1,381 research outputs found

    IPA: Integrated predictive gene signature from gene expression based breast cancer patient samples

    Full text link
    Background: Novel predictive markers are needed to accurately diagnose the breast cancer patients so they do not need to undergo any unnecessary aggressive therapies. Various gene expression studies based predictive gene signatureshave generated in the recent past to predict the binary estrogen-receptor subclass or to predict the therapy response subclass. However, the existing algorithms comes with many limitations, including low predictive performances over multiple cohorts of patients and non-significant or limited biological roles associated with thepredictive gene signatures. Therefore, the aim of this study is to develop novel predictive markers with improved performances.Methods: We propose a novel prediction algorithm called IPA to construct a predictive gene signature for performing multiple prediction tasks of predicting estrogen-receptor based binary subclass and predicting chemotherapy response (neoadjuvantly) based binary subclass. The constructed gene signature with considering multiple classification techniques was used to evaluate the algorithm performance on multiple cohorts of breast cancer patients.Results: The evaluation on multiple validation cohorts demonstrated that proposed algorithm achieved stable and high performance to perform prediction tasks, with consideration given to any classification techniques. We show that the predictive gene signature of our proposed algorithm reflects the mechanisms underlying the estrogen-receptors or response to therapy with significant greater biological interpretations, compared with the other existing algorithm

    Identifying Network-Biomarkers of Breast Cancer Survivability

    Get PDF
    One of the key challenges of breast cancer research is to predict whether a patient identified with specific subtype or treated with a specific therapy is going to survive or die. Current studies find small subsets of gene biomarkers able to accurately predict the response to therapy. In these studies, the selected genes are not necessarily functionally related, and hence, they may not correctly indicate the molecular mechanism behind breast cancer survivability. Also, several studies have shown there is a very low overlap between the different respective biomarkers subsets for the same cancer disease. To improve the robustness of classification performance and stability of detected biomarkers, recent methods take existing knowledge on relations between genes into account in the classifier, by aggregating functionality related genes to produce discriminative gene subnetworks called network-biomarkers. In this paper, given a breast cancer dataset of patients with different subtypes treated with a given therapy drug, we devised network-based machine learning approach by integrating protein protein interaction network (PPI) with gene expression data (1) to identify the network-biomarkers of breast cancer survivability a) based on subtypes and b) based on therapy and (2) to predict the survivability of breast cancer patients a) based on subtypes b) treated with a therapy drug. We used the concept of seed gene for identification of network-biomarkers with distance 2, 3 and 4 from seed gene protein and our method found distance 3 and 44 are the distance that gives us best result for identifying survivability of breast cancer patient based on subtype and therapy respectively. To solve the class imbalance problem in some subtypes, we implemented ADASYN. We obtained best classification performance using random forest where the geometric mean, F1-measure and accuracy are respectively 0.867, 0.850 and 87.00% for subtype specific study, and 0.829, 0.807 and 83.77%, for therapy specific

    Machine Learning Approaches for Cancer Analysis

    Get PDF
    In addition, we propose many machine learning models that serve as contributions to solve a biological problem. First, we present Zseq, a linear time method that identifies the most informative genomic sequences and reduces the number of biased sequences, sequence duplications, and ambiguous nucleotides. Zseq finds the complexity of the sequences by counting the number of unique k-mers in each sequence as its corresponding score and also takes into the account other factors, such as ambiguous nucleotides or high GC-content percentage in k-mers. Based on a z-score threshold, Zseq sweeps through the sequences again and filters those with a z-score less than the user-defined threshold. Zseq is able to provide a better mapping rate; it reduces the number of ambiguous bases significantly in comparison with other methods. Evaluation of the filtered reads has been conducted by aligning the reads and assembling the transcripts using the reference genome as well as de novo assembly. The assembled transcripts show a better discriminative ability to separate cancer and normal samples in comparison with another state-of-the-art method. Studying the abundance of select mRNA species throughout prostate cancer progression may provide some insight into the molecular mechanisms that advance the disease. In the second contribution of this dissertation, we reveal that the combination of proper clustering, distance function and Index validation for clusters are suitable in identifying outlier transcripts, which show different trending than the majority of the transcripts, the trending of the transcript is the abundance throughout different stages of prostate cancer. We compare this model with standard hierarchical time-series clustering method based on Euclidean distance. Using time-series profile hierarchical clustering methods, we identified stage-specific mRNA species termed outlier transcripts that exhibit unique trending patterns as compared to most other transcripts during disease progression. This method is able to identify those outliers rather than finding patterns among the trending transcripts compared to the hierarchical clustering method based on Euclidean distance. A wet-lab experiment on a biomarker (CAM2G gene) confirmed the result of the computational model. Genes related to these outlier transcripts were found to be strongly associated with cancer, and in particular, prostate cancer. Further investigation of these outlier transcripts in prostate cancer may identify them as potential stage-specific biomarkers that can predict the progression of the disease. Breast cancer, on the other hand, is a widespread type of cancer in females and accounts for a lot of cancer cases and deaths in the world. Identifying the subtype of breast cancer plays a crucial role in selecting the best treatment. In the third contribution, we propose an optimized hierarchical classification model that is used to predict the breast cancer subtype. Suitable filter feature selection methods and new hybrid feature selection methods are utilized to find discriminative genes. Our proposed model achieves 100% accuracy for predicting the breast cancer subtypes using the same or even fewer genes. Studying breast cancer survivability among different patients who received various treatments may help understand the relationship between the survivability and treatment therapy based on gene expression. In the fourth contribution, we have built a classifier system that predicts whether a given breast cancer patient who underwent some form of treatment, which is either hormone therapy, radiotherapy, or surgery will survive beyond five years after the treatment therapy. Our classifier is a tree-based hierarchical approach that partitions breast cancer patients based on survivability classes; each node in the tree is associated with a treatment therapy and finds a predictive subset of genes that can best predict whether a given patient will survive after that particular treatment. We applied our tree-based method to a gene expression dataset that consists of 347 treated breast cancer patients and identified potential biomarker subsets with prediction accuracies ranging from 80.9% to 100%. We have further investigated the roles of many biomarkers through the literature. Studying gene expression through various time intervals of breast cancer survival may provide insights into the recovery of the patients. Discovery of gene indicators can be a crucial step in predicting survivability and handling of breast cancer patients. In the fifth contribution, we propose a hierarchical clustering method to separate dissimilar groups of genes in time-series data as outliers. These isolated outliers, genes that trend differently from other genes, can serve as potential biomarkers of breast cancer survivability. In the last contribution, we introduce a method that uses machine learning techniques to identify transcripts that correlate with prostate cancer development and progression. We have isolated transcripts that have the potential to serve as prognostic indicators and may have significant value in guiding treatment decisions. Our study also supports PTGFR, NREP, scaRNA22, DOCK9, FLVCR2, IK2F3, USP13, and CLASP1 as potential biomarkers to predict prostate cancer progression, especially between stage II and subsequent stages of the disease

    Pathway-Based Genomics Prediction using Generalized Elastic Net.

    Get PDF
    We present a novel regularization scheme called The Generalized Elastic Net (GELnet) that incorporates gene pathway information into feature selection. The proposed formulation is applicable to a wide variety of problems in which the interpretation of predictive features using known molecular interactions is desired. The method naturally steers solutions toward sets of mechanistically interlinked genes. Using experiments on synthetic data, we demonstrate that pathway-guided results maintain, and often improve, the accuracy of predictors even in cases where the full gene network is unknown. We apply the method to predict the drug response of breast cancer cell lines. GELnet is able to reveal genetic determinants of sensitivity and resistance for several compounds. In particular, for an EGFR/HER2 inhibitor, it finds a possible trans-differentiation resistance mechanism missed by the corresponding pathway agnostic approach

    Feature Selection and Molecular Classification of Cancer Using Genetic Programming

    Get PDF
    AbstractDespite important advances in microarray-based molecular classification of tumors, its application in clinical settings remains formidable. This is in part due to the limitation of current analysis programs in discovering robust biomarkers and developing classifiers with a practical set of genes. Genetic programming (GP) is a type of machine learning technique that uses evolutionary algorithm to simulate natural selection as well as population dynamics, hence leading to simple and comprehensible classifiers. Here we applied GP to cancer expression profiling data to select feature genes and build molecular classifiers by mathematical integration of these genes. Analysis of thousands of GP classifiers generated for a prostate cancer data set revealed repetitive use of a set of highly discriminative feature genes, many of which are known to be disease associated. GP classifiers often comprise five or less genes and successfully predict cancer types and subtypes. More importantly, GP classifiers generated in one study are able to predict samples from an independent study, which may have used different microarray platforms. In addition, GP yielded classification accuracy better than or similar to conventional classification methods. Furthermore, the mathematical expression of GP classifiers provides insights into relationships between classifier genes. Taken together, our results demonstrate that GP may be valuable for generating effective classifiers containing a practical set of genes for diagnostic/ prognostic cancer classification

    Optimisation Models for Pathway Activity Inference in Cancer

    Get PDF
    BACKGROUND: With advances in high-throughput technologies, there has been an enormous increase in data related to profiling the activity of molecules in disease. While such data provide more comprehensive information on cellular actions, their large volume and complexity pose difficulty in accurate classification of disease phenotypes. Therefore, novel modelling methods that can improve accuracy while offering interpretable means of analysis are required. Biological pathways can be used to incorporate a priori knowledge of biological interactions to decrease data dimensionality and increase the biological interpretability of machine learning models. METHODOLOGY: A mathematical optimisation model is proposed for pathway activity inference towards precise disease phenotype prediction and is applied to RNA-Seq datasets. The model is based on mixed-integer linear programming (MILP) mathematical optimisation principles and infers pathway activity as the linear combination of pathway member gene expression, multiplying expression values with model-determined gene weights that are optimised to maximise discrimination of phenotype classes and minimise incorrect sample allocation. RESULTS: The model is evaluated on the transcriptome of breast and colorectal cancer, and exhibits solution results of good optimality as well as good prediction performance on related cancer subtypes. Two baseline pathway activity inference methods and three advanced methods are used for comparison. Sample prediction accuracy, robustness against noise expression data, and survival analysis suggest competitive prediction performance of our model while providing interpretability and insight on key pathways and genes. Overall, our work demonstrates that the flexible nature of mathematical programming lends itself well to developing efficient computational strategies for pathway activity inference and disease subtype prediction

    Prediction potential of candidate biomarker sets identified and validated on gene expression data from multiple datasets

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Independently derived expression profiles of the same biological condition often have few genes in common. In this study, we created populations of expression profiles from publicly available microarray datasets of cancer (breast, lymphoma and renal) samples linked to clinical information with an iterative machine learning algorithm. ROC curves were used to assess the prediction error of each profile for classification. We compared the prediction error of profiles correlated with molecular phenotype against profiles correlated with relapse-free status. Prediction error of profiles identified with supervised univariate feature selection algorithms were compared to profiles selected randomly from a) all genes on the microarray platform and b) a list of known disease-related genes (a priori selection). We also determined the relevance of expression profiles on test arrays from independent datasets, measured on either the same or different microarray platforms.</p> <p>Results</p> <p>Highly discriminative expression profiles were produced on both simulated gene expression data and expression data from breast cancer and lymphoma datasets on the basis of ER and BCL-6 expression, respectively. Use of relapse-free status to identify profiles for prognosis prediction resulted in poorly discriminative decision rules. Supervised feature selection resulted in more accurate classifications than random or a priori selection, however, the difference in prediction error decreased as the number of features increased. These results held when decision rules were applied across-datasets to samples profiled on the same microarray platform.</p> <p>Conclusion</p> <p>Our results show that many gene sets predict molecular phenotypes accurately. Given this, expression profiles identified using different training datasets should be expected to show little agreement. In addition, we demonstrate the difficulty in predicting relapse directly from microarray data using supervised machine learning approaches. These findings are relevant to the use of molecular profiling for the identification of candidate biomarker panels.</p

    Inferring Pathway Activity toward Precise Disease Classification

    Get PDF
    The advent of microarray technology has made it possible to classify disease states based on gene expression profiles of patients. Typically, marker genes are selected by measuring the power of their expression profiles to discriminate among patients of different disease states. However, expression-based classification can be challenging in complex diseases due to factors such as cellular heterogeneity within a tissue sample and genetic heterogeneity across patients. A promising technique for coping with these challenges is to incorporate pathway information into the disease classification procedure in order to classify disease based on the activity of entire signaling pathways or protein complexes rather than on the expression levels of individual genes or proteins. We propose a new classification method based on pathway activities inferred for each patient. For each pathway, an activity level is summarized from the gene expression levels of its condition-responsive genes (CORGs), defined as the subset of genes in the pathway whose combined expression delivers optimal discriminative power for the disease phenotype. We show that classifiers using pathway activity achieve better performance than classifiers based on individual gene expression, for both simple and complex case-control studies including differentiation of perturbed from non-perturbed cells and subtyping of several different kinds of cancer. Moreover, the new method outperforms several previous approaches that use a static (i.e., non-conditional) definition of pathways. Within a pathway, the identified CORGs may facilitate the development of better diagnostic markers and the discovery of core alterations in human disease
    • …
    corecore