Molecular pathology/genetics of thymic tumors: extended abstract
Extended Abstract

Molecular pathology/genetics of thymic tumors: extended abstract

Yosuke Yamada1,2*, Alexander Marx3

1Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; 2Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; 3Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany

*The affiliation 2: from February 1st, 2024.

Correspondence to: Yosuke Yamada, MD, PhD. Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan. Email: yyamada@kuhp.kyoto-u.ac.jp.

Received: 13 November 2023; Accepted: 06 March 2024; Published online: 24 May 2024.

doi: 10.21037/med-23-55


Genetics of thymic epithelial tumors (TETs)

Comprehensive profiling of genetic or chromosomal abnormalities and gene expression has significantly advanced our molecular understanding of TETs. The most valuable discovery in TET genetics was made by Petrini et al. (1), who found that TETs can harbor the general transcription factor IIi (GTF2I) L424H variant: GTF2I(NM_032999.4):c.1271T>A p.(Leu424His). The significance of this finding is highlighted by two observations. First, this variant is common to TETs. The original study reported that the frequency in type A, AB, B1, B2, and B3 thymomas, and thymic carcinoma is 82%, 74%, 32%, 22%, 21%, and 8%, respectively (1). This high frequency contrasts with the fact that TETs have the fewest variants among cancers in humans (2-4). The study conducted as part of The Cancer Genome Atlas (TCGA) project confirmed the subtype dependency because the variant was observed in 100% and 72% of type A and type AB thymomas, respectively, but in 0% to 10% of type B thymomas and thymic carcinomas (2). Second, GTF2I L424H is virtually specific to TETs. Among 10,844 tumors from 31 TCGA studies for non-TETs, only 2 tumors harbored GTF2I L424H (3,4). These findings suggest that GTF2I L424H is biologically relevant in a thymus-specific manner.

Two recent in vivo mouse studies successfully addressed this hypothesis. Giorgetti et al. induced Gtf2i L384H with the Foxn1 promoter into thymic epithelial cells (TECs) and demonstrated that this caused developmental abnormalities of the thymus, such that the border between the cortex and medulla became obscured (5). He et al., the group that first reported GTF2I variant in TETs, established a Foxn1-dependent GTF2I L424H knock-in model and demonstrated that this caused tumor-like expansion of the thymus through proliferation of keratin-positive epithelial cells (6). These in vivo studies have demonstrated the biological significance of the GTF2I variant, and future studies may address the possibility that this variant could be a treatment target. Alternatively, these models could be improved by inducing the transgenes not congenitally but conditionally in adult mice.

Radovich et al. conducted the most comprehensive genomic profiling of TETs to date, as part of the TCGA study cited above (2). According to the study, HRAS, NRAS, and TP53 can be recurrently mutated in TETs, although in minor proportions. One drawback of the TCGA study is that the number of cases may not be sufficient, especially for minor thymoma subtypes and thymic carcinoma. On the other hand, some recent studies enrolled many cases for genetic studies by utilizing gene panel testing, a method that is gaining popularity. Girard et al. genetically analyzed 274 advanced-stage TETs (7). As found with the TCGA dataset, they reported few genomic alterations in thymomas. That study included many thymic carcinomas (n=144) with detailed histological subtypes and noted that even thymic carcinoma generally harbors few variants. A new finding was that CDKN2A and CDKN2B were relatively frequently mutated. A recent study from Japan enrolled the largest number of cases (n=794) (8) and reported results that overlapped substantially with that of Girard et al. (7). Collectively, we can summarize representative TET genetics as follows: (I) GTF2I L424H occurs often in type A and AB thymomas; (II) HRAS variants might be expected in type A thymoma, but the frequency is unconvincing; (III) TP53, CDKN2A, and CDKN2B variants are seen in about 30% of thymic carcinomas; (IV) druggable KIT variants can be detected in around 10% of thymic carcinomas (9); and (V) tumor mutation burden-high and microsatellite instability-high statuses are observed in less than 10% of thymic carcinomas, and these may be useful biomarkers for applying immune checkpoint inhibitors. Thus, it is true that TETs generally lack druggable variants; however, because TETs are rare and establishing treatment strategies based on big data is challenging, each case can be treated based on its unique features, which can be identified through genomic profiling. One example is a thymic carcinoma case that harbored a PI3KCA variant and substantially responded to everolimus (7). Another important finding about TET genetics is that thymomas rarely harbor gene fusions relevant to prognosis or diagnosis. KMT2A-MAML2 fusion in rare type B2 and B3 thymomas and YAP1-MAML2 fusion in metaplastic thymoma should be mentioned here (10,11).


Molecular pathology of TETs

The TCGA project (2) has played a fundamental role in this topic. By combining five omics platforms, it showed that TETs can be divided into four molecular subtypes: A (type A thymoma)-like, AB-like, B-like, and C (thymic carcinoma)-like, which are correlated with histotypes. Of note, the B-like and C-like clusters are entirely separate. A recent review indicated almost no histogenetic link between type B3 thymoma and thymic carcinoma (12). In contrast, recent studies, with the help of the TCGA dataset, have suggested that thymic carcinoma may have a phenotype related to medullary TECs. Our group proposed that thymic carcinoma often exhibits an expression profile similar to that of tuft cells, a unique subset of mTECs (13,14), and another group showed that thymic carcinoma can express AIRE, the representative mTEC gene (15). The functional and therapeutic relevance of this medullary characteristic has not been addressed, and future studies should answer these questions to advance our understanding of TET histogenesis.


Potential role of epigenetics linking genetics and molecular pathology in TETs: closing remarks

The relevance of epigenetic signatures in cancer has been appreciated (16). Indeed, the TCGA study of TETs demonstrated that methylation status alone is highly correlated with histological subtype (2,17). In addition, studies by Wang et al. (18) have indicated that epigenetic regulatory genes, such as BAP1, SETD2, and ASXL1, are recurrently mutated in thymic carcinomas, which was confirmed by a large Japanese study (8). Therefore, future in-depth epigenetic studies might contribute to a more comprehensive understanding of TETs. Accordingly, the significance of loss of 16q, the most common chromosomal abnormality in thymic carcinoma, should be investigated not only because the locus may contain functionally relevant genes for carcinogenesis, but because chromosomal abnormalities can cause global epigenetic alterations (19).


Acknowledgments

Funding: This work was supported by JSPS KAKENHI (Grant Number: JP 21K06902).


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editors (Malgorzata Szolkowska, Chul Kim, Mohammad Ashraghi, and Claudio Silva) for “The Series Dedicated to the 13th International Thymic Malignancy Interest Group Annual Meeting (ITMIG 2023)” published in Mediastinum. The article has undergone external peer review.

Peer Review File: Available at https://med.amegroups.com/article/view/10.21037/med-23-55/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://med.amegroups.com/article/view/10.21037/med-23-55/coif). “The Series Dedicated to the 13th International Thymic Malignancy Interest Group Annual Meeting (ITMIG 2023)” was commissioned by the editorial office without any funding or sponsorship. Y.Y. reports funding from JSPS KAKENHI (Grant Number: JP 21K06902). The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Petrini I, Meltzer PS, Kim IK, et al. A specific missense mutation in GTF2I occurs at high frequency in thymic epithelial tumors. Nat Genet 2014;46:844-9. [Crossref] [PubMed]
  2. Radovich M, Pickering CR, Felau I, et al. The Integrated Genomic Landscape of Thymic Epithelial Tumors. Cancer Cell 2018;33:244-258.e10. [Crossref] [PubMed]
  3. 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;2:401-4. [Crossref] [PubMed]
  4. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. [Crossref] [PubMed]
  5. Giorgetti OB, Nusser A, Boehm T. Human thymoma-associated mutation of the GTF2I transcription factor impairs thymic epithelial progenitor differentiation in mice. Commun Biol 2022;5:1037. [Crossref] [PubMed]
  6. He Y, Kim IK, Bian J, et al. A Knock-In Mouse Model of Thymoma With the GTF2I L424H Mutation. J Thorac Oncol 2022;17:1375-86. [Crossref] [PubMed]
  7. Girard N, Basse C, Schrock A, et al. Comprehensive Genomic Profiling of 274 Thymic Epithelial Tumors Unveils Oncogenic Pathways and Predictive Biomarkers. Oncologist 2022;27:919-29. [Crossref] [PubMed]
  8. Kurokawa K, Shukuya T, Greenstein RA, et al. Genomic characterization of thymic epithelial tumors in a real-world dataset. ESMO Open 2023;8:101627. [Crossref] [PubMed]
  9. Ströbel P, Hartmann M, Jakob A, et al. Thymic carcinoma with overexpression of mutated KIT and the response to imatinib. N Engl J Med 2004;350:2625-6. [Crossref] [PubMed]
  10. Vivero M, Davineni P, Nardi V, et al. Metaplastic thymoma: a distinctive thymic neoplasm characterized by YAP1-MAML2 gene fusions. Mod Pathol 2020;33:560-5. [Crossref] [PubMed]
  11. Massoth LR, Hung YP, Dias-Santagata D, et al. Pan-Cancer Landscape Analysis Reveals Recurrent KMT2A-MAML2 Gene Fusion in Aggressive Histologic Subtypes of Thymoma. JCO Precis Oncol 2020;4:PO.19.00288.
  12. Roden AC, Ahmad U, Cardillo GThymic Carcinomas-A Concise Multidisciplinary Update on Recent Developments From the Thymic Carcinoma Working Group of the International Thymic Malignancy Interest Group, et al. J Thorac Oncol 2022;17:637-50. [Crossref] [PubMed]
  13. Yamada Y, Simon-Keller K, Belharazem-Vitacolonnna D, et al. A Tuft Cell-Like Signature Is Highly Prevalent in Thymic Squamous Cell Carcinoma and Delineates New Molecular Subsets Among the Major Lung Cancer Histotypes. J Thorac Oncol 2021;16:1003-16. [Crossref] [PubMed]
  14. Yamada Y, Sugimoto A, Hoki M, et al. POU2F3 beyond thymic carcinomas: expression across the spectrum of thymomas hints to medullary differentiation in type A thymoma. Virchows Arch 2022;480:843-51. [Crossref] [PubMed]
  15. Matsumoto M, Ohmura T, Hanibuchi Y, et al. AIRE illuminates the feature of medullary thymic epithelial cells in thymic carcinoma. Cancer Med 2023;12:9843-8. [Crossref] [PubMed]
  16. Hanahan D. Hallmarks of Cancer: New Dimensions. Cancer Discov 2022;12:31-46. [Crossref] [PubMed]
  17. Yamada Y, Bohnenberger H, Kriegsmann M, et al. Tuft cell-like carcinomas: novel cancer subsets present in multiple organs sharing a unique gene expression signature. Br J Cancer 2022;127:1876-85. [Crossref] [PubMed]
  18. Wang Y, Thomas A, Lau C, et al. Mutations of epigenetic regulatory genes are common in thymic carcinomas. Sci Rep 2014;4:7336. [Crossref] [PubMed]
  19. Letourneau A, Santoni FA, Bonilla X, et al. Domains of genome-wide gene expression dysregulation in Down's syndrome. Nature 2014;508:345-50. [Crossref] [PubMed]
doi: 10.21037/med-23-55
Cite this article as: Yamada Y, Marx A. Molecular pathology/genetics of thymic tumors: extended abstract. Mediastinum 2024;8:16.

Download Citation