Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 15 February 2019
Sec. Hematologic Malignancies

Time to Treatment Prediction in Chronic Lymphocytic Leukemia Based on New Transcriptional Patterns

\r\nAdrin Mosquera Orgueira,,*Adrián Mosquera Orgueira1,2,3*Beatriz Antelo Rodríguez,,Beatriz Antelo Rodríguez1,2,3Natalia Alonso Vence,Natalia Alonso Vence1,2ngeles Bendaa Lpez,Ángeles Bendaña López1,2Jos ngel Díaz Arias,José Ángel Díaz Arias1,2Nicols Díaz VarelaNicolás Díaz Varela2Marta Sonia Gonzlez Prez,Marta Sonia González Pérez1,2Manuel Mateo Prez Encinas,Manuel Mateo Pérez Encinas1,3Jos Luis Bello Lpez,,José Luis Bello López1,2,3
  • 1Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
  • 2Division of Hematology, Complexo Hospitalario Universitario de Santiago de Compostela, SERGAS, Santiago de Compostela, Spain
  • 3Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain

Chronic lymphocytic leukemia (CLL) is the most frequent lymphoproliferative syndrome in western countries. CLL evolution is frequently indolent, and treatment is mostly reserved for those patients with signs or symptoms of disease progression. In this work, we used RNA sequencing data from the International Cancer Genome Consortium CLL cohort to determine new gene expression patterns that correlate with clinical evolution.We determined that a 290-gene expression signature, in addition to immunoglobulin heavy chain variable region (IGHV) mutation status, stratifies patients into four groups with notably different time to first treatment. This finding was confirmed in an independent cohort. Similarly, we present a machine learning algorithm that predicts the need for treatment within the first 5 years following diagnosis using expression data from 2,198 genes. This predictor achieved 90% precision and 89% accuracy when classifying independent CLL cases. Our findings indicate that CLL progression risk largely correlates with particular transcriptomic patterns and paves the way for the identification of high-risk patients who might benefit from prompt therapy following diagnosis.

Introduction

Chronic lymphocytic leukemia (CLL) is a low-grade B-cell lymphoproliferative disease with an estimated yearly incidence in western countries of about 6.9 cases per 100,000 people (1) and remarkable variation between races. The incidence of CLL is higher in men than in women and it increases progressively from the age of 35 until the last decades of life (2). Currently, CLL treatment is delayed until disease progression (bone marrow failure, organomegaly, general symptoms, or high-grade lymphoma transformation) and in the case of refractory autoimmune phenomena (3, 4). Nevertheless, with the advent of new targeted treatments such as ibrutinib (5), idelalisib (6), and venetoclax (7), it is tempting to speculate that some individuals could benefit from early intervention immediately following diagnosis, when the tumoral mass is smaller and patients have a better physical condition. Thus, improved risk stratification for patients with CLL is needed.

Recent advances in CLL genomics have discovered new drivers of disease, many of which are associated with a different clinical evolution. Deletions (6p21, 6q15, 11q, 14q24, 15q15, 17p, 18p, and 20p; gains in 2p16, 5q24, and 8q24), trisomy 12, and gene mutations (TP53, ATM, NOTCH1, SF3B1, BIRC3, BRAF, POT1, ZNF292, NFKB2, MGA, IRF4, DDX3X, ZMYM3, and FUBP1)have been repeatedly observed in the CLL genome and are linked to rapid disease progression (8) Nevertheless, immunoglobulin heavy chain variable region (IGHV) mutation status, which is an indirect measure of the tumor lymphocytes' maturation stage (9), is among the most important single predictive factor known to date (10). IGHV unmutated patients show remarkably worse prognosis than IGHV mutated patients (10, 11) and only a few other genomic factors have proven to be associated with clinical evolution independent of this variable. Lymphocyte maturation is such an important indicator that DNA methylation status has been used to classify CLL into three different groups that resemble different B cell maturation stages (naive B cell, intermediate, and memory B cell). This classification was shown to outperform IGHV status at predicting time to first treatment (TTT) (12).

Mutations, genomic aberrations, and DNA methylation patterns induce transcriptomic changes that can be measured using RNA sequencing (RNAseq), a technique that offers an opportunity to identify new biomarkers for disease progression and drug response prediction (1315). In fact, previous efforts to improve CLL risk stratification based on RNAseq data have demonstrated impressive results (16), but the clinical application is difficult due to the expense of extensive technical and bioinformatics efforts. Therefore, there is a need for smaller transcriptomics patterns correlated with disease evolution for medical use.

In this study, we performed machine-learning based Gaussian mixture model clustering on a subgroup of genes significantly associated with TTT in order to identify transcriptional clusters with clinical implications. We studied TTT due to the lack of treatment uniformity in the International Cancer Genome Consortium (ICGC) CLL cohort and because it is a variable associated with overall survival (17). We tested our results on a 196 patient cohort and validated its clinical significance in an independent 79 patient cohort. The overall results delineated two IGHV-independent transcriptional clusters that stratify patients according to their risk of treatment initiation. Furthermore, we demonstrated that machine learning algorithms using gene expression data can predict patient need for treatment in the first 5 years following diagnosis. We anticipate that our findings will improve the identification of high-risk CLL patients following diagnosis.

Materials and Methods

Data Sources and Patient Characteristics

We applied for access to the ICGC's CLL sequencing data (18) deposited in the European Genome-Phenome Database (EGA) (19). The Data Access Committee approved access to this data under DACO-1040945. Two CLL RNA-seq cohorts were uploaded in two stages with the following accession codes: EGAD00001001443 and EGAD00001000258.

The first cohort (EGAD00001001443, hereafter study cohort) contains RNAseq data and from CLL-purified cells of 196 individuals along with clinical data. The cohort was composed of 169 CLL, 22 monoclonal B cell lymphocytosis (MBL), and five small lymphocytic lymphoma (SLL) samples. There were 132 IGHV mutated cases and 64 IGHV unmutated cases in 119 males and 77 females. By staging at diagnosis, there were 22 MBL cases, 151 Binet Stage A cases, 14 Binet Stage B cases, and 8 Binet C stage cases.

The second cohort (EGAD00001000258, hereafter validation cohort) is composed of RNAseq data of CLL-purified cells from 98 individuals, of which 79 (55 males and 24 females) have publicly available phenotypic information. In this cohort there were 72 CLL, 4 SLL, and 3 MBL samples. 45 of the patients had mutated IGHV and 34 had unmutated IGHV. By staging at diagnosis, there were 3 MBL, 72 Binet Stage A, 3 Binet Stage B, and 1 Binet Stage C cases.

A summary of the patient characteristics of both cohorts can be consulted in Table 1.

TABLE 1
www.frontiersin.org

Table 1. Patient characteristics for the test and validation cohorts.

Data Preprocessing and Alignment

RNAseq paired-end data were obtained from Illumina paired-end sequencing performed by the ICGC CLL consortium as described by Ferreira et al. (16) Illumina adapters were removed using cutadapt (20) and alignment to the human reference genome (GRCh37) was performed using Hisat2 (21) with default specifications. We used the Hisat2-provided Hierarchical Graph FM index for GRCh37 with SNP and Ensembl transcript information. Bam files were sorted and indexed using samtools (22).

Gene Expression Estimation

RNAseq bam files were processed in R (23) according to the RNAseq gene expression protocol developed by Love et al. (24) Briefly, bam files were read using Rsamtools, (25) followed by gene-level expression estimation using the SummarizeOverlaps function from the GenomicAlignments package. (26) Gene models in GTF format were downloaded from Ensembl (GRCh37.75 version) (27). Genes with a median read count below one were discarded.

Statistical Analysis

We analyzed gene expression association with CLL's TTT using cox regression implemented in the survival package (28, 29). In this model we included the covariates donor sex and CLL stage (MBL, Binet Stage A, Binet Stage B, and Binet Stage C). Time to Treatment was calculated as the period between CLL diagnosis and the initiation of the first treatment for CLL. The day of last follow-up was used for right censoring the data of patients with incomplete follow-up.

Clustering was performed using the Mclust package (30) with default parameters. Briefly, Mclust infers the likeliest data clusters based on Gaussian Mixture Modeling fitted by an Expectation-Maximization (GMM-EM) algorithm.

Those genes with significant association with TTT in the study cohort (cox regression false discovery rate [FDR] below 5%) were selected as our initial list of genes. Variable selection was performed by adding one new gene in p-value ascending order to the model (starting with the first two most significant genes until reaching the top at 2,198 genes [FDR < 5%]) and computing the most likely clusters. For the sake of simplicity, we discarded the 25% least variable genes, the 50% least expressed genes and those with a high (>0.9) Spearman's rank correlation with any other gene in the input data. In the case of a highly correlated pair of genes, the one with the lowest p-value was discarded. In each iteration we forced Mclust to calculate the two most likely groups of samples in our data, and to select the best model according to the maximal Bayesian Information Criterion (BIC). Association with TTT calculated using cox regression (survival package), including IGHV mutation status as covariate in each iteration. P-value adjustment was performed with the Bonferroni method.

Machine Learning Ensembl Construction

For IGHV status and need of treatment at 5 years prediction we ran boosted trees analysis using BigML applications (31) with a 2,000 tree node threshold. We chose 5 years due to the following reasons: (1) it is important to differ which patients will have progression in the first years since diagnosis; and (2) the number of cases progressing in earlier years was too small in order to train a good classificator. Varying percentages of learning rates were tested. The best model was selected based on receiver operating characteristic (ROC) curves, Precision-Recall curves, and Kolmogorov-Smirnov statistics.

Results

Genes Associated With Time to Treatment and Clusterization

A cox regression model was constructed with gene expression, donor sex and CLL stage at diagnosis as independent variables. 2,198 genes were found to be significantly associated with TTT (FDR < 5%) in the study cohort.

Patient clusterization based on gene expression data using a GMM-EM algorithm retrieved 19 sets of genes that clustered samples into two groups with significant associations with TTT when adjusted for IGHV status (Bonferroni-adjusted p-value < 0.01) (Supplemental Table 1). The most significant cluster (cluster 2) contained 290 transcripts (Figure 1, Supplemental Table 2) and achieved an association p-value of 6.4 × 10−7 (Bonferroni p-value 1.4 × 10−3) with the TTT variable adjusted for IGHV mutation status (Figure 2). A significant association was confirmed in the validation cohort (IGHV adjusted p-value 3.05 × 10−3) (Figures 2, 3).

FIGURE 1
www.frontiersin.org

Figure 1. Heatmap showing the rank-transformed distribution of expression values for the 290 genes in the study cohort. Red-labeled samples on the left bar petain to C1 and blue-labeled samples pertain to C2.

FIGURE 2
www.frontiersin.org

Figure 2. Kaplan-Meier survival plots. The upper plots show the association of C1 (red curve) and C2 (blue curve) with TTT in the study (left) and validation cohorts (right). Corresponding p-values are 1.7 × 10−6 and 1.3 × 10−4. The lower plots show the association with TTT stratified by IGHV mutation status in the study (left) and validation cohorts (right). The blue line indicates C2 samples with mutated IGHV, the purple line indicates C2 samples with unmutated IGHV, the red line indicates C1 samples with mutated IGHV, and the green line refers to C1 samples with unmutated IGHV.

FIGURE 3
www.frontiersin.org

Figure 3. Heatmap showing the rank-transformed distribution of expression values for the 290 genes in the validation cohort. Red-labeled samples on the left bar petain to C1 and blue-labeled samples pertain to C2.

According to the selected classificator, patients in cluster two (C2) had a more favorable prognosis than patients in cluster one (C1) (Hazard Ratios (HR) of−1.70 and−1.41 in the test and validation cohorts, respectively), independently of IGHV mutation status. Among the study cohort, roughly 36.7% of patients belonged to C2, while 34.1% of patients in the validation cohort clustered within C2. C2 involved 51.5% of IGHV-mutated patients and 6.4% of IGHV-unmutated patients in the study cohort, as well as 55.5% of IGHV-mutated patients and 5.8% of IGHV-unmutated patients in the validation cohort.

Machine Learning for Treatment Free Survival Prediction

We were interested in a machine learning (ML) classifier that could predict which patients would require CLL therapy in the first years following diagnosis. We constructed model ensembles with all genes associated with TTT in Cox regression at a FDR of 5%. We also tested different learning rates (0.5, 1, 2.5, 5, and 10%). 222 patients had a follow-up period >5years or had been treated in the first 5 years following diagnosis, and we divided them into a training set (80% of patients, composed of 146 patients from the study cohort and 31 patients from the validation cohort) and a test set (20% of patients, composed of 45 patients from the validation cohort). ROC AUC and Precision-Recall AUC plots were evaluated to select the best results.

The best model used a 2.5% learning rate and 2,000 tree nodes. It achieved 90% precision at identifying patients that needed treatment in 5 years with 69.23% recall, and 88.57% precision at identifying those patients that did not require treatment in 5 years with 96.88% recall. We only detected 1 false positive case (3.1% False Positive Rate) and 4 false negatives (30% False Negative Rate). Average precision was 89.29%, accuracy was 88.89% and ROC area under the curve (AUC) was 0.911 (Figure 4 and Table 2). Precision-Recall AUC was 0.860 and 0.959 for predicting which patients would or would not need treatment within this period, respectively. In each case, the results mostly overlapped with the area under the convex hull (AUCH).

FIGURE 4
www.frontiersin.org

Figure 4. ROC curve of the boosted-tree Ensembl model for 5 year treatment need prediction (upper left). Kolmogorov-Smirnov plot for the same model (upper right). Precision-Recall plot for the 5 year not-treated (lower left) and treated (lower right) patients according to the same model. White dots in each graph indicate the probability threshold (in this case 50%), which is the point reflecting the best classification accuracy of the patients.

TABLE 2
www.frontiersin.org

Table 2. Confusion Matrix for the boosted-tree Ensembl model predicting the 5 year need of treatment.

Discussion

The main aim of this study was to identify new transcriptomic patterns in order to improve CLL patient risk stratification. We used the GMM-EM algorithm to stratify patients in two clusters with remarkably different clinical behavior based on the expression of 290 genes, and we observed that this pattern was independent of IGHV mutation status. Interestingly, we identified a group of CLL patients with mutated IGHV and a low-risk transcriptomic profile that only need treatment in approximately 25% of the cases during disease evolution. Two additional groups (one composed of patients with mutated IGHV and a high-risk transcriptomic profile and the second composed of unmutated IGHV patients with a low-risk transcriptomic profile) have similar intermediate evolution, while a final group (composed of patients with unmutated IGHV and an adverse transcriptomic profile) has the highest probability of treatment need in the first years following diagnosis. These results are concordant with previous reports in the field. For example, Yepes et al. (32) reported a division of CLL cases in two groups based on microarray transcriptome characterization through unsupervised clustering analysis, which was validated in 4 independent cohorts. Similarly, Friedman et al. (33) described a 180 probe classifier based on microarray data that also divided two clusters of CLL patients independently of IGHV mutation status. Our findings are also similar to those published by Ferreira et al. (16), who described two gene expression clusters that show IGHV mutation-independent association with TTT using an early release of the ICGC CLL cohort. Nevertheless, there are remarkable differences between our analysis and that of Ferreira et al, Yepes et al. and Friedman et al. Firstly, our clusterization is based on a transcriptional pattern of a small subgroup of genes that facilitates its future applicability, whilst those of Ferreira et al. and Yepes et al. are based on whole transcriptome analysis. Secondly, our classifier is based on RNAseq data, a technology that has outperformed microarray analysis in most fields. With the use RNAseq it will be possible to couple transcriptome clusterization with targeted gene mutation detection, stereotyped B cell receptor expression or IGHV hypermutation status analysis.

We also describe a novel artificial intelligence algorithm that can predict a CLL patient's need for therapy during the first 5 years following diagnosis with high precision and accuracy. This is in line with other ML applications to oncologic malignancies that are starting to change paradigms in patient risk stratification and drug response prediction. For example, Aziz et al. (34) recently reported the identification of a ML model that integrates clinical and genomic data from patients with myelodysplastic syndrome (MDS). This model outperformed all commonly used prediction models in the field of MDS. Similarly, Yousefi et al. (35) used bayesian-optimized deep learning for survival prediction in pan-cancer analysis, showing not only better performance than other state-of-the-art methods, but also improved predictability of cancer survival through transfer learning in different types of cancer genomic data. Thus, it is likely that ML-driven algorithms applied to genomic and transcriptomic data will be used in the near future for the identification of “smoldering” CLLs that may benefit from early intervention.

RNAseq is a powerful technique that can sequence the whole transcriptome at an increasingly lower cost. Targeted RNAseq is being developed for clinical application, with the additional possibility of testing for gene mutations and fusion genes in the same technique. Therefore, defining reproducible gene expression patterns with clinical implications is a strategy that can close the gap between research and the clinical practice. Here we present patterns of gene expression that can improve CLL patient risk stratification with a relatively small set of the transcriptome. These results may pave the way for the design of new treatment strategies involving early CLL treatment in high-risk patients before disease progression.

Author Contributions

AMO designed the study performed research. AMO, BAR, JDA, and JBL analyzed the data. AMO wrote the paper. NAV, ABL, NDV, MGP, and MPE reviewed the paper.

Funding

The publication costs associated with this manuscript have been paid by Roche Pharmaceuticals. The funder played no role in the study design, the collection, analysis or interpretation of data, the writing of this paper or the decision to submit it for publication.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The authors thank the ICGC consortium for sharing the genomic data and the Centro de Supercomputación de Galicia (CESGA) for approving the access to informatics facilities with technical support. We would also like to thank BigML Inc., particularly Francisco J. Martín, for access to machine learning applications. The content of this paper is part of the doctoral thesis of AMO to obtain a PhD at the Department of Medicine, University of Santiago de Compostela.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2019.00079/full#supplementary-material

Supplemental Table 1. This table represents the original 19 clusters with Bonferroni p-values < 0.01.

Supplemental Table 2. List of the 290 genes used for C1 vs. C2 clusterization.

References

1. Zhao Y, Wang Y, Ma S. Racial differences in four leukemia subtypes: comprehensive descriptive epidemiology. Sci Rep. (2018) 8:548. doi: 10.1038/s41598-017-19081-4

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Dores GM, Anderson WF, Curtis RE, Landgren O, Ostroumova E, Bluhm EC, et al. Chronic lymphocytic leukaemia and small lymphocytic lymphoma: overview of the descriptive epidemiology. Br J Haematol. (2007) 139:809–19. doi: 10.1111/j.1365-2141.2007.06856.x

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Mozas P, Rivas-Delgado A, Baumann T, Villamor N, Ortiz-Maldonado V, Aymerich M, et al. Analysis of criteria for treatment initiation in patients with progressive chronic lymphocytic leukemia. Blood Cancer J. (2018) 8:10. doi: 10.1038/s41408-017-0044-5

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Eichhorst B, Robak T, Montserrat E, Ghia P, Hillmen P, Hallek M, et al. Chronic lymphocytic leukaemia: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. (2015) 26 (Suppl. 5):v78–84. doi: 10.1093/annonc/mdv303

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, et al. Ibrutinib as Initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. (2015) 373:2425–37. doi: 10.1056/NEJMoa1509388

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Brown JR, Byrd JC, Coutre SE, Benson DM, Flinn IW, Wagner-Johnston ND, et al. Idelalisib, an inhibitor of phosphatidylinositol 3-kinase p110δ, for relapsed/refractory chronic lymphocytic leukemia. Blood (2014) 123:3390–7. doi: 10.1182/blood-2013-11-535047

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Roberts AW, Davids MS, Pagel JM, Kahl BS, Puvvada SD, Gerecitano JF, et al. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. (2016) 374:311–22. doi: 10.1056/NEJMoa1513257

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Puente XS, Beà S, Valdés-Mas R, Villamor N, Gutiérrez-Abril J, Martín-Subero JI, et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature (2015) 526:519–24. doi: 10.1038/nature14666

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Oakes CC, Seifert M, Assenov Y, Gu L, Przekopowitz M, Ruppert AS, et al. DNA methylation dynamics during B cell maturation underlie a continuum of disease phenotypes in chronic lymphocytic leukemia. Nat Genet. (2016) 48:253–64. doi: 10.1038/ng.3488

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Damle RN, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, et al. Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood (1999) 94:1840–7.

PubMed Abstract | Google Scholar

11. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood (1999) 94:1848–54.

PubMed Abstract | Google Scholar

12. Queirós AC, Villamor N, Clot G, Martinez-Trillos A, Kulis M, Navarro A, et al. A B-cell epigenetic signature defines three biologic subgroups of chronic lymphocytic leukemia with clinical impact. Leukemia (2015) 29:598–605. doi: 10.1038/leu.2014.252

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Maag JLV, Fisher OM, Levert-Mignon A, Kaczorowski DC, Thomas ML, Hussey DJ, et al. Novel aberrations uncovered in barrett's esophagus and esophageal adenocarcinoma using whole transcriptome sequencing. Mol Cancer Res. (2017) 15:1558–69. doi: 10.1158/1541-7786.MCR-17-0332

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Wang Q, Gan H, Chen C, Sun Y, Chen J, Xu M, et al. Identification and validation of a 44-gene expression signature for the classification of renal cell carcinomas. J Exp Clin Cancer Res. (2017) 36:176. doi: 10.1186/s13046-017-0651-9

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Zhang YH, Huang T, Chen L, Xu Y, Hu Y, Hu LD, et al. Identifying and analyzing different cancer subtypes using RNA-seq data of blood platelets. Oncotarget (2017) 8:87494–511. doi: 10.18632/oncotarget.20903

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Ferreira PG, Jares P, Rico D, Gómez-López G, Martínez-Trillos A, Villamor N, et al. Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome Res. (2014) 24:212–26. doi: 10.1101/gr.152132.112

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Ammann EM, Shanafelt TD, Wright KB, McDowell BD, Link BK, Chrischilles EA. Updating survival estimates in patients with chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL) based on treatment-free interval length. Leuk Lymphoma (2018) 59:643–9. doi: 10.1080/10428194.2017.1349905

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Ramsay AJ, Martínez-Trillos A, Jares P, Rodríguez D, Kwarciak A, Quesada V. Next-generation sequencing reveals the secrets of the chronic lymphocytic leukemia genome. Clin Transl Oncol. (2013) 15:3–8. doi: 10.1007/s12094-012-0922-z

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Lappalainen I, Almeida-King J, Kumanduri V, Senf A, Spalding JD, Ur-Rehman S, et al. The European Genome-phenome Archive of human data consented for biomedical research. Nat Genet. (2015) 47:692–5. doi: 10.1038/ng.3312

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Martin M. Cutadapt Removes Adapter Sequences From High-Throughput Sequencing Reads. EMBnet.journal, [S.l.], v. 17, n. 1, p. pp. 10-12, may. (2011) ISSN 2226-6089. Available online at: < http://journal.embnet.org/index.php/embnetjournal/article/view/200> (accessed February 15, 2018).

21. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. (2015) 12:357–60. doi: 10.1038/nmeth.3317

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics (2009) 25:2078–9. doi: 10.1093/bioinformatics/btp352

PubMed Abstract | CrossRef Full Text | Google Scholar

23. R Development Core Team R: A Language and Environment for Statistical Computing. Vienna, Austria: the R Foundation for Statistical Computing. ISBN: 3-900051-07-0. Available online at: http://www.R-project.org/ (2011)

PubMed Abstract

24. Love MI, Anders S, Kim V, Huber W. RNA-seq Workflow: Gene-level Exploratory Analysis and Differential Expression. Bioconductor. Available online at: https://www.bioconductor.org/help/workflows/rnaseqGene/ (2017)

PubMed Abstract

25. Morgan M, Pagès H, Obenchain V, Hayden N. Rsamtools: Binary alignment (BAM), FASTA, Variant Call (BCF), and Tabix File Import. R package version 1.30.0, Available online at: http://bioconductor.org/packages/release/bioc/html/Rsamtools.html (2017)

26. Lawrence M, Huber W, Pagès H, Aboyoun P, Carlson M, Gentleman R., et al. Software for computing and annotating genomic ranges. PLoS ComputBiol. (2013) 9:e1003118. doi: 10.1371/journal.pcbi.1003118

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Yates A, Akanni W, Amode MR, Barrell D, Billis K, Carvalho-Silva D, et al. Ensembl 2016. Nucleic Acids Res. (2016) 44(D1):D710–6. doi: 10.1093/nar/gkv1157

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Therneau T (2015). A Package for Survival Analysis in S. version 2.38, Available online at: https://CRAN.R-project.org/package=survival

29. Therneau TM, Grambsch PM. Modeling Survival Data: Extending the Cox Model. New York, NY: Springer (2000) doi: 10.1007/978-1-4757-3294-8

CrossRef Full Text | Google Scholar

30. Scrucca L, Fop M, Murphy TB, Raftery AE. mclust 5: clustering, classification and density estimation using gaussian finite mixture models. R J. (2016) 8:289–317.

PubMed Abstract | Google Scholar

31. BigML is Machine Learning made easy, viewed, 15 February 2018, Available, online at: https://bigml.com

32. Yepes S, Torres MM, Andrade RE. Clustering of expression data in chronic lymphocytic leukemia reveals new molecular subdivisions. PLoS ONE (2015) 10:e0137132. doi: 10.1371/journal.pone.0137132

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Friedman DR, Weinberg JB, Barry WT, Goodman BK, Volkheimer AD, Bond KM, et al. A genomic approach to improve prognosis and predict therapeutic response in chronic lymphocytic leukemia. Clin Cancer Res. (2009) 15:6947–55. doi: 10.1158/1078-0432.CCR-09-1132

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Nazha A, Komrokji RS, Barnard J, Al-Issa K., Padron E., Madanat Y. F, et al. A personalized prediction model to risk stratify patients with myelodysplastic syndromes (MDS). Blood (2017) 130(Suppl 1):160.

Google Scholar

35. Yousefi S, Amrollahi F, Amgad M, Dong C, Lewis JE, Song C, et al. Predicting clinical outcomes from large scale cancer genomic profiles with deep survival models. Sci Rep. (2017) 7:11707. doi: 10.1038/s41598-017-11817-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: chronic lymphocytic leukemia, time to treatment prediction, gene expression, RNAseq, machine learning, prognostic factors, IGHV

Citation: Mosquera Orgueira A, Antelo Rodríguez B, Alonso Vence N, Bendaña López Á, Díaz Arias JÁ, Díaz Varela N, González Pérez MS, Pérez Encinas MM and Bello López JL (2019) Time to Treatment Prediction in Chronic Lymphocytic Leukemia Based on New Transcriptional Patterns. Front. Oncol. 9:79. doi: 10.3389/fonc.2019.00079

Received: 23 November 2018; Accepted: 29 January 2019;
Published: 15 February 2019.

Edited by:

Adam Finn Binder, Thomas Jefferson University, United States

Reviewed by:

Michael Diamantidis, University Hospital of Larissa, Greece
Jerry Polesel, Centro di Riferimento Oncologico di Aviano (IRCCS), Italy

Copyright © 2019 Mosquera Orgueira, Antelo Rodríguez, Alonso Vence, Bendaña López, Díaz Arias, Díaz Varela, González Pérez, Pérez Encinas and Bello López. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Adrián Mosquera Orgueira, adrian.mosquera@live.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.