Login / Signup

Genetic drivers of oncogenic pathways in molecular subgroups of peripheral T-cell lymphoma.

Tayla B HeavicanAlyssa BouskaJiayu YuWaseem LoneCatalina AmadorQiang GongWeiwei ZhangYuping LiBhavana J DaveMaarja-Liisa NairismägiTimothy C GreinerJulie VoseDennis D WeisenburgerCynthia LachelChao WangKai FuJadd M StevensSoon Thye LimChoon Kiat OngRandy D GascoyneEdoardo MissiagliaFrancois LemonnierCorinne HaiounSylvia HartmannMartin Bjerregård PedersenMaria Antonella LaginestraRyan A WilcoxBin Tean TehNoriaki YoshidaKoichi OhshimaMasao SetoAndreas RosenwaldGerman OttElias CampoLisa M RimszaElaine S JaffeRita M BrazielFrancesco d'AmoreGiorgio InghiramiFrancesco BertoniLaurence de LevalPhilippe GaulardLouis M StaudtTimothy W McKeithanStefano PileriWing C ChanJaveed Iqbal
Published in: Blood (2019)
Peripheral T-cell lymphoma (PTCL) is a group of complex clinicopathological entities, often associated with an aggressive clinical course. Angioimmunoblastic T-cell lymphoma (AITL) and PTCL-not otherwise specified (PTCL-NOS) are the 2 most frequent categories, accounting for >50% of PTCLs. Gene expression profiling (GEP) defined molecular signatures for AITL and delineated biological and prognostic subgroups within PTCL-NOS (PTCL-GATA3 and PTCL-TBX21). Genomic copy number (CN) analysis and targeted sequencing of these molecular subgroups revealed unique CN abnormalities (CNAs) and oncogenic pathways, indicating distinct oncogenic evolution. PTCL-GATA3 exhibited greater genomic complexity that was characterized by frequent loss or mutation of tumor suppressor genes targeting the CDKN2A /B-TP53 axis and PTEN-PI3K pathways. Co-occurring gains/amplifications of STAT3 and MYC occurred in PTCL-GATA3. Several CNAs, in particular loss of CDKN2A, exhibited prognostic significance in PTCL-NOS as a single entity and in the PTCL-GATA3 subgroup. The PTCL-TBX21 subgroup had fewer CNAs, primarily targeting cytotoxic effector genes, and was enriched in mutations of genes regulating DNA methylation. CNAs affecting metabolic processes regulating RNA/protein degradation and T-cell receptor signaling were common in both subgroups. AITL showed lower genomic complexity compared with other PTCL entities, with frequent co-occurring gains of chromosome 5 (chr5) and chr21 that were significantly associated with IDH2 R172 mutation. CN losses were enriched in genes regulating PI3K-AKT-mTOR signaling in cases without IDH2 mutation. Overall, we demonstrated that novel GEP-defined PTCL subgroups likely evolve by distinct genetic pathways and provided biological rationale for therapies that may be investigated in future clinical trials.
Keyphrases