Next Article in Journal
Sex Difference in Disease-Related Adverse Events Post-Diagnosis of Lung Cancer Brain Metastases in Medicare Individuals ≥ 66 Years of Age
Previous Article in Journal
Using mHealth Technology to Evaluate Daily Symptom Burden among Adult Survivors of Childhood Cancer: A Feasibility Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Prevalence of Germline Pathogenic Variants in Renal Cancer Predisposition Genes in a Population-Based Study of Renal Cell Carcinoma

1
Cancer Epidemiology Division, Cancer Council Victoria, East Melbourne, VIC 3002, Australia
2
Burnet Institute, Melbourne, VIC 3004, Australia
3
Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3010, Australia
4
Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
5
School of Public Health, The University of Queensland, Herston, QLD 4006, Australia
6
Melbourne Bioinformatics, The University of Melbourne, Melbourne, VIC 3010, Australia
7
Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, VIC 3010, Australia
8
Victoria Comprehensive Cancer Centre, The University of Melbourne Centre for Cancer Research, Melbourne, VIC 3010, Australia
9
Department of Surgery, Royal Melbourne Hospital, Melbourne Medical School, The University of Melbourne, Melbourne, VIC 3010, Australia
10
Department of Medicine, Melbourne Medical School, The University of Melbourne, Melbourne, VIC 3010, Australia
11
Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
12
Hereditary Cancer Centre, Nelune Comprehensive Cancer Centre, Prince of Wales Hospital, Sydney, NSW 2031, Australia
13
Division of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
14
Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Melbourne, VIC 3010, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cancers 2024, 16(17), 2985; https://doi.org/10.3390/cancers16172985
Submission received: 18 June 2024 / Revised: 21 August 2024 / Accepted: 22 August 2024 / Published: 27 August 2024

Abstract

:

Simple Summary

Studies of genetic predisposition to renal cell carcinoma (RCC) have been highly variable in design and have reported similarly highly variable rates of cases identified as carrying pathogenic variants in cancer susceptibility genes (4–26%). The aim of our study was to conduct a population-based study of genetic predisposition to RCC. We conducted a 21-gene panel sequencing study and identified that 18/1029 participants (1.7%) carried a pathogenic or likely pathogenic (PLP) variant. Genes with PLP variants included BAP1, FH, FLCN, MITF, MSH6, SDHB, TSC1, and VHL. This study provides further evidence that family history alone may not be sufficient for identifying all individuals who are at increased genetic risk of renal cell carcinoma, and further research is urgently needed to understand how to target genetic testing to identify those at high genetic risk of kidney cancer.

Abstract

Renal cell carcinoma (RCC) has been associated with germline pathogenic or likely pathogenic (PLP) variants in recognised cancer susceptibility genes. Studies of RCC using gene panel sequencing have been highly variable in terms of study design, genes included, and reported prevalence of PLP variant carriers (4–26%). Studies that restricted their analysis to established RCC predisposition genes identified variants in 1–6% of cases. This work assessed the prevalence of clinically actionable PLP variants in renal cancer predisposition genes in an Australian population-based sample of RCC cases. Germline DNA from 1029 individuals diagnosed with RCC who were recruited through the Victoria and Queensland cancer registries were screened using a custom amplicon-based panel of 21 genes. Mean age at cancer diagnosis was 60 ± 10 years, and two-thirds (690, 67%) of the participants were men. Eighteen participants (1.7%) were found to carry a PLP variant. Genes with PLP variants included BAP1, FH, FLCN, MITF, MSH6, SDHB, TSC1, and VHL. Most carriers of PLP variants did not report a family history of the disease. Further exploration of the clinical utility of gene panel susceptibility testing for all RCCs is warranted.

1. Introduction

Renal cell carcinoma (RCC) accounts for 2% of cancer diagnoses worldwide and is associated with a mortality rate of 25% within 5 years [1]. Identification of those at greatest risk and early diagnosis through screening are important for improved survival because approximately 13% of cases are diagnosed with metastatic disease [2].
RCC aetiology is partially explained by hereditary factors, with 6% of cases reporting a family history of kidney cancer [3]. Several heritable renal cancer syndromes are clinically described (including von Hippel–Lindau Syndrome, Birt–Hogg–Dubé Syndrome, Hereditary Leiomyomatosis and RCC, Hereditary Papillary RCC, Cowden Syndrome, and Tuberous Sclerosis), all of which are associated with pathogenic or likely pathogenic (PLP) variants in genes that display an autosomal dominant inheritance.
Genetic epidemiology studies of RCC have been heterogenous in terms of design (hospital-based or commercial genetics service; retrospective or cross-sectional), the number of genes investigated (n = 19 to 808), and the prevalence of PLP variants observed (4–26%) [4,5,6,7,8,9,10,11]. These studies investigated cancer susceptibility genes that are not clinically actionable for RCC, including CHEK2 and BRCA2. In studies that restricted their analysis to established RCC predisposition genes (BAP1, EPCAM, FH, FLCN, MET, MITF, MLH1, MSH2, MSH6, PMS2, PTEN, SDHA, SDHB, SDHC, SDHD, TP53, TSC1, TSC2, and VHL), PLP variants were identified in 1–6% of individuals tested [4,5,6,8,11,12]. However, these estimates may not reflect population estimates because the studies oversampled individuals with a suspected genetic aetiology for their RCC.
In Australia, there are 21 genes that are considered clinically actionable in the context of RCC by most clinical genetics services. These include genes associated with RCC syndromic diseases (CDC73, FH, FLCN, MET, PTEN, SDHB, SDHC, SDHD, TSC1, TSC2, and VHL), Lynch Syndrome (EPCAM, MLH1, MSH2, MSH6, PMS2), Li-Fraumeni Syndrome (TP53), and cancer predisposition genes (BAP1, MITF, POLD1, and POLE).
This study investigated the prevalence of PLP variants in these 21 genes in a population-based sample of RCC cases. The study provides estimates that are applicable to all individuals with RCC, irrespective of family history.

2. Materials and Methods

2.1. Study Participants

Participants were from the population-based family case-control component [13] of the Consortium for the Investigation of Renal Malignancies (CONFIRM) study. Participant characteristics are described in Table 1. Non-carriers were defined as individuals without a clinically actionable PLP variant in a gene included in our panel design.
Participants were recruited from the Victorian and Queensland Cancer Registries. The study included Australian residents diagnosed between 2009 and 2015 with a histologically confirmed RCC (ICD-10 C64) at age 18–74 years. Eligible participants had tumours at least 2cm in diameter, no previous diagnosis of RCC, and were able to complete the study questionnaires in English.
Once an individual was identified as eligible by the cancer registry, the treating doctor was informed by the registry of the intention to invite the patient to participate in the study. A letter of invitation, participant information sheet, and study consent form were sent to the prospective participant, unless the treating doctor declined involvement of their patient.

2.2. Data and Biological Sample Collection

Participants were asked to complete epidemiological questionnaires that included a section on family history of kidney cancer. Eighty-five percent of sequenced participants were recruited within 12 months of diagnosis.
All participants were invited to provide a blood sample (or saliva if preferred) from which DNA was extracted for sequencing.

2.3. Mutation Screening

Amplicon-based massively parallel sequencing of the coding regions and proximal intron–exon junctions of 21 genes (BAP1, NM_004656.4; CDC73, NM_024529.5; EPCAM, NM_002354.3; FH, NM_000143.4; FLCN, NM_144997.7; MET, NM_001127500.3; MITF, NM_000248.4; MLH1, NM_000249.4; MSH2, NM_000251.3; MSH6, NM_000179.3; PMS2, NM_000535.7; POLD1, NM_002691.4; POLE, NM_006231.4; PTEN, NM_000314.8; SDHB, NM_003000.3; SDHC, NM_003001.5; SDHD, NM_003002.4; TP53, NM_000546.6; TSC1, NM_000368.5; TSC2, NM_000548.5, and VHL, NM_000551.4) was performed on germline-derived DNA from 1071 individuals using the Hi-Plex protocol (Nguyen-Dumont et al., 2015 Breast Cancer Res Treat). Massively parallel sequencing (150 bp paired-end) was performed on a MiSeq (Illumina, San Diego, CA, USA). Mapping to the human reference build hg19 and variant calling were performed as described in Nguyen-Dumont et al., 2015 [14]. A DNA sample was considered to have been successfully sequenced when at least 80% of the targeted regions were covered at a read depth of at least 50X.

2.4. Variant Prioritisation and Classification

Variant annotation and filtering were performed using the variant analysis software Varseq (version 2.3.0, Golden Helix, Bozeman, MT, USA), consistent with those applied in exome/genome studies. Variants were selected for further investigation if they were considered genuine (observed at a total read depth ≥ 50X and a variant allele fraction of ≥0.2) and rare (minor allele frequency < 0.01, or absent, in gnomAD exomes and genomes). Variants were considered PLP if they were classified in ClinVar (accessed 2 March 2023) as PLP with a review status of at least “2 stars”. When variants were classified as PLP in ClinVar with a review status of “1 star”, or were absent from ClinVar but predicted to be loss-of-function, then variant classification was performed according to the American College of Medical Genetics (ACMG) criteria [15], including updated recommendations by the Association for Clinical Genomic Science [16], the Cancer Variant Interpretation Group UK [17], and the ClinGen Sequence Variant Interpretation Working Group [18,19,20].
PLP variants were manually assessed using Integrative Genomics Viewer (IGV) for errors in alignment, and sequencing or mapping artefacts were excluded. All PLP variants were validated by Sanger sequencing at the Australian Genome Research Facility or the Garvan Institute for Medical Research (Sydney, Australia).
The number of rare variants of uncertain significance (VUSs) in this sample of participants was calculated by counting the number of variants that were neither classified as (i) PLP according to the criteria described above, nor (ii) benign or likely benign according to ClinVar (at least “1 star”). These variants were not inspected in IGV.

2.5. Statistical Analyses

All p-values were calculated using Fisher’s exact test; a p-value <0.05 was considered significant.

3. Results

Of the 3113 individuals diagnosed with RCC who were invited to participate in CONFIRM, 1424 provided consent. Of those, 1071 had germline DNA available for screening, and 1029 were successfully sequenced with an average of 97% of the targeted regions covered at a minimum of 50X. Mean age (±standard deviation) at cancer diagnosis was 60 ± 10 years, and two-thirds (690, 67%) were men (Table 1).
Eighteen cases (1.7%) were found to carry a PLP variant (Table 2). The mean age at diagnosis of RCC for these cases was 58 ± 10 years and eleven (61%) were men (Table 1). The gene with the most PLP variant carriers identified was MITF with five individuals carrying NM_000248.4:c.952G>A; p.Glu318Lys (Table 2). Other genes with PLP variants included FLCN, FH, MSH6, VHL, BAP1, SDHB, and TSC1. No PLP variants were identified in CDC73, EPCAM, MET, MLH1, MSH2, PMS2, POLD1, POLE, PTEN, SDHC, SDHD, TP53, or TSC2. Three hundred and forty-six VUSs were identified in 289 participants (28%).
Family history data were available for 897 (87%) of the 1029 participants (15 PLP carriers, 882 non-carriers), of whom 769 reported if any of their first-degree relatives had kidney cancer (12 PLP carriers, 757 non-carriers), and 752 reported if any of their second-degree relatives had kidney cancer (12 PLP carriers, 740 non-carriers). Of PLP variant carriers, only 1 of 12 (8%) had a first-degree relative with kidney cancer compared with 54 of 757 (7%) non-carriers (p = 0.59). Also, 1 of 12 (8%) PLP carriers had a second-degree relative with kidney cancer compared with 34 of 740 (5%) non-carriers (p = 0.44) (Table 1). These two PLP carriers who reported a family history (carriers 4 and 7, Table 2) both had a PLP variant in FLCN. In total, 86 of 882 (10%) non-carriers had a family history of RCC.
Histopathological presentations for PLP variant carriers included seven (39%) with clear cell RCC, four (22%) with chromophobe RCC, three (17%) with papillary RCC, and four (22%) with RCC (not otherwise specified) (Table 2). Presentations for non-PLP variant carriers included 606 (60%) with clear cell RCC, 96 (9%) with chromophobe RCC, 107 (11%) with papillary RCC, 186 (18%) with RCC (not otherwise specified), and 15 (1%) with other subtypes of RCC.

4. Discussion

This study provides an important population-based assessment of the prevalence of clinically actionable PLP germline variants in RCC in Australia. PLP variants were identified in 1.7% of participants, consistent with previous studies that identified 1–6% were carriers of PLP variants in an RCC predisposition gene [4,5,6,8,11,12]. Unlike most previous studies, this study did not oversample for individuals with a likely genetic aetiology [7,8,11,12], or recruit from clinical settings that limit analysis and relevance to a subset of the population [4,6,9,10].
PLP variants were identified in 8 of 21 established RCC predisposition genes included in our panel design, with variants most commonly observed in MITF. The PLP variant MITF p.Glu318Lys was observed in 0.5% of participants and has been previously reported to be associated with an increased risk of RCC [21]. In a retrospective study, Nguyen et al., 2017 reported this variant in 0.7% of 1235 individuals with kidney cancer, the majority of whom did not report a family history of kidney cancer [12]. Although there are no well-described clinical features of RCC associated with MITF p.Glu318Lys, the histopathology predominantly observed with this variant is either clear cell or papillary RCC [12].
Most individuals who were identified as carrying a PLP variant had no reported family history of RCC, indicating that they, and possibly their relatives, are unaware of their genetic predisposition to RCC. In Australia, genetic counselling and germline panel testing are recommended for individuals with a RCC diagnosis who have a suspected genetic aetiology based on their clinical presentation, including age of onset and family history of RCC. Genetic testing offered more broadly may therefore be useful to identify those at genetically increased risk of RCC, irrespective of family history. Furthermore, predictive testing for PLP variants may be offered to family members to enable them to engage in clinical care adapted to their hereditary risk, of which the family were hitherto unaware.
Conversely, most participants (98%) who reported a family history were not identified as carrying a PLP variant, so the possible hereditary cause of their RCC remains undetermined. It is possible these individuals carry a variant in a gene that is not yet demonstrated to be associated with RCC. Indeed, a recent report from Yngvadottir et al., 2022 provided some evidence for an association between RCC risk and PLP variants in CHEK2, which was not included in our panel design [22].
The prevalence of PLP variants in the 21 genes included in this study could be higher than reported here. The estimate of VUS in this study was high (n = 346) due to the difficulty in interpreting these variants; most were rare missense variants with currently insufficient evidence of pathogenicity for classification as PLP.
For instance, classification of a splice variant, BAP1 NM_004656.4:c.783+2T>C, was challenging under current ACMG guidelines. This variant is located in the donor splice site of intron 9. The in silico tool, SpliceAI, predicted a cryptic donor site 2bp downstream, which, if engaged, creates a transcript with four additional nucleotides resulting in a frameshift event. This prediction was used in the assessment of the variant for PVS1 and to support the LP classification of the variant. However, donor site GT>GC variants can be problematic because an estimated 15–18% are partially functional and give rise to normal transcripts [23]. It is also unclear to what extent adjacent splice site motifs could influence splicing in this region. In the future, RNA sequencing may be useful to assess the potential spliceogenic impact of this variant.
Two variants classified as VUSs in this study, according to current ACMG criteria, were classified as PLP in ClinVar with a review status of 1 star. These were in FH (NM_000143.4:c.1196G>A; p.Ser399Asn) and SDHD (NM_003002.4:c.278_280dupATT; p.Tyr93dup). Neither ClinVar entry provided sufficient details about affected participants to be able to curate these variants as PLP according to current ACMG standards.
The significant strength of this study is its population-based design; however, because individuals were recruited from cancer registries rather than clinical settings, we do not have detailed clinical information about features that could possibly accompany the genetic syndromes and assist variant classification, especially for variants identified in FLCN, FH, and TSC1.
Some types of genomic variation cannot be detected by the gene panel design and technology applied, including deep intronic, UTR, structural, or copy-number variants. This limitation is true for many technologies applied in previous studies, and thus our understanding of these types of genomic variants in these genes is likely to be incomplete. However, large deletions (10–13 kb) have been identified in VHL in cohorts of RCC cases [22] that, if present, remain undetected in our study. It is also likely that further RCC susceptibility genes exist that have not yet been identified/validated and thus were not included in this study (e.g., CHEK2) [22].
Furthermore, not all individuals with RCC reported in the registries were eligible, nor consented, to participate in this study. Some individuals may have declined participation because they had worse health outcomes that were associated with carrying a PLP variant.

5. Conclusions

This study indicates that the proportion of individuals with RCC who would benefit from short-read gene panel testing is less than 2%. This study provides further evidence that (i) family history alone may not be sufficient for identifying all individuals who are at increased genetic risk of RCC and (ii) further research is urgently needed to identify and characterise additional genetic risk factors for RCC.

Author Contributions

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

Funding

Victorian Cancer Agency (grant number EO109_36, MCRF21029); The National Health and Medical Research Council (GNT1074383, GNT1025879, GNT2011329, GNT2017325); Cancer Council Victoria (GNT1066612).

Institutional Review Board Statement

The study was approved by the Human Research Ethics Committee (HREC) of Cancer Council Victoria (HREC No. 0912) and Metro South Queensland (HREC/11/QPAH/271). The study was performed in accordance with the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this report can be requested via PEDIGREE. https://www.cancervic.org.au/research/epidemiology/pedigree.

Acknowledgments

We thank the participants of the CONFIRM study. We thank Helen Tsimiklis for biological material management and Fleur Hammet for supporting the gene panel testing for this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Padala, S.A.; Barsouk, A.; Thandra, K.C.; Saginala, K.; Mohammed, A.; Vakiti, A.; Rawla, P.; Barsouk, A. Epidemiology of Renal Cell Carcinoma. World J. Oncol. 2020, 11, 79–87. [Google Scholar] [CrossRef] [PubMed]
  2. Monda, S.M.; Lui, H.T.; Pratsinis, M.A.; Chandrasekar, T.; Evans, C.P.; Dall’Era, M.A. The Metastatic Risk of Renal Cell Carcinoma by Primary Tumor Size and Subtype. Eur. Urol. Open Sci. 2023, 52, 137–144. [Google Scholar] [CrossRef] [PubMed]
  3. Clague, J.; Lin, J.; Cassidy, A.; Matin, S.; Tannir, N.M.; Tamboli, P.; Wood, C.G.; Wu, X. Family history and risk of renal cell carcinoma: Results from a case-control study and systematic meta-analysis. Cancer Epidemiol. Biomarkers Prev. 2009, 18, 801–807. [Google Scholar] [CrossRef]
  4. Carlo, M.I.; Mukherjee, S.; Mandelker, D.; Vijai, J.; Kemel, Y.; Zhang, L.; Knezevic, A.; Patil, S.; Ceyhan-Birsoy, O.; Huang, K.-C.; et al. Prevalence of Germline Mutations in Cancer Susceptibility Genes in Patients With Advanced Renal Cell Carcinoma. JAMA Oncol. 2018, 4, 1228–1235. [Google Scholar] [CrossRef]
  5. Han, S.H.; Camp, S.Y.; Chu, H.; Collins, R.; Gillani, R.; Park, J.; Bakouny, Z.; Ricker, C.A.; Reardon, B.; Moore, N.; et al. Integrative Analysis of Germline Rare Variants in Clear and Non–Clear Cell Renal Cell Carcinoma. Eur. Urol. Open Sci. 2024, 62, 107–122. [Google Scholar] [CrossRef]
  6. Sekine, Y.; Iwasaki, Y.; Aoi, T.; Endo, M.; Hirata, M.; Kamatani, Y.; Matsuda, K.; Sugano, K.; Yoshida, T.; Murakami, Y.; et al. Different risk genes contribute to clear cell and non-clear cell renal cell carcinoma in 1532 Japanese patients and 5996 controls. Hum. Mol. Genet. 2022, 31, 1962–1969. [Google Scholar] [CrossRef] [PubMed]
  7. Lincoln, S.E.; Nussbaum, R.L.; Kurian, A.W.; Nielsen, S.M.; Das, K.; Michalski, S.; Yang, S.; Ngo, N.; Blanco, A.; Esplin, E.D. Yield and Utility of Germline Testing Following Tumor Sequencing in Patients With Cancer. JAMA Netw. Open 2020, 3, e2019452. [Google Scholar] [CrossRef]
  8. Hartman, T.R.; Demidova, E.V.; Lesh, R.W.; Hoang, L.; Richardson, M.; Forman, A.; Kessler, L.; Speare, V.; Golemis, E.A.; Hall, M.J.; et al. Prevalence of pathogenic variants in DNA damage response and repair genes in patients undergoing cancer risk assessment and reporting a personal history of early-onset renal cancer. Sci. Rep. 2020, 10, 13518. [Google Scholar] [CrossRef]
  9. Smith, P.S.; West, H.; Whitworth, J.; Castle, B.; Sansbury, F.H.; Warren, A.Y.; Woodward, E.R.; Tischkowitz, M.; Maher, E.R. Pathogenic germline variants in patients with features of hereditary renal cell carcinoma: Evidence for further locus heterogeneity. Genes Chromosom. Cancer 2021, 60, 5–16. [Google Scholar] [CrossRef]
  10. Meng, H.; Jiang, X.; Cui, J.; Yin, G.; Shi, B.; Liu, Q.; Xuan, H.; Wang, Y. Genomic Analysis Reveals Novel Specific Metastatic Mutations in Chinese Clear Cell Renal Cell Carcinoma. BioMed Res. Int. 2020, 2020, 2495157. [Google Scholar] [CrossRef]
  11. Kong, W.; Yang, T.; Wen, X.; Mu, Z.; Zhao, C.; Han, S.; Tian, J.; Zhang, X.; Zhou, T.; Zhang, Y.; et al. Germline Mutation Landscape and Associated Clinical Characteristics in Chinese Patients With Renal Cell Carcinoma. Front. Oncol. 2021, 11, 737547. [Google Scholar] [CrossRef]
  12. Nguyen, K.A.; Syed, J.S.; Espenschied, C.R.; LaDuca, H.; Bhagat, A.M.; Suarez-Sarmiento, A.; O’Rourke, T.K.; Brierley, K.L.; Hofstatter, E.W.; Shuch, B. Advances in the diagnosis of hereditary kidney cancer: Initial results of a multigene panel test. Cancer 2017, 123, 4363–4371. [Google Scholar] [CrossRef]
  13. Bruinsma, F.; Jordan, S.; Bassett, J.; Severi, G.; MacInnis, R.; Walsh, J.; Aitken, T.; Jenkins, M.; Carroll, R.; Jefford, M.; et al. Analgesic use and the risk of renal cell carcinoma–Findings from the Consortium for the Investigation of Renal Malignancies (CONFIRM) study. Cancer Epidemiol. 2021, 75, 102036. [Google Scholar] [CrossRef]
  14. Nguyen-Dumont, T.; Hammet, F.; Mahmoodi, M.; Tsimiklis, H.; Teo, Z.L.; Li, R.; Pope, B.J.; Terry, M.B.; Buys, S.S.; Daly, M.; et al. Mutation screening of PALB2 in clinically ascertained families from the Breast Cancer Family Registry. Breast Cancer Res. Treat. 2015, 149, 547–554. [Google Scholar] [CrossRef] [PubMed]
  15. Richards, S.; Aziz, N.; Bale, S.; Bick, D.; Das, S.; Gastier-Foster, J.; Grody, W.W.; 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. 2015, 17, 405–424. [Google Scholar] [CrossRef] [PubMed]
  16. Ellard, S.; Baple, E.L.; Callaway, A.; Berry, I.; Forrester, N.; Turnbull, C.; Owens, M.; Eccles, D.M.; Abbs, S.; Scott, R.; et al. ACGS Best Practice Guidelines for Variant Classification in Rare Disease 2020. Available online: https://www.acgs.uk.com/media/11631/uk-practice-guidelines-for-variant-classification-v4-01-2020.pdf (accessed on 2 November 2023).
  17. Garrett, A.; Callaway, A.; Durkie, M.; Cubuk, C.; Alikian, M.; Burghel, G.J.; Robinson, R.; Izatt, L.; Talukdar, S.; Side, L.; et al. Cancer Variant Interpretation Group UK (CanVIG-UK): An exemplar national subspecialty multidisciplinary network. J. Med. Genet. 2020, 57, 829–834. [Google Scholar] [CrossRef]
  18. Brnich, S.E.; Abou Tayoun, A.N.; Couch, F.J.; Cutting, G.R.; Greenblatt, M.S.; Heinen, C.D.; Kanavy, D.M.; Luo, X.; McNulty, S.M.; Starita, L.M.; et al. Recommendations for application of the functional evidence PS3/BS3 criterion using the ACMG/AMP sequence variant interpretation framework. Genome Med. 2019, 12, 3. [Google Scholar] [CrossRef] [PubMed]
  19. Ghosh, R.; Harrison, S.M.; Rehm, H.L.; Plon, S.E.; Biesecker, L.G.; ClinGen Sequence Variant Interpretation Working Group. Updated recommendation for the benign stand-alone ACMG/AMP criterion. Hum. Mutat. 2018, 39, 1525–1530. [Google Scholar] [CrossRef]
  20. Abou Tayoun, A.N.; Pesaran, T.; DiStefano, M.T.; Oza, A.; Rehm, H.L.; Biesecker, L.G.; Harrison, S.M.; ClinGen Sequence Variant Interpretation Working Group (ClinGen SVI). Recommendations for interpreting the loss of function PVS1 ACMG/AMP variant criterion. Hum. Mutat. 2018, 39, 1517–1524. [Google Scholar] [CrossRef]
  21. Bertolotto, C.; Lesueur, F.; Giuliano, S.; Strub, T.; De Lichy, M.; Bille, K.; Dessen, P.; D’Hayer, B.; Mohamdi, H.; Remenieras, A.; et al. A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 2011, 480, 94–98. [Google Scholar] [CrossRef]
  22. Yngvadottir, B.; Andreou, A.; Bassaganyas, L.; Larionov, A.; Cornish, A.J.; Chubb, D.; Saunders, C.N.; Smith, P.S.; Zhang, H.; Cole, Y.; et al. Frequency of pathogenic germline variants in cancer susceptibility genes in 1336 renal cell carcinoma cases. Hum. Mol. Genet. 2022, 31, 3001–3011. [Google Scholar] [CrossRef]
  23. Lin, J.; Tang, X.; Boulling, A.; Zou, W.; Masson, E.; Fichou, Y.; Raud, L.; Le Tertre, M.; Deng, S.; Berlivet, I.; et al. First estimate of the scale of canonical 5′ splice site GT>GC variants capable of generating wild-type transcripts. Hum. Mutat. 2019, 40, 1856–1873. [Google Scholar] [CrossRef]
Table 1. Characteristics of cases with renal cell carcinoma participating in CONFIRM.
Table 1. Characteristics of cases with renal cell carcinoma participating in CONFIRM.
PLP Variant CarriersNon-CarriersAll Tested Individuals (100%)
Participants: N (%)18 (1.7%)1011 (98.3%)1029
Sex: N (%)
Male11 (2%)679 (98%)690
Female7 (2%)332 (98%)339
Age at diagnosis: mean ± SD58 ± 1060 ± 1060 ± 10
Histology: N (%)
Clear cell7 (1%)606 (99%)613
Chromophobe4 (4%)96 (96%)100
Papillary3 (3%)107 (97%)110
Not otherwise specified4 (2%)186 (98%)190
Other015 (100%)15
Unknown01 (100%)1
Family history: N (%) p-value
First-degree relative with RCC p = 0.59
Yes1 (2%)54 (98%)
No11 (2%)703 (98%)
Missing6 (2%)254 (98%)
Second-degree relative with RCC p = 0.44
Yes1 (3%)34 (97%)
No11 (2%)706 (98%)
Missing6 (2%)271 (98%)
Table 2. Characteristics of carriers of pathogenic or likely pathogenic variants in CONFIRM.
Table 2. Characteristics of carriers of pathogenic or likely pathogenic variants in CONFIRM.
CarrierGeneHGVS c. bHGVS p. bSexAge of Diagnosis
(Years)
Affected
First-Degree
Relative
Affected
Second-Degree
Relative
Histology
1BAP1NM_004656.4:c.783+2T>C Female54UnknownUnknownclear cell
2FHNM_000143.4:c.698G>Ap.Arg233HisMale54NoNoNOS
3FHNM_000143.4:c.575C>Tp.Pro192LeuMale56UnknownUnknownclear cell
4 aFLCNNM_144997.7:c.1432+1G>A Male66YesUnknownpapillary
5 aFLCNNM_144997.7:c.1432+1G>A Male47UnknownUnknownchromophobe
6FLCNNM_144997.7:c.1318_1334dupp.Leu449Glnfs*25Female62NoNoNOS
7FLCNNM_144997.7:c.469_471delTTCp.Phe157delFemale62NoYeschromophobe
8MITFNM_000248.4:c.952G>Ap.Glu318LysMale63UnknownNopapillary
9MITFNM_000248.4:c.952G>Ap.Glu318LysMale74NoNoclear cell
10MITFNM_000248.4:c.952G>Ap.Glu318LysMale60UnknownNoclear cell
11MITFNM_000248.4:c.952G>Ap.Glu318LysMale72NoNoclear cell
12MITFNM_000248.4:c.952G>Ap.Glu318LysFemale73NoUnknownNOS
13MSH6NM_000179.3:c.3469G>Ap.Gly1157SerMale56UnknownNochromophobe
14MSH6NM_000179.3:c.2057G>Ap.Gly686AspFemale45NoNopapillary
15SDHBNM_003000.3:c.505C>Tp.Gln169TerMale47NoUnknownNOS
16TSC1NM_000368.5:c.589delTp.Cys197Alafs*13Female58NoNochromophobe
17VHLNM_000551.4:c.548C>Ap.Ser183TerFemale67NoNoclear cell
18VHLNM_000551.4:c.556G>Tp.Glu186TerMale35NoNoclear cell
a Variant carriers 4 and 5 are first-degree relatives (parent–child). b Variant nomenclature according to the Human Genome Variation Society (HGVS), HGVS.c for coding DNA, and HGVS.p for protein variants.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bruinsma, F.; Harraka, P.; Jordan, S.; Park, D.J.; Pope, B.; Steen, J.; Milne, R.L.; Giles, G.G.; Winship, I.; Tucker, K.M.; et al. Prevalence of Germline Pathogenic Variants in Renal Cancer Predisposition Genes in a Population-Based Study of Renal Cell Carcinoma. Cancers 2024, 16, 2985. https://doi.org/10.3390/cancers16172985

AMA Style

Bruinsma F, Harraka P, Jordan S, Park DJ, Pope B, Steen J, Milne RL, Giles GG, Winship I, Tucker KM, et al. Prevalence of Germline Pathogenic Variants in Renal Cancer Predisposition Genes in a Population-Based Study of Renal Cell Carcinoma. Cancers. 2024; 16(17):2985. https://doi.org/10.3390/cancers16172985

Chicago/Turabian Style

Bruinsma, Fiona, Philip Harraka, Susan Jordan, Daniel J. Park, Bernard Pope, Jason Steen, Roger L. Milne, Graham G. Giles, Ingrid Winship, Katherine M. Tucker, and et al. 2024. "Prevalence of Germline Pathogenic Variants in Renal Cancer Predisposition Genes in a Population-Based Study of Renal Cell Carcinoma" Cancers 16, no. 17: 2985. https://doi.org/10.3390/cancers16172985

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop