scholarly journals 19P Single sample predictor of non-small cell lung cancer histology based on gene expression analysis of archival tissue

2018 ◽  
Vol 13 (4) ◽  
pp. S10
Author(s):  
A. Karlsson ◽  
H. Cirenajwis ◽  
K. Ericson-Lindqvist ◽  
C. Reuterswärd ◽  
M. Jönsson ◽  
...  
Genomics Data ◽  
2015 ◽  
Vol 6 ◽  
pp. 145-148 ◽  
Author(s):  
Erin A. Marshall ◽  
Kevin W. Ng ◽  
Christine Anderson ◽  
Roland Hubaux ◽  
Kelsie L. Thu ◽  
...  

2003 ◽  
Vol 64 (10) ◽  
pp. S116
Author(s):  
Paolo Fardin ◽  
Jurg Bahler ◽  
Paolo Capanni ◽  
Erika Inglese ◽  
Annamaria Ricciardi ◽  
...  

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.


Sign in / Sign up

Export Citation Format

Share Document