All content on this site is intended for healthcare professionals only. By acknowledging this message and accessing the information on this website you are confirming that you are a Healthcare Professional. If you are a patient or carer, please visit Know AML.

The AML Hub uses cookies on this website. They help us give you the best online experience. By continuing to use our website without changing your cookie settings, you agree to our use of cookies in accordance with our updated Cookie Policy

Introducing

Now you can personalise
your AML Hub experience!

Bookmark content to read later

Select your specific areas of interest

View content recommended for you

Find out more
  TRANSLATE

The AML Hub website uses a third-party service provided by Google that dynamically translates web content. Translations are machine generated, so may not be an exact or complete translation, and the AML Hub cannot guarantee the accuracy of translated content. The AML Hub and its employees will not be liable for any direct, indirect, or consequential damages (even if foreseeable) resulting from use of the Google Translate feature. For further support with Google Translate, visit Google Translate Help.

Steering CommitteeAbout UsNewsletterContact
LOADING
You're logged in! Click here any time to manage your account or log out.
LOADING
You're logged in! Click here any time to manage your account or log out.
2019-10-21T12:26:27.000Z

SOHO 2019 | FLT3 inhibitors in AML

Oct 21, 2019
Share:

Bookmark this article

FMS-like tyrosine kinase 3 (FLT3) is a type III receptor tyrosine kinase expressed by hematopoietic progenitor cells.1 Specific binding of its ligand, FLT3-ligand (FLT3L), leads to the activation of multiple downstream signaling pathways.1 FLT3 gene mutations are one of the most common genetic abnormalities in acute myeloid leukemia (AML).2 Internal tandem duplications (ITD) and tyrosine kinase domain (TKD) mutations of FLT3 occur in approximately 30% of all cases of AML.3 FLT3-ITD is the most common FLT3 mutation and presents with high leukemic burden and has poor prognosis with a significant impact on disease management for patients with AML.3

At the 2019 meeting of the Society of Hematologic Oncology (SOHO), Catherine Smith of the University of California San Francisco, US, discussed therapeutic targeting of FLT3 in AML, presenting the current landscape of approved and investigational FLT3 inhibitors.4

Midostaurin

Midostaurin is a first-generation, small-molecule tyrosine kinase inhibitor (TKI) and is approved by the FDA and EMA for the treatment of FLT3-mutated AML.5,6  The RATIFY study (NCT00651261) was an international, randomized, placebo-controlled, phase III trial of midostaurin in combination with chemotherapy (previously reported by the AML Global portal) which demonstrated that the addition of midostaurin to standard chemotherapy significantly prolonged overall survival (OS) and event-free survival (EFS) in patients with AML and a FLT3 mutation. In this study, 717 newly-diagnosed patients with FLT3-positive AML (median age = 47.9 years, range 18–59 years) were randomly assigned to receive induction and consolidation chemotherapy with intravenous daunorubicin and cytarabine plus either placebo (n= 357) or midostaurin (n = 360) 50 mg orally twice daily on days 8–21 of each cycle of induction and consolidation chemotherapy.7 OS was significantly longer in the midostaurin group than in the placebo group (hazard ratio [HR] for death, 0.78; p= 0.009), as was EFS (HR for event or death, 0.78; p =0.002). These results led to induction chemotherapy with midostaurin as the new standard of care for adult patients with newly diagnosed FLT3-mutant AML.

Quizartinib

Quizartinib is a second-generation class III receptor TKI with improved selectivity and potency, compared with the first-generation FLT3 inhibitors. The initial open-label international multicenter single-arm phase II trial (NCT00989261) evaluated quizartinib in patients with morphologically documented primary AML or secondary AML (previously reported by the AML Global Portal).8 Patients were divided into two groups as follows: cohort one included 157 patients (age ≥ 60 years) with relapsed/refractory (R/R) AML within one year after first-line therapy; cohort two included 176 patients (age ≥ 18 years) with R/R AML following salvage chemotherapy or hematopoietic stem cell transplantation (HSCT). Quizartinib was administered once daily as an oral solution in 28-day treatment cycles. Endpoints were complete response (CR) and composite CR (CRc), defined as the combination of CR, CR with incomplete platelet recovery (CRp), and CR with incomplete hematologic recovery (CRi). In this study, 56% of FLT3-ITD-positive patients in cohort one and 46% of FLT3-ITD-positive patients in cohort two achieved CRc.

The pivotal phase III QuANTUM-R study (NCT02039726) evaluated quizartinib monotherapy compared with salvage chemotherapy in patients with R/R FLT3-ITD AML (previously reported by the AML Global portal).9 Patients were refractory or relapsed after ≥ 1 cycle of a standard anthracycline-containing or mitoxantrone-containing AML therapy with a duration of response (DOR) ≤ 6 months, with or without prior allogeneic hematopoietic stem cell transplant (allo-HSCT). Randomization was 2:1 to receive quizartinib 60 mg (n= 245) or one of three preselected investigator’s choice SC regimens (n= 122): low-dose cytarabine (LoDAC); mitoxantrone, etoposide, and intermediate-dose cytarabine (MEC); or fludarabine, cytarabine, and granulocyte-colony stimulating factor (G-CSF) with idarubicin (FLAG-IDA). At a median follow-up of 23.5 months, the median OS was 6.2 months (95% CI, 5.3–7.2) for quizartinib compared with 4.7 months (95% CI, 4.0–5.5) with SC (HR, 0.76; 95% CI, 0.58-0.98; p= 0.02).

Quizartinib is currently under review by the EMA, however, in May 2019 the FDA Oncologic Drug Advisory Committee did not support approval of quizartinib for the treatment of adults with FLT3-ITD-positive R/R AML.10

Gilteritinib

Gilteritinib is a potent, oral type I FLT3 inhibitor. Data from a phase I/II dose-escalation study (NCT02421939; previously reported by the AML Global Portal) revealed that gilteritinib monotherapy was well tolerated and generated frequent, prolonged, clinically important responses in FLT3-mutated patients with R/R AML.

In the phase III ADMIRAL study (NCT02421939), patients with confirmed FLT3-mutant AML (FLT3-ITD and/or FLT3-TKD D835 or I836 mutations) refractory to induction chemotherapy or in untreated first relapse were randomized (2:1) to receive 120 mg/day gilteritinib or pre-randomization selected SC: LoDAC, azacytidine (AZA), MEC, or FLAG-IDA.11 Patients randomized to gilteritinib had significantly longer OS (9.3 months) compared with SC (5.6 months; HR for death=0.637; p= 0.0007). The CR/CRh rates for gilteritinib and SC were 34.0% and 15.3%, respectively (p= 0.0001); CR rates were 21.1% and 10.5%. The median duration of response was 11 months in the gilteritinib arm and 1.8 months in the SC arm.

Based on interim analysis of the ADMIRAL trial, In November 2018 the FDA approved gilteritinib for treatment of adult patients with R/R AML with an FLT3-ITD or D835/I836 mutation.12 Then in September 2019, the EMA CHMP recommended that gilteritinib is granted Market Authorisation in Europe for the treatment of FLT3-mutated R/R AML.13 The results of the ADMIRAL trial changed the treatment paradigm for salvage therapy of R/R FLT3 mutant AML and gilteritinib has become the new standard of care.

Conclusions

The approvals of midostaurin in the frontline setting and gilteritinib in the salvage setting represent a new standard of care for patients with FLT3-mutated AML. However, despite this progress, many questions still remain regarding how best to incorporate the FLT3 inhibitors into the treatment of AML as well as the ubiquitous clinical problem of development of resistance to FLT3 inhibitors.

  1. Levis M. FLT3/ITD AML and the law of unintended consequences. Blood 2011 117:6987-6990; DOI:https://doi.org/10.1182/blood-2011-03-340273
  2. Small D. Am Soc Hematol Educ Program. 2006;178–84.
  3. Daver N et al., Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. DOI: 2019;33:299–312
  4. Smith C. Use of FLT3 Inhibitors in AML. Oral presentation. 2019 Sep 12. SOHO annual meeting, Houston, US.
  5. Rydapt Summary of Product Characteristics.  Available at:https://www.ema.europa.eu/en/documents/product-information/rydapt-epar-product-information_en.pdf [Assessed 01 October 2019]
  6. Rydapt Prescribing Information. Available at:https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/207997s000lbl.pdf [Assessed 01 October 2019]
  7. Stone R et al., Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N Engl J Med. 2017;377:454–64
  8. Cortes J et al., Quizartinib, an FLT3 inhibitor, as monotherapy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2018;19:889–903. DOI:https://doi.org/10.1016/S1470-2045(18)30240-7
  9. Cortes J et al., Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019;20:984–997. DOI:10.1016/S1470-2045(19)30150-0
  10. FDA Briefing Document Oncologic Drugs Advisory Committee (ODAC) Meeting May 14, 2019 NDA 212166 Quizartinib Applicant: Daiichi-Sankyo, Inc. https://www.fda.gov/media/124896/download [Accessed 25 September 2019]
  11. Perl A et al., Abstract CT184: Gilteritinib significantly prolongs overall survival in patients with FLT3-mutated (FLT3mut+) relapsed/refractory (R/R) acute myeloid leukemia (AML): Results from the Phase III ADMIRAL trial. Cancer Res. 2019;79:13 Supplement DOI: 10.1158/1538-7445.AM2019-CT184
  12. FDA approves gilteritinib for relapsed or refractory acute myeloid leukemia (AML) with a FLT3 mutatation. https://www.fda.gov/drugs/fda-approves-gilteritinib-relapsed-or-refractory-acute-myeloid-leukemia-aml-flt3-mutatation. [Accessed 25 September 2019]
  13. European Medicines Agency: Xospata.https://www.ema.europa.eu/en/medicines/human/summaries-opinion/xospata. [Accessed 25 September, 2019]

Newsletter

Subscribe to get the best content related to AML delivered to your inbox