scholarly journals Multiclonal complexity of pediatric acute lymphoblastic leukemia and the prognostic relevance of subclonal mutations

Haematologica ◽  
2020 ◽  
pp. 0-0
Author(s):  
Željko Antić ◽  
Jiangyan Yu ◽  
Simon V. Van Reijmersdal ◽  
Anke Van Dijk ◽  
Linde Dekker ◽  
...  

Genomic studies of pediatric acute lymphoblastic leukemia (ALL) have shown remarkable heterogeneity in initial diagnosis, with multiple (sub)clones harboring lesions in relapse-associated genes. However, the clinical relevance of these subclonal alterations remains unclear. We assessed the clinical relevance and prognostic value of subclonal alterations in the relapse-associated genes IKZF1, CREBBP, KRAS, NRAS, PTPN11, TP53, NT5C2, and WHSC1 in 503 ALL cases. Using Molecular Inversion Probe sequencing and breakpoint-spanning PCR we reliably detected alterations below 1% allele frequency. We identified 660 genomic alterations in 285 diagnosis samples of which 495 (75%) were subclonal. RAS pathway mutations were common, particularly in minor subclones, and comparisons between RAS hotspot mutations revealed differences in their capacity to drive clonal expansion in ALL. We did not find an association of subclonal alterations with unfavorable outcome. Particularly for IKZF1, an established prognostic marker in ALL, all clonal but none of the subclonal alterations were preserved at relapse. We conclude that, for the genes tested, there is no basis to consider subclonal alterations detected at diagnosis for risk group stratification of ALL treatment.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2507-2507
Author(s):  
Smadar Avigad ◽  
Iedan RN Verly ◽  
Gertjan J.L. Kaspers ◽  
Jacqueline Cloos ◽  
Anat Ohali ◽  
...  

Abstract Abstract 2507 Aim: microRNAs (miRNAs) have been implicated in many malignancies. Our aim was to identify specific miRNAs that can predict risk of relapse in pediatric acute lymphoblastic leukemia (ALL) patients treated according to BFM protocols already at diagnosis. The current risk group stratification is based on the amount of minimal residual disease (MRD) assessed at specific time points by real time quantitative PCR (RQ-PCR). Material and methods: Following miRNA expression analysis, we decided to focus on miR-151-5p and miR-451 that significantly correlated with known prognostic factors in ALL. Validation was performed by measuring the expression levels of miR-151-5p and miR-451 by RQ-PCR on bone marrow samples at diagnosis of 101 B-lineage ALL patients excluding Philadelphia positive patients. Results: Low expression of miR-151-5p, miR-451 or of both together resulted in significantly worse relapse free survival (RFS) (43%, 58% and 38%, respectively) compared to RFS rates when either or both miRNAs were highly expressed (81%, 75% and 75%, respectively) (p=0.001, 0.044 and 0.001, respectively). Moreover, following PCR-MRD stratification, low expression of miR-451 or both miRNAs remained significant within the PCR-MRD medium risk group (p=0.00001) and within the standard group for both miRNAs (p=0.024). Multivariate Cox regression analysis identified low expression of both miRNAs as an independent prognostic marker with 11.7 fold increased risk for relapse (p=0.002). After excluding patients harboring the adverse genetic markers Ikaros deletion and P2RY8-CRLF2 rearrangement, a patient expressing low levels of both miRs had a 35 fold risk to relapse (p=0.005). Analyzing a non-BFM treated B-lineage ALL cohort from The Netherlands, both miRNAs significantly correlated with outcome (p=0.003). Conclusion: Our results identify miR-151-5p and miR-451 as novel biomarkers for outcome in pediatric B-lineage ALL patients, regardless of treatment protocol. This may allow earlier and improved risk group stratification already at diagnosis, enabling exploration of early therapeutic interventions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2608-2608
Author(s):  
Jiangyan Yu ◽  
Simon V. van Reijmersdal ◽  
Alexander Hoischen ◽  
Edwin Sonneveld ◽  
Peter M. Hoogerbrugge ◽  
...  

Abstract Introduction: Recent work has demonstrated that relapses in pediatric acute lymphoblastic leukemia (ALL) can arise from minor subclones present at diagnosis. Several genes have been associated with therapy resistance in these subclones, including the Ras pathway genes KRAS, NRAS, and PTPN11, the H3K36 methyltransferase NSD2 (WHSC1), and the 5'-nucleotidase NT5C2. Retrospective backtracking of these relapse-associated alterations has demonstrated that these alterations are frequently present at time of diagnosis in minor subclones, sometimes in less than a few percent of the cells. The prognostic value of subclonal alterations in these genes at time of diagnosis, however, is less well understood. Prospective screening of subclonal mutations, without prior knowledge of the mutation status, requires extra specificity and sensitivity. Accurate quantification of the subclonal burden of these mutations will provide potential for following the subclonal dynamics during early stages of treatment, and could be informative for adapting therapy. The aim of this study was to develop a targeted next generation sequencing assay to perform quantitative detection of subclonal mutations in the selected genes. We used single molecule molecular inversion probes (smMIPs), an approach that applies single molecule tagging to correct for amplification biases (Hiatt et al., Genome Research. 2013, 23: 843-854), an artifact that becomes relevant in case of low mosaic mutations. Method: We designed a pool of 77 smMIP oligonucleotides targeting the coding sequences of five genes associated with therapy resistance in BCP-ALL, including KRAS, NRAS, PTPN11, NT5C2, and WHSC1. The smMIPs tiled a total of 4124bp of genomic sequence, including hotspot regions of the genes. To demonstrate the potential of this method, we applied this newly designed smMIP panel on 22 BCP-ALL diagnosis samples to retrospective backtrack mutations in KRAS (n=11), NRAS (n=8) and PTPN11 (n=3) that were previously characterized at relapse. We used 100ng of genomic DNA per sample as input, which is the equivalent of 15,000 haploid copies. Sequencing was performed on the Illumina NextSeq platform with pair-end sequencing, data were analyzed by SeqNext v4.2.2. Result: The average read depth obtained varied per gene from 30,081x (NRAS) to 65,749x (PTPN11). Sequencing reads with the same molecular tag were clustered into one tag-defined read group, in which random errors caused by library construction and sequencing were eliminated. These so-called single molecule consensus reads (smc-reads) were comprised of, on average, 139 individual sequencing reads. Using the smMIP approach, 19 out of the 22 Ras pathway mutations identified at relapse were detectable at diagnosis, of which 10 had a low mutant allele frequency (varying from 0.52-8.31%), which is in line with our previous ultra-deep backtracking result. Taking advantage of the known position of the mutations at relapse, we established the noise level in the diagnosis samples by analyzing variant calls outside the hotspot regions. The noise level was varied between samples from 0.03% to 0.24% (average 0.06%). Based on these background settings, we subsequently searched for novel mutations and identified 1 mutation in NT5C2 (p.P534S, 0.38%), 2 hotspot mutations in WHSC1 (p.E1099K, 0.17% and 0.27%), as well as many additional subclonal mutations in KRAS, NRAS and PTPN11. The latter finding suggests the presence of multiple Ras-mutated subclones in individual cases, of which only a subset survive from chemotherapy and grow out in the relapse clone. Conclusions: Taken together, single molecule tagging based smMIP technology allows the accurate detection of low mosaic mutations. These findings illustrate the need for the current ongoing prospective mutation screens in unbiased cohorts of diagnosis samples to determine the prognostic value of subclonal mutations in these five genes on the risk of relapse. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3793-3793
Author(s):  
Isaac Yaniv ◽  
Asaf Lebel ◽  
Keren Shichrur ◽  
Oshrit Kordi ◽  
Anat Ohali ◽  
...  

Abstract Aim: microRNAs (miRs) have been implicated in many malignancies. Our aim was to identify specific miRNAs that can predict risk of relapse in pediatric acute lymphoblastic leukemia (ALL) patients treated on BFM protocols, better than the current risk group stratification. Currently, the main method for risk group stratification is based on the amount of minimal residual disease (MRD) assessed at specific time points by real time quantitative PCR (RQ-PCR). Material and methods: Following microRNA expression analysis performed on 48 bone marrow samples at diagnosis, we focused on several miRs that correlated with at least 3 of the established prognostic markers in ALL to be validated in a cohort of 138 B-lineage ALL samples. Of the miRs studied, down regulated miR-151-5p and miR-451 and upregulated miR-1290 significantly correlated with outcome. Results: Low expression of miR-151-5p, miR-451 or of both together resulted in significantly worse relapse free survival (RFS) (56%, 63% and 47%, respectively) compared to RFS rates when either or both miRs were highly expressed (80%, 78% and 78%, respectively) (p=0.007, 0.042 and 0.002, respectively). High expression of miR-1290 resulted in worse RFS compared to those that expressed low levels (54% versus 81%; p=0.014). When combining the 3 miRs, a patient expressing low levels of both miRs and/or high levels of miR-1290 had a RFS of 54% (p=0.004). Furthermore, the expression of the 3 miRs could distinguish within the TEL-AML1 negative cohort two groups; one with 80% and the other with 44% RFS (p=0.004). Multivariate Cox regression analysis identified low expression of both miRs and/or high expression of miR-1290 as an independent prognostic marker in the PCR-MRD non-high risk cohort. Patients expressing abnormal levels of the 3 miRs had a 4.47-fold increased risk for relapse (p=0.037). Deletion of IKZF1 gene is a known adverse prognostic marker in B-lineage ALL. In 85 patients, IKZF1 status was analyzed and 9 patients were found to harbor the deletion. The expression of the 3 miRs could still identify an adverse group of patients with 69% RFS within the group with no deletion (p=0.004). When adding IKZF1 and TEL-AML1 status to the Multivariate analysis in addition to PCR-MRD, the expression of the 3 miRs remained a significant independent marker with a 24 fold increased risk for relapse (p=0.011). Figure 1 Figure 1. Conclusion: Our results identify the combination of miR-151-5p, miR-451 and miR-1290 as a novel biomarker for outcome in pediatric B-lineage ALL patients, already at time of diagnosis. This may lead to improved risk group stratification and enable early therapeutic intervention that may result in better RFS. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 272-272 ◽  
Author(s):  
Esmé Waanders ◽  
Vincent H.J. van der elden ◽  
Ellen van der Schoot ◽  
Frank N. van Leeuwen ◽  
Simon V. van Reijmersdal ◽  
...  

Abstract Abstract 272 The response to therapy as determined by minimal residual disease (MRD) is currently used for stratification in treatment protocols for pediatric acute lymphoblastic leukemia (ALL). Even though MRD classification clearly identifies patients at low or at high risk for relapse, it also results in a large intermediate group (50 to 60% of patients), which still contains approximately half of all relapse cases. To improve risk stratification, we evaluated the added value of the IKZF1 alteration status, recently identified as a prognostic factor, in precursor-B-ALL patients. In an unbiased cohort of 131 uniformly treated precursor-B-ALL patients, we determined MRD levels at 42 and 84 days after treatment initiation using RQ-PCR analysis of Ig/TCR rearrangements. Based on these levels, patients were divided into three groups: MRD-Low (MRD-L), MRD-Medium (MRD-M) and MRD-High (MRD-H). IKZF1 alterations at diagnosis were determined using multiplex ligation-dependent probe amplification and genomic sequencing. We confirmed the strong prognostic significance of MRD classification, which was independent of IKZF1 status. Importantly, in the large MRD-M group (n=81; 62% of patients) containing 46% of the relapsed patients, IKZF1 alteration status identified 8 out of 11 relapsed patients (72%). The 9 year relapse-free survival (RFS) for IKZF1 mutated patients in this MRD-M group was 27% compared to 96% for patients wild-type for IKZF1 (P<0.001). Based on these results, we defined a new parameter integrating both MRD and IKZF1 status. The favorable risk group included patients classified as MRD-L or MRD-M with IKZF1 wild-type (n=104; 5 relapses), whereas the high risk group consisted of MRD-H patients or MRD-M patients with IKZF1 alterations (n=27; 19 relapses). This parameter showed stronger prognostic value than each of the established risk factors alone (Hazard Ratio[95%CI]: 24.98[8.29-75.31]). Importantly, whereas MRD and IKZF1 status alone identified only 46% and 54% of relapses, respectively, their integrated use allowed prediction of 79% of all relapses with 93% specificity. In conclusion: The use of a new parameter integrating MRD and IKZF1 status results in an unprecedented sensitivity in upfront relapse prediction and has a high potential for future risk stratification, particularly for patients originally classified as non-high-risk, such as the large group of MRD-M patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3539-3539 ◽  
Author(s):  
Inés Gómez-Seguí ◽  
Esperanza Such ◽  
Jose Cervera ◽  
Pascual Fernandez ◽  
Lurdes Zamora ◽  
...  

Abstract Abstract 3539 Background: Microdeletions of genes involved in B lymphopoiesis and cell-cycle regulation, such as CDKN2A/B, PAX5, IKZF1, ETV6, RB1, BTG1 and EBF1 have been reported as a frequent event in pediatric acute lymphoblastic leukemia (ALL). Whether these findings are found in adulthood and the possible differences with childhood ALL, as well as its prognostic implication, are still unknown. Aims: To assess the differences between two cohorts of children and adults diagnosed with ALL on the frequency of deletions in these genes and their relationship with clinical data and prognosis. Methods: We studied 70 children and 83 adults diagnosed with ALL with available DNA sample at diagnosis. In children, median age was 4y. (1 – 14), median leukocytes 10.3×109/L (0.7 – 675) and the cytogenetic risk distribution was 42(39%), 30(27%) and 12(11%) for favourable [t(12;21) and hyperdiploidy], intermediate (normal karyotype and miscellaneous) and high risk [t(9;22), t(4;11), hypodiploid and complex karyotype], respectively. In adults, median age was 38y. (15 – 85), median leukocytes 16.8×109/L (1 – 371) and 29(35%) patients belonged to the high risk cytogenetic group. We performed Multiplex Ligation Probe Amplification (MLPA) using SALSA kit P335-A1 (MRC-Holland). PCR products were separated on an ABIPRISM 310 DNA Analyzer and analyzed using GeneMapper v3.2 (Applied Biosystems). Results: Frequency of deletions in the studied genes was similar in children and adults, except for IKZF1 deletions that were more frequent in adults (P<.001) (Table 1). In children, ETV6 deletions occurred more frequently in patients with t(12;21) (67% of patients with deletion vs. 17% without, P <.001); CDKN2A/B deletions were found in patients assigned to the intermediate cytogenetic risk group (59% of patients with deletion vs. 23% without, P =.028); and the three cases with RB1 deletions were found in patients with hypodiploidy (P <.001). In adults, ETV6 and CDKN2A/B deletions occurred more frequently in women (67% vs. 39%, P =.022 and 77% vs. 42%, P =.021, for patients with and without deletions, respectively); PAX5 and IKZF1 deletions appeared more frequently in patients with >30×109/L leukocytes (60% vs. 27%, P =.032 and 52% vs. 21%, P =.007, for patients with and without deletions, respectively); besides, PAX5 deletions occurred in patients who belonged to the standard cytogenetic risk group (55% vs. 6% for patients with and without deletions, P <.001). In the pediatric cohort, the leukocytes >30×109/L and the cytogenetic risk group were the variables that reached statistical significance for both overall survival (OS) and relapse free survival (RFS) and also age >10y. for OS, but in the multivariate analyses, just the cytogenetic risk classification remained significant [HR: 4 (CI 95%: 1.6 – 10), P =. 004 for OS and HR: 3.5 (CI 95%: 1.7 – 7.2), P =. 001 for RFS]. In the adult cohort, multivariate analysis for OS including all significant variables in the univariate analysis (age >60y, karyotype, CDKN2A/B and ETV6 deletions) showed as independent variables: age >60y. [HR: 4.3 (CI 95%: 2.1 – 8.6), P<. 001] and CDKN2A/B deletions [HR: 2.6 (CI 95%: 1.4 – 5.3), P=. 004]. Similarly, taking into account karyotype, CDKN2A/B and ETV6 deletions for the RFS multivariate analyses, just ETV6 deletions arose as an independent factor [HR: 3.8 (CI 95%: 1.5 – 9.4), P=. 004]. In fact, having CDKN2A/B and/or ETV6 deletions conferred a worse prognosis to patients in both standard risk cytogenetic group (3y. RFS: 45% vs. 70% for patients with and without deletions, respectively; P =.049) and high risk cytogenetic group (3y. RFS: 14% vs. 66% for patients with and without deletions, respectively; P =.025). Conclusions: This study shows the high incidence of deletions in genes of cell-cycle and B-lymphopoiesis in adult and pediatric ALL. However, the biological and prognostic implications of these deletions seem to differ between both patient groups: while cytogenetics was the strongest variable for risk assessment in children, gene microdeletions in CDKN2A/B and ETV6 added a prognostic value to karyotype in our adult cohort. Fundings: AP-194/10, R06/0020/0031, BES2008–008053, CM10/00321, CM09/00038, and CA08/00141. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shumaila Sayyab ◽  
Anders Lundmark ◽  
Malin Larsson ◽  
Markus Ringnér ◽  
Sara Nystedt ◽  
...  

AbstractThe mechanisms driving clonal heterogeneity and evolution in relapsed pediatric acute lymphoblastic leukemia (ALL) are not fully understood. We performed whole genome sequencing of samples collected at diagnosis, relapse(s) and remission from 29 Nordic patients. Somatic point mutations and large-scale structural variants were called using individually matched remission samples as controls, and allelic expression of the mutations was assessed in ALL cells using RNA-sequencing. We observed an increased burden of somatic mutations at relapse, compared to diagnosis, and at second relapse compared to first relapse. In addition to 29 known ALL driver genes, of which nine genes carried recurrent protein-coding mutations in our sample set, we identified putative non-protein coding mutations in regulatory regions of seven additional genes that have not previously been described in ALL. Cluster analysis of hundreds of somatic mutations per sample revealed three distinct evolutionary trajectories during ALL progression from diagnosis to relapse. The evolutionary trajectories provide insight into the mutational mechanisms leading relapse in ALL and could offer biomarkers for improved risk prediction in individual patients.


2021 ◽  
Vol 10 (9) ◽  
pp. 1926
Author(s):  
Hiroto Inaba ◽  
Ching-Hon Pui

The outcomes of pediatric acute lymphoblastic leukemia (ALL) have improved remarkably during the last five decades. Such improvements were made possible by the incorporation of new diagnostic technologies, the effective administration of conventional chemotherapeutic agents, and the provision of better supportive care. With the 5-year survival rates now exceeding 90% in high-income countries, the goal for the next decade is to improve survival further toward 100% and to minimize treatment-related adverse effects. Based on genome-wide analyses, especially RNA-sequencing analyses, ALL can be classified into more than 20 B-lineage subtypes and more than 10 T-lineage subtypes with prognostic and therapeutic implications. Response to treatment is another critical prognostic factor, and detailed analysis of minimal residual disease can detect levels as low as one ALL cell among 1 million total cells. Such detailed analysis can facilitate the rational use of molecular targeted therapy and immunotherapy, which have emerged as new treatment strategies that can replace or reduce the use of conventional chemotherapy.


Sign in / Sign up

Export Citation Format

Share Document