Abstract
The genetics of nephroblastoma (Wilms tumor) occurring in adults is largely unknown, as studies have largely been limited to isolated case reports. We, therefore, studied 14 adult Wilms tumors for genetic alterations, using expanded targeted sequencing on 11 cases. The patients ranged from 17 to 46 years of age (mean and median, 31 y), and there were 8 males and 6 females. Five Wilms tumors harbored BRAF V600E mutations. All of these had better-differentiated areas identical to metanephric adenoma, as has previously been described. In 3 such cases, microdissection studies revealed that the BRAF V600E mutation was present in both the metanephric adenoma and Wilms tumor areas; however, additional genetic alterations (including TERT promoter mutations in 2 cases, ASLX1/ATR mutations in 1 other case) were limited to the Wilms tumor component. These findings suggest that the Wilms tumor developed from the metanephric adenoma. Other adult Wilms tumors harbored genetic alterations previously reported in the more common pediatric Wilms tumors, including WT1 mutations (2 cases), ASLX1 mutations (3 additional cases), NSD2 mutation (1 additional case), and 11p loss (3 cases). In summary, a significant subset of adult Wilms tumors (specifically those of epithelial type with differentiated areas) harbor targetable BRAF V600E mutations and appear to arise from metanephric adenomas as a consequence of additional acquired genetic alterations. Other adult Wilms tumors often harbor genetic alterations found in their more common pediatric counterparts, suggesting at least some similarities in their pathogenesis.
Keywords: renal neoplasm, BRAF, metanephric, Wilms
Approximately 3% of Wilms tumor (nephroblastoma) occur in adults,1 and these have long been problematic for oncologists. For many years, the general consensus has been that adult Wilms tumors are more aggressive than their pediatric counterparts.2–6 However, several factors could account for this difference. First, adult Wilms may not be treated as expeditiously and appropriately as pediatric cases. There is frequently a delay in diagnosis of this typically pediatric tumor in adults, where it comprises <1% of all renal tumors.7 Moreover, adults with Wilms tumor are often not treated on standardized protocols developed by the National Wilms Tumor Study Group (now COG) or the Société International d’Oncologie Pédiatrique (SIOP). Along these lines, treatment of adult Wilms tumors according to standardized pediatric protocols has been shown to improve outcomes.7–10 Second, adult Wilms tumors have a greater tendency to present at an advanced stage relative to pediatric cases, perhaps reflecting the delay in seeking medical attention. Third, unlike pediatric Wilms tumors, adult Wilms tumors have not been shown to arise in association with nephrogenic rests or be shown to be associated with developmental conditions associated with Wilms tumor, potentially reflecting differing biology. Fourth, other recently described more aggressive lesions (such as renal Ewing sarcoma11,12 or synovial sarcoma13) have historically been misdiagnosed as adult Wilms tumor, contributing to the observed poor outcome.
The genetics of adult Wilms tumors has remained largely unstudied, limited to isolated case reports frequently assessing only a few genetic alterations.14 Along these lines, in a recent study, our group found that a subset of Wilms tumors with areas morphologically resembling metanephric adenoma harbored BRAF V600E mutations that are typical of metanephric adenoma15 and not previously found in Wilms tumor.16 One pediatric patient with metastatic disease had a complete response to combined targeted RAF/MEK inhibitors dabrafenib and trametinib.16,17 While these cases were identified based on morphologic features, 2 of the 4 cases occurred in adults, raising the possibility that BRAF V600E mutations may be more frequent in adult Wilms tumors. In the current study, we perform a more comprehensive genetic analysis of 14 adult Wilms tumors, including expanded targeted sequencing of 11 of these cases.
MATERIALS AND METHODS
Institutional Review Board Approval
This study was approved by the Institutional Review Boards at our respective institutions.
Cases
We identified all cases of nephroblastoma (Wilms tumor) in patients at or above the age of 17 years in the consultation and teaching files of 2 authors (P.A., J.I.E.) at Johns Hopkins Hospital and the institutional files of another hospital (Memorial Sloan-Kettering Cancer Center). We acknowledge that patients aged 17 to 20 could be considered older adolescents, but they fall far outside the typical age range for Wilms tumor (ages 3 to 6 y), and this age group has been included in prior adult Wilms tumor cohorts.3–6 Cases included in our prior study16 were excluded. None of the cases in this study have previously been published. Cases were reviewed by one author (P.A.) to verify the diagnosis of Wilms tumor, and cases were included in the study if additional formalin-fixed paraffin-embedded (FFPE) material was available for genetic analysis.
Molecular Techniques
DNA and RNA sequencing were performed by the Genomics and Molecular Pathology Core at UT Southwestern Medical Center on all cases except cases 5, 6, and 12. Briefly, tumor hematoxylin and eosin slides were examined and marked by 2 pathologists (P.A. and D.N.P.) for subsequent macrodissection, nucleic acid isolation, and molecular testing. Areas enriched with tumor were then scraped from adjacent 5-μm thick FFPE sections. Adjacent normal tissue was separately isolated and processed for select cases when available. Extraction and purification were performed using Qiagen Allprep kits (Qiagen, Germantown, MD). Libraries were prepared using KAPA Hyperplus kits (Roche Sequencing and Life Science Kapa Biosystems, Wilmington, MA) with genomic regions of interest captured by custom DNA probes covering all exons of over 1425 cancer-related genes. The libraries were sequenced to an average unique read depth of > ×600 (95% of exons at or above ×100) using Sequencing by Synthesis (SBS) paired-end cluster generation on the Illumina NextSeq. 550 platform (Illumina Inc., San Diego, CA). Sequence reads were aligned to reference genome GRCh38, and subsequent analyses were performed using custom germline, somatic, and mRNA bioinformatics pipelines run on the UTSW BioHigh Performance Computer cluster and optimized for detection of single nucleotide variants, indels, and known gene fusions. Minor allele frequency limit of detection for the assay: single nucleotide variants—5%; indels and known gene fusions—10%. Variants in exons with <×100 coverage are not reported.
Somatic variants were identified on the basis of their variant allele frequencies and relative absence in matched normal tissue (if available) and/or presence at frequencies below 0.5% in databases of germline variants (dbSNP, gnomAD). Variants were classified according to the Association for Molecular Pathology/American Society for Clinical Oncology/College of American Pathologists guidelines,18 and all variant calls were inspected using Integrated Genomics Viewer, version 2.3.4 (IGV; Broad Institute, MIT Harvard, Cambridge, MA) before reporting.
Copy number variants were detected using CNVKit and an internally derived panel of normal FFPE tissue samples.
Adequate tissue for expanded targeted sequencing was not available for cases 5, 6, and 12. For these cases, BRAF mutation status alone was assessed by using pyrosequencing to detect mutations at codons 600 and 601 or AmpliSeq Cancer Hotspot Panel (v2) (Life Technologies, Carlsbad, CA) to detect mutations within exons 11 and 15 as described previously.19,20
RESULTS
Cases
The clinicopathologic and genetic features of the cases are summarized in Table 1. The patients ranged from 17 to 46 years of age (mean and median, 31 y), and there were 8 males and 6 females. Tumors were generally large—sizes ranged from 2.5 to 23 cm (mean, 11.8 cm; median, 11 cm)—and often presented at an advanced stage. Of 13 cases with staging information, 6 were stage 1, 2 were stage 2, 3 were stage 3, and 2 were stage 4.
TABLE 1.
Adult Wilms Tumor Cases in This Study
| Case No. | Age/Sex | Size/Stage | Histology | IHC | Genetic Alterations of Interest |
|---|---|---|---|---|---|
|
| |||||
| 1 | 38/F | 17 cm/stage 1 | Epithelial-predominant triphasic with MA-like areas, rim of calcified MA; hyaline calcified nodule | WT1+, EMA−, CK7− | MA and WT: BRAF V600E, NOTCH1 G842del, STYK1 T100fs WT only: TERT c.-124G- > A, LTBP N1187fs, WHSC1(NSD2) C846Y, 1q gain (MDM4), 17q loss (BRCA1) |
| 2 | 30/F | 3 cm/stage 1 | Epithelial predominant with MA-like areas; hyaline calcified nodule | WT1+ | MA and WT areas: BRAF V600E WT only: TERT c.-124G- > A, CREB1 R284* |
| 3 | 46/M | 6 cm/stage 2 | Epithelial predominant with MA-like areas | WT1+ CK7− | MA and WT areas: BRAF V600E WT only: ATR c.2078+1G> A, ASXL1 G660fs |
| 4 | 44/F | 11 cm/stage 1 | Epithelial predominant with MA-like areas; hyaline calcified nodule | WT1+, PAX8+; CK7− | BRAF V600E, CTCF T400fs, EZH2 R313Q, 1q gain (MDM4), 12p/q gain (KRAS, GLI1, CDK4, MDM2) |
| 5 | 40/M | 2.5 cm/stage 1 | Epithelial predominant with MA-like areas; hyaline calcified nodule | WT1+ | BRAF V600E+ |
| 6 | 35/F | NA | Epithelial | WT1+, PAX8+; EMA− | BRAF − |
| 7 | 32/M | 11 cm/stage 3 | Epithelial predominant, not anaplastic | PAX8+ WT1− | TP53 R175H, BCOR N1459S, BCORL1 R1266*, ASXL1 c.1086–2A > T, KRAS G12V, ERBB3 S1094*, 4p loss (WHSC1/NSD2), 11p LOH (WT1), 12p gain (KRAS), 22p/q loss (BRD1, CHEK2, SMARCB1) |
| 8 | 35/M | 23 cm/stage 3 | Blastemal predominant, triphasic | WT1+ | BRCA1 R1720W, TBX3 E255fs, ASXL1 R596fs, MELK Q67*, 2p/q gain (ASXLX2, ERBB4, GLI2, MYCN), 3q gain (TERC, TP63), 7p/q gain (BRAF, MET), 8p/q gain (MYC, TERF1, WHSC1L1), 12p/q gain (KRAS, GLI1, CDK4, MDM2, TBX3), 16p/q gain (TERF2), 20p/q gain (ASXL1) |
| 9 | 17/M | 19 cm/stage 1 | Epithelial predominant, serpentine, and nodular blastema | WT1+ | 4p loss (WHSC1/NSD2); 11p loss (WT1); 8p loss (WHSC1L1); 3p loss (BAP1); 16p/q loss (CTCF, FANCA); 1p loss (MTOR); 19p gain (BRD4), 19q loss (POLD1) |
| 10 | 20/F | 17 cm/stage 4 (lung metastasis) | Blastemal predominant, nodular | WT1+ | 1q loss, 9q loss (FANCC, PTCH1, XPA), 10p/q loss (RET), 11p loss (CENPU) |
| 11 | 19/M | 14 cm/stage 2 | Epithelial-blastemal | WT1+ | ATM R248*, WHSC1(NSD2) E1099K, KAT6B W606fs, BIRC6 L2121F |
| 12 | 28/F | Stage 4 (brain metastasis) | Epithelial predominant | WT1+ | BRAF − |
| 13 | 23/M | 10 cm/stage 3 | Triphasic with skeletal muscle but prominent diffuse blastemal pattern | WT1+, EMA− | WT1 H235fs, ASXL1 G645fs, CCDC88C L47fs, CHD2 L1018R, KMTD2 L389fs, 1q gain (MDM4), 2p/q gain (ASXL2, ERBB4, GLI2, MYCN), 12p/q gain (KRAS, GLI1, CDK4, MDM2) |
| 14 | 29/M | 8.5 cm/stage 1 | Blastemal-epithelial | WT1+ | WT1 D436Y, SPOP R354C, KMT2A T2230I |
Bolded genes are of special interest.
Stop codon.
F indicates female; fs, frameshift; IHC, immunohistochemistry; M, male; MA, metanephric adenoma; NA, not applicable.
Histologic Features
Among the 14 cases, 9 were epithelial predominant, 3 were blastemal predominant, and 2 were an even mixture of epithelial and blastemal elements. There were no stromal predominant neoplasms. By immunohistochemistry, 13 of the 14 Wilms tumors were immunoreactive for WT1 protein. The background kidney demonstrated no specific abnormities. Nephrogenic rests were not identified in the surrounding renal parenchyma in any case.
Genetics
Overall, 5 Wilms tumors harbored BRAF V600E mutations. Other adult Wilms tumors harbored genetic alterations previously reported in the more common pediatric Wilms tumors, including WT1 mutations (2 cases), ASLX1 mutations (3 additional cases not including one of the BRAF V600E-mutated cases), NSD2 mutation (1 additional case not including one of the BRAF V600E-mutated cases), and 11p loss (3 cases) (Fig. 1). Comprehensive data is presented in Supplementary Table 1 (Supplemental Digital Content 1, http://links.lww.com/PAS/B319).21–41
FIGURE 1.

Next-generation sequencing of adult Wilms tumors. A, Epithelial-predominant Wilms tumor with metanephric adenoma (MA)-like areas (cases 1 to 4) consistently harbored BRAF V600E mutations in both the MA and Wilms tumor components (cases 1 to 3) with additional genomic alterations involving Wnt and/or TGFβ signaling, telomere maintenance, DNA damage response, apoptosis, or histone modification/chromatin remodeling/DNA methylation in the Wilms tumor areas. The remaining adult Wilms tumor cases without identifiable MA-like areas (cases 7 to 11, 13 to 14) demonstrated genetic alterations in similar pathways as well as receptor-mediated signaling associated with receptor tyrosine kinases. B, Large chromosomal alterations were detected in most but not all cases of adult Wilms tumor. Both chromosomal gains, particularly involving 1q and 12p/q, and losses were noted. †Matched normal not sequenced. ‡Also detected in MA-like area. Note: WHSC1 is also known as NSD2. CN indicates copy number; LOH, loss of heterozygosity; MA, metanephric adenoma.
Correlation of Histologic Features With Genetics
As previously found by our group,16 the BRAF V600E-mutated Wilms tumors all had distinctive histology—in addition to epithelial-predominant Wilms tumor areas featuring mitotically active columnar-shaped cells with primitive (finely dispersed but hyperchromatic) chromatin, there was a mitotically inactive differentiated tubular component associated with calcifications that is typical of metanephric adenoma (Figs. 2–4). Four of the 5 BRAF V600E-mutated cases had large hyalinized nodules associated with calcification adjacent to the metanephric adenoma component. In 3 of these cases, the Wilms tumor area formed a discrete expansile nodule, such that it could be dissected apart from the metanephric adenoma areas and the 2 components sequenced separately. In each case, both components harbored the BRAF V600E mutation. However, the Wilms tumor component from each case harbored additional genetic alterations not found in the metanephric adenoma component. There were additional genomic alterations involving Wnt and/or TGFβ signaling, telomere maintenance, DNA damage response, apoptosis, or histone modification/chromatin remodeling/DNA methylation. Of note, in cases 1 and 2, the Wilms tumor component had an additional TERT promoter mutation. In case 3, ATR and ASXL1 mutations were identified only in the Wilms tumor areas. Analysis of telomere lengths in the 2 neoplasms with TERT mutations revealed short telomeres in both the metanephric adenoma and Wilms tumor components (Supplemental Fig. 1, Supplemental Digital Content 2, http://links.lww.com/PAS/B320). No difference between the 2 components could be discerned.
FIGURE 2.

Case 1: Wilms tumor arising from metanephric adenoma. This whole-mount slide section demonstrates a nodule of epithelial-predominant Wilms tumor (right) arising in the background of a metanephric adenoma (bottom) associated with extensive sclerosis (left) (A). At intermediate power, the transition between the differentiated metanephric adenoma (left) and the epithelial Wilms tumor (right) is sharp (B). At higher magnification, one can appreciate the mitotically inactive metanephric adenoma cells (left) and the mitotically active columnar-shaped epithelial Wilms tumor cells (right) (C). The metanephric adenoma areas have the typical solid and focally papillary architecture of metanephric adenoma, set in the hyalinized stroma (D). At high power, the metanephric adenoma cells have minimal cytoplasm and show no mitotic activity (E). In contrast, the Wilms tumor areas have foci of stroma in addition to the epithelial areas and are associated with necrosis (lower left) (F). At high power, the columnar epithelial cells demonstrate mitotic activity (G). In this case, while both areas harbored BRAF V600E mutations, the Wilms tumor area demonstrated TERT promoter and WHSC1(NSD2) mutations (the latter 2 not found in the metanephric adenoma areas).
FIGURE 4.

Case 3: Wilms tumor arising from metanephric adenoma. This whole-mount section shows a primitive neoplasm which abuts the renal sinus of the native kidney at the left (A). One can appreciate the nodule of epithelial Wilms tumor (white star) separated by a capsule from the adjacent metanephric adenoma area (black star) (A). The metanephric adenoma area abuts the cortical type parenchyma adjacent to the renal pelvic urothelium and demonstrates the typical un-encapsulated border and intermingling with native nephrons (B). At higher power, the primitive metanephric adenoma cells are mitotically inactive and composed of small tubules which touch the adjacent non-neoplastic renal tubules (C). The sclerotic metanephric adenoma area (left) transitions into the nodule of epithelial-predominant Wilms tumor (right) (D). High magnification of the epithelial Wilms tumor area demonstrates tubules formed by cells with primitive columnar nuclei showing mitotic activity (E). In this case, while both areas harbored BRAF V600E mutations, the epithelial Wilms tumor demonstrated ASXL1 and ATR mutations (which were not found in the metanephric adenoma area).
The remaining adult Wilms tumors demonstrated the typical morphology seen in their pediatric counterparts (Fig. 5). They harbored genomic alterations involving Wnt and/or TGFβ signaling, telomere maintenance, DNA damage response, apoptosis, histone modification/chromatin remodeling/DNA methylation, as well as receptor-mediated signaling associated with receptor tyrosine kinases. In case 13, a triphasic neoplasm with a prominent diffuse blastemal component, the diffuse blastemal component demonstrated striking angioinvasion within the renal parenchyma and a positive renal vein margin. This neoplasm harbored a WT1 mutation. Cases 9 and 10 had prominent nodular blastemal patterns typically seen in pediatric Wilms tumor, and both demonstrated loss of portions of chromosome 11p. The TP53-mutated Wilms tumor (case 7) was epithelial predominant but did not demonstrate anaplasia. The BRCA1-mutated Wilms tumor (case 8) demonstrated the typical triphasic morphology of the Wilms tumor.
FIGURE 5.

Additional adult Wilms tumors associated with mutations in genes commonly altered in pediatric Wilms tumor. Case 13, Adult Wilms tumor with WT1 mutation. This neoplasm had a predominant blastemal pattern and permeated native nephrons (A). Focal areas of well-formed tubules were present in addition to spindled stroma (B). Case 14, Adult Wilms tumor associated with WT1 mutation. This neoplasm was predominantly composed of blastemal cells and primitive tubules (C). The neoplasm demonstrated diffuse nuclear labeling for WT1 (D). Case 7, Adult Wilms tumor associated with TP53, BCOR, BCORL1, ASXL1, and KRAS mutations and 11p loss of heterozygosity. This favorable histology neoplasm was predominantly blastemal and epithelial and separated from the kidney by a fibrous capsule (E). The neoplasm demonstrates the typical primitive round cell blastemal and tubular patterns of Wilms tumor (F). Case 8, Adult Wilms tumor associated with a BRCA1 mutation. The neoplasm demonstrates triphasic morphology with blastemal, stromal, and epithelial elements (G). At high power, the primitive cytology of the 3 elements is typical of Wilms tumor (H).
DISCUSSION
Accumulated data now show that adult Wilms tumors differ in many respects from their pediatric counterparts. First, adult Wilms tumors are rarely associated with developmental syndromes such as the WAGR (Wilms, aniridia, genitourinary malformations, and intellectual disability), Denys-Drash (Wilms tumor, glomerular disease, gonadal dysgenesis), or Beckwith Wiedemann (overgrowth and predisposition to neoplasia) syndromes. One adult Wilms tumor patient has been reported to have a germline mutation in WT1, but no clear evidence of a syndrome was reported.42 One other case was reported in a patient with cryptorchidism and hypospadias, but genetic testing was not performed.43 In contrast, ~15% of pediatric Wilms tumors are syndromic. Second, unlike pediatric Wilms tumors, adult Wilms tumors have not been shown to arise from nephrogenic rests, the established precursor of Wilms tumors.1,3,5,6,9,10,44 While most studies of adult Wilms tumors have not mentioned nephrogenic rests specifically, there have been no cases reported of adult Wilms tumor arising in association with nephrogenic rests, which is consistent with our findings in this study. This also fits data on bilateral Wilms tumors, which are consistently associated with nephrogenic rests. Only one in over 200 adult Wilms tumors reported in large studies has been bilateral, while ~6% of pediatric Wilms tumors are bilateral (Supplemental Table 2, Supplemental Digital Content 3, http://links.lww.com/PAS/B321). This difference has been corroborated by the SEER database and is statistically significant.4 Third, adult Wilms tumors have been associated with diminished prognosis relative to pediatric cases, in large part due to a more frequent presentation at an advanced stage, delay in therapy, and use of nonstandardized therapeutic protocols. Along these lines, the prognosis for Wilms tumor in adults is improved when cases are treated according to pediatric protocols.
The most common genetic alterations/dysregulated pathways in pediatric Wilms tumor include IGF2 overexpression, WNT pathway alterations (including WT1 and CTNNB1 mutations), and microRNA processor mutations. Whether the genomics of adult Wilms tumor is distinct from its more common pediatric counterpart is not currently known. Studies have generally been limited to a few cases assessing only a few genetic alterations. One study did not find β-catenin mutations in 4 adult Wilms tumors, whereas β-catenin mutations were found in 15% of pediatric Wilms tumors.45 A single nucleotide polymorphism array of a single Wilms tumor in a 71-year-old female showed greater genomic complexity than typically seen in a pediatric Wilms tumor.46 Hence, the purpose of our study was to address this issue.
We found that the majority of adult Wilms tumor cases harbored genetic alterations which are well-known to be associated with pediatric Wilms tumors.47,48 These included mutations in WT1 and loss of heterozygosity at chromosome 11p, which are among the more common changes found in pediatric Wilms tumors. We also found recurrent mutations in genes less commonly associated with pediatric Wilms tumor, such as the ASXL transcriptional regulator one (ASXL1) gene.49,50 The latter is one of the more commonly mutated genes in clonal hematopoiesis, myeloid malignancies, and small cell lung carcinoma and is a known epigenetic regulator which binds to BAP1. Mutations in ASXL1 give rise to the Bohring-Opitz syndrome, a rare developmental condition associated with facial abnormalities, unusual posture, intellectual disability, and an association with Wilms tumor.51,52 We additionally identified in 2 cases mutations in the NSD2 gene (also widely known as WHSC1), which has previously been shown to be deleted or amplified in <2% of pediatric Wilms tumors but not mutated.47,48 The NSD2 gene encodes the NSD2 histone lysine methyltransferase, a regulator of development which methylates the H3K36 residue of histones to promote an open chromatin state that favors gene transcription. NSD2 mutations are found in 5% to 10% of several common adult cancers such as colonic or gastric adenocarcinoma, urothelial carcinoma, and melanoma, but far less commonly in pediatric solid tumors.53,54 Mutations in the transcriptional corepressors BCOR and BCORL1, which we found in one of our adult cases, have also been found in pediatric Wilms tumor.55 Hence, a significant proportion of adult Wilms tumors have molecular alterations similar to their pediatric counterparts, suggesting that they may be driven by similar molecular pathways. One limitation of our study is that we used targeted sequencing of 1425 cancer-related genes rather than whole-genome sequencing, so not all genetic alterations would be discovered. However, our platform is better suited to FFPE tissues of variable age, in large part due to the depth of sequencing.
We also identified a high proportion of BRAF V600E mutations in our adult Wilms tumor cohort (5/14, or 36%). These neoplasms had a morphology similar to that reported in our original study describing BRAF V600E-mutated Wilms tumors – the neoplasms contained areas morphologically identical to differentiated metanephric adenoma (which characteristically harbors BRAF V600E mutations) in addition to more proliferative, overtly malignant areas which are morphologically identical to Wilms tumor.16 Of note, all 5 neoplasms with BRAF V600E mutations in this cohort presented with localized disease (stages 1 or 2), while only 2 of the 8 other cases did so. We previously reported the BRAF V600E mutation in 4 metanephric adenoma-epithelial Wilms tumor overlap lesions, 2 of which were pediatric cases and 2 of which were adult cases. The skewing toward adult cases in this small morphologically selected cohort suggested the possibility that BRAF V600E-mutated Wilms tumors might be more prevalent in the adult population, and our current study appears to support this hypothesis. One confounding factor is that most of our cases were derived from our consultation files, so it is likely that the adult Wilms tumor cases we studied were skewed towards those with unusual morphology (such as having metanephric adenoma-like areas). In fact, 2 of the 5 adult BRAF-mutated cases were sent to us due to morphologic overlap with cases in our prior study.16 Removing these cases from consideration, the frequency of BRAF-mutated Wilms tumors in adults in our study would be 3 of 12 or 25%. As we and others have previously described, identification of BRAF V600E mutations in Wilms tumors has therapeutic significance, as these tumors may be treated with highly effective BRAF/MEK inhibitor therapy. In addition to the child we reported with metastatic disease who had a durable complete response, another recent report described a 51-year-old male with relapsed metastatic adult Wilms tumor harboring a BRAF V600E mutation who had a prolonged and dramatic response to the BRAF inhibitor dabrafenib.56 While the histology was not illustrated, the tumor was described as predominantly epithelial and, in fact, was initially misdiagnosed as papillary renal cell carcinoma (RCC), suggesting that it was a lesion similar to those we described herein and previously.16
Interestingly, one prior genetic study analyzing the genomics of adult Wilms tumor identified isochromosome 17q as a recurrent finding in 3 of 5 cases.57 Since the BRAF gene maps to this chromosome, one could hypothesize that these neoplasms harbored BRAF V600E mutations which were amplified by the isochromosome 17q. Unfortunately, BRAF sequencing was not performed on these neoplasms, and the illustrated morphology does not address whether these lesions harbored a differentiated metanephric adenoma-like component.
Given the known association of metanephric adenoma-like areas with Wilms tumor and their common underlying BRAF V600E mutation, 2 hypotheses could explain the findings. The first possibility is that epithelial-predominant BRAF V600E-mutated Wilms tumors focally differentiate to resemble metanephric adenoma. This pathway has always seemed most likely, given that preoperative treatment is known to induce some epithelial Wilms tumors to differentiate and resemble metanephric adenoma58 and treated differentiated stromal Wilms tumors can mimic metanephric stromal tumors.59,60 Another related precedent is that chemotherapy can induce another embryonal neoplasm of childhood, neuroblastoma, to differentiate and resemble ganglioneuroma. The second possibility, for which there has previously been no support, is that a BRAF V600E-mutated metanephric adenoma could undergo malignant transformation to become a Wilms tumor. While there is no clinical precedent for this, as metanephric adenomas are clinically benign, it should be noted that almost all metanephric adenomas are completely excised by partial or complete nephrectomy, so their untreated natural history is in fact not known. Our study provides molecular evidence which supports the hypotheses that BRAF V600E-mutated metanephric adenomas may progress to Wilms tumors, as the Wilms tumors areas of all 3 neoplasms in which microdissection could be performed harbored not only BRAF V600E mutations but also additional genomic alterations not found in the metanephric adenoma component. These findings are compatible with those reported by Chami et al,61 who found the same BRAF V600E mutation in the metanephric adenoma and papillary renal cell components of a composite metanephric adenofibroma-papillary RCC case. It is more logical to conceptualize this case as the transformation of a component of an underlying BRAF V600E differentiated composite metanephric neoplasm into a malignancy (papillary RCC) than to think of it as differentiation of papillary RCC (which is characterized by multiple distinctive genetic alterations) into metanephric adenoma. The molecular findings in our study are also supported by morphology, as the expansile nodules of Wilms tumors as illustrated in Figures 2–4 are more consistent with Wilms tumors arising from metanephric adenoma than a Wilms tumor demonstrating maturation, in which foci of maturation would be predicted to be more randomly dispersed.
Interestingly, 2 of our combined Wilms tumor-metanephric adenoma cases had TERT promoter mutations limited to the Wilms tumor component. High telomerase RNA expression has been associated with poor prognoses in Wilms tumor,62 and TERT promoter mutations are thought to synergize with BRAF alterations in thyroid cancer to promote an aggressive phenotype.63 Correlating with these findings, we did find shortening of telomeres in the Wilms tumors associated with these secondary TERT promoter mutations as well as in the metanephric adenoma components. This is consistent with a proposed model in which TERT promoter mutations do not prevent bulk telomere length shortening in neoplasms but instead restore the length of critically shortened telomeres allowing these cells to survive, thereby promoting genetic instability.64 The short telomere length of the metanephric adenoma component also matches the observation that most precancerous lesions have short telomeres but are telomerase negative.65–67
While the concept of progression of metanephric adenoma to Wilms tumor proposed in our study is a novel one, we note that Chan et al68 recently reported a single BRAF V600E-mutated metanephric adenoma associated with sarcomatous transformation. The sarcoma lacked morphologic features of the Wilms tumor, and both the metanephric adenoma and sarcomatous components had BRAF, EIF1AX, and TERT promoter mutations. However, the sarcomatous component contained a deep deletion of CDKN2A (which encodes for p16) and MYC amplification, while the metanephric adenoma component lacked these alterations. We did not identify loss of p16 protein immunoreactivity in the Wilms tumor components of our composite neoplasms (not shown). However, taken together with our study, these findings suggest that metanephric adenomas have some potential for malignant transformation and should be excised if possible and that if not, there should be active surveillance of such lesions. This risk would seem to be low since small foci of Wilms tumor are not commonly seen in large metanephric adenomas. Finally, we note that recent single-cell sequencing-based analyses of Wilms tumor have revealed a distinct cellular origin with transcriptional profiles overlapping with aberrant nephrogenesis; whether adult Wilms tumor display such evolutionary phenotypes is likely to be a subject of future studies.69
Finally, given the absence of association with nephrogenic rests and association with metanephric adenoma, one could question whether BRAF-mutated Wilms tumors should be considered Wilms tumors at all or, instead, a separate entity such as “malignant metanephric adenoma.” We believe that these cases should be considered adult Wilms tumors, given their morphologic identity to pediatric epithelial-predominant Wilms tumors, the presence of blastemal, and stroma in selected cases (as illustrated in Figs. 2F–G of this manuscript and figure 7 of reference 16), and shared WT1 immunoreactivity. It is well-known that Wilms tumors arise by diverse genetic pathways, such as IGF2 overexpression or WT1 mutation/CTNNB1 mutation.55 In our view, BRAF mutation represents another pathway which leads to the phenotype of Wilms tumor.
In conclusion, adult Wilms tumors harbor some of the same genetic alterations found in their more common pediatric counterparts. A higher percentage harbor BRAF V600E mutations and appear to arise from metanephric adenomas.
Supplementary Material
FIGURE 3.

Case 2: Wilms tumor arising from metanephric adenoma. In the whole-mount section, the nodule of epithelial-predominant Wilms tumor is surrounded by a rim of adjacent metanephric adenoma, most prominent to the left (A). The transition of the calcified metanephric adenoma, which abuts the native kidney, and the epithelial Wilms tumor is abrupt and associated with a fibrous capsule (B). At high power, the metanephric adenoma demonstrates the typical small tubules associated with psammomatous calcifications which directly abut the native nephrons (C, D). At high power, the epithelial Wilms tumor demonstrates elongated tubules formed by nuclei with primitive columnar nuclei, associated with prominent mitotic activity (E). In this case, while both areas harbored BRAF V600E mutations, the Wilms tumor area demonstrated an additional TERT promoter mutation not found in the metanephric adenoma area.
ACKNOWLEDGMENTS
The authors thank Norman Barker, MA, MS, RBP, for expert photographic assistance. They also thank David Brinker, MD (Towson, MD) and Frank Ingram, MD (Charlotte, NC) for providing case material.
Source of Funding:
Supported in part by the Dahan Fund (P.A.) and Joey’s Wings (P.A.).
Footnotes
Conflicts of Interest: The authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article.
Supplemental Digital Content is available for this article. Direct URL citations appear in the printed text and are provided in the HTML and PDF versions of this article on the journal’s website, www.ajsp.com.
REFERENCES
- 1.Reinhard H, Aliani S, Ruebe C, et al. Wilms’ tumor in adults: results of the Society of Pediatric Oncology (SIOP) 93–01/Society for Pediatric Oncology and Hematology (GPOH) Study. J Clin Oncol. 2004;22:4500–4506. [DOI] [PubMed] [Google Scholar]
- 2.Tripathi S, Mishra A, Popat VC, et al. Wilms’ tumor in adults-conventional and unconventional presentations of a rare entity with a review of literature. J Kidney Cancer VHL. 2021;8:40–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Terenziani M, Spreafico F, Collini P, et al. Adult Wilms tumor: a monoinstitutional experience and a review of the literature. Cancer. 2004;15:289–293. [DOI] [PubMed] [Google Scholar]
- 4.Ali AN, Diaz R, Shu HK, et al. A surveillance, epidemiology and end results (SEER) program comparison of adult and pediatric Wilms tumor. Cancer. 2011;1:2541–2551. [DOI] [PubMed] [Google Scholar]
- 5.Byrd RL, Evans AE, D’Angio GJ. Adult Wilms tumor: effect of combined therapy on survival. J Urol. 1982;127:648–651. [DOI] [PubMed] [Google Scholar]
- 6.Mitry E, Ciccolallo L, Coleman MP, et al. Incidence of and survival from Wilms tumour in adults in Europe: data from EUROCARE study. Eur J Cancer. 2006;42:2363–2368. [DOI] [PubMed] [Google Scholar]
- 7.Segers H, van den Heuvel-Eibrink MM, Coppes MJ, et al. The SIOP-RTSG and the COG-Renal Tumour Committee. Management of adults with Wilms tumor: recommendations based on international consensus. Expert Rev Anticancer Ther. 2011;11:1105–1113. [DOI] [PubMed] [Google Scholar]
- 8.Huszno J, Starzyczyn-Stota D, Jaworska M, et al. Adult Wilms tumor—diagnosis and current therapy. Cent European J Urol. 2013;66: 39–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kalapurakal JA, Nan B, Norkool P, et al. Treatment outcomes in adults with favorable histologic type Wilms tumor-an update from the National Wilms Tumor Study Group. Int J Radiation Oncology Biol Phys. 2004;60:1379–1384. [DOI] [PubMed] [Google Scholar]
- 10.Arrigo S, Beckwith JB, Sharples K, et al. Better survival after combined modality care for adults with Wilms tumor. A report from the National Wilms Tumor Study. Cancer. 1990;1:827–830. [DOI] [PubMed] [Google Scholar]
- 11.Quezado M, Benjamin DR, Tsokos M. EWS/FLI-1 fusion transcripts in three peripheral primitive neuroectodermal tumors of the kidney. Hum Pathol. 1997;28:767–771. [DOI] [PubMed] [Google Scholar]
- 12.Jimenez RE, Folpe AL, Lapham RL, et al. Primary Ewing’s sarcoma/primitive neuroectodermal tumor of the kidney. A clinicopathologic and immunohistochemical analysis of 11 cases. Am J Stag Pathol. 2002;26:320–327. [DOI] [PubMed] [Google Scholar]
- 13.Argani P, Faria PA, Epstein JI, et al. Primary renal synovial sarcoma. Morphologic and molecular delineation of an entity previously included among embryonal sarcomas of the kidney. Am J Surg Pathol. 2000;24:1087–1096. [DOI] [PubMed] [Google Scholar]
- 14.Spreafico F, Ferrari A, Mascarin M, et al. Wilms tumor, medulloblastoma, and rhabdomyosarcoma in adult patient: lessons learned from the pediatric experience. Cancer and Metastasis Rev. 2019;38:683–694. [DOI] [PubMed] [Google Scholar]
- 15.Choueiri TK, Cheville J, Palescandolo E, et al. BRAF mutations in metanephric adenoma of the kidney. Eur Urol. 2012;62:917–922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Wobker SE, Matoso A, Pratilas CA, et al. Metanephric adenoma-epithelial Wilms tumor overlap lesions. Am J Surg Pathol. 2019;43: 1157–1159. [DOI] [PubMed] [Google Scholar]
- 17.Obasaju P, Shahab S, Dunn E, et al. BRAF V600E-mutated metastatic pediatric Wilms tumor with complete response to targeted RAF/MEK inhibition. Cold Spring Harb Mol Case Stud. 2020;1:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Li MM, Datto M, Duncavage EJ, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017;19:4–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Dudley JC, Gurda GT, Tseng LH, et al. Tumor cellularity as a quality assurance measure for accurate clinical detection of BRAF mutations in melanoma. Mol Diagn Ther. 2014;18:409–418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Carter J, Tseng LH, Zheng G, et al. Non-p.V600E BRAF mutations are common using a more sensitive and broad detection tool. Am J Clin Pathol. 2015;144:620–628. [DOI] [PubMed] [Google Scholar]
- 21.Zhang J, Lu J, Chen Y, et al. WHSC1 promotes wnt/β-catenin signaling in a FoxMl-dependent manner facilitating proliferation, invasion and epithelial-mesenchymal transition in breast cancer. J Recept Signal Transduct Res. 2020;40:410–418. [DOI] [PubMed] [Google Scholar]
- 22.Chen Q, Zheng PS, Yang WT. EZH2-mediated repression of GSK-3β and TP53 promotes Wnt/β-catenin signaling-dependent cell expansion in cervical carcinoma. Oncotarget. 2016;7:36115–36129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhou CC, Xiong QC, Zhu XX, et al. AFF1 and AFF4 differentially regulate the osteogenic differentiation of human MSCs. Bone Res. 2017;5:17044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Li B, Xu X, Bin X, et al. Ectopic expression of MELK in oral squamous cell carcinoma and its correlation with epithelial mesenchymal transition. Aging. 2021;13:13048–13060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dominguez-Brauer C, Khatun R, Elia AJ, et al. E3 ubiquitin ligase Mule targets β-catenin under conditions of hyperactive Wnt signaling. Proc Natl Acad Sci U S A. 2017;114:E1148–E1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ekici AB, Hilfinger D, Jatzwauk M, et al. Disturbed Wnt signalling due to a mutation in CCDC88C causes an autosomal recessive nonsyndromic hydrocephalus with medial diverticulum. Mol Svndromol. 2010;1:99–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Corbin M, de Reyniès A, Rickman DS, et al. WNT/beta-catenin pathway activation in Wilms tumors: a unifying mechanism with multiple entries? Genes Chromosomes Cancer. 2009;48:816–827. [DOI] [PubMed] [Google Scholar]
- 28.Warner DR, Greene RM, Pisano MM. Cross-talk between the TGFbeta and Wnt signaling pathways in murine embryonic maxillary mesenchymal cells. FEBS Lett. 2005;579:3539–3546. [DOI] [PubMed] [Google Scholar]
- 29.Pelullo M, Zema S, Nardozza F, et al. Wnt, Notch, and TGF-β pathways impinge on hedgehog signaling complexity: an open window on cancer. Front Genet. 2019;10:711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen X, Wang C, Jiang Y, et al. Bcl-3 promotes Wnt signaling by maintaining the acetylation of β-catenin at lysine 49 in colorectal cancer. Signal Transduct Target Ther. 2020;5:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chen X, Cao X, Sun X, et al. Bcl-3 regulates TGFβ signaling by stabilizing Smad3 during breast cancer pulmonary metastasis. Cell Death Dis. 2016;7:e2508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Caspi M, Perry G, Skalka N, et al. Aldolase positively regulates of the canonical Wnt signaling pathway. Mol Cancer. 2014;13:164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Romero G, von Zastrow M, Friedman PA. Role of PDZ proteins in regulating trafficking, signaling, and function of GPCRs: means, motif, and opportunity. Adv Pharmacol. 2011;62:279–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li XL, Huang YY, Yuan TJ, et al. STYK1 promotes the malignant progression of laryngeal squamous cell carcinoma through targeting TGF-β1. Eur Rev Med Pharmacol Sci. 2020;24:6166–6174. [DOI] [PubMed] [Google Scholar]
- 35.Koga K, Yokoi H, Mori K, et al. MicroRNA-26a inhibits TGFβ-induced extracellular matrix protein expression in podocytes by targeting CTGF and is downregulated in diabetic nephropathy. Diabetologia. 2015;58:2169–2180. [DOI] [PubMed] [Google Scholar]
- 36.Wang SE, Xiang B, Guix M, et al. Transforming growth factor beta engages TACE and ErbB3 to activate phosphatidylinositol-3 kinase/Akt in ErbB2-overexpressing breast cancer and desensitizes cells to trastuzumab. Mol Cell Biol. 2008;28:5605–5620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Black LE, Longo JF, Carroll SL. Mechanisms of receptor tyrosine-protein kinase ErbB-3 (ERBB3) action in human neoplasia. Am J Pathol. 2019;189:1898–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Seong HA, Manoharan R, Ha H. Zinc finger protein ZPR9 functions as an activator of AMPK-related serine/threonine kinase MPK38/MELK involved in ASK1/TGF-β/p53 signaling pathways. Sci Rep. 2017;7:42502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tang LY, Heller M, Meng Z, et al. Transforming Growth Factor-β (TGF-β) directly activates the JAK1-STAT3 Axis to induce hepatic fibrosis in coordination with the SMAD pathway. J Biol Chem. 2017;292:4302–4312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Williams RD, Chagtai T, Alcaide-German M, et al. Multiple mechanisms of MYCN dysregulation in Wilms tumour. Oncotarget. 2015;6:7232–7243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.McDonald JM, Douglass EC, Fisher R, et al. Linkage of familial Wilms’ tumor predisposition to chromosome 19 and a two-locus model for the etiology of familial tumors. Cancer Res. 1998;58: 1387–1390. [PubMed] [Google Scholar]
- 42.Trappe RU, Hippek RH, Plotkin M, et al. Effective use of high-dose chemotherapy and autologous stem cell rescue for relapsed adult Wilms tumor and a novel alteration in intron 1 of the WT2 gene. J Pediatr Hematol Oncol. 2004;26:82–83. [PubMed] [Google Scholar]
- 43.Hsiao HL, Chang TH, Wu WJ, et al. Adult Wilms tumor with hypospadias and cryptorchidism: a case report. Kaohsiung J Med Sci. 2007;23:584–589. [PubMed] [Google Scholar]
- 44.Huser J, Grignon DJ, Ro JY, et al. Adult Wilms’ tumor: a clinicopathologic study of 11 cases. Mod Pathol. 1990;3:321–326. [PubMed] [Google Scholar]
- 45.Su MC, Huang WC, Lien HC. Beta-catenin expression and mutation in adult and pediatric Wilms tumors. APMIS. 2008;116:771–778. [DOI] [PubMed] [Google Scholar]
- 46.Karlsson J, Mengelbier JH, Elfving P, et al. High-resolution genomic profiling of an adult Wilms tumor: evidence for a pathogenesis distinct from corresponding pediatric tumors. Virchows Arch. 2011; 459:547–553. [DOI] [PubMed] [Google Scholar]
- 47.Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:l1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sashida G, Oshima M, Iwama A. Deregulated polycomb functions in myeloproliferative neoplasms. Int J Hematol. 2019;110:170–178. [DOI] [PubMed] [Google Scholar]
- 50.Asada S, Fujino T, Goyama S, et al. The role of ASXL1 in hematopoiesis and myeloid malignancies. Cell Mol Life Sci. 2019;76:2511–2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Russell B, Johnston J, Biesecker LG, et al. Clinical management of patients with ASXL1 mutations and Bohring-Opitz syndrome, emphasizing the need for Wilms tumor surveillance. Am J Med Genet. 2015;167A: 2122–2131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Cao L, Li R, Wu X. The Functions and Mechanisms of PR-DUB in Malignancy. Front Mol Biosci. 2021;8:657150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Vougiouklakis T, Hamamoto R, Nakamura Y, et al. The NSD family of protein methyltransferases in human cancer. Epigenomics. 2015;7:863–874. [DOI] [PubMed] [Google Scholar]
- 54.Bennett RL, Swaroop A, Troche C, et al. The role of nuclear receptor-binding SET domain family histone lysine methyltransferases in cancer. Cold Spring Harb Perspect Med. 2017;7:a026708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Treger TD, Chowdhury T, Pritchard-Jones K, et al. The genetic changes of Wilms tumour. Nat Rev Nephrol. 2019;15:240–251. [DOI] [PubMed] [Google Scholar]
- 56.de vries-Brilland M, Sionneau B, Dutriaux C, et al. Successful treatment of metastatic adult Wilms tumor with anti-BRAF treatment: a case report and a brief review of the literature. Clin Genitourin Cancer. 2019;17:e721–e723. [DOI] [PubMed] [Google Scholar]
- 57.Rubin BP, Pins MR, Nielsen G, et al. Isochromosome 7q in adult Wilms tumors: diagnostic and pathogenetic implication. Am J Surg Pathol. 2000;24:1663–1669. [DOI] [PubMed] [Google Scholar]
- 58.Argani P Metanephric neoplasms: The hyperdifferentiated, benign end of the Wilms tumor spectrum? Clin Lab Med. 2005;25:379–392. [DOI] [PubMed] [Google Scholar]
- 59.Arroyo MR, Green DM, Perlman EJ, et al. The spectrum of metanephric adenofibroma and related lesions: clinicopathologic study of 25 cases from the National Wilms Tumor Study Group Pathology Center. Am J Surg Pathol. 2001;25:433–444. [DOI] [PubMed] [Google Scholar]
- 60.Argani P, Beckwith JB. Metanephric stromal tumor: report of 31 cases of a distinctive pediatric renal neoplasm. Am J Surg Pathol. 2000;24:917–926. [DOI] [PubMed] [Google Scholar]
- 61.Chami R, Yin M, Marrano P, et al. BRAF mutations in pediatric metanephric tumors. Hum Pathol. 2015;46:1153–1161. [DOI] [PubMed] [Google Scholar]
- 62.Diniz G, Aktas S, Turedi A, et al. Telomerase reverse transcriptase catalytic subunit expression and proliferation index in Wilms tumor. Tumor Biol. 2011;32:761–767. [DOI] [PubMed] [Google Scholar]
- 63.Song YS, Park YJ. Mechanisms of TERT reactivation and its interaction with BARFV600E. Endocrinol Metab. 2020;35:515–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chiba K, Lorbeer FK, Shain AH, et al. Mutations in the promoter of the telomerase gene TERT contribute to tumorigenesis by a two-step mechanism. Science. 2017;357:1416–1420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Meeker AK, Hicks JL, Icobuzio-Donahue CA, et al. Telomere length abnormalities occur early in the initiation of epithelial carcinogenesis. Clin Cancer Res. 2004;10:3317–3326. [DOI] [PubMed] [Google Scholar]
- 66.Hansel DE, Meeker AK, Hicks J, et al. Telomere length variation in biliary tract metaplasia, dysplasia, and carcinoma. Mod Pathol. 2006;19:772–779. [DOI] [PubMed] [Google Scholar]
- 67.Meeker AK, Hicks JL, Gabrielson E, et al. Telomere shortening occurs in subsets of normal breast epithelium as well as in situ and invasive carcinoma. Am J Pathol. 2004;164:925–935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chan E, Stohr BA, Croom NA, et al. Molecular characterisation of metanephric adenomas beyond BRAF: genetic evidence for potential malignant evolution. Histopathology. 2020;76:1084–1090. [DOI] [PubMed] [Google Scholar]
- 69.Young MD, Mitchell TJ, Vieira Braga FA, et al. Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science. 2018;361:594–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
