Updates in AML Maintenance Therapy

CME

Updates in AML Treatment: Multidisciplinary Strategies to Optimize Maintenance Therapy

Physicians: Maximum of 1.00 AMA PRA Category 1 Credit

Released: November 22, 2023

Expiration: November 21, 2024

Brian A. Jonas
Brian A. Jonas, MD, PhD, FACP

Activity

Progress
1 2
Course Completed

Introduction

In this module, Brian A. Jonas, MD, PhD, FACP, reviews the latest data on the evolving role of maintenance therapy following chemotherapy or transplant to mitigate the risk of relapse for patients with acute myeloid leukemia (AML).

Please note that the slide thumbnails in this activity link to the PowerPoint slideset associated with this program, which also can be found here. The slideset can be downloaded by clicking on any of the thumbnails within the activity.

Clinical Care Options plans to measure the educational impact of this activity. Some questions will be asked twice: once at the beginning of the activity and once again after the discussion that informs the best choice. Your responses will be aggregated for analysis, and your specific responses will not be shared. Thank you in advance for helping us assess the impact of this education.

For those providing patient care, how many patients with AML do you see in a typical month?

 In patients with AML, which of the following characteristics is associated with a higher risk for relapse?

A 58-year-old male was diagnosed with AML 4 months ago. He immediately received 7+3 (anthracycline- and cytarabine-based induction chemotherapy) and achieved remission with full count recovery on Day 31. After discussing the advantages and disadvantages of consolidative allogeneic hematopoietic stem cell transplant (alloHSCT) in first remission for intermediate-risk AML, the patient opted for consolidation and received 3 cycles of intermediate-dose cytarabine. Postcycle bone marrow biopsy showed ongoing complete remission (CR) without measurable residual disease (MRD). The patient has declined transplantation and enrollment on a clinical trial.

What treatment would you recommend for this patient?

A 59-year-old female with hypertension and hepatic steatosis presented with bruising and epistaxis. Her labs reveal a white blood cell count of 150,000/µL, hemoglobin of 7.0 g/dL, platelet count of 8,000/µL, circulating immature cells, and absolute neutrophil count of 600/µL. Bone marrow biopsy shows 65% abnormal monocytic myeloblasts, consistent with AML. Cytogenetic analysis showed normal female karyotype, and mutational analysis showed FLT3-ITD (variant allele frequency of 35%) and IDH2 mutations. 


She receives a cycle of induction chemotherapy plus midostaurin, experiences brisk count recovery, achieves remission, and goes on to receive alloHSCT. She asks what options are available to improve her likelihood of survival following transplantation.

Which of the following would be the most suitable approach to the management of this patient following transplantation?

A patient with newly diagnosed FLT3-ITD‒positive AML received quizartinib plus 7+3 during induction followed by high-dose cytarabine plus quizartinib consolidation and quizartinib maintenance therapy. In cycle 11 of quizartinib maintenance, the patient has an ECG that shows QT prolongation of 492 ms (grade 2).

Which of the following is the most appropriate strategy for managing this patient’s QT prolongation?

2022 ELN Risk Categorization for AML

Before discussing the role of maintenance therapy in the management of AML, it is important to discuss how we identify patients who have a high risk of relapse. One of the long-standing ways that we do this is through risk categorization.

The most commonly used system for risk categorization, I would argue, is the European LeukemiaNet (ELN) system, which was most recently updated in 2022.1 The 2022 ELN risk stratification system for AML is based on patients’ genetics or molecular mutations and the cytogenetic features of their disease. This information is usually obtained at the time of diagnosis and often checked throughout the time of their treatment course.

According to the ELN system, patients with AML are categorized as having favorable, intermediate, or adverse risk. For example, mutated NPM1 on its own is considered favorable risk, whereas an NPM1 mutation with an accompanying FLT3-ITD mutation is considered intermediate risk. Examples of genetic abnormalities associated with adverse risk include RUNX1, ASXL1, and EZH2.

Patients who are favorable risk typically have a lower risk of relapse and often are treated with approaches that do not require bone marrow or stem cell transplant. Patients who are intermediate or adverse risk often will receive alloHSCT as part of their treatment course because of their increased risk of relapse.

Validation of ELN 2022 Risk Stratification of AML

This is a recent study that validated ELN 2022 by looking at relapse-free survival (RFS) and OS according to risk group.2 This shows that the patients with AML stratify nicely into the 3 groups of favorable, intermediate, and adverse risk in both the RFS curves and the OS curves.

 When you look at RFS on the left side, you will see that after 5 years, approximately 85% of patients who have an adverse risk in cytogenetics (orange curve) are going to have either relapsed or died. So, this is definitely a group with an increased risk of relapse.

Measurable Residual Disease

With morphologic analysis, a pathologist typically can measure on the level of 1 leukemic cell per 100 cells.3 Detecting levels below that requires testing that is more sensitive—and that is where MRD testing comes into play. MRD is a way for us to detect AML below the level of detection of a typical pathology assay, and this can be done using numerous modalities, including flow cytometry‒based systems and quantitative real-time polymerase chain reaction, which can detect down to levels of 1 leukemic cell per 10,000 cells or lower depending on the technique and marker. There also are emerging next-generation sequencing‒based techniques and droplet digital polymerase chain reaction, which can detect levels even lower than the typical MRD by flow cytometry.

In relation to relapse, the idea is that patients with MRD after initial treatment are more likely to relapse, and patients who have no MRD or levels that are below the limit of detection of our assays are less likely to relapse.

MRD and Survival in AML: Meta-analysis of 81 Publications

Here are the results of a meta-analysis of many studies looking at MRD status at induction or during or after consolidation and the impact of survival and relapse.4 You can see on the OS curves on the left and the disease-free survival (DFS) curves on the right that patients who are MRD negative fare much better than patients who are MRD positive (5-year OS: 68% vs 34%; 5-year DFS: 64% vs 25%). There is a big difference in these curves favoring the MRD-negative population.

So, back to the original concept of this section, identifying patients who are more likely to relapse: These would be patients who have higher-risk disease status at baseline and patients who are MRD positive.

Recommended Postchemotherapy Maintenance

The goal of initial induction is to bring AML into remission, and treatment can vary greatly depending on patient characteristics. Fit patients typically will be treated with intensive chemotherapy with the addition of targeted therapy (eg, FLT3) as clinically appropriate.5 Patients ineligible for intensive chemotherapy usually are prescribed prescribed a hypomethylating agent (HMA) plus venetoclax, targeted therapy with or without an HMA, or low-dose cytarabine with glasdegib. After remission, patients may go on to receive consolidation therapy consisting of limited-duration chemotherapy or alloHSCT. In some situations, maintenance therapy may be used after consolidation during a longer period of time to reduce the risk of relapse.

Before getting into the data, I’d like to begin with a summary of the most recent National Comprehensive Cancer Network (NCCN) guidelines on postchemotherapy maintenance for patients with AML. Patients with intermediate- or adverse-risk disease who received intensive chemotherapy and who have their AML in remission with no transplant planned are recommended to receive maintenance therapy until progressive disease or unacceptable toxicity. Patients could complete no consolidation, some consolidation, or a recommended consolidation course. Currently, the category 1 recommendation for maintenance therapy in this setting is oral azacitidine. There are also data for the use of azacitidine or decitabine, the more conventional HMAs, as postchemotherapy maintenance. It is important to note that oral azacitidine is not intended to replace consolidation chemotherapy or transplant. There are no data to suggest that it can be used in lieu of those approaches. Those approaches should still be done if the patient is a candidate.

For patients with a history of FLT3-ITD mutation who received quizartinib as frontline therapy and who do not have alloHSCT planned, there is now a recommendation for the use of quizartinib maintenance therapy.

HOVON97: Azacitidine Maintenance After Intensive Chemotherapy

First, let’s look at the data from the randomized phase III HOVON97 study, which looked at azacitidine maintenance vs observation after intense chemotherapy in 116 patients.6 Median DFS favored azacitidine maintenance vs observation, with a median DFS of 15.9 months vs 10.3 months (P = .04). OS, on the other hand, was not statistically significantly different between the 2 arms (P = .21), although there was a numerical advantage in the azacitidine maintenance arm vs observation (12-month estimated OS: 82% vs 63%).

QUAZAR AML-001 Oral Aza in AML: Study Design

The HOVON97 data suggested a beneficial role for HMA maintenance in some patients after chemotherapy. Subsequently, an oral azacitidine formulation was developed and tested in the QUAZAR AML-001 trial.

QUAZAR AML-001 was a randomized, double-blind, placebo-controlled phase III trial in 472 patients 55 years of age and older with AML and intermediate or poor cytogenetics who had achieved a CR/CRi after intensive chemotherapy with or without consolidation and were not eligible for transplant.7,8

Patients were randomized to receive either oral azacitidine 300 mg daily for 14 days every 4 weeks or placebo for 14 days every 4 weeks, and they had to begin therapy within 4 months of achieving a CR.

Response was assessed every 3 cycles, and if patients achieved or maintained a CR/CRi, they would continue treatment. If there was a residual blast population of 5% to 15%, they could have an optional treatment escalation, including both azacitidine and placebo, and if they had 15% or more blasts, they would have to discontinue treatment. Patients otherwise would continue receiving treatment until death, withdrawal of consent, and/or loss to follow-up.

The primary endpoint for QUAZAR AML-001 was OS, and the key secondary endpoint was RFS.

QUAZAR AML-001 Oral Aza in AML: Long-term Overall Survival

QUAZAR AML-001 met its primary endpoint with a superior median OS for oral azacitidine vs placebo of 24.7 months vs 14.8 months (HR: 0.7; P = .0009).7 At 3 years, the estimated OS was 37.4% vs 27.9%, which was an absolute difference of approximately 10%. These results led to the FDA approval of oral azacitidine for continued treatment of adult patients with AML who achieved first CR/CRi following intensive induction chemotherapy and are not able to complete intensive curative therapy.9

Looking at subgroups, patients with NPM1-mutated AML did quite well on the oral azacitidine maintenance, with a significantly longer median OS vs placebo (47.2 months vs 15.9 months).10 NPM1 mutation status is overall prognostically favorable and independently predictive of increased OS with oral azacitidine.

QUAZAR AML-001: OS for Patients With MRD-Positive Disease at Baseline

Patients who were MRD positive at baseline had improved survival with oral azacitidine vs placebo (14.6 months vs 10.4 months).11 Something that was interesting about this data set is that there were patients who actually responded—in the sense that they were able to clear their MRD while they were receiving oral azacitidine. In fact, some cleared their MRD while receiving placebo. The MRD clearance rate was 37% with oral azacitidine and 19% with placebo. Many of these responses were early (≤6 months), although there were responses seen after 6 months of treatment

QUAZAR AML-001: Safety

In terms of safety, there were more adverse events (AEs) in the oral azacitidine arm compared with the placebo arm.8 The AEs that stand out included gastrointestinal (GI) toxicities such as nausea, vomiting, and diarrhea, as well as hematologic AEs such as neutropenia, thrombocytopenia, anemia, febrile neutropenia, and leukopenia. Hematologic malignancies were associated with more grade 3/4 AEs. Of note, although the GI toxicities were more numerous, there were not many grade 3/4 toxicities, but the grade 3/4 events generally were higher than what was seen in the placebo arm.

In QUAZAR AML-001, AEs led to discontinuation in 13% of patients receiving oral azacitidine vs 4% receiving placebo. Similarly, there were more treatment-emergent AEs (TEAEs) that led to dose reduction (16% vs 3%) or dose interruption (43% vs 17%) on the oral azacitidine arm vs the placebo arm.

Managing Key AEs With Oral Azacitidine

Here are the recommendations for managing neutropenia and thrombocytopenia with oral azacitidine treatment.9 When the neutrophil count is <500 cells/mL, the recommendation is to hold oral azacitidine until improved to >500 cells/mL, and then one can resume at the same dose. For febrile neutropenia with neutrophil count <1000 cells/mL, treatment should be held and then resume after resolution. If febrile neutropenia occurs in 2 consecutive cycles, then this prompts a dose reduction of the oral azacitidine to 200 mg. If the febrile neutropenia persists after dose reduction, the recommendation is to reduce the dosing cycle by 7 days. After adjustment in dose and cycle length, persistent febrile neutropenia would prompt a discontinuation of the oral azacitidine. There are similar recommendations for thrombocytopenia with platelets <50 cells/mL with bleeding. On the first occurrence, oral azacitidine should be held and then resumed after resolution. If the thrombocytopenia occurs in 2 consecutive cycles, the treatment needs to be held and then resumed at 200 mg after resolution, and the treatment cycle should be shortened by 7 days. If thrombocytopenia happens again, then treatment should be discontinued.

In practice, GI toxicity can be a bothersome AE. The recommended management approaches are similar to what I described for cytopenias. If one develops grade 3/4 GI toxicity, then you would need to hold the drug and resume at the same dose once it resolves to grade 1. If it recurs, then reduce the dose to 200 mg, and if it recurs again, drop to a 7-day cycle. If the severe GI toxicity persists despite these measures, oral azacitidine will need to be discontinued. In my own practice, I give patients an antiemetic before each dose of oral azacitidine to reduce the chance that patients will experience nausea or vomiting. Loperamide can be useful for patients who have diarrhea.

QuANTUM-First: Quizartinib + Chemotherapy in Newly Diagnosed FLT3-ITD+ AML

I mentioned earlier that the NCCN guidelines now recommend the use of quizartinib as maintenance therapy, and that is based on the recently reported QuANTUM-First study.

QuANTUM-First was a randomized, double-blind, placebo-controlled phase III trial that looked at quizartinib plus chemotherapy in 539 patients 18-75 years of age with newly diagnosed FLT3-ITD‒positive AML.12,13 Patients received up to 2 cycles of induction chemotherapy with cytarabine and daunorubicin or idarubicin—the classic 7+3 regimen—with quizartinib or placebo given on Days 8-21 of each cycle. The patients with a CR could get up to 4 cycles of high-dose cytarabine consolidation with quizartinib (or placebo) and/or transplant per investigator discretion and institution policies. Then, patients who either finished consolidation or went to transplant could continue dosing of quizartinib monotherapy for up to 3 years or 36 cycles.

The primary endpoint of the study was OS. Secondary endpoints included event-free survival, response rates, and MRD negativity. Other exploratory endpoints included RFS and duration of CR.

QuANTUM-First: OS (Primary Endpoint)

QuANTUM-First met its primary endpoint of improved OS. The blue line represents quizartinib, with a median OS of 31.9 months vs 15.9 months with placebo (HR: 0.78; P = .032). The absolute difference was 16.8 months.

These data led to FDA approval of quizartinib for patients with newly diagnosed AML with FLT3-ITD mutation, in combination with standard cytarabine and anthracycline induction and cytarabine consolidation, and as maintenance monotherapy after consolidation chemotherapy.14

QuANTUM-First Post Hoc Analysis*: OS in Patients With CR1 With or Without Transplant

This is a post hoc analysis of OS in patients with CR with or without transplant. This is a descriptive analysis, but it is interesting.13

On the left is the OS curve for patients who have achieved a CR and received a transplant on first remission. On the right are patients who had a CR who did not receive a transplant on first remission. Both figures are numerically in favor of the quizartinib arm, with HRs of approximately 0.6 in both analyses. This suggests that 36 months of quizartinib is providing benefit with or without transplant. We will discuss additional data on posttransplant quizartinib maintenance later in this module.

QuANTUM-First: Safety

Quizartinib, like all drugs, has some toxicity. On the QuANTUM-First trial, there was more neutropenia in the quizartinib arm vs the placebo arm (20% vs 10% all grades; 18% vs 9% grade ≥3) and more febrile neutropenia (44% vs 42% all grades; 43% vs 41% grade ≥3).12,13

These are some potential cardiac AEs specific to quizartinib. There was more QTc prolongation with quizartinib compared with placebo. In total, 13.6% of patients receiving quizartinib had any form of treatment-emergent QTc prolongation compared with 4.1% of patients receiving placebo. Approximately 2.3% of patients receiving quizartinib had QTc that was >500 ms, for example, and 10% had a QTc increase from baseline of >60 ms. Other cardiac TEAEs reported with quizartinib include cardiac arrest/ventricular fibrillation and ventricular tachycardia, which occurred in <1% of patients.

In this trial, 2 patients had cardiac arrest with recorded ventricular fibrillation in the setting of severe hypokalemia while receiving quizartinib, and 1 patient who received quizartinib died in their sleep.

QTc prolongation led to treatment discontinuation for 2 patients receiving quizartinib.

Managing Cardiac AEs With Quizartinib

Quizartinib has a new approval, and the QT prolongation and other cardiac AEs are potentially fatal, so it is important for healthcare professionals to be aware of the monitoring and management recommendations.

ECGs should be performed prior to initiation of induction or consolidation chemotherapy, once weekly, and then as clinically indicated. ECGs also are recommended prior to initiation of maintenance therapy, then once weekly for at least the first month and following dose initiation and escalation. To manage QT prolongation, dose modification is not needed unless QTcF is >480 ms. For QTcF 481-500 ms, we would reduce but not interrupt the quizartinib dose. If QTcF is >500 ms, we would interrupt the quizartinib and then resume at a lower dose until QTcF is <450 ms. If QTcF >500 occurs more than once, we would permanently discontinue the drug.

It is also important to note that quizartinib has a Risk Evaluation and Mitigation Strategy program that healthcare professionals need to enroll in because of the risk of QT prolongation.

Select Ongoing Postinduction Chemotherapy Maintenance Trials

I would like to wrap up this section with a review of a few ongoing postinduction chemotherapy maintenance trials. This is by no means every trial that is out there, but it is an interesting group.

Several trials are evaluating targeted therapies as maintenance following induction chemotherapy. The FLT3 inhibitor gilteritinib is being compared with midostaurin in the phase III HOVON 156 AML trial (NCT04027309) and as monotherapy maintenance in the phase III GOSSAMER trial (NCT02927262). Quizartinib maintenance is being evaluated in patients with untreated non–FLT3-ITD AML in a phase II trial. Crenolanib is being compared with midostaurin in a phase III trial of patients with newly diagnosed FLT3-mutated AML (NCT03258931). For newly diagnosed IDH-mutated AML, the HOVON 150 AML trial (NCT03839771) will be evaluating the efficacy of the IDH inhibitors ivosidenib or enasidenib in combination with induction and consolidation chemotherapy and then as maintenance therapy. I am interested to see how these trials will turn out.

Another interesting trial is a phase I trial (NCT05010772) that is aiming to personalize maintenance based on the patient’s specific genotype. This trial will combine decitabine with venetoclax if there are no particular mutations. Patients will receive decitabine with gilteritinib if they have a FLT3 mutation or decitabine plus ivosidenib or enasidenib if they have an IDH1 or IDH2 mutation, respectively. So, this is kind of a personalized medicine approach to maintenance, and it will be interesting to see how this trial pans out.

There is also a question about whether we can improve on oral azacitidine outcomes. The VIALE-M trial (NCT04102020)—where the M stands for maintenance—is ongoing. VIALE-M is comparing venetoclax plus oral azacitidine vs oral azacitidine alone in patients with newly diagnosed AML in CR/CRi after intensive induction and consolidation chemotherapy. The primary endpoints are safety and RFS. VIALE-M is active and no longer recruiting, so data are eagerly anticipated.

Guideline-Recommended Posttransplant Maintenance

Next, I want to discuss maintenance in the posttransplant setting. The current NCCN guidelines recommend the use of FLT3 inhibitors as maintenance therapy for patients who underwent alloHSCT, are in remission, and have a history of a FLT3 mutation.5 The panel recommends consideration of sorafenib, midostaurin, gilteritinib, and quizartinib. Of note, quizartinib is only for patients with FLT3-ITD‒positive disease.

HMAs as Posttransplant Maintenance

There is literature on using HMAs as posttransplant maintenance, but the data have not translated into standard practice.

Oral decitabine and IV azacitidine have been investigated in posttransplant maintenance. An early study evaluating IV azacitidine compared with supportive care has shown improvements in RFS and OS, but the results were not statistically significant.15 Oral azacitidine also has shown promise in a single-arm study. Other favorable studies have shown reduction in the rate of relapse with decitabine,16 but again it has not necessarily led to widespread use of these drugs in the posttransplant setting.

One interesting study is the RELAZA2 trial, which looked at patients who developed kind of an MRD relapse state, either post induction or post transplant. This study showed that the initiation of azacitidine could delay or prevent frank hematologic relapse.17

SORMAIN: Sorafenib Maintenance After AlloHSCT in FLT3-ITD AML: Study Design

There are several studies in the literature showing the benefit of the FLT3 inhibitor sorafenib in the posttransplant setting for patients with AML. One of these trials was the SORMAIN trial. SORMAIN was a randomized, double-blind phase II trial that evaluated sorafenib maintenance in 83 patients with FLT3-ITD‒positive AML who underwent alloHSCT.18 Patients 60-100 days post transplant and in remission with normal peripheral blood counts were randomized to receive sorafenib 2 x 400 mg daily or placebo for up to 24 months. We also should note that this study was closed early because of slow accrual.

The primary endpoint for this trial was RFS, which was defined as death from any cause or relapse of AML. Secondary endpoints included OS, RFS and OS in patients with NPM1-mutated vs wild-type disease, RFS and OS based on FLT3-ITD ratio, safety, and biomarker analysis.

SORMAIN: RFS and OS

The primary endpoint of RFS improvement was met despite the study being stopped early because of slow accrual. The median RFS was not reached vs 30.9 months for sorafenib vs placebo. The HR was 0.39 (P = .013), and this was statistically significant in favor of sorafenib vs placebo. The 24-month RFS estimates were 85% for sorafenib vs 53.3% for placebo.

Median OS was not reached in both arms of the trial, with an HR of 0.516 in favor of sorafenib (P = .0855). The 24-month estimated OS with sorafenib was 90.5% vs 66.2% in the placebo arm (HR: 0.241; P = .007).

So, this really is a positive trial showing a benefit to sorafenib maintenance in patients with FLT3-ITD‒positive AML. Sorafenib can be difficult to use, and dose reductions due to AEs are common. On the SORMAIN trial, 48% of patients receiving sorafenib vs 40% receiving placebo had dose reductions, and 22% vs 5% of patients had treatment discontinuation due to AEs. Common AEs on the sorafenib arm were graft-vs-host disease (GvHD), infections, electrolyte alterations, and GI or skin toxicity. The most common drug-related AEs were electrolyte alterations (7.1%), GI toxicities (4.8%), skin toxicities (4.8%), and infections (2.4%).

Sorafenib Maintenance in FLT3-ITD+ AML After AlloHSCT: 5-Yr Follow-up

A study similar to SORMAIN was performed in China. This was an open-label, randomized phase III trial comparing sorafenib 400 mg twice daily with placebo from 30-60 days post transplant.19,20

The 5-year follow-up of this trial was published recently, and it was also positive.20 The 5-year OS HR of 0.55 was statistically significant (P = .011) in favor of sorafenib vs placebo. The cumulative incidence of relapse was lower with sorafenib (5-year HR: 0.33; P = .0003), and the leukemia-free survival was also better for sorafenib vs placebo (5-year HR: 0.47; P = .0007).

As I mentioned with the SORMAIN study, AEs can be problematic with sorafenib. In this trial, there were more AEs with sorafenib vs placebo. Common AEs include infections, GvHD, and hematologic toxicities. Overall, however, this is a second study showing that sorafenib maintenance improves outcomes for patients with FLT3-ITD‒positive AML after alloHSCT.

RADIUS: Midostaurin Maintenance After AlloHSCT in FLT3-ITD+ AML

RADIUS was another randomized phase II trial that enrolled patients with FLT3-ITD‒positive AML in CR1 after alloHSCT. This trial compared midostaurin plus standard-of-care maintenance with standard of care alone after alloHSCT. Patients received maintenance for up to 12 cycles, and the primary endpoint was RFS 18 months post alloHSCT. Secondary endpoints of importance were OS, RFS at 24 months post alloHSCT, and safety.

This was a negative trial. The midostaurin arm potentially had a numerical benefit, with 18-month RFS 89% vs 76% (HR: 0.46), but this was not a statistically significant difference (P = .2655). The story was similar for OS. The estimated 24-month OS was 85% for the midostaurin arm vs 76% for standard of care (HR: 0.58; P = .3418).

QuANTUM-First: OS in Patients Who Received AlloHSCT in CR1 by MRD Status

As I mentioned earlier, there are data for the use of quizartinib in the posttransplant setting. This again comes from the QuANTUM-First trial, where patients continued to receive quizartinib for up to 36 cycles as maintenance.12 Here are the results of a post hoc analysis showing OS based on MRD status in patients who received quizartinib and alloHSCT.21 In patients who were MRD negative, the median OS was not reached for both quizartinib and placebo, but the results were numerically in favor of quizartinib (HR: 0.7). In patients who were MRD positive, the OS again favored quizartinib vs placebo (HR: 0.47), but again—similar to patients who were MRD negative—the median survival was not reached for either arm.

MORPHO: Gilteritinib vs Placebo as Posttransplant Maintenance in FLT3-ITD+ AML

MORPHO was an important study presented at the European Hematology Association 2023 Congress. This multicenter, randomized, double-blind, placebo-controlled phase III trial evaluated gilteritinib vs placebo as posttransplant maintenance in FLT3-ITD‒positive AML.22 Patients in CR who were >30 but <90 days after transplant were randomized after transplant to receive either gilteritinib 120 mg/day or placebo for 24 months.

The primary endpoint was RFS, and key secondary endpoints included OS, event-free survival, incidence of GvHD, safety, and nonrelapse mortality. Marrow samples were collected for MRD analysis at various time points during the study.

MORPHO: RFS

The primary endpoint for the study (RFS) was not met. There was a benefit numerically to gilteritinib, as seen in the figure, but this was not statistically significant (HR: 0.679; P = .0518). The secondary endpoint of OS for the entire population also was not met (HR: 0.846; P = .4394).

When one looks at RFS by MRD status going into the transplant or going into treatment, the data are in favor of gilteritinib vs placebo (HR: 0.515; P = .0065) for patients who were MRD positive. For patients who were MRD negative, on the other hand, there was no benefit to the gilteritinib maintenance (P = .5750).

In terms of safety, there were more TEAEs with gilteritinib. Neutrophils were decreased more frequently (42.1% vs 15.8%), and there was more chronic GvHD (52.2% vs 42.1%). There were also more TEAEs leading to discontinuation of gilteritinib.

Choosing Between FLT3 Inhibitors

All the FLT3 inhibitors recommended by the NCCN as posttransplant maintenance are being used off-label with no FDA approval in this setting. So, this leaves the question of how to choose between the available inhibitors.

For me, this decision is largely based on the strength of the efficacy data, as well as the prior treatment. In my opinion, switching FLT3 inhibitors between the pre- and posttransplant setting is not ideal, but I feel comfortable doing it in some cases. In the first-line setting, patients with FLT3-positive AML will receive midostaurin or quizartinib. For a patient who initially received midostaurin, I think it makes a lot of sense to switch them to sorafenib in the posttransplant setting. The RADIUS trial, which evaluated midostaurin maintenance after transplant, did not meet its primary endpoint, and I do not find the data compelling enough to warrant its use. On the other hand, the data are very compelling for sorafenib based on the SORMAIN trial and the Chinese study mentioned earlier.18,20 At my institution, we primarily use sorafenib as posttransplant maintenance for patients with FLT3-ITD–positive AML. In the postchemotherapy setting, I would continue to give  a patient quizartinib maintenance if they received it initially.

Because of the improved safety profile and potency, many experts will lean toward the use of a second-generation FLT3 inhibitor such as quizartinib or gilteritinib. The data from the MORPHO trial—although primary endpoints were not met—are compelling for the use of gilteritinib for patients who are MRD positive post transplant. If I had a patient with MRD-positive AML, I would feel comfortable prescribing gilteritinib maintenance. I also would feel comfortable using quizartinib for posttransplant maintenance for patients who initially were treated with quizartinib based on the data from QuANTUM-First.12,21

Select Ongoing Trials in Posttransplant Maintenance

Let’s finish with a review of a few interesting studies in the posttransplant maintenance setting for AML. Again, a lot of these trials are testing targeted therapies, as this is an exciting area being investigated. Crenolanib, another FLT3 inhibitor, is currently being investigated in a phase II trial (NCT02400255). IDH inhibitors also are being studied in the posttransplant maintenance setting, and some potentially interesting data are emerging from these studies. A phase I trial of enasidenib posttransplant maintenance in IDH2-mutated AML reported a 2-year progression-free survival of 69%,23 and a phase I trial of ivosidenib posttransplant maintenance in IDH1-mutated AML reported a 2-year progression-free survival of 81%.24 These data are early but show promise, and we look forward to later-phase trials investigating these inhibitors.

Studies are still ongoing with HMAs in the posttransplant setting. The phase III AMADEUS study (NCT04173533) is investigating oral azacitidine vs placebo in patients in first or second CR. A phase II trial (NCT04128501) is evaluating the efficacy of azacitidine plus venetoclax in various leukemias (NCT04128501).

The phase III VIALE-T trial (NCT04161885) is also of interest. This trial, which is still recruiting, is evaluating venetoclax plus azacitidine posttransplant maintenance. Enrolled patients will plan to receive or have received alloHSCT within the past 60 days and have <10% blasts before pretransplant conditioning and <5% blasts post transplant. This trial will have 2 parts. Part 1 will be used to establish the recommended phase III dose for the combination of venetoclax plus azacitidine and best supportive care. The primary endpoint for part 1 is dose-limiting toxicity. In part 2, enrolled patients will be randomized to receive venetoclax plus azacitidine with best supportive care vs best supportive care alone for up to 24 cycles. The primary endpoint for part 2 is OS, and secondary endpoints are morphologic and composite RFS, GvHD rate, GvHD-free RFS, MRD-negativity rate, and quality of life. It will be interesting to see whether combination therapy will have a role in the posttransplant setting.

These are some interesting studies, and I think their results will be eagerly anticipated in the event that they might expand the patient populations who are offered posttransplant maintenance.

Shared Decision-making

One important question that remains about maintenance therapy is how long to continue with treatment. Right now, there is no definitive answer to this question, so we must take patient preference into consideration. It can be challenging for patients to come to terms with the possibility of receiving oral chemotherapy for the rest of their lives. Most patients do not desire that course, especially as these agents come with toxicities that they must deal with as long as they continue therapy. I would like to see a trial evaluating a fixed period (eg, 2 years) of oral azacitidine maintenance therapy to understand how much treatment is necessary to mitigate the risk of relapse. Although there are benefits to these treatments, we risk overtreating patients. I think having a finite treatment length in mind will make a lot of patients more comfortable receiving maintenance therapy.

In my practice, whether to initiate maintenance therapy, with which drug, and for how long are shared decisions I make with the patient. Beyond the indications and data for specific agents, patient preferences, goals, and beliefs are a vital part of the discussion and shared decision-making.

Conclusion

Maintenance therapy in AML is an evolving field. An increasing number of drugs are being considered standard of care today. One of the big advances was the QUAZAR AML-001 data, which led to the FDA approval of oral azacitidine as postchemotherapy maintenance for patients who are ineligible for or who decline transplant.9 There also have been great advances in the use of FLT3 inhibitors (sorafenib, quizartinib, and gilteritinib) post transplant, but we are still waiting on an FDA approval in this setting.

Of course, there also are exciting new prospects for emerging maintenance therapies. The IDH inhibitors ivosidenib and enasidenib have shown promise. There is also a potential role for venetoclax-based combinations as postchemotherapy or posttransplant maintenance therapy. In addition to these, several approaches are being explored—including immunotherapy, CAR T-cell therapies, and novel targeted agents—to decrease the risk of relapse after induction therapy or alloHSCT.

 In patients with AML, which of the following characteristics is associated with a higher risk for relapse?

A 58-year-old male was diagnosed with AML 4 months ago. He immediately received 7+3 (anthracycline- and cytarabine-based induction chemotherapy) and achieved remission with full count recovery on Day 31. After discussing the advantages and disadvantages of consolidative alloHSCT in first remission for intermediate-risk AML, the patient opted for consolidation and received 3 cycles of intermediate-dose cytarabine. Postcycle bone marrow biopsy showed ongoing CR without MRD. The patient has declined transplantation and enrollment on a clinical trial.

What treatment would you recommend for this patient?

A 59-year-old female with hypertension and hepatic steatosis presented with bruising and epistaxis. Her labs reveal a white blood cell count of 150,000/µL, hemoglobin of 7.0 g/dL, platelet count of 8,000/µL, circulating immature cells, and absolute neutrophil count of 600/µL. Bone marrow biopsy shows 65% abnormal monocytic myeloblasts, consistent with AML. Cytogenetic analysis showed normal female karyotype, and mutational analysis showed FLT3-ITD (variant allele frequency of 35%) and IDH2 mutations.


She receives a cycle of induction chemotherapy plus midostaurin, experiences brisk count recovery, achieves remission, and goes on to receive alloHSCT. She asks what options are available to improve her likelihood of survival following transplantation.

Which of the following would be the most suitable approach to the management of this patient following transplantation?

A patient with newly diagnosed FLT3-ITD‒positive AML received quizartinib plus 7+3 during induction followed by high-dose cytarabine plus quizartinib consolidation and quizartinib maintenance therapy. In cycle 11 of quizartinib maintenance, the patient has an ECG that shows QT prolongation of 492 ms (grade 2).

Which of the following is the most appropriate strategy for managing this patient’s QT prolongation?