This PDF is available for free download from a site hosted by Medknow Publications. ( Review Article

Similar documents
A RESOURCE MANUAL MANAGEMENT RETINOBLASTOMA LOW & MIDDLE RESOURCE SETTINGS

The Egyptian Journal of Hospital Medicine (October 2018) Vol. 73 (9), Page

Eye-Preserving Therapy in Retinoblastoma: Prolonged Primary Chemotherapy Alone or Combined with Local Therapy

Early detection of Retinoblastoma in children. Max Mantik

Tiffany L. Kruger, D.O. Children s Hospital of Michigan Wayne State University/Kresge Eye Institute

Retinoblastoma: A Review of Current Treatment Strategies

Retinoblastoma. Retinoblastoma

EVIDENCE BASED MANAGEMENT FOR Retinoblastoma

INDIAN COUNCIL OF MEDICAL RESEARCH INDIAN RETINOBLASTOMA GROUP

CLINICAL SCIENCES. with thermotherapy or cryotherapy is an important

CHEMOREDUCTION FOR RETINOBLASTOMA: ANALYSIS OF TUMOR CONTROL AND RISKS FOR RECURRENCE IN 457 TUMORS

Postenucleation adjuvant chemotherapy with vincristine, etoposide, and carboplatin for the treatment of high-risk retinoblastoma.

Financial Disclosures

RETINOBLASTOMA. Executive Summary

Bilateral Retinoblastoma Joseph Junewick, MD FACR

1. FBE,UEC, LFT 2. USS of eyes confirm intraocular origin, possible calcifications. 3. CT(if available) of eyes and head

Pediatric Ocular Sonography

Gene Expression Profiling has been proposed as a method of risk stratification for uveal melanoma.

Retinoblastoma. Protocol applies to retinoblastoma only.

Incidence. Clinical Manifestations

CATARACT IN RETINOBLASTOMA

Pseudohypopyon in Retinoblastoma. Choroidal Nevus. Masquerade Syndromes. Vision pathways. Flat with uniform color

Advances in Ocular Imaging

Bilateral retinoblastoma in early infancy

Outcomes of Cataract Surgery Following Treatment for Retinoblastoma

Neuroblastoma. Elizabeth Roberts. Data Coordinator CIBMTR Data Managers Mentor. Tandem Meeting February 18

What is so special about Retinoblastoma?

Retina Center of Oklahoma Sam S. Dahr, M.D. Adult Intraocular Tumors

Retinoblastoma. all information provided by:

Retinoblastoma in Nepal: case report and review

SOLID TUMOURS IN CHILDHOOD

All India Institute of Medical Sciences, New Delhi, INDIA. Department of Pediatric Surgery, Medical Oncology, and Radiology

Retinoblastoma, the most common intraocular. Utility of Pupillary Dilation for Detecting Leukocoria in Patients With Retinoblastoma

CLINICAL SCIENCES. Histopathologic Findings in Eyes With Retinoblastoma Treated Only With Chemoreduction

J of Evolution of Med and Dent Sci/ eissn , pissn / Vol. 4/ Issue 55/ July 09, 2015 Page 9665

Uveal Melanoma. Protocol applies to malignant melanoma of the uvea.

Retinoblastoma. At-A-Glance

THORACIC MALIGNANCIES

MANAGEMENT OF RETINOBLASTOMA

Vitreoretinal surgical management In ocular oncology

Complicated Cataract to Intraocular Tumors, Beware of the unexpected

PEDIATRIC ORBITAL TUMORS RADIOTHERAPY PLANNING

Financial Disclosures. The Eye in Neoplastic Disease. Course Goal. We wish to acknowledge and thank: Tumor Definition

From a suspicious cystic pineal gland to pineoblastoma in a patient with familial unilateral retinoblastoma

Retinoblastoma in Great Britain : incidence,

Retinoblastoma: clinical picture and grouping at the time of first presentation

Retinoblastoma (Rb) is currently considered to be a highly

Current therapy and recent advances in the management of retinoblastoma

Retinoblastoma Survival in Iran: 10 Years Experience of a Referral Center

Characteristic Ultrasonographic Findings of Choroidal Tumors

Outline. Brief history and principles of ophthalmic ultrasound. Types of ocular ultrasound. Examination techniques. Types of Ultrasound

Isolated Eye Relapse of Acute Lymphocy tic Leukemia during Intensification Therapy with Vincristine and Prednisolone

Indocyanine Green-Enhanced Transpupillary Thermotherapy for Retinoblastoma: Analysis of 42 Tumors

Primary Intravenous Chemotherapy for Group D Retinoblastoma: A 13-Year Retrospective Analysis

Solid Tumour Section Review

Retinoblastoma: Exploring New Horizons

Metastatic retinoblastoma: 20-year single institution experience

Dr. Lim, maybe we should start by you telling us a little about yourself and what exactly you do.

A Parent s Guide to Understanding. Retinoblastoma

Protocol for the Examination of Specimens From Patients With Retinoblastoma

Treatment of Retinoblastoma: The Role of External Beam Radiotherapy

Proton Radiation Therapy of Ocular Melanoma at PSI

Metastatic retinoblastoma of the parotid and submandibular glands: a rare case report

Role of Ultrasound Biomicroscopy in the Management of Retinoblastoma

ANNEX 1 OBJECTIVES. At the completion of the training period, the fellow should be able to:

Case Study. Monocular Malignant Melanoma

A Handbook for Families. Retinoblastoma ONCOLOGY SERIES

Case Report Tackling a Recurrent Pinealoblastoma

AMORE (Ablative surgery, MOulage technique brachytherapy and REconstruction) for childhood head and neck rhabdomyosarcoma Buwalda, J.

Recent Developments in Retinoblastoma

Effective local and systemic therapy is necessary for the cure of Ewing tumor Most chemotherapy regimens are a combination of cyclophosphamide,

North of Scotland Cancer Network Clinical Management Guideline for Carcinoma of the Uterine Cervix

Corporate Medical Policy

Transvitreal Fine Needle Aspiration Biopsy of Choroidal Melanoma via Pars Plana Vitrectomy

Acute: Symptoms that start and worsen quickly but do not last over a long period of time.

Cancers involving the eye are rare compared with lung, prostate,

Secondary Tumors After Hereditary Retinoblastoma: A Case of Orbital Leiomyosarcoma 50 Years After Initial Enucleation and Radiation Therapy

Ruthenium-106 plaque brachytherapy in the primary management of ocular medulloepithelioma

CLINICAL SCIENCES. Clinicopathologic Features of Retinoblastoma After Primary Chemoreduction

Houston, Texas, USA. Key Words. Retinoblastoma Chemotherapy Genetics of retinoblastoma Second malignancies Animal models

Ocular warning signs in GP practice: Paediatric Eye Pointers

NEW YORK UNIVERSITY SCHOOL OF MEDICINE DEPARTMENT OF OPHTHALMOLOGY EDUCATIONAL OBJECTIVES AND GOALS

MD (Ophthalmology) May 2007 Examination Paper I MD (Ophthalmology) May 2007 Examination Paper II

Retinoblastoma - A 20 year review from the University Hospital Kuala Lumpur

Dr.Dafalla Ahmed Babiker Jazan University

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors

Systemic and ocular follow-up after conservative management of an intraocular tumor

2. The clinician will know how to manage common pediatric ocular diseases

A Patient s Guide to Diabetic Retinopathy

Retinoblastoma. A Parent s Guide to Understanding. Education Understanding Retinoblastoma

Radiotherapy of choroidal metastases 1

Carlo Mosci. Ocular Oncology Service Galliera Hospital Genova Italy (

Case Rep Oncol 2012;5: DOI: /

Doppler ultrasound of the abdomen and pelvis, and color Doppler

FEP Medical Policy Manual

Retinoblastoma associated orbital cellulitis

CLINICAL PRACTICE GUIDELINES SIOP-PODC Recommendations for Graduated-Intensity Treatment of Retinoblastoma in Developing Countries

Moncef Khairallah, MD

CLINICAL SCIENCES. Intra-arterial Chemotherapy for Retinoblastoma. Report No. 1, Control of Retinal Tumors, Subretinal Seeds, and Vitreous Seeds

Transcription:

Review Article Full text online at http://www.jiaps.com Genetics and management of retinoblastoma Sameer Bakhshi, Radhika Bakhshi* Department of Medical Oncology, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, *Department of Bio-Medical Sciences, Shaheed Raj Guru College of Applied Sciences, Delhi University, New Delhi, India Correspondence: Dr. Sameer Bakhshi, Department of Medical Oncology, Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi - 110 029, India. E-mail: sambakh@hotmail.com ABSTRACT Retinoblastoma is the most common intraocular tumor in childhood. In majority of early stage retinoblastoma, the eyeballs as well as vision can be preserved with chemotherapy and local intraocular therapy with laser or photocoagulation. However, more than half the patients in India and other developing nations present in advanced stage of the disease. This article reviews the genetics, clinical approach, and treatment options for retinoblastoma focussing on advances in chemotherapy for intraocular retinoblastoma (chemoreduction), as well as improvement in survival in advanced retinoblastoma with surgery, chemotherapy, radiotherapy, and bone marrow transplantation. KEY WORDS: Childhood, genetics, retinal blastoma Retinoblastoma originates from the retinal neuroepithelium that can differentiate into almost any type of outer or inner retinal cell, including photoreceptors. It is the most common intraocular tumor in childhood, occurring in 1 of 17000 to 24000 live births, independent of race and sex. Approximately, 80% of the cases occur before 4 years of age while 40% of cases occur during infancy. This article reviews the genetics, clinical approach, and treatment options for retinoblastoma focussing on advances in survival in extraocular retinoblastoma with chemotherapy and bone marrow transplantation. GENETICS OF RETINOBLASTOMA Retinoblastoma may occur as nonhereditary or hereditary tumor. Tumors in nonhereditary retinoblastoma (60% cases) are typically solitary and unilateral with no family history and no detectable chromosomal abnormalities. Although <10% of cases of retinoblastoma have a positive family history, about 40% of retinoblastoma are of hereditary origin caused by a germline mutation in RB1 gene on chromosome 13q14. This is due to the majority being the result of a de novo mutation in the RB1 gene. Amongst those with hereditary retinoblastoma (40% cases), 25% cases have bilateral disease while 15% have unilateral disease. Hereditary RB1 mutations are found in all cells thereby increasing risk of other cancers, in particular, osteosarcoma and malignant melanoma. Inherited retinoblastoma is transmitted as an autosomal dominant trait with high but incomplete penetrance. Children of patients with hereditary retinoblastoma have a one in two chance of carrying the germ cell mutation and for those who are carriers, the probability of developing retinoblastoma is very close to 90% if parents have bilateral retinoblastoma but probably less if they have the unilateral form. [1] The probability of developing retinoblastoma in offsprings and siblings of patients is higher than in the general population because of which they need genetic counseling [Table 1] so as to effectively counsel and diagnose retinoblastoma early in high risk subjects. In bilateral cases or those with a positive family Table 1: Risk of developing retinoblastoma in siblings and off springs of patients Subjects Probability of disease % Subjects with carriers of RB1 gene mutation 90 Offspring of patient 45 Sibling of patient (if either parent is affected) 45 Sibling of patient with bilateral disease 2 Sibling of patient with unilateral disease 1 109

history, one can directly perform the genetic studies on peripheral blood sample as they would always be due to germline mutation in RB1 gene. In unilateral cases and those with a negative family history, there is only 15%-20% chance that somatic cells would also carry the mutation. Therefore, in these cases, genetic studies for RB1 gene mutation are initially performed in the tumor specimen, and subsequently, once the mutation has been identified, peripheral blood is screened to determine if it is a germline mutation (hereditary retinoblastoma) or non hereditary retinoblastoma [Figure 1]. If genetic testing is not pursued, then tumor surveillance for all siblings of the affected patient is recommended, which includes examination under anesthesia (EUA) at birth and every four months until the age of 4 years. On the contrary, if genetic testing demonstrates that the sibling did not inherit RB1 gene mutation, the surveillance and anesthesia required for eye examinations can be avoided, thereby decreasing costs and potential morbidity. Further, in unilateral retinoblastoma, if it can be demonstrated that the child does not carry a germline RB1 gene mutation, then (a) the child is not at substantial risk for second malignancies; (b) radiation therapy is associated with less hazards and; (c) the parents and eventually the child have a negligible risk of having another child with retinoblastoma. CLINICAL PRESENTATION The median age of diagnosis is 2 years. Bilateral retinoblastoma occurs earlier than unilateral disease. Majority of retinoblastoma in developed nations Unilateral retinoblastoma Screen tumor (enucleated specimen) for RB1 gene mutations Present Hereditary retinoblastoma presents as an intraocular disease wherein 90% of cases are curable. However, in the developing nations, still 65% of patients may present with extraocular disease. An unpublished data from our center showed that mean age of presentation of unilateral retinoblastoma is 34 months, while that of bilateral disease is 24 months, which is higher than that in the west. The mean duration of symptoms was 8 months prior to visiting the tertiary care center, and 49% of patients presented with extraocular disease. This may be due to the lack of awareness, delayed referrals, and/or more aggressive disease biology. From a therapeutic point of view, retinoblastoma presentation may be divided into the following subtypes: A. Early stage intraocular disease (preservation of eye possible) presents most commonly with leucocoria (white reflex with an ophthalmoscope) followed by strabismus. Therefore, a pediatrician should always include the documentation of a red reflex with an ophthalmoscope in examination of newborn and at subsequent well baby visits of a child. The absence of red reflex or strabismus merits an examination by an ophthalmologist in order to detect early stage intraocular retinoblastoma. B. Advanced intraocular retinoblastoma (needs enucleation) presents with rubeosis iridis, hyphema, hypopyon, and glaucoma. The potential diagnosis of retinoblastoma should always be investigated in a child presenting with spontaneous hyphema in the absence of trauma or who presents with signs of endophthalmitis. Pain is not a feature of intaocular retinoblastoma unless secondary glaucoma or inflammation is present. C. Extraocular retinoblastoma presents with proptosis, preauricular lymphadenopathy, bone pain, features suggestive of bone marrow or central nervous system involvement. Investigations The diagnostic work up includes EUA of the eye and ultrasonography and CT scan of the orbit and head to confirm the diagnosis of retinoblastoma, and to detect ectopic disease in the pineal gland. The clinical presentation and ancillary radiological findings are typical for retinoblastoma in majority of the patients, and a tissue biopsy is not required for confirmation prior to the therapy. However, a fine needle aspiration biopsy may be performed in select cases of pediatric ophthalmological disease where the diagnosis is in question. Metastatic work up includes cerebrospinal fluid examination for malignant cells and bone marrow biopsy and bone scan in those with extraocular disease. Intraocular retinoblastoma is traditionally classified by Reese-Ellsworth (RE) [2] classification [Table 2], whereas intraocular and extraocular retinoblastoma is classified according to Grabowski-Abramson [3] staging [Table 3]. Absent Non hereditary retinoblastoma Screen peripheral blood for same RB1 gene mutation Sibling Both parents Present Absent Present Absent Screen child with 4 monthly EUA till 4 years No follow-up Hereditary germline mutation Acquired germline mutation Figure 1: Algorithm for genetic studies in unilateral retinoblastoma 110

MODES OF THERAPY IN RETINOBLASTOMA The aim of treatment in retinoblastoma is to cure the patient with preservation of vision; the second aim is to minimize the long-term effects of therapy. With advances in therapy, the survival has risen from 30% in 1930 s to nearly 95% in 1990 s for non-metastatic retinoblastoma. However, untreated retinoblastoma is always fatal. The major therapies that have resulted in this improved survival are enucleation and external beam radiation therapy (EBRT), both of which are associated with significant morbidity. A. Enucleation cures localized retinoblastoma but at the cost of loss of sight. Apart from its obvious adverse physiologic and psychological effects, enucleation can be associated with chronic local effects such as discharge from orbit, contraction of socket, and extrusion of implant. Thus, there is a need for relatively non-invasive focal ophthalmological therapies. B. EBRT provides good local control in retinoblastoma Table 2: Reese-Ellsworth classification of retinoblastoma Group I - Very favorable prognosis A. Solitary tumor, less than 4 disc diameters (dd)* in size, at or behind the equator B. Multiple tumors, less than 4 dd in size, all at or behind the equator Group II - Favorable prognosis A. Solitary lesion 4-10 dd in size, at or behind the equator B. Multiple tumors, 4-10 dd in size, behind the equator Group III- Doubtful prognosis A. Any lesion anterior to the equator B. Solitary tumors larger than 10 dd behind the equator Group IV - Unfavorable prognosis A. Multiple tumors, some larger than 10 dd B. Any lesion extending anteriorly to the ora serrata Group V - Very unfavorable prognosis A. Massive tumors involving over half the retina B. Vitreous seeding *One disc diameter = 1.6 mm when used in conjunction with local non-invasive ophthalmological therapies. However, it has significant local side effects such as xerophthalmia, cataract, retinopathy, and keratopathy; it often does not spare vision. It adversely affects midface growth in 90% of the patients. The risk of secondary non-ocular malignant tumors increases 6-fold after EBRT especially in those with germline mutation of RB1. [4-6] Patients carrying RB1 germline mutation have a 35% cumulative risk of secondary cancers in the radiation field by the age of 30 years, whereas those who do not have RB1 mutation have a risk of 6%. This effect may be age dependent with the greatest risk in those retinoblastoma patients with hereditary disease treated under-1 year of age. The cumulative risk of death from secondary tumors is 26% at 40 years of age. Plaque radiotherapy avoids long-term complications of EBRT, but this cannot be used in large tumors, tumors with vitreous seeding, or tumors at posterior pole. C. Chemotherapy is one of the possible treatment modalities, which is free from long-term effects of radiation. Drugs commonly used in retinoblastoma include vincristine, adriamycin, idarubicin, cyclophosphamide, cisplatin, carboplatin, and etoposide. Carboplatin is preferred over cisplatin because of its reduced ototoxicity and nephrotoxicity profile as compared to cisplatin. An increased incidence of secondary primary tumors has been attributed to the use of cyclophosphamide and etoposide in children with RB1 mutations. Recently, combination of vincristine, etoposide, and carboplatin (VEC) is the preferred drug combination [Table 4]. Carboplatin has good penetration in eye, brain, and bone marrow, which are two potential sites for metastatic disease in retinoblastoma. Multidrug resistance is caused by overexpression of membrane-associated energy-dependent drug efflux pump, the P-glycoprotein. The P-glycoprotein is encoded by mdr1 gene and cells with multidrug resistance often show amplification of this gene. Although, the effect of P-glycoprotein is reversible Table 3: Grabowski and Abramson staging for intra- and extra-ocular retinoblastoma Intraocular disease a. Retinal disease b. Extension to lamina cribrosa c. Uveal extension Orbital disease a. Orbital tumor 1. Scattered episcleral cells 2. Orbital invasion b. Optic nerve 1. Invasion upto cut end 2. Invasion beyond the cut end Intracranial metastasis a. Positive cerebrospinal fl uid alone b. Mass lesion in the central nervous system Hematogenous metastasis a. Positive bone marrow alone b. Focal bone lesions with/without bone marrow disease Table 4: Chemotherapy vincristine, etoposide, and carboplatin protocol A. Drugs Vincristine 1.5 mg/m 2 day 1 (0.05 mg/kg for children <3 years and max dose 2 mg) Carboplatin 560 mg/m 2 day 1 (18.6 mg/kg for children <3 years) Etoposide 150 mg/m 2 days 1 and 2 (5 mg/kg for children <3 years) B. Cycles Every 3-4 weeks; Ensure ANC >1000 and platelets >100,000/mm 3 C. Number of Cycles 2-6 cycles for chemoreduction 6 cycles for chemoprevention 6-12 cycles for systemic disease 111

with high concentrations of cyclosporine and might even be diminished in future by using MoAbs against P-glycoprotein, role of cyclosporine in reversing drug resistance in patients with retinoblastoma remains unclear in absence of a randomized trial. [7] Chemotherapy is used in retinoblastoma in three settings: intraocular RB, micrometastatic RB, and overt dissemination. CHEMOTHERAPY IN INTRAOCULAR RETINOBLASTOMA (CHEMOREDUCTION) Chemoreduction is the use of chemotherapy to shrink the tumor so that local treatment can be delivered to a smaller volume and cause less morbidity. This technique has been employed in an effort to avoid or at least delay EBRT and enucleation for children with retinoblastoma, especially those with bilateral disease. The great advantage of chemoreduction in retinoblastoma seems to be the ability to move the tumor margins away from visually vital structures, such as optic disc and foveola. Retinal tumors generally respond rapidly to chemoreduction; residual tumors can thereafter be destroyed without vision loss by using adjuvant local brachytherapy, photocoagulation, cryocoagulation, and/or laser therapy. Eyes with additional vitreous seeds or subretinal seeds are managed differently using chemoreduction without focal consolidation treatments because the number of seeds is usually far beyond the capability of focal treatment methods and the multitude of tiny seeds typically respond with regression, calcification, and often complete disappearance after several months of treatment. In applying this strategy in the management of bilateral retinoblastoma, it seems appropriate to conserve both eyes at first. The decision to enucleate one eye can be postponed at least until early response to primary chemotherapy has been assessed. It is then easier to judge which eye is salvageable and which not. Chemotherapy is administered every 3-4 weekly for 2-6 cycles. Patient requires tedious monitoring every 2-3 weekly by examination under anesthesia of the affected eye. Chemotherapy by itself is not sufficient to cure retinoblastoma, and so, additional focal treatment is mandatory since histopathological evaluation still reveals viable proliferating tumor cells after 2-6 cycles. [8] At any suggestion of tumor progression, the eye needs enucleation or EBRT. A regimen with fewer than 6 cycles is less effective in preventing enucleation or EBRT, especially in eyes with Reese-Ellsworth (RE) stages IV and V. [9] Efficacy of chemoreduction An early volume reduction of around 50% after two courses of VEC can usually be expected. [10] The overall salvage of eyes in the chemoreduction studies is about 80%. EBRT was added in an additional 30% of the salvaged eyes. Thus, approximately 50-60% of affected eyes treated with chemoreduction are successfully preserved with avoidance of EBRT or enucleation. [11] The rate of globe preservation is best with less advanced eyes (85%), such as those in RE groups I to IV whereas with more advanced eyes, such as those in RE group V, preservation is less successful at <50%. In a study by Shields et al., [12] chemoreduction using six cycles of VEC offers satisfactory retinoblastoma control for RE groups I-IV eyes, with treatment failure necessitating additional EBRT in only 10% of eyes and enucleation in 15% of eyes at 5-year follow-up. Patients with RE group V eyes required EBRT in 47% and enucleation in 53% at 5 years. Thus, all localized intraocular retinoblastoma have a potential for eye preservation using chemoreduction [Figure 2]. However, any evidence for potential micrometastatic disease or overt metastatic disease should be excluded from chemoreduction [Table 5]. Recurrence of retinoblastoma following chemoreduction The mean interval from discontinuation of chemoreduction to first recurrence of retinal tumor was 4 Evidence of micro or Overt metastatic disease Exclude from chemoreduction Intraocular retinoblastoma Metastatic work-up Orbital and head scan Bone marrow biopsy CSF cytology Reduction in size Focal ophthalmological therapy CT for total of 6 cycles Localized intraocular disease CT 2-6 cycles Assess every 3 weekly Close follow up for recurrence for Retinal and subretinal tumors: Upto 3 years Vitreous seeding: Upto 5 years Progressive disease Enucleation Figure 2: Algorithm for management of intraocular retinoblastoma 112

Table 5: Exclusion criteria for treatment with chemoreduction Table 6: Post-enucleation specimen (histopathological criteria for chemoprevention) Biomicroscopic evidence of iris neovascularization Neovascular glaucoma Tumor invasion into the anterior chamber, iris, optic nerve and/or choroid Extraocular disease as documented by clinical, ultrasonographic, and neuroimaging modalities If vitrectomy is performed for an eye with unsuspected retinoblastoma months, recurrence of vitreous seeds was 2 months, and recurrence of subretinal seeds was 2 months. [13] Thus, monitoring of the eye is especially critical following chemoreduction to detect recurrence. It is reassuring to know that most children manifest their recurrent retinal tumors and subretinal seeds by 3 years after treatment with little recurrence thereafter; accordingly, followup can be adjusted for this time interval. Vitreous seed recurrence, however, continues to be a problem up to 5 years after treatment and potentially longer; therefore, patients with vitreous seeds at initial examination might require cautious ocular examination for many years following treatment. At 5-year follow-up, the recurrence rates for intraretinal tumors, vitreous seeds, and subretinal seeds were seen in 24%, 50%, and 62% of eyes, respectively. [13] Those at greatest risk for retinal tumor recurrence are eyes with tumor-associated subretinal seeds surrounding the base of the tumor. Patients at greatest risk for vitreous or subretinal seed recurrence are those who, at initial examination are younger, had large tumor dimensions, and had tumor-associated subretinal seeds. All children receiving a chemoreduction protocol should be monitored by retinoblastoma specialist who is able to detect minute recurrences and capable of treating the recurrences. CHEMOTHERAPY FOR RETINOBLASTOMA POST- ENUCLEATION (CHEMOPREVENTION) With improved understanding of risk factors predictive of metastasis and availability of effective chemotherapy regimens for intraocular retinoblastoma, it would seem logical to consider chemotherapy following enucleation to prevent metastasis in high-risk cases; this is referred to as chemoprevention. Various histopathological factors have been identified as potential risk factors for retinoblastoma [Table 6]; however, there is some controversy as to whether choroidal involvement alone is a significant risk factor for metastases. [14-16] The current strategy is to give 6 cycles VEC to prevent metastases, and the rate of metastasis is significantly reduced in the group receiving chemoprevention as compared with the group that did not receive chemotherapy (4% vs. 24%). Indications for chemoprevention Anterior chamber seeding Iris infi ltration Ciliary body infi ltration Massive choroidal infi ltration Invasion of optic nerve lamina cribrosa Retrolaminar optic nerve invasion Invasion of optic nerve transection* Scleral extension* Indications for no additional chemotherapy Intraretinal extension Prelaminar optic nerve invasion *These require additional EBRT as this is considered as extraocular disease limited to orbit Kaplan-Meier estimates showed that 96% of patients who received adjuvant therapy would remain free of metastasis at 10 years post-enucleation compared with 76% of those who did not receive adjuvant therapy. [15] Chemoprevention following vitrectomy Retinoblastoma may present with atypical features such as vitreous hemorrhage or signs of vitreous inflammation, particularly in older children. Vitrectomy should be avoided in these cases until the possibility of underlying retinoblastoma is excluded. If vitrectomy is performed in an eye with unsuspected retinoblastoma, enucleation combined with chemotherapy, radiotherapy or both without delay is advised to prevent systemic tumor dissemination. [17] Chemotherapy for extraocular retinoblastoma Metastatic retinoblastoma is seen in less than 10% cases in developed nations whereas almost two-third of cases of retinoblastoma in developing countries. Chemotherapy is indicated in all these situations and is used in the following two fashions: 1. Conventional chemotherapy wherein the same drugs are used as is used in chemoreduction or chemoprevention, but for a longer duration of 6-12 months. 2. High dose chemotherapy (HDT) wherein after initial conventional chemotherapy, the patient is consolidated with high doses of the same agents and bone marrow rescued with an autologous stem cell transplantation (ASCT). Two different subgroups of patients with extraocular retinoblastoma with different outcome can be distinguished [Figure 3] as follows: A. Extraocular disease limited to orbit alone (invasion upto or beyond the cut end of optic nerve; scleral invasion upto the orbital contents) or with concomitant lymph node invasion. These patients have a 5-year progression free survival of >80% using initial exenteration followed by intensive chemotherapy 113

Metastatic Disease Orbital and head CT scan Bone marrow biopsy Bone scan CSF cytology metastatic disease cannot be cured with conventional chemotherapy in majority of the cases; however, HDT with ASCT appears to be a promising therapy for such cases. Retinoblastoma with CNS metastases continues to have dismal prognosis despite HDT with SCT and/or cranial radiation. Orbital and/or pre-auricular nodal disease Distant metastatic disease REFERENCES Orbital Exenteration Neoadjuvant CT CT 2-4 cycles, local RT and/or surgery CT and RT Limited Surgery CR or PR Progressive Disease Figure 3: Algorithm for management of metastatic retinoblastoma and radiotherapy. [18] Similar results have also been obtained using initial neoadjuvant chemotherapy followed by limited surgery (enucleation or resection of residual orbital mass) and adjuvant therapy and radiotherapy. [19] Comparable results have been reported using HDT with ASCT. Thus, HDT in these two situations seems to be a therapeutic alternative with the advantage of shorter duration of therapy. [20] Further, most of the failures or recurrences are in the CNS, and thus, intrathecal chemotherapy is used by some centers along with chemotherapy. B. Those with systemic and/or CNS dissemination (bones, bone marrow, positive CSF cytology, or mass lesion in brain) are seldom cured with conventional chemotherapy. However, HDT using carboplatin, etoposide, and cyclophosphamide is effective in patients with chemosensitive retinoblastoma patients with distant metastatic disease, except those with CNS disease. [21] Prognosis is extremely poor in those with CNS disease. CNS irradiation, as is currently employed, does not cure CNS disease. Role of intrathecal therapy using methotrexate, cytosine arabinoside, and hydrocortisone, as is employed in CNS leukemia, is debatable. Thus, more effective therapeutic strategies are required to cure CNS disease in retinoblastoma. CONCLUSION Adjuvant CT and RT HDT with ASCT Alternate therapy Retinoblastoma is a chemosensitive disease but cannot be cured with chemotherapy alone. It is a very effective mode of therapy in preserving vision and the long-term complications of enucleation and EBRT, especially in intraocular retinoblastoma. Metastatic retinoblastoma to the orbit can be treated with good results with combination chemotherapy, radiotherapy, and possibly, conservative eye surgery as well. Distant 1. Draper GJ, Sanders BM, Brownbill PA, Hawkins MM. Patterns of risk of hereditary retinoblastoma and applications to genetic counseling. Br J Cancer 1992;66:211-9. 2. Reese AB, Ellsworth RM. The evaluation and current concept of retinoblastoma therapy. Trans Am Acad Ophthalmol Otolaryngol 1963;67:164-72. 3. Grabowski EF, Abramson DH. Intraocular and extraocular retinoblastoma. Hematol Oncol Clin North Am 1987;1:721-35. 4. Hasegawa T, Matsuno Y, Niki T, Hirohashi S, Shimoda T, Takayama J, et al. Second primary rhabdomyosarcomas in patients with bilateral retinoblastoma: A clinicopathologic and immunohistochemical study. Am J Surg Pathol 1998;22:1351-60. 5. Mohney BG, Robertson DM, Schomberg PJ, Hodge DO. Second nonocular tumors in survivors of heritable retinoblastoma and prior radiation therapy. Am J Ophthalmol 1998;126:269-77. 6. Imhof SM, Moll AC, Hofman P, Mourits MP, Schipper J, Tan KE. Second primary tumours in hereditary- and nonhereditary retinoblastoma patients treated with megavoltage external beam irradiation. Doc Ophthalmol 1997;93:337-44. 7. Gallie BL, Budnig A, DeBoer G, Thiessen JJ, Koren G, Verjee Z, et al. Chemotherapy with focal therapy can cure retinoblastoma without radiotherapy. Arch Ophthalmol 1996;114:1321-8. 8. Bechrakis NE, Bornfeld N, Schueler A, Coupland SE, Henze G, Foerster MH. Clinicopathologic features of retinoblastoma after primary chemoreduction. Arch Ophthalmol 1998;116:887-93. 9. Gunduz K, Shields CL, Shields JA, Meadows AT, Gross N, Cater J, et al. The outcome of chemoreduction treatment in patients with Reese-Ellsworth group V retinoblastoma. Arch Ophthalmol 1998;116:1613-7. 10. Shields CL, Shields JA, Needle M, de Potter P, Kheterpal S, Hamada A, et al. Combined chemoreduction and adjuvant treatment for intraocular retinoblastoma. Ophthalmology 1997;104:2101-11. 11. Schouten-Van Meeteren AY, Moll AC, Imhof SM, Veerman AJ. Overview: Chemotherapy for retinoblastoma: An expanding area of clinical research. Med Pediatr Oncol 2002;38:428-38. 12. Shields CL, Honavar SG, Meadows AT, Shields JA, Demirci H, Singh A, et al. Chemoreduction plus focal therapy for retinoblastoma: Factors predictive of need for treatment with external beam radiotherapy or enucleation. Am J Ophthalmol 2002;133:657-64. 13. Shields CL, Honavar SG, Shields JA, Demirci H, Meadows AT, Naduvilath TJ. Factors predictive of recurrence of retinal tumors, vitreous seeds and subretinal seeds following chemoreduction for retinoblastoma. Arch Ophthalmol 2002;120:460-4. 14. Finger PT, Harbour JW, Karcioglu ZA. Risk factors for metastases in retinoblastoma. Surv Ophthalmol 2002;47:1-16. 15. Honavar SG, Singh AD, Shields CL, Meadows AT, Demirci H, Cater J, et al. Post enucleation adjuvant therapy in high-risk retinoblastoma. Arch Ophthalmol 2002;120:923-31. 16. Bellaton E, Bertozzi AI, Behar C, Chastagner P, Brisse H, Sainte- Rose C, et al. Neoadjuvant chemotherapy for extensive unilateral retinoblastoma. Br J Ophthalmol 2003;87:327-9. 17. Shields CL, Honavar S, Shields JA, Demirci H, Meadows AT. Vitrectomy in eyes with unsuspected retinoblastoma. Ophthalmology 2000;107:2250-5. 18. Doz F, Neuenschwander S, Plantaz D, Courbon B, Gentet JC, Bouffet E, et al. Etoposide and carboplatin in extraocular retinoblastoma: A study by the Societe Francaise d Oncologie 114

Pediatrique. J Clin Oncol 1995;13:902-9. 19. Chantada G, Fandino A, Casak S, Manzitti J, Raslawski E, Schvartzman E. Treatment of overt extraocular retinoblastoma. Med Pediatr Oncol 2003;40:158-61. 20. Namouni F, Doz F, Tanguy ML, Quintana E, Michon J, Pacquement H, et al. High-dose chemotherapy with carboplatin, etoposide and cyclophosphamide followed by a haematopoietic stem cell rescue in patients with high-risk retinoblastoma: A SFOP and SFGM study. Eur J Cancer 1997;33:2368-75. 21. Rodriguez-Galindo C, Wilson MW, Haik BG, Lipson MJ, Cain A, Merchant TE, et al. Treatment of metastatic retinoblastoma. Ophthalmology 2003;110:1237-40. Source of Support: Nil, Conflict of Interest: None declared. Author Help: Online Submission of the Manuscripts Articles can be submitted online from http://www.journalonweb.com. For online submission articles should be prepared in two files (first page file and article file). Images should be submitted separately. 1) First Page File: Prepare the title page, covering letter, acknowledgement, etc., using a word processor program. All information which can reveal your identity should be here. Use text/rtf/doc/pdf files. Do not zip the files. 2) Article file: The main text of the article, beginning from Abstract till References (including tables) should be in this file. Do not include any information (such as acknowledgement, your names in page headers, etc.) in this file. Use text/rtf/doc/pdf files. Do not zip the files. Limit the file size to 400 kb. Do not incorporate images in the file. If file size is large, graphs can be submitted as images separately without incorporating them in the article file to reduce the size of the file. 3) Images: Submit good quality colour images. Each image should be less than 400 kb in size. Size of the image can be reduced by decreasing the actual height and width of the images (keep up to about 4 inches) or by reducing the quality of image. All image formats (jpeg, tiff, gif, bmp, png, eps, etc.) are acceptable; jpeg is most suitable. The image quality should be good enough to judge the scientific value of the image. Always retain a good quality, high resolution image for print purpose. This high resolution image should be sent to the editorial office at the time of sending a revised article. 4) Legends: Legends for the figures/images should be included at the end of the article file. 115