Molecular subtypes of non-small-cell lung cancer

2013 ◽  
pp. 6-20
Author(s):  
Thomas E Stinchcombe
2019 ◽  
Vol 19 (7) ◽  
pp. 415-415 ◽  
Author(s):  
Charles M. Rudin ◽  
John T. Poirier ◽  
Lauren Averett Byers ◽  
Caroline Dive ◽  
Afshin Dowlati ◽  
...  

Nature Cancer ◽  
2021 ◽  
Vol 2 (11) ◽  
pp. 1224-1242
Author(s):  
Janne Lehtiö ◽  
Taner Arslan ◽  
Ioannis Siavelis ◽  
Yanbo Pan ◽  
Fabio Socciarelli ◽  
...  

2014 ◽  
Vol 20 (2 Supplement) ◽  
pp. B27-B27 ◽  
Author(s):  
Akshata Ramrao Udyavar ◽  
Megan Hoeksema ◽  
Kirsten Diggins ◽  
Jonathan Irish ◽  
Pierre P. Massion ◽  
...  

2019 ◽  
Author(s):  
Ayushi S. Patel ◽  
Seungyeul Yoo ◽  
Ranran Kong ◽  
Takashi Sato ◽  
Abhilasha Sinha ◽  
...  

AbstractComprehensive genomic analyses of small cell lung cancer (SCLC), the most aggressive form of lung cancer, have revealed near universal loss of tumor suppressors (RB1 and TP53) and frequent genomic amplification of all three MYC family members. The amplification of each Myc family member is mutually exclusive; hence it had been long suggested that they are functionally equivalent. However, their expression has more recently been associated with specific neuroendocrine markers and distinct histopathology. In this study, we explored a novel role of c-Myc and L-Myc as lineage determining factors contributing to SCLC molecular subtypes and histology. Integrated analyses of a gene regulatory network generated from mRNA expression of primary SCLC tumor and chromatin state profiling of SCLC cell lines showed that Myc family members impart distinct transcriptional programs associated with lineage state; wherein the L-Myc signature was enriched for neuronal pathways while the c-Myc signature was enriched for Notch signaling and epithelial-to-mesenchymal transition. We investigated the functional redundancy and distinction of c-Myc and L-Myc, and noted the insufficiency of L-Myc to induce lineage switch in contrast to the potential of c-Myc to induce trans-differentiation. c-Myc rewires the Myc-accessible landscape and activates neuron al repressor, Rest to mediate transition from ASCL1-SCLC to NeuroD1-SCLC characterized by distinct LCNEC-like histopathology. Collectively, our findings reveal a previously undescribed role of historically defined general oncogenes, c-Myc and L-Myc, for regulating lineage plasticity across molecular subtypes as well as histological subclasses.


2019 ◽  
Vol 30 ◽  
pp. vii6-vii7
Author(s):  
C. Xu ◽  
W. Wang ◽  
Q. Zhang ◽  
W. Zhuang ◽  
Y. Zhu ◽  
...  

2019 ◽  
Vol 19 (5) ◽  
pp. 289-297 ◽  
Author(s):  
Charles M. Rudin ◽  
John T. Poirier ◽  
Lauren Averett Byers ◽  
Caroline Dive ◽  
Afshin Dowlati ◽  
...  

2019 ◽  
Vol 21 (2) ◽  
pp. 729-740 ◽  
Author(s):  
Helena Cirenajwis ◽  
Martin Lauss ◽  
Maria Planck ◽  
Johan Vallon-Christersson ◽  
Johan Staaf

Abstract The development of multigene classifiers for cancer prognosis, treatment prediction, molecular subtypes or clinicopathological groups has been a cornerstone in transcriptomic analyses of human malignancies for nearly two decades. However, many reported classifiers are critically limited by different preprocessing needs like normalization and data centering. In response, a new breed of classifiers, single sample predictors (SSPs), has emerged. SSPs classify samples in an N-of-1 fashion, relying on, e.g. gene rules comparing expression values within a sample. To date, several methods have been reported, but there is a lack of head-to-head performance comparison for typical cancer classification problems, representing an unmet methodological need in cancer bioinformatics. To resolve this need, we performed an evaluation of two SSPs [k-top-scoring pair classifier (kTSP) and absolute intrinsic molecular subtyping (AIMS)] for two case examples of different magnitude of difficulty in non-small cell lung cancer: gene expression–based classification of (i) tumor histology and (ii) molecular subtype. Through the analysis of ~2000 lung cancer samples for each case example (n = 1918 and n = 2106, respectively), we compared the performance of the methods for different sample compositions, training data set sizes, gene expression platforms and gene rule selections. Three main conclusions are drawn from the comparisons: both methods are platform independent, they select largely overlapping gene rules associated with actual underlying tumor biology and, for large training data sets, they behave interchangeably performance-wise. While SSPs like AIMS and kTSP offer new possibilities to move gene expression signatures/predictors closer to a clinical context, they are still importantly limited by the difficultness of the classification problem at hand.


Oncogene ◽  
2016 ◽  
Vol 36 (10) ◽  
pp. 1384-1393 ◽  
Author(s):  
F Chen ◽  
Y Zhang ◽  
E Parra ◽  
J Rodriguez ◽  
C Behrens ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document