2020
DOI: 10.1093/bioinformatics/btaa442
|View full text |Cite
|
Sign up to set email alerts
|

AITL: Adversarial Inductive Transfer Learning with input and output space adaptation for pharmacogenomics

Abstract: Motivation The goal of pharmacogenomics is to predict drug response in patients using their single- or multi-omics data. A major challenge is that clinical data (i.e. patients) with drug response outcome is very limited, creating a need for transfer learning to bridge the gap between large pre-clinical pharmacogenomics datasets (e.g. cancer cell lines), as a source domain, and clinical datasets as a target domain. Two major discrepancies exist between pre-clinical and clinical datasets: (i) i… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

0
41
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
6
1
1

Relationship

0
8

Authors

Journals

citations
Cited by 34 publications
(41 citation statements)
references
References 36 publications
(70 reference statements)
0
41
0
Order By: Relevance
“…Recent studies have proposed models with adversarial networks by training both in vitro and in vivo datasets and obtained increased performance compared with that of models trained using only in vitro datasets [51]. This approach is helpful when appropriate in vivo datasets for a given drug are available.…”
Section: Discussionmentioning
confidence: 99%
“…Recent studies have proposed models with adversarial networks by training both in vitro and in vivo datasets and obtained increased performance compared with that of models trained using only in vitro datasets [51]. This approach is helpful when appropriate in vivo datasets for a given drug are available.…”
Section: Discussionmentioning
confidence: 99%
“…As the authors of AITL (see below) point out, considering in-vitro to in-vivo translation: "Two major discrepancies exist between preclinical and clinical datasets: (i) in the input space, the gene expression data, due to difference in the basic biology, and (ii) in the output space, the different measures of the drug response. Therefore, training a computational model on cell lines and testing it on patients violates the i.i.d assumption that train and test data are from the same distribution" (Sharifi-Noghabi et al, 2020). Other frequently encountered gaps hindering transfer are the species gap and the complex relationships between the molecular entities that may be measured (see above).…”
Section: Molecular Data and Similarity Of Molecular Processesmentioning
confidence: 99%
“…By default, we follow Sharifi-Noghabi et al (2020) in adopting the terminology of Pan and Yang (2010), which is a widely cited review and reference in the field of transfer learning. In their paper, transfer learning is defined in terms of source and target domains (of features with probability distributions associated with these), as well as source and target tasks (mapping features to labels using predictors) so that the predictor in the target domain uses some knowledge from the source domain.…”
Section: Transfer Learning Terminology and Examplesmentioning
confidence: 99%
“…Previous works ( Geeleher et al , 2014 , 2017 ; Sakellaropoulos et al , 2019 ) assumed that batch effects were the main origin of differences to correct for between models, without directly addressing biological variations. Recently, some methods have sought to use domain adaptation (DA) techniques to bridge the in vitro to in vivo gap ( Mourragui et al , 2019 , 2020 ; Sharifi-Noghabi et al , 2020 ).…”
Section: Introductionmentioning
confidence: 99%
“…A refinement of this idea, TRANSACT ( Mourragui et al , 2020 ), uses Kernel-PCA based sub-space alignment to further capture non-linear relationships between samples from in vitro and in vivo domains. However, to learn the similarity between domains, existing DA methods either do not take into account the conditional distributions ( and for drug response Y given gene expression X in source s and target t ), obtaining a subset of shared features that might be unrelated to drug response ( Mourragui et al , 2019 , 2020 ), or rely on the covariate-shift assumption ( Sharifi-Noghabi et al , 2020 ), where marginal distributions for features ( and , for tasks/domains s and t ) are allowed to vary while the conditional distribution for drug response is assumed to be the same ( ) ( Kouw and Loog, 2021 ; Zhao et al , 2019 ). This assumption can often lead to NT ( Rampášek, 2020 ; Zhao et al , 2019 ) when e.g.…”
Section: Introductionmentioning
confidence: 99%