777 research outputs found

    Stage-Specific Predictive Models for Cancer Survivability

    Get PDF
    Survivability of cancer strongly depends on the stage of cancer. In most previous works, machine learning survivability prediction models for a particular cancer, were trained and evaluated together on all stages of the cancer. In this work, we trained and evaluated survivability prediction models for five major cancers, together on all stages and separately for every stage. We named these models joint and stage-specific models respectively. The obtained results for the cancers which we investigated reveal that, the best model to predict the survivability of the cancer for one specific stage is the model which is specifically built for that stage. Additionally, we saw that for every stage of cancer, the most important features to predict survivability, differed from other stages. By evaluating the models separately on different stages we found that their performance differed on different stages. We also found that evaluating the models together on all stages, as was done in past, is misleading because it overestimates performance

    Applications of Machine Learning in Cancer Prediction and Prognosis

    Get PDF
    Machine learning is a branch of artificial intelligence that employs a variety of statistical, probabilistic and optimization techniques that allows computers to “learn” from past examples and to detect hard-to-discern patterns from large, noisy or complex data sets. This capability is particularly well-suited to medical applications, especially those that depend on complex proteomic and genomic measurements. As a result, machine learning is frequently used in cancer diagnosis and detection. More recently machine learning has been applied to cancer prognosis and prediction. This latter approach is particularly interesting as it is part of a growing trend towards personalized, predictive medicine. In assembling this review we conducted a broad survey of the different types of machine learning methods being used, the types of data being integrated and the performance of these methods in cancer prediction and prognosis. A number of trends are noted, including a growing dependence on protein biomarkers and microarray data, a strong bias towards applications in prostate and breast cancer, and a heavy reliance on “older” technologies such artificial neural networks (ANNs) instead of more recently developed or more easily interpretable machine learning methods. A number of published studies also appear to lack an appropriate level of validation or testing. Among the better designed and validated studies it is clear that machine learning methods can be used to substantially (15–25%) improve the accuracy of predicting cancer susceptibility, recurrence and mortality. At a more fundamental level, it is also evident that machine learning is also helping to improve our basic understanding of cancer development and progression

    Predicting breast cancer risk, recurrence and survivability

    Full text link
    This thesis focuses on predicting breast cancer at early stages by using machine learning algorithms based on biological datasets. The accuracy of those algorithms has been improved to enable the physicians to enhance the success of treatment, thus saving lives and avoiding several further medical tests

    Gene expression profiling of breast cancer survivability by pooled cDNA microarray analysis using logistic regression, artificial neural networks and decision trees

    Get PDF
    BACKGROUND: Microarray technology can acquire information about thousands of genes simultaneously. We analyzed published breast cancer microarray databases to predict five-year recurrence and compared the performance of three data mining algorithms of artificial neural networks (ANN), decision trees (DT) and logistic regression (LR) and two composite models of DT-ANN and DT-LR. The collection of microarray datasets from the Gene Expression Omnibus, four breast cancer datasets were pooled for predicting five-year breast cancer relapse. After data compilation, 757 subjects, 5 clinical variables and 13,452 genetic variables were aggregated. The bootstrap method, Mann–Whitney U test and 20-fold cross-validation were performed to investigate candidate genes with 100 most-significant p-values. The predictive powers of DT, LR and ANN models were assessed using accuracy and the area under ROC curve. The associated genes were evaluated using Cox regression. RESULTS: The DT models exhibited the lowest predictive power and the poorest extrapolation when applied to the test samples. The ANN models displayed the best predictive power and showed the best extrapolation. The 21 most-associated genes, as determined by integration of each model, were analyzed using Cox regression with a 3.53-fold (95% CI: 2.24-5.58) increased risk of breast cancer five-year recurrence… CONCLUSIONS: The 21 selected genes can predict breast cancer recurrence. Among these genes, CCNB1, PLK1 and TOP2A are in the cell cycle G2/M DNA damage checkpoint pathway. Oncologists can offer the genetic information for patients when understanding the gene expression profiles on breast cancer recurrence

    Breast cancer data analysis for survivability studies and prediction

    Full text link
    © 2017 Elsevier B.V. Background Breast cancer is the most common cancer affecting females worldwide. Breast cancer survivability prediction is challenging and a complex research task. Existing approaches engage statistical methods or supervised machine learning to assess/predict the survival prospects of patients. Objective The main objectives of this paper is to develop a robust data analytical model which can assist in (i) a better understanding of breast cancer survivability in presence of missing data, (ii) providing better insights into factors associated with patient survivability, and (iii) establishing cohorts of patients that share similar properties. Methods Unsupervised data mining methods viz. the self-organising map (SOM) and density-based spatial clustering of applications with noise (DBSCAN) is used to create patient cohort clusters. These clusters, with associated patterns, were used to train multilayer perceptron (MLP) model for improved patient survivability analysis. A large dataset available from SEER program is used in this study to identify patterns associated with the survivability of breast cancer patients. Information gain was computed for the purpose of variable selection. All of these methods are data-driven and require little (if any) input from users or experts. Results SOM consolidated patients into cohorts of patients with similar properties. From this, DBSCAN identified and extracted nine cohorts (clusters). It is found that patients in each of the nine clusters have different survivability time. The separation of patients into clusters improved the overall survival prediction accuracy based on MLP and revealed intricate conditions that affect the accuracy of a prediction. Conclusions A new, entirely data driven approach based on unsupervised learning methods improves understanding and helps identify patterns associated with the survivability of patient. The results of the analysis can be used to segment the historical patient data into clusters or subsets, which share common variable values and survivability. The survivability prediction accuracy of a MLP is improved by using identified patient cohorts as opposed to using raw historical data. Analysis of variable values in each cohort provide better insights into survivability of a particular subgroup of breast cancer patients

    Machine learning application in personalised lung cancer recurrence and survivability prediction

    Get PDF
    Machine learning is an important artificial intelligence technique that is widely applied in cancer diagnosis and detection. More recently, with the rise of personalised and precision medicine, there is a growing trend towards machine learning applications for prognosis prediction. However, to date, building reliable prediction models of cancer outcomes in everyday clinical practice is still a hurdle. In this work, we integrate genomic, clinical and demographic data of lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) patients from The Cancer Genome Atlas (TCGA) and introduce copy number variation (CNV) and mutation information of 15 selected genes to generate predictive models for recurrence and survivability. We compare the accuracy and benefits of three well-established machine learning algorithms: decision tree methods, neural networks and support vector machines. Although the accuracy of predictive models using the decision tree method has no significant advantage, the tree models reveal the most important predictors among genomic information (e.g. KRAS, EGFR, TP53), clinical status (e.g. TNM stage and radiotherapy) and demographics (e.g. age and gender) and how they influence the prediction of recurrence and survivability for both early stage LUAD and LUSC. The machine learning models have the potential to help clinicians to make personalised decisions on aspects such as follow-up timeline and to assist with personalised planning of future social care needs

    Methods to Improve the Prediction Accuracy and Performance of Ensemble Models

    Get PDF
    The application of ensemble predictive models has been an important research area in predicting medical diagnostics, engineering diagnostics, and other related smart devices and related technologies. Most of the current predictive models are complex and not reliable despite numerous efforts in the past by the research community. The performance accuracy of the predictive models have not always been realised due to many factors such as complexity and class imbalance. Therefore there is a need to improve the predictive accuracy of current ensemble models and to enhance their applications and reliability and non-visual predictive tools. The research work presented in this thesis has adopted a pragmatic phased approach to propose and develop new ensemble models using multiple methods and validated the methods through rigorous testing and implementation in different phases. The first phase comprises of empirical investigations on standalone and ensemble algorithms that were carried out to ascertain their performance effects on complexity and simplicity of the classifiers. The second phase comprises of an improved ensemble model based on the integration of Extended Kalman Filter (EKF), Radial Basis Function Network (RBFN) and AdaBoost algorithms. The third phase comprises of an extended model based on early stop concepts, AdaBoost algorithm, and statistical performance of the training samples to minimize overfitting performance of the proposed model. The fourth phase comprises of an enhanced analytical multivariate logistic regression predictive model developed to minimize the complexity and improve prediction accuracy of logistic regression model. To facilitate the practical application of the proposed models; an ensemble non-invasive analytical tool is proposed and developed. The tool links the gap between theoretical concepts and practical application of theories to predict breast cancer survivability. The empirical findings suggested that: (1) increasing the complexity and topology of algorithms does not necessarily lead to a better algorithmic performance, (2) boosting by resampling performs slightly better than boosting by reweighting, (3) the prediction accuracy of the proposed ensemble EKF-RBFN-AdaBoost model performed better than several established ensemble models, (4) the proposed early stopped model converges faster and minimizes overfitting better compare with other models, (5) the proposed multivariate logistic regression concept minimizes the complexity models (6) the performance of the proposed analytical non-invasive tool performed comparatively better than many of the benchmark analytical tools used in predicting breast cancers and diabetics ailments. The research contributions to ensemble practice are: (1) the integration and development of EKF, RBFN and AdaBoost algorithms as an ensemble model, (2) the development and validation of ensemble model based on early stop concepts, AdaBoost, and statistical concepts of the training samples, (3) the development and validation of predictive logistic regression model based on breast cancer, and (4) the development and validation of a non-invasive breast cancer analytic tools based on the proposed and developed predictive models in this thesis. To validate prediction accuracy of ensemble models, in this thesis the proposed models were applied in modelling breast cancer survivability and diabetics’ diagnostic tasks. In comparison with other established models the simulation results of the models showed improved predictive accuracy. The research outlines the benefits of the proposed models, whilst proposes new directions for future work that could further extend and improve the proposed models discussed in this thesis

    Integration of Data Mining Classification Techniques and Ensemble Learning for Predicting the Type of Breast Cancer Recurrence

    Get PDF
    Conservative surgery plus radiotherapy is an alternative to radical mastectomy in the early stages of breast cancer, presenting equivalent survival rates. Data mining facilitates to manage the data and provide the useful medical progression and treatment of cancerous conditions as these methods can help to reduce the number of false positive and false negative decisions. Various machine learning techniques can be used to support the doctors in effective and accurate decision making. In this paper, various classifiers have been tested for the prediction of type of breast cancer recurrence and the results show that neural networks outperform others

    Comparison of machine learning algorithms for the prediction of five-year survival in oral squamous cell carcinoma

    Get PDF
    BACKGROUND/AIM Machine learning analyses of cancer outcomes for oral cancer remain sparse compared to other types of cancer like breast or lung. The purpose of the present study was to compare the performance of machine learning algorithms in the prediction of global, recurrence-free five-year survival in oral cancer patients based on clinical and histopathological data.METHODS Data were gathered retrospectively from 416 patients with oral squamous cell carcinoma. The data set was divided into training and test data set (75:25 split). Training performance of five machine learning algorithms (Logistic regression, K-nearest neighbours, Naïve Bayes, Decision tree and Random forest classifiers) for prediction was assessed by k-fold cross-validation. Variables used in the machine learning models were age, sex, pain symptoms, grade of lesion, lymphovascular invasion, extracapsular extension, perineural invasion, bone invasion and type of treatment. Variable importance was assessed and model performance on the testing data was assessed using receiver operating characteristic curves, accuracy, sensitivity, specificity and F1 score.RESULTS The best performing model was the Decision tree classifier, followed by the Logistic Regression model (accuracy 76% and 60%, respectively). The Naïve Bayes model did not display any predictive value with 0% specificity.CONCLUSIONS Machine learning presents a promising and accessible toolset for improving prediction of oral cancer outcomes. Our findings add to a growing body of evidence that Decision tree models are useful in models in predicting OSCC outcomes. We would advise that future similar studies explore a variety of machine learning models including Logistic regression to help evaluate model performance.</p

    Discriminative and informative subspace assessment with categorical and numerical outcomes

    Get PDF
    CEECIND/01399/2017Pattern discovery and subspace clustering play a central role in the biological domain, supporting for instance putative regulatory module discovery from omics data for both descriptive and predictive ends. In the presence of target variables (e.g. phenotypes), regulatory patterns should further satisfy delineate discriminative power properties, well-established in the presence of categorical outcomes, yet largely disregarded for numerical outcomes, such as risk profiles and quantitative phenotypes. DISA (Discriminative and Informative Subspace Assessment), a Python software package, is proposed to evaluate patterns in the presence of numerical outcomes using well-established measures together with a novel principle able to statistically assess the correlation gain of the subspace against the overall space. Results confirm the possibility to soundly extend discriminative criteria towards numerical outcomes without the drawbacks well-associated with discretization procedures. Results from four case studies confirm the validity and relevance of the proposed methods, further unveiling critical directions for research on biotechnology and biomedicine. Availability: DISA is freely available at https://github.com/JupitersMight/DISA under the MIT license.publishersversionpublishe
    corecore