Skip to main content

Machine learning and systems genomics approaches for multi-omics data

Abstract

In light of recent advances in biomedical computing, big data science, and precision medicine, there is a mammoth demand for establishing algorithms in machine learning and systems genomics (MLSG), together with multi-omics data, to weigh probable phenotype-genotype relationships. Software frameworks in MLSG are extensively employed to analyze hundreds of thousands of multi-omics data by high-throughput technologies. In this study, we reviewed the MLSG software frameworks and future directions with respect to multi-omics data analysis and integration. Our review was targeted at researching recent approaches and technical solutions for the MLSG software frameworks using multi-omics platforms.

Background

Over the past few years, researchers and scientists have made remarkable progress in the interdisciplinary fields of precision medicine, data mining and predictive algorithms, bioinformatics, and computational medicine [1]. Machine learning and systems genomics (MLSG) approaches integrate multiple data types from multi-omics data by using data mining and predictive algorithms, pointing out that the MLSG approaches can support a more meaningful interpretation of phenotype-genotype relationships than an analysis using only a single data type. Therefore, there is an acute need for development of the MLSG software frameworks that can generate prediction of a given quantitative or categorical phenotype using next-generation multi-omic data [2].

Precision medicine, an emerging field of medicine, is becoming the cornerstone of medical practices with prospects of the customization of healthcare, which means medical decisions, practices, and treatments are tailored to individual patients [3]. The use of genomic biomarkers, such as multi-omics data, has played a major role in precision medicine in oncology and other chronic diseases such as asthma [4], mental disorders [5, 6], and diabetes [7–9]. More specifically, patients are divided into groups by genetic variability and other biomarkers so that medications may be tailored to individual patients with similar or related genetic characteristics [10, 11]. For example, accumulating evidence reveals that selected single nucleotide polymorphisms (SNPs) could be used as genetic markers to influence clinical treatment response and adverse drug reactions for antidepressants in patients with major depressive disorder [12–14]. With the advent of technology in multi-omics approaches such as genomics, proteomics, metabolomics, and epigenomics, we are able to employ materials or devices that can interact with biological systems at the molecular level and then target different molecules with high precision.

In big data science, machine learning methods are computer algorithms that can automatically learn to recognize complex patterns based on empirical data [15, 16]. The goal of an machine learning method is to enable an algorithm to learn from data of the past or present and use that knowledge to make predictions or decisions for unknown future events [17, 18]. In the general terms, the workflow for an machine learning method consists of three phases including build the model from example inputs, evaluate and tune the model, and then put the model into production in prediction-making. Some of the best-known algorithms in machine learning methods include naive Bayes [19], C4.5 decision tree [20], artificial neural networks (ANNs) [21–23], support vector machine (SVM) [24], k-Means [25], k-nearest neighbors (kNN) [26], and regression [27, 28]. There were some key emerging diagnostics studies for various diseases and treatments of significance for public health with consideration of machine learning methods, including applications in mental health [29–33], cancer [34–38], and pharmacogenetics [39–41].

In this review, we surveyed the MLSG software frameworks that could enable definite assessment of the phenotype-genotype interplay status by using multi-omics platforms. The MLSG software frameworks encompass the model-based integration (MBI), concatenation-based integration (CBI), and transformation-based integration (TBI) approaches (Table 1). Furthermore, we investigated some potential data reduction and feature selection approaches that can be leveraged together with the MLSG software frameworks. Finally, we summarized the future perspectives with respect to the MLSG approaches.

Table 1 Summary, strength, and limitation of each method of machine learning and systems genomics (MLSG) software frameworks

Model-based integration approach

First, we explored the MBI approach, which generates multiple models using different data types as training sets, and then generates a final model from the multiple models created during the training phase (Fig. 1). One advantage of the MBI approach is that this approach can merge predictive models from different data types and each data type can be assembled from a different set of patients with same phenotype [42].

Fig. 1
figure 1

A flowchart for the model-based integration (MBI) software framework

In order to identify interactions between different levels of genomic data associated with certain disease or phenotype (for example, survival in ovarian cancer), the MBI approach can integrate multi-omics data, including, but not limited to, miRNA, methylation, gene expression, and copy number variation data. The MBI approach can then conduct the final multi-dimensional model from a particular machine learning algorithm (for example, Bayesian networks) with variables from the best models of each individual genomic dataset. Next, the MBI approach can compare the predictive power of the integration model with the one of the individual model from single level of genomic data to see whether the integration model can show the improvement. Finally, the MBI approach can obtain the best multi-dimensional model of all variables from multi-omics dimension as well as a balanced accuracy for the final model.

In the literature, the MBI approach encompasses the following computational frameworks for constructing a model: a majority voting approach [43], an ensemble classifier approach [44], and probabilistic causal networks [45]. In addition, we can employ the Analysis Tool for Heritable and Environmental Network Associations methodology, which is a suite of analysis tools for integrating multi-omics data [46].

Probabilistic causal network framework

In order to integrate highly dissimilar types of data, we can leverage Bayesian networks that are one type of probabilistic causal networks [47]. Bayesian networks are directed acyclic graphs where the edges of the graph are represented by conditional probabilities, which define the distribution of states of each node given the state of its parents [47]. In Bayesian networks, each node characterizes a quantitative trait that can be a genomic factor (such as variation in DNA, gene expression, methylation, metabolite, and protein). These conditional probabilities represent not only relationships between genomic factors, but also the stochastic nature of these relationships. By assuming the observed data as a function of our prior belief, the Bayes formula is used to determine the likelihood of a Bayesian network model. Because the number of potential network structures grows super-exponentially with the number of nodes, it is infeasible to find the best model by an exhaustive search of all possible structures. Therefore, we can utilize Monte Carlo Markov Chain simulation [48] to pinpoint probably a huge amount of different plausible Bayesian networks, which are then integrated to accomplish a consensus network model. In the beginning, there is a null network. Then, slight arbitrary changes are made to the network by flipping, adding, or deleting individual edges. Ultimately, accepting those changes will lead to an overall improvement by fitting the network to the data. In order to avoid over-fitting owing to the addition of new parameters, the Bayesian information criterion score [49] can be employed to assess whether a change improves the network model.

Ensemble classifier framework

In order to reduce the variance caused by the distinctiveness of a single genomic factor, Shen and Chou employed ensemble classifier models to integrate multiple classifiers, where each of those classifiers was based on individual genomic factor [50]. Thus, ensemble classifier models were able to obtain a more concrete concept in classification than a single classifier. The final output of the ensemble classifier model was the weighted fusion of the outputs generated by the individual basic classifiers. The weighted factor was assigned with the value of the success rate obtained by the individual basic classifier. Here, Shen and Chou adopted the optimized evidence-theoretic K-nearest-neighbors algorithm for the basic classifier [50].

Concatenation-based integration approach

Second, we investigated the CBI approach, which combines multiple data matrices for each dataset into one large input matrix before constructing a model (Fig. 2). One advantage of the CBI approach is that, after we determine how to combine all of the variables into one matrix, it is relatively simple to employ a variety of machine learning methods for analyzing continuous or categorical data [42].

Fig. 2
figure 2

A flowchart for the concatenation-based integration (CBI) software framework

In the literature, the CBI approach encompasses the following computational frameworks for constructing a model: Bayesian networks [51], multivariate Cox LASSO models [52], grammatical evolution neural networks [53], iCluster [54], Bayesian correlated clustering [55], and Bayesian consensus clustering [56]. In addition, We can consider some of the best-known machine learning algorithms including naive Bayes [19], C4.5 decision tree [20], ANNs [21–23], SVM [24], k-Means [25], kNN [26], and regression [27, 28]. Depending on the number of variables in the data matrix, we can also employ data reduction and feature selection methods as described below.

In order to assess response to cancer therapeutics such as gemcitabine, Fridley et al. employed a Bayesian integrative model, which combines the ideas of Bayesian pathway analysis with Bayesian variable selection using stochastic search variable selection [51]. They employed two various high-throughput multi-omics datasets, such as mRNA expression and SNPs data, which were integrated into one large input matrix [51]. Fridley et al. reported that the Bayesian integrative model had greater sensitivity to detect genomic effects in the drug gemcitabine, as compared to the traditional single data type analysis [51].

Furthermore, instead of a single data type, Shen et al. implemented the iCluster framework to carry out cancer subtype discovery in glioblastoma using three multi-omics data types such as copy number data, mRNA expression data, and methylation data [54]. The iCluster framework is a CBI method that can simultaneously accomplish both data integration and dimension reduction to combine multi-omics data into one large input matrix [54]. Shen et al. revealed three distinct integrated tumor subtypes by using iCluster and multi-omics data [54].

Transformation-based integration approach

Third, we assessed the TBI approach, which transforms each dataset into an intermediate form, such as a graph or a kernel matrix, and then merges multiple graphs or kernels into one before constructing a model (Fig. 3). One advantage of the TBI approach is that this approach can be employed to integrate a variety of continuous or categorical data values if the data contain unique variables such as patient identifiers for linking multi-omics data types [42].

Fig. 3
figure 3

A flowchart for the transformation-based integration (TBI) software framework

In the literature, the TBI approach encompasses the following statistical frameworks for constructing a model: a kernel-based integration method [57] and a graph-based semi-supervised learning method [58]. The TBI approach investigates whether there is a relevant intermediate representation, such as a kernel or graph, for each multi-omics data type.

In order to find metabolic consequences underlying body weight change, Wahl et al. implemented a weighted correlation network approach [59], which was inferred using the Gaussian graphical model [60]. Instead of a single data type, they leveraged two different high-throughput multi-omics datasets, such as serum metabolomics and whole blood gene expression [59]. Wahl et al. first clustered multi-omics data into intermediate forms, namely modules of closely connected molecules, and then constructed a partial correlation network from the modules. Their analysis revealed that four metabolite and two gene expression modules were significantly associated with body weight change, indicating an association of long-term weight change with serum metabolite concentrations [59].

Data reduction and feature selection approach

Accounting for models is not a trivial task because even a relatively small set of factors results in the large number of possible models [61]. For example, if we study 10 factors, then these 10 factors yield 210 possible models. The purpose of data reduction and feature selection approaches is to find a subset of factors that maximizes the performance of the prediction model, depending on how these methods incorporate the feature selection search with the classification algorithms. There are two data reduction and feature selection approaches including extrinsic approaches (which use information external to the data set itself) and intrinsic approaches (which use the data set and some analytical technique for filtering). The extrinsic approaches, such as Biofilter [62], employ prior knowledge that is accessible in the public domain. Additionally, the intrinsic approaches encompass factor analysis [63], ReliefF [64], chi-square statistics, principal component analysis [65], and genetic algorithms [66].

Furthermore, a hybrid approach, which combines the information-gain method and the chi-squared method, is designed to reduce bias introduced by each of the methods [67]. Each feature is measured and ranked according to its merit in both methods. The measurement of the merit for the two methods is defined as follows. The information-gain method measures the decrease in the entropy of a given feature provided by another feature, and the chi-squared method is based on Pearson chi-squared statistic to measure divergence from the expected distribution. Next, all features are sorted by their average rank across these two methods. After the features are ranked, the classifiers are utilized to add one feature at a time based on its individual ranking and then select the desired number of the top ranked features that provides the best predictive performance, respectively.

Moreover, in a wrapper-based feature selection approach, the feature selection algorithm acts as a wrapper around the classification algorithm. The wrapper-based feature selection approach conducts best-first search for a good subset using the classification algorithm itself as part of the function for evaluating feature subsets [68, 69]. Best first search starts with empty set of features and searches forward to select possible subsets of features by greedy hill-climbing augmented with a backtracking technique [18].

Future perspective

The MLSG modeling is essential to root out the false positive candidate genes discovered at the current association analyses by using meta-analysis, epistasis analysis, and pathway models [13]. Using multi-omics data not only could take care of missing information from any single data source, but also could help bridge the gap between phenotypes and more comprehensive biological regulation models [70]. In future research, models in MLSG will be established to predict the probability of drug efficacy to guide clinicians in choosing medications. In order to establish models for predicting drug efficacy, techniques in MLSG may provide a plausible way to predict drug efficacy in therapy. Finally, data analysis and integration in MLSG may play a key role in weighing gene–gene and gene–environment interactions.

Conclusions

In this study, we reviewed several recent findings and relevant studies in terms of the MLSG software frameworks. The work also underscores the importance of techniques in MLSG to track down a greater diversity of populations in the clinical settings of diseases and their treatments. In fact, facilitating the MLSG tools based on multi-omics data plays a pivotal role, economically and clinically, in predicting the possible outcomes of diseases and treatments. Future research using the MLSG approaches is needed in order to weigh the interplay among clinical factors and multi-omics data.

Abbreviations

ANNs:

Artificial neural networks

CBI:

Concatenation-based integration

kNN:

k-nearest neighbors

MBI:

Model-based integration

MLSG:

Machine learning and systems genomics

SNPs:

Single nucleotide polymorphisms

SVM:

Support vector machine

TBI:

Transformation-based integration

References

  1. Katsanis SH, Javitt J, Hudson K. A case study of personalized medicine. Science. 2008;v320(4):53–4.

    Article  Google Scholar 

  2. Snyderman R. Personalized health care: from theory to practice. Biotechnol J. 2012;v7:973–9.

    Article  Google Scholar 

  3. Lin E. Novel drug therapies and diagnostics for personalized medicine and nanomedicine in genome science, nanoscience, and molecular engineering. Pharma Regul Aff. 2012;1:e116.

    Google Scholar 

  4. Lin E, Lin CG, Wang JY, Wu LS. Gene-gene interactions among genetic variants from seven candidate genes with pediatric asthma in a Taiwanese population. Curr Topics Genet. 2009;3:83–8.

    Google Scholar 

  5. Lin E, Hong CJ, Hwang JP, Liou YJ, Yang CH, Cheng D, et al. Gene-gene interactions of the brain-derived neurotrophic-factor and neurotrophic tyrosine kinase receptor 2 genes in geriatric depression. Rejuvenation Res. 2009;12(6):387–93.

    Article  CAS  PubMed  Google Scholar 

  6. Lin E, Tsai SJ. Gene-gene interactions in a context of individual variability in antipsychotic drug pharmacogenomics. Curr Pharmacogenomics Person Med. 2011;9:323–31.

    Article  CAS  Google Scholar 

  7. Lin E, Pei D, Huang YJ, Hsieh CH, Wu LS. Gene-gene interactions among genetic variants from obesity candidate genes for nonobese and obese populations in type 2 diabetes. Genet Test Mol Biomarkers. 2009;13(4):485–93.

    Article  CAS  PubMed  Google Scholar 

  8. Wu LS, Hsieh CH, Pei D, Hung YJ, Kuo SW, Lin E. Association and interaction analyses of genetic variants in ADIPOQ, ENPP1, GHSR, PPARgamma and TCF7L2 genes for diabetic nephropathy in a Taiwanese population with type 2 diabetes. Nephrol Dial Transplant. 2009;24(11):3360–6.

    Article  CAS  PubMed  Google Scholar 

  9. Wang CH, Ke WS, Lin E. Evaluation of the ENPP1 and PLIN single nucleotide polymorphisms with type 2 diabetes in a Taiwanese population: evidence for replication and gene-gene interaction. J Investig Med. 2012;60(8):1169–73.

    Article  CAS  PubMed  Google Scholar 

  10. Lin E, Tsai SJ. Novel diagnostics R&D for public health and personalized medicine in Taiwan: current state, challenges and opportunities. Curr Pharmacogenomics Person Med. 2012;10:239–46.

    Article  CAS  Google Scholar 

  11. Lin E, Hwang Y, Tzeng CM. A case study of the utility of the HapMap database for pharmacogenomic haplotype analysis in the Taiwanese population. Mol Diagn Ther. 2006;10:367–70.

    Article  CAS  PubMed  Google Scholar 

  12. Lin E, Chen PS. Pharmacogenomics with antidepressants in the STAR*D study. Pharmacogenomics. 2008;9:935–46.

    Article  CAS  PubMed  Google Scholar 

  13. Lin E, Lane HY. Genome-wide association studies in pharmacogenomics of antidepressants. Pharmacogenomics. 2015;16(5):555–66.

    Article  CAS  PubMed  Google Scholar 

  14. Lin E, Tsai SJ. Genome-wide microarray analysis of gene expression profiling in major depression and antidepressant therapy. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:334–40.

    Article  CAS  PubMed  Google Scholar 

  15. Kononenko I. Machine learning for medical diagnosis: history, state of the art and perspective. Artif Intell Med. 2001;23(1):89–109.

    Article  CAS  PubMed  Google Scholar 

  16. Lane HY, Tsai GE, Lin E. Assessing gene-gene interactions in pharmacogenomics. Mol Diagn Ther. 2012;16(1):15–27.

    Article  CAS  PubMed  Google Scholar 

  17. Landset S, Khoshgoftaar TM, Richter AN, Hasanin T. A survey of open source tools for machine learning with big data in the hadoop ecosystem. J Big Data. 2015;2:24.

    Article  Google Scholar 

  18. Lin E, Tsai SJ. Machine learning and predictive algorithms for personalized medicine: from physiology to treatment. In: Turnbull A, editor. Personalized medicine. New York: Nova Science Publishers (in press).

  19. Domingos P, Pazzani M. On the optimality of the simple Bayesian classifier under zero–one loss. Mach Learn. 1997;29:103–37.

    Article  Google Scholar 

  20. Quinlan JR. C4.5: programs for machine learning. San Francisco: Morgan Kaufmann Publishers; 1993.

    Google Scholar 

  21. Kung SY, Hwang JN. Neural networks for intelligent multimedia processing. Proc IEEE. 1998;86:1244–72.

    Article  Google Scholar 

  22. Bishop CM. Neural networks for pattern recognition. Oxford: Clarendon; 1995.

    Google Scholar 

  23. Rumelhart DE, Hinton GE, William RJ. Learning internal representation by error propagation. In: Parallel distributed processing: explorations in the microstructure of cognition. Cambridge: MIT Press; 1996. p. 318–62.

    Google Scholar 

  24. Vapnik V. The nature of statistical learning theory. New York: Springer; 1995.

    Book  Google Scholar 

  25. Lloyd SP. Least squares quantization in PCM. IEEE Trans Inform Theory (Special Issue on Quantization). 1982;IT-28:129–37.

    Article  Google Scholar 

  26. Altman NS. An introduction to kernel and nearest-neighbor nonparametric regression. Am Stat. 1992;46(3):175–85.

    Google Scholar 

  27. Friedman J, Hastie T, Tibshirani R. Regularization paths for generalized linear models via coordinate descent. J Stat Softw. 2010;33(1):1–22.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zou H, Hastie T. Regularization and variable selection via the elastic net. J R Stat Soc Series B Stat Methodol. 2005;67(suppl):301–20.

    Article  Google Scholar 

  29. Kessler RC, Warner CH, Ivany C, Petukhova MV, Rose S, Bromet EJ, et al. Predicting suicides after psychiatric hospitalization in US army soldiers. JAMA Psychiatry. 2015;72(1):49–57.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Huang LC, Hsu SY, Lin E. A comparison of classification methods for predicting chronic fatigue syndrome based on genetic data. J Transl Med. 2009;7:81.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Lin E, Chen PS, Yang YK, Lee IH, Yeh TL, Gean PW, et al. Modeling short-term antidepressant responsiveness with artificial neural networks. Open Access Bioinform. 2010;2:55–60.

    Article  CAS  Google Scholar 

  32. Lin E, Tsai SJ. Genetics and suicide. In: Courtet P, editor. Understanding suicide - from diagnosis to personalized treatment. Switzerland: Springer; 2016.

    Google Scholar 

  33. Lin E, Hsu SY. Gender differences and pharmacogenomics with antidepressants in depression. In: Hernandez P, Alonso S, editors. Women and Depression. New York: Nova; 2009.

    Google Scholar 

  34. Kim W, Kim KS, Lee JE, Noh DY, Kim SW, Jung YS, et al. Development of novel breast cancer recurrence prediction model using support vector machine. J Breast Cancer. 2012;15(2):230–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Tseng CJ, Lu CJ, Chang CC, Chen GD. Application of machine learning to predict the recurrence-proneness for cervical cancer. Neural Comput & Applic. 2014;24(6):1311–6.

    Article  Google Scholar 

  36. Chen YC, Ke WC, Chiu HW. Risk classification of cancer survival using ANN with gene expression data from multiple laboratories. Comput Biol Med. 2014;48:1–7.

    Article  PubMed  Google Scholar 

  37. Chang SW, Abdul-Kareem S, Merican AF, Zain RB. Oral cancer prognosis based on clinicopathologic and genomic markers using a hybrid of feature selection and machine learning methods. BMC Bioinformatics. 2013;14:170.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Rosado P, Lequerica-Fernández P, Villallaín L, Peña I, Sanchez-Lasheras F, de Vicente JC. Survival model in oral squamous cell carcinoma based on clinicopathological parameters, molecular markers and support vector machines. Expert Syst Appl. 2013;40(12):4770–6.

    Article  Google Scholar 

  39. Lin E, Hwang Y, Wang SC, Gu ZJ, Chen EY. An artificial neural network approach to the drug efficacy of interferon treatments. Pharmacogenomics. 2006;7:1017–24.

    Article  CAS  PubMed  Google Scholar 

  40. Ke WS, Hwang Y, Lin E. Pharmacogenomics of drug efficacy in the interferon treatment of chronic hepatitis C using classification algorithms. Adv Appl Bioinform Chem. 2010;3:39–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Lin E, Hwang Y. A support vector machine approach to assess drug efficacy of interferon-alpha and ribavirin combination therapy. Mol Diagn Ther. 2008;12:219–23.

    Article  PubMed  Google Scholar 

  42. Ritchie MD, Holzinger ER, Li R, Pendergrass SA, Kim D. Methods of integrating data to uncover genotype-phenotype interactions. Nat Rev Genet. 2015;16(2):85–97.

    Article  CAS  PubMed  Google Scholar 

  43. Draghici S, Potter RB. Predicting HIV drug resistance with neural networks. Bioinformatics. 2003;19:98–107.

    Article  CAS  PubMed  Google Scholar 

  44. Akavia UD, Litvin O, Kim J, Sanchez-Garcia F, Kotliar D, Causton HC, et al. An integrated approach to uncover drivers of cancer. Cell. 2010;143:1005–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhu J, Sova P, Xu Q, Dombek KM, Xu EY, Vu H, et al. Stitching together multiple data dimensions reveals interacting metabolomic and transcriptomic networks that modulate cell regulation. PLoS Biol. 2012;10:e1001301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim D, Li R, Dudek SM, Ritchie MD. ATHENA: identifying interactions between different levels of genomic data associated with cancer clinical outcomes using grammatical evolution neural network. BioData Min. 2013;6:23.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Pearl J. Probabilistic reasoning in intelligent systems: networks of plausible inference. San Mateo: Morgan Kaufmann Publishers; 1988.

    Google Scholar 

  48. Madigan D, York J. Bayesian graphical models for discrete data. Int Stat Rev. 1995;63:215–32.

    Article  Google Scholar 

  49. Schwarz G. Estimating the dimension of a model. Ann Stat. 1978;6:461–4.

    Article  Google Scholar 

  50. Shen HB, Chou KC. Ensemble classifier for protein fold pattern recognition. Bioinformatics. 2006;22(14):1717–22.

    Article  CAS  PubMed  Google Scholar 

  51. Fridley BL, Lund S, Jenkins GD, Wang LA. Bayesian integrative genomic model for pathway analysis of complex traits. Genet Epidemiol. 2012;36:352–9.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mankoo PK, Shen R, Schultz N, Levine DA, Sander C. Time to recurrence and survival in serous ovarian tumors predicted from integrated genomic profiles. PLoS ONE. 2011;6:e24709.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Holzinger ER, Dudek SM, Frase AT, Pendergrass SA, Ritchie MD. ATHENA: the analysis tool for heritable and environmental network associations. Bioinformatics. 2014;30:698–705.

    Article  CAS  PubMed  Google Scholar 

  54. Shen R, Mo Q, Schultz N, Seshan VE, Olshen AB, Huse J, et al. Integrative subtype discovery in glioblastoma using iCluster. PLoS ONE. 2012;7:e35236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kirk P, Griffin JE, Savage RS, Ghahramani Z, Wild DL. Bayesian correlated clustering to integrate multiple datasets. Bioinformatics. 2012;28:3290–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Lock EF, Dunson DB. Bayesian consensus clustering. Bioinformatics. 2013;29:2610–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lanckriet GRG, De Bie T, Cristianini N, Jordan MI, Noble WS. A statistical framework for genomic data fusion. Bioinformatics. 2004;20:2626–35.

    Article  CAS  PubMed  Google Scholar 

  58. Shin H, Lisewski AM, Lichtarge O. Graph sharpening plus graph integration: a synergy that improves protein functional classification. Bioinformatics. 2007;23:3217–24.

    Article  CAS  PubMed  Google Scholar 

  59. Wahl S, Vogt S, Stückler F, Krumsiek J, Bartel J, Kacprowski T, et al. Multi-omic signature of body weight change: results from a population-based cohort study. BMC Med. 2015;13:48.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Krumsiek J, Suhre K, Illig T, Adamski J, Theis FJ. Gaussian graphical modeling reconstructs pathway reactions from high-throughput metabolomics data. BMC Syst Biol. 2011;5:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Lin E, Huang LC. Identification of significant genes in genomics using Bayesian variable selection methods. Adv Appl Bioinform Chem. 2008;1:13–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Bush WS, Dudek SM, Ritchie MD. Biofilter: a knowledge-integration system for the multi-locus analysis of genome-wide association studies. Pac Symp Biocomput. 2009;368–379.

  63. Hastie T, Tibshirani R, Friedman J. The elements of statistical learning: data mining, inference, and prediction. New York: Springer; 2001.

    Book  Google Scholar 

  64. Greene CS, Penrod NM, Kiralis J, Moore JH. Spatially uniform ReliefF (SURF) for computationally efficient filtering of gene–gene interactions. BioData Min. 2009;2:5.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Zou H, Hastie T, Tibshirani R. Sparse principal component analysis. J Comput Graph Stat. 2006;15:265–86.

    Article  Google Scholar 

  66. Holland JH. Genetic algorithms. Sci Am. 1992;267:66–72.

    Article  Google Scholar 

  67. Saeys Y, Inza I, Larrañaga P. A review of feature selection techniques in bioinformatics. Bioinformatics. 2007;23:2507–17.

    Article  CAS  PubMed  Google Scholar 

  68. Kohavi R, John GH. Wrappers for feature subset selection. Artif Intell. 1997;97:273–324.

    Article  Google Scholar 

  69. Lin E, Hwang Y, Liang KH, Chen EY. Pattern-recognition techniques with haplotype analysis in pharmacogenomics. Pharmacogenomics. 2007;8(1):75–83.

    Article  CAS  PubMed  Google Scholar 

  70. Leung MKK, Delong A, Alipanahi B, Frey BJ. Machine learning in genomic medicine: a review of computational problems and data sets. Proc IEEE. 2016;104(1):176–97.

    Article  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the Ministry of Economic Affairs in Taiwan (SBIR Grant S099000280249-154), Taiwan Ministry of Health and Welfare Clinical Trial and Research Center of Excellence (MOHW105-TDU-B-212-133019), and China Medical University Hospital, Taiwan (DMR-101-091 and DMR-102-069). The funding supports had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

Not applicable.

Authors’ contributions

The present manuscript was drafted by EL and revised by EL and HYL. Both authors read and approved the final manuscript.

Competing interests

Both authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Hsien-Yuan Lane.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, E., Lane, HY. Machine learning and systems genomics approaches for multi-omics data. Biomark Res 5, 2 (2017). https://doi.org/10.1186/s40364-017-0082-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40364-017-0082-y

Keywords