51
|
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. [PMID: 32847550 PMCID: PMC7448309 DOI: 10.1186/s12885-020-07314-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/18/2020] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to determine the efficacy and complications of intravitreal chemotherapy-assisted endoresection for refractory International Classification of Retinoblastoma (ICRB) group D retinoblastoma in monocular patients. Methods In this retrospective case series, intravitreal chemotherapy-assisted endoresection by pars plana vitrectomy was performed in 11 eyes with refractory ICRB group D retinoblastoma unresponsive to standard therapies in monocular patients. Results Across a mean follow-up period of 42.7 months, globe salvage was attained in all 11 eyes (100%). There were no cases of extra-ocular tumour seeding or remote metastasis. In 9 eyes (81.8%), tumour control was achieved with one pars plana vitrectomy; in 2 cases (18.2%), repeated treatment, such as laser therapy, intravitreal chemotherapy or a second pars plana vitrectomy, was needed. Retinal reattachment was achieved in all 4 eyes (100%) with previous retinal detachment. Four eyes (36.4%) required subsequent cataract surgery due to secondary cataract. Ten eyes (90.9%) had improvement in best-corrected visual acuity at the last follow-up. Conclusion Intravitreal chemotherapy-assisted endoresection appears to be a safe and effective globe-salvaging method for refractory group D retinoblastoma. It is a promising alternative to enucleation and a supplementary therapeutic strategy for those unresponsive to standard therapies, especially for the monocular retinoblastoma patients.
Collapse
Affiliation(s)
- Xiling Yu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Xueke Li
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Yue Xing
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Siduo Lu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Silvia Tanumiharjo
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China
| | - Jin Ma
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
52
|
Retinoblastoma: Etiology, Modeling, and Treatment. Cancers (Basel) 2020; 12:cancers12082304. [PMID: 32824373 PMCID: PMC7465685 DOI: 10.3390/cancers12082304] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022] Open
Abstract
Retinoblastoma is a retinal cancer that is initiated in response to biallelic loss of RB1 in almost all cases, together with other genetic/epigenetic changes culminating in the development of cancer. RB1 deficiency makes the retinoblastoma cell-of-origin extremely susceptible to cancerous transformation, and the tumor cell-of-origin appears to depend on the developmental stage and species. These are important to establish reliable preclinical models to study the disease and develop therapies. Although retinoblastoma is the most curable pediatric cancer with a high survival rate, advanced tumors limit globe salvage and are often associated with high-risk histopathological features predictive of dissemination. The advent of chemotherapy has improved treatment outcomes, which is effective for globe preservation with new routes of targeted drug delivery. However, molecularly targeted therapeutics with more effectiveness and less toxicity are needed. Here, we review the current knowledge concerning retinoblastoma genesis with particular attention to the genomic and transcriptomic landscapes with correlations to clinicopathological characteristics, as well as the retinoblastoma cell-of-origin and current disease models. We further discuss current treatments, clinicopathological correlations, which assist in guiding treatment and may facilitate globe preservation, and finally we discuss targeted therapeutics for future treatments.
Collapse
|
53
|
Camp DA, Dalvin LA, Schwendeman R, Lim LAS, Shields CL. Outcomes of neonatal retinoblastoma in pre-chemotherapy and chemotherapy eras. Indian J Ophthalmol 2020; 67:1997-2004. [PMID: 31755437 PMCID: PMC6896534 DOI: 10.4103/ijo.ijo_634_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Purpose To quantify outcomes for neonatal retinoblastoma patients treated during the pre-chemotherapy (1980-1994) and chemotherapy (1995-2018) eras. Methods Retrospective review of retinoblastoma patients diagnosed within the first 28 days of life between 1/1/1980 and 11/30/2018. Student's t-test, Chi-square, and Fisher's exact test were performed to compare treatments and outcomes by era. Results There were 68 patients with neonatal retinoblastoma (12% unilateral and 88% bilateral). According to era (pre-chemotherapy vs. chemotherapy), the number of treated patients was 26 (38%) vs. 42 (62%). Primary treatment was external beam radiotherapy (50% vs. 1%,P < 0.001), plaque radiotherapy (17% vs. 0%,P < 0.001), focal treatment (transpupillary thermotherapy or cryotherapy) only (21% vs. 14%,P= 0.33), intravenous chemotherapy (0% vs. 81%,P < 0.001), enucleation (10% vs. 4%,P= 0.26), or exenteration (2% vs. 0%,P= 0.37). Outcomes included tumor control (79% vs. 94%,P= 0.02), globe salvage (75% vs. 91%,P= 0.02), final gross visual acuity for salvaged eyes 20/200 or better (66% vs. 89%,P < 0.01), and death (19% vs. 0%,P < 0.01). Conclusion Chemotherapy advancements for neonatal retinoblastoma have improved tumor control, globe salvage, visual acuity, and patient survival.
Collapse
Affiliation(s)
- David A Camp
- Ocular Oncology Service, Wills Eye Hospital, Philadelphia, PA, USA
| | - Lauren A Dalvin
- Ocular Oncology Service, Wills Eye Hospital, Philadelphia, PA; Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | - Li-Anne S Lim
- Ocular Oncology Service, Wills Eye Hospital, Philadelphia, PA, USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Philadelphia, PA, USA
| |
Collapse
|
54
|
Dalvin LA, Bas Z, Tadepalli S, Rao R, Vaidya S, Pacheco R, Shields CL. Risk Factors for Tumor Recurrence Following Primary Intravenous Chemotherapy (Chemoreduction) for Retinoblastoma in 869 Eyes of 551 Patients. J Pediatr Ophthalmol Strabismus 2020; 57:224-234. [PMID: 32687206 DOI: 10.3928/01913913-20200417-01] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 03/30/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE To identify risk factors for retinoblastoma recurrence following chemoreduction. METHODS This was a retrospective review of patients with retinoblastoma treated from 1994 to 2019 using chemoreduction with analysis for recurrence using Kaplan-Meier, Cox regression, and logistic regression. RESULTS There were 869 eyes of 551 patients with retinoblastoma treated with chemoreduction. Follow-up in 556 eyes revealed main solid tumor recurrence (n = 355, 64%), subretinal seed recurrence (n = 244, 44%), vitreous seed recurrence (n = 162, 29%), and/or new tumor (n = 118, 21%) requiring management with focal therapy (transpupillary thermotherapy, cryotherapy) (n = 294, 53%), intra-arterial chemotherapy (n = 125, 22%), intravitreal chemotherapy (n = 36, 6%), plaque radiotherapy (n = 120, 22%), external beam radiotherapy (n = 57, 10%), and/or enucleation (n = 49, 9%). Of all recurrences, 62% were detected by 1 year, 86% by 2 years, 94% by 3 years, 98% by 5 years, 99% by 10 years, and 100% by 15 years. Risk factors for recurrence on multivariate analysis included younger patient age at presentation (odds ratio [OR] = 1.02 [1.00 to 1.04] per 1 month decrease, P = .02), greater International Classification of Retinoblastoma group (OR = 1.24 [1.05 to 1.47] per 1 more advanced group, P = .01), shorter tumor distance to optic disc (OR = 1.11 [1.01 to 1.21] per 1 mm decrease, P = .03), and presence of subretinal seeds (OR = 1.66 [1.09 to 2.53], P = .02). CONCLUSIONS Retinoblastoma recurrence after chemoreduction is usually detected within the first 3 years following treatment. Younger patients with more advanced, posteriorly located tumors and subretinal seeds at presentation are at increased risk, but recurrence can often be managed with globe-sparing therapy. [J Pediatr Ophthalmol Strabismus. 2020;57(4):224-234.].
Collapse
|
55
|
Lee JS, Kim JY, Jung C, Woo SJ. Iatrogenic ophthalmic artery occlusion and retinal artery occlusion. Prog Retin Eye Res 2020; 78:100848. [PMID: 32165219 DOI: 10.1016/j.preteyeres.2020.100848] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 01/19/2023]
Abstract
Iatrogenic ophthalmic artery occlusion (IOAO) is a rare but devastating ophthalmic disease that may cause sudden and permanent visual loss. Understanding the possible etiologic modalities and pathogenic mechanisms of IOAO may prevent its occurrence. There are numerous medical etiologies of IOAO, including cosmetic facial filler injection, intravascular procedures, intravitreal gas or drug injection, retrobulbar anesthesia, intraarterial chemotherapy in retinoblastoma. Non-ocular surgeries and vascular events in arteries that are not directly associated with the ophthalmic artery, can also cause IOAO. Since IOAO has a limited number of treatment modalities, which lead to poor final visual prognosis, it is imperative to acknowledge the information regarding medical procedures that are etiologically associated with IOAO. We accumulated all searchable and available IOAO case reports (our cases and previous reported cases from the literature), classified them according to their mechanisms of pathogenesis, and summarized treatment options and responses of each of the causes. Various sporadic cases of IOAO can be categorized into three mechanisms as follows: intravascular event, orbital compartment syndrome, and increased intraocular pressure. Embolic IOAO, which is considered the primary cause of the condition, was classified into three subgroups according to the pathway of embolic movement (retrograde pathway, anterograde pathway, pathway through collateral channels). Despite the practical limitations of treating spontaneous (non-iatrogenic) retinal artery occlusion, this article will contribute in predicting and improving the prognosis of IOAO by recognizing the treatable factors. Furthermore, it is expected to provide clues to future research associated with the treatment of retinal artery occlusion.
Collapse
Affiliation(s)
- Jong Suk Lee
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jun Yup Kim
- Department of Neurology, Seoul National University College of Medicine, Cerebrovascular Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Cheolkyu Jung
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Se Joon Woo
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea.
| |
Collapse
|
56
|
Sun J, Xi HY, Shao Q, Liu QH. Biomarkers in retinoblastoma. Int J Ophthalmol 2020; 13:325-341. [PMID: 32090044 DOI: 10.18240/ijo.2020.02.18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma (RB) is the most common intraocular malignancy of childhood caused by inactivation of the Rb genes. The prognosis of RB is better with an earlier diagnosis. Many diagnostic approaches and appropriate clinical treatments have been developed to improve clinical outcomes. However, limitations exist when utilizing current methods. Recently, many studies have identified identify new RB biomarkers which can be used in diagnosis, as prognostic indicators and may contribute to understanding the pathogenesis of RB and help determine specific treatment strategies. This review focuses on recent advances in the discovery of RB biomarkers and discusses their clinical utility and challenges from areas such as epigenetics, proteomics and radiogenomics.
Collapse
Affiliation(s)
- Jie Sun
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Hui-Yu Xi
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.,Department of Ophthalmology, Xuzhou First People's Hospital of Xuzhou Medical University, Xuzhou Eye Research Institute, Xuzhou 221002, Jiangsu Province, China
| | - Qing Shao
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Qing-Huai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
57
|
Ancona-Lezama D, Dalvin LA, Lucio-Alvarez JA, Jabbour P, Shields CL. OPHTHALMIC VASCULAR EVENTS AFTER INTRA‐ARTERIAL CHEMOTHERAPY FOR RETINOBLASTOMA. Retina 2019; 39:2264-2272. [DOI: 10.1097/iae.0000000000002315] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
58
|
Depression, Anxiety, and Stress in Parents of Patients With Retinoblastoma. Am J Ophthalmol 2019; 207:130-143. [PMID: 31163135 DOI: 10.1016/j.ajo.2019.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/25/2019] [Accepted: 05/24/2019] [Indexed: 11/20/2022]
Abstract
PURPOSE To assess depression, anxiety, and stress in parents of patients with retinoblastoma and to evaluate the impact of unifocal vs multifocal retinoblastoma. METHODS A cross-sectional, self-reported psychological assessment of parents of patients with retinoblastoma at a tertiary care ocular oncology center was performed. The Beck Depression Inventory-II (BDI), Beck Anxiety Inventory (BAI), The Parental Stress Index 4-Short Form, and a retinoblastoma Knowledge Assessment questionnaire were administered. Descriptive statistics for outcomes and comparative analyses were made. RESULTS There were 138 parents of children with retinoblastoma (unifocal: n = 77, multifocal: n = 61). Overall, parents displayed mild, moderate, or severe depression (BDI) (n = 37, 26.7%); mild, moderate, or severe anxiety (BAI) (n = 49, 35.8%), and stress scores within normal limits (n = 138, 100%). A comparison (unifocal vs multifocal) revealed parents of children with multifocal retinoblastoma with severe depression (1.4% vs 10.2%, P < .02), and no differences in anxiety or stress. Factors associated with moderate or severe parental depression included previous history of depression (30.0% vs 3.9%, P < .001) and factors for moderate or severe anxiety included previous history of depression (33.3% vs 8.6%, P < .001), parent highest level of education at high school or less vs college or beyond (29.2% vs 10.9%, P = .031), and parental report of "child developmental delay" (31.5% vs 11.3%, P = .019). CONCLUSIONS The majority of parents displayed minimal depression (73.3%), anxiety (64.2%), or stress (100%). However, severe depression is more often found in those whose children have multifocal disease, and previous history of depression and less education can impact psychological function. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
|
59
|
Patnaik SK, Halder N, Chawla B, Maithani D, Thavaraj V, Biswas NR, Velpandian T. Comparison of ocular pharmacokinetics of etoposide and its nanoemulsion after subtenon administration in rabbits. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2018-0108. [PMID: 31494629 DOI: 10.1515/jbcpp-2018-0108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/12/2019] [Indexed: 06/10/2023]
Abstract
Background Subtenon anticancer drugs are given as an adjunct to systemic chemotherapy for conditions like retinoblatoma. This study evaluated the ocular kinetics of nano-emulsion formulation of etoposide (NanoEt) and compared it with an equal dose of commercially available alcohol-based etoposide formulation in healthy rabbits. Methods A nanoemulsion formulation of NanoEt was developed and then evaluated for its ocular kinetics by subtenon administration in healthy rabbits. After the sterile subtenon administration of the drug, the eyes were enucleated after CO2 euthanasia at time intervals of 2 h, 6 h, 12 h, and 24 h, and ocular tissues, blood, and plasma were separated. The concentration of etoposide in the ocular tissues and blood was quantified using liquid chromatography tandem mass spectrometry (LC MS/MS). Results This study found that subtenon injection of NanoEt showed 24 times higher concentration in rabbit retina compared to an equal dose of conventional marketed formulation. Based on the ocular tissue bioavailability calculations (AUC0-24), the present study revealed that the formulation enhanced 90% ocular bioavailability of etoposide, when it was injected in the form of nano-emulsion in most of the tissues. Conclusions NanoEt has better bioavailability compared to the commercial alcohol-based formulation for subtenon injection. Low systemic exposure showed further advantage for its projected use in retinoblastoma (Rb) as an adjunct therapy. Further studies in Rb animal models are required to evaluate its safety and efficacy, for its clinical utility.
Collapse
Affiliation(s)
| | - Nabanita Halder
- Ocular Pharmacology and Pharmacy Division, Dr. R.P Centre, AIIMS, New Delhi, India
| | - Bhavna Chawla
- Department of Ophthalmology, AIIMS, New Delhi, India
| | - Deepti Maithani
- School of Pharmaceutical Sciences, Shobhit University, Uttar Pradesh, India
| | | | | | | |
Collapse
|
60
|
Manjandavida FP, Stathopoulos C, Zhang J, Honavar SG, Shields CL. Intra-arterial chemotherapy in retinoblastoma - A paradigm change. Indian J Ophthalmol 2019; 67:740-754. [PMID: 31124482 PMCID: PMC6552585 DOI: 10.4103/ijo.ijo_866_19] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intra-arterial chemotherapy (IAC), also known as superselective ophthalmic artery chemotherapy or chemosurgery, is currently widely accepted as one of the primary treatment modalities for intraocular retinoblastoma worldwide. Following the introduction of the technique in 1998, IAC has evolved over the past decades to be safer and more effective. Accumulated evidence shows that IAC is more effective in providing eye salvage in group D and E retinoblastoma as compared to conventional systemic intravenous chemotherapy (IVC). In contrast to IVC, IAC has the added benefits of reduced overall treatment duration and minimal systemic toxicity. This review provides a comprehensive update on the history, technique, indications, contraindications, and outcome of IAC. We have also identified the strengths, weaknesses, opportunities and threats (SWOT analysis) of the technique in this review.
Collapse
Affiliation(s)
- Fairooz P Manjandavida
- Ocular Oncology Service, HORUS Specialty Eye Care and Prabha Eye Clinic, Bangalore, India; Women's and Children's Hospital, Guangzhou, China
| | | | - Jing Zhang
- Women's and Children's Hospital, Guangzhou, China
| | | | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
61
|
LncRNA TP73-AS1 down-regulates miR-139-3p to promote retinoblastoma cell proliferation. Biosci Rep 2019; 39:BSR20190475. [PMID: 31015368 PMCID: PMC6509052 DOI: 10.1042/bsr20190475] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 01/18/2023] Open
Abstract
Our study aimed to investigate the role of long non-coding RNAs (lncRNA) TP73-AS1 in retinoblastoma (Rb). In the present study, we found that TP73-AS1 was up-regulated, while miR-139–3p was down-regulated in Rb. TP73-AS1 and miR-139-3p were inversely correlated in Rb tissues. In cells of Rb cell lines, overexpression of miR-139-3p failed to affect TP73-AS1, while TP73-AS1 overexpression caused the down-regulated miR-139-3p. TP73-AS1 overexpression caused promoted proliferation of Rb cells but showed no significant effects on cell migration and invasion. miR-139-3p overexpression played an opposite role and attenuated the effects of TP73-AS1 overexpression. Therefore, lncRNA TP73-AS1 may down-regulate miR-139-3p to promote Rb cell proliferation.
Collapse
|
62
|
Chueh HW. Current Assessment and Management of Retinoblastoma. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2019. [DOI: 10.15264/cpho.2019.26.1.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Hee Won Chueh
- Department of Pediatrics, Dong-A University Hospital, Dong-A University School of Medicine, Busan, Korea
| |
Collapse
|
63
|
Sioufi K, Say EAT, Ferenczy SC, Leahey AM, Shields CL. OPTICAL COHERENCE TOMOGRAPHY ANGIOGRAPHY FINDINGS OF DEEP CAPILLARY PLEXUS MICROISCHEMIA AFTER INTRAVENOUS CHEMOTHERAPY FOR RETINOBLASTOMA. Retina 2019; 39:371-378. [DOI: 10.1097/iae.0000000000001973] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
64
|
Wang L, Lyu X, Ma Y, Wu F, Wang L. MicroRNA‑504 targets AEG‑1 and inhibits cell proliferation and invasion in retinoblastoma. Mol Med Rep 2019; 19:2935-2942. [PMID: 30720088 DOI: 10.3892/mmr.2019.9923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/14/2018] [Indexed: 11/06/2022] Open
Abstract
The dysregulation of microRNAs (miRNAs/miRs) has become increasingly recognized as a primary feature of retinoblastoma (RB). Furthermore, miRNAs have been demonstrated to be involved in the occurrence and development of RB. Therefore, it is crucial to investigate the expression profile and roles of miRNAs in RB in order to identify potential therapeutic targets to treat patients with RB. The expression profile and biological roles of miRNA‑504 (miR‑504) have been reported in numerous types of human cancer; however, the roles of miR‑504 in RB remain unknown. In the present study, it was demonstrated that miR‑504 expression was significantly decreased in RB tissues and cell lines. Functional analysis identified that resumption of miR‑504 expression suppressed cell proliferation and invasion in RB. Furthermore, astrocyte elevated gene‑1 (AEG‑1) was determined to be a direct target of miR‑504 in RB, and a negative correlation between miR‑504 and AEG‑1 mRNA expression levels was observed in RB tissues. Additionally, the tumor‑suppressing effects of miR‑504 overexpression in RB cells could be rescued by AEG‑1 upregulation. In conclusion, these results indicated a significant role of the miR‑504/AEG‑1 pathway in inhibiting the aggressiveness of RB, suggesting that this miRNA may be employed as a therapeutic target for the treatment of patients with this disease.
Collapse
Affiliation(s)
- Lina Wang
- Department of Ophthalmology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xueman Lyu
- Department of Ophthalmology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yunqing Ma
- Department of Intensive Care Unit, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Fei Wu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ling Wang
- Department of Ophthalmology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
65
|
Abstract
Purpose: Refractory or recurrent vitreous seeds account for a large proportion of failure of eye salvage in retinoblastoma. The purpose of this study is to evaluate the efficacy of periocular topotecan (POT) in the management of vitreous seeds in retinoblastoma. Methods: Retrospective, interventional study of patients with retinoblastoma with vitreous seeds who received POT concurrent with intravenous chemotherapy (IVC). Results: Thirty-eight eyes of 35 patients received POT. Five eyes (13%) belonged to International Classification of Retinoblastoma group C, 23 eyes (61%) belonged to group D, and 10 eyes (26%) belonged to group E. Primary treatment included IVC with a combination of carboplatin, etoposide, and vincristine for a mean of 6 cycles (median 6; range 6–9). Concurrent to IVC from the fourth cycle onward, all patients received POT. Focal vitreous seeds were present in 20 eyes (53%) which received a mean of 3 injections (median 3; range 1–7). Diffuse vitreous seeds were present in 18 eyes (47%) which received a mean of 4 injections (median 5; range 1–7). At a mean follow-up of 8.5 months (median 5 months; range 1–15 months), regression of focal and diffuse vitreous seeds was achieved in 16 eyes (80%) and 8 eyes (44%), respectively. In all, 24 eyes (63%) had complete remission of vitreous seeds with POT given concurrently with IVC. Eye salvage was possible in 19 eyes (95%) with focal vitreous seeds and 12 eyes (68%) with diffuse VS. Enucleation was necessary for persistent vitreous seeds and viable tumor in five eyes (13%), viable tumor alone in one eye (0.02%), and recurrent vitreous seeds in one eye (0.02%). None of the patients developed systemic metastasis. Conclusion: POT administered concurrent with IVC is safe and effective in the initial management of vitreous seeds.
Collapse
Affiliation(s)
- Purnima R Sthapit
- National Retinoblastoma Foundation, Department of Ophthalmic and Facial Plastic Surgery and Ocular Oncology, Centre for Sight Superspeciality Eye Hospital, Hyderabad, Telangana, India
| | - Raksha Rao
- National Retinoblastoma Foundation, Department of Ophthalmic and Facial Plastic Surgery and Ocular Oncology, Centre for Sight Superspeciality Eye Hospital, Hyderabad, Telangana, India
| | - Santosh G Honavar
- National Retinoblastoma Foundation, Department of Ophthalmic and Facial Plastic Surgery and Ocular Oncology, Centre for Sight Superspeciality Eye Hospital, Hyderabad, Telangana, India
| |
Collapse
|
66
|
Feasibility of intra-arterial chemotherapy for retinoblastoma: experiences in a large single center cohort study. Neuroradiology 2019; 61:351-357. [DOI: 10.1007/s00234-019-02153-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
|
67
|
|
68
|
TREFOIL FACTOR FAMILY 1 EXPRESSION CORRELATES WITH CLINICAL OUTCOME IN PATIENTS WITH RETINOBLASTOMA. Retina 2018; 38:2422-2428. [DOI: 10.1097/iae.0000000000001881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
69
|
Dalvin LA, Ancona-Lezama D, Lucio-Alvarez JA, Masoomian B, Jabbour P, Shields CL. Ophthalmic Vascular Events after Primary Unilateral Intra-arterial Chemotherapy for Retinoblastoma in Early and Recent Eras. Ophthalmology 2018; 125:1803-1811. [DOI: 10.1016/j.ophtha.2018.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/18/2018] [Accepted: 05/09/2018] [Indexed: 02/02/2023] Open
|
70
|
Tang LJ, Zhou LJ, Zhang WX, Lin JY, Li YP, Yang HS, Zhang P. Expression of multidrug-resistance associated proteins in human retinoblastoma treated by primary enucleation. Int J Ophthalmol 2018; 11:1463-1466. [PMID: 30225219 DOI: 10.18240/ijo.2018.09.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/09/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To reveal the expression of multidrug-resistance associated proteins: glutathione-S-transferase π (GSTπ), P-glycoprotein (P-gp) and vault protein lung resistance protein (LRP) in retinoblastoma (RB) without any conservative treatment before primary enucleation and to correlate this expression with histopathological tumor features. METHODS A total of 42 specimens of RB undergone primary enucleation were selected for the research. Sections from the formalin-fixed, paraffin-embedded specimens were stained with HE and immunohistochemistry to detect the expression of GSTπ, P-gp and LRP. RESULTS GSTπ was expressed in 39/42 (92.86%) RBs and in 9/9 (100%) well-differentiated RBs. P-gp/GSTπ was found in 30 (71.42%) of 42 RBs. Totally 9 (21.43%) tumors were well differentiated and 33 (78.57%) were poorly differentiated. Totally 15 (35.71%) eyes had optic nerve (ON) tumor invasion, 36 (85.71%) had choroidal tumor invasion, and 14 (33.33%) had simultaneous choroidal and ON invasion. There was no statistically significant relationship between P-gp, GSTπ, LRP positivity and the degree of ocular layer tumor invasion and ON tumor invasion (P>0.05). CONCLUSION RB intrinsically expresses GSTπ, P-gp and LRP. GSTπ expression is positive in 100% well-differentiation ones, so in which way it is correlated with differentiation. But the other two proteins expressions are not related to tumor differentiation and to the degree of tumor invasion. GSTπ may be a new target of chemotherapy in RB.
Collapse
Affiliation(s)
- Li-Juan Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Li-Jun Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Wen-Xin Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Jian-Yan Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Yong-Ping Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Hua-Sheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, Guangdong Province, China
| |
Collapse
|
71
|
Spencer MA, Welch RJ, Shields CL. Hand-held Optical Coherence Tomography Monitoring of Submillimeter Retinoblastoma Treated with Indocyanine Green-enhanced Transpupillary Therapy. Middle East Afr J Ophthalmol 2018; 25:108-110. [PMID: 30122857 PMCID: PMC6071346 DOI: 10.4103/meajo.meajo_280_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Over recent years, hand-held optical coherence tomography (HH-OCT) has become critical for retinoblastoma diagnosis and management. We report precise HH-OCT findings in a case of sub-millimeter retinoblastoma treated with foveal-sparing indocyanine green-enhanced transpupillary thermotherapy (ICG-TTT). A 2-month-old Caucasian female with bilateral Group B retinoblastoma showed two recurrent macular tumors in the right eye, demonstrating 88 μm and 37 μm of growth to 344 μm and 413 μm in thickness, respectively, on HH-OCT. Each was treated with additional intravenous chemotherapy and foveal-sparing ICG-TTT. Tumor regression to 154 μm and 224 μm was documented on HH-OCT and maintained on follow-up. HH-OCT is vital in confirming clinical findings and influencing management decisions in retinoblastoma. In this case, HH-OCT precisely documented submillimeter retinoblastoma recurrence and treatment response.
Collapse
Affiliation(s)
- Meredith A Spencer
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - R Joel Welch
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
72
|
Yu MD, Dalvin LA, Welch RJ, Shields CL. Precision Intravitreal Chemotherapy for Localized Vitreous Seeding of Retinoblastoma. Ocul Oncol Pathol 2018; 5:284-289. [PMID: 31367592 DOI: 10.1159/000491432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/20/2018] [Indexed: 11/19/2022] Open
Abstract
Purpose To describe and evaluate the effectivity of a novel precision intravitreal injection technique for select cases of localized vitreous seeding in retinoblastoma. Methods Patients with localized vitreous seeds from retinoblastoma received intravitreal chemotherapy via a precision injection technique (P-IVitC) intended to optimize local delivery of melphalan to target seeds. The needle was inserted trans-pars plana and chemotherapy deposited within 3 mm of the target seed(s) under indirect ophthalmoscopic guidance. Without subsequent globe manipulation, the head was then positioned to promote gravity-dependent settling of chemotherapy. Results There were 8 eyes of 8 patients treated with P-IVitC for active vitreous seeds following adequate solid tumor control with intravenous chemotherapy (n = 3, 38%) or intra-arterial chemotherapy (n = 5, 63%). Viable vitreous seeds were primarily solitary spheres (n = 6, 75%) and measured an average of 2.7 mm in diameter. Vitreous seed resolution (n = 8, 100%) was noted after a mean of 2.6 injections (median 2; range 1-3) of 20 μg melphalan. There was no recurrence at a mean follow-up of 10.0 months. Only 1 eye had an associated complication (focal retinal pigment epithelial mottling, n = 1, 13%). Conclusions With direct delivery of chemotherapy to within 3-4 mm of vitreous seed activity, P-IVitC provided complete control of localized vitreous seeds with minimal toxicity.
Collapse
Affiliation(s)
- Michael D Yu
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lauren A Dalvin
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - R Joel Welch
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
73
|
Dalvin LA, Kumari M, Essuman VA, Shohelly Shipa S, Ancona-Lezama D, Lucio-Alvarez JA, Jabbour P, Shields CL. Primary Intra-Arterial Chemotherapy for Retinoblastoma in the Intravitreal Chemotherapy Era: Five Years of Experience. Ocul Oncol Pathol 2018; 5:139-146. [PMID: 30976594 DOI: 10.1159/000491580] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/26/2018] [Indexed: 11/19/2022] Open
Abstract
Purpose To report our 5-year experience with intra-arterial chemotherapy (IAC) in the intravitreal chemotherapy (IvitC) era. Methods Retrospective review of retinoblastoma treated with primary unilateral IAC in the IvitC era (2012-2017). Results There were 34 eyes treated with IAC alone versus 20 eyes treated with IAC plus IvitC for vitreous seeds. IAC (IAC alone vs. IAC plus IvitC) consisted of melphalan (41 vs. 10%) or melphalan plus topotecan (59 vs. 90%, p = 0.03). IvitC consisted of melphalan (60%) or melphalan plus topotecan (40%). Tumor control and globe salvage were achieved in 100% of group B and C eyes without IvitC. Despite more extensive vitreous seeds in the IvitC group (p < 0.01), comparison of IAC alone versus IAC plus IvitC revealed no difference in tumor control for group D (88 vs. 69%, p = 0.36) or group E (67 vs. 100%, p = 0.25) and no difference in globe salvage for group D (88 vs. 69%, p = 0.36) or group E (58 vs. 57%, p = 0.39). Conclusions IAC is effective as primary therapy for unilateral group B, C, D, and E retinoblastoma. IvitC is an important adjuvant therapy to achieve comparable globe salvage rates for group D and E eyes with persistent active vitreous seeds.
Collapse
Affiliation(s)
- Lauren A Dalvin
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mamta Kumari
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vera Adobea Essuman
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Ophthalmology Unit, Department of Surgery, School of Medicine and Dentistry, University of Ghana, Accra, Ghana
| | - Shormin Shohelly Shipa
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - David Ancona-Lezama
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - J Antonio Lucio-Alvarez
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Pascal Jabbour
- Department of Endovascular Neurosurgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
74
|
Shang Y. LncRNA THOR acts as a retinoblastoma promoter through enhancing the combination of c-myc mRNA and IGF2BP1 protein. Biomed Pharmacother 2018; 106:1243-1249. [PMID: 30119193 DOI: 10.1016/j.biopha.2018.07.052] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 02/09/2023] Open
Abstract
Long non-coding RNA (lncRNA) THOR is an extremely conserved lncRNA with specifically expressed in testis while widespreadly exist in human multiple cancer tissues. The high expression of it significantly promotes the occurrence and progression of melanoma, non-small cell lung cancer, osteosarcoma and renal cell carcinoma. However, the expression pattern and effects of lncRNA THOR in the progression of retinoblastoma remain unclear. As a result, this study was conducted to discovery the expression and roles of lncRNA THOR in the malignant phenotype transformation of retinoblastoma cells, as well as its underlying mechanism. Our results demonstrated that lncRNA THOR was over-expressed in the retina tissues from retinoblastoma patients and retinoblastoma Y79 and WERI-Rb1 cell lines. Down-regulation of lncRNA THOR with siRNA significantly repressed cell growth, migration and S phase accumulation, while induced cell apoptosis and G1 phase reduction and reduced the expression of c-myc. Besides, knockdown of c-myc promoted cell apoptosis and suppressed cell proliferation. Furthermore, RNA pull down and PIP assays showed that up-regulation of lncRNA THOR enhanced the combination of IGF2BP1 protein and c-myc RNA. And lncRNA THOR up-regulation obviously increased the tumorigenesis of Y79 cells in vivo. In conclusion, this study makes clear that lncRNA THOR is up-regulated in retinoblastoma, and its over-expression significantly enhances the malignant phenotype transformation of retinoblastoma cells through up-regulating c-myc expression via enhancing its combination with TGF2BP1 protein. Overall, our study illustrates that lncRNA THOR/c-myc molecular cascade might be another potent target for retinoblastoma treatment.
Collapse
Affiliation(s)
- Yamin Shang
- Department of Pediatrics, Huaihe Hospital of Henan University, Kaifeng, 475000, Henan, China.
| |
Collapse
|
75
|
Abramson DH, Ji X, Francis JH, Catalanotti F, Brodie SE, Habib L. Intravitreal chemotherapy in retinoblastoma: expanded use beyond intravitreal seeds. Br J Ophthalmol 2018; 103:488-493. [PMID: 29875233 DOI: 10.1136/bjophthalmol-2018-312037] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 05/09/2018] [Accepted: 05/12/2018] [Indexed: 11/04/2022]
Abstract
BACKGROUND/AIMS Ophthalmic artery chemosurgery (OAC) has changed the face of retinoblastoma treatment and led to a higher rate of globe salvage. The introduction of intravitreal chemotherapy (IVitC) has further enhanced globe salvage with increased success in treatment of intravitreal seeds. Our group has seen success at treating non-vitreous disease that is refractory to OAC using IVitC. This study was undertaken to quantify and report on this success. METHODS A retrospective review was used to identify patients treated with IVitC for indications other than vitreous seeds from two centres. The indication, prior and concurrent treatment, response time and duration of treatment were documented. Kaplan-Meier estimates were used to evaluate ocular and recurrence-free survival. Ocular toxicity was evaluated using the 30 Hz flicker electroretinogram (ERG). Continuous and categorical variables were compared with Student's t-test and χ2 test, respectively. RESULTS Fifty-six eyes from 52 retinoblastoma patients were identified. There were no disease-related or treatment-related deaths. One patient developed a second primary malignancy (pinealoblastoma) and subsequent leptomeningeal spread. Ninety-eight per cent of the eyes showed clinical regression. Recurrence was seen in 14.3%. Of the recurrences, five occurred in retinal tumours and three in subretinal seeds. The Kaplan-Meier estimated risk of recurrence in all patients treated was 83.5% (95% CI 7.9 to 14.1) at 10 months. The mean change in ERG over treatment course was -17.7 μV. CONCLUSIONS Intravitreal chemotherapy is successful for the treatment of subretinal seeds and recurrent retinal tumours and could be considered as adjunctive therapy in globe-sparing treatment of retinoblastoma.
Collapse
Affiliation(s)
- David H Abramson
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA.,Department of Ophthalmology, Weill Cornell University, New York, USA
| | - Xunda Ji
- Department of Ophthalmology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jasmine H Francis
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA.,Department of Ophthalmology, Weill Cornell University, New York, USA
| | | | - Scott E Brodie
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA.,Department of Ophthalmology, NYU School of Medicine, New York, USA
| | - Larissa Habib
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
76
|
Jansen RW, de Jong MC, Kooi IE, Sirin S, Göricke S, Brisse HJ, Maeder P, Galluzzi P, van der Valk P, Cloos J, Eekhout I, Castelijns JA, Moll AC, Dorsman JC, de Graaf P. MR Imaging Features of Retinoblastoma: Association with Gene Expression Profiles. Radiology 2018; 288:506-515. [PMID: 29714679 DOI: 10.1148/radiol.2018172000] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose To identify associations between magnetic resonance (MR) imaging features and gene expression in retinoblastoma. Materials and Methods A retinoblastoma MR imaging atlas was validated by using anonymized MR images from referral centers in Essen, Germany, and Paris, France. Images were from 39 patients with retinoblastoma (16 male and 18 female patients [the sex in five patients was unknown]; age range, 5-90 months; inclusion criterion: pretreatment MR imaging). This atlas was used to compare MR imaging features with genome-wide messenger RNA (mRNA) expression data from 60 consecutive patients obtained from 1995 to 2012 (35 male patients [58%]; age range, 2-69 months; inclusion criteria: pretreatment MR imaging, genome-wide mRNA expression data available). Imaging pathway associations were analyzed by means of gene enrichment. In addition, imaging features were compared with a predefined gene expression signature of photoreceptorness. Statistical analysis was performed with generalized linear modeling of radiology traits on normalized log2-transformed expression values. P values were corrected for multiple hypothesis testing. Results Radiogenomic analysis revealed 1336 differentially expressed genes for qualitative imaging features (threshold P = .05 after multiple hypothesis correction). Loss of photoreceptorness gene expression correlated with advanced stage imaging features, including multiple lesions (P = .03) and greater eye size (P < .001). The number of lesions on MR images was associated with expression of MYCN (P = .04). A newly defined radiophenotype of diffuse-growing, plaque-shaped, multifocal tumors displayed overexpression of SERTAD3 (P = .003, P = .049, and P = .06, respectively), a protein that stimulates cell growth by activating the E2F network. Conclusion Radiogenomic biomarkers can potentially help predict molecular features, such as photoreceptorness loss, that indicate tumor progression. Results imply a possible role for radiogenomics in future staging and treatment decision making in retinoblastoma.
Collapse
Affiliation(s)
- Robin W Jansen
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Marcus C de Jong
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Irsan E Kooi
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Selma Sirin
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Sophia Göricke
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Hervé J Brisse
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Philippe Maeder
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Paolo Galluzzi
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Paul van der Valk
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Jacqueline Cloos
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Iris Eekhout
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Jonas A Castelijns
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Annette C Moll
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Josephine C Dorsman
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| | - Pim de Graaf
- From the Departments of Radiology and Nuclear Medicine (R.W.J., M.C.d.J., J.A.C., P.d.G.), Clinical Genetics (I.E.K., J.C.D.), Ophthalmology (A.C.M.), Pathology (P.v.d.V.), Pediatric Oncology (J.C.), and Epidemiology and Biostatistics (I.E.), VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands; European Retinoblastoma Imaging Collaboration (ERIC) (R.W.J., M.C.d.J., S.S., S.G., H.J.B., P.M., P.G., J.A.C., P.d.G.); Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (S.S., S.G.); Department of Radiology, Institut Curie, Paris, France and Paris Sciences et Lettres Research University, Paris, France (H.J.B.); Department of Radiology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne, Lausanne, Switzerland (P.M.); and Unit of Neuroimaging and Neurointervention, Department of Neurosciences, Siena University Hospital, Siena, Italy (P.G.)
| |
Collapse
|
77
|
Galangin inhibits the cell progression and induces cell apoptosis through activating PTEN and Caspase-3 pathways in retinoblastoma. Biomed Pharmacother 2018; 97:851-863. [DOI: 10.1016/j.biopha.2017.09.144] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/11/2017] [Accepted: 09/26/2017] [Indexed: 12/17/2022] Open
|
78
|
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. [PMID: 28646513 DOI: 10.1002/pbc.26696] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND To evaluate outcomes of Group D retinoblastoma (Rb) eyes during the intravitreal melphalan era. PROCEDURE Retrospective chart review of patients diagnosed with Group D Rb from 2011 to 2016 was done. Overall, 76 Group D eyes of 68 patients were included; salvage therapy included systemic chemoreduction with vincristine, etoposide, and carboplatin with local consolidation, followed by intravitreal injection of melphalan for recurrent or persistent seeding. External beam radiation was not used as a treatment modality. Primary outcome measurement was globe salvage. RESULTS Of 76 Group D eyes, 24 were enucleated primarily and 52 were treated with intent to salvage the globe. Systemic chemoreduction salvaged 25 of 52 eyes (48%). Tumor recurrences were diagnosed in 27 eyes (52%); five with massive retinal recurrences underwent enucleation and 22 were treated with intravitreal melphalan injection. Of the 22 injected eyes, 14 (64%) were salvaged and eight required enucleation primarily for retinal recurrences. Success in eradicating vitreous seeds was 100%. The Kaplan-Meier 3-year survival estimate for treated eyes is 76.5% (95% CI: 61.4-86.3). Median follow-up for the group of 76 Group D eyes was 29.5 months (SD 17.9 months). CONCLUSION During a 6-year period that included the initiation of intravitreal melphalan at our institution, the salvage rate of treated Group D eyes was 75% (39/52 eyes). Intravitreal melphalan was utilized for ocular salvage in 42% (22/52 eyes). Systemic chemoreduction combined with intravitreal melphalan for seeding demonstrated a high overall salvage rate for Group D eyes in this cohort.
Collapse
Affiliation(s)
- Jesse L Berry
- USC Roski Eye Institute, Los Angeles, California.,The Vision Center at Children's Hospital Los Angeles, Los Angeles, California
| | - Sona Shah
- USC Roski Eye Institute, Los Angeles, California.,The Vision Center at Children's Hospital Los Angeles, Los Angeles, California
| | - Mercy Bechtold
- USC Roski Eye Institute, Los Angeles, California.,The Vision Center at Children's Hospital Los Angeles, Los Angeles, California
| | - Emily Zolfaghari
- The Vision Center at Children's Hospital Los Angeles, Los Angeles, California
| | - Rima Jubran
- The Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, California
| | - Jonathan W Kim
- USC Roski Eye Institute, Los Angeles, California.,The Vision Center at Children's Hospital Los Angeles, Los Angeles, California
| |
Collapse
|
79
|
Busch M, Papior D, Stephan H, Dünker N. Characterization of etoposide- and cisplatin-chemoresistant retinoblastoma cell lines. Oncol Rep 2017; 39:160-172. [PMID: 29192327 PMCID: PMC5783599 DOI: 10.3892/or.2017.6100] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022] Open
Abstract
Retinoblastoma (RB) is the most common malignant intraocular tumor in early childhood. Imminent chemotherapy resistance diminishes the clinical-therapeutic options and emphasizes the necessity for new therapeutic approaches. The present study aimed at characterizing and comparing etoposide and cisplatin-resistant human RB cell lines with regard to changes in proliferation and apoptosis levels, anchorage independent growth behavior in vitro as well as tumor formation capacity in vivo. The proliferation rates were significantly increased in the etoposide-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 reflecting significantly higher growth kinetics compared to the parental controls. In line with these findings in in vivo chicken chorioallantoic (CAM) assays, etoposide-resistant cell lines generated significantly increased numbers of tumors with higher tumor weights compared to their parental counterparts. In contrast to etoposide, the cisplatin-resistant RB cell lines Y-79, WERI-Rb1 and RB-355 displayed significantly increased apoptosis rates and reduced proliferation rates resulting in significantly decreased growth kinetics. Tumor formation capacity of cisplatin-resistant cell lines did not significantly change, and in comparison with parental controls cisplatin-resistant Y-79 cells displayed significantly reduced tumor weight. Soft agarose assays indicated that anchorage-independent growth of all chemotherapy-resistant cell lines analyzed was significantly decreased. Summarizing, one can state that etoposide-resistant RB cells behave more aggressively than the tumor cells of origin and potentially represent a risk factor for local relapse, while cisplatin-resistant cells show a significantly decreased tumorigenic potential.
Collapse
Affiliation(s)
- Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| | - David Papior
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Harald Stephan
- Division of Haematology and Oncology, Children's Hospital, University of Duisburg-Essen, D-45122 Essen, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, D-45122 Essen, Germany
| |
Collapse
|
80
|
Yarovaya V, Sioufi K, Shields CL. Parafoveolar retinoblastoma regression with foveal preservation following intra-arterial chemotherapy documented on hand-held optical coherence tomography in a newborn. Int J Retina Vitreous 2017; 3:43. [PMID: 29158915 PMCID: PMC5682635 DOI: 10.1186/s40942-017-0098-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/09/2017] [Indexed: 11/21/2022] Open
Abstract
Background Optical coherence tomography (OCT) has become an invaluable tool in retinoblastoma management, providing submillimeter visualization of tumor control following treatment. Herein, we document OCT-detection of a subtle tumor recurrence, allowing early intervention and achieving foveal microanatomy preservation. Case presentation A 3-week-old girl was diagnosed with bilateral familial retinoblastoma, classified as group D in the right eye (OD) and group B in the left eye (OS), and treated with intravenous chemoreduction. At 6-months follow-up, the right eye was under control, but the left eye revealed a subtle juxtafoveal tumor recurrence, documented on handheld OCT (HH-OCT) and measuring 2750 µm in diameter and 792 µm in thickness. Treatment with intraarterial chemotherapy (IAC) using 1 cycle of melphalan 5 mg was performed and complete tumor control was achieved, leaving a flat, concave scar 663 µm from the intact foveola and measuring 2750 µm in diameter and 120 µm in thickness. Foveal microanatomy OS was preserved on HH-OCT. The findings remained stable at 2 years following IAC. Conclusions HH-OCT is an important tool in retinoblastoma management. In this case, HH-OCT allowed for early detection of retinoblastoma recurrence, before foveal invasion. Following treatment with IAC, complete tumor regression was noted and foveal microanatomy remained intact.
Collapse
Affiliation(s)
- Vera Yarovaya
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, 840 Walnut Street, Suite 1440, 14th Floor, Philadelphia, PA 19107 USA
| | - Kareem Sioufi
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, 840 Walnut Street, Suite 1440, 14th Floor, Philadelphia, PA 19107 USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, 840 Walnut Street, Suite 1440, 14th Floor, Philadelphia, PA 19107 USA
| |
Collapse
|
81
|
Stenfelt S, Blixt MKE, All-Ericsson C, Hallböök F, Boije H. Heterogeneity in retinoblastoma: a tale of molecules and models. Clin Transl Med 2017; 6:42. [PMID: 29124525 PMCID: PMC5680409 DOI: 10.1186/s40169-017-0173-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 10/26/2017] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma, an intraocular pediatric cancer, develops in the embryonic retina following biallelic loss of RB1. However, there is a wide range of genetic and epigenetic changes that can affect RB1 resulting in different clinical outcomes. In addition, other transformations, such as MYCN amplification, generate particularly aggressive tumors, which may or may not be RB1 independent. Recognizing the cellular characteristics required for tumor development, by identifying the elusive cell-of-origin for retinoblastoma, would help us understand the development of these tumors. In this review we summarize the heterogeneity reported in retinoblastoma on a molecular, cellular and tissue level. We also discuss the challenging heterogeneity in current retinoblastoma models and suggest future platforms that could contribute to improved understanding of tumor initiation, progression and metastasis in retinoblastoma, which may ultimately lead to more patient-specific treatments.
Collapse
Affiliation(s)
- Sonya Stenfelt
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | - Maria K E Blixt
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | | | - Finn Hallböök
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden
| | - Henrik Boije
- Department of Neuroscience, Uppsala University, 75124, Uppsala, Sweden.
| |
Collapse
|
82
|
|
83
|
Regional intra-arterial steroid treatment in 120 patients with steroid-resistant or -dependent GvHD. Bone Marrow Transplant 2017. [PMID: 28650453 DOI: 10.1038/bmt.2017.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
GvHD results in death in the majority of steroid-resistant patients. This report assesses the safety and efficacy of two regional intra-arterial steroid (IAS) treatment protocols in the largest published cohort of patients with resistant/dependent hepatic and/or gastrointestinal GvHD, as well as identification of predictors of response to IAS and survival. One hundred and twenty patients with hepatic, gastrointestinal GvHD or both were given IAS. Gastrointestinal initial response (IR) and complete response (CR) were documented in 67.9% and 47.6%, respectively, whereas hepatic IR/CR in 54.9% and 33.3%, respectively. The predictors of gastrointestinal CR were lower peak GvHD and steroid-dependent (SD) GvHD. The predictors for hepatic CR were male patient, reduced intensity conditioning and SD GvHD. Twenty-six of the 120 patients (21.6%) are currently alive (median follow-up for the survivors 91.5 months). The 12 months' overall survival is 30% with no treatment-associated deaths. Predictors of 12 months' survival were as follows: first transplant, age<20 years, non-TBI regimen and GvHD CR. Shorter time to gastrointestinal IR but not time to hepatic IR was associated with improved 12 months' survival. IAS appears to be safe and effective. Gastrointestinal treatment is more effective than hepatic treatment. In our study, we conclude our current recommendations for IAS treatment.
Collapse
|
84
|
SOST silencing promotes proliferation and invasion and reduces apoptosis of retinoblastoma cells by activating Wnt/β-catenin signaling pathway. Gene Ther 2017; 24:399-407. [DOI: 10.1038/gt.2017.31] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 04/20/2017] [Accepted: 04/26/2017] [Indexed: 12/25/2022]
|
85
|
Magan T, Khoo CTL, Jabbour PM, Shields CL. INTRAARTERIAL CHEMOTHERAPY FOR RETINOBLASTOMA IN A 2-MONTH-OLD INFANT. Retin Cases Brief Rep 2017; 11:24-26. [PMID: 26756523 DOI: 10.1097/icb.0000000000000279] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
PURPOSE Intraarterial chemotherapy for retinoblastoma is usually reserved for infants aged 3 months or older because of the intricacy of the newborn vascular anatomy making the procedure technically challenging. The authors report a successful case of intraarterial chemotherapy performed in a 2-month-old infant using a minimal exposure approach. METHODS Case report. RESULTS A 2-month-old infant presented with leukocoria and was subsequently diagnosed with an exophytic Group D retinoblastoma in the right eye. The infant received melphalan 3 mg delivered into the ostium of the ophthalmic artery of the right eye under fluoroscopic guidance. Examination under anesthesia a month later showed complete tumor regression to a calcified Type I scar. After a second cycle of intraarterial chemotherapy, no further treatment was necessary. There were no complications. CONCLUSION Intraarterial chemotherapy is generally used for retinoblastoma in infants aged 3 months or older. The patient was successfully catheterized and treated at 2 months of age, with complete tumor regression after a single chemotherapy dose. Thus, in expert hands, intraarterial chemotherapy can be considered in such young infants.
Collapse
Affiliation(s)
- Tejal Magan
- *Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, Pennsylvania; †St George's, University of London, London, United Kingdom; ‡Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; and §Division of Neurovascular and Endovascular Surgery, Neurosurgery Department, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
86
|
Long non-coding RNA HOTAIR regulates proliferation and invasion via activating Notch signalling pathway in retinoblastoma. J Biosci 2017; 41:677-687. [PMID: 27966488 DOI: 10.1007/s12038-016-9636-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Retinoblastoma is the most frequently occurring tumour in the eyes in early childhood. Novel targets that are important for the diagnosis or treatment of retinoblastoma could be valuable in increasing the survival rate of patients affected by this disease. Long non-coding RNAs (lncRNAs) are a recently discovered type of RNAs with no proteincoding function; yet it has become increasingly clear that lncRNAs are responsible for important gene regulatory functions in various diseases. In this study, the expression of lncRNA HOTAIR was measured by qRT-PCR, and HOTAIR expression was found to be significantly upregulated in human retinoblastomas tissues as compared with that in paracancerous tissues. Knockdown of HOTAIR restricted the proliferation and invasion of the more invasive retinoblastoma Y79 cells, and led to G0/G1 arrest, possibly through inhibiting phospho-RB1, RB1 and CCNE. Furthermore, we found that the Notch signalling pathway was activated abnormally in retinoblastoma cell lines, while knockdown of HOTAIR attenuated the endogenous Notch signalling pathway in vitro and in vivo. In addition, knockdown of HOTAIR could inhibit the tumour progression in a xenograft model of retinoblastoma. In summary, our findings indicate that HOTAIR may play important roles in retinoblastoma progression via Notch pathway. HOTAIR has the potential to enhance the development of novel targeted diagnostic and therapeutic approaches for retinoblastoma.
Collapse
|
87
|
Gonzalez-Montpetit ME, Samara WA, Magrath GN, Shields CL. Detection of Minimally Visible Recurrent Retinoblastoma by Hand-held Spectral-Domain Optical Coherence Tomography. J Pediatr Ophthalmol Strabismus 2017; 54:e6-e8. [PMID: 28196267 DOI: 10.3928/01913913-20170201-03] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 11/03/2016] [Indexed: 11/20/2022]
Abstract
A 2-month-old male infant with bilateral familial retinoblastoma was treated with intravenous chemotherapy and focal adjuvant therapy. At 5 months of follow-up, fundus examination and ultrasonography disclosed no recurrence; however, hand-held spectral-domain optical coherence tomography (SD-OCT) demonstrated subclinical recurrence within a previous regression scar. Subsequent treatment led to flat scar. Hand-held SD-OCT can be a useful tool for detection of subclinical recurrent retinoblastoma. [J Pediatr Ophthalmol Strabismus. 2017;54:e6-e8.].
Collapse
|
88
|
Stathopoulos C, Say EAT, Shields CL. Intra-arterial and Intravitreal Chemotherapy for Retinoblastoma. CURRENT OPHTHALMOLOGY REPORTS 2017. [DOI: 10.1007/s40135-017-0123-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
89
|
Long noncoding RNA BDNF-AS is a potential biomarker and regulates cancer development in human retinoblastoma. Biochem Biophys Res Commun 2017; 497:1142-1148. [PMID: 28131827 DOI: 10.1016/j.bbrc.2017.01.134] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Long non-coding RNAs (lncRNA) have been shown to play important roles in human cancer. We examined expression, prognostic potential and functional roles of lncRNA, brain-derived neurotrophic factor antisense (BDNF-AS) in human retinoblastoma (RB). METHODS BDNF-AS expression in RB tumors was characterized according to the clinicopathological parameters of patients. BDNF-AS mRNA level was compared between RB tumors and normal retinas, as well as RB cell lines and normal retinal epithelial cells. RB patients' overall survival was compared between those with low and high BDNF-AS tumor expressions. Statistical analysis was performed to examine the independence of BDNF-AS being cancer biomarker in RB. In Y79 and WERI-Rb-1 cells, BDNF-AS was upregulated. It's effect on cancer proliferation, migration and cell-cycle transition were assessed. RESULTS BDNF-AS is downregulated in RB tumors and cell lines. Low BDNF-AS expression in RB tumors is correlated with patients' advanced clinical stage and tumor differentiation status. Low BDNF-AS expression is associated with shorter overall survival and may be acting as an independent marker in RB. In Y79 and WERI-Rb-1 cells, forced overexpression of BDNF-AS inhibited cancer proliferation and migration. It also induced cell-cycle arrest at G0/G1 phase by downregulating CDC42, Cyclin E and BDNF. CONCLUSION BDNF-AS is lowly expressed, and may be used as a prognostic biomarker in RB. Upregulating BDNF-AS has inhibitory effect on RB development, probably through the suppression of cell-cycle transition.
Collapse
|
90
|
Shields CL, Shields JA. The American Society of Retina Specialists 2016 Founders Award Lecture. ACTA ACUST UNITED AC 2017. [DOI: 10.1177/2474126416677898] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Purpose: To review the current state of the art of retinal tumors with respect to clinical features, imaging, and management. Methods: Review of published literature and personal experience from an ocular oncology service. Results: There are several tumors that arise from the sensory retina including those of glial origin (astrocytic hamartoma, acquired astrocytoma, and solitary circumscribed retinal astrocytic proliferation), neural origin (retinoblastoma), and vascular origin (hemangioblastoma, cavernous hemangioma, racemose hemangioma, and vasoproliferative tumor). Most retinal tumors are benign with the exception of retinoblastoma and retinal metastasis. Some retinal tumors are associated with systemic oculoneurocutaneous syndromes such as tuberous sclerosis complex (TSC), 13q deletion syndrome, Von Hippel–Lindau disease, and systemic cavernous and racemose hemangioma syndromes with ocular, cutaneous, neurologic, and other findings. Regarding management, recent data indicate that retinal astrocytic hamartoma and retinal achromic patch are key to the diagnosis of TSC and imply greater risk for brain and kidney tumors. Some children with TSC respond to mammalian target of rapamycin inhibitors, controlling brain, kidney, and eye tumors. Children with retinoblastoma are all screened genetically, and systemic evaluation is provided for detection of metastasis and secondary-related tumors. Newer chemotherapy regimens by intravenous or intra-arterial routes have revolutionized retinoblastoma control. Retinal vascular tumors are uncommon but can have serious systemic implications. Therefore, imaging of the brain for related cerebellar hemangioblastoma, midbrain racemose hemangioma, or multifocal cavernomas is performed. Genetic testing for each disease can be revealing. Conclusions: Retinal tumors are often benign, can be recognized by clinical and imaging features, and can be associated with important systemic syndromes.
Collapse
Affiliation(s)
- Carol L. Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jerry A. Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
91
|
Shao Y, Yu Y, Zong R, Quyang L, He H, Zhou Q, Pei C. Erlotinib has tumor inhibitory effect in human retinoblastoma cells. Biomed Pharmacother 2016; 85:479-485. [PMID: 27899256 DOI: 10.1016/j.biopha.2016.11.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
AIM In this study, we explored the effect of erlotinib on the development of retinoblastoma (RB) cells both in vitro and in vivo. METHOD RB cell lines, Y79 and WERI cells were treated with various concentrations of erlotinib in vitro to assess their cytotoxic profiles. In vitro proliferation, cell-cycle transition and migration were compared between RB cells treated with erlotinib and cells without erlotinib treatment. In in vivo tumorigenicity assay, mice were injected with Y79 cells and orally fed with erlotinib for 28days. The effect of erlotinib on in vivo tumor grafts was then assessed. Western blot analysis on EGFR, ERK, AKT proteins and their phosphorylated proteins was also performed to assess molecular signaling pathways of associated with erlotinib in RB cells. RESULTS In vitro erlotinib treatment induced cytotoxicity in Y79 and WERI cells in dose-dependent manner. While Y79 and WERI cells were treated with erlotinib close to EC50 concentrations for 3days, RB proliferation, cell-cycle transition and migration were all significantly inhibited. In in vivo tumorigenicity assay, oral induction of erlotinib also dramatically reduced the growth of Y79 tumor grafts. Western blot demonstrated that, in in vitro RB cells, erlotinib did not alter the protein expression levels of EGFR, ERK or AKT, but significantly reduced the expressions of phosphorylated EGFR, ERK and AKT proteins. CONCLUSION Erlotinib was shown to have tumor suppressive effect on RB growth in vitro and in vivo, possibly through the inhibition on EGFR, ERG/AKT signaling pathways.
Collapse
Affiliation(s)
- Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Province clinical ophthalmology Institute, Nanchang, Jiangxi Province, 330006, China.
| | - Yao Yu
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Province clinical ophthalmology Institute, Nanchang, Jiangxi Province, 330006, China; Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi Province, 330009, China
| | - Rongrong Zong
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian Province, 361102, China
| | - Luowa Quyang
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian Province, 361102, China
| | - Hui He
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian Province, 361102, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Province clinical ophthalmology Institute, Nanchang, Jiangxi Province, 330006, China.
| | - Chonggang Pei
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Province clinical ophthalmology Institute, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
92
|
Ravindran S. Carol Shields, MD. Ophthalmology 2016; 123:2444-2445. [PMID: 27871392 DOI: 10.1016/j.ophtha.2016.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022] Open
|
93
|
Shields CL, Alset AE, Say EAT, 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; 53:275-84. [PMID: 27486728 DOI: 10.3928/01913913-20160719-04] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 05/03/2016] [Indexed: 11/20/2022]
Abstract
PURPOSE To compare outcomes of intra-arterial chemotherapy for retinoblastoma as primary therapy before (Era I) and during (Era II) the intravitreal chemotherapy era. METHODS In this retrospective interventional case series at a tertiary referral center, 66 eyes of 66 patients with untreated unilateral retinoblastoma were used. intraarterial chemotherapy into the ophthalmic artery under fluoroscopic guidance was performed using melphalan in every case, with additional topotecan as necessary. Intravitreal chemotherapy using melphalan and/or topotecan was employed as needed for active vitreous seeding. Globe salvage was measured based on the International Classification of Retinoblastoma (ICRB) during two eras. RESULTS The two eras encompassed 2008 to 2012 (intraarterial chemotherapy alone, Era I) and 2012 to 2015 (intraarterial chemotherapy plus intravitreal chemotherapy, Era II). Over this period, there were 66 patients with unilateral untreated retinoblastoma treated with primary intra-arterial chemotherapy. A comparison of features (Era I vs Era II) revealed no significant difference in mean patient age (24 vs 24 months), ICRB groups, mean largest tumor diameter (19 vs 17 mm), mean largest tumor thickness (10 vs 10 mm), vitreous seed presence (56% vs 59%), subretinal seed presence (67% vs 62%), retinal detachment (70% vs 66%), or vitreous hemorrhage (0% vs 5%). There was no significant difference in mean number of intra-arterial chemotherapy cycles (3 vs 3.1) or intraarterial chemotherapy dosages. Following therapy, there was a significant difference (Era I vs Era II) in the need for enucleation overall (44% vs 15%, P = .012), especially for group E eyes (75% vs 27%, P = .039). Four of the eyes that initiated therapy in Era I later required intravitreal chemotherapy during Era II. The enucleation rate was 0% for groups B and C in both eras and non-significant for group D (23% vs 13%). There were no patients with stroke, seizure, limb ischemia, extraocular tumor extension, secondary leukemia, metastasis, or death. CONCLUSIONS The current era of retinoblastoma management using intra-arterial chemotherapy plus additional intravitreal chemotherapy (as needed for vitreous seeding) has improved globe salvage in eyes with advanced retinoblastoma. [J Pediatr Ophthalmol Strabismus. 2016;53(5):275-284.].
Collapse
|
94
|
Chuluunbat T, Jamiyanjav B, Munkhuu B, Bazarsad U, Molom A, Kao LY, Wu WC. Retinoblastoma in Mongolia: Clinical characteristics and survival from 1987 to 2014. Taiwan J Ophthalmol 2016; 6:79-84. [PMID: 29018716 PMCID: PMC5602694 DOI: 10.1016/j.tjo.2016.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/07/2016] [Accepted: 04/09/2016] [Indexed: 11/16/2022] Open
Abstract
Background/Purpose: This study aims to describe the clinical characteristics and treatment outcome of retinoblastoma in Mongolian children. Methods: Data of all children diagnosed with retinoblastoma at the National Center for Maternal and Child Health of Mongolia from 1987 to January 2014 were reviewed retrospectively. The ICRB classification was used. Survival characteristics of the cohort were analyzed. Results: Retinoblastoma was diagnosed in 79 eyes of 64 cases during the study period. Median age of diagnosis was 24.5 ± 15.8 months. There were no differences in sex ratio, and 15 cases (23%) were bilateral. Forty-three (67%) patients were from rural areas. The more frequent clinical presentations were leukocoria in 50 (78%) patients, strabismus in 24 (38%) patients, and glaucoma in 21 (33%) patients. Sixty-one (95%) patients were diagnosed with Classification D or worse when presented to us. Due to late diagnosis in the majority of cases, unilateral and bilateral enucleations were performed in 48 (61%) eyes and 24 (30%) eyes, respectively; exenteration was done in three (4%) eyes. Fifty-two (81%) patients received chemotherapy and 13 (8.3%) patients underwent external beam radiation after enucleation. At the time of last follow-up, 52 (81%) patients were alive, five (8%) patients were dead, and seven (11%) patients had lost to follow-up or unknown vital status. The mean follow-up period was 121.5 months (range, 12–360 months). In five cases with immunohistochemistry analysis in the eye specimen, neuron-specific enolase-, Ki-67 protein-, and B-cell lymphoma 2-positive cells were found in all five (100%) cases and Rb protein was detected in three (60%) cases. Conclusion: Retinoblastoma in Mongolia is frequently diagnosed at late stages and has a poor outcome. These data show the importance of early pediatric eye examinations and better treatment of retino-blastoma in children younger than 3 years in Mongolia.
Collapse
Affiliation(s)
- Tsengelmaa Chuluunbat
- Department of Ophthalmology, National Center for Maternal and Child Health, Mongolia
| | - Baasankhuu Jamiyanjav
- Department of Ophthalmology, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Baylag Munkhuu
- Department of Ophthalmology, National Center for Maternal and Child Health, Mongolia
| | - Uranchimeg Bazarsad
- Department of Pathology, National Center of Pathology, Ulaanbaatar, Mongolia
| | - Altankhuu Molom
- Department of Ophthalmology, National Center for Maternal and Child Health, Mongolia
| | - Ling-Yuh Kao
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Wei-Chi Wu
- Department of Ophthalmology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
95
|
Affiliation(s)
- Pia R. Mendoza
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| | - Hans E. Grossniklaus
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
96
|
Shields CL, Jorge R, Say EAT, 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. [PMID: 26765038 DOI: 10.1097/apo.0000000000000172] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE The objective of this study was to compare outcomes after intravenous chemotherapy (IVC) versus intra-arterial chemotherapy (IAC) for unilateral retinoblastoma. DESIGN A retrospective comparative interventional case series. METHODS Patients with unilateral retinoblastoma managed with either IVC using vincristine, etoposide, and carboplatin or IAC using melphalan with or without topotecan with a minimum of 1-year follow-up were compared. The primary outcome measure was globe salvage. RESULTS Of 91 patients with unilateral retinoblastoma, IVC was employed in 42 (46%) cases and IAC in 49 (54%). By comparison (IVC vs IAC), patients in the IAC group had greater mean tumor diameter (14 vs 18 mm, P < 0.001) and thickness (7 vs 10 mm, P = 0.001), greater percentage with active vitreous seeds (29% vs 55%, P = 0.01), and greater total retinal detachment (10% vs 43%, P < 0.001). There were no cases of group A in either treatment arm. Globe salvage was not significantly different in groups B, C, or E, but there was significantly improved globe salvage with IAC for group D (48% vs 91%, P = 0.004). Control was significantly better with IAC for solid tumor (62% vs 92%, P = 0.002), subretinal seeds (31% vs 86%, P = 0.006), and vitreous seeds (25% vs 74%, P = 0.006). There were no patients with pinealoblastoma, second cancer, metastasis, or death in either group. CONCLUSIONS For unilateral retinoblastoma, IAC provided significantly superior globe salvage compared with IVC for group D eyes. In addition, IAC provided significantly superior control for solid tumor, subretinal seeds, and vitreous seeds.
Collapse
Affiliation(s)
- Carol L Shields
- From the *Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA; †Ribeirão Preto Medical School, University of São Paulo, Department of Ophthalmology, Otorhinolaryngology, and Head and Neck Surgery, São Paulo, Brazil; ‡Nemours Center for Cancer and Blood Disorders, Nemours/Alfred I. DuPont Hospital for Children at Thomas Jefferson University; §Pediatric Oncology Department, The Children's Hospital of Philadelphia, University of Pennsylvania; and ¶Department of Neurovascular and Endovascular Surgery, Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Abstract
Retinoblastoma is the most common primary intraocular malignancy of childhood. It typically presents with leukocoria or strabismus. In later stages of the disease, the child may exhibit proptosis, buphthalmos, or hypopyon. The pathognomonic molecular aberration is a loss of function mutation in the RB1 gene on chromosome 13q. The degree of tumor involvement within the eye is defined by its group. Grouping was historically done with Reese-Ellsworth System. Recent therapeutic advances have led to the development of a new grouping system, the International Classification of Retinoblastoma (ICRB). In cases of extraocular extension and metastatic disease, the degree of tumor involvement outside of the eye is defined by its stage. Retinoblastoma is staged using the International Retinoblastoma Staging System (IRSS). Children with intraocular retinoblastoma have an excellent overall and ocular survival. In order to avoid the morbidity of enucleation and external beam radiation, treatments for isolated intraocular retinoblastoma have progressively moved toward targeted local modalities. Patients with extraocular involvement, such as those with trilateral retinoblastoma, have a poorer prognosis. The majority of these higher stage patients are now able to be cured with combination chemotherapy.
Collapse
Affiliation(s)
- Michael V Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ira J Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
98
|
Yanık Ö, Gündüz K, Yavuz K, Taçyıldız N, Ünal E. Chemotherapy in Retinoblastoma: Current Approaches. Turk J Ophthalmol 2015; 45:259-267. [PMID: 27800245 PMCID: PMC5082265 DOI: 10.4274/tjo.06888] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/10/2015] [Indexed: 12/01/2022] Open
Abstract
Retinoblastoma (RB) is the most common childhood malignant intraocular tumor. Although enucleation and external beam radiotherapy have been historically used, today the most commonly used eye-sparing approach is chemotherapy. Chemotherapy can be used in both intraocular and extraocular RB cases. Chemotherapeutic agents may be applied in different ways, including systemic, subconjunctival, intra-arterial and intravitreal routes. The main purposes of application of systemic therapy are to reduce the tumor size for local treatment (chemoreduction), or to reduce the risk of metastasis after enucleation surgery (adjuvant therapy). Intra-arterial chemotherapy with the current name “super-selective intra-arterial infusion therapy” could be applied as primary therapy in tumors confined to the retina or as a secondary method in tumor recurrence. The most important advantage of intra-arterial therapy is the prevention of systemic chemotherapy complications. Intravitreal chemotherapy is administered in the presence of persistent or recurrent vitreous seeding. The term “extraocular RB” includes orbital invasion and metastatic disease. Current treatment for orbital invasion is neoadjuvant chemotherapy followed by surgical enucleation and adjuvant chemotherapy and radiotherapy after surgery. In metastatic disease, regional lymph node involvement, distant metastases, and/or central nervous system (CNS) involvement may occur. Among them, CNS involvement has the worst prognosis, remaining at almost 100% mortality. In metastatic disease, high-dose salvage chemotherapy and autologous hematopoietic stem cell rescue therapy are the possible treatment options; radiotherapy could also be added to the protocol according to the side of involvement.
Collapse
Affiliation(s)
- Özge Yanık
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Kaan Gündüz
- Ankara University Faculty of Medicine, Department of Ophthalmology, Ankara, Turkey
| | - Kıvılcım Yavuz
- Ankara University Faculty of Medicine, Department of Radiology, Ankara, Turkey
| | - Nurdan Taçyıldız
- Ankara University Faculty of Medicine, Department of Pediatrics, Ankara, Turkey
| | - Emel Ünal
- Ankara University Faculty of Medicine, Department of Pediatrics, Ankara, Turkey
| |
Collapse
|
99
|
Traine PG, Schedler KJ, Rodrigues EB. Clinical Presentation and Genetic Paradigm of Diffuse Infiltrating Retinoblastoma: A Review. Ocul Oncol Pathol 2015; 2:128-32. [PMID: 27239450 DOI: 10.1159/000441528] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/05/2015] [Indexed: 11/19/2022] Open
Abstract
Retinoblastoma is the most common childhood cancer. Thanks to modern technology and good medical access, mortality in Europe has decreased to about 5%. Diffuse infiltrating retinoblastoma is a very rare subtype of this neoplasm and is characterized by its atypical growth pattern. Diffuse infiltrating retinoblastoma may mimic other more innocuous diseases and may therefore be misdiagnosed. The purpose of this paper was to provide a short review of the main symptoms of diffuse infiltrating retinoblastoma presenting to the ophthalmologist and give a comparison to typical retinoblastoma. The second purpose was to set up a discussion of the genetic paradigm of diffuse infiltrating retinoblastoma. It has often been described to occur sporadically; however, in the last years, it has been shown that it might be heritable. A literature search concerning diffuse infiltrating retinoblastoma considering English, German and Spanish cases and case series identified 77 patients. Moreover, an overview of general data, main symptoms, clinical findings and initial working diagnoses or referral diagnoses is given. Males were significantly more often affected than females. Diffuse infiltrating retinoblastoma can be heritable. Genetic analysis should be offered to the patient and relatives. Interdisciplinary medical follow-up care is needed to detect associated cancers.
Collapse
Affiliation(s)
- Peter G Traine
- Department of Ophthalmology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Eduardo B Rodrigues
- Department of Ophthalmology and Visual Sciences, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
100
|
Hasanreisoglu M, Saktanasate J, Schwendeman R, Shields JA, Shields CL. Indocyanine Green-Enhanced Transpupillary Thermotherapy for Retinoblastoma: Analysis of 42 Tumors. J Pediatr Ophthalmol Strabismus 2015; 52:348-54. [PMID: 26584748 DOI: 10.3928/01913913-20150929-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/13/2015] [Indexed: 11/20/2022]
Abstract
PURPOSE To evaluate the efficacy of indocyanine green-enhanced transpupillary thermotherapy (ICG-TTT) for retinoblastoma that shows suboptimal response to conventional treatments. METHODS A single center, retrospective chart review. The technique involved ICG infusion (range: 0.3 to 0.5 mg/kg) 1 minute prior to applying TTT using the indirect ophthalmoscope technique with a spot size of 1.2 mm. RESULTS There were 42 retinoblastomas in 30 eyes of 21 patients treated with ICG-TTT. The reasons for ICG enhancement included suboptimal response to standard TTT (n = 31, 74%), recurrence after standard TTT (n = 3, 7%), or minimally pigmented fundus with poor standard TTT uptake (n = 8, 19%). The mean patient age at treatment was 12 months (median: 11.6 months, range: 3 to 31 months). The mean tumor base was 3.5 mm (median: 3 mm), mean tumor thickness was 2.5 mm (median; 2 mm), mean distance to the foveola was 2.6 mm (median: 3 mm), and mean distance to the optic disc was 2.2 mm (median: 0.75 mm). Treatment parameters included a spot size of 1.2 mm, mean power of 760 mW (median: 800 mW, range: 400 to 1,200 mW), and mean duration of 4 minutes (median: 4 minutes, range: 0.5 to 14 minutes). Following a median of 2 sessions (range: 1 to 5 sessions) of ICG-TTT, 33 (79%) tumors demonstrated complete regression. The mean tumor thickness postoperatively was 1.7 mm. Two (5%) tumors showed minimal regression after ICG-TTT. During a mean follow-up of 46 months (median: 33 months), tumor recurrence after ICG-TTT developed in 7 (17%) cases at a mean interval of 7 months. Local complications of ICG-TTT included focal paraxial cataract (n = 2, 7%), iris atrophy (n = 1, 3%), and transient retinal hemorrhage (n = 2, 7%). Systemic problems included ICG allergy (n = 1, 5%). Overall, tumor control and globe salvage was achieved in all 30 (100%) eyes. There were no metastatic events. CONCLUSIONS ICG-TTT is an effective alternative for reti-noblastoma control, particularly for small tumors that show suboptimal response to standard
Collapse
|