Non-internal tandem duplication (ITD), non-tyrosine kinase domain (TKD) FLT3 mutations in myeloid malignancies: A brief report of 10 patients

2018 ◽  
Vol 72 ◽  
pp. 1-4
Author(s):  
Alexandra Higgins ◽  
Abhishek A. Mangaonkar ◽  
Mehrdad Hefazi ◽  
David Viswanatha ◽  
Pedro Horna ◽  
...  
2014 ◽  
Vol 6 (1) ◽  
pp. e2014038 ◽  
Author(s):  
Ghaleb Elyamany ◽  
Mohamed Awad ◽  
Omar Alsuhaibani ◽  
Kamal Fadalla ◽  
Omer Al Sharif ◽  
...  

The fms-like tyrosine kinase 3 (FLT3) gene is a member of the class III receptor tyrosine kinase family, mutations of FLT3 were first described in 1997 and account for the most frequent molecular mutations in acute myeloid leukemia. No data currently exist regarding FLT3 mutations in Saudi acute lymphoblastic leukemia (ALL) patients as no study has reported for FLT3 mutations in Saudi ALL patients. In this retrospective study, we have examined a cohort of 77 ALL patients, to determine the prevalence of FLT3 mutations and the possible prognostic relevance of these mutations in ALL patients and did correlations to other biologic factors, such as karyotype, molecular mutations, and leukocyte count. FLT3 internal tandem duplication (ITD) mutations and point mutation in tyrosine kinase domain (D835) mutations were analyzed in ALL patients at diagnosis by polymerase chain reaction (PCR). 2 cases (2.6%, 2/77) were positive for FLT3 mutations, one was found to have FLT3/ITD and other was found to have FLT3/D835. Our findings suggest that FLT3 mutations were not common in Saudi ALL and did not affect clinical outcome.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5118-5118 ◽  
Author(s):  
Alissa Marhäll ◽  
Thomas Fischer ◽  
Florian H. Heidel ◽  
Julhash U. Kazi ◽  
Lars Rönnstrand

Abstract Up to 30% of patients with acute myeloid leukemia (AML) harbor a mutation in FMS like tyrosine kinase 3 (FLT3). This mutation is not only the most frequent, but also the most clinically challenging, because it is associated with increased risk of relapse and poor overall survival. The most common oncogenic FLT3 mutation is the internal tandem duplication (ITD) in the juxtamembrane domain (ITD-JM). Other less frequently occurring mutations include ITD mutations in the tyrosine kinase domain (ITD-TDK) and point mutations in the TKD (e. g. D835Y-TKD). Use of FLT3 inhibitors has shown initial promise, though an acquired resistance remains a problem. A need therefore remains for additional therapeutic targets, and in order to identify these we need to have a better understanding of the mechanisms by which the FLT3 mutations drive leukemogenesis. Although ITD-JM and D835Y-TKD mutations have been studied extensively, the role of the recently identified ITD-TKD remains poorly understood. In this study we compared the ITD-TKD domain mutations with other well-studied FLT3 mutations. We observed that transfection of cytokine-dependent Ba/F3 cells with ITD-TKD is sufficient to induce formation of colonies in semisolid medium in the absence of cytokines. Number and size of colonies were comparable to that of ITD-JM, while D835Y-TKD transfected cells failed to form colonies suggesting that the ITD-TKD mutations have stronger transforming potential than other TKD mutations. Similar to colony formation assays, proliferation and cell survival was significantly higher in ITD-TKD transfected cells compared to cells transfected with D835Y-TKD. ITD-TKD selectively enhanced STAT5 and AKT phosphorylation while ERK1/2 and p38 phosphorylation remained unchanged. Collectively our data suggest that ITD-TKD is a more potent oncogenic mutant compared to the TKD mutants. Disclosures Fischer: Novartis: Consultancy, Honoraria.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2514-2514
Author(s):  
Sabine Kayser ◽  
Richard F. Schlenk ◽  
Frank Breitenbücher ◽  
Martina Porebski ◽  
Kerstin Wittke ◽  
...  

Abstract Background: Activating FLT3 internal tandem duplication mutations (FLT3-ITDs) occur in approximately 30% of acute myeloid leukemia (AML) patients. Expression of the FLT3-ITD receptor results in autophosphorylation of FLT3 and subsequent activation of downstream signaling. Clinically, FLT3-ITDs are associated with a dismal clinical outcome; previous explorative analyses suggest that not only FLT3-ITD per se but also the mutant/wild-type allelic ratio and/or the length of the FLT3-ITD provide prognostic information. Aims: To determine ITD insertion sites and length in FLT3-ITD mutated AML and to correlate these findings with clinical outcome. Methods: In 241 patients, DNA-based amplification of the FLT3-ITD mutation was followed by DHPLC-based separation of FLT3 mutant and wild-type fragments. Single mutated fragments were collected by a fragment collector, reamplified and sequenced. Patients [16 to 60 years of age] were entered on 3 consecutive AMLSG treatment trials [AML HD93, AML HD98A, AMLSG 07-04] all including intensive therapy. Results: Thirty-four (14.1) of the 241 patients had more than one ITD (two ITDs n=29, three ITDs n=3, four ITDs n=2). In total, 282 ITDs were analyzed. The median length was 52 nucleotides (range 15–180). For further correlations we grouped ITD integration sites according to the functional regions of FLT3: JM-domain (JMD) [amino acid (AA) 572–603, patients n =141, ITDs n=148], hinge region of JMD [AA 604–609, patients n=45, ITDs n=48], beta-1-sheet of the tyrosine kinase domain-1 (TKD1) [AA 610–615, patients n=69, ITDs n=73], and the remaining region of TKD1 [AA >615, patients n=13, ITDs n=13]. Interestingly, ITD length was strongly correlated to functional regions with shortest ITDs being present in the JMD and longest in the TKD1 (p<0.001). Clinical data were available in 239 patients showing no differences in patient characteristics (age, WBC etc.); frequencies of cooperating class II mutations (NPM1-mut n=137, CEBPA-mut n=12, MLL-PTD n=21) were equally distributed among the functionally categorized groups. The logistic regression model on induction success (IS) revealed ITD integration sites in the beta-1-sheet (odds ratio (OR) 0.25, p=0.01) and in the remaining region of TKD1 (OR 0.14, p=0.007) as well as logarithm of WBC count (OR 0.36, p=0.002) and NPM1 mutations (OR 2.04, p=0.04) as significant variables. ITD insertion in the beta-1-sheet was also significant in the Cox regression analysis on overall survival (OS) (Hazard Ratio (HR) 2.7, p=0.01). In univariable analyses, event free survival (EFS) and OS were significantly inferior in patients with ITD in the beta-1-sheet (p=0.005 and p=0.001). Of note, the proportions of patients receiving an allogeneic transplantation were comparable in both groups (61% and 67%, respectively). In multivariable analyses, neither length of ITD nor mutant/wild-type allelic ratio had an impact on OS. Conclusions: FLT3-ITD insertion sites seems to be an important prognostic marker for induction failure, EFS and OS. Therefore, not only FLT3-ITD mutation status but also ITD integration site should be prospectively analyzed in future clinical trials, in particular in the context of treatment with FLT3-tyrosine kinase inhibitors.


Blood ◽  
2009 ◽  
Vol 114 (12) ◽  
pp. 2386-2392 ◽  
Author(s):  
Sabine Kayser ◽  
Richard F. Schlenk ◽  
Martina Correa Londono ◽  
Frank Breitenbuecher ◽  
Kerstin Wittke ◽  
...  

AbstractTo evaluate internal tandem duplication (ITD) insertion sites and length as well as their clinical impact in younger adult patients with FLT3-ITD–positive acute myeloid leukemia (AML), sequencing after DNA-based amplification was performed in diagnostic samples from 241 FLT3-ITD–mutated patients. All patients were treated on 3 German-Austrian AML Study Group protocols. Thirty-four of the 241 patients had more than 1 ITD, leading to a total of 282 ITDs; the median ITD length was 48 nucleotides (range, 15-180 nucleotides). ITD integration sites were categorized according to functional regions of the FLT3 receptor: juxtamembrane domain (JMD), n = 148; JMD hinge region, n = 48; beta1-sheet of the tyrosine kinase domain-1 (TKD1), n = 73; remaining TKD1 region, n = 13. ITD length was strongly correlated with functional regions (P < .001). In multivariable analyses, ITD integration site in the beta1-sheet was identified as an unfavorable prognostic factor for achievement of a complete remission (odds ratio, 0.22; P = .01), relapse-free survival (hazard ratio, 1.86; P < .001), and overall survival (hazard ratio, 1.59; P = .008). ITD insertion site in the beta1-sheet appears to be an important unfavorable prognostic factor in young adult patients with FLT3-ITD–positive AML. The clinical trials described herein have been registered as follows: AML HD93 (already published in 2003), AML HD98A (NCT00146120; http://www.ClinicalTrials.gov), and AMLSG 07-04 (NCT00151242; http://www.ClinicalTrials.gov).


2019 ◽  
Vol 15 (34) ◽  
pp. 3885-3894 ◽  
Author(s):  
Shilpa Paul ◽  
Adam J DiPippo ◽  
Farhad Ravandi ◽  
Tapan M Kadia

FLT3 mutations, characterized by an internal-tandem duplication or missense mutations in the tyrosine kinase domain, are observed in a third of patients with newly diagnosed acute myeloid leukemia. FLT3-ITD mutations are associated with high relapse rates and short overall survival with conventional chemotherapy. Several tyrosine kinase inhibitors targeting FLT3 have been developed in an effort to improve survival and therapeutic options. This review focuses on quizartinib, a second-generation FLT3 inhibitor that has demonstrated efficacy and safety as a single agent and in combination with chemotherapy. We discuss its clinical development as well as its place in the treatment of FLT3-mutated acute myeloid leukemia among the other FLT3 inhibtors currently available and its mechanisms of resistance.


2020 ◽  
Vol 4 (3) ◽  
pp. 514-524 ◽  
Author(s):  
Theodore C. Tarver ◽  
Jason E. Hill ◽  
Leena Rahmat ◽  
Alexander E. Perl ◽  
Erkut Bahceci ◽  
...  

Abstract Gilteritinib is the first FMS-like tyrosine kinase 3 (FLT3) tyrosine kinase inhibitor (TKI) approved as monotherapy in acute myeloid leukemia with FLT3 internal tandem duplication and D835/I836 tyrosine kinase domain (TKD) mutations. Sequencing studies in patients have uncovered less common, noncanonical (NC) mutations in FLT3 and have implicated secondary TKD mutations in FLT3 TKI resistance. We report that gilteritinib is active against FLT3 NC and TKI resistance-causing mutations in vitro. A mutagenesis screen identified FLT3 F691L, Y693C/N, and G697S as mutations that confer moderate resistance to gilteritinib in vitro. Analysis of patients treated with gilteritinib revealed that 2/9 patients with preexisting NC FLT3 mutations responded and that secondary TKD mutations are acquired in a minority (5/31) of patients treated with gilteritinib. Four of 5 patients developed F691L mutations (all treated at &lt;200 mg). These studies suggest that gilteritinib has broad activity against FLT3 mutations and limited vulnerability to resistance-causing FLT3 TKD mutations, particularly when used at higher doses.


Sign in / Sign up

Export Citation Format

Share Document