Review Article

Split Viewer

Blood Res 2022; 57(S1):

Published online April 30, 2022

https://doi.org/10.5045/br.2022.2022017

© The Korean Society of Hematology

FLT3 mutations in acute myeloid leukemia: a review focusing on clinically applicable drugs

Jae-Sook Ahn1,2, Hyeoung-Joon Kim1,2

1Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University, Gwangju,
2Genomic Research Center for Hematopoietic Diseases, Chonnam National University Hwasun Hospital, Hwasun, Korea

Correspondence to : Jae-Sook Ahn, M.D., Ph.D.
Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun 58128, Korea
E-mail: f0115@jnu.ac.kr

Received: January 19, 2022; Revised: February 28, 2022; Accepted: February 28, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

FMS-like tyrosine kinase 3 (FLT3) mutations, the most frequently detected genetic aberrations in patients with acute myeloid leukemia (AML), are identified in approximately 30% of patients with newly diagnosed AML and are more common in patients with normal karyotypes. Since the discovery of FLT3 mutations in AML, clinical trials have been actively conducted in patients with FLT3 mutated AML, and FLT3 inhibitors have been introduced into clinical practice. The current standard treatment for patients with newly diagnosed FLT3-mutated AML is 7+3 induction chemotherapy combined with midostaurin. Additionally, gilteritinib is more effective than salvage chemotherapy for relapsed or refractory FLT3-mutated AML. Ongoing trials are expected to provide additional treatment options depending on the disease state and patient vulnerability. This review summarizes information on clinically available FLT3 inhibitors for the management of AML with FLT3 mutations.


Keywords: Acute myeloid leukemia, FLT3-ITD, FLT3-TKD, Tyrosine kinase inhibitor, Gilteritinib, Midostaurin

Article

Review Article

Blood Res 2022; 57(S1): S32-S36

Published online April 30, 2022 https://doi.org/10.5045/br.2022.2022017

Copyright © The Korean Society of Hematology.

FLT3 mutations in acute myeloid leukemia: a review focusing on clinically applicable drugs

Jae-Sook Ahn1,2, Hyeoung-Joon Kim1,2

1Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University, Gwangju,
2Genomic Research Center for Hematopoietic Diseases, Chonnam National University Hwasun Hospital, Hwasun, Korea

Correspondence to:Jae-Sook Ahn, M.D., Ph.D.
Department of Internal Medicine, Chonnam National University Hwasun Hospital, Chonnam National University, 322 Seoyang-ro, Hwasun-eup, Hwasun-gun, Hwasun 58128, Korea
E-mail: f0115@jnu.ac.kr

Received: January 19, 2022; Revised: February 28, 2022; Accepted: February 28, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

FMS-like tyrosine kinase 3 (FLT3) mutations, the most frequently detected genetic aberrations in patients with acute myeloid leukemia (AML), are identified in approximately 30% of patients with newly diagnosed AML and are more common in patients with normal karyotypes. Since the discovery of FLT3 mutations in AML, clinical trials have been actively conducted in patients with FLT3 mutated AML, and FLT3 inhibitors have been introduced into clinical practice. The current standard treatment for patients with newly diagnosed FLT3-mutated AML is 7+3 induction chemotherapy combined with midostaurin. Additionally, gilteritinib is more effective than salvage chemotherapy for relapsed or refractory FLT3-mutated AML. Ongoing trials are expected to provide additional treatment options depending on the disease state and patient vulnerability. This review summarizes information on clinically available FLT3 inhibitors for the management of AML with FLT3 mutations.

Keywords: Acute myeloid leukemia, FLT3-ITD, FLT3-TKD, Tyrosine kinase inhibitor, Gilteritinib, Midostaurin

Table 1 . Summary of clinically applicable FLT3 inhibitors for FLT3-mutated AML..

Patient eligibilityDisease statusDrugTarget mutated lesionRepresentative trialUsageBenefitApprovalIn Korea
Intensive induction eligibleNewly diagnosedMidostaurinFLT3-ITD/TKDRATIFY (Phase3) [7]Combination with 7+3 induction and consolidation chemotherapyMedian OS (74.7 vs. 25.6 mo), P=0.009FDA, EMAAvailable
MaintenanceMidostaurinFLT3-ITD/TKDRATIFY (Phase3) [7]Maintain until relapse for up to 12 monthsas the extension of RATIFY trialEMANot available
Post-HCT maintenanceSorafenibFLT3-ITDSORMAIN (Phase 2) [9]Maintain untilrelapse for up to 24 months2-year RFS (85 vs. 53%), P=0.002Off-labelNot available
Relapsed or refractoryGilteritinibFLT3-ITD/TKDADMIRAL (Phase 3)MonotherapyMedian OS (9.3 vs. 5.6 mo), P<0.001FDA, EMAAvailable
Intensive induction ineligibleNewly diagnosedSorafenibFLT3-ITDNCT02196857 (Phase 2) and NCT01254890 Phase 1/2) [35]Combination with azacitidineMedian OS (8.3 mo)Off-labelNot available
Relapsed or refractorySorafenibFLT3-ITDNCT01254890 [36]Combination with azacitidineResponse rate: 46%Off-labelNot available

Abbreviations: AML, acute myeloid leukemia; EMA, European Medicines Agency; FDA, Food and Drug Administration; FLT3, FMS-like tyrosine kinase 3; HCT, hematopoietic cell transplantation; ITD, internal tandem duplication; mo, months; OS, overall survival; RFS, relapse-free survival; TKD, tyrosine kinase domain..


Blood Res
Volume 59 2024

Stats or Metrics

Share this article on

  • line

Related articles in BR

Blood Research

pISSN 2287-979X
eISSN 2288-0011
qr-code Download