319 research outputs found

    Simple and Effective Visual Models for Gene Expression Cancer Diagnostics

    Get PDF
    In the paper we show that diagnostic classes in cancer gene expression data sets, which most often include thousands of features (genes), may be effectively separated with simple two-dimensional plots such as scatterplot and radviz graph. The principal innovation proposed in the paper is a method called VizRank, which is able to score and identify the best among possibly millions of candidate projections for visualizations. Compared to recently much applied techniques in the field of cancer genomics that include neural networks, support vector machines and various ensemble-based approaches, VizRank is fast and finds visualization models that can be easily examined and interpreted by domain experts. Our experiments on a number of gene expression data sets show that VizRank was always able to find data visualizations with a small number of (two to seven) genes and excellent class separation. In addition to providing grounds for gene expression cancer diagnosis, VizRank and its visualizations also identify small sets of relevant genes, uncover interesting gene interactions and point to outliers and potential misclassifications in cancer data sets

    Finding minimum gene subsets with heuristic breadth-first search algorithm for robust tumor classification

    Get PDF
    Background: Previous studies on tumor classification based on gene expression profiles suggest that gene selection plays a key role in improving the classification performance. Moreover, finding important tumor-related genes with the highest accuracy is a very important task because these genes might serve as tumor biomarkers, which is of great benefit to not only tumor molecular diagnosis but also drug development. Results: This paper proposes a novel gene selection method with rich biomedical meaning based on Heuristic Breadth-first Search Algorithm (HBSA) to find as many optimal gene subsets as possible. Due to the curse of dimensionality, this type of method could suffer from over-fitting and selection bias problems. To address these potential problems, a HBSA-based ensemble classifier is constructed using majority voting strategy from individual classifiers constructed by the selected gene subsets, and a novel HBSA-based gene ranking method is designed to find important tumor-related genes by measuring the significance of genes using their occurrence frequencies in the selected gene subsets. The experimental results on nine tumor datasets including three pairs of cross-platform datasets indicate that the proposed method can not only obtain better generalization performance but also find many important tumor-related genes. Conclusions: It is found that the frequencies of the selected genes follow a power-law distribution, indicating that only a few top-ranked genes can be used as potential diagnosis biomarkers. Moreover, the top-ranked genes leading to very high prediction accuracy are closely related to specific tumor subtype and even hub genes. Compared with other related methods, the proposed method can achieve higher prediction accuracy with fewer genes. Moreover, they are further justified by analyzing the top-ranked genes in the context of individual gene function, biological pathway, and protein-protein interaction network. Keywords: Gene expression profiles; Gene selection; Tumor classification; Heuristic breadth-first search; Power-law distributio

    Finding minimum gene subsets with heuristic breadth-first search algorithm for robust tumor classification

    Get PDF
    Background: Previous studies on tumor classification based on gene expression profiles suggest that gene selection plays a key role in improving the classification performance. Moreover, finding important tumor-related genes with the highest accuracy is a very important task because these genes might serve as tumor biomarkers, which is of great benefit to not only tumor molecular diagnosis but also drug development. Results: This paper proposes a novel gene selection method with rich biomedical meaning based on Heuristic Breadth-first Search Algorithm (HBSA) to find as many optimal gene subsets as possible. Due to the curse of dimensionality, this type of method could suffer from over-fitting and selection bias problems. To address these potential problems, a HBSA-based ensemble classifier is constructed using majority voting strategy from individual classifiers constructed by the selected gene subsets, and a novel HBSA-based gene ranking method is designed to find important tumor-related genes by measuring the significance of genes using their occurrence frequencies in the selected gene subsets. The experimental results on nine tumor datasets including three pairs of cross-platform datasets indicate that the proposed method can not only obtain better generalization performance but also find many important tumor-related genes. Conclusions: It is found that the frequencies of the selected genes follow a power-law distribution, indicating that only a few top-ranked genes can be used as potential diagnosis biomarkers. Moreover, the top-ranked genes leading to very high prediction accuracy are closely related to specific tumor subtype and even hub genes. Compared with other related methods, the proposed method can achieve higher prediction accuracy with fewer genes. Moreover, they are further justified by analyzing the top-ranked genes in the context of individual gene function, biological pathway, and protein-protein interaction network. Keywords: Gene expression profiles; Gene selection; Tumor classification; Heuristic breadth-first search; Power-law distributio

    Unsupervised multiple kernel learning approaches for integrating molecular cancer patient data

    Get PDF
    Cancer is the second leading cause of death worldwide. A characteristic of this disease is its complexity leading to a wide variety of genetic and molecular aberrations in the tumors. This heterogeneity necessitates personalized therapies for the patients. However, currently defined cancer subtypes used in clinical practice for treatment decision-making are based on relatively few selected markers and thus provide only a coarse classifcation of tumors. The increased availability in multi-omics data measured for cancer patients now offers the possibility of defining more informed cancer subtypes. Such a more fine-grained characterization of cancer subtypes harbors the potential of substantially expanding treatment options in personalized cancer therapy. In this thesis, we identify comprehensive cancer subtypes using multidimensional data. For this purpose, we apply and extend unsupervised multiple kernel learning methods. Three challenges of unsupervised multiple kernel learning are addressed: robustness, applicability, and interpretability. First, we show that regularization of the multiple kernel graph embedding framework, which enables the implementation of dimensionality reduction techniques, can increase the stability of the resulting patient subgroups. This improvement is especially beneficial for data sets with a small number of samples. Second, we adapt the objective function of kernel principal component analysis to enable the application of multiple kernel learning in combination with this widely used dimensionality reduction technique. Third, we improve the interpretability of kernel learning procedures by performing feature clustering prior to integrating the data via multiple kernel learning. On the basis of these clusters, we derive a score indicating the impact of a feature cluster on a patient cluster, thereby facilitating further analysis of the cluster-specific biological properties. All three procedures are successfully tested on real-world cancer data. Comparing our newly derived methodologies to established methods provides evidence that our work offers novel and beneficial ways of identifying patient subgroups and gaining insights into medically relevant characteristics of cancer subtypes.Krebs ist eine der häufigsten Todesursachen weltweit. Krebs ist gekennzeichnet durch seine Komplexität, die zu vielen verschiedenen genetischen und molekularen Aberrationen im Tumor führt. Die Unterschiede zwischen Tumoren erfordern personalisierte Therapien für die einzelnen Patienten. Die Krebssubtypen, die derzeit zur Behandlungsplanung in der klinischen Praxis verwendet werden, basieren auf relativ wenigen, genetischen oder molekularen Markern und können daher nur eine grobe Unterteilung der Tumoren liefern. Die zunehmende Verfügbarkeit von Multi-Omics-Daten für Krebspatienten ermöglicht die Neudefinition von fundierteren Krebssubtypen, die wiederum zu spezifischeren Behandlungen für Krebspatienten führen könnten. In dieser Dissertation identifizieren wir neue, potentielle Krebssubtypen basierend auf Multi-Omics-Daten. Hierfür verwenden wir unüberwachtes Multiple Kernel Learning, welches in der Lage ist mehrere Datentypen miteinander zu kombinieren. Drei Herausforderungen des unüberwachten Multiple Kernel Learnings werden adressiert: Robustheit, Anwendbarkeit und Interpretierbarkeit. Zunächst zeigen wir, dass die zusätzliche Regularisierung des Multiple Kernel Learning Frameworks zur Implementierung verschiedener Dimensionsreduktionstechniken die Stabilität der identifizierten Patientengruppen erhöht. Diese Robustheit ist besonders vorteilhaft für Datensätze mit einer geringen Anzahl von Proben. Zweitens passen wir die Zielfunktion der kernbasierten Hauptkomponentenanalyse an, um eine integrative Version dieser weit verbreiteten Dimensionsreduktionstechnik zu ermöglichen. Drittens verbessern wir die Interpretierbarkeit von kernbasierten Lernprozeduren, indem wir verwendete Merkmale in homogene Gruppen unterteilen bevor wir die Daten integrieren. Mit Hilfe dieser Gruppen definieren wir eine Bewertungsfunktion, die die weitere Auswertung der biologischen Eigenschaften von Patientengruppen erleichtert. Alle drei Verfahren werden an realen Krebsdaten getestet. Den Vergleich unserer Methodik mit etablierten Methoden weist nach, dass unsere Arbeit neue und nützliche Möglichkeiten bietet, um integrative Patientengruppen zu identifizieren und Einblicke in medizinisch relevante Eigenschaften von Krebssubtypen zu erhalten

    Accurate molecular classification of cancer using simple rules

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>One intractable problem with using microarray data analysis for cancer classification is how to reduce the extremely high-dimensionality gene feature data to remove the effects of noise. Feature selection is often used to address this problem by selecting informative genes from among thousands or tens of thousands of genes. However, most of the existing methods of microarray-based cancer classification utilize too many genes to achieve accurate classification, which often hampers the interpretability of the models. For a better understanding of the classification results, it is desirable to develop simpler rule-based models with as few marker genes as possible.</p> <p>Methods</p> <p>We screened a small number of informative single genes and gene pairs on the basis of their depended degrees proposed in rough sets. Applying the decision rules induced by the selected genes or gene pairs, we constructed cancer classifiers. We tested the efficacy of the classifiers by leave-one-out cross-validation (LOOCV) of training sets and classification of independent test sets.</p> <p>Results</p> <p>We applied our methods to five cancerous gene expression datasets: leukemia (acute lymphoblastic leukemia [ALL] vs. acute myeloid leukemia [AML]), lung cancer, prostate cancer, breast cancer, and leukemia (ALL vs. mixed-lineage leukemia [MLL] vs. AML). Accurate classification outcomes were obtained by utilizing just one or two genes. Some genes that correlated closely with the pathogenesis of relevant cancers were identified. In terms of both classification performance and algorithm simplicity, our approach outperformed or at least matched existing methods.</p> <p>Conclusion</p> <p>In cancerous gene expression datasets, a small number of genes, even one or two if selected correctly, is capable of achieving an ideal cancer classification effect. This finding also means that very simple rules may perform well for cancerous class prediction.</p

    3D radiomics predicts EGFR mutation, exon-19 deletion and exon-21 L858R mutation in lung adenocarcinoma

    Get PDF
    Background: To establish a radiomic approach to identify epidermal growth factor receptor (EGFR) mutation status in lung adenocarcinoma patients based on CT images, and to distinguish exon-19 deletion and exon-21 L858R mutation. Methods: Two hundred sixty-three patients who underwent pre-surgical contrast-enhanced CT and molecular testing were included, and randomly divided into the training (80%) and test (20%) cohort. Tumor images were three-dimensionally segmented to extract 1,672 radiomic features. Clinical features (age, gender, and smoking history) were added to build classification models together with radiomic features. Subsequently, the top-10 most relevant features were used to establish classifiers. For the classifying tasks including EGFR mutation, exon-19 deletion, and exon-21 L858R mutation, four logistic regression models were established for each task. Results: The training and test cohort consisted of 210 and 53 patients, respectively. Among the established models, the highest accuracy and sensitivity among the four models were 75.5% (61.7-86.2%) and 92.9% (76.5-99.1%) to classify EGFR mutation, respectively. The highest specificity values were 86.7% (69.3-96.2%) and 70.4% (49.8-86.3%) to classify exon-19 deletion and exon-21 L858R mutation, respectively. Conclusions: CT radiomics can sensitively identify the presence of EGFR mutation, and increase the certainty of distinguishing exon-19 deletion and exon-21 L858R mutation in lung adenocarcinoma patients. CT radiomics may become a helpful non-invasive biomarker to select EGFR mutation patients for invasive sampling

    Analysis of large-scale molecular biological data using self-organizing maps

    Get PDF
    Modern high-throughput technologies such as microarrays, next generation sequencing and mass spectrometry provide huge amounts of data per measurement and challenge traditional analyses. New strategies of data processing, visualization and functional analysis are inevitable. This thesis presents an approach which applies a machine learning technique known as self organizing maps (SOMs). SOMs enable the parallel sample- and feature-centered view of molecular phenotypes combined with strong visualization and second-level analysis capabilities. We developed a comprehensive analysis and visualization pipeline based on SOMs. The unsupervised SOM mapping projects the initially high number of features, such as gene expression profiles, to meta-feature clusters of similar and hence potentially co-regulated single features. This reduction of dimension is attained by the re-weighting of primary information and does not entail a loss of primary information in contrast to simple filtering approaches. The meta-data provided by the SOM algorithm is visualized in terms of intuitive mosaic portraits. Sample-specific and common properties shared between samples emerge as a handful of localized spots in the portraits collecting groups of co-regulated and co-expressed meta-features. This characteristic color patterns reflect the data landscape of each sample and promote immediate identification of (meta-)features of interest. It will be demonstrated that SOM portraits transform large and heterogeneous sets of molecular biological data into an atlas of sample-specific texture maps which can be directly compared in terms of similarities and dissimilarities. Spot-clusters of correlated meta-features can be extracted from the SOM portraits in a subsequent step of aggregation. This spot-clustering effectively enables reduction of the dimensionality of the data in two subsequent steps towards a handful of signature modules in an unsupervised fashion. Furthermore we demonstrate that analysis techniques provide enhanced resolution if applied to the meta-features. The improved discrimination power of meta-features in downstream analyses such as hierarchical clustering, independent component analysis or pairwise correlation analysis is ascribed to essentially two facts: Firstly, the set of meta-features better represents the diversity of patterns and modes inherent in the data and secondly, it also possesses the better signal-to-noise characteristics as a comparable collection of single features. Additionally to the pattern-driven feature selection in the SOM portraits, we apply statistical measures to detect significantly differential features between sample classes. Implementation of scoring measurements supplements the basal SOM algorithm. Further, two variants of functional enrichment analyses are introduced which link sample specific patterns of the meta-feature landscape with biological knowledge and support functional interpretation of the data based on the ‘guilt by association’ principle. Finally, case studies selected from different ‘OMIC’ realms are presented in this thesis. In particular, molecular phenotype data derived from expression microarrays (mRNA, miRNA), sequencing (DNA methylation, histone modification patterns) or mass spectrometry (proteome), and also genotype data (SNP-microarrays) is analyzed. It is shown that the SOM analysis pipeline implies strong application capabilities and covers a broad range of potential purposes ranging from time series and treatment-vs.-control experiments to discrimination of samples according to genotypic, phenotypic or taxonomic classifications

    Nephroblastoma in MRI Data

    Get PDF
    The main objective of this work is the mathematical analysis of nephroblastoma in MRI sequences. At the beginning we provide two different datasets for segmentation and classification. Based on the first dataset, we analyze the current clinical practice regarding therapy planning on the basis of annotations of a single radiologist. We can show with our benchmark that this approach is not optimal and that there may be significant differences between human annotators and even radiologists. In addition, we demonstrate that the approximation of the tumor shape currently used is too coarse granular and thus prone to errors. We address this problem and develop a method for interactive segmentation that allows an intuitive and accurate annotation of the tumor. While the first part of this thesis is mainly concerned with the segmentation of Wilms’ tumors, the second part deals with the reliability of diagnosis and the planning of the course of therapy. The second data set we compiled allows us to develop a method that dramatically improves the differential diagnosis between nephroblastoma and its precursor lesion nephroblastomatosis. Finally, we can show that even the standard MRI modality for Wilms’ tumors is sufficient to estimate the developmental tendencies of nephroblastoma under chemotherapy

    Identification of triple negative breast cancer genes using rough set based feature selection algorithm & ensemble classifier

    Get PDF
    In recent decades, microarray datasets have played an important role in triple negative breast cancer (TNBC) detection. Microarray data classification is a challenging process due to the presence of numerous redundant and irrelevant features. Therefore, feature selection becomes irreplaceable in this research field that eliminates non-required feature vectors from the system. The selection of an optimal number of features significantly reduces the NP hard problem, so a rough set-based feature selection algorithm is used in this manuscript for selecting the optimal feature values. Initially, the datasets related to TNBC are acquired from gene expression omnibuses like GSE45827, GSE76275, GSE65194, GSE3744, GSE21653, and GSE7904. Then, a robust multi-array average technique is used for eliminating the outlier samples of TNBC/non-TNBC which helps enhancing classification performance. Further, the pre-processed microarray data are fed to a rough set theory for optimal gene selection, and then the selected genes are given as the inputs to the ensemble classification technique for classifying low-risk genes (non-TNBC) and high-risk genes (TNBC). The experimental evaluation showed that the ensemble-based rough set model obtained a mean accuracy of 97.24%, which superior related to other comparative machine learning techniques.Web of Science12art. no. 5

    Class discovery via feature selection in unsupervised settings

    Full text link
    Identifying genes linked to the appearance of certain types of cancers and their phenotypes is a well-known and challenging problem in bioinformatics. Discovering marker genes which, upon genetic mutation, drive the proliferation of different types and subtypes of cancer is critical for the development of advanced tests and therapies that will specifically identify, target, and treat certain cancers. Therefore, it is crucial to find methods that are successful in recovering "cancer-critical genes" from the (usually much larger) set of all genes in the human genome. We approach this problem in the statistical context as a feature (or variable) selection problem for clustering, in the case where the number of important features is typically small (or rare) and the signal of each important feature is typically minimal (or weak). Genetic datasets typically consist of hundreds of samples (n) each with tens of thousands gene-level measurements (p), resulting in the well-known statistical "large p small n" problem. The class or cluster identification is based on the clinical information associated with the type or subtype of the cancer (either known or unknown) for each individual. We discuss and develop novel feature ranking methods, which complement and build upon current methods in the field. These ranking methods are used to select features which contain the most significant information for clustering. Retaining only a small set of useful features based on this ranking aids in both a reduction in data dimensionality, as well as the identification of a set of genes that are crucial in understanding cancer subtypes. In this paper, we present an outline of cutting-edge feature selection methods, and provide a detailed explanation of our own contributions to the field. We explain both the practical properties and theoretical advantages of the new tools that we have developed. Additionally, we explore a well-developed case study applying these new feature selection methods to different levels of genetic data to explore their practical implementation within the field of bioinformatics
    corecore