5,704 research outputs found

    Correcting for selection bias via cross-validation in the classification of microarray data

    Full text link
    There is increasing interest in the use of diagnostic rules based on microarray data. These rules are formed by considering the expression levels of thousands of genes in tissue samples taken on patients of known classification with respect to a number of classes, representing, say, disease status or treatment strategy. As the final versions of these rules are usually based on a small subset of the available genes, there is a selection bias that has to be corrected for in the estimation of the associated error rates. We consider the problem using cross-validation. In particular, we present explicit formulae that are useful in explaining the layers of validation that have to be performed in order to avoid improperly cross-validated estimates.Comment: Published in at http://dx.doi.org/10.1214/193940307000000284 the IMS Collections (http://www.imstat.org/publications/imscollections.htm) by the Institute of Mathematical Statistics (http://www.imstat.org

    The influence of feature selection methods on accuracy, stability and interpretability of molecular signatures

    Get PDF
    Motivation: Biomarker discovery from high-dimensional data is a crucial problem with enormous applications in biology and medicine. It is also extremely challenging from a statistical viewpoint, but surprisingly few studies have investigated the relative strengths and weaknesses of the plethora of existing feature selection methods. Methods: We compare 32 feature selection methods on 4 public gene expression datasets for breast cancer prognosis, in terms of predictive performance, stability and functional interpretability of the signatures they produce. Results: We observe that the feature selection method has a significant influence on the accuracy, stability and interpretability of signatures. Simple filter methods generally outperform more complex embedded or wrapper methods, and ensemble feature selection has generally no positive effect. Overall a simple Student's t-test seems to provide the best results. Availability: Code and data are publicly available at http://cbio.ensmp.fr/~ahaury/

    Machine Learning and Integrative Analysis of Biomedical Big Data.

    Get PDF
    Recent developments in high-throughput technologies have accelerated the accumulation of massive amounts of omics data from multiple sources: genome, epigenome, transcriptome, proteome, metabolome, etc. Traditionally, data from each source (e.g., genome) is analyzed in isolation using statistical and machine learning (ML) methods. Integrative analysis of multi-omics and clinical data is key to new biomedical discoveries and advancements in precision medicine. However, data integration poses new computational challenges as well as exacerbates the ones associated with single-omics studies. Specialized computational approaches are required to effectively and efficiently perform integrative analysis of biomedical data acquired from diverse modalities. In this review, we discuss state-of-the-art ML-based approaches for tackling five specific computational challenges associated with integrative analysis: curse of dimensionality, data heterogeneity, missing data, class imbalance and scalability issues

    Development of a simple artificial intelligence method to accurately subtype breast cancers based on gene expression barcodes

    Get PDF
    >Magister Scientiae - MScINTRODUCTION: Breast cancer is a highly heterogeneous disease. The complexity of achieving an accurate diagnosis and an effective treatment regimen lies within this heterogeneity. Subtypes of the disease are not simply molecular, i.e. hormone receptor over-expression or absence, but the tumour itself is heterogeneous in terms of tissue of origin, metastases, and histopathological variability. Accurate tumour classification vastly improves treatment decisions, patient outcomes and 5-year survival rates. Gene expression studies aided by transcriptomic technologies such as microarrays and next-generation sequencing (e.g. RNA-Sequencing) have aided oncology researcher and clinician understanding of the complex molecular portraits of malignant breast tumours. Mechanisms governing cancers, which include tumorigenesis, gene fusions, gene over-expression and suppression, cellular process and pathway involvementinvolvement, have been elucidated through comprehensive analyses of the cancer transcriptome. Over the past 20 years, gene expression signatures, discovered with both microarray and RNA-Seq have reached clinical and commercial application through the development of tests such as Mammaprint®, OncotypeDX®, and FoundationOne® CDx, all which focus on chemotherapy sensitivity, prediction of cancer recurrence, and tumour mutational level. The Gene Expression Barcode (GExB) algorithm was developed to allow for easy interpretation and integration of microarray data through data normalization with frozen RMA (fRMA) preprocessing and conversion of relative gene expression to a sequence of 1's and 0's. Unfortunately, the algorithm has not yet been developed for RNA-Seq data. However, implementation of the GExB with feature-selection would contribute to a machine-learning based robust breast cancer and subtype classifier. METHODOLOGY: For microarray data, we applied the GExB algorithm to generate barcodes for normal breast and breast tumour samples. A two-class classifier for malignancy was developed through feature-selection on barcoded samples by selecting for genes with 85% stable absence or presence within a tissue type, and differentially stable between tissues. A multi-class feature-selection method was employed to identify genes with variable expression in one subtype, but 80% stable absence or presence in all other subtypes, i.e. 80% in n-1 subtypes. For RNA-Seq data, a barcoding method needed to be developed which could mimic the GExB algorithm for microarray data. A z-score-to-barcode method was implemented and differential gene expression analysis with selection of the top 100 genes as informative features for classification purposes. The accuracy and discriminatory capability of both microarray-based gene signatures and the RNA-Seq-based gene signatures was assessed through unsupervised and supervised machine-learning algorithms, i.e., K-means and Hierarchical clustering, as well as binary and multi-class Support Vector Machine (SVM) implementations. RESULTS: The GExB-FS method for microarray data yielded an 85-probe and 346-probe informative set for two-class and multi-class classifiers, respectively. The two-class classifier predicted samples as either normal or malignant with 100% accuracy and the multi-class classifier predicted molecular subtype with 96.5% accuracy with SVM. Combining RNA-Seq DE analysis for feature-selection with the z-score-to-barcode method, resulted in a two-class classifier for malignancy, and a multi-class classifier for normal-from-healthy, normal-adjacent-tumour (from cancer patients), and breast tumour samples with 100% accuracy. Most notably, a normal-adjacent-tumour gene expression signature emerged, which differentiated it from normal breast tissues in healthy individuals. CONCLUSION: A potentially novel method for microarray and RNA-Seq data transformation, feature selection and classifier development was established. The universal application of the microarray signatures and validity of the z-score-to-barcode method was proven with 95% accurate classification of RNA-Seq barcoded samples with a microarray discovered gene expression signature. The results from this comprehensive study into the discovery of robust gene expression signatures holds immense potential for further R&F towards implementation at the clinical endpoint, and translation to simpler and cost-effective laboratory methods such as qtPCR-based tests

    Google Goes Cancer: Improving Outcome Prediction for Cancer Patients by Network-Based Ranking of Marker Genes

    Get PDF
    Predicting the clinical outcome of cancer patients based on the expression of marker genes in their tumors has received increasing interest in the past decade. Accurate predictors of outcome and response to therapy could be used to personalize and thereby improve therapy. However, state of the art methods used so far often found marker genes with limited prediction accuracy, limited reproducibility, and unclear biological relevance. To address this problem, we developed a novel computational approach to identify genes prognostic for outcome that couples gene expression measurements from primary tumor samples with a network of known relationships between the genes. Our approach ranks genes according to their prognostic relevance using both expression and network information in a manner similar to Google's PageRank. We applied this method to gene expression profiles which we obtained from 30 patients with pancreatic cancer, and identified seven candidate marker genes prognostic for outcome. Compared to genes found with state of the art methods, such as Pearson correlation of gene expression with survival time, we improve the prediction accuracy by up to 7%. Accuracies were assessed using support vector machine classifiers and Monte Carlo cross-validation. We then validated the prognostic value of our seven candidate markers using immunohistochemistry on an independent set of 412 pancreatic cancer samples. Notably, signatures derived from our candidate markers were independently predictive of outcome and superior to established clinical prognostic factors such as grade, tumor size, and nodal status. As the amount of genomic data of individual tumors grows rapidly, our algorithm meets the need for powerful computational approaches that are key to exploit these data for personalized cancer therapies in clinical practice

    Rank discriminants for predicting phenotypes from RNA expression

    Get PDF
    Statistical methods for analyzing large-scale biomolecular data are commonplace in computational biology. A notable example is phenotype prediction from gene expression data, for instance, detecting human cancers, differentiating subtypes and predicting clinical outcomes. Still, clinical applications remain scarce. One reason is that the complexity of the decision rules that emerge from standard statistical learning impedes biological understanding, in particular, any mechanistic interpretation. Here we explore decision rules for binary classification utilizing only the ordering of expression among several genes; the basic building blocks are then two-gene expression comparisons. The simplest example, just one comparison, is the TSP classifier, which has appeared in a variety of cancer-related discovery studies. Decision rules based on multiple comparisons can better accommodate class heterogeneity, and thereby increase accuracy, and might provide a link with biological mechanism. We consider a general framework ("rank-in-context") for designing discriminant functions, including a data-driven selection of the number and identity of the genes in the support ("context"). We then specialize to two examples: voting among several pairs and comparing the median expression in two groups of genes. Comprehensive experiments assess accuracy relative to other, more complex, methods, and reinforce earlier observations that simple classifiers are competitive.Comment: Published in at http://dx.doi.org/10.1214/14-AOAS738 the Annals of Applied Statistics (http://www.imstat.org/aoas/) by the Institute of Mathematical Statistics (http://www.imstat.org

    Assessment of SVM Reliability for Microarray Data Analysis

    Get PDF
    The goal of our research is to provide techniques that can assess and validate the results of SVM-based analysis of microarray data. We present preliminary results of the effect of mislabeled training samples. We conducted several systematic experiments on artificial and real medical data using SVMs. We systematically flipped the labels of a fraction of the training data. We show that a relatively small number of mislabeled examples can dramatically decrease the performance as visualized on the ROC graphs. This phenomenon persists even if the dimensionality of the input space is drastically decreased, by using for example feature selection. Moreover we show that for SVM recursive feature elimination, even a small fraction of mislabeled samples can completely change the resulting set of genes. This work is an extended version of the previous paper [MBN04]

    Boosting the concordance index for survival data - a unified framework to derive and evaluate biomarker combinations

    Get PDF
    The development of molecular signatures for the prediction of time-to-event outcomes is a methodologically challenging task in bioinformatics and biostatistics. Although there are numerous approaches for the derivation of marker combinations and their evaluation, the underlying methodology often suffers from the problem that different optimization criteria are mixed during the feature selection, estimation and evaluation steps. This might result in marker combinations that are only suboptimal regarding the evaluation criterion of interest. To address this issue, we propose a unified framework to derive and evaluate biomarker combinations. Our approach is based on the concordance index for time-to-event data, which is a non-parametric measure to quantify the discrimatory power of a prediction rule. Specifically, we propose a component-wise boosting algorithm that results in linear biomarker combinations that are optimal with respect to a smoothed version of the concordance index. We investigate the performance of our algorithm in a large-scale simulation study and in two molecular data sets for the prediction of survival in breast cancer patients. Our numerical results show that the new approach is not only methodologically sound but can also lead to a higher discriminatory power than traditional approaches for the derivation of gene signatures.Comment: revised manuscript - added simulation study, additional result

    From Correlation to Causality: Does Network Information improve Cancer Outcome Prediction?

    Get PDF
    Motivation: Disease progression in cancer can vary substantially between patients. Yet, patients often receive the same treatment. Recently, there has been much work on predicting disease progression and patient outcome variables from gene expression in order to personalize treatment options. A widely used approach is high-throughput experiments that aim to explore predictive signature genes which would provide identification of clinical outcome of diseases. Microarray data analysis helps to reveal underlying biological mechanisms of tumor progression, metastasis, and drug-resistance in cancer studies. Despite first diagnostic kits in the market, there are open problems such as the choice of random gene signatures or noisy expression data. The experimental or computational noise in data and limited tissue samples collected from patients might furthermore reduce the predictive power and biological interpretability of such signature genes. Nevertheless, signature genes predicted by different studies generally represent poor similarity; even for the same type of cancer. Integration of network information with gene expression data could provide more efficient signatures for outcome prediction in cancer studies. One approach to deal with these problems employs gene-gene relationships and ranks genes using the random surfer model of Google's PageRank algorithm. Unfortunately, the majority of published network-based approaches solely tested their methods on a small amount of datasets, questioning the general applicability of network-based methods for outcome prediction. Methods: In this thesis, I provide a comprehensive and systematically evaluation of a network-based outcome prediction approach -- NetRank - a PageRank derivative -- applied on several types of gene expression cancer data and four different types of networks. The algorithm identifies a signature gene set for a specific cancer type by incorporating gene network information with given expression data. To assess the performance of NetRank, I created a benchmark dataset collection comprising 25 cancer outcome prediction datasets from literature and one in-house dataset. Results: NetRank performs significantly better than classical methods such as foldchange or t-test as it improves the prediction performance in average for 7%. Besides, we are approaching the accuracy level of the authors' signatures by applying a relatively unbiased but fully automated process for biomarker discovery. Despite an order of magnitude difference in network size, a regulatory, a protein-protein interaction and two predicted networks perform equally well. Signatures as published by the authors and the signatures generated with classical methods do not overlap -- not even for the same cancer type -- whereas the network-based signatures strongly overlap. I analyze and discuss these overlapping genes in terms of the Hallmarks of cancer and in particular single out six transcription factors and seven proteins and discuss their specific role in cancer progression. Furthermore several tests are conducted for the identification of a Universal Cancer Signature. No Universal Cancer Signature could be identified so far, but a cancer-specific combination of general master regulators with specific cancer genes could be discovered that achieves the best results for all cancer types. As NetRank offers a great value for cancer outcome prediction, first steps for a secure usage of NetRank in a public cloud are described. Conclusion: Experimental evaluation of network-based methods on a gene expression benchmark dataset suggests that these methods are especially suited for outcome prediction as they overcome the problems of random gene signatures and noisy expression data. Through the combination of network information with gene expression data, network-based methods identify highly similar signatures over all cancer types, in contrast to classical methods that fail to identify highly common gene sets across the same cancer types. In general allows the integration of additional information in gene expression analysis the identification of more reliable, accurate and reproducible biomarkers and provides a deeper understanding of processes occurring in cancer development and progression.:1 Definition of Open Problems 2 Introduction 2.1 Problems in cancer outcome prediction 2.2 Network-based cancer outcome prediction 2.3 Universal Cancer Signature 3 Methods 3.1 NetRank algorithm 3.2 Preprocessing and filtering of the microarray data 3.3 Accuracy 3.4 Signature similarity 3.5 Classical approaches 3.6 Random signatures 3.7 Networks 3.8 Direct neighbor method 3.9 Dataset extraction 4 Performance of NetRank 4.1 Benchmark dataset for evaluation 4.2 The influence of NetRank parameters 4.3 Evaluation of NetRank 4.4 General findings 4.5 Computational complexity of NetRank 4.6 Discussion 5 Universal Cancer Signature 5.1 Signature overlap – a sign for Universal Cancer Signature 5.2 NetRank genes are highly connected and confirmed in literature 5.3 Hallmarks of Cancer 5.4 Testing possible Universal Cancer Signatures 5.5 Conclusion 6 Cloud-based Biomarker Discovery 6.1 Introduction to secure Cloud computing 6.2 Cancer outcome prediction 6.3 Security analysis 6.4 Conclusion 7 Contributions and Conclusion
    • …
    corecore