A Narrative Review - Therapy Options and Therapy Failure in Retinoblastoma

 

Argyrios Chronopoulosa    Neele Babstb    Christian Schiemenzb    James S. Schutza    

Ludwig M. Heindlc,d    Mahdy Ranjbarb    Vinodh Kakkasseryb

 

aDepartment of Ophthalmology, Hospital of Ludwigshafen, Akademisches Lehrkrankenhaus der Johannes-Gutenberg-Universität Mainz und der Medizinischen Fakultät Mannheim der Universität Heidelberg, Ludwigshafen am Rhein, Germany, bDepartment of Ophthalmology, University of Lübeck, Lübeck, Germany, cDepartment of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany, dCentrum für Integrierte Onkologie (CIO) Aachen-Bonn-Köln-Düsseldorf, Köln, Deutschland

 

 

 

 

Key Words

Retinoblastoma • Therapy • Therapy failure • Enucleation • Metastasis

 

Abstract

Retinoblastoma (RB) management has evolved over the last three decades. Goals of modern RB treatment are first to protect life and prevent metastatic disease, then preservation of the globe and useful vision. With modern treatment protocols and early disease detection success rates can reach up to 100% of disease-free-globe and eye preservation. Treatment of advanced cases remains complex, requiring aggressive chemotherapy or/and external beam radiation. Treatment protocols are extremely diverse and dependent on local resources thus success rates are variable. Here we review narratively current treatment protocols and failure rates based on a PubMed search using keywords of retinoblastoma, retinoblastoma seed, retinoblastoma treatment, enucleation.

 

 

Introduction

 

Retinoblastoma (RB) is the most common ocular malignancy in childhood and is lethal if untreated. Modern treatment protocols have evolved in the last three decades resulting in substantially improved treatment with up to almost a high percentage of disease-free, globe-salvaging survival rate when disease is identified in its still intraocular stage [1-5]. Goals of modern treatment aim to protect life by preventing metastatic disease, followed by globe preservation, and when possible preservation of useful vision. Advanced treatment algorithms aim for additional improvement in globe preservation even in advanced cases and are possible only in centers that can provide comprehensive treatment.

Chemoreduction and focal treatment methods are not uniformly successful in controlling intraocular RB. Chemoreduction failure has been defined as unresponsive or recurrent RB. Local recurrence can occur at three anatomic locations: as retinal tumor, as vitreous seeds, and as subretinal seeds. Eyes that fail chemoreduction eventually require external beam radiotherapy (EBR) which can induce secondary central nervous system malignancy. Delayed diagnosis and treatment may also lead to extraocular metastasis, with central nervous system involvement having particularly poor prognosis [6, 7]. As therapy protocols are extremely diverse and vary a lot depending on region and available resources, the objective of this review is to describe current treatment protocols with respect to their success rate and causes of therapy failure in RB.

 

 

Study selection and data extraction

 

For this narrative review, we performed a search on Medline and PubMed using the key words: Retinoblastoma, retinoblastoma seed, retinoblastoma treatment, "retinoblastoma and enucleation". Titles and abstracts of identified studies were independently reviewed by two investigators (AC and VK) and a full-text review was carried out on potentially eligible studies to evaluate inclusion. The following data were collected: publication year, study design, diagnosis of RB, type of RB, treatment, follow-up, visual and anatomic outcomes, adverse events. Visual outcome was reported as best corrected visual acuity (BCVA) change. Anatomic outcome included the type and extension of RB, laterality and type of recurrence, incidence, and localization of recurrence. When a study provided only individual patient data because of a case series design, means were calculated. The searches yielded a total of 8853 records which were screened for eligibility. 146 articles received full-text evaluation and inclusion in the analysis. Among the 146 included studies, 12 reported general findings on classification, incidence, and mortality. The rest reported on different RB treatments.

 

 

Incidence of retinoblastoma

 

RB is the most common pediatric primary intraocular malignancy with an incidence of approximately 1 in 15,000 to 1 in 23,000 live births [8]. It represents 3% of childhood cancers [9]. Unilateral cases are usually diagnosed at an average age of 18 months, bilateral cases slightly earlier at an average age of about 12 months. Most of newly diagnosed cases (94%) are sporadic and only 6% are familial. There is no apparent predisposition for race, sex, or laterality. Most of RBs are unilateral; 25–35% of cases demonstrate bilateral involvement. In rare instances, the tumor is first recognized at birth, in the teens, or even in adulthood [10].

Despite the ongoing, advanced globe sparing therapies, modern literature lacks information on incidence of metastases and metastatic deaths from RB. In USA for example survival rates have increased following improved therapies from the 1970s to 2010, with 5-year overall survival reported as 93.7% for 1975–1979 up to 97% presently [11]. A large retrospective study in 2019 for unilateral RB reported an overall metastatic rate of 3.9% and death rate of 1.75% [12]. A similar UK retrospective study from 2009 reported survival rates for unilateral cases at 93% at 1 year and 86% at 20 years and for bilateral RB 86% at 1 year and 80% at 20 years [13]. Furthermore, in 2019 a large registry study reported an overall survival rate of 95.2 [14].

 

 

Therapy of retinoblastoma

 

Management of RB is in constant evolution and treatment varies among different centers worldwide but highest success rates are reported from centers that can provide comprehensive therapy.

Systemic chemotherapy combined with adjunctive local treatment such as laser photocoagulation, transpupillary thermotherapy, cryotherapy, and brachytherapy has been widely used in the treatment of RB. The most common regimen for systemic chemotherapy is vincristine, carboplatin, and etoposide (VEC regimen). The ophthalmic artery infusion chemotherapy technique has been introduced to provide a targeted therapy with better tissue response while minimizing systemic toxicity and has been refined and is now known as direct intra-arterial chemotherapy (IAC) for treating advanced intraocular RB [15, 16]. A commonly used drug in IAC is melphalan with or without the combination of topotecan and/or carboplatin [17]. Intravitreal chemotherapy injection (IViC) is a further option for targeted localized treatment and has been mainly reported for treating severe and monocular cases with recurrent RB and extensive vitreous seeding [18]. Vitreous seeds represent a challenge in the management of RB because the avascular vitreous cavity has poor penetration of systemic chemotherapy. Even with IAC, approximately 30% of eyes are removed because of the presence of vitreous seeds [19, 20]. All treatment options have been summarized in Table 1.

 

Table 1. Treatment option for intraocular retinoblastoma

 

 

Intra-arterial chemotherapy

 

Since 2008, treatment of children with RB by selective ophthalmic artery chemotherapy, IAC, has become the first-line treatment for advanced and recurrent intra-ocular RB, significantly reducing the need for enucleation and minimizing systemic chemotherapy toxicity [16, 21-23]. Specialized centers offer IAC for RB [24]; however, the worldwide use of primary IAC in advanced RB ranges from 0% to 100% with some centers enucleating 100% of ‘D’ eyes, while others treat 100% of ‘D’ eyes [25]. A retrospective analysis of 73 patients demonstrated that IAC is an effective and safe treatment for RB, with a technical success rate of 98.5% and an overall ocular preservation rate of 78.5%. Although some patients experienced minor side effects, major complications did not occur [26]. In another retrospective study, globe salvage was achieved in 72% of primary-treated patients and in 62% of secondary-treated patients [24]. Only advanced Group D or E cases were treated in this way with an attempt to salvage the eye and minimize risk of metastatic disease, which did not occur in any of the treated cases. Complete control was achieved in 80% of the cases with recurrent solid tumor, in 55% with recurrent subretinal seeds, and in 43% with recurrent vitreous seeds [27]. Similar results with IAC were reported in a study with fewer patients; globe salvage was achieved in 72% of primary-treated cases and in 62% of secondary-treated cases with a mean follow-up of 19 months [28]. Specifically, primary therapy achieved globe salvage for group B (100%), group C (100%), group D (94%), and group E (36%) [24]. Salvation of two-thirds of eyes with advanced intra-ocular RB managed primarily with IAC without need for enucleation or external beam radiotherapy has been reported [25]. More orbital recurrences in a group primarily treated with enucleation (7.9%) than in the IAC group (1.3%) were reported in a single-institution retrospective study of advanced intraocular RB[29].

 

 

Combined intra-arterial chemotherapy and intravitreal chemotherapy

 

While IAC is effective for the majority of group B, C, and D eyes, the efficacy of IAC alone is limited in eyes with extensive vitreous seeds [20, 24, 30-35]. The addition of adjuvant (IViC) has been found to be effective for eyes with vitreous seeds to achieve comparable globe salvage rates for group D and E eyes with persistent active vitreous seeds [36-42]. With IViC group E eyes have improved significantly to 73% compared to 25% in the era of IAC alone [35]. Combined treatment has been reported to be effective and safe for the treatment of advanced unilateral RB achieving a high control of both the retinal tumor and vitreous seeds, particularly cloud vitreous seeds, with only one recurrent case after the initial treatment; globe salvage rate was in 93.3% with no severe systemic complications occurring and no extraocular tumor extension or metastasis during the follow-up period [43].

 

 

Combined intra-arterial chemotherapy and intravenous chemotherapy

 

Intravenous chemotherapy (IVC) in the treatment of RB was first used in the mid-90s and unexpectedly achieved a globe salvage rate of >90% for eyes in groups A–C [44-47]. Additionally, there is evidence that IVC as adjuvant postoperative treatment can effectively prevent metastasis in high-risk patients who have undergone enucleation [48]. However, a major obstacle to the use of IVC is the blood–retina barrier, which limits the entry of chemotherapeutic agents into the eye, reducing their efficacy while causing systemic adverse effects. Since 2006, IAC has become a common treatment method for managing RB due to its targeted application and higher local efficacy with lower overall systemic sideeffects [15, 49]. This modality has been shown to control relatively advanced tumors that would previously have required enucleation or would have failed the first-line treatment [3, 19, 20, 24-26, 32]. No additional effect of IVC combined with IAC was demonstrated in a study follow-up period of 3-year in comparison to primary IAC for treating advanced intraocular RB [50].

A study of the effectiveness of IAC, IVC and their combination in the management of group D RB demonstrated that in group D patients, primary IAC achieved ocular survival in 76.7% of eyes with less need for local consolidation treatment, whereas following primary IVC, ocular survival was 43.2% [51].

 

 

Intra-arterial chemotherapy in advanced stages

 

IAC improved ocular survival rates for eyes with advanced RB (naive or not) saving the majority of eyes with advanced RB in a 9-year period study [52]. 46% of advanced eyes in preverbal children treated with IAC achieved “fair” to “excellent” ERG amplitudes at last follow-up but future research is needed to determine the actual vision functional gains in this population [34].

 

 

Primary enucleation

 

Enucleation has been performed for RB for more than 200 years [53, 54]. Survival after enucleation for RB was 5% in 1869, 17% in 1897 and 57% in 1916 [12]. Further advances allowed some unilateral RB to be managed without primary enucleation with survival rates of 63% and 70%, and finally 92% by the end of the 20th century [55-57]. Eventually, advanced globe sparing therapies allowed some unilateral eyes to be salvaged, often with useful vision and without compromising patient survival [52]. Enucleation is still performed in every RB center worldwide but there has been a lack of recent literature on incidence of metastases and metastatic deaths following enucleation surgery. Primary enucleation for unilateral RB is still associated with a 3.9% chance of metastases within a year, mostly in patients with higher risk histopathology [12].

In a single-institution retrospective study, advanced intraocular RB primarily treated with enucleation had more frequent orbital recurrences [58]. IAC for advanced intraocular RB was not found to increase the chance of orbital recurrence, metastatic disease, or death compared with primary enucleation [29].

Berry et al. studied the risk of metastasis and orbital recurrence in advanced RB eyes treated with systemic chemoreduction versus primary enucleation and reported for both incidence of orbital recurrence and metastatic disease <1%. Globe salvage therapy with systemic chemoreduction and subsequent enucleation for poor response does not increase the risk of metastatic disease or orbital recurrence [59].

 

 

Primary intravenous chemotherapy (IVC) in combination with further treatment

 

For unilateral RB, IAC provided significantly superior globe salvage rate compared to IVC for group D eyes and provided significantly superior control for solid tumor, subretinal seeds, and vitreous seeds [60]. For particular locations such as macular RBs IVC plus adjuvant foveal-sparing thermotherapy provides tumor control of 83% by 4 years, and this appears to be better than IVC alone, which provides control of 65% by 4 years [61]. Tumors most destined for recurrence are small tumors [61]. The management of group E RB can be particularly difficult. For unilateral E RB or when the opposite eye has minimal tumor, enucleation with salvation of the partner eye with potential vision should be sought [44, 62]. Options for management in these cases include IVC, EBR, or a combination IVC and EBR [63]. Importantly group E RB managed with IVC and EBR showed significantly less need for enucleation or therapeutic radiotherapy than eyes treated with IVC alone. These findings merit further study [63, 64]. Furthermore, chemotherapy in group E eyes before enucleation performed more than 3 months after diagnosis possibly masks evidence of extraocular extension, increasing the risk of metastatic death from reduced surveillance of high-risk disease [65]. Currently, EBR is rarely performed, as retrospective analysis has shown a higher rate of secondary CNS malignancies after this treatment.

 

 

Intra-arterial chemotherapy as secondary therapy

 

IAC, as mentioned earlier, was initially developed as first-line therapy for the treatment of unilateral advanced RB but is also used as second-line therapy for relapsed RB [15, 27]. In a retrospective study, globe salvage was achieved in 72% of primary-treated patients and in 62% of secondary-treated patients [24]. Further studies have demonstrated that globe salvage rate is higher (93.3%) when IAC is used as primary therapy than when it is used as secondary therapy (79%), which is consistent with previous reports [32, 66]. IAC globe salvage is lower in patients previously treated with IVC, especially previous failed treatment as most RBs showed an initial response to intravenous chemotherapy but eventually progressed [32, 66]. This could be partially explained by multidrug resistance observed in some studies [67, 68].

Advanced D or E eyes treated with primary IVC had no metastasis in a 2 year follow up period. However, in these cases, recurrence made the secondary use of IAC necessary and complete control was achieved in 80% of the recurrent solid tumor cohort, in 55% with recurrent subretinal seeds, and in 43% with recurrent vitreous seeds [27].

Another study suggests that IAC, whether primary or secondary, is effective and fairly safe for the management of advanced RB in infants aged < 3 months; however, adverse events related to intra-arterial injection were reported and the visual outcomes cannot be neglected and require further investigation [69].

A report of the efficacy of a second course of IAC noted a 24-month ocular survival at 83% and progression-free survival at 26% [70]. Consequently, such eyes are still ‘‘at risk’’ for tumor progression even after the second round of IAC as 74% of patients demonstrated vitreous seeding requiring treatment. In other series, tumor control was achieved in 9 of 12 eyes (75%), with tumor progression requiring treatment in 5 (42%) using intravitreal melphalan for vitreous seeding and cryotherapy with or without plaque radiotherapy for subretinal seeding. Such cases warrant a close monitoring and focal therapy when appropriate [42, 71, 72].

 

 

Comparison between intra-arterial chemotherapy and intravenous chemotherapy

 

IVC was compared to IAC for unilateral RB and IAC was found to provide significantly superior globe salvage for group D eyes as well as superior control for solid tumor, subretinal seeds, and vitreous seeds [60].

Similar results have been reported with globe retention at 100% when IAC was the first-line therapy compared with 57% in the IVC group [73]. No metastases or deaths occurred in either group. IAC was associated with fewer side effects, faster response rates, fewer relapses, and better visual acuity than IVC treatment. There were differences between the two groups with respect to the consolidating treatments used and differences in the time frame due to availability of IVC. Despite this, the results suggest that eyes treated with first line IAC will have shorter treatment period, better ocular survival, and visual acuity than first line-treatment with IVC [73].

 

 

Comparison between intra-arterial chemotherapy and enucleation

 

Eyes treated initially with IAC have been shown to have ocular event-free survival rates at 2 years of greater than 80% despite their advanced stage [32]. These findings have been replicated at multiple centers, demonstrating that IAC as a primary therapy or with intravenous chemotherapy as a bridge may achieve globe salvage and tumor control in a high percentage of eyes with group D and E RB [24, 32, 74, 75]. IAC also has been shown to be useful in the treatment of vitreous seeds and to prevent development of new intraocular tumors, suggesting that it may treat ophthalmoscopically undetectable tumors present at initial diagnosis [19, 76].

While the multiple benefits and favorable risk profile of IAC have been documented in the literature, the incidence of orbital recurrence in patients treated with this modality was described in a single-institution retrospective study of advanced intraocular RB; there were more orbital recurrences in the group primarily treated with enucleation than with IAC. On the other hand, ophthalmic artery intra-arterial chemotherapy for advanced intraocular RB was not found to increase the chance of orbital recurrence, metastatic disease, or death compared with primary enucleation [58].

 

 

Focal therapy options

 

The use of thermal energy in conjunction with systemic chemotherapy has been shown to be synergistic [77-79]. In a transgenic murine RB cell line, the cytocidal interaction of heat and carboplatin were found to be superior to each of the monotherapies [80]. Laser treatment alone for small tumors, and combined with cryotherapy and chemotherapy for larger tumors, is effective in the treatment of RB. Complications of focal therapy can most often be avoided by using the minimal effective laser power.

 

 

Intravitreal chemotherapy

 

The use of IViC has reported to have a salvage rate of 51% [81]; however, unstable chromosomes in RB cells have high mitotic index, causing them to infiltrate rapidly in neighboring accessible tissue (vitreous, subretinal space, anterior chamber) which has been a concern [82, 83]. Intravitreal chemotherapy gained greater acceptance when an anti-reflux technique with needle track sterilization was introduced so there is minimal chance of extraocular spreading of tumor cells and very promising outcomes in globe salvage with advanced disease [84].

So far intravitreal therapy has been extensively tested and compared to other therapies. Intravitreal melphalan appears to be promising for group D RB with resistant vitreous seeds [85].

The management of vitreous seeds and subretinal seeds is problematic with respect to final prognosis and eye salvage [86, 87]. Salvage rates vary from 100% for group A eyes, 96% for group B eyes, 90% for group C eyes, and 48% for group D eyes [44]. The lower rate for group D eyes is usually due to active vitreous seeding [47]. Chemotherapy cannot reach therapeutic vitreous levels making treatment of vitreous seeds a challenge [88]. Intravitreal thiotepa, melphalan, and methotrexate have been used for RB [89, 90]. Melphalan was the most efficient drug among 12 tested in vitro and structurally nontoxic to the retina in vivo [90]. Therefore, intravitreal melphalan has emerged as a treatment for active vitreous seeds with considerable success [71, 91].

Treating vitreous seed clouds with IAC and intravitreous/periocular chemotherapy has resulted in a shorter regression time compared with IAC alone, fewer recurrences, fewer retreatments, and fewer enucleations [92]. The salvage rate for Group D eyes was 75% with intravitreal melphalan reaching ocular salvage in 42% over 6 years; systemic chemoreduction combined with intravitreal melphalan for seeding demonstrated a high overall salvage rate for Group D eyes in this cohort [39].

Intravitreal chemotherapy-assisted endoresection appears to be a safe and effective globe-salvaging method for refractory D RBs and may be a promising alternative to enucleation for those unresponsive to standard therapies, especially for monocular RB patients [93]. This surgical technique has to further be tested to see if extraocular metastasis occurs.

 

 

Sub-Tenon's chemotherapy

 

Studies have also more convenient routes of local chemotherapy such as sub-tenon’s injections as an alternative to intravitreal administration. Experimental work has already demonstrated that periocular carboplatin quickly enters the vitreous reaching high concentrations, recommended for the management of Group C and Group D eyes [94]. Carboplatin sub-tenon treatment for RB with secondary vitreous seeds is safe and can salvage 30.3% of eyes without EBR with no statistically significant difference to eyes treated with intravitreal melphalan [95].

 

 

External beam radiotherapy (EBR)

 

EBR since 1954 was first eye salvage therapy for advanced intraocular RB and was used extensively as primary treatment [96]. Although quite effective in controlling tumor growth, enucleation was ultimately needed due to radiation complications. Further, there was also a lifelong increased risk of secondary malignancies in children with genetic predisposition (e.g. germline RB1 gene mutation) and cosmetic problems from orbital bone growth retardation [97, 98]. Consequently, EBR was successively replaced by systemic chemotherapy combined with focal therapy and reserved as a salvage option for treatment failures [99]. However, the presence of vitreous seeds, stage migration during the course of chemotherapy, as well as good vision in the other eye may not justify the risk profile of EBR [5, 100, 101].

Given the high radiation sensitivity of RB, proton beam therapy (PBT) could be a valuable rescue therapy to preserve a RB eye with a visual function in otherwise treatment resistant RB cases. PBT has distinct advantages over photon radiation therapy reducing collateral tissue damage but is not itself complications free [102, 103]. An eye preservation rate up to 90% has been demonstrated with a fairly good survival rate and low in-field recurrences even after 8 years of follow-up [104]. A lifelong risk of subsequent primary tumors cannot be excluded thus consistent monitoring is warranted.

 

 

Secondary enucleation with high dose intravenous chemotherapy

 

Eyes that fail chemoreduction therapy eventually require EBR and/or enucleation. Compared with Reese-Ellsworth classification (Table 2) groups I to IV, group V disease requires more often external beam radiotherapy and enucleation after chemoreduction [5, 105]. Chemoreduction failure was defined as unresponsive or recurrent disease. Unresponsive disease in 10 of 105 (9.5%) study eyes was treated with enucleation after the second chemoreduction treatment [5]. Careful handling and interpretation of histopathology of eyes with RB is necessary to assign metastatic risk [106]. Eyes with less than 3 mm choroidal invasion and without prelaminar/laminar optic nerve invasion show no recurrence and may warrant no adjuvant chemotherapy [106]. In contrast, greater than 3 mm peripapillary choroidal invasion with 1.5 mm or greater of post laminar optic nerve invasion have the poorest outcomes, supporting the need for a more intensive adjuvant chemotherapy regimen. Strict criteria for adjuvant therapy may improve outcomes of children who undergo enucleation at diagnosis and may avoid unnecessary adjuvant chemotherapy for those who are not at risk for recurrence [106].

 

Table 2. Reese-Ellsworth classification [105]

 

 

Treatment failure rate

 

Ravindran et al. reported in a big review study an enucleation rate of 32% after intra-arterial chemotherapy [107]. The rates of new tumor development/recurrences following all types of treatment have ranged from 6% to 45% [108-112]. The rate of tumor recurrence following IVC without focal treatment ranges from 35% to 45%, while the rate following chemoreduction with consolidative treatment has been lower at 17% to 18% [61, 111]. Recurrence after IAC, with consolidation, has been reported as low as 0% to 23% [16, 24, 32]. The results suggest that IAC has advantages in eyes with advanced disease, including groups D, IV, and V, but not in groups A-C, E, and I-III eyes. And although the studies cannot be directly compared due to different study design the overall success rate is higher in IAC compared to IVC and there seems to be no significant difference with respect to tumor recurrence and metastasis disease [50].

 

 

International Intraocular Retinoblastoma Classification and Intraocular Classification of Retinoblastoma Classification

 

When intravenous chemotherapy for intraocular RB was introduced in the 1990s, a new precise classification scheme for staging RB was adopted, the International Intraocular RB Classification (IIRC) [47]. Subsequently it has been modified as the Intraocular Classification of RB (ICRB) with refined definitions of the advanced groups D and E and has been found to successfully predict the outcome of intravenous chemotherapy (Table 3) [44].

 

Table 3. Classification system for Intraocular Retinoblastoma [44,47]

 

 

Risk factors of unfavorable outcome

 

Chemoreduction failure is defined as a treatment unresponsive or recurrent RB. Risk factors associated with low tumor response and increased need for enucleation include older age at diagnosis, greater tumor thickness, presence of vitreous seeds, subretinal fluid at baseline and retinal tumor recurrence after chemoreduction [5]. Low-income level was found to be an independent, significant predictive factor for advanced disease [1]. Extraocular/transscleral tumor extension with orbital tissue involvement as well as presence of tumor at the optic nerve represent high-risk features for recurrence [1, 113, 114]. Although anterior segment involvement has been considered a high-risk feature, recent retrospective data suggest no further treatment necessity for this [115]. Specifically, compared with high-income countries (HIC), RB patients from low-income countries had a 10.3-fold risk and 9.3-fold mortality and a 2.2-fold risk and 1.6-fold risk for local treatment failure respectively [116]. Histopathologic analysis of enucleated eyes from low to medium income countries (LMIC) and upper to middle income countries (UMIC) revealed that patients with high-risk histopathology (pT3 and pT4) fared worse in comparison with HIC [116].

 

 

Therapy failure in retinoblastoma

 

In HIC, most children with limited intraocular disease are cured by a combined chemotherapy and focal treatment regime [24] or enucleation for orbital and metastatic disease followed by pathology risk–adapted adjuvant chemotherapy [117-121]. In contrast, in LMICs, RB presents in an already advanced disease stage with ocular salvage rarely possible and patients suffering on incurable metastatic disease [122-127]. Finally, a considerable proportion of patients refuse enucleation and abandon treatment [128]. Abandonment of therapy is defined as the failure to return to treatment for 4 weeks or more [129]. Hence, most patients die at this stage if they do not undergo enucleation [126, 130]. This challenge requires multilevel approaches (both on cultural and medicolegal levels), seldomly available in LMICs, critically affecting survival [122, 130]. The Central American Association of Pediatric Oncology reported the results of its first RB study (AHOPCA I), on very advanced disease presentation with a 5-year overall survival (OS) rate of 0.48±0.04 [126]. In this cohort, 45 of 171 patients had unilateral Reese-Ellsworth classification group I disease, and 16% abandoned therapy. Over subsequent years, the participating institutions embarked on a regionwide initiative (AHOPCA II) aimed at enhancing diagnosis and establishing a risk-adapted therapy that involved group discussion of all patients. Herein, we reviewed the results of a risk-adapted approach for patients with advanced intraocular unilateral RB. The standard approach of upfront enucleation and adjuvant therapy adjusted to pathology risk factors was modified for patients who presented with buphthalmus and for those who refused upfront enucleation or were at risk of abandonment to include neoadjuvant chemotherapy and delayed enucleation. Enucleation also carries a risk of rupture, causing contamination of the orbit and increasing the risk of relapse [123, 130-134].

 

 

Therapy failure in retinoblastoma: Group D/E tumors

 

With external beam irradiation the treatment mainstay just 25 years ago clinicians worldwide began using systemic chemotherapy instead for intraocular RB. Success rates vary widely depending mainly on disease stage for A to C stage eyes. D stage eyes managed initially with systemic chemotherapy also vary but most fall in the 25–30% range [135-138]. More advanced E stage eyes are mostly enucleated [138, 139]. Direct comparison of salvage rates described in the literature is difficult since different versions for classification are in use.

According to the Reese-Ellsworth classification, globe salvage with chemoreduction is achieved in 93% of group I, 88% of group II, 83% of group III, 62% of group IV, and 43% of group V cases [44]. According to the more recent International Classification of Retinoblastoma, globe salvage with IVC is achieved in 100% of group A, 93% of group B, 90% of group C, and 47% of group D cases [44]. Group E was not included in these analyses because most ocular oncologists have avoided use of IVC for group E RB because of the anticipated high risk of failure. Furthermore, the previously used external beam radiation, although very effective in reducing tumor size salvaging globes and vison has been progressively abandoned due to increased rate of secondary metastasis rate, ocular complications as well as developmental abnormalities of soft tissue and bony orbit/scall in children [36, 140]. Thus, most eyes with group E RB have been managed with enucleation.

However, in the recent years direct ophthalmic artery infusion, also called ophthalmic artery chemosurgery (IAC), a technique in which chemotherapy is locally delivered to the eye by a micro-catheter and advanced from the femoral artery into the ophthalmic artery has given an new wind in the treatment of simple but also advanced cases mostly avoiding the need for enucleation or external beam irradiation or systemic chemotherapy [34]. Furthermore, the metastatic rate drops down to 1.5%, even lower than the 5% described in the current literature [34]. Shields and colleagues have also shown successful treatment results in advanced eyes. With a mean follow-up of 19 months in naïve patients the ocular survival is 94% in D eyes and 36% in E eyes [24]. In addition, Tuncer and colleagues recently reported a 62.9% 2-year ocular survival in D eyes also treated with IAC [25]. Similarly, Kiratli et al. demonstrated that group D patients, primary IAC achieved ocular survival in 76.7% of eyes, and less need for local consolidation treatments, whereas following primary IVC ocular survival was only 43.2% [51].

Munier et al. on the other hand demonstrated globe retention at 100% with IAC as first-line therapy and intravitreal chemotherapy (IViC) in 2008 with primary enucleation dropping from 56% to 5% [73]. Shields et al. also tried intravenous chemoreduction IVC followed by IAC and found that advanced RB (D and E) in which enucleation would be the alternative, primary systemic (IVC) followed by secondary focal IAC provides globe salvage in 57% of the eyes and with no metastatic event [27].

Berry et al. reported the rates of extraocular relapse for patients with advanced Group D and E RB eyes treated and followed over an approximate 20-year [59]. The eyes were grouped according to whether primary treatment was enucleation versus attempted salvage with systemic chemoreduction, and the main outcomes assessed were whether the incidence of orbital recurrence, metastatic disease, and death varied by initial treatment modality. Overall, the incidence of these outcome measures was 1% or less in both groups, which is lower than what has been reported previously in similar retrospective analyses [29, 58, 65, 141].

Finally, the option of subtenon chemotherapy has also been explored with carboplatin plus chemotherapy with carboplatin/etoposide/vincristine with only partial effectiveness in Group D and unacceptable ocular toxicities [142].

 

 

Therapy failure in retinoblastoma: optic nerve invasion

 

Overt metastatic spread of the tumor beyond the globe, such as post laminar optic nerve invasion, indicates a poorer prognosis [143]. Tumor in orbital tissues and at the cut end of the optic nerve following enucleation represent high-risk features for recurrence [113, 114, 144]. In such cases intensive/aggressive intervention with chemotherapy and often radiation is required [113, 145, 146]. In such advanced cases enucleation is a treatment option, with a cure rate of more than 90% [114, 147-150]. Chevez-Barrios et al. have further defined high risk criteria and need for adjunct therapy [106]. On the basis of their results, less than 3 mm choroidal and any prelaminar/laminar optic nerve invasion show no recurrence and may warrant no adjuvant chemotherapy [106]. However, the combination of extensive involvement of both choroid and post laminar optic nerve invasion (i.e. 3 mm invasion in the peripapillary posterior choroid in combination with 1.5 mm post laminar optic nerve invasion) represent the highest risk of recurrence and have a poorer prognosis [106, 151-153]. In such cases a higher degree of vigilance and a more aggressive adjuvant treatment is warranted in this scenario. Furthermore, the overall prognosis of unilateral RB patients with intracranial segment of retrobulbar optic nerve invasion was poor [106]. In such cases chemotherapy and surgical treatment were necessary, but more attention should be paid to radiotherapy and intrathecal injection for improving prognosis [144].

Finally, bilateral RB cases remain challenging for ophthalmologists and pediatric oncologists despite new therapeutic strategies for eye preservation. Bernal-Diaz evaluated treatment outcomes in 88 consecutive patients (96 eyes) with bilateral RB who underwent eye salvage treatment at a single-center prior to chemotherapy with eye preservation, long-term tumor control, and functional visual acuity maintained in many children and adolescent RB survivors [154]. Seventy-eight eyes (81%) were salvaged, seven patients (8%) required brachytherapy and 34 patients (39%) underwent EBRT. Thirty-three of 78 preserved eyes (42%) achieved normal visual acuity. The overall survival rate was 86% after a mean follow-up of 10 years [154]. Weng et al. reported similar results with globe salvage rate at 73% for group D eyes and 44% for group E eyes with vision-preserving therapy [64].

 

 

Conclusion

 

RB is a mostly curable cancer if diagnosed and treated early. However, proper management of RB is complex. Eye and vision preservation is not guaranteed at all. The overall goal of RB management is to prevent the progression of the disease and the development of higher-risk pathologic factors that increase the risk of metastatic spread. Advances in treatment protocols and implementation of new technology have enabled a shift in treatment emphasis from enucleation in organ preservation. As each case is unique, treatment regimens must be carefully customized for varying disease presentations, available equipment, and regional culture or traditions.

Furthermore, toxicity of chemotherapy regardless of delivery routes is still a limiting factor, indicating the need for novel targeted drugs. Molecularly targeted drugs may be more effective and less toxic, and the effects of drugs would be potentiated by local delivery. This may not only save lives, but may also spare eye(s), some sight, and the quality of life of RB survivors.

 

 

Acknowledgements

 

Conceptualization, A.C. and V.K.; methodology, A.C., N.B., C.S., J.S. and V.K.; software, A.C, L.M.H. and M.R.; validation, A.C., L.M.H., M.R. and V.K.; formal analysis, N.B. and V.K.; investigation, L.K.I. and N.B.; resources, A.C and V.K.; data curation, N.B., L.K.I. and P.C.R.; writing—original draft preparation, A.C, J.S. and V.K.; writing—review and editing, N.B., C.S., L.M.H. and M.R.; visualization, N.B., C.S., L.M.H. and M.R.; supervision, A.C. and V.K.; project administration, A.C. and V.K.; funding acquisition, A.C. and V.K.. All authors have read and agreed to the published version of the manuscript.

 

 

Disclosure Statement

 

The authors declare that no conflicts of interest exist.

 

 

References

 

1 Global Retinoblastoma Study Group, Fabian ID, Abdallah E, Abdullahi SU, Abdulqader RA, Adamou Boubacar S, Ademola-Popoola DS, Adio A, Afshar AR, Aggarwal P, Aghaji AE, Ahmad A, Akib MNR, Al Harby L, Al Ani MH, Alakbarova A, Portabella SA, Al-Badri SAF, Alcasabas APA, Al-Dahmash SA, et al.: Global Retinoblastoma Presentation and Analysis by National Income Level. JAMA Oncol 2020;6:685-695.
https://doi.org/10.1001/jamaoncol.2019.6716

 

2 Scelfo C, Francis JH, Khetan V, Jenkins T, Marr B, Abramson DH, Shields CL, Pe'er J, Munier F, Berry J, Harbour JW, Yarovoy A, Lucena E, Murray TG, Bhagia P, Paysse E, Tuncer S, Chantada GL, Moll AC, Ushakova T, et al.: An international survey of classification and treatment choices for group D retinoblastoma. Int J Ophthalmol 2017;10:961-967.

 

3 Abramson DH, Daniels AB, Marr BP, Francis JH, Brodie SE, Dunkel IJ, Gobin YP: Intra-Arterial Chemotherapy (Ophthalmic Artery Chemosurgery) for Group D Retinoblastoma. PLoS One 2016;11:e0146582.
https://doi.org/10.1371/journal.pone.0146582

 

4 Shields JA, Shields CL, Sivalingam V: Decreasing frequency of enucleation in patients with retinoblastoma. Am J Ophthalmol 1989;108:185-188.
https://doi.org/10.1016/0002-9394(89)90015-9

 

5 Gunduz K, Gunalp I, Yalcindag N, Unal E, Tacyildiz N, Erden E, Geyik PO: Causes of chemoreduction failure in retinoblastoma and analysis of associated factors leading to eventual treatment with external beam radiotherapy and enucleation. Ophthalmology 2004;111:1917-1924.
https://doi.org/10.1016/j.ophtha.2004.04.016

 

6 Huang D, Zhang Y, Zhang W, Wang Y, Zhang P, Hong L, Zhou Y, Han T, Zhi T: Study on clinical therapeutic effect including symptoms, eye preservation rate, and follow-up of 684 children with retinoblastoma. Eur J Ophthalmol 2013;23:532-538.
https://doi.org/10.5301/ejo.5000245

 

7 Dimaras H, Kimani K, Dimba EA, Gronsdahl P, White A, Chan HS, Gallie BL: Retinoblastoma. Lancet 2012;379:1436-1446.
https://doi.org/10.1016/S0140-6736(11)61137-9

 

8 Saunders BM DG, Kingston JE.: Retinoblastoma in Great Britain 1969-1980. Br J Ophthalmol 1988;72:576-583.
https://doi.org/10.1136/bjo.72.8.576

 

9 Stiller CA, Parkin DM: Geographic and ethnic variations in the incidence of childhood cancer. Br Med Bull 1996;52:682-703.
https://doi.org/10.1093/oxfordjournals.bmb.a011577

 

10 Shields CL, Shields JA, Shah P: Retinoblastoma in older children. Ophthalmology 1991;98:395-399.
https://doi.org/10.1016/S0161-6420(91)32283-8

 

11 Tamboli D, Topham A, Singh N, Singh AD: Retinoblastoma: A SEER Dataset Evaluation for Treatment Patterns, Survival, and Second Malignant Neoplasms. Am J Ophthalmol 2015;160:953-958.
https://doi.org/10.1016/j.ajo.2015.07.037

 

12 Lu JE, Francis JH, Dunkel IJ, Shields CL, Yu MD, Berry JL, Kogachi K, Skalet AH, Miller AK, Santapuram PR, Daniels AB, Abramson DH: Metastases and death rates after primary enucleation of unilateral retinoblastoma in the USA 2007-2017. Br J Ophthalmol 2019;103:1272-1277.
https://doi.org/10.1136/bjophthalmol-2018-312915

 

13 MacCarthy A, Birch JM, Draper GJ, Hungerford JL, Kingston JE, Kroll ME, Stiller CA, Vincent TJ, Murphy MF: Retinoblastoma: treatment and survival in Great Britain 1963 to 2002. Br J Ophthalmol 2009;93:38-39.
https://doi.org/10.1136/bjo.2008.139626

 

14 Siegel RL, Miller KD, Jemal A: Cancer statistics, 2019. CA Cancer J Clin 2019;69:7-34.
https://doi.org/10.3322/caac.21551

 

15 Yamane T, Kaneko A, Mohri M: The technique of ophthalmic arterial infusion therapy for patients with intraocular retinoblastoma. Int J Clin Oncol 2004;9:69-73.
https://doi.org/10.1007/s10147-004-0392-6

 

16 Abramson DH, Dunkel IJ, Brodie SE, Kim JW, Gobin YP: A phase I/II study of direct intraarterial (ophthalmic artery) chemotherapy with melphalan for intraocular retinoblastoma initial results. Ophthalmology 2008;115:1398-1404, 1404 e1391.
https://doi.org/10.1016/j.ophtha.2007.12.014

 

17 Munier FL, Beck-Popovic M, Chantada GL, Cobrinik D, Kivela TT, Lohmann D, Maeder P, Moll AC, Carcaboso AM, Moulin A, Schaiquevich P, Bergin C, Dyson PJ, Houghton S, Puccinelli F, Vial Y, Gaillard MC, Stathopoulos C: Conservative management of retinoblastoma: Challenging orthodoxy without compromising the state of metastatic grace. "Alive, with good vision and no comorbidity". Prog Retin Eye Res 2019;73:100764.
https://doi.org/10.1016/j.preteyeres.2019.05.005

 

18 Seregard S, Kock E, af Trampe E: Intravitreal chemotherapy for recurrent retinoblastoma in an only eye. Br J Ophthalmol 1995;79:194-195.
https://doi.org/10.1136/bjo.79.2.194

 

19 Abramson DH, Marr BP, Dunkel IJ, Brodie S, Zabor EC, Driscoll SJ, Gobin YP: Intra-arterial chemotherapy for retinoblastoma in eyes with vitreous and/or subretinal seeding: 2-year results. Br J Ophthalmol 2012;96:499-502.
https://doi.org/10.1136/bjophthalmol-2011-300498

 

20 Shields CL, Bianciotto CG, Jabbour P, Ramasubramanian A, Lally SE, Griffin GC, Rosenwasser R, Shields JA: Intra-arterial chemotherapy for retinoblastoma: report No. 1, control of retinal tumors, subretinal seeds, and vitreous seeds. Arch Ophthalmol 2011;129:1399-1406.
https://doi.org/10.1001/archophthalmol.2011.150

 

21 Kaneko A, Takayama J, Matsuoka H, Inomata M, Moori M: Chemo-thermotherapy was successful in two cases of recurrence of intraocular retinoblastoma after irradiation. Jpn J Clin Ophthalmol 1990;44:289-292.

 

22 Makoto M: The technique of selective ophthalmic arterial in fusion for conservative treatment of recurrent retinoblastoma. J Keio Med Sco 1993:679-687.

 

23 Ueda M, Tanabe J, Inomata M, Kaneko A, Kimura T: [Study on conservative treatment of retinoblastoma--effect of intravitreal injection of melphalan on the rabbit retina]. Nippon Ganka Gakkai Zasshi 1995;99:1230-1235.

 

24 Shields CL, Manjandavida FP, Lally SE, Pieretti G, Arepalli SA, Caywood EH, Jabbour P, Shields JA: Intra-arterial chemotherapy for retinoblastoma in 70 eyes: outcomes based on the international classification of retinoblastoma. Ophthalmology 2014;121:1453-1460.
https://doi.org/10.1016/j.ophtha.2014.01.026

 

25 Tuncer S, Sencer S, Kebudi R, Tanyildiz B, Cebeci Z, Aydin K: Superselective intra-arterial chemotherapy in the primary management of advanced intra-ocular retinoblastoma: first 4-year experience from a single institution in Turkey. Acta Ophthalmol 2016;94:e644-e651.
https://doi.org/10.1111/aos.13077

 

26 Chen M, Jiang H, Zhang J, Shen G, Jiang Y, Li H, Liu Z: Outcome of intra-arterial chemotherapy for retinoblastoma and its influencing factors: a retrospective study. Acta Ophthalmol 2017;95:613-618.
https://doi.org/10.1111/aos.13333

 

27 Shields CL, Kaliki S, Al-Dahmash S, Rojanaporn D, Leahey A, Griffin G, Jabbour P, Shields JA: Management of advanced retinoblastoma with intravenous chemotherapy then intra-arterial chemotherapy as alternative to enucleation. Retina 2013;33:2103-2109.
https://doi.org/10.1097/IAE.0b013e318295f783

 

28 Liu CC, Mohmood A, Hamzah N, Lau JH, Khaliddin N, Rahmat J: Intra-arterial chemotherapy for retinoblastoma: Our first three-and-a-half years' experience in Malaysia. PLoS One 2020;15:e0232249.
https://doi.org/10.1371/journal.pone.0232249

 

29 Yannuzzi NA, Francis JH, Marr BP, Belinsky I, Dunkel IJ, Gobin YP, Abramson DH: Enucleation vs Ophthalmic Artery Chemosurgery for Advanced Intraocular Retinoblastoma: A Retrospective Analysis. JAMA Ophthalmol 2015;133:1062-1066.
https://doi.org/10.1001/jamaophthalmol.2015.2243

 

30 Shields CL, Lally SE, Leahey AM, Jabbour PM, Caywood EH, Schwendeman R, Shields JA: Targeted retinoblastoma management: when to use intravenous, intra-arterial, periocular, and intravitreal chemotherapy. Curr Opin Ophthalmol 2014;25:374-385.
https://doi.org/10.1097/ICU.0000000000000091

 

31 Shields CL, Bianciotto CG, Jabbour P, Griffin GC, Ramasubramanian A, Rosenwasser R, Shields JA: Intra-arterial chemotherapy for retinoblastoma: report No. 2, treatment complications. Arch Ophthalmol 2011;129:1407-1415.
https://doi.org/10.1001/archophthalmol.2011.151

 

32 Gobin YP, Dunkel IJ, Marr BP, Brodie SE, Abramson DH: Intra-arterial chemotherapy for the management of retinoblastoma: four-year experience. Arch Ophthalmol 2011;129:732-737.
https://doi.org/10.1001/archophthalmol.2011.5

 

33 Abramson DH, Fabius AW, Issa R, Francis JH, Marr BP, Dunkel IJ, Gobin YP: Advanced Unilateral Retinoblastoma: The Impact of Ophthalmic Artery Chemosurgery on Enucleation Rate and Patient Survival at MSKCC. PLoS One 2015;10:e0145436.
https://doi.org/10.1371/journal.pone.0145436

 

34 Abramson DH, Fabius AW, Francis JH, Marr BP, Dunkel IJ, Brodie SE, Escuder A, Gobin YP: Ophthalmic artery chemosurgery for eyes with advanced retinoblastoma. Ophthalmic Genet 2017;38:16-21.
https://doi.org/10.1080/13816810.2016.1244695

 

35 Shields CL, Alset AE, Say EA, Caywood E, Jabbour P, Shields JA: Retinoblastoma Control With Primary Intra-arterial Chemotherapy: Outcomes Before and During the Intravitreal Chemotherapy Era. J Pediatr Ophthalmol Strabismus 2016;53:275-284.
https://doi.org/10.3928/01913913-20160719-04

 

36 Shields CL, Fulco EM, Arias JD, Alarcon C, Pellegrini M, Rishi P, Kaliki S, Bianciotto CG, Shields JA: Retinoblastoma frontiers with intravenous, intra-arterial, periocular, and intravitreal chemotherapy. Eye (Lond) 2013;27:253-264.
https://doi.org/10.1038/eye.2012.175

 

37 Francis JH, Abramson DH, Gaillard MC, Marr BP, Beck-Popovic M, Munier FL: The classification of vitreous seeds in retinoblastoma and response to intravitreal melphalan. Ophthalmology 2015;122:1173-1179.
https://doi.org/10.1016/j.ophtha.2015.01.017

 

38 Francis JH, Marr BP, Abramson DH: Classification of Vitreous Seeds in Retinoblastoma: Correlations with Patient, Tumor, and Treatment Characteristics. Ophthalmology 2016;123:1601-1605.
https://doi.org/10.1016/j.ophtha.2016.02.036

 

39 Berry JL, Shah S, Bechtold M, Zolfaghari E, Jubran R, Kim JW: Long-term outcomes of Group D retinoblastoma eyes during the intravitreal melphalan era. Pediatr Blood Cancer 2017; DOI: 10.1002/pbc.26696.
https://doi.org/10.1002/pbc.26696

 

40 Ghassemi F, Shields CL, Ghadimi H, Khodabandeh A, Roohipoor R: Combined intravitreal melphalan and topotecan for refractory or recurrent vitreous seeding from retinoblastoma. JAMA Ophthalmol 2014;132:936-941.
https://doi.org/10.1001/jamaophthalmol.2014.414

 

41 Shields CL, Douglass AM, Beggache M, Say EA, Shields JA: INTRAVITREOUS CHEMOTHERAPY FOR ACTIVE VITREOUS SEEDING FROM RETINOBLASTOMA: Outcomes After 192 Consecutive Injections. The 2015 Howard Naquin Lecture. Retina 2016;36:1184-1190.
https://doi.org/10.1097/IAE.0000000000000903

 

42 Shields CL, Manjandavida FP, Arepalli S, Kaliki S, Lally SE, Shields JA: Intravitreal melphalan for persistent or recurrent retinoblastoma vitreous seeds: preliminary results. JAMA Ophthalmol 2014;132:319-325.
https://doi.org/10.1001/jamaophthalmol.2013.7666

 

43 Liang TY, Zhu XY, Hua XM, Ji XD, Zhao PQ: Combined intra-arterial chemotherapy and intravitreal melphalan for the treatment of advanced unilateral retinoblastoma. In J Ophthalmol 2020;13:257-262.
https://doi.org/10.18240/ijo.2020.02.09

 

44 Shields CL, Mashayekhi A, Au AK, Czyz C, Leahey A, Meadows AT, Shields JA: The International Classification of Retinoblastoma predicts chemoreduction success. Ophthalmology 2006;113:2276-2280.
https://doi.org/10.1016/j.ophtha.2006.06.018

 

45 Shin JY, Kim JH, Yu YS, Khwarg SI, Choung HK, Shin HY, Ahn HS: Eye-preserving therapy in retinoblastoma: prolonged primary chemotherapy alone or combined with local therapy. Korean J Ophthalmol 2010;24:219-224.
https://doi.org/10.3341/kjo.2010.24.4.219

 

46 Yousef YA, Nazzal RM, Khalil MB, Deebajah R, Mehyar M, Hajja S, Mohammad M, Al Jabary R, Jaradat I, Sultan I, Al Nawaiseh I: Management outcome(s) in eyes with retinoblastoma previously inadequately treated with systemic chemotherapy alone without focal therapy. Oman J Ophthalmol 2017;10:70-75.

 

47 Linn Murphree A: Intraocular retinoblastoma: the case for a new group classification. Ophthalmol Clin North Am 2005;18:41-53, viii.
https://doi.org/10.1016/j.ohc.2004.11.003

 

48 Kaliki S, Shields CL, Shah SU, Eagle RC, Jr., Shields JA, Leahey A: Postenucleation adjuvant chemotherapy with vincristine, etoposide, and carboplatin for the treatment of high-risk retinoblastoma. Arch Ophthalmol 2011;129:1422-1427.
https://doi.org/10.1001/archophthalmol.2011.289

 

49 Mendoza PR, Grossniklaus HE: Therapeutic Options for Retinoblastoma. Cancer Control 2016;23:99-109.
https://doi.org/10.1177/107327481602300203

 

50 Chen Q, Zhang B, Dong Y, Mo X, Zhang L, Huang W, Jiang H, Xia J, Zhang S: Comparison between intravenous chemotherapy and intra-arterial chemotherapy for retinoblastoma: a meta-analysis. BMC Cancer 2018;18:486.
https://doi.org/10.1186/s12885-018-4406-6

 

51 Kiratli H, Koc I, Inam O, Varan A, Akyuz C: Retrospective analysis of primarily treated group D retinoblastoma. Graefes Arch Clin Exp Ophthalmol 2018;256:2225-2231.
https://doi.org/10.1007/s00417-018-4051-4

 

52 Abramson DH: Retinoblastoma: saving life with vision. Annu Rev Med 2014;65:171-184.
https://doi.org/10.1146/annurev-med-061312-123455

 

53 Albert DM: Historic review of retinoblastoma. Ophthalmology 1987;94:654-662.
https://doi.org/10.1016/S0161-6420(87)33407-4

 

54 Dunphy EB: The Story of Retinoblastoma. Trans Am Acad Ophthalmol Otolaryngol 1964;68:249-264.

 

55 Reese AB, Hyman GA, Tapley ND, Forrest AW: The treatment of retinoblastoma by x-ray and triethylene melamine. AMA Arch Ophthalmol 1958;60:897-906.
https://doi.org/10.1001/archopht.1958.00940080917010

 

56 Ellsworth RM: The practical management of retinoblastoma. Trans Am Ophthalmol Soc 1969;67:462-534.

 

57 Broaddus E, Topham A, Singh AD: Survival with retinoblastoma in the USA: 1975-2004. Br J Ophthalmol 2009;93:24-27.
https://doi.org/10.1136/bjo.2008.143842

 

58 Yannuzzi NA, Francis JH, Abramson DH: Incidence of Orbital Recurrence After Enucleation or Ophthalmic Artery Chemosurgery for Advanced Intraocular Retinoblastoma--Reply. JAMA Ophthalmol 2016;134:114-115.
https://doi.org/10.1001/jamaophthalmol.2015.4581

 

59 Berry JL, Kogachi K, Aziz HA, McGovern K, Zolfaghari E, Murphree AL, Jubran R, Kim JW: Risk of metastasis and orbital recurrence in advanced retinoblastoma eyes treated with systemic chemoreduction versus primary enucleation. Pediatr Blood Cancer 2017; DOI: 10.1002/pbc.26270.
https://doi.org/10.1002/pbc.26270

 

60 Shields CL, Jorge R, Say EA, Magrath G, Alset A, Caywood E, Leahey AM, Jabbour P, Shields JA: Unilateral Retinoblastoma Managed With Intravenous Chemotherapy Versus Intra-Arterial Chemotherapy. Outcomes Based on the International Classification of Retinoblastoma. Asia Pac J Ophthalmol (Phila) 2016;5:97-103.
https://doi.org/10.1097/APO.0000000000000172

 

61 Shields CL, Mashayekhi A, Cater J, Shelil A, Ness S, Meadows AT, Shields JA: Macular retinoblastoma managed with chemoreduction: analysis of tumor control with or without adjuvant thermotherapy in 68 tumors. Arch Ophthalmol 2005;123:765-773.
https://doi.org/10.1001/archopht.123.6.765

 

62 Shields CL, Shields JA: Basic understanding of current classification and management of retinoblastoma. Curr Opin Ophthalmol 2006;17:228-234.
https://doi.org/10.1097/01.icu.0000193079.55240.18

 

63 Shields CL, Ramasubramanian A, Thangappan A, Hartzell K, Leahey A, Meadows AT, Shields JA: Chemoreduction for group E retinoblastoma: comparison of chemoreduction alone versus chemoreduction plus low-dose external radiotherapy in 76 eyes. Ophthalmology 2009;116:544-551.e1.
https://doi.org/10.1016/j.ophtha.2008.10.014

 

64 Weng PY, Chen SH, Kao LY, Tsai YJ, Yang SH, Tseng CK, Tsay PK, Jaing TH: Clinical spectrum and treatment outcome of retinoblastoma with Group D and E diseases: A single institution retrospective review. Medicine (Baltimore) 2020;99:e22201.
https://doi.org/10.1097/MD.0000000000022201

 

65 Zhao J, Dimaras H, Massey C, Xu X, Huang D, Li B, Chan HS, Gallie BL: 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.
https://doi.org/10.1200/JCO.2010.32.5332

 

66 Zanaty M, Barros G, Chalouhi N, Starke RM, Manasseh P, Tjoumakaris SI, Shields CL, Hasan D, Bulsara K, Rosenwasser RH, Jabbour P: Update on intra-arterial chemotherapy for retinoblastoma. ScientificWorldJournal 2014;2014:869604.
https://doi.org/10.1155/2014/869604

 

67 Gallie BL, Budning A, DeBoer G, Thiessen JJ, Koren G, Verjee Z, Ling V, Chan HS: Chemotherapy with focal therapy can cure intraocular retinoblastoma without radiotherapy. Arch Ophthalmol 1996;114:1321-1328.
https://doi.org/10.1001/archopht.1996.01100140521001

 

68 Chan HS, Lu Y, Grogan TM, Haddad G, Hipfner DR, Cole SP, Deeley RG, Ling V, Gallie BL: Multidrug resistance protein (MRP) expression in retinoblastoma correlates with the rare failure of chemotherapy despite cyclosporine for reversal of P-glycoprotein. Cancer Res 1997;57:2325-2330.

 

69 Chen Q, Zhang B, Dong Y, Mo X, Zhang L, Xia J, Zhang J, Zhang S: Intra-arterial chemotherapy as primary or secondary treatment for infants diagnosed with advanced retinoblastoma before 3 months of age. BMC Cancer 2019;19:693.
https://doi.org/10.1186/s12885-019-5844-5

 

70 Francis JH, Abramson DH, Gobin YP, Marr BP, Tendler I, Brodie SE, Dunkel IJ: Efficacy and toxicity of second-course ophthalmic artery chemosurgery for retinoblastoma. Ophthalmology 2015;122:1016-1022.
https://doi.org/10.1016/j.ophtha.2014.11.029

 

71 Munier FL, Gaillard MC, Balmer A, Soliman S, Podilsky G, Moulin AP, Beck-Popovic M: Intravitreal chemotherapy for vitreous disease in retinoblastoma revisited: from prohibition to conditional indications. Br J Ophthalmol 2012;96:1078-1083.
https://doi.org/10.1136/bjophthalmol-2011-301450

 

72 Ghassemi F, Shields CL: Intravitreal melphalan for refractory or recurrent vitreous seeding from retinoblastoma. Arch Ophthalmol 2012;130:1268-1271.
https://doi.org/10.1001/archophthalmol.2012.1983

 

73 Munier FL, Mosimann P, Puccinelli F, Gaillard MC, Stathopoulos C, Houghton S, Bergin C, Beck-Popovic M: First-line intra-arterial versus intravenous chemotherapy in unilateral sporadic group D retinoblastoma: evidence of better visual outcomes, ocular survival and shorter time to success with intra-arterial delivery from retrospective review of 20 years of treatment. Br J Ophthalmol 2017;101:1086-1093.
https://doi.org/10.1136/bjophthalmol-2016-309298

 

74 Suzuki S, Yamane T, Mohri M, Kaneko A: Selective ophthalmic arterial injection therapy for intraocular retinoblastoma: the long-term prognosis. Ophthalmology 2011;118:2081-2087.
https://doi.org/10.1016/j.ophtha.2011.03.013

 

75 Peterson EC, Elhammady MS, Quintero-Wolfe S, Murray TG, Aziz-Sultan MA: Selective ophthalmic artery infusion of chemotherapy for advanced intraocular retinoblastoma: initial experience with 17 tumors. J Neurosurg 2011;114:1603-1608.
https://doi.org/10.3171/2011.1.JNS10466

 

76 Abramson DH, Francis JH, Dunkel IJ, Marr BP, Brodie SE, Gobin YP: Ophthalmic artery chemosurgery for retinoblastoma prevents new intraocular tumors. Ophthalmology 2013;120:560-565.
https://doi.org/10.1016/j.ophtha.2012.08.023

 

77 Da Silva VF, Raaphorst GP, Goyal R, Feeley M: Drug cytotoxicity at elevated temperature. In vitro study on the U-87MG glioma cell line. J Neurosurg 1987;67:885-888.
https://doi.org/10.3171/jns.1987.67.6.0885

 

78 Herman TS, Teicher BA: Summary of studies adding systemic chemotherapy to local hyperthermia and radiation. Int J Hyperthermia 1994;10:443-449.
https://doi.org/10.3109/02656739409010290

 

79 Tapazoglou E, Cohen JD, Schmitt CL, Khatana A, Sapareto SA, Robins HI: Whole body hyperthermia and carboplatin: cytotoxicity for murine leukaemia and normal marrow. Br J Cancer 1991;64:528-530.
https://doi.org/10.1038/bjc.1991.343

 

80 Murray TG, Cicciarelli N, McCabe CM, Ksander B, Feuer W, Schiffman J, Mieler WF, O'Brien JM: In vitro efficacy of carboplatin and hyperthermia in a murine retinoblastoma cell line. Invest Ophthalmol Vis Sci 1997;38:2516-2522.

 

81 Suzuki S, Aihara Y, Fujiwara M, Sano S, Kaneko A: Intravitreal injection of melphalan for intraocular retinoblastoma. Jpn J Ophthalmol 2015;59:164-172.
https://doi.org/10.1007/s10384-015-0378-0

 

82 Coschi CH, Martens AL, Ritchie K, Francis SM, Chakrabarti S, Berube NG, Dick FA: Mitotic chromosome condensation mediated by the retinoblastoma protein is tumor-suppressive. Genes Dev 2010;24:1351-1363.
https://doi.org/10.1101/gad.1917610

 

83 Balmer A, Zografos L, Munier F: Diagnosis and current management of retinoblastoma. Oncogene 2006;25:5341-5349.
https://doi.org/10.1038/sj.onc.1209622

 

84 Munier FL, Soliman S, Moulin AP, Gaillard MC, Balmer A, Beck-Popovic M: Profiling safety of intravitreal injections for retinoblastoma using an anti-reflux procedure and sterilisation of the needle track. Br J Ophthalmol 2012;96:1084-1087.
https://doi.org/10.1136/bjophthalmol-2011-301016

 

85 Amin S, Rizvi F, Zia N, Ali A, Hamid A, Kumari B: Outcomes of Group D Retinoblastoma With Resistant Vitreous Seeds After Integration of Intravitreal Chemotherapy to the Treatment Protocol. Cureus 2020;12:e11757.
https://doi.org/10.7759/cureus.11757

 

86 Shields CL, Honavar SG, Meadows AT, Shields JA, Demirci H, Singh A, Friedman DL, Naduvilath TJ: Chemoreduction plus focal therapy for retinoblastoma: factors predictive of need for treatment with external beam radiotherapy or enucleation. Am J Ophthalmol 2002;133:657-664.
https://doi.org/10.1016/S0002-9394(02)01348-X

 

87 Amemiya T, Yoshida H, Ishigooka H: Vitreous seeds in retinoblastoma, clinical significance and ultrastructure. Albrecht Von Graefes Arch Klin Exp Ophthalmol 1979;211:205-213.
https://doi.org/10.1007/BF02387425

 

88 Gombos DS, Cauchi PA, Hungerford JL, Addison P, Coen PG, Kingston JE: Vitreous relapse following primary chemotherapy for retinoblastoma: is adjuvant diode laser a risk factor? Br J Ophthalmol 2006;90:1168-1172.
https://doi.org/10.1136/bjo.2006.091223

 

89 Smith SJ, Smith BD, Mohney BG: Ocular side effects following intravitreal injection therapy for retinoblastoma: a systematic review. Br J Ophthalmol 2014;98:292-297.
https://doi.org/10.1136/bjophthalmol-2013-303885

 

90 Kaneko A, Suzuki S: Eye-preservation treatment of retinoblastoma with vitreous seeding. Jpn J Clin Oncol 2003;33:601-607.
https://doi.org/10.1093/jjco/hyg113

 

91 Yousef YA, Noureldin AM, Sultan I, Deebajah R, Al-Hussaini M, Shawagfeh M, Mehyar M, Mohammad M, Jaradat I, AlNawaiseh I: Intravitreal Melphalan Chemotherapy for Vitreous Seeds in Retinoblastoma. J Ophthalmol 2020;2020:8628525.
https://doi.org/10.1155/2020/8628525

 

92 Francis JH, Iyer S, Gobin YP, Brodie SE, Abramson DH: retinoblastoma Vitreous Seed Clouds (Class 3): A Comparison of Treatment with Ophthalmic Artery Chemosurgery with or without Intravitreous and Periocular Chemotherapy. Ophthalmology 2017;124:1548-1555.
https://doi.org/10.1016/j.ophtha.2017.04.010

 

93 Yu X, Li X, Xing Y, Lu S, Tanumiharjo S, Ma J: Effectiveness of intravitreal chemotherapy-assisted endoresection in monocular patients with group D retinoblastoma. BMC Cancer 2020;20:808.
https://doi.org/10.1186/s12885-020-07314-1

 

94 Marr BP, Dunkel IJ, Linker A, Abramson DH: Periocular carboplatin for retinoblastoma: long-term report (12 years) on efficacy and toxicity. Br J Ophthalmol 2012;96:881-883.
https://doi.org/10.1136/bjophthalmol-2011-300517

 

95 Said AMA, Aly MG, Rashed HO, Rady AM: Safety and efficacy of posterior sub-Tenon's carboplatin injection versus intravitreal melphalan therapy in the management of retinoblastoma with secondary vitreous seeds. Int J Ophthalmol 2018;11:445-455.

 

96 Reese AB, Hyman GA, Merriam GR, Jr., Forrest AW, Kligerman MM: Treatment of retinoblastoma by radiation and triethylenemelamine. AMA Arch Ophthalmol 1954;53:505-513.
https://doi.org/10.1001/archopht.1955.00930010507007

 

97 Servodidio CA, Abramson DH: Acute and long-term effects of radiation therapy to the eye in children. Cancer Nurs 1993;16:371-381.
https://doi.org/10.1097/00002820-199310000-00006

 

98 Eng C, Li FP, Abramson DH, Ellsworth RM, Wong FL, Goldman MB, Seddon J, Tarbell N, Boice JD, Jr.: Mortality from second tumors among long-term survivors of retinoblastoma. J Natl Cancer Inst 1993;85:1121-1128.
https://doi.org/10.1093/jnci/85.14.1121

 

99 Ferris FL, 3rd, Chew EY: A new era for the treatment of retinoblastoma. Arch Ophthalmol 1996;114:1412.
https://doi.org/10.1001/archopht.1996.01100140612015

 

100 Yousef YA, Mohammad M, Jaradat I, Shatnawi R, Banat S, Mehyar M, Al-Nawaiseh I: The role of external beam radiation therapy for retinoblastoma after failure of combined chemoreduction and focal consolidation therapy. Ophthalmic Genet 2020;41:20-25.
https://doi.org/10.1080/13816810.2020.1719519

 

101 Yousef YA, Mohammad M, Jaradat I, Shatnawi R, Mehyar M, Al-Nawaiseh I: Prognostic Factors for Eye Globe Salvage by External Beam Radiation Therapy for Resistant Intraocular retinoblastoma. Oman J Ophthalmol 2020;13:123-128.
https://doi.org/10.4103/ojo.OJO_250_2019

 

102 Jung EH, Kim JH, Kim JY, Jo DH, Yu YS: Outcomes of Proton Beam Radiation Therapy for Retinoblastoma With Vitreous Seeds. J Pediatr Hematol Oncol 2018;40:569-573.
https://doi.org/10.1097/MPH.0000000000001176

 

103 Sethi RV, Shih HA, Yeap BY, Mouw KW, Petersen R, Kim DY, Munzenrider JE, Grabowski E, Rodriguez-Galindo C, Yock TI, Tarbell NJ, Marcus KJ, Mukai S, MacDonald SM: Second nonocular tumors among survivors of retinoblastoma treated with contemporary photon and proton radiotherapy. Cancer 2014;120:126-133.
https://doi.org/10.1002/cncr.28387

 

104 Biewald E, Kiefer T, Geismar D, Schluter S, Manthey A, Westekemper H, Wulff J, Timmermann B, Ketteler P, Schonberger S, Metz KA, Ting S, Goricke S, Bechrakis NE, Bornfeld N: Feasibility of Proton Beam Therapy as a Rescue Therapy in Heavily Pre-Treated Retinoblastoma Eyes. Cancers (Basel) 2021;13:1862.
https://doi.org/10.3390/cancers13081862

 

105 Reese AB, Ellsworth RM: The evaluation and current concept of retinoblastoma therapy. Trans Am Acad Ophthalmol Otolaryngol 1963;67:164-172.

 

106 Chevez-Barrios P, Eagle RC, Jr., Krailo M, Piao J, Albert DM, Gao Y, Vemuganti G, Ali MJ, Khetan V, Honavar SG, O'Brien J, Leahey AM, Matthay K, Meadows A, Chintagumpala M: Study of Unilateral retinoblastoma With and Without Histopathologic High-Risk Features and the Role of Adjuvant Chemotherapy: A Children's Oncology Group Study. J Clin Oncol 2019;37:2883-2891.
https://doi.org/10.1200/JCO.18.01808

 

107 Ravindran K, Dalvin LA, Pulido JS, Brinjikji W: Intra-arterial chemotherapy for retinoblastoma: an updated systematic review and meta-analysis. J Neurointerv Surg 2019;11:1266-1272.
https://doi.org/10.1136/neurintsurg-2019-014909

 

108 Abramson DH, Gamell LS, Ellsworth RM, Kruger EF, Servodidio CA, Turner L, Sussman D: Unilateral retinoblastoma: new intraocular tumours after treatment. Br J Ophthalmol 1994;78:698-701.
https://doi.org/10.1136/bjo.78.9.698

 

109 Abramson DH, Greenfield DS, Ellsworth RM: Bilateral retinoblastoma. Correlations between age at diagnosis and time course for new intraocular tumors. Ophthalmic Paediatr Genet 1992;13:1-7.
https://doi.org/10.3109/13816819209070046

 

110 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;120:460-464.
https://doi.org/10.1001/archopht.120.4.460

 

111 Shields CL, Shelil A, Cater J, Meadows AT, Shields JA: Development of new retinoblastomas after 6 cycles of chemoreduction for retinoblastoma in 162 eyes of 106 consecutive patients. Arch Ophthalmol 2003;121:1571-1576.
https://doi.org/10.1001/archopht.121.11.1571

 

112 Shields CL, Mashayekhi A, Cater J, Shelil A, Meadows AT, Shields JA: Chemoreduction for retinoblastoma: analysis of tumor control and risks for recurrence in 457 tumors. Trans Am Ophthalmol Soc 2004;102:35-44; discussion 44-35.

 

113 Chantada GL, Dunkel IJ, de Davila MT, Abramson DH: Retinoblastoma patients with high risk ocular pathological features: who needs adjuvant therapy? Br J Ophthalmol 2004;88:1069-1073.
https://doi.org/10.1136/bjo.2003.037044

 

114 Messmer EP, Heinrich T, Hopping W, de Sutter E, Havers W, Sauerwein W: Risk factors for metastases in patients with retinoblastoma. Ophthalmology 1991;98:136-141.
https://doi.org/10.1016/S0161-6420(91)32325-X

 

115 Sreelakshmi KV, Chandra A, Krishnakumar S, Natarajan V, Khetan V: Anterior Chamber Invasion in Retinoblastoma: Not an Indication for Adjuvant Chemotherapy. Invest Ophthalmol Vis Sci 2017;58:4654-4661.
https://doi.org/10.1167/iovs.17-22111

 

116 Tomar AS, Finger PT, Gallie B, Kivela TT, Mallipatna A, Zhang C, Zhao J, Wilson MW, Brenna RC, Burges M, Kim J, Khetan V, Ganesan S, Yarovoy A, Yarovaya V, Kotova E, Yousef YA, Nummi K, Ushakova TL, Yugay OV, et al.: Global Retinoblastoma Treatment Outcomes: Association with National Income Level. Ophthalmology 2021;128:740-753.
https://doi.org/10.1016/j.ophtha.2020.09.032

 

117 Shields CL, Shields JA, Baez K, Cater JR, De Potter P: Optic nerve invasion of retinoblastoma. Metastatic potential and clinical risk factors. Cancer 1994;73:692-698.
https://doi.org/10.1002/1097-0142(19940201)73:3<692::AID-CNCR2820730331>3.0.CO;2-8

 

118 Shields CL, Shields JA, Baez KA, Cater J, De Potter PV: Choroidal invasion of retinoblastoma: metastatic potential and clinical risk factors. Br J Ophthalmol 1993;77:544-548.
https://doi.org/10.1136/bjo.77.9.544

 

119 Chantada GL, Fandino AC, Raslawski EC, Manzitti J, de Davila MT, Casak SJ, Scopinaro MJ, Schvartzman E: Experience with chemoreduction and focal therapy for intraocular retinoblastoma in a developing country. Pediatr Blood Cancer 2005;44:455-460.
https://doi.org/10.1002/pbc.20259

 

120 Sullivan EM, Wilson MW, Billups CA, Wu J, Merchant TE, Brennan RC, Haik BG, Shulkin B, Free TM, Given V, Rodriguez-Galindo C, Qaddoumi I: Pathologic risk-based adjuvant chemotherapy for unilateral retinoblastoma following enucleation. J Pediatr Hematol Oncol 2014;36:e335-340.
https://doi.org/10.1097/MPH.0000000000000141

 

121 Kunkele A, Wilm J, Holdt M, Lohmann D, Bornfeld N, Eggert A, Temming P, Schulte JH: Neoadjuvant/adjuvant treatment of high-risk retinoblastoma: a report from the German Retinoblastoma Referral Centre. Br J Ophthalmol 2015;99:949-953.
https://doi.org/10.1136/bjophthalmol-2014-306222

 

122 Sitorus RS, Moll AC, Suhardjono S, Simangunsong LS, Riono P, Imhof S, Volker-Dieben HJ: The effect of therapy refusal against medical advice in retinoblastoma patients in a setting where treatment delays are common. Ophthalmic Genet 2009;30:31-36.
https://doi.org/10.1080/13816810802464320

 

123 Chantada G, Fandino A, Manzitti J, Urrutia L, Schvartzman E: Late diagnosis of retinoblastoma in a developing country. Arch Dis Child 1999;80:171-174.
https://doi.org/10.1136/adc.80.2.171

 

124 Nyamori JM, Kimani K, Njuguna MW, Dimaras H: Retinoblastoma referral pattern in Kenya. Middle East Afr J Ophthalmol 2014;21:321-327.
https://doi.org/10.4103/0974-9233.142270

 

125 Bowman RJ, Mafwiri M, Luthert P, Luande J, Wood M: Outcome of retinoblastoma in east Africa. Pediatr Blood Cancer 2008;50:160-162.
https://doi.org/10.1002/pbc.21080

 

126 Luna-Fineman S, Barnoya M, Bonilla M, Fu L, Baez F, Rodriguez-Galindo C: Retinoblastoma in Central America: report from the Central American Association of Pediatric Hematology Oncology (AHOPCA). Pediatr Blood Cancer 2012;58:545-550.
https://doi.org/10.1002/pbc.23307

 

127 Chawla B, Hasan F, Azad R, Seth R, Upadhyay AD, Pathy S, Pandey RM: Clinical presentation and survival of retinoblastoma in Indian children. Br J Ophthalmol 2016;100:172-178.
https://doi.org/10.1136/bjophthalmol-2015-306672

 

128 Friedrich P, Lam CG, Itriago E, Perez R, Ribeiro RC, Arora RS: Magnitude of Treatment Abandonment in Childhood Cancer. PLoS One 2015;10:e0135230.
https://doi.org/10.1371/journal.pone.0135230

 

129 Mostert S, Arora RS, Arreola M, Bagai P, Friedrich P, Gupta S, Kaur G, Koodiyedath B, Kulkarni K, Lam CG, Luna-Fineman S, Pizer B, Rivas S, Rossell N, Sitaresmi MN, Tsimicalis A, Weaver M, Ribeiro RC: Abandonment of treatment for childhood cancer: position statement of a SIOP PODC Working Group. Lancet Oncol 2011;12:719-720.
https://doi.org/10.1016/S1470-2045(11)70128-0

 

130 Olteanu C, Dimaras H: Enucleation Refusal for Retinoblastoma: A Global Study. Ophthalmic Genet 2016;37:137-143.
https://doi.org/10.3109/13816810.2014.937543

 

131 Bellaton E, Bertozzi AI, Behar C, Chastagner P, Brisse H, Sainte-Rose C, Doz F, Desjardins L: Neoadjuvant chemotherapy for extensive unilateral retinoblastoma. Br J Ophthalmol 2003;87:327-329.
https://doi.org/10.1136/bjo.87.3.327

 

132 Chantada GL, Gonzalez A, Fandino A, de Davila MT, Demirdjian G, Scopinaro M, Abramson D: Some clinical findings at presentation can predict high-risk pathology features in unilateral retinoblastoma. J Pediatr Hematol Oncol 2009;31:325-329.
https://doi.org/10.1097/MPH.0b013e3181923cc5

 

133 Haimi M, Peretz Nahum M, Ben Arush MW: Delay in diagnosis of children with cancer: a retrospective study of 315 children. Pediatr Hematol Oncol 2004;21:37-48.
https://doi.org/10.1080/pho.21.1.37.48

 

134 Leander C, Fu LC, Pena A, Howard SC, Rodriguez-Galindo C, Wilimas JA, Ribeiro RC, Haik B: Impact of an education program on late diagnosis of retinoblastoma in Honduras. Pediatr Blood Cancer 2007;49:817-819.
https://doi.org/10.1002/pbc.21052

 

135 Chan HS, Gallie BL, Munier FL, Beck Popovic M: Chemotherapy for retinoblastoma. Ophthalmol Clin North Am 2005;18:55-63, viii.
https://doi.org/10.1016/j.ohc.2004.11.002

 

136 Abramson DH, Schefler AC: Update on retinoblastoma. Retina 2004;24:828-848.
https://doi.org/10.1097/00006982-200412000-00002

 

137 Chung SE, Sa HS, Koo HH, Yoo KH, Sung KW, Ham DI: Clinical manifestations and treatment of retinoblastoma in Korea. Br J Ophthalmol 2008;92:1180-1184.
https://doi.org/10.1136/bjo.2008.140046

 

138 Gunduz K, Kose K, Kurt RA, Suren E, Tacyildiz N, Dincaslan H, Unal E, Erden E, Heper AO: Retinoblastoma in Turkey: results from a tertiary care center in Ankara. J Pediatr Ophthalmol Strabismus 2013;50:296-303.
https://doi.org/10.3928/01913913-20130730-02

 

139 Lim FP, Soh SY, Iyer JV, Tan AM, Swati H, Quah BL: Clinical profile, management, and outcome of retinoblastoma in singapore. J Pediatr Ophthalmol Strabismus 2013;50:106-112.
https://doi.org/10.3928/01913913-20121211-01

 

140 Kim JY, Park Y: Treatment of Retinoblastoma: The Role of External Beam Radiotherapy. Yonsei Med J 2015;56:1478-1491.
https://doi.org/10.3349/ymj.2015.56.6.1478

 

141 Kaliki S, Shields CL, Rojanaporn D, Al-Dahmash S, McLaughlin JP, Shields JA, Eagle RC, Jr.: High-risk retinoblastoma based on international classification of retinoblastoma: analysis of 519 enucleated eyes. Ophthalmology 2013;120:997-1003.
https://doi.org/10.1016/j.ophtha.2012.10.044

 

142 Jubran RF, Villablanca JG, Krailo M, Piao J, Huang L, Murphree AL, O'Brien J, Gombos D, Shields CL, Meadows A, Chintagumpala M: A single-arm study of systemic and sub-Tenon chemotherapy for Groups C and D intraocular retinoblastoma: A Children's Oncology Group study (ARET 0231). Pediatr Blood Cancer 2020;67:e28502.
https://doi.org/10.1002/pbc.28502

 

143 Kim U, Rathi G, Chowdhary G, Srinavasan KG, Shanthi R, Krishna RSP: Accuracy of preoperative imaging in predicting optic nerve invasion in retinoblastoma: A retrospective study. Indian J Ophthalmol 2019;67:2019-2022.
https://doi.org/10.4103/ijo.IJO_1611_18

 

144 Wang YZ, Zhang Y, Huang DS, Shi JT, Ma JM, Li B, Xu XL, Zhou Y, Gu HL: Clinical characteristics, treatment and prognosis of children with unilateral retinoblastoma and intracranial segment of Retrobulbar optic nerve invasion. BMC Ophthalmol 2021;21:38.
https://doi.org/10.1186/s12886-020-01768-4

 

145 Honavar SG, Singh AD, Shields CL, Meadows AT, Demirci H, Cater J, Shields JA: Postenucleation adjuvant therapy in high-risk retinoblastoma. Arch Ophthalmol 2002;120:923-931.
https://doi.org/10.1001/archopht.120.7.923

 

146 Uusitalo MS, Van Quill KR, Scott IU, Matthay KK, Murray TG, O'Brien JM: Evaluation of chemoprophylaxis in patients with unilateral retinoblastoma with high-risk features on histopathologic examination. Arch Ophthalmol 2001;119:41-48.

 

147 Kopelman JE, McLean IW, Rosenberg SH: Multivariate analysis of risk factors for metastasis in retinoblastoma treated by enucleation. Ophthalmology 1987;94:371-377.
https://doi.org/10.1016/S0161-6420(87)33436-0

 

148 Magramm I, Abramson DH, Ellsworth RM: Optic nerve involvement in retinoblastoma. Ophthalmology 1989;96:217-222.
https://doi.org/10.1016/S0161-6420(89)32910-1

 

149 Haik BG, Dunleavy SA, Cooke C, Ellsworth RM, Abramson DH, Smith ME, Karcioglu ZA: Retinoblastoma with anterior chamber extension. Ophthalmology 1987;94:367-370.
https://doi.org/10.1016/S0161-6420(87)33437-2

 

150 Khelfaoui F, Validire P, Auperin A, Quintana E, Michon J, Pacquement H, Desjardins L, Asselain B, Schlienger P, Vielh P, et al.: Histopathologic risk factors in retinoblastoma: a retrospective study of 172 patients treated in a single institution. Cancer 1996;77:1206-1213.
https://doi.org/10.1002/(SICI)1097-0142(19960315)77:6<1206::AID-CNCR30>3.0.CO;2-3

 

151 Chantada GL, Fandino AC, Guitter MR, Raslawski EC, Dominguez JL, Manzitti J, de Davila MT, Zubizarreta P, Scopinaro M: Results of a prospective study for the treatment of unilateral retinoblastoma. Pediatr Blood Cancer 2010;55:60-66.
https://doi.org/10.1002/pbc.22503

 

152 Perez V, Sampor C, Rey G, Parareda-Salles A, Kopp K, Dabezies AP, Dufort G, Zelter M, Lopez JP, Urbieta M, Alcalde-Ruiz E, Catala-Mora J, Sunol M, Ossandon D, Fandino AC, Croxatto JO, de Davila MTG, Reaman G, Ravindranath Y, Chantada GL: Treatment of Nonmetastatic Unilateral Retinoblastoma in Children. JAMA Ophthalmol 2018;136:747-752.
https://doi.org/10.1001/jamaophthalmol.2018.1501

 

153 Sastre X, Chantada GL, Doz F, Wilson MW, de Davila MT, Rodriguez-Galindo C, Chintagumpala M, Chevez-Barrios P, International Retinoblastoma Staging Working G: 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.
https://doi.org/10.5858/133.8.1199

 

154 Del Carmen Bernal-Diaz Z, Murillo-Maldonado MA, Perez-Villanueva H, Reyes-Lopez A, Ramirez-Ortiz MA: Ocular preservation in patients with bilateral retinoblastoma before chemotherapy in situ era: A report from a Mexican Retinoblastoma Reference Hospital. Pediatr Blood Cancer 2020;67:e28625.
https://doi.org/10.1002/pbc.28625