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
Filter
  • Medicine  (93)
  • 1
    In: The American Journal of Human Genetics, Elsevier BV, Vol. 91, No. 3 ( 2012-09), p. 422-434
    Type of Medium: Online Resource
    ISSN: 0002-9297
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2012
    detail.hit.zdb_id: 1473813-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: The American Journal of Human Genetics, Elsevier BV, Vol. 91, No. 3 ( 2012-09), p. 408-421
    Type of Medium: Online Resource
    ISSN: 0002-9297
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2012
    detail.hit.zdb_id: 1473813-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: The American Journal of Human Genetics, Elsevier BV, Vol. 88, No. 4 ( 2011-04), p. 458-468
    Type of Medium: Online Resource
    ISSN: 0002-9297
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2011
    detail.hit.zdb_id: 1473813-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cell, Elsevier BV, Vol. 182, No. 2 ( 2020-07), p. 417-428.e13
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2020
    detail.hit.zdb_id: 187009-9
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 42-42
    Abstract: Both genome wide association studies (GWAS) of common variation and exome wide association studies (EXWAS) of rare variation have successfully identified disease susceptibility variants for a variety of diseases. One GWAS of inherited susceptibility to Acute Myeloid Leukemia (AML) has been conducted, but no EXWAS have been performed to measure risk of AML attributable to low-frequency constitutional genetic variation. We performed the first EXWAS of risk of AML as a nested case-control study in the DISCOVeRY-BMT (Determining the Influence of Susceptibility Conveying Variants Related to one-Year mortality after BMT) cohorts. The DISCOVeRY-BMT parent study examined transplant-related mortality in leukemia patients undergoing unrelated donor allogeneic BMT. To identify low frequency variants and genes contributing to increased susceptibility to AML we used genotype data from the Illumina HumanExome BeadChip typed in the DISCOVeRY-BMT cohorts; the HumanExome BeadChip contains 242,901 variants, which are mainly protein-coding variants. The optimal sequence kernel association test (SKAT-O) was used to analyze gene-level associations with risk of AML. These gene-based tests evaluate the cumulative effects of multiple single gene variants on risk of AML. Analyses were performed in all European American AML cases and two subtypes: 1) de novo AML, 2) de novo AML with normal cytogenetics. Models were adjusted for age at transplant and principal components to control for population stratification. For gene-based tests at least 2 variants with minor allele frequency (MAF) ≤ 5%, were required to be present in the gene. This yielded a total of 13,687 genes tested, and a Bonferroni corrected significance level of P 〈 3.65 x 10-6. Association tests were performed in 1,189 AML cases reported to CIBMTR 2000-08 (Cohort 1) and 327 AML cases reported to CIBMTR from 2009-11 (Cohort 2). Controls in Cohorts 1 (n=1,986) and 2 (n= 515) were 10/10 HLA-matched unrelated donors who passed a comprehensive medical exam and deemed healthy. We used metaSKAT to combine Cohorts 1 and 2 and obtain p-values of association with AML. We present the results of gene-level tests significant in both cohorts. The likely pathogenicity of these variants was determined in silico using SIFT, PolyPhen and MutationTaster. Patient characteristics are in Table 1. DNMT3A, on chromosome 2, was associated in the gene-based test with risk of AML (Pmeta=1.70x10-9, Table 2). Three missense variants at MAF 〈 1% comprise both overall AML and de novo AML gene-based association: exm177559 (Asn- 〉 Ser), exm177507 (Arg- 〉 His), and exm177543 (Arg- 〉 Trp). Normal cytogenetics de novo AML gene-based assocations consisted of only 2 of these variants: exm177559 and exm177507 (Table 2). While prevalence of exm177507 is 〈 1% for all AML cases, in de novo AML with normal cytogenetics the MAF was higher at 3%. The other 2 variants had a MAF 〈 1% irrespective of subtype. Somatically, DNMT3A is most frequently mutated in hematologic malignancies, with 〉 30% of de novo AML cases with a normal karyotype and 〉 10% of MDS patients having DNMT3A mutations. Although these are germline gene associations all three of the variants found have been reported somatically in hematologic malignancies. In 200 AML cases from The Cancer Genome Atlas (TCGA) p.R882H (represented as exm177507 on the exome chip) was a frequent somatic mutation (25%). Exm177543 (p.R635W) and exm177559 (p.N501S) are reported in the Catalogue of Somatic Mutations in Cancer (COSMIC) as somatic mutations involved in hematopoietic and lymphoid tissue in both cell lines and humans. Exm177507 and exm177543 show evidence of pathogenicity in all three in silico tools, while exm177559 was reported as deleterious and disease causing by Sift and MutationTaster, respectively. Our results show that multiple potentially pathogenic missense germline variants in DNMT3A comprise the gene-based association with AML, specifically de novo AML with normal cytogenetics. Given the functional nature of these variants it is possible germline risk stratification could be informative in determining AML risk, and subsequently development of AML harboring DNMT3A mutations. Confirmation of these findings in additional cohorts could have implications for individualized risk screening, prediction and prognosis. Additional cytogenetic subgroup analyses, including treatment-related AML, are underway. Disclosures Hahn: Novartis: Equity Ownership; NIH: Research Funding. McCarthy:Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; The Binding Site: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Honoraria, Membership on an entity's Board of Directors or advisory committees; Gamida Cell: Honoraria, Membership on an entity's Board of Directors or advisory committees. Sucheston-Campbell:NIH/NCI: 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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 71-71
    Abstract: Over the past decade, there have been numerous published candidate gene association studies of non-HLA variants, genes and/or pathways in relation to BMT survival outcomes. Most of these studies investigated a small number of candidate single nucleotide polymorphisms (SNPs), chosen based on known gene function, in a limited number of cases without replication. Here we test the association of previously reported SNPs with survival outcomes (overall survival (OS), progression-free survival (PFS), death due to disease (DD) or transplant-related mortality (TRM)) after URD BMT using data from the DISCOVeRY-BMT (Determining the Influence of Susceptibility Conveying Variants Related to one-Year mortality after BMT) study which included 2,052 European descent recipient-unrelated donor pairs (Cohort 1) and 763 recipient-unrelated donor pairs (Cohort 1) reported to the Center for International Blood and Marrow Transplant Research (CIBMTR) between 2000-2009 and 2009-2011, respectively. DISCOVeRY-BMT is a Genome Wide Association Study (GWAS) that includes over 9 million typed or imputed SNPs. This represents the largest study to date and is well powered to test both SNP and gene-based association hypotheses. We conducted an extensive literature review of PubMed using MESH terms related to transplant and hematologic disease which yielded 76 candidate gene association studies that investigated a total of 46 genes with any survival outcome. Of the 46 genes, 11 genes (CTLA4, CYP2B6, CYP2C19, GSTM1, GSTT1, IL23R, IL6, MTHFR, NOD2/CARD15, TNFSRF1B, and VDR) were studied in at least 2 publications. A total of 16 SNPs in 8 of these genes reported a significant (P 〈 0.05) association with at least one survival outcome (OS, PFS, DD or TRM). We tested these 16 SNPs for an association with the survival outcome originally reported in each article (Table 1). All association models included age at BMT, diagnosis (AML, ALL, MDS), disease status at BMT, cell source (blood, bone marrow) and year of BMT. P-values from Cohorts 1 and 2 were combined using a fixed-effects model in METAL software. Since the candidate gene studies have a gene-based hypothesis, we also performed gene-level testing using the VErsatile Gene-Based Association Study (VEGAS2) software to measure the aggregate effect of all available typed and imputed SNPs from our GWAS data in these 8 genes. The candidate gene association studies included heterogeneous patient populations of autologous and/or allogeneic (related or unrelated donors) BMT for a variety of hematologic diseases. Most candidate gene studies performed genotyping on both donors and recipients, and SNPs were tested either independently for donor or recipient genomes, or considering the total number of variant alleles the recipient-donor pair had at a given locus (0-4) (Table 1). The 16 SNPs from the previous candidate gene association studies were not significantly associated at a nominal P 〈 0.05 to either the survival outcome reported in the original published paper, or to any survival outcome (Table 1). Likewise, the VEGAS2 gene-based analysis did not reveal any statistically significant associations for any of the 8 genes at P 〈 0.05. Our study also corroborated prior reports of SNPs and genes that were not associated with survival outcomes. Candidate gene association studies use a priori biological knowledge to select genes that are likely associated with outcome and subsequently SNPs within the gene thought likely to affect function based on knowledge at the time of the investigation. Our findings suggest that neither the genetic variation tested in these candidate genes, nor other individual SNPs in the gene independently or collectively are not associated with survival outcomes in recipients of URD BMT. Our failure to replicate the prior candidate gene studies may be due to these SNPs being dependent on the underlying disease (ie. AML, ALL, etc), the donor relation (related, unrelated, autologous), or that there is no true association of these SNPs with the outcomes studied. #These authors contributed equally Table 1. Table 1. Disclosures McCarthy: Gamida Cell: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Honoraria, Membership on an entity's Board of Directors or advisory committees; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; The Binding Site: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees. Hahn:Novartis: Equity Ownership; NIH: Research Funding. Sucheston-Campbell:NIH/NCI: 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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 131, No. 22 ( 2018-05-31), p. 2490-2499
    Abstract: We conducted the first exome-wide association study between germ line variant genotype and survival outcomes after unrelated-donor BMT. A number of novel genes were found to significantly affect survival outcomes.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 61-61
    Abstract: Transplant-related mortality (TRM) is the largest limiting factor to successful URD-BMT as curative therapy. Identification of non-HLA genetic factors in either recipients or donors could improve BMT outcomes through better matching at these loci. We performed a genome-wide association study (GWAS), named DISCOVeRY-BMT (Determining the Influence of Susceptibility COnveying Variants Related to one-Year mortality after BMT) of 1-year TRM in 3,532 patients treated for AML, ALL or MDS (recipients) reported to CIBMTR from 2000-2011 and their HLA-matched URD (donors); donor DNA was available for 98% of HCT recipients. Cohort 1 includes 2,240 donor-recipient pairs; patients received a 10/10 HLA URD-BMT from 2000-08. Cohort 2 includes 823 donor-recipient pairs; patients received either a 10/10 HLA URD-BMT from 2009-11 or 8/8 (but 〈 10/10) HLA URD-BMT. Genotyping of recipient and donor DNA was performed using the Illumina HumanOmniExpress-24 BeadChip containing 729,293 single nucleotide polymorphisms (SNPs) at University of Southern California. Due to the small number of non-European individuals, we report analyses of European American recipients only. After quality control, Cohort 1 includes 2,052 donor-recipient pairs and Cohort 2 includes 763 donor-recipient pairs typed at 637,655 SNPs. For each SNP, a shared genotype variable was created to capture donor-recipient allele sharing. The shared genotype was assigned a value of 0 if the recipient and donor have the same genotype at a given SNP and a value of 1 if they differ by 1 or 2 alleles. This idea, that a "matched" genotype can impact risk of TRM, is similar to the idea behind HLA matching. For all survival analyses we accommodated the competing risk of death due to disease; covariates including age, body mass index, and graft type (blood or marrow) were included in all analyses. Analyses of the shared genotype variable with TRM were run for all diseases together and excluding ALL (AML+MDS). P-values for each cohort were combined using METAL software with weights proportional to the square root of the number of cases. We report on results for combined P-value (Pmeta) 〈 5 x 10-8. In analyses of AML+MDS donor-recipient pairs four typed SNPs, rs9884653, rs16850885, rs1246576 and rs10014791, spanning 865,040 base pairs in and near MTHFD2L and EPGN on chromosome 4q13.3 were significant at Pmeta 〈 5 x 10-8; several SNPs in this region approach genome wide significance level. The most significant SNP, rs16850885 (P=7.6 x 10-7 in Cohort 1, P=6 x10-3 in Cohort 2, Pmeta =1.8 x 10-8) at 74,304,423 bp, places recipients who differ from their donors by at least one allele (5.2% of patients in Cohort 1 and 3.9% in Cohort 2) at a 2.6 fold increased risk of TRM (Tables 1 and 2). It is in perfect linkage disequilibrium (r2=1) with rs9884653 at 74,280,426 and rs10014791 at 75,145,466; rs1246576 is in strong linkage disequilibrium with rs16850885 (r2 =.91). All four SNPs shows hazard ratios (HRs) of similar magnitude across both cohorts for patients who differ from their donors by at least 1 allele. These SNPs are not associated with death due to disease in either cohort. The difference was not allele specific; of those patients who died of TRM with at least one allele difference approximately 50% of the donors provided the less common A allele (donor A/G or A/A with recipient G/G) and 50% of the recipients had the A allele (recipient A/G with a G/G donor). Analyses of the donors and recipients separately each show some evidence of association with the minor allele A at rs16850885 (Table 2), but it is patients who differ from their donors at this locus who have greatest risk of TRM. A previous GWAS showed that individuals with at least one copy of the A allele at rs16850885 have lower levels of secreted IL-1β following small pox vaccination compared with those who are homozygous GG (P=7.3 x 10-9). IL-1β, a pro-inflammatory cytokine secreted early in the inflammatory response, may be contributing to the risk of TRM through the combination of initiation and maintenance of host tissue inßammation and donor cell inßammatory response. Our study, DISCOVeRY-BMT, is the largest GWAS of TRM HLA-matched URD-BMT. Further confirmation of these findings in a third cohort may aid in donor selection. Disclosures Sucheston-Campbell: NIH/NHLBI: Research Funding. McCarthy:The Binding Site: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Hahn:NIH/NHLBI: Research Funding; Novartis: Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2621-2621
    Abstract: Inherited genetic polymorphisms in ARID5B, GATA3, and IKZF1 have been reported to be associated with ALL and vary by cytogenetic subgroups. For example, ARID5B variants are associated with hyperdiploid B-ALL in pediatric patients, which occurs in approximately 30% of ALL patients and is a marker of good prognosis. In contrast, a variant in GATA3 (rs3824662) has been consistently associated with ALL regardless of cytogenetics. While associations by subtype and across age have been explored in ALL patients, the effect of sex on the association of germline variants with ALL has not been tested, despite the higher incidence and worse prognosis of ALL in males than females. We tested the association of previously reported ALL variants in a high-risk ALL population of URD-BMT recipients, assessing the association of these variants with different cytogenetic subgroups and for heterogeneity of effect by sex. Genotyping was performed using the Illumina Omni-Express BeadChip containing approximately 730,000 single nucleotide polymorphisms (SNPs). After quality control, cohort 1 (C1) and cohort 2 (C2) included 364 (C1) and 82 (C2) B-ALL cases of continental European ancestry and 2,229 (C1) and 809 (C2) controls (unrelated healthy donors). The sample size of non-European cases was not adequate for genetic analyses. Cases were a subset of the genome wide association study (GWAS) named Determining the Influence of Susceptibility COnveying Variants Related to one-Year mortality after BMT (DISCOVeRY-BMT) a parent GWAS study in collaboration with the Center for International Blood and Marrow Transplant Research. We tested SNPs within GATA3, IKZF1 and ARID5B for associations with disease status using logistic regression assuming an additive model adjusted for age. Analyses were done separately for the following B-ALL subgroups: hyperdiploid negative ALL, Ph-negative ALL, Abnormal cytogenetics and Normal cytogenetics. P-values for each cohort were combined using METAL software with weights proportional to the square root of the sample size(Pmeta). Sex-stratified analyses were performed using all B-ALL cases and by subgroup. Tests for heterogeneity using Cochran's Q method and I² statistic were used to calculate the percentage of variation across sex and cytogenetic group, abnormal versus normal). The study population consisted mostly of non-hyperdiploid and Ph negative B-ALL. The GATA3 variant conferred a 78% (P=2.9x10-10) and 82% (P=0.001) increased odds of B-ALL in C1 and C2, respectively, Pmeta=1.7x10-12 (Table 1). The GATA3 SNP was significantly associated, Pmeta 〈 5x10-8, with all subgroups except abnormal cytogenetics ALL. The SNP in IKZF1 wasassociated (Pmeta 〈 5x10-8) with B-ALL overall, Ph negative and normal cytogenetics ALL, with some evidence of heterogeneity in odds ratios (ORs) between normal and abnormal cytogenetics B-ALL in both cohorts. The ARID5B variant, previously shown to be strongly associated with pediatric ALL, showed weak or no evidence of association (Table 1). IKZF1 (rs11980379) associations were stronger across all associated subgroups in males (Table 2). When comparing ORs calculated for males and females in there was evidence of significant heterogeneity (Table 2) for B-ALL overall (P=0.02), non-hyperdiploid (P =0.02) and normal cytogenetics (P=0.004). GATA3 showed consistent association, with replication, across the cytogenetic subgroups analyzed. IKZF1 variant demonstrated differences by sex, with significant associations between this SNP and B-ALL, observed primarily in males, regardless of cytogenetics. Thus, sex-specific associations were not an artifact of underlying cytogenetic distributions between males and females. The variation between males and females indicates the IKZF1 variant may impact ALL risk differently in males and females, potentially explaining both the overall greater risk of ALL in males and the risk of ALL with worse outcomes. ARID5B associations have previously been associated with hyperdiploid ALL, however the majority of our high-risk study population is non-hyperdiploid, so ARID5B may be a marker of good risk ALL which is rarely treated with URD-BMT. Disclosures Hahn: Novartis: Equity Ownership; NIH/NHLBI: Research Funding. McCarthy:Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; The Binding Site: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Honoraria, Membership on an entity's Board of Directors or advisory committees. Sucheston-Campbell:NIH/NHLBI: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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. 126, No. 23 ( 2015-12-03), p. 397-397
    Abstract: While survival outcomes after HLA-matched unrelated donor (URD) blood and marrow transplant (BMT) have significantly improved over the last 2 decades, about 40% of patients die of various causes before one-year post-URD BMT. We performed a genome-wide association study (GWAS) named DISCOVeRY-BMT (Determining the Influence of Susceptibility COnveying Variants Related to one-Year mortality after BMT) of overall (OS) and progression-free (PFS) survival in 3,532 patients treated for AML, ALL or MDS and reported to CIBMTR from 2000-2011 and their 8/8 HLA-matched URDs. Cohort 1 consisted of 2,609 patients with 10/10 HLA-matched URD BMT from 2000-08; Cohort 2 consisted of 923 patients with 10/10 HLA-matched URD BMT from 2009-11 and 8/8 (but 〈 10/10) HLA-matched URD BMT from 2000-11. Genotyping on recipients and donors was performed using the HumanOmniExpress-24 BeadChip (Illumina, San Diego, CA), containing approximately 730,000 single nucleotide polymorphisms (SNPs); the CEU reference panel from the 1000 Genomes project (March 2012 release) was used to impute additional genotypes not available on the GWAS chip or that failed typing. Samples were pre-phased with SHAPEIT and genotypes inferred using IMPUTE2. After quality control, Cohort 1 included 2,108 recipients and 2,052 donors and Cohort 2 included 773 recipients and 760 donors of European-American ancestry typed at 637,655 markers and ~9 million imputed SNPs. Each SNP was measured for association with OS and PFS using Cox proportional hazard models in R. All models included age at BMT, diagnosis (AML, ALL, MDS), disease status at BMT (early, intermediate, advanced), cell source (blood, marrow) and year of BMT. P-values were combined using METAL software with weights proportional to the square root of the number of cases. We report results for a combined P -value (Pmeta) 〈 5x10-8. The T allele of rs9990017 in recipients is significantly associated with better OS (Table) yielding about a 30% decreased hazard of all causes of death including disease relapse, graft-versus-host disease, infection and organ failure. Analysis of the effect of donor genotype at this SNP shows no evidence of association with OS or PFS. The strongest imputed association in this region (info score 〉 .98) was rs9857765 with the T allele at this locus conferring a significantly improved OS in both cohorts (Table). Two SNPs (rs9845520 and rs9839074) in strong linkage disequilibrium (r2 =.94) with both rs9857765 and rs9990017, are predicted to affect transcription factor binding (Regulomedb score=2b) of the highly-conserved gene MBNL1 and the common allele in rs9839074 is predicted to be in an important position in the GATA1 (erythroid development) motif and a binding site for CEBPB, STAT3 and FOS across multiple cell lines. MBNL1 protein is expressed in most blood and marrow hematopoietic cells, while MBNL1 mRNA is expressed in many organs. CEBPB regulates immune response genes, is required for normal macrophage function/differentiation and can interact with (among others) glucocorticoid receptor, IL-6, TNF-alpha and transporter proteins conferring multi-drug resistance (ABCC2, ABCB1). Normal activation of STAT3 is required for self-renewal of embryonic stem cells and differentiation of TH17 cells, while constitutive activation of STAT3 is associated with poor prognosis in acute leukemia. FOS is involved with CD16+ signaling in NK cells, vitamin D receptor gene regulation in osteoporosis, and proliferation of hematopoietic cells, among other roles. A region on 13q34 showed evidence of association with PFS; recipients whose donor had a T allele at rs11617176 showed improved PFS compared to those whose donors had the C allele (see Table). This region contains variants identified in other GWAS of mean platelet volume, coronary artery disease, interstitial lung disease, response to tocilizumab in rheumatoid arthritis patients and renal transplant outcomes, among others. Our study, DISCOVeRY-BMT, is the first GWAS for URD BMT survival outcomes. Confirmation of these findings in a third cohort, genotyping of imputed SNPs, and further studies of the functional consequences of these SNPs may provide more individualized risk prediction and prognosis, while confirmation of rs11617176 may aid in donor selection. Table 1. Table 1. Disclosures Hahn: Novartis: Equity Ownership; NIH/NHLBI: Research Funding. McCarthy:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Honoraria, Membership on an entity's Board of Directors or advisory committees; The Binding Site: Honoraria, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Honoraria, Membership on an entity's Board of Directors or advisory committees; Onyx: Honoraria, Membership on an entity's Board of Directors or advisory committees; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees. Sucheston-Campbell:NIH/NHLBI: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    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...