Está en la página 1de 20

Author's Accepted Manuscript

Retinoblastoma: An update
Kushal S. Delhiwala MD, V.P. Indu MD,
Kaustubh Mulay MD, Vikas Khetan MD

www.elsevier.com/locate/serndb

PII: S0740-2570(15)00142-2
DOI: http://dx.doi.org/10.1053/j.semdp.2015.10.007
Reference: YSDIA50433

To appear in: Seminars in Diagnostic Pathology

Cite this article as: Kushal S. Delhiwala MD, V.P. Indu MD, Kaustubh
Mulay MD, Vikas Khetan MD, Retinoblastoma: An update, Seminars in
Diagnostic Pathology, http://dx.doi.org/10.1053/j.semdp.2015.10.007

This is a PDF file of an unedited manuscript that has been accepted for
publication. As a service to our customers we are providing this early
version of the manuscript. The manuscript will undergo copyediting,
typesetting, and review of the resulting galley proof before it is published in
its final citable form. Please note that during the production process errors
may be discovered which could affect the content, and all legal disclaimers
that apply to the journal pertain.
Delhiwala et al. Page 1

RETINOBLASTOMA: AN UPDATE
Kushal S. Delhiwala, MD1

Indu VP, MD1

Kaustubh Mulay, MD2

Vikas Khetan, MD1,3

1 – Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India.

2 – National Reporting Centre for Ophthalmic Pathology, Centre For Sight, Hyderabad

3 - Ocular Oncology Services and Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya,

Chennai, Tamil Nadu, India

Corresponding Author

Vikas Khetan, MD, Ocular Oncology Services and ShriBhagwanMahavirVitreoretinal Services,

SankaraNethralaya, Chennai - 600 006, Tamil Nadu, India (E-mail: drkhetan@yahoo.com)

Abstract

Retinoblastoma is the most common ocular malignancy in children, and is initiated by

mutation of the RB1 gene. The tumor may be unilateral or bilateral and can be

inherited. Overall survival, eye salvage, and preservation of vision are largely

dependent on the stage of disease at presentation. Despite a recently-enhanced

understanding of the etiology of retinoblastoma, the mortality associated with it remains

high worldwide. This may relate to a continuing lack of awareness of the lesion by

laypersons, and unavailability of modern treatment facilities. Adverse outcomes are


Delhiwala et al. Page 2

also caused by the occurrence of secondary malignancies after treatment of

retinoblastoma in childhood. Early diagnosis, multidisciplinary treatment, and genetic

counselling are all priorities in the management of this tumor.

Key Words: Retinoblastoma; childhood neoplasms; ocular tumors; heritable neoplasms

Retinoblastoma (RB), which can be considered as a prototype of inherited

cancers, is the second most common intraocular malignancy after uveal melanoma. It is

the principal tumor seen in the eyes of children, in whom it represents 4% of all pediatric

malignancies.1 Despite being an aggressive disease, retinoblastoma has been

associated with good survival rates because of early detection and improving treatment

methods.2,3 Identification of genetic mutations that are associated with RB, recognition

of high-risk histopathologic factors, and aggressive multidisciplinary therapy have

contributed to better survival, eye salvage, and potential preservation of vision.

History, Epidemiology, & Classification

Retinoblastoma was first described by Pawius in 1597.4 Wardrop (1809) named it

“fungus hematoides,” and Virchow considered RB to be a glioma of the retina in 1864.4

The presence of rosettes in the tumor at a histological level prompted Flexner, in 1891,

and then Wintersteiner, in 1897, to designate RB as a neuroepithelioma.4 In 1926, the

American Ophthalmological Society agreed upon the term ‘retinoblastoma’ after a

consensus was reached that the tumor likely originated from the proliferation of

retinoblasts.5
Delhiwala et al. Page 3

The incidence of retinoblastoma has been constant worldwide, at 1 case per

15,000-20,000 live births; that figure corresponds to 9000 new cases each year.6,7 The

populations of Asia and Africa account for the majority of these.6 RB in those continents

is also associated with high mortality rates, which are likely attributable to delayed

detection and inadequate treatment facilities. No significant racial or gender

predilection has been reported; bilaterality is seen in 25-35% cases.8 Sporadic

examples are unilateral, whereas familial tumors may either be unilateral or bilateral.9

The average age at diagnosis is 18 months-- 24 months for unilateral disease and 12

months for bilateral.10

Genetics

Of all newly diagnosed cases, 6% are inherited and the rest are sporadic.8,9

Retinoblastoma was the first disease in which a genetic basis for cancer was

identified—biallelic deactivation of the RB1 gene.6 In 1971, Knudson proposed the “2-

hit hypothesis” for RB development, and suggested that 2 mutational events were

necessary to that process.10 In familial cases, the initial event is a germline mutation,

found in all cells of the affected offspring; the second “hit” occurs during the

development of somatic retinal cells. By contrast, both mutations are somatic in nature

in sporadic tumors. This paradigm may also explain the high risk of secondary non-

ocular malignancies that is seen in patients with familial bilateral retinoblastoma.10,11

The RB1 gene has been mapped to the long arm of chromosome 13 (13q14).12

It is a negative cell-cycle regulator, maintaining a balance between cell proliferation and

development.13 RB-phosphoprotein, the gene product. acts to interrupt the transition

between the G1- and S-phases of the cell cycle. Its active form, pRB, is
Delhiwala et al. Page 4

hypophosphorylated and seen in resting cells; on the other hand, proliferating cells

contain hyper-phosphorylated pRB (ppRB). Phosphorylation is inhibited by cyclin

dependent kinases (CDK). pRB blocks cell division by binding to transcription factor

E2F and preventing the transcription of cell cycle-related genes that are active in the

G1-S phase transition; ppRB has an opposing activity.

The loss of RB1 function initiates the formation of retinoblastoma but is

inadequate for complete development of that tumor; other genetic and epigenetic events

are necessary to that process.14 There is increasing evidence for a role of oncogenes

such as MDM4, KIF14, MYCN, DEK, and E2F3, and the tumor suppressor gene CDH11

in retinoblastomagenesis.12 Comparative genomic hybridization (CGH) has identified

gains in chromosomes 1q32, 2p24, 6p22, and losses in 13q and 16q22-24 in

retinoblastoma.12,15 Dysregulated genes in the insulin and JAK/STAT pathways have

likewise been identified.16 Epigenetic events in retinoblastoma include hypermethylation

of RB1 promoters; genes in the RAS association domain; MGMT; p16INK4; MSH6;

CD44; PAX5; GATA5; TP53; VHL; and GSTP1.12 Micro RNAs (miRNAs) may also be

differentially expressed in retinoblastoma.12,17

Matrix metalloproteinases (MMPs)-- which play a role in tumor invasion,

degradation of stroma, and angiogenesis-- are modified in retinoblastomas. Although

MMP-2 is observed in differentiated tumors, the expression of TIMP-1 and TIMP-2

predominates in primitive blastomatous neoplasms.18 Epithelial cell adhesion molecule

(EpCAM) appears to show greater expression in invasive tumors as compared with non-

invasive examples.19
Delhiwala et al. Page 5

Clinical Manifestations

The clinical features of retinoblastoma largely depend on the stage of the disease

at presentation. Leucocoria is the commonest initial finding (seen in 60% of cases)

followed by strabismus (20%).20 Others include a painful blind eye, diminished vision,

orbital cellulitis, unilateral mydriasis, heterochromia iridis, and hyphema.21 The tumor

growth pattern may be endophytic (growth into the vitreous cavity) (Figure 1), exophytic

(towards the sub-retinal space) or diffusely infiltrative, causing placoid retinal thickness

rather than a discrete mass. Proptosis may be seen in advanced cases with orbital

extension. Atypical presentations include phthisis bulbi, vitreous haemorrhage, and

pseudohypopyon.22

Grouping & Staging

Grouping of RB is a clinical system to prognosticate the likelihood of ocular

salvage; staging is used to estimate the likelihood of survival. The Resse-Ellsworth

classification was introduced prior to the use of indirect ophthalmoscopy; it applies to

patients treated by means other than enucleation, and its components are

complicated.23 The more recently-developed International Classification of Intraocular

Retinoblastoma (ICIOR) scheme correlates grading with current treatment modalities

and is preferred. Two versions of that system are available.24,25 The TNM staging

scheme, developed by the American Joint Committee on Cancer (AJCC) and Union

Internationale Contre le Cancer (UICC), includes clinical and histopathological data

(Table 1).
Delhiwala et al. Page 6

Histopathology of Retinoblastoma

Retinoblastoma is thought to arise from totipotential blasts in the retina.

Microscopically, it is usually characterized by alternating necrotic and viable areas. As

true of other small round-cell tumors, the neoplastic elements in retinoblastoma contain

only scanty cytoplasm and have deeply basophilic nuclei that show molding on one

another (Figure 2). Because retinoblastoma commonly outgrows it blood supply,

perivascular pseudorosettes and zonal necrosis are frequently seen. Flexner-

Wintersteiner and Homer-Wright rosettes (Figures 3 & 4) are present in differentiated

tumors, but glial differentiation is rare.

Pitfalls in Diagnosis

For the accurate detection of invasive features, it is important that both central

and peripheral calottes of the eyeball should be sampled. Peripheral foci of choroidal,

scleral, or extraocular invasion (Figures 5 & 6) may be missed if only a central calotte is

examined. Invasion of tumor cells into the choroid may cause them to be compressed,

resembling lymphocytes. Immunostaining for CD45 will resolve any confusion on that

point. Destruction of optic nerve axons by the tumor also can cause secondary

astrocytic proliferation, resulting in a misdiagnosis of true optic nerve invasion by RB

(Figure 7).

Effects of Irradiation or Chemotherapy

After successful responses to therapy, retinoblastoma may appear as amorphous

debris in a glial network. Relapsed lesions seen after treatment may be retinocytoma-

like histologically,sometimes with internal fleurettes.


Delhiwala et al. Page 7

High Risk Factors (HRFs)

Clinical HRFs

Older age at presentation, hyphema, pseudohypopyon, orbital cellulitis,

staphyloma, elevated intraocular pressure, buphthalmos, and delayed enucleation after

diagnosis are associated with a poor prognosis.26-28 Modification of staging data,

especially regarding extraocular extension, by neoadjuvant chemotherapy may result in

inappropriately non-aggressive subsequent management according to a recent study.27

Histopathologic HRFs

Extraretinal extension (ERE) may still be seen in retinoblastoma cases. In

developing countries, up to 50% of affected eyes show ERE.29,30 This finding includes

spread of tumor to the choroid, iris &ciliary body, anterior chamber, sclera, and optic

nerve. Optic nerve involvement may be parenchymal or leptomeningeal.

The extent of uveal invasion correlates with the risk of extraocular relapse.31

Choroidal invasion may be focal (< 3 mm in any dimension) or massive (> 3 mm in any

dimension). The risk of extraocular disease in children with massive choroidal invasion

is 6%,32 and they have a lower probability of overall event-free survival as compared to

those with focal choroidal invasion.32 Pre-lamina cribrosa optic nerve invasion is not

associated with a significant risk of extraocular relapse. However, retrolaminar optic

nerve invasion is indeed a HRF and it necessitates the use of adjuvant chemotherapy.33

The risk is further increased when retrolaminar optic invasion is present along with

massive choroidal invasion or scleral compromise.34 Also, the concurrent presence of

focal choroidal invasion and pre-laminar optic nerve involvement is associated with a

greater risk of relapse than is either of those factors independently.35 Scleral invasion
Delhiwala et al. Page 8

with neo-vascularization of the iris and neovascular glaucoma is likewise regarded as a

HRF.36

Recent Advances

In the recent past, the expression of several proteins has been studied in

retinoblastoma. High expression of high mobility group Box – 1 protein (HMGB1), polo-

like kinase-1 (PLK1), CDC25A, CDC25B, and bcl2, and the loss of mitochondrial

complex –1 have been linked to high histologic grade and invasiveness in RB.37-41 The

coexpression of bax and p53 is correlated with choroidal invasion.41,42 On the other

hand, a loss of mitochondrial complexes III, IV and V has been associated with better

survival.38

Differential Diagnosis

Clinical differential diagnoses for RB include Coats’ disease, toxocariasis,

persistent hyperplastic primary vitreous (PHPV), retinopathy of prematurity (ROP),

retinal dysplasia, and medulloepithelioma. Coats’ disease is the most frequently

encountered among those possibilities. Microscopically, it is characterized by

telangiectatic retinal vessels and lipid-rich sub-retinal exudates with numerous

macrophages (Figure 5). The histological diagnosis of retinoblastoma is usually

straightforward, especially with knowledge of the clinical findings and imaging studies.

It is appropriate to consider any intraocular, small round-cell tumor in a child under 5-

years of age as a retinoblastoma, unless proven otherwise. The diagnostic separation of

poorly differentiated forms of medulloepithelioma and retinoblastoma is difficult, and no

immunostains can distinguish between them. Moreover, although retinoblastomas are

typically posterior in location and medulloepitheliomas are anterior, the converse of that
Delhiwala et al. Page 9

localization has been reported.43,44 Other histological diagnostic considerations include

rhabdomyosarcoma, leukemia, lymphoma, Ewing sarcoma/primitive neuroectodermal

tumor (PNET), and neuroblastoma. An immunohistochemical panel comprising CD45,

myeloperoxidase, CD34, CD43, CD117, desmin, myogenin, CD99, and FLI1 is useful in

that context.

Secondary Malignancies

Children with retinoblastoma, especially the hereditary form, have a significantly

risk of developing another malignancy later in life.45,46 It increases from 5% in the first

10 years after diagnosis to 44% at 45 years of age.45,46 Although the cumulative

incidence of a secondary malignancy is 35% in children who are irradiated, it is only 6%

in those who are not.46 Osteosarcoma is the commonest secondary tumor in those

children, but other reported lesions include rhabdomyosarcoma, neuroblastoma,

chondrosarcoma, leukemia, sebaceous carcinoma, squamous cell carcinoma, and

malignant melanoma.46 One study also reported a heightened incidence of secondary

acute myelogenous leukemia in patients with RB who were treated with topoisomerase

II inhibitors.47
Delhiwala et al. Page 10

Treatment

The primary goal of treatment in patients with retinoblastoma is the preservation

of life; ocular salvage and preservation of vision are secondary. Management depends

on the age at diagnosis, tumor location, staging, overall patient fitness, and cost-

effectiveness of the therapeutic regimen. A multidisciplinary approach is best, involving

the ophthalmologist, oncologist, radiation physicist, pathologist, and ocularist.

Small equatorial and peripheral tumors up to 4 mm in basal diameter and 2 mm

in thickness are treated with cryotherapy.8,22 Posterior tumors of similar dimensions are

managedwith photocoagulation.8,22 Plaque brachytherapy, using ruthenium 106 and

iodine 125, is reserved for tumors up to16 mm in basal diameter and 8 mm in thickness.

With recent advances in chemotherapy, external radiotherapy is now used infrequently.

Advanced retinoblastomas are treated with enucleation followed by chemotherapy, if

HRFs are present.

Conclusions

Retinoblastoma is an aggressive but curable intraocular tumor, and its overall

management has included dramatic changes in recent years. The pathologist plays a

key role in management by careful assessment for histopathologic HRFs in enucleated

eyes.
Delhiwala et al. Page 11

References

1. Shields CL, Shields JA. Basic understanding of current classification and


management of retinoblastoma. Curr Opin Ophthalmol. 2006;17:228-234.
2. Young JL, Smith MA, Roffers SD, Liff JM, Bunin GR. Retinoblastoma. In: Ries
LA, Smith MA, Gurney JG, Linet M, Tamra T, Young JL, et al., editors. Cancer
Incidence and Survival among Children and Adolescents: United States SEER
Program 1975-1995. Maryland: National Cancer Institute, SEER Program; 2012.
3. Ramasubramanian A, Shields CL, editors. New Delhi, India: Jaypee Brothers
Medical Publishers; 2012. Epidemiology and magnitude of the problem.
Retinoblastoma; pp. 10–5.
4. Albert DM; Historic review of retinoblastoma. Ophthalmology 1987; 94: 654-62.
5. Jackson E; Report of the committee to investigate and revise the classification of
certain retinal conditions. Trans Ophthalmol Soc 1926; 24: 38-9.
6. Dimaras H, Kimani K, Dimba EA, Gronsdahl P, White A, Chan HS, Gallie BL.
Lancet. 2012 Apr 14; 379(9824):1436-46.
7. Kivela T. The epidemiological challenge of the most frequent eye cancer:
retinoblastoma, an issue of birth and death. Br J Ophthalmol 2009; 93:1129–31
8. Shields JA, Shields CL. Intraocular tumors – A text and Atlas. Philadelphia, PA,
USA, WB Saunders Company, 1992.
9. Shields CL, Shields JA. Diagnosis and management of retinoblastoma. Cancer
Control. 2004;11:317-327.
10. Mutation and cancer: statistical study of retinoblastoma. Knudson AG Jr. Proc
Natl Acad Sci U S A. 1971 Apr; 68(4):820-3.
11. A general theory of carcinogenesis. Comings DE. Proc Natl Acad Sci U S A.
1973 Dec; 70(12):3324-8
12. Thériault BL, Dimaras H, Gallie BL, Corson TW. The genomic landscape of
retinoblastoma: a review. Clinical & experimental ophthalmology. 2014;42(1):33-
52.
13. Yun, J., Li, Y., Xu, C.T. and Pan, B.R. (2011) Epidemiol-ogy and Rb1 gene of
retinoblastoma. International Jour-nal of Ophthalmology, 4, 103-109.
14. Dimaras H, Khetan V, Halliday W, Orlic M, Prigoda NL, Piovesan B, Marrano P,
Corson TW, Eagle RC Jr, Squire JA, Gallie BL. Loss of RB1 induces non-
proliferative retinoma: increasing genomic instability correlates with progression
to retinoblastoma. Hum Mol Genet. 2008 May 15; 17:1363-72.
15. Corson TW, Gallie BL. One hit, two hits, three hits, more? Genomic changes in
the development of retinoblastoma. Genes Chromosomes Cancer. 2007; 46:617-
34.
16. Chakraborty S, Khare S, Dorairaj SK, Prabhakaran VC, Prakash DR, Kumar A.
Identification of genes associated with tumorigenesis of retinoblastoma by
microarray analysis. Genomics. 2007 Sep; 90:344-53.
17. Martin J, Bryar P, Mets M, et al. Differentially expressed miRNAs in
retinoblastoma. Gene. 2013;512:294-299.
Delhiwala et al. Page 12

18. Adithi M, Nalini V, Kandalam M, Krishnakumar S. Expression of matrix


metalloproteinases and their inhibitors in retinoblastoma. J Pediatr Hematol
Oncol. 2007; 29:399-405.
19. Krishnakumar S, Mohan A, Mallikarjuna K, Venkatesan N, Biswas J,
Shanmugam MP, Ren-Heidenreich L. EpCAM expression in retinoblastoma: a
novel molecular target for therapy. Invest Ophthalmol Vis Sci. 2004; 45:4247-50.
20. Balmer A, Zografos L, Munier F. Diagnosis and current management of
retinoblastoma. Oncogene. 2006;25:5341-5349.
21. Abramson DH, Frank CM, Susman M, Whalen MP, Dunkel IJ, Boyd NW 3rd.
Presenting signs of retinoblastoma. J Pediatr 1998; 132:505-8.
22. Murthy R, Honavar SG, Naik MN, Reddy VA. Retinoblastoma. In: Dutta LC, ed.
Modern Ophthalmology . New Delhi, India, Jaypee Brothers;2004: 849-59.
23. Ellsworth RM. The practical management of retinoblastoma. Trans Am
Ophthalmol Soc 1969; 67: 462-534.
24. Shields CL, Shields JA. Basic under standing of current classification and
management of retinoblastoma. Curr Opin Ophthalmol.2006;17:228-234.
25. Chantada G, Doz F, Antoneli CB, et al A proposal for an international
retinoblastoma staging system Pediatr Blood Cancer 2006:47;801-805.
26. Kashyap S, Meel R, Pushker N, et al. Clinical predictors of high risk
histopathology in retinoblastoma. Pediatr Blood Cancer. 2012; 58:356-361.
27. Zhao J, Dimaras H, Massey C, et al. Pre-enucleation chemotherapy for eyes
severely affected by retinoblastoma masks risk of tumor extension and increases
death from metastasis. J Clin Oncol. 2011; 29:845-851.
28. Chantada GL, Gonzalez A, Fandino A, et al. Some clinical findings at
presentation can predict high-risk pathology features in unilateral retinoblastoma.
J Pediatr Hematol Oncol. 2009; 31:325-329.
29. Gupta R, Vemuganti GK, Reddy VA, Honavar SG. Histopathologic risk factors in
retinoblastoma in India. Arch Pathol Lab Med. 2009;133:1210-1214.
30. Kaliki S, Shields CL, Rojanoporn D, et al. High-risk retinoblastoma based on
international classification of retinoblastoma: analysis of 519 enucleated eyes.
Ophthalmology. 2013;120:997-1003.
31. Sastre X, Chantada GL, Doz F, et al. Proceedings of the consensus meetings
from the International Retinoblastoma Staging Working Group on the pathology
guidelines for the examination of enucleated eyes and evaluation of prognostic
risk factors in retinoblastoma. Arch Pathol Lab Med. 2009;133:1199-1202.
32. Bosaleh A, Sampor C, Solemou V, et al. Outcome of children with retinoblastoma
and isolated choroidal invasion. Arch Ophthalmol. 2011;129:1416-1421.
33. Aerts I, Sastre-Garau X, Savignoni A, et al. Results of a multicentre prospective
study on the postoperative treatment of unilateral retinoblastoma after primary
enucleation. J Clin Oncol. 2013;31:1458-63.
34. Chantada GL, Dunkel IJ, de Davila MT, Abramson DH. Retinoblastoma patients
with high risk ocular pathological features: who needs adjuvant chemotherapy?
Br J Ophthalmol. 2004;88:1069-1073.
Delhiwala et al. Page 13

35. Kaliki S, Shields CL, Shah SU, Eagle RC Jr, Shields JA, Leahey A. Post
enucleation adjuvant chemotherapy with vincristine, etoposide and carboplatin
for treatment of high-risk retinoblastoma. Arch Ophthalmol. 2011;129:1422-1427.
36. Eagle RC Jr. High-risk features and tumor differentiation in retinoblastoma: a
retrospective histopathologic study. Arch Pathol Lab Med. 2009; 133:1203-1209.
37. Singh MK, Singh L, Pushker N, Sen S, Sharma A, Chauhan FA, Kashyap S.
Correlation of High Mobility Group Box-1 Protein (HMGB1) with
Clinicopathological Parameters in Primary Retinoblastoma. Pathol Oncol Res.
2015;21:1237-42.
38. Singh L, Saini N, Bakhshi S, et al. Prognostic significance of mitochondrial
oxidative phosphorylation complexes: Therapeutic target in the treatment of
retinoblastoma. Mitochondrion. 2015;23:55-63.
39. Singh L, Pushker N, Sen S, Singh MK, Chauhan FA, Kashyap S. Prognostic
significance of polo-like kinases in retinoblastoma: correlation with patient
outcome, clinical and histopathological parameters. Clin Experiment Ophthalmol.
2015;43:550-7.
40. Singh L, Pushker N, Sen S, et al. Expression of CDC25A and CDC25B
phosphatase proteins in human retinoblastoma and its correlation with
clinicopathological parameters. Br J Ophthalmol. 2015;99:457-463.
41. Singh L, Pushker N, Saini N, et al. Expression of pro-apoptotic Bax and anti-
apoptotic Bcl-2 proteins in human retinoblastoma. Clin Experiment Ophthalmol.
2015;43:259-267.
42. Rao S, Sobti P, Khurana N, Kamlesh. High risk histomorphological features in
retinoblastoma and their association with p53 expression: an Indian experience.
In J Ophthalmol. 2014;62:1069-1071.
43. Grossniklaus HE, Dhaliwal RS, Martin DF. Diffuse anterior retinoblastoma.
Retina.1998;18:238–241.
44. Hamburg A. Medulloepithelioma arising from the posterior pole.
Ophthalmologica. 1980;181:152-159.
45. Moll AC, Imhof SM, Bouter LM, Tan KE. Second primary tumors in patients with
retinoblastoma. A review of the literature. Ophthalmic Genet. 1997;18:27–34.
46. Abramson DH, Melson MR, Dunkel IJ. Third (fourth and fifth) nonocular tumors in
survivors of retinoblastoma. Ophthalmology. 2001;108:1868–1876.
47. Gombos DS, Hungerford J, Abramson DH, et al. Secondary acute myelogenous
leukemia in patients with retinoblastoma: is chemotherapy a factor?
Ophthalmology. 2007; 114:1378-83.
Delhiwala et al. Page 14

Figure Legends

Figure 1: Ophthalmoscopic (left) and gross-pathologic (right) images of retinoblastoma.

Figure 2: Retinoblastoma of the left eye in a young child, producing proptosis (left); leukocoria

was also evident. Histologically, the mass comprises a small round-cell neoplasm with foci of

dystrophic calcification (right).

Figure 3: The microscopic formation of rosettes is evident in this retinoblastoma.

Figure 4: Flexner-Wintersteiner rosettes in a retinoblastoma.

Figure 5: Scleral invasion by retinoblastoma is shown here.

Figure 6: Invasion of the choroid (left) and ciliary body (right) is present in this example of

retinoblastoma.

Figure 7: Invasion of the optic nerve by retinoblastoma.


Delhiwala et al. Page 15

Table 1: AJCC –TNM staging system for retinoblastoma

Primary Tumor (pT )


pTX Primary tumor cannot be assessed
pT0 No evidence of primary tumor
pT1 Tumor confi ned to eye with no optic nerve or choroidal invasion
pT2 Tumor with minimal optic nerve and/or choroidal invasion:
pT2a Tumor superfi cially invades optic nerve head but does not extend past lamina
cribrosa or tumor exhibits focal choroidal invasion
pT2b Tumor superfi cially invades optic nerve head but does not extend past lamina
cribrosa and exhibits
focal choroidal invasion
pT3 Tumor with signifi cant optic nerve and/or choroidal invasion:
pT3a Tumor invades optic nerve past lamina cribrosa but not to surgical resection line
or tumor exhibits
massive choroidal invasion
pT3b Tumor invades optic nerve past lamina cribrosa but not to surgical resection line
and exhibits
massive choroidal invasion
pT4 Tumor invades optic nerve to resection line or exhibits extra-ocular extension
elsewhere
pT4a Tumor invades optic nerve to resection line but no extra-ocular extension identifi
ed
pT4b Tumor invades optic nerve to resection line and extra-ocular extension identified

Regional Lymph Nodes (pN )


pNX Regional lymph nodes cannot be assessed
pN0 No regional lymph node involvement
pN1 Regional lymph node involvement (preauricular, cervical)
N2 Distant lymph node involvement

Metastasis (pM )
cM0 No metastasis
pM1 Metastasis to sites other than CNS
pM1a Single lesion
pM1b Multiple lesions
pM1c CNS metastasis
pM1d Discrete mass(es) without leptomeningeal and/or CSF involvement
pM1e Leptomeningeal and/or CSF involvement
Delhiwala et al. Page 16
Delhiwala et al. Page 17
Delhiwala et al. Page 18
Delhiwala et al. Page 19

También podría gustarte