Clinical evaluation of RB1 genetic testing reveals novel mutations in Vietnamese patients with retinoblastoma

  • Authors:
    • Chinh Quoc Hoang
    • Hong-Quan Duong
    • Nguyen Thanh Nguyen
    • Sy Anh Hao Nguyen
    • Cuong Nguyen
    • Bo Duy Nguyen
    • Lan Tuyet Phung
    • Dung Thuy Nguyen
    • Chau Thi Minh Pham
    • Trang Le Doan
    • Mai Hoang Tran
  • View Affiliations

  • Published online on: July 3, 2021     https://doi.org/10.3892/mco.2021.2344
  • Article Number: 182
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Clinical evaluation of the genetic testing strategy is essential for ensuring the correct determination of mutation carriers. The current study retrospectively analyzed genetic and clinicopathological data from 62 Vietnamese patients with retinoblastoma (RB) referred to the Vinmec Hi‑Tech Center for RB transcriptional corepressor 1 (RB1) genetic testing between 2017 and 2019. The present study aimed to evaluate the sensitivity of the Next Generation Sequencing (NGS) method to identify novel RB1 mutations, and to consider using age at diagnosis as a risk factor. Genomic DNA was analyzed with custom panel based targeted NGS. NGS was performed on the Beijing Genomics Institute (BGI) sequencing platform, and pathogenic or likely pathogenic variants were confirmed by Sanger sequencing, quantitative PCR (qPCR) or Multiplex Ligation‑dependent Probe Amplification assay (MLPA). Constitutional RB1 variants were identified in 100% (25/25) of the bilateral cases, while several common previously reported RB1 mutations were also recorded. In addition, in Vietnamese patients with RB, nine novel RB1 mutations were identified. Children aged between 0‑36 months were more likely to be RB1 carriers compared with those aged >36 months. The current findings indicated that the NGS method implemented in the Vinmec Hi‑Tech Center was highly accurate, and age at diagnosis may be used to assess the risk of hereditary RB. Furthermore, the newly identified RB1 mutations may provide additional data to improve the current understanding of the mechanisms underlying RB1 inactivation and the development of rapid assays for detecting RB1 mutations. Overall, the present study suggested that NGS may be applied for detecting germline RB1 mutations in routine clinical practice.

Introduction

Retinoblastoma (RB) is the most common intraocular malignancy, characterized by high mortality if not detected early and treated promptly. Early diagnosis and intervention play a key role in the successful treatment of RB (1). Delayed diagnosis of RB for >6 months from the first clinical sign has been reported to be associated with a mortality rate of ~70% (2). Therefore, although in patients with RB diagnosed at stage A of the disease the eyes or vision can be salvaged, currently, there is not an effective treatment approach for those diagnosed at stage E (3,4).

RB is considered as a monogenetic hereditary type of cancer since 97% of RB cases are caused by the disruption of the RB transcriptional corepressor 1 (RB1) tumor suppressor gene (5). The RB1-encoded protein (pRB) acts as a scaffold protein, which interacts with other proteins to regulate multiple cellular processes essential for cell fate and function. Consequently, RB1 deficiency may predispose cells to tumorigenesis. In fact, it has been reported that RB1 inactivation is detected in several types of cancer. Since pRB interacts with other proteins through cyclin folds in the N-terminus and pocket domain, and intrinsically disordered structures in the C-terminus, a wide spectrum of mutations dispersed throughout the RB1 gene has been identified in patients with cancer (6).

It has been also reported that 45% of patients with RB are suffering from the inherited type of the disease (RB1 carriers), where the first allele of RB1 is mutated during preconception or shortly after conception, predisposing the child to retinal tumorigenesis (7). The remaining RB1 allele is functionally lost during retinal development, promoting the initiation of RB in either both eyes (bilateral RB) or one eye (unilateral RB) with multifocal tumors. ‘Non-heritable or sporadic RB’ (non-RB1 carriers) is commonly referred to as patients without germline RB1 mutations, who usually present with unifocal tumors in one eye (8). Patients with hereditary RB exhibit a worse prognosis, are sensitive to certain treatments, have a high risk of developing second primary malignancies and can pass the mutations on to their offspring (3,9). Therefore, identifying germline RB1 mutations is of great importance for implementing the appropriate treatment approach, and assessing the risk of developing second primary malignancies, both secondary RB and other primary malignancies, in patients with RB, and the risk of RB onset in the patient's relatives.

Genetic testing and counseling (GTC) are recommended for all patients with RB and are integrated in the management of RB in developed countries (10), thus resulting in high survival rate of patients with RB and cost-effective medical treatments for RB (11,12). GTC for patients with bilateral or familial RB is straightforward. However, for unilateral, non-familial RB cases, GTC is not as easy due to the low risk of RB1 mutation carriers, and the inefficient detection of germline RB1 mosaicism (13). Genetic testing for RB1 is a time-consuming and expensive procedure given the large size of the RB1 gene, which can be inactivated by multiple mutations, and the absence of mutational hotspots (14,15). Direct sequencing is widely applied for detecting RB1 mutations, however, this method is not recommended for identifying low allelic-fraction variants (16). For the detection of these types of variants, PCR can be applied only when the mutations are already known. In addition, Multiplex Ligation-dependent Probe Amplification (MLPA), quantitative PCR (qPCR) or array Comparative Genomic Hybridization (aCGH) can identify large RB1 rearrangements. The combination of the aforementioned methods is essential for detecting all possible RB1 mutations (17-19). Recently, Next Generation Sequencing (NGS) has been implemented as rapid and effective strategy for identification of RB1 mutations since all variations can be detected in a single test, thus providing a number of advantages, including high sensitivity and cost-effectiveness (20-25). In Vietnam, Sanger sequencing coupled with MLPA (SS-MLPA) could detect germline RB1 mutations in 82-84% of bilateral cases (26,27). Nevertheless, the sensitivity of NGS in a routine clinical practice remains unknown.

The two-hit hypothesis suggests that patients with bilateral or unilateral multifocal RB, and/or diagnosed at an early age are more likely to carry germline RB1 mutations (28). This hypothesis is supported by clinical data. Therefore, a study demonstrated that up to 100% of patients with bilateral carried germline RB1 mutations, and their age at diagnosis was 10 months younger compared with that of the unilateral cases (13). Bilateral and unilateral eye diseases account for 40 and 60% of all RB cases (7), respectively. All bilateral RB cases are considered heritable, whereas ~15% of unilateral cases carry constitutional RB1 mutations (29,30). Since age at diagnosis is younger in bilateral cases, previous studies have associated age at diagnosis with germline RB1 status in order to predict age associated with increased risk of patients with unilateral disease being RB1 carriers. Unfortunately, these studies yielded conflicting results (31-34).

The current study retrospectively analyzed the clinicopathological and genetic data of patients with RB to evaluate the sensitivity of NGS for detecting constitutional RB1 variants, to detect novel germline RB1 mutations, and to consider age at diagnosis as a risk factor for patients being RB1 carriers.

Materials and methods

Patients and samples

In the current retrospective study, a total of 62 patients with RB were included, who were referred to Department of Cancer Research, Vinmec Research Institute of Stem Cell and Gene Technology and the Department of Medical Genetics, Vinmec Hi-Tech Center for genetic testing between 2017 and 2019. Signed informed consents were obtained from parents/caregivers of all subjects and the study was approved by the Vinmec's Institutional Review Board. All data, including age at diagnosis, sex, tumor stages, laterality and family history were retrieved from the patients' medical records. Genetic testing for germline RB1 variants was performed at the Beijing Genomics Institute (BGI), HongKong. The workflow for analyzing germline RB1 variants is illustrated in Fig. 1.

Preparation of tissue samples, NGS and variant calling

Genomic DNA (gDNA) was extracted from peripheral blood mononuclear cells and its concentration was quantified using a Qubit Fluorometer (Thermo Fisher Scientific, Inc.). Subsequently, gDNA was fragmented, indexed and amplified. The RB1 promoter and all its exons, plus 20 nucleotides proximal to either 5' or 3' of each exon, were captured by a BGI chip. Library size and quantity were verified using Qubit® 2.0 (Thermo Fisher Scientific, Inc.) and an Agilent Bioanalyzer 2100 (Agilent Technologies, Inc.). Sequencing was performed on the BGIseq platform. All samples identified with pathogenic or likely pathogenic variants were confirmed by Sanger Sequencing, qPCR or MLPA.

Bioinformatics analysis

The sequence reads were aligned to the human reference genome (GRCh37/Hg19) using the Burrows-Wheeler Aligner. Single nucleotide variants, insertion/deletion variants (InDel) and copy number variations were identified using the BGI internal NGS software (BGISEQ-500).

Variant annotation

Interpretation of germline variants followed the American College of Medical Genetics and Genomics (ACMG) standards and guidelines (35). The variants were designated according to nomenclature and the recommendations of the Human Genome Variation Society (36). Public databases, including Clinvar (37), Universal Mutation Database (38), Leiden Open Variation Database (LOVD) (39) and ARUP (40) were used for data analysis. The SIFT (41), POLYPHEN2(42) and Mutation taster (43) tools were applied to predict deleterious mutations, while variants were classified using the Varsome (44) and InterVar (45) classification tools.

Identification of novel variants

All variants identified by the BGI laboratory were annotated with the Ensemble Variant Effect Predictor (EVEP) tool (46). Variants with existing ID were checked on corresponding databases. The unmatched variants and those without existing ID were screened on PubMed. Variants not previously reported in public databases and PubMed were considered as novel ones. The allele frequency of the identified novel variants was investigated in four databases, including the 1000 Genomes Project (browser.1000genomes.org), the Exome Sequencing Project (esp.gs.washington.edu/drupal), the Genome Aggregation Database and the Vietnamese Genetic Variation Database (47). Deleterious novel variants were evaluated by Combined Annotation Dependent Depletion (CADD) (48) and EVEP, while their pathogenicity was interpreted using VarSome.

Statistical analysis

The differences between the characteristics of RB1 and non-RB1 carriers were compared using a χ2 test, Fisher's exact test or Student's t-test. Fisher's exact test and odds ratio (OR) were calculated using calculators provided by the Social Science Statistics website (49) and MedCalc statistical software (50), respectively.

Results

Characteristics of RB1and non-RB1 carriers

A total of 25 bilateral and 37 unilateral RB cases were included in the present study. There was no statistically significant difference in the total number of RB1 and non-RB1 carriers, as well as in sex distribution between the two groups. However, the mean age at diagnosis of RB1 carriers was significantly younger compared with that of non-RB1 carriers (22.14 vs. 32.66 months). Although 80% of all patients were diagnosed with stage E RB, the distribution of the RB stages was not statistically different between RB1 and non-RB1 carriers. Additionally, the proportion of RB1 carriers was notably higher in patients with bilateral RB and significantly decreased in the unilateral cases. Constitutional RB1 mutations were detected in 100% (25/25) and 27% (10/37) of patients with bilateral and unilateral RB, respectively (Tables I and II), suggesting a sensitivity rate of 100% for detecting germline RB1 variants using the NGS technology.

Table I

Summary of germline RB1 mutations identified in Vietnamese patients with retinoblastoma.

Table I

Summary of germline RB1 mutations identified in Vietnamese patients with retinoblastoma.

Case IDFamily historyMonths at diagnosisLateralitySexStagingRB1 germline mutationsMolecular consequencesInterpretationExon/IntronPreviously reported (Refs.)
VinRB01Yes48BilFC/EEx1_27 DelLarge rearrangementPathogenicExon 1-27(53,61-63)
VinRB02No38BilMA/EEx1_27 DelLarge rearrangementPathogenicExon 1-27(53,61-63)
VinRB03No25BilFE/CEx1_27 DelLarge rearrangementPathogenicExon 1-27(53,61-63)
VinRB04No10BilFE/AEx18_23 DupLarge rearrangementPathogenicExon 18-23(64)
VinRB05No30BilFD/EEx12_dup & c.83 C>GLarge rearrangementPathogenic & VUSExon 12 & Exon 1Novel & rs776175164
VinRB06No3BilME/Bc.210_211insAGFrameshiftPathogenicExon 2Novel
VinRB07No31BilMA/Ec.515_516insAFrameshiftPathogenicExon 5Novel
VinRB08No6BilMB/Ec.380G>AMissensePathogenicExon 3COSV57302882
VinRB09No18BilME/Cc.1072C>TNonsensePathogenicExon 11rs121913301
VinRB10No36BilMD/E c.1403_1404insAFrameshiftPathogenicExon 15Novel
VinRB11No7BilFE/Bc.1735C>TNonsensePathogenicExon 18rs121913305
VinRB12Yes6BilMB/Bc.1735C>TNonsensePathogenicExon 18rs121913305
VinRB13No5BilFB/Ec.2359C>TNonsensePathogenicExon 23rs137853293
VinRB14No1BilMB/Bc.306T>ANonsensePathogenicExon 3Novel
VinRB15No26BilFE/Ec.958C>TNonsensePathogenicExon 10rs121913300
VinRB16No29BilFE/Ec.958C>TNonsensePathogenicExon 10rs121913300
VinRB17No31BilMB/E c.1953_c.1960+1delTAAAAAAGGSpliceLikely pathogenicExon 19Novel
VinRB18No19BilFB/Ec.1128-1G>ASplicePathogenicIntron 11COSV57294096
VinRB19No20BilMB/Ec.1215+1G>ASplicePathogenicIntron 12rs587776783
VinRB20No2BilFB/Bc.1696-1G>ASplicePathogenicIntron 17COSV57329272
VinRB21No3BilFB/B c.2520+1_2520+4delGTGASplicePathogenicIntron 24rs1131690858
VinRB22No22BilME/Bc.607+1G>ASplicePathogenicIntron 6COSV57310480
VinRB23No14BilMD/Dc.607+1G>TSplicePathogenicIntron 6COSV57310480
VinRB24No54UniM/Ec.2107-2A>CSpliceLikely pathogenicIntron 20Novel
VinRB25No21BilME/Bc.719-6_719-2del TTACASpliceLikely pathogenicIntron 7Novel
VinRB26No30UniFEc.1981C>TMissensePathogenicExon 20rs137853294
VinRB27No19UniFE/Ex24 DelLarge rearrangementPathogenicExon 24(65)
VinRB28No14UniME/ c.1953_1954insAFrameshiftPathogenicExon 19rs1566234123
VinRB29Yes2UniM/Bc.958C>TNonsensePathogenicExon 10rs121913300
VinRB30No74UniF/Dc.1072C>TNonsensePathogenicExon 11rs121913301
VinRB31No17UniFE/c.1303G>TNonsenseLikely pathogenicExon 13COSV57313162
VinRB32Yes34UniME/c.1735C>TNonsensePathogenicExon 18rs121913305
VinRB33No21BilFD/Ec.958C>TNonsensePathogenicExon 10rs121913300
VinRB34No24UniF/Ec.2106+1G>CSpliceLikely pathogenicIntron 20Novel
VinRB35No35UniMD/c.264+1G>CSplicePathogenicIntron 2COSV57317171
VinRB36No14UniFE/No    
VinRB37No10UniF/ENo    
VinRB38No11UniME/No    
VinRB39No22UniFE/No    
VinRB40No30UniME/No    
VinRB41No24UniF/ENo    
VinRB42No25UniFE/No    
VinRB43No57UniM/DNo    
VinRB44No25UniME/No    
VinRB45No17UniMENo    
VinRB46No31UniM/ENo    
VinRB47No22UniME/No    
VinRB48No108UniFE/No    
VinRB49No59UniFE/No    
VinRB50No84UniF/ENo    
VinRB51No8UniME/No    
VinRB52No10UniMD/No    
VinRB53No8UniFE/No    
VinRB54No44UniME/No    
VinRB55No23UniME/No    
VinRB56No49UniFD/No    
VinRB57No21UniFD/No    
VinRB58No56UniME/No    
VinRB59No41UniFE/No    
VinRB60No33UniME/No    
VinRB61No42UniME/No    
VinRB62No8UniF/ENo    

[i] RB stages were classified according to the International Classification of Intraocular Retinoblastoma (66) as follows: A, small post-equatorial tumor confined to the retina; B, foveal tumor confined to the retina; C, retinal tumor surrounded by a cuff of subretinal seeds; D, diffuse vitreous seeding; E, large tumor touching the crystalline lens, resulting in elevated intraocular pressure and expanded globe. F, female; M, male; Bil, bilateral; Uni, unilateral; RB1, RB transcriptional corepressor 1; Ex, exon; Ins, insertion; Del, deletion; Dup, duplication; VUS, variants of uncertain significance.

Table II

Clinicopathological distribution of RB1 (n=35) and non-RB1 (n=27) carriers by family history, laterality, sex, age at diagnosis and stage of RB tumorigenesis.

Table II

Clinicopathological distribution of RB1 (n=35) and non-RB1 (n=27) carriers by family history, laterality, sex, age at diagnosis and stage of RB tumorigenesis.

CharacteristicsRB1 carriers (n=35)Non-RB1 carriers (n=27)Both (n=62)P-value
Family history, n (%)4 (100.0)0 (0.0)4 (100.0) 
Laterality, n (%)   0.001a
     Bilateral RB25 (100.0)0 (0.0)25 (100.0) 
     Unilateral RB10 (27.0)27 (73.0)37 (100.0) 
Sex, n (%)   0.87b
     Female17 (57.0)13 (43.0)30 (100.0) 
     Male18 (56.0)14 (44.0)32 (100.0) 
Mean age, months22.1432.67 0.04c
RB stage, n (%)   0.80a
     A0 (0.0)0 (0.0)0 (0.0) 
     B4 (11.4)1 (3.7)5 (8.0) 
     C0 (0.0)0 (0.0)0 (0.0) 
     D3 (8.6)4 (14.8)7 (11.3) 
     E28 (80.0)22 (81.5)50 (80.6) 

[i] aCalculated using Fisher's exact test.

[ii] bCalculated using χ2 test.

[iii] cCalculated using Student's t-test. RB stages were classified according to the International Classification of Intraocular Retinoblastoma (66) as follows: A, small post-equatorial tumor confined to the retina; B, foveal tumor confined to the retina; C, retinal tumor surrounded by a cuff of subretinal seeds; D, diffuse vitreous seeding; E, large tumor touching the crystalline lens, resulting in elevated intraocular pressure and expanded globe. RB, retinoblastoma; RB1, RB transcriptional corepressor 1.

Novel RB1 germline mutations

A total of 28 distinct variants, including four recurrent and 24 non-recurrent ones, were identified in 56% (35/62) of patients with. RB. In addition, the four recurrent mutations were found in 33% (12/35) of RB1 carriers. Point and small InDel mutations in RB1 were dispersed along the gene. However, large rearrangements were only identified in or nearby the pocket domain. The majority of mutations (26/28) were located in the N-terminus or pocket domain of pRB (Fig. 2). The EVEP tool predicted that all these variations, except one (c.83C>G), exerted a highly disruptive effect in pRB and were classified as pathogenic variants using the Clinvar, Cosmic or Varsome databases. The c.83C>G mutation was concurrently found with an exon 12 duplication in one patient with bilateral RB. Nonsense and slice mutations were the two most frequent mutations, identified in 34 (12/35) and 31% (11/35) of all RB1 carriers, respectively. Additionally, large rearrangements, frameshift and missense mutations were detected in 17 (6/35), 11.4 (4/35) and 8.6% (3/35) of RB1 carriers, respectively (Tables I and III).

Table III

A total of 28 distinct RB1 germline alterations, including 9 novel ones, were identified in 35 patients with retinoblastoma.

Table III

A total of 28 distinct RB1 germline alterations, including 9 novel ones, were identified in 35 patients with retinoblastoma.

RB1 germline variantsMolecular consequencesAmino acid alterationsClinical significanceExisting variationsNo. of carriersSubunits
c.210_211insAGFrameshift p.(Ala74Glufs*4)PathogenicNewly identified1N-terminus
c.306T>ANonsensep.(Cys102Ter)PathogenicNewly identified1N-terminus
c.515_516insAFrameshift p.(Tyr173Ilefs*12)PathogenicNewly identified1N-terminus
c.719-6_7192 delTTACASplice-PathogenicNewly identified1N-terminus
Ex12 DupLarge rearrangement-PathogenicNewly identified1Pocket
c.1403_1404insA c.1953_c.1960+1Frameshift p.(Ser469Ilefs*6)PathogenicNewly identified1Pocket
delTAAAAAAGGSplice-PathogenicNewly identified1Pocket
c.2106+1G>CSplice-PathogenicNewly identified1Pocket
c.2107-2A>CSplice-PathogenicNewly identified1Pocket
c.83C>G (concurrent with Ex12dup)Missensep.(Pro28Arg)VUSrs7761751641N-terminus
c.1215+1G>ASplice-Pathogenicrs5877767831Pocket
c.1953_1954insAFrameshiftp.(V654Sfs*14)Pathogenicrs15662341231Pocket
c.1981C>TMissensep.(Arg661Trp)Pathogenicrs1378532941Pocket
c.2359C>TNonsensep.(Arg787Ter)Pathogenicrs1378532931Pocket
c.1735C>TNonsensep.(Arg579Ter)Pathogenicrs1219133053Pocket
c.1072C>TNonsensep.(Arg358Ter)Pathogenicrs1219133012N-terminus
c.958C>TNonsensep.(Arg320Ter)Pathogenicrs1219133004N-terminus
c.2520+1_2520+4delGTGASplice-Pathogenicrs11316908581C-terminus
Ex1_27 DelLarge rearrangement-Pathogenic(53,61-63)3Whole gene
Ex18_23 DupLarge rearrangement-Pathogenic(64)1Pocket
Ex24 DelLarge rearrangement-Pathogenic(65)1C-terminus
c.1696-1G>ASplice-PathogenicCOSV573292721Pocket
c.264+1G>CSplice-PathogenicCOSV573171711N-terminus
c.1303G>TNonsensep.(Gly435Ter)PathogenicCOSV573131621Pocket
c.607+1G>ASplice-PathogenicCOSV573104801N-terminus
c.607+1G>TSplice-PathogenicCOSV573104801N-terminus
c.380G>AMissensep.(Ser127Asn)PathogenicCOSV573028821N-terminus
c.1128-1G>ASplice-PathogenicCOSV572940961Pocket

[i] RB1, RB transcriptional corepressor 1; Ex, exon; Ins, insertion; Del, deletion; Dup, duplication.

The novel variants were defined by screening 28 RB1 mutations into the EVEP tool. Among them, 16 RB1 alterations exerted Variation ID on the Clinvar, Cosmic or LOVD databases. For the remaining 12 unidentified variants, screening on the PubMed platform was carried out. The analysis revealed three large rearrangement, including Ex1_27 DEL, Ex24 DEL and Ex13_18 DUP. Therefore, nine variants were considered as novel. These nine variations, including one large rearrangement, one nonsense, four slice and three frameshift mutations, were all null mutations, identified in seven patients with bilateral and two with unilateral RB. These variants were located at either the pocket (5/9) or N-terminus (4/9) domain. Furthermore, the EVEP tool predicted that these variants could have a significant disruptive effect on pRB, and all, except EX12 DUP, were also predicted to be among the top 1% of the most deleterious variants in the human genome by CADD. Finally, these mutations were not found in polymorphism databases, and were classified as pathogenic by VarSome (Table III; Fig. 2).

Association between age at diagnosis and the genetic status of patients with RB

The proportion of RB1 and non-RB1 carriers with an age at diagnosis of 0-36 and >36 months is presented in Table IV. It was found that the proportion of RB1 carriers with age at diagnosis of 0-36 months was notably higher compared with those of >36 months. Consequently, the relative risk of children aged between 0-36 months being RB1 carriers was significantly higher than that of children >36 months (63.3 vs. 36%; OR=3.88; 95% CI=1.04-14.4; Table IV). In terms of unilateral cases only, although the number of RB1 carriers was reduced compared with the non-RB1 ones, the difference between the two groups was not statistically significant. Accordingly, the relative risk of children aged between 0-36 months suffering from the inherited form of the disease was not markedly different compared with children >36 months of age (30.8 vs. 18%; OR=2; 95% CI=0.35=11.4; Table IV).

Table IV

Distribution of RB1 and non-RB1 carriers diagnosed at 0-36 vs. >36 months.

Table IV

Distribution of RB1 and non-RB1 carriers diagnosed at 0-36 vs. >36 months.

A, All patients with RB
CharacteristicRB1 carriers, n=35Non-RB1 carriers, n=27TotalP-value
Age, n (%)   0.036a
     0-36 months31 (63.3)18 (36.7)49 (100.0) 
     >36 months4 (36.0)9 (64.0)13 (100.0) 
OR, 0-36 vs. >36 months3.88 (95% CI, 1.04-14.40)0.26 (95% CI, 0.07-0.96) 0.040
B, Patients with unilateral RB only
CharacteristicRB1 carriers, n=10Non-RB1 carriers, n=27TotalP-value
Age, n (%)  0.620.043a
     0-36 months8 (30.8)18 (69.2)26 (100.0) 
     >36 months2 (18.0)9 (82.0)11 (100.0) 
OR, 0-36 vs. >36 months2 (95% CI, 0.35-11.40)0.5 (95% CI, 0.09-2.86) 0.043

[i] aCalculated using Fisher's exact test. OR, odds ratio; RB, retinoblastoma; RB1, RB transcriptional corepressor 1.

Discussion

Integrated RB1 genetic testing for the management of RB may reduce RB-associated mortality and treatment costs. However, the test sensitivity rate should be ≥90% to ensure a negative result would indicate a low risk of hereditary RB. In the current study, patients who were diagnosed with bilateral RB were used as a positive control group to evaluate the sensitivity of NGS in routine clinical practice. The detection rate of NGS was 100% for patients with bilateral RB, while all common RB1 mutation types were detected. Additionally, nine novel pathogenic mutations were identified. The present study also supported the potential use of age at diagnosis as a risk factor for inherited RB.

Recently, NGS is considered a highly sensitive and efficient approach for the detection of RB1 mutations due to increasing use of NGS in gene mutation analysis of RB; especially screening and identification of RB1 mutations with NGS substantially benefits the prepotency, early diagnosis and treatment of retinoblastoma (20-25). Furthermore, the sensitivity rate of NGS in the present study was similar with that reported to previous studies. For example, Li et al (51) detected germline RB1 mutations in 100% (19/19) of patients with bilateral RB, which was consistent with previously reported laboratory data. Additionally, Singh et al (52) demonstrated a detection rate of 100% (22/22) for bilateral cases, following validation of data using the TruSight Cancer Sequencing Panel (Illumina, Inc.), with a sensitivity rate of 98.2%, specificity of 100%, and reproducibility of 99.5%. Furthermore, Grotta et al (53) showed a determination rate of 96% (28/29) for patients with bilateral RB when combining NGS and aCGH. Herein, the detected RB1 variants had common characteristics with RB1 alterations commonly observed in patients with RB. For instance, mutations were not identified in the exons 26 and 27 of the RB1 gene and were scattered along the RB1 gene. Another study revealed that null mutations, including large rearrangements, nonsense, splicing and frameshift mutations were detected at high frequency, whereas missense mutations exhibited a low frequency (6). Overall, the aforementioned data indicated that NGS could be considered as an accurate method for detecting germline RB1 mutations.

The detection rate of germline RB1 mutations in Vietnamese patients with RB using SS-MLPA has been previously reported. Therefore, Kiet et al (26) and Nguyen et al (27) detected germline RB1 mutations in 84 (21/25) and 82% (9/11) of patients with bilateral RB, respectively. However, the detection rates were lower compared with those reported in other Asian studies applying the same approach. For example, Tomar et al (18) and He et al (54) revealed a detection rate of 94 (17/18) and 92% (36/39) in Singaporean and Chinese patients with bilateral RB, respectively. In addition, Rojanaporn et al (55) and Mohd Khalid et al (56) also reported a detection rate of germline RB1 mutations of 92 (25/27) and 100% (7/7) in Thai and Malaysian patients with bilateral RB, respectively.

In the present study nine novel RB1 mutations were identified in the cyclin fold-contained subunits of the pRB. The allele frequencies of these null mutations were <0.0001% in the public databases, and had not been recorded in the genetic polymorphism database consisting of 400 healthy Vietnamese individuals (47). The low frequency of these variations in the general population indicated that these mutations could be eliminated during evolution due to their disadvantages. Additionally, the CADD tool predicted that all these alterations, except one (EX12 DUP), were among the top 1% of deleterious variants in the human genome. CADD integrates multiple annotations, including genomic features, gene-annotation models, evolutionary and epigenetic features, and gene functional predictions to generate a single, quantitative scoring system. These scores are then used to rank the deleterious effect of a given variant (48,57). For example, a CADD score of 20 or 30 suggests that a variant is among the top 1% or 0.1% of deleterious variants in the human genome, respectively. Consequently, VarSome (44), a tool for implementing the ACMG standards, is used to classify variants as pathogenic ones when they meet very strong evidence of pathogenicity (PSV1). The PSV1 criterion assumes that certain null mutations, such as nonsense, frameshift, splice sites of +/-1 or 2, initiation codon and exon deletions, can lead to a complete absence of the gene product due to impaired transcription or nonsense-mediated decay of an altered transcript (35). The predicted null variants in a gene, whose loss of function is the known mechanism underlying the development of a particular disease, such as RB1 for RB, can be considered as the common cause of the disease. Devarajan et al (58) also applied the stringent criteria for defining pathogenic variants and reported no false-positive results during the detection of constitutional RB1 variants.

The two-hit hypothesis suggests that in patients with hereditary RB the tumors are formed at younger ages compared with sporadic cases (28). In 1998, Zajaczek et al (31) reported four patients with unilateral RB, who were diagnosed with germline RB1 mutations at the age of <19 months. This initial evidence supported the hypothesis that age at diagnosis could differentiate the hereditary from the sporadic form of unilateral RB. However, further studies with larger sample sizes did not reveal any association between age at diagnosis and germline RB1 status in patients with unilateral RB (32-34). In addition, Tomar et al (18) showed that Singaporean individuals diagnosed with RB at 0-36 months of age had a 53% risk of being RB1 carriers, whereas those >36 months had 8% risk of suffering from hereditary RB. The results of the present study were consistent with those of the previous one, suggesting that children diagnosed with RB at 0-36 months of age were more likely to be RB1 carriers compared with those aged >36 months. Nevertheless, similar analyses in patients with unilateral RB did not reveal significant difference in the risk of hereditary RB.

The present study revealed three clinical implications. Firstly, the results supported that the NGS method implemented at the Vinmec Hi-Tech Center was highly reliable in detecting germline RB1 variants. Secondly, this study could provide novel insights into the mechanisms underlying RB1 inactivation. Finally, the current findings suggested that age at diagnosis could be considered as a risk factor for hereditary RB.

Nevertheless, there are some limitations in the present retrospective study, including the small sample sizes and delay in diagnosis. Therefore, the results could not be generalizable to all patients with RB. The number of patients with bilateral RB, who underwent RB1 testing in each of the aforementioned studies was <30. Furthermore, the small sample size and different detection strategies used to identify germline RB1 mutations could result in variations in the detection rate between NGS and SS-MPLA. However, previous studies revealed that NGS-based methods could detect germline RB1 mutations in patients with RB, whose constitutional RB1 mutations could not be detected by either Sanger sequencing nor MPLA (59,60). In addition, the lack of statistical significance in the relative risk of patients with unilateral RB diagnosed at the age of 0-36 and those >36 months being carriers of RB1 could be also due to the small number of unilateral cases.

In conclusion, the results of the present study indicated that NGS could be considered a reliable method for screening for constitutional RB1 mutations, and age at diagnosis could be used to assess the risk of hereditary RB. Furthermore, the newly identified RB1 mutations could provide useful information for an in-depth understanding of the mechanisms underlying RB1 inactivation, and for the development of rapid assays for detecting RB1 mutations. Altogether, the current study suggested that NGS could be used for detecting germline RB1 mutations in routine clinical practice.

Acknowledgements

The authors would like to thank Professor Paolo Boffetta (Icahn School of Medicine, Mount Sinai School of Medicine, Tisch Cancer Institute, New York City, NY, USA) for the critical review of this manuscript.

Funding

No funding was received.

Availability of data and materials

The FASTQ data that support the findings of this study are available from the Beijing Genomics Institute (BGI) laboratory (Hong Kong) but restrictions apply to the availability of these data, which were used under license for the current study, and so are not publicly available. Data are however available from the authors upon reasonable request and with permission of the BGI laboratory.

Authors' contributions

CQH and HQD conceived and designed the study. BDN, LTP, DTN, CTMP and TLD recruited and referred patients to the study and helped with their follow-up. HQD, CQH, NTN, SAHN, CN, BDN, LTP, DTN, CTMP, TLD and MHT performed the experiments. HQD, CQH, NTN, SAHN, CN and MHT analyzed and interpreted the results. CQH and HQD wrote the manuscript. HQD and CQH are responsible for confirming the authenticity of the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Signed informed consents were obtained from parents/caregivers of all subjects, and the study was approved by the Vinmec's Institutional Review Board (Vinmec Healthcare System, Hanoi, Vietnam).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Canty CA: Retinoblastoma: An overview for advanced practice nurses. J Am Acad Nurse Pract. 21:149–155. 2009.PubMed/NCBI View Article : Google Scholar

2 

Aerts I, Lumbroso-Le Rouic L, Gauthier-Villars M, Brisse H, Doz F and Desjardis L: Retinoblastoma. Orphanet J Rare Dis. 1(31)2006.PubMed/NCBI View Article : Google Scholar

3 

Kim JY and Park Y: Treatment of retinoblastoma: The role of external beam radiotherapy. Yonsei Med J. 56:1478–1491. 2015.PubMed/NCBI View Article : Google Scholar

4 

Abramson DH: Retinoblastoma: Saving life with vision. Annu Rev Med. 65:171–184. 2014.PubMed/NCBI View Article : Google Scholar

5 

Soliman SE, Racher H, Zhang C, MacDonald H and Gallie BL: Genetics and molecular diagnostics in retinoblastoma-an update. Asia Pac J Ophthalmol (Phila). 6:197–207. 2017.PubMed/NCBI View Article : Google Scholar

6 

Valverde JR, Alonso J, Palacios I and Pestaňa A: RB1 gene mutation up-date, a meta-analysis based on 932 reported mutations available in a searchable database. BMC Genet. 6(53)2005.PubMed/NCBI View Article : Google Scholar

7 

Draper GJ, Sanders BM, Brownbill PA and Hawkins MH: Patterns of risk of hereditary retinoblastoma and applications to genetic counselling. Br J Cancer. 66:211–219. 1992.PubMed/NCBI View Article : Google Scholar

8 

Rushlow D, Piovesan B, Zhang K, Prigoda-Lee NL, Marchong MN, Clark RD and Gallie BL: Detection of mosaic RB1 mutations in families with retinoblastoma. Hum Mutat. 30:842–851. 2009.PubMed/NCBI View Article : Google Scholar

9 

Ali A, Kletke S, Gallie B and Lam WC: Retinoblastoma for pediatric ophthalmologists. Asia Pac J Ophthalmol (Phila). 7:160–168. 2018.PubMed/NCBI View Article : Google Scholar

10 

Canadian Retinoblastoma Society. National retinoblastoma strategy Canadian guidelines for care. Can J Ophthalmol. 44 (Suppl 2):S9–S47. 2009.PubMed/NCBI View Article : Google Scholar

11 

Dhar SU, Chintagumpala M, Noll C, Chévez-Barrios P, Paysse EA and Plon SE: Outcomes of integrating genetics in management of patients with retinoblastoma. Arch Ophthalmol. 129:1428–1434. 2011.PubMed/NCBI View Article : Google Scholar

12 

Joseph B, Shanmugam MP, Srinivasan MK and Kumaramanickavel G: Retinoblastoma: Genetic testing versus conventional clinical screening in India. Mol Diagn. 8:237–243. 2004.PubMed/NCBI View Article : Google Scholar

13 

Berry JL, Kim JW, Damato BE and Singh AD (eds): Clinical Ophthalmic Oncology: Retinoblastoma. Springer, p303, 2019.

14 

Ali MJ, Parsam VL, Honavar SG, Kannabiran C, Vemuganti GK and Reddy VA: RB1 gene mutations in retinoblastoma and its clinical correlation. Saudi J Ophthalmol. 24:119–123. 2010.PubMed/NCBI View Article : Google Scholar

15 

Lohmann DR: RB1 gene mutations in retinoblastoma. Hum Mutat. 14:283–288. 1999.PubMed/NCBI View Article : Google Scholar

16 

Ellison G, Donald E, McWalter G, Knight L, Fletcher L, Sherwood J, Cantarini M, Orr M and Speake G: A comparison of ARMS and DNA sequencing for mutation analysis in clinical biopsy samples. J Exp Clin Cancer Res. 29(132)2010.PubMed/NCBI View Article : Google Scholar

17 

Richter S, Vandezande K, Chen N, Zhang K, Sutherland J, Anderson J, Han L, Panton R, Branco P and Gallie B: Sensitive and efficient detection of RB1 gene mutations enhances care for families with retinoblastoma. Am J Hum Genet. 72:253–269. 2003.PubMed/NCBI View Article : Google Scholar

18 

Tomar S, Sethi R, Sundar G, Quah TC, Quah BL and Lai PS: Mutation spectrum of RB1 mutations in retinoblastoma cases from Singapore with implications for genetic management and counselling. PLoS One. 6(e0178776)2017.PubMed/NCBI View Article : Google Scholar

19 

Thirumalairaj K, Abraham A, Devarajan B, Gaikwad N, Kim U, Muthukkaruppan V and Vanniarajan A: A stepwise strategy for rapid and cost-effective RB1 screening in Indian retinoblastoma patients. J Hum Genet. 60:547–552. 2015.PubMed/NCBI View Article : Google Scholar

20 

Mehyar M, Mosallam M, Tbakhi A, Saab A, Sultan I, Deebajah R, Jaradat I, AlJabari R, Mohammad M, AlNawaiseh I, et al: Impact of RB1 gene mutation type in retinoblastoma patients on clinical presentation and management outcome. Hematol Oncol Stem Cell Ther. 13:152–159. 2020.PubMed/NCBI View Article : Google Scholar

21 

Zhang Z, Xiao YS, Shen R, Jiang HC, Tan L, Li RQ, Yang XH, Gu HY, He WJ and Ma J: Next generation sequencing of RB1 gene for the molecular diagnosis of ethnic minority with retinoblastoma in Yunnan. BMC Med Genet. 21(230)2020.PubMed/NCBI View Article : Google Scholar

22 

Xu L, Shen L, Polski A, Prabakar RK, Shah R, Jubran R, Kim JW, Biegel J, Kuhn P, Cobrinik D, et al: Simultaneous identification of clinically relevant RB1 mutations and copy number alterations in aqueous humor of retinoblastoma eyes. Ophthalmic Genet. 41:526–532. 2020.PubMed/NCBI View Article : Google Scholar

23 

Chai P, Luo Y, Yu J, Li Y, Yang J, Zhuang A, Fan J, Han M and Jia R: Clinical characteristics and germline mutation spectrum of RB1 in Chinese patients with retinoblastoma: A dual-center study of 145 patients. Exp Eye Res. 205(108456)2021.PubMed/NCBI View Article : Google Scholar

24 

Zou Y, Li J, Hua P, Liang T, Ji X and Zhao P: Spectrum of germline mutations in RB1 in Chinese patients with retinoblastoma: Application of targeted next-generation sequencing. Mol Vis. 27:1–16. 2021.PubMed/NCBI

25 

Francis JH, Richards AL, Mandelker DL, Berger MF, Walsh MF, Dunkel IJ, Donoghue MTA and Abramson DH: Molecular changes in retinoblastoma beyond RB1: Findings from next-generation sequencing. Cancers (Basel). 13(149)2021.PubMed/NCBI View Article : Google Scholar

26 

Kiet NC, Khuong LT, Minh DD, Nguyen The Vinh, Quan NHM, Xinh PT, Trang NNC, Luan NT, Khai NM and Vu HA: Spectrum of mutations in the RB1 gene in Vietnamese patients with retinoblastoma. Mol Vis. 25:215–221. 2019.PubMed/NCBI

27 

Nguyen HH, Nguyen HTT, Vu NP, Le QT, Pham CM, Huyen TT, Manh H, Pham HLB, Nguyen TD, Le HTT and Van Nong H: Mutational screening of germline RB1 gene in Vietnamese patients with retinoblastoma reveals three novel mutations. Mol Vis. 24:231–238. 2018.PubMed/NCBI

28 

Knudson AG Jr: Mutation and cancer: Statistical study of retinoblastoma. Proc Natl Acad Sci USA. 68:820–823. 1971.PubMed/NCBI View Article : Google Scholar

29 

Dimaras H, Kimani K, Dimba EA, Gronsdahl P, White A, Chan HS and Ballie BL: Retinoblastoma. Lancet. 379:1436–1446. 2012.PubMed/NCBI View Article : Google Scholar

30 

Lohmann DL and Gallie BL: Retinoblastoma: Revisiting the model prototype of inherited cancer. Am J Med Genet C Semin Med Genet. 129C:23–28. 2004.PubMed/NCBI View Article : Google Scholar

31 

Zajaczek S, Jakubowska A, Kurzawski G, Krzystolik Z and Lubiński J: Age at diagnosis to discriminate those patients for whom constitutional DNA sequencing is appropriate in sporadic unilateral retinoblastoma. Eur J Cancer. 34:1919–1921. 1998.PubMed/NCBI View Article : Google Scholar

32 

Schüler A, Weber S, Neuhäuser M, Jurklies C, Lehnert T, Heimann H, Rudolph G, Jöckel KH, Bornfeld N and Lohmann DR: Age at diagnosis of isolated unilateral retinoblastoma does not distinguish patients with and without a constitutional RB1 gene mutation but is influenced by a parent-of-origin effect. Eur J Cancer. 41:735–740. 2005.PubMed/NCBI View Article : Google Scholar

33 

Lohmann DR, Gerick M, Brandt B, Oelschläger U, Lorenz B, Passarge E and Horsthemke B: Constitutional RB1-gene mutations in patients with isolated unilateral retinoblastoma. Am J Hum Genet. 61:282–294. 1997.PubMed/NCBI View Article : Google Scholar

34 

Berry JL, Lewis L, Zolfaghari E, Green S, Le BHA, Lee TC, Murphree AL, Kim JW and Jubran R: Lack of correlation between age at diagnosis and RB1 mutations for unilateral retinoblastoma: The importance of genetic testing. Ophthalmic Genet. 39:407–409. 2018.PubMed/NCBI View Article : Google Scholar

35 

Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Brody WW, Hegde M, Lyon E, Spector E, et al: Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American college of medical genetics and genomics and the association for molecular pathology. Genet Med. 17:405–424. 2015.PubMed/NCBI View Article : Google Scholar

36 

den Dunnen JT, Dalgleish R, Maglott DR, Hart RK, Greenblatt MS, McGowan-Jordan J, Roux AF, Smith T, Antonarakis SE and Taschner PE: HGVS recommendations for the description of sequence variants: 2016 update. Hum Mutat. 37:564–569. 2016.PubMed/NCBI View Article : Google Scholar

37 

Landrum MJ, Lee JM, Riley GR, Jang W, Rubinstein WS, Church DM and Maglott DR: ClinVar: Public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 42:D980–D985. 2014.PubMed/NCBI View Article : Google Scholar

38 

Béroud C, Collod-Béroud G, Boileau C, Soussi T and Junien C: UMD (Universal mutation database): A generic software to build and analyze locus-specific databases. Hum Mutat. 15:86–94. 2000.PubMed/NCBI View Article : Google Scholar

39 

Fokkema IF, Taschner PE, Schaafsma GC, Celli J, Laros JF and den Dunnen JT: LOVD v.2.0: The next generation in gene variant databases. Hum Mutat. 32:557–563. 2011.PubMed/NCBI View Article : Google Scholar

40 

ARUP Scientific Resource for Research and Education. Available from: https://arup.utah.edu/.

41 

Vaser R, Adusumalli S, Leng SN, Sikic M and Ng PC: SIFT missense predictions for genomes. Nat Protoc. 11:1–9. 2016.PubMed/NCBI View Article : Google Scholar

42 

Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P, Kondrashov AS and Sunyaev SR: A method and server for predicting damaging missense mutations. Nat Methods. 7:248–249. 2010.PubMed/NCBI View Article : Google Scholar

43 

Schwarz JM, Cooper DN, Schuelke M and Seelow D: MutationTaster2: Mutation prediction for the deep-sequencing age. Nat Methods. 11:361–362. 2014.PubMed/NCBI View Article : Google Scholar

44 

Kopanos C, Tsiolkas V, Kouris A, Chapple CE, Aguilera MA, Meyer R and Massouras A: VarSome: The human genomic variant search engine. Bioinformatics. 35:1978–1980. 2019.PubMed/NCBI View Article : Google Scholar

45 

Li Q and Wang K: InterVar: Clinical interpretation of genetic variants by the 2015 ACMG-AMP guidelines. Am J Hum Genet. 100:267–280. 2017.PubMed/NCBI View Article : Google Scholar

46 

McLaren W, Gil L, Hunt SE, Riat HS, Ritchie GS, Thormann A, Flicek P and Cunningham F: The ensembl variant effect predictor. Genome Biol. 17(122)2016.PubMed/NCBI View Article : Google Scholar

47 

Le VS, Tran KT, Bui HTP, Le HTT, Nguyen CD, Do DH, Ly HTT, Pham LTD, Dao LTM and Nguyen LT: A Vietnamese human genetic variation database. Hum Mutat. 40:1664–1675. 2019.PubMed/NCBI View Article : Google Scholar

48 

Rentzsch P, Witten D, Cooper GM, Shendure J and Kircher M: CADD: Predicting the deleteriousness of variants throughout the human genome. Nucleic Acids Res. 47:D886–D894. 2019.PubMed/NCBI View Article : Google Scholar

49 

Social Science Statistics. Available from: https://www.socscistatistics.com/.

50 

Schoonjans F, Zalata A, Depuydt CE and Comhaire FH: MedCalc: A new computer program for medical statistics. Comput Methods Programs Biomed. 48:257–262. 1995.PubMed/NCBI View Article : Google Scholar

51 

Li WL, Buckley J, Sanchez-Lara PA, Maglinte DT, Viduetsky L, Tatarinova TV, Aparicio JG, Kim JW, Au M, Ostrow D, et al: A rapid and sensitive next-generation sequencing method to detect RB1 mutations improves care for retinoblastoma patients and their families. J Mol Diagn. 18:480–493. 2016.PubMed/NCBI View Article : Google Scholar

52 

Singh J, Mishra A, Pandian AJ, Mallipatna AC, Khetan V, Sripriya S, Kapoor S, Agarwal S, Sankaran S, Katragadda S, et al: Next-generation sequencing-based method shows increased mutation detection sensitivity in an Indian retinoblastoma cohort. Mol Vis. 22:1036–1047. 2016.PubMed/NCBI

53 

Grotta S, D'Elia G, Scavelli R, Genovese S, Surace C, Sirleto P, Cozza R, Romanzo A, De lois MA, Valente P, et al: Advantages of a next generation sequencing targeted approach for the molecular diagnosis of retinoblastoma. BMC Cancer. 15(841)2015.PubMed/NCBI View Article : Google Scholar

54 

He MY, An Y, Gao YJ, Qian XW, Li G and Qian J: Screening of RB1 gene mutations in Chinese patients with retinoblastoma and preliminary exploration of genotype-phenotype correlations. Mol Vis. 20:545–552. 2014.PubMed/NCBI

55 

Rojanaporn D, Boontawon T, Chareonsirisuthigul T, Thanapanpanich O, Attaseth T, Saengwimol D, Anurathapan U, Sujirakul T, Kaewkhaw R and Hongeng S: Spectrum of germline RB1 mutations and clinical manifestations in retinoblastoma patients from Thailand. Mol Vis. 24:778–788. 2018.PubMed/NCBI

56 

Mohd Khalid MK, Yakob Y, Md Yasin R, Teik K, Siew CG, Rahmat J, Ramasamy S and Alagaratnam J: Spectrum of germ-line RB1 gene mutations in Malaysian patients with retinoblastoma. Mol Vis. 21:1185–1190. 2015.PubMed/NCBI

57 

Kircher M, Witten DM, Jain P, O'Roak BJ, Cooper GM and Shendure J: A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet. 46:310–315. 2014.PubMed/NCBI View Article : Google Scholar

58 

Devarajan B, Prakash L, Kannan TR, Abraham AA, Kim U, Muthukkaruppan V and Vanniarajan A: Targeted next generation sequencing of RB1 gene for the molecular diagnosis of Retinoblastoma. BMC Cancer. 15(320)2015.PubMed/NCBI View Article : Google Scholar

59 

Chen Z, Moran K, Richards-Yutz J, Toorens E, Gerhart D, Ganguly T, Shields CL and Ganguly A: Enhanced sensitivity for detection of low-level germline mosaic RB1 mutations in sporadic retinoblastoma cases using deep semiconductor sequencing. Hum Mutat. 35:384–391. 2014.PubMed/NCBI View Article : Google Scholar

60 

Rodríguez-Martín C, Robledo C, Gómez-Mariano G, Monzón S, Sastre A, Abelairas J, Sábado C, Martín-Begué N, Ferreres JC, Fernández-Teijeiro A, et al: Frequency of low-level and high-level mosaicism in sporadic retinoblastoma: Genotype-phenotype relationships. J Hum Genet. 65:165–174. 2020.PubMed/NCBI View Article : Google Scholar

61 

Parsam VL, Kannabiran C, Honavar S, Vemuganti GK and Ali MJ: A comprehensive, sensitive and economical approach for the detection of mutations in the RB1 gene in retinoblastoma. J Genet. 88:517–527. 2009.PubMed/NCBI View Article : Google Scholar

62 

Parma D, Ferrer M, Luce L, Giliberto F and Szijan I: RB1 gene mutations in Argentine retinoblastoma patients. Implications for genetic counseling. PLoS One. 12(e0189736)2017.PubMed/NCBI View Article : Google Scholar

63 

Lan X, Xu W, Tang X, Ye H, Song X, Lin L, Ren X, Yu G, Zhang H and Wu S: Spectrum of RB1 germline mutations and clinical features in unrelated Chinese patients with retinoblastoma. Front Genet. 11(142)2020.PubMed/NCBI View Article : Google Scholar

64 

Berge EO, Knappskog S, Lillehaug JR and Lønning PE: Alterations of the retinoblastoma gene in metastatic breast cancer. Clin Exp Metastasis. 28:319–326. 2011.PubMed/NCBI View Article : Google Scholar

65 

Kato MV, Ishizaki K, Toguchida J, Kaneko A, Takayama J, Tanooka H, Kato T, Shimuzu T and Sasaki MS: Mutations in the retinoblastoma gene and their expression in somatic and tumor cells of patients with hereditary retinoblastoma. Hum Mutat. 3:44–51. 1994.PubMed/NCBI View Article : Google Scholar

66 

Fabian ID, Reddy A and Sagoo MS: Classification and staging of retinoblastoma. Community Eye Health. 31:11–13. 2018.PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 15 Issue 3

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hoang CQ, Duong H, Nguyen NT, Nguyen SA, Nguyen C, Nguyen BD, Phung LT, Nguyen DT, Pham CT, Le Doan T, Le Doan T, et al: Clinical evaluation of RB1 genetic testing reveals novel mutations in Vietnamese patients with retinoblastoma. Mol Clin Oncol 15: 182, 2021
APA
Hoang, C.Q., Duong, H., Nguyen, N.T., Nguyen, S.A., Nguyen, C., Nguyen, B.D. ... Tran, M.H. (2021). Clinical evaluation of RB1 genetic testing reveals novel mutations in Vietnamese patients with retinoblastoma. Molecular and Clinical Oncology, 15, 182. https://doi.org/10.3892/mco.2021.2344
MLA
Hoang, C. Q., Duong, H., Nguyen, N. T., Nguyen, S. A., Nguyen, C., Nguyen, B. D., Phung, L. T., Nguyen, D. T., Pham, C. T., Le Doan, T., Tran, M. H."Clinical evaluation of RB1 genetic testing reveals novel mutations in Vietnamese patients with retinoblastoma". Molecular and Clinical Oncology 15.3 (2021): 182.
Chicago
Hoang, C. Q., Duong, H., Nguyen, N. T., Nguyen, S. A., Nguyen, C., Nguyen, B. D., Phung, L. T., Nguyen, D. T., Pham, C. T., Le Doan, T., Tran, M. H."Clinical evaluation of RB1 genetic testing reveals novel mutations in Vietnamese patients with retinoblastoma". Molecular and Clinical Oncology 15, no. 3 (2021): 182. https://doi.org/10.3892/mco.2021.2344