internal tandem duplication
Recently Published Documents


TOTAL DOCUMENTS

349
(FIVE YEARS 70)

H-INDEX

50
(FIVE YEARS 8)

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Jarno Kivioja ◽  
Disha Malani ◽  
Ashwini Kumar ◽  
Mika Kontro ◽  
Alun Parsons ◽  
...  

AbstractFLT3 internal tandem duplication (FLT3-ITD) is a frequent mutation in acute myeloid leukemia (AML) and remains a strong prognostic factor due to high rate of disease recurrence. Several FLT3-targeted agents have been developed, but determinants of variable responses to these agents remain understudied. Here, we investigated the role FLT3-ITD allelic ratio (ITD-AR), ITD length, and associated gene expression signatures on FLT3 inhibitor response in adult AML. We performed fragment analysis, ex vivo drug testing, and next generation sequencing (RNA, exome) to 119 samples from 87 AML patients and 13 healthy bone marrow controls. We found that ex vivo response to FLT3 inhibitors is significantly associated with ITD-AR, but not with ITD length. Interestingly, we found that the HLF gene is overexpressed in FLT3-ITD+ AML and associated with ITD-AR. The retrospective analysis of AML patients treated with FLT3 inhibitor sorafenib showed that patients with high HLF expression and ITD-AR had better clinical response to therapy compared to those with low ITD-AR and HLF expression. Thus, our findings suggest that FLT3 ITD-AR together with increased HLF expression play a role in variable FLT3 inhibitor responses observed in FLT3-ITD+ AML patients.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2315-2315
Author(s):  
Philip Connor ◽  
Linyi Fan ◽  
Stanley Gill ◽  
Jason E. Hill ◽  
Nisha Philipose ◽  
...  

Abstract Background : Acute myeloid leukemia (AML) accounts for ~18% of all childhood leukemias (Puumala SE, et al. Pediatr Blood Cancer. 2013;60(5):728-733). Gilteritinib, an oral FMS-like tyrosine kinase 3 (FLT3) inhibitor, has demonstrated efficacy with favorable tolerability in clinical trials of adults with FLT3-mutated relapsed or refractory (R/R) AML, including internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutations (Perl AE, et al. N Engl J Med. 2019;381(18):1728-1740), leading to approval for the treatment of adults with R/R AML in the United States and many other countries/regions. Since the clinical progression of FLT3 ITD AML appears to be similar in children and adults, it is expected that the clinical benefit of gilteritinib in pediatric patients with FLT3 ITD R/R AML should be similar to that demonstrated in adults. We describe an ongoing phase 1, multicenter, open-label, single-arm, dose-escalation study (ClinicalTrials.gov identifier: NCT04240002) investigating the combination of gilteritinib with chemotherapy in children, adolescents, and young adults with FLT3 ITD R/R AML, which will inform the dose for the phase 2 dose-expansion portion of the study. Study Design and Methods : This phase 1 dose-escalation study will enroll patients 6 months to <21 years of age with FLT3 ITD and/or TKD-mutated AML (determined using institutional assay). Key inclusion and exclusion criteria include patients who are in first or greater relapse or are refractory to induction therapy with no more than 1 attempt at remission induction and (a) fully recovered from the acute toxic effects of all prior therapy, (b) have a Karnofsky (if ≥16 years) or Lansky (if <16 years) score ≥50, and (c) been ≥90 days relapsed since hematopoietic stem cell transplantation with no active graft-versus-host disease, if relevant. Patients will undergo screening, including complete physical exam, subject-reported symptoms, and areas of disease (Figure). Patients will be enrolled in 1 of the 3 following groups: group 1 (2 to <21 years), group 2 (1 to <2 years), and group 3 (6 months to <1 year). Gilteritinib will be administered once daily starting from day 8 through day 21 of a 28-day cycle at the starting dose of 2 mg/kg/day for Group 1 and 1 mg/kg/day for Groups 2 and 3, with escalation to 2 or 3 mg/kg/day (the latter to be evaluated only if there is lack of toxicity or acceptable dose-limiting toxicity [DLT] and lack of sufficient gilteritinib activity observed at 2 mg/kg/day). Patients in all groups will receive FLAG (fludarabine, 30 mg/m 2/day; cytarabine, 2000 mg/m 2/day; granulocyte-colony stimulating factor, 5 μg/kg/day) on days −1 to 5 and prophylactic single intrathecal cytarabine (age-adjusted dose). Patients who complete 2 cycles of treatment will have the option to participate in long-term treatment (LTT) with gilteritinib for ≤2 years. An end of treatment (EOT) visit will occur 7 days after the last dose of gilteritinib or before initiation of another anticancer therapy. A follow-up visit will be performed 28 days after the EOT visit in patients who are ineligible for LTT. Patients eligible for LTT will undergo a follow-up visit 28 days after the LTT EOT visit. Follow-up for survival will be performed every 3 months for ≤2 years from the 28-day follow-up visit. Study End Points : The primary end point is identification of the maximum tolerated dose and/or recommended phase 2 dose based on the DLT observed in treatment cycle 1 and biologic activity according to plasma inhibitory activity (PIA). The secondary end points are inhibition of phosphorylated FLT3, gilteritinib pharmacokinetics, safety, tolerability, toxicity, event-free survival, overall survival, minimal residual disease assessment, and acceptability and palatability assessment of the formulation. The exploratory end points are correlation of clinical responses to gilteritinib with FLT3 PIA levels, correlation of FLT3 PIA levels and clinical responses to gilteritinib therapy with FLT3 ligand levels before and after exposure to FLAG chemotherapy, and mechanisms of innate and acquired resistance to gilteritinib. All data will be summarized with descriptive statistics for continuous end points and frequency/percentage for categorical end points. Figure 1 Figure 1. Disclosures Connor: Astellas Pharma Inc.: Research Funding. Fan: Astellas Pharma Global Development: Current Employment. Gill: Astellas Pharma Global Development: Current Employment. Hill: Astellas Pharma Global Development: Current Employment; Ligacept, LLC: Current holder of individual stocks in a privately-held company, Other: Stockholder. Philipose: Astellas Pharma Global Development: Current Employment. Delgado: Astellas Pharma Global Development: Current Employment. Tiu: Astellas Pharma, Inc.: Current Employment. Reinhardt: Abbvie: Other: Advisory board; Eusa: Other: Advisory board; Astellas Pharma Inc.: Research Funding; Janssen: Other: Advisory board; BluebirdBio: Other: Advisory board; BMS: Other: Advisory board; Novartis: Other: Advisory board; JAZZ: Other: Advisory board. OffLabel Disclosure: New Indication


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Arnault Tauziède-Espariat ◽  
Gaëlle Pierron ◽  
Delphine Guillemot ◽  
Dorian Bochaton ◽  
Sarah Watson ◽  
...  

2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Sakinah A. Thiryayi ◽  
Helen Ettler ◽  
Emily A. Goebel ◽  
Michel Prefontaine ◽  
Tara A. Paton ◽  
...  

2021 ◽  
pp. 1-11
Author(s):  
Huda F. Ebian ◽  
Sherin Elshorbagy ◽  
Haitham Mohamed ◽  
Ahmad Embaby ◽  
Tarek Khamis ◽  
...  

BACKGROUND: Both of Fms-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) and Additional Sex Comb-like 1 (ASXL1) mutations are frequent and early genetic alteration events in acute myeloid leukemia (AML) patients. These genetic alterations may be associated with unfavorable prognosis. OBJECTIVE: Up to our knowledge, this is the first study performed to evaluate the clinical implication and prognostic significance of FLT3-ITD and ASXL1 mutations and their coexistence on the outcome of Egyptian AML patients. METHODS: Our study included 83 patients with AML who were subjected to immunophenotyping and detection of FLT3-ITD and ASXL1 gene mutation by polymerase chain reaction (PCR) and real-time PCR, respectively. RESULTS: FLT3-ITD and ASXL1 mutations were detected in 20.5% and 18.1% of AML patients respectively. Seven patients (8.4%) had co-expression of both genes’ mutation. FLT3-ITD mutation was significantly higher in younger age, higher WBCs count and poor cytogenetic risk patients (P= 0.01, < 0.001 and 0.008 respectively). ASXL1 mutation was significantly higher in intermediate cytogenetic risk patients (P= 0.2). The mean period of survival and relapse free survival (RFS) were significantly reduced in FLT3-ITD and ASXL1 mutations compared with their non-mutant types (P= 0.01 and 0.03 respectively). Both mutations were independent risk factors for overall survival (OS) and (RFS) in univariate and multivariate analysis in AML patients. CONCLUSION: FLT3-ITD and ASXL1 gene mutations or their coexistence can predict a poor prognosis in AML patients.


Sign in / Sign up

Export Citation Format

Share Document