Insights into molecular aspects and targeted therapy of thymic carcinoma: a narrative review
Review Article

Insights into molecular aspects and targeted therapy of thymic carcinoma: a narrative review

Tadashi Sakane1,2 ORCID logo, Hiroshi Haneda1, Katsuhiro Okuda2

1Department of Thoracic Surgery, Nagoya City University West Medical Center, Nagoya, Japan; 2Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Tadashi Sakane, MD, PhD. Department of Thoracic Surgery, Nagoya City University West Medical Center, 1-1-1 Hirate-cho, Kita-ku, Nagoya 462-8508, Japan; Department of Thoracic and Pediatric Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan. Email: t.sakane@med.nagoya-cu.ac.jp.

Background and Objective: Thymic carcinomas are rare tumors derived from thymic epithelial cells. Owing to their rarity, the search for molecular biology has been conducted in combination with thymoma as one histological subtype, and only a few studies have exclusively focused on thymic carcinoma. Currently, no therapy is more effective than complete surgical resection, and the development of novel therapies, including targeted therapies, is hampered. In this review, we summarize the knowledge regarding altered genes and pathways in thymic carcinoma with recent preclinical and clinical targeted therapies.

Methods: We conducted a narrative review of the relevant English literature available in PubMed and Google Scholar on genomic characteristics and targeted therapies for thymic carcinoma.

Key Content and Findings: Although the literature consists of a relatively small series, it suggests that the frequently involved genes or pathways associated with thymic carcinoma are tumor suppressor genes, including TP53 and CDKN2A/B, and the receptor tyrosine kinase pathway. Targeted therapy demonstrated antitumor activity with encouraging results. However, potential predictive biomarkers have not been identified and the response to these therapies appears to be irrelevant to gene alterations.

Conclusions: Some studies have revealed the molecular characteristics of thymic carcinoma, although the results of these studies have shown a different pattern of gene alterations. The further accumulation of data would be helpful in revealing the genomic landscape and establishing molecular-targeted therapies.

Keywords: Thymic carcinoma; molecular profile; targeted therapies


Received: 31 October 2023; Accepted: 21 December 2023; Published online: 21 March 2024.

doi: 10.21037/med-23-48


Introduction

Thymic carcinoma is an extremely rare thymic neoplasm, accounting for approximately 10% of thymic epithelial tumors (TETs) (1). In addition to their rarity, thymic carcinomas include various histological subtypes, with squamous cell carcinoma being the most common (2). Thymic carcinomas exhibit more aggressive behavior and a higher metastatic potential than thymomas (3). The median overall survival (OS) is 6.6 years, with 5- and 10-year OS rates of 60% and 40%, respectively. The prognosis for advanced disease, which accounts for approximately 70–75% of all cases, is miserable; the 5-year OS rates were 63% for stage III, 42% for stage IVa and 30% for IVb (4,5).

The factors associated with the development of TETs remain unknown; however, the understanding of the aberrant gene pathways involved in TETs has been gradually improving over the last decade, largely through the advent of next-generation sequencing (NGS) technologies. Previous studies have found that different histological subtypes of TETs exhibit different molecular profiles (6-10). In thymomas, a significant and recurrent missense mutation in the general transcription factor IIi (GTF2I) have been identified in type A and AB subtypes, which is reputed to drive their growth (6,8). In thymic carcinomas, owing to the rarity of these tumors and their histological heterogeneity, the results of studies show a different pattern of molecular aberrations with only a few significantly and recurrently mutated genes. Accordingly, data on their biology and clinical behavior are limited. In this review, we discuss the recent advances in the investigation of the molecular characteristics of thymic carcinoma and the development of potential targeted therapies. We present this article in accordance with the Narrative Review reporting checklist (available at https://med.amegroups.com/article/view/10.21037/med-23-48/rc).


Methods

An extended review of the relevant literature in PubMed and Google Scholar was conducted, using different combinations of search terms, including ‘thymic carcinoma’, ‘thymic epithelial tumor’, ‘gene’ or ‘genetic’, ‘mutation’ or ‘aberration’ or ‘alteration’, ‘molecular’, ‘targeted’ or ‘molecular targeting’ and ‘therapy’. The types of articles included in the search criteria were meta-analyses, systematic reviews, prospective studies, retrospective studies, case studies, and previous related reviews. Additional papers were identified by reviewing the reference lists of relevant publications. Publications with incomplete or irrelevant data, and those written in languages other than English were excluded. The search strategy is presented in Table 1.

Table 1

Search strategy summary

Items Specification
Date of search 15th July–10th August 2023
Databases and other sources searched PubMed and Google Scholar
Search terms used ‘thymic carcinoma’, ‘thymic epithelial tumor’, ‘gene’ or ‘genetic’, ‘mutation’ or ‘aberration’ or ‘alteration’, ‘molecular’, ‘targeted’ or ‘molecular targeting’ and ‘therapy’
Timeframe Date unrestricted to August 2023
Inclusion and exclusion criteria Inclusion criteria: (I) English language; (II) meta-analyses, systematic reviews, prospective studies, retrospective studies, case studies, and previous related reviews
Exclusion criteria: studies with incomplete or irrelevant data
Selection process One author compiled a list of eligible studies followed by review by all authors to determine suitability

Genetic alterations in thymic carcinoma

Tumor suppressor genes

In addition to the two early reports by Hirabayashi et al. and Tateyama et al. that showed a high frequency TP53 point mutations in thymic carcinoma, Wang et al. and Moreira et al. reported that TP53 mutations were exclusively observed in thymic carcinoma and were associated with aggressive behavior (7,11-13). Petrini et al. also identified recurrent mutations in TP53 in thymic carcinoma (6). Several studies have found a high frequency of TP53 in thymic carcinoma (7.7–25.7%), some of which showed that the presence of TP53 mutations was associated with a poor prognosis (14-22). A recent study conducted by Girard et al., which included the largest cohort, identified TP53 mutations in 25.9% of 174 thymic carcinoma cases (9).

CDKN2A and CDKN2B, located on chromosome 9p21, encode p16 and p15, respectively, which act by inhibiting CDK4 and CDK6, and are negative regulators of cell cycle progression (23,24). In thymic carcinoma, Aesif et al. examined the expression of p16 by immunohistochemistry (IHC) and cytogenetic abnormalities of CDKN2A by fluorescence in situ hybridization (FISH) (25). They reported that 53.8% (14/26) of the cases showed the expression of p16 and 19.0% (4/21) had homozygous deletion of CDKN2A, suggesting that the loss of p16 expression and homozygous deletion of CDKN2A could be predictors of a poor prognosis. Another study reported that copy number aberrations of CDKN2A and CDKN2B are associated with a worse prognosis in thymic carcinoma (26). Two recent NGS analyses with large cohorts showed similar results: the mutation frequencies of CDKN2A and CDKN2B were high: approximately 40% for CDKN2A and approximately 25% for CDKN2B (9,27).

CYLD and BAP1 are both tumor suppressor genes, and mutations in these genes have been detected in 8.5–18.8% and 8.2–12.5% of thymic carcinomas, respectively (6,7,9,13,27,28). According to the results of a phase 2 study of pembrolizumab by Giaccone et al., there were five patients with a CYLD mutation (12.2%) among 41 patients with thymic carcinoma, and these five patients exhibited high expression of programmed death-ligand 1 (PD-L1), three of whom had a complete response (CR) or partial response (PR) (29). He et al. characterized the genomic profiles of ten patients with thymic carcinoma who received pembrolizumab and identified that alterations in CYLD were promising predictors of a response to pembrolizumab (30). Meanwhile, they found that mutations in BAP1, which were also correlated with the expression of PD-L1, were promising predictors of pembrolizumab resistance (30). Angirekula et al. demonstrated that 11.4% of thymic carcinomas had lost the nuclear expression of BAP1, and that the loss of BAP1 expression may help distinguish thymomas from thymic carcinomas (31).

Receptor tyrosine kinases

The epidermal growth factor receptor (EGFR) is frequently mutated and/or overexpressed in different types of human cancers and is a target of multiple cancer therapies (32). Several studies investigated the EGFR expression levels in thymic carcinoma using IHC and reported that EGFR was overexpressed in 20.0–100.0% of cases (33-39). However, EGFR mutations are rare in thymic carcinomas (18,21,35-37,40-42).

KIT plays a major role in the development and maintenance of gastrointestinal stromal tumors (GISTs). Since Pan et al. found that thymic carcinoma frequently overexpressed KIT, whereas thymoma was found to be consistently negative for KIT by a systematic survey using a tissue array technique, alterations or expression of KIT in thymic carcinoma have been well-demonstrated in the literature (43). Immunohistochemical KIT positivity is found in 50.0–88.2%, although KIT mutations are relatively rare (19,33,37,44-46). The expression of KIT has been associated with activating mutations in exons 9, 11, 13, and 17 of KIT. KIT and PDGFRA are highly homologous and activate similar downstream signal transduction pathways (47). PDGFRA mutation, which is also considered to be a major driver gene of GISTs, is reported to occur in 0.0–5.6% of thymic carcinomas (19,42,44).

HER-2/neu is a proto-oncogene, and gene amplification and the overexpression of HER-2 have been demonstrated to be targets for several cancers (48). Pan et al. found that 47.1% (8/17) of thymic carcinoma overexpressed HER-2 by IHC, while no evidence of gene amplification was detected by FISH (49). According to a study conducted by Weissferdt et al., the significant immunohistochemical expression of HER-2 was observed in 58.3% (14/24) of cases, while 4.2% (1/24) showed HER-2/neu gene amplification, and 75.0% (18/24) exhibited increased HER-2/neu gene copy numbers (39). Genetic alterations of HER-2/neu are rare (9,42).

Insulin-like growth factor 1 receptor (IGF-1R) is a transmembrane receptor involved in cancer development, metastasis, and therapeutic resistance (50). Zucali et al. analyzed the IGF-1R expression in eight cases of thymic carcinoma by IHC, and seven cases (87.5%) were positive for IGF-1R (51). They also found that the expression of IGF-1R was significantly more common in aggressive histological subtypes than in indolent ones. Meanwhile, IGF-1R mutations have been reported to be rare, with a frequency of less than 8.3% (9,14,21).

FGFR3 encodes a member of the FGFR family (52). Aberrant FGFR signaling has been reported in many cancers, including breast cancer and colorectal cancer, and contributes to oncogenesis, tumor progression, and resistance to anticancer therapies (53,54). Asselta et al. performed an NGS analysis targeting the hotspot regions of 50 oncogenes and tumor suppressor genes and found five FGFR3 mutations in four (26.7%) out of 15 patients with thymic carcinoma (46). In this study, FGFR3 was the most frequently mutated gene, and patients carrying FGFR3 mutations showed significantly better survival. Enkner et al. reported, based on NGS with a gene panel, that 5.7% (2/35) of cases of thymic carcinoma harbored FGFR3 mutations; other NGS studies did not identify any FGFR3 mutations (7,15-17,19).

Rat sarcoma virus (RAS)/mitogen-activated protein kinase (MAPK) cascade

EGFR-mediated activation of the canonical RAS/MAPK signaling cascade is responsible for cell proliferation and death. Gene mutations in this cascade are rare in thymic carcinoma. To date, only a few studies have identified low mutation rates of genes of the RAS family, including HRAS, KRAS, NRAS, as well as RAF genes, including ARAF and BRAF (9,18-20,42,46).

PI3K/Akt/mammalian target of rapamycin (mTOR) signaling pathway

The frequency of gene alterations in this signaling pathway in thymic carcinoma is low. Alberobello et al. first reported mutations in the subunits of PI3K using thymic carcinoma cell lines (55). Several studies that used NGS have reported that mutations of PI3K, Akt, mTOR, and TSC1/2 were detected in 0.0–5.0% of thymic carcinoma (6,8,9,14,17,27,46).

PTEN gene

PTEN is a tumor suppressor gene that plays a role in growth and survival and is a negative regulator of the PI3K/Akt signaling pathway (56). There have only been a few reports on PTEN expression or PTEN mutations. Masunaga et al. analyzed TET samples, including four cases of thymic carcinoma, for the expression of PTEN, PTEN exon mutations, and PTEN promoter methylation (57). They found that the PTEN protein was immunohistochemically expressed in all thymic carcinoma cases; however, they did not detect PTEN mutations. Enkner et al. reported that the expression of PTEN was found in 30 of 31 thymic carcinoma cases (96.8%), 14 (45.2%) of which showed high expression levels (15).

GTF2I gene

Previous reports have revealed that the GTF2I point mutation (L424H) was the most frequent mutation in thymomas, especially in indolent type A and AB thymomas (6,8). However, GTF2I mutations have not been identified in thymic carcinoma (6,8,58).


Tumor mutation burden (TMB) and microsatellite instability (MSI) in thymic carcinoma

Multiple biomarkers related to immune checkpoint inhibitors (ICIs), as well as the immunohistochemical detection of PD-L1 in tumor cells, have been identified. The TMB and MSI have been clinically used in several oncologic cases.

The TMB refers to the total number of somatic non-synonymous mutations in a particular region of the tumor genome. A TMB of ≥10 mutations per megabase was reported to predict a better response to ICIs in non-small cell lung cancer (NSCLC) (59). According to the comprehensive genomic analysis of TET in The Cancer Genome Atlas (TCGA) Project, the TMB of thymic carcinoma was high (21.29 mutations per megabase), while TETs had the lowest average TMB (0.48 mutations per megabase) among adult cancers (8). Hou et al. also found a similar trend, in that the TMB of thymic carcinoma was significantly higher than that of thymoma (58). In contrast, two studies from China reported that the average TMB value of patients with thymic carcinoma was 0.72 and 0.66 mutations per megabase (10,60). According to a recent study by Kurokawa et al., who examined data from a cohort of TET cases in the United States using the Foundation Medicine Inc. research database, the prevalence of TMB-high cases was 7.0% (27). Conforti et al. conducted a phase 2 study of the combination of the anti-PD-L1 inhibitor avelumab with the anti-angiogenesis drug axitinib in patients with advanced thymic carcinoma (n=27), B3 thymoma (n=3), and mixed-type thymic carcinoma and B3 thymoma (n=2) (61). Although this population was not limited to patients with thymic carcinoma, there was a positive association between a higher TMB and the response rate.

Microsatellites are short tandem repeats scattered throughout the genome and are prone to a high mutation rate. MSI is defined as a hypermutable phenotype that occurs in genomic microsatellites in the presence of a deficient DNA mismatch repair machinery (62). Several clinical trials have revealed that patients with MSI-high colorectal cancer benefit from ICI treatment (63). The data on MSI in thymic carcinoma are limited. According to a study by Kurokawa et al., MSI-high cases accounted for 2.3% of thymic carcinoma cases, and Girard et al. reported that no MSI-high cases were found in 174 thymic carcinoma cases (9,27).


Targeted therapy in thymic carcinoma

Despite significant research efforts, the development of new drugs for thymic carcinoma is slow. Lenvatinib was approved in 2021 on the basis of a phase 2 trial, the REMORA study (64). Lenvatinib is a multitargeted kinase inhibitor of VEGFR, FGFR, KIT, and other kinases (64). The REMORA study assessed the activity of lenvatinib as a second-line treatment in 42 patients with advanced or metastatic thymic carcinoma and showed that 38.1% of patients had PR and 57.1% had stable disease (SD) with a median progression-free survival (PFS) of 9.3 months, which could be considered as the most promising results for previously advanced or metastatic thymic carcinoma. Currently, predictive biomarkers for lenvatinib activity have not been identified. Tsukaguchi et al. reported a lenvatinib-refractory thymic mucinous adenocarcinoma, whose PIK3CA mutation could be associated with resistance to lenvatinib (65).

Activating mutations in KIT and PDGFRA in GISTs are related to the response to the KIT inhibitor imatinib (66). Despite the rare KIT mutations in thymic carcinoma, several studies found that KIT-mutated thymic carcinoma showed a significant clinical response to imatinib (67,68). Sunitinib and sorafenib are multitarget tyrosine kinase inhibitors (TKIs) of KIT and other kinases. Thomas et al. observed a PR to sunitinib in 26.1% (6/23) of patients with thymic carcinoma and a SD in 65.2% (15/23); disease control was achieved in 91.3% (21/23) (69). Recently, Proto et al. conducted a phase 2 trial of sunitinib in patients with thymic carcinoma and found that 3.6% of the patients had CR, 17.9% had PR and 67.9% had SD; the objective response rate (ORR) was 21.4% and the disease control rate was 89.3% (70). At present, the correlations between the response to sunitinib and KIT mutation status are uncertain. Pagano et al. retrospectively evaluated sorafenib activity in five patients with metastatic thymic carcinoma, and reported that two patients (40.0%) achieved PR and two (40.0%) achieved SD (44). They also reported that sorafenib activity seemed independent from the KIT and PDGFRA mutation status. Perrino et al. reported the results of the Resound Trial, which examined the efficacy of regorafenib in seven patients with thymic carcinoma (71). Regorafenib potentially inhibits angiogenic and stromal receptor tyrosine kinases, VEGFR1-3, tyrosine kinase with immunoglobulin-like and EGF-like domains 2, and PDGFRB, which have been approved by the Food and Drug Administration for the treatment of colorectal cancer and GIST. SD was observed in six patients (85.7%) and progressive disease (PD) was observed in one patient (14.3%); the response was not satisfactory (71). Anlotinib is a new oral multitarget TKI targeting VEGFR1-3, FGFR1-4, PDGF-A and -B, and KIT (72). Several retrospective studies have examined the efficacy and safety of anlotinib in patients with relapsed or refractory TET (73,74). Wang et al. reported an ORR of 41.1% and a median PFS of 6 months (74).

Zucali et al. conducted a phase 2 study of everolimus, a potent oral mTOR inhibitor, in 18 patients with thymic carcinoma (75). Disease control was achieved in 77.8% of the patients (CR, n=1; PR, n=2; SD, n=11); the median PFS was 5.6 months and the median OS was 14.7 months. Hellyer et al. performed NGS with a 130-gene targeted panel on samples from 12 TET patients, including three with thymic carcinoma; however, they failed to identify correlations between detectable tumor mutations and everolimus activity (76). Predictive biomarkers for everolimus remain unclear.

Aesif et al. reported that CDK4/6 inhibitors may be considered for targeted therapy (25). Recently, Jung et al. conducted a phase 2 trial of palbociclib, an oral inhibitor of CDK4/6, in patients with recurrent or refractory advanced TETs, including 23 cases of thymic carcinoma (77). The PFS at 6 months was 52.2% and the median PFS and OS were 9.2 and 25.6 months, respectively. Two patients (8.7%) achieved PR, 16 (69.8%) achieved SD, and 18 (78.3%) achieved disease control.

Rajan et al. investigated the efficacy of cixutumumab, a fully human IgG1 monoclonal antibody that targets IGF-1R, in patients with TETs (78). The thymic carcinoma cohort was closed after enrolling 12 patients due to lack of activity. Five (41.7%) of 12 patients had SD and seven (58.3%) patients had PD; there were no objective responses and the disease control rate was 41.7%, with a median time to progression of 1.7 months and a median survival of 8.4 months. The tumor expression of IGF-1R did not appear as a good biomarker predictive response to anti-IGF treatment, as well as the raise of serum IGF-1 level.

EGFR-TKIs, a standard treatment modality for EGFR-mutated NSCLC, have not been proven to be effective in thymic carcinoma, although only a few case reports have described the clinical activity of EGFR-TKIs (79,80). In 2005, Kurup et al. conducted a phase 2 study of gefitinib and failed to demonstrate any activity in seven cases of thymic carcinoma (81). In 2008, Bedano et al. performed a phase 2 study of erlotinib plus bevacizumab in seven cases with thymic carcinoma, and reported that it was associated with a limited response (82).

Somatostatin (SST) is a naturally occurring peptide composed of 14 amino acids. Among the five SST receptors identified, the most common SST receptor expressed in human tumors is the SST2 subtype, which is visualized using radionuclide octreotide scintigraphy. The octapeptide SST analog has a high affinity for a selective SST subtype receptor (SST2). Palmieri et al. and Loehrer et al. have conducted phase 2 trials of octreotide alone or with prednisone in patients with refractory or unresectable, advanced TETs who were positive in an octreotide scan (83,84). In these studies, the ORRs of the entire TET cohort were 37.5% and 30.3%, respectively; however, thymic carcinoma treatment did not produce an objective response. Kirzinger et al. conducted another phase 2 trial of octreotide in combination with prednisone in 17 patients with primary or locally recurrent unresectable TETs, including two patients with thymic carcinoma (85). In this trial, one patient had SD and one had PD.

The wild-type Wilms tumor gene, WT1 is expressed in various types of neoplasms and has been considered to be a tumor suppressor (86,87). In recent years, WT1 has been identified as a target antigen for tumor-specific immunotherapy. Oji et al. conducted a phase 2 study of cancer immunotherapy with the WT1 peptide vaccine in patients with advanced TET, including nine patients with thymic carcinoma, which overexpressed the WT1 protein in tumor cells (88). Unfortunately, no patients achieved a CR or PR; 75.0% of patients with thymic carcinoma had SD and the remaining 25.0% of patients had PD without serious adverse events. Autoimmune complications related to thymoma, pure red cell aplasia, and myasthenia gravis occurred in two of four patients with thymoma.


Conclusions

Thymic carcinomas have a distinct genomic landscape characterized by a high prevalence of specific genes and a high TMB. Despite the rarity and histological heterogeneity of these tumors, several studies have revealed significant molecular alterations. However, there have been few suitable alterations for targeted therapy and the identified alterations seem to have little correlation with activity. Most clinical trials for thymic carcinomas have been conducted in combination with thymoma, although thymic carcinomas exhibit different biological behavior from thymoma in genetic, clinical, and immunological aspects. Continued data sharing and international collaborations would be helpful in better understanding the genomic landscape, leading to molecular targeted therapies.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://med.amegroups.com/article/view/10.21037/med-23-48/rc

Peer Review File: Available at https://med.amegroups.com/article/view/10.21037/med-23-48/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://med.amegroups.com/article/view/10.21037/med-23-48/coif). The authors have no 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. Ruffini E, Detterbeck F, Van Raemdonck D, et al. Thymic carcinoma: a cohort study of patients from the European society of thoracic surgeons database. J Thorac Oncol 2014;9:541-8. [Crossref] [PubMed]
  2. Marx A, Chan JKC, Chalabreysse L, et al. The 2021 WHO Classification of Tumors of the Thymus and Mediastinum: What Is New in Thymic Epithelial, Germ Cell, and Mesenchymal Tumors? J Thorac Oncol 2022;17:200-13. [Crossref] [PubMed]
  3. Kim DJ, Yang WI, Choi SS, et al. Prognostic and clinical relevance of the World Health Organization schema for the classification of thymic epithelial tumors: a clinicopathologic study of 108 patients and literature review. Chest 2005;127:755-61. [Crossref] [PubMed]
  4. Weksler B, Dhupar R, Parikh V, et al. Thymic carcinoma: a multivariate analysis of factors predictive of survival in 290 patients. Ann Thorac Surg 2013;95:299-303. [Crossref] [PubMed]
  5. Ahmad U, Yao X, Detterbeck F, et al. Thymic carcinoma outcomes and prognosis: results of an international analysis. J Thorac Cardiovasc Surg 2015;149:95-100, 101.e1-2.
  6. 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]
  7. 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]
  8. 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]
  9. 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]
  10. Yang J, Zhang B, Guan W, et al. Molecular genetic characteristics of thymic epithelial tumors with distinct histological subtypes. Cancer Med 2023;12:10575-86. [Crossref] [PubMed]
  11. Hirabayashi H, Fujii Y, Sakaguchi M, et al. p16INK4, pRB, p53 and cyclin D1 expression and hypermethylation of CDKN2 gene in thymoma and thymic carcinoma. Int J Cancer 1997;73:639-44. [Crossref] [PubMed]
  12. Tateyama H, Eimoto T, Tada T, et al. p53 protein expression and p53 gene mutation in thymic epithelial tumors. An immunohistochemical and DNA sequencing study. Am J Clin Pathol 1995;104:375-81. [Crossref] [PubMed]
  13. Moreira AL, Won HH, McMillan R, et al. Massively parallel sequencing identifies recurrent mutations in TP53 in thymic carcinoma associated with poor prognosis. J Thorac Oncol 2015;10:373-80. [Crossref] [PubMed]
  14. Shitara M, Okuda K, Suzuki A, et al. Genetic profiling of thymic carcinoma using targeted next-generation sequencing. Lung Cancer 2014;86:174-9. [Crossref] [PubMed]
  15. Enkner F, Pichlhöfer B, Zaharie AT, et al. Molecular Profiling of Thymoma and Thymic Carcinoma: Genetic Differences and Potential Novel Therapeutic Targets. Pathol Oncol Res 2017;23:551-64. [Crossref] [PubMed]
  16. Asao T, Fujiwara Y, Sunami K, et al. Medical treatment involving investigational drugs and genetic profile of thymic carcinoma. Lung Cancer 2016;93:77-81. [Crossref] [PubMed]
  17. Saito M, Fujiwara Y, Asao T, et al. The genomic and epigenomic landscape in thymic carcinoma. Carcinogenesis 2017;38:1084-91. [Crossref] [PubMed]
  18. Sakane T, Murase T, Okuda K, et al. A mutation analysis of the EGFR pathway genes, RAS, EGFR, PIK3CA, AKT1 and BRAF, and TP53 gene in thymic carcinoma and thymoma type A/B3. Histopathology 2019;75:755-66. [Crossref] [PubMed]
  19. Sakane T, Sakamoto Y, Masaki A, et al. Mutation Profile of Thymic Carcinoma and Thymic Neuroendocrine Tumor by Targeted Next-generation Sequencing. Clin Lung Cancer 2021;22:92-99.e4. [Crossref] [PubMed]
  20. Wang H, Xu X, Luo L, et al. Mutational landscape of thymic epithelial tumors in a Chinese population: insights into potential clinical implications. Gland Surg 2021;10:1410-7. [Crossref] [PubMed]
  21. Xu S, Li X, Zhang H, et al. Frequent Genetic Alterations and Their Clinical Significance in Patients With Thymic Epithelial Tumors. Front Oncol 2021;11:667148. [Crossref] [PubMed]
  22. Szpechcinski A, Szolkowska M, Winiarski S, et al. Targeted Next-Generation Sequencing of Thymic Epithelial Tumours Revealed Pathogenic Variants in KIT, ERBB2, KRAS, and TP53 in 30% of Thymic Carcinomas. Cancers (Basel) 2022;14:3388. [Crossref] [PubMed]
  23. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochim Biophys Acta 1998;1378:F115-77. [PubMed]
  24. Krimpenfort P, Ijpenberg A, Song JY, et al. p15Ink4b is a critical tumour suppressor in the absence of p16Ink4a. Nature 2007;448:943-6. [Crossref] [PubMed]
  25. Aesif SW, Aubry MC, Yi ES, et al. Loss of p16INK4A Expression and Homozygous CDKN2A Deletion Are Associated with Worse Outcome and Younger Age in Thymic Carcinomas. J Thorac Oncol 2017;12:860-71. [Crossref] [PubMed]
  26. Petrini I, Meltzer PS, Zucali PA, et al. Copy number aberrations of BCL2 and CDKN2A/B identified by array-CGH in thymic epithelial tumors. Cell Death Dis 2012;3:e351. [Crossref] [PubMed]
  27. 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]
  28. Laitman Y, Newberg J, Molho RB, et al. The spectrum of tumors harboring BAP1 gene alterations. Cancer Genet 2021;256-257:31-5. [Crossref] [PubMed]
  29. Giaccone G, Kim C, Thompson J, et al. Pembrolizumab in patients with thymic carcinoma: a single-arm, single-centre, phase 2 study. Lancet Oncol 2018;19:347-55. [Crossref] [PubMed]
  30. He Y, Ramesh A, Gusev Y, et al. Molecular predictors of response to pembrolizumab in thymic carcinoma. Cell Rep Med 2021;2:100392. [Crossref] [PubMed]
  31. Angirekula M, Chang SY, Jenkins SM, et al. CD117, BAP1, MTAP, and TdT Is a Useful Immunohistochemical Panel to Distinguish Thymoma from Thymic Carcinoma. Cancers (Basel) 2022;14:2299. [Crossref] [PubMed]
  32. Yarden Y, Pines G. The ERBB network: at last, cancer therapy meets systems biology. Nat Rev Cancer 2012;12:553-63. [Crossref] [PubMed]
  33. Girard N, Shen R, Guo T, et al. Comprehensive genomic analysis reveals clinically relevant molecular distinctions between thymic carcinomas and thymomas. Clin Cancer Res 2009;15:6790-9. [Crossref] [PubMed]
  34. Henley JD, Koukoulis GK, Loehrer PJ Sr. Epidermal growth factor receptor expression in invasive thymoma. J Cancer Res Clin Oncol 2002;128:167-70. [Crossref] [PubMed]
  35. Suzuki E, Sasaki H, Kawano O, et al. Expression and mutation statuses of epidermal growth factor receptor in thymic epithelial tumors. Jpn J Clin Oncol 2006;36:351-6. [Crossref] [PubMed]
  36. Meister M, Schirmacher P, Dienemann H, et al. Mutational status of the epidermal growth factor receptor (EGFR) gene in thymomas and thymic carcinomas. Cancer Lett 2007;248:186-91. [Crossref] [PubMed]
  37. Yoh K, Nishiwaki Y, Ishii G, et al. Mutational status of EGFR and KIT in thymoma and thymic carcinoma. Lung Cancer 2008;62:316-20. [Crossref] [PubMed]
  38. Aisner SC, Dahlberg S, Hameed MR, et al. Epidermal growth factor receptor, C-kit, and Her2/neu immunostaining in advanced or recurrent thymic epithelial neoplasms staged according to the 2004 World Health Organization in patients treated with octreotide and prednisone: an Eastern Cooperative Oncology Group study. J Thorac Oncol 2010;5:885-92. [Crossref] [PubMed]
  39. Weissferdt A, Lin H, Woods D, et al. HER family receptor and ligand status in thymic carcinoma. Lung Cancer 2012;77:515-21. [Crossref] [PubMed]
  40. Avci N, Cecener G, Deligonul A, et al. Molecular markers for patients with thymic malignancies: not feasible at present? Asian Pac J Cancer Prev 2014;15:3457-60. [Crossref] [PubMed]
  41. Zhan P, Chen X, Wu XY, et al. Mutation analysis of the EGFR gene and its downstream signaling pathway in thymic carcinoma patients from a Chinese Han population. Clin Respir J 2018;12:601-7. [Crossref] [PubMed]
  42. Tiseo M, Damato A, Longo L, et al. Analysis of a panel of druggable gene mutations and of ALK and PD-L1 expression in a series of thymic epithelial tumors (TETs). Lung Cancer 2017;104:24-30. [Crossref] [PubMed]
  43. Pan CC, Chen PC, Chiang H. KIT (CD117) is frequently overexpressed in thymic carcinomas but is absent in thymomas. J Pathol 2004;202:375-81. [Crossref] [PubMed]
  44. Pagano M, Sierra NM, Panebianco M, et al. Sorafenib efficacy in thymic carcinomas seems not to require c-KIT or PDGFR-alpha mutations. Anticancer Res 2014;34:5105-10. [PubMed]
  45. Terzi N, Yilmaz I, Batur S, et al. C-KIT mutation in thymic carcinomas. Pol J Pathol 2020;71:120-6. [Crossref] [PubMed]
  46. Asselta R, Di Tommaso L, Perrino M, et al. Mutation profile and immunoscore signature in thymic carcinomas: An exploratory study and review of the literature. Thorac Cancer 2021;12:1271-8. [Crossref] [PubMed]
  47. Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors. Science 2003;299:708-10. [Crossref] [PubMed]
  48. Moasser MM. The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene 2007;26:6469-87. [Crossref] [PubMed]
  49. Pan CC, Chen PC, Wang LS, et al. Expression of apoptosis-related markers and HER-2/neu in thymic epithelial tumours. Histopathology 2003;43:165-72. [Crossref] [PubMed]
  50. Wang P, Mak VC, Cheung LW. Drugging IGF-1R in cancer: New insights and emerging opportunities. Genes Dis 2023;10:199-211. [Crossref] [PubMed]
  51. Zucali PA, Petrini I, Lorenzi E, et al. Insulin-like growth factor-1 receptor and phosphorylated AKT-serine 473 expression in 132 resected thymomas and thymic carcinomas. Cancer 2010;116:4686-95. [Crossref] [PubMed]
  52. Ornitz DM, Marie PJ. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev 2002;16:1446-65. [Crossref] [PubMed]
  53. Chew NJ, Nguyen EV, Su SP, et al. FGFR3 signaling and function in triple negative breast cancer. Cell Commun Signal 2020;18:13. [Crossref] [PubMed]
  54. Dienstmann R, Rodon J, Prat A, et al. Genomic aberrations in the FGFR pathway: opportunities for targeted therapies in solid tumors. Ann Oncol 2014;25:552-63. [Crossref] [PubMed]
  55. Alberobello AT, Wang Y, Beerkens FJ, et al. PI3K as a Potential Therapeutic Target in Thymic Epithelial Tumors. J Thorac Oncol 2016;11:1345-56. [Crossref] [PubMed]
  56. Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol 2009;4:127-50. [Crossref] [PubMed]
  57. Masunaga A, Omatsu M, Kunimura T, et al. Expression of PTEN and its pseudogene PTENP1, and promoter methylation of PTEN in non-tumourous thymus and thymic tumours. J Clin Pathol 2017;70:690-6. [Crossref] [PubMed]
  58. Hou X, Lin S, Liu Y, et al. Analysis of the tumor microenvironment and mutation burden identifies prognostic features in thymic epithelial tumors. Am J Cancer Res 2022;12:2387-96. [PubMed]
  59. Ready N, Hellmann MD, Awad MM, et al. First-Line Nivolumab Plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer (CheckMate 568): Outcomes by Programmed Death Ligand 1 and Tumor Mutational Burden as Biomarkers. J Clin Oncol 2019;37:992-1000. [Crossref] [PubMed]
  60. Yang W, Chen S, Cheng X, et al. Characteristics of genomic mutations and signaling pathway alterations in thymic epithelial tumors. Ann Transl Med 2021;9:1659. [Crossref] [PubMed]
  61. Conforti F, Zucali PA, Pala L, et al. Avelumab plus axitinib in unresectable or metastatic type B3 thymomas and thymic carcinomas (CAVEATT): a single-arm, multicentre, phase 2 trial. Lancet Oncol 2022;23:1287-96. [Crossref] [PubMed]
  62. Baretti M, Le DT. DNA mismatch repair in cancer. Pharmacol Ther 2018;189:45-62. [Crossref] [PubMed]
  63. Zhao P, Li L, Jiang X, et al. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J Hematol Oncol 2019;12:54. [Crossref] [PubMed]
  64. Sato J, Satouchi M, Itoh S, et al. Lenvatinib in patients with advanced or metastatic thymic carcinoma (REMORA): a multicentre, phase 2 trial. Lancet Oncol 2020;21:843-50. [Crossref] [PubMed]
  65. Tsukaguchi A, Ihara S, Yasuoka H, et al. Lenvatinib-refractory thymic mucinous carcinoma with PIK3CA mutation. Int Cancer Conf J 2022;12:36-40. [Crossref] [PubMed]
  66. Joensuu H, Wardelmann E, Sihto H, et al. Effect of KIT and PDGFRA Mutations on Survival in Patients With Gastrointestinal Stromal Tumors Treated With Adjuvant Imatinib: An Exploratory Analysis of a Randomized Clinical Trial. JAMA Oncol 2017;3:602-9. [Crossref] [PubMed]
  67. 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]
  68. Buti S, Donini M, Sergio P, et al. Impressive response with imatinib in a heavily pretreated patient with metastatic c-KIT mutated thymic carcinoma. J Clin Oncol 2011;29:e803-5. [Crossref] [PubMed]
  69. Thomas A, Rajan A, Berman A, et al. Sunitinib in patients with chemotherapy-refractory thymoma and thymic carcinoma: an open-label phase 2 trial. Lancet Oncol 2015;16:177-86. [Crossref] [PubMed]
  70. Proto C, Manglaviti S, Lo Russo G, et al. STYLE (NCT03449173): A Phase 2 Trial of Sunitinib in Patients With Type B3 Thymoma or Thymic Carcinoma in Second and Further Lines. J Thorac Oncol 2023;18:1070-81. [Crossref] [PubMed]
  71. Perrino M, De Pas T, Bozzarelli S, et al. Resound Trial: A phase 2 study of regorafenib in patients with thymoma (type B2-B3) and thymic carcinoma previously treated with chemotherapy. Cancer 2022;128:719-26. [Crossref] [PubMed]
  72. Syed YY. Anlotinib: First Global Approval. Drugs 2018;78:1057-62. [Crossref] [PubMed]
  73. Li S, Zhou H, Zhang X, et al. The Efficacy and Safety of Anlotinib Alone and in Combination with Other Drugs in Previously Treated Advanced Thymic Epithelia Tumors: A Retrospective Analysis. Recent Pat Anticancer Drug Discov 2023;18:528-37. [Crossref] [PubMed]
  74. Wang CL, Zhao YZ, Zhang Q, et al. Anlotinib in patients with relapsed or refractory thymic epithelial tumors: a study of 50 cases. Anticancer Drugs 2023;34:852-6. [Crossref] [PubMed]
  75. Zucali PA, De Pas T, Palmieri G, et al. Phase II Study of Everolimus in Patients With Thymoma and Thymic Carcinoma Previously Treated With Cisplatin-Based Chemotherapy. J Clin Oncol 2018;36:342-9. [Crossref] [PubMed]
  76. Hellyer JA, Ouseph MM, Padda SK, et al. Everolimus in the treatment of metastatic thymic epithelial tumors. Lung Cancer 2020;149:97-102. [Crossref] [PubMed]
  77. Jung HA, Kim M, Kim HS, et al. A Phase 2 Study of Palbociclib for Recurrent or Refractory Advanced Thymic Epithelial Tumors (KCSG LU17-21). J Thorac Oncol 2023;18:223-31. [Crossref] [PubMed]
  78. Rajan A, Carter CA, Berman A, et al. Cixutumumab for patients with recurrent or refractory advanced thymic epithelial tumours: a multicentre, open-label, phase 2 trial. Lancet Oncol 2014;15:191-200. [Crossref] [PubMed]
  79. Pedersini R, Vattemi E, Lusso MR, et al. Erlotinib in advanced well-differentiated thymic carcinoma with overexpression of EGFR: a case report. Tumori 2008;94:849-52. [Crossref] [PubMed]
  80. Brown LA. Successful Use of Erlotinib in Treating Recurrent Thymic Carcinoma: A Case Report. World J Oncol 2013;4:214-6. [Crossref] [PubMed]
  81. Kurup A, Burns M, Dropcho S, et al. Phase II study of gefitinib treatment in advanced thymic malignancies. J Clin Oncol 2005;23:7068. [Crossref]
  82. Bedano PM, Perkins S, Burns M, et al. A phase II trial of erlotinib plus bevacizumab in patients with recurrent thymoma or thymic carcinoma. J Clin Oncol 2008;26:19087. [Crossref]
  83. Palmieri G, Montella L, Martignetti A, et al. Somatostatin analogs and prednisone in advanced refractory thymic tumors. Cancer 2002;94:1414-20. [Crossref] [PubMed]
  84. Loehrer PJ Sr, Wang W, Johnson DH, et al. Octreotide alone or with prednisone in patients with advanced thymoma and thymic carcinoma: an Eastern Cooperative Oncology Group Phase II Trial. J Clin Oncol 2004;22:293-9. [Crossref] [PubMed]
  85. Kirzinger L, Boy S, Marienhagen J, et al. Octreotide LAR and Prednisone as Neoadjuvant Treatment in Patients with Primary or Locally Recurrent Unresectable Thymic Tumors: A Phase II Study. PLoS One 2016;11:e0168215. [Crossref] [PubMed]
  86. Call KM, Glaser T, Ito CY, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell 1990;60:509-20. [Crossref] [PubMed]
  87. Oka Y, Tsuboi A, Elisseeva OA, et al. WT1 as a novel target antigen for cancer immunotherapy. Curr Cancer Drug Targets 2002;2:45-54. [Crossref] [PubMed]
  88. Oji Y, Inoue M, Takeda Y, et al. WT1 peptide-based immunotherapy for advanced thymic epithelial malignancies. Int J Cancer 2018;142:2375-82. [Crossref] [PubMed]
doi: 10.21037/med-23-48
Cite this article as: Sakane T, Haneda H, Okuda K. Insights into molecular aspects and targeted therapy of thymic carcinoma: a narrative review. Mediastinum 2024;8:36.

Download Citation