Gilteritinib Approved in Europe for Relapsed/Refractory FLT3+ AML

Article

The European Commission has approved gilteritinib as a single agent for the treatment of relapsed/refractory patients with FLT3-mutant acute myeloid leukemia.

Giovanni Martinelli, MD, Institute of Hematology, S.Orsola-Malpighi University Hospital, Bologna, Italy

Giovanni Martinelli, MD, Institute of Hematology, S.Orsola-Malpighi University Hospital, Bologna, Italy

Giovanni Martinelli, MD

The European Commission has approved gilteritinib (Xospata) as a single agent for the treatment of relapsed/refractory patients with FLT3-mutant acute myeloid leukemia (AML).1

The approval is based on findings from the phase III ADMIRAL trial, which showed that the median overall survival (OS) in patients who were treated with gilteritinib was 9.3 months (95% CI, 7.7-10.7) compared with 5.6 months in those who received salvage chemotherapy (95% CI, 4.7-7.3), which translated to a 36% reduction in the risk of death (HR, 0.64; 95% CI, 0.49-0.83;  P  = .0004).2  At 1 year, the OS rates were 37% for gilteritinib and 17% for chemotherapy, respectively.

“AML is a rare cancer and patients with a FLT3 mutation have a particularly poor prognosis, with a median survival of less than six months following treatment with salvage chemotherapy,” study investigator Giovanni Martinelli, MD, Institute of Hematology, S.Orsola-Malpighi University Hospital, Bologna, Italy, stated in a press release. “Gilteritinib is a new and clinically meaningful treatment option that provides a welcome advance for patients and health care professionals across the European Union.” 

The European Commission’s decision follows a positive opinion granted in September 2019 by the European Medicines Agency’s Committee for Medicinal Products for Human Use. The European approval allows gilteritinib to be administered in all European Union member countries, as well as Iceland, Norway, and Liechtenstein.

In the international, phase III ADMIRAL study, 371 adult patients with  FLT3-mutant relapsed/refractory AML were randomized 2:1 to receive gilteritinib at 120 mg daily (n = 247) or salvage chemotherapy (n = 124). Patients in both arms then underwent hematopoietic stem cell transplantation (HSCT), but only patients in the gilteritinib arm then resumed treatment with the  FLT3  inhibitor; crossover was not permitted. Additionally, salvage chemotherapy was selected prior to randomization and could be one of the following regimens: mitoxantrone, etoposide, and cytarabine; fludarabine, cytarabine, idarubicin, and G-CSF; low-dose cytarabine; and azacitidine.

To be eligible for enrollment, patients with AML must have also harbored a &#8239;FLT3-ITD or&#8239; FLT3-TKD mutation, a mean of triplicate Fridericia-corrected QT interval <450 milliseconds at screening based on central reading, and be refractory to induction chemotherapy or in untreated first relapse.

Baseline characteristics were comparable between arms. Overall, the median age was 62 (range, 19-85) and 54% of patients were female. The majority of patients had intermediate-risk cytogenetics (73%) and 88% of patients had &#8239;FLT3-ITD mutations. Additionally, 20% of patients had underwent prior HSCT, and 82% had received upfront intensive chemotherapy. Thirty-nine percent of patients had primary refractory AML without HSCT and 27% relapsed ≤6 months after composite complete remission (CRc).

The coprimary endpoints were OS and CR/CRh rate; secondary endpoints were event-free survival, CR rate, leukemia-free survival, duration of remission (DOR), CRc rate, transplantation rate, brief fatigue inventory, CRh rate, transfusion conversion rate, and transfusion maintenance rate.

Results showed that the complete remission (CR)/complete remission with partial hematologic recovery (CRh), CR with incomplete hematologic recovery, and CR with incomplete platelet recovery rates with gilteritinib compared with salvage chemotherapy were 21% versus 11%, 13% versus 5%, 26% versus 11%, and 8% versus 0%, respectively. Additionally, the CRc rate was 54% with gilteritinib and 22% with salvage therapy.

The overall response rates were 26% with salvage chemotherapy and 68% with gilteritinib, which comprised a 13% partial response rate.

Moreover, the median duration of gilteritinib exposure was significantly longer than that of salvage therapy, at 4.1 months (range, 0.1-29.1) and 0.9 months (range, 0.2-7.1), respectively. The median time to achieve CRc was 1.8 months (95% CI, 0.9-9.95) with gilteritinib and 1.1 months (95% CI, 0.8-2.9) with salvage treatment. The median DOR with gilteritinib was also significantly longer at 11.0 months compared with 1.8 months with salvage therapy.

Regarding safety, gilteritinib's tolerability profile is based on 319 patients with relapsed/refractory AML in 3 clinical trials: NCT02421939, NCT02014558, and NCT02181660.

Adverse events (AEs) that led to death occurred in 2% of patients on gilteritinib, which included cardiac arrest (1%) and differentiation syndrome and pancreatitis (n = 1 case each). The most frequent (≥5%) nonhematological serious AEs were fever (13%), dyspnea (9%), renal impairment (8%), transaminase increased (6%) and noninfectious diarrhea (5%). Moreover, the most frequent (≥5%) grade ≥3 nonhematological AEs reported in patients were transaminase increased (21%), dyspnea (12%), hypotension (7%), mucositis (7%), myalgia/arthralgia (7%), and fatigue/malaise (6%).

“Today’s approval marks a significant advance for patients living with relapsed or refractory, FLT3 mutation-positive acute myeloid leukemia,” said Andrew Krivoshik, MD, PhD, senior vice president and global therapeutic area head, Oncology Development, Astellas, the developer of gilteritinib. “We look forward to working with health authorities across the EU to bring gilteritinib to patients who need it the most, as soon as possible.”

Gilteritinib was initially approved by the FDA in November 2018; the decision was based on an interim analysis of CR/CRh rate, duration of CR/CRh, and rate of conversion from transfusion dependence to transfusion independence. The label was updated in May 2019 to include the OS data from the ADMIRAL trial.

References

  1. European Commission Approves Astellas’ XOSPATA&#8239;(gilteritinib) as a Monotherapy for Patients with Relapsed or Refractory Acute Myeloid Leukemia with a FLT3 Mutation [news release]. Tokyo, Japan: Astellas Pharma; October 25, 2019. prn.to/31Kb3RN. Accessed October 25, 2019.
  2. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib significantly prolongs overall survival in patients with &#8239;FLT3-mutated (FLT3mut+) relapsed/refractory (R/R) acute myeloid leukemia (AML): results from the Phase III ADMIRAL trial. Cancer Res. 2019;79(13 suppl; abstr CT184). doi: 10.1158/1538-7445.AM2019-CT184.
Related Videos
Catherine C. Coombs, MD, associate clinical professor, medicine, University of California, Irvine School of Medicine
Jorge J. Castillo, MD,
Catherine C. Coombs, MD, associate clinical professor, medicine, University of California, Irvine School of Medicine
Alessandra Ferrajoli, MD
Dipti Patel-Donnelly, MD, Johns Hopkins
Jasmin M. Zain, MD
Andrew Ip, MD
Sagar S. Patel, MD