Next Article in Journal
CAR-NK Cells Targeting HER1 (EGFR) Show Efficient Anti-Tumor Activity against Head and Neck Squamous Cell Carcinoma (HNSCC)
Next Article in Special Issue
IGF1R Contributes to Cell Proliferation in ALK-Mutated Neuroblastoma with Preference for Activating the PI3K-AKT Signaling Pathway
Previous Article in Journal
Development Trajectories of Fatigue, Quality of Life, and the Ability to Work among Colorectal Cancer Patients in the First Year after Rehabilitation—First Results of the MIRANDA Study
Previous Article in Special Issue
PRAME Is a Novel Target of Tumor-Intrinsic Gas6/Axl Activation and Promotes Cancer Cell Invasion in Hepatocellular Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Large, Nested Variant of Urothelial Carcinoma Is Enriched with Activating Mutations in Fibroblast Growth Factor Receptor-3 among Other Targetable Mutations

by
Yaser Gamallat
1,2,†,
Mitra Afsharpad
1,†,
Soufiane El Hallani
3,
Christopher A. Maher
4,
Nimira Alimohamed
5,
Eric Hyndman
6 and
Tarek A. Bismar
1,2,5,7,*
1
Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
2
Department of Oncology, Biochemistry and Molecular Biology, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
3
Alberta Precision Laboratory, Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
4
Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
5
Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, AB T2N 4N2, Canada
6
Department of Surgery and Urology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 2T9, Canada
7
Alberta Precision Laboratory, Rockyview General Hospital, Departments of Pathology and Laboratory Medicine, Calgary, AB T2V 1P9, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(12), 3167; https://doi.org/10.3390/cancers15123167
Submission received: 8 April 2023 / Revised: 6 June 2023 / Accepted: 9 June 2023 / Published: 13 June 2023

Abstract

:

Simple Summary

Urothelial carcinoma of the large, nested variant is a specific histological morphology subtype of urothelial carcinoma. Although it is a rare variant, it requires specific attention due to its bland histology and the fact that it may potentially be missed in routine biopsies. In this study, we identify Fibroblast Growth Factor Receptor-3 (FGFR-3) as the most common mutation present in this subtype among other potential targetable mutations. All our cases of this variant also harbored other potentially actionable mutations in other genes, which could also be amenable to novel targeted therapy. Patients with this variant may benefit from additional molecular screening to identify potential therapeutic targets that could improve the clinical outcome of such patients.

Abstract

The large, nested variant of urothelial carcinoma (LNVUC) is characterized by bland histomorphology mimicking that of benign von Brunn nests. In the current study, we aimed to investigate the Fibroblast Growth Factor Receptor-3 (FGFR-3) activation and missense mutation in 38 cases, including 6 cases diagnosed with LNVUC and 32 with metastatic invasive urothelial carcinoma (UC). Initially, six formalin-fixed paraffin-embedded (FFPE) tissue samples of the LNVUC were subjected to whole-exome sequencing (WES), and then we performed targeted sequencing on 32 cases of metastatic invasive UC of various morphological subtypes, which were interrogated for the FGFR3. Our results revealed 3/6 (50%) LNVUC cases evaluated by WES in our study showed an activating mutation in FGFR-3, 33% showed an activating mutation in PIK3CA, and 17% showed activating mutation in GNAS or MRE11. Additionally, 33% of cases showed a truncating mutation in CDKN1B. All LNVUC in our study that harbored the FGFR-3 mutation showed additional activating or truncating mutations in other genes. Overall, 6/32 (18.75%) cases of random metastatic invasive UC showed missense mutations of the FGFR-3 gene. The LNVUC variant showed the higher incidence of FGFR-3 mutations compared to other types of mutations. Additionally, all LNVUC cases show additional activating or truncating mutations in other genes, thus being amenable to novel targeted therapy.

1. Introduction

Urothelial carcinoma is the most common pathological (histological) type of bladder cancer [1]. Approximately 75% of cases are classified as pure urothelial carcinoma and 25% as variants of mixed or less common variants such as squamous and adenocarcinoma [2]. The recent 2022 edition of WHO recognized several of these urothelial carcinoma variants as having important prognostic and predictive value [3].
The nested variant of urothelial carcinoma is a very rare subtype that is characterized by bland nests of neoplastic urothelial cells that are often seen invading muscularis propria [2]. The large, nested variant (LNVUC) is defined as a spectrum of the nested variant and is characterized by larger nests of invasive neoplastic urothelial cells [4]. The nested variant of urothelial carcinoma, and the large, nested variant as a spectrum, was first described in 1979 but was not recognized by the WHO until 2004. The bland histomorphology of this variant can mimic those of a benign von Brunn nests, leading to misdiagnosis, particularly in small biopsies, causing a delay in recognizing this lesion and therefore the treatment of patients [5]. Studies have shown that despite bland histomorphology, this variant is associated with poor outcome [6,7]. Moreover, at the time of diagnosis, approximately 69% of nested variants are in advanced stages (pT3/4), and 19% have nodal involvement. As with all variants subtypes of urothelial carcinoma, the risk of recurrence and progression of the nested variant is increased based on many factors including the presence of a residual tumor and in situ component [8], which, understandably, can be easily missed in this variant due to the bland morphological appearance.
Fibroblast growth factor receptor-3 (FGFR-3) is one of the four highly conserved fibroblast growth factor receptor tyrosine kinases (RTKs) and has been shown to have a role in tumor growth and survival regulations [9,10,11]. FGFR-3 has been implicated in multiple malignant neoplasms including multiple myeloma, urothelial and cervical carcinoma [12,13,14]. Despite this, several studies have focused on FGFR-3 as therapeutic and prognostic marker in urothelial carcinoma [10,15]. The clinical significance of FGFR mutations is highlighted with the development of the pan-FGFR inhibitor, Erdafitinib, and Pembrolizumab, which was approved by the Food and Drug Administration [13,16,17]. This has provided new hope in the management and treatment of malignancies with FGFR-3 mutations [18].
Studies have shown FGFR-3 mutation to be among the most common mutated oncogene in urothelial carcinoma overall [19]. FGFR-3 mutations have been reported in about 75% of noninvasive urothelial carcinoma and about 15–20% of high-grade tumors [20]. However, most studies have focused on the presence of this mutation in the plasmacytoid variant of urothelial carcinoma, another rare and aggressive variant of urothelial carcinoma [21,22,23], but only a few have focused on the nested variant.
This study is designed to characterize the molecular backgrounds of the large, nested variant of urothelial carcinoma, in the hope of identifying potential targetable mutations linking this specific histomorphology with specific genetic mutations.

2. Material and Methods

2.1. Patient Samples

In the current study, we recruited 38 metastatic UC samples of which 6 cases were diagnosed with LNVUC at Alberta Precision labs/University of Calgary Cumming School of Medicine between 2015 and 2019. Of these 6 cases, 2 belonged to the same patient (cases 2 and 3), who developed a primary LNVUC in the lower urothelial system (bladder) which was treated with partial cystectomy, and subsequently, after this diagnosis, he developed another primary LNVUC in the upper urothelial system (ureter) which was treated with bilateral nephroureterectomy. These cases were re-reviewed by an experienced genitourinary pathologist to confirm the diagnosis of this variant and select the most suitable areas for sequencing. The remaining 32 cases of metastatic invasive urothelial carcinoma, including 3 cases of the LNVUC, were also analyzed for FGFR-3 target seq.
All cases were classified based on the 2016 edition of the World Health Organization Classification of Tumors of the Urinary System. LNVUC cases demographic data, stage at the time of diagnosis, treatments and outcomes up to date were documented for each case (Table 1), and further demographics about the 32 cases of metastatic invasive urothelial carcinoma cases are provided in Table 2.

2.2. DNA Extraction and Whole-Exome Sequencing

Pathologically reviewed samples confirmed the diagnosis and determined tumor content, and marked tumor areas on hematoxylin and eosin (H&E) slides used to accurately obtain tissue samples from formalin-fixed paraffin-embedded (FFPE) samples. Briefly, the study pathologists cored or scrolled the selected tumor areas. Then tumor DNA was extracted using the QIAamp DNA FFPE Tissue kit (Qiagen, Cat # 56404, Hilden, Germany). Germline DNA was extracted from normal kidney tissue adjacent to the tumor. Qubit used to quantify the DNA using Qubit DNA HS assay (Life Technologies, Carlsbad, CA, USA). Whole-exome sequencing (WES) was carried out by The Centre for Applied Genomics (TCAG), Toronto, ON, Canada. Briefly, 750 ng of DNA was used for WES exome library preparation and sequencing using SureSelect XT Human All Exon V5 Kit (Agilent Technologies, Santa Clara, CA, USA). Genomic DNA was fragmented to 200-bp on average using a Covaris LE220 instrument. Prior to ligation, sheared DNA was end-repaired, and the 3′ ends were adenylated on adapters with overhang-T. Then, the genomic library was amplified by PCR using 10 cycles and hybridized with biotinylated probes that target exonic regions; the enriched exome libraries were amplified by an additional 8 cycles of PCR. The exomic libraries were validated using DNA High-Sensitivity chips on a Bioanalyzer 2100 (Agilent Technologies) for size and by qPCR using the Kapa Library Quantification Illumina/ABI Prism Kit protocol (KAPA Biosystems) for quantities. Exome libraries were pooled and sequenced with the TruSeq SBS sequencing chemistry using a V4 high throughput flowcell on a HiSeq 2500 platform (Illumina Inc., San Diego, CA, USA), as per Illumina’s recommended protocol.

2.3. Data Alignment and Validation

Around 6–8 gigabases of raw paired end data of 126-bases were generated per exome library. Reads were aligned to the hg19 build human reference genome using BWA (version 0.5.9). PCR duplicates were marked using picard-tools-1.108, and local re-alignment and base recalibration were performed using GATK 1.1-28. Variants (SNV, indel) were called using GATK UnifiedGenotyper 1.1-28. An Annovar-based pipeline was used for adding gene-based, feature-based and frequency-based annotations for variant filtering and prioritization [24].

2.4. Targeted Sequencing for FGFR Gene Fusions

Targeted seq testing was performed at Cancer Genetics Clinic, Jewish General Hospital, using an NGS panel which analyzes both DNA and RNA extracted from FFPE material and detects sequence changes involving the following FGFR Fusion proteins: Driver genes partner genes FGFR1: ADAM32, BAG4, BCR, CNTRL, CPSF6, CUX1, ERVK3-1, FGFR1OP, FGFR1OP2, FN1, LRRFIP1, MYO18A, NTM, PLAG1, RANBP2, SQSTM1, TACC1, TPR, TRIM24, WHSC1L1, ZMYM2, ZNF703 FGFR2: AFF3, AHCYL1, BICC1, CASP7, CCAR2, CCDC6, CD44, CIT, COL14A1, CREB5, CTNNB1, FAM76A, KCTD1, MGEA5, NOL4, OFD1, PARK2, PDHX, PPHLN1, SHTN1, SLC45A3, SNX19, TACC3, TXLNA, USP10 FGFR3: AES, BAIAP2L1, ELAVL3, ETV6, FBXO28, JAKMIP1, TACC3.

3. Results

Whole-exome sequencing results (Table 3) for the six cases of the large, nested variant of urothelial carcinoma showed 3 cases (50%) to harbor positive activating mutation in FGFR-3. Two cases showed an activating mutation in PIK3CA (33%), and one showed activating mutations in GNAS (17%), and another case showed mutations in MRE11 (17%).
A truncating mutation in CDKN1B was seen in two cases (33%), and truncating mutations in CDKN2A, ARID1B, ARID1A and KDM6A was seen only in one case each (17%). It was interesting to note that cases that showed activating mutations in FGFR-3 also showed additional activating and truncating mutations in other genes including PIK3CA. Detailed genetic mutations detected in the six cases studied are presented in Table 3.
All our cases that harbored FGFR-3 mutations showed additional activating or truncating mutations. One case showed simultaneous mutations in FGFR-3, PIK3CA, CDKN1B, ARID1B and PPP2R1A. Another case showed mutations in FGFR-3 as well as MRE11 and KDM66, and an additional case had simultaneous mutations in FGFR-3, CDKN1B and CDKN2A. Of the cases that did not harbor any FGFR-3 mutations, one case showed simultaneous mutations in PIK3CA, ARID1A and GNAS (Table 3).
As exhibited in Table 2, FGFR-3 mutation fusions by targeted sequencing were assessed in the 32 cases, including three cases of the six WES-performed samples series, which did not show FGFR-3 mutations by WES. Of those, 6/32 cases (18%) were positive for FGFR-3 missense mutations (two S249C, three Y373C and one G370C mutations). Three out of six cases exhibited dedifferentiated histology (poorly differentiated or squamous/sarcomatoid differentiation), five of six were from metastatic sites, and one was from the upper urothelial tract. However, no significant association was noted to specific histopathological morphology or metastatic site.

4. Discussion

Our results indicates that the FGFR-3 mutation is among the most common mutated oncogene in urothelial carcinoma, and it is even more common in the LNVUC. In our small study, we report 50% of cases as having a positive activating mutation in FGFR-3.
Additionally, we observed that many cases of LNVUC harbor simultaneous multiple activating mutations. In our series, four out of six (66%) LNVUC cases showed multiple activating mutations in oncogenes and truncating mutations in tumor suppressors, simultaneously. Of note, one patient who had two separate primaries of bladder and renal pelvis LNVUC demonstrated different mutational landscape between the two tumors, thus supporting different mutational landscape even in same-patient tumors, based on location.
Based on the targeted FGFR-3 sequencing of 32 cases of metastatic invasive urothelial carcinoma, 16% of cases showed FGFR-3 missense mutations. None of the cases that did not harbor FGFR-3 mutations by whole-exome sequencing showed FGFR-3 mutations, raising the possibility that point mutations in FGFR-3 gene are likely more frequent in LNVUC.
To illustrate our results in relation to published reports using genomic sequencing data, we added a review table for FGFR-3 analysis and comparison table analysis of two studies, including a study by Pietzak et al. [25] and the TCGA data, to compare the rate of specific mutations across urothelial carcinomas.
As demonstrated in our data, in non-muscle-invasive urothelial carcinoma, our results showed a slight but non-significantly higher incidence of FGFR-3 mutation, whereas compared to data provided by TCGA, the incident of FGFR-3 mutations in our variant was significantly higher compared to unselected variants of urothelial carcinoma. The incidence of CDKN1B, GNAS, MRE11 and PPP2R1A mutations was also significantly higher in our cases compared to both studies.
Similarly, other studies characterizing non-muscle-invasive and muscle-invasive high-grade urothelial carcinoma reported similar incidence of FGFR-3 mutations, ranging from 11% to 52% suggesting that the rate of FGFR-3 mutations may vary significantly depending on the methods used, site of assessment and variants of urothelial carcinoma included in the studies [26,27,28,29] (Table 4).
As in our study, Weyerer et al. [5] focused mainly on the large, nested variant of urothelial carcinoma, but they reported that 97% of their pure nested variants showed FGFR-3 mutation, whereas only 13% of the mixed tumor variant harbored this mutation. Their finding raises the possibility of different neoplastic pathways for mixed and pure LNVUC in their study.
It is also important to note that LNVUC and advanced UC shows a response to pembrolizumab especially with the recurrent LNVUC; however, our study does not focus on therapeutic strategies but the incidence associated with the presence of FGFR3 mutations [17,18].
Overall, all of these studies document a high incidence of FGFR-3 mutations in urothelial carcinoma, and they support that the LNVUC variant may exhibit an even higher incidence of FGFR-3 mutations especially in more pure histological type. Additionally, the rate of FGFR-3 mutations varies depending on the samples analyzed, whether it is resection or TURBT as well as histological grade, patient’s demographics, and underlying risk factors such as history of smoking. Finally, the studies document that FGFR-3 mutations usually occur in association with other activating or truncating mutations, especially in the PI3CKA pathway, and that LUCNV may harbor simultaneous activating and truncating mutations making them amenable for targeted therapy.

5. Conclusions

Our study provides further evidence of the promising role for FGFR-3 in the diagnosis and treatment of the large, nested variant of urothelial carcinoma, possibly implicating other targetable pathways compared to random unselected variants of urothelial carcinoma. We do acknowledge the limitations of our study, including small sample size and the fact that most cases were those of TURB. To better evaluate the role of these mutations in this rare variant of urothelial carcinoma, more studies, with a larger number of cases, focused on histomorphology, grade, stage as well as patient demographics and prognosis should be designed.

Author Contributions

Conceptualization, T.A.B.; Formal analysis, S.E.H. and C.A.M.; Resources, E.H.; Data curation, N.A.; Writing—original draft, M.A.; Writing—review & editing, Y.G. All authors have read and agreed to the published version of the manuscript.

Funding

Funding in part was supported by APL resident training grant # RS20-603.

Institutional Review Board Statement

The study was approved by the University of Calgary Cumming School of Medicine Ethics Review Board # HREBA CC16-0723 and in accordance with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Informed Consent Statement

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

Data Availability Statement

The data can be shared up on request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xu, N.; Yao, Z.; Shang, G.; Ye, D.; Wang, H.; Zhang, H.; Qu, Y.; Xu, F.; Wang, Y.; Qin, Z.; et al. Integrated Proteogenomic Characterization of Urothelial Carcinoma of the Bladder. J. Hematol. Oncol. 2022, 15, 76. [Google Scholar] [CrossRef] [PubMed]
  2. Lobo, N.; Shariat, S.; Guo, C.; Fernandez, M.; Kassouf, W.; Choudhury, A.; Gao, J.; Williams, S.; Galsky, M.; Taylor, J.A., 3rd; et al. What is the significance of variant histology in urothelial carcinoma? Eur. Urol. Focus 2020, 6, 653–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Netto, G.J.; Amin, M.; Berney, D.; Comperat, E.; Gill, A.; Hartmann, A.; Menon, S.; Raspollini, M.; Rubin, M.; Srigley, J.; et al. The 2022 world health organization classification of tumors of the urinary system and male genital organs-part b: Prostate and urinary tract tumors. Eur. Urol. 2022, 82, 469–482. [Google Scholar] [CrossRef] [PubMed]
  4. Comperat, E.; McKenney, J.; Hartmann, A.; Hes, O.; Bertz, S.; Varinot, J.; Brimo, F. Large nested variant of urothelial carcinoma: A clinicopathological study of 36 cases. Histopathology 2017, 71, 703–710. [Google Scholar] [CrossRef]
  5. Weyerer, V.; Eckstein, M.; Comperat, E.; Juette, H.; Gaisa, N.; Allory, Y.; Stohr, R.; Wullich, B.; Roupret, M.; Hartmann, A.; et al. Pure large nested variant of urothelial carcinoma (lnuc) is the prototype of an fgfr3 mutated aggressive urothelial carcinoma with luminal-papillary phenotype. Cancers 2020, 12, 763. [Google Scholar] [CrossRef] [Green Version]
  6. Levy, D.R.; Cheng, L. The expanding molecular and mutational landscape of nested variant of urothelial carcinoma. Histopathology 2020, 76, 638–639. [Google Scholar] [CrossRef]
  7. Linder, B.J.; Frank, I.; Cheville, J.; Thompson, R.; Thapa, P.; Tarrell, R.; Boorjian, S.A. Outcomes following radical cystectomy for nested variant of urothelial carcinoma: A matched cohort analysis. J. Urol. 2013, 189, 1670–1675. [Google Scholar] [CrossRef]
  8. Gontero, P.; Sylvester, R.; Pisano, F.; Joniau, S.; Eeckt, K.V.; Serretta, V.; Larre, S.; Di Stasi, S.; Van Rhijn, B.; Witjes, A.; et al. Prognostic factors and risk groups in t1g3 non-muscle-invasive bladder cancer patients initially treated with bacillus calmette-guerin: Results of a retrospective multicenter study of 2451 patients. Eur. Urol. 2015, 67, 74–82. [Google Scholar] [CrossRef] [Green Version]
  9. Kardoust Parizi, M.; Margulis, V.; Lotan, Y.; Mori, K.; Shariat, S.F. Fibroblast growth factor receptor: A systematic review and meta-analysis of prognostic value and therapeutic options in patients with urothelial bladder carcinoma. Urol. Oncol. 2021, 39, 409–421. [Google Scholar] [CrossRef]
  10. Kacew, A.; Sweis, R.F. Fgfr3 alterations in the era of immunotherapy for urothelial bladder cancer. Front. Immunol. 2020, 11, 575258. [Google Scholar] [CrossRef]
  11. Guancial, E.A.; Werner, L.; Bellmunt, J.; Bamias, A.; Choueiri, T.; Ross, R.; Schutz, F.; Park, R.; O’Brien, R.J.; Hirsch, M.; et al. Fgfr3 expression in primary and metastatic urothelial carcinoma of the bladder. Cancer Med. 2014, 3, 835–844. [Google Scholar] [CrossRef] [PubMed]
  12. Rezzola, S.; Ronca, R.; Loda, A.; Nawaz, M.; Tobia, C.; Paganini, G.; Maccarinelli, F.; Giacomini, A.; Semeraro, F.; Mor, M.; et al. The autocrine fgf/fgfr system in both skin and uveal melanoma: Fgf trapping as a possible therapeutic approach. Cancers 2019, 11, 1305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Qin, Q.; Patel, V.; Galsky, M.D. Urothelial carcinoma: The development of fgfr inhibitors in combination with immune checkpoint inhibitors. Expert Rev. Anticancer Ther. 2020, 20, 503–512. [Google Scholar] [CrossRef] [PubMed]
  14. Carneiro, B.A.; Elvin, J.; Kamath, S.; Ali, S.; Paintal, A.; Restrepo, A.; Berry, E.; Giles, F.; Johnson, M.L. Fgfr3-tacc3: A novel gene fusion in cervical cancer. Gynecol. Oncol. Rep. 2015, 13, 53–56. [Google Scholar] [CrossRef]
  15. Brunelli, M.; Tafuri, A.; Cima, L.; Cerruto, M.; Milella, M.; Zivi, A.; Buti, S.; Bersanelli, M.; Fornarini, G.; Vellone, V.; et al. Mdm2 gene amplification as selection tool for innovative targeted approaches in pd-l1 positive or negative muscle-invasive urothelial bladder carcinoma. J. Clin. Pathol. 2022, 75, 39–44. [Google Scholar] [CrossRef]
  16. King, G.; Javle, M. Fgfr Inhibitors: Clinical activity and development in the treatment of cholangiocarcinoma. Curr. Oncol. Rep. 2021, 23, 108. [Google Scholar] [CrossRef]
  17. Fukuta, K.; Izaki, H.; Shiozaki, K.; Nakanishi, R.; Inai, T.; Kataoka, H.; Kudo, E.; Kanda, K. Complete response to pembrolizumab in recurrent nested variant of urothelial carcinoma. IJU Case Rep. 2021, 4, 310–313. [Google Scholar] [CrossRef]
  18. Aragon-Ching, J.B. Pembrolizumab use in bladder cancer: A tale of two trials. Nat. Rev. Urol. 2021, 18, 577–578. [Google Scholar] [CrossRef]
  19. Al-Obaidy, K.I.; Cheng, L. Fibroblast growth factor receptor (fgfr) gene: Pathogenesis and treatment implications in urothelial carcinoma of the bladder. J. Clin. Pathol. 2021, 74, 491–495. [Google Scholar] [CrossRef]
  20. van Rhijn, B.W.G.; Mertens, L.; Mayr, R.; Bostrom, P.; Real, F.; Zwarthoff, E.; Boormans, J.; Abas, C.; van Leenders, G.; Gotz, S.; et al. Fgfr3 mutation status and fgfr3 expression in a large bladder cancer cohort treated by radical cystectomy: Implications for anti-fgfr3 treatment?(dagger). Eur. Urol. 2020, 78, 682–687. [Google Scholar] [CrossRef]
  21. Zengin, Z.B.; Chehrazi-Raffle, A.; Salgia, N.; Muddasani, R.; Ali, S.; Meza, L.; Pal, S.K. Targeted therapies: Expanding the role of fgfr3 inhibition in urothelial carcinoma. Urol. Oncol. 2022, 40, 25–36. [Google Scholar] [CrossRef] [PubMed]
  22. Sood, S.; Paner, G.P. Plasmacytoid urothelial carcinoma: An unusual variant that warrants aggressive management and critical distinction on transurethral resections. Arch. Pathol. Lab. Med. 2019, 143, 1562–1567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Teo, M.Y.; Al-Ahmadie, H.; Seier, K.; Tully, C.; Regazzi, A.; Pietzak, E.; Solit, D.; Tickoo, S.; Reuter, V.; Cha, E.; et al. Correction: Natural history, response to systemic therapy, and genomic landscape of plasmacytoid urothelial carcinoma. Br. J. Cancer 2022, 126, 1236. [Google Scholar] [CrossRef]
  24. Wang, K.; Li, M.; Hakonarson, H. Annovar: Functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010, 38, e164. [Google Scholar] [CrossRef]
  25. Pietzak, E.J.; Bagrodia, A.; Cha, E.; Drill, E.; Iyer, G.; Isharwal, S.; Ostrovnaya, I.; Baez, P.; Li, Q.; Berger, M.; et al. Next-generation sequencing of nonmuscle invasive bladder cancer reveals potential biomarkers and rational therapeutic targets. Eur. Urol. 2017, 72, 952–959. [Google Scholar] [CrossRef]
  26. Iyer, G.; Milowsky, M.I. Fibroblast growth factor receptor-3 in urothelial tumorigenesis. Urol. Oncol. 2013, 31, 303–311. [Google Scholar] [CrossRef]
  27. Downes, M.R.; Weening, B.; van Rhijn, B.; Have, C.; Treurniet, K.; van der Kwast, T.H. Analysis of papillary urothelial carcinomas of the bladder with grade heterogeneity: Supportive evidence for an early role of cdkn2a deletions in the fgfr3 pathway. Histopathology 2017, 70, 281–289. [Google Scholar] [CrossRef]
  28. Al-Ahmadie, H.A.; Iyer, G.; Janakiraman, M.; Lin, O.; Heguy, A.; Tickoo, S.; Fine, S.; Gopalan, A.; Chen, Y.; Balar, A.; et al. Somatic mutation of fibroblast growth factor receptor-3 (fgfr3) defines a distinct morphological subtype of high-grade urothelial carcinoma. J. Pathol. 2011, 224, 270–279. [Google Scholar] [CrossRef] [Green Version]
  29. Pouessel, D.; Neuzillet, Y.; Mertens, L.; van der Heijden, M.; de Jong, J.; Sanders, J.; Peters, D.; Leroy, K.; Manceau, A.; Maille, P.; et al. Tumor heterogeneity of fibroblast growth factor receptor 3 (fgfr3) mutations in invasive bladder cancer: Implications for perioperative anti-fgfr3 treatment. Ann. Oncol. 2016, 27, 1311–1316. [Google Scholar] [CrossRef] [PubMed]
  30. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014, 507, 315–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Demographics of the studied cases.
Table 1. Demographics of the studied cases.
CaseSexAge at the Time of DxDiagnosisCISStage at the Time of DxInitial TreatmentProgression
1M74Invasive high-grade urothelial carcinoma with features of large, nested variantNopT3Partial cystectomy and 4
cycles of cisplatin and gemcitabine back in January 2016
Progression to stage IV with bone med in 2019
2M48Papillary and inverted urothelial carcinoma with features of large, nested variant of urothelial carcinomaNopT2bRadical cystectomyProgression with second primary LNVUC
3M49High-grade papillary urothelial carcinoma of kidneyNopTaBilateral nephroureterectomyOngoing treatment
4M57High-grade, high volume invasive urothelial carcinoma, with nested areas (predominantly papillary)YesNot doneBladder preservation
Chemotherapy (cisplatin) and radiation, 2018.
Pembrolizumab from 2019
No known progression
5M61High-grade urothelial carcinoma, nested variantNopT2Neoadjuvant chemotherapy with cisplatin and gemcitabine November 2018–2019
cystectomy in 2020
No known progression
6F73Invasive urothelial carcinoma, large, nested variantYespT3aAdjuvant chemotherapy Jan 2019No known progression
Table 2. Site of tumor primary/metastasis and mutational status of the FGFR-3 on the additional 32 cases of metastatic invasive urothelial carcinoma for FGFR-3 using targeted seq.
Table 2. Site of tumor primary/metastasis and mutational status of the FGFR-3 on the additional 32 cases of metastatic invasive urothelial carcinoma for FGFR-3 using targeted seq.
Site of MetastasisPrimary DiagnosisAgeGenderMutation
1LungHigh-grade invasive urothelial carcinoma with focal sarcomatoid differentiation79MNegative
2ProstateHigh-grade invasive urothelial carcinoma with features of large, nested variant infiltrating into muscularis propria and bladder neck60MNegative
3PenileHigh-grade invasive urothelial carcinoma78MNegative
4Right humerusInvasive high-grade urothelial carcinoma72FNegative
5Prostatic urethraHigh-grade invasive urothelial carcinoma arising from the prostatic urethra61MFGFR-3 S249C
6Renal pelvisInvasive high-grade papillary urothelial carcinoma, squamous differentiation present71MFGFR-3 Y373C
7LiverInvasive high-grade papillary urothelial carcinoma88MNegative
8LungNoninvasive high-grade papillary urothelial carcinoma.71MFGFR-3 G370C
9Lymph nodeHigh-grade invasive urothelial carcinoma68MFGFR-3 Y373C
10Lymph nodeHigh-grade invasive urothelial carcinoma with extensive squamous differentiation60FNegative
11KidneyHigh-grade urothelial carcinoma (HGUC)74MNegative
12LungInvasive high-grade urothelial carcinoma66MNegative
13Lymph node, bone, lung and liverHigh-grade invasive urothelial carcinoma with focal sarcomatoid differentiation79MNegative
14Facial bonePoorly differentiated malignant cells present, compatible with a poorly differentiated carcinoma49MFGFR-3 Y373C
15Lymph node and liverHigh-grade papillary urothelial carcinoma with squamous differentiation65MNegative
16Lymph nodeUrothelial carcinoma, with prominent intraductal spread77MNegative
17ProstateHigh-grade invasive urothelial carcinoma with focal sarcomatoid differentiation,78MFGFR-3 S249C
18Lymph nodes and peritoneumHigh-grade urothelial carcinoma with divergent differentiation74MNegative
19BoneHigh-grade invasive urothelial carcinoma69FNegative
20Lymph node (para-aortic)Invasive high-grade urothelial carcinoma61MNegative
21Lymph node (retroperitoneal)High-grade invasive urothelial carcinoma arising from the prostatic66MNegative
22KidneyInvasive high-grade papillary urothelial carcinoma, sarcomatoid differentiation present35FNegative
23Liver and boneInvasive high-grade papillary urothelial carcinoma66MNegative
24Pelvic soft tissueNoninvasive high-grade papillary urothelial carcinoma.64MNegative
25Retroperitoneal soft tissueHigh-grade invasive urothelial carcinoma75FNegative
26LungHigh-grade invasive urothelial carcinoma with extensive squamous differentiation68MNegative
27Retroperitoneal soft tissueHigh-grade urothelial carcinoma (HGUC):74MNegative
28Retroperitoneal soft tissueInvasive high-grade urothelial carcinoma62MNegative
29Lymph nodeHigh-grade invasive urothelial carcinoma with focal sarcomatoid59MNegative
30None (from the first series)High-grade, high-volume invasive urothelial carcinoma, with nested areas (predominantly papillary)57MNegative
31None (from the first series)High-grade urothelial carcinoma, nested variant61MNegative
32None (from the first series)Invasive urothelial carcinoma, large, nested variant73FNegative
Table 3. The genetic mutations detected in the 6 cases of LNVUC via WES.
Table 3. The genetic mutations detected in the 6 cases of LNVUC via WES.
GenesFrequencyCase 1Case 2 *Case 3 *Case 4Case 5Case 6
FGFR-350%
PIK3CA33%
CDKN1B33%
CDKN2A17%
ARID1B17%
ARID1A17%
GNAS17%
MRE1117%
KDM6A17%
PPP2R1A17%
BRD717%
CCDC17517%
CFTR17%
CNTLN17%
CRHR217%
FKBP1517%
GPRASP117%
KCNQ317%
KRTAP24-117%
KRTAP24-117%
LOC10012908317%
LRP817%
MAGED117%
MBD617%
OR2T217%
OR2T3517%
OR6P117%
OR6P117%
PRR3017%
PRR3017%
RABGGTA17%
RBM1017%
RREB117%
RYR117%
SIPA1L117%
SMOX17%
STX1017%
TMC717%
ZNF56017%
ZNF56017%
* Cases from same patient (case 2, bladder; case 3, renal pelvis). Green Boxes: Activating mutation in oncogenes; Orange Boxes: Truncating mutation in tumor suppressors.
Table 4. Data provided on the incidence of FGFR-3 mutation in our study and reviewed studies.
Table 4. Data provided on the incidence of FGFR-3 mutation in our study and reviewed studies.
StudyMethod UsedPatient PopulationFGFR-3 Mutation
Our studyWhole-genome sequencingInvasive LNVUC diagnosed on both TURB and cystectomy50%
Target sequencingMetastatic urothelial carcinoma regardless of variant16%
Pietrzak et al. [25]Targeted NGSNon-muscle-invasive UC49%
The Cancer Genome Atlas (TCGA) 2014 [30]Whole-exome sequencingHigh-grade muscle-invasive urothelial bladder carcinomas13%
Downes et al. [27]PCR and SNaPshot methodologyPapillary urothelial carcinoma52%
Iyer et al. [26]Review articleNon-muscle-invasive UCActivating mutation 70%
Muscle-invasive UCOverexpression 40%
Al-Ahmadie et al. [28]Sanger sequencing and MALDI–TOF mass spectrometryHGUC17%
LGUC84%
Pouessel et al. [29]PCR-SnaPshot methodT1-TURB UC38%
T2-TURB UC30%
LN + UC5%
Weyerer et al. [5]SnaPshot analysisPure LNVUC94%
Mixed LNVUC14.2%
Overall LNVUC73.9%
Large, nested urothelial carcinoma (LNVUC), urothelial carcinoma (UC), lymph node (LN), transurethral resection of the bladder (TURB), polymerase chain reaction (PCR).
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gamallat, Y.; Afsharpad, M.; El Hallani, S.; Maher, C.A.; Alimohamed, N.; Hyndman, E.; Bismar, T.A. Large, Nested Variant of Urothelial Carcinoma Is Enriched with Activating Mutations in Fibroblast Growth Factor Receptor-3 among Other Targetable Mutations. Cancers 2023, 15, 3167. https://doi.org/10.3390/cancers15123167

AMA Style

Gamallat Y, Afsharpad M, El Hallani S, Maher CA, Alimohamed N, Hyndman E, Bismar TA. Large, Nested Variant of Urothelial Carcinoma Is Enriched with Activating Mutations in Fibroblast Growth Factor Receptor-3 among Other Targetable Mutations. Cancers. 2023; 15(12):3167. https://doi.org/10.3390/cancers15123167

Chicago/Turabian Style

Gamallat, Yaser, Mitra Afsharpad, Soufiane El Hallani, Christopher A. Maher, Nimira Alimohamed, Eric Hyndman, and Tarek A. Bismar. 2023. "Large, Nested Variant of Urothelial Carcinoma Is Enriched with Activating Mutations in Fibroblast Growth Factor Receptor-3 among Other Targetable Mutations" Cancers 15, no. 12: 3167. https://doi.org/10.3390/cancers15123167

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

Article Metrics

Back to TopTop