GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood Advances, American Society of Hematology, Vol. 5, No. 23 ( 2021-12-14), p. 5420-5428
    Abstract: The effect of genetic variation on second malignant neoplasms (SMNs) remains unclear. First, we identified the pathogenic germline variants in cancer-predisposing genes among 15 children with SMNs after childhood leukemia/lymphoma using whole-exome sequencing. Because the prevalence was low, we focused on the association between SMNs and NUDT15 in primary acute lymphoblastic leukemia (ALL) cases. NUDT15 is one of the 6-mercaptopurine (6-MP) metabolic genes, and its variants are common in East Asian individuals. The prevalence of NUDT15 hypomorphic variants was higher in patients with SMNs (n = 14; 42.9%) than in the general population in the gnomAD database (19.7%; P = .042). In the validation study with a cohort of 438 unselected patients with ALL, the cumulative incidence of SMNs was significantly higher among those with (3.0%; 95% confidence interval [CI], 0.6% to 9.4%) than among those without NUDT15 variants (0.3%; 95% CI, 0.0% to 1.5%; P = .045). The 6-MP dose administered to patients with ALL with a NUDT15 variant was higher than that given to those without SMNs (P = .045). The 6-MP–related mutational signature was observed in SMN specimens after 6-MP exposure. In cells exposed to 6-MP, a higher level of 6-MP induced DNA damage in NUDT15-knockdown induced pluripotent stem cells. Our study indicates that NUDT15 variants may confer a risk of SMNs after treatment with 6-MP in patients with ALL.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2021
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 22, No. Supplement_3 ( 2020-12-04), p. iii277-iii278
    Abstract: Genetic hallmark of atypical teratoid/rhabdoid tumor (AT/RT) is loss-of-function variants or deletions in SMARCB1 gene on 22q11.2 chromosome, which is common to extracranial malignant rhabdoid tumors (MRT). Previous studies demonstrated that approximately one-thirds of AT/RT and extracranial MRT patients harbored germline SMARCB1 variants as the rhabdoid tumor predisposing syndrome. We studied herein intensive analysis of the SMARCB1 gene in AT/RT and extracranial MRT patients focusing on prevalence of germline genetic variants. PROCEDURE: In total, 16 patients were included. Both tumor-derived DNA and germline DNA were obtained from all patients. First, screening for SMARCB1 alterations in the tumor specimens was done by direct sequencing, ddPCR and SNP array analysis. Then, analysis of germline DNA samples focusing on the genomic abnormalities detected in the paired tumors in each case was performed. RESULTS In eight of 16 cases (50%), genomic alterations observed in the tumor-derived DNA were also detected in the germline DNA. It is worth noting that three patients had germline mosaicism. Two of three patients had mosaic deletion, including SMARCB1 region, and the average copy number of the deleted region in the SMARCB1 gene in the germline was 1.60 and 1.76. For another patient, the fraction of SMARCB1 variants in normal cells was as low as 1.7%. CONCLUSIONS Approximately half the MRT cases in this study had SMARCB1 germline alterations. Considering the presence of low-frequency mosaicisms which conventional methods might overlook, inherited germline variants in predisposition genes are more important than previously assumed for the pathogenesis of pediatric cancers.
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2020
    detail.hit.zdb_id: 2094060-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 1530-1530
    Abstract: Introduction Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous disease caused by various genetic alterations. Some prognosis-associated chromosomal aberrations and gene mutations such as t(8;21), inv(16), monosomy 7, and FLT3-ITD have been adopted for risk stratification. Although treatment outcomes have improved via stratification therapy, relapse and mortality are still observed in 40% and 30% patients, respectively. Patients with an intermediate risk with no favorable or recurrent factors are considered to exhibit varied biology and outcomes. Further studies are warranted to evaluate the accuracy of prognosis in these patients. Methods Among 369 patients with de novo AML participated in the Japanese AML-05 study conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group during 2006-2010, 77 patients including 59 with normal karyotype-AML (NK-AML) and 18 with complex karyotype-AML (CK-AML) were enrolled. Targeted sequencing was performed using a 343-gene custom panel and next-generation sequencer. Reportedly, these 343 genes are associated with hematopoietic malignancy or solid tumor pathogenesis. Correlations among gene mutations, other cytogenetic alterations, and clinical characteristics were investigated. Results In all, 187 mutations in 61 genes (average: 2.42 mutations/patient) were detected, and 72 patients (93%) had at least one genetic mutation. Among patients with NK-AML, 51 (86%) had one of the following driver mutations: FLT3-ITD, KMT2A-PTD, CEBPA, or NPM. Interestingly, internal tandem duplication (ITD) of BCOR (BCOR-ITD) was detected along with several novel ITDs in patients with unclear AML pathogenesis. Among patients with CK-AML, the following mutations regarding myelodysplastic syndrome (MDS) pathogenesis were detected: TP53: 3 (17%), JAK2: 2 (11%), ASXL1: 2 (11%), U2AF1: 1 (7%), SF3A1: 1 (7%), RUNX1: 2 (11%), and BCOR/BCORL1: 3 (17%). Consequently, 15 of 18 (83%) patients with CK-AML had some genetic mutations related to MDS. Eight types of transcription factor mutations and five of epigenetic factor mutations were detected in 10 patients. Eight of these (80%) relapsed or died. Three RUNX1 rearrangements (RUNX1-CBFA2T2, RUNX1-CBFA2T3, and RUNX1-FNBP1) and other fusions (PICALM-MLL10 and MYB-GATA1) were detected in patients with CK-AML. Interestingly, these patients had a low transcription factor or epigenetic factor mutation number; all of them survived without relapse. Discussion We detected several novel ITDs other than FLT3-ITD in patients with NK-AML. KIT-ITD was reported in adult and pediatric patients with AML; BCOR-ITD was linked to the pathogenesis of pediatric clear-cell sarcoma of the kidney. However, the clinical significance of ITDs, other than FLT3-ITD, has not been revealed in AML, for which further studies are being planned. This study identified the characteristic genetic background (i.e., MDS) in patients with CK-AML. Most patients with CK-AML (17/18; 94%) were diagnosed as AML with MDS related changes, according to World Health Organization classification. Fifteen of those (88%) had genetic mutations related to MDS pathogenesis. Furthermore, several RUNX1 rearrangements were detected in patients with CK-AML. Reportedly, RUNX1-CBFA2T2 and RUNX1-CBFA2T3 are recurrent fusions in adult AML. Particularly, RUNX1-CBFA2T3 has a gene expression profile similar to that of RUNX1-RUNX1T1, which may explain the favorable outcome in patients with such rearrangements. Despite the small sample size of this study, the findings indicate two major subgroups of pediatric CK-AML: 1) CK-AML with MDS-related genetic mutations (i.e., transcription factor and epigenetic factor mutations) linked to poor outcomes and 2) CK-AML with few of these mutations and with fusion genes (e.g., RUNX1) linked to favorable outcomes. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2850-2850
    Abstract: Background: Pediatric acute myeloid leukemia (AML) comprises approximately 20% of pediatric leukemia, representing one of the major therapeutic challenges in pediatric oncology with a current overall survival rate of less than 70%. The pathogenesis of AML is heterogeneous and can be caused by various chromosomal aberrations, gene mutations/epigenetic modifications, and deregulated/overregulated gene expressions, leading to increased proliferation and decreased hematopoietic progenitor cell differentiation. Recurrent chromosomal structural aberrations [e.g., t(8;21), inv(16), and MLL-rearrangements] have been well established as diagnostic and prognostic markers of AML. Furthermore, recurrent mutations in FLT3, KIT, NPM1, and CEBPA have been reported in both adult and pediatric AML. Recently, massively parallel sequencing enabled the discovery of recurrent mutations in DNMT3A, TET2, and IDH, which are clinically useful for the prediction of the prognosis. However, these mutations are rare in pediatric AML, suggesting that other genetic alterations exist in pediatric AML. In contrast, recent reports have described NUP98-NSD1 fusion as an adverse AML prognostic marker and PRDM16 (also known as MEL1) as the representative overexpressed gene in patients harboring NUP98-NSD1 fusion. Intriguingly, PRDM16 overexpression occurs in nearly one-quarter of all children, with AML involving NUP98-NSD1-negative patients. Moreover, this overexpression is enriched in specimens with other high-risk lesions (e.g., FLT3-ITD, NUP98-NSD1, and MLL-PTD). Patients and Methods: To reveal a complete registry of gene rearrangements and other genetic lesions in pediatric AML with a normal karyotype, we performed transcriptome analysis (RNA sequencing) of 61 of 70 de novo pediatric AML patients with a normal karyotype using Illumina HiSeq 2000. We could not perform RNA sequencing in nine patients because of a lack of RNA quantity or quality. Among the 70 AML patients with a normal karyotype, 33 patients overexpressed PRDM16, which was found to be strongly associated with a poor prognosis in our previous studies. All patients were enrolled and treated with AML-05 in the study conducted by the Japan Pediatric Leukemia/Lymphoma Study Group (JPLSG). We also analyzed the known genetic mutations associated with these patients using the data derived from RNA sequencing. Results: A total of 144 candidate gene rearrangements, which were not observed in normal samples, were identified in 51 of 61 samples. Many of the recurrent gene rearrangements identified in this study involved previously reported targets in AML, including NUP98-NSD1, NUP98-JARID1A, CBFA2T3-GLIS2, MLL-MLLT10, and MLL-MLLT3. However, several gene rearrangements were newly identified in the current study, including MLL-SEPT6, HOXA10-HOXA-AS3, PRDM16-SKI, and CUL1-EZH2. We have also performed the validation of these novel gene rearrangements using Sanger sequencing. Most of these gene rearrangements were found in patients with a high expressionof PRDM16. In contrast, CEBPA mutations were frequently observed in patients with a low expression of PRDM16. Known gene alterations, such as FLT3-ITD and MLL-PTD, and mutations of the RAS, KIT, CEBPA, WT1, and NPM1genes were also detected using RNA sequencing. Conclusion: RNA sequencing unmasked a complexity of gene rearrangements and mutations in pediatric AML genomes. Our results indicate that a subset of pediatric AML represents a discrete entity that could be discriminated from adult counterparts, regarding the spectrum of gene rearrangements and mutations. In the present study, we identified at least one potential gene rearrangement or driver mutation in nearly all AML samples, including some novel fusion genes. These findings suggest that gene rearrangements in conjunction with mutations also play essential roles in pediatric AML. Disclosures Ogawa: Kan research institute: Consultancy, Research Funding; Takeda Pharmaceuticals: Consultancy, Research Funding; Sumitomo Dainippon Pharma: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 9134-9135
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2727-2727
    Abstract: [Background] Customized gene panel sequencing is commonly used in cancer research to detect point mutations and small insertions or deletions. This technique can also be used to detect copy number alterations (CNAs) in tumor samples based on the comparison of sequence depth of targeted regions between samples and variant allele frequencies of germline single nucleotide polymorphisms (SNPs). CNAs in TP53 and CDKN2A/2B are frequently found in various types of leukemias. TP53 loss was reported to be a frequent CNA with poor prognosis in adult AML, and CDKN2A/2B alterations were found to be frequent in adult and pediatric acute lymphoblastic leukemia. However, the molecular and clinical profiles of the CNAs of these genes remain unclear in pediatric AML. [Patients and Methods] We analyzed clinical samples from 331 pediatric AML patients in the AML-05 trial, which was conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group. Targeted sequencing was performed using a 343-gene custom panel and next-generation sequencer. These 343 genes are reportedly associated with hematopoietic malignancy or solid tumor pathogenesis. Copy number analysis was performed using the in-house pipeline CNACS. Additionally, a SNP array was performed for 40 of the 331 patients, including 34 patients with complex karyotype, to detect CNAs of TP53. [Results] TP53 alterations were identified in 7 (2.1%) of 331 patients. TP53 losses and mutations were detected in 7 and 4 patients, respectively, and all the patients with TP53 mutations concurrently had TP53 losses. Seven patients with TP53 losses were identified by CNACS, although only 4 patients with TP53 losses were found by SNP array. As for the cytogenetics of 7 patients with TP53 losses, six carried a complex karyotype, lacking FAB-M7 morphology. Accordingly, 6 (28.6%) of 21 non-FAB-M7 patients with complex karyotype had TP53 losses. Fusion genes and gene mutations were rare in patients with TP53 losses (NUP98-KDM5A, n=1; KIT, n=1; NRAS, n=1; CEBPA, n=1; ASXL1, n=1), and no patients with core-binding factor AML (CBF-AML) or KMT2A-rearrangements had TP53 losses. In survival analyses, patients with TP53 losses had significantly worse overall survival, event-free survival, and cumulative incidence of relapse rates than patients with wildtype TP53 (14.3% vs 71.4%, p 〈 0.001, 0% vs 54.4%, p 〈 0.001, and 100% vs 43.1%, p 〈 0.001, respectively) (Figure). Among the 7 patients with TP53 losses, 2 had induction failure, and the remaining 5 relapsed. As a result, 6 patients died. CDKN2A/2B alterations were identified in 10 patients (3.0%) (9 losses and 1 mutation). Three patients with CDKN2A/2B losses had simultaneous TP53 alterations. Unlike TP53 alterations, CDKN2A/2B alterations were found in patients with CBF-AML and those with FAB-M7. In survival analyses, no significant difference was observed between patients with or without CDKN2A/2B alterations. [Discussion] TP53 losses were identified in approximately 2% of patients with pediatric AML. However, these alterations were found to be frequent in patients with complex karyotypes, particularly in those with non-FAB-M7 (28.6%). Furthermore, patients with TP53 losses had very poor prognosis. These findings indicate the necessity of risk stratification using TP53 status for pediatric AML. The efficacy of hematopoietic stem cell transplantation (HSCT) for the treatment of patients with TP53 losses remains unclear because 6 of 7 patients with TP53 losses received HSCT in this study. Moreover, because some of the identified TP53 mutations were possible germline mutations, HSCT may increase the incidence rate of the secondary cancer in patients with TP53 mutations. Thus, the efficacy of new agents (e.g., azacitizine and gemtuzumab ozogamicin) should be evaluated in future clinical trials for the intensification of chemotherapy for these patients. Meanwhile, the clinical and molecular profiles of CDKN2A/2B alterations were not fully identified in this study. However, the high incidence of CDKN2A/2B alterations in patients with TP53 losses indicates that the correlation between these gene alterations is essential for oncogenesis in a subset of pediatric AML patients. Figure Disclosures Ogawa: ChordiaTherapeutics, Inc.: Consultancy, Equity Ownership; Kan Research Laboratory, Inc.: Consultancy; RegCell Corporation: Equity Ownership; Asahi Genomics: Equity Ownership; Qiagen Corporation: Patents & Royalties; Dainippon-Sumitomo Pharmaceutical, Inc.: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 16 ( 2018-08-15), p. 4452-4458
    Abstract: Translocations of retinoic acid receptor-α (RARA), typically PML–RARA, are a genetic hallmark of acute promyelocytic leukemia (APL). However, because a small fraction of APL lack translocations of RARA, we focused here on APL cases without RARA translocation to elucidate the molecular etiology of RARA-negative APL. We performed whole-genome sequencing, PCR, and FISH for five APL cases without RARA translocations. Four of five RARA-negative APL cases had translocations involving retinoic acid receptor-β (RARB) translocations, and TBL1XR1–RARB was identified as an in-frame fusion in three cases; one case had an RARB rearrangement detected by FISH, although the partner gene could not be identified. When transduced in cell lines, TBL1XR1–RARB homodimerized and diminished transcriptional activity for the retinoic acid receptor pathway in a dominant-negative manner. TBL1XR1–RARB enhanced the replating capacity of mouse bone marrow cells and inhibited myeloid maturation of human cord blood cells as PML–RARA did. However, the response of APL with RARB translocation to retinoids was attenuated compared with that of PML–RARA, an observation in line with the clinical resistance of RARB-positive APL to ATRA. Our results demonstrate that the majority of RARA-negative APL have RARB translocations, thereby forming a novel, distinct subgroup of APL. TBL1XR1–RARB as an oncogenic protein exerts effects similar to those of PML–RARA, underpinning the importance of retinoic acid pathway alterations in the pathogenesis of APL. Significance: These findings report a novel and distinct genetic subtype of acute promyelocytic leukemia (APL) by illustrating that the majority of APL without RARA translocations harbor RARB translocations. Cancer Res; 78(16); 4452–8. ©2018 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Pediatric Blood & Cancer, Wiley, Vol. 64, No. 9 ( 2017-09)
    Abstract: Previous studies have reported several cases of juvenile myelomonocytic leukemia (JMML) developing blastic transformation during an indolent clinical course, but the underlying mechanism of transformation is still not well understood. In this report, we describe a case of JMML with blastic transformation possibly caused by additional copy number gains of the KRAS mutant allele. We have discovered that the copy number gain of the mutant allele is an additional possible cause of blastic transformation in JMML.
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2697-2697
    Abstract: Introduction Treatment outcomes of pediatric acute myeloid leukemia (AML) have improved via stratification therapy. However, relapse and mortality are still observed in 40% and 30% patients, respectively. Recently, comprehensive analyses using next-generation sequencing have revealed novel genetic alterations in adult AML patients, but insufficient analyses have been performed in pediatric AML patients. Further studies are warranted to evaluate the relationship between a more detailed genetic background and prognosis in pediatric AML patients. Methods We performed targeted sequencing using a 343-gene custom panel and next-generation sequencer in 302 pediatric patients with de novo AML who participated in the Japanese AML-05 study conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group from 2006 to 2010. One hundred and twenty patients with core binding factor (CBF)-AML, 52 with KMT2A rearrangements, 56 with normal karyotype (NK), 18 with complex karyotype (CK), and 57 with other karyotypes except for patients with acute megakaryocytic leukemia were enrolled. Correlations among gene mutations, other cytogenetic alterations, and clinical characteristics were investigated. Results and Discussion We found the following 6 recurrent gene mutations, which have not been frequently reported in pediatric AML: KMT2C (n = 13, 4.3%), PHF6 (n = 6, 2.0%), MGA (n = 13, 4.3%), TET2 (n = 11, 3.6%), JAK3 (n = 12, 4.0%), and GATA2 (n = 11, 3.6%). KMT2C mutations have been reported in adult and pediatric patients with CBF-AML and in a few patients with other types of AML; however, their clinical significance has not been revealed. Six of 13 patients with KMT2C mutations were found in CBF-AML, whereas the other 7 patients were detected in non CBF-AML (1 patient in KMT2A-rearrangement, 1 in CK, and 5 in other karyotypes). Loss of function mutations in KMT2C have also been reported to be associated with chemotherapy resistance in pediatric AML patients. Although no significant differences were found, 4 of 13 patients with KMT2C mutations did not achieve complete remission (p = .072), and 3 patients relapsed in 〈 12 months. Significant differences were observed in event-free survival (EFS) between patients with and without KMT2C mutations (3-year EFS 36.9% vs. 56.8%; p = .019). In adult AML, PHF6 mutations have been detected in 3% of patients and have been reported as poor prognostic factors. In 6 patients with PHF6 mutations, 3 patients had a normal karyotype, and 2 patients had a complex karyotype. No PHF6 mutations were detected in AML patients with CBF or KMT2A rearrangements. PHF6 mutations were significantly frequently detected in AML patients with myelodysplasia-related changes (4/6, p = .008). Although 5 of 6 patients with PHF6 mutations archived complete remission, finally, 5 patients relapsed or died. There were significant differences in EFS between patients with or without PHF6 mutations (3-year EFS 16.7% vs. 58.2%; p = .041). MGA mutations have been reported thus far in a few AML patients. MGA is a negative regulator of MYC signaling; thus, loss of functions of MGA is considered to contribute to tumorigenesis. However, the clinical significance of MGA mutations has not been reported. MGA mutations were significantly frequently detected in AML patients with FAB M2 (7/13, p = .020). In our cohort, 2 of 3 AML patients with DEK-NUP214 had MGA mutations (p = .004). We did not find correlations between MGA mutations and prognosis (3-year OS 83.3% vs. 75.5%; p= .488, 3-year EFS 66.7% vs. 56.7%; p = .507) TET2 mutations have been found in 8%-19% of adult AML cases and are considered poor prognostic factors. In previous reports, TET2 mutations were also detected in 3.8% of pediatric AML patients, but because of the small number of patients, the association between TET2 mutations and prognosis has not been revealed. TET2 mutations were significantly frequently detected in older AML patients (median age 13.95 vs. 9.0 years; p = .022) and in AML patients with NK (5/11, p = .035). Contrary to previous reports on adult AML, TET2 mutations were not associated with prognosis (3-year OS 77.9% vs. 75.9%; p= .688, 3-year EFS 54.5% vs. 57.4%; p = .907). This suggested that TET2 mutations have a different clinical significance between adult and pediatric AML patients. In conclusion, mutations in the KMT2C and PHF6 genes were associated with poor prognosis, but TET2 mutations were not. Further analysis should be needed about TET2 mutations. Disclosures Ogawa: ChordiaTherapeutics, Inc.: Consultancy, Equity Ownership; Kan Research Laboratory, Inc.: Consultancy; RegCell Corporation: Equity Ownership; Asahi Genomics: Equity Ownership; Qiagen Corporation: Patents & Royalties; Dainippon-Sumitomo Pharmaceutical, Inc.: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 1024-1024
    Abstract: Background; Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous disease. Currently, a number of gene mutations have been implicated in the pathogenesis of both adult and pediatric AML, including mutations of CEBPA,NPM1, DNMT3A, IDH1/2, TET2 and EZH2 in addition to RAS, KIT and FLT3, because the recent development of massively parallel sequencing technologies. We have performed whole-exome sequencing of paired tumor-normal DNA from 19 patients, and identified 80 somatic mutations or 4.2 mutations per sample. Many of the recurrent mutations identified in this study involved previously reported targets in adult AML, such as FLT3, CEBPA, KIT, CBL, NRAS, WT1, BCORL1, EZH2, and major cohesin components including SMC3 and RAD21. In addition to these mutations, we also identified disease-associated candidate genes of ASXL2, PAX5 and others. Recently, recurrent somatic mutations in ASXL1 have been reported to occur in patients with adult AML, and to be associated with adverse outcome. Another study suggested that ASXL1 or ASXL2 mutations were associated with a high incidence of relapse. To reveal the significance of these mutations, we performed mutational analysis of ASXL1 and ASXL2 in 184 pediatric AML patients. Methods; Between 2006 and 2010, 485 de novo pediatric AML patients aged 〈 18 years old participated in the Japanese AML-05 study conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG). Among them, 369 samples were available. We analyzed the first half of these samples which were registered in the order (184 samples). To estimate the frequency and prognostic impact of the ASXL1 and ASXL2 mutations in pediatric AML, we performed targeted sequencing of ASXL1 (exon 12) and ASXL2 (exon 12) genes using next-generation sequencer in 184 de novo AML patients including 51 patients with t(8;21). We validated the mutations in ASXL1 and ASXL2 by Sanger sequencing. Furthermore, we investigated the correlation among these mutations, other cytogenetic alterations and clinical characteristics. Results and Discussion; ASXL1 mutations were identified in 4 of 184 de novo pediatric AML patients (2.2 %) and all 4 ASXL1 mutation positive patients harbored t(8;21). Two of them relapsed, and one died after relapsed. On the other hand, ASXL2 mutations were identified in 10 of 184 de novo pediatric AML patients (5.4%) and 6 of them harbored t(8;21). Five of these 10 patients relapsed, and 2 of them died after relapsed. Especially, all of 4 patients without t(8;21) relapsed (2 in M5a and 2 with CBFA2T3-GLIS2 in M7), and 2 died after relapsed. Although only one ASXL2 patients with t(8;21) relapsed (1/6 or 17%), no ASXL2 positive patients with t(8;21) died. ASXL2 mutations were more observed in AML patients with t(8;21) (11.8%, 6/51), but not in 13 patients with inv(16). Overall survival of the patients with or without ASXL2 mutations were 80% and 66.7% (p=0.54), respectively. ASXL1 and ASXL2 mutations were mutually exclusive in this study. Conclusion; Ten of 184 patients (5.4%) had mutations of ASXL2 in pediatric AML, and the outcome of ASXL2 mutant patients with t(8;21) was favorable. Among the 51 pediatric AML patients with t(8;21), ASXL2 mutations were detected in 6 (11.8%) patients. All of them have been survived, suggesting that ASXL2 mutations in patients with t(8;21) may be associated with favorable prognosis in pediatric AML in contrast to adult AML. On the other hand, 4 (2.2%) of 184 patients had ASXL1 mutations, and all of them were t(8;21). In these 4 patients, 2 of them relapsed, and one died after relapsed. Although the number of patients is too small, ASXL1 mutations were not considered to be associated with favorable outcome. Both ASXL1 and ASXL2 mutations were detected at high frequency among pediatric AML patients with t(8;21) and mutual exclusive. As we consider that further study will be needed to clarify the significance of these mutations, we are now analyzing mutations in other exons of ASXL2, and would like to report these data in the annual meeting. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...