02.12.2016 Views

Journal Thoracic Oncology

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

WCLC2016-Abstract-Book_vF-WEB_revNov17-1

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Abstracts <strong>Journal</strong> of <strong>Thoracic</strong> <strong>Oncology</strong> • Volume 12 Issue S1 January 2017<br />

On the contrary, tumors from never smokers demonstrate a much lower<br />

mutational burden (0.8-1 mutations/Mb) and are enriched for C>T transitions.<br />

1,2<br />

Single nucleotide variations (SNVs) and copy number alterations (CNAs)<br />

While both LUAD and SQCC show frequent inactivation of the tumor<br />

suppressors TP53 and CDKN2A, these alterations are considerably more<br />

common in SQCCs. CDKN2A harbors the loci for two isoforms, p14ARF and<br />

p16INK4A, and is inactivated in SQCC through homozygous deletion (29%),<br />

methylation (21%), inactivating mutations (18%), or exon 1b skipping (4%).<br />

1,2 These findings indicate a strong selective pressure for the loss of these<br />

tumor suppressors in NSCLC. The pattern of oncogenic alterations varies<br />

considerably between LUAD and SQCC. While LUADs typically showed<br />

activating RTK/RAS/RAF pathway mutations, these mutations are highly<br />

infrequent in SQCCs - which predominantly showed alterations in oxidative<br />

stress response (NFE2L2, KEAP1 and CUL3) and squamous differentiation<br />

pathways (SOX2, TP63, NOTCH1, etc.) in 44% of samples. 1,2KRAS is the most<br />

commonly mutated oncogene in LUAD, followed by EGFR, BRAF, PIK3CA, and<br />

MET. The majority of EGFR mutations in LUAD are targetable (L858R or exon 19<br />

deletion) with tyrosine kinase inhibitors (TKIs). 1 In contrast, such alterations<br />

are absent in SQCC. Two SQCC samples however demonstrated L861Q<br />

mutations in EGFR, which are potentially targetable with TKIs. 1,2 Although<br />

SQCC and LUAD shared several CNAs at the chromosomal arm level,<br />

amplification of 3q was frequent in SQCC. This region harbors important<br />

oncogenes such as SOX2, PIK3CA, and TP63. LUADs frequently showed<br />

amplifications in genes such as NKX2-1, TERT, MDM2, KRAS, and<br />

EGFR. 1,2 Oncogenic activation of kinases such as ALK, ROS1, and RET through<br />

rearrangement has been well described in LUAD, and these fusions are<br />

targetable with TKIs. These fusions were seen in 1-2% (ALK : 3/230, ROS1:<br />

4/230, and RET: 2/230 samples) of LUADs. 1,2 Transcriptome analysis<br />

Deregulated splicing can be a consequence of mutations that alter splice-sites<br />

within a gene or splicing factors. Mutations in the proto-oncogene MET that<br />

lead to exon 14 skipping, and abnormal splicing of proto-oncogenes such as<br />

CTNNB1 as a result of U2AF1 mutation have been described in LUAD. 1<br />

Transcriptome analyses have also enabled a reclassification of LUADs and<br />

SQCCs into three and four distinct subtypes, respectively. LUAD samples can<br />

be categorized as terminal respiratory unit (enriched for EGFR mutations and<br />

fusions; favorable prognosis), proximal-inflammatory (NF1 and TP53<br />

co-mutation), or proximal-proliferative (KRAS and STK11 alterations) subtypes.<br />

Similarly, SQCCs can be classified as classical, basal, secretory, or primitive.<br />

Alterations in genes that participate in the oxidative stress response<br />

pathway, hypermethylation, and chromosomal instability are characteristic<br />

of the classical subtype (associated with heavy smoking and poor prognosis).<br />

1,2<br />

Key pathogenic alterations TCGA analysis revealed alterations in well<br />

known oncogenic drivers involving RAS signaling pathway in 62% of LUAD..<br />

These samples with readily identifiable oncogenic driver alterations were<br />

collectively labeled ‘oncogene-positive’. Additional analyses of the ‘oncogenenegative’<br />

sample cohort showed enrichment for RIT1, and NF1 mutations.<br />

Given the role of RIT1 and NF1 in RTK/RAS/RAF signaling, samples with these<br />

mutations were reclassified as oncogene positive, increasing the overall<br />

percentage of oncogene positive samples in LUAD to 76%. Nearly 69% of SQCC<br />

samples showed alterations in genes regulating PI3K/AKT, or RTK/RAS<br />

signaling. 1,2 The inability to readily identify an oncogenic driver in nearly a<br />

third of sequenced lung cancer samples highlights the need for greater<br />

powering of subsequent studies to identify novel low frequency genomic<br />

alterations. For instance, previously uncharacterized alterations in the RTK/<br />

RAS/RAF pathway were observed in RASA1, SOS1 in the updated TCGA analysis<br />

which analyzed a much larger cohort of samples. 9 Overall, despite showing a<br />

few similarities between LUAD and SQCC, investigators of TCGA reported<br />

prominent differences between the genomic landscapes of these subtypes.<br />

These subtypes have more of their alterations in common with other cancers<br />

than with one another. SQCCs more closely resembled head and neck<br />

squamous cell and bladder cancer, while LUAD resembled glioblastoma<br />

multiforme and colorectal cancer in this regard. 9 Immunotherapies The vast<br />

majority of lung cancers do not harbor alterations that are targetable by TKIs.<br />

1,2<br />

Immune checkpoint inhibitors are approved for use in patients with<br />

metastatic NSCLC. There is a clear need to develop optimal predictive<br />

biomarkers to identify those who are likely to respond to immune checkpoint<br />

inhibitors. Mutational burden has been correlated with better response to<br />

checkpoint inhibitors. Furthermore, using exome and transcriptome<br />

sequencing and sophisticated bioinformatics, it is now possible to identify<br />

mutated and expressed genes that could potentially serve as a trigger for<br />

immune response (so called neoantigens) once immune checkpoints like<br />

programmed death-1 or programmed death ligand-1 are inhibited.. Swanton<br />

and colleagues performed a neoantigen and clonality analysis on TCGA<br />

samples to examine characteristics such as neoantigen burden and<br />

intratumor heterogeneity (ITH), and their impact on survival. In LUAD, a<br />

higher neoantigen burden was significantly associated with longer survival.<br />

Although not statistically significant, there was a trend towards longer<br />

survival in molecularly homogeneous tumors (

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!