Radiol Oncol 2022; 56(1): 92-101. doi: 10.2478/raon-2021-0050 92 research article Clinical impacts of copy number variations in B-cell differentiation and cell cycle control genes in pediatric B-cell acute lymphoblastic leukemia: a single centre experience Klementina Crepinsek1,2, Gasper Marinsek1, Marko Kavcic2,3, Tomaž Prelog3, Lidija Kitanovski3, Janez Jazbec2,3, Marusa Debeljak1,2 1 Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia 2 Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia 3 Department of Oncology and Haematology, University Children’s Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia Radiol Oncol 2022; 56(1): 92-101. Received 7 September 2021 Accepted 5 November 2021 Correspondence to: Assist. Prof. Maruša Debeljak, Ph.D., Clinical Institute for Special Laboratory Diagnostics, University Children’s Hospital, University Medical Centre Ljubljana, Vrazov trg 1, 1000 Ljubljana, Slovenia. E-mail: marusa.debeljak@kclj.si Disclosure: No potential conflicts of interest were disclosed. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). Background. IKZF1 gene deletions have been identified as a poor prognostic factor in pediatric B-cell acute lympho- blastic leukemia (B-ALL), especially in the presence of co-occurring deletions (IKZF1plus profile). This study aimed to determine the frequency of IKZF1 deletions and deletions in other B-cell differentiation and cell cycle control genes, and their prognostic impact in Slovenian pediatric B-ALL patients. Patients and methods. We studied a cohort of 99 patients diagnosed with B-ALL from January 2012 to December 2020 and treated according to the ALL IC-BFM 2009 protocol. Eighty-eight bone marrow or peripheral blood samples were analysed for copy number variations (CNVs) using the SALSA MLPA P335 ALL-IKZF1 probemix. Results. At least one CNV was detected in more than 65% of analysed samples. The most frequently altered genes were PAX5 and CDKN2A/B (30.7%, 26.1%, and 25.0%, respectively). Deletions in IKZF1 were present in 18.2% of analysed samples and were associated with an inferior 5-year event-free survival (EFS; 54.8% vs. 85.9%, p = 0.016). The IKZF1plus profile was identified in 12.5% of the analysed samples, and these patients had an inferior 5-year EFS than those with deletions in IKZF1 only and those without deletions (50.8% vs. 75.0% vs. 85.9%, respectively, p = 0.049). Overall survival (OS) was also worse in patients with the IKZF1plus profile than those with deletions in IKZF1 only and those without dele- tions (5-year OS 76.2% vs. 100% vs. 93.0%, respectively). However, the difference between the groups was not statisti- cally significant. Conclusions. Our results are in concordance with the results obtained in larger cooperative clinical trials. Copy num- ber variations analysis using the SALSA MLPA kit is a reliable tool for initial diagnostic approach in children with B-ALL, even in smaller institutions in low- and middle-income countries. Key words: B-acute lymphoblastic leukemia; IKZF1 deletions; IKZF1plus; MLPA; pediatric; copy number variations (CNVs) Introduction Improvements in risk stratification and new thera- peutic approaches have dramatically improved treatment outcomes in pediatric B-cell acute lymphoblastic leukemia (B-ALL). In developed countries, the overall survival for these patients is approaching 90%.1,2 Nevertheless, some genetic Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL 93 subtypes still imply poor outcomes, and 10–20% of patients experience a relapse that is often ac- companied by treatment resistance and failure.3,4 Therefore, the need for new diagnostic and prog- nostic markers remains of paramount importance. In the previous years, deletions in the IKZF1 gene have been identified as an important predic- tor of relapse in B-ALL.5-9 IKZF1 gene is located on chromosome 7p12.2 and consists of 8 exons, and of those, exons 2–8 are protein-coding. The gene codes for the transcription factor IKAROS, which regulates the expression of genes that control cell cycle progression and cell survival. It is involved in the development of all lymphoid lineages, especial- ly in the differentiation of B-progenitor cells. Exons 4–6 encode four zinc finger DNA-binding domains that are essential for the tumour-suppressive func- tion of IKAROS, and exon 8 encodes two zinc fin- gers that are responsible for the homo- or heter- odimerization of IKAROS.10,11 Genomic deletions in IKZF1 occur in around 15% of pediatric B-ALL cases.5,6,8,12-14 Their occurrence is exceptionally high in BCR-ABL1-positive (70%)15,16 and BCR-ABL1-like (40%)13,17 B-ALL, and have been associated with poor treatment response and an increased risk of relapse.5–9 Deletions are most common in exons 4–7 or affect the whole gene, however, other less com- mon lesions may also be present (e. g. deletions in exons 2–8, 2–7), and they are all associated with an unfavourable outcome in pediatric B-ALL.6,18 Therefore, some study groups on B-ALL treatment have decided to include IKZF1 deletion status into their risk stratification protocols. Others, however, did not, as there was hesitation on whether the prognostic impact of these deletions was strong enough to justify treatment intensification.14,19,20 Recently, another, minimal residual disease (MRD) dependent prognostic profile IKZF1plus with an immensely poor prognostic value was identified. This profile is defined by additional de- letions in genes involved in cell differentiation and cell cycle regulation. IKZF1 deletions that co-occur with deletions in CDKN2A, CDKN2B, PAX5, or the PAR1 region (deletions of CSF2RA and IL3RA, but not CRLF2) in the absence of ERG deletions are as- sociated with the worst event-free and overall sur- vival.21 This profile is already being used in the cur- rent AIEOP-BFM ALL 2017 trial as a high-risk cri- terion.20 Copy number variations (CNVs) in some aforementioned genes (PAX5, CDKN2A, CDKN2B) also seem to be independently associated with poor prognosis, however, the results remain con- flicting.19,22-25 The inclusion of the CNV status of these genes may significantly improve risk strati- fication in B-ALL, but more studies are needed to elucidate their true prognostic effect. Deletions in IKZF1 are mostly observed in high- risk pediatric ALL subtypes. Many studies have confirmed the association of IKZF1 deletions and IKZF1plus profile with poor treatment outcomes. In Slovenia, no studies have been done yet to deter- mine the frequency of IKZF1 deletions and CNVs in other cell differentiation and cell cycle regula- tion genes in pediatric B-ALL patients and treat- ment outcomes for these patients. Due to the im- portance of these alterations in the prognosis and choosing the best treatment approach, it is of great importance to determine their presence. Therefore, the study aimed to analyze bone marrow samples from Slovenian pediatric patients diagnosed with B-ALL from January 2012 to December 2020 for the presence of these CNVs and to determine their prognostic value. Patients and methods Patients and samples In total, 99 children with B-ALL that were treated at the University Children’s Hospital, University Medical Centre Ljubljana between January 2012 and December 2020 according to the ALL IC-BFM 2009 protocol were included in this study. Diagnoses were established following standard clinical, cyto- morphological, and immunological criteria. We obtained bone marrow samples for 92 pa- tients as part of the diagnostic procedure before starting treatment. For 7 patients, bone marrow samples were not available, therefore, peripheral blood samples were obtained for the analysis. For four patients, there was no sufficient material avail- able to perform the multiplex ligation-dependent probe amplification (MLPA) assay, 3 samples con- tained less than 40% of blasts and were excluded from analysis, and for an additional four, the assay failed due to poor sample quality. Therefore, data analysis was performed on 88 patient samples (82 bone marrow and 6 peripheral blood). The bone marrow samples contained 77.4 ± 16.7% of blast in average, and for the peripheral blood samples this value was 77.8 ± 16.0%. For survival analysis, patient samples from the year 2020 were excluded, due to the short follow-up period. Therefore, the survival analysis was carried out on 72 patients diagnosed between January 2012 and December 2019. Informed consent was obtained from all sub- jects involved in the study, or their parents. The Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL94 study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics Committee of the Republic of Slovenia (ref- erence number KME 51/03/11). DNA extraction Genomic DNA was extracted from bone marrow or peripheral blood samples using the FlexiGene DNA Kit 250 (QIAGEN®, Hilden, Germany) according to the manufacturer’s instructions. All samples were quantified using DS-11 FX+ Spectrophotometer (DeNovix, Wilmington, USA), and stored at 4°C. Before analysis, the DNA con- centration was established at 25 ± 1 ng/μL for the MLPA assay. Analysis of copy number alterations DNA was analysed for copy number alterations us- ing the SALSA MLPA P335 ALL-IKZF1 probemix, according to the manufacturer’s instructions (MRC Holland, Amsterdam, the Netherlands). The P335 probemix allows for the detection of deletions and duplications in B-cell differentiation and cell cycle control genes (IKZF1, CDKN2A/B, PAX5, EBF1, ETV6, BTG1, and RB1), as well as in genes from the X/Y PAR1 region (CRLF2, CSF2RA, SHOX, IL3RA, and P2RY8). It also contains 13 reference probes that function as internal controls. Additionally, analysis for copy number alterations for the deter- mination of ERG status was carried out on samples that carried deletions in IKZF1 and at least one ad- ditional gene (namely CDKN2A, CDKN2B, PAX5, CSF2RA, and IL3RA) with the SALSA MLPA P327 iAMP21-ERG probemix. The P327 probemix is used for the detection of deletions, duplications or amplifications of specific sequences on chromo- some 21, including intragenic deletions of ERG. It contains 59 MLPA probes that bind to several regions on chromosome 21, including the ERG gene, and 13 reference probes. DNA samples from healthy donors were used as controls. MLPA reactions were carried out on a 96-well PCR thermocycler SimpliAmp Thermal Cycler (Applied Biosystems, Thermo Fisher, Massachusetts, USA), and the products were sepa- rated by capillary electrophoresis on an ABI-3500 genetic analyser (Applied Biosystems, Thermo Fisher, Massachusetts, USA). The resulting peak in- tensities were analysed using Coffalyser software (MRC-Holland) which performed the intrasample and intersample normalization of the peaks with the manufacturer’s reference probes and normal control DNA, respectively. Values above 1.3 were considered as gain, between 1.3 and 0.75 normal, between 0.75 and 0.25 heterozygous loss, and be- low 0.25 homozygous loss. Statistical analysis All statistical analyses were performed using SPSS 22.0 software (IBM Corp., Armonk, NY, USA). TABLE 1. The demographic and clinical characteristics of Slovenian B-ALL patients included in the study Characteristic Nr. of patients 99 Sex Male 54 (54.5%) Female 45 (45.5%) Primary genetic abnormalities ETV6-RUNX1 28 (28.3%) BCR-ABL1 7 (7.1%) KMT2A rearrangements 4 (4.0%) TCF3-PBX1 4 (4.0%) Hyperdiploidy 27 (27.3%) Hypodiploidy 3 (3.0%) iAMP21 2 (2.0%) No recurrent abnormalities 24 (24.2%) Age at diagnosis < 1 3 (3.0%) 1–5 57 (57.6%) ≥ 6 39 (39.4%) Risk group Standard risk 17 (17.2%) Intermediate risk 59 (59.6%) High risk 23 (23.2%) FC- minimal residual disease Day 15 < 0.1% 35 (35.4%) 0.1–10% 48 (48.5%) > 10% 13 (13.1%) Unknown 3 (3.0%) Day 33 < 0.01% 73 (73.7%) 0.01–1% 20 (20.2%) > 1% 3 (3.0%) Unknown 3 (3.0%) Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL 95 carried additional deletions in CDKN2A, CDKN2B and PAX5, three in CDKN2A and CDKN2B, one in only CDKN2B, and one had deletions in the PAR1 region. ERG deletions were not found in any of these 11 samples. The comparison of patient characteristics de- pending on IKZF1 deletion is summarized in FIGURE 2. Primary genetic alterations in patients with IKZF1 deletions. FIGURE 1. Prevalence of ALL subtypes in the Slovenian pediatric B-ALL cohort. FIGURE 3. The number of CNVs present in Slovenian B-ALL samples. Event-free survival (EFS; defined as the time be- tween diagnosis and relapse or death) and overall survival (OS; defined as time between diagnosis and death or last follow-up) were analysed using the Kaplan-Meier method and the differences be- tween multiple groups were analysed using the log- rank test. Multivariate analysis was performed us- ing a Cox regression model, which was adjusted for other risk factors, namely sex, age at diagnosis, and risk group (HR vs. non-HR). Comparisons of cat- egorical values were carried out using the Fischer’s exact test, and for the comparison of numerical val- ues, the Mann–Whitney U-test was used. The sig- nificance level for all the tests was 5% (p < 0.05 was considered to be statistically significant). Results Study group The cohort included 54 males and 45 females (N = 99), with median age 4 years (range from 1 day to 23 years). Of these, 75 harboured recurrent ge- netic abnormalities (28 ETV6-RUNX1, 27 hyperdip- loid karyotype, 7 BCR-ABL1, 4 KMT2A rearrange- ments, 4 TCF3-PBX1, 3 hypodiploid karyotype and 2 iAMP21), while no genetic alterations were identified in 24 patients (Figure 1). Based on age, white blood cell (WBC) count at diagnosis, blast cell counts on day 8, genetic abnormalities, and MRD at days 15 and 33, 17 patients were classified as standard risk (SR), 59 as intermediate risk (IR), and 23 as high risk (HR). The main patient charac- teristics are summed up in Table 1. Detection and analysis of IKZF1 deletions by MLPA Altogether, 92 patient samples and 5 controls were analysed by MLPA using the SALSA MLPA P335 ALL-IKZF1 probemix. Four patient samples failed the MLPA analysis. Out of the remaining 88 sam- ples, IKZF1 deletions were found in 16 (18.2%). The most common was the deletion of the whole gene, which was observed in eight patients (50%), oth- ers were focal deletions. The second most common deletion was the deletion of exons 4–8, which was found in four patients (25%). This deletion was de- scribed as rare in other studies. Other deletions that were also detected in our cohort were the deletion of exons 2–8 (two patients; 12.5%), 4–7 and 5 (both found in one patient; 6.3%). Eleven patients with IKZF1 deletion had additional deletions present, which put them in the IKZF1plus group. Of these, six Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL96 Table 2. Of the 16 patients that harboured IKZF1 deletions, five patients had no recurrent genetic alterations. The presence of primary recurrent ge- netic abnormalities found in patients with IKZF1 deletions is shown in Figure 2. IKZF1 deletions were significantly more common in Ph+ patients than in Ph– patients (6/7, 85.7% vs. 10/81, 12.3%, p TABLE 2. Patients’ characteristics and response to treatment according to IKZF1 deletion status in 91 Slovenian pediatric B-ALL patients Characteristic IKZF1 status No IKZF1 deletion IKZF1 deletion only IKZF1 plus Nr. of patients 72 5 11 Sex Male 34 (47.2%) 5 (100%) 9 (81.8%) Female 38 (52.8%) 0 (0.0%) 2 (18.2%) Primary genetic abnormalities ETV6-RUNX1 24 (33.3%) 0 (0.0%) 1 (9.1%) BCR-ABL1 1 (1.4%) 2 (40.0%) 4 (36.4%) KMT2A rearrangements 4 (5.6%) 0 (0.0%) 0 (0.0%) TCF3-PBX1 3 (4.2%) 0 (0.0%) 1 (9.1%) Hyperdiploidy 20 (27.8%) 2 (40.0%) 1 (9.1%) Hypodiploidy 2 (2.8%) 0 (0.0%) 1 (9.1%) iAMP21 1 (1.4%) 0 (0.0%) 0 (0.0%) No recurrent abnormalities 18 (25.0%) 1 (20.0%) 3 (27.3%) Age at diagnosis < 1 3 (4.2%) 0 (0.0%) 0 (0.0%) 1–5 41 (56.9%) 2 (40.0%) 5 (45.5%) ≥ 6 28 (38.9%) 3 (60.0%) 6 (54.5%) Risk group Standard risk 13 (18.1%) 0 (0.0%) 0 (0.0%) Intermediate risk 46 (63.9%) 2 (40.0%) 4 (36.4%) High risk 13 (18.1%) 3 (60.0%) 7 (63.6%) FC- minimal residual disease Day 15 < 0.1% 30 (41.7%) 0 (0.0%) 1 (9.1%) 0.1–10% 33 (45.8%) 2 (40.0%) 6 (54.5%) > 10% 6 (8.3%) 3 (60.0%) 4 (36.4%) Unknown 3 (4.2%) 0 (0.0%) 0 (0.0%) Day 33 < 0.01% 56 (77.8%) 0 (0.0%) 8 (72.7%) 0.01–1% 13 (18.1%) 3 (60.0%) 2 (18.2%) > 1% 2 (2.8%) 1 (20.0%) 0 (0.0%) Unknown 1 (1.4%) 1 (20.0%) 1 (9.1%) = 0.0001), and so was the presence of the IKZF1plus profile (4/7, 57.1% vs. 7/81, 8.6%, p = 0.004). The IKZF1 deletions were significantly more common in males than in females (p = 0.0045), how- ever, the presence of the IKZF1plus profile did not significantly differ between the sexes (p = 0.0607). Patients with IKZF1 deletions were also older at diagnosis than those without deletions (median age 6 years, interquartile range (IQR) = 8 years vs. median age 4 years, IQR = 3.25 years, p = 0.052), and so were the patients with IKZF1plus profile in comparison to those who did not have this profile (median age 6 years, IQR = 8 years vs. median age 4 years, IQR = 4, p = 0.136), however the differenc- es were not statistically significant. Patients with IKZF1 deletions showed higher blast count values on the 8th day of chemotherapy treatment (medi- an blast count 252 blasts/μL, IQR = 2018.5 blasts/ μL vs. median blast cell count 17 blasts/μL, IQR = 192,5 blasts/μL, p = 0.0005), higher values of MRD on day 15 (median MRD15 6.15% IQR = 30.67% vs. median MRD15 0.184%, IQR = 1.24%, p = 0.0005), and 33 of treatment (median MRD33 0.001%, IQR = 0.15% vs. median MRD33 0.000%, IQR = 0.01%, p = 0.033) compared to those without deletions. Similarly, blast cell count and MRD15 values of patients with the IKZF1plus profile were higher compared to patients without the profile (median blast cell counts 224 blasts/μL, IQR = 2112 blasts/ μL vs. median blast cell count 24 blasts/μL, IQR = 220 blasts/μL, p = 0.006; median MRD15 3.8%, IQR = 17.6% vs. median MRD15 0.24%, IQR = 2.23%, p = 0.030), while the difference was not significant for MRD33. The deletions were also more common in the HR group than in the IR (10/23, 43.5% vs. 6/52, 11.5%, p = 0.0032) and the SR group (10/23, 43.5% vs. 0/13, 0%, p = 0.0045). Patients in the HR group also exhibited the IKZF1plus profile more of- ten than those in the IR (7/23, 30.4% vs. 4/52, 7.7%, p = 0.0160) and the SR group (7/23, 30.4% vs. 0/13, 0%, p = 0.0294). In our cohort, 13 patients (14.8%) experienced an event (either relapse or death). Among these, four patients had no recurrent genetic alterations, three patients carried the ETV6-RUNX1 fusion gene, two the BCR-ABL1 fusion gene, two were hyper- diploid, one had a KMT2A rearrangement and one carried the TCF3-PBX1 fusion gene. One patient was classified as SR, eight as IR and four as HR. In this group, five patients (5/13, 38.5%) carried an IKZF1 deletion, and of those, four had additional deletions, but lacked the ERG deletions, which met the criteria for the IKZF1plus profile. The presence of IKZF1 deletions and the IKZF1plus profile was Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL 97 higher in the group of patients who experienced an event in comparison to those who did not, however the difference did not reach statistical significance (5/13 vs. 11/75, p = 0.055 and 4/13 vs. 7/75, p = 0.053, respectively). Detection and analysis of other gene deletions and duplications by MLPA The SALSA MLPA P335 ALL-IKZF1 probemix can detect deletions or duplications in the following B-cell differentiation and cell cycle control genes: IKZF1, EBF1, CDKN2A/B, PAX5, ETV6, BTG1, RB1, and in the PAR1 region (SHOX area, CRLF2, CSF2RA, IL3RA and P2RY8 genes). At least one CNV was detected in 60 patient samples (68.2%). Of those, 60% carried three or more CNVs, 28.3% carried only one CNV, and 11.7% carried two CNVs (Figure 3). The most common CNVs were those in the PAX5 gene that were present in 30.7% of analysed samples, followed by CDKN2A and CDKN2B that were altered in 26.1% and 25.0% of analysed sam- ples, respectively. In these genes, deletions were more common than amplifications. CNVs were also very common in the PAR1 region, 22.7% of analysed samples had at least one CNV present in this region, and in these genes, amplifications were observed more often than deletions. Detailed information about CNVs in all analysed genes are shown in Figure 4. Prognostic significance of IKZF1 deletions First, the patients were divided into two groups, a group with and a group without IKZF1 deletions. The 5-year EFS was significantly worse for patients harbouring IKZF1 deletions, compared to those without the deletions (54.8% vs. 85.9%, p = 0.016) (Figure 5A). The 5-year OS was slightly worse as well for these patients, although the difference was not statistically significant (81.5% vs. 93.0%, p = 0.295) (Figure 5B). The 5-year EFS and OS were also compared be- tween groups with no IKZF1 deletions, with dele- tions in the IKZF1 gene only and with the IKZF1plus profile. The difference in EFS between groups was statistically significant (p = 0.049). Pairwise com- parison showed that patients in group IKZF1plus had significantly poorer EFS in comparison to those in group with no IKZF1 deletions (5-year EFS 50.8% vs. 85.9%, p = 0.016), while the difference was not significant between other groups (Figure 6A). The OS analysis between groups showed no differ- ences between the groups (Figure 6B). A multivariate Cox regression model was ap- plied to this data to see, whether after adjusting for other relevant risk factors, IKZF1 deletions profile retained a prognostic impact on event-free sur- vival. We included the following variables in the model: sex, age at diagnosis, risk group (HR vs. nonHR) and the presence of IKZF1 deletions. The overall model showed borderline significance (p = 0.05). When each separate variable was inspected, it was observed that both sex and the presence of the IKZF1 deletions showed a certain trend (males having poorer survival than females (HR = 1.44), and those with the IKZF1 deletions having poorer survival than those without the deletions (HR = 1.15)), however, they did not reach significance (p = 0.072 and p = 0.078, respectively). Similar results were obtained when this analysis was applied for the IKZF1plus profile. This can most likely be attrib- uted to the small sample size of our sample for sur- vival analysis. FIGURE 4. Frequency of copy number variations: (A) Gene deletions in the cohort. (B) Gene amplifications in the cohort. BTG1 = BTG anti-proliferation factor 1; CDKN2A/2B = cyclin dependent kinase inhibitor 2A/2B; CRLF2 = cytokine receptor-like factor 2; CSF2RA = colony-stimulating factor 2 receptor α subunit; EBF1 = early B-cell factor 1; ETV6 = ETS variant 6; IKZF1 = IKAROS family zinc finger 1; IL3RA = interleukin 3 receptor subunit α; JAK2 = Janus kinase 2; PAX5 = paired box 5; P2RY8 = purinergic receptor P2Y8; SHOX = short-stature homeobox gene; RB1 = RB transcriptional corepressor 1 A B Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL98 We further looked at the group of patients, clas- sified as non-high risk (either SR or IR). In this group, six patients carried the IKZF1 deletions, and of those, two patients experienced an event (2/6, 33.3%), and among the patients without the deletions, seven experienced an event (7/59, 11.9%) (p = 0.1907). The 5-year EFS for those without the deletions was 83.6%, while it was only 50% for those with the deletion. Once again, there is a cer- tain trend to be seen, however, the difference was not statistically significant (p = 0.114). We also examined patients with the IKZF1plus profile in the non-HR group. The frequency of events was higher amongst the patients with the profile com- pared to those without it (2/4, 50% vs. 7/61, 11.5%, p = 0.0890), and their EFS was poorer, although not significantly (50.0% vs. 83.7%, p = 0.087). FIGURE 5. (A) Event-free survival in patients with or without IKZF1 deletions (5-year event-free survival [EFS] 54.8% vs. 85.9%, p = 0.016). (B) Overall survival in patients with or without IKZF1 deletions (5-year overall survival [OS] 81.5% vs. 93.0%, p = 0.295). FIGURE 6. (A) Event-free survival in patients without IKZF1 deletions, with IKZF1 deletions only, and those with the IKZF1plus profile (5-year EFS 85.9% vs. 75.0% vs. 50.8%, p = 0.049). (B) Overall survival in patients without IKZF1 deletions, with IKZF1 deletions only and those with the IKZF1plus profile (5-year OS 93.0% vs. 100% vs. 76.2%, p = 0.290). A A B B Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL 99 Discussion Various alterations in genes involved in cell dif- ferentiation and cell cycle regulation are a hall- mark of B-ALL. The role of deletions in the IKZF1 gene in B-ALL has previously been described as very prognostically revealing and predictive for relapse.8,21 Due to their association with poorer treatment outcomes, it is important to detect them to adjust the treatment protocol accordingly. Moreover, alterations in other genes, such as PAX5 and CDKN2A/2B, are also often present. However, information regarding their prognostic impact is still rather ambiguous19,22,23,26, therefore, more stud- ies need to be carried out to define their true value for patient risk stratification. Our study was carried out on a smaller (88 patients), yet consecutive, unselected, and well- controlled population of pediatric patients with B-ALL. This is the first report about CNVs in cell differentiation and cell cycle regulation genes in Slovenian pediatric B-ALL patients. In our co- hort, IKZF1 deletions were identified in 18.2% of analysed samples. This is in concordance with reports of IKZF1 deletions ranging from 10.7 to 15.9% in other studies that analysed unselected co- horts.6,8,12–14,21 The IKZF1 deletions were more com- mon in the HR group when compared to the IR and SR groups, as was also shown in the study done by Dörge et al.6 Six out of seven (86%) patients with the BCR-ABL1 translocation also carried deletions in the IKZF1 gene, four of those had the IKZF1plus profile. As it was previously described, these dele- tions are very commonly, however not exclusively, present in BCR-ABL1 ALL.12,15,16,21 Some studies ex- cluded patients with the BCR-ABL1 translocation and determined the IKZF1 status only in the BCR- ABL1-negative patients. These studies reported fre- quencies of IKZF1 deletions from 9.4 to 16%.7,27-30 Our results show that 12.3% of patients without BCR-ABL1 in this cohort carried IKZF1 deletions, which is again similar to previously published re- sults. The most common IKZF1 deletion in our cohort was, as was also reported by other studies, the de- letion of the whole gene (50%). Interestingly, how- ever, the second most common deletion was that of exons 4–8 (25%), whereas this deletion was de- scribed as rare in other studies. The second most common deletion reported by other groups was that of exons 4–7 6,8,18,31 which results in a dominant- negative isoform (IK6). This deletion occurred in only one patient in our cohort. Nevertheless, all IKZF1 deletions, including the rare ones, have an important prognostic impact.18 Therefore, all should be considered when selecting the appropri- ate treatment. In our cohort, IKZF1 deletions and the IKZF1plus profile were more common amongst males and were associated with higher blast values on day 8 of chemotherapy, higher values of MRD15 and MRD33, and were more often found in the HR group. The presence of IKZF1 deletions and the IKZF1plus profile was higher amongst the patients who experienced an event (relapse or death) than those who did not, albeit this difference was not significant. Two of the patients who carried these deletions and experience a relapse also had the BCR-ABL1 fusion gene, which placed them in the HR group. However, three others with these dele- tions and an event did not exhibit genetic altera- tions that would predict a poorer outcome – one patient carried the ETV6-RUNX1 fusion, one was hyperdiploid and one had no recurrent genetic abnormalities. This shows that alterations in cell differentiation and cell cycle regulation genes may play a crucial role in disease development even in patients with primary genetic alterations that are thought to be favourable, which is in concordance with reports of these deletions being an independ- ent prognostic factor.6 Previously published studies have shown a poorer event-free and overall survival of patients with IKZF1 deletions in comparison to those with- out such aberrations6,8,12,14,16,30, and Stanulla et al. discovered that patients with additional muta- tions in certain genes that define the IKZF1plus pro- file have even more dismal outcomes.21 Our study similarly confirmed the inferior event-free survival for patients with IKZF1 deletions and the IKZF1plus profile, however, the overall survival did not sig- nificantly differ between the groups. After adjust- ing for confounding factors, our data did not con- firm the independent prognostic role of the IKZF1 deletions, as well as IKZF1plus, on EFS. However, the trend of poorer EFS in patients with the IKZF1 deletions and the IKZF1plus profile was observed. These discrepancies, as well as the high proportion of males presenting with the IKZF1 mutations, are most likely the result our small sample size and short follow-up duration. When inspecting non- high-risk patients with the IKZF1plus profile, the relative frequency of events among the patients with the profile was higher, and there was a trend of poorer EFS for these patients. The dismal out- comes for patients with the IKZF1plus profile have previously been confirmed by other studies, and this profile is currently already being used in cer- Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL100 tain treatment protocols20, and more are likely to follow. The CNV analysis of other genes showed that the most common alterations were in the PAX5 gene (30.7%), CDKN2A, and CDKN2B (26.1% and 25.0%, respectively). However, deletions in CDKN2A/2B were more common than deletions in PAX5 which was also observed by Öfverholm et al.31 and Mullighan et al.9 Deletions in PAX5 have previ- ously been reported as much more common than intragenic amplifications in this gene, and the latter have mostly been reported only in isolated cases.31-34 However, more recently, Schwab et al.23 showed that PAX5 amplifications occur in around 1% of B-ALL cases, out of which 40% experienced a relapse, sug- gesting that these alterations may play an important role in leukemogenesis. Interestingly, in our cohort, the amplifications were even more common, as they occurred in 10.2% of all B-ALL cases. Out of 27 patients with CNVs in PAX5, 9 (33.3%) carried amplifications. As already suggested by Schwab et al.23, more studies need to be conducted to evalu- ate the prognostic impact of PAX5 amplifications. The PAX5 deletions were also more frequent in our cohort (20.5%) than previously described (10%).35,36 Amongst the samples with PAX5 deletion, a sizable amount (61.1%) also carried the CDKN2A/2B dele- tions. The frequent co-occurrence of these deletions has previously been described by Kim et al.37 In our cohort, amplifications in the PAR1 region were observed quite frequently. Altogether, 17 pa- tients (19.3%) had at least one gene amplification in this region. This is due to the fact that our cohort is unselected, and therefore also includes patients with hyperdiploidy. In hyperdiploidy, gains in the X chromosome are very common (present in 70% of hyperdiploid childhood B-ALL cases)38, and indeed, 14 out of our 17 patients with amplifica- tions in PAR1 had a hyperdiploid karyotype. This karyotype is associated with a favourable outcome. However, the hyperdiploid patients in our cohort did not have a significantly better 5-year EFS, and the same was seen for the patients with amplifica- tions in the PAR1 region that were identified with MLPA. Two patients carried deletions in this re- gion (namely in CSF2RA, IL3RA and P2RY8 genes), which resulted in the formation of the P2RY8- CRLF2 fusion gene. This was confirmed with fluo- rescence in situ hybridization. While the P2RY8- CRLF2 fusion is associated with a poorer treatment response and outcomes39, the two patients in our cohort did not experience an event. Despite the limitations of our study due to a lower number of analyzed samples and relatively short follow-up period, it produced results that are in concordance with the results obtained in larger co- operative clinical trials. We have shown that it is possible to provide comparable results regarding the presence of certain CNVs and their prognostic value in pediatric B-ALL patients even within a single-center experience. This study is only a start- ing point for the more comprehensive screening of patients diagnosed with B-ALL in Slovenia that we have planned for the future and will enable us to better evaluate and treat these patients. Acknowledgments The research was supported by the Slovenian Research Agency (ARRS) grant number P3-0343. References 1. Roberts KG, Reshmi SC, Harvey RC, Chen IM, Patel K, Stonerock E, et al. Genomic and outcome analyses of Ph-like ALL in NCI standard-risk patients: a report from the children’s oncology group. Blood 2018; 132: 815-24. doi: 10.1182/blood-2018-04-841676 2. Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ, et al. 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 2012; 30: 1663-9. doi: 10.1200/JCO.2011.37.8018 3. Ward E, DeSantis C, Robbins A, Kohler B, Jemal A. Childhood and adolescent cancer statistics, 2014. CA Cancer J Clin 2014; 64: 83-103. doi: 10.3322/ caac.21219 4. Einsiedel HG, Von Stackelberg A, Hartmann R, Fengler R, Schrappe M, Janka- Schaub G, et al. Long-term outcome in children with relapsed ALL by risk- stratified salvage therapy: results of trial Acute Lymphoblastic Leukemia- Relapse Study of the Berlin-Frankfurt-Münster Group 87. J Clin Oncol 2005; 23: 7942-50. doi: 10.1200/JCO.2005.01.1031 5. Olsson L, Ivanov Öfverholm II, Norén-Nyström U, Zachariadis V, Nordlund J, Sjögren H, et al. The clinical impact of IKZF1 deletions in paediatric B-cell precursor acute lymphoblastic leukaemia is independent of minimal residual disease stratification in Nordic Society for Paediatric Haematology and Oncology treatment protocols used between 1992 aand 1993. Br J Haematol 2015; 170: 847-58. doi: 10.1111/bjh.13514 6. Dörge P, Meissner B, Zimmermann M, Möricke A, Schrauder A, Bouquin JP, et al. IKZF1 deletion is an independent predictor of outcome in pedi- atric acute lymphoblastic leukemia treated according to the ALL-BFM 2000 protocol. Haematologica 2013; 98: 428-32. doi: 10.3324/haema- tol.2011.056135 7. Asai D, Imamura T, Suenobu S, Saito A, Hasegawa D, Deguchi T, et al. IKZF 1 deletion is associated with a poor outcome in pediatric B-cell precursor acute lymphoblastic leukemia in Japan. Cancer Med 2013; 2: 412-9. doi: 10.1002/cam4.87 8. Kuiper RP, Waanders E, Van Der Velden VHJ, Van Reijmersdal SV, Venkatachalam R, Scheijen B, et al. IKZF1 deletions predict relapse in uni- formly treated pediatric precursor B-ALL. Leukemia 2010; 24: 1258-64. doi: 10.1038/leu.2010.87 9. Mullighan CG, Su X, Zhang J, Radtke I, Phillips LAA, Miller CB, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 2009; 360: 470-80. doi: 10.1056/NEJMoa0808253 10. Georgopoulos K, Bigby M, Wang JH, Molnar A, Wu P, Winandy S, et al. The ikaros gene is required for the development of all lymphoid lineages. Cell 1994; 79: 143-56. doi: 10.1016/0092-8674(94)90407-3 Radiol Oncol 2022; 56(1): 92-101. Crepinsek K et al. / Copy number variations in pediatric B-ALL 101 11. Molnar A, Wu P, Largespada DA, Vortkamp A, Scherer S, Copeland NG, et al. The Ikaros gene encodes a family of lymphocyte-restricted zinc finger DNA binding proteins, highly conserved in human and mouse. J Immunol 1996; 156: 585-92. PMID: 8543809 12. Yang YL, Hung CC, Chen JS, Lin KH, Jou ST, Hsiao CC, et al. IKZF1 deletions predict a poor prognosis in children with B-cell progenitor acute lympho- blastic leukemia: a multicenter analysis in Taiwan. Cancer Sci 2011; 102: 1874-81. doi: 10.1111/j.1349-7006.2011.02031.x 13. Van Der Veer A, Waanders E, Pieters R, Willemse ME, Van Reijmersdal S V., Russell LJ, et al. Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in chil- dren with B-cell precursor ALL. Blood 2013; 122: 2622-9. doi: 10.1182/ blood-2012-10-462358 14. Moorman A V., Enshaei A, Schwab C, Wade R, Chilton L, Elliott A, et al. A novel integrated cytogenetic and genomic classification refines risk stratifi- cation in pediatric acute lymphoblastic leukemia. Blood 2014; 124: 1434-44. doi: 10.1182/blood-2014-03-562918 15. Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 2008; 453: 110-14. doi: 10.1038/nature06866 16. Van Der Veer A, Zaliova M, Mottadelli F, De Lorenzo P, Te Kronnie G, Harrison CJ, et al. IKZF1 status as a prognostic feature in BCR-ABL1-positive childhood ALL. Blood 2014; 123: 1691-8. doi: 10.1182/blood-2013-06-509794 17. Roberts KG. The biology of Philadelphia chromosome-like ALL. Best Pract Res Clin Haematol 2017; 30: 212-21. doi: 10.1016/j.beha.2017.07.003 18. Boer JM, Van Der Veer A, Rizopoulos D, Fiocco M, Sonneveld E, De Groot- Kruseman HA, et al. Prognostic value of rare IKZF1 deletion in childhood B-cell precursor acute lymphoblastic leukemia: an international collabora- tive study. Leukemia 2016; 30: 32-38. doi: 10.1038/leu.2015.199 19. Steeghs EMP, Boer JM, Hoogkamer AQ, Boeree A, de Haas V, de Groot- Kruseman HA, et al. Copy number alterations in B-cell development genes, drug resistance, and clinical outcome in pediatric B-cell precursor acute lymphoblastic leukemia. Sci Rep 2019; 9: 1-11. doi: 10.1038/s41598-019- 41078-4 20. Stanulla M, Cavé H, Moorman A V. IKZF1 deletions in pediatric acute lymphoblastic leukemia: still a poor prognostic marker? Blood 2020; 135: 252-60. doi: 10.1182/blood.2019000813 21. Stanulla M, Dagdan E, Zaliova M, Möricke A, Palmi C, Cazzaniga G, et al. IKZF1 plus defines a new minimal residual disease-dependent very-poor prognostic profile in pediatric b-cell precursor acute lymphoblastic leuke- mia. J Clin Oncol 2018; 36: 1240-9. doi: 10.1200/JCO.2017.74.3617 22. Gu Z, Churchman ML, Roberts KG, Moore I, Zhou X, Nakitandwe J, et al. PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia. Nat Genet 2019; 51: 296-307. doi: 10.1038/s41588-018-0315-5 23. Schwab C, Nebral K, Chilton L, Leschi C, Waanders E, Boer JM, et al. Intragenic amplification of PAX5: a novel subgroup in B-cell precursor acute lymphoblastic leukemia? Blood Adv 2017; 1: 1473-7. doi: 10.1182/bloodad- vances.2017006734 24. Zhang W, Kuang P, Liu T. Prognostic significance of CDKN2A/B deletions in acute lymphoblastic leukaemia: a meta-analysis. Ann Med 2019; 51: 28-40. doi: 10.1080/07853890.2018.1564359 25. Carrasco Salas P, Fernández L, Vela M, Bueno D, González B, Valentín J, et al. The role of CDKN2A/B deletions in pediatric acute lympho- blastic leukemia. Pediatr Hematol Oncol 2016; 33: 415-22. doi: 10.1080/08880018.2016.1251518 26. Zhang W, Kuang P, Liu T. Prognostic significance of CDKN2A/B deletions in acute lymphoblastic leukaemia: a meta-analysis. Ann Med 2019; 51: 28-40. doi: 10.1080/07853890.2018.1564359 27. Yamashita Y, Shimada A, Yamada T, Yamaji K, Hori T, Tsurusawa M, et al. IKZF1 and CRLF2 gene alterations correlate with poor prognosis in japanese BCR-ABL1-negative high-risk B-cell precursor acute. Pediatr Blood Cancer 2013; 60: 1587-92. doi: 10.1002/pbc 28. Palmi C, Valsecchi MG, Longinotti G, Silvestri D, Carrino V, Conter V, et al. What is the relevance of Ikaros gene deletions as a prognostic marker in pediatric Philadelphia-negative B-cell precursor acute lympho- blastic leukemia? Haematologica 2013; 98: 1226-31. doi: 10.3324/haema- tol.2012.075432 29. Clappier E, Grardel N, Bakkus M, Rapion J, De Moerloose B, Kastner P, et al. IKZF1 deletion is an independent prognostic marker in childhood B-cell precursor acute lymphoblastic leukemia, and distinguishes patients benefit- ing from pulses during maintenance therapy: results of the EORTC Children’s Leukemia Group study 58951. Leukemia 2015; 29: 2154-61. doi: 10.1038/ leu.2015.134 30. Vrooman LM, Blonquist TM, Harris MH, Stevenson KE, Place AE, Hunt SK, et al. Refining risk classification in childhood b acute lymphoblastic leukemia: results of DFCI ALL consortium protocol 05-001. Blood Adv 2018; 2: 1449- 58. doi: 10.1182/bloodadvances.2018016584 31. Öfverholm I, Tran AN, Heyman M, Zachariadis V, Nordenskjöld M, Nordgren A, et al. Impact of IKZF1 deletions and PAX5 amplifications in pediatric B-cell precursor ALL treated according to NOPHO protocols. Leukemia 2013; 27: 1936-9. doi: 10.1038/leu.2013.92 32. Schwab CJ, Jones LR, Morrison H, Ryan SL, Yigittop H, Schouten JP, et al. Evaluation of multiplex ligation-dependent probe amplification as a method for the detection of copy number abnormalities in B-cell precursor acute lymphoblastic leukemia. Genes Chromosom Cancer 2010; 49: 1104-13. doi: 10.1002/gcc 33. Schwab CJ, Chilton L, Morrison H, Jones L, Al-Shehhi H, Erhorn A, et al. Genes commonly deleted in childhood B-cell precursor acute lymph- oblastic leukemia: association with cytogenetics and clinical features. Haematologica 2013; 98: 1081-8. doi: 10.3324/haematol.2013.085175 34. Mullighan CG, Goorha S, Radtke I, Miller CB, Coustan-Smith E, Dalton JD, et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 2007; 446: 758-64. doi: 10.1038/nature05690 35. Kim M, Choi JE, She CJ, Hwang SM, Shin HY, Ahn HS, et al. PAX5 deletion is common and concurrently occurs with CDKN2A deletion in B-lineage acute lymphoblastic leukemia. Blood Cells, Mol Dis 2011; 47: 62-6. doi: 10.1016/j. bcmd.2011.04.003 36. Kawamata N, Ogawa S, Zimmermann M, Sanada M, Hemminki K, Yamatomo G, et al. Rearrangement and deletion of the PAX5 gene in pediatric acute B-cell lineage lymphoblastic leukemia. Blood 2007; 110: 981. doi: 10.1182/ blood.V110.11.981.981 37. Kim M, Choi JE, She CJ, Hwang SM, Shin HY, Ahn HS, et al. PAX5 deletion is common and concurrently occurs with CDKN2A deletion in B-lineage acute lymphoblastic leukemia. Blood Cells, Mol Dis 2011; 47: 62-6. doi: 10.1016/j. bcmd.2011.04.003 38. Paulsson K, Johansson B. High Hyperdiploid childhood acute lymphoblas- tic leukemia. Genes Chromosom Cancer 2009; 48: 637-60. doi: 10.1002/ gcc.20671 39. Palmi C, Vendramini E, Silvestri D, Longinotti G, Frison D, Cario G, et al. Poor prognosis for P2RY8-CRLF2 fusion but not for CRLF2 over-expression in chil- dren with intermediate risk B-cell precursor acute lymphoblastic leukemia. Leukemia 2012; 26: 2245-53. doi: 10.1038/leu.2012.101