Identification of biomarkers for the prediction of relapse‑free survival in pediatric B‑precursor acute lymphoblastic leukemia

  • Authors:
    • Wei Jing
    • Jing Li
  • View Affiliations

  • Published online on: November 2, 2018     https://doi.org/10.3892/or.2018.6846
  • Pages: 659-667
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

B‑precursor acute lymphoblastic leukemia (B‑ALL) is the most common cancer diagnosed in children and adolescents. Despite the fact that the 5‑year survival rate has increased from 60 to 90%, approximately a quarter of children suffer from relapse with poor outcome. To improve the clinical management of B‑ALL, there is an urgent need for prognostic biomarkers for the prediction of B‑ALL outcomes. In the present study, we performed a comprehensive analysis of the gene expression data of 456 samples from five independent cohorts. We first sought to identify B‑ALL‑associated genes by differential gene expression analysis by applying linear models. Then, the statistical modelling was applied to identify candidates related to relapse‑free survival. We identified a total of 1,273 B‑ALL‑associated genes that have functions relevant to chemokine signaling. From these genes, 59 genes were identified as prognostic biomarkers. Based on expression patterns of these genes, we successfully distinguished high‑ and low‑risk groups of B‑ALL patients (log-rank test, P‑value=0.025). We further investigated the 59‑gene expression levels in ALL chemotherapy‑treated cohorts and identified 4 genes as potential drug targets associated with drug sensitivity. Our results provided a novel biomarker panel. By leveraging the large scale of data and statistical modelling, we believe this 59‑gene biomarker could help to unveil the mechanisms underlying B‑ALL progression and become potential drug targets.

Introduction

B-precursor acute lymphoblastic leukemia (B-ALL) is the most common cancer diagnosed in children and adolescents (1,2). One of the major causes of mortality is relapse despite intensive multi-agent chemotherapy (3). For the past two decades, several studies have reported that molecular abnormalities including TP53 mutations (4), deletion of INK4A/ARF (5) and TEL deletion (6) contribute to B-ALL relapse. However, the pathogenesis and biological mechanisms underlying relapsed ALL remain largely unknown. Thus, we sought to provide novel insights by identifying prognostic biomarkers from genome-wide expression profiling data generated by DNA microarrays.

Microarray technology has been developed more than a decade ago and is widely used in biomedical and clinical research. This high-throughput strategy enables profiling genome-wide expression simultaneously. Previously, based on committee neural networks, the leukemia gene expression data can be subcategorized into B-cell acute lymphoblastic leukemia, T-cell acute lymphoblastic leukemia and acute myeloid leukemia (7). Unsupervised hierarchal clustering of ALL gene expression could be used to reveal unique clusters with distinct cytogenetic, genomic and transcriptomic characterizations (8). By comparing gene expression profiles of specimens at the time of diagnosis vs. at relapse, or early- vs. late-relapse, several biological pathways such as cell cycle regulation, WNT and mitogen-activated protein kinase pathways (9,10) were identified to contribute to ALL relapse.

However, these findings lack connections to clinical practice, as the prediction of prognosis plays a crucial role in facilitating clinical decision-making. The purpose of this study was to develop a prognostic biomarker from gene expression profiles. Unlike the previously published study (11), we integrated data sets from multiple cohorts and implemented a comprehensive computational pipeline to identify a 59-gene biomarker that could serve as a B-ALL prognostic biomarker in practical applications.

Materials and methods

Gene expression data and preprocessing

We collected raw microarray data from two cohorts including TARGET-ALL study (11) and Microarray Innovations in LEukemia (MILE) (12,13). For the TAGET-ALL study, 207 high-risk B-precursor ALL patients between March 15, 2000 and April 25, 2003 were recruited from the Children's Oncology Group (COG) Clinical Trial P9906. RNA was first purified from samples with >80% blasts (131 bone marrow, 76 peripheral blood) and then hybridized to Affymetrix Human Genome U133 Plus 2.0 Array. The raw intensity *.CEL files were retrieved from NCBI Gene Expression Omnibus (http://www.ncbi.nih.gov/geo) under the accession number GSE11877. For the MILE study, there were in total 2,096 blood or bone marrow samples of acute and chronic leukemia patients from 11 participant centers. In this study, we restricted to take 74 non-leukemia healthy samples as controls. Consistent with the TARGET-ALL study, Affymetrix Human Genome U133 Plus 2.0 Array was also used for the gene expression profiling.

For the discovery of prognostic biomarkers, we used the gene expression data set from one previously published study (14), where 80 samples collected at the time of diagnosis were considered in this study. The RNA was hybridized to Affymetrix HG-U133A oligonucleotide microarrays. The raw intensity *CEL files were retrieved from EBI ArrayExpress (https://www.ebi.ac.uk/arrayexpress/) under the accession number E-MTAB-1216.

The raw intensity *.CEL files were preprocessed using the frma (15) package in R 3.4.1 (https://www.r-project.org) environment. Briefly, the frma package converted raw probe-level intensities into background-corrected gene-level intensities. The data was then normalized based on the estimation of probe-specific effects, which allowed us to combine data from various cohorts without batch effects.

Identification of B-ALL-associated genes

We sought to identify B-ALL-associated genes by performing differential gene expression analysis between B-ALL (n=207) and healthy samples (n=74). The R package limma (16) package was used to conduct the differential gene expression analysis. The expression data was first Log2 transformed and then fitted into linear models with Empirical Bayesian methods for the analysis. We filtered the results at Log2 fold-change ≥2, or ≤-2 and B-statistics ≥4.6, which indicates that the probabilities of the genes were differentially expressed were >99%. After filtering, 1,273 genes were identified. The gene set enrichment analysis was then conducted using the R package clusterProfiler (17). The enrichment P-value was Benjamini & Hochberg adjusted.

Prognostic biomarker identification for relapse-free survival (RFS)

For the development of prognostic gene signature, the 1,273 B-ALL-associated genes were first fitted with the Cox proportional hazards model in 80 samples. The prognostic significance of each gene test was assessed by log-rank test. We selected 59 at the cut-off P-value ≤0.05. The hierarchal clustering algorithm was then applied to the 59-gene expression profiles and the patients were divided into high- and low-risk groups. The Kaplan Meier-plot and log-rank test were used to test the prognostic significance of the two groups.

Results

In the present study, we sought to identify B-ALL RFS biomarkers using transcriptome data. We hypothesized that the biomarkers may be involved in B-ALL pathogenesis, thus the gene differential expression analysis was conducted by comparing 207 B-ALL samples with 74 healthy normal blood, or bone marrow samples. After applying the linear models and stringent cut-off [Log fold-change ≥2, ≤-2, and false discovery rate (FDR) ≤0.01], a total of 1,273 genes were identified as ALL-associated genes as they were significantly upregulated or downregulated in the ALL samples (Fig. 1).

The Gene Ontology (GO) enrichment analysis revealed that the B-ALL-associated genes were involved in various biological processes (Table I). Surprisingly, we found that the B-ALL-associated genes were most significantly enriched in the biological processes that respond to bacterium (GO:0009617). We further categorized these genes and found that they were involved in cytokine-cytokine receptor interaction pathway (Fig. 2). These genes include chemokine ligands (CCL5, CXCL1, CXCL16, CXCL2, CXCL3, CXCL8), tumor necrosis factor receptor superfamily (FAS, TNFRSF10C, TNFRSF1B, TNFSF8), interleukins (IL-18, CXCL8) and interleukin receptors (IL-10RA, IL-6R). These findings agreed with previous studies on B-ALL pathogenesis. Chemokines and their receptors are vital in many cellular activities such as migrations, targeting developing and mature leukocytes (18). It was previously reported that the CXCR5-CXCL13 axis plays a vital role in chronic lymphocytic leukemia (CLL) (19). There are also other chemokines such as CCR7, CXCR4 and CXCR5 (20), CXCR3 (21), CCL25 (22) and the CXCR4-CXCL12 axis (23) that have been identified as potential targets in leukemia treatment.

Table I.

Gene Ontology enrichment results of ALL-related genes.

Table I.

Gene Ontology enrichment results of ALL-related genes.

Gene OntologyBiological processP-value
GO:0009617Response to bacterium 1.39×10−14
GO:0050900Leukocyte migration 2.31×10−10
GO:0002274Myeloid leukocyte activation 7.56×10−10
GO:0006778 Porphyrin-containing compound metabolic process 7.71×10−10
GO:0042168Heme metabolic process 8.40×10−10
GO:0002237Response to molecule of bacterial origin 2.55×10−9
GO:0006783Heme biosynthetic process 3.53×10−9
GO:0032496Response to lipopolysaccharide 3.53×10−9
GO:0046501Protoporphyrinogen IX metabolic process 3.53×10−9
GO:0042742Defense response to bacterium 4.61×10−9

Gene OntologyCellular componentP-value

GO:0030141Secretory granule 1.95×10−6
GO:0099503Secretory vesicle 5.15×10−5
GO:0098857Membrane microdomain 5.15×10−5
GO:0030667Secretory granule membrane 5.15×10−5
GO:0031091Platelet alpha granule 6.73×10−5
GO:0045121Membrane raft 6.73×10−5
GO:0098589Membrane region 7.00×10−4
GO:0031092Platelet alpha granule membrane 9.60×10−4
GO:0098552Side of membrane 9.60×10−4
GO:0009897External side of plasma membrane 1.30×10−3

Gene OntologyMolecular functionP-value

GO:0030246Carbohydrate binding 3.98×10−5
GO:0008329Signaling pattern recognition receptor activity 2.94×10−4
GO:0038187Pattern recognition receptor activity 2.94×10−4
GO:0017171Serine hydrolase activity 2.94×10−4
GO:0008236Serine-type peptidase activity 4.53×10−4
GO:0042379Chemokine receptor binding 6.71×10−4
GO:0004252Serine-type endopeptidase activity 1.14×10−3
GO:0019865Immunoglobulin binding 1.14×10−3
GO:0004601Peroxidase activity 2.59×10−3
GO:0016684Oxidoreductase activity, acting on peroxide as acceptor 4.99×10−3

[i] Top 10 significantly enriched Gene Ontology (Biological process, Cellular component, Molecular function) terms of the identified B-ALL-associated genes. P-values were Benjamini & Hochberg adjusted. B-ALL, B-precursor acute lymphoblastic leukemia.

Then, we applied Cox proportional hazards modelling to the B-ALL-associated genes and selected 59 top ranked genes based on log-rank test P-values as B-ALL RFS genes (Table II). After applying the hierarchal clustering to this 59-gene profile, the high- (n=20), and low-risk (n=60) samples were identified from the B-ALL cohort with significant (log-rank test P=0.025) different relapse-free survival outcome (Fig. 3). The B-ALL RFS biomarkers included genes from various families, such as glycosyltransferases (C1GALT1 and B4GALT5), RAS oncogene GTPases (RAB27B and RAB7A), nuclear hormone receptors (NR3C1 and RORA), RNA binding proteins (RBM6 and U2SURP) and Zinc finger proteins (PLAGL2, SP1 and ZNF91). We also found that among these biomarkers, 4 genes, B4GALT5, CDK6, PDZD8 and RAB7A, were candidates that were associated with chemotherapy response (Fig. 4) in two independent ALL cohorts, GSE19143 (n=52) (24) and GSE13280 (n=44) (25) where patients were treated with prednisolone. Indeed, the B4GALT family is involved in mediating drug resistance in human leukemia cells by regulating the Hedgehog pathway (26). This suggest that the 59-gene biomarker could also indicate drug sensitivity in ALL treatment.

Table II.

List of 59 prognostic genes for RFS in ALL.

Table II.

List of 59 prognostic genes for RFS in ALL.

Gene symbolGene IDChromosome locationGene nameHR (95% CI)P-value
ADAM987548p11.22ADAM metallopeptidase domain 90.51 (0.27–0.94) 2.83×10−2
AOAH3137p14.2Acyloxyacyl hydrolase2.06 (1.12–3.78) 1.63×10−2
APP35121q21.3Amyloid β precursor protein0.53 (0.34–0.84) 5.49×10−3
B4GALT5933420q13.13 β-1,4-galactosyltransferase 50.43 (0.20–0.95) 4.53×10−2
BSG68219p13.3Basigin (Ok blood group)2.05 (1.05–4.00) 3.37×10−2
C1GALT1569137p22.1-p21.3Core 1 synthase, glycoprotein-N-acetylgalactosamine 3-β-galactosyltransferase 17.40 (1.68–32.52) 7.47×10−3
CDK610217q21.2Cyclin-dependent kinase 63.06 (1.14–8.24) 2.71×10−2
CHPT15699412q23.2Choline phosphotransferase 10.41 (0.24–0.69) 5.35×10−4
CKAP22658613q14.3Cytoskeleton associated protein 20.42 (0.19–0.97) 4.20×10−2
CLC117819q13.2Charcot-Leyden crystal galectin1.52 (1.02–2.29) 3.43×10−2
CLINT196855q33.3Clathrin interactor 10.28 (0.09–0.88) 2.72×10−2
CLU11918p21.1Clusterin12.51 (1.36–115.07) 2.42×10−2
COL17A1130810q25.1Collagen type XVII α 1 chain18.63 (1.35–256.93) 2.69×10−2
CRTAP104913p22.3Cartilage associated protein0.20 (0.05–0.82) 2.71×10−2
CTSA547620q13.12Cathepsin A2.55 (1.22–5.30) 1.33×10−2
DACH1160213q21.33Dachshund family transcription factor 12.04 (1.26–3.31) 1.68×10−3
DNAJB94189 7q31.1;14q24.2-q24.3DnaJ heat shock protein family (Hsp40) member B90.40 (0.18–0.91) 3.24×10−2
DNTT179110q24.1DNA nucleotidylexotransferase0.82 (0.67–0.99) 3.74×10−2
ECRP64333214q11.2Ribonuclease A family member 2 pseudogene2.77 (1.20–6.40) 1.27×10−2
FCGR1B22101p11.2Fc fragment of IgG receptor Ib1.79 (1.18–2.70) 1.96×10−3
GALNT10555685q33.2Polypeptide N-acetylgalactosaminyl transferase 104.01 (1.46–11.02) 6.34×10−3
GPI282119q13.11Glucose-6-phosphate isomerase2.37 (1.03–5.43) 4.16×10−2
JCHAIN35124q13.3Joining chain of multimeric IgA and IgM1.39 (1.13–1.71) 1.10×10−3
KYNU89422q22.2Kynureninase2.33 (1.10–4.93) 1.88×10−2
LPP40263q27.3-q28LIM domain containing preferred translocation partner in lipoma6.42 (1.19–34.48) 2.92×10−2
MICALL2797787p22.3MICAL-like 219.04 (3.85–94.09) 2.70×10−4
MME43113q25.2Membrane metalloendopeptidase0.83 (0.69–1.00) 4.42×10−2
MTMR11109031q21.2Myotubularin related protein 111.65 (1.01–2.70) 3.78×10−2
MZT2B800972q21.1Mitotic spindle organizing protein 2B14.10 (3.05–65.23) 5.59×10−4
NR3C129085q31.3Nuclear receptor subfamily 3 group C member 10.56 (0.36–0.86) 8.11×10−3
NRBF22998210q21.3Nuclear receptor binding factor 20.28 (0.10–0.74) 1.00×10−2
PDZD811898710q25.3-q26.11PDZ domain containing 80.42 (0.21–0.85) 1.49×10−2
PGRMC2104244q28.2Progesterone receptor membrane component 20.37 (0.15–0.92) 3.05×10−2
PIP4K2C7983712q13.3 Phosphatidylinositol-5-phosphate 4-kinase type 2 γ2.36 (1.01–5.48) 4.44×10−2
PLAGL2532620q11.21PLAG1 like zinc finger 20.13 (0.02–0.84) 2.93×10−2
PLD153373q26.31Phospholipase D149.97 (3.91–637.92) 2.28×10−3
PRR115577117q22Proline rich 118.79 (1.49–51.87) 1.56×10−2
PRSS211094216p13.3Protease, serine 211.88 (1.18–2.99) 5.29×10−3
PTAFR57241p35.3Platelet activating factor receptor4.12 (1.20–14.20) 2.46×10−2
QKI94446q26QKI, KH domain containing RNA binding0.50 (0.26–0.98) 4.47×10−2
RAB27B587418q21.2RAB27B, member RAS oncogene family112.03 (3.02–4156.89) 1.05×10−2
RAB7A78793q21.3RAB7A, member RAS oncogene family0.02 (0.00–0.39) 8.24×10−3
RBM6101803p21.31RNA binding motif protein 63.35 (1.08–10.37) 3.49×10−2
RNASE3603714q11.2Ribonuclease A family member 32.49 (1.58–3.92) 1.89×10−6
RORA609515q22.2RAR-related orphan receptor A2.29 (1.12–4.66) 2.06×10−2
RRAGD585286q15Ras-related GTP binding D0.41 (0.23–0.73) 1.96×10−3
S100A1062811q21.3S100 calcium binding protein A101.57 (1.10 to 2.24) 1.18×10−2
SAR1B511285q31.1Secretion associated Ras related GTPase 1B0.42 (0.18 to 0.97) 4.49×10−2
CAVIN284362q32.3Caveolae associated protein 213.41 (1.33 to 135.33) 2.71×10−2
SLAMF7578231q23.3SLAM family member 75.91 (1.01 to 34.70) 4.79×10−2
SLC25A16803410q21.3Solute carrier family 25 member 1693.59 (1.42 to 6174.64) 3.47×10−2
SOCS3902117q25.3Suppressor of cytokine signaling 36.70 (1.35 to 33.25) 2.00×10−2
SORT16272 1p13.3;1p21.3-p13.1Sortilin 15.74 (1.24 to 26.54) 2.28×10−2
SP1666712q13.13Sp1 transcription factor0.10 (0.01 to 0.65) 1.60×10−2
STEAP3552402q14.2STEAP3 metalloreductase3.08 (1.00 to 9.43) 4.95×10−2
TOB21076622q13.2Transducer of ERBB2, 20.38 (0.17 to 0.85) 2.01×10−2
U2SURP233503q23U2 snRNP associated SURP domain containing0.31 (0.13 to 0.77) 1.17×10−2
UBE2D1732110q21.1Ubiquitin conjugating enzyme E2 D10.15 (0.02 to 0.95) 4.14×10−2
ZNF91764419p12Zinc finger protein 910.59 (0.35 to 0.99) 4.80×10−2

[i] Fifty-nine genes associated with B-ALL relapse-free survival. HR, hazard ratio; CI, confidence interval; B-ALL, B-precursor acute lymphoblastic leukemia; RFS, recurrence-free survival.

Discussion

The major challenge in clinical treatments of B-precursor acute lymphoblastic leukemia (B-ALL) is relapse after chemotherapy; thus or the development of alternative treatments is critical. Here, we demonstrated the ability of gene expression profiling to reveal not only biological mechanisms but also clinical diagnostic markers.

Among the 59 genes, several genes have been characterized as being involved in the progression of leukemia and solid tumors. For instance, MME, which is also known as CD10, or CALLA, encodes a type II transmembrane glycoprotein and a common acute lymphocytic leukemia antigen. It is an important cell surface marker in the diagnosis of human acute lymphocytic leukemia (27) has been well-documented in many leukemia-related studies (2832). SOCS3 is a member of the suppressor of cytokine signaling (SOCS) family and negatively regulates JAK2 kinase. Its altered expression is associated with leukemia (33,34) and solid tumors including melanoma (3538), cervical cancer (3941), renal cell carcinoma (4245), prostate cancer (46,47) and gastric cancer (48). CDK6 is a serine/threonine protein that is important for cell cycle G1 phase progression and G1/S transition. CDK6 is dysregulated or disrupted in many types of cancer (4954), and it is previously reported to be in a three-way rearrangement including other two elements: MLL and AF-4 in a case of infant ALL (55). This indicates the biological relevance of the 59-gene biomarker to B-ALL and could propose new directions for investigations.

However, we do realize that there is merely a one gene (JCHAIN known as IGJ) overlap of our 59-gene biomarker with a previously published 38-gene classifier (11). We reasoned that this discordance is due to differences in the analysis strategy. While in the previous study, the biomarker selection was implemented directly on all of the microarray probe sets, we pre-filtered the candidate list to B-ALL-associated genes by differential gene expression analysis. We also optimized the process of preprocessing the microarray data by using up-to-date algorithms and software, which could lead to higher confidence in producing final results.

In conclusion, our systematic approach provided an intriguing guideline for the identification of B-ALL prognostic biomarkers and revealed their potential roles in chemosensitivity. Further investigations are expected to validate the performances of these biomarkers before being applied to clinical management. As the RNA-seq technologies are trending in transcriptome profiling, we will also collect RNA-seq data to re-train and optimize our model. We will also try to reduce the number of biomarkers while maintaining the predictive power, so that the application in clinical management is more feasible.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used during the present study are available from NCBI GEO Database with corresponding accession numbers.

Authors' contributions

WJ and JL conceived and designed the study. WJ collected the data and performed the data analysis. WJ and JL wrote and edited the manuscript. Both authors read and approved the manuscript and agreed to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors do not have any competing interests.

References

1 

Locatelli F, Schrappe M, Bernardo ME and Rutella S: How I treat relapsed childhood acute lymphoblastic leukemia. Blood. 120:2807–2816. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Bhojwani D and Pui CH: Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol. 14:e205–e217. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, Reaman GH and Carroll WL: Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: A report from the children's oncology group. J Clin Oncol. 30:1663–1669. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Zhu YM, Foroni L, McQuaker IG, Papaioannou M, Haynes A and Russell HH: Mechanisms of relapse in acute leukaemia: Involvement of p53 mutated subclones in disease progression in acute lymphoblastic leukaemia. Br J Cancer. 79:1151–1157. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Maloney KW, McGavran L, Odom LF and Hunger SP: Acquisition of p16INK4A and p15INK4B gene abnormalities between initial diagnosis and relapse in children with acute lymphoblastic leukemia. Blood. 93:2380–2385. 1999.PubMed/NCBI

6 

Zuna J: TEL deletion analysis supports a novel view of relapse in childhood acute lymphoblastic leukemia. Clin Cancer Res. 10:5355–5360. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Sewak MS, Reddy NP and Duan ZH: Gene expression based leukemia sub-classification using committee neural networks. Bioinform Biol Insights. 3:89–98. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Harvey RC, Mullighan CG, Wang X, Dobbin KK, Davidson GS, Bedrick EJ, Chen IM, Atlas SR, Kang H, Ar K, et al: Identification of novel cluster groups in pediatric high-risk B-precursor acute lymphoblastic leukemia with gene expression profiling: Correlation with genome-wide DNA copy number alterations, clinical characteristics, and outcome. Blood. 116:4874–4884. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Hogan LE, Meyer JA, Yang J, Wang J, Wong N, Yang W, Condos G, Hunger SP, Raetz E, Saffery R, et al: Integrated genomic analysis of relapsed childhood acute lymphoblastic leukemia reveals therapeutic strategies. Blood. 118:5218–5226. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Staal FJ, van der Burg M, Wessels LF, Barendregt BH, Baert MR, van den Burg CM, van Huffel C, Langerak AW, van der Velden VH, Reinders MJ, et al: DNA microarrays for comparison of gene expression profiles between diagnosis and relapse in precursor-B acute lymphoblastic leukemia: Choice of technique and purification influence the identification of potential diagnostic markers. Leukemia. 17:1324–1332. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Kang H, Chen IM, Wilson CS, Bedrick EJ, Harvey RC, Atlas SR, Devidas M, Mullighan CG, Wang X, Murphy M, et al: Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. Blood. 115:1394–1405. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Kohlmann A, Kipps TJ, Rassenti LZ, Downing JR, Shurtleff SA, Mills KI, Gilkes AF, Hofmann WK, Basso G, Dell'orto MC, et al: An international standardization programme towards the application of gene expression profiling in routine leukaemia diagnostics: The microarray innovations in LEukemia study prephase. Br J Haematol. 142:802–807. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Haferlach T, Kohlmann A, Wieczorek L, Basso G, Kronnie GT, Béné MC, De Vos J, Hernández JM, Hofmann WK, Mills KI, et al: Clinical utility of microarray-based gene expression profiling in the diagnosis and subclassification of leukemia: Report from the international microarray innovations in leukemia Study Group. J Clin Oncol. 28:pp. 2529–2537. 2010, View Article : Google Scholar : PubMed/NCBI

14 

Beesley AH, Cummings AJ, Freitas JR, Hoffmann K, Firth MJ, Ford J, de Klerk NH and Kees UR: The gene expression signature of relapse in paediatric acute lymphoblastic leukaemia: Implications for mechanisms of therapy failure. Br J Haematol. 131:447–456. 2005. View Article : Google Scholar : PubMed/NCBI

15 

McCall MN, Jaffee HA and Irizarry RA: fRMA ST Frozen robust multiarray analysis for Affymetrix Exon and Gene ST arrays. Bioinformatics. 28:3153–3154. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

17 

Yu G, Wang LG, Han Y and He QY: clusterProfiler: An R package for comparing biological themes among gene clusters. Omi A J Integr Biol. 16:284–287. 2012. View Article : Google Scholar

18 

Campbell DJ, Kim CH and Butcher EC: Chemokines in the systemic organization of immunity. Immunol Rev. 195:58–71. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Burkle A, Niedermeier M, Schmitt-Graff A, Wierda WG, Keating MJ and Burger JA: Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia. Blood. 110:3316–3325. 2007. View Article : Google Scholar : PubMed/NCBI

20 

López-Giral S, Quintana NE, Cabrerizo M, Alfonso-Pérez M, Sala-Valdés M, De Soria VG, Fernández-Rañada JM, Fernández-Ruiz E and Muñoz C: Chemokine receptors that mediate B cell homing to secondary lymphoid tissues are highly expressed in B cell chronic lymphocytic leukemia and non-Hodgkin lymphomas with widespread nodular dissemination. J Leukoc Biol. 76:462–471. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Jones D, Benjamin RJ, Shahsafaei A and Dorfman DM: The chemokine receptor CXCR3 is expressed in a subset of B-cell lymphomas and is a marker of B-cell chronic lymphocytic leukemia. Blood. 95:627–32. 2000.PubMed/NCBI

22 

Qiuping Z, Jei X, Youxin J, Wei J, Chun L, Jin W, Qun W, Yan L, Chunsong H, Mingzhen Y, et al: CC chemokine ligand 25 enhances resistance to apoptosis in CD4+ T cells from patients with T-cell lineage acute and chronic lymphocytic leukemia by means of livin activation. Cancer Res. 64:7579–7587. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Burger JA and Peled A: CXCR4 antagonists: Targeting the microenvironment in leukemia and other cancers. Leukemia. 23:43–52. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Stam RW, Den Boer ML, Schneider P, de Boer J, Hagelstein J, Valsecchi MG, de Lorenzo P, Sallan SE, Brady HJ, Armstrong SA, et al: Association of high-level MCL-1 expression with in vitro and in vivo prednisone resistance in MLL-rearranged infant acute lymphoblastic leukemia. Blood. 115:1018–1125. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Spijkers-Hagelstein JA, Schneider P, Hulleman E, de Boer J, Williams O, Pieters R and Stam RW: Elevated S100A8/S100A9 expression causes glucocorticoid resistance in MLL-rearranged infant acute lymphoblastic leukemia. Leukemia. 26:1255–1165. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Zhou H, Ma H, Wei W, Ji D, Song X, Sun J, Zhang J and Jia L: B4GALT family mediates the multidrug resistance of human leukemia cells by regulating the hedgehog pathway and the expression of p-glycoprotein and multidrug resistance-associated protein 1. Cell Death Dis. 4:e6542013. View Article : Google Scholar : PubMed/NCBI

27 

Epstein J, Xiao HQ and He XY: Markers of multiple hematopoietic-cell lineages in multiple myeloma. N Engl J Med. 322:664–668. 1990. View Article : Google Scholar : PubMed/NCBI

28 

Dunphy CH, Gardner LJ, Evans HL and Javadi N: CD15+ acute lymphoblastic leukemia and subsequent monoblastic leukemia: Persistence of t(4;11) abnormality and B-cell gene rearrangement. Arch Pathol Lab Med. 125:1227–1230. 2001.PubMed/NCBI

29 

Rozovskaia T, Feinstein E, Mor O, Foa R, Blechman J, Nakamura T, Croce CM, Cimino G and Canaani E: Upregulation of Meis1 and HoxA9 in acute lymphocytic leukemias with the t(4:11) abnormality. Oncogene. 20:874–878. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Burmeister T, Meyer C, Schwartz S, Hofmann J, Molkentin M, Kowarz E, Schneider B, Raff T, Reinhardt R, Gökbuget N, et al: The MLL recombinome of adult CD10-negative B-cell precursor acute lymphoblastic leukemia: Results from the GMALL study group. Blood. 113:4011–4015. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Gleissner B, Goekbuget N, Rieder H, Arnold R, Schwartz S, Diedrich H, Schoch C, Heinze B, Fonatsch C, Bartram CR, et al: CD10 pre-B acute lymphoblastic leukemia (ALL) is a distinct high-risk subgroup of adult ALL associated with a high frequency of MLL aberrations: Results of the german multicenter trials for adult ALL (GMALL). Blood. 106:4054–4056. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Ha K, Hozumi N, Hrincu A and Gelfand EW: Lineage specific classification of leukaemia: Results of the analysis of sixty cases of childhood leukaemia. Br J Haematol. 61:237–249. 1985. View Article : Google Scholar : PubMed/NCBI

33 

Suzuki R, Sakamoto H, Yasukawa H, Masuhara M, Wakioka T, Sasaki A, Yuge K, Komiya S, Inoue A and Yoshimura A: CI S3 and JAB have different regulatory roles in interleukin-6 mediated differentiation and STAT3 activation in M1 leukemia cells. Oncogene. 17:2271–2278. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Chen C, Yu K, Yan QX, Xing CY, Chen Y, Yan Z, Shi YF, Zhao KW and Gao SM: Pure curcumin increases the expression of SOCS1 and SOCS3 in myeloproliferative neoplasms through suppressing class I histone deacetylases. Carcinogenesis. 34:1442–1449. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Komyod W, Böhm M, Metze D, Heinrich PC and Behrmann I: Constitutive suppressor of cytokine signaling 3 expression confers a growth advantage to a human melanoma cell line. Mol Cancer Res. 5:271–281. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Lesinski GB, Zimmerer JM, Kreiner M, Trefry J, Bill MA, Young GS, Becknell B and Carson WE III: Modulation of SOCS protein expression influences the interferon responsiveness of human melanoma cells. BMC Cancer. 10:1422010. View Article : Google Scholar : PubMed/NCBI

37 

Tokita T, Maesawa C, Kimura T, Kotani K, Takahashi K, Akasaka T and Masuda T: Methylation status of the SOCS3 gene in human malignant melanomas. Int J Oncol. 30:689–694. 2007.PubMed/NCBI

38 

Fojtova M, Boudny V, Kovarik A, Lauerova L, Adamkova L, Souckova K, Jarkovsky J and Kovarik J: Development of IFN-gamma resistance is associated with attenuation of SOCS genes induction and constitutive expression of SOCS 3 in melanoma cells. Br J Cancer. 97:231–237. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Shivapurkar N, Sherman ME, Stastny V, Echebiri C, Rader JS, Nayar R, Bonfiglio TA, Gazdar AF and Wang SS: Evaluation of candidate methylation markers to detect cervical neoplasia. Gynecol Oncol. 107:549–553. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Zhang P, Yang B, Yao YY, Zhong LX, Chen XY, Kong QY, Wu ML, Li C, Li H and Liu J: PIA S3SH P2 and SOCS3 Expression patterns in Cervical Cancers: Relevance with activation and resveratrol-caused inactivation of STAT3 signaling. Gynecol Oncol. 139:529–535. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Kim MH, Kim MS, Kim W, Kang MA, Cacalano NA, Kang SB, Shin YJ and Jeong JH: Suppressor of cytokine signaling (SOCS) genes are silenced by DNA hypermethylation and histone deacetylation and regulate response to radiotherapy in cervical cancer cells. PLoS One. 10:e01231332015. View Article : Google Scholar : PubMed/NCBI

42 

Urbschat A, Stumpf S, Hänze J, Paulus P, Maier TJ, Weipert C, Hofmann R and Hegele A: Expression of the anti-inflammatory suppressor of cytokine signaling 3 (SOCS3) in human clear cell renal cell carcinoma. Tumour Biol. 37:9649–9656. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Stofas A, Levidou G, Piperi C, Adamopoulos C, Dalagiorgou G, Bamias A, Karadimou A, Lainakis GA, Papadoukakis S, Stravodimos K, et al: The role of CXC-chemokine receptor CXCR2 and suppressor of cytokine signaling-3 (SOCS-3) in renal cell carcinoma. BMC Cancer. 14:1492014. View Article : Google Scholar : PubMed/NCBI

44 

Tomita S, Ishibashi K, Hashimoto K, Sugino T, Yanagida T, Kushida N, Shishido K, Aikawa K, Sato Y, Suzutani T, et al: Suppression of SOCS3 increases susceptibility of renal cell carcinoma to interferon-α. Cancer Sci. 102:57–63. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Oguro T, Ishibashi K, Sugino T, Hashimoto K, Tomita S, Takahashi N, Yanagida T, Haga N, Aikawa K, Suzutani T, et al: Humanised antihuman IL-6R antibody with interferon inhibits renal cell carcinoma cell growth in vitro and in vivo through suppressed SOCS3 expression. Eur J Cancer. 49:1715–1724. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Neuwirt H, Puhr M, Cavarretta IT, Mitterberger M, Hobisch A and Culig Z: Suppressor of cytokine signalling-3 is up-regulated by androgen in prostate cancer cell lines and inhibits androgen-mediated proliferation and secretion. Endocr Relat Cancer. 14:1007–1019. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Kneitz B, Krebs M, Kalogirou C, Schubert M, Joniau S, van Poppel H, Lerut E, Kneitz S, Scholz CJ, Ströbel P, et al: Survival in patients with high-risk prostate cancer is predicted by miR-221, which regulates proliferation, apoptosis, and invasion of prostate cancer cells by inhibiting IRF2 and SOCS3. Cancer Res. 74:2591–2603. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Li G, Xu J, Wang Z, Yuan Y, Li Y, Cai S and He Y: Low expression of SOCS-1 and SOCS-3 is a poor prognostic indicator for gastric cancer patients. J Cancer Res Clin Oncol. 141:443–452. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Lien HC, Lin CW, Huang PH, Chang ML and Hsu SM: Expression of cyclin-dependent kinase 6 (cdk6) and frequent loss of CD44 in nasal-nasopharyngeal NK/T-cell lymphomas: Comparison with CD56-negative peripheral T-cell lymphomas. Lab Invest. 80:893–900. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Lee YH, Judge AD, Seo D, Kitade M, Gómez-Quiroz LE, Ishikawa T, Andersen JB, Kim BK, Marquardt JU, Raggi C, et al: Molecular targeting of CSN5 in human hepatocellular carcinoma: A mechanism of therapeutic response. Oncogene. 30:4175–4184. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Ouyang Q, Chen G, Zhou J, Li L, Dong Z, Yang R, Xu L, Cui H, Xu M and Yi L: Neurotensin signaling stimulates glioblastoma cell proliferation by upregulating c-Myc and inhibiting miR-29b-1 and miR-129-3p. Neuro Oncol. 18:216–226. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Easton J, Wei T, Lahti JM and Kidd VJ: Disruption of the cyclin D/cyclin-dependent kinase/INK4/retinoblastoma protein regulatory pathway in human neuroblastoma. Cancer Res. 58:2624–2632. 1998.PubMed/NCBI

53 

Brandi G, Paiardini M, Cervasi B, Fiorucci C, Filippone P, De Marco C, Zaffaroni N and Magnani M: A new indole-3- carbinol tetrameric derivative inhibits cyclin-dependent kinase 6 expression, and induces G1 cell cycle arrest in both estrogen-dependent and estrogen-independent breast cancer cell lines. Cancer Res. 63:4028–4036. 2003.PubMed/NCBI

54 

Ye Y, Yang H, Grossman HB, Dinney C, Wu X and Gu J: Genetic variants in cell cycle control pathway confer susceptibility to bladder cancer. Cancer. 112:2467–2474. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Raffini LJ, Slater DJ, Rappaport EF, Lo Nigro L, Cheung NK, Biegel JA, Nowell PC, Lange BJ and Felix CA: Panhandle and reverse-panhandle PCR enable cloning of der(11) and der(other) genomic breakpoint junctions of MLL translocations and identify complex translocation of MLL, AF-4, and CDK6. Proc Natl Acad Sci USA. 99:4568–4573. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jing W and Jing W: Identification of biomarkers for the prediction of relapse‑free survival in pediatric B‑precursor acute lymphoblastic leukemia. Oncol Rep 41: 659-667, 2019
APA
Jing, W., & Jing, W. (2019). Identification of biomarkers for the prediction of relapse‑free survival in pediatric B‑precursor acute lymphoblastic leukemia. Oncology Reports, 41, 659-667. https://doi.org/10.3892/or.2018.6846
MLA
Jing, W., Li, J."Identification of biomarkers for the prediction of relapse‑free survival in pediatric B‑precursor acute lymphoblastic leukemia". Oncology Reports 41.1 (2019): 659-667.
Chicago
Jing, W., Li, J."Identification of biomarkers for the prediction of relapse‑free survival in pediatric B‑precursor acute lymphoblastic leukemia". Oncology Reports 41, no. 1 (2019): 659-667. https://doi.org/10.3892/or.2018.6846