Abstract
Primary renal manifestations of diffuse large B-cell lymphoma (PR-DLBCL) represent an exceptionally rare variant of the most common type of non-Hodgkin lymphoma (NHL). Insights into PR-DLBCL pathogenesis have been limited to small case series and methodologically limited approaches. The mechanisms driving lymphomagenesis within an organ lacking an intrinsic lymphatic niche and its proclivity for dissemination to immune-privileged sites, including testes and central nervous system, remain poorly understood. To decode the genetic and transcriptional framework of PR-DLBCL, we utilized whole exome sequencing, array-based somatic copy number alterations analysis, and RNA sequencing. Hereby we characterize the most extensive cohort of PR-DLBCL published, comprising 34 samples from 30 patients. Despite significant mutational heterogeneity with a broad distribution among molecular clusters, we observed a strong unifying enrichment in deleterious MHC class I and II aberrations and loss of CDKN2A at a frequency similar to primary large B-cell lymphoma of immune privileged sites (IP-LBCL) alongside significant transcriptional deregulation of interferon signaling and MYC targets in MHC class I-deficient cases.
Our integrative assessment of PR-DLBCL biology expands the molecular understanding of this rare variant including similarities with IP-LBCL as an intriguing explanation for its clinical behavior and tropism. Our observations may inform future risk-adapted therapeutic approaches.
Introduction
Diffuse large B-cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma (NHL) and accounts for 30 – 40% of newly diagnosed lymphomas 1. Extranodal manifestations in both the adrenal glands and the kidneys constitute long-standing risk factors for developing simultaneous or secondary CNS manifestations 2. Secondary renal involvement is recurrently observed in advanced-stage non-Hodgkin lymphoma (NHL), whereas primary renal (PR-) NHL manifestations are notably less frequent, leading to skepticism regarding the existence of primary renal lymphoma for an extended period 2–6. Population-based datasets suggest an age-adjusted incidence of 0.035/100,000 with an increasing tendency, a median age of approximately 70 years at diagnosis, a male predominance, and mostly unilateral tumors. Among PR-NHL, DLBCL appears to be the most common histology followed by marginal zone (MZL) and follicular lymphoma (FL) 7, 8. Clinical differentiation of PR-NHL from other renal malignancies including renal cell carcinoma poses a long-standing challenge, which led to a substantial number of PR-NHL patients undergoing curatively intended resection 9. In recent years, the introduction of FDG-PET-CTs into routine clinical practice has enabled a more reliable characterization, particularly in patients with more aggressive PR-NHL10.
Insufficient characterization of pathogenetic mechanisms in indolent PR-DLBCL derives from limited data, primarily comprised of small case series and a lack of in-depth molecular characterization of patients and samples 11. Molecular drivers of lymphomas originating from organs lacking an intrinsic lymphatic niche, such as the kidney, remain widely elusive apart from individual case studies 12. To elucidate the genetic and transcriptional landscape and drivers of PR-DLBCL, we conducted a comprehensive investigation employing whole exome sequencing (WES), array-based analysis of somatic copy number alterations (SCNA), and RNA sequencing (RNA-seq) to unravel both molecular properties of this rare entity as well as the cellular composition of its tumor microenvironment (TME).
Materials and methods
Case selection and clinicopathological assessment
For this retrospective analysis, we reviewed our institutional archive for cases of histologically confirmed primary renal DLBCL between January 2001 and December 2021. Only such cases without concurrent non-renal lymphoma manifestations at diagnosis were included in the study, which led to a large subset of samples obtained by complete renal resection under the suspicion of non-metastatic renal cell carcinoma (22/34 samples; 65%). The study was approved by the ethics committee of the University of Lübeck (reference-no 18-356) and conducted per the declaration of Helsinki. Patients provided written informed consent regarding routine diagnostic and academic assessment, including genomic studies. Histopathological work-up was performed as described and diagnosis was confirmed following the 5th edition WHO classification of hematopoietic and lymphoid tumors and the ICC criteria and yielded 34 cases of PR-DLBCL for which sufficient formalin-fixed paraffin-embedded (FFPE) tissue samples were available for subsequent molecular analysis 1, 13, 14.
Delineation of the molecular composition of the study cohort
Extraction of nucleic acids, WES and RNA-seq alongside detection of Somatic Copy Number Alterations employing the OncoScan CNV array (ThermoFisher was performed as described, and Raw fastq files have been added to the European Genome-phenome archive (EGA) under the previous accession number EGA50000000386 13, 15. OncoScan Array data has been deposited in Gene Expression Omnibus (GEO) under accession number GSE270422.
Whole exome data processing and variant calling
The raw sequencing data in FASTQ format was processed using the NFORE workflow (NEXTFLOW v23.10.1) SAREK (v3.3.2) for variant calling against the GRCh38 reference genome 16, 17. Initially, reads were trimmed for adapter sequences and quality using FASTP (v0.23.4), followed by alignment with BWA MEM2 (v2.2.1) 18, 19. Subsequent steps included mate-pair information correction, removal of PCR duplicates, and base quality recalibration utilizing PICARD TOOLS, GATK (v4.4.0.0), and dbSNP v146 20, 21. Finally, variant calling was executed in tumor-only mode using MUTECT2 with GNOMAD (r2.1.1) as the reference for known germline variants 22, 23. Variants were left aligned (GATK LEFTALIGNANDTRIMVARIANTS) and only variants with a PASS filter flag were kept for variant annotation using VARIANT EFFECT PREDICTOR (VEP v111, GRCh38; adding CADD v1.6, dbNSFP v4.1a, and GNOMAD r3.0 as additional annotations) 24–26. Next, annotations were converted into MAF format using VCF2MAF (V1.6.21) (DOI:10.5281/ZENODO.593251); coverage was extracted directly from the vcf INFO field. Potential FFPE artifacts in the variant data were identified using two approaches. First, strand orientation biases (mutations just found on one strand, F1R2 or F2R1) were detected by SOBDETECTOR (v1.0.4) and potential artifacts were removed 27. Next, a classifier detecting the origin of mutation in formalin-fixation paraffin-embedding (FFPE) samples was applied (R-package EXCERNO v0.1.0) 28. Variants annotated with an FFPE-like signature were removed. Potential germline variants in the data were removed using GATKs 1000g Panel of Normals. The top 20 frequently mutated genes (FLAGS)29 were removed from further analysis and the remaining somatic variants were filtered as follows: minimum coverage of 50, minimum alternative allele coverage of 5, minimum variant allele frequency of 10%, and only variants with a frequency < 0.1% in 1000 genomes, GNOMAD, or ExAC were considered for subsequent downstream analysis; variants were required to have a COSMIC or dbSNP ID. High-impact variants (CADD score > 20) in candidate genes (see below) were filtered as such that minimum coverage of 10, minimum alternative coverage of 4, and minimum variant allele frequency of 10% was required. Candidate genes were defined as follows: (a) listed as tumor suppressor or oncogene according to Vogelstein et al. 30, (b) genes from the Lymphgen algorithm, (c) DLBCL genes described by Chapuy et al., (d) MHC genes according to Harmonizome (v3.0) 31–33. A brief graphical representation of the bioinformatics workflow for WES data processing is provided in Supplementary Figure S1.
Detection of Somatic Copy Number Alterations
Somatic copy number alterations were detected using OncoScan CNV assays (ThermoFisher). Raw data (CEL files) were processed using the EACON package (v0.3.6-2) with SEQUENZA as segmentation algorithm 34. L2R files in CBS format were used as input for GISTIC (v2.0.22) to identify regions significantly amplified or deleted across all samples (confidence level 0.90, focal length cutoff 0.5, q-value threshold 0.1) 34, 35.
For details on transcriptome data quantification, fusion detection, statistical analysis, and pseudonymization, please see Supplementary Materials and Methods.
Results
Clinicopathological Characteristics and Cell-of-Origin in PR-DLBCL
We collected 34 PR-DLBCL samples from 30 patients with sufficient FFPE tissue samples for in-depth molecular characterization (Figure 1A). The median age of the study group was 66.5 years, 13 patients were female and 17 were male. No iatrogenic immunosuppression, immunodeficiency, and history or current presence of other lymphomas were reported in any of the patients. In particular, only patients without suspicion of concurrent lymphoma manifestations were eligible for the study. This is mirrored in the exceptionally high frequency of cases that underwent surgical resection of the kidney (22/34 cases; 65%). Baseline clinicopathological data are summarized in Supplementary Table 1.
Structural variants in BCL2, BCL6, and MYC were assessed successfully in 19, 18, and 17 patients by FISH break-apart probes. In cases without sufficient tissue for FISH but with RNA-seq data, we screened for high-confidence BCL2/BCL6/MYC fusions (Figure 1 B). Here, we observed aberrations in five (26 %), two (11 %), and three (18%) cases respectively, indicative of a slightly elevated frequency of these aberrations in comparison to DLBCL (NOS) of other primary manifestations. In addition, we observed two cases with ETV6-Ig fusions, previously described in PCNSL another case of an ETV6:PAX5 fusion respectively, previously described to lead to similar ETV6 activation in B-cell precursor acute lymphoblastic leukemia 36, 37. Manually curated oncogenic fusions derived from RNA-seq data are summarized in Supplementary Table 2. By immunohistochemistry according to the Hans et al. algorithm in all samples with sufficient tissue available after DNA/RNA extraction, we observed the majority of cases to be of non-GCB type (18/30; 60%), whereas only a minor subset was of GCB type (11/30; 37%), and one case could not be classified due to insufficient immunoreactivity 38. Only one case was EBV-associated (>50% positive tumor cells by EBER-ISH). To further unravel the cell of origin, we performed RNA-seq on samples from 30 patients and applied the approach proposed by Wright et al. and found 15/30 cases (50%) to be of ABC-subtype. In comparison, 9/30 (30%) were GCB-DLBCL and six remaining cases (20%) remained unclassifiable 31, 39 (Figure 1 C).
The mutational landscape of PR-DLBCL
To elucidate the mutational landscape of PR-DLBCL and to screen for mutational drivers of the subgroup’s peculiar tropism, we performed WES on FFPE tissue samples from 34 patients (Figure 1 D).
Adhering to stringent variant filtering, outlined above, oncogenic mutations were identified in all PR-DLBCL cases. In total 3,178 presumably deleterious mutations, affecting 1,388 genes were observed. Additionally, 25,457 somatic copy number aberrations (SCNAs) were detected. SNVs and indels comprised 11% of these mutations, of which 2,673 were missense (84%), 333 nonsense (10%) and 70 indel mutations (2%). Mutations affecting splice sites represented 3% of estimated somatic mutations and one of the observed mutations was a non-stop mutation (Supplementary Table 3). At an intermediate-low median tumor mutational burden (TMB) of 2.45 mutations/Mb (range 0.18 – 5.76), we observed no cases of microsatellite instability as could be expected in aggressive B-cell lymphomas. Variants per variant class as well as variants per sample including information on variant subtypes are provided in Supplementary Figure S2.
The most commonly mutated genes included chromatin-remodeling genes CREBBP and KMT2D both in 56% of cases with > 20% nonsense mutations, followed by NOTCH signaling-associated genes SPEN and NOTCH2 (both 50%) with nonsense mutations in 47% and 18% respectively. Overall, we observed an enrichment in IL6/JAK/STAT3 signaling mutations including STAT3, PIM1, and MYD88. While STAT3 mutations were distributed throughout the entire gene, we observed mutational impairment of pre-described hotspots in MYD88 and PIM1 (all restricted to the kinase domain) (Figure 2). Other biological processes targeted by deleterious mutations in PR-DLBCL included the epigenetic regulation of gene expression (SF3B1, KMT2D, and EZH2), as well as B-cell receptor and RTK-RAS signaling (CARD11, PRDM1, and BRAF). The distribution of mutation onto the candidate driver genes referenced above is displayed in Figure 2. In a more global approach, we assessed the impairment of key biological processes through an enrichment analysis of mutations against previously described oncogenetic gene sets and against the IL6/JAK/STAT3 signaling pathway (Figure 3) 40. Hereby we uncovered a relatively wide distribution within a given set (Figure 3A). Intriguingly, the most commonly affected gene sets included NOTCH, RTK-RAS, and IL6/JAK/STAT signaling at 97%, 94%, and 91%, respectively (Figure 3 B, C). Beyond this, we observed mutational impairment of PI3K signaling in 79% of cases, suggesting a significant role in PR-DLBCL pathogenesis. Correlating information on cell of origin and mutational patterns, we observed a statistically significant enrichment in mutations affecting TNFAIP3 and PIM1 among non-GCB cases. BCL2 rearranged cases showed significantly more DNMT1 and SMARCA4 mutations (Supplementary Figure S3). We additionally screened for recurrent mutations in canonical tumor suppressor genes (TSGs) and found deleterious mutations in 33/34 (97%) cases. Among the most common targets, in addition to CREBBP and KMT2D, we observed mutations in TP53 in 15% of cases.
Genome-wide analysis of somatic copy-number variations in primary renal DLBCL
To unravel the genome-wide landscape of SCNA in PR-DLBCL, we analyzed 29 samples successfully on the Illumina OncoScan array and subsequently employed GISTIC (v2.0.22) to identify regions significantly amplified or deleted across all samples. Arguably the most striking observation was a broad arm-level deletion spanning a large region on chromosome 6 in 48% of evaluable cases (Figure 4A). Candidate genes, previously implicated in lymphoma pathogenesis located in this vast region included PRDM1, NFKBIE, HLA-A, HLA-B, and ARID1B. Beyond this aberration, the genome-wide CNV landscape of PR-DLBCL is dominated by CNVs affecting 3p12.1 and leading to copy number losses of VHL, SETD2, and others (17%), 9p21.3 encompassing the region encoding for TSG CDKN2A (38%), as well as the NOTCH1 region at 9q34.3 (28%). Focal amplifications were observed at 1q23.1, 3p12.3, 3q29, and 22q11.23. Candidate genes among significant deletions and amplifications are provided in Supplementary Table 4 and Figure 4B. To assess the characteristics of PR-DLBCL in contrast to transcriptionally defined subtypes of DLBCL (ABC/GCB) and primary CNS lymphoma (PCNSL) as well as primary testicular large B-cell lymphoma (PTLBL) together representing the subgroup of primary large B-cell lymphomas of immune-privileged sites. We conducted a comparative analysis drawing CNV data from two previously published large-scale genome-wide studies (Figure 4 C) 41, 42. Apart from a focal 3q29 amplification, none of the previously reported copy number gains were observed at a significant level in our cohort. At the same time and of particular interest, we observed strong similarities between PR-DLBCL, PCNSL, and PTLBL regarding deletions at 6p21.33, 6q21, and 6q23.3. Candidate gene deletions previously implicated in post-germinal differentiation and strong CNS tropism located at these loci include HLA-B and PRDM1. Moreover, we identified the above-mentioned CDKN2A deletions in PR-DLBCL at a frequency more similar to PCNSL/PTLBL than an all-comer DLBCL cohort. Additionally, we observed a significant impact of SCNAs on gene-expression profiles which is shown exemplarily for TSGs (Supplementary Figure S4).
Molecular clusters in primary renal DLBCL
Building on our observations from WES, CNV analysis, RNA-seq, alongside structural variants identified by FISH for BCL2, BCL6, and MYC, we performed an integrative analysis on our entire cohort of PR-DLBCL samples to achieve an allocation to the previously defined molecular clusters (A53, N1, BN2, EZB, and MCD) based on the probabilistic LymphGen 2.0 classifier 31. Molecular data for an integrative analysis of sufficient sensitivity and specificity was available in 30 cases. Unlike previous observations in PCNSL/PTLBL, only one case resembled MCD-DLBCL. The most common molecular subtypes were ST2 (eight cases, 27%) and EZB (five cases, 17%). Of note, no double- or triple-hit lymphomas including MYC+ EZB cases were observed. Only two cases were BN2-DLBCL (7%). While a significant overlap between signatures with compound-cluster allocation was observed in eight cases, six cases were found to exhibit a constellation of findings leading to a classification as “other” (Figure 1 D and Supplementary Table 5).
Immune-escape mechanisms in primary renal DLBCL as a driver of tropism
Following the characterization of the mutational profiles and molecular clusters that displayed the significant heterogeneity outlined above, we sought to understand the underlying mechanism behind the exceptional renal tropism in our present cohort. Building on our observation of a significant overlap between PR-DLBCL, PCNSL, and PTLBL, in particular regarding copy number losses affecting HLA-B as a hallmark gene of MHC Class I, we sought to assess the extent to which the clinical behavior of PR-DLBCL might be explained by immune escape strategies. Integrating all available datasets, we identified deleterious MHC Class I lesions in 72% of cases, including HLA-A and HLA-B deletions in 48% and deleterious B2M mutations in 21% of evaluable patients (Figure 5A, B). An additional fraction of 33% of patients exhibited deleterious aberrations within the MHC class II machinery and 60% of patients additionally exhibited molecular hits in immediate interaction partners of MHC class I or II required for adequate immune response including del 19p13.3 and mutations affecting CD70, CIITA, and CD58 (Figure 5A, C).
Having identified the large-scale 6p25.2-q27 deletion outlined above as a potential hallmark of aggressive B-cell lymphomas with renal tropism, we went on to assess the transcriptional profile of cases carrying said aberration and observed a significantly elevated expression of several candidate genes (Figure 5D and Supplementary Table 6) including CXCL10 as well as strong enrichment for interferon alpha and gamma response, MYC targets, JAK/STAT signaling, and others (Figure 5E and Supplementary Table 7). In particular, we found downstream NFkB activation via TNFalpha signaling. In addition, several metabolic processes including glycolysis, fatty acid metabolism, and oxidative phosphorylation appear to be particularly engaged compared to non-MHC-class I-deleted cases.
Subsequent analysis of our cohort deconvoluting cell states and their assembly into communities of co-association patterns in the form of lymphoma ecotypes (LE) employing the lymphoma EcoTyper revealed a relatively diverse composition of both the tumor as well as its microenvironment in PR-DLBCL 43. Despite the limited number of cases, we observed a trend towards enrichment in prognostically adverse ABC-DLBCL-associated lymphoma ecotypes (LE) 2 and 4 dominated by B-cells, plasma cells, and dendritic cells as well as NK cells, CD4 and CD8 positive T-cells alongside regulatory T-cells, respectively in del 6p25.2-q27 cases. Moreover, a trend towards enrichment of the T follicular helper cells dominated LE5 was observed in non-del 6p25.2-q27 cases 43. The distribution of ecotypes on cases is depicted in Figure 6 (Supplementary Table 8).
Discussion
Primary renal DLBCL poses a rare phenomenon associated with adverse clinical features and outcomes, including a so far unexplained CNS tropism leading to significant mortality, especially in relapsed and/or refractory cases 6. This study was initiated to unravel the molecular underpinnings of primary renal disease, to allow for an understanding as to why and how the disease originates in an organ without a genuine lymphatic niche, and to explain the clinically observed tendency of early CNS progression. To this end, we conducted an integrative analysis employing WES, RNA-seq and array-based SCNV analysis on the largest cohort of PR-DLBCL studied to date.
Hereby, we made the following noteworthy observations. First, we provide a detailed molecular landscape of PR-DLBCL and observe a strong degree of heterogeneity in terms of molecular clusters according to the LymphGen algorithm 31. Contrary to our initial expectation, we observed a relatively low frequency of MYD88 mutations. Therefore only one case that was allocated to the characteristic MCD cluster, which is otherwise recurrently observed in both primary and secondary CNS lymphoma as well as testicular lymphoma. Tropism for immune-privileged sites appears to be driven by immune escape strategies, which may be augmented by activating mutations in the B-cell receptor signaling cascade 31, 32, 44, 45. Beyond this, we saw a high frequency of SPEN mutations previously described to be associated with early progression and inferior outcome in an all-comer DLBCL cohort 46. Significant deregulation of TSGs was further validated via an integrative analysis of SCNAs and transcriptional abundance. Among the genes most commonly affected by mutations within our cohort, potentially targetable vulnerabilities included recurrent mutations in genes like EZH2 for which clinically active inhibitors have been described previously, including combinations with BCL2 inhibitors in patients with co-occurrence of EZH2 and BCL2 aberrations, which we observed in our cohort as well. At the same time, recurrent CARD11 mutations, downstream of Bruton’s tyrosine kinase (BTK) may render therapeutic approaches, employing BTK inhibitors inefficient in a clinically meaningful subset of patients47–49. Given the peculiar tropism of PR-DLBCL, the abundance of STAT3 mutations appears striking. In addition to the known cooperative interplay between STAT3 and the NFkB signaling cascade in post-germinal DLBCL, this may further be explained as a selection advantage throughout clonal evolution. In keeping with previous observations that linked STAT3 mutations and its elevated activity to the modulation of the abundance and function of regulatory T cells in the TME of solid cancers, this suggests a therapeutical perspective involving STAT3 inhibition which might be able to target the TME and its permissive impact on the immunological niche in PR-DLBCL 50, 51. An additional candidate driver for early progression and CNS dissemination is the relatively high rate of MYC aberrations in more than 17% of evaluable cases 52.
Second, we describe a profile of SCNA and mutations leading to significant impairment of MHC class I/II families and its immediately associated genes in approx. 95% of cases in the cohort, which significantly exceeds observations in all-comer DLBCL cohorts 32, 53. From these observations, we deduce a strong overlap between PR-DLBCL and large B-cell lymphomas of immune-privileged sites (IP-LBCL) regarding SCNAs. At the same time, PR-DLBCL apparently lacks the characteristic mutational profile resulting in tonic B-cell receptor signaling usually associated with this newly described entity 1, 54. It is tempting to speculate on a theory of a first and second hit in the central nervous dissemination of PR-DLBCL acquiring the typical driver mutations in a second step, following the initial presence of strong immune-escape mechanisms 55. To investigate this in future studies, paired analyses of renal and CNS tumors will be required. Deleterious aberrations in MHC class I components and related genes constitute well-documented phenomena in DLBCL. However, the frequency at which this is observed in the present study, extending to highly recurrent alterations of MHC class II genes poses a significant enrichment 32. Moreover, this aligns with previous observations in primary CNS lymphoma and testicular lymphoma regarding deletions of the HLA and CDKN2A loci and extends well beyond previous reports in ABC-DLBCL by both immunohistochemistry and gene expression profiling 41. Further in keeping with pathogenetically similar trajectories between PR-DLBCL and other IP-LBCL, we observed 10% of cases harboring ETV6 rearrangements previously reported as recurrent, oncogenic structural variants and drivers of a post-germinal phenotype in PCNSL 36.
Third, we describe a characteristic transcriptional profile in cases harboring MHC class I SCNAs as a newly described hallmark of primary renal DLBCL. It is predominantly characterized by a strong enrichment for interferon alpha and gamma responses, MYC targets, and JAK/STAT signaling and downstream NFkB activation via TNF-alpha signaling. All of these mechanisms are known drivers, especially of ABC-DLBCL. TNF signaling was recently shown to aberrantly remodel the fibroblastic reticular cell (FRC) network and contribute to a tolerogenic TME hindering therapeutic efficacy of immune-based therapies 51,56. Further, several metabolic processes including glycolysis, fatty acid metabolism, and oxidative phosphorylation appear particularly engaged. This could be particularly interesting in future targeted therapeutical approaches exploiting this dependence, as recently described57. In addition, we observed a trend towards enrichment of LE2- and LE4-high states in our predominantly ABC-like group of MHC class I deleted tumors, in keeping with previous observations from all-comer DLBCL cohorts where these lymphoma ecotypes were associated with inferior clinical outcomes 43. The trend towards more LE5-high tumors in the non-MHC class I deleted cases with its underlying cell states being more prevalent in healthy lymphoid tissues integrates seamlessly with this notion.
The study presents several limitations, inherent to the retrospective design. Despite this being the largest cohort of PR-DLBCL to date the sample size remains limited and samples for the study were collected over more than two decades. This results in significant heterogeneity in terms of first- and later-line treatment approaches and limited follow-up information. These factors prohibit a meaningful integrative analysis of molecular findings and clinical endpoints. In addition, there is a potential for cases with occult non-renal primary manifestations, which may have been missed by diagnosing physicians. However, considering that secondary renal manifestations typically occur in advanced-stage disease, the likelihood of a significant fraction of patients harboring secondary renal manifestations is relatively low. In summary, we present the largest molecular study on PR-DLBCL to date, untangle the genomic and transcriptional heterogeneity of the disease, and uncover evidence of significant pathogenetic overlap between PR-DLBCL and IP-LBCL. Our observations may inform future risk-adapted therapeutic approaches.
Data Availability
All data produced in the present work are contained in the manuscript
Declarations
Ethics approval and consent to participate
This retrospective study was approved by the ethics committee of the University of Lübeck (reference-no 18-356) and conducted in accordance with the declaration of Helsinki. Patients have provided written informed consent regarding routine diagnostic and academic assessment, including genomic studies of their biopsy specimens alongside the transfer of their clinical data.
Availability of data and material
Raw fastq files have been deposited in the European genome-phenome archive (EGA) under the accession number EGA50000000386. OncoScan Array data has been deposited in Gene Expression Omnibus (GEO) under accession number GSE270422.
Funding
This work was supported by generous funding by the Wilhelm Sander-Stiftung through a project grant (NG; Grant-Nr.: 2021.150.1). HB and AK acknowledge support from the BMBF project OUTLIVE-CRC (FKZ 01KD2103A).
Author contributions
Study concept: NG, ACF, HM
Data collection: NG, AK, VvK, PL, MK, HW, VB, SS, KK, HM, SS, KS, AA, ACF
Data analysis and creation of figures and tables: AK, NG, ACF, HB, NvB,
Initial Draft of manuscript: NG.
Critical revision and approval of final version: all authors.
Acknowledgements
The authors would like to thank Tanja Oeltermann for her skilled technical assistance. AK and HB acknowledge computational support from the OMICS compute cluster at the University of Lübeck.
Footnotes
Competing Interests: Niklas Gebauer received travel support from Beigene, Janssen, and Roche as well as honoraria from Roche, Takeda, Janssen, Menarini Stemline, and AstraZeneca.
↵** Shared senior authorship