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, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 41-41
    Abstract: While there have been extensive studies to define the roles of recurrent somatic mutations in AML, the contribution of germline variants to AML initiation and progression is less well established. DNA repair disorders often predispose patients to developing myeloid malignancies. In particular, biallelic mutations affecting FANC genes cause the recessive heritable bone marrow failure syndrome Fanconi Anemia (FA), which is associated with 〉 800-fold increased risk of progression to AML. A recent explosion of cancer predisposition studies has also revealed the importance of germlineFANC variants in elevated cancer risk (Cancer Treat Rev 2012; 38:89). To investigate the role of FANC gene variants in AML we have performed a case-control study, analyzing rare, deleterious somatic and germline variants for the 19 FANC genes in adult AML and healthy controls cohorts. Whole exome sequencing was performed on diagnosis samples from 131 adult Caucasian AML patients from two major Australian centers, and a cohort of 329 healthy females. We identified rare Tier 1 variants using a minor allele frequency (MAF) 〈 0.001, as reported in common dbSNP137, 1000 Genome and NHLBI-ESP project databases. Combined Annotation Dependent Depletion algorithm (CADD, Nat Genet 2014; 46: 310) 〉 10 was used to filter for FANC gene variants with high probability of pathogenicity. Sanger sequencing of matched tumour/non-tumour DNA showed the large majority of variants tested to be germline (90%), consistent with previous studies reporting that somatic FANC genes variants are extremely rare in AML ( 〈 1%). Overall, we identified 52 FANC gene variants in 44 cases with 34% of AML cases carrying one or more variant. For independent validation we determined the presence of somatic and germline FANC variants in the TCGA AML cohort using an identical pipeline and filtering analysis. In line with our results, we found that 36% of TCGA AML patients carry at least one germline FANC variant. We investigated known disease-causing (D-C) variants in these two AML cohorts using the FA (FAMutdb) and breast cancer (kConFab and BIC) mutation databases. We found 8 D-C FANC variants in the Australian AML cohort and 5 in the TCGA cohort, with 1 variant present in both cohorts. Moreover, the frequency of D-C variants in our cohort of females with AML (n=51) is 13.7%, while the frequency in the healthy female cohort is 4.5%, comparable to that reported in the ESP database for female European-Americans (2.1%, Hum Mol Genet 2014; 23: 6815). Accordingly, we determined that deleterious FANC germline variants confer a significant increased risk of AML (P=0.018, OR=3.3 for the Australian AML cohort). Finally, we performed mutational burden analysis to investigate enrichment of variants associated with particular FANC genes across the AML cohort. This revealed a significant enrichment of FANCL variants in AML vs healthy controls (P=0.008, Figure 1). FANCL is the enzymatic component of the FA core complex that monoubiquitinates the FANCD2/I heterodimer initiating DNA repair, and its down-regulation has been linked to AML (Oncogene 2016; doi:10.1038). Several FANCL variants, found in our AML cohort, affect the catalytic RING domain and are of particular interest. These include a D-C null variant present in 2 patients, a frame shift variant in 2 patients who presented with AML at a very early age (27 and 46 years old), and a variant affecting a critical conserved residue required for monoubiquitination of FANCD2/I. In conclusion, we show enrichment of rare potentially deleterious FANC gene mutations in AML, associated with a 3-fold increased risk of developing the disease. We hypothesise that, in hematopoietic stem/progenitor cells, these variants confer a subtle defect in interstrand cross-link repair leading to an increased accumulation of mutations and subsequent development of AML. Consistent with this there have been several reports of defective DNA damage repair and increased sensitivity to DNA damaging agents in cells from FANC carriers compared to normal controls (Nat Commun 2014; 5:5496; Mutagenesis 2009; 24:67). Importantly, it is possible to target defects in several DNA repair pathways, and our finding identifies a group of AML patients who may benefit from approaches that target defective FA and homologous recombination pathways. Figure 1. A significant increase mutational burden of FANCL was observed in our AML cohort (line represents P=0.05). Figure 1. A significant increase mutational burden of FANCL was observed in our AML cohort (line represents P=0.05). Disclosures Gill: Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1684-1684
    Abstract: The role of metabolic dysfunction in the initiation and progression of AML is still to be determined. Next-generation sequencing studies in AML have identified somatic mutations in mitochondrially encoded subunits of the mitochondrial respiratory chain (MRC; N Engl J Med, 2013; 368:2059). Recurrent somatic mutations in nuclear MRC genes are yet to be reported, but recurrent founding mutations occur in nuclear genes encoding the TCA cycle enzymes Isocitrate Dehydrogenase 1 and 2 (IDH1, IDH2). IDH1 and IDH2 mutant AML samples display sensitivity to BCL-2 inhibition (Nat Med, 2015; 21:178), and reduced proliferative capacity has been observed for IDH1 mutant samples following glutaminase inhibition (Exp Hematol, 2014; 42:247). In glioma, IDH2 mutant tumors show enrichment of oxidative phosphorylation (OXPHOS) related gene sets (J Exp Clin Cancer Res, 2016;35:86). Additionally, germline succinate dehydrogenase (MRC Complex-II) mutations have been associated with hereditary paragangliomas (Science, 2000; 287:848). We therefore investigated the nature and frequency of rare germline and somatic mutations affecting nuclear MRC genes in an adult AML cohort, and profiled the metabolic phenotypes of primary BM AML samples. Whole-exome sequencing was performed on 145 diagnosis (Dx) adult BM AML samples. Variants were filtered against dbSNP137, 1000 genome and NHLBI-ESP with a focus on rare germline/somatic variants with a minor allele frequency 〈 0.005. A total of 62 variants affecting the 39 nuclear MRC Complex-I (CI) genes were found in 52 samples. Across the MRC (85 nuclear genes), we identified 140 variants in 95 samples. Confirmation of the germline/somatic status of identified variants is ongoing, with 20 / 21 variants tested being germline in origin. Case-control burden analysis (Am J Hum Genet, 2013; 92:841) was performed to compare variants in the AML cohort with those in an ethnically matched healthy control cohort (n = 329; PLoS Genet, 2011; 7:e1001372). This showed enrichment in the AML cases for variants across all 5 MRC complexes (p = 0.04; Burden analysis, Bonferroni adjusted), and for variants affecting the β-complex of CI (14 genes; p = 0.01; Burden analysis, Bonferroni adjusted). We further investigated the association of CI variants with genes commonly mutated in AML. This revealed a significant under-representation of IDH1 mutations in AML with CI variants (p = 0.036; Fisher's exact; Figure 1). Additionally, for a panel of AML samples (n = 117), we used the BioMark (Fluidigm) qRT-PCR platform to profile the expression of all nuclear CI genes and a panel of AML oncogenes and tumor suppressors. This revealed a significant negative correlation (p 〈 0.05; Pearson's correlation, Bonferroni adjusted) between the expression of all nuclear CI genes and that of IDH2, which is independent of underlying mutation or genetic alterations, and which is not observed for IDH1 expression. Finally, Seahorse metabolic profiling showed that IDH1 mutant samples were found to be utilizing their glycolytic pathway at maximal capacity with no glycolytic reserve while simultaneouslydisplaying significantly reduced glycolytic rates (p 〈 0.05; T-test, Bonferroni adjusted) when compared to IDH2 mutant AML samples, AML samples lacking recurrent AML mutations (WT-AML), and normal bone marrow (NBM) controls. Both IDH1 and IDH2 mutant samples showed significantly deficient OXPHOS reserve capacity when compared to NBM and WT-AML samples (p 〈 0.05; T-test, Bonferroni adjusted). Ongoing investigations will determine the metabolic phenotype of CI mutant samples. In summary, we observed significant enrichment of rare variants affecting the MRC and CI genes in an adult AML cohort compared to healthy controls, highlighting a potential role for altered cellular energetics in AML pre-disposition. The significant under-representation of somatic IDH1 mutations in AML with rare germline/somatic CI mutations raises the possibility that these CI variants induce functional consequences that mimic those associated with somatic IDH1 mutation. The distinct glycolytic profiles of IDH1 and IDH2 mutant samples, and the selective negative correlation of CI gene expression with IDH2 mRNA expression, suggests that distinct metabolic phenotypes maybe associated with perturbations to IDH1 and IDH2. Ongoing investigations will compare the metabolic phenotype of samples with CI variants to that of IDH1 and IDH2 mutant samples. Disclosures Gill: Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 3
    In: Blood Cancer Journal, Springer Science and Business Media LLC, Vol. 8, No. 6 ( 2018-06-01)
    Type of Medium: Online Resource
    ISSN: 2044-5385
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2600560-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 698-698
    Abstract: Introduction - AML is a complex group of malignancies, with heterogeneity in morphology, cytogenetics, molecular characteristics, aggressiveness and importantly, in its response to treatment and survival outcomes. Next generation sequencing by the Cancer Genome Atlas Research Network analysed 200 primary AML cases and identified 23 genes that display recurrent somatic mutations at varying frequency in AML (NEJM 368(22):2059-2074). Defects in DNA repair are frequently identified in treatment-related AML and inherited mutations in genes of DNA repair pathways predispose patients to myeloid malignancies. For example, biallelic mutations in FANC genes, which cause the recessive heritable bone marrow failure syndrome Fanconi Anaemia (FA) are associated with high risk of progression to AML and other cancers (Kutler et al.Blood, 101:1249-1256), suggesting a potential involvement of FANC gene mutations in AML pathogenesis. Methods - In this study we present a two-stage approach to gene discovery in AML: initial unbiased whole genome sequence (WGS) and whole exome sequence (WES) analysis of tumour DNA from a cytogenetically normal AML case at diagnosis and relapse, and corresponding germ-line DNA (prepared from mesenchymal stromal cells). Potential oncogenic mutations and changes associated with disease progression were identified. WES of a further 96 diagnostic AML samples further defined recurrent mutations and allowed identification of affected functional groups and networks in AML. Results – WGS and WES were performed on diagnosis, non-haematopoietic and relapse samples from an index AML patient. Somatic SNVs and indels unique to the tumour samples include a number of variants in genes previously reported as recurrently somatically mutated in AML including FLT3, WT1 and IDH2. Somatic mutations in genes not previously associated with AML were also identified including a mutation in FANCD2 (p.S1412N) present in the index AML tumour DNA at diagnosis and at relapse. Variants in genes recurrently mutated at low frequency in AML can also be disease drivers, however separating such genes from the background level of mutation in AML requires analysis across multiple samples, and sequencing studies to determine recurrence and/or mutations in proteins involved in the same functional pathway or complex. STRING-db v9.05 (Franceschini et al. NAR, 2013(41), Database issue) was used to identify a larger network of proteins, including and associated with the FANC genes, involved in homologous recombination-mediated DNA repair. Known somatic mutations from other AML studies were mapped onto this network; as shown in Figure 1 multiple genes in this extended network are affected by somatic mutation in AML suggesting a potential role in pathogenesis. Analysis of our WES data from diagnosis samples from a further 96 Australian AML cases identified an additional two somatic mutations in genes from the extended STRING-db v9.05 FANC network. In total we identified 18 mutations in the 16 classified FANC genes and 8 variants in the BLM complex as shown in Figure 2. Two of the germline FANC gene mutations, FANCM-Q13333fs and FANCD2-R926X, are known pathogenic mutations in FA. Patients with mutations in the 8 FANC genes of the core complex form a distinct subset from those with mutations in the other 8 FANC genes. 5 of the 8 patients with mutations in the BLM complex also form a separate group while BLM complex mutations are present in 2 patients that also have FANC mutations. For the two patients with acquired changes the allele frequency for these FANC mutations is greater than 25% suggesting an early origin in disease. Discussion. Our findings suggest that germline and somatic mutations affecting function of the FANC DNA repair pathway may be a recurrent abnormality in AML, potentially contributing to leukaemogenesis. FANC/BLM gene mutations frequently co-exist with mutations in DNMT3A and DNMT1; 46% of the patients with DNMT3A/DNMT1 mutations are also mutant for FANC or BLM complex genes representing significant over-representation (p = 0.021). Within the group of FANC and BLM patients there is also significant under-representation of FLT3-ITD mutations and mutations in N-RAS and K-RAS (p = 0.051), raising the possibility that defects in homologous DNA repair may favour cooperation with alternative signalling pathways. Figure 1 Figure 1. Figure 2 Figure 2. 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...