely when surgical technique should be necessarily
chemotherapy is contraindicated. 10 practiced.11 It is important not to acci-
dentally perforate the eye. The sclera is Figure 18. Ruthenium 106 plaque
The major problems with external beam thin at the site of muscle insertions and Figure 19. Osteosarcoma of the frontal bone
radiotherapy are the stunting of the the rectus muscles have to be hooked in a 20-year-old patient with bilateral retino-
orbital growth, dry eye, cataract, radia- delicately. It is important to obtain a blastoma who had undergone external beam
tion retinopathy and optic neuropathy. long optic nerve stump, ideally more radiotherapy at 1-year age

15
Enucleation

Table 5. Special Considerations for tents of the superior orbital fissure),


Enucleation in Retinoblastoma the blades of the scissors are opened to
a. Minimal manipulation
engage the optic nerve, and the nerve
is transected with one bold cut. This
b. Avoid perforation of the eye maneuver generally provides at least
Harvest long (> 15 mm) optic nerve 15 mm long optic nerve stump. 11 Enu-
c.
stump cleation spoon and heavy enucleation
Inspect the enucleated eye for scissors limit space for maneuverability
d. macroscopic extraocular extension and and may result in a shorter optic nerve
optic nerve involvement stump. In addition, one should be care-
e. Harvest fresh tissue for genetic studies ful not to accidentally perforate the eye
during enucleation. The enucleated
Avoid biointegrated implant if
f.
postoperative radiotherapy is necessary
eyeball is inspected for optic nerve (Fig-
ure 20) or extraocular extension (Figure
than 15 mm, but never less than 10 mm 21) of tumor.
(Figure 20). 11 Eyes manifesting tumor necrosis with
aseptic orbital cellulitis pose specific
Certain steps can be taken to obtain problem. These patients should be im-
about 15 mm long optic nerve stump in aged to rule out extraocular extension.
all cases of advanced retinoblastoma. 11 Enucleation is best performed when
Gentle traction can be applied by the the inflammation is resolved. 11 A brief
traction sutures applied to recti muscle course of preoperative oral and topi-
stumps prior to transecting the optic cal steroids help control inflammation.
nerve. As an alternative to the traction Patients with retinoblastoma present-
sutures, medial or lateral rectus muscle ing as phthisis bulbi need imaging to
stumps may be kept long and traction exclude extraocular and optic nerve
exerted with an artery clamp. A 15-de- extension. 11 Phthisis generally results
gree curved and blunt-tipped tenotomy following spontaneous tumor necrosis
scissors is introduced from the lateral and an episode of aseptic intraocular
aspect (or a straight scissors from the and orbital inflammation. Enucleation
medial aspect) and the optic nerve is in these cases is often complicated by
palpated with the closed tip of the scis- excessive peribulbar fibrosis and intra-
sors while maintaining gentle traction operative bleeding. 11
on the eyeball. The scissors is moved
posteriorly to touch the orbital apex Placement of an orbital implant follow-
while “strumming” the optic nerve. The ing enucleation for retinoblastoma is
scissors is lifted by 3 or 4 millimeters off the current standard of care. The or-
the orbital apex (to preserve the con- bital implant promotes orbital growth,

Figure 20 Figure 22 A

Figure 21 Figure 22 B
Figure 20. Enucleated eyeball showing 18 mm
optic nerve stump. Note the proximal portion
of the optic nerve is thickened indicating tu-
mor infiltration.
Figure 21. Enucleated eyeball showing extras-
cleral tumor extension
Figure 22. Retinoblastoma in the right eye fol-
lowing enucleation with orbital implant by the
myoconjunctival technique (22 A). Excellent
cosmesis follwing fitting of a custom ocular
prosthesis (22 B).
16
Chemotherapy

provides better cosmesis and enhances for tumor, subretinal seed and vitreous
prosthesis motility. The implants could seed recurrence, and failure of chem-
be non-integrated (polymethyl meth- oreduction leading to external beam
acrylate or silicon) or bio-integrated (hy- radiotherapy and/or enucleation have
droxyapatite or porous polyethylene). been identified. 7, 8 Chemoreduction of-
Placement of a biointegrated implant fers satisfactory tumor control for Reese
is generally avoided if post-operative Ellsworth groups I-IV eyes, with treat-
adjuvant radiotherapy is considered ment failure necessitating additional
necessary. 11 Although most implants external beam radiotherapy in only 10%
structurally tolerate radiotherapy well, and enucleation in 15% at 5-year follow-
up. Patients with Reese Ellsworth group
implant vascularization may be com-
V eyes require external beam radiother-
promised by radiotherapy thus increas- apy in 47% and enucleation in 53% at
ing the risk of implant exposure. Use of 5 years. 7, 8 Chemoreduction is an option
myoconjunctival technique and custom for selected eyes with unilateral retino-
ocular prosthesis have optimized pros- blastoma. 9
thesis motility and static cosmesis (Fig-
ure 22). Figure 23 shows a juxtapapillary tumor
regressed with chemoreduction alone.
Chemotherapy Transpupillary thermotherapy was not
performed because of the crucial loca-
Chemoreduction, defined as the process tion. Figure 24, 25 and 26 show that
of reduction in the tumor volume with the resulting scar with chemoreduction
chemotherapy, has become an integral was much smaller than the original tu-
part of the current management of mor with the foveola fully exposed, thus
retinoblastoma.32 Chemotherapy alone maximizing visual potential. With the
is however not curative and must be modified protocol that we use specifi-
associated with intensive local therapy. cally for advanced retinoblastoma, our
Chemoreduction coupled with focal eye salvage rates are 100% for Reese
therapy can minimize the need for enu- Ellsworth groups 1-3, 90% for group D
cleation or external beam radiotherapy and 75% for group E (Table 7).
without significant systemic toxicity.
It is important to be aware of the adverse
Chemoreduction in combination with effects and interactions of chemothera-
focal therapy is now extensively used in peutic agents, which include myelosup-
the primary management of retinoblas- pression, febrile episodes, neurotoxicity
toma. 33-36 There are different protocols and non-specific gastrointestinal toxic-
in chemotherapy. The commonly used ity. Chemotherapy should be given only
drugs are vincristine, etoposide and car- under the supervision of an experienced
boplatin, for 6 cycles. 7-10 (Table 6) Stand- pediatric oncologist.
ard dose chemoreduction is provided in
ICIOR groups A-C. 10 In high dose chem-
Figure 23 A
oreduction, the dose of etoposide and
carboplatin is increased. This is indicated
in ICIOR groups D tumors. 10

With chemoreduction and sequential lo-


cal therapy, it is now possible to salvage
many an eye and maximize residual vi-
sion. Chemoreduction is most successful
for tumors without associated subretinal
fluid or vitreous seeding. 7,8 Risk factors

Table 6. Chemoreduction Regimen and Doses


for Intraocular Retinoblastoma
Figure 23 B
Day 1: Vincristine + Etoposide + Carboplatin
Day 2: Etoposide
Standard dose (3 weekly, 6 cycles): Vincris-
tine 1.5 mg/m2 (0.05 mg/kg for children < 36
months of age and maximum dose < 2mg),
Etoposide 150 mg/m2 (5 mg/kg for children
< 36 months of age), Carboplatin 560 mg/m2
(18.6 mg/kg for children < 36 months of age)
High-dose (3 weekly, 6-12 cycles): Vincristine Figure 23. Juxtapapillary retinoblastoma in a 6-
0.025 mg/Kg, Etoposide 12 mg/Kg, Carbopla- month-old child (23 A), completely regressed
tin 28 mg/Kg with 6 cycles of chemoreduction alone (23 B)
17
Chemotherapy

Figure 24 A Figure 24 B

Figure 25 A Figure 25 B

Figure 26 A Figure 26 B

Table 7. Eye Salvage Rates with External Beam Radiotherapy Vs Chemoreduction


Reese Ellsworth Ellsworth, 1977 Hungerford, 1995 Shields, 2003 LVPEI, 2005
Group EBRT EBRT Chemoreduction Chemoreduction*
I 91% 100% 100% 100%
II 83% 84% 100% 100%
III 82% 82% 100% 100%
IV 62% 43% 75% 90%
V 29% 66% 50% 75%
* High dose chemotherapy for group V, periocular chemotherapy for VB

Factors predictive of recurrence of retinal tu- PANTS: There were 158 eyes with 364 tumors
mors, vitreous seeds, and subretinal seeds fol- in 103 consecutive patients with retinoblasto-
Figure 24. Multifocal retinoblastoma (24 A) lowing chemoreduction for retinoblastoma ma managed with chemoreduction between
following chemoreduction and transpupillary Shields CL, Honavar SG, Shields JA, Demirci H, June 1994 and August 1999. INTERVENTION:
Meadows AT, Naduvilath TJ All patients received treatment for retinoblas-
thermotherapy (24 B). Note flat scars that are toma with 6 cycles of chemoreduction using
much smaller than the original tumor. Arch Ophthalmol. 2002;120:460-4 vincristine, etoposide, and carboplatin com-
Figure 25. Multifocal retinoblastoma (25 A) bined with focal treatment (cryotherapy, ther-
OBJECTIVE: To identify the clinical features motherapy, or plaque radiotherapy) for each
regressed following chemoreduction and retinal tumor. MAIN OUTCOME MEASURES:
of eyes with retinoblastomas that predict the
transpupillary thermotherapy (25 B) recurrence of retinal tumors, vitreous seeds, The 3 main outcome measures included recur-
Figure 26. A juxtapapillary retinal tumor in a and subretinal seeds following treatment with rence of retinal tumors, recurrence of vitre-
chemoreduction. DESIGN: Prospective nonran- ous seeds, and recurrence of subretinal seeds.
9-month-old child (26 A) completely regressed The clinical features at the initial examination
domized single-center clinical trial. SETTING:
with 6 cycles of chemoreduction and transpu- Ocular oncology service at Wills Eye Hospital of were analyzed for their association with the
pillary thermotherapy (26 B). Note the com- Thomas Jefferson University (Philadelphia, Pa) main outcome measures using a series of Cox
pletely exposed fovea following treatment, in conjunction with the division of oncology at proportional hazards regressions. RESULTS: All
thus maximizing visual potential. Children’s Hospital of Philadelphia. PARTICI- retinal tumors, vitreous seeds, and subretinal

18
Chemotherapy

seeds showed an initial favorable response of I-IV eyes and 47% of RE group V eyes. Multivari-
regression during this treatment regimen. Us- ate factors predictive of treatment with exter-
ing Kaplan-Meier estimates, at least 1 retinal nal beam radiotherapy included non-Caucasian
tumor recurrence per eye was found in 37% of
eyes at 1 year, 51% at 3 years, and no further race, male sex, and RE group V disease. Failure
increase at 5 years. By multivariate analysis, of chemoreduction and the need for treatment
the only factor predictive of retinal tumor re- with enucleation occurred in 13% eyes at 1 year,
currence was the presence of tumor-associated 29% at 3 years, and 34% at 5 years. More spe-
subretinal seeds at the initial examination. Of cifically, enucleation was necessary in 15% of
the 54 eyes that had vitreous seeds at the ini- RE groups I-IV eyes at 5 years and in 53% of
tial examination, vitreous seed recurrence was RE group V at 5 years. Multivariate factors pre-
found in 26% of eyes at 1 year, 46% at 3 years, dictive of treatment with enucleation included
and 50% at 5 years. By univariate analysis, the patient age older than 12 months, single tumor
only factor predictive of vitreous seed recur- in eye, and tumor proximity to foveola within
rence was the presence of tumor-associated 2 mm. Overall, of the 158 eyes, 50% required
subretinal seeds at the initial examination. Of external beam radiotherapy or enucleation and
the 71 eyes that had subretinal seeds at the 50% were successfully managed without these
initial examination, subretinal seed recurrence treatments. No patient developed retinoblas-
was detected in 53% of eyes at 1 year, 62% at toma metastasis, pinealoblastoma, or second
3 years, and no further increase at 5 years. By
multivariate analysis, factors predictive of sub- malignant neoplasms over the 5-year follow
retinal seed recurrence included a tumor base up. CONCLUSIONS: Chemoreduction offers sat-
greater than 15 mm and a patient age of 12 isfactory retinoblastoma control for RE groups
months or younger at diagnosis. There were I-IV eyes, with treatment failure necessitating
no patients who developed retinoblastoma additional external beam radiotherapy in only
metastasis, pinealoblastoma, or second malig- 10% of eyes and enucleation in 15% of eyes at
nant neoplasms. CONCLUSIONS: Chemoreduc- 5-year follow-up. Patients with RE group V eyes
tion combined with focal therapy is effective require external beam radiotherapy in 47% and
for selected eyes with retinoblastomas. Eyes enucleation in 53% at 5 years.
with subretinal seeds at initial examination
are at particular risk for recurrence of retinal
tumor and vitreous seeds. Younger patients
with large tumors are at risk for recurrence Periocular Chemotherapy
of subretinal seeds. Retinal tumor and subreti-
nal seed recurrence seems to manifest within Carboplatin delivered deep posterior
3 years of follow-up. Close follow-up of all subtenon has been demonstrated to
patients treated with chemoreduction is war-
ranted. be efficacious in the management of
Reese Ellsworth Group VB retinoblas-
Chemoreduction plus focal therapy for retino-
blastoma: factors predictive of need for treat- toma with vitreous seeds because it
ment with external beam radiotherapy or enu- can penetrate the sclera and achieve
cleation effective concentrations in the vitreous
Shields CL, Honavar SG, Meadows AT, Shields cavity. This modality is currently under
JA, Demirci H, Singh A, Friedman DL, Naduvi- trial. Our early results have shown that
lath TJ
periocular chemotherapy achieves 70%
Am J Ophthalmol. 2002;133:657-64
eye salvage in patients with retinoblas-
PURPOSE: To report the results of chemore- toma with diffuse vitreous seeds (Fig-
duction and focal therapy for retinoblastoma
with determination of factors predictive of the ure 27).37
need for treatment with external beam radio-
therapy or enucleation. DESIGN: Interventional
case series. METHODS: One-hundred three pa- Figure 27 A
tients with retinoblastoma (158 eyes with 364
tumors) at the Ocular Oncology Service at Wills
Eye Hospital of Thomas Jefferson University in
conjunction with the Division of Oncology at
Children’s Hospital of Philadelphia from June
1994 to August 1999 were enrolled for this pro-
spective clinical trial. The patients received treat-
ment for retinoblastoma with six planned cycles
(one cycle per month) of chemoreduction using
vincristine, etoposide, and carboplatin com-
bined with focal treatments (cryotherapy, ther-
motherapy, or plaque radiotherapy). The two
main outcome measures after chemoreduction
and focal therapy were the need for external
beam radiotherapy and the need for enuclea-
tion. The clinical features at the time of patient
presentation were analyzed for impact on the
main outcome measures using a series of Cox Figure 27 B
proportional hazards regressions. RESULTS: Us-
ing Reese-Ellsworth (RE) staging for retinoblas-
toma, there were nine (6%) eyes with group I
disease, 26 (16%) eyes with group II disease, 16
(10%) eyes with group III disease, 32 (20%) eyes
with group IV disease, and 75 (48%) eyes with
group V retinoblastoma. All eyes showed ini-
tial favorable response with tumor regression.
The median follow-up was 28 months (range,
2-63 months). Failure of chemoreduction and Figure 27. Retinoblastoma with massive vitre-
need for treatment with external beam radio-
ous seeds (Figure 27 A). Following 6 cycles of
therapy occurred in 25% of eyes at 1 year, 27%
at 3 years, and no further increase at 5 years. high-dose chemoreduction and periocular car-
More specifically, external beam radiotherapy boplatin injection, the tumour and the vitreous
was necessary at 5 years in 10% of RE groups seeds show complete regression (Figure 27 B).
19
High-Risk Retinoblastoma

Periocular carboplatin as an adjunctive therapy High–Risk Factors


in the treatment of advanced retinoblastoma
Honavar SG, Shome D, Naik M, Reddy VAP None of the clinical high-risk factors
seem to strongly correlate with mortal-
Proceedings of the XII International Congress of
ity. Recent studies have evaluated the
Ocular Oncology, Vancouver, Canada, 2005
role of histopathologic high-risk factors
Purpose: To evaluate the role of periocular car- identified following enucleation. The
boplatin injections as an adjunctive therapy in
identification of frequency and signifi-
retinoblastoma patients, with vitreous seeds.
Methods: We studied our first 10 patients with cance of high-risk histopathologic fac-
group V B (Reese-Ellsworth classification) retin- tors (Figures 28-31) that can reliably
oblastoma who received deep posterior sub- predict metastasis is vital for patient
tenon injections of carboplatin as an adjunctive selection for adjuvant therapy. Several
therapy in addition to systemic chemotherapy. studies have addressed this issue. 39, 41-49
We compared the results with a cohort of 10 It is now generally agreed that massive
patients with similar severity of disease who
received similar therapy except that they did
choroidal infiltration, retrolaminar op-
not receive periocular carboplatin injections as tic nerve invasion, invasion of the optic
a part of therapy protocol. The main outcome nerve to transection, scleral infiltration,
measures studied were tumor regression and fi- and extrascleral extension are the risk
nal visual outcome. Results: In the study group, factors that are predictive of metastasis
7 patients (70%) showed complete regression of (Table 8). 39, 41-49
tumor while 3 patients (30%) had progression
of tumor and needed enucleation. In the con-
trol group, 3 patients (30%) responded to ther-
apy and 7 patients (70%) required enucleation. Table 8. Histopathologic High-risk Factors
Although the above result was not statistically Predictive of Metastasis
significant (p=0.1789), it was highly significant
1. Anterior chamber seeding
clinically (40% absolute benefit increase). Side
effects included transient periocular inflamma- 2. Iris infiltration
tion in all. Conclusion: Periocular carboplatin
has a potential beneficial role as an adjunctive 3. Ciliary body infiltration
therapy in the treatment of advanced intraocu-
lar retinoblastoma. 4. Massive choroidal infiltration
Invasion of the optic nerve lamina
Follow-up Schedule 5.
cribrosa

The usual protocol is to schedule the 6. Retrolaminar optic nerve invasion


first examination 3–6 weeks after the
7. Invasion of optic nerve transection
initial therapy. In cases where chemore-
duction therapy has been administered, 8. Scleral infiltration
the examination should be done every
3 weeks with each cycle of chemother- 9. Extrascleral extension
apy. Patients under focal therapy are
evaluated and treated every 4-8 weeks
until complete tumor regression. Fol- The reported occurrence of ante-
lowing tumor regression, subsequent rior chamber seeding (7%), 45 massive
examination should be 3 monthly for choroidal infiltration (12-23%), 43-49 in-
the first year, 6 monthly for three years vasion of optic nerve lamina cribrosa
or until the child attains 6 years of age, (6-7%),43-49 retrolaminar optic nerve
and yearly thereafter. invasion (6-12%),43-49 invasion of optic
nerve transection (1-25%),43-49 scleral
infiltration (1-8%),43-49 and extrascleral
High Risk Retinoblastoma extension (2-13%),43-49 widely vary even
in developed countries. Vemuganti
Systemic metastasis is the main cause and associates have reported that 21%
for mortality in patients with retino- of the 76 eyes enucleated for advanced
blastoma. Although the life prognosis retinoblastoma in India had anterior
of patients with retinoblastoma has chamber seeding, 54% had massive
dramatically improved in the last three choroidal infiltration, 46% had optic
decades, with a reported survival of nerve invasion at or beyond the lamina
more than 90% in developed coun- cribrosa and 7% had scleral infiltration
tries,38 mortality is still as high as 50% in or extrascleral extension. 12 It is appar-
the developing nations. 39, 40 Reduction ent that the incidence of histopatho-
in the rate of systemic metastasis by logic risk factors is strikingly high in
identification of high-risk factors and developing countries compared to the
appropriate adjuvant therapy may help published data from developed coun-
improve survival. tries.
20
Adjuvant Therapy

Figure 28 Postenucleation adjuvant therapy in high-risk


retinoblastoma
Honavar SG, Singh AD, Shields CL, Meadows
AT, Demirci H, Cater J, Shields JA
Arch Ophthalmol. 2002;120:923-31
PURPOSE: The main purpose of this study was
to determine the efficacy of postenucleation
adjuvant therapy in preventing metastasis in
cases of high-risk retinoblastoma. METHODS:
This was a retrospective, nonrandomized com-
parative study. Of 1020 consecutive patients
with retinoblastoma had were managed at a
referral center between January 1974 and De-
cember 1999, 80 (8%) of those analyzed had
unilateral sporadic cases that were treated by
primary enucleation and that had high-risk
Figure 29 characteristics for metastasis on histopathol-
ogy reports (anterior chamber seeding, iris
infiltration, ciliary body infiltration, massive
choroidal infiltration, invasion of optic nerve
lamina cribrosa, retrolaminar optic nerve in-
vasion, invasion of optic nerve transection,
scleral infiltration, and extrascleral extension).
The main outcome measure was the develop-
ment of metastasis at a minimum follow-up
period of 12 months. RESULTS: There were 44
male and 36 female patients, with age ranging
from 1 day to 16 years (median, 33 months).
A single histopathologic high-risk characteristic
was present in 50 patients (62.5%). Thirty pa-
tients (37.5%) manifested 2 or more high-risk
characteristics. Forty-six patients (58%) had
received postenucleation adjuvant therapy
(chemotherapy with or without orbital exter-
Figure 30 nal beam radiotherapy). Adjuvant therapy was
not administered in 34 patients (42%). Metas-
tasis occurred in 10 patients (13%) at a median
of 9 months (range, 6-57 months) following
enucleation. Eight (80%) of those who devel-
oped metastasis had not received adjuvant
therapy. A significant difference (P =.02) was
found in the incidence of metastasis between
the group that had received adjuvant therapy
(4%; 2/46) and the group that had not (24%;
8/34). The beneficial effect of adjuvant thera-
py was statistically significant in subgroups of
patients with massive choroidal infiltration (P
=.04) or retrolaminar optic nerve invasion (P
=.04). There were no adjuvant therapy-related
serious systemic complications. CONCLUSION:
Postenucleation adjuvant therapy is safe and
Figure 31 effective in significantly reducing the occur-
rence of metastasis in patients with retino-
blastoma manifesting histopathologic high-risk
characteristics.

enucleation. The study used specific


predetermined histopathologic charac-
teristics for patient selection. A mini-
mum follow-up of 1 year was allowed
to include metastatic events that gen-
erally occur at a mean of 9 months fol-
lowing enucleation. 13, 50 The incidence
of metastasis was 4% in those who re-
ceived adjuvant therapy compared to
24% in those who did not. The study Figure 28. Histopathology of retinoblastoma
Adjuvant Therapy found that administration of adjuvant showing anterior chamber seeding, iris infiltra-
tion, trabecular meshwork infiltration and cili-
therapy significantly reduced the risk ary body invasion
Studies on the efficacy of adjuvant of metastasis in patients with high-risk
therapy to minimize the risk of metas- Figure 29. Histopathology of retinoblastoma
histopathologic characteristics. showing massive choroidal infiltration, scleral
tasis initiated in the 1970s were marked
infiltration and extrascleral extension
by variable results and provided no firm Our current practice is to administer
recommendation. 18 A recent study with Figure 30. Histopathology of retinoblastoma
6 cycles of a combination of carbopla- showing infiltration of the optic nerve beyond
a long-term follow-up provides useful tin, etoposide and vincristine (identical the lamina cribrosa
information. 13, 50 It included a subset of to the protocol used for chemoreduc- Figure 31. Histopathology of retinoblastoma
patients with unilateral sporadic retin- tion of intraocular retinoblastoma) in showing optic nerve infiltration to the level of
oblastoma who underwent primary patients with histopathologic high-risk transection
21
Orbital Retinoblastoma

characteristics. All patients with exten-


sion of retinoblastoma up to the level of Figure 32 A
optic nerve transection, scleral infiltra-
tion, and extrascleral extension receive
high dose chemotherapy for 12 cycles
and fractionated 4500 to 5000 cGy or-
bital external beam radiotherapy.

Orbital Retinoblastoma
Orbital retinoblastoma is rare in devel-
oped countries. Ellsworth observed a
steady decline in the incidence of orbit-
al retinoblastoma in his large series of Figure 32 B
1160 patients collected over 50 years.51
Orbital retinoblastoma is relatively more
common in the developing countries. In
a recent large multi-center study from
Mexico, 18% of 500 patients presented
with an orbital retinoblastoma. 52 A Tai-
wanese group reported that 36% (42 of
116) of their patients manifested with
orbital retinoblastoma. 53 The incidence
is higher (40%, 19 of 43) in Nepal, with
proptosis being the most common clini-
cal manifestation of retinoblastoma. 54
Figure 33 A

Clinical Manifestations
There are several clinical presentations
of orbital retinoblastoma.

a. Primary Orbital Retinoblastoma

Primary orbital retinoblastoma refers


to clinically or radiologically detected
orbital extension of an intraocular
retinoblastoma at the initial clinical
presentation, with or without proptosis Figure 33 B
or a fungating mass (Figure 32). Silent
proptosis without significant orbital and
periocular inflammation in a patient
with manifest intraocular tumor is the
characteristic presentation. Proptosis
with inflammation generally indicates
reactive sterile orbital cellulitis second-
ary to intraocular tumor necrosis.

b. Secondary Orbital Retinoblastoma


Orbital recurrence following uncom-
in an eye with unsuspected intraocular
plicated enucleation for intraocular
retinoblastoma should be considered as
retinoblastoma is named secondary
accidental orbital retinoblastoma and
orbital retinoblastoma (Figure 33). Un-
managed as such (Figure 34).
explained displacement, bulge or extru-
sion of a previously well-fitting conform- d. Overt Orbital Retinoblastoma
Figure 32. Primary orbital retinoblastoma er or a prosthesis is an ominous finding
manifesting with proptosis (32 A). Computed
tomography scan shows massive orbital tumor
suggestive of orbital recurrence. Previously unrecognized extrascleral or
(32 B). optic nerve extension discovered dur-
Figure 33. Secondary orbital retinoblastoma c. Accidental Orbital Retinoblastoma ing enucleation qualifies as overt orbit-
following enucleation, manifesting with extru- al retinoblastoma. Pale pink to cherry
sion of the prosthetic eye (33 A). CT can shows Inadvertent perforation, fine-needle red episcleral nodule, generally in jux-
an orbital tumor (33 B). aspiration biopsy or intraocular surgery tapapillary location or at the site of
22
Orbital Retinoblastoma

aspiration biopsy if involved, imaging


Figure 34 A of the orbit and brain, chest x-ray, ul-
trasonography of the abdomen, bone
marrow biopsy and cerebrospinal fluid
cytology are necessary to stage the
disease. Technetium-99 bone scan and
positron-emission tomography coupled
with computed tomography may be
useful modalities of the early detec-
tion of subclinical systemic metastases.
Orbital biopsy is rarely required, and
should be considered specifically when
a child presents with an orbital mass
Figure 34 B following enucleation or evisceration
where the primary histopathology is
unavailable.
Management of Orbital
Retinoblastoma
Primary orbital retinoblastoma has
been managed in the past with orbital
exenteration, chemotherapy or exter-
nal beam radiotherapy in isolation or
in sequential combination with vari-
able results. 55-60 It is well known that
vortex veins, may be visualized during local treatments have a limited effect
enucleation. An enlarged and inelastic on the course of this advanced disease.
optic nerve with or without nodular op- Orbital exenteration alone is unlikely
tic nerve sheath are clinical indicators to achieve complete surgical clearance
of optic nerve extension of retinoblas- and prevent secondary relapses; exter-
toma that should be recognized during nal beam radiotherapy does not gener-
enucleation. ally prevent systemic metastasis; and
chemotherapy alone may not eradicate
e. Microscopic Orbital Retinoblastoma residual orbital disease. 55-60 Therefore,
a combination therapy is considered to
In several instances, orbital extension be more effective. We have developed
of retinoblastoma may not be clinically a treatment protocol comprising of ini-
evident and may only be microscopic. tial three drug (Vincristine, Etoposide,
Detection of full-thickness scleral infil- Carboplatin) high dose chemotherapy
tration, extrascleral extension and inva- (3-6 cycles) followed by surgery (enu-
sion of the optic nerve transection on cleation, extended enucleation or or-
histopathologic evaluation of an eye bital exenteration as appropriate), or-
enucleated for intraocular retinoblasto- bital radiotherapy, and additional 12
ma are unequivocal features of orbital cycle standard dose chemotherapy.17
retinoblastoma. Tumor cells in choroi- In 6 carefully selected cases without
dal and scleral emissaria and optic nerve
sheath indicate possible orbital exten- Management of orbital retinoblastoma
sion mandating further serial sections Honavar SG, Reddy VAP, Murthy R, Naik M,
Vemuganti GK
and detailed histopathologic analysis.
Proceedings of the XI International Congress
Diagnostic Evaluation of Ocular Oncology, Hyderabad, India, 2004
PURPOSE: Retinoblastoma with orbital exten-
A thorough clinical evaluation paying sion carries poor life prognosis. We aimed to
evaluate the results of a multimodality treat-
attention to the subtle signs of orbital ment protocol in such cases. METHODS: Pro-
retinoblastoma is necessary. Magnetic spective clinical trial of an initial 3 cycle high
resonance imaging preferably, or com- dose chemotherapy followed sequentially by
enucleation, orbital radiotherapy and extended
puted tomography scan of the orbit 12 cycle chemotherapy in 6 consecutive pa-
and brain in axial and coronal orien- tients having retinoblastoma with orbital ex-
tation with 2-mm slice thickness helps tension. RESULTS: All the patients were free
if local recurrence or systemic metastasis at Figure 34. A child with retinoblastoma misdi-
confirm the presence of orbital retino- a mean follow-up of 12 months and achieved
an acceptable cosmesis. Transient bone mar- agnosed as traumatic hyphema in the left eye
blastoma and determine its extent. Sys- and treated with hyphema drainage without
row suppression was a common complication
temic evaluation, including a detailed (75%). CONCLUSION: Early results of a mul- a baseline ultrasonograpy evaluation presents
physical examination, palpation of the timodal treatment protocol for retinoblastoma after 1 year with extraocular extension (34 A)
regional lymph nodes and fine needle with orbital extension are encouraging. and regional lymph node metastasis (34 B)
23
Orbital Retinoblastoma

sis at a mean follow-up of 36 months


Figure 35 A (range 12-102 months) and achieved
acceptable cosmetic outcome (Figure
35). 17
Our treatment protocol outlined for pri-
mary orbital retinoblastoma is currently
under evaluation for secondary orbital
retinoblastoma and early results have
been very encouraging. Surgical inter-
vention in such cases may be limited to
excision of the residual orbital mass or
an orbital exenteration depending on
the extent of the residual tumor after
Figure 35 B the initial 3-6 cycles of high-dose chem-
otherapy.
All eyes that have undergone an in-
traocular surgery for unsuspected retin-
oblastoma should be promptly enucle-
ated. 16 Conjunctiva overlying the ports
with about 4 mm clear margins should
be included en-bloc with enucleation.
Random orbital biopsy may be also ob-
tained, but there is no data to support
its utility. If immediate enucleation is
not logistically possible, then the vit-
Figure 35 C rectomy ports or the surgical incision
should be subjected to triple-freeze-
Intraocular surgery after treatment of retino-
blastoma
Honavar SG, Shields CL, Shields JA, Demirci
H, Naduvilath TJ
Arch Ophthalmol. 2001;119:1613-21
OBJECTIVES: To analyze the results of in-
traocular surgery in patients treated for
retinoblastoma and to assess the ocular and
systemic outcomes. DESIGN: Retrospective
noncomparative case series. PATIENTS: Forty-
five consecutive patients who underwent an
introcular surgery after treatment for retino-
Figure 35 D blastoma. MAIN OUTCOME MEASURES:
(1) Recurrence of retinoblastoma, (2) need
for enucleation, and (3) systemic metastasis.
Overall outcome was defined as favorable in
the absence of any of these measures and
unfavorable in the presence of 1 or more. RE-
SULTS: Thirty-four patients (76%) underwent
a single procedure of cataract surgery, a scle-
ral buckling procedure, or pars plana vitrec-
tomy and 11 (24%) underwent a combination
of 2 or more surgical procedures. In all, 16 pa-
tients (36%) achieved final visual acuity better
than 20/200. Unfavorable outcomes included
recurrence of retinoblastoma in 14 patients
(31%), enucleation in 16 (36%), and systemic
metastasis in 3 (7%). Five patients (20%) who
underwent cataract surgery, 5 (63%) who un-
derwent a scleral buckling procedure, and 9
intracranial extension and systemic me- (75%) who underwent pars plana vitrectomy
tastasis, there was dramatic resolution manifested an unfavorable outcome. The me-
dian interval between completion of treatment
of orbital involvement. All the involved for retinoblastoma and intraocular surgery
eyes became phthisical after 3 cycles of was 26 months in patients with a favorable
outcome vs 6 months in those with an unfa-
high dose chemotherapy. No clinically vorable outcome. CONCLUSIONS: Intraocular
Figure 35. A child with primary orbital retino- apparent residual orbital tumor was surgery after treatment for retinoblastoma
blastoma (35 A), showing massive orbital tumor found during enucleation. All patients may be justified in certain exceptional clinical
situations. Cataract surgery is safe and effec-
on computed tomography scan (35 B). Follow- completed the treatment protocol of tive in most cases. However, the need for a
ing 12 cycles of high-dose chemotherapy, ex- orbital external beam radiotherapy, scleral buckling procedure and pars plana vit-
tended enucleation and orbital external beam rectomy may be associated with a higher risk
radiotherapy (35 C). The child is alive and well
and additional 12 cycle standard chem- for recurrence of retinoblastoma, enucleation,
and wears a custom ocular prosthesis 3 years otherapy. All the patients were free of and systemic metastasis, and a cautious ap-
following completion of treatment (35 D). local recurrence or systemic metasta- proach is warranted.

24
Metastatic Retinoblastoma

Vitrectomy in eyes with unsuspected retino- cific analysis of the sites of sclerotomy
blastoma ports or the cataract wound for tumor
Shields CL, Honavar S, Shields JA, Demirci H, cells. There are specific guidelines for
Meadows AT planned intraocular surgery in patients
Ophthalmology 2000;107:2250-5 with treated retinoblastoma. 15
OBJECTIVE: To analyze patient management
and prognosis after vitrectomy in eyes with If an extraocular extension is macro-
unsuspected retinoblastoma. DESIGN: Ret- scopically visualized during enucleation,
rospective, noncomparative case series. PAR-
TICIPANTS: Eleven consecutive patients who special precaution is taken to excise it
had undergone vitrectomy on an eye with un- completely along with the eyeball, pref-
suspected retinoblastoma. MAIN OUTCOME erably along with the layer of Tenon’s
MEASURES: The two main outcome meas-
ures were ultimate patient management and capsule intact in the involved area. 11
the development of retinoblastoma metasta-
sis. RESULTS: Of more than 900 consecutive All patients with accidental, overt or
patients with retinoblastoma managed on the microscopic orbital retinoblastoma un-
Ocular Oncology Service at Wills Eye Hospital
in Philadelphia, 11 (1%) had prior vitrectomy dergo baseline systemic evaluation to
in an eye with viable tumor before referral to rule out metastasis. Orbital external
us for suspected retinoblastoma. The main pr-
eoperative diagnoses included vitreous hem- beam radiotherapy (fractionated 45-50
orrhage in seven patients (64%), toxocariasis Gy) and 12 cycles of high dose chemo-
in two patients (18%), toxoplasmosis in one therapy is recommended. 16
patient (9%), and endophthalmitis in one pa-
tient (9%). In no case was retinoblastoma sus-
pected before vitrectomy. The mean patient
age at vitrectomy was 6 years. Retinoblasto-
Metastatic
ma was later suspected during vitrectomy in
two patients (18%), on cytologic examination
Retinoblastoma
of the vitrectomy specimen in eight patients
(73%), and after referral in one patient (9%). Metastatic disease at the time of retino-
The mean interval between vitrectomy and blastoma diagnosis is very rare. There-
referral to us was 23 days. On examination, fore, staging procedures such as bone
the globe was classified as Reese-Ellsworth
group Vb in all 11 patients (100%). Anterior scans, lumbar puncture, and bone mar-
chamber tumor cells were clinically visible in row aspirations at the initial presenta-
four eyes (36%), hyphema in two eyes (18%), tion are generally not recommended.
and iris neovascularization in two eyes (18%).
Retinoblastoma cells were visualized in the vit- The common sites for local spread and
reous in seven eyes (64%) and not visualized metastasis include orbital and regional
in four eyes (36%) that had vitreous blood.
Enucleation was necessary in all 11 patients lymph node extension, central nerv-
(100%). Adjuvant treatment was delivered in ous system metastasis, and systemic
10 patients (91%), using orbital radiotherapy metastasis to bone and bone marrow.
in nine patients (82%) and chemotherapy in
nine patients (82%). Histopathologic evidence Metastasis in retinoblastoma usually
of retinoblastoma invasion was documented occurs within one year of diagnosis of
in the episclera (two eyes; 18%), anterior the retinoblastoma. If there is no meta-
chamber (seven eyes; 64%), iris (five eyes;
45%), ciliary body (five eyes; 45%), choroid static disease within 5 years of retino-
(three eyes; 27%), and optic nerve (four eyes; blastoma diagnosis, the child is usually
36%; prelaminar, two eyes; postlaminar, two considered cured.
eyes). The vitrectomy ports, Tenon’s fascia,
cut end of the optic nerve, and orbit were free
of tumor. Of the 10 patients who received pro- Metastatic retinoblastoma is reported
phylactic chemotherapy, radiotherapy, or both to develop in fewer than 10% of pa-
in addition to enucleation for prevention of
retinoblastoma metastasis, none (0%) experi- tients in advanced countries. However,
enced metastasis or orbital recurrence during it is a major contributor to retinoblas-
the mean follow-up of 7 years (range, 0.2-24 toma related mortality in developing
years) from the time of retinoblastoma diag-
nosis. However, one patient was referred to nations. Until recently, the prognosis
us after the development of metastatic retino- with metastatic retinoblastoma was
blastoma, and despite aggressive chemother- poor. Conventional dose chemothera-
apy and radiotherapy after enucleation, died
24 months later. CONCLUSIONS: Retinoblas- py using vincristine, doxorubicin, cyclo-
toma may present with atypical features such phosphamide, cisplatin, and etoposide
as vitreous hemorrhage or signs of vitreous combined with radiation therapy has
inflammation, particularly in older children. Vit-
rectomy should be avoided in these cases un- yielded only a few survivors. Dismal
til the possibility of underlying retinoblastoma results with conventional therapy has
is excluded. If vitrectomy is performed in an
eye with unsuspected retinoblastoma, enucle- prompted the use of high dose chemo-
ation combined with adjuvant chemotherapy, therapy with hematopoietic stem cell
radiotherapy, or both without delay is advised rescue. Twenty-five patients with extra
to prevent systemic tumor dissemination.
ocular disease or invasion of the cut end
of optic nerve received high-dose chem-
thaw cryotherapy and enucleation otherapy including carboplatin, etopo-
should be performed at the earliest side, and cyclophosphamide followed
possible convenience. Histopathologic by autologous hematopoietic stem cell
evaluation of such eyes may include spe- rescue. The three year disease-free sur-
25
Conclusion

vival was 67%. 60,61 All except one pa- gene in Indian patients with retinoblas-
tient with central nervous system dis- toma. Ophthalmic Genet. 2002;23:121-
ease died. The main side effects were 8.
hematological, mucositis, diarrhoea,
ototoxicity, and cardiac toxicity. Over- 6. Kiran VS, Kannabiran C, Chakravar-
all the response rate suggested that thi K, Vemuganti GK, Honavar SG. Mu-
the treatment regimen was promising tational screening of the RB1 gene in
in patients with bone or bone marrow Indian patients with retinoblastoma re-
involvement, but not in patients with veals eight novel and several recurrent
central nervous system disease. mutations. Hum Mutat. 2003;22:339.

7. Shields CL, Honavar SG, Shields JA,


Conclusion Demirci H, Meadows AT, Naduvilath
TJ. Factors predictive of recurrence of
There has been a dramatic change in retinal tumors, vitreous seeds, and sub-
the overall management of retinoblast- retinal seeds following chemoreduction
oma in the last decade. Specific genetic for retinoblastoma. Arch Ophthalmol.
protocols have been able to make pre- 2002;120:460-4.
natal diagnosis of retinoblastoma. Early
diagnosis and advancements in focal 8. Shields CL, Honavar SG, Meadows
therapy have resulted in improved eye AT, Shields JA, Demirci H, Singh A,
and vision salvage. Chemoreduction has Friedman DL, Naduvilath TJ. Chemore-
become the standard of care for the duction plus focal therapy for retino-
management of moderately advanced blastoma: factors predictive of need for
intraocular retinoblastoma. Periocular treatment with external beam radio-
chemotherapy is now an additional use- therapy or enucleation. Am J Ophthal-
ful tool in salvaging eyes with vitreous mol. 2002;133:657-64.
seeds. Enucleation continues to be the
preferred primary treatment approach 9. Shields CL, Honavar SG, Meadows
in unilateral advanced retinoblastoma. AT, Shields JA, Demirci H, Naduvilath
Post-enucelation protocol, including TJ. Chemoreduction for unilateral
identification of histopathologic high- retinoblastoma. Arch Ophthalmol.
risk characteristics and provision of 2002;120:1653-8.
adjuvant therapy has resulted in sub-
stantial reduction in the incidence of 10. Murthy R, Honavar SG, Naik MN,
systemic metastasis. The vexing orbital Reddy VA. Retinoblastoma. In: Dutta
retinoblastoma now seems to have a LC, ed. Modern Ophthalmology . New
cure finally with the aggressive multi- Delhi, India, Jaypee Brothers; 2004:849-
modal approach. Future holds promise 859.
for further advancement in focal thera- 11. Honavar SG, Singh AD. Manage-
py and targeted drug delivery. ment of advanced retinoblastoma.
Ophthalmol Clin North Am. 2005;18:65-
References 73.

1. Bishop JO, Madsen EC: Retinoblas- 12. Vemuganti G, Honavar SG, John R.
toma. Review of current status. Surv Clinicopathological profile of retino-
Ophthalmol 19: 342-366, 1975. blastoma in Asian Indians. Invest Oph-
thalmol Vis Sci 2000; 41(S):790.
2. Shields JA, Shields CL. Intraocular
tumors – A text and Atlas. Philadel- 13. Honavar SG, Singh AD, Shields CL,
phia, PA, USA, WB Saunders Company, Meadows A, Shields JA. Does adjuvant
1992. chemotherapy prevent metastasis in
high-risk retinoblastoma? Investigative
3. Albert DM: Historic review of retino- Ophthalmology and Visual Science,
blastoma. Ophthalmology 94; 654-662, 2000, 41(S):790
1987.
14. Honavar SG, Rajeev B. Needle Tract
4. Jackson E: Report of the committee Tumor Cell Seeding Following Fine Nee-
to investigate and revise the classifica- dle Aspiration Biopsy for Retinoblasto-
tion of certain retinal conditions. Trans ma. Investigative Ophthalmology and
Am Ophthalmol Soc 24:38-39, 1926 Visual Science 1998; 39: S 658.

5. Ata-ur-Rasheed M, Vemuganti G, Ho- 15. Honavar SG, Shields CL, Shields JA,
navar S, Ahmed N, Hasnain S, Kanna- Demirci H, Naduvilath TJ. Intraocular
biran C. Mutational analysis of the RB1 surgery after treatment of retinoblast-
26
References

oma. Arch Ophthalmol. 2001;119:1613- eye technique. Br J Ophthalmol. 1995;


21. 79: 112-117.

16. Shields CL, Honavar S, Shields JA, 30. Abramson DH, Frank CM. Second
Demirci H, Meadows AT. Vitrectomy in nonocular tumors in survivors of bilat-
eyes with unsuspected retinoblastoma. eral retinoblastoma; a possible age ef-
Ophthalmology. 2000;107:2250-5. fect on radiation-related risk. Ophthal-
mology. 1998; 105: 573-580.
17. Honavar SG, Reddy VAP, Murthy R,
Naik M, Vemuganti GK. Management 31. Shields JA, Shields CL, Sivalingam V.
of orbital retinoblastoma. XI Interna- Decreasing frequency of enucleation
tional Congress of Ocular Oncology. in patients with retinoblastoma. Am J
Hyderabad, India, 2004. pp. 51. Ophthalmol 1989; 108:185-8.

18. Stallard HB: The conservative treat- 32. Ferris FL, 3rd, Chew EY. A new era
ment of retinoblastoma. Trans Am Op- for the treatment of retinoblastoma.
ththalmol Soc UK 1962; 82:473-534. Arch Ophthalmol 1996; 114:1412.

19. Murphree AL, Benedict WF: Retino- 33. Kingston JE, Hungerford JL, Ma-
blastoma: Clues to human oncogenesis. dreperla SA, Plowman PN. Results of
Science 1984: 223:1028-1033. combined chemotherapy and radio-
therapy for advanced intraocular retin-
20. Knudson AG: Mutation and cancer: oblastoma. Archives of Ophthalmology
Statistical study of retinoblastoma. Proc 1996; 114:1339-43.
Natl Acad Sci, USA 1971:68: 820-823.
34. Gallie BL, Budning A, DeBoer G,
21. Abramson DH, Frank CM, Susman Thiessen JJ, Koren G, Verjee Z, et al.
M, Whalen MP, Dunkel IJ, Boyd NW Chemotherapy with focal therapy can
3rd. Presenting signs of retinoblastoma. cure intraocular retinoblastoma with-
J Pediatr. 1998; 132:505-8. out radiotherapy. Arch Ophthalmol
1996; 114:1321-8.
22. Ellsworth RM. The practical man-
agement of retinoblastoma. Trans Am 35. Murphree AL, Villablanca JG,
Ophthalmol Soc 1969: 67: 462-534. Deegan WF, 3rd, Sato JK, Malogolowkin
M, Fisher A, et al. Chemotherapy plus
23. Linn Murphree A. Intraocular retin- local treatment in the management of
oblastoma: the case for a new group intraocular retinoblastoma. Arch Oph-
classification. Ophthalmol Clin North thalmol 1996; 114:1348-56.
Am. 2005;18:41-53.
36. Shields CL, De Potter P, Himelstein
24. Shields CL, Shields JA. Basic under- BP, Shields JA, Meadows AT, Maris JM.
standing of current classification and Chemoreduction in the initial manage-
management of retinoblastoma. Curr ment of intraocular retinoblastoma.
Opin Ophthalmol.2006;17:228-34. Arch Ophthalmol 1996; 114:1330-8.
25. Chantada G, Doz F, Antoneli CB, 37. Honavar SG, Shome D, Reddy VAP.
et al A proposal for an international Periocular carboplatin in the manage-
retinoblastoma staging system Pediatr ment of advanced intraocular retin-
Blood Cancer 2006:47;801-805 oblastoma. Proceedings of the XII
Intrernational Congress of Ocular On-
26. Shields CL, Santos MC, Diniz W et
cology, Vancouver, Canada, 2005.
al. Thermotherapy for retinoblastoma.
Arch Ophthalmol 1999; 117: 885-893. 38. Abramson DH, Niksarli K, Ellsworth
27. Shields CL, Shields JA, Cater J et RM, Servodidio CA. Changing trends in
al. Plaque radiotherapy for retinoblas- the management of retinoblastoma:
toma, long term tumor control and 1951-1965 vs 1966-1980. J Pediatr Oph-
treatment complications in 208 tumors. thalmol Strabismus 1994; 31:32-7.
Ophthalmology 2001;108: 2116-2121.
39. Singh AD, Shields CL, Shields JA.
28. Ellsworth RM. Retinoblastoma. Prognostic factors in retinoblastoma. J
Modern problems in ophthalmology. Pediatr Ophthalmol Strab 2000; 37:134-
1977; 96:1826-1830. 41.

29. Hungerford JL, Toma NMG, Plow- 40. Ajaiyeoba IA, Akang EE, Campbell
man PN, Kingston JE. External beam ra- OB, Olurin IO, Aghadiuno PU. Retino-
diotherapy for retinoblastoma: I, whole blastomas in Ibadan: treatment and
27
References

prognosis. West African Journal of from the Mexican Retinoblastoma


Medicine 1993; 12:223-7. Group. Br J Ophthalmol 2004;88:1074-
1077.
41. Kingston JE, Hungerford JL, Plow-
man PN. Chemotherapy in metastatic 53. Kao LY, Su WW, Lin YW. Retino-
retinoblastoma. Ophthalmic Paediatr blastoma in Taiwan: survival and clinical
Genet 1987; 8:69-72. characteristics 1978-2000. Jpn J Oph-
thalmol 2002;46:577-580.
42. White L. Chemotherapy for retino-
blastoma: where do we go from here? 54. Badhu B, Sah SP, Thakur SK, et al.
Ophthalmic Pediatr and Genet 1991; Clinical presentation of retinoblastoma
12:115-30. in Eastern Nepal. Clin Experiment Oph-
thalmol 2005;33:386-389.
43. Kopelman JE, McLean IW, Rosen-
berg SH. Multivariate analysis of risk 55. Pratt CB, Crom DB, Howarth C. The
factors for metastasis in retinoblastoma use of chemotherapy in extraocular
treated by enucleation. Ophthalmology retinoblastoma. Med and Pediatr Oncol
1987; 94:371-7. 1985;13:330-333.

44. Magramm I, Abramson DH, 56. Kiratli H, Bilgic S, Ozerdem U. Man-


Ellsworth RM. Optic nerve involvement agement of massive orbital involvement
in retinoblastoma. Ophthalmology of intraocular retinoblastoma. Ophthal-
1989; 96:217-22. mology 1998;105:322-326.

45. Messmer EP, Heinrich T, Hopping 57. Goble RR, McKenzie J, Kingston JE,
W, de Sutter E, Havers W, Sauerwein W. et al. Orbital recurrence of retinoblas-
Risk factors for metastases in patients toma successfully treated by combined
with retinoblastoma. Ophthalmology therapy. Br J Ophthalmol 1990;74:97-
1991; 98:136-41. 98.

46. Shields CL, Shields JA, Baez KA, et a. 58. Doz F, Khelfaoui F, Mosseri V, et
Choroidal invasion of retinoblastoma: al. The role of chemotherapy in orbital
Metastatic potential and clinical risk involvement of retinoblastoma. The ex-
factors. Br J Ophthalmol 1993; 77:544- perience of a single institution with 33
8. patients. Cancer 1994;74:722-732.

47. Shields CL, Shields JA, Baez K, Cater 59. Antoneli CB, Steinhorst F, de Cas-
JR, De Potter P. Optic nerve invasion of sia Braga Ribeiro K, et al. Extraocular
retinoblastoma. Metastatic potential retinoblastoma: a 13-year experience.
and clinical risk factors. Cancer 1994; Cancer 2003;98:1292-1298.
73:692-8.
60. Dunkel IJ, Aledo A, Kernan NA,
48. Chantada GL, de Silva MTG, Fand- Kushner B, Bayer L, Gollamudi SV, et
ino A, et a. Retinoblastoma with low al. Successful treatment of metastatic
risk for extraocular relapse. Ophthalmic retinoblastoma. Cancer 2000; 89:2117-
Genet 1999; 20:133-40. 21.

49. Khelfaoui F, Validire P, Auperin A, 61. Rodriguez-Galindo C, Wilson MW,


Quintana E, Michon J, Pacquement H, Haik BG, Lipson MJ, Cain A, Merchant
et al. Histopathologic risk factors in TE, et al. Treatment of metastatic
retinoblastoma: a retrospective study retinoblastoma. Ophthalmology 2003;
of 172 patients treated in a single insti- 110:1237-40.
tution. Cancer 1996; 77:1206-13.

50. Honavar SG, Singh AD, Shields CL,


Demirci H, Smith AF, Shields JA. Post-
enucleation prophylactic chemotherapy
in high- risk retinoblastoma. Arch Oph-
thalmol 2002; 120:923-31.

51. Ellsworth RM. Orbital retinoblas-


toma. Trans Am Ophthalmol Soc
1974;72:79-88.

52. Leal-Leal C, Flores-Rojo M, Medina-


Sanson A, et al. A multicentre report
28
Sponsors

An educational initiative by

29
LVPEI

LV PRASAD EYE The institute’s coordinated and inter-


linked functions of patient care, train-
INSTITUTE ing, research, rehabilitation, commu-
nity eye care and product development
serve a spectrum of population in need
of diverse eye care services and a large
network of practitioners and students.
A web of national and international
partnerships provides a solid frame-
work of support.

Underpinning all the efforts of LVPEI


are three dominant themes - efficiency,
equity and excellence. The focus has
been on extending equitable and effi-
cient eye care to the underprivileged.
The Institute’s charter aims at providing
50% of its surgical services free of cost
to the economically disadvantaged.

LVPEI is a World Health Organization


Collaborating Center for Prevention
of Blindness, engaged in creating eye
health models for underserved areas
of the developing world. In collabora-
tion with the World Health Organiza-
tion and the International Agency for
the Prevention of Blindness, LVPEI has
designed and implemented many inno-
vative community eye health programs.

LV Prasad Eye Institute, located in Hy- The cornerstone of progress in medical


derabad, India, is a world-class eye hos- science is research and LVPEI collabo-
pital and research, training and rehabil- rates with premier institutions globally
itation centre. Since its establishment in in this area. LVPEI research program
1987, LVPEI has grown into a centre of takes pride in putting cutting edge
excellence in the field of eye health. technology to clinical application.

LV Prasad Eye Institute


Kallam Anji Reddy Campus
LV Prasad Marg, Banjara Hills
Hyderabad 500034, India
Telephone +91-40-30612345
Fax +91-40-23548271
e-mail care@lvpei.org
http:\\www.lvpei.org
30
SightSavers

a result of which India is the first


ever country globally to have a na-
tional programme for the control
of blindness as early as 1976.
Every 5 seconds one person in our world • Promoting technology by revo-
goes blind and a child goes blind every lutionizing the affordability of
minute. 75% of all blindness around cataract surgery in the developing
the world could be easily avoided. 90% world by contributing to the estab-
of children who are blind don’t go to lishment of a unit in India in the 90s
school. to produce high-quality, low-cost
intra-ocular lens, sutures and initi-
Sightsavers International is a leading
ating the production unit for cor-
global development agency working in
nea preservation for eye donation
33 countries to combat blindness, re-
and transplantation.
store sight and work for the rights and
needs of those with irreversible visual • Pioneering of models such as in-
impairment. India has one-fourth of tegrated education for visually
the world’s reported 161 million blind impaired children and community
and severely visually impaired people. based rehabilitation for inclusion
Sightsavers in India works with 90 local of blind and low vision persons into
partners across 18 states in alignment the mainstream, and comprehen-
with the Government of India’s nation- sive eye services which aims at pro-
al plan and global movements such as viding holistic eye services within
VISION 2020: The Right to Sight and communities.
the Global Campaign for Education to
address this mammoth issue. Despite the enormous strides made by
the achievements in India in the area
UK-based Sightsavers International’s of blindness control and inclusion of
work in India, which started in 1966, has disabled people, a lot more needs to
supported the treatment of millions of be done. The stark fact remains that to-
persons with eye disorders and brought day more people than ever are threat-
eye services to some of the least served ened with blindness. The World Health
areas of the country. In addition, many Organisation has estimated that the
thousands of irreversibly blind people world’s blindness burden could double
have received rehabilitation and educa- by the year 2020 and that an additional
tional support to enable them to lead 100 million people will go blind unless
lives of independence and dignity. urgent action is taken.
Path-breaking milestones in Sightsav- Sightsavers is committed to eradicating
ers’ challenging but immensely reward- avoidable blindness in India and to pro-
ing journey in India over the last four moting inclusion. Future plans include
decades include: further strengthening its work in the
areas of awareness raising, tackling
• The establishment of the National serious eye conditions such as diabetic
Programme for the Control of Blind- retinopathy, glaucoma, refractive error
ness in India: Sightsavers founder and blinding disorders among children,
Sir John Wilson was one of the few and advocating for the education of
who strongly advocated with the irreversibly visually impaired children,
Government of India to include and the rights and needs of blind and
blindness in the national plan, as low vision persons.

Information
Ranjish Kattady
Regional Information &
Liaison Officer
Sightsavers International
India Region
email rkattady@sightsavers.org
www.sightsavers.org
31
Ocular Oncology Service
LV Prasad Eye Institute
Kallam Anji Reddy Campus
LV Prasad Marg, Banjara Hills
Hyderabad 500034, India
Telephone +91-40-30612632
Fax +91-40-23548271
e-mail oncology@lvpei.org

Das könnte Ihnen auch gefallen