RT Journal Article SR Electronic T1 Integrative Analysis of Germline Rare Variants in Clear and Non-Clear Cell Renal Cell Carcinoma JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2023.01.18.23284664 DO 10.1101/2023.01.18.23284664 A1 Seunghun Han A1 Sabrina Y. Camp A1 Hoyin Chu A1 Ryan Collins A1 Riaz Gillani A1 Jihye Park A1 Ziad Bakouny A1 Cora A. Ricker A1 Brendan Reardon A1 Nicholas Moore A1 Eric Kofman A1 Chris Labaki A1 David Braun A1 Toni K. Choueiri A1 Saud H. AlDubayan A1 Eliezer M. Van Allen YR 2023 UL http://medrxiv.org/content/early/2023/01/19/2023.01.18.23284664.abstract AB IMPORTANCE RCC encompasses a set of histologically distinct cancers with a high estimated genetic heritability, of which only a portion is currently explained. Previous rare germline variant studies in RCC have usually pooled clear and non-clear cell RCCs and have not adequately accounted for population stratification that may significantly impact the interpretation and discovery of certain candidate risk genes.OBJECTIVE To evaluate the enrichment of germline PVs in established cancer-predisposing genes (CPGs) in clear cell and non-clear cell RCC patients compared to cancer-free controls using approaches that account for population stratification and to identify unconventional types of germline RCC risk variants that confer an increased risk of developing RCC.DESIGN, SETTING, AND PARTICIPANTS In 1,436 unselected RCC patients with sufficient data quality, we systematically identified rare germline PVs, cryptic splice variants, and copy number variants (CNVs). From this unselected cohort, 1,356 patients were ancestry-matched with 16,512 cancer-free controls, and gene-level enrichment of rare germline PVs were assessed in 143 CPGs, followed by an investigation of somatic events in matching tumor samples.MAIN OUTCOMES AND MEASURES Gene-level burden of rare germline PVs, identification of secondary somatic events accompanying the germline PVs, and characterization of less-explored types of rare germline PVs in RCC patients.RESULTS In clear cell RCC (n = 976 patients), patients exhibited significantly higher prevalence of PVs in VHL compared to controls (OR: 39.1, 95% CI: 7.01-218.07, p-value:4.95e-05, q-value:0.00584). In non-clear cell RCC (n = 380 patients), patients carried enriched burden of PVs in FH (OR: 77.9, 95% CI: 18.68-324.97, p-value:1.55e-08, q-value: 1.83e-06) and MET (OR: 1.98e11, 95% CI: 0-inf, p-value: 2.07e-05, q-value: 3.50e-07). In a CHEK2-focused analysis with European cases and controls, clear cell RCC patients (n=906 European patients) harbored nominal enrichment of the previously reported low-penetrance CHEK2 variants, p.Ile157Thr (OR:1.84, 95% CI: 1.00-3.36, p-value:0.049) and p.Ser428Phe (OR:5.20, 95% CI: 1.00-26.40, p-value:0.045) while non-clear cell RCC patients (n=295 European patients) exhibited nominal enrichment of CHEK2 LOF germline PVs (OR: 3.51, 95% CI: 1.10-11.10, p-value: 0.033). RCC patients with germline PVs in FH, MET, and VHL exhibited significantly earlier age of cancer onset compared to patients without any germline PVs in CPGs (Mean: 46.0 vs 60.2 years old, Tukey adjusted p-value < 0.0001), and more than half had secondary somatic events affecting the same gene (n=10/15, 66.7%, 95% CI: 38.7-87.0%). Conversely, patients with rare germline PVs in CHEK2 exhibited a similar age of disease onset to patients without any identified germline PVs in CPGs (Mean: 60.1 vs 60.2 years old, Tukey adjusted p-value: 0.99), and only 30.4% of the patients carried secondary somatic events in CHEK2 (n=7/23, 95% CI: 14.1-53.0%). Finally, rare pathogenic germline cryptic splice variants underexplored in RCC were identified in SDHA and TSC1, and rare pathogenic germline CNVs were found in 18 patients, including CNVs in FH, SDHA, and VHL.CONCLUSIONS AND RELEVANCE This systematic analysis supports the existing link between several RCC risk genes and elevated RCC risk manifesting in earlier age of RCC onset. Our analysis calls for caution when assessing the role of germline PVs in CHEK2 due to the burden of founder variants with varying population frequency in different ancestry groups. It also broadens the definition of the RCC germline landscape of pathogenicity to incorporate previously understudied types of germline variants, such as cryptic splice variants and CNVs.Question Can we improve the assessment of germline genetic risk determinants for clear cell and non-clear cell renal cell carcinoma (RCC) with approaches that are aware to population-stratification and RCC histological subtypes?Findings In this systematic case-control study of 1,356 RCC patients and 16,512 ancestry-matched cancer-free controls strictly controlling for population stratification, clear-cell RCC patients exhibited a significantly higher prevalence of rare germline pathogenic variants (PVs) in VHL, and non-clear cell RCC patients carried significantly more rare germline PVs in FH and MET. European clear-cell RCC patients harbored a nominally significant enrichment of two low-penetrance CHEK2 variants (p.Ser428Phe and p.Ile157Thr) while European non-clear RCC patients carried a nominally significant enrichment of rare germline loss-of-function (LOF) variants in CHEK2. Subsequent somatic analyses identified secondary somatic events in genes significantly enriched for germline PVs (VHL, FH, MET), and these variant carriers presented with earlier age of disease onset, but CHEK2 germline variant carriers harbored relatively fewer somatic events in CHEK2 and did not present with earlier age of onset. Finally, we identified 6 RCC patients with rare germline cryptic splice and copy number variants that impacted known kidney cancer risk genes, increasing the diagnostic yield of pathogenic variants in RCC risk genes from 2.1% to 2.5%.Meaning Clear and non-clear RCCs have distinct germline pathogenic variant enrichment patterns and somatic variants. Accurate risk assessment of CHEK2 in RCC requires careful adjustment for population stratification. In addition, previously underappreciated forms of germline variants may explain a portion of the missing heritability in RCC.Competing Interest StatementE.M.V.A. holds consulting roles with Tango Therapeutics, Genome Medical, Genomic Life, Enara Bio, Manifold Bio, Monte Rosa, Novartis Institute for Biomedical Research, Riva Therapeutics and Serinus Bio; he receives research support from Novartis, Bristol-Myers Squibb and Sanofi; he has equity in Tango Therapeutics, Genome Medical, Genomic Life, Syapse, Enara Bio, Manifold Bio, Microsoft, Monte Rosa, Riva Therapeutics and Serinus Bio; he has filed institutional patents on chromatin mutations, immunotherapy response, and methods for clinical interpretation. T.K.C. reports institutional and personal, paid and/or unpaid support for research, advisory boards, consultancy, and honoraria from: Alkermes, AstraZeneca, Aravive, Aveo, Bayer, Bristol Myers-Squibb, Calithera, Circle Pharma, Eisai, EMD Serono, Exelixis, GlaxoSmithKline, IQVA, Infinity, Ipsen, Jansen, Kanaph, Lilly, Merck, Nikang, Nuscan, Novartis, Pfizer, Roche, Sanofi/Aventis, Surface Oncology, Takeda, Tempest, Up-To-Date, CME events (Peerview, OncLive, MJH and others), outside the submitted work. Institutional patents filed on molecular alterations and immunotherapy response/toxicity, and ctDNA. Equity: Tempest, Pionyr, Osel, Precede Bio. CureResponse. Committees: NCCN, GU Steering Committee, ASCO/ESMO, ACCRU, KidneyCan. Medical writing and editorial assistance support may have been funded by Communications companies in part. No speakers bureau. Mentored several non-US citizens on research projects with potential funding (in part) from non-US sources/Foreign Components. The institution (Dana-Farber Cancer Institute) may have received additional independent funding of drug companies or/and royalties potentially involved in research around the subject matter. T. K. C is also supported in part by the Dana-Farber/Harvard Cancer Center Kidney SPORE (2P50CA101942-16) and Program 5P30CA006516-56, the Kohlberg Chair at Harvard Medical School and the Trust Family, Michael Brigham, Pan Mass Challenge, Hinda and Arthur Marcus Fund and Loker Pinard Funds for Kidney Cancer Research at DFCI. D.A.B. reports nonfinancial support from Bristol Myers Squibb, honoraria from LM Education/Exchange Services, advisory board fees from Exelixis and AVEO, personal fees from Charles River Associates, Schlesinger Associates, Imprint Science, Insight Strategy, Trinity Group, Cancer Expert Now, Adnovate Strategies, MDedge, CancerNetwork, Catenion, OncLive, Cello Health BioConsulting, PWW Consulting, Haymarket Medical Network, Aptitude Health, ASCO Post/Harborside, Targeted Oncology, AbbVie, and research support from Exelixis and AstraZeneca, outside of the submitted work. R.G. has equity in Google, Microsoft, Amazon, Apple, Moderna, Pfizer, and Vertex Pharmaceuticals. B.R. has filed institutional patents on methods for clinical interpretation. Z.B. receives research support from Bristol Myers Squibb and imCORE/Genentech. He also reports honoraria from UpToDate. C.L. receives research funding from imCORE/Genentech outside the submitted work. The other authors declare no competing interests.Funding StatementThis work was supported by The National Institutes of Health R37CA222574 (E.M.V), R01CA227388 (E.M.V), R50CA265182 (J.P.), Mark Foundation Emerging Leader Award, the Department of Defense Physician Research Award (W81XWH-21-1-0084, PC200150) (S.H.A), and the Department of Defense Idea Development Award - Early-Career Investigator (KC210042/W81XWH-22-1-0455) (S.H.A), Alexs Lemonade Stand Foundation Young Investigator Grant (R.G.), and the Wong Family Award in Translational Oncology (R.G.). The funding organizations were not responsible for the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:IRB#20-293 of Dana Farber Cancer Institute gave ethical approval for this workI confirm that all necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived, and that any patient/participant/sample identifiers included were not known to anyone (e.g., hospital staff, patients or participants themselves) outside the research group so cannot be used to identify individuals.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesAll computation tools and packages in this study are publicly available. The docker image containing all GATK tools is available at (https://hub.docker.com/r/broadinstitute/gatk/). The docker image containing the germline variant detection tool, DeepVariant can be found at (https://hub.docker.com/r/google/deepvariant). Tools and detailed usage for SpliceAI (https://github.com/Illumina/SpliceAI) and GATK-gCNV (https://github.com/theisaacwong/talkowski/tree/master/gCNV) can be found on the respective GitHub pages. All raw sequencing data for TCGA studies can be accessed with controlled access on the GDC data portal (https://portal.gdc.cancer.gov/) with approval. All raw sequencing data for the ICGC study can be accessed with controlled access on the ICGC data portal (https://dcc.icgc.org/) with approval. All raw sequencing data for CHECKMATE clinical studies and Genentech study can be downloaded from European Genome Phenome Archive (Dataset ID: EGAD00001001023) with approval. All raw sequencing data for cancer free control samples can be accessed on dbGAP Autism Sequencing Consortium (ASC) (dbGAP:phs000298.v4.p3), Framingham Cohort (dbGAP:phs000007.v32.p1), MESA Cohort (dbGAP: phs000209.v13.p3), NHLBI GO ESP: Lung Cohorts Exome Sequencing Project (dbGAP: phs000291.v2.p1). In house exome data for controls is available upon request.