Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

ITMIG DEFINITIONS AND POLICIES

4UBUFPGUIF"SUPG(FOFUJD"MUFSBUJPOTJO5IZNJD
&QJUIFMJBM5VNPST
Arun Rajan, MD,* Nicolas Girard, MD, PhD,† and Alexander Marx, MD‡

Abstract: The rapid advent of technology in recent years has resulted typically outnumber and tightly intermingle with the neoplas-
in a substantial increase in our knowledge of the molecular underpin- tic epithelial cells and largely preclude classical or array-based
nings of thymic epithelial tumors. In addition to previously described comparative genomic hybridization (CGH) approaches except
chromosomal aberrations and alterations in DNA methylation, genome in rare cases in which epithelial content is unusually high or in
sequencing has helped unravel hitherto unknown mutations in these which thymocytes can be removed through short-term cell cul-
tumors. Attempts are also being made to develop gene signatures to help ture of neoplastic epithelial cells.8–11 Fluorescence in situ hybrid-
in the identification of patients likely to benefit from adjuvant therapy. ization analyses have been sparse as well.8,12,13 However, with
Some of the recently identified genetic alterations have the potential the advent of newer and more sensitive techniques in molecular
to serve as targets for biological therapy, thus opening newer avenues biology (e.g., next generation sequencing) and bioinformatics,
for treatment of thymic epithelial tumors and increasing the number of there has been and will be an incremental increase in the avail-
effective options for treatment of recurrent or refractory disease. ability of high-quality data that has helped in expanding our
understanding of the molecular pathogenesis of these diseases.
Key Words: Thymoma, Thymic carcinoma, Methylation, Chromosomal Development of newer therapies based on an understanding of
abnormalities, Gene expression, Sequencing, Gene signatures genomic alterations could potentially revolutionize treatment of
(J Thorac Oncol. 2014;9: S131–S136) TETs and result in an improvement in clinical outcomes. This
review provides an overview of genetic alterations associated
with TETs with a focus on recent discoveries that could have an
impact on the management of thymic cancers.
T hymic epithelial tumors (TETs) are rare mediastinal
cancers that have traditionally been treated using a mul-
timodality approach consisting of surgery, radiotherapy, and
CHROMOSOMAL ABNORMALITIES
Copy number (CN) gains and losses of various chromo-
platinum-based chemotherapy.1 Effective treatment options
somes have been well described in TETs, and the frequency
are limited for patients with recurrent disease. The World
of these changes is higher in the more aggressive histological
Health Organization (WHO) classification is the most widely
subtypes. Based on the pattern of chromosomal changes, TETs
used system for the histological classification of TETs.2 Based
can be clustered into various groups, demonstrating a correla-
on thymic epithelial cell morphology, the degree of thymocyte
tion between genetic findings and WHO-based histology, with
involvement and the extent of epithelial atypia TETs are clas-
overlapping alterations between type AB and B2 thymoma,
sified into the following subtypes: A, AB, B1, B2, B3, and C.
B2 and B3 thymoma, and B3 thymomas and thymic carcino-
Efforts such as The Cancer Genome Atlas (TCGA)
mas.8–11,14 In short, 6q25.2–25.3 loss is observed in all sub-
research network have helped in providing a deep understanding
types except B1 thymomas (a paucity of B1 thymomas among
of the molecular alterations associated with a variety of tumors.3–7
cases analyzed might have had a bearing on this result); 1q
In comparison, until recently, scant information was available
gains are common in type B2, B3 thymomas and thymic car-
on genomic changes associated with TETs. Major challenges in
cinomas; in addition, thymic carcinomas present with 1q, 4, 5,
obtaining genetic data still exist, especially from the more com-
7, 8, 9q, 12, 15, 17q, 18, 20 gains, and 3p, 6, 6p23, 9p, 13q,
mon thymoma subtypes (AB, B1, and B2) due to the presence
14, 16q, 17p losses.9–11,14 The biological significance of these
of a significant number of non-neoplastic thymocytes. The latter
changes is yet to be determined in the vast majority of cases.
However, it is known that the 6p23 region encompasses the
*Thoracic and Gastrointestinal Oncology Branch, National Cancer Institute, FOXC1 tumor suppressor gene, and chromosomal loss is cor-
Bethesda, MD; †Respiratory Medicine, Thoracic Oncology Service, related with lower protein expression. Patients with FOXC1-
Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France; and negative tumors had a shorter time to progression and a trend
‡Institute of Pathology, University Medical Centre Mannheim, University
of Heidelberg, Mannheim, Germany.
for a shorter disease-related survival.15
Disclosure: The authors declare no conflict of interest.
Address for correspondence: Arun Rajan, MD, Thoracic and Gastrointestinal ALTERATIONS IN DNA METHYLATION
Oncology Branch, National Cancer Institute, 10 Center Drive, 10-CRC, Silencing of tumor suppressor genes such as FHIT,
Room 4-5330, Bethesda, MD 20892. E-mail. rajana@mail.nih.gov
Copyright © 2014 by the International Association for the Study of Lung
MLH1, and E-cad by promoter hypermethylation has been
Cancer described in TETs.16 These changes along with DNA hypo-
ISSN: 1556-0864/14/0909-S131 methylation have been correlated with Masaoka stage and

Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFS S131


Rajan et al. Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFS

WHO subtype of the TET. Other genes affected by aber- of COL1A1 was decreased 13-fold. This is the first study to
rant methylation include DAP-K, p16, MGMT, and HPP1.17 attempt a correlation between expression of individual genes
MGMT methylation and loss of its protein expression has and clinical factors in TETs. Validation of data obtained from
been reported to be significantly more frequent in thymic car- this study has the potential to identify newer therapeutics tar-
cinoma than in thymoma (74% versus 29%) and in advanced gets such as AKR1B10.24
TETs (stages III/IV) compared with early stage tumors (stages
I/II).18 Methylation of the promoter region of CDKN2 has Gene Signatures
been observed in up to 14% of thymomas and 25% of thymic Traditional prognostic factors associated with TETs
carcinomas.19 Immunohistochemical analysis of 132 TETs have included Masaoka stage, histological subtype, and the
demonstrated CN loss of CDKN2A accompanied by lack completeness of surgical resection.25 Most attempts to identify
of expression of its related protein p16INK4 and identified prognostic and predictive biomarkers have focused on correla-
tumors with poor prognosis.20 tions with the stage and histological subtype of disease.9,26–28
A deeper understanding of alterations in DNA methyla- The first study to attempt a correlation between gene expres-
tion can have important therapeutic implications. For example, sion and the risk of development of metastases was reported
MGMT hypermethylation is associated with a greater degree in 2013.29 In a retrospective study using archival samples of
of sensitivity to alkylating agents.21 Dysregulation of the thymomas from multiple institutions, gene expression levels
cyclin-dependant kinase pathway can potentially be offset by were used to categorize tumors into two groups based on the
using cyclin-dependent kinase inhibitors such as milciclib. An probability of developing metastases. This study included
ongoing phase II study is evaluating the efficacy of milciclib 111 cases of thymoma (36 in the training set and 75 in the
in thymic carcinoma (NCT01011439). Forty-three patients validation set). Based on the prediction analysis of microar-
have been enrolled and treated to date including 26 patients rays (PAMs), 19 differentially expressed genes and four refer-
with thymic carcinoma and nine patients with B3 thymoma. ence genes were selected for quantitative reverse transcriptase
Of 30 evaluable patients, 14 have met the prespecified primary polymerase chain reaction analysis. The primary end point of
end point of progression-free survival at 3 months (PFS-3 rate the study was to determine whether a gene signature could
= 47%; 95% confidence interval [CI] 28.3–65.7%) including predict 5- and 10-year metastasis-free survival (MFS). A nine-
one partial response.22 gene signature was identified that could identify patients at
low- and high-risk for metastasis with 5- and 10-year MFS of
GENOMIC PROFILING 77% and 26% (p = 0.0047) and 97% and 30% (p = 0.0004)
in the training and validation sets, respectively. In univariate
Gene Expression Analysis Cox regression analysis, the nine-gene signature was a strong
In an effort to overcome the limitations associated with predictor of the development of metastasis with a hazard ratio
traditional histological classification of TETs, efforts have (HR) of 8.33 (95% CI = 1.99–34.9; p = 0.004) compared with
been made to correlate the results of gene expression deter- a HR of 2.51 (95% CI = 1.22–5.15; p = 0.012) for presence
mined by whole genome gene expression analysis with clinical of residual disease and a HR of 2.09 (95% CI = 0.95–4.59;
parameters such as stage of disease. Badve et al.23 conducted p = 0.067) for Masaoka stage. In multivariate analysis, the
a retrospective analysis in 34 patients with type AB and B thy- nine-gene signature was the only independent predictor of the
moma using RNA extracted from fresh frozen tumors. Ten of risk of developing metastasis (HR 5.26, 95% CI = 1.12–24.8;
34 patients had received chemotherapy and eight patients had p = 0.036). Genes with increased expression in tumors with
received radiation therapy. Eight patients were known to have a higher metastatic potential included AKR1B10, JPH1, and
metastatic disease either at diagnosis or developed it during the NGB. Conversely, genes with decreased expression in tumors
follow-up period. In addition, although all patients underwent with high metastatic potential included DACT3, SLC9A2,
surgery with curative intent, eight patients developed disease PDGFRL, FCGBP, PRRX1, and SERPINF1.
relapse during the follow-up period. After gene expression The practical implications of development of prognos-
analysis using Partek Genomics Suite and Ingenuity Pathways tic biomarkers such as the gene signature illustrated above
Analysis, four molecular clusters of tumors were identified are the identification of patients at high risk for recurrence
by unsupervised cluster analysis of 8260 genes that showed who might benefit from administration of appropriate adju-
unique gene expression profiles. Supervised cluster analysis vant therapy and the potential to identify novel drug targets
using 15 genes with the highest positive and negative fold for patients at higher risk for development of unresectable,
changes helped in the identification of three clusters of his- metastatic disease. However, significant challenges need to be
tologically heterogeneous tumors. Analysis of clinical param- overcome before widespread adoption of gene signatures in
eters such as stage of disease and presence of metastases did clinical practice, especially for rare diseases like TETs. These
not show a correlation with either histological subtype or the include the need for validation of results in a prospective set-
newly identified gene expression clusters. However, ingenuity ting, a clear demonstration of benefit over widely used clini-
pathway analysis resulted in the identification of differentially cal parameters such as Masaoka stage, histological subtype
expressed genes in patients with or without metastatic dis- and completeness of resection, and the ease of performing the
ease. In tumors obtained from the metastasis-positive group, gene expression assay. Some of the limitations that might oth-
there was a sixfold and threefold increase in the expression erwise preclude mass validation of a gene signature for TETs
of AKR1B10 and JPH1, respectively, whereas expression can be surmounted by performing prospective trials involving

S132 Copyright © 2014 by the International Association for the Study of Lung Cancer
Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFSGenetic Alterations in Thymic Epithelial Tumors

multiple institutions or in a cooperative group setting to maxi- clinical trials evaluating EGFR inhibitors have yielded disap-
mize accrual of cases. pointing results.34,35
Despite these observations, a few recently completed
Whole Genome and Transcriptome Sequencing clinical trials of targeted therapy in TETs have yielded prom-
The first reported TET to be analyzed using whole ising results. The multikinase inhibitor, sunitinib, has been
genome and transcriptome sequencing involved a surgi- evaluated in a phase II clinical trial in TETs. Among 23 evalu-
cally resected stage IVA, B3 thymoma.30 In addition to able patients with thymic carcinoma, sunitinib demonstrated
well-described changes in gene CN, a number of previously a response rate of 26% and disease control rate of 91%.36
unreported observations were made including a translocation The response to sunitinib was independent of the mutational
involving chromosome 11 and the X chromosome, t(11;X), status of KIT and seems to be partly related to an antiangio-
and mutations in BCOR, FBN3, PCNXL3, PHF15, SFXN3, genic effect. Cixutumumab, a monoclonal antibody–directed
SRGAP1, VN1R5, and WDR70. The pathogenic role of FBN3, against the insulin-like growth factor-1 receptor, has been
PCNXL3, SFXN3, SRGAP1, VN1R5, and WDR70 is unclear. evaluated in a phase II trial and demonstrated a 89% disease
However, PHF15 belongs to the PHF protein family, several control rate in patients with relapsed or refractory thymoma.37
members of which affect chromatin-mediated gene regulation Results from selected clinical trials of targeted therapy for
and BCOR is a corepressor of several transcription factors. TETs are presented in Table 1.34–41
The mutation rate of 0.72/Mb seen in this case (including sin- In the aforementioned trials, patient selection was not
gle nucleotide variations and insertions/deletions) was com- based on the presence of targetable mutations. Expression of
parable with that observed in more aggressive malignancies potential drug targets such as the vascular endothelial growth
such as breast cancer and myelogenous leukemia. However, factor receptor and the insulin-like growth factor 1-recep-
no mutations were observed in well-characterized cancer tor that could potentially explain the activity of drugs like
genes. This report highlights the fact that TETs have a unique sunitinib and cixutumumab are being evaluated in TETs
mutational profile that is not detectable by gene panels used (Table 2).12,13,42–50 Greater use of genomic analysis to optimize
for molecular profiling of common solid tumors, and strate- patient selection based on the presence of targetable mutations
gies such as whole exome sequencing might be better suited has the potential to improve upon the results obtained so far
to identify recurrent mutations in this rare tumor type, some of with targeted therapy.
which may represent targets for treatment.
Role of Newer Methods of Genomic and
IDENTIFICATION OF DRUG TARGETS Integrative Analysis to Identify Drug Targets
Initial experiences of the use of targeted therapy in Because strategies to identify genetic mutations often
TETs have been disappointing. The reasons for these negative seen in other solid tumors have not yielded promising results
results have become clear with the advent of technology and for TETs, newer panels of tests need to be developed that
the relative ease of performing mutational analysis in recent focus on identifying genetic alterations that might be unique
years. However, a subset of patients with TETs do benefit from to TETs.
targeted therapy, highlighting the need to gain greater insight Recently, differentially expressed miRNAs have been
into TET biology to help select patients for targeted therapy. identified in TETs. Upregulation of miR-21-5p, miR-34b-5p,
These concepts are illustrated by the following examples. and miR-141-3p, as well as down-regulation of miR-486-5p
KIT overexpression is frequently observed in thy- and miR-145-5p has been observed in TETs in comparison
mic carcinomas (75–80%), whereas KIT gene mutations are with normal tissue.51 miR-145-5p down-regulation is thought
found in less than 10% of cases.9,31 Based on the type of KIT to lead to EGFR signaling pathway overexpression. miR-
mutation, TETs demonstrate varying degrees of sensitivity to 128, miR-142-5p, and miR-181c-5p are also differentially
tyrosine kinase inhibitors. In order of sensitivity to sunitinib expressed in thymoma versus thymic carcinoma. Interestingly,
and imatinib, these mutations include: W557R, V559A, and among the miRNAs down-regulated in thymic carcinoma,
V560del (all on exon 11 and highly sensitive to sunitinib and some putatively target BIRC3, SCYA20, and MYC.52
imatinib); Y553N and D579del (both on exon 11 and highly Differential expression of antiapoptotic genes has also been
sensitive to imatinib); E490K on exon 9 (highly sensitive to identified in B3 thymoma and squamous cell carcinoma of the
sunitinib and moderately sensitive to imatinib); H697Y on thymus. These results have raised the possibility of therapeu-
exon 14 (highly sensitive to sunitinib and low sensitivity to tic intervention with drugs targeting inhibitors of apoptosis
imatinib); L576P on exon 11 (moderate sensitivity to sunitinib and BIRC3.52 In another study, miR-515 overexpression was
and low sensitivity to imatinib); and D820E on exon 17 (resis- observed in type A-AB thymomas; this may result in PTEN
tant to sunitinib and imatinib).1 The rarity and spectrum of KIT inhibition, which could potentially be targeted by PIK3CA/
mutations in thymic carcinoma explains the lack of activity of AKT inhibitors.53
imatinib in phase II trials where patient selection was based Array CGH in 59 TETs reported recurrent CN gain of
on histology (B3 thymomas and thymic carcinomas) or KIT BCL2, providing further evidence for the role of an antiapop-
staining by immunohistochemistry and not genotyping.32,33 A totic strategy as a major hallmark of TETs.20 Moreover, TET
similar pattern of overexpression of epidermal growth factor cell lines were reported to be sensitive to siRNA knockdown
receptor (EGFR) and a paucity of sensitizing mutations in the of the antiapoptotic molecules BCL2 and MCL1. Gx15-070, a
EGFR gene has been observed in TETs.31 Not surprisingly, pan-BCL2 inhibitor, induced autophagy-dependent necrosis in

Copyright © 2014 by the International Association for the Study of Lung Cancer S133
Rajan et al. Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFS

TABLE 1. 4FMFDUFE$MJOJDBM5SJBMTPG5BSHFUFE5IFSBQJFTGPS5IZNJD&QJUIFMJBM5VNPST


Time to Progression Overall Survival
Intervention Evaluable Patients Responses (months) (months) Reference
Gefitinib 26 1 4 NR 34

Thymoma 19 NR NR NR
Thymic carcinoma 7 NR NR NR
Erlotinib + bevacizumab 18 0 NR Not reached 35

Thymoma 11 0 NR Not reached


Thymic carcinoma 7 0 NR Not reached
Sunitinib 39 7 NR NR 36

Thymoma 16 1 8.5a Not reached


Thymic carcinoma 23 6 6.7a 16.3
Cixutumumab 49 5 8.2 16.2 37

Thymoma 37 5 9.9 27.5


Thymic carcinoma 12 0 1.7 8.4
Imatinib 15 0 3a Nor reached 38

Thymoma 12 0 NRa NR
Thymic carcinoma 3 0 NRa NR
Saracatinib 21 0 NRa NR 39

Thymoma 14 NR 3.4a Not reached


Thymic carcinoma 7 NR 1.4a Not reached
Belinostat 40 2 NR NR 40

Thymoma 24 2 11.4 Not reached


Thymic carcinoma 16 0 2.7 12.4
Everolimus 35 4 12.1a 24 41

Thymoma 23 1 NRa NR
Thymic carcinoma 12 3 NRa NR
Progression-free survival.
a

NR, Not reported.

TABLE 2. (FOF"NQMJmDBUJPOBOE1SPUFJO0WFSFYQSFTTJPOJO


IMPACT OF THE GENOMICS REVOLUTION AND
5IZNJD&QJUIFMJBM5VNPST LESSONS FOR CLINICAL PRACTICE
In view of the rarity of TETs, translation of preclinical
Thymic
Gene Thymoma Carcinoma References observations into newer therapies poses unique challenges.
One approach to validate the concept of personalized medi-
EGFR 13, 42, 43
cine in thymic malignancies is the development of open-label,
Amplification 20% 25% multicenter phase II trials using high-throughput genome anal-
Overexpression 23% 67–100% ysis (multiplex mutation sequencing, CGH arrays, next gen-
HER2 12, 44
eration sequencing) as a therapeutic decision tool to identify
Amplification 6% 0–4% patients for targeted therapy directed against specific molecu-
Overexpression 53–58% lar alterations identified in their tumors. TETs have been inte-
KIT 45–47
grated into a few clinical trials using this approach such as
Overexpression 0–4% 46–86% the recently completed CUSTOM trial (NCT01306045), the
IGF-1R 48, 49
first prospective study of molecular profiling to determine eli-
Overexpression 14–43% 86–88% gibility for targeted therapy in patients with TETs.56 Tumors
VEGF 50 were analyzed for molecular changes including mutations in
Overexpression 32% 63% EGFR, KRAS, HRAS, NRAS, BRAF, PIK3CA, AKT, PTEN,
ERBB2, KIT, and PDGFRA, amplification of HER2 or ALK
IGF-1R, insulin-like growth factor-1 receptor.
translocations in a CLIA-certified laboratory, and patients
were assigned to one of five treatment groups on the basis
TET cells through a mechanism involving mammalian target of these changes. This study showed that the genomic altera-
of rapamycin (mTOR) pathways, and inhibited TET xenograft tions described above were uncommon in TETs and included
growth. Hence, mTOR targeting may represent a clinically HER2 amplification in 7.7% of samples analyzed, and a
relevant approach for treatment of advanced TETs.54,55 Table 3 mutational frequency of 4.7% in HRAS and 1.4% in PIK3CA
summarizes selected molecular alterations in TETs. and EGFR genes. This trial confirms the uniqueness of the

S134 Copyright © 2014 by the International Association for the Study of Lung Cancer
Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFSGenetic Alterations in Thymic Epithelial Tumors

TABLE 3. 4FMFDUFE.PMFDVMBS"MUFSBUJPOTJO5IZNJD&QJUIFMJBM5VNPST


Chromosomal
WHO Arm-Level Chromosomal Mutations and Copy Number Changes RNAs
Type Aberrations Translocations Affecting Selected Genes Methylation MicroRNAs Reference
A 6q25 loss HRAS (G13V) miR-515 upregulation 9–11, 14, 15, 53

6p23 loss (FOXC1) (targets PTEN)


AB 6q25 loss
6p23 loss (FOXC1)
7p15 loss
B1 1p, 2q, 3q, 6q losses 9–11, 14

B2 6q25 loss KRAS (G12A) 9–11, 14, 15

6p23 loss (FOXC1)


1q gain
B3 6q25 loss t(11;X) BCL2 copy number gains (18q21.33) 9–11, 14, 15, 20, 30

6p23 loss (FOXC1) MCL1 copy number gain


11q4 losses CDKN2A/B copy number losses (9p21.3)
1q gain BCOR
PHF15
TSCC 6q25 loss KIT mutations: E490K (exon 9), Y553N, MGMT miR-142-5p 1, 9–11, 14, 15, 17–19, 20, 51

6p23 loss (FOXC1) W557R, V559A, V560del, L576P, CDKN2 down-regulation


9p, 13q, 16q, losses P577-D579del (exon 11), H697Y (targets MYC)
1q, 9q, 17q gains (exon 14), D820E (exon 17)
KRAS (G12V)
BCL2 copy number gains (18q21.33)
MCL1 copy number gain
CDKN2A/B copy number losses (9p21.3)
TSCC, Thymic squamous cell carcinoma.

mutational profile of TETs and the inability of gene panels incorporate these data into future clinical trials to help select
designed for other solid tumors to detect mutations in TETs patients for appropriate interventions with an aim to improve
that can help identify patients for targeted therapy. Another response to treatment and survival.
approach is to promote the enrolment of patients with refrac-
tory TETs in appropriate phase I trials, which may lead to
the identification of new molecular pathways of therapeutic REFERENCES
1. Girard N. Thymic epithelial tumours: from basic principles to individual-
interest. For example, the mTOR pathway has emerged as ised treatment strategies. Eur Respir Rev 2013;22:75–87.
a potential area of interest after tumor responses observed 2. Kuo TT. Tumors of the thymus. In Travis WD, Brambilla E, Muller-
in phase I trials reported by several groups.54,55 In addition, Hermelink HK, et al., editors. World Health Organization Classification
disease stabilization as a result of treatment with the mTOR of Tumors: Pathology and Genetics of Tumors of the Lung, Pleura,
inhibitor, everolimus, in combination with somatostatin ana- Thymus and Heart. Lyon, France: IARC Press, 2004. Pp. 146–248.
3. Cancer Genome Atlas Research Network. Comprehensive genomic char-
logs has been described in a case series of two patients with acterization of squamous cell lung cancers. Nature 2012;489:519–25.
thymoma.57 An ongoing phase II clinical trial is evaluating 4. Cancer Genome Atlas Research Network. Genomic and epigenomic
the role of the mTOR inhibitor, everolimus, in patients with landscapes of adult de novo acute myeloid leukemia. N Engl J Med
previously treated TETs (NCT02049047). Preliminary results 2013;368:2059–2074.
5. Cancer Genome Atlas Network. Comprehensive molecular portraits of
show a disease control rate of 71%, median PFS of 12 months,
human breast tumours. Nature 2012;490:61–70.
and overall survival of 24 months.41 These novel approaches 6. Cancer Genome Atlas Network. Comprehensive molecular characteriza-
have the potential to help in the successful translation of the tion of urothelial bladder carcinoma. Nature 2014;507:315–322.
increasing volume of data generated by newer genomic tech- 7. Cancer Genome Atlas Network. Integrated genomic analyses of ovarian
nologies and help in the development of newer therapies for carcinoma. Nature 2011;474:609–615.
8. Zettl A, Ströbel P, Wagner K, et al. Recurrent genetic aberrations in thy-
patients with TETs. moma and thymic carcinoma. Am J Pathol 2000;157:257–266.
9. Girard N, Shen R, Guo T, et al. Comprehensive genomic analysis reveals
CONCLUSIONS clinically relevant molecular distinctions between thymic carcinomas and
thymomas. Clin Cancer Res 2009;15:6790–6799.
Rapid advances in technology have helped uncover 10. Penzel R, Hoegel J, Schmitz W, et al. Clusters of chromosomal imbal-
novel genetic alterations in TETs and resulted in an increas- ances in thymic epithelial tumours are associated with the WHO clas-
ing recognition that TETs have a unique molecular profile sification and the staging system according to Masaoka. Int J Cancer
compared with other solid tumors. These new discoveries 2003;105:494–498.
have helped in gaining greater insight into thymic tumor biol- 11. Inoue M, Starostik P, Zettl A, et al. Correlating genetic aberrations with
World Health Organization-defined histology and stage across the spec-
ogy and have resulted in a steady increase in the identifica- trum of thymomas. Cancer Res 2003;63:3708–3715.
tion of molecular alterations that have the potential to serve 12. Weissferdt A, Lin H, Woods D, et al. HER family receptor and ligand
as biomarkers or drug targets. Attempts should be made to status in thymic carcinoma. Lung Cancer 2012;77:515–521.

Copyright © 2014 by the International Association for the Study of Lung Cancer S135
Rajan et al. Journal of Thoracic Oncology ® t 7PMVNF /VNCFS 4VQQMFNFOU 4FQUFNCFS

13. Ionescu DN, Sasatomi E, Cieply K, Nola M, Dacic S. Protein expression 37. Rajan A, Carter CA, Berman A, et al. Cixutumumab for patients with
and gene amplification of epidermal growth factor receptor in thymomas. recurrent or refractory advanced thymic epithelial tumours: a multicentre,
Cancer 2005;103:630–636. open-label, phase 2 trial. Lancet Oncol 2014;15:191–200.
14. Lee GY, Yang WI, Jeung HC, et al. Genome-wide genetic aberrations of 38. Palmieri G, Marino M, Buonerba C, et al. Imatinib mesylate in thymic epi-
thymoma using cDNA microarray based comparative genomic hybridiza- thelial malignancies. Cancer Chemother Pharmacol 2012;69:309–315.
tion. BMC Genomics 2007;8:305. 39. Wakelee HA, Gubens MA, Burns M, et al. A phase II study of saracatinib
15. Petrini I, Wang Y, Zucali PA, et al. Copy number aberrations of genes (AZD0530), a Src inhibitor, administered orally daily to patients with
regulating normal thymus development in thymic epithelial tumors. Clin advanced thymic malignancies. Chicago Multidisciplinary Symposium in
Cancer Res 2013;19:1960–1971. Thoracic Oncology, Poster 152, 2010.
16. Chen C, Yin N, Yin B, Lu Q. DNA methylation in thoracic neoplasms. 40. Giaccone G, Rajan A, Berman A, et al. Phase II study of belinostat in
Cancer Lett 2011;301:7–16. patients with recurrent or refractory advanced thymic epithelial tumors. J
17. Suzuki M, Chen H, Shigematsu H, et al. Aberrant methylation: common Clin Oncol 2011;29:2052–2059.
in thymic carcinomas, rare in thymomas. Oncol Rep 2005;14:1621–1624. 41. Zucali PA, De Pas T, Palmieri G, et al. Phase II study of everolimus
18. Mokhtar M, Kondo K, Namura T, et al. Methylation and expression profiles in patients with thymoma and thymic carcinoma previously treated
of MGMT gene in thymic epithelial tumors. Lung Cancer 2014;83:279–287. with cisplatin-based chemotherapy. J Clin Oncol (Meeting Abstracts)
19. Hirabayashi H, Fujii Y, Sakaguchi M, et al. p16INK4, pRB, p53 and 2014;32:7527.
cyclin D1 expression and hypermethylation of CDKN2 gene in thymoma 42. Hayashi Y, Ishii N, Obayashi C, et al. Thymoma: tumour type related to
and thymic carcinoma. Int J Cancer 1997;73:639–644. expression of epidermal growth factor (EGF), EGF-receptor, p53, v-erb
20. Petrini I, Meltzer PS, Zucali PA, et al. Copy number aberrations of BCL2 B and ras p21. Virchows Arch 1995;426:43–50.
and CDKN2A/B identified by array-CGH in thymic epithelial tumors. 43. Suzuki E, Sasaki H, Kawano O, et al. Expression and mutation statuses of
Cell Death Dis 2012;3:e351. epidermal growth factor receptor in thymic epithelial tumors. Jpn J Clin
21. Fahrer J, Kaina B. O6-methylguanine-DNA methyltransferase in Oncol 2006;36:351–356.
the defense against N-nitroso compounds and colorectal cancer. 44. Pan CC, Chen PC, Wang LS, Lee JY, Chiang H. Expression of apop-
Carcinogenesis 2013;34:2435–2442. tosis-related markers and HER-2/neu in thymic epithelial tumours.
22. Besse B, Garassino M, Rajan A, et al. A phase II study of milciclib (PHA- Histopathology 2003;43:165–172.
848125AC) in patients (pts) with thymic carcinoma (TC). . J Clin Oncol 45. Pan CC, Chen PC, Chiang H. KIT (CD117) is frequently overex-
(Meeting Abstracts) 2014;32:7526. pressed in thymic carcinomas but is absent in thymomas. J Pathol
23. Badve S, Goswami C, Gökmen-Polar Y, et al. Molecular analysis of thy- 2004;202:375–381.
moma. PLoS One 2012;7:e42669. 46. Petrini I, Zucali PA, Lee HS, et al. Expression and mutational status of
24. Tammali R, Srivastava SK, Ramana KV. Targeting aldose reductase for c-kit in thymic epithelial tumors. J Thorac Oncol 2010;5:1447–1453.
the treatment of cancer. Curr Cancer Drug Targets 2011;11:560–571. 47. Nakagawa K, Matsuno Y, Kunitoh H, Maeshima A, Asamura H, Tsuchiya
25. Ruffini E, Detterbeck F, Van Raemdonck D, et al. Tumours of the thymus: a R. Immunohistochemical KIT (CD117) expression in thymic epithelial
cohort study of prognostic factors from the European Society of Thoracic tumors. Chest 2005;128:140–144.
Surgeons database. Eur J Cardiothorac Surg 2014 [Epub ahead of print]. 48. Girard N, Teruya-Feldstein J, Payabyab EC, et al. Insulin-like growth
26. Sasaki H, Ide N, Yukiue H, et al. Arg and DAP3 expression was correlated factor-1 receptor expression in thymic malignancies. J Thorac Oncol
with human thymoma stage. Clin Exp Metastasis 2004;21:507–513. 2010;5:1439–1446.
27. Takahashi E, Tateyama H, Akatsu H, et al. Expression of matrix metal- 49. Zucali PA, Petrini I, Lorenzi E, et al. Insulin-like growth factor-1 receptor
loproteinases 2 and 7 in tumor cells correlates with the World Health and phosphorylated AKT-serine 473 expression in 132 resected thymo-
Organization classification subtype and clinical stage of thymic epithelial mas and thymic carcinomas. Cancer 2010;116:4686–4695.
tumors. Hum Pathol 2003;34:1253–1258. 50. Tomita M, Matsuzaki Y, Edagawa M, et al. Correlation between tumor
28. Sasaki H, Ide N, Fukai I, Kiriyama M, Yamakawa Y, Fujii Y. Gene expres- angiogenesis and invasiveness in thymic epithelial tumors. J Thorac
sion analysis of human thymoma correlates with tumor stage. Int J Cardiovasc Surg 2002;124:493–498.
Cancer 2002;101:342–347. 51. Ganci F, Vico C, Korita E, et al. MicroRNA expression profiling of thy-
29. Gökmen-Polar Y, Cook RW, Goswami CP, et al. A gene signature to deter- mic epithelial tumors. Lung Cancer 2014;85:197–204.
mine metastatic behavior in thymomas. PLoS One 2013;8:e66047. 52. Huang B, Belharazem D, Li L, et al. Anti-apoptotic signature in thy-
30. Petrini I, Rajan A, Pham T, et al. Whole genome and transcriptome mic squamous cell carcinomas—functional relevance of anti-apoptotic
sequencing of a B3 thymoma. PLoS One 2013;8:e60572. BIRC3 expression in the thymic carcinoma cell line 1889c. Front Oncol
31. Girard N. Thymic tumors: relevant molecular data in the clinic. J Thorac 2013;3:316.
Oncol 2010;5:S291–S295. 53. Radovich M, Porter RF, Conces M, et al. Next-generation sequencing of
32. Giaccone G, Rajan A, Ruijter R, Smit E, van Groeningen C, Hogendoorn thymic malignancies. J Clin Oncol (Meeting Abstracts) 2012;30:7032.
PC. Imatinib mesylate in patients with WHO B3 thymomas and thymic 54. Wheler J, Hong D, Swisher SG, et al. Thymoma patients treated in a phase
carcinomas. J Thorac Oncol 2009;4:1270–1273. I clinic at MD Anderson Cancer Center: responses to mTOR inhibitors
33. Salter JT, Lewis D, Yiannoutsos C, Loehrer PJ, Sr., Risley L, Chiorean and molecular analyses. Oncotarget 2013;4:890–898.
EG. Imatinib for the treatment of thymic carcinoma. J Clin Oncol 55. Kossai M, Duchemann B, Caramella C, et al. Antitumor activity in
(Meeting Abstracts) 2008;26:8116. advanced cancer patients with thymic malignancies enrolled in early clin-
34. Kurup A, Burns M, Dropcho S, Pao W, Loehrer PJ. Phase II study of gefi- ical drug development program (phase I trials) at Institut Gustave Roussy.
tinib treatment in advanced thymic malignancies. J Clin Oncol (Meeting J Clin Oncol (Meeting Abstracts) 2013;31:7604.
Abstracts) 2005;23:7068. 56. Giaccone G, Lopez-Chavez A, Thomas A, et al. Custom (Molecular
35. Bedano PM, Perkins S, Burns M, et al. A phase II trial of erlotinib plus Profiling and Targeted Therapy for Advanced Non-Small Cell Lung
bevacizumab in patients with recurrent thymoma or thymic carcinoma. J Cancer, Small Cell Lung Cancer, and Thymic Malignancies) trial. J Clin
Clin Oncol (Meeting Abstracts) 2008;26:19087. Oncol (Meeting Abstracts) 2013;31:7513.
36. Thomas A, Rajan A, Berman AW, et al. Phase II trial of sunitinib in 57. Palmieri G, Buonerba C, Federico P, et al. Everolimus plus long-acting
patients with thymic epithelial tumors (TET). J Clin Oncol (Meeting somatostatin analogs in thymic epithelial malignancies. World J Clin
Abstracts) 2014;32:7525. Oncol 2012;3:111–115.

S136 Copyright © 2014 by the International Association for the Study of Lung Cancer

You might also like