Open Access

Panel‑based next‑generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings

  • Authors:
    • Hisashi Ishida
    • Akihiro Iguchi
    • Michinori Aoe
    • Ritsuo Nishiuchi
    • Takehiro Matsubara
    • Dai Keino
    • Masashi Sanada
    • Akira Shimada
  • View Affiliations

  • Published online on: August 28, 2020     https://doi.org/10.3892/br.2020.1353
  • Article Number: 46
  • Copyright: © Ishida et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute myeloid leukemia (AML) accounts for ~20% of pediatric leukemia cases. The prognosis of pediatric AML has been improved in recent decades, but it trails that of most other types of pediatric cancer, with mortality rates of 30‑40%. Consequently, newer more targeted drugs are required for incorporation into treatment plans. These newer drugs selectively target AML cells with specific gene alterations. However, there are significant differences in genetic alterations between adult and pediatric patients with AML. In the present study, inexpensive and rapid next‑generation sequencing (NGS) of >150 cancer‑related genes was performed for matched diagnostic, remission and relapse (if any) samples from 27 pediatric patients with AML. In this analysis, seven genes were recurrently mutated. KRAS was mutated in seven patients, NRAS was mutated in three patients, and KIT, GATA1, WT1, PTPN11, JAK3 and FLT3 were each mutated in two patients. Among patients with relapsed AML, six harbored KRAS mutations at diagnosis; however, four of these patients lost these mutations at relapse. Additionally, two genetic alterations (FLT3‑ITD and TP53 alterations) were detected among patients who eventually relapsed, and these mutations are reported to be adverse prognostic factors for adult patients with AML. This panel‑based, targeted sequencing approach may be useful in determining the genetic background of pediatric AML and improving the prediction of treatment response and detection of potentially targetable gene alterations. RAS pathway mutations were highly unstable at relapse; therefore, these mutations should be chosen as a target with caution. Incorporating this panel‑based NGS approach into the clinical setting may allow for a patient‑oriented strategy of precision treatment for childhood AML.

Introduction

Acute myeloid leukemia (AML) accounts for ~20% of all cases of pediatric leukemia (1). Although the prognosis of pediatric AML has improved in recent decades, it trails that of other types of pediatric cancer, as 30-40% of children with AML eventually succumb to the disease (1-3). The treatment strategies for pediatric AML include intensive multimodal chemotherapy with or without stem cell transplantation, and cytarabine and anthracyclines have remained the primary choices of chemotherapy for >30 years (1,2,4,5). As clinical outcomes have not improved, even with intensive contemporary chemotherapeutic regimens and/or stem cell transplantation, newer targeted drugs are required for incorporation into treatment plans (5,6).

Recently, various newer targeted therapies have emerged, most of which target AML cells with specific genetic alterations (7). Among them, FLT3 inhibitors, such as midostaurin and gilteritinib, and IDH inhibitors, such as enasidenib and ivosidenib have already been approved for use in clinical settings (7). These newer drugs selectively target AML cells with specific features; hence, the genomic characterization of AML cells is becoming increasingly important in the clinical setting. Using next-generation sequencing (NGS), several studies have reported on the value of performing NGS for adult patients with AML (8-12). However, there are comparatively fewer reports focusing on childhood AML, particularly in cases of relapsed AML (13,14).

In the present study, panel-based, targeted NGS for the molecular characterization of AML cells from pediatric patients was retrospectively performed. The objective of this study was to determine whether it was possible to obtain clinically useful information for children with AML through this NGS approach.

Materials and methods

Patients

A total of 27 children aged 0-18 years who were diagnosed with AML between January 2000 and December 2017 in Okayama University Hospital, Kochi Health Sciences Center, St. Marianna University School of Medicine Hospital or Hokkaido University Hospital were enrolled in the present study. The ratio of boys to girls was 17:10 in the present cohort and the median age at diagnosis was 6 years (range, 0 months to 15 years). The treatment protocols were diverse, including those from the Japanese Pediatric Leukemia/Lymphoma Study Group AML-99(15), AML-05(16) and AML-12 studies, and are listed in Table I. The standard chimeric fusion gene screening varied has changed over time in Japan. AML-05 protocol included the PCR-based detection of eight frequent chimeric gene fusions; RUNX1-RUNXT1, CBFB-MYH11, KMT2A-MLLT3, KMT2A-MLLT4, KMT2A-MLLT1, FUS-ERG, NUP98-HOXA9 and PML-RARA, and the AML-12 protocol included eight gene fusions; RUNX1-RUNXT1, CBFB-MYH11, KMT2A-MLLT3, KMT2A-MLLT4, BCR-ABL, FUS-ERG, NUP98-NSD1 and PML-RARA. In our previous study, the clinical courses of UPN 6 and UPN 25 was reported (17,18). The present study was performed in accordance with the Declaration of Helsinki and other applicable guidelines (19-21). The present study was approved by the Institutional Ethics Committee of Okayama University Hospital, and informed consent was obtained the patients and/or their legal guardians.

Table I

Clinical information of the analyzed patients.

Table I

Clinical information of the analyzed patients.

A, Relapsed AML
UPNAgeSexDisease subtypeChimeric geneKaryotypingInitial therapyaSCT at CR1Duration of CR1, monthsKaryotype at relapseOutcomeInstitute
112 yearsMFAB M5KMT2A-MLLT346, XY, add(11)(q23), der(21)t(1;21) (q11;q11.2) [9/20], 46, sl, der(19)t(11;19) (q13;p13) [11/20]AML 05No2447, XY, add(11) (q23), +marlAliveOU
24 yearsMFAB M6 (RAEB)None46, XY, inv(9) (p11q13)OtherYes746, XY, t(2;5)(q33;q31), add(6)(q21), t(6;12)(q13;p13), inv(9)(p11q13)AliveOU
35 monthsMFAB M7 (non-Down-syndrome)None49, XY, +4, ?t(4;5)(q21;q15), del(12)(p?), +19, +22AML 12No1350, XY, +4, ?t(4;5)(q21;q15), +8, del(12)(p?), +19, +22AliveOU
410 yearsMFAB M5None48, XY, +16, +19AML 05No946, XY, add(7)(q32), ?t(10;11)(p12;q23)Succumbed to diseaseOU
54 yearsFFAB M7 (non-Down-syndrome)None46, XX, add(5) (p13), del(6)(q), add(12)(p11.2)OtherNo2246, XX, add(5)(p13), del(6)(q), add(12)(p11.2)AliveOU
612 yearsFFAB M2None46, XXAML 05Yes7NASuccumbed to diseaseKHSC
77 yearsFFAB M2None46, XXAML 99No1046, XXSuccumbed to diseaseHU
81 yearMFAB M7 (Down-syndrome)None47, XY, +21OtherNo1245, XY, -10, der(17) t(17;18)(p11;q11), -18, +21Dead (CR)HU
913 yearsMFAB M4NA46, XY, t(6;11) (q27;q23)AML 99No1846, XY, t(6;11)(q27;q23)Succumbed to diseaseHU
101 yearMFAB M5bNA47, XY, t(9;11) (p22;q23), +mar1AML 99No6NAAliveHU
1114 yearsMFAB M1None46, XY, del(9)(q?)AML 05No1846, XYAliveHU
1211 yearsFT/MyeloidNone46, XX, t(1;2) (p31;p16)AML 05No1346, XX, t(1;2)(p31;p16)AliveHU
135 yearsMFAB M7 (non-Down-syndrome)None47, XY, del(9) (q12q34), del(12)(p12), +21OtherYes1947, XY, +8, del(9)(q12q34), t(10;17)(q11.2;q25), del(12)(p12)Succumbed to diseaseHU
145 yearsFFAB M4None46, XX, der(2) t(11;10;2)(q21; q11.2;q37), der(10) add(10)(p11.2) t(11;10;2), der(11) t(11;10;2)AML 05No2446, XXSuccumbed to diseaseHU
1514 yearsMT/MyeloidNone47, XY, del(4)(q?), +22, incAML05No447,XY, del(4)(q?), 22AliveSMU
B, Non-relapsed AML
UPNAgeSexDisease subtypeChimeric geneKaryotypingInitial therapyaSCT at CR1Duration of CR1, monthsKaryotype at relapseOutcomeInstitute
1610 yearsMFAB M3PML-RARA46, XY, t(15;17) (q22;q11~21)AML99No--AliveOU
179 yearsFFAB M3PML-RARA46, XX, t(15;17) (q22;q12)AML 99No--AliveOU
188 monthsFFAB M5aKMT2A-MLLT1046, XX, t(10;11) (p12;q23)AML 12Yes--AliveOU
1913 yearsMFAB M4EoCBFB-MYH1147, XY, +8, inv(16)(p13.1q22)AML 05No--AliveOU
207 yearsMFAB M2RUNX1-RUNXT145, X, -Y, t(8;21) (q22;q22)AML 05No--AliveOU
2115 yearsMAML with myelodysplasia-related changesNone46, XYOtherYes--Dead (CR)OU
226 yearsFFAB M2CBFB-MYH1146, XX, inv(16) (p13.1q22)AML 05No--AliveOU
235 yearsFFAB M2RUNX1-RUNXT146, XX, t(8;21) (q22;q22)AML 05No--AliveOU
243 yearsMFAB M2RUNX1-RUNXT146, XY, t(1;21;8) (p36;q22;q22), del(9)(q13q22)AML 99No--AliveOU
250 monthsFFAB M5KAT6A-CREBBP t(8;16)(p11;p13)AML 99No--AliveOU
2611 monthsMFAB M5KMT2A-MLLT346, XYAML 99No--AliveOU
271 yearMFAB M2RUNX1-RUNXT146, XY, t(8;21)(q22;q22), del(9)(q?), add(11)(q23)AML 99No--AliveOU

[i] aDetails regarding treatment protocols are described in references (15,16). UPN, unique patient number; AML, acute myeloid leukemia; SCT, stem cell transplantation; CR1, first complete remission; OU, Okayama University Hospital; HU, Hokkaido University Hospital; SMU, St. Marianna University School of Medicine Hospital; KHSC, Kochi Health Sciences Center.

DNA isolation

Somatic DNA was obtained from bone marrow samples at diagnosis and each episode of relapse, whereas germline DNA was obtained from a buccal swab or peripheral blood in CR status. DNA was extracted using a QIAamp DNA Blood Mini kit (Qiagen, Inc.) and quantified using a NanoDrop 2000 (Thermo Fisher Scientific, Inc.) and a Qubit® 2.0 Fluorometer (Thermo Fisher Scientific, Inc.) according to the manufacturers' protocol.

Targeted NGS approach

Targeted sequencing of >150 cancer-related genes was performed as described previously (22-24). The targeted gene lists are shown in Table SI and patient allocation is shown in Table SII. These gene panels were generated using an online design tool for HaloPlex (Agilent Technologies, Inc.), and target enrichment was performed using the HaloPlex standard protocol. Samples were then sequenced using MiSeq (Illumina, Inc.). Read alignment to the hg19 reference genome was performed using Burrows-Wheeler Aligner (bio-bwa.sourceforge.net) and variant calling was performed using SureCall version 3.0 (Agilent Technologies, Inc.).

Variant prioritization and assessment of pathogenicity

Synonymous or non-coding variants and single nucleotide polymorphisms reported with a frequency of >1% in various databases (dbSNP, 1000gp and Human Genetic Variation Database) were excluded. Variant bases that had >5 reads in each sample were used for the next step. Genetic variations that were constantly detected from diagnostic, remission and relapse samples with a variant allele frequency (VAF) ≥0.2 were regarded as candidate germline alterations. Genetic alterations that were rarely detected, or not detected at all, from remission samples but were detected from diagnostic and/or relapse samples with a VAF ≥0.05 were regarded as candidate somatic alterations with reference to a previous study (25). To exclude the possibility of false-positive findings, differences in VAF between normal and diagnostic/relapse samples were assessed using a Fisher's exact test. P<0.01 was considered to indicate a statistically significant difference. Finally, for germline and somatic alterations, the read quality was checked using IGV software version 2.3 (Broad Institute).

Results

Patient characteristics

The clinical information of the analyzed patients is shown in Table I. All patients were Japanese. The present cohort included one patient with Down-syndrome (UPN 8) and no other patients had a known underlying congenital condition. Remission samples were obtained by buccal smear from patient No. 2 and the sample collection was performed during remission. All relapsed patients lost their first remission within 24 months; 6 out of 15 patients experienced their relapse within 12 months, which is considered to be an adverse prognostic factor for survival (26).

Descriptive results from sequencing runs

The average number of total reads was 1,917,277 (range, 998,341-4,333,332) and the average read length was 116-136. The read depth in analyzable target regions per sample ranged from 168-757x. In total, 70.49-97.51% of analyzable regions were covered by at least 20 reads, 66.72-95.24% were covered by at least 50 reads and 58.15-91.05% were covered by at least 100 reads. These quality metrics data were obtained from analysis using SureCall version 3.0 software (Agilent Technologies, Inc.) and the details are shown in Table SII.

The results of NGS are shown in Table II. A total of 26 single nucleotide variations (SNVs) and insertions/deletions (indels) were identified at diagnosis, and 22 SNVs and indels at relapse for 15 patients with relapsed AML, as well as 12 SNVs and indels for the leukemia samples of 12 patients without relapse.

Table II

Gene alterations detected at diagnosis and relapse.

Table II

Gene alterations detected at diagnosis and relapse.

A, Relapsed AML
UPNDisease subtypeSNVs at diagnosis (VAF)SNVs at relapse (VAF)
1FAB M5KRASp.G12V (0.296)None
2FAB M6 (RAEB) PTPN11p.G60V(0.83) PTPN11 p.V45L (0.10) PTPN11p.G60V(0.44)
3FAB M7 (non-Down-syndrome)KRAS p.A146T (0.09)None
4FAB M5NoneNone
5FAB M7 (non-Down-syndrome)GATA1p.R270W (0.43) KRAS p.G12A (0.43) IKZF1 p.F154Y (0.38)a IKZF1p.L161H (0.38)a GATA1p.R270W(0.23) KRAS p.G12A (0.19) IKZF1p.F154Y(0.17)a IKZF1p.F161H(0.17)a
6FAB M2FLT3-ITD (0.572)FLT3-ITD (0.689)
7FAB M2PTPN11 p.I479F (0.40) + c.1473-1481dela PTPN11p.I479F (0.35) + c.1473-1481dela
8FAB M7 (non-Down-syndrome)GATA1 p.P50fs (0.30)GATA1 p.P50fs (0.05)
9FAB M4KRAS p.G12V (0.41) SMARCA4p.R979Q (0.06)aMLH1p.A586S (0.06)a
10FAB M5bKRAS p.G13D (0.32) RUNX1p.Q390fs(0.2)
11FAB M1GATA2 p.R362Q (0.11) CEBPAp.Q312HR (0.99)a CEBPAp.D63fs(0.37)a CEBPAp.Q312HR (0.39)a
12T/MyeloidJAK3 p.L857P (0.10) NOTCH1p.V1721E (0.33) STAT5Bp.N642H(0.11) NF1 p.L2317H (0.05)aJAK3 p.L857P (0.13) NOTCH1p.V1721E (0.33) IL7R p.L243GTARCV (0.10)a VHL p.L85fs (0.11)
13FAB M7 (non-Down-syndrome)NoneNone
14FAB M4U2AF1 p.R35L (0.53) KRAS p.G12D (0.37)KRAS p.G12D (0.23)
15T/MyeloidTP53 p.K164E (0.998) PHF6 p.R274fs (0.724) NPM1 p.L287fs (0.579)TP53 p.K164E (0.858) PHF6p.R274fs (0.611) NPM1 p.L287fs (0.53) MPLp.A486V(0.064)a
B, Non-relapsed AML
UPNDisease subtypeSNVs at diagnosis (VAF)SNVs at relapse (VAF)
16FAB M3None-
17FAB M3None-
18FAB M5aNone-
19FAB M4EoKIT p.D816Y (0.28)-
  WT1 p.H448Y (0.22)a-
  NRAS p.Q61K (0.08)-
20FAB M2JAK3 p.M511I (0.13)-
21AML with myelodysplasia-related changesNone-
22FAB M2NRAS p.G13D (0.37)-
  WT1 p.D447N (0.34)-
23FAB M2NRAS p.Q61K (0.25)-
24FAB M2KRAS p.G12D (0.52)-
25FAB M5None-
26FAB M5FLT3 p.D839G (0.21)-
  FLT3 p.Y591D (0.08)-
  FLT3 p.D839N (0.07)-
27FAB M2KIT p.N822K (0.43)-

[i] aAlterations not reported in Catalogue of Somatic Mutations in Cancer. UPN, unique patient number; AML, acute myeloid leukemia; VAF, variable allele frequency; SNV, single nucleotide variation.

Somatic genetic alterations at diagnosis and AML subtypes

In the present study, seven genes were recurrently mutated. KRAS was mutated in 7 patients, NRAS was mutated in 3 patients, and KIT, GATA1, WT1, PTPN11, JAK3 and FLT3 were each mutated in 2 patients. As previously reported, KIT was mutated in patients with core-binding factor AML (UPN 19 and UPN 27), and GATA1 was mutated in patients with acute megakaryoblastic leukemia with or without Down-syndrome (UPN 5 and UPN 8) (27-29). The present cohort included 2 patients with mixed-phenotype acute leukemia, and these patients harbored mutations previously reported in a larger study (30). Other mutations were not apparently associated with a specific type of AML. Most detected mutations have been reported in the Catalogue Of Somatic Mutations In Cancer database (cancer.sanger.ac.uk/cosmic), but some alterations were not. These mutations were thought to be variants of unknown significance and are indicated with a superscripted letter in Table II.

Prognostic genetic alterations detected at diagnosis

According to the European LeukemiaNet (ELN) guideline, several factors are associated with the prognosis of AML (31). Among these, two high-risk genetic alterations (FLT3-ITD in patient 6 and TP53 alteration in patient 15) and one low-risk genetic alteration (CEBPA mutation in patient 11) were added to the known cytogenetic risk factors (Table III). According to the guidelines, RUNX1 or ASXL1 alterations are indicated not to be used as adverse prognostic markers if they are present concurrently with favorable-risk AML subtypes. However, TP53 alterations are regarded as an independent adverse prognostic factor, thus UPN 15 was placed in the ELN high-risk group. The gene alterations which were defined in the ELN guideline or recurrently detected in the present study are summarized in Fig. 1.

Table III

Prognostic genetic alterations at diagnosis according to the European LeukemiaNet guidelines.

Table III

Prognostic genetic alterations at diagnosis according to the European LeukemiaNet guidelines.

UPNLow-risk featuresIntermediate risk featuresHigh-risk features.
1No KMT2A-MLLT3Complex karyotype
2NoNoNo
3NoNoComplex karyotype
4NoNoNo
5NoNoComplex karyotype
6NoNo FLT3-ITD
7NoNoNo
8NoNoNo
9NoNo t(6;11)(q27;q23)
10No t(9;11)(p22;q23)No
11CEBPA p.Q312HRNoNo
12NoNoNo
13NoNoComplex karyotype
14NoNoComplex karyotype
15NoNoTP53 p.K164E
16 PML-RARANoNo
17 PML-RARANoNo
18NoNo KMT2A-MLLT10
19 CBFB-MYH11NoNo
20 RUNX1-RUNXT1NoNo
21NoNoNo
22 CBFB-MYH11NoNo
23 RUNX1-RUNXT1NoNo
24 RUNX1-RUNXT1NoNo
25NoNoNo
26No KMT2A-MLLT3No
27 RUNX1-RUNXT1NoNo

[i] UPN, unique patient number.

Mutational changes between diagnosis and relapse

A total of 15 patients who experienced relapse were analyzed. Among these, six harbored KRAS mutations at diagnosis. However, four of the six patients lost these mutations at relapse. None of the patients gained new RAS pathway mutations at relapse.

UPN 11 had a CEBPA p.Q312HR insertion-type alteration at diagnosis, and this alteration appeared to be homozygous as its VAF was notably high (0.99). At relapse however, the VAF of CEBPA p.Q312HR alteration decreased, and a new p.D53fs alteration appeared, leading to a ‘double hit’ status.

Germline variations

In the present study, matched samples at diagnosis, remission and relapse (if any) among patients with childhood AML was analyzed, and this approach enabled detection of germline variations. Although candidates of germline variations were detected in 8 patients, none were regarded as pathogenic or likely pathogenic according to published recommendations (19,21). These candidate genes are listed in Table SIII.

Discussion

In the present study, matched samples obtained from pediatric patients with AML at diagnosis, remission and relapse (if any) were analyzed using a panel-based NGS method. Several studies have reported the utility of NGS for analysis of AML in adult patients (8-12). However, there are comparatively fewer reports focusing on childhood AML, particularly in cases of relapsed AML (13,14).

The utility of NGS should be discussed separately in adult and pediatric patients. As was shown in the present study, there are significant differences in genetic alterations between adult and pediatric patients with AML. Whereas mutations in epigenetic components or spliceosome complexes are common among adults with AML (11), these mutations were notably less common amongst children with AML, based on the results of the present study. However, large structural aberrations such as chromosomal translocations are more common amongst children with AML (11,14). As a result, disease stratification guidelines or newer drugs developed for adults with AML may not always be suitable for children.

The panel-based NGS strategy has been used several times in our previous studies for hematological malignancies, and the results of this method have been compared with conventional approaches, such as the multiplex ligation-dependent probe amplification or Sanger sequencing in our previous studies (22-24). Based on the panel-based NGS approach, several prognostic genetic alterations for patients with AML were detected in the present study. The ELN guidelines identified several cytogenetic alterations and a smaller number of genetic mutations as prognostic factors. In the present cohort, additional cytogenetic risk factors possessed prognostic implications than the genetic mutations. Using this approach, two high-risk genetic mutations were detected amongst patients who relapsed but did not possess any cytogenetic risk factors (UPN 6 and UPN 15). Conversely, none of the non-relapsed patients possessed adverse prognostic genetic mutations, and non-relapsed patients were enriched in low risk genetic alternations defined in the ELN guidelines. Thus, it may be possible to more accurately predict the prognosis of patients with AML using this approach. However, as noted above, there are significant differences in genetic alterations between adult and pediatric patients with AML. Previous larger studies suggested that current guidelines, including the ELN guidelines, are not adequate for children with AML (14,32,33), thus, there is a need for the development of a pediatric-specific guidelines for more precise stratification.

FLT3-ITD has a prognostic impact in pediatric AML (34,35), and this alteration was detected using the panel-based NGS method. However, the clinical impact of FLT3-ITD has been reported to be modulated by other sequence aberrations (WT1 mutation, NUP98-NSD1 or NPMI mutations for adult AML) (14,36). In this context, panel-based sequencing may be more useful than conventional approaches that detect only FLT3-ITD. However, the ELN guidelines recommend the use of DNA fragment analysis to determine the ratio of FLT3-ITD and prognosis. To confirm the usefulness of the NGS approach, a direct comparison of standard procedures and the NGS approach is required.

The number of genes that should be assessed has increased; however, one large study found that a limited number of genes are recurrently mutated in pediatric patients with AML (14); where several genetic analytical methods, including whole-genome and targeted DNA sequencing were performed. Mutations in only 5 genes (FLT3, NPM1, WT1, CEBPA and KIT) were present in >5% of patients, and <40 gene mutations were reported in >2% of patients. This previous study illustrated the need to focus on these 40 genes to detect recurrent gene mutations in AML cells obtained from pediatric patients, and that panel-based sequencing is an ideal approach in a clinical setting. Furthermore, Morita et al (37) reported that the clearance of somatic mutation at remission was associated with significantly improved survival and a lower risk of relapse. This strategy requires a sufficient read depth to detect mutations with a VAF <1%; contrarily, the approach used in the present study could not reach that read depth due to low throughput and relatively high number of targeted genes. Limiting the number of targeted genes to 40 recurrently mutated genes will increase the read depth and potentially enable detection of gene alterations with lower VAFs.

The panel-based approach used in the present study also offers potentially useful information regarding targetable genetic alterations. Cytotoxic chemotherapy primarily based on cytarabine and anthracyclines with or without stem cell transplantation has long been the mainstay of AML treatment (1). The curative rate of pediatric AML steadily improves with increasing doses of these drugs; however, this leads to substantial treatment-related complications in vulnerable pediatric populations (1,16,26). Hence, newer targeted therapies are desired. These targeted drugs include midostaurin, gilteritinib, enasidenib and ivosidenib, which target leukemic cells with specific genetic alterations such as FLT3, KIT, MEK, DOT1L or BET alterations (5,6); hence, genomic characterization of AML cells is becoming increasingly important in the clinical setting. Among these potentially targetable gene alterations, 6 patients had RAS pathway mutations at the time of AML diagnosis. However, in the present cohort, a notably high percentage of patients (four out of six cases, 66.7%) lost the KRAS mutations at relapse. Thus, RAS pathway mutations should be chosen with caution as treatment targets.

The present study has several limitations. First, the study was retrospective, and it was not possible to clarify whether dose increases in patients who had high-risk features would improve their prognoses. Second, the method used in the present study has a disadvantage of comparatively low throughput. Third, potential false positives were excluded, thus several relevant genetic alterations may have been missed. To detect minor clones with low variant frequencies at diagnosis, the number of genes to be targeted should be limited, as noted above. The genetic events including TP53 loss of heterozygosity in UPN 15 should have also been illustrated using other experiments, such as fluorescent in situ hybridization, but this could not be achieved due to the sample availability and quality. Relatively small patient numbers is another disadvantage of the present study. Furthermore, the ELN guidelines were established for adult patients; hence its validity in a larger cohort of pediatric patients with AML requires validation.

In summary, the panel-based targeted sequencing approach used in the present study may be useful for revealing the genetic background of pediatric AML, and may facilitate the precise prediction of patient prognosis and detection of druggable gene alterations. Incorporating this method into the clinical setting may enable a patient-oriented precision strategy for treatment of childhood AML.

Supplementary Material

HaloPlex custom panels used in the present study.
Quality metrics of next-generation sequencing.
Candidate germline variants detected in the present study.

Acknowledgements

Not applicable.

Funding

This present study was supported by grants from MEXT/JSPS KAKENHI (grant no. JP 20K08157).

Availability of data and materials

The datasets generated and/or analyzed during the present study could not be submitted to a public curated database as the informed consent obtained does not include unrestricted disclosure of sequencing data. Instead, these data are available from the corresponding author upon reasonable request.

Authors' contributions

HI and AS wrote the manuscript. HI, MA, TM, MS and AS performed the genetic analysis and interpreted the results. HI, AI, RN, DK and AS performed patient care and collected the clinical data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by The Institutional Review Board of Okayama University Hospital (Okayama, Japan). Informed consent was obtained from the patients and/or their legal guardians.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Taga T, Tomizawa D, Takahashi H and Adachi S: Acute myeloid leukemia in children: Current status and future directions. Pediatr Int. 58:71–80. 2016.PubMed/NCBI View Article : Google Scholar

2 

Rasche M, Zimmermann M, Borschel L, Bourquin JP, Dworzak M, Klingebiel T, Lehrnbecher T, Creutzig U, Klusmann JH and Reinhardt D: Successes and challenges in the treatment of pediatric acute myeloid leukemia: A retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia. 32:2167–2177. 2018.PubMed/NCBI View Article : Google Scholar

3 

Pikman Y and Stegmaier K: Targeted therapy for fusion-driven high-risk acute leukemia. Blood. 132:1241–1247. 2018.PubMed/NCBI View Article : Google Scholar

4 

Dohner H, Weisdorf DJ and Bloomfield CD: Acute myeloid leukemia. N Engl J Med. 373:1136–1152. 2015.PubMed/NCBI View Article : Google Scholar

5 

Zwaan CM, Kolb EA, Reinhardt D, Abrahamsson J, Adachi S, Aplenc R, De Bont ES, De Moerloose B, Dworzak M, Gibson BE, et al: Collaborative efforts driving progress in pediatric acute myeloid leukemia. J Clin Oncol. 33:2949–2962. 2015.PubMed/NCBI View Article : Google Scholar

6 

Kolb EA and Meshinchi S: Acute myeloid leukemia in children and adolescents: Identification of new molecular targets brings promise of new therapies. Hematology. 2015:507–513. 2015.PubMed/NCBI View Article : Google Scholar

7 

Kayser S and Levis MJ: Advances in targeted therapy for acute myeloid leukaemia. Br J Haematol. 180:484–500. 2018.PubMed/NCBI View Article : Google Scholar

8 

Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS, Ritchey JK, Young MA, Lamprecht T, McLellan MD, et al: Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 481:506–510. 2012.PubMed/NCBI View Article : Google Scholar

9 

Wong TN, Ramsingh G, Young AL, Miller CA, Touma W, Welch JS, Lamprecht TL, Shen D, Hundal J, Fulton RS, et al: Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 518:552–555. 2015.PubMed/NCBI View Article : Google Scholar

10 

Quek L, Ferguson P, Metzner M, Ahmed I, Kennedy A, Garnett C, Jeffries S, Walter C, Piechocki K, Timbs A, et al: Mutational analysis of disease relapse in patients allografted for acute myeloid leukemia. Blood Adv. 1:193–204. 2016.PubMed/NCBI View Article : Google Scholar

11 

Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, et al: Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 374:2209–2221. 2016.PubMed/NCBI View Article : Google Scholar

12 

Greif PA, Hartmann L, Vosberg S, Stief SM, Mattes R, Hellmann I, Metzeler KH, Herold T, Bamopoulos SA, Kerbs P, et al: Evolution of cytogenetically normal acute myeloid leukemia during therapy and relapse: An exome sequencing study of 50 patients. Clin Cancer Res. 24:1716–1726. 2018.PubMed/NCBI View Article : Google Scholar

13 

Shiba N, Yoshida K, Shiraishi Y, Okuno Y, Yamato G, Hara Y, Nagata Y, Chiba K, Tanaka H, Terui K, et al: Whole-exome sequencing reveals the spectrum of gene mutations and the clonal evolution patterns in paediatric acute myeloid leukaemia. Br J Haematol. 175:476–489. 2016.PubMed/NCBI View Article : Google Scholar

14 

Bolouri H, Farrar JE, Triche T Jr, Ries RE, Lim EL, Alonzo TA, Ma Y, Moore R, Mungall AJ, Marra MA, et al: The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med. 24:103–112. 2018.PubMed/NCBI View Article : Google Scholar

15 

Tsukimoto I, Tawa A, Horibe K, Tabuchi K, Kigasawa H, Tsuchida M, Yabe H, Nakayama H, Kudo K, Kobayashi R, et al: Risk-stratified therapy and the intensive use of cytarabine improves the outcome in childhood acute myeloid leukemia: The AML99 trial from the Japanese childhood AML cooperative study group. J Clin Oncol. 27:4007–4013. 2009.PubMed/NCBI View Article : Google Scholar

16 

Tomizawa D, Tawa A, Watanabe T, Saito AM, Kudo K, Taga T, Iwamoto S, Shimada A, Terui K, Moritake H, et al: Excess treatment reduction including anthracyclines results in higher incidence of relapse in core binding factor acute myeloid leukemia in children. Leukemia. 27:2413–2416. 2013.PubMed/NCBI View Article : Google Scholar

17 

Hanada T, Kanamitsu K, Chayama K, Miyamura T, Kanazawa Y, Muraoka M, Washio K, Imada M, Kageyama M, Takeuchi A, et al: A Long-term survivor after congenital acute myeloid leukemia with t(8 ; 16)(p11 ; p13). Acta Med Okayama. 70:31–35. 2016.PubMed/NCBI View Article : Google Scholar

18 

Iwasaki Y, Nishiuchi R, Aoe M, Takahashi T, Watanabe H, Tokorotani C, Kikkawa K and himada A: Positive minimal residual disease of FLT3-ITD before hematopoietic stem cell transplantation resulted in a poor prognosis of an acute myeloid leukemia. Acta Med Okayama. 71:79–83. 2017.PubMed/NCBI View Article : Google Scholar

19 

Green RC, Berg JS, Grody WW, Kalia SS, Korf BR, Martin CL, McGuire AL, Nussbaum RL, O'Daniel JM, Ormond KE, et al: ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing. Genet Med. 15:565–574. 2013.PubMed/NCBI View Article : Google Scholar

20 

Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al: Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 17:405–423. 2015.PubMed/NCBI View Article : Google Scholar

21 

Kalia SS, Adelman K, Bale SJ, Chung WK, Eng C, Evans JP, Herman GE, Hufnagel SB, Klein TE, Korf BR, et al: Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): A policy statement of the american college of medical genetics and genomics. Genet Med. 19:249–255. 2017.PubMed/NCBI View Article : Google Scholar

22 

Ishida H, Kanamitsu K, Washio K, Muraoka M, Sakakibara K, Matsubara T, Kanzaki H and Shimada A: Relapsed infant MLL-rearranged acute lymphoblastic leukemia with additional genetic alterations. Pediatr Blood Cancer. 63:2059–2060. 2016.PubMed/NCBI View Article : Google Scholar

23 

Aoe M, Ishida H, Matsubara T, Karakawa S, Kawaguchi H, Fujiwara K, Kanamitsu K, Washio K, Okada K, Shibakura M and Shimada A: Simultaneous detection of ABL1 mutation and IKZF1 deletion in Philadelphia chromosome-positive acute lymphoblastic leukemia using a customized target enrichment system panel. Int J Lab Hematol. 40:427–436. 2018.PubMed/NCBI View Article : Google Scholar

24 

Ishida H, Iguchi A, Aoe M, Takahashi T, Tamefusa K, Kanamitsu K, Fujiwara K, Washio K, Matsubara T, Tsukahara H, et al: Panel-based next-generation sequencing identifies prognostic and actionable genes in childhood acute lymphoblastic leukemia and is suitable for clinical sequencing. Ann Hematol. 98:657–668. 2019.PubMed/NCBI View Article : Google Scholar

25 

Sunami K, Ichikawa H, Kubo T, Kato M, Fujiwara Y, Shimomura A, Koyama T, Kakishima H, Kitami M, Matsushita H, et al: Feasibility and utility of a panel testing for 114 cancer-associated genes in a clinical setting: A hospital-based study. Cancer Sci. 110:1480–1490. 2019.PubMed/NCBI View Article : Google Scholar

26 

Nakayama H, Tabuchi K, Tawa A, Tsukimoto I, Tsuchida M, Morimoto A, Yabe H, Horibe K, Hanada R, Imaizumi M, et al: Outcome of children with relapsed acute myeloid leukemia following initial therapy under the AML99 protocol. Int J Hematol. 100:171–179. 2014.PubMed/NCBI View Article : Google Scholar

27 

Duployez N, Marceau-Renaut A, Boissel N, Petit A, Bucci M, Geffroy S, Lapillonne H, Renneville A, Ragu C, Figeac M, et al: Comprehensive mutational profiling of core binding factor acute myeloid leukemia. Blood. 127:2451–2459. 2016.PubMed/NCBI View Article : Google Scholar

28 

Faber ZJ, Chen X, Gedman AL, Boggs K, Cheng J, Ma J, Radtke I, Chao JR, Walsh MP, Song G, et al: The genomic landscape of core-binding factor acute myeloid leukemias. Nat Genet. 48:1551–1556. 2016.PubMed/NCBI View Article : Google Scholar

29 

de Rooij JDE, Branstetter C, Ma J, Li Y, Walsh MP, Cheng J, Obulkasim A, Dang J, Easton J, Verboon LJ, et al: Pediatric non-Down syndrome acute megakaryoblastic leukemia is characterized by distinct genomic subsets with varying outcomes. Nat Genet. 49:451–456. 2017.PubMed/NCBI View Article : Google Scholar

30 

Alexander TB, Gu Z, Iacobucci I, Dickerson K, Choi JK, Xu B, Payne-Turner D, Yoshihara H, Loh ML, Horan J, et al: The genetic basis and cell of origin of mixed phenotype acute leukaemia. Nature. 562:373–379. 2018.PubMed/NCBI View Article : Google Scholar

31 

Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, et al: Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 129:424–447. 2017.PubMed/NCBI View Article : Google Scholar

32 

Marceau-Renaut A, Duployez N, Ducourneau B, Labopin M, Petit A, Rousseau A, Geffroy S, Bucci M, Cuccuini W, Fenneteau O, et al: Molecular profiling defines distinct prognostic subgroups in childhood AML. Hemasphere. 2(e31)2018.PubMed/NCBI View Article : Google Scholar

33 

Shiba N, Yoshida K, Hara Y, Yamato G, Shiraishi Y, Matsuo H, Okuno Y, Chiba K, Tanaka H, Kaburagi T, et al: Transcriptome analysis offers a comprehensive illustration of the genetic background of pediatric acute myeloid leukemia. Blood Adv. 3:3157–3169. 2019.PubMed/NCBI View Article : Google Scholar

34 

Meshinchi S, Alonzo TA, Stirewalt DL, Zwaan M, Zimmerman M, Reinhardt D, Kaspers GJ, Heerema NA, Gerbing R, Lange BJ and Radich JP: Clinical implications of FLT3 mutations in pediatric AML. Blood. 108:3654–3661. 2006.PubMed/NCBI View Article : Google Scholar

35 

Shimada A, Taki T, Tabuchi K, Taketani T, Hanada R, Tawa A, Tsuchida M, Horibe K, Tsukimoto I and Hayashi Y: Tandem duplications of MLL and FLT3 are correlated with poor prognoses in pediatric acute myeloid leukemia: A study of the Japanese childhood AML cooperative study group. Pediatr Blood Cancer. 50:264–269. 2008.PubMed/NCBI View Article : Google Scholar

36 

Ivey A, Hills RK, Simpson MA, Jovanovic JV, Gilkes A, Grech A, Patel Y, Bhudia N, Farah H, Mason J, et al: Assessment of minimal residual disease in Standard-risk AML. N Engl J Med. 374:422–433. 2016.PubMed/NCBI View Article : Google Scholar

37 

Morita K, Kantarjian HM, Wang F, Yan Y, Bueso-Ramos C, Sasaki K, Issa GC, Wang S, Jorgensen J, Song X, et al: Clearance of somatic mutations at remission and the risk of relapse in acute myeloid leukemia. J Clin Oncol. 36:1788–1797. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2020
Volume 13 Issue 5

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ishida H, Iguchi A, Aoe M, Nishiuchi R, Matsubara T, Keino D, Sanada M and Shimada A: Panel‑based next‑generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings. Biomed Rep 13: 46, 2020
APA
Ishida, H., Iguchi, A., Aoe, M., Nishiuchi, R., Matsubara, T., Keino, D. ... Shimada, A. (2020). Panel‑based next‑generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings. Biomedical Reports, 13, 46. https://doi.org/10.3892/br.2020.1353
MLA
Ishida, H., Iguchi, A., Aoe, M., Nishiuchi, R., Matsubara, T., Keino, D., Sanada, M., Shimada, A."Panel‑based next‑generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings". Biomedical Reports 13.5 (2020): 46.
Chicago
Ishida, H., Iguchi, A., Aoe, M., Nishiuchi, R., Matsubara, T., Keino, D., Sanada, M., Shimada, A."Panel‑based next‑generation sequencing facilitates the characterization of childhood acute myeloid leukemia in clinical settings". Biomedical Reports 13, no. 5 (2020): 46. https://doi.org/10.3892/br.2020.1353