1
|
Zhang Y, Mao L, Jiang A, Liu J, Lu Y, Yao C, Huang G. PRMT1 mediates the proliferation of Y79 retinoblastoma cells by regulating the p53/p21/CDC2/cyclin B pathway. Exp Eye Res 2024; 247:110040. [PMID: 39134132 DOI: 10.1016/j.exer.2024.110040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Retinoblastoma (RB) is the most common intraocular malignancy among children and presents a certain mortality risk, especially in low- and middle-income countries. Clarifying the molecular mechanisms underlying the onset and progression of retinoblastoma is vital for devising effective cancer treatment approaches. PRMT1, a major type I PRMT, plays significant roles in cancer development. However, its expression and role in retinoblastoma are still unclear. Our research revealed a marked increase in PRMT1 levels in both retinoblastoma tissues and Y79 cells. The overexpression of PRMT1 in Y79 cells promoted their growth and cell cycle progression. Conversely, the suppression of PRMT1 hindered the growth of Y79 cells and impeded cell cycle progression. Mechanistically, PRMT1 mediated the growth of Y79 retinoblastoma cells by targeting the p53/p21/CDC2/Cyclin B pathway. Additionally, the ability of PRMT1 knockdown to suppress cell proliferation was also observed in vivo. Overall, PRMT1 could function as a potential target for therapeutic treatment in individuals with retinoblastoma.
Collapse
Affiliation(s)
- Yanyan Zhang
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Ophthalmology, Jiangxi Provincial People's Hospital & the First Affiliated Hospital of Nanchang Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Longbing Mao
- Department of Ophthalmology, Shangrao Eye's Hospital, The First Affiliated Hospital of Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Alan Jiang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jingchao Liu
- Medical Department of Graduate School, Nanchang University, Nanchang, Jiangxi, China
| | - Yongan Lu
- Medical Department of Graduate School, Nanchang University, Nanchang, Jiangxi, China
| | - Chunyue Yao
- Medical Department of Graduate School, Nanchang University, Nanchang, Jiangxi, China
| | - Guofu Huang
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Vempuluru VS, Maniar A, Bakal K, Kaliki S. Role of MYCN in retinoblastoma: A review of current literature. Surv Ophthalmol 2024; 69:697-706. [PMID: 38796108 DOI: 10.1016/j.survophthal.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Chromosomal abnormalities that involve the MYCN gene are rare; however, it is one of the most commonly mutated genes in retinoblastoma (RB) after the RB1 gene. MYCN is amplified in approximately 1-9 % of all RB tumors. It plays a role in RB oncogenesis via many mechanisms, including synergism with RB1 deletion, positive feedback with MDM2, upregulation of cell cycle regulating genes, upregulation of miRNA, and upregulation of glucose metabolism. MYCN amplifications are not mutually exclusive and can occur even in the presence of RB1 gene mutations. Clinically, RB1+/+MYCNA tumors present as sporadic, unilateral, advanced tumors in very young children and tend to follow an aggressive course. Magnetic resonance imaging features include peripheral tumor location, placoid configuration, retinal folding, tumor-associated hemorrhage, and anterior chamber enhancement. Genetic testing for MYCNA is especially recommended in patients with unilateral RB where genetic blood testing and tumor tissue show a lack of RB1 mutation. MYCN-targeted therapies are evolving and hold promise for the future.
Collapse
Affiliation(s)
- Vijitha S Vempuluru
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Arpita Maniar
- Duke Eye Center, Duke University, Durham, NC 27705, USA
| | - Komal Bakal
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, L V Prasad Eye Institute, Hyderabad 500034, India.
| |
Collapse
|
3
|
Zheng J, Li T, Ye H, Jiang Z, Jiang W, Yang H, Wu Z, Xie Z. Comprehensive identification of pathogenic variants in retinoblastoma by long- and short-read sequencing. Cancer Lett 2024; 598:217121. [PMID: 39009069 DOI: 10.1016/j.canlet.2024.217121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 06/16/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Retinoblastoma (RB) is the most common intraocular malignancy in childhood. The causal variants in RB are mostly characterized by previously used short-read sequencing (SRS) analysis, which has technical limitations in identifying structural variants (SVs) and phasing information. Long-read sequencing (LRS) technology has advantages over SRS in detecting SVs, phased genetic variants, and methylation. In this study, we comprehensively characterized the genetic landscape of RB using combinatorial LRS and SRS of 16 RB tumors and 16 matched blood samples. We detected a total of 232 somatic SVs, with an average of 14.5 SVs per sample across the whole genome in our cohort. We identified 20 distinct pathogenic variants disrupting RB1 gene, including three novel small variants and five somatic SVs. We found more somatic SVs were detected from LRS than SRS (140 vs. 122) in RB samples with WGS data, particularly the insertions (18 vs. 1). Furthermore, our analysis shows that, with the exception of one sample who lacked the methylation data, all samples presented biallelic inactivation of RB1 in various forms, including two cases with the biallelic hypermethylated promoter and four cases with compound heterozygous mutations which were missing in SRS analysis. By inferring relative timing of somatic events, we reveal the genetic progression that RB1 disruption early and followed by copy number changes, including amplifications of Chr2p and deletions of Chr16q, during RB tumorigenesis. Altogether, we characterize the comprehensive genetic landscape of RB, providing novel insights into the genetic alterations and mechanisms contributing to RB initiation and development. Our work also establishes a framework to analyze genomic landscape of cancers based on LRS data.
Collapse
Affiliation(s)
- Jingjing Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Tong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huijing Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zehang Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenbing Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Huasheng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Zhikun Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Ryl T, Afanasyeva E, Hartmann T, Schwermer M, Schneider M, Schröder C, Wagemanns M, Bister A, Kanber D, Steenpass L, Schramm K, Jones B, Jones DTW, Biewald E, Astrahantseff K, Hanenberg H, Rahmann S, Lohmann DR, Schramm A, Ketteler P. A MYCN-driven de-differentiation profile identifies a subgroup of aggressive retinoblastoma. Commun Biol 2024; 7:919. [PMID: 39079981 PMCID: PMC11289481 DOI: 10.1038/s42003-024-06596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Retinoblastoma are childhood eye tumors arising from retinal precursor cells. Two distinct retinoblastoma subtypes with different clinical behavior have been described based on gene expression and methylation profiling. Using consensus clustering of DNA methylation analysis from 61 retinoblastomas, we identify a MYCN-driven cluster of subtype 2 retinoblastomas characterized by DNA hypomethylation and high expression of genes involved in protein synthesis. Subtype 2 retinoblastomas outside the MYCN-driven cluster are characterized by high expression of genes from mesodermal development, including NKX2-5. Knockdown of MYCN expression in retinoblastoma cell models causes growth arrest and reactivates a subtype 1-specific photoreceptor signature. These molecular changes suggest that removing the driving force of MYCN oncogenic activity rescues molecular circuitry driving subtype 1 biology. The MYCN-RB gene signature generated from the cell models better identifies MYCN-driven retinoblastoma than MYCN amplification and can identify cases that may benefit from MYCN-targeted therapy. MYCN drives tumor progression in a molecularly defined retinoblastoma subgroup, and inhibiting MYCN activity could restore a more differentiated and less aggressive tumor biology.
Collapse
Affiliation(s)
- Tatsiana Ryl
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Elena Afanasyeva
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Till Hartmann
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Schwermer
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Markus Schneider
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Christopher Schröder
- Algorithms for Reproducible Bioinformatics, Genome Informatics, Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maren Wagemanns
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Laura Steenpass
- Human and Animal Cell Lines, Leibniz Institute DSMZ German Collection of Microorganisms and Cell Cultures, 38124, Braunschweig, Germany
| | - Kathrin Schramm
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Barbara Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - David T W Jones
- Division of Pediatric Glioma Research, Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, University of Heidelberg, Heidelberg, Germany
| | - Eva Biewald
- Department of Ophthalmology, Medical Faculty, University of Duisburg-Essen, 45147, Essen, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Oncology and Hematology, Charité - University Medicine Berlin, Berlin, Germany
| | - Helmut Hanenberg
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Sven Rahmann
- Algorithmic Bioinformatics, Center for Bioinformatics Saar and Saarland University, Saarland Informatics Campus, Saarbrücken, Germany
| | - Dietmar R Lohmann
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Alexander Schramm
- Laboratory for Molecular Oncology, Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany.
- Institute of Human Genetics, University Hospital Essen, University Duisburg Essen, Essen, Germany.
| |
Collapse
|
5
|
Wang W, Zhang A, Li Y, Wang D, Chen L, Li Q, Chen J, Li H, Sun S, Pan M, Zhou W, Wu H. Clinical, pathological, and molecular features of central nervous system tumors with BCOR internal tandem duplication. Pathol Res Pract 2024; 259:155367. [PMID: 38797130 DOI: 10.1016/j.prp.2024.155367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Central nervous system tumor with BCOR internal tandem duplication (CNS tumor with BCOR-ITD) constitutes a molecularly distinct entity, characterized by internal tandem duplication within exon 15 of the BCOR transcriptional co-repressor gene (BCOR-ITD). The current study aimed to elucidate the clinical, pathological, and molecular attributes of CNS tumors with BCOR-ITD and explore their putative cellular origin. This study cohort comprised four pediatric cases, aged 23 months to 13 years at initial presentation. Magnetic resonance imaging revealed large, well-circumscribed intra-CNS masses localized heterogeneously throughout the CNS. Microscopically, tumors were composed of spindle to ovoid cells, exhibiting perivascular pseudorosettes and palisading necrosis, but lacking microvascular proliferation. Immunohistochemical staining showed diffuse tumor cell expression of BCOR, CD56, CD99, vimentin, and the stem cell markers PAX6, SOX2, CD133 and Nestin, alongside focal positivity for Olig-2, S100, SOX10, Syn and NeuN. Molecularly, all cases harbored BCOR-ITDs ranging from 87 to 119 base pairs in length, including one case with two distinct ITDs. Notably, the ITDs were interrupted by unique 1-3 bp insertions in all cases. In summary, CNS tumors with BCOR-ITD exhibit characteristic clinical, pathological, and molecular features detectable through BCOR immunohistochemistry and confirmatory molecular analyses. Their expression of stem cell markers raises the possibility of an origin from neuroepithelial stem cells rather than representing true embryonal neoplasms.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Anli Zhang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yujie Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Daizhong Wang
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Lian Chen
- Department of Pathology, Children's Hospital of Fudan University, Shanghai, China
| | - Qianqian Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Chen
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Heng Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Sibai Sun
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Minhong Pan
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Wenchao Zhou
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Haibo Wu
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
6
|
Pareek A, Kumar D, Pareek A, Gupta MM, Jeandet P, Ratan Y, Jain V, Kamal MA, Saboor M, Ashraf GM, Chuturgoon A. Retinoblastoma: An update on genetic origin, classification, conventional to next-generation treatment strategies. Heliyon 2024; 10:e32844. [PMID: 38975183 PMCID: PMC11226919 DOI: 10.1016/j.heliyon.2024.e32844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
The most prevalent paediatric vision-threatening medical condition, retinoblastoma (RB), has been a global concern for a long time. Several conventional therapies, such as systemic chemotherapy and focal therapy, have been used for curative purposes; however, the search for tumour eradication with the least impact on surrounding tissues is still ongoing. This review focuses on the genetic origin, classification, conventional treatment modalities, and their combination with nano-scale delivery systems for active tumour targeting. In addition, the review also delves into ongoing clinical trials and patents, as well as emerging therapies such as gene therapy and immunotherapy for the treatment of RB. Understanding the role of genetics in the development of RB has refined its treatment strategy according to the genetic type. New approaches such as nanostructured drug delivery systems, galenic preparations, nutlin-3a, histone deacetylase inhibitors, N-MYC inhibitors, pentoxifylline, immunotherapy, gene therapy, etc. discussed in this review, have the potential to circumvent the limitations of conventional therapies and improve treatment outcomes for RB. In summary, this review highlights the importance and need for novel approaches as alternative therapies that would ultimately displace the shortcomings associated with conventional therapies and reduce the enucleation rate, thereby preserving global vision in the affected paediatric population.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Deepanjali Kumar
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Madan Mohan Gupta
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine 3303, Trinidad and Tobago
| | - Philippe Jeandet
- Research Unit Induced Resistance and Plant Bioprotection - USC INRAe 1488, University of Reims, PO Box 1039, 51687, Reims, France
| | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur, 313001, India
| | - Mohammad Amjad Kamal
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, West China School of Nursing, Frontiers Science Centre for Disease-related Molecular Network, West China Hospital, Sichuan University, China
- King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW, 2770, Australia
| | - Muhammad Saboor
- Department of Medical Laboratory Science, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Science, College of Health Sciences, and Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa
| |
Collapse
|
7
|
Price EA, Sagoo MS, Reddy MA, Onadim Z. An overview of RB1 transcript alterations detected during retinoblastoma genetic screening. Ophthalmic Genet 2024; 45:235-245. [PMID: 37932244 DOI: 10.1080/13816810.2023.2270570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/24/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Identification of pathogenic RB1 variants aids in the clinical management of families with retinoblastoma. We routinely screen DNA for RB1 variants, but transcript analysis can also be used for variant screening, and to help decide variant pathogenicity. DNA was screened by conformation analysis followed by Sanger sequencing. Large deletion/insertions were detected by polymorphism analysis, MLPA and quantitative-PCR. Methylation-specific PCR was used to detect hypermethylation. RNA screening was performed when a DNA pathogenic variant was missing, or to determine effects on splicing.Two hundred and thirteen small coding variants were predicted to affect splicing in 207 patients. Splice donor (sd) variants were nearly twice as frequent as splice acceptor (sa) with the most affected positions being sd + 1 and sa-1. Some missense and nonsense codons altered splicing, while some splice consensus variants did not. Large deletion/insertions can disrupt splicing, but RNA analysis showed that some of these are more complex than indicated by DNA testing. RNA screening found pathogenic variants in 53.8% of samples where DNA analysis did not. RB1 splicing is altered by changes at consensus splice sites, some missense and nonsense codons, deep intronic changes and large deletion/insertions. Common alternatively spliced transcripts may complicate analysis. An effective molecular screening strategy would include RNA analysis to help determine pathogenicity.
Collapse
Affiliation(s)
- Elizabeth A Price
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
| | - Mandeep S Sagoo
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, Institute of Ophthalmology, University College London, London, UK
| | - M Ashwin Reddy
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK
- Faculty of Medicine, Queen Mary University of London, London, UK
| | - Zerrin Onadim
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
- Faculty of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Nag A, Khetan V. Retinoblastoma - A comprehensive review, update and recent advances. Indian J Ophthalmol 2024; 72:778-788. [PMID: 38804799 PMCID: PMC11232864 DOI: 10.4103/ijo.ijo_2414_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/28/2023] [Accepted: 12/02/2023] [Indexed: 05/29/2024] Open
Abstract
Retinoblastoma is the most common pediatric ocular malignancy. It is triggered by a biallelic mutation in the RB1 gene or MYCN oncogene amplification. Retinoblastomas can be unilateral (60%-70%) or bilateral (30%-40%); bilateral tumors are always heritable and present at an earlier age as compared to unilateral ones (18-24 months vs. 36 months in India). High prevalence rates, delayed presentation, and inaccessibility to healthcare lead to worse outcomes in developing countries. The past few decades have seen a paradigm change in the treatment of retinoblastomas, shifting from enucleation and external beam radiotherapy to less aggressive modalities for eye salvage. Multimodality treatment is now the standard of care and includes intraarterial or intravenous chemotherapy along with focal consolidation therapies such as transpupillary thermotherapy, cryotherapy, and laser photocoagulation. Intravitreal and intracameral chemotherapy can help in controlling intraocular seeds. Advanced extraocular or metastatic tumors still have a poor prognosis. Genetic testing, counseling, and screening of at-risk family members must be incorporated as essential parts of management. A better understanding of the genetics and molecular basis of retinoblastoma has opened up the path for potential targeted therapy in the future. Novel recent advances such as liquid biopsy, prenatal diagnosis, prognostic biomarkers, tylectomy, and chemoplaque point to promising future directions.
Collapse
Affiliation(s)
- Adwaita Nag
- Ocular Oncology Service, Department of Ophthalmology and Vision Sciences, University of Toronto, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Vikas Khetan
- Formerly at Shri Bhagwan Mahavir Vitreoretinal Services, Sankara Nethralaya, Chennai, Tamil Nadu, India
- Professor, Department of Ophthalmology, Flaum Eye Institute, Rochester, NY, USA
| |
Collapse
|
9
|
Zhou L, Tong Y, Ho BM, Li J, Chan HYE, Zhang T, Du L, He JN, Chen LJ, Tham CC, Yam JC, Pang CP, Chu WK. Etiology including epigenetic defects of retinoblastoma. Asia Pac J Ophthalmol (Phila) 2024:100072. [PMID: 38789041 DOI: 10.1016/j.apjo.2024.100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/09/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Retinoblastoma (RB), originating from the developing retina, is an aggressive intraocular malignant neoplasm in childhood. Biallelic loss of RB1 is conventionally considered a prerequisite for initiating RB development in most RB cases. Additional genetic mutations arising from genome instability following RB1 mutations are proposed to be required to promote RB development. Recent advancements in high throughput sequencing technologies allow a deeper and more comprehensive understanding of the etiology of RB that additional genetic alterations following RB1 biallelic loss are rare, yet epigenetic changes driven by RB1 loss emerge as a critical contributor promoting RB tumorigenesis. Multiple epigenetic regulators have been found to be dysregulated and to contribute to RB development, including noncoding RNAs, DNA methylations, RNA modifications, chromatin conformations, and histone modifications. A full understanding of the roles of genetic and epigenetic alterations in RB formation is crucial in facilitating the translation of these findings into effective treatment strategies for RB. In this review, we summarize current knowledge concerning genetic defects and epigenetic dysregulations in RB, aiming to help understand their links and roles in RB tumorigenesis.
Collapse
Affiliation(s)
- Linbin Zhou
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Yan Tong
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Bo Man Ho
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiahui Li
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Hoi Ying Emily Chan
- Medicine Programme Global Physician-Leadership Stream, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Tian Zhang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Lin Du
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jing Na He
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Li Jia Chen
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Clement C Tham
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jason C Yam
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Chi Pui Pang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| | - Wai Kit Chu
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China; Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
10
|
Zhou M, Tang J, Fan J, Wen X, Shen J, Jia R, Chai P, Fan X. Recent progress in retinoblastoma: Pathogenesis, presentation, diagnosis and management. Asia Pac J Ophthalmol (Phila) 2024; 13:100058. [PMID: 38615905 DOI: 10.1016/j.apjo.2024.100058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/16/2024] Open
Abstract
Retinoblastoma, the primary ocular malignancy in pediatric patients, poses a substantial threat to mortality without prompt and effective management. The prognosis for survival and preservation of visual acuity hinges upon the disease severity at the time of initial diagnosis. Notably, retinoblastoma has played a crucial role in unraveling the genetic foundations of oncogenesis. The process of tumorigenesis commonly begins with the occurrence of biallelic mutation in the RB1 tumor suppressor gene, which is then followed by a cascade of genetic and epigenetic alterations that correspond to the clinical stage and pathological features of the tumor. The RB1 gene, recognized as a tumor suppressor, encodes the retinoblastoma protein, which plays a vital role in governing cellular replication through interactions with E2F transcription factors and chromatin remodeling proteins. The diagnosis and treatment of retinoblastoma necessitate consideration of numerous factors, including disease staging, germline mutation status, family psychosocial factors, and the resources available within the institution. This review has systematically compiled and categorized the latest developments in the diagnosis and treatment of retinoblastoma which enhanced the quality of care for this pediatric malignancy.
Collapse
Affiliation(s)
- Min Zhou
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Jieling Tang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Jiayan Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Xuyang Wen
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Jianfeng Shen
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China
| | - Peiwei Chai
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China.
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 20025, People's Republic of China.
| |
Collapse
|
11
|
Krupina K, Goginashvili A, Cleveland DW. Scrambling the genome in cancer: causes and consequences of complex chromosome rearrangements. Nat Rev Genet 2024; 25:196-210. [PMID: 37938738 PMCID: PMC10922386 DOI: 10.1038/s41576-023-00663-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 11/09/2023]
Abstract
Complex chromosome rearrangements, known as chromoanagenesis, are widespread in cancer. Based on large-scale DNA sequencing of human tumours, the most frequent type of complex chromosome rearrangement is chromothripsis, a massive, localized and clustered rearrangement of one (or a few) chromosomes seemingly acquired in a single event. Chromothripsis can be initiated by mitotic errors that produce a micronucleus encapsulating a single chromosome or chromosomal fragment. Rupture of the unstable micronuclear envelope exposes its chromatin to cytosolic nucleases and induces chromothriptic shattering. Found in up to half of tumours included in pan-cancer genomic analyses, chromothriptic rearrangements can contribute to tumorigenesis through inactivation of tumour suppressor genes, activation of proto-oncogenes, or gene amplification through the production of self-propagating extrachromosomal circular DNAs encoding oncogenes or genes conferring anticancer drug resistance. Here, we discuss what has been learned about the mechanisms that enable these complex genomic rearrangements and their consequences in cancer.
Collapse
Affiliation(s)
- Ksenia Krupina
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Alexander Goginashvili
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Don W Cleveland
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Zhang H, Konjusha D, Rafati N, Tararuk T, Hallböök F. Inhibition of high level E2F in a RB1 proficient MYCN overexpressing chicken retinoblastoma model normalizes neoplastic behaviour. Cell Oncol (Dordr) 2024; 47:209-227. [PMID: 37606819 PMCID: PMC10899388 DOI: 10.1007/s13402-023-00863-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/23/2023] Open
Abstract
PURPOSE Retinoblastoma, a childhood cancer, is most frequently caused by bi-allelic inactivation of RB1 gene. However, other oncogenic mutations such as MYCN amplification can induce retinoblastoma with proficient RB1. Previously, we established RB1-proficient MYCN-overexpressing retinoblastoma models both in human organoids and chicken. Here, we investigate the regulatory events in MYCN-induced retinoblastoma carcinogenesis based on the model in chicken. METHODS MYCN transformed retinal cells in culture were obtained from in vivo MYCN electroporated chicken embryo retina. The expression profiles were analysed by RNA sequencing. Chemical treatments, qRT-PCR, flow cytometry, immunohisto- and immunocytochemistry and western blot were applied to study the properties and function of these cells. RESULTS The expression profile of MYCN-transformed retinal cells in culture showed cone photoreceptor progenitor signature and robustly increased levels of E2Fs. This expression profile was consistently observed in long-term culture. Chemical treatments confirmed RB1 proficiency in these cells. The cells were insensitive to p53 activation but inhibition of E2f efficiently induced cell cycle arrest followed by apoptosis. CONCLUSION In conclusion, with proficient RB1, MYCN-induced high level of E2F expression dysregulates the cell cycle and contributes to retinoblastoma carcinogenesis. The increased level of E2f renders the cells to adopt a similar mechanistic phenotype to a RB1-deficient tumour.
Collapse
Affiliation(s)
- Hanzhao Zhang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dardan Konjusha
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Nima Rafati
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Tatsiana Tararuk
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Finn Hallböök
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85, Uppsala, Sweden.
| |
Collapse
|
13
|
Akdeniz Odemis D, Kebudi R, Bayramova J, Kilic Erciyas S, Kuru Turkcan G, Tuncer SB, Sukruoglu Erdogan O, Celik B, Kurt Gultaslar B, Buyukkapu Bay S, Tuncer S, Yazici H. RB1 gene mutations and genetic spectrum in retinoblastoma cases. Medicine (Baltimore) 2023; 102:e35068. [PMID: 37682130 PMCID: PMC10489529 DOI: 10.1097/md.0000000000035068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023] Open
Abstract
The aim of the study was to investigate the frequency and types of mutations on the retinoblastoma gene (RB1 gene) in Turkish population. RB1 gene mutation analysis was performed in a total of 219 individuals (122 probands with retinoblastoma, 14 family members with retinoblastoma and 83 clinically healthy family members). All 27 exons and close intronic regions of the RB1 gene were sequenced for small deletions and insertions using both the Sanger sequencing or NGS methods, and the large deletions and duplications were investigated using the MLPA analysis and CNV algorithm. The bilateral/trilateral retinoblastoma rate was 66% in the study population. The general frequency of RB1 gene mutation in the germline of the patients with retinoblastoma was 41.9%. Approximately 51.5% of the patients were diagnosed earlier than 12 months old, and de novo mutation was found in 32.4% of the patients. Germline small genetic rearrangement mutations were detected in 78.9% of patients and LGRs were detected in 21.1% of patients. An association was detected between the eye color of the RB patients and RB1 mutations. 8 of the mutations detected in the RB1 gene were novel in the study.
Collapse
Affiliation(s)
- Demet Akdeniz Odemis
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Rejin Kebudi
- Istanbul University, Oncology Institute, Division of Pediatric Hematology-Oncology, Istanbul, Türkiye
| | - Jamila Bayramova
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Seda Kilic Erciyas
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Gozde Kuru Turkcan
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
- Halic University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, Istanbul, Türkiye
| | - Seref Bugra Tuncer
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Ozge Sukruoglu Erdogan
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Betul Celik
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Busra Kurt Gultaslar
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
| | - Sema Buyukkapu Bay
- Istanbul University, Oncology Institute, Division of Pediatric Hematology-Oncology, Istanbul, Türkiye
| | - Samuray Tuncer
- Istanbul University, Istanbul Medical Faculty, Department of Ophthalmology, Istanbul, Türkiye
| | - Hulya Yazici
- Istanbul University, Oncology Institute, Department of Basic Oncology, Division of Cancer Genetics, Istanbul, Türkiye
- Istanbul Arel University, Istanbul Arel Medical Faculty, Department of Medical Biology and Genetics, Istanbul, Türkiye
| |
Collapse
|
14
|
Rathore S, Verma A, Ratna R, Marwa N, Ghiya Y, Honavar SG, Tiwari A, Das S, Varshney A. Retinoblastoma: A review of the molecular basis of tumor development and its clinical correlation in shaping future targeted treatment strategies. Indian J Ophthalmol 2023; 71:2662-2676. [PMID: 37417104 PMCID: PMC10491038 DOI: 10.4103/ijo.ijo_3172_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/25/2023] [Accepted: 05/21/2023] [Indexed: 07/08/2023] Open
Abstract
Retinoblastoma is a retinal cancer that affects children and is the most prevalent intraocular tumor worldwide. Despite tremendous breakthroughs in our understanding of the fundamental mechanisms that regulate progression of retinoblastoma, the development of targeted therapeutics for retinoblastoma has lagged. Our review highlights the current developments in the genetic, epigenetic, transcriptomic, and proteomic landscapes of retinoblastoma. We also discuss their clinical relevance and potential implications for future therapeutic development, with the aim to create a frontline multimodal therapy for retinoblastoma.
Collapse
Affiliation(s)
- Shruti Rathore
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Aman Verma
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Ria Ratna
- Ocular Genetics Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Navjot Marwa
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Yagya Ghiya
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Santosh G Honavar
- Ophthalmic Plastic Surgery, Orbit and Ocular Oncology, Centre for Sight, Hyderbad, Telangana, India
| | - Anil Tiwari
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Sima Das
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| | - Akhil Varshney
- Ocular Oncology Services, Dr. Shroff’s Charity Eye Hospital, New Delhi, India
| |
Collapse
|
15
|
Stachelek K, Harutyunyan N, Lee S, Beck A, Kim J, Xu L, Berry JL, Nagiel A, Reynolds CP, Murphree AL, Lee TC, Aparicio JG, Cobrinik D. Non-synonymous, synonymous, and non-coding nucleotide variants contribute to recurrently altered biological processes during retinoblastoma progression. Genes Chromosomes Cancer 2023; 62:275-289. [PMID: 36550020 PMCID: PMC10006380 DOI: 10.1002/gcc.23120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Retinoblastomas form in response to biallelic RB1 mutations or MYCN amplification and progress to more aggressive and therapy-resistant phenotypes through accumulation of secondary genomic changes. Progression-related changes include recurrent somatic copy number alterations and typically non-recurrent nucleotide variants, including synonymous and non-coding variants, whose significance has been unclear. To determine if nucleotide variants recurrently affect specific biological processes, we identified altered genes and over-represented variant gene ontologies in 168 exome or whole-genome-sequenced retinoblastomas and 12 tumor-matched cell lines. In addition to RB1 mutations, MYCN amplification, and established retinoblastoma somatic copy number alterations, the analyses revealed enrichment of variant genes related to diverse biological processes including histone monoubiquitination, mRNA processing (P) body assembly, and mitotic sister chromatid segregation and cytokinesis. Importantly, non-coding and synonymous variants increased the enrichment significance of each over-represented biological process term. To assess the effects of such mutations, we examined the consequences of a 3' UTR variant of PCGF3 (a BCOR-binding component of Polycomb repressive complex I), dual 3' UTR variants of CDC14B (a regulator of sister chromatid segregation), and a synonymous variant of DYNC1H1 (a regulator of P-body assembly). One PCGF3 and one of two CDC14B 3' UTR variants impaired gene expression whereas a base-edited DYNC1H1 synonymous variant altered protease sensitivity and stability. Retinoblastoma cell lines retained only ~50% of variants detected in tumors and enriched for new variants affecting p53 signaling. These findings reveal potentially important differences in retinoblastoma cell lines and tumors and implicate synonymous and non-coding variants, along with non-synonymous variants, in retinoblastoma oncogenesis.
Collapse
Affiliation(s)
- Kevin Stachelek
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Cancer Biology and Genomics Program, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Narine Harutyunyan
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Susan Lee
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Assaf Beck
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Jonathan Kim
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Liya Xu
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jesse L. Berry
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Aaron Nagiel
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - C. Patrick Reynolds
- Department of Pediatrics and Cancer Center, Texas Tech University Health Sciences Center, School of Medicine, Lubbock, TX
| | - A. Linn Murphree
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Thomas C. Lee
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jennifer G. Aparicio
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
| | - David Cobrinik
- The Vision Center and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA
- Department of Ophthalmology and Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
16
|
Jeyaprakash K, Thirumalairaj K, Kim U, Muthukkaruppan V, Vanniarajan A. RB1 transcript analysis detects novel splicing aberration in retinoblastoma. Pediatr Blood Cancer 2023; 70:e30290. [PMID: 36916769 DOI: 10.1002/pbc.30290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/16/2023]
Affiliation(s)
- Kumar Jeyaprakash
- Department of Molecular Genetics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India.,Department of Molecular Biology, Aravind Medical Research Foundation, Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India
| | - Kannan Thirumalairaj
- Department of Molecular Genetics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Usha Kim
- Department of Orbit, Oculoplasty and Oncology, Aravind Eye Hospital, Madurai, Tamil Nadu, India
| | - Veerappan Muthukkaruppan
- Department of Stem Cell Biology and Immunology, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India
| | - Ayyasamy Vanniarajan
- Department of Molecular Genetics, Aravind Medical Research Foundation, Madurai, Tamil Nadu, India.,Department of Molecular Biology, Aravind Medical Research Foundation, Affiliated to Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
17
|
Bisht S, Chawla B, Kumar A, Vijayan V, Kumar M, Sharma P, Dada R. Identification of novel genes by targeted exome sequencing in Retinoblastoma. Ophthalmic Genet 2022; 43:771-788. [PMID: 35930312 DOI: 10.1080/13816810.2022.2106497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Retinoblastoma (RB) is initiated by mutation in both alleles of RB1 gene. However, few cases may occur even in the absence of RB1 mutation suggesting the role of genes other than RB1. METHODOLOGY The current study was planned to utilize targeted exome sequencing in Indian RB patients affected with unilateral non-familial RB. 75 unilateral RB patients below 5 years of age were enrolled. Genomic DNA was extracted from blood and tumor tissue. From peripheral blood DNA, all coding and exon/intron regions were amplified using PCR and direct sequencing. Cases which did not harbor pathogenic variants in peripheral blood DNA were further screened for mutations in their tumor tissue DNA using targeted exome sequencing. Three pathogenicity prediction tools (Mutation Taster, SIFT, and PolyPhen-2) were used to determine the pathogenicity of non-synonymous variations. An in-house bioinformatics pipeline was devised for the mutation screening by targeted exome sequencing. Protein modeling studies were also done to predict the effect of the mutations on the protein structure and function. RESULTS Using the mentioned approach, we found two novel variants (g.69673_69674insT and g.48373314C>A) in RB1 gene in peripheral blood DNA. We also found novel variants in eight genes (RB1, ACAD11, GPR151, KCNA1, OTOR, SOX30, ARL11, and MYCT1) that may be associated with RB pathogenesis. CONCLUSION The present study expands our current knowledge regarding the genomic landscape of RB and also highlights the importance of NGS technologies to detect genes and novel variants that may play an important role in cancer initiation, progression, and prognosis.
Collapse
Affiliation(s)
- Shilpa Bisht
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Bhavna Chawla
- Ocular Oncology Service, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Amit Kumar
- Computational Genomics Centre, Indian Council of Medical Research, New Delhi, India
| | - Viswanathan Vijayan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Rima Dada
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
18
|
E2F1/CKS2/PTEN signaling axis regulates malignant phenotypes in pediatric retinoblastoma. Cell Death Dis 2022; 13:784. [PMID: 36096885 PMCID: PMC9468144 DOI: 10.1038/s41419-022-05222-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2022] [Accepted: 08/31/2022] [Indexed: 01/21/2023]
Abstract
Retinoblastoma (RB) is the most common pediatric intraocular malignancy and is a serious vision- and life-threatening disease. The biallelic mutation of the retinoblastoma gene RB1 is the initial event in the malignant transformation of RB, but the exact molecular mechanism is still unclear. E2F transcription factors can be activated by RB1 loss of function and lead to uncontrolled cell division. Among E2F family numbers, E2F1 has higher expression abundance than E2F2 and E2F3 in RB clinical samples. By integrating E2F1 ChIP-seq data, RNA-seq profiling from RB samples and RNA-seq profiling upon E2F1 knockdown, together with pathway analysis, literature searching and experimental validation, we identified Cyclin-dependent kinases regulatory subunit 2 (CKS2) as a novel regulator in regulating tumor-associated phenotypes in RB. CKS2 exhibited aberrantly higher expression in RB. Depletion of CKS2 in Y79 retinoblastoma cell line led to reduced cell proliferation, delayed DNA replication and decreased clonogenic growth. Downregulation of CKS2 also slowed tumor xenograft growth in nude mice. Importantly, reversed expression of CKS2 rescued cancer-associated phenotypes. Mechanistically, transcription factor E2F1 enhanced CKS2 expression through binding to its promoter and CKS2 regulated the cancer-associated PI3K-AKT pathway. This study discovered E2F1/CKS2/PTEN signaling axis regulates malignant phenotypes in pediatric retinoblastoma, and CKS2 may serve as a potential therapeutic target for this disease.
Collapse
|
19
|
MYCN induces cell-specific tumorigenic growth in RB1-proficient human retinal organoid and chicken retina models of retinoblastoma. Oncogenesis 2022; 11:34. [PMID: 35729105 PMCID: PMC9213451 DOI: 10.1038/s41389-022-00409-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022] Open
Abstract
Retinoblastoma is a rare, intraocular paediatric cancer that originates in the neural retina and is most frequently caused by bi-allelic loss of RB1 gene function. Other oncogenic mutations, such as amplification and increased expression of the MYCN gene, have been found even with proficient RB1 function. In this study, we investigated whether MYCN over-expression can drive carcinogenesis independently of RB1 loss-of-function mutations. The aim was to elucidate the events that result in carcinogenesis and identify the cancer cell-of-origin. We used the chicken retina, a well-established model for studying retinal neurogenesis, and established human embryonic stem cell-derived retinal organoids as model systems. We over-expressed MYCN by electroporation of piggyBac genome-integrating expression vectors. We found that over-expression of MYCN induced tumorigenic growth with high frequency in RB1-proficient chicken retinas and human organoids. In both systems, the tumorigenic cells expressed markers for undifferentiated cone photoreceptor/horizontal cell progenitors. The over-expression resulted in metastatic retinoblastoma within 7–9 weeks in chicken. Cells expressing MYCN could be grown in vitro and, when orthotopically injected, formed tumours that infiltrated the sclera and optic nerve and expressed markers for cone progenitors. Investigation of the tumour cell phenotype determined that the potential for neoplastic growth was embryonic stage-dependent and featured a cell-specific resistance to apoptosis in the cone/horizontal cell lineage, but not in ganglion or amacrine cells. We conclude that MYCN over-expression is sufficient to drive tumorigenesis and that a cell-specific resistance to apoptosis in the cone/horizontal cell lineage mediates the cancer phenotype. ![]()
Collapse
|
20
|
Lee C, Kim J. Genome maintenance in retinoblastoma: Implications for therapeutic vulnerabilities (Review). Oncol Lett 2022; 23:192. [PMID: 35527780 PMCID: PMC9073582 DOI: 10.3892/ol.2022.13312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/08/2022] [Indexed: 11/19/2022] Open
Abstract
Retinoblastoma (RB) is a pediatric ocular malignancy that is initiated mostly by biallelic inactivation of the RB transcriptional corepressor 1 (RB1) tumor suppressor gene in the developing retina. Unlike the prevailing prediction based on multiple studies involving RB1 gene disruption in experimental models, human RB tumors have been demonstrated to possess a relatively stable genome, characterized by a low mutation rate and a few recurrent chromosomal alterations related to somatic copy number changes. This suggests that RB may harbor heightened genome maintenance mechanisms to counteract or compensate for the risk of massive genome instability, which can potentially be driven by the early RB1 loss as a tumor-initiating event. Although the genome maintenance mechanisms might have been evolved to promote RB cell survival by preventing lethal genomic defects, emerging evidence suggests that the dependency of RB cells on these mechanisms also exposes their unique vulnerability to chemotherapy, particularly when the genome maintenance machineries are tumor cell-specific. This review summarizes the genome maintenance mechanisms identified in RB, including findings on the roles of chromatin regulators in DNA damage response/repair and protein factors involved in maintaining chromosome stability and promoting survival in RB. In addition, advantages and challenges for exploiting these therapeutic vulnerabilities in RB are discussed.
Collapse
Affiliation(s)
- Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Jong Kim
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‑sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
21
|
Chai P, Jia R, Li Y, Zhou C, Gu X, Yang L, Shi H, Tian H, Lin H, Yu J, Zhuang A, Ge S, Jia R, Fan X. Regulation of epigenetic homeostasis in uveal melanoma and retinoblastoma. Prog Retin Eye Res 2021; 89:101030. [PMID: 34861419 DOI: 10.1016/j.preteyeres.2021.101030] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Uveal melanoma (UM) and retinoblastoma (RB), which cause blindness and even death, are the most frequently observed primary intraocular malignancies in adults and children, respectively. Epigenetic studies have shown that changes in the epigenome contribute to the rapid progression of both UM and RB following classic genetic changes. The loss of epigenetic homeostasis plays an important role in oncogenesis by disrupting the normal patterns of gene expression. The targetable nature of epigenetic modifications provides a unique opportunity to optimize treatment paradigms and establish new therapeutic options for both UM and RB with these aberrant epigenetic modifications. We aimed to review the research findings regarding relevant epigenetic changes in UM and RB. Herein, we 1) summarize the literature, with an emphasis on epigenetic alterations, including DNA methylation, histone modifications, RNA modifications, noncoding RNAs and an abnormal chromosomal architecture; 2) elaborate on the regulatory role of epigenetic modifications in biological processes during tumorigenesis; and 3) propose promising therapeutic candidates for epigenetic targets and update the list of epigenetic drugs for the treatment of UM and RB. In summary, we endeavour to depict the epigenetic landscape of primary intraocular malignancy tumorigenesis and provide potential epigenetic targets in the treatment of these tumours.
Collapse
Affiliation(s)
- Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Ruobing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Yongyun Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Chuandi Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Hanhan Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Huimin Lin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Jie Yu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025, PR China.
| |
Collapse
|
22
|
Sradhanjali S, Rout P, Tripathy D, Kaliki S, Rath S, Modak R, Mittal R, Chowdary TK, Reddy MM. The Oncogene MYCN Modulates Glycolytic and Invasive Genes to Enhance Cell Viability and Migration in Human Retinoblastoma. Cancers (Basel) 2021; 13:cancers13205248. [PMID: 34680394 PMCID: PMC8533785 DOI: 10.3390/cancers13205248] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022] Open
Abstract
Retinoblastoma is usually initiated by biallelic RB1 gene inactivation. In addition, MYCN copy number alterations also contribute to RB pathogenesis. However, MYCN expression, its role in disease progression and correlation with RB histological risk factors are not well understood. We studied the expression of MYCN in enucleated RB patient specimens by immunohistochemistry. MYCN is overexpressed in RB compared to control retina. Our microarray gene expression analysis followed by qRT-PCR validation revealed that genes involved in glucose metabolism and migration are significantly downregulated in MYCN knockdown cells. Further, targeting MYCN in RB cells using small molecule compounds or shRNAs led to decreased cell survival and migration, increased apoptosis and cell cycle arrest, suggesting that MYCN inhibition can be a potential therapeutic strategy. We also noted that MYCN inhibition results in reduction in glucose uptake, lactate production, ROS levels and gelatinolytic activity of active-MMP9, explaining a possible mechanism of MYCN in RB. Taking clues from our findings, we tested a combination treatment of RB cells with carboplatin and MYCN inhibitors to find enhanced therapeutic efficacy compared to single drug treatment. Thus, MYCN inhibition can be a potential therapeutic strategy in combination with existing chemotherapy drugs to restrict tumor cell growth in RB.
Collapse
Affiliation(s)
- Swatishree Sradhanjali
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India; (S.S.); (P.R.)
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, Odisha, India;
| | - Padmalochan Rout
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India; (S.S.); (P.R.)
- Novo Nordisk, Bangalore 560066, Karnataka, India
| | - Devjyoti Tripathy
- Ophthalmic Plastics, Orbit and Ocular Oncology Service, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India; (D.T.); (S.R.)
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad 500034, Telangana, India;
| | - Suryasnata Rath
- Ophthalmic Plastics, Orbit and Ocular Oncology Service, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India; (D.T.); (S.R.)
| | - Rahul Modak
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, Odisha, India;
| | - Ruchi Mittal
- Kanupriya Dalmia Ophthalmic Pathology Laboratory, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India;
- Department of Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar 751024, Odisha, India
| | - Tirumala Kumar Chowdary
- School of Biological Sciences, National Institute of Science Education and Research, Homi Bhabha National Institute, Bhubaneswar 752050, Odisha, India;
| | - Mamatha M. Reddy
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Bhubaneswar 751024, Odisha, India; (S.S.); (P.R.)
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, Odisha, India;
- Correspondence: or ; Tel.: +91-674-3987175
| |
Collapse
|
23
|
Investigation of Key Signaling Pathways Associating miR-204 and Common Retinopathies. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5568113. [PMID: 34646884 PMCID: PMC8505061 DOI: 10.1155/2021/5568113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 06/15/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a large group of small noncoding RNAs that work in multiple cellular pathways. miR-204, as one of the key axes in the development, maintenance, and pathogenesis of the retina, plays several roles by modulating its target genes. This study was aimed at evaluating the target genes of miR-204 involved in the development and progression of common retinopathies such as glaucoma, retinoblastoma, and age-related macular degeneration. In this study, three datasets related to retinopathies (GSE50195, GSE27276, and GSE97508) were selected from Gene Expression Omnibus. miR-204 target genes were isolated from TargeScan. The shares between retinopathy and miR-204 target genes were then categorized. Using Enrichr and STRING, we highlighted the signaling pathways and the relationships between the proteins. SHC1 events in ERBB2, adherent junction's interactions, NGF signaling via TRKA from the plasma membrane, IRF3-mediated activation of type 1 IFN, pathways in upregulated genes and G0 and early G1, RORA-activated gene expression, PERK-regulated gene expression, adherent junction's interactions, and CREB phosphorylation pathways in downregulated genes were identified in glaucoma, retinoblastoma, and age-related macular degeneration. WEE1, SMC2, HMGB1, RRM2, and POLA1 proteins were also observed to be involved in the progression and invasion of retinoblastoma; SLC24A2 and DTX4 in age-related macular degeneration; and EPHB6, EFNB3, and SHC1 in glaucoma. Continuous bioinformatics analysis has shown that miR-204 has a significant presence and expression in retinal tissue, and approximately 293 genes are controlled and regulated by miR-204 in this tissue; also, target genes of miR-204 have the potential to develop various retinopathies; thus, a study of related target genes can provide appropriate treatment strategies in the future.
Collapse
|
24
|
Wang S, C Ordonez-Rubiano S, Dhiman A, Jiao G, Strohmier BP, Krusemark CJ, Dykhuizen EC. Polycomb group proteins in cancer: multifaceted functions and strategies for modulation. NAR Cancer 2021; 3:zcab039. [PMID: 34617019 PMCID: PMC8489530 DOI: 10.1093/narcan/zcab039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/24/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Polycomb repressive complexes (PRCs) are a heterogenous collection of dozens, if not hundreds, of protein complexes composed of various combinations of subunits. PRCs are transcriptional repressors important for cell-type specificity during development, and as such, are commonly mis-regulated in cancer. PRCs are broadly characterized as PRC1 with histone ubiquitin ligase activity, or PRC2 with histone methyltransferase activity; however, the mechanism by which individual PRCs, particularly the highly diverse set of PRC1s, alter gene expression has not always been clear. Here we review the current understanding of how PRCs act, both individually and together, to establish and maintain gene repression, the biochemical contribution of individual PRC subunits, the mis-regulation of PRC function in different cancers, and the current strategies for modulating PRC activity. Increased mechanistic understanding of PRC function, as well as cancer-specific roles for individual PRC subunits, will uncover better targets and strategies for cancer therapies.
Collapse
Affiliation(s)
- Sijie Wang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Sandra C Ordonez-Rubiano
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Alisha Dhiman
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Guanming Jiao
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Brayden P Strohmier
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Casey J Krusemark
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University and Purdue University Center for Cancer Research, 201 S. University St., West Lafayette, IN 47907 USA
| |
Collapse
|
25
|
Poly ADP Ribose Polymerase Inhibitor Olaparib Targeting Microhomology End Joining in Retinoblastoma Protein Defective Cancer: Analysis of the Retinoblastoma Cell-Killing Effects by Olaparib after Inducing Double-Strand Breaks. Int J Mol Sci 2021; 22:ijms221910687. [PMID: 34639028 PMCID: PMC8508856 DOI: 10.3390/ijms221910687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Retinoblastoma is the most common intraocular cancer in childhood. Loss of function in both copies of the RB1 gene is the causal mutation of retinoblastoma. Current treatment for retinoblastoma includes the use of chemotherapeutic agents, such as the DNA damaging agent etoposide, which is a topoisomerase II poison that mainly generates DNA double-strand breaks (DSBs) and genome instability. Unfaithful repairing of DSBs could lead to secondary cancers and serious side effects. Previously, we found that RB knocked-down mammalian cells depend on a highly mutagenic pathway, the micro-homology mediated end joining (MMEJ) pathway, to repair DSBs. Poly ADP ribose polymerase 1 (PARP1) is a major protein in promoting the MMEJ pathway. In this study, we explored the effects of olaparib, a PARP inhibitor, in killing retinoblastoma cells. Retinoblastoma cell line Y79 and primary retinoblastoma cells expressed the cone-rod homeobox protein (CRX), a photoreceptor-specific marker. No detectable RB expression was found in these cells. The co-treatment of olaparib and etoposide led to enhanced cell death in both the Y79 cells and the primary retinoblastoma cells. Our results demonstrated the killing effects in retinoblastoma cells by PARP inhibitor olaparib after inducing DNA double-strand breaks. The use of olaparib in combination with etoposide could improve the cell-killing effects. Thus, lower dosages of etoposide can be used to treat retinoblastoma, which would potentially lead to a lower level of DSBs and a relatively more stable genome.
Collapse
|
26
|
Liu J, Ottaviani D, Sefta M, Desbrousses C, Chapeaublanc E, Aschero R, Sirab N, Lubieniecki F, Lamas G, Tonon L, Dehainault C, Hua C, Fréneaux P, Reichman S, Karboul N, Biton A, Mirabal-Ortega L, Larcher M, Brulard C, Arrufat S, Nicolas A, Elarouci N, Popova T, Némati F, Decaudin D, Gentien D, Baulande S, Mariani O, Dufour F, Guibert S, Vallot C, Rouic LLL, Matet A, Desjardins L, Pascual-Pasto G, Suñol M, Catala-Mora J, Llano GC, Couturier J, Barillot E, Schaiquevich P, Gauthier-Villars M, Stoppa-Lyonnet D, Golmard L, Houdayer C, Brisse H, Bernard-Pierrot I, Letouzé E, Viari A, Saule S, Sastre-Garau X, Doz F, Carcaboso AM, Cassoux N, Pouponnot C, Goureau O, Chantada G, de Reyniès A, Aerts I, Radvanyi F. A high-risk retinoblastoma subtype with stemness features, dedifferentiated cone states and neuronal/ganglion cell gene expression. Nat Commun 2021; 12:5578. [PMID: 34552068 PMCID: PMC8458383 DOI: 10.1038/s41467-021-25792-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma is the most frequent intraocular malignancy in children, originating from a maturing cone precursor in the developing retina. Little is known on the molecular basis underlying the biological and clinical behavior of this cancer. Here, using multi-omics data, we demonstrate the existence of two retinoblastoma subtypes. Subtype 1, of earlier onset, includes most of the heritable forms. It harbors few genetic alterations other than the initiating RB1 inactivation and corresponds to differentiated tumors expressing mature cone markers. By contrast, subtype 2 tumors harbor frequent recurrent genetic alterations including MYCN-amplification. They express markers of less differentiated cone together with neuronal/ganglion cell markers with marked inter- and intra-tumor heterogeneity. The cone dedifferentiation in subtype 2 is associated with stemness features including low immune and interferon response, E2F and MYC/MYCN activation and a higher propensity for metastasis. The recognition of these two subtypes, one maintaining a cone-differentiated state, and the other, more aggressive, associated with cone dedifferentiation and expression of neuronal markers, opens up important biological and clinical perspectives for retinoblastomas.
Collapse
Affiliation(s)
- Jing Liu
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Daniela Ottaviani
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Meriem Sefta
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Céline Desbrousses
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Elodie Chapeaublanc
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Rosario Aschero
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Nanor Sirab
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Fabiana Lubieniecki
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Gabriela Lamas
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Laurie Tonon
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Catherine Dehainault
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France
| | - Clément Hua
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Paul Fréneaux
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Narjesse Karboul
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Anne Biton
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France ,grid.428999.70000 0001 2353 6535Present Address: Institut Pasteur – Hub Bioinformatique et Biostatistique – C3BI, USR 3756 IP CNRS, 75015 Paris, France
| | - Liliana Mirabal-Ortega
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Magalie Larcher
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Céline Brulard
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.411777.30000 0004 1765 1563Present Address: INSERM U930, CHU Bretonneau, 37000 Tours, France
| | - Sandrine Arrufat
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - André Nicolas
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Nabila Elarouci
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Tatiana Popova
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Fariba Némati
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Didier Decaudin
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - David Gentien
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Sylvain Baulande
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, NGS Platform, 75005 Paris, France
| | - Odette Mariani
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Florent Dufour
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Sylvain Guibert
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Céline Vallot
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Livia Lumbroso-Le Rouic
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Alexandre Matet
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Laurence Desjardins
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Guillem Pascual-Pasto
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Mariona Suñol
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Pathology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jaume Catala-Mora
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Ophthalmology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Genoveva Correa Llano
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jérôme Couturier
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Emmanuel Barillot
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France
| | - Paula Schaiquevich
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Marion Gauthier-Villars
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Dominique Stoppa-Lyonnet
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Lisa Golmard
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Claude Houdayer
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France ,grid.41724.34Present Address: Department of Genetics, Rouen University Hospital, 76000 Rouen, France
| | - Hervé Brisse
- grid.418596.70000 0004 0639 6384Département d’Imagerie Médicale, Institut Curie, 75005 Paris, France
| | - Isabelle Bernard-Pierrot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Eric Letouzé
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Universités, INSERM, 75006 Paris, France ,grid.508487.60000 0004 7885 7602Functional Genomics of Solid Tumors, équipe labellisée Ligue Contre le Cancer, Université de Paris, Université Paris 13, Paris, France
| | - Alain Viari
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Simon Saule
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Xavier Sastre-Garau
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.414145.10000 0004 1765 2136Present Address: Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France
| | - François Doz
- grid.508487.60000 0004 7885 7602Université de Paris, Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - Angel M. Carcaboso
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Nathalie Cassoux
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Celio Pouponnot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Guillermo Chantada
- grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Aurélien de Reyniès
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Isabelle Aerts
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - François Radvanyi
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| |
Collapse
|
27
|
Gargallo P, Oltra S, Balaguer J, Barranco H, Yáñez Y, Segura V, Juan-Ribelles A, Calabria I, Llavador M, Castel V, Cañete A. Retinoblastoma and mosaic 13q deletion: a case report. Int J Retina Vitreous 2021; 7:50. [PMID: 34479642 PMCID: PMC8418035 DOI: 10.1186/s40942-021-00321-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with 13q-syndrome are at risk of retinoblastoma when the RB1 gene, located in the chromosomal band 13q14.2, is deleted. This syndrome is frequently associated with congenital malformations and developmental delay, although these signs could be mild. Mosaic 13q-deletion patients have been previously reported in the literature; their phenotype is variable, and they may not be recognized. CASE PRESENTATION Retinoblastoma diagnosed in a child with 13q-mosaicism confirmed in blood, oral mucosa, healthy retina and retinoblastoma. A second RB1 hit is present exclusively in the retinoblastoma sample (RB1 c.958C>T p.Arg320Ter). Other detected molecular events in retinoblastoma are 6p12.3pter gain and 6q25.3qter loss. Clinical examination is unremarkable except for clinodactyly of the right fifth finger. DISCUSSION AND CONCLUSIONS We describe a case of mosaic 13q deletion syndrome affected by retinoblastoma. Molecular data obtained from the tumor analysis are similar to previous data available about this malignancy. High clinical suspicion is essential for an adequate diagnosis of mosaic cases.
Collapse
Affiliation(s)
- Pablo Gargallo
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain. .,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.
| | - Silvestre Oltra
- Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain
| | - Julia Balaguer
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| | - Honorio Barranco
- Pediatric Ophthalmology Unit, University Hospital La Fe Hospital, Valencia, Spain
| | - Yania Yáñez
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Vanessa Segura
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Antonio Juan-Ribelles
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| | - Inés Calabria
- Oncology Department, Imegen-Health in Code Group, Valencia, Spain
| | | | - Victoria Castel
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Adela Cañete
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| |
Collapse
|
28
|
Retinoblastoma from human stem cell-derived retinal organoids. Nat Commun 2021; 12:4535. [PMID: 34315877 PMCID: PMC8316454 DOI: 10.1038/s41467-021-24781-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma is a childhood cancer of the developing retina that initiates with biallelic inactivation of the RB1 gene. Children with germline mutations in RB1 have a high likelihood of developing retinoblastoma and other malignancies later in life. Genetically engineered mouse models of retinoblastoma share some similarities with human retinoblastoma but there are differences in their cellular differentiation. To develop a laboratory model of human retinoblastoma formation, we make induced pluripotent stem cells (iPSCs) from 15 participants with germline RB1 mutations. Each of the stem cell lines is validated, characterized and then differentiated into retina using a 3-dimensional organoid culture system. After 45 days in culture, the retinal organoids are dissociated and injected into the vitreous of eyes of immunocompromised mice to support retinoblastoma tumor growth. Retinoblastomas formed from retinal organoids made from patient-derived iPSCs have molecular, cellular and genomic features indistinguishable from human retinoblastomas. This model of human cancer based on patient-derived iPSCs with germline cancer predisposing mutations provides valuable insights into the cellular origins of this debilitating childhood disease as well as the mechanism of tumorigenesis following RB1 gene inactivation. Retinoblastoma is a heritable pediatric cancer driven by mutations in RB1. Here, the authors demonstrate the first patient derived model of retinoblastoma using iPSCs from patients with germline mutations in RB1.
Collapse
|
29
|
Jiménez I, Frouin É, Chicard M, Dehainault C, Le Gall J, Benoist C, Gauthier A, Lapouble E, Houdayer C, Radvanyi F, Bernard V, Brisse HJ, Gauthier-Villars M, Stoppa-Lyonnet D, Baulande S, Cassoux N, Lumbroso L, Matet A, Aerts I, Renault V, Doz F, Golmard L, Delattre O, Schleiermacher G. Molecular diagnosis of retinoblastoma by circulating tumor DNA analysis. Eur J Cancer 2021; 154:277-287. [PMID: 34298378 DOI: 10.1016/j.ejca.2021.05.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/11/2021] [Accepted: 05/27/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE The analysis of circulating tumor DNA (ctDNA), a fraction of total cell-free DNA (cfDNA), might be of special interest in retinoblastoma patients. Because the accessibility to tumor tissue is very limited in these patients, either for histopathological diagnosis of suspicious intraocular masses (biopsies are proscribed) or for somatic RB1 studies and genetic counseling (due to current successful conservative approaches), we aim to validate the detection of ctDNA in plasma of non-hereditary retinoblastoma patients by molecular analysis of RB1 gene. EXPERIMENTAL DESIGN In a cohort of 19 intraocular unilateral non-hereditary retinoblastoma patients for whom a plasma sample was available at diagnosis, we performed high-deep next-generation sequencing (NGS) of RB1 in cfDNA. Two different bioinformatics/statistics approaches were applied depending on whether the somatic RB1 status was available or not. RESULTS Median plasma sample volume was 600 μL [100-1000]; median cfDNA plasma concentration was 119 [38-1980] and 27 [11-653] ng/mL at diagnosis and after complete remission, respectively. In the subgroup of patients with known somatic RB1 alterations (n = 11), seven of nine somatic mutations were detected (median allele fraction: 6.7%). In patients without identified somatic RB1 alterations (n = 8), six candidate variants were identified for seven patients. CONCLUSIONS Despite small tumor size, blood-ocular barrier, poor ctDNA blood release and limited plasma sample volumes, we confirm that it is possible to detect ctDNA with high-deep NGS in plasma from patients with intraocular non-hereditary retinoblastoma. This may aid in diagnosis of suspicious cases, family genetic counseling or follow-up of residual intraocular disease.
Collapse
Affiliation(s)
- Irene Jiménez
- SiRIC RTOP « Recherche Translationelle en Oncologie Pédiatrique », Translational Research Department, PSL Research University, Institut Curie Research Center, Paris, France; INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, Institut Curie Research Center, Paris, France; SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Éléonore Frouin
- Clinical Bioinformatics, PSL Research University, Institut Curie, Paris, France
| | - Mathieu Chicard
- SiRIC RTOP « Recherche Translationelle en Oncologie Pédiatrique », Translational Research Department, PSL Research University, Institut Curie Research Center, Paris, France; INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, Institut Curie Research Center, Paris, France; SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | | | - Jessica Le Gall
- Department of Genetics, PSL Research University, Institut Curie, Paris, France
| | - Camille Benoist
- Clinical Bioinformatics, PSL Research University, Institut Curie, Paris, France
| | - Arnaud Gauthier
- Pathology Department, PSL Research University, Institut Curie, Paris, France
| | - Eve Lapouble
- Somatic Genetics Unit, PSL Research University, Institut Curie, Paris, France
| | - Claude Houdayer
- INSERM U1245, Normandie University, UNIROUEN, Normandy Centre for Genomic and Personalized Medicine and Rouen University Hospital, Department of Genetics, Rouen, France
| | - François Radvanyi
- CNRS, UMR144, Equipe Labellisée Ligue Contre le Cancer, Institut Curie, PSL Research University, Paris, France
| | - Virginie Bernard
- Centre Hospitalier Universitaire Grenoble-Alpes, Grenoble, France
| | - Hervé J Brisse
- Imaging Department, PSL Research University, Institut Curie, Paris, France
| | | | | | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, PSL Research University, Research Center, Institut Curie, Paris, France
| | - Nathalie Cassoux
- SiRIC RTOP « Recherche Translationelle en Oncologie Pédiatrique », Translational Research Department, PSL Research University, Institut Curie Research Center, Paris, France; SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France; Université de Paris, Paris, France
| | | | - Alexandre Matet
- Ocular Oncology Service, Institut Curie, Paris, France; Université de Paris, Paris, France
| | - Isabelle Aerts
- SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Victor Renault
- Clinical Bioinformatics, PSL Research University, Institut Curie, Paris, France
| | - François Doz
- SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France; Université de Paris, Paris, France
| | - Lisa Golmard
- Department of Genetics, PSL Research University, Institut Curie, Paris, France
| | - Olivier Delattre
- INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France; SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Gudrun Schleiermacher
- SiRIC RTOP « Recherche Translationelle en Oncologie Pédiatrique », Translational Research Department, PSL Research University, Institut Curie Research Center, Paris, France; INSERM U830, Equipe Labellisée Ligue Contre le Cancer, PSL Research University, Institut Curie Research Center, Paris, France; Department of Translational Research, Institut Curie Research Center, Paris, France; SIREDO Center: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France.
| |
Collapse
|
30
|
Price EA, Patel R, Scheimberg I, Kotiloglu Karaa E, Sagoo MS, Reddy MA, Onadim Z. MYCN amplification levels in primary retinoblastoma tumors analyzed by Multiple Ligation-dependent Probe Amplification. Ophthalmic Genet 2021; 42:604-611. [PMID: 34003079 DOI: 10.1080/13816810.2021.1923038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Retinoblastoma (Rb) is a childhood tumor of the developing retina where predisposition is caused by RB1 pathogenic variants. MYCN amplification (MYCNA) has been implicated in around 2% of sporadic unilateral Rb tumors with no detectable RB1 variants. We audited data from tumors collected between 1993 and 2019 to determine if this is the case for patients treated at Barts Health NHS Trust, and how often it occurred alongside RB1 variants. Materials and methods: Screening for MYCNA was carried out by Multiple Ligation Probe Analysis of tumor and blood samples collected for RB1 genetic screening. The cohort consisted of 149 tumors, of which 114 had matched blood samples. Results: 10/149 (6.7%) tumors were positive for MYCNA in a population containing a disproportionate number of cases negative for RB1 pathogenic variants. Of 65 unbiased tumors collected from 2014 to 2019, 2 (3.1%) had MYCNA. All MYCNA samples were from sporadic, unilateral patients and 3/10 (30%) had RB1 pathogenic variants. MYCNA was not detected in any blood sample. No MYCNA tumor had 6p gain which is usually a common alteration in Rbs. Conclusions: MYCNA occurs in a small fraction of Rbs and can occur in the presence of pathogenic RB1 variants. However, where it occurs alongside RB1 alterations, the age of onset appears to be later. MYCNA has yet to be seen as a heritable change. In sporadic cases with early diagnosis, Rbs with no RB1 pathogenic variant identified should be tested for MYCNA. Conversely, tumors with MYCNA should still be screened for RB1 pathogenic variants.
Collapse
Affiliation(s)
- Elizabeth A Price
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
| | - Roopal Patel
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
| | | | | | - Mandeep S Sagoo
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK.,NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK
| | - M Ashwin Reddy
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Zerrin Onadim
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
| |
Collapse
|
31
|
Saengwimol D, Chittavanich P, Laosillapacharoen N, Srimongkol A, Chaitankar V, Rojanaporn D, Aroonroch R, Suktitipat B, Saisawang C, Svasti S, Hongeng S, Kaewkhaw R. Silencing of the Long Noncoding RNA MYCNOS1 Suppresses Activity of MYCN-Amplified Retinoblastoma Without RB1 Mutation. Invest Ophthalmol Vis Sci 2021; 61:8. [PMID: 33270844 PMCID: PMC7718827 DOI: 10.1167/iovs.61.14.8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Purpose MYCNOS (MYCN opposite strand) is co-amplified with MYCN in pediatric cancers, including retinoblastoma. MYCNOS encodes several RNA variants whose functions have not been elucidated in retinoblastoma. Thus, we attempted to identify MYCNOS variants in retinoblastoma and aimed to decipher the role of MYCNOS variant 1 (MYCNOS1) on the activity of MYCN-amplified retinoblastoma. Methods The profiles of MYCNOS variants and MYCN status were determined in 17 retinoblastoma tissues, cell lines, retinas, and retinal organoids. A functional study of MYCNOS1 expression was conducted in patient-derived tumor cells and in retinoblastoma cell lines via short hairpin RNA-mediated gene silencing. We carried out MYCN expression, cell viability, cell cycle, apoptosis, soft agar colony formation, and transwell assays to examine the role of MYCNOS1 in MYCN and cell behaviors. We analyzed a transcriptome of MYCN-amplified retinoblastoma cells deficient for MYCNOS1 and, finally, tested the responses of these cells to chemotherapeutic agents. Results Expression of MYCNOS1 was associated with the expression and copy number of MYCN. Knockdown of MYCNOS1 caused instability of the MYCN protein, leading to cell cycle arrest and impaired proliferation and chemotaxis-directed migration in MYCN-amplified retinoblastoma cells in which RB1 was intact. MYCNOS1 expression was associated with gene signatures of photoreceptor cells and epithelial–mesenchymal transition. MYCNOS1 silencing enhanced the response of retinoblastoma cells to topotecan but not carboplatin. Conclusions MYCNOS1 supports progression of retinoblastoma. Inhibition of MYCNOS1 expression may be necessary to suppress MYCN activity when treating MYCN-amplified cancers without RB1 mutation.
Collapse
Affiliation(s)
- Duangporn Saengwimol
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pamorn Chittavanich
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Natanan Laosillapacharoen
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Atthapol Srimongkol
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Vijender Chaitankar
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Duangnate Rojanaporn
- Department of Ophthalmology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rangsima Aroonroch
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Bhoom Suktitipat
- Department of Biochemistry, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand.,Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom, Thailand
| | - Chonticha Saisawang
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Saovaros Svasti
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Rossukon Kaewkhaw
- Section of Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
32
|
Schieffer KM, Feldman AZ, Kautto EA, McGrath S, Miller AR, Hernandez-Gonzalez ME, LaHaye S, Miller KE, Koboldt DC, Brennan P, Kelly B, Wetzel A, Agarwal V, Shatara M, Conley S, Rodriguez DP, Abu-Arja R, Shaikhkhalil A, Snuderl M, Orr BA, Finlay JL, Osorio DS, Drapeau AI, Leonard JR, Pierson CR, White P, Magrini V, Mardis ER, Wilson RK, Cottrell CE, Boué DR. Molecular classification of a complex structural rearrangement of the RB1 locus in an infant with sporadic, isolated, intracranial, sellar region retinoblastoma. Acta Neuropathol Commun 2021; 9:61. [PMID: 33827698 PMCID: PMC8025529 DOI: 10.1186/s40478-021-01164-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/19/2021] [Indexed: 12/15/2022] Open
Abstract
Retinoblastoma is a childhood cancer of the retina involving germline or somatic alterations of the RB Transcriptional Corepressor 1 gene, RB1. Rare cases of sellar-suprasellar region retinoblastoma without evidence of ocular or pineal tumors have been described. A nine-month-old male presented with a sellar-suprasellar region mass. Histopathology showed an embryonal tumor with focal Flexner-Wintersteiner-like rosettes and loss of retinoblastoma protein (RB1) expression by immunohistochemistry. DNA array-based methylation profiling confidently classified the tumor as pineoblastoma group A/intracranial retinoblastoma. The patient was subsequently enrolled on an institutional translational cancer research protocol and underwent comprehensive molecular profiling, including paired tumor/normal exome and genome sequencing and RNA-sequencing of the tumor. Additionally, Pacific Biosciences (PacBio) Single Molecule Real Time (SMRT) sequencing was performed from comparator normal and disease-involved tissue to resolve complex structural variations. RNA-sequencing revealed multiple fusions clustered within 13q14.1-q21.3, including a novel in-frame fusion of RB1-SIAH3 predicted to prematurely truncate the RB1 protein. SMRT sequencing revealed a complex structural rearrangement spanning 13q14.11-q31.3, including two somatic structural variants within intron 17 of RB1. These events corresponded to the RB1-SIAH3 fusion and a novel RB1 rearrangement expected to correlate with the complete absence of RB1 protein expression. Comprehensive molecular analysis, including DNA array-based methylation profiling and sequencing-based methodologies, were critical for classification and understanding the complex mechanism of RB1 inactivation in this diagnostically challenging tumor.
Collapse
|
33
|
Davies HR, Broad KD, Onadim Z, Price EA, Zou X, Sheriff I, Karaa EK, Scheimberg I, Reddy MA, Sagoo MS, Ohnuma SI, Nik-Zainal S. Whole-Genome Sequencing of Retinoblastoma Reveals the Diversity of Rearrangements Disrupting RB1 and Uncovers a Treatment-Related Mutational Signature. Cancers (Basel) 2021; 13:754. [PMID: 33670346 PMCID: PMC7918943 DOI: 10.3390/cancers13040754] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
The development of retinoblastoma is thought to require pathological genetic changes in both alleles of the RB1 gene. However, cases exist where RB1 mutations are undetectable, suggesting alternative pathways to malignancy. We used whole-genome sequencing (WGS) and transcriptomics to investigate the landscape of sporadic retinoblastomas derived from twenty patients, sought RB1 and other driver mutations and investigated mutational signatures. At least one RB1 mutation was identified in all retinoblastomas, including new mutations in addition to those previously identified by clinical screening. Ten tumours carried structural rearrangements involving RB1 ranging from relatively simple to extremely complex rearrangement patterns, including a chromothripsis-like pattern in one tumour. Bilateral tumours obtained from one patient harboured conserved germline but divergent somatic RB1 mutations, indicating independent evolution. Mutational signature analysis showed predominance of signatures associated with cell division, an absence of ultraviolet-related DNA damage and a profound platinum-related mutational signature in a chemotherapy-exposed tumour. Most RB1 mutations are identifiable by clinical screening. However, the increased resolution and ability to detect otherwise elusive rearrangements by WGS have important repercussions on clinical management and advice on recurrence risks.
Collapse
Affiliation(s)
- Helen R. Davies
- Academic Department of Medical Genetics, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (H.R.D.); (X.Z.)
- MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Kevin D. Broad
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Zerrin Onadim
- Retinoblastoma Genetic Screening Unit, The Royal London Hospital, Barts Health NHS Trust, London E1 1FR, UK; (Z.O.); (E.A.P.)
| | - Elizabeth A. Price
- Retinoblastoma Genetic Screening Unit, The Royal London Hospital, Barts Health NHS Trust, London E1 1FR, UK; (Z.O.); (E.A.P.)
| | - Xueqing Zou
- Academic Department of Medical Genetics, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (H.R.D.); (X.Z.)
- MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Ibrahim Sheriff
- Retinoblastoma Service, Royal London Hospital, Barts Health Trust, London E1 1FR, UK; (I.S.); (M.A.R.)
| | - Esin Kotiloğlu Karaa
- Pathology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1FR, UK; (E.K.K.); (I.S.)
| | - Irene Scheimberg
- Pathology Department, Royal London Hospital, Barts Health NHS Trust, London E1 1FR, UK; (E.K.K.); (I.S.)
| | - M. Ashwin Reddy
- Retinoblastoma Service, Royal London Hospital, Barts Health Trust, London E1 1FR, UK; (I.S.); (M.A.R.)
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, Institute of Ophthalmology, University College London, London EC1V 2PD, UK
| | - Mandeep S. Sagoo
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
- Retinoblastoma Service, Royal London Hospital, Barts Health Trust, London E1 1FR, UK; (I.S.); (M.A.R.)
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, Institute of Ophthalmology, University College London, London EC1V 2PD, UK
| | - Shin-ichi Ohnuma
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Serena Nik-Zainal
- Academic Department of Medical Genetics, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK; (H.R.D.); (X.Z.)
- MRC Cancer Unit, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0XZ, UK
| |
Collapse
|
34
|
Ortega V, Mendiola C, Rodriguez J, Ehman W, Qian YW, Velagaleti G. Bi-allelic amplification of ATM gene in blastoid variant of mantle cell lymphoma: a novel mechanism of inactivation due to chromoanagenesis? Mol Cytogenet 2021; 14:8. [PMID: 33541390 PMCID: PMC7863528 DOI: 10.1186/s13039-020-00526-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/28/2020] [Indexed: 11/10/2022] Open
Abstract
Background Mantle cell lymphoma (MCL) is derived from naïve CD5+ B-cells with the cytogenetic hallmark translocation 11;14. The presence of additional abnormalities is associated with blastoid variants in MCL (BMCL) and confers a poor prognosis. Many of these tumors also show deletion or loss of heterozygosity (LOH) of the ATM gene and biallelic ATM inactivation show significantly higher chromosomal imbalances. Case presentation Here we report a 52 year-old male who presented to the clinic with worsening dyspnea, fever, chills, diffuse lymphadenopathy, splenomegaly and leukocytosis with blastoid cells circulating in blood. The bone marrow aspirate showed about 40% abnormal blast-looking cells and biopsy revealed a remarkable lymphoid infiltrate. The patient was diagnosed with blastoid variant mantle cell lymphoma (BMCL). Chromosome analysis on bone marrow showed a complex karyotype. FISH analysis from B-cell lymphoma panel showed bi-allelic amplification of ATM gene. Other abnormalities were present including CCND1/IGH fusion, confirming the MCL diagnosis, in addition to RB1 and p53 deletion. High resolution SNP-microarray studies showed complex copy number changes, especially on chromosomes 7 and 11, consistent with chromoanagenesis. Microarray studies also showed LOH at the ATM locus indicating the amplification seen on FISH is not biallelic. Conclusion To the best of our knowledge, ATM gene amplification is not previously reported in BMCL and our case suggests a novel mechanism of ATM inactivation caused by chromoanagenesis resulting in mutant allele specific imbalance with copy number gain.
Collapse
Affiliation(s)
- Veronica Ortega
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Christina Mendiola
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Juana Rodriguez
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - William Ehman
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - You-Wen Qian
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Gopalrao Velagaleti
- Department of Pathology and Laboratory Medicine, UT Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
35
|
Molecular Changes in Retinoblastoma beyond RB1: Findings from Next-Generation Sequencing. Cancers (Basel) 2021; 13:cancers13010149. [PMID: 33466343 PMCID: PMC7796332 DOI: 10.3390/cancers13010149] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/25/2020] [Accepted: 12/30/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The gene causing retinoblastoma was the first tumor suppressor cloned (1986) and because retinoblastoma is the classic example of autosomal dominant inheritance, there has been little research on non-RB1 alterations in tumors and the impact these alterations have on growth patterns in the eye, metastases and predilection for non-ocular cancers. This study interrogated enucleated retinoblastoma specimens using a MSK-IMPACT clinical next-generation sequencing panel with the aim to correlate them with clinicopathologic characteristics. We found that vitreous seeding (the main reason for eye removal) correlates with copy number variations, specifically 1q gains and 16q loss. We also found that somatic BCOR mutations correlate with propensity for metastasis and this offers a molecular pathway for monitoring high risk tumors. In addition, the finding that 11% of these retinoblastoma patients have additional germline mutations (on other chromosomes) that predispose them to a different host of cancers throughout their lives enables more targeted and specific screening strategies. Abstract This investigation uses hybridization capture-based next-generation sequencing to deepen our understanding of genetics that underlie retinoblastoma. Eighty-three enucleated retinoblastoma specimens were evaluated using a MSK-IMPACT clinical next-generation sequencing panel to evaluate both somatic and germline alterations. Somatic copy number variations (CNVs) were also identified. Genetic profiles were correlated to clinicopathologic characteristics. RB1 inactivation was found in 79 (97.5%) patients. All specimens had additional molecular alterations. The most common non-RB1 gene alteration was BCOR in 19 (22.9%). Five (11.0%) had pathogenic germline mutations in other non-RB1 cancer predisposition genes. Significant clinicopathologic correlations included: vitreous seeds associated with 1q gains and 16q loss of heterozygosity (BH-corrected p-value = 0.008, 0.004; OR = 12.6, 26.7, respectively). BCOR mutations were associated with poor prognosis, specifically metastases-free survival (MFS) (nominal p-value 0.03). Furthermore, retinoblastoma patients can have non-RB1 germline mutations in other cancer-associated genes. No two specimens had the identical genetic profile, emphasizing the individuality of tumors with the same clinical diagnosis.
Collapse
|
36
|
Clinical, Genomic, and Pharmacological Study of MYCN-Amplified RB1 Wild-Type Metastatic Retinoblastoma. Cancers (Basel) 2020; 12:cancers12092714. [PMID: 32971811 PMCID: PMC7565107 DOI: 10.3390/cancers12092714] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/11/2020] [Accepted: 08/23/2020] [Indexed: 12/15/2022] Open
Abstract
An uncommon subgroup of unilateral retinoblastomas with highly aggressive histological features, lacking aberrations in RB1 gene with high-level amplification of MYCN (MCYNamplRB1+/+) has only been described as intra-ocular cases treated with initial enucleation. Here, we present a comprehensive clinical, genomic, and pharmacological analysis of two cases of MCYNamplRB1+/+ with orbital and cervical lymph node involvement, but no central nervous system spread, rapidly progressing to fatal disease due to chemoresistance. Both patients showed in common MYCN high amplification and chromosome 16q and 17p loss. A somatic mutation in TP53, in homozygosis by LOH, and high chromosomal instability leading to aneuploidy was identified in the primary ocular tumor and sites of dissemination of one patient. High-throughput pharmacological screening was performed in a primary cell line derived from the lymph node dissemination of one case. This cell line showed resistance to broad spectrum chemotherapy consistent with the patient's poor response but sensitivity to the synergistic effects of panobinostat-bortezomib and carboplatin-panobinostat associations. From these cells we established a cell line derived xenograft model that closely recapitulated the tumor dissemination pattern of the patient and served to evaluate whether triple chemotherapy significantly prolonged survival of the animals. We report novel genomic alterations in two cases of metastatic MCYNamplRB1+/+ that may be associated with chemotherapy resistance and in vitro/in vivo models that serve as basis for tailoring therapy in these cases.
Collapse
|
37
|
Retinoblastoma: Etiology, Modeling, and Treatment. Cancers (Basel) 2020; 12:cancers12082304. [PMID: 32824373 PMCID: PMC7465685 DOI: 10.3390/cancers12082304] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [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
|
38
|
Zhang S, Zeng T, Hu B, Zhang YH, Feng K, Chen L, Niu Z, Li J, Huang T, Cai YD. Discriminating Origin Tissues of Tumor Cell Lines by Methylation Signatures and Dys-Methylated Rules. Front Bioeng Biotechnol 2020; 8:507. [PMID: 32528944 PMCID: PMC7264161 DOI: 10.3389/fbioe.2020.00507] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
DNA methylation is an essential epigenetic modification for multiple biological processes. DNA methylation in mammals acts as an epigenetic mark of transcriptional repression. Aberrant levels of DNA methylation can be observed in various types of tumor cells. Thus, DNA methylation has attracted considerable attention among researchers to provide new and feasible tumor therapies. Conventional studies considered single-gene methylation or specific loci as biomarkers for tumorigenesis. However, genome-scale methylated modification has not been completely investigated. Thus, we proposed and compared two novel computational approaches based on multiple machine learning algorithms for the qualitative and quantitative analyses of methylation-associated genes and their dys-methylated patterns. This study contributes to the identification of novel effective genes and the establishment of optimal quantitative rules for aberrant methylation distinguishing tumor cells with different origin tissues.
Collapse
Affiliation(s)
- Shiqi Zhang
- School of Life Sciences, Shanghai University, Shanghai, China.,Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Tao Zeng
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | - Bin Hu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Yu-Hang Zhang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kaiyan Feng
- Department of Computer Science, Guangdong AIB Polytechnic, Guangzhou, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Zhibin Niu
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Jianhao Li
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
39
|
Jiménez I, Laé M, Tanguy ML, Savignoni A, Gauthier-Villars M, Desjardins L, Cassoux N, Dendale R, Rodriguez J, Doz F, Brisse HJ, Aerts I. Craniofacial second primary tumors in patients with germline retinoblastoma previously treated with external beam radiotherapy: A retrospective institutional analysis. Pediatr Blood Cancer 2020; 67:e28158. [PMID: 31904159 DOI: 10.1002/pbc.28158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/08/2022]
Abstract
BACKGROUND The long-term survival of germline retinoblastoma patients is decreased due to the risk of second primary tumors (SPTs) that occur years after the diagnosis of retinoblastoma. This risk is related to genetic predisposition and other factors, such as the treatment of retinoblastoma by external beam radiotherapy (EBRT). PROCEDURE We studied the incidence, risk factors, and prognosis of specific craniofacial SPTs developed within the margins of radiation field in a cohort of 209 patients with germline retinoblastoma treated with EBRT at our institution between 1977 and 2010. Clinical characteristics, survival, incidence, and histology of craniofacial SPTs were recorded. RESULTS Fifty-three of the 209 patients developed 60 distinct craniofacial SPTs in irradiated field with a median time from EBRT of 16.9 years (4-35) and a median follow-up of 24.8 years (5.3-40). Osteosarcoma (33.3%) and undifferentiated sarcoma (23.3%) were the more prevalent histological entities. Benign tumors (16.7%) also occurred. The cumulative incidence of craniofacial SPTs reached 32.6% at 35 years after EBRT, and the median survival after diagnosis was five years. In our series, irradiation under 12 months of age, bilateral EBRT, or previous treatment of retinoblastoma with chemotherapy did not significantly increase the risk of craniofacial SPTs. CONCLUSIONS This work presents a strong argument to avoid EBRT in the management of retinoblastoma and emphasizes the high risk and poor prognosis of specific craniofacial SPTs. This study also points to the question of the need and benefits of special programs for early detection of craniofacial SPTs in survivors of irradiated germline retinoblastoma.
Collapse
Affiliation(s)
- Irene Jiménez
- Institut Curie, SIREDO Oncology Center (Care, Innovation and Research for children and AYA with Cancer), PSL Research University, Paris, France
| | - Marick Laé
- Institut Curie, Department of Pathology, PSL Research University, Paris, France.,Centre Henri Becquerel, Department of Pathology, INSERM U1245, UNIROUEN, University of Normandie, Rouen, France
| | - Marie-Laure Tanguy
- Institut Curie, Biostatistics Department, PSL Research University, Paris, France
| | - Alexia Savignoni
- Institut Curie, Biostatistics Department, PSL Research University, Paris, France
| | | | - Laurence Desjardins
- Institut Curie, Ocular Oncology Service, PSL Research University, Paris, France
| | - Nathalie Cassoux
- Institut Curie, Ocular Oncology Service, PSL Research University, Paris, France.,Université de Paris, Paris, France
| | - Rémi Dendale
- Institut Curie, Radiation Oncology Service, PSL Research University, Paris, France
| | - Joseph Rodriguez
- Hôpital Jean Bernard, Department of Surgery, Head and Neck Surgery, Valenciennes, France
| | - François Doz
- Université de Paris, Paris, France.,Institut Curie, SIREDO Oncology Center (Care, Innovation and Research for Children and AYA with Cancer), Paris, France
| | - Hervé J Brisse
- Institut Curie, Imaging Department, PSL Research University, Paris, France
| | - Isabelle Aerts
- Institut Curie, SIREDO Oncology Center (Care, Innovation and Research for children and AYA with Cancer), PSL Research University, Paris, France
| |
Collapse
|
40
|
Pascual-Pasto G, Bazan-Peregrino M, Olaciregui NG, Restrepo-Perdomo CA, Mato-Berciano A, Ottaviani D, Weber K, Correa G, Paco S, Vila-Ubach M, Cuadrado-Vilanova M, Castillo-Ecija H, Botteri G, Garcia-Gerique L, Moreno-Gilabert H, Gimenez-Alejandre M, Alonso-Lopez P, Farrera-Sal M, Torres-Manjon S, Ramos-Lozano D, Moreno R, Aerts I, Doz F, Cassoux N, Chapeaublanc E, Torrebadell M, Roldan M, König A, Suñol M, Claverol J, Lavarino C, Carmen de T, Fu L, Radvanyi F, Munier FL, Catalá-Mora J, Mora J, Alemany R, Cascalló M, Chantada GL, Carcaboso AM. Therapeutic targeting of the RB1 pathway in retinoblastoma with the oncolytic adenovirus VCN-01. Sci Transl Med 2020; 11:11/476/eaat9321. [PMID: 30674657 DOI: 10.1126/scitranslmed.aat9321] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/22/2018] [Indexed: 12/12/2022]
Abstract
Retinoblastoma is a pediatric solid tumor of the retina activated upon homozygous inactivation of the tumor suppressor RB1 VCN-01 is an oncolytic adenovirus designed to replicate selectively in tumor cells with high abundance of free E2F-1, a consequence of a dysfunctional RB1 pathway. Thus, we reasoned that VCN-01 could provide targeted therapeutic activity against even chemoresistant retinoblastoma. In vitro, VCN-01 effectively killed patient-derived retinoblastoma models. In mice, intravitreous administration of VCN-01 in retinoblastoma xenografts induced tumor necrosis, improved ocular survival compared with standard-of-care chemotherapy, and prevented micrometastatic dissemination into the brain. In juvenile immunocompetent rabbits, VCN-01 did not replicate in retinas, induced minor local side effects, and only leaked slightly and for a short time into the blood. Initial phase 1 data in patients showed the feasibility of the administration of intravitreous VCN-01 and resulted in antitumor activity in retinoblastoma vitreous seeds and evidence of viral replication markers in tumor cells. The treatment caused local vitreous inflammation but no systemic complications. Thus, oncolytic adenoviruses targeting RB1 might provide a tumor-selective and chemotherapy-independent treatment option for retinoblastoma.
Collapse
Affiliation(s)
- Guillem Pascual-Pasto
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | - Nagore G Olaciregui
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | | | - Daniela Ottaviani
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - Klaus Weber
- AnaPath GmbH, Oberbuchsiten 4625, Switzerland
| | - Genoveva Correa
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Sonia Paco
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Monica Vila-Ubach
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Maria Cuadrado-Vilanova
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Helena Castillo-Ecija
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Gaia Botteri
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Laura Garcia-Gerique
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Helena Moreno-Gilabert
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | | | | | | | - Silvia Torres-Manjon
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Dolores Ramos-Lozano
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Isabelle Aerts
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - François Doz
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Paris Descartes University, 75006 Paris, France
| | - Nathalie Cassoux
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Paris Descartes University, 75006 Paris, France.,Institut Curie, Ophthalmic Oncology, 75248 Paris, France
| | - Elodie Chapeaublanc
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | - Montserrat Torrebadell
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Monica Roldan
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pathology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Andrés König
- Vivotecnia Research S.L., Tres Cantos, Madrid 28760, Spain
| | - Mariona Suñol
- Pathology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Joana Claverol
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Clinical Trials Unit, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Cinzia Lavarino
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Torres Carmen de
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Ligia Fu
- Pediatric Hematology-Oncology, Hospital Escuela Universitario, Tegucigalpa, Honduras
| | - François Radvanyi
- Institut Curie, CNRS, UMR144, SIREDO Oncology Center, 75248 Paris, France.,Institut Curie, PSL Research University, 75248 Paris, France
| | | | | | - Jaume Mora
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| | - Ramón Alemany
- Translational Research Laboratory, IDIBELL-Institut Catala d'Oncologia, 08908, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Manel Cascalló
- VCN Biosciences, Sant Cugat del Valles, Barcelona 08174, Spain
| | - Guillermo L Chantada
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain.,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain.,Hospital de Pediatria JP Garrahan, Buenos Aires 1245, Argentina.,CONICET, Buenos Aires 1245, Argentina
| | - Angel M Carcaboso
- Institut de Recerca Sant Joan de Deu, Barcelona 08950, Spain. .,Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona 08950, Spain
| |
Collapse
|
41
|
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: 4.5] [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
|
42
|
Two Cases of Oculofaciocardiodental (OFCD) Syndrome due to X-Linked BCOR Mutations Presenting with Infantile Hemangiomas: Phenotypic Overlap with PHACE Syndrome. Case Rep Genet 2020; 2019:9382640. [PMID: 31956451 PMCID: PMC6949664 DOI: 10.1155/2019/9382640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/07/2019] [Indexed: 01/16/2023] Open
Abstract
Background Oculofaciocardiodental (OFCD) syndrome is due to mutations in BCOR (BCL-6 corepressor). OFCD has phenotypic overlaps with PHACE syndrome (Posterior fossa anomalies, Hemangioma, Arterial anomalies, Cardiac defects, Eye anomalies). Infantile hemangiomas are a key diagnostic criterion for PHACE, but not for OFCD. A previous study reported two cases of infantile hemangiomas in OFCD, but the authors could not exclude chance association. Case Presentation We describe two novel cases of female patients (one initially diagnosed with PHACE syndrome), both of whom had infantile hemangiomas. Ophthalmological findings were consistent with oculofaciocardiodental (OFCD) syndrome. Upon genetic testing, these two females were determined to have X-linked BCOR mutations confirming OFCD syndrome diagnoses. Conclusion These case reports add support to the hypothesis that infantile hemangiomas may be a feature of OFCD. BCOR may potentially be within a pathway of genes involved in PHACE syndrome and/or in infantile hemangioma formation.
Collapse
|
43
|
Afshar AR, Pekmezci M, Bloomer MM, Cadenas NJ, Stevers M, Banerjee A, Roy R, Olshen AB, Van Ziffle J, Onodera C, Devine WP, Grenert JP, Bastian BC, Solomon DA, Damato BE. Next-Generation Sequencing of Retinoblastoma Identifies Pathogenic Alterations beyond RB1 Inactivation That Correlate with Aggressive Histopathologic Features. Ophthalmology 2019; 127:804-813. [PMID: 32139107 DOI: 10.1016/j.ophtha.2019.12.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To determine the usefulness of a comprehensive, targeted-capture next-generation sequencing (NGS) assay for the clinical management of children undergoing enucleation for retinoblastoma. DESIGN Cohort study. PARTICIPANTS Thirty-two children with retinoblastoma. METHODS We performed targeted NGS using the UCSF500 Cancer Panel (University of California, San Francisco, San Francisco, CA) on formalin-fixed, paraffin-embedded tumor tissue along with constitutional DNA isolated from peripheral blood, buccal swab, or uninvolved optic nerve. Peripheral blood samples were also sent to a commercial laboratory for germline RB1 mutation testing. MAIN OUTCOME MEASURES Presence or absence of germline RB1 mutation or deletion, tumor genetic profile, and association of genetic alterations with clinicopathologic features. RESULTS Germline mutation or deletion of the RB1 gene was identified in all children with bilateral retinoblastoma (n = 12), and these NGS results were 100% concordant with commercial germline RB1 mutation analysis. In tumor tissue tested with NGS, biallelic inactivation of RB1 was identified in 28 tumors and focal MYCN amplification was identified in 4 tumors (2 with wild-type RB1 and 2 with biallelic RB1 inactivation). Additional likely pathogenic alterations beyond RB1 were identified in 13 tumors (41%), several of which have not been reported previously in retinoblastoma. These included focal amplifications of MDM4 and RAF1, as well as damaging mutations involving BCOR, ARID1A, MGA, FAT1, and ATRX. The presence of additional likely pathogenetic mutations beyond RB1 inactivation was associated with aggressive histopathologic features, including higher histologic grade and anaplasia, and also with both unilateral and sporadic disease. CONCLUSIONS Comprehensive NGS analysis reliably detects relevant mutations, amplifications, and chromosomal copy number changes in retinoblastoma. The presence of genetic alterations beyond RB1 inactivation correlates with aggressive histopathologic features.
Collapse
Affiliation(s)
- Armin R Afshar
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California.
| | - Melike Pekmezci
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Michele M Bloomer
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Nicola J Cadenas
- Cancer Genetics and Prevention Program, University of California, San Francisco, San Francisco, California
| | - Meredith Stevers
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Anuradha Banerjee
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Division of Hematology-Oncology, Department of Pediatrics, University of California, San Francisco, San Francisco, California
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Computational Biology and Informatics, University of California, San Francisco, San Francisco, California
| | - Adam B Olshen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Computational Biology and Informatics, University of California, San Francisco, San Francisco, California; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Jessica Van Ziffle
- Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California
| | - Courtney Onodera
- Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California
| | - W Patrick Devine
- Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California
| | - James P Grenert
- Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California
| | - Boris C Bastian
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California; Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - David A Solomon
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California; Department of Pathology, University of California, San Francisco, San Francisco, California; Clinical Cancer Genomics Laboratory, University of California, San Francisco, San Francisco, California
| | - Bertil E Damato
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
44
|
Berry NK, Scott RJ, Rowlings P, Enjeti AK. Clinical use of SNP-microarrays for the detection of genome-wide changes in haematological malignancies. Crit Rev Oncol Hematol 2019; 142:58-67. [PMID: 31377433 DOI: 10.1016/j.critrevonc.2019.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/17/2022] Open
Abstract
Single nucleotide polymorphism (SNP) microarrays are commonly used for the clinical investigation of constitutional genomic disorders; however, their adoption for investigating somatic changes is being recognised. With increasing importance being placed on defining the cancer genome, a shift in technology is imperative at a clinical level. Microarray platforms have the potential to become frontline testing, replacing or complementing standard investigations such as FISH or karyotype. This 'molecular karyotype approach' exemplified by SNP-microarrays has distinct advantages in the investigation of several haematological malignancies. A growing body of literature, including guidelines, has shown support for the use of SNP-microarrays in the clinical laboratory to aid in a more accurate definition of the cancer genome. Understanding the benefits of this technology along with discussing the barriers to its implementation is necessary for the development and incorporation of SNP-microarrays in a clinical laboratory for the investigation of haematological malignancies.
Collapse
Affiliation(s)
- Nadine K Berry
- Department of Haematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Biomedical Sciences and Pharmacy, University of Newcastle, New South Wales, Australia; Department of Molecular Medicine, NSW Health Pathology, Newcastle, New South Wales, Australia.
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, New South Wales, Australia; Department of Molecular Medicine, NSW Health Pathology, Newcastle, New South Wales, Australia
| | - Philip Rowlings
- Department of Haematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Medicine and Public Health, University Newcastle, New South Wales, Australia
| | - Anoop K Enjeti
- Department of Haematology, Calvary Mater Hospital, Newcastle, New South Wales, Australia; School of Medicine and Public Health, University Newcastle, New South Wales, Australia
| |
Collapse
|
45
|
Akdeniz D, Tuncer SB, Kebudi R, Celik B, Kuru G, Kilic S, Sukruoglu Erdogan O, Avsar M, Buyukkapu Bay S, Tuncer S, Yazici H. Investigation of new candidate genes in retinoblastoma using the TruSight One "clinical exome" gene panel. Mol Genet Genomic Med 2019; 7:e785. [PMID: 31207142 PMCID: PMC6687622 DOI: 10.1002/mgg3.785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/07/2019] [Accepted: 05/17/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Retinoblastoma (Rb) is the most prevalent intraocular pediatric malignancy of the retina. Significant genetic factors are known to have a role in the development of Rb. METHODS Here, we report the mutation status of 4813 clinically significant genes in six patients with noncarrier of RB1 gene mutation and having normal RB1 promoter methylation from three families having higher risk for developing Rb in the study. RESULTS A total of 27 variants were detected in the study. Heterozygous missense variants c.1162G > A (p.Gly388Arg) in the FGFR4 gene; c.559C > T (p.Pro187Ser) in the NQO1 gene were identified. The family based evaluation of the variants showed that the variant, c.714T > G (p.Tyr238Ter), in the CLEC7A gene in first family; the variant, c.55C > T (p.Arg19Ter), in the APOC3 gene and the variant, c.1171C > T (p.Gln391Ter), in the MUTYH gene in second family; and the variant, c.211G > A (p.Gly71Arg), in the UGT1A1 gene in the third family, were found statistically significant (p < 0.05). CONCLUSION This study might be an important report on emphazing the mutational status of other genes in patients without RB1 gene mutations and having high risk for developing Rb. The study also indicates the interaction between the retinoic acid pathway and Rb oncogenesis for the first time.
Collapse
Affiliation(s)
- Demet Akdeniz
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Seref Bugra Tuncer
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Rejin Kebudi
- Division of Pediatric Hematology-Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey.,Division of Pediatric Hematology-Oncology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Betul Celik
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Gozde Kuru
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Seda Kilic
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Ozge Sukruoglu Erdogan
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Mukaddes Avsar
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Sema Buyukkapu Bay
- Division of Pediatric Hematology-Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| | - Samuray Tuncer
- Istanbul Medical Faculty, Department of Ophthalmology, Istanbul University, Istanbul, Turkey
| | - Hulya Yazici
- Division of Cancer Genetics, Department of Basic Oncology, Oncology Institute, Istanbul University, Istanbul, Turkey
| |
Collapse
|
46
|
Munier FL, Beck-Popovic M, Chantada GL, Cobrinik D, Kivelä TT, Lohmann D, Maeder P, Moll AC, Carcaboso AM, Moulin A, Schaiquevich P, Bergin C, Dyson PJ, Houghton S, Puccinelli F, Vial Y, Gaillard MC, Stathopoulos C. Conservative management of retinoblastoma: Challenging orthodoxy without compromising the state of metastatic grace. "Alive, with good vision and no comorbidity". Prog Retin Eye Res 2019; 73:100764. [PMID: 31173880 DOI: 10.1016/j.preteyeres.2019.05.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/25/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Retinoblastoma is lethal by metastasis if left untreated, so the primary goal of therapy is to preserve life, with ocular survival, visual preservation and quality of life as secondary aims. Historically, enucleation was the first successful therapeutic approach to decrease mortality, followed over 100 years ago by the first eye salvage attempts with radiotherapy. This led to the empiric delineation of a window for conservative management subject to a "state of metastatic grace" never to be violated. Over the last two decades, conservative management of retinoblastoma witnessed an impressive acceleration of improvements, culminating in two major paradigm shifts in therapeutic strategy. Firstly, the introduction of systemic chemotherapy and focal treatments in the late 1990s enabled radiotherapy to be progressively abandoned. Around 10 years later, the advent of chemotherapy in situ, with the capitalization of new routes of targeted drug delivery, namely intra-arterial, intravitreal and now intracameral injections, allowed significant increase in eye preservation rate, definitive eradication of radiotherapy and reduction of systemic chemotherapy. Here we intend to review the relevant knowledge susceptible to improve the conservative management of retinoblastoma in compliance with the "state of metastatic grace", with particular attention to (i) reviewing how new imaging modalities impact the frontiers of conservative management, (ii) dissecting retinoblastoma genesis, growth patterns, and intraocular routes of tumor propagation, (iii) assessing major therapeutic changes and trends, (iv) proposing a classification of relapsing retinoblastoma, (v) examining treatable/preventable disease-related or treatment-induced complications, and (vi) appraising new therapeutic targets and concepts, as well as liquid biopsy potentiality.
Collapse
Affiliation(s)
- Francis L Munier
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland.
| | - Maja Beck-Popovic
- Unit of Pediatric Hematology-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Guillermo L Chantada
- Hemato-Oncology Service, Hospital JP Garrahan, Buenos Aires, Argentina; Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - David Cobrinik
- The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; USC Roski Eye Institute, Department of Biochemistry & Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Tero T Kivelä
- Department of Ophthalmology, Ocular Oncology and Pediatric Ophthalmology Services, Helsinki University Hospital, Helsinki, Finland
| | - Dietmar Lohmann
- Eye Oncogenetics Research Group, Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Philippe Maeder
- Unit of Neuroradiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Annette C Moll
- UMC, Vrije Universiteit Amsterdam, Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Angel Montero Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Alexandre Moulin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paula Schaiquevich
- Unit of Clinical Pharmacokinetics, Hospital de Pediatria JP Garrahan, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Ciara Bergin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Susan Houghton
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Francesco Puccinelli
- Interventional Neuroradiology Unit, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Yvan Vial
- Materno-Fetal Medicine Unit, Woman-Mother-Child Department, University Hospital of Lausanne, Switzerland
| | - Marie-Claire Gaillard
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Christina Stathopoulos
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
47
|
Abstract
BCOR is a gene that encodes for an epigenetic regulator involved in the specification of cell differentiation and body structure development and takes part in the noncanonical polycomb repressive complex 1. This review provides a comprehensive summary of BCOR’s involvement in oncology, illustrating that various BCOR aberrations, such as the internal tandem duplications of the PCGF Ub-like fold discriminator domain and different gene fusions (mainly BCOR–CCNB3, BCOR–MAML3 and ZC3H7B–BCOR), represent driver elements of various sarcomas such as clear cell sarcoma of the kidney, primitive mesenchymal myxoid tumor of infancy, small round blue cell sarcoma, endometrial stromal sarcoma and histologically heterogeneous CNS neoplasms group with similar genomic methylation patterns known as CNS-HGNET-BCOR. Furthermore, other BCOR alterations (often loss of function mutations) recur in a large variety of mesenchymal, epithelial, neural and hematological tumors, suggesting a central role in cancer evolution.
Collapse
Affiliation(s)
- Annalisa Astolfi
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Michele Fiore
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Fraia Melchionda
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Valentina Indio
- 'Giorgio Prodi' Cancer Research Center, University of Bologna, 40138 Bologna, Italy
| | - Salvatore N Bertuccio
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| | - Andrea Pession
- Pediatric Oncology & Hematology Unit 'Lalla Seràgnoli', S Orsola-Malpighi Hospital, 40138 Bologna, Italy.,Department of Medical & Surgical Sciences, University of Bologna, S Orsola-Malpighi Hospital, 40138 Bologna, Italy
| |
Collapse
|
48
|
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
|
49
|
Zhang L, Dong Y, Wang Y, Gao J, Lv J, Sun J, Li M, Wang M, Zhao Z, Wang J, Xu W. Long non-coding RNAs in ocular diseases: new and potential therapeutic targets. FEBS J 2019; 286:2261-2272. [PMID: 30927500 DOI: 10.1111/febs.14827] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/21/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Long non-coding RNAs (lncRNAs) are non-protein coding transcripts containing more than 200 nucleotides. In the past, lncRNAs were considered as 'transcript noise' or 'pseudogenes' and were thus ignored. However, in recent years, lncRNAs have been proven to regulate gene expression at the epigenetic, transcriptional and translational level, and thereby influence cell proliferation, apoptosis, viability, immune response and oxidative stress. Furthermore, increasing evidence points to their involvement in different diseases, including cancer and heart diseases. Recently, lncRNAs were shown to be differentially expressed in ocular tissues and play a significant role in the pathogenesis of ophthalmological disorders such as glaucoma, corneal diseases, cataract, diabetic retinopathy, proliferative vitreoretinopathy and ocular tumors. In this review, we summarize the classification and mechanisms of known lncRNAs, while detailing their biological functions and roles in ocular diseases. Moreover, we provide a concise review of the clinical relevance of lncRNAs as novel, potential therapeutic targets in the treatment of eye diseases.
Collapse
Affiliation(s)
- Lixia Zhang
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Yanhan Dong
- Institute for Translational Medicine, Qingdao University, China
| | - Yujie Wang
- The Clinical Laboratory of Qingdao Municipal Hospital, China
| | - Jinning Gao
- Institute for Translational Medicine, Qingdao University, China
| | - Jiayi Lv
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Jingguo Sun
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Mengjie Li
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Meng Wang
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Zhihong Zhao
- Department of Inspection, The Medical Faculty of Qingdao University, China
| | - Jianxun Wang
- Institute for Translational Medicine, Qingdao University, China
| | - Wenhua Xu
- Department of Inspection, The Medical Faculty of Qingdao University, China
| |
Collapse
|
50
|
Pellestor F. Chromoanagenesis: cataclysms behind complex chromosomal rearrangements. Mol Cytogenet 2019; 12:6. [PMID: 30805029 PMCID: PMC6371609 DOI: 10.1186/s13039-019-0415-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background During the last decade, genome sequencing projects in cancer genomes as well as in patients with congenital diseases and healthy individuals have led to the identification of new types of massive chromosomal rearrangements arising during single chaotic cellular events. These unanticipated catastrophic phenomenon are termed chromothripsis, chromoanasynthesis and chromoplexis., and are grouped under the name of “chromoanagenesis”. Results For each process, several specific features have been described, allowing each phenomenon to be distinguished from each other and to understand its mechanism of formation and to better understand its aetiology. Thus, chromothripsis derives from chromosome shattering followed by the random restitching of chromosomal fragments with low copy-number change whereas chromoanasynthesis results from erroneous DNA replication of a chromosome through serial fork stalling and template switching with variable copy-number gains, and chromoplexy refers to the occurrence of multiple inter-and intra-chromosomal translocations and deletions with little or no copy-number alterations in prostate cancer. Cumulating data and experimental models have shown that chromothripsis and chromoanasynthesis may essentially result from lagging chromosome encapsulated in micronuclei or telomere attrition and end-to-end telomere fusion. Conclusion The concept of chromanagenesis has provided new insight into the aetiology of complex structural rearrangements, the connection between defective cell cycle progression and genomic instability, and the complexity of cancer evolution. Increasing reported chromoanagenesis events suggest that these chaotic mechanisms are probably much more frequent than anticipated.
Collapse
Affiliation(s)
- Franck Pellestor
- Unit of Chromosomal Genetics, Department of Medical Genetics, Arnaud de Villeneuve Hospital, Montpellier CHRU, 371, avenue du Doyen Gaston Giraud, 34295 Montpellier cedex 5, France.,INSERM 1183 Unit «Genome and Stem Cell Plasticity in Development and Aging », Institute of Regenerative Medicine and Biotherapies, St Eloi Hospital, Montpellier, France
| |
Collapse
|