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
  • American Association for Cancer Research (AACR)  (20)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 8 ( 2016-04-15), p. 2197-2205
    Abstract: The genomic and clinical information used to develop and implement therapeutic approaches for acute myelogenous leukemia (AML) originated primarily from adult patients and has been generalized to patients with pediatric AML. However, age-specific molecular alterations are becoming more evident and may signify the need to age-stratify treatment regimens. The NCI/COG TARGET-AML initiative used whole exome capture sequencing (WXS) to interrogate the genomic landscape of matched trios representing specimens collected upon diagnosis, remission, and relapse from 20 cases of de novo childhood AML. One hundred forty-five somatic variants at diagnosis (median 6 mutations/patient) and 149 variants at relapse (median 6.5 mutations) were identified and verified by orthogonal methodologies. Recurrent somatic variants [in (greater than or equal to) 2 patients] were identified for 10 genes (FLT3, NRAS, PTPN11, WT1, TET2, DHX15, DHX30, KIT, ETV6, KRAS), with variable persistence at relapse. The variant allele fraction (VAF), used to measure the prevalence of somatic mutations, varied widely at diagnosis. Mutations that persisted from diagnosis to relapse had a significantly higher diagnostic VAF compared with those that resolved at relapse (median VAF 0.43 vs. 0.24, P & lt; 0.001). Further analysis revealed that 90% of the diagnostic variants with VAF & gt;0.4 persisted to relapse compared with 28% with VAF & lt;0.2 (P & lt; 0.001). This study demonstrates significant variability in the mutational profile and clonal evolution of pediatric AML from diagnosis to relapse. Furthermore, mutations with high VAF at diagnosis, representing variants shared across a leukemic clonal structure, may constrain the genomic landscape at relapse and help to define key pathways for therapeutic targeting. Cancer Res; 76(8); 2197–205. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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 ...
  • 2
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 23 ( 2008-12-01), p. 7896-7899
    Abstract: Purpose: FAS is a cell surface receptor involved in apoptotic signal transmission. Deregulation of this pathway results in down-regulation of apoptosis and subsequent persistence of a malignant clone. A single nucleotide polymorphism resulting in guanine-to-adenine transition in the FAS promoter region (position -1377) is thought to reduce stimulatory protein 1 transcription factor binding and decrease FAS expression. Previous work has shown increased risk of developing acute myeloid leukemia (AML) in adult patients with a variant allele at this site. The same authors have shown that the presence of an adenine residue rather than a guanine residue at −1,377 bp significantly attenuates transcription factor stimulatory protein 1 binding and may contribute to a reduction in FAS expression and ultimately to the enrichment of apoptosis-resistant clones in AML. We hypothesized that FAS genotype by altering susceptibility to apoptosis might affect outcome of childhood AML therapy. Experimental Design: Four hundred forty-four children treated for de novo AML on a uniform protocol were genotyped for FAS 1377. Results: There were no significant differences in overall survival, event-free survival, treatment-related mortality, or relapse rate between patients with FAS 1377GG genotype versus 1377GA/1377AA genotypes. Conclusions: FAS 1377 genotype does not alter outcome of de novo AML in children.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 8 ( 2016-04-15), p. 1951-1957
    Abstract: Purpose: Gemtuzumab ozogamicin (GO), a calicheamicin-conjugated mAb against CD33, has been used in the treatment of acute myeloid leukemia (AML). We evaluated the impact of the addition of GO to standard chemotherapy and hematopoietic stem cell transplant (HCT) in patients with FLT3/ITD. Experimental Design: We analyzed children with FLT3/ITD–positive AML (n = 183) treated on two consecutive Children's Oncology Group AML trials (NCT00070174 and NCT00372593). Outcomes were assessed for FLT3/ITD patients receiving standard chemotherapy with or without GO (GO vs. No-GO, respectively), and the impact of consolidation HCT for high-risk FLT3/ITD patients [high FLT3/ITD allelic ratio (ITD-AR)]. Results: For all FLT3/ITD patients, complete remission (CR) rates for the GO versus No-GO cohorts were identical (64% vs. 64%; P = 0.98). Relapse rate (RR) after initial CR was 37% for GO recipients versus 59% for No-GO recipients (P = 0.02), disease-free survival (DFS) was similar (47% vs. 41%; P = 0.45), with higher treatment-related mortality (TRM) in GO recipients (16% vs. 0%; P = 0.008). Among high-risk FLT3/ITD patients with high ITD-AR, those who received HCT in first CR with prior exposure to GO had a significant reduction in RR (15% vs. 53%; P = 0.007), with a corresponding DFS of 65% versus 40% (P = 0.08), and higher TRM (19% vs. 7%; P = 0.08). Conclusions: CD33 targeting with HCT consolidation may be an important therapeutic strategy in high-risk FLT3/ITD AML and its efficacy and associated toxicity warrant further investigation. Clin Cancer Res; 22(8); 1951–7. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-93-LB-93
    Abstract: Acute myeloid leukemia (AML) is a heterogeneous group of disorders with significant genomic complexity that contributes to variable clinical responses and poor outcomes despite intensive therapy. Although genomic alterations, including karyotypic alterations and somatic mutations have been identified in most patients with AML, their utility has been limited by an inability to accurately predict outcome in the majority of patients. There is also little information on the molecular evolution of AML from diagnosis to relapse. To define the genomic profile of relapse in AML and evaluate the extent of clonal evolution from diagnosis to relapse, we performed exome capture sequencing in matched trios of specimens (diagnostic, remission and relapse specimens) from 22 patients with AML (64 specimens) who were treated on COG clinical trials and lacked previously known high risk features. In the 22 matched diagnostic and 20 relapse cases, 729 somatic variants were identified with a variant allelic frequency ranging from 1% to 94% in diagnostic (N=384) or relapse (N=345) specimens (33.1 mutations/patient) of which 372 were verified with secondary deep sequencing of targeted regions. A verification rate of 86% in mutations with & gt;20% variant allelic frequency was achieved. 335 somatic mutations (median15.5 mutations/patient, range 8-42 mutations/patient) were observed in the 20 cases with diagnostic and relapse specimens. Of these 335 somatic mutations, 221 mutations (66%) in 193 genes were identified in the diagnostic specimens, of which 107 mutations (49%) in 101 genes were present at disease recurrence. In addition to the 107 mutations that persisted from diagnosis to relapse, there was emergence of an additional 114 mutations in 106 genes at the time of relapse, possibly due to clonal selection of a rare pre-existing clone. Somatic mutations in 17 genes were detected in more than one patient at either diagnosis or relapse or both. Of the mutations that were present at both diagnosis and relapse, only 2 genes (KIT and TET2) were mutated in more than one patient, highlighting the paucity of common, novel relapse-associated mutations in the studied cohort. In addition evaluation of copy number (CN) alterations by SNP genotyping in the diagnostic and relapse specimens revealed similar clonal evolution with apparent resolution of diagnostic CN variations and appearance of novel somatic CN alterations in relapse specimens. The study results highlight the genomic complexity of childhood AML, and suggest significant clonal evolution from diagnosis to relapse, with resolution of mutations associated with chemotherapy sensitive clones. Newly evolved mutations may represent rare, chemotherapy resistant clones present at diagnosis, which are selected by exposure to chemotherapy, and may cooperate with other mutations to lead to therapy resistance and poor outcome. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-93. doi:1538-7445.AM2012-LB-93
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 631-631
    Abstract: Background: Although genomic changes identified by clinical tumor sequencing may be present in the germline, precision oncology trials have generally not incorporated germline testing and reporting. We describe our experience with clinical reporting of matched tumor and germline results from the NCI-COG Pediatric Molecular Analysis for Therapy Choice (MATCH) trial (NCT03155620). Design/Method: Patients age 1 to 21 years old with treatment-refractory solid tumors, non-Hodgkin lymphomas, or histiocytic disorders were eligible. DNA and RNA extracted from FFPE tumors were sequenced using Oncomine Assay v.3 with reporting of potentially actionable fusions, amplifications, and mutations, including loss of function variants in tumor suppressor genes. Germline reporting focused on 36 genes from the same DNA panel that convey adult or pediatric cancer susceptibility but does not include all cancer susceptibility genes (e.g. APC, DICER1). Deletions and splice site variants were not reported. Results: As of June 2020, 1009 patients had been enrolled from 132 COG sites, with tumor and germline testing complete for 868 patients to date. Overall, 62 (7.1%) germline reports included a pathogenic or likely pathogenic germline variant (termed germline findings) in 18 cancer susceptibility genes. This frequency was similar in patients with solid tumors (46/633, 7.3%), CNS tumors (14/204, 6.9%), and lymphomas/histiocytoses (2/31, 6.5%). Variant(s) of potential germline significance were identified in 25% of tumor reports; of these, 74% (163/221) had no germline findings. The proportion of germline findings for genes with & gt;10 reported tumor variants varied: 32% (7/22) in NF1, 25% (3/12) in RB1, 16% (17/108) in TP53 and 0% in ALK (0/21) and PTEN (0/12). Of note, 82% (14/17) of tumor variants in breast cancer susceptibility genes (BRCA1/2, CHEK2 and PALB2) had germline findings. On study intake forms, 25% of patients with germline findings had a “known genetic disease” reported. Oncologists caring for patients with a germline finding were then queried about prior knowledge of a genetic diagnosis. Of 24 respondents, 13 (54%) reported no prior molecular diagnosis of the identified condition. Conclusions: Coordinated germline and tumor testing revealed clinically relevant cancer susceptibility variants across a spectrum of genes in 7% of pediatric patients with treatment-refractory cancers. This is likely an underestimate of germline findings given test limitations. The parallel tumor/normal reporting approach minimized the need for targeted reflex genetic testing in 19% of study participants and provided new information on cancer susceptibility in germline positive patients to half of the responding oncologists. The NCI-COG Pediatric MATCH trial reporting process can serve as a model for precision oncology trials and clinical tumor profiling. Citation Format: Sarah Scollon, Sharon E. Plon, Steven Joffe, Jaclyn A. Biegel, Shashikant Kulkarni, George Miles, David Patton, Brent Coffey, Paul M. Williams, Gregory J. Tsongalis, Mark J. Routbort, Julie M. Gastier-Foster, Lauren Saguilig, Jin Piao, Todd A. Alonzo, Katherine A. Janeway, Peter C. Adamson, Margaret Mooney, James V. Tricoli, Nita L. Seibel, Donald W. Parsons. Germline cancer predisposition results from the National Cancer Institute - Children's Oncology Group (NCI-COG) Pediatric MATCH Trial [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 631.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 16 ( 2019-08-15), p. 5038-5048
    Abstract: KIT mutations (KIT+) are common in core binding factor (CBF) AML and have been associated with varying prognostic significance. We sought to define the functional and clinical significance of distinct KIT mutations in CBF pediatric AML. Experimental Design: Following transfection of exon 17 (E17) and exon 8 (E8) mutations into HEK293 and Ba/F3 cells, KIT phosphorylation, cytokine-independent growth, and response to tyrosine kinase inhibitors (TKI) were evaluated. Clinical outcomes of patients treated on COG AAML0531 (NCT01407757), a phase III study of gemtuzumab ozogamicin (GO), were analyzed according to mutation status [KIT+ vs. wild-type KIT (KIT−)] and mutation location (E8 vs. E17). Results: KIT mutations were detected in 63 of 205 patients (31%); 22 (35%) involved only E8, 32 (51%) only E17, 6 (10%) both exons, and 3 (5%) alternative exons. Functional studies demonstrated that E17, but not E8, mutations result in aberrant KIT phosphorylation and growth. TKI exposure significantly affected growth of E17, but not E8, transfected cells. Patients with KIT+ CBF AML had overall survival similar to those with KIT− (78% vs. 81%, P = 0.905) but higher relapse rates (RR = 43% vs. 21%; P = 0.005). E17 KIT+ outcomes were inferior to KIT− patients [disease-free survival (DFS), 51% vs. 73%, P = 0.027; RR = 21% vs. 46%, P = 0.007)], although gemtuzumab ozogamicin abrogated this negative prognostic impact. E8 mutations lacked significant prognostic effect, and GO failed to significantly improve outcome. Conclusions: E17 mutations affect prognosis in CBF AML, as well as response to GO and TKIs; thus, clinical trials using both agents should be considered for KIT+ patients.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2699-2699
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2699-2699
    Abstract: Background: Despite substantial increases in therapy intensity, the overall survival of pediatric acute myeloid leukemia (AML) is still guarded, with survival rates of approximately 60%. This indicates the need for new therapeutic strategies, as well as improved risk stratification. Chemotherapies target proteins rather than genetic events, yet little is known about the proteomic landscape in pediatric AML. This study provides a global assessment of pediatric AML protein expression and correlates protein expression with outcome. Methods: A reverse phase protein array (RPPA) probed with 298 validated antibodies was performed to determine protein expression in ‘‘bulk'' (CD3-/19-) AML cells from 505 diagnostic pediatric AML patients who participated in the Children's Oncology Group AAML1031 phase 3 clinical trial. Proteomic profiling was applied in the context of 31 protein functional groups (PFG) (e.g., cell cycle, apoptosis) to analyze their expression in relation to related proteins. Progeny clustering was performed to identify patients with correlated protein expression patterns within each PFG (protein cluster). Block clustering searched for protein clusters that recurrently co-occurred (protein constellation), and for subgroups of patients that expressed similar combinations of protein constellations (patient signatures). Signatures were correlated with patient and disease characteristics. Results: For each PFG, protein clusters (n=120) could be discerned that showed different protein expression states. From this we constructed 11 protein constellations and 10 patient signatures. Signatures were correlated with event-free survival (EFS) when we combined signatures into favorable (Sig. 4, 8), intermediate (Sig. 6, 7, 9) and unfavorable (Sig. 1-3, 5, 10) groups (p=0.01). Other significant clinical correlations between signatures included CEPBA (40% in Sig. 6, vs. 9% overall, p & lt;0.001), MRD status (high in Sig. 2 vs. low in Sig. 6+7, p=0.006) and several laboratory features. Proteins that were significantly altered compared to normal CD34+ cells were identified for each signature. From this list, 20 proteins were recognized as universally downregulated (CDKN1A, PPP2R2A) and only PIK3CA was universally upregulated. Many druggable proteins showed association with specific protein signatures: high KIT (Sig. 1, 2, 6), high BCL2 (Sig. 1, 2, 6, 9) and high NPM1 (Sig. 1, 2, 6, 9). Conclusion: We studied the proteomic landscape in 505 pediatric AML patients, and identified 10 protein signatures based on 11 protein constellations. We identified signatures that did well with ADE therapy vs. signatures that did not. Recognition of deregulated proteins could help to select drugs that could potentially improve individualized therapies for the latter signatures. Citation Format: Fieke W. Hoff, Yihua Qiu, Wendy Hu, Amina A. Qutub, Alan S. Gamis, Richard Aplenc, E Anders Kolb, Todd A. Alonzo, Eveline SJM de Bont, Terzah M. Horton, Steven M. Kornblau. Proteomic landscape of de novo pediatric acute myeloid leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2699.
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 14 ( 2015-07-15), p. 3187-3195
    Abstract: Purpose: Exploratory gene expression array analyses suggested multimerin-1 (MMRN1) to be a predictive biomarker in acute myelogenous leukemia (AML). Following up on these studies, we evaluated the role of MMRN1 expression as outcome predictor in two recent Children's Oncology Group trials. Experimental Design: We retrospectively quantified MMRN1 expression in 183 participants of AAML03P1 and 750 participants of AAML0531 by reverse-transcriptase PCR and correlated expression levels with disease characteristics and clinical outcome. Results: In AAML03P1, the highest quartile of MMRN1 expression (expression ≥0.5 relative to β-glucuronidase; n = 45) was associated with inferior event-free survival (EFS; P & lt; 0.002) and higher relapse risk (P & lt; 0.004). In AAML0531, in which we quantified MMRN1 mRNA for validation, patients with relative MMRN1 expression ≥0.5 (n = 160) less likely achieved remission (67% vs. 77%, P = 0.006), and more frequently had minimal residual disease (43% vs. 24%, P = 0.001) after one induction course. They had inferior overall survival (OS; 44% ± 9% vs. 69% ± 4% at 5 years; P & lt; 0.001) and EFS (32% ± 8% vs. 54% ± 4% at 5 years; P & lt; 0.001) and higher relapse risk (57% ± 10% vs. 35% ± 5% at 5 years; P & lt; 0.001). These differences were partly attributable to the fact that patients with high MMRN1 expression less likely had cytogenetic/molecular low-risk disease (P & lt; 0.001) than those with low MMRN1 expression. Nevertheless, after multivariable adjustment, high MMRN1 expression remained statistically significantly associated with shorter OS (HR, 1.57; 95% confidence interval, 1.17–2.12; P = 0.003) and EFS (HR, 1.34; 1.04–1.73; P = 0.025), and higher relapse risk (HR, 1.40; 1.01–1.94; P = 0.044). Conclusions: Together, our studies identify MMRN1 expression as a novel biomarker that may refine AML risk stratification. Clin Cancer Res; 21(14); 3187–95. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3479-3479
    Abstract: Childhood AML is an aggressive disease with high rates of failures and poor survival. We have demonstrated that the molecular landscape of AML in children is distinct, and co-occurrence of variants modulate outcomes. Recent discovery of tandem duplication (TD) of the UBTF gene in AML, with enrichment in FLT3-ITD has implicated yet another mutation whose cooperation with FLT3-ITD may modify outcome. Here, we provide a comprehensive evaluation of UBTF-TD in de novo AML and define its clinical implications within FLT3-ITD patients. Initial interrogation of transcriptome data from 1,158 children enrolled on COG AAML1031 identified 50 cases of UBTF-TD (4.3%). Overwhelming majority of UBTF-TD cases were observed in FLT3-ITD cases (77%), vs. that of 1.2% in those without FLT3-ITD (p & lt;0.001). Given extreme enrichment of UBTF-TD in FLT3-ITD, we inquired whether cooperation of UBTF-TD and FLT3-ITD creates a distinct clinical entity. To this end we screened diagnostic DNA from 400 FLT3-ITD patients treated on three consecutive CCG/COG trials (COG AAML1031, COG AAML0531, and CCG-2961) by PCR and fragment analysis. UBTF-TD was identified in 61 FLT3-ITD cases (15.3%). The data presented here forth focuses on evaluation of implications of UBTF-TD in FLT3-ITD positive patients only. Within the FLT3-ITD patients, initial correlation of UBTF-TD with demographics, disease characteristics, and associated genomic variants was conducted. Patients with and without UBTF-TD had a similar median age at diagnosis (p=0.322), lower diagnostic WBC (p=0.010) and higher marrow blast % (p & lt;0.001). There was a stark paucity of cooperating variants that commonly co-occur with FLT3-ITD, with a single NPM1 mutation (1.6% vs. 29%, p & lt;0.001) and no NUP98 fusions (0% vs. 23%, p & lt;0.001). There was a significant enrichment of WT1 mutations, with 45% UBTF-TD patients with a WT1 mutation (FLT3-ITD/UBTF-TD/WT1), vs. 11% in UBTF-WT (p & lt;0.001). Trisomy 8 (Tri8) was seen in 15% of UBTF-TD. Patients with UBTF-TD had a lower CR rate (44% vs. 60%, p = 0.018), and Higher MRD rate (38% vs. 21%, p & lt;0.001). Patients with and without UBTF-TD had an EFS of 28% vs. 42% (p=0.047) with a corresponding OS of 40% and 57% (p=0.019). Given enrichment of WT1 mutations and Tri8 in patients with UBTF-TD, we studied the outcome UBTF-TD patients in the context of these two variants. FLT3-ITD/UBTF-TD/WT1 patients had a 5-year EFS of 17% vs. 38% for similar patients without WT1 mutations (p=0.0062). Patients with UBTF-TD with additional Tri8 had a similarly poor outcome with an EFS of 23% with a corresponding OS of 33%, providing a distinct high risk UBTF-TD cohort (+WT1 or Tri8), whereas the remaining UBTF-TD patients had a more favorable outcome with EFS and OS of 64% and 86%, respectively (p & lt;0.0001, and p & lt;0.0001). UBTF-TD is a novel genomic entity with high enrichment in patients with FLT3-ITD and a distinct clinical outcome driven by cooperating WT1 mutation and Tri8. Citation Format: Leila Robinson, Amanda Leonti, Todd A. Alonzo, Yi-Cheng Wang, Michele S. Redell, Rhonda E. Ries, Jenny L. Smith, Tiffany A. Hylkema, Quy Le, E Anders Kolb, Richard Aplenc, Xiaotu Ma, Jeffrey Klco, Katherine Tarlock, Soheil Meshinchi. UBTF tandem duplications (UBTF-TD) in childhood AML: Enrichment in FLT3-ITD and association with clinical outcome [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3479.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1921-1921
    Abstract: Polymorphisms in DNA damage repair (DDR) genes could serve as predictors of response to gemtuzumab ozogamicin (GO), a CD33 targeting antibody conjugated to the DNA damaging agent calicheamicin approved for use in immunotherapy for acute myeloid leukemia (AML). GO is internalized upon binding to the myeloid cell surface antigen CD33, allowing calicheamicin-mediated DNA damage and subsequent apoptosis. Following concerns over safety, GO was withdrawn in 2010 but was re-approved in 2017 with the emergence of new evidence of its clinical efficacy and safety. In clinical trials, significantly improved event free survival (EFS), disease free survival (DFS), and relapse risk (RR) were observed in a subset of AML patients with the addition of GO to conventional AML chemotherapy. Combined with low overall survival (OS) and high RR with the relatively unchanged conventional AML chemotherapy, GO usage is anticipated to increase. We used samples from the Children's Oncology Group (COG) trial AAML0531 to determine the association between polymorphism in DDR genes and OS, EFS, DFS, and RR with GO treatment as the primary end points. In this trial, newly diagnosed AML patients were randomized to receive either the standard 5 course chemotherapy alone (Arm A) or with two doses of GO (3mg/m2/dose) during induction 1 and intensification 2 (Arm B). Briefly, genomic DNA samples from a total of 947 patients from Arm A and Arm B were genotyped for 131 SNPs across 43 key genes involved in calicheamicin-mediated DDR. Overall, 28 SNPs in 21 genes were significantly associated with OS, EFS, DFS, and RR. Of these, 21 SNPs in 16 genes were associated with response to GO treatment (Arm B; p ≤ 0.05) and not with conventional AML chemotherapy (Arm A; p ≥ 0.05). The top 5 SNPs occurred within DDB2, XPA, XPC, and XRCC1 – genes involved in the nucleotide excision repair (NER) DDR pathway. The intronic variation rs830083 (G & gt;C) in DDB2 had the poorest outcome (OS Cox p = 0.01; EFS cox p = 0.004; DFS p = 0.000; RR reg p = 0.0006). Furthermore, rs830083 was also most significantly associated with standard risk patients (DFS p = 0.003), EFS (p=0.01) and RR (p = 0.0006). DDB2 has been reported to play a critical role in apoptotic processes post-DNA damage induction by regulating p53, lending further importance to the association between rs830083 and poor response to treatment. In conclusion, the identification of several variants associated with treatment outcomes in our study underscores the importance of a pathway-directed SNP-based candidate gene approach to identify personalized approaches for AML treatment. Specifically, screening DDB2 SNPs could be applied in patient selection and predicting response to GO treatment. Citation Format: Vivek M. Shastri, Lata Chauhan, Todd A. Alonzo, Yi-Cheng Wang, Richard Aplenc, Betsy A. Hirsch, Alan S. Gamis, Soheil Meshinchi, Jatinder K. Lamba. The DDB2 intronic variation rs830083 is a strong predictor of gemtuzumab ozogamicin response in AML patients: A calicheamicin pathway-directed analysis from the COG-AAML0531 trial [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 1921.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...