Purpose:

We report our experience with next-generation sequencing to characterize the landscape of actionable genomic alterations in renal cell carcinoma (RCC).

Experimental Design:

A query of our institutional clinical sequencing database (MSK-IMPACT) was performed that included tumor samples from 38,468 individuals across all cancer types. Somatic variations were annotated using a precision knowledge database (OncoKB) and the available clinical data stratified by level of evidence. Alterations associated with response to immune-checkpoint blockade (ICB) were analyzed separately; these included DNA mismatch repair (MMR) gene alterations, tumor mutational burden (TMB), and microsatellite instability (MSI). Data from The Cancer Genome Atlas (TCGA) consortium as well as public data from several clinical trials in metastatic RCC were used for validation purposes. Multiregional sequencing data from the TRAcking Cancer Evolution through Therapy (TRACERx) RENAL cohort were used to assess the clonality of somatic mutations.

Results:

Of the 753 individuals with RCC identified in the MSK-IMPACT cohort, 115 showed evidence of targetable alterations, which represented a prevalence of 15.3% [95% confidence interval (CI), 12.7%–17.8%). When stratified by levels of evidence, the alterations identified corresponded to levels 2 (11.3%), 3A (5.2%), and 3B (83.5%). A low prevalence was recapitulated in the TCGA cohort at 9.1% (95% CI, 6.9%–11.2%). Copy-number variations predominated in papillary RCC tumors, largely due to amplifications in the MET gene. Notably, higher rates of actionability were found in individuals with metastatic disease (stage IV) compared with those with localized disease (OR, 2.50; 95% CI, 1.16–6.16; Fisher's P = 0.01). On the other hand, the prevalence of alterations associated with response to ICB therapy was found to be approximately 5% in both the MSK-IMPACT and TCGA cohorts and no associations with disease stage were identified (OR, 1.35; 95% CI, 0.46–5.40; P = 0.8). Finally, multiregional sequencing revealed that the vast majority of actionable mutations occurred later during tumor evolution and were only present subclonally in RCC tumors.

Conclusions:

RCC harbors a low prevalence of clinically actionable alterations compared with other tumors and the evidence supporting their clinical use is limited. These aberrations were found to be more common in advanced disease and seem to occur later during tumor evolution. Our study provides new insights on the role of targeted therapies for RCC and highlights the need for additional research to improve treatment selection using genomic profiling.

Translational Relevance

The central premise of precision oncology is the ability to select a therapy specific to the molecular characteristics of a given patient's tumor. One approach to achieving this individualized therapy goal is to determine the genomic alterations present in a tumor that can predict response to specific drugs. Although the evaluation of somatic DNA alterations has proved useful in the management of tumors such as melanoma, lung, breast, and prostate cancer, its utility has been limited in renal cell carcinoma (RCC). Our study describes the landscape of actionable alterations in different malignancies using next-generation sequencing data from several patient cohorts, with emphasis in RCC. Somatic aberrations identified in cancer genes were annotated using OncoKB, a curated precision oncology knowledge database projecting drug action ability based on available clinical evidence; this tool adheres to the FDA recommendations and expert panel guidelines to categorize clinical data. The landscape of actionable mutations, copy-number variations and fusions, as well as their association with relevant clinical parameters is explored. The relative timing of these alterations during tumor evolution and their role in metastatic progression are further expanded upon. Our results suggest a largely subclonal presence of actionable alterations as well as enrichment in advanced disease. Finally, assessing for microsatellite instability, mutational burden, and mismatch repair deficiency revealed that there is only a small subset of tumors containing features predictive of response to immune-checkpoint blockade therapy. Although the prevalence of actionable somatic alterations with therapeutic implications was markedly low in RCC and the alterations observed were mostly associated with response in other indications, our data suggest a potential role for genomic biomarkers in RCC. Clinical sequencing currently has limited utility in guiding therapeutic decisions in RCC, and further research is required to understand the potential role of these tools in disease management.

A recent surge in the understanding of the molecular and genetic underpinnings of several malignancies has led to the ability to precisely target mutations and aberrant pathways in patients with cancer, thus harkening the era of “precision oncology.” Prospective clinical sequencing has etched its way into the treatment algorithms and guidelines of many cancer types, as the benefits of such sequencing efforts have significantly improved outcomes in genetically defined populations (1). The gradually increasing shift toward personalized medicine likewise highlights the disparate benefits achieved by molecular profiling of specific cancers. Although the benefits of tumor genotyping are clear in certain tumors, they are less apparent and sometimes absent in others (2–6).

Renal cell carcinoma (RCC) represents one such cancer with seemingly fewer benefits reaped from genomic innovations. Despite therapeutic advances stemming from insight into the fundamental dysregulation of the HIF1a pathway, long-term survival is not achieved for the majority of patients with metastatic disease (7). Thus, ongoing efforts to exact and refine prognostic models and treatment paradigms in RCC have progressively integrated molecular and genotypic information (8–13). Although the advent of tyrosine kinase inhibitors (TKI) and immune-checkpoint blockade (ICB) therapy represent significant therapeutic advances in RCC, there are currently no known biomarkers of response to therapy in this disease (14).

We report our institutional experience in a cohort of 753 RCCs profiled with a targeted next-generation sequencing (NGS) panel, summarizing the frequency and evidence of actionability for all identified somatic alterations that have been reported to be associated with therapy response in any cancer type. Given that the therapies in question are mostly relevant in the metastatic setting, we expand our analyses to explore different cohorts and look for associations between prevalence of these alterations and disease stage. We further leverage data from other landmark sequencing efforts to validate our findings and explore the role of these alterations during tumor evolution, with emphasis on metastatic disease.

Ethical considerations and patient selection

All research-related activities were preapproved by the institutional ethics review board at Memorial Sloan Kettering Cancer Center. All the individuals who participated provided written informed consent to have their tissue profiled and their clinical data shared in a nonidentifiable manner. All activities related to this study were conducted in accordance with the principles of Good Clinical Practice and the Declaration of Helsinki. Individuals with a diagnosis of more than one cancer type were excluded from the analysis. In addition, in the RCC cohort, patients with multifocal RCCs or benign renal tumors (i.e., angiomyolipoma or oncocytoma) were excluded from the analysis.

Targeted NGS assay

Sequencing was performed at a Clinical Laboratory Improvement Amendments (CLIA)–certified laboratory using MSK-IMPACT, a targeted NGS panel approved by the FDA for the study of solid tumors (15). The panel captures the exons of 341 to 468 cancer genes (varies by version, Supplementary Table S1), a set of known single-nucleotide polymorphisms (SNP) tiled across the genome used for copy-number estimation, as well as the degree of microsatellite instability (MSI; via MSIsensor; ref. 16). Different thresholds have been proposed to determine the degree of MSI in tumors (e.g., >3; refs. 16, 17). To minimize false positives, we used a more restrictive threshold that has been reported in previous colorectal cancer studies. Individuals with matched-normal samples available whose tumors had MSIsensor scores ≥10 were considered to show MSI. The types of samples included in the analysis consisted of both primary and metastatic tumor specimens. For individuals with more than one specimen available, all the unique somatic events identified across all samples were considered. Individuals included were categorized as having localized (stage I–III) or metastatic disease (stage IV) according to the American Joint Committee on Cancer 8th edition. Raw data were processed and analyzed using a previously validated bioinformatics pipeline (15) that includes mutation and fusion detection as well as conventional copy-number estimation (via GISTIC 2.0; ref. 18).

Somatic alteration actionability: OncoKB

Only somatic aberrations were considered in the analysis. Individual gene-level events were categorized according to their therapeutic implications using the publicly available Precision Oncology Knowledge Base (OncoKB) through its Python implementation (https://github.com/oncokb/oncokb-annotator/). This clinical support tool distills information from the published literature, projecting drug actionability based on the available clinical evidence. It is updated on a continuous basis, including emerging biomarker data for FDA-approved regimens but also those still under clinical investigation.

The OncoKB workflow has been previously described in detail (Supplementary Fig. S1; ref. 19). Briefly, alterations are first identified from public databases. Clinical therapeutic implications of alterations are then curated from several public resources; finally, OncoKB annotations are vetted by a committee of clinicians and cancer biologists across a number of disease management teams (based on current guidelines) before being made publicly available (http://oncokb.org). A level of evidence classification system was developed, and potentially actionable alterations are assigned to one of four levels based on the strength of evidence that the alteration is an FDA-recognized or standard care biomarker predictive of response to an FDA-approved drug (level 1), a standard care biomarker recommended by expert panels predictive of a response to an FDA-approved drug (level 2), has compelling clinical evidence supporting the biomarker being predictive of response to a drug (level 3A), or alterations that are considered predictive of response based on promising clinical data to FDA-approved or investigational agents being tested in clinical trials in another indication (level 3B). Only alterations with therapeutic implications corresponding to OncoKB levels of evidence 1 to 3B were considered “actionable” and included in this analysis. The OncoKB database query was performed on December 28th, 2020.

Two additional open-source knowledge databases were queried for comparison purposes (Supplementary Fig. S1C), these included the Cancer Genome Interpreter database (20) and the Clinical Interpretation of Variants in Cancer (CIViC) database (21). The data from both platforms were queried up to January 2021. All the mutations identified in RCC samples from the MSK-IMPACT cohort were used for this analysis (n variants = 1,808). We could confirm that OncoKB reliably captured all the actionable variants listed in the other databases. The only five variants not detected by OncoKB likely represent false positives; these consisted of truncating mutations in oncogenes that CIViC captured due to the nomenclature being nonspecific. In this particular case, the mutations in those genes were logged as potentially oncogenic but OncoKB does not consider truncating variants in oncogenes as such.

DNA mismatch repair deficiency analysis

For a subset of tumors with available allele-specific copy-number data an in-depth genomic analysis was performed with the goal of showing potential biallelic inactivation events (Supplementary Fig. S2, “RCC copy-number cohort”). Mutations and copy-number alterations across the four genes approved by the FDA to determine mismatch repair (MMR) deficiency (i.e., MLH1, MSH2, MSH6, and PMS2) were evaluated. Samples included in this analysis had a tumor purity ≥20%, which allows for allele-specific resolution during copy-number estimation. The rest of the samples not included in the study had purity values between 10% and 20% that is suboptimal for these purposes, but still allows assessment of imbalanced events such as chromosomal gains. Allele-specific estimates allow for the integration of mutation and CN data to better detect “double-hits” in tumor suppressors. Tumors displaying heterozygous (i.e., single copy) losses in MMR genes and no evidence mutations in the remaining allele were not considered. These same genes were also investigated when replicating the analysis in the validation cohorts. The analysis included mutation and allele-specific copy-number estimation using FACETS v0.5.6 (22) through a publicly available R package (https://github.com/mskcc/facets/).

Tumor mutational burden

Tumor mutational burden (TMB) was defined as the number of nonsynonymous somatic mutations per DNA megabase (mut/Mb). Tumors with ≥10 mut/Mb were considered to have high TMB (TMBhi); this was based on the recent FDA approval for pembrolizumab in solid tumors (23). Because the samples included in the study were profiled using a variety of sequencing assays, the results were normalized by the total length of the panel (in Mb) for comparison purposes. The total length of the IMPACT sequencing panels used in the analysis were 0.9 Mb (IMPACT341), 1.1 (IMPACT 410), and 1.3 Mb (IMPACT468). Tumor samples profiled via exome-wide sequencing (see Materials and Methods, result validation) were normalized considering a panel length of 36.8 Mb (24).

Result validation in the TCGA KIPAN, record-3, COMPARZ, and IMmotion150 cohorts.

Sequencing data from The Cancer Genome Atlas (TCGA) pan-kidney cohort (KIPAN) were used for validation. This comprises a total of 695 individuals with clear cell (n = 356), papillary (n = 274), and chromophobe RCC (n = 65) whose primary tumors were profiled using exome sequencing. Data obtained from the publicly available NCI Pancancer Atlas website (https://gdc.cancer.gov/about-data/publications/pancanatlas) were used for analysis. Although the original TCGA publications included a total of 860 individuals, only a subset of the samples was later found to have high-quality data and thus included in the Pancancer Atlas studies. Only mutation and conventional CN data were available for the TCGA. This cohort consisted exclusively of primary samples; however, 10% of the individuals had stage IV (metastatic) disease at the time of diagnosis/profiling.

In addition, sequencing data from several randomized clinical trials in patients with metastatic RCC were obtained from the original investigators and used for validation purposes; only somatic mutation data were available for these cohorts. Although all individuals had clinical evidence of metastasis, data on the sample type profiled (primary vs. metastases) were only available for some. RECORD-3 was a phase 2 clinical trial comparing the efficacy of sequential therapy with everolimus and sunitinib in metastatic RCC (25). Tumor samples from these individuals were sequenced using the IMPACT341 panel and the majority (94%) consisted of primary tumors (12). Similarly, data from COMPARZ (26), a phase 3 randomized clinical trial comparing the efficacy of sunitinib versus pazopanib in metastatic clear cell RCC were analyzed; only primary tumor samples from the individuals enrolled in the study were profiled using the IMPACT410 panel (8). Finally, we included data from IMmotion150, a more recent phase 2 trial of sunitinib versus atezolizumab versus the combination of atezolizumab plus bevacizumab in metastatic RCC (27). The tumor samples from this study were profiled using exome sequencing, but data on the type of sample used were not available for review (9).

Clonality of targetable alterations in clear cell RCC

Multiregional sequencing data from the TRACERx RENAL cohort (28) were used to assess the clonality of the most frequent targetable alterations identified. In this landmark study, multiple tumor regions from 101 patients were profiled with a targeted NGS panel that captures approximately 110 well-known RCC drivers. Only somatic mutations were considered in this analysis. The same clonality definitions from the original publication were used [i.e., mutations had to be present in every region at a cancer-cell fraction (CCF) ≥50% to be considered clonal]. CCF is a measure of relative abundance that represents the proportion of cancer cells in the specimen bearing a specific variant. Only nonsynonymous somatic mutations at or above 5% CCF were reported in the TRACERx studies and therefore considered in the analysis.

Statistical analyses

All statistical analyses were performed using the R platform v4.0.0. Results were reported as point estimates along with their 95% confidence intervals. Hypothesis tests were always two-sided and a P value below 0.05 was used to define statistical significance. Differences in proportions were evaluated using the prop.test() function that considers the χ2 distribution with k−1 degrees of freedom. Fisher's exact tests and odds ratios were estimated using the fisher.test() function. Both functions are contained in the R package “stats.” Odds ratios were estimated considering the hypergeometric distribution from a given 2 × 2 table while assuming fixed marginals (i.e., conditional maximum likelihood estimate). Multivariable logistic regression analysis was performed to evaluate the factors associated with increased odds of encountering a targetable alteration or biomarker of ICB response in RCC, results were presented in the form of forest plots using the R package “forestmodel.”

After excluding individuals with multiple cancer types, the pan-cancer MSK-IMPACT cohort consisted of 37,978 patients (Supplementary Fig. S2). The final RCC cohort consisted of 753 patients, of which 15.3% (95% CI, 12.7%–17.8%) harbored at least one targetable alteration in their tumor specimen (exclusive of ICB-associated features). On the other hand, tumors such as melanoma (67.2%), breast (66.3%), and bladder carcinomas (62.9%) showed high a prevalence of actionable alterations (Fig. 1). These high actionability rates are largely driven by alterations in single genes that are recurrent in specific cancer types (e.g., BRCA2 and BRAF alterations in breast cancer and melanoma, respectively). None of the targetable alterations identified in RCC consisted of FDA-recognized biomarkers (level 1), and most of the evidence (83.5%, level 3B) originated from studies in other tumor types. The majority of individuals in the RCC cohort had tumors of clear cell histology (59.7%), followed by papillary (8.6%), and chromophobe (5.7%). Remaining subtypes, grouped as “other,” included fumarate hydratase (FH)–deficient, succinate dehydrogenase (SDH)–deficient, collecting duct, translocation-associated, and unclassified RCC. Notably, this cohort consisted mostly of patients with stage IV disease (62.8%), and 36.7% of samples originated from metastatic sites (Supplementary Fig. S3).

Figure 1.

Actionable alterations identified in the MSK-IMPACT pan-cancer cohort. The top 15 commonest cancers in MSK-IMPACT are shown, ranked from highest to lowest prevalence of actionable genomic alterations (exclusive of ICB-associated features). The prevalence of actionable events in a given cancer type is inherent to the tumor biology and the role that certain well-known actionable events play in tumor evolution. NSCLC, non–small cell lung cancer; VUS, variant of unknown significance.

Figure 1.

Actionable alterations identified in the MSK-IMPACT pan-cancer cohort. The top 15 commonest cancers in MSK-IMPACT are shown, ranked from highest to lowest prevalence of actionable genomic alterations (exclusive of ICB-associated features). The prevalence of actionable events in a given cancer type is inherent to the tumor biology and the role that certain well-known actionable events play in tumor evolution. NSCLC, non–small cell lung cancer; VUS, variant of unknown significance.

Close modal

The prevalence of targetable alterations was compared among histologic subtypes (Fig. 2A). Although chromophobe tumors appeared to have a lower prevalence of actionable alterations, no significant differences were observed in the rate of actionability among the different subtypes (clear cell = 16.3%; 95% CI, 12.9–19.7), papillary = 15.4% (95% CI, 6.6–24.2), chromophobe = 7.0% (95% CI, 0–14.6), other = 14.7% (95% CI, 9.8–19.7), χ2 d.f. = 3, P = 0.4). Papillary tumors exhibited the largest proportion of alterations with level 2 OncoKB evidence (12.3%), a result that was driven by a higher frequency of gains/amplifications involving the MET locus (Ch 7q31). In clear cell tumors, mutations were more common and the prevalence of targetable alterations were level 2 (0.9%), level 3A (1.3%), and level 3B (14.1%). Thus, not only was the overall frequency of actionable genomic alterations low across RCC but the evidence supporting their use was rather weak, with complete absence of level 1 targets (not considering alterations associated with response to ICB therapy). Regarding the genes involved (Fig. 2B), these included TSC1 (23.5%), PIK3CA (21.7%), ATM (14.8%), and MET (12.2%). Supplementary Fig. S4 and Supplementary Table S2 present a summary of the gene-level actionable alterations identified.

Figure 2.

Targetable genomic alterations in RCC. A, Proportion of individuals with any given targetable alteration, broken down by RCC subtype and highest level of evidence identified (left). The pie charts (right) show the type of somatic alterations observed in each subtype. B, Genes bearing targetable alterations identified in the RCC cohort, the bars represent the relative frequency of druggable alterations in each gene (N = 753). VUS, Variant of unknown significance; CNV, copy-number variation.

Figure 2.

Targetable genomic alterations in RCC. A, Proportion of individuals with any given targetable alteration, broken down by RCC subtype and highest level of evidence identified (left). The pie charts (right) show the type of somatic alterations observed in each subtype. B, Genes bearing targetable alterations identified in the RCC cohort, the bars represent the relative frequency of druggable alterations in each gene (N = 753). VUS, Variant of unknown significance; CNV, copy-number variation.

Close modal

The prevalence of somatic alterations predictive of response to ICB therapy in other solid-tumor malignancies was assessed in the MSK-IMPACT “RCC copy-number” cohort (Fig. 3). The alterations evaluated included TMB, MSI, as well as genomic alterations in MMR genes. The median TMB was 3.1 mut/Mb and only 1.1% of individuals showed TMBhi tumors (95% CI, 0.3–1.9). Notably, only one individual with RCC showed microsatellite-instability and consisted of a chromophobe RCC tumor. It is worth noting that other studies have used more lenient definitions for tumors with MSI (16, 17). When applying these parameters to our data, we observed that an MSI score threshold of 3.0 increases the number of MSI tumors from 1 to 18 (2.4%). The prevalence of somatic MMR gene alterations was found to be 4% (95% CI, 2.5–5.5%). Of these, the majority (80.8%) consisted of biallelic inactivation events, which included homozygous deletions or a combination of mutation and heterozygous loss, whereas the rest showed only mutations. The majority of mismatch repair deficiency (dMMR) alterations were found in clear cell RCC tumors. The combined prevalence of all biomarkers of response to ICB therapy (i.e., MSI or TMBhi or dMMR) in the MSK-IMPACT cohort was 5.0% (95% CI, 3.3–6.6). Of the tumors that were found to bear a biomarker of response to ICB, the majority (75%) were due to alterations in MMR genes, and only 21.9% of patients had TMBhi tumors.

Figure 3.

Spectrum of somatic alterations associated with response to immune-checkpoint blockade (ICB). A, Prevalence of mutations and copy-number alterations resulting in deficient mismatch-repair tumors (dMMR) in the “RCC copy-number” cohort (n = 645). B, Detailed representation of the mutation and copy-number alterations in tumors identified as mismatch repair deficient. The top panel represents the RCC subtype in question. C, Distribution of tumor mutational burden (nonsynonymous mutations per DNA megabase). D, Proportion of patients with somatic alterations associated with response to ICB therapy. dMMR: deficient mismatch repair; MSI: microsatellite instability; TMBhi: high tumor mutational burden. VUS: Variant of unknown significance. WT: wild-type.

Figure 3.

Spectrum of somatic alterations associated with response to immune-checkpoint blockade (ICB). A, Prevalence of mutations and copy-number alterations resulting in deficient mismatch-repair tumors (dMMR) in the “RCC copy-number” cohort (n = 645). B, Detailed representation of the mutation and copy-number alterations in tumors identified as mismatch repair deficient. The top panel represents the RCC subtype in question. C, Distribution of tumor mutational burden (nonsynonymous mutations per DNA megabase). D, Proportion of patients with somatic alterations associated with response to ICB therapy. dMMR: deficient mismatch repair; MSI: microsatellite instability; TMBhi: high tumor mutational burden. VUS: Variant of unknown significance. WT: wild-type.

Close modal
Figure 4.

Prevalence of actionable genomic alterations in RCC tumors from the TCGA cohort. A, Prevalence of targetable alterations across all RCC subtypes (left) and broken down by histologic subtype (right). B, Targetable somatic alterations by gene. C, Prevalence of alterations associated with response to immune-checkpoint blockade in the TCGA cohort (microsatellite data not available). VUS, variant of unknown significance; CNV, copy-number variation; dMMR, deficient mismatch repair; TMBhi, high tumor mutational burden. TCGA RCC cohort definitions, KIPAN: pan-kidney; KIRC: clear cell; KIRP: papillary; KICH: chromophobe.

Figure 4.

Prevalence of actionable genomic alterations in RCC tumors from the TCGA cohort. A, Prevalence of targetable alterations across all RCC subtypes (left) and broken down by histologic subtype (right). B, Targetable somatic alterations by gene. C, Prevalence of alterations associated with response to immune-checkpoint blockade in the TCGA cohort (microsatellite data not available). VUS, variant of unknown significance; CNV, copy-number variation; dMMR, deficient mismatch repair; TMBhi, high tumor mutational burden. TCGA RCC cohort definitions, KIPAN: pan-kidney; KIRC: clear cell; KIRP: papillary; KICH: chromophobe.

Close modal

The TCGA KIPAN cohort used for validation purposes included 695 individuals with clear cell (KIRC), papillary (KIRP), and chromophobe (KICH) RCC tumors. Of note, all of these samples were obtained from primary tumors and the vast majority of the individuals (89.9%) had not developed metastatic disease at the time the TCGA analysis was reported. Notably, this cohort contained a higher frequency of papillary tumors compared with the MSK-IMPACT cohort (39.4 vs. 8.6, difference = 30.8%; 95% CI, 26.5–35.1; P < 0.001). Which could be due to differences in the criteria used to define papillary histology in the two studies (i.e., TCGA includes both papillary RCC type 1 and 2). The frequency of targetable alterations in the TCGA KIPAN cohort was found to be 9.1% (95% CI, 4.0%–7.1%), significantly lower than the MSK-IMPACT cohort (difference = 4.5%; 95% CI, 1.1–7.9; χ2 d.f. = 1; P = 0.009; excluding fusions). This difference was mainly driven by clear cell RCC tumors (difference = 7.2%; 95% CI, 2.5–11.9; χ2 d.f. = 1; P = 0.003). The most commonly altered genes were very similar in both studies, with the top four targetable genes being ATM, TSC1, MET, and PIK3CA, suggesting an overrepresentation of mTOR pathway alterations. We observed that the proportion of actionable alterations in this pathway (i.e., considering only TSC1, TSC2, PIK3CA or MTOR) was higher in MSK-IMPACT compared with the TCGA (difference = 20.7%; 95% CI, 4.6–36.8; χ2 d.f. = 1; P = 0.01). Finally, for the TCGA cohort the frequency of alterations associated with response to ICB therapy in other cancers (i.e., dMMR/TMBhi) was 5.2% (95% CI, 3.5–6.8), nearly identical to the MSK-IMPACT cohort (difference = 0.4%; 95% CI, 2.1–2.9; χ2 d.f. = 1; P = 0.9). Only 1.9% of tumors were found to be TMBhi in the TCGA cohort.

Next, we explored the association between the presence of actionable alterations and metastasis. The odds of finding at least one actionable alteration were found to be significantly higher in patients with metastatic disease (OR, 2.50; 95% CI, 1.16–6.16; Fisher's; P = 0.01). Notably, this difference was found to be due to disease stage and did not appear to be confounded by the type of sample profiled (Fig. 5A). On the contrary, no differences were observed in the prevalence of ICB-associated biomarkers (i.e., TMBhi or MSI or dMMR) between individuals with localized and metastatic disease (OR, 1.35; 95% CI, 0.46; 5.40; P = 0.8); with primary tumors and metastatic samples showing similar results (OR, 0.97; 95% CI, 0.44–2.21; P = 0.99). Mutation data from several clinical trials as well as from the TCGA and MSK-IMPACT cohorts were aggregated and a total of 1434 individuals with metastatic RCC were analyzed. Of these, 10.9% (95% CI, 9.29–12.5) showed at least one actionable alteration and 3.3% (95% CI, 2.4–4.2) were either dMMR/TMBhi. Although between-cohort differences were observed in the prevalence of actionable alterations, no differences were seen in the prevalence of dMMR/TMBhi tumors (Fig. 5B).

Figure 5.

Spectrum of actionable somatic alterations in metastatic RCC. A, Prevalence of somatic alterations in the “RCC copy-number” cohort (n = 645) identified as targetable (left) or associated with response to immune-checkpoint blockade (ICB; right), results are broken down into three groups based on sample type and disease stage. B, Prevalence of somatic mutations identified as actionable (left) or associated with ICB therapy response (right) among different cohorts of patients with metastatic RCC. The overall proportion of alterations among all individuals is shown (N = 1,434), error bars represent the 95% confidence intervals. dMMR, deficient mismatch repair; MSI, microsatellite instability; TMBhi, high tumor mutational burden; N.S., not significant; *, two-proportion z-test; P < 0.05.

Figure 5.

Spectrum of actionable somatic alterations in metastatic RCC. A, Prevalence of somatic alterations in the “RCC copy-number” cohort (n = 645) identified as targetable (left) or associated with response to immune-checkpoint blockade (ICB; right), results are broken down into three groups based on sample type and disease stage. B, Prevalence of somatic mutations identified as actionable (left) or associated with ICB therapy response (right) among different cohorts of patients with metastatic RCC. The overall proportion of alterations among all individuals is shown (N = 1,434), error bars represent the 95% confidence intervals. dMMR, deficient mismatch repair; MSI, microsatellite instability; TMBhi, high tumor mutational burden; N.S., not significant; *, two-proportion z-test; P < 0.05.

Close modal

Multivariable logistic regression analysis was used to evaluate the association between disease stage (i.e., metastatic/stage IV vs. localized/stages I–III) and presence of actionable alterations while adjusting for histologic subtype (Supplementary Fig. S5). In the MSK-IMPACT cohort, stage IV disease was found to be significantly associated with higher odds of finding an actionable alteration (OR, 1.79; 95% CI, 1.08–3.15; P = 0.03) but not a biomarker of response to ICB therapy (OR, 1.20; 95% CI, 0.45–4.16; P = 0.7). When attempting to reproduce these analyses in the TCGA cohort, no significant associations were observed with either targetable alterations (OR, 1.42; 95% CI, 0.6–3.01; P = 0.4) nor biomarkers of ICB response (OR, 1.89; 95% CI, 0.68–4.49; P = 0.2).

Finally, we assessed the relative timing of the most frequent actionable mutations during tumor evolution using multiregional sequencing data from the TRACERx RENAL cohort. The final cohort included 96 clear cell patients with RCC, with a total of 1,146 regions profiled (Fig. 6A). Individuals with multifocal tumors were excluded from the analysis (n = 5). The frequency of targetable alterations in this cohort was found to be 18.8% (95% CI, 10.9–26.6), a result comparable with the MSK-IMPACT cohort (−4.2%; 95% CI, 13.3–4.9; χ2d.f. = 1; P = 0.4). However, in only 4.2% (95% CI, 0.2–8.2) of the tumors these mutations were found to be clonal and thus present throughout the entire tumor (Fig. 6B). All the subclonal events identified were observed in specific regions of the primary tumor and none was private to metastatic lesions.

Figure 6.

Clonality of targetable mutations assessed via multiregional sequencing. A, Number of tumor regions profiled per tumor in the TRACERx RENAL cohort (N individuals = 96; N regions = 1,146). B, Prevalence of targetable mutations broken down by level of evidence and clonal status. C, Clonality of targetable alterations identified, separated by gene (only ∼100 genes profiled). VUS, variant of unknown significance.

Figure 6.

Clonality of targetable mutations assessed via multiregional sequencing. A, Number of tumor regions profiled per tumor in the TRACERx RENAL cohort (N individuals = 96; N regions = 1,146). B, Prevalence of targetable mutations broken down by level of evidence and clonal status. C, Clonality of targetable alterations identified, separated by gene (only ∼100 genes profiled). VUS, variant of unknown significance.

Close modal

Using a large prospective clinical sequencing database of more than 38,000 patients with any cancer type, we demonstrate a 15.3% prevalence of actionable alterations in RCC. Our analysis of the TCGA RCC cohort confirms this low frequency of targetable alterations in a separate set of 695 patients and shows a near-identical, low prevalence of biomarkers previously proposed to associate with response to ICB therapy across solid-tumor malignancies. We show that actionable alterations tend to be enriched in advanced RCC and, by using multiregional sequencing data, we demonstrate that many of these mutations tend to be subclonal.

Although current treatment paradigms for patients with RCC use targeted therapies, they are not directed against specific actionable genomic alterations. Several large-scale sequencing efforts by the TCGA and others have characterized the genomic landscape of RCC (29–31). However, other than insights in molecular diagnostics, few practical clinical applications of comprehensive genetic profiling can be extracted from these analyses and a paucity of data exists on the clinical actionability of these alterations. An earlier report of our experience with MSK-IMPACT by Carlo and colleagues (32) detailed the frequency of clinically actionable mutations across a cohort of 115 patients with non-clear cell RCC, showing 15.3% of tumors harboring clinically actionable somatic alterations. Here, we expand on the spectrum of alterations in RCC, with the goal of providing new insights into the role of genomic actionability in this context.

Results are introduced at the pan-cancer level for comparative purposes, but formal testing on the differences between cancer types was purposely not performed due to the large number of potential confounders (e.g., distribution of histologic subtypes, disease stage, disease frequency, therapeutic context, etc.). For example, it is expected that tumors whose targetable alterations correspond to cancer initiation events will show higher rates of actionability (e.g., mutations in BRCA1/2 in ovarian/breast cancer or BRAF in melanoma). Further analyses on cancer types other than RCC are outside the scope of this study. By exploring the association between actionable alterations and other clinical variables we made important observations, such as an obvious enrichment of MET amplifications in papillary RCC, which have been shown to be associated with response to crizotinib (33, 34). We also noted a higher prevalence of targetable alterations in patients with metastatic disease. However, the association with advanced stage could not be replicated in the TCGA cohort (stage I–III vs. stage IV; difference 2.9%; 95% CI, 5.6–11.5; P = 0.6) and it is unclear whether factors such as study-specific tumor selection factors, might have influenced the results (e.g., clear cell papillary tumors, a distinct and indolent molecular entity, are known to be included in the KIRC cohort; ref. 35).

The majority of the targetable alterations identified were found in four key components of the PI3K/mTOR pathway (i.e., MTOR, PIK3CA, TSC1, TSC2). These accounted for 55.6% of all targetable alterations in the MSK-IMPACT renal cohort and 34.9% in the TCGA KIPAN cohort. Randomized clinical trials showing clinical benefit of the mTOR inhibitors temsirolimus and everolimus in metastatic RCC ultimately led to regulatory agency approval (36–38). Although earlier outlier studies from our group and others identified mTOR and TSC1 as candidate biomarkers in a limited number of cases with exceptional response to rapalog therapy, these findings could not be confirmed in an unselected population (39–41). Interestingly, a significant difference was observed in the rate of actionable targetable alterations in mTOR pathway genes between the MSK-IMPACT and TCGA cohorts. These results could be due at least in part to the differences in the distribution of disease stage and histologic subtypes in the two studies, such as the overrepresentation of metastatic clear cell RCC in the MSK-IMPACT cohort.

Biomarkers of response to ICB therapy were explored separately throughout the study. Somatic mutations in the MMR genes MLH1, MSH2, MSH6, and PMS2 lead to dMMR, genetic hypermutability, and a resultant MSI phenotype (42). MSI tumors have been shown to be susceptible to the antiprogrammed death 1 immune checkpoint inhibitor pembrolizumab (43), resulting in the FDA-approval in 2017 of pembrolizumab for the treatment of MSI or dMMR solid tumors in pretreated patients (14). As further indication of a paradigm shift toward personalized medicine, this particular approval was unique in that it was the first site-agnostic anticancer therapy approved for use based on a biomarker, and it was followed by an additional approval for TMBhi tumors (23). Cohort studies ranging from 51 to 339 patients with predominantly localized RCC report a 0.7% to 5.9% prevalence of MSI tumors (44–46). These newly recognized biomarkers might represent an emerging change in paradigm for guiding therapeutic decisions in RCC, as the identification of specific genomic changes in tumors may render patients eligible for basket trials regardless of tumor type or location. To our knowledge, this analysis represents the largest MSI analysis in RCC to date, which has particular implications for immunotherapy-eligible patients in the metastatic setting.

Among the MSK-IMPACT RCC cohort, only one patient out of 753, or 0.13%, harbored an MSI tumor. Although this is a rather low value, it falls within the confidence interval of previously published estimates, which often include a 0% rate. On the other hand, we did use a strict MSI cutoff, which might be a contributing factor. Because of the rarity of this feature in RCC, there is a paucity of data regarding optimal definitions and further research is warranted to optimize these definitions for kidney cancer. In terms of the rate of TMBhi tumors, we found that only 1.1% of RCC tumors showed 10 muts/Mb, which is consistent with prior pan-cancer reports that place RCC in the lower end of the TMB spectrum (47, 48). It is important to note that the cohorts analyzed in this study were profiled using different sequencing platforms and bioinformatics pipelines. Nevertheless, a strong correlation between TMB values obtained with targeted panels and exome sequencing has previously been reported (49, 50) and TMB measurements from these assays have successfully been used to predict outcomes in the ICB therapy setting (48).

With regards to the prevalence of MMR gene alterations observed, approximately 4% of tumors ultimately harbored mutations in MLH1, MSH2, MSH6, or PMS2. However, it should be noted that IHC would be needed to confirm loss of one or more of these proteins and label these tumors as dMMR according to the recent FDA-approval. Although an attempt was made to determine the number of tumors that had “double hits” in these genes, results should be interpreted with caution as dMMR can arise from other molecular mechanisms that were not considered in the study, such as epigenetic silencing (51). When aggregating the data, we observed a prevalence of dMMR–TMBhi–MSI tumors of 5% and no differences among any of the cohorts evaluated were observed. It is important to note that only mutations were considered in the validation sets, which will likely result in an underestimation of the true prevalence at the population level. A recent retrospective analysis of DNA damage repair (DDR) gene mutations (which include MMR genes) in RCC revealed a 19% prevalence rate in 229 patients with metastatic clear cell RCC, which was associated with longer overall survival in patients receiving immune-oncology and not TKI therapies (52). However, the larger set of genes and the different DNA repair processes considered complicate the interpretation of these findings. Although further research is needed to explore the role of these alterations in ICB response prediction, our data suggest that they could be valuable in a small subset of individuals.

The rarity of targetable alterations and the unknown degree to which identified alterations respond to genomically targeted therapies likely do not justify the widespread adoption of clinical sequencing in RCC currently. Nevertheless, we and others have shown that mutations in RCC can have prognostic value even though they are limited in guiding therapy (13, 53), which could lead to new efforts in the adoption of this technology in clinical practice. As Tannock and colleagues posit, factors predicated around the cost of large-scale genomic analyses, fostering collaborative efforts to limit waste of resources, and tumor-specific factors such as intratumoral heterogeneity, must be brought to the forefront of any efforts seeking to integrate costly tests into routine care (54).

Intratumoral heterogeneity in RCC is significant, and may explain the modest responses observed with targeted antiangiogenic therapies despite near-ubiquitous VHL gene inactivation and resultant angiogenic dysregulation in clear cell RCC (31, 55). Although detailing the clinical responses to genomically targeted therapies was not the goal of the current study, we did assess the clonality of druggable alterations in the TRACERx RENAL cohort (28). On the rare instances when a targetable alteration is identified, these alterations were found to be mostly subclonal (i.e., present only in a subset of tumor cells), suggesting that single-region profiling is inadequate in capturing the full spectrum of these alterations. Notably, all the subclonal alterations identified were private to the primary tumors—a finding that if validated could have important implications in the treatment of metastatic disease. MMR genes were not sequenced and MSI data were not available on this research endeavor, limiting our ability to assess ICB-associated alterations in this cohort.

We recognize several limitations to our study. Data from MSK-IMPACT are based on the genetic profile of a single region of tumor that may potentially limit the ability to detect subclonal targetable alterations (56, 57). As a result, the true prevalence of targetable alterations in RCC might have been underestimated. This is particularly important in the metastatic disease setting, where the genomic composition of primary tumors might differ significantly from that of metastatic deposits (58). Notably, the absence of metastatic sample profiling in the TCGA cohort, together with the abundance of such specimens in the MSK-IMPACT cohort, might have affected our results. For example, a potential undersampling of targetable events in the TCGA study (which included only primaries) could help explain the differences observed in the prevalence of targetable alterations by disease stage in these two cohorts.

Nevertheless, analysis of a single tumor sample reflects current clinical sequencing efforts, and the results reported in our study are likely a reflection of the rates of targetable alterations that will be encountered in clinical practice. In addition, bioinformatic tools have inherent limitations and biases, which inevitably result in batch effects and between-cohort differences that are difficult to account for. Given the differences that exist in sequencing platforms, bioinformatic processing and patient selection between the different cohorts studied, we did not perform formal statistical testing to compare the prevalence of actionable alterations or biomarkers of ICB response in our study.

Although several precision oncology knowledge databases exist and other investigators have reported a higher targetable variant detection rate by using more than one tool to annotate variants (59), we used OncoKB in this study for various reasons. Compared with other tools that rely on an online community to distill clinical actionability data, this platform is curated by a panel of experts from a world-leading institution in cancer genomics research. Furthermore, its frequent updates and strict adherence to expert panel guidelines to categorize clinical data make it extremely useful for practical purposes. Many other knowledge databases are in early phases of development or lack sufficient information to support treatment decisions across distinct tumor types. Importantly, OncoKB is freely accessible with standardized, interpretable formatting intended for clinicians of all knowledge levels; in fact, it is currently being considered for FDA approval for use as a clinical decision-support tool. We must emphasize that the definitions of actionability of most of the alterations described in our cohort (level 3B) are based on clinical evidence extracted from other cancer types. Although the targets in question have not been specifically studied in the RCC population, these results are included for descriptive purposes and are meant to be hypothesis-generating, not to be interpreted as definitive proof of response prediction in RCC.

Finally, it is important to acknowledge that the prevalence of actionable alterations will almost certainly change over time with the recognition of new biomarkers of response and the development of novel targeted therapeutics, thus the need for an up-to-date resource that is vetted with the highest clinical standards. For example, targeting truncal molecular alterations in RCC (i.e., occurring early during tumor evolution) has been proposed as a logical strategy to treat to clear cell tumors, which are known to be in their vast majority VHL deficient (29). Exploitation of the consequential upregulation of HIF2a for therapeutic purposes is a topic of active research. In fact, belzutifan (MK-6482), a small-molecule HIF2a inhibitor, has demonstrated promising antitumor activity and safety in a phase 1/2 trial of previously treated patients with VHL-associated RCC (NCT03401788) and a randomized phase 3 trial comparing belzutifan to everolimus is currently underway (NCT04195750). Significant advances in these areas could represent a paradigm shift in the degree of actionability of RCC. However, additional research would also be needed to determine whether VHL deficiency in itself could constitute a reliable biomarker of response to these agents.

Conclusions

The present study provides important new insights into the landscape and clinical significance of targetable alterations in RCC, representing a first step toward the optimization of personalized cancer therapy while hopefully encouraging further research into biomarkers of response in this context. We demonstrate a substantially lower prevalence of actionable alterations in RCC. Druggable alterations identified were nearly universally subclonal, suggesting that routine clinical sequencing outside of concerted, academic efforts to be assays of low clinical yield. Although some cancers have reaped demonstrable benefits from the current genomic revolution, the same benefits have not been yet observed in kidney cancer, necessitating further efforts to identify the precise clinical role of genomic tumor profiling in this setting.

C.H. Lee reports grants and personal fees from Bristol Myers Squibb; grants and nonfinancial support from Calithera; grants, personal fees, and nonfinancial support from Eisai; grants from Eli Lilly; grants and personal fees from Exelixis, Merck, and Pfizer; personal fees from Amgen, AiCME, Intellisphere, and Research to Practice outside the submitted work. J.C. Durack reports other support from Adient Medical, Verix Medical, and Serpex Medical outside the submitted work. T.A. Chan reports other support from Gritstone Bio and grants from Illumina, Nysnobio, and Pfizer outside the submitted work. R.J. Motzer reports grants and personal fees from Pfizer, Novartis, Exelixis, Genentech; personal fees from Incyte, grants and personal fees from Roche; grants from Bristol Myers Squibb; and personal fees from AstraZeneca, EMD Serome, and Aveo outside the submitted work. M.H. Voss reports grants and personal fees from Pfizer and personal fees from Corvus, Exelixis, Eisai, Calitehrap, Merck, Aveo, and Chengdu outside the submitted work. No disclosures were reported by the other authors.

K. Attalla: Conceptualization, data curation, formal analysis, investigation, writing–original draft. R.G. DiNatale: Conceptualization, data curation, software, formal analysis, investigation, visualization, writing–review and editing. P.M. Rappold: Supervision, writing–review and editing. C.J. Fong: Data curation, software, formal analysis, methodology. F. Sanchez-Vega: Data curation, software, formal analysis, investigation. A.W. Silagy: Resources, data curation, formal analysis. S. Weng: Data curation, formal analysis. J. Coleman: Supervision, project administration. C.-H. Lee: Data curation, formal analysis, supervision. M.I. Carlo: Data curation, supervision, writing–review and editing. J.C. Durack: Resources, data curation. S.B. Solomon: Resources, data curation. V.E. Reuter: Resources, data curation. P. Russo: Resources, data curation, supervision. T.A. Chan: Supervision. R.J. Motzer: Supervision, writing–review and editing. N.D. Schultz: Supervision, writing–review and editing. E. Reznik: Software, formal analysis, methodology. M.H. Voss: Resources, supervision, writing–review and editing. A.A. Hakimi: Resources, supervision, funding acquisition, writing–review and editing.

We thank Charles Swanton, Samra Turajlic, Nicholas McGranahan, Kevin Litchfield, and all the members of the TRACERx consortium for facilitating the mutation data used in the study. We thank the Reznik and Chan Laboratory members for helpful discussions. We thank the staff and physicians of the MSK Department of Medicine Kidney Program and the Urology Service for helpful suggestions. We acknowledge the use of the Integrated Genomics Operation Core, funded by the NIH/NCI Cancer Center Support grant (CCSG, P30 CA008748), Cycle for Survival, and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. This work was in part supported by grants NIH R35 CA232097 and DOD KC180165 (to T.A. Chan), as well as the DOD Kidney Cancer Research Program W81XWH-18–1-0318 and the Kidney Cancer Association Young Investigator Award (to E. Reznik). This work was also supported by the Mellnikoff Fund (to T.A. Chan) and the Weiss Family Fund (to A.A. Hakimi and R.J. Motzer) as well as the Ruth L. Kirschstein National Research Service Award T32CA082088 (to K. Attalla).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Schwaederle
M
,
Zhao
M
,
Lee
JJ
,
Eggermont
AM
,
Schilsky
RL
,
Mendelsohn
J
, et al
.
Impact of precision medicine in diverse cancers: a meta-analysis of phase ii clinical trials
.
J Clin Oncol
2015
;
33
:
3817
25
.
2.
Drilon
A
,
Laetsch
TW
,
Kummar
S
,
DuBois
SG
,
Lassen
UN
,
Demetri
GD
, et al
.
Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children
.
N Engl J Med
2018
;
378
:
731
9
.
3.
Hyman
DM
,
Puzanov
I
,
Subbiah
V
,
Faris
JE
,
Chau
I
,
Blay
J-Y
, et al
.
Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations
.
N Engl J Med
2015
;
373
:
726
36
.
4.
Montemurro
F
,
Valabrega
G
,
Aglietta
M
.
Trastuzumab treatment in breast cancer
.
N Engl J Med.
2006
;
2186
.
5.
Lott
JP
.
Vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
1449
50
.
6.
Chapman
PB
,
Hauschild
A
,
Robert
C
,
Haanen
JB
,
Ascierto
P
,
Larkin
J
, et al
.
Improved survival with vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
364
:
2507
16
.
7.
Siegel
RL
,
Miller
KD
,
Jemal
A
.
Cancer statistics, 2020
.
CA Cancer J Clin
2020
;
70
:
7
30
.
8.
Hakimi
AA
,
Voss
MH
,
Kuo
F
,
Sanchez
A
,
Liu
M
,
Nixon
BG
, et al
.
Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: data from a randomized phase III trial
.
Cancer Discov
2019
;
9
:
510
25
.
9.
McDermott
DF
,
Huseni
MA
,
Atkins
MB
,
Motzer
RJ
,
Rini
BI
,
Escudier
B
, et al
.
Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma
.
Nat Med
2018
;
24
:
749
57
.
10.
Miao
D
,
Margolis
CA
,
Vokes
NI
,
Liu
D
,
Taylor-Weiner
A
,
Wankowicz
SM
, et al
.
Genomic correlates of response to immune-checkpoint blockade in microsatellite-sSupplementary table solid tumors
.
Nat Genet
2018
;
50
:
1271
81
.
11.
Miao
D
,
Margolis
CA
,
Gao
W
,
Voss
MH
,
Li
W
,
Martini
DJ
, et al
.
Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma
.
Science
2018
;
359
:
801
6
.
12.
Hsieh
JJ
,
Chen
D
,
Wang
PI
,
Marker
M
,
Redzematovic
A
,
Chen
Y-B
, et al
.
Genomic biomarkers of a randomized trial comparing first-line everolimus and sunitinib in patients with metastatic renal cell carcinoma
.
Eur Urol
2017
;
71
:
405
14
.
13.
Voss
MH
,
Reising
A
,
Cheng
Y
,
Patel
P
,
Marker
M
,
Kuo
F
, et al
.
Genomically annotated risk model for advanced renal-cell carcinoma: a retrospective cohort study
.
Lancet Oncol
2018
;
19
:
1688
98
.
14.
Havel
JJ
,
Chowell
D
,
Chan
TA
.
The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy
.
Nat Rev Cancer
2019
;
19
:
133
50
.
15.
Cheng
DT
,
Mitchell
TN
,
Zehir
A
,
Shah
RH
,
Benayed
R
,
Syed
A
, et al
.
Memorial sloan kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology
.
J Mol Diagn
2015
;
17
:
251
64
.
16.
Niu
B
,
Ye
K
,
Zhang
Q
,
Lu
C
,
Xie
M
,
McLellan
MD
, et al
.
MSIsensor: microsatellite instability detection using paired tumor-normal sequence data
.
Bioinformatics
2014
;
30
:
1015
6
.
17.
Middha
S
,
Zhang
L
,
Nafa
K
,
Jayakumaran
G
,
Wong
D
,
Kim
HR
, et al
.
Reliable pan-cancer microsatellite instability assessment by using targeted next-generation sequencing data
.
JCO Precis Oncol
2017
;
2017
:
PO.17.00084
.
18.
Mermel
CH
,
Schumacher
SE
,
Hill
B
,
Meyerson
ML
,
Beroukhim
R
,
Getz
G
.
GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers
.
Genome Biol
2011
;
12
:
R41
.
19.
Chakravarty
D
,
Gao
J
,
Phillips
SM
,
Kundra
R
,
Zhang
H
,
Wang
J
, et al
.
OncoKB: a precision oncology knowledge base
.
JCO Precis Oncol
2017
;
2017
:
PO.17.00011
.
20.
Tamborero
D
,
Rubio-Perez
C
,
Deu-Pons
J
,
Schroeder
MP
,
Vivancos
A
,
Rovira
A
, et al
.
Cancer genome interpreter annotates the biological and clinical relevance of tumor alterations
.
Genome Med
2018
;
10
:
25
.
21.
Griffith
M
,
Spies
NC
,
Krysiak
K
,
McMichael
JF
,
Coffman
AC
,
Danos
AM
, et al
.
CIViC is a community knowledgebase for expert crowdsourcing the clinical interpretation of variants in cancer
.
Nat Genet
2017
;
49
:
170
4
.
22.
Shen
R
,
Seshan
VE
.
FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing
.
Nucleic Acids Res
2016
;
44
:
e131
.
23.
Subbiah
V
,
Solit
DB
,
Chan
TA
,
Kurzrock
R
.
The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: a decision centered on empowering patients and their physicians
.
Ann Oncol
2020
;
31
:
1115
8
.
24.
Marcon
J
,
DiNatale
RG
,
Sanchez
A
,
Kotecha
RR
,
Gupta
S
,
Kuo
F
, et al
.
Comprehensive genomic analysis of translocation renal cell carcinoma reveals copy-number variations as drivers of disease progression
.
Clin Cancer Res
2020
;
26
:
3629
40
.
25.
Knox
JJ
,
Barrios
CH
,
Kim
TM
,
Cosgriff
T
,
Srimuninnimit
V
,
Pittman
K
, et al
.
Final overall survival analysis for the phase II RECORD-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic RCC
.
Ann Oncol
2018
;
29
:
2269
.
26.
Motzer
RJ
,
Hutson
TE
,
Cella
D
,
Reeves
J
,
Hawkins
R
,
Guo
J
, et al
.
Pazopanib versus sunitinib in metastatic renal-cell carcinoma
.
N Engl J Med
2013
;
369
:
722
31
.
27.
Pal
SK
,
McDermott
DF
,
Atkins
MB
,
Escudier
B
,
Rini
BI
,
Motzer
RJ
, et al
.
Patient-reported outcomes in a phase 2 study comparing atezolizumab alone or with bevacizumab vs sunitinib in previously untreated metastatic renal cell carcinoma
.
BJU Int
2020
;
126
:
73
82
.
28.
Turajlic
S
,
Xu
H
,
Litchfield
K
,
Rowan
A
,
Horswell
S
,
Chambers
T
, et al
.
Deterministic evolutionary trajectories influence primary tumor growth: TRACERx renal
.
Cell
2018
;
173
:
595
610
.e11.
29.
Cancer Genome Atlas Research Network
,
Linehan
WM
,
Spellman
PT
,
Ricketts
CJ
,
Creighton
CJ
,
Fei
SS
, et al
.
Comprehensive molecular characterization of papillary renal-cell carcinoma
.
N Engl J Med
2016
;
374
:
135
45
.
30.
Davis
CF
,
Ricketts
CJ
,
Wang
M
,
Yang
L
,
Cherniack
AD
,
Shen
H
, et al
.
The somatic genomic landscape of chromophobe renal cell carcinoma
.
Cancer Cell
2014
;
26
:
319
30
.
31.
Cancer Genome Atlas Research Network
.
Comprehensive molecular characterization of clear cell renal cell carcinoma
.
Nature
2013
;
499
:
43
9
.
32.
Carlo
MI
,
Khan
N
,
Zehir
A
,
Patil
S
,
Ged
Y
,
Redzematovic
A
, et al
.
Comprehensive genomic analysis of metastatic non-clear-cell renal cell carcinoma to identify therapeutic targets
.
JCO Precis Oncol
2019
;
3
:
PO.18.00372
.
33.
Schuller
AG
,
Barry
ER
,
Jones
RDO
,
Henry
RE
,
Frigault
MM
,
Beran
G
, et al
.
The met inhibitor AZD6094 (savolitinib, HMPL-504) induces regression in papillary renal cell carcinoma patient-derived xenograft models
.
Clin Cancer Res
2015
;
21
:
2811
9
.
34.
Rochigneux
P
,
Thomassin-Piana
J
,
Laibe
S
,
Brunelle
S
,
Salem
N
,
Escudier
B
, et al
.
Long-term efficacy of crizotinib in a metastatic papillary renal carcinoma with MET amplification: a case report and literature review
.
BMC Cancer
2018
;
18
:
1159
.
35.
Weng
S
,
DiNatale
RG
,
Silagy
A
,
Mano
R
,
Attalla
K
,
Kashani
M
, et al
.
The clinicopathologic and molecular landscape of clear cell papillary renal cell carcinoma: implications in diagnosis and management
.
Eur Urol
2021
;
79
:
468
77
.
36.
Motzer
RJ
,
Escudier
B
,
Oudard
S
,
Hutson
TE
,
Porta
C
,
Bracarda
S
, et al
.
Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors
.
Cancer
2010
;
116
:
4256
65
.
37.
Motzer
RJ
,
Hutson
TE
,
Glen
H
,
Michaelson
MD
,
Molina
A
,
Eisen
T
, et al
.
Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial
.
Lancet Oncol
2015
;
16
:
1473
82
.
38.
Hutson
TE
,
Escudier
B
,
Esteban
E
,
Bjarnason
GA
,
Lim
HY
,
Pittman
KB
, et al
.
Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2014
;
32
:
760
7
.
39.
Voss
MH
,
Hakimi
AA
,
Pham
CG
,
Brannon
AR
,
Chen
Y-B
,
Cunha
LF
, et al
.
Tumor genetic analyses of patients with metastatic renal cell carcinoma and extended benefit from mTOR inhibitor therapy
.
Clin Cancer Res
2014
;
20
:
1955
64
.
40.
Voss
MH
,
Chen
D
,
Reising
A
,
Marker
M
,
Shi
J
,
Xu
J
, et al
.
PTEN Expression, not mutation status in, or, correlates with the outcome on everolimus in patients with renal cell carcinoma treated on the randomized RECORD-3 trial
.
Clin Cancer Res
2019
;
25
:
506
14
.
41.
Nassar
AH
,
Hamieh
L
,
Gray
KP
,
Thorner
AR
,
Fay
AP
,
Lasseter
KD
, et al
.
Mutations and response to rapalogs in patients with metastatic renal cell carcinoma
.
Mol Cancer Ther
2020
;
19
:
690
6
.
42.
Ionov
Y
,
Peinado
MA
,
Malkhosyan
S
,
Shibata
D
,
Perucho
M
.
Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis
.
Nature
1993
;
363
:
558
61
.
43.
Asaoka
Y
,
Ijichi
H
,
Koike
K
.
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
373
:
1979
.
44.
Altavilla
G
,
Fassan
M
,
Busatto
G
,
Orsolan
M
,
Giacomelli
L
.
Microsatellite instability and hMLH1 and hMSH2 expression in renal tumors
.
Oncol Rep
2010
;
24
:
927
32
.
45.
Stoehr
C
,
Burger
M
,
Stoehr
R
,
Bertz
S
,
Ruemmele
P
,
Hofstaedter
F
, et al
.
Mismatch repair proteins hMLH1 and hMSH2 are differently expressed in the three main subtypes of sporadic renal cell carcinoma
.
Pathobiology
2012
;
79
:
162
8
.
46.
Cortes-Ciriano
I
,
Lee
S
,
Park
W-Y
,
Kim
T-M
,
Park
PJ
.
A molecular portrait of microsatellite instability across multiple cancers
.
Nat Commun
2017
;
8
:
15180
.
47.
Wu
H-X
,
Wang
Z-X
,
Zhao
Q
,
Chen
D-L
,
He
M-M
,
Yang
L-P
, et al
.
Tumor mutational and indel burden: a systematic pan-cancer evaluation as prognostic biomarkers
.
Ann Transl Med
2019
;
7
:
640
.
48.
Samstein
RM
,
Lee
C-H
,
Shoushtari
AN
,
Hellmann
MD
,
Shen
R
,
Janjigian
YY
, et al
.
Tumor mutational load predicts survival after immunotherapy across multiple cancer types
.
Nat Genet
2019
;
51
:
202
6
.
49.
Heydt
C
,
Rehker
J
,
Pappesch
R
,
Buhl
T
,
Ball
M
,
Siebolts
U
, et al
.
Analysis of tumor mutational burden: correlation of five large gene panels with whole exome sequencing
.
Sci Rep
2020
;
10
:
11387
.
50.
Bevins
N
,
Sun
S
,
Gaieb
Z
,
Thorson
JA
,
Murray
SS
.
Comparison of commonly used solid tumor targeted gene sequencing panels for estimating tumor mutation burden shows analytical and prognostic concordance within the cancer genome atlas cohort
.
J Immunother Cancer
2020
;
8
:
e000613
.
51.
Herman
JG
,
Umar
A
,
Polyak
K
,
Graff
JR
,
Ahuja
N
,
Issa
JP
, et al
.
Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma
.
Proc Natl Acad Sci U S A
1998
;
95
:
6870
5
.
52.
Ged
Y
,
Chaim
JL
,
DiNatale
RG
,
Knezevic
A
,
Kotecha
RR
,
Carlo
MI
, et al
.
DNA damage repair pathway alterations in metastatic clear cell renal cell carcinoma and implications on systemic therapy
.
J Immunother Cancer
2020
;
8
:
e000230
.
53.
Kapur
P
,
Peña-Llopis
S
,
Christie
A
,
Zhrebker
L
,
Pavía-Jiménez
A
,
Rathmell
WK
, et al
.
Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation
.
Lancet Oncol
2013
;
14
:
159
67
.
54.
Tannock
IF
,
Hickman
JA
.
Limits to personalized cancer medicine
.
New Engl J Med
2016
;
375
:
1289
94
.
55.
Linehan
WM
,
Lerman
MI
,
Zbar
B
.
Identification of the von Hippel-Lindau (VHL) gene. Its role in renal cancer
.
JAMA
1995
;
273
:
564
70
.
56.
Gerlinger
M
,
Rowan
AJ
,
Horswell
S
,
Math
M
,
Larkin
J
,
Endesfelder
D
, et al
.
Intratumor heterogeneity and branched evolution revealed by multiregion sequencing
.
N Engl J Med
2012
;
366
:
883
92
.
57.
Turajlic
S
,
Sottoriva
A
,
Graham
T
,
Swanton
C
.
Resolving genetic heterogeneity in cancer
.
Nat Rev Genet
2019
;
20
:
404
16
.
58.
Becerra
MF
,
Reznik
E
,
Redzematovic
A
,
Tennenbaum
DM
,
Kashan
M
,
Ghanaat
M
, et al
.
Comparative genomic profiling of matched primary and metastatic tumors in renal cell carcinoma
.
Eur Urol Focus
2018
;
4
:
986
94
.
59.
Wagner
AH
,
Walsh
B
,
Mayfield
G
,
Tamborero
D
,
Sonkin
D
,
Krysiak
K
, et al
.
A harmonized meta-knowledgebase of clinical interpretations of somatic genomic variants in cancer
.
Nat Genet
2020
;
52
:
448
57
.

Supplementary data