CE / CME
Physician Assistants/Physician Associates: 1.00 AAPA Category 1 CME credit
Nurses: 1.00 Nursing contact hour
Physicians: maximum of 1.00 AMA PRA Category 1 Credit™
Pharmacists: 1.00 contact hour (0.1 CEUs)
Released: March 06, 2024
Expiration: March 05, 2025
QuANTUM-First MRD Analysis: Background and Study Design
QuANTUM-First was a randomized, double-blind, placebo-controlled phase III trial examining whether addition of the second-generation FLT3 inhibitor quizartinib to intensive chemotherapy conveyed benefit for younger (aged 75 years and younger), fit patients with newly diagnosed FLT3-ITD‒mutant AML. In contrast to the RATIFY trial of midostaurin added to chemotherapy published in 2017, which included patients with either ITD or TKD mutations,1 QuANTUM-First was restricted to those with ITD because of quizartinib selectively inhibiting ITD. The treatment regimen was a standard cytarabine and daunorubicin/idarubicin induction plus quizartinib or placebo, followed by high-dose cytarabine consolidation, again with quizartinib or placebo. Patients who achieved responses were allowed to proceed to allogeneic stem cell transplantation (alloSCT) followed by quizartinib or placebo maintenance for up to 3 months.
Previously published primary analysis at a median follow-up of 39.2 months showed that the median overall survival (OS) was significantly prolonged at 31.9 months with quizartinib vs 15.1 months with placebo (HR: 0.78; 95% CI: 0.62-0.98; P = .032).2 This analysis examined the prognostic value of FLT3-ITD measurable residual disease (MRD) status on survival.3
QuANTUM-First MRD Analysis: Sample Collection
MRD was assessed via bone marrow sampling at the time of screening and at sequential time points throughout the study until the end of treatment. Analyses attempted to determine whether obtaining MRD-negative or lower disease at certain time points correlated with clinical outcomes and whether the addition of quizartinib had any impact on the depth of response in terms of MRD at those time points. In addition, because FLT3-ITD mutations come in many forms, the investigators examined whether the presence of multiple ITDs or the length of the internal tandem duplication correlated with outcomes and whether quizartinib treatment had any impact on this correlation.3
QuANTUM-First MRD Analysis: Baseline Characteristics
QuANTUM-First differed from the prior RATIFY trial, which enrolled patients 59 years of age and younger,1 by including older patients up to 75 years of age. However, the majority (>60%) of the patients on the QuANTUM-First trial still were younger than 60 years of age. The Eastern Cooperative Oncology Group (ECOG) performance status ranged from 0-2. The majority of patients had baseline FLT3 mutational assessments and MRD analyses based on bone marrow samples.3
QuANTUM-First MRD Analysis: Association Between FLT3-ITD MRD Decrease Over Time and OS
The first analysis investigated whether MRD level correlates with OS regardless of treatment. As anticipated from a common sense point of view, patients who became MRD negative, meaning no measurable residual disease, clearly had improved OS at all time points (after induction, during treatment, and after the final consolidation).
On the right-hand side of the table, the number of patients with MRD-negative status is presented for the separate treatment arms. Quizartinib treatment led to more patients with MRD-negative status at all time points, ending with 55.8% of patients compared with 41.2% of patients with placebo after the last round of consolidation.3
QuANTUM-First MRD Analysis: OS by MRD (Cutoff Zero)
Looking at OS at all time points in both treatment arms, patients who received quizartinib had prolonged survival compared with placebo regardless of MRD status at any time point. Of patients who achieved MRD-zero status, survival was extended with quizartinib vs placebo (HR: 0.753; 95% CI: 0.432-1.311), although the median was not reached for either treatment arm.3
QuANTUM-First MRD Analysis: OS by ITD Insertion Length
Longer tandem duplications were associated with shortened survival across treatment arms (median: 17.0 months vs 40.7 months for those with greater or less than median ITD length, respectively). The benefit of quizartinib on survival was observed regardless of ITD insertion length.3
QuANTUM-First MRD Analysis: Conclusions and Clinical Implications
The investigators concluded that this analysis substantiates the prognostic value of FLT3-ITD‒specific MRD measurement in patients with FLT3-ITD receiving intensive chemotherapy. Achieving MRD negativity at any time point during or after treatment led to longer OS for all patients, but the addition of quizartinib was more likely to yield MRD negativity, a deeper level of MRD negativity, and improved survival compared with intensive chemotherapy alone. In addition, multiple and/or longer ITDs were associated with worse OS, and these patients also experienced survival benefits with the addition of quizartinib to chemotherapy.
Quizartinib has been approved by the FDA as an addition to intensive chemotherapy and represents the new standard of care for younger, fitter patients with newly diagnosed AML with FLT3-ITD. MRD testing for AML is not commercially available and not routinely used in practice at this time, but moving forward it may become standard of care based on these and other data demonstrating its prognostic value. MRD assessment of a specific molecular lesion, such as FLT3-ITD in this case, may be the best way to monitor disease status independently from standard pathological remission. It remains to be seen how this can be integrated into practice.
STOP-VEN: Background and Study Design
The next study investigated patients with AML who are unfit for intensive chemotherapy, for whom standard of care remains venetoclax plus azacitidine based on results of the phase III VIALE-A trial.4,5 VIALE-A found that in patients aged 75 years and older or those otherwise considered unfit, venetoclax plus azacitidine led to a 67% ORR and a median OS of 14.7 months. This is a very effective regimen, but continued administration may lead to prolonged cytopenias, which are especially a concern given the older, frail nature of these patients. Long-term results of VIALE-A with a median 43 months of follow-up raised the question of whether continued venetoclax plus azacitidine treatment necessarily resulted in the best outcomes, or whether patients in response could have better quality of life with early treatment discontinuation. The ability to discontinue treatment also would have a positive impact on cost and convenience. This possibility would parallel current practice in CML, where many patients can stop BCR-ABL‒targeted tyrosine kinase inhibitor (TKI) treatment and continue to do well.
The study designed to answer this question was STOP-VEN, a retrospective trial of 84 patients with newly diagnosed or R/R AML who were in remission. Enrolled patients were treated at French Innovative Leukemia Organization centers or the Moffitt Cancer Center in Tampa, Florida, between 2018 and 2023 and were retrospectively analyzed.6
Eligible patients had achieved a complete response (CR), CR with incomplete count recovery (CRi), or clearing of morphologic blasts) with venetoclax plus azacitidine and then discontinued venetoclax and/or azacitidine for ≥3 months. Primary endpoints were treatment-free remission and OS.
STOP-VEN: Baseline Characteristics
The trial population was typical of patients who receive venetoclax plus azacitidine, with a median age of 75 years and a range of up to 89 years. Approximately one half had de novo disease, and cell counts were low at the time of discontinuation. Many patients had known mutations, including FLT3-ITD and TP53, which are associated with worse outcomes with this treatment regimen.6
STOP-VEN: Venetoclax Plus Azacitidine Treatment and Response
Patients received a median of 4 cycles of venetoclax plus azacitidine, with the overwhelming majority achieving CR or CRi. Many patients were in MRD-negative CR, allowing investigators to examine the value of achieving a deep response in patients receiving a less-intensive treatment regimen. The primary reason for discontinuation was hematologic toxicities, followed by patient preference.6
STOP-VEN: Efficacy
The median treatment-free time period was 16 months for newly diagnosed patients and 10 months for patients with R/R disease. Median OS was very prolonged: 44 months in newly diagnosed patients and 19 months in patients with R/R disease.
For those who had achieved an MRD-negative response in the newly diagnosed setting, the majority were still alive, with a 2-year survival rate of 80%. That is impressive given that median OS in VIALE-A was 14 months. It also is promising to see that when relapsed patients were rechallenged with venetoclax plus azacitidine, 27% of newly diagnosed patients and 40% of patients with R/R disease were able to successfully obtain remission with that retreatment.6
STOP-VEN: Multivariate Analysis
The multivariate analysis shows that newly diagnosed patients had better outcomes than patients with R/R disease. Mutational analyses are shown, but these were in small patient numbers.6
STOP-VEN: Conclusions and Clinical Implications
In this retrospective study, discontinuation of venetoclax plus azacitidine in primarily MRD negative‒responding patients with AML was associated with treatment-free remission intervals of 16 and 10 months for newly diagnosed and R/R AML, respectively. These lengths of time are meaningful in terms of the quality of life for patients without having to come in for frequent transfusions, chemotherapy, and count checks.
Median OS was also impressive in this retrospective group: 44 months in newly diagnosed patients, with 80% alive at 2 years, and 31 months in patients with R/R disease. Based on these data, it may be feasible to stop venetoclax plus azacitidine treatment, although this should be confirmed in a placebo-controlled trial. Looking forward, I would consider specific characteristics such as mutational profile and clinical response to select the best candidates for this approach. The possibility of discontinuing treatment would be very appealing to many of our patients.
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Background and Study Design
The QuANTUM-First trial discussed earlier explored the addition of a FLT3 inhibitor to an intensive chemotherapy backbone for patients with FLT3-mutated disease. This next study applies a similar triplet approach to patients with FLT3-mutated disease who are unfit or who have relapsed following intensive chemotherapy by adding FLT3 inhibition to a nonintensive regimen.
This single-arm, multicohort phase I/II trial performed at the MD Anderson Cancer Center examined such a combination.7 The trial population included unfit patients with newly diagnosed FLT3-mutated AML, as well as patients with R/R FLT3-mutated disease. The triplet induction included decitabine for 10 days, venetoclax for 21 days, and quizartinib for 28 days initially, with an amendment later in the study reducing quizartinib to 14 days. Consolidation was more truncated, with 5 days of decitabine, 14 days of venetoclax, and quizartinib between 14 and 28 days, depending on tolerability.
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Baseline Characteristics
Patients in the frontline cohort were older, with a median age of 70 years, and those in the R/R cohort were younger, with a median age of 59 years. Of note, only 14 patients were treated in the frontline cohort, so that should be considered when interpreting the results. Of the patients with R/R disease, the median number of previous therapies received was 3 (range 1-5). Most of these patients (56%) had previously received venetoclax plus a hypomethylating agent, and 83% of patients had previously received ≥1 prior FLT3 inhibitor. Therefore, in the R/R cohort, the study tested the triplet combination regimen in patients who had previously been exposed to a hypomethylating agent, venetoclax, and/or a FLT3 inhibitor.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: DLTs
It was determined in the dose-finding phase that 40 mg of quizartinib led to significant toxicity, with grade 4 myelosuppression in the first 2 patients treated; 30 mg was selected as the quizartinib dose moving forward, and 100% response was seen in 6 patients treated with the triplet at this dose.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Efficacy
Of the 43 patients in the R/R cohort, 65% achieved CRc, with most of these being morphologic leukemia-free state (34%) or CRi (19%). In addition, approximately one third of the patients achieved MRD negativity when determined by flow cytometry or polymerase chain reaction (PCR). In the subgroup analysis of patients with R/R FLT3-ITD mutation‒positive AML, 63% of those with prior exposure to gilteritinib achieved CRc, and 72% of those without any previous exposure to gilteritinib achieved CRc. The CRc rate was >50% regardless of prior exposure to a hypomethylating agent plus venetoclax or the RAS/MAPK, DNMT3A, and NPM1 mutation status.
Among the 14 patients with newly diagnosed disease, the CRc rate was 100%, including a CR rate of 79% and a CRi rate of 21%. The MRD negativity rate in this small cohort of 14 patients was impressive at 75% when determined by flow cytometry and 86% when assessed by PCR.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: R/R Cohort OS by Subgroup
Looking more closely at subgroup results in the R/R setting, OS is unsurprisingly greater in patients who showed a response: a median of 19 months for those with CR and 8.1 months for those with any type of response. Of note, meaningful survival outcomes were seen in patients who had previously been treated with venetoclax and hypomethylating agents or with gilteritinib.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Frontline Cohort Outcomes
Of 14 patients treated in the frontline cohort, there were 4 deaths and 2 relapses; 8 of the 10 patients still alive at the time of analysis did not go on to stem cell transplant, meaning they had been able to continue on some level of consolidation or maintenance therapy.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Safety
The major adverse event (AE), as touched on previously in the dose-finding phase, was myelosuppression. In patients treated with 28 days of quizartinib, cytopenias were seen that lasted 42-43 days, on average. For those treated with quizartinib reduced to 14 days, myelosuppression was improved to median durations of 35 or 36 days. This shorter duration of quizartinib did not appear to decrease efficacy.7
Quizartinib, Venetoclax, and Decitabine in FLT3-ITD AML: Conclusions and Clinical Implications
This phase I/II trial showed very promising results in line with other published findings with gilteritinib, supporting efficacy of triplet therapy combining a FLT3 inhibitor with venetoclax and a hypomethylating agent. Careful monitoring is needed in light of the risk of myelosuppression, but the 100% ORRs in newly diagnosed patients is incredibly promising. I would not consider this regimen ready for standard clinical use until further clinical trial evidence can determine the safest regimen and the optimal management of associated cytopenias, but this combination should be further developed for patients with FLT3-mutated AML.
STIMULUS-AML2: Background and Study Design
This next study returns to the importance of MRD assessment in determining the best setting to treat AML. Many healthcare professionals do not want to wait until overt relapse, because many times at that point, the disease is so clinically aggressive and treatment refractory that it is very challenging to get the leukemia back into remission. If MRD positivity is detected early, then treatment can potentially be pursued at that point to achieve MRD negativity instead of waiting until overt relapse. The other setting in which the MRD status is important is when planning to perform alloSCT, which can be curative for many patients. There are concerns about patients who clinically appear cured after transplant but are still MRD positive, suggesting the risk of imminent relapse and the need to act early to eliminate residual disease to prevent overt recurrence after transplant.
Sabatolimab is an antibody targeting TIM-3, which is expressed on leukemic stem cells and blasts and on immune cells in the marrow microenvironment.8 It is hypothesized that inhibiting TIM-3 could alter the adaptive and innate microenvironment to enhance mechanisms of disease control after transplant in myeloid diseases. The hypothesis is that sabatolimab monotherapy may be effective in the MRD setting, just as blinatumomab is used for MRD-positive patients with acute lymphoblastic leukemia to enhance donor immune cells to exert graft vs leukemia effects and eradicate residual AML cells.9
In STIMULUS-AML2, patients with AML who had achieved hematologic CR after alloSCT but still had evidence of MRD were treated with sabatolimab at several doses post transplant to discover whether this could eradicate MRD or prevent overt relapse.8 The data recently presented are from the safety run-in in which patients received 400 mg or 800 mg of the anti‒TIM-3 antibody sabatolimab. Primary endpoints were relapse-free survival (RFS) and safety.
STIMULUS-AML2: Baseline Characteristics
Data from 21 patients were presented, divided almost equally between dose levels. These patients were mostly younger, as would be anticipated as candidates for alloSCT, and patients had an array of genetic risk levels. Of importance, a majority were MRD positive at the time of transplant, with the concern that they would be MRD positive post transplant with a high risk for disease relapse.8
STIMULUS-AML2: Baseline Local MRD Assessment
Patients were required to demonstrate MRD by local assessment at least 60 days post transplant. Most of these were performed with PCR; fewer used flow cytometry, and 2 patients had chimerism analysis.8
STIMULUS-AML2: Safety
The single dose of sabatolimab was very well-tolerated. Approximately 30% of patients experienced any treatment-related AEs. No acute or chronic graft vs host disease (GVHD) was seen; 1 patient had a myocarditis event that was considered a DLT at the 800-mg dose, and 5 patients died from overt relapse of AML.8
STIMULUS-AML2: Treatment-Related AEs
Treatment-related AEs in these 21 patients included grade 3 neutropenia and thrombocytopenia in 1 patient, the 1 incidence of myocarditis mentioned above, and 1 low-grade instance each of folliculitis and peripheral neuropathy.8
STIMULUS-AML2: Preliminary Efficacy
Preliminary efficacy data are intriguing, showing that one third of patients remain on treatment and in hematologic CR at the time of data collection. Response does not seem to be dose dependent, as the number of relapse-free patients was similar for each dose level. Many of the patients who remained disease free continued to receive additional cycles of sabatolimab in the following year.8
STIMULUS-AML2: Conclusions and Clinical Implications
These early results suggest that sabatolimab monotherapy is very well-tolerated. No exacerbations of GVHD occurred, and there may be some enhancement or maintenance of a graft vs leukemic effect. The trial is moving forward into the expansion phase at the 800-mg dose.
Because only approximately one third of patients remained in remission after >1 year of treatment, there is a question of whether this immune activity potentially could be enhanced with an antidisease agent, such as the addition of azacitidine in the dose-expansion phase. In any case, posttransplant MRD positivity is a disease setting of unmet clinical need, and it would be helpful to have a treatment, in this scenario, to offer these patients.
AUGMENT-101: Background and Study Design
There has been growing excitement regarding menin inhibitors, which are novel targeted therapies for AML with KMT2A rearrangements (formerly known as mixed-lineage leukemia or MLL rearrangements). These agents act by blocking the activation of the pathogenic epigenetic complex resulting from this mutation. The leading menin inhibitor in clinical development is revumenib, formerly known as SNDX-5613. Data reported here are from patients treated with the recommended revumenib dose of 163 mg twice daily in the phase II expansion of AUGMENT-101.10 At ASH 2023, results from a cohort of 94 patients with R/R acute leukemias harboring KMT2Ar mutations were reported.
AUGMENT-101: Baseline Characteristics
Out of the 57 patients treated with revumenib in the efficacy population, 86% had AML, 12% had ALL, and 1 patient had mixed phenotype leukemia. The majority were older patients, but 13 patients (23%) were younger than 18 years of age. These patients had received a median of 2 prior lines of therapy, with 46% having received ≥3 prior lines of therapy; 75% had received prior venetoclax-based regimens, and almost one half had received prior alloSCT. Overall, this is a prognostically poor patient population.10
AUGMENT-101: CR/CRh Rate (Primary Endpoint) and Additional Efficacy Findings
In these prognostically poor patients, ORR was 63%, which breaks down into a CR rate of 18%, CR/CRh/CR with incomplete platelet recovery/CRi rate of 44%, and morphologic leukemia-free state of 18%. The median time to response was <2 months. The median OS (without censoring for transplant) in the efficacy population was 8 months, with a range of approximately 4-11 months.10
AUGMENT-101: CR/CRh Rate by Subgroup
The CR/CRh rate was identical regardless of prior stem cell transplantation at 23%. This rate was higher in patients who had received 1 prior line of therapy compared with those who had received ≥2 lines of therapy (47% vs 12%-14%), suggesting that this agent may be more efficacious in the upfront setting than in patients who are heavily pretreated.10
AUGMENT-101: Response by KMT2A Rearrangement
Response rates were analyzed by type of KMT2A rearrangement, but no clear pattern of association was seen. Patient numbers were quite small in each group.10
AUGMENT-101: Duration of Response
The median duration of CR/CRh was approximately 6 months. In these patients with R/R disease, the ability to proceed to alloSCT may be the best predictor of long-term response. Here, 14 of 36 patients were able to proceed to transplant; one half of those patients restarted revumenib treatment in the posttransplant setting.10
AUGMENT-101: Safety
The phase I study of revumenib found some evidence of QTc prolongation, which was considered the DLT.11 At the recommended phase II dose used here, this still occurred at any grade in 26% of patients and grade ≥3 in 14% of patients.10 Differentiation syndrome also emerged in this cohort, at any grade in 28% of patients and grade ≥3 in 16% of patients. The differentiation syndrome is likely due to the underlying mechanism of action of menin inhibitors, similar to that first seen with IDH inhibitors in APL. These 2 AEs are the ones to watch for carefully with this agent.
AUGMENT-101: Conclusions and Clinical Implications
The future for revumenib looks very promising; it has been submitted to the FDA for consideration as the first menin inhibitor for the treatment of AML. The response rates seen in KMT2Ar disease in this trial are consistent with other approved drugs in the R/R setting, such as gilteritinib and IDH1/IDH2 inhibitors. Similarly, OS of 6 months is in the same range as that seen with approved agents. Revumenib is also continuing development for patients with NPM1-mutant AML. Preliminary data from a phase I trial of another menin inhibitor, JNJ-75276617, also were reported at ASH 2023. The study demonstrated comparable response rates with somewhat lower incidence of differentiation syndrome in patients with R/R AML with KMT2A rearrangements or NPM1 mutations, supporting the efficacy of this class of drugs in these AML subsets.12
ASCEND-CML: Background
TKIs are the preferred treatments for chronic-phase CML.13,14 The first-generation TKI imatinib or the second-generation TKIs dasatinib, nilotinib, and bosutinib currently are approved for the upfront treatment of patients with newly diagnosed chronic-phase CML. The third-generation TKIs, ponatinib and asciminib, are approved for patients with resistant CML or those with disease progression after ≥2 prior TKIs.
Asciminib currently is approved for adult patients with Philadelphia chromosome‒positive CML in the chronic phase who were previously treated with ≥2 TKIs, as well as patients with Philadelphia chromosome‒positive CML in the chronic phase with the T315I mutation of BCR-ABL.15 The question is, if asciminib demonstrates efficacy as salvage therapy for resistant disease, should it be moved to the frontline setting? To address this question, the ASCEND-CML trial was performed.
ASCEND-CML: Study Design
ASCEND-CML was a prospective, open-label phase II study using asciminib at 40 mg twice daily in 101 patients with newly diagnosed chronic-phase CML.
Responses were evaluated in 3-month intervals, primarily by P210 BCR::ABL1. Early molecular response was defined as BCR::ABL1 <10% by Month 3, and major molecular response was defined as BCR::ABL1 <0.1% by Month 12. For patients who achieved a major molecular response, the asciminib dose was changed to 80 mg once daily, whereas those with a suboptimal response received asciminib 80 mg twice daily. For those with treatment failure, asciminib was administered in combination with imatinib, dasatinib, or nilotinib.
Coprimary endpoints for ASCEND-CML were early molecular response and major molecular response.16
ASCEND-CML: Baseline Characteristics
The participants enrolled on ASCEND-CML were mostly middle-aged male patients. The majority of these patients had low-risk (67%) or intermediate-risk (28%) disease at the time of their diagnosis. The median follow-up for this report was relatively short at 23 months, which is why we have the 3- and 12-month milestones.16
ASCEND-CML: Molecular Response at 3 Mo (Coprimary Endpoint)
At 3 months, 93% of patients achieved an early molecular response, and 48% of patients achieved a major molecular response.16
ASCEND-CML: Molecular Response at 12 Mo (Coprimary Endpoint)
At 12 months, the major molecular response rate had gone up to 78%. Almost one third of patients had a 4.5-log reduction in BCR::ABL1 at that 12-month time point (BCR::ABL1 ≤0.0032%). The cumulative instance of major molecular response at 12 months was approximately 78% and at 24 months was approximately 87%.16
ASCEND-CML: Safety and Discontinuations
Out of 101 patients, 6 discontinued treatment because of intolerance, and a small number of patients were noted to have therapeutic resistance. These individuals were taken off the study mostly because of the presence of mutations or lack of clinical response.16
ASCEND-CML: Conclusions and Clinical Implications
The early molecular response of 93%, as well as the fact that approximately one half of patients achieved a major molecular response at 3 months, demonstrates the potency of this agent in the frontline setting. This trial was not randomized to compare asciminib with imatinib or any of the second-generation TKIs, but these data look very promising compared with other TKIs in the frontline setting.16
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL—Background
The next study focuses on APL, a rare and distinct subset of AML.17
Standard-of-care induction therapy for newly diagnosed APL is all-trans retinoic acid with arsenic trioxide with or without chemotherapy.18 A pertinent issue with treating APL is the need to give IV arsenic trioxide 5 times per week for 4 weeks during induction and for 5 weeks during consolidation.18,19 Furthermore, lack of availability and logistics of administering daily IV arsenic trioxide for 5 weeks per cycle in resource-limited countries also can be highly challenging. An oral arsenic trioxide was developed in Hong Kong,20 and one interesting study presented at ASH this year evaluated the efficacy of an all-oral arsenic-based regimen for the treatment of newly diagnosed APL.21
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL: Study Design
This study enrolled 125 patients with standard- or high-risk newly diagnosed APL. During induction, patients with standard-risk APL were assigned to receive arsenic trioxide/all-trans retinoic acid/ascorbic acid (AAA) with daunorubicin, and those with high-risk APL received AAA only. Both groups subsequently received AAA for consolidation and maintenance.
Primary endpoints included OS, RFS, and safety. The secondary endpoint was molecular response via molecular monitoring of bone marrow and peripheral blood after each cycle, every 8 weeks during maintenance, and every 3 months for 2 years after maintenance completion (NCT03624270).21
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL: Baseline Characteristics
The median age of patients enrolled on this trial was 49 years, and most patients had classical APL pathology. Nine patients had CNS involvement, and most (74%) participants had standard-risk APL.21
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL: Efficacy Outcomes
At a median follow-up of 31 months, all patients on this trial achieved CR; 3-year OS and RFS rates were 97% each.21
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL: Toxicities
The most common AEs were grade 1 headache and grade 2 hepatotoxicity, including transaminitis. Grade 3 or 4 AEs were rare in this trial, and there were no reports of treatment-related discontinuations or death. Of interest, all patients who developed APL differentiation syndrome within 14 days of treatment initiation responded fully to dexamethasone.21
All-Oral Arsenic Trioxide–Based Induction in Newly Diagnosed APL: Conclusions and Clinical Implications
Regardless of risk category and age, this regimen was highly effective among patients with newly diagnosed APL, with 3-year OS and RFS rates of 97% each. There was a low incidence of high-grade AEs, and the therapy was well-tolerated.21
In this trial, there did not appear to be a difference in efficacy among patients receiving IV arsenic vs oral arsenic. With this, an oral arsenic‒based formulation is something to look forward to in the future. To expand on this study, an ongoing phase II trial in Asia is evaluating AAA in the first-line setting (NCT04687176).
COMMANDS: Full Analysis of Phase III Trial of Luspatercept vs Epoetin Alfa for ESA-Naive, Transfusion-Dependent, Lower-Risk MDS
In 2023, luspatercept was approved by the FDA for the treatment of ESA-naive anemia in adult patients with very low‒ to intermediate-risk MDS who may require regular RBC transfusions.22 This approval was based on the interim results of the phase III COMMANDS trial, which showed an improved rate of RBC-TI with luspatercept compared with epoetin alfa in patients with lower-risk MDS based on the revised International Prognostic Scoring System (IPSS-R).23 At ASH 2023, we saw the reports of the full efficacy and safety analysis of the COMMANDS trial.24
COMMANDS: Study Design
This study enrolled 363 patients with ESA-naive, very low‒, low-, or intermediate-risk MDS who required regular transfusions. The standard of care for this patient population typically is an erythropoietic agent, and this trial randomized patients to receive either luspatercept or epoetin alfa. Treatment continued until progressive disease or lack of benefit according to the International Working Group 2006 criteria. The primary endpoint was RBC-TI for >12 weeks with a concurrent mean hemoglobin increase of ≥1.5 g/dL up to Week 24, and key secondary endpoints included safety and erythroid hematologic improvement (NCT03682536).24
COMMANDS: Baseline Characteristics
The arms of this trial were well-balanced. The median age of enrolled patients was 74 years, and most patients (approximately 63%) had a transfusion burden of <3 units/8 weeks. There was a preponderance of patients with SF3B1-mutated MDS enrolled on this trial. Approximately 63% of patients on the luspatercept arm and 56% of those on the epoetin alfa arm harbored SF3B1 mutations. In addition, approximately 72% to 73% of patients on both arms were positive for ringed sideroblasts in the marrow. Of note, these biological features are known to correlate with high response to luspatercept therapy.
COMMANDS: Primary Endpoint by Patient Population
When looking at the intention-to-treat population, luspatercept resulted in a significantly higher rate of RBC-TI for ≥12 weeks with a concurrent hemoglobin increase of ≥1.5 g/dL compared with epoetin alfa (60.4% vs 34.8%; P <.0001). The superiority of luspatercept vs epoetin alfa in achieving RBC-TI was clearly seen in patients with SF3B1-mutated MDS (70.2% vs 32.7%) and those with ring sideroblasts (65.4% vs 29.2%), and this trend was consistent irrespective of baseline serum erythropoietin level.24
COMMANDS: Key Secondary Endpoints
In addition, patients with low- and intermediate-risk MDS treated with luspatercept demonstrated superior response rates vs epoetin alfa in all key secondary trial endpoints, including RBC-TI at ≥12 weeks and ≥24 weeks of treatment. Luspatercept therapy also was associated with 74.2% of patients who achieved hematologic improvement in the erythroid lineage at ≥8 weeks vs 53% of patients who received epoetin alfa (P <.0001).24
COMMANDS: Median Duration of RBC-TI ≥12 Weeks by Patient Population
As expected, luspatercept was more effective in patients who had SF3B1 mutations or the presence of ring sideroblasts. The mean duration of RBC-TI was 126.6 weeks and 89.7 weeks in the luspatercept and ESA arms, respectively.24
COMMANDS: Safety
The median duration of treatment in the luspatercept and epoetin alfa arms was 51.3 and 37.0 weeks, respectively. There were more treatment-emergent AEs among patients receiving luspatercept, as well as more AEs in general. Over time, rates of fatigue and asthenia decreased, and events were mostly grade 1/2 in the luspatercept arm. Of interest, there were a similar number of deaths. Overall, luspatercept was considered to be a safe agent in this setting.24
COMMANDS: Conclusions and Clinical Implications
In the full analysis of the phase III COMMANDS study, patients with ESA-naive, transfusion-dependent, lower-risk MDS received enhanced benefit with administration of luspatercept vs epoetin alfa. This was independent of transfusion burden, serum erythropoietin level, and the presence of SF3B1 mutation. This study led to the recent approval of luspatercept as a first-line growth factor administration in this patient population, and the results remain consistent with this full analysis.24
In the frontline setting, luspatercept currently is approved for anemia without previous ESA use (ESA naive) in adult patients with very low‒ to intermediate-risk MDS who may require regular RBC transfusions. In the COMMANDS trial, RBC-TI with a concurrent hemoglobin increase ≥1.5 g/dL was achieved by 70.2% of patients with MDS harboring SF3B1 mutations and 65.4% of patients with ring sideroblasts upon treatment with luspatercept compared with 32.7% and 29.2%, respectively, with epoetin alfa. Among patients lacking this mutation or those without ring sideroblasts, the response rates were similar between luspatercept and epoetin alfa. However, the fact that luspatercept is administered every 3 weeks and epoetin alfa requires weekly to biweekly injections supports a potential benefit to patients in terms of a reduced number of clinic visits and injections with luspatercept. For these reasons, luspatercept is likely to replace epoetin alfa as the preferred frontline therapy for all patients with low- to intermediate-risk MDS requiring RBC transfusions, including those harboring SF3B1 mutations and those with ring sideroblasts, in the near future.
Of importance, some patients with low- to intermediate-risk transfusion-dependent MDS may prefer treatment with an ESA rather than luspatercept. Such patients include individuals who develop an allergic reaction to and/or experience AEs with luspatercept, those with chronic renal disease with especially low serum erythropoietin levels, or those who prefer a weekly/biweekly schedule and/or already are responding to ESA therapy.
Phase III IMerge Subgroup Analysis: Background
There is an unmet need among patients with RBC transfusion‒dependent lower-risk MDS who did not respond to or are ineligible for ESAs. Previous data from the phase III IMerge trial showed that imetelstat—a telomerase inhibitor—offers durable transfusion independence and disease-modifying activity among patients with lower-risk MDS.25 At ASH 2023, we saw the results of subgroup analysis evaluating the efficacy of imetelstat across different MDS cytogenetic risk categories.26,27
IMerge Subgroup Analysis: Study Design
IMerge was an international, double-blind phase III trial that enrolled 178 patients with low- or intermediate-1‒risk MDS who were RBC transfusion dependent and refractory to or relapsed on an ESA or erythropoietin. Participants were randomly assigned to receive imetelstat or placebo. This particular analysis evaluated the rates of RBC-TI across risk categories using the IPSS, the IPSS-R, or the modified IPSS (IPSS-M) (NCT02598661).26,27
IMerge Subgroup Analysis: IPSS Risk Category at Baseline
This outlines the variety of the IPSS, IPSS-R, and IPSS-M risk scores of patients enrolled on this trial. The majority of patients were characterized as having low-risk MDS (67% by IPSS, 75% by IPSS-R, and 63% by IPSS-M).26,27
IMerge Subgroup Analysis: Rates of Durable Transfusion Independence
As mentioned previously, durable RBC-TI was observed over time with imetelstat treatment. RBC-TI of ≥8 weeks was 40% with imetelstat vs 15% with placebo (P <.0008). Of note, 83% of patients who responded at 8 weeks of treatment achieved RBC-TI.26,27
IMerge Subgroup Analysis: RBC-TI by IPSS Risk Category
Imetelstat demonstrated a significant improvement in RBC-TI vs placebo, regardless of IPSS risk category. For patients with IPSS low risk, RBC-TI was 28.8% vs 5.1% at ≥24 weeks (P = .003). For patients with IPSS intermediate-1 risk, RBC-TI was 39.5% vs 4.8% at ≥8 weeks (P = .004) and 26.3% vs 0% at ≥24 weeks (P = .009).26
IMerge Subgroup Analysis: RBC-TI by IPSS-R Risk Category
Across IPSS-R subgroups, the transfusion-independence benefit with imetelstat still holds true compared with placebo. Furthermore, the rate of response among patients with intermediate-risk disease was similar to that of patients with low-risk disease. No responses were seen among patients with intermediate-risk disease who were receiving placebo. Patients with very low‒ or high-risk disease were not assessed because of low patient numbers.26
IMerge Subgroup Analysis: RBC-TI by IPSS-R Cytogenetic Risk Category
When stratifying by cytogenic risk, imetelstat also increased the proportion of patients with RBC-TI, regardless of IPSS-R risk category.26
IMerge Subgroup Analysis: RBC-TI by IPSS-M Risk Category
RBC-TI continued to be improved across all IPSS-M risk categories, and one third of patients with higher-risk MDS experienced RBC-TI of ≥8 weeks.26,27
IMerge Subgroup Analysis: Conclusions and Clinical Implications
Here, we saw that imetelstat increased rates of RBC-TI in patients with lower-risk, transfusion-dependent MDS who were refractory to, relapsed on, or ineligible for ESAs, regardless of IPSS risk category.26,27 Data also suggest that imetelstat could be a disease-modifying agent, as evidenced by the fact that many of those patients treated on the IMerge trial had decreased variant allele frequency in many of the mutations that are associated with MDS.25,27 For patients who have become resistant to luspatercept or ESAs, I think treatment with a disease-modifying agent such as imetelstat might be the way to go. These phase III trial results with imetelstat will be submitted to the FDA for New Drug Approval as a follow-up therapy for patients with MDS with previous failure on growth factors.
Of note, the patients enrolled on the IMerge trial were individuals with low- or intermediate-1‒risk MDS who were RBC transfusion dependent and R/R to an ESA. I would caution that for these patients who have previously responded to an ESA and are now failing treatment, it is crucial to perform repeat bone marrow biopsies prior to initiating imetelstat to rule out the possibility that the underlying disease biology has progressed to high-grade MDS and/or AML. Findings of increased blasts ranging from 10% to 19% (AML/MDS) or AML (>20%) should prompt healthcare professionals to offer hypomethylating agent therapy (azacitidine or decitabine) and consider referral for possible alloSCT rather than initiating imetelstat.