We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Systematic ReviewOpen Accesscc iconby iconnc iconnd icon

Therapies for acute myeloid leukemia in patients ineligible for standard induction chemotherapy: a systematic review

    Michael Heuser

    *Author for correspondence:

    E-mail Address: heuser.michael@mg-hannover.de

    Department of Hematology, Hemostasis, Oncology & Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany

    , , ,
    Olga M Klibanov

    IQVIA, Medical and Patient Communications, Parsippany, NJ 07054, USA

    , &
    Published Online:https://doi.org/10.2217/fon-2022-1286

    Abstract

    Aim: To review clinical evidence for current and emerging treatments for patients with acute myeloid leukemia (AML) who are ineligible for first-line induction chemotherapy. Methods: A systematic literature review was performed (28 October 2021) to identify clinical outcomes including overall survival (OS), event-free survival (EFS), relapse-free survival (RFS) and adverse events (AEs). Results: Of 233 references that met prespecified criteria, 26 studies were included. Adding targeted therapies (venetoclax/ivosidenib) to hypomethylating agents (HMAs) yielded better OS hazard ratios (HRs) (0.44–0.66) and EFS HRs (0.33–0.63) compared with other agents. AEs were more frequent with combination therapies than control arms, except with ivosidenib plus azacitidine. Conclusion: Targeted therapy combined with a HMA shows the most promising results in this difficult-to-treat population.

    Plain language summary

    Acute myeloid leukemia (AML) is a type of cancer of the bone marrow and blood that leads to overproduction of immature white blood cells. High-dose (intensive) chemotherapy is usually the first treatment option for AML. However, more than half of people newly diagnosed with AML cannot receive the recommended initial intensive chemotherapy due to older age or poor health. Treatment with low-dose cytarabine (LDAC) and hypomethylating agents (HMAs), such as azacitidine, is key for such people. We reviewed 26 clinical trials looking into available and developing treatment options for people who cannot have the recommended initial chemotherapy. The review found evidence that combining LDAC or HMA with a targeted therapy can improve survival. In AML, new therapies (such as ivosidenib, venetoclax and glasdegib) ‘target’ specific changes in the genes of cancer cells to slow or stop their division and growth. The greatest improvement in survival was seen in clinical trials where targeted therapies were added to azacitidine or LDAC. Targeted therapies may result in certain side effects that require regular monitoring. To provide patients with the benefits of targeted therapies they need to undergo genetic testing at the time of diagnosis. Tests to determine an individual's specific gene changes allows clinicians to develop personalised treatment plans with available targeted therapies. This shows promise in improving survival for people with AML who cannot receive initial intensive chemotherapy.

    Acute myeloid leukemia (AML) represents a group of clonal hematopoietic stem cell disorders in which both failure to differentiate and over-proliferation in the stem cell compartment result in accumulation of non-functional cells, termed myeloblasts [1]. It is an aggressive hematopoietic malignancy and, if left untreated, patients die of infection or bleeding, usually within weeks. However, some older adults may have a slower progressive clinical course [2]. In adults, AML is the most common type of acute leukemia and has the shortest 5-year overall survival (OS) rate at 24%, with the mortality risk increasing with age [3].

    In adults and children, AML is the second most common type of leukemia, making up 31% of total adult leukemia cases, and primarily affects the elderly (>65 years of age), with the average age at diagnosis of 68 years [1,4]. In 2022, it was estimated that AML will be diagnosed in 20,050 people of all ages in the USA [4]. As of 2017, the incidence of AML in Europe (France, Germany, Italy, Spain and the UK) was 3.5 per 100,000 individuals, with the reported prevalence in these five European countries ranging from approximately 2300 to 5000 cases [5].

    AML treatment depends on disease subtype, patient age and overall patient health. Subject to patient fitness and eligibility, first-line treatment of AML in newly diagnosed patients involves induction and consolidation chemotherapy [6,7]. However, fewer than 50% of patients are candidates for first-line induction chemotherapy (IC) [8,9] due to older age, comorbidities, or Eastern Cooperative Oncology Group (ECOG) performance status [6,7], and only 10–20% of patients older than 80 years receive first-line IC [10].

    Various treatment-decision algorithms have been developed to assess fitness for standard IC in newly diagnosed patients with AML [11–14]. Due to the complexity of this disease and poor outcomes in older patients, using one or more of these algorithms to identify patients who are not fit for standard IC can help facilitate discussions between clinicians and patients, thereby optimising personalised medicine.

    For newly diagnosed patients with AML ineligible for first-line IC, the hypomethylating agents (HMAs) azacitidine and decitabine are the mainstay of therapy [6,7]. However, new data have emerged regarding the important role that cytogenetics and molecular abnormalities play in the risk stratification and management of patients with AML [15,16], and novel therapies targeting genetic mutations have resulted in improved survival rates in this difficult-to-treat population. Several mutations have previously been shown to affect overall survival (OS) in AML, including those in the NPM1 gene, CCAAT/CEBPA gene, FMS-related tyrosine kinase 3 gene (FLT3) [17], and, more recently, the IDH gene [6,18]. Mutations in IDH (IDH1 and IDH2) are among the most common genetic mutations in AML, detected in approximately 20% of patients [19]. IDH1 mutations in AML are present in 6–9% of patients, with a higher frequency of 8–18% among patients with normal karyotype AML (NK-AML) [18,20–23]. Multiple studies have examined the impact of IDH1/IDH2 mutations on prognosis, but the current data do not yet warrant their designation to one of the European LeukemiaNet (ELN) prognosis groups [15]. The IDH mutation does, however, serve as a potential target for new AML drug development, and the emerging use of IDH inhibitors may impact the prognostic outcome of patients with IDH1/IDH2-mutated (mIDH1/mIDH2) AML [15]. Screening for genetic mutations has become a standard part of the initial clinical workup in AML [6,7,15].

    Treatment of AML is heterogeneous worldwide due to variable drug approval status, reimbursement practices, treatment guidelines, and definitions of ineligibility for IC. In the USA, the National Comprehensive Cancer Network (NCCN) guidelines recommend adding venetoclax (B-cell lymphoma [BCL]-2 inhibitor) to HMAs as the preferred therapy for patients who are ≥60 years old and ineligible for IC [6]. For patients with mIDH1 AML who are ≥60 years old and who are not candidates for IC or decline IC, the preferred regimens include HMA plus venetoclax, ivosidenib plus azacitidine, and ivosidenib monotherapy [6]. For patients with mIDH2 AML, NCCN guidelines recommend HMA plus venetoclax or enasidenib monotherapy, and for those with the FLT3 mutation, they recommend HMA plus venetoclax [6].

    In Europe, the preferred regimens for this population according to the 2022 ELN AML recommendations include HMA plus venetoclax, low-dose cytarabine (LDAC) plus venetoclax, ivosidenib plus azacitidine for patients with mIDH1, and best supportive care (BSC) [15]. In Europe, ivosidenib received an orphan drug designation by the EMA for the treatment of AML in 2016 [24], and a marketing authorisation application was submitted to the EMA in March 2022 for this drug to be used in combination with azacitidine in patients with mIDH1 AML [15,25].

    With the rapidly expanding landscape of therapies for AML, understanding the efficacy and safety outcomes of available therapies can assist in decision-making when managing this patient population. The objective of this systematic literature review (SLR) was to identify and qualitatively assess the clinical efficacy and safety evidence for current and emerging treatments for patients with newly diagnosed AML who are ineligible for first-line IC.

    Materials & methods

    Study design

    This SLR was conducted in accordance with the recommendations of the Cochrane Handbook for Systematic Reviews and Interventions [26], the general principles of the Centre for Reviews and Dissemination (University of York) guidance [27] for undertaking reviews in healthcare, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [28], and the methods for systematic reviews as specified by the National Institute for Health and Care Excellence [29].

    Literature search

    A systematic literature search was conducted using patient/population, intervention, comparison and outcomes (PICOS) terms to identify English-language publications of clinical trials (any phase) that were published and indexed in the Embase (1974 to 28 October 2021), MEDLINE (1946 to 28 October 2021) and Cochrane Central Register of Controlled Trials (CENTRAL) (1991 to 28 October 2021) databases. The search targeted publications that evaluated therapeutic interventions in adults (≥18 years old) newly diagnosed with AML/secondary AML (sAML) and ineligible for standard IC. The search strategy combined free text words and indexing terms (e.g. medical subject headings [MeSH] terms for MEDLINE and EMTREE terms for Embase) relevant to AML and study designs with Boolean operators (e.g. ‘and’, ‘or’, ‘not’), using Embase-, MEDLINE-, and CENTRAL-specific search syntax. The complete literature search strategy and terms are described in the supplemental material (Supplementary Tables 1–5).

    Interventions of interest included chemotherapy (decitabine, azacitidine, FLUGA [fludarabine, cytarabine and filgrastim], sapacitabine, ATRA [all-trans retinoic acid], LDAC, GRASPA [L-asparaginase encapsulated in red blood cells, eryaspase] and guadecitabine), immunotherapy (lenalidomide, durvalumab, talacotuzumab, lintuzumab and gemtuzumab ozogamicin), targeted therapy (venetoclax, enasidenib, gilteritinib, ivosidenib, volasertib, glasdegib, barasertib [AZD1152] and tipifarnib), and BSC.

    Reference lists from recently published SLRs and meta-analyses in AML were also evaluated to ensure that all relevant evidence was captured. In addition, abstracts published between 1 January 2016 and 31 December 2021 from the following congresses were searched separately to identify relevant studies that are registered as completed but not yet published: European Society for Medical Oncology, American Society of Clinical Oncology, International Society for Pharmacoeconomics and Outcomes Research, European Haematology Association and American Society of Haematology.

    Literature reviews & study selection

    All abstracts identified in the literature searches were reviewed independently by two reviewers, and a sample of the abstracts was quality checked by a third independent reviewer. The reviewers then assessed the full texts of the included publications against the same eligibility criteria applied during the abstract review.

    Trials were excluded if the study participants included patients who were eligible for first-line IC. Studies were excluded if they were performed in animals, patients <18 years of age, or patients with acute promyelocytic leukemia, or if they described interventions other than pharmacological interventions or BSC. Additionally, studies were excluded if they did not provide data on specific outcomes of interest (e.g. OS, event-free survival [EFS], relapse-free survival [RFS], disease-free survival [DFS], complete response [CR], partial response [PR] and adverse events [AEs]) or were not clinical trials (e.g. case studies, editorials, pilot studies and pharmacokinetic studies).

    Data extraction & interpretation

    Outcomes of interest were extracted from full-text studies by using a standardised data extraction template in MS Excel®. They were organised for HMAs, LDAC, BSC, and ‘other’ (if they could not be categorised in the former three groups) as the four comparative treatment arms compared with chemotherapy, immunotherapy and targeted therapy. Comparative statistical analyses were not performed. If the publication did not specify the data for OS, EFS and PFS rates, these data were extracted from the Kaplan–Meier curves in the publications using WebPlotDigitizer [30]. At the time of writing, the most recent publication was used to report data of studies for which updated results were published after the dates of the systematic literature search.

    Results

    Selection of publications

    A total of 4503 references were identified from electronic databases (MEDLINE: 828; Embase: 2629; CENTRAL: 1046). Following the removal of duplicate records, screening against the eligibility criteria, abstract title screening, and full-text review, 184 publications were included. Manual screening of relevant congresses and clinical trial registries as well as reference checks of other published reviews resulted in the identification of 49 additional records that met the inclusion criteria. Of all included articles, 26 unique studies [31–56] reported in 60 publications [31–90] were prioritised for data extraction. The prioritisation was based on study design, prioritising randomised controlled trials (RCTs) and studies with a sample size of 20 participants or greater. Figure 1 depicts the PRISMA flow diagram of the study selection process.

    Figure 1. PRISMA flow diagram of the study selection process.

    EMB: Evidence Based Medicine; SLR: Systematic literature review; TLR: Targeted literature review.

    Key characteristics of the 26 included RCTs [31–56] are described in Supplementary Table 6. Of the 26 RCTs, nine (35%) were phase II/IIb trials [32,36,37,39,40,46,47,52,56], 12 (46%) were phase III trials [33,38,41–45,49,51,53–55], four (15%) were phase II/III trials [31,34,35,50], and one (4%) did not report study phase [48].

    The total studied population size included in this SLR was approximately 7000 patients with AML who were ineligible for standard IC. The baseline characteristics of these patients are presented in Supplementary Table 7. Most of the studies included patients ≥75 years of age. The median age was reported in 23 studies and ranged from 64 [48] to 80 years [47]. In all studies except one [52], more men were included than women. The proportion of men ranged from 45 [52] to 80% [47]. Five studies [37,41,43,58,91] reported the race/ethnicity of enrolled patients; the majority of patients were White (range: 16 [91] to 100% [37]), followed by Asian (range: 2 [43] to 26% [91]), Black (range: 0.44 [43] to 2.2% [58]) and other/not reported (range: 2 [43] to 62% [91]).

    Survival outcomes

    Of the 26 identified clinical trials, all [31–56] reported data on OS, 12 trials [38–42,46,49–51,53–55] reported data on EFS, and six trials [34,35,40–42,53] reported data on RFS (Supplementary Table 8). Although EFS is a frequently reported end point in AML trials, not all trials used a uniform definition of EFS. Most commonly, EFS was defined as time from randomisation to disease progression, treatment failure, relapse, or death [38,39,46,49,50,53,55]. The definition of treatment failure varied among studies, with the VIALE-A trial defining it as failure to achieve CR or <5% bone marrow blasts after at least six cycles of treatment [38], and the AGILE trial defining it as lack of CR by week 24 [49]. The BI 1230.4 study only included disease progression or death in the EFS definition [40], the ASTRAL-1 trial reported EFS results as part of a post hoc analysis [68], and two trials (POLO-AML-2 and NCT02752035) did not define EFS in the publications [41,54].

    Figure 2 illustrates the OS results for the nine trials [33,37,38,40,46,53,78,82,91] that demonstrated statistically significant differences in the OS outcome between study arms, and Figure 3 illustrates EFS results for five [38,40,53,82,91] of these nine trials that also reported EFS results.

    Figure 2. Comparison of mOS outcomes among trials with statistically significant differences between treatment groups.

    *Added to placebo.

    AGlLE specifically enrolled an mlDH1 population.

    Two additional experimental groups included in the DEClDER trial – VAL + DEC and ATRA + DEC – are not included here; HRs and p-values reflect improvements in mOS in all three experimental groups vs DEC.

    ATRA: All-trans retinoic acid; AZA: Azacitidine; BSC: Best supportive care; DEC: Decitabine; FLUGA: Fudarabine, cytarabine and filgrastim; GLAS: Glasdegib; GO: Gemtuzumab ozogamicin; HMA: Hypomethylating agent; HR: Hazard ratio; lVO: Ivosidenib; LDAC: Low-dose cytarabine; mOS: Median overall survival; n: Sample size; OS: Overall survival; TC: Treatment choice; VAL: Valproate; VEN: Venetoclax; VOL: Volasertib.

    Figure 3. Comparison of median event-free survival outcomes among trials with statistically significant differences between treatment groups.

    *Added to placebo.

    AGlLE specifically enrolled an mlDH1 population.

    AZA: Azacitidine; DEC: Decitabine; EFS: Event-free survival; FLUGA: Fludarabine, cytarabine and filgrastim; HMA: Hypomethylating agent; HR: Hazard ratio; lVO: Ivosidenib; LDAC: Low-dose cytarabine; mEFS: Median event-free survival; n: Sample size; VEN: Venetoclax; VOL: Volasertib.

    Hypomethylating agents

    In ten of the 26 studies, HMAs (azacitidine or decitabine) were compared with other interventions, including other chemotherapies (FLUGA [53], sapacitabine [44], valproate plus ATRA [46]), immunotherapies (lenalidomide [47], durvalumab [56], talacotuzumab [50]), and targeted therapies (venetoclax [38], enasidenib [39], gilteritinib [54], ivosidenib [49]).

    Of the three studies [44,46,53] that compared HMAs with other chemotherapies, two [46,53] showed that treatment with HMAs significantly improved survival; one trial showed that combining a HMA with sapacitabine did not result in a significant improvement in survival [44]. In the PETHEMA-FLUGAZA trial [53], azacitidine treatment resulted in a median OS (mOS) of 9.8 months compared with 4.1 months with FLUGA (p = 0.005). Median EFS (mEFS) was also significantly longer with azacitidine compared with FLUGA (4.9 months vs 3.0 months; p = 0.001), but the increase in median RFS (mRFS) duration with azacitidine compared with FLUGA did not reach statistical significance (12.1 months vs 7.9 months; p = 0.43). In a subgroup analysis of patients with genomic variations, the mOS did not differ between the treatment groups among patients with mIDH1 (Supplementary Table 8) and, in the IDH2 subpopulation, the OS benefit with azacitidine was similar to the overall population (Supplementary Table 8). Although the number of observations in this subanalysis was limited (n = 34 patients with mIDH1 AML; n = 36 patients with mIDH2 AML), the trial reported a numerically lower mOS in the azacytidine-treated group with mIDH1 AML (7.0 months; 95% CI: 0–20.4) compared with mIDH2 AML (12.0 months; 95% CI: 2.3–21.7) [61].

    The DECIDER trial [46] reported a mOS with decitabine of 4.8 months, compared with 6.1 months with decitabine plus valproate (hazard ratio [HR] = 0.85, 95% CI: 0.57–1.28), 8.4 months with decitabine plus ATRA (HR = 0.58, 95% CI: 0.37–0.91), and 7.7 months with decitabine plus ATRA plus valproate (HR = 0.62; 95% CI: 0.4–0.95). An exploratory analysis [70] showed that adding ATRA to decitabine monotherapy in a subgroup of patients with 20–30% bone marrow blasts resulted in a significant OS benefit (ATRA: 12.5 months vs no ATRA: 7.6 months; HR = 0.47, 95% CI: 0.24–0.94; p = 0.032).

    In the SEAMLESS trial [44], no improvement in mOS was seen when comparing the combination of decitabine and sapacitabine with decitabine monotherapy (Supplementary Table 8).

    Three studies [47,50,56] that evaluated outcomes when immunotherapies (lenalidomide, durvalumab and talacotuzumab) were added to HMAs did not show survival benefits compared with HMA monotherapy (Supplementary Table 8).

    Of the ten studies with HMAs as the comparator therapy, four [38,39,54,91] evaluated the addition of targeted therapy to an HMA. In the VIALE-A trial, the combination of venetoclax plus azacitidine significantly increased the mOS by approximately 5 months compared with azacitidine plus placebo (14.7 months vs 9.6 months; HR = 0.66; 95% CI: 0.52–0.85; p < 0.001); similarly, the mEFS increased by almost 3 months (9.8 months vs 7.0 months; HR = 0.63; 95% CI: 0.50–0.80; p < 0.001). Compared with azacitidine plus placebo, OS and EFS rates improved consistently over 12, 18 and 24 months in patients who received venetoclax plus azacitidine (Supplementary Table 8). In a subgroup of 89 patients with IDH1 or IDH2 mutations at baseline, OS rates at 12 months were also significantly improved in the venetoclax plus azacitidine group compared with the azacitidine plus placebo group (66.8% vs 35.7%; HR = 0.35; 95% CI: 0.20–0.60; p < 0.001). In a subgroup of 34 patients with mIDH1 AML, the hazard of death was lower in the venetoclax plus azacitidine group compared with the azacitidine plus placebo group (HR = 0.28; 95% CI: 0.12–0.65).

    The AGILE trial [49] investigated the combination of ivosidenib plus azacitidine in previously untreated patients with mIDH1 AML ineligible for induction chemotherapy. A 16-month improvement in mOS was seen with ivosidenib plus azacitidine compared with azacitidine plus placebo (24.0 months vs 7.9 months; HR = 0.44; 95% CI: 0.27–0.73; p = 0.001); a 19-month improvement was observed in mEFS (22.9 months vs 4.1 months; HR = 0.39; 95% CI: 0.24–0.64; p < 0.001). Similarly, OS and EFS rates continually improved over 24 months in patients who received ivosidenib plus azacitidine compared with those who received azacitidine plus placebo (Supplementary Table 8).

    In contrast to the VIALE-A and AGILE trials, mOS and EFS durations were not significantly different on adding enasidenib to azacitidine in a population with mIDH2 AML [39] or adding gilteritinib to azacitidine in patients with FLT3-mutated (mFLT3) AML compared with azacitidine alone (Supplementary Table 8).

    Low-dose cytarabine

    In ten of the 26 studies, LDAC monotherapy (or in combination with placebo) was compared with LDAC in combination with chemotherapy [34,35], immunotherapy [36,52], targeted therapy [31,37,40,41,55] or ‘other’ therapy (GRASPA) [32].

    No significant differences were seen in two-year OS or RFS when LDAC monotherapy was compared with chemotherapy consisting of clofarabine [35] or sapacitabine [34] (Supplementary Table 8).

    The five studies that evaluated adding a targeted therapy to LDAC reported significant improvements in survival compared with LDAC monotherapy [31,37,40,41,55] (Supplementary Table 8). Two trials [40,41] investigated the combination of volasertib with LDAC in comparison with LDAC monotherapy (with or without placebo) but reported dissimilar results. In the BI 1230.4 trial [40], the mOS and EFS durations were significantly longer with volasertib plus LDAC than with LDAC monotherapy (OS: 8.0 months vs 5.2 months; HR = 0.63; 95% CI: 0.4–1.0; p = 0.047; EFS: 5.6 months vs 2.3 months; HR = 0.57; 95% CI: 0.35–0.92; p = 0.021). In contrast, the POLO-AML-2 trial [41] showed no significant difference in mOS or EFS between volasertib plus LDAC and LDAC plus placebo (OS: 5.6 months vs 6.5 months; HR = 0.97; 95% CI: 0.8–1.2; p = 0.76; EFS: 3.3 months vs 2.8 months; HR = 0.96; 95% CI: 0.8–1.2; p = 0.67). In both trials, the RFS duration with volasertib plus LDAC was longer than with LDAC alone, but p-values were not reported for this outcome [40,41].

    In the VIALE-C trial [55,82], the final analysis [82] showed that venetoclax plus LDAC resulted in significantly longer mOS and mEFS durations compared with LDAC plus placebo (OS: 8.4 months vs 4.1 months; HR = 0.7; 95% CI: 0.50–0.98; p = 0.04; EFS: 4.9 months vs 2.1 months; p = 0.002). A subgroup analysis [55] of 33 patients with an mIDH1/2 demonstrated a numerical improvement in mOS with venetoclax plus LDAC compared with LDAC plus placebo (10.8 vs 9.0), but the sample size was small, and the differences were not statistically significant.

    In the BRIGHT AML 1003 trial [37], glasdegib plus LDAC resulted in significantly improved mOS versus LDAC alone in the primary analysis (8.3 months vs 4.3 months; HR = 0.46; 95% CI: 0.35–0.62; p = 0.0002); these benefits were maintained in the final post hoc analysis [67]. In a small subgroup of patients with mIDH1de novo AML (n = 8), the mOS duration with glasdegib plus LDAC (n = 6) was 7.8 months (95% CI: 3.3–27.1 months) compared with 1.6 months (95% CI: 1.3–1.9 months) with LDAC alone (n = 2). In two patients with mIDH1 sAML, the mOS duration with glasdegib plus LDAC was 13.8 months (95% CI: 3.1–24.4 months); there were no patients with mIDH1 sAML assigned to the LDAC alone group. In eight patients with mIDH2de novo AML, the mOS duration was 5.0 months (95% CI: 1.1–23.1 months) with glasdegib plus LDAC; there were no patients with mIDH2de novo AML assigned to the LDAC alone group. In three patients with mIDH2 sAML, the mOS duration was 4.6 months (95% CI: 2.2–7.1 months) compared with 5.8 months (95% CI: 4.5–6.5 months) with LDAC alone (n = 4).

    The SPARK-AML1 study demonstrated that the addition of targeted therapy – barasertib – to LDAC numerically improved the mOS compared with LDAC monotherapy, but the sample size was small (n = 77) and statistical significance was not reached (8.2 months vs 4.5 months; HR = 0.88; 95% CI: 0.49–1.58; p = 0.663).

    Combining LDAC with immunotherapy did not improve survival outcomes in the trials evaluating lenalidomide [36] or lintuzumab [52] (Supplementary Table 8).

    LDAC was also evaluated with GRASPA (erythrocyte encapsulated l-asparaginase), for the treatment of AML in the ENFORCE 1 trial [32]; the OS duration was statistically similar compared with LDAC monotherapy (Supplementary Table 8).

    Best supportive care

    Limited data were available comparing BSC with other treatment strategies. In the two studies [33,43] where BSC was a comparator group, BSC included administration of blood product transfusions, antimicrobials and other therapies for the symptomatic treatment of AML and its complications, according to institutional policies; hydroxyurea was permitted in both studies. In the AML-19 trial [33], a longer mOS duration was seen in the gemtuzumab ozogamicin group compared with the BSC group (4.9 months vs 3.6 months; HR = 0.69; 95% CI: 0.53–0.90; p = 0.005). In the FIGHT-AML-301 trial [43], targeted therapy – tipifarnib – resulted in similar mOS duration as in the BSC group (Supplementary Table 8).

    Other interventions

    During data extraction, the interventions in four trials [42,45,48,51] could not be categorised in one of the above groups and were, therefore, categorised as ‘other’. All four studies compared HMAs, LDAC, or BSC in one of the treatment arms. Three [42,45,51] compared HMA monotherapy with treatment choice (TC) and one trial [48] compared LDAC with BSC.

    In the DACO-016 trial, TC included BSC or LDAC, and although statistical differences in OS duration were not detected in the intention-to-treat (ITT) analysis [45], the increase in OS duration with decitabine therapy compared with TC therapy reached statistical significance in the ad hoc mature analysis (7.7 months vs 5.0 months; HR = 0.82; 95% CI: 0.68–0.99; p = 0.03) [78]. OS rates were also significantly higher in the decitabine arm compared with the TC arm at 18 months (p = 0.027) and 24 months (p = 0.019) in the post hoc analysis [77].

    In the AZA-AML-001 trial [42], 488 patients with AML and >30% BM blasts were randomised to the azacitidine arm or the ‘combined conventional care regimen (CCR)’, which included BSC, LDAC, or standard IC. Numerically, but not statistically significantly, higher mOS and mEFS durations were seen in the azacitidine arm compared with the combined CCR arm (Supplementary Table 8). However, the mRFS duration was slightly shorter with azacitidine (9.3 months vs 10.5 months; p = 0.6). Subgroup analyses for each of the treatments in the CCR group showed that azacitidine monotherapy resulted in significantly longer mOS compared with BSC, but not compared with LDAC or IC (Supplementary Table 8) [42]. Subgroup analysis [64] of patients with mIDH2 AML showed slight improvements in OS rates at 18 and 24 months in the azacitidine arm; however, improvements in OS were not seen in patients with mIDH1 AML, with numerically lower OS rates at 12 months with azacitidine in the mIDH1 AML group (49.2%) compared with the mIDH2 AML group (60.5%). Sample sizes were very small in these subgroups with mIDH AML that received azacitidine (n = 6 patients with mIDH1 AML; n = 21 with mIDH2 AML) (Supplementary Table 8).

    In the ASTRAL-1 trial [51], mOS duration and EFS rates were similar in the guadecitabine and TC (azacitidine, decitabine, or LDAC) arms [51,68], and mOS duration was similar in a subgroup analysis [85] of the different treatments within the TC arm (Supplementary Table 8).

    A small study compared LDAC with BSC in 60 patients with AML and showed that the mOS duration was 8.4 months in the BSC group but was not reached in the LDAC group (Supplementary Table 8).

    Transfusion independence outcomes

    Of the 26 identified clinical trials, seven [37–39,42,45,49,55] reported transfusion independence (TI) rates after intervention, either in the main publication [38,39,42,55] or in an associated paper [65,79,81,84,91]. These seven trials used variable definitions for TI. All seven studies defined TI as no transfusions for 56 consecutive days, but two (VIALE-A [38] and BRIGHT AML 1003 [79]) evaluated TI in all randomised patients, two trials (AG221-AML-005 [39], AZA-AML-001 [42,91]) reported TI only among patients who were transfusion dependent at baseline, and three trials (AGILE [91], VIALE-C [55,82,84], and DACO-016 [65]) reported TI for all randomised patients as well as patients who were transfusion dependent at baseline. Of these seven trials, five [38,65,79,81,91] reported statistically significant differences in OS between study arms, and the TI results for these five trials are illustrated in Figure 4.

    Figure 4. Post-baseline transfusion-independence rates in trials that showed statistically significant differences in median overall survival.

    *n, all patients/number of patients with baseline RBC transfusion dependence/number of patients with baseline PLT transfusion dependence/number of patients with baseline RBC and/or PLT transfusion dependence.

    Added to placebo.

    AGILE specifically enrolled an mIDH1 population. §6-month follow-up to primary data cut-off.

    AZA: Azacitidine; DEC: Decitabine; GLAS: Glasdegib; HMA: Hypomethylating agent; IVO: Ivosidenib; LDAC: Low-dose cytarabine; NP: Not performed; NR: Not reported; PLT: Platelet; RBC: Red blood cell; TC: Treatment choice; VEN: Venetoclax.

    In three of the seven trials that evaluated TI, HMA (azacitidine) was compared with targeted therapy [38,39,91]. The VIALE-A study [38], which included all patients in the TI definition, reported similar baseline transfusion-dependence rates in the venetoclax plus azacitidine (red blood cells [RBC]: 50.0%; platelets [PLT]: 24.0%) and azacitidine plus placebo (RBC: 52.0%; PLT: 22.0%) groups. After treatment, RBC and PLT TI rates in the venetoclax plus azacitidine groups were significantly higher than the rates in the azacitidine plus placebo group RBC: 59.8% (95% CI: 53.9–65.5) versus 35.2% (95% CI: 27.4–43.5); p < 0.001; PLT: 68.5% (95% CI: 62.8–73.9) versus 49.7% (95% CI: 41.3–58.1; p < 0.001; Figure 4).

    The AGILE trial [91] reported similar baseline RBC and/or PLT transfusion-dependence rates of 54% in the ivosidenib plus azacitidine and placebo plus azacitidine groups in patients with mIDH1 AML. Among this group of transfusion-dependent patients, ivosidenib plus azacitidine resulted in a significantly higher rate of RBC and/or PLT TI compared with placebo plus azacitidine (46.2% vs 17.5%; p = 0.006). An evaluation of all patients (regardless of baseline transfusion status) showed a numerically greater proportion of patients in the ivosidenib plus azacitidine group experiencing RBC and/or PLT TI compared with the placebo plus azacitidine group (62.5% vs 51.4%; p = 0.21) (Figure 4).

    The AG221-AML-005 trial [39] showed mixed results in patients with mIDH2 AML. A post hoc analysis showed that, among those who were transfusion dependent at baseline, a higher proportion of patients experienced RBC TI in the enasidenib plus azacitidine group than in the azacitidine monotherapy group (59% vs 41%; p-value not reported). In contrast, the proportion of patients that converted to PLT TI was lower in the enasidenib plus azacitidine group than the azacitidine monotherapy group (47% vs 63%; p-value not reported).

    Two of the seven studies evaluating TI compared LDAC to targeted therapy [55,79,82,84]. Both studies demonstrated higher TI rates in patients treated with LDAC in combination with targeted therapy compared with LDAC with or without placebo. In the VIALE-C study, the primary analysis showed that the RBC and PLT TI rates were significantly higher in patients treated with venetoclax plus LDAC compared with those treated with LDAC plus placebo [RBC: 41% (95% CI: 32–49) versus 18% (95% CI: 10–29); p = 0.001; PLT: 48% (95% CI: 39–56) versus 32% (95% CI: 22–45); p = 0.04)] [55], with similar results seen in the 6-month follow-up analysis [82]. The 6-month analysis also reported TI rates in the subgroup of patients who were transfusion dependent at baseline, showing numerically higher RBC and PLT TI rates in the venetoclax plus LDAC group compared with the LDAC plus placebo group [RBC: 40.4% (95% CI: 30.9–50.5) versus 16.7% (95% CI: 7.9–29.3); PLT: 28.8% (95% CI: 17.1–43.1) versus 12.5% (95% CI: 2.7–32.4)]; statistical analysis was not performed for this subgroup (Figure 4). In a cohort of 27 Japanese patients in the VIALE-C trial, post-baseline RBC and PLT TI rates were similar in both arms, likely due to the small sample size [84].

    In the post hoc analysis of the BRIGHT AML 1003 trial [79], higher rates of RBC and PLT TI were seen among patients treated with glasdegib plus LDAC compared with those treated with LDAC alone (RBC: 36.0% vs 8.0%; PLT: 47.0% vs 14.0%; p-values not reported) (Figure 4).

    In the DACO-016 study, RBC and PLT TI rates were significantly higher in patients randomised to decitabine compared with those randomised to TC (RBC: 30.0% vs 19.0%; p = 0.0058; PLT: 48.0% vs 35.0%; p = 0.0028)] [65]. In the subgroup of patients with transfusion dependence at baseline, post-baseline RBC and PLT TI rates were also significantly higher in the decitabine group compared with the TC group (RBC: 26.0% vs 13.0%; p = 0.0026; PLT: 31.0% vs 13.0%; p = 0.0069; Figure 4).

    In the AZA-AML-001 trial, a numerically higher proportion of patients in the azacitidine arm achieved RBC transfusion independence compared with CCR (38.5% vs 27.6%) as well as PLT TI (40.6% vs 29.3%) [42].

    Safety

    Of the 26 trials included in the SLR, six [34–36,48,54,56] did not report rates of all-cause or treatment-emergent/treatment-related AEs (TEAEs/TRAEs; Supplementary Table 9). Figure 5 illustrates the incidence of grade ≥3 key AEs (febrile neutropenia, pneumonia and QT interval prolongation) reported in the nine clinical trials [33,37,38,40,45,46,53,55,91] with statistically significant differences in mOS between study arms.

    Figure 5. Key adverse events in trials that showed statistically significant differences in median OS.

    *Reported in ≥10% of patients.

    Added to placebo. ‡AGILE specifically enrolled an mIDH1 population.

    §Two additional experimental groups included in the DECIDER trial – VAL + DEC and ATRA + DEC – are not included here.

    AE: Adverse event; ATRA: All-trans retinoic acid; AZA: Azacitidine; BSC: Best supportive care; DEC: Decitabine; FLUGA: Fludarabine, cytarabine and filgrastim, GLAS: Glasdegib; GO: Gemtuzumab ozogamicin; HMA: Hypomethylating agent; IVO: Ivosidenib; LDAC: Low-dose cytarabine; n: Sample size; NR: Not reported; OS: Overall survival; TC: Treatment choice; TEAEs: Treatment-emergent AE; VAL: Valproate; VEN: Venetoclax; VOL: Volasertib.

    HMAs

    Safety end points were reported in three trials [44,46,53] that compared an HMA with chemotherapy. In all three trials, the most common haematologic toxicities were neutropenia, febrile neutropenia and thrombocytopenia. Pneumonia was the most common non-haematologic toxicity reported in these studies (Supplementary Table 9).

    Of the three studies [47,50,56] that compared HMA monotherapy with immunotherapies, two [47,50] reported no relevant difference in safety outcomes between the arms (Supplementary Table 9), and one study [56] did not report rates of AEs.

    Of the four studies that compared HMA plus targeted therapy with HMA monotherapy (or with placebo), two trials reported higher rates of AEs in the targeted therapy arm compared with the HMA arm [38,39]. The other two trials reported similar safety outcomes in both arms [49,54]. In the VIALE-A trial [38], the most frequently reported grade ≥3 haematologic AEs in the azacitidine plus venetoclax and azacitidine plus placebo groups were thrombocytopenia, neutropenia, and febrile neutropenia, and the most common grade ≥3 non-haematologic AEs were hypokalaemia and pneumonia (Supplementary Table 9).

    In the AG221-AML-005 trial [39], neutropenia, thrombocytopenia, and anaemia were the most common grade 3–4 TRAEs (Supplementary Table 9). There was no incidence of differentiation syndrome in the azacitidine monotherapy group, but 12 (18%) patients in the combination group experienced 14 such events. The median time to onset of differentiation syndrome was 28.5 days (inter-quartile range [IQR]: 17.0–34.5); all 14 events resolved within a median of 11.5 days (IQR: 7.0–19.0).

    In the AGILE trial [91], the rates of AEs were similar between the ivosidenib plus azacitidine arm and the azacitidine plus placebo arm. Febrile neutropenia, neutropenia, anaemia, thrombocytopenia and pneumonia were the most common grade ≥3 TEAEs in both arms (Supplementary Table 9). The proportion of patients with infections was lower in the ivosidenib plus azacitidine group (any grade: 28%; grade ≥3: 21%) than the azacitidine plus placebo group (any grade: 49%; grade ≥3: 30%). The rates of febrile neutropenia were also lower in the ivosidenib plus azacitidine group (any grade: 28%; grade ≥3: 28%) than the azacitidine plus placebo group (any grade: 34%; grade ≥3: 34%). Consistent with these observations, an increase in the absolute neutrophil count (ANC) was noted from baseline over time in the ivosidenib plus azacitidine arm, particularly during the first treatment cycle. QT interval prolongation was reported more frequently with ivosidenib plus azacitidine (any grade: 20%; grade ≥3 10%) than with azacitidine plus placebo (any grade: 7%; grade ≥3: 3%). Differentiation syndrome was seen more frequently in the ivosidenib plus azacitidine group any grade: 14% (10 events); grade 3: 4% (3 events); grade 4: 0%) than the azacitidine plus placebo group [any grade: 8% (6 events); grade 3: 3% (2 events); grade 4: (1% (one event)], with a median onset of 19.5 days (range, 3.0–33.0).

    In a study comparing gilteritinib plus azacitidine with azacitidine monotherapy in patients with mFLT3 AML [54], rates of AEs were similar between arms (Supplementary Table 9).

    Low-dose cytarabine

    Two trials compared LDAC monotherapy (or plus placebo) with LDAC plus immunotherapy [36,52]. The LI-1 trial found that the rate of AEs was higher with lenalidomide plus LDAC than with LDAC monotherapy, including thrombotic events [36] (Supplementary Table 9). In contrast, the trial comparing lintuzumab plus LDAC with LDAC plus placebo showed no difference in the rates of AEs, except for a higher frequency of infusion-related reactions with lintuzumab [52] (Supplementary Table 9).

    All five trials that compared LDAC monotherapy (or with placebo) with LDAC plus targeted therapy reported higher rates of AEs in the combination therapy arms [31,37,40,41,55] (Supplementary Table 9). In the VIALE-C trial [55], the rates of AEs were higher in the venetoclax plus LDAC group compared with the LDAC plus placebo group. In the primary [55] and final [82] analyses, the most common haematologic grade ≥3 TEAEs in the venetoclax plus LDAC and LDAC plus placebo were febrile neutropenia, neutropenia, thrombocytopenia and anaemia (Supplementary Table 9).

    In the BRIGHT AML 1003 trial [37] the rates of AEs were higher in the glasdegib plus LDAC arm than the LDAC monotherapy arm, with anaemia, thrombocytopenia and febrile neutropenia as the most common haematologic grade ≥3 TEAEs. Grade 3 QTc prolongation (QTc ≥500 ms) was seen in 5/83 (6.0%%) patients treated with glasdegib plus LDAC and in 2/17 (11.8%) patients treated with LDAC monotherapy.

    In the SPARK-AML1 trial [31], the incidence of grade ≥3 AEs was higher in the barasertib arm (83%) than the LDAC monotherapy arm (69%).

    In the ENFORCE 1 trial, serious AEs were reported by 91.4% of patients receiving GRASPA plus LDAC and by 82.1% of patients receiving LDAC monotherapy [32] (Supplementary Table 9).

    Best supportive care

    In the two studies [33,43] where BSC was a comparator group, the AML-19 trial [33] reported no differences in the rate of AEs between the immunotherapy, gemtuzumab ozogamicin and BSC arms (Supplementary Table 9). In contrast, the FIGHT-AML-301 trial [43] showed higher rates of AEs with tipifarnib, particularly grade 4 neutropenia and thrombocytopenia as well as grade 3 or 4 diarrhoea, than with BSC (Supplementary Table 9).

    Other interventions

    Of the four trials [42,45,48,51] that compared other treatment strategies in patients with AML, three [42,45,51] reported safety outcomes. In the DACO-016 [45] and ASTRAL-1 [51] trials, the rate of AEs was higher in the decitabine (DACO-016) and guadecitabine (ASTRAL-1) arms compared with the TC arms (Supplementary Table 9). Conversely, comparable safety was seen in the azacitidine and CCR arms in the AZA-AML-001 trial (Supplementary Table 9) [42].

    Risk of bias

    The quality of the 26 included RCTs was assessed using the revised Cochrane Risk of Bias tool for RCTs (RoB 2.0) [92]. The risk of bias was assessed in five distinct domains, with each answer leading to judgements of ‘low risk of bias’, ‘some concerns’, or ‘high risk of bias’ (Figure 6). Concerning overall study risk of bias, 11 studies [33,38,41–43,45,46,49,50,52,55] showed a low risk, ten studies [34,35,37,39,40,44,47,48,51,53] showed some concerns, and five studies [31,32,36,54,56] showed a high risk of bias.

    Figure 6. Risk of bias.

    Discussion

    Multiple treatment strategies have been investigated for patients with newly diagnosed AML who are ineligible for chemotherapy. Studies that evaluated the strategy of combining LDAC with novel therapies showed that adding immunotherapy to LDAC did not provide any added survival benefit [36,52]; adding targeted therapy, such as venetoclax or glasdegib, to LDAC improved survival rates and TI rates but resulted in higher rates of AEs [37,40,67,82].

    Although not meeting the inclusion criteria for this SLR, it should be noted that data from a phase II single-arm trial showed that combining LDAC with cladribine (a purine nucleoside and inhibitor of S-adenosylhomocysteine hydrolase), alternating with decitabine, resulted in a mOS of 13.8 months and DFS of 10.8 months in patients with AML who were ≥70 years old and unfit for standard IC [93]. This regimen, which was generally well tolerated, may offer another treatment option for this population.

    Trials that compared novel therapies with BSC reported improved survival with immuno-chemotherapy (gemtuzumab ozogamicin) [33], but not targeted therapy (tipifarnib) [43]. Evaluation of other treatment strategies showed that OS improved significantly with decitabine compared with TC (supportive care or LDAC) [78], but no significant improvement was observed with azacitidine compared with BSC [42].

    HMAs are the mainstay of therapy, with some additional strategies showing survival benefits, such as adding FLUGA or ATRA chemotherapy to HMAs, but the toxicity rates in this difficult-to-treat patient population are unacceptably high [46,53].

    Of the newer treatments, adding immunotherapies to HMA therapy did not add any survival benefit [47,56]. Interestingly, allogeneic haematopoietic cell transplantation (alloHCT) is the most commonly used and best-established form of immunotherapy in AML, resulting in the longest OS duration in patients who are candidates for the procedure [6,7,15]. Unlike immunotherapy drugs, which are used in the setting of a very dysfunctional immune system in the presence of AML cells, alloHCT results in instalment of healthy immune and T cells from a donor, leading to a graft-versus-leukemia (GvL) response [94–96]. To improve the efficacy of immunotherapy drugs for patients with AML who are not candidates for alloHCT, novel cell-surface targets will need to be identified and evaluated for efficacy and toxicity for use in AML.

    Two new targeted therapies, venetoclax and ivosidenib, have shown the most promising and consistent beneficial survival results when added to an HMA [38,91]. In the VIALE-A study [38], venetoclax plus azacitidine was studied in 431 patients with AML ineligible for IC, with a subgroup of 34 patients with the IDH1 mutation. Although a reduction in the risk of death was shown with venetoclax and azacitidine compared with azacitidine plus placebo in the subgroup with mIDH1 AML, the results should be interpreted with caution due to the small sample size, lack of stratification for mutations at baseline, and lack of statistical testing for interaction between the mutations and survival. In a pooled analysis [97] of the VIALE-A trial and a single-arm phase Ib study that evaluated venetoclax with an HMA (azacitidine or decitabine), the mOS in the 44 patients with mIDH1 AML was 15.2 months (95% CI: 7.0–not estimable) in the combination therapy group compared with 2.2 months (95% CI: 1.1–5.6) in the HMA monotherapy group from the VIALE-A study (HR = 0.19; 95% CI: 0.08–0.44). Due to the drawbacks of pooling data, such as differences in study design, lack of a control group in the phase I trial and, therefore, a limited number of patients in the azacitidine monotherapy group, and the small sample sizes in the IDH1/2 molecular subgroups, the results of this analysis should be interpreted with caution.

    The low incidence of IDH1 mutations in patients with AML makes it challenging to conduct phase 3 studies in this population. The AGILE trial [91] evaluated ivosidenib plus azacitidine in 146 patients with mIDH1 AML and is the only phase III study in the AML population that exclusively enrolled patients with this mutation. The results from this trial showed significant increases in the duration of mOS and mEFS as well as improvements in the rates of OS and EFS at 24 months with ivosidenib plus azacitidine compared with azacitidine plus placebo. Compared with the VIALE-A trial, where the HR for OS in all randomised patients was 0.66 (95% CI: 0.52–0.85; p < 0.001) [38], the AGILE trial demonstrated a HR of 0.44 (95% CI: 0.27–0.73; p = 0.001) in the mIDH1 population [91], representing an increase in mOS by approximately 5 months when adding venetoclax to azacitidine and approximately 16 months when adding ivosidenib to azacitidine. Similarly, the HR for EFS was 0.63 (95% CI: 0.50–0.80; p < 0.001) in the VIALE-A trial [38] and 0.33 (95% CI: 0.16–0.69; p = 0.002) in the AGILE trial [91], representing an increase in the mEFS duration by approximately 3 months with venetoclax plus azacitidine and by approximately 19 months with ivosidenib plus azacitidine. This significant survival benefit with ivosidenib plus azacitidine may be partially due to the synergy seen in a preclinical trial with the two drugs [98]. The simultaneous administration of ivosidenib and azacitidine leads to inhibition of leukemia stem cells through suppression of mitogen-activated protein kinase and RB/E2F signalling, which are needed for cell proliferation and survival, resulting in a depletion of leukemia cells in mice by 7960-fold with this combination compared with azacitidine alone [98].

    Interestingly, the phase II AG221-AML-005 trial [39], conducted specifically in the mIDH2 AML population, had a comparable control group of azacitidine plus placebo as the phase III AGILE trial [91] performed in the mIDH1 AML population and the pooled analysis that included the VIALE-A and a phase 1 trial with venetoclax plus azacitidine [97]; however, the median survival durations in the control group were notably longer in the AG221-AML-005 trial (mOS: 22.0 months, 95% CI: 14.6–not reached; mEFS: 15.9, 95% CI: 13.0–not reached) compared with that in the AGILE trial (mOS: 7.9 months, 95% CI: 4.1–11.3; mEFS: 4.1, 95% CI: 2.7–6.8) and the subpopulation of patients with mIDH2 AML in the pooled analysis of the VIALE-A and phase I trial with venetoclax and azacitidine (mOS: 13.0 months, 95% CI: 3.8–15.8). It is not clear why such differences in survival were seen in the control groups of these three trials, but it may be due to the difference in the studied populations. In contrast to the AGILE trial, which enrolled patients with mIDH1 AML, the AG221-AML-005 trial enrolled patients with mIDH2 AML, and some data suggest that mIDH2 is an independent favourable prognostic factor in patients with AML [99,100]. The small sample size of 33 patients with mIDH2 AML in the pooled analysis of the VIALE-A and phase 1 trials with venetoclax and azacitidine may have contributed to the shorter mOS compared with that seen in the AG221-AML-005 trial.

    One efficacy end point that has not been well characterised in AML studies is TI, despite evidence from other haematologic cancers (e.g. myelodysplastic syndromes) showing that transfusion dependence significantly increases the economic, clinical, and quality of life (QoL) burden in patients [101]. Study cross-comparisons and interpretation of these data are challenging due to the various assessment methods used across studies and some trials [42,79] not reporting baseline transfusion-dependence data.

    The added risk of toxicities with combination therapy requires careful consideration, since adding an additional drug generally may result in increased toxicity, as was seen in clinical trials that evaluated adding certain targeted drugs to an HMA [38,39] and to LDAC [31,37,40,41,55], or adding immunotherapy to LDAC [36]. In the VIALE-A study, a higher rate of serious AEs was reported with the combination of venetoclax and azacitidine compared with azacitidine plus placebo, with febrile neutropenia and pneumonia being the most reported serious AEs [38]. Interestingly, the rates for these two AEs were lower in the ivosidenib plus azacitidine group than in the azacitidine plus placebo group in the AGILE trial, and the rate of infections was lower in the ivosidenib plus azacitidine group. These findings are likely due to the increase in the ANC and leucocytosis seen with ivosidenib. This increase in neutrophils has not been observed with the other targeted therapies studied in patients with AML but has been noted in some studies with ivosidenib [102,103]. A bone marrow analysis in a phase I trial of patients with mIDH1 AML showed that ivosidenib induced myeloid differentiation and trilineage haematopoietic recovery without an intervening period of bone marrow aplasia, indicating that functional neutrophils are involved in haematopoietic recovery, consistent with ivosidenib's mechanism of action of differentiation [102].

    The incidence of differentiation syndrome reported with ivosidenib plus azacitidine in the AGILE study [91] was similar to that seen with enasidenib plus azacitidine in the AG221-AML-005 trial [39], with supportive care resulting in resolution of this adverse event. QTc interval prolongation has been noted with targeted therapies [104], and grade ≥3 QTc prolongation (QTc >500 ms) was seen in the AGILE trial [91] and the BRIGHT AML 1003 trial [37]. Torsades de pointes was not seen in these studies, but the risk of QTc prolongation should be considered when initiating targeted therapies, and electrolyte levels should be closely monitored throughout the duration of treatment.

    As the landscape of AML therapies evolves, health-related quality of life (HRQoL) is a crucial consideration in the overall management of AML, since the goal of therapy is not just to prolong survival, but to optimise QoL and minimise the risk of cytopenia-related complications [15]. As HRQoL scores in adults with AML have been reported to be significantly lower than in the population norms and lower than those in many other cancers [105,106], this element becomes a great challenge and is of utmost importance in older patients with AML who are ineligible for standard IC [107].

    Interpreting HRQoL outcomes is challenging due to the various instruments used to capture the physical, emotional, social and other domains. Additionally, the study population evaluable for HRQoL in AML studies is often smaller than the ITT population, diminishing over the study period due to the high mortality rate. Our SLR identified only four trials [38,55,63,91] that reported HRQoL data with targeted therapies. In the AG221-AML-005 trial, treatment with enasidenib plus azacitidine did not result in improvement of HRQoL outcomes [39]. In the VIALE-C study, better improvements from baseline in fatigue and global health status were seen in the venetoclax plus LDAC arm compared with the LDAC plus placebo arm, and there was a trend of longer time to deterioration in the venetoclax plus LDAC arm across each subscale [55]. In the VIALE-A study, there were no differences from baseline in the QoL measures between the venetoclax plus azacitidine group and the azacitidine plus placebo group [38]. In the AGILE trial, improvements from baseline across all European Organisation for Research and Treatment of Cancer QLQ-C30 subscales were noted in the ivosidenib plus azacitidine arm [91]. As targeted therapies lead to prolonged survival in patients with AML, it will be important to assess the long-term tolerance of these agents and to compare the HRQoL outcomes associated with these therapies.

    The limitations associated with SLRs should be considered when interpreting these findings. Characteristic to all SLRs, some risks include missing relevant publications due to improper indexing within databases, restricting this SLR to English-language publications, or articles published after the searches were performed. Three major databases were searched for data extraction, along with relevant congress proceedings, to avoid missing relevant publications. It is challenging to make cross-study comparisons or generalised conclusions with SLRs due to different outcomes reported across studies. Variability in the baseline characteristics of enrolled patients can also make it challenging to compare outcomes across studies, although the key baseline characteristics that may have affected survival outcomes (age, cytogenics, and AML type) were similar across the nine trials [33,37,38,40,45,46,53,55,91] that demonstrated statistically significant differences in the OS outcome between study arms (Supplementary Figures 1–3). AEs are not uniformly reported in the trials included in our SLR. Nine trials [37,39,42–47,55] did not report rates of all-cause AEs, creating a potential gap and challenge in comparing AEs across studies. Additionally, two of the trials [39,43] reported rates of TRAEs instead of TEAEs. Lastly, five trials [31,32,36,54,56] showed a high risk of bias, and not all 26 included trials reported on certain outcomes of interest, such as TI and HRQoL outcomes.

    Conclusion

    Patients with AML who are ineligible for IC have historically had poor outcomes with the available treatment options. Novel therapies have improved clinical outcomes in this patient population, with targeted therapy combined with an HMA showing the most promising results. Adding ivosidenib or venetoclax to azacitidine or adding volasertib, venetoclax or glasdegib to LDAC significantly improved OS. In line with the most recent NCCN and ELN guidelines [6,15], ivosidenib in combination with azacitidine in mIDH1 AML and venetoclax in combination with azacitidine in the general AML population showed the most promising results. Some treatment-specific AEs highlighted in this SLR require attention during treatment, and management recommendations have been developed for these. Other important factors to consider for treatment decisions include patient's genetic characteristics, comorbidities, fitness, and social support. New treatment options have been and will continue to be incorporated in treatment guidelines for AML. The efficacy of the new treatment combinations for second-line treatment of patients ineligible for IC is largely unknown and has not been evaluated in this review. Thus, the sequential efficacy of treatments cannot be considered for the choice of the first-line treatment. However, targetable mutations may get lost at relapse in a variable proportion of patients, which may preclude the use of the targeting agent at relapse. Successful development of targeted agents in AML supports the approach of dissecting AML into well-described genetic subgroups and developing treatment strategies for each subgroup individually.

    Summary points
    • Treatment options for newly diagnosed patients with acute myeloid leukemia (AML) ineligible for induction chemotherapy are expanding.

    • Of the novel therapies, targeted drugs (venetoclax [BCL-2 inhibitor] and ivosidenib [IDH1 inhibitor]) have shown the most consistent and highest survival benefits, when combined with azacitidine.

    • Screening for genetic mutations is now a standard part of the initial clinical workup in AML.

    • IDH1 mutations in AML are rare, making it challenging to conduct phase III clinical trials in patients with mIDH1 AML.

    • Ivosidenib is the only targeted drug that has been studied (in combination with azacitidine) specifically in patients with mIDH1 AML in a phase III trial; it showed a significant improvement in overall survival by 16 months compared with the control group of azacitidine plus placebo.

    • Venetoclax plus azacitidine showed an improvement of 5 months compared with the control group of azacitidine plus placebo in a post hoc analysis of patients with mIDH1 in a phase III trial.

    • Added toxicities should be considered when using combination therapies in patients with AML.

    • Patients initiating on combination therapies should be closely monitored as they have an increased risk of bone marrow suppression and infections, such as pneumonia.

    • Unlike the other studies with combination therapies, the trial comparing ivosidenib plus azacitidine with azacitidine plus placebo showed similar frequencies of adverse events in the two groups, as well as a decrease in the frequency of infectious complications with ivosidenib plus azacitidine, possibly due to the increase in absolute neutrophil count that is seen with ivosidenib.

    • Patients initiating on targeted therapies such as ivosidenib, glasdegib, gilteritinib, and enasidenib should be monitored for differentiation syndrome as well as QTc prolongation.

    Supplementary data

    An infographic accompanies this paper. To view or download this infographic in your browser please click here: https://www.futuremedicine.com/doi/suppl/10.2217/fon-2022-1286

    Author contributions

    All authors contributed to the conception or design of this work or the acquisition, analysis, or interpretation of data for the work. All authors contributed to drafting or revising this manuscript for important intellectual content. All authors provided their final approval of the version to be published. All authors agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

    Financial & competing interests disclosure

    M Heuser: Research funding: Abbvie, Astellas, BergenBio, BMS, Bayer Pharma AG, Daiichi Sankyo, Jazz Pharmaceuticals, Karyopharm, Novartis, Pfizer, PinotBio, Roche; Honoraria: Abbvie, Jazz Pharmaceuticals, Janssen, Novartis; Consulting: Abbvie, BMS, Daiichi Sankyo, Jazz Pharmaceuticals, Novartis, Pfizer, PinotBio, Roche, Tolremo. C Fernandez is an employee of Servier. O Hauch is a consultant for Servier. OM Klibanov is an employee of IQVIA. T Chaudhary is an employee of IQVIA. V Rives is an employee of Servier. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Medical writing support was provided by L Huber and R Kurtkoti of IQVIA, which was funded by Servier, Suresnes, France, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3).

    Ethical disclosure

    This was a systematic literature review; no institutional review board approval was required.

    Data sharing statement

    This manuscript does not report original results of a clinical trial or secondary results of clinical trial data. The protocol for this SLR, including data collection form and data extracted from included studies, was developed and can be provided upon request to the corresponding author.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1. De Kouchkovsky I, Abdul-Hay M. Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J. 6(7), e441 (2016).
    • 2. Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood 106(4), 1154–1163 (2005).
    • 3. Shallis RM, Wang R, Davidoff A, Ma X, Zeidan AM. Epidemiology of acute myeloid leukemia: recent progress and enduring challenges. Blood Rev. 36, 70–87 (2019).
    • 4. Cancer.net. Leukemia - Acute Myeloid - AML: statistics. www.cancer.net/cancer-types/leukemia-acute-myeloid-aml/statistics (Accessed August 23, 2022).
    • 5. The Economist Intelligence Unit. Acute Myeloid leukemia: mapping the policy response to an acute cancer in France, Germany, Italy, Spain and the UK. https://worldcancerinitiative.economist.com/pdf/acute-myeloid-leukemia/acute-myeloid-leukemia-report.pdf (Accessed 23 August 2022).
    • 6. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology. Acute Myeloid Leukemia, version 2.2022-June 14, 2022. www.nccn.org/professionals/physician_gls/pdf/aml.pdf (Accessed 23 August 2022).
    • 7. Heuser M, Ofran Y, Boissel N et al. Acute myeloid leukemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 31(6), 697–712 (2020).
    • 8. Meyers J, Yu Y, Kaye JA, Davis KL. Medicare fee-for-service enrollees with primary acute myeloid leukemia: an analysis of treatment patterns, survival, and healthcare resource utilization and costs. Appl. Health. Econ. Health Policy 11(3), 275–286 (2013).
    • 9. Oran B, Weisdorf DJ. Survival for older patients with acute myeloid leukemia: a population-based study. Haematologica 97(12), 1916–1924 (2012).
    • 10. Walter RB, Estey EH. Management of older or unfit patients with acute myeloid leukemia. Leukemia 29(4), 770–775 (2015).
    • 11. Ferrara F, Barosi G, Venditti A et al. Consensus-based definition of unfitness to intensive and non-intensive chemotherapy in acute myeloid leukemia: a project of SIE, SIES and GITMO group on a new tool for therapy decision making. Leukemia 27(5), 997–999 (2013).
    • 12. Krug U, Rollig C, Koschmieder A et al. Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukemia: a web-based application for prediction of outcomes. Lancet 376(9757), 2000–2008 (2010).
    • 13. Walter RB, Othus M, Borthakur G et al. Prediction of early death after induction therapy for newly diagnosed acute myeloid leukemia with pretreatment risk scores: a novel paradigm for treatment assignment. J. Clin. Oncol. 29(33), 4417–4423 (2011).
    • 14. Sorror ML, Storer BE, Fathi AT et al. Development and validation of a novel acute myeloid leukemia-composite model to estimate risks of mortality. JAMA Oncol. 3(12), 1675–1682 (2017).
    • 15. Dohner H, Wei AH, Appelbaum FR et al. Diagnosis and management of AML in adults: 2022 ELN recommendations from an International Expert Panel. Blood doi:10.1182/blood.2022016867 (2022).
    • 16. Elnahass YH, Badawy RH, Elrefaey FA et al. IDH mutations in AML patients; a higher association with intermediate risk cytogenetics. Asian Pac. J. Cancer Prev. 21(3), 721–725 (2020).
    • 17. Schlenk RF, Dohner K, Krauter J et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 358(18), 1909–1918 (2008).
    • 18. Abbas S, Lugthart S, Kavelaars FG et al. Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood 116(12), 2122–2126 (2010).
    • 19. Medeiros BC, Fathi AT, Dinardo CD, Pollyea DA, Chan SM, Swords R. Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia 31(2), 272–281 (2017).
    • 20. Chou WC, Hou HA, Chen CY et al. Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation. Blood 115(14), 2749–2754 (2010).
    • 21. Mardis ER, Ding L, Dooling DJ et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361(11), 1058–1066 (2009).
    • 22. Chotirat S, Thongnoppakhun W, Promsuwicha O, Boonthimat C, Auewarakul CU. Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients. J. Hematol. Oncol. 5, 5 (2012).
    • 23. Wagner K, Damm F, Gohring G et al. Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J. Clin. Oncol. 28(14), 2356–2364 (2010).
    • 24. European Medicines Agency. Public summary of opinion on orphan designation: ivosidenib for the treatment of acute myeloid leukemia. www.ema.europa.eu/en/documents/orphan-designation/eu/3/16/1802-public-summary-opinion-orphan-designation-ivosidenib-treatment-acute-myeloid-leukemia_en.pdf (Accessed 23 August 2022).
    • 25. PR Newswire. Servier submits a marketing authorization application to the European Medicines Agency (EMA) for TIBSOVO® (ivosidenib tablets) for patients with IDH1-mutated acute myeloid leukemia (AML) and cholangiocarcinoma. www.prnewswire.com/news-releases/servier-submits-a-marketing-authorization-application-to-the-european-medicines-agency-ema-for-tibsovo-ivosidenib-tablets-for-patients-with-idh1-mutated-acute-myeloid-leukemia-aml-and-cholangiocarcinoma-301500159.html (Accessed August 23, 2022).
    • 26. Higgins JPT, Thomas J, Chandler J et al. Cochrane Handbook for Systematic Reviews of Interventions version 6.3 (updated February 2022). Cochrane (2022).
    • 27. Centre for Reviews and Dissemination, University of York. Systematic reviews: CRD's guidance for undertaking reviews in health care. www.york.ac.uk/media/crd/Systematic_Reviews.pdf (Accessed 24 August 2022).
    • 28. Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLOS Med. 6(7), e1000097 (2009).
    • 29. National Institute for Health and Care Excellence. Single technology appraisal and highly specialised technologies evaluation: user guide for company evidence submission template. www.nice.org.uk/process/pmg24/chapter/clinical-effectiveness#quality-assessment-of-the-relevant-clinical-effectiveness-evidence (Accessed 24 August 2022).
    • 30. Github. WebPlotDigitizer. https://apps.automeris.io/wpd/ (Accessed 12 September 2022).
    • 31. Kantarjian HM, Martinelli G, Jabbour EJ et al. Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer 119(14), 2611–2619 (2013).
    • 32. EU Clinical Trials Register. A multicenter, open, randomized, controlled phase IIb trial evaluating efficacy and tolerability of GRASPA (L-asparaginase encapsulated in red blood cells, eryaspase) plus low-dose cytarabine versus low-dosecytarabine alone, in treatment of newly diagnosed acute myeloid leukemia (AML) elderly patients, unfit forintensive chemotherapy. ENFORCE 1 study. www.clinicaltrialsregister.eu/ctr-search/trial/2012-002026-78/results (Accessed 25 August 2022).
    • 33. Amadori S, Suciu S, Selleslag D et al. Gemtuzumab ozogamicin versus best supportive care in older patients with newly diagnosed acute myeloid leukemia unsuitable for intensive chemotherapy: results of the randomized phase III EORTC-GIMEMA AML-19 trial. J. Clin. Oncol. 34(9), 972–979 (2016).
    • 34. Burnett AK, Russell N, Hills RK et al. A randomised comparison of the novel nucleoside analogue sapacitabine with low-dose cytarabine in older patients with acute myeloid leukemia. Leukemia 29(6), 1312–1319 (2015).
    • 35. Burnett AK, Russell NH, Hunter AE et al. Clofarabine doubles the response rate in older patients with acute myeloid leukemia but does not improve survival. Blood 122(8), 1384–1394 (2013).
    • 36. Copland M, Ariti C, Thomas I et al. A randomised evaluation of low-dose cytarabine arabinoside plus lenalidomide versus single-agent low-dose cytarabine arabinoside in older patients with acute myeloid leukemia: results from the LI-1 trial. Blood 138(Suppl. 1), 1266 (2021).
    • 37. Cortes JE, Heidel FH, Hellmann A et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia 33(2), 379–389 (2019). •• A phase II clinical trial describing the efficacy and safety of glasdegib plus low-dose cytarabine (LDAC) compared to LDAC monotherapy in patients with acute myeloid leukemia (AML), showing significantly longer median duration of overall survival (OS) with combination therapy compared to control.
    • 38. Dinardo CD, Jonas BA, Pullarkat V et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N. Engl. J. Med. 383(7), 617–629 (2020). •• A phase III clinical trial describing the efficacy and safety of venetoclax plus azacitidine compared to azacitidine plus placebo in patients with AML, showing an increase in median OS (mOS) of 5 months and median event-free survival (mEFS) of 3 months with combination therapy compared to the control group.
    • 39. Dinardo CD, Schuh AC, Stein EM et al. Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 22(11), 1597–1608 (2021). •• A phase II clinical trial describing the efficacy and safety of enasidenib plus azacitidine compared to azacitidine monotherapy in patients with IDH2m AML, showing no significant survival benefits with combination therapy compared to the control group.
    • 40. Dohner H, Lubbert M, Fiedler W et al. Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy. Blood 124(9), 1426–1433 (2014).
    • 41. Dohner H, Symeonidis A, Deeren D et al. Adjunctive volasertib in patients with acute myeloid leukemia not eligible for standard induction therapy: a randomized, phase 3 trial. Hemasphere 5(8), e617 (2021).
    • 42. Dombret H, Seymour JF, Butrym A et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 126(3), 291–299 (2015).
    • 43. Harousseau JL, Martinelli G, Jedrzejczak WW et al. A randomized phase 3 study of tipifarnib compared with best supportive care, including hydroxyurea, in the treatment of newly diagnosed acute myeloid leukemia in patients 70 years or older. Blood 114(6), 1166–1173 (2009).
    • 44. Kantarjian HM, Begna KH, Altman JK et al. Results of a randomized phase 3 study of oral sapacitabine in elderly patients with newly diagnosed acute myeloid leukemia (SEAMLESS). Cancer 127(23), 4421–4431 (2021).
    • 45. Kantarjian HM, Thomas XG, Dmoszynska A et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J. Clin. Oncol. 30(21), 2670–2677 (2012). •• A phase III clinical trial describing the efficacy and safety of decitabine compared to treatment choice (supportive care or LDAC) in patients with AML, showing significantly longer median duration of OS with decitabine compared to treatment choice.
    • 46. Lubbert M, Grishina O, Schmoor C et al. Valproate and retinoic acid in combination with decitabine in elderly nonfit patients with acute myeloid leukemia: results of a multicenter, randomized, 2 × 2, phase II trial. J. Clin. Oncol. 38(3), 257–270 (2020).
    • 47. Medeiros BC, Mccaul K, Kambhampati S et al. Randomized study of continuous high-dose lenalidomide, sequential azacitidine and lenalidomide, or azacitidine in persons 65 years and over with newly-diagnosed acute myeloid leukemia. Haematologica 103(1), 101–106 (2018).
    • 48. Mohammed AA, Elsayed FM. Low-dose cytarabine versus best supportive care for patients with acute myeloid leukemia unfit for the standard of care: egyptian centre experience. Middle East J. Cancer 12(3), 415–421 (2021).
    • 49. Montesinos P, Recher C, Vives S et al. AGILE: a global, randomized, double-blind, phase 3 study of ivosidenib + azacitidine versus placebo + azacitidine in patients with newly diagnosed acute myeloid leukemia with an IDH1 mutation. 63rd American Society of Hematology (ASH) Annual Meeting doi:10.1182/blood-2021-147805 (2021).
    • 50. Montesinos P, Roboz GJ, Bulabois CE et al. Safety and efficacy of talacotuzumab plus decitabine or decitabine alone in patients with acute myeloid leukemia not eligible for chemotherapy: results from a multicenter, randomized, phase 2/3 study. Leukemia 35(1), 62–74 (2021).
    • 51. Roboz GJ, Döhner H, Gobbi M et al. Results from a global randomized phase 3 study of guadecitabine (G) vs treatment choice (TC) in 815 patients with treatment naïve (TN) AML unfit for intensive chemotherapy (IC) ASTRAL-1 study: analysis by number of cycles. Blood 134(Suppl. 1), 2591 (2019).
    • 52. Sekeres MA, Lancet JE, Wood BL et al. Randomized phase IIb study of low-dose cytarabine and lintuzumab versus low-dose cytarabine and placebo in older adults with untreated acute myeloid leukemia. Haematologica 98(1), 119–128 (2013).
    • 53. Vives S, Martinez-Cuadron D, Bergua Burgues J et al. A phase 3 trial of azacitidine versus a semi-intensive fludarabine and cytarabine schedule in older patients with untreated acute myeloid leukemia. Cancer 127(12), 2003–2014 (2021). •• A phase III clinical trial describing the efficacy and safety of fudarabine, cytarabine and filgrastim (FLUGA) plus azacitidine compared to azacitidine monotherapy in patients with AML, showing significantly longer median duration of OS and EFS with azacitidine compared to FLUGA.
    • 54. Wang ES, Montesinos P, Minden MD et al. Phase 3, open-label, randomized study of gilteritinib and azacitidine vs azacitidine for newly diagnosed FLT3-mutated acute myeloid leukemia in patients ineligible for intensive induction chemotherapy. Blood 138(Suppl. 1), 700 (2021).
    • 55. Wei AH, Montesinos P, Ivanov V et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood 135(24), 2137–2145 (2020). •• A phase III clinical trial describing the efficacy and safety of venetoclax plus LDAC compared to LDAC monotherapy in patients with AML, showing significantly longer median duration of OS and EFS at the final analysis with combination therapy compared to control.
    • 56. Zeidan AM, Cavenagh J, Voso MT et al. Efficacy and safety of azacitidine (AZA) in combination with the anti-PD-L1 durvalumab (durva) for the front-line treatment of older patients (pts) with acute myeloid leukemia (AML) who are unfit for intensive chemotherapy (IC) and pts with higher-risk myelodysplastic syndromes (HR-MDS): results from a large, international, randomized phase 2 study. Blood 134(Suppl. 1), 829 (2019).
    • 57. Fenaux P, Gobbi M, Kropf PL et al. Results of ASTRAL-1 study, a phase 3 randomized trial of guadecitabine (G) vs treatment choice (TC) in treatment naïve acute myeloid leukemia (TN-AML) not eligible for intensive chemotherapy (IC). Hemasphere 3(Suppl. 1), 394–395 (2019).
    • 58. ClinicalTrials.gov. SGI-110 in adults with untreated acute myeloid leukemia (AML), not considered candidates for intensive remission induction. www.clinicaltrials.gov/ct2/show/NCT02348489 (Accessed 25 August 2022).
    • 59. Amadori S, Suciu S, Selleslag D et al. Improved overall survival with gemtuzumab ozogamicin (GO) compared with best supportive care (BSC) in elderly patients with untreated acute myeloid leukemia (AML) not considered fit for intensive chemotherapy: final results from the randomized phase III study (AML-19) of the EORTC and Gimema Leukemia Groups. Blood 124(21), 619 (2014).
    • 60. Arthur C, Cermak J, Delaunay J et al. Post hoc analysis of the relationship between baseline white blood cell count and survival outcome in a randomized phase III trial of decitabine in older patients with newly diagnosed acute myeloid leukemia. J. Blood Med. 6, 25–29 (2015).
    • 61. Ayala R, Rapado I, Onecha E et al. The mutational landscape of acute myeloid leukemia predicts responses and outcomes in elderly patients from the PETHEMA-FLUGAZA phase 3 clinical trial. Cancers (Basel) 13(10), (2021).
    • 62. Becker H, Schmoor C, Grishina O et al. Randomized phase II study of all-trans retinoic acid and valproic acid added to decitabine in newly diagnosed elderly AML patients (DECIDER trial): predictive impact of TP53 status. Blood 138(Suppl. 1), 2380 (2021).
    • 63. Dinardo CD, Dohner H, Zeidan AM et al. Health-related quality of life (HRQoL) during treatment with enasidenib (ENA) plus azacitidine (AZA) in patients with newly diagnosed mutant IDH2 (m IDH2) acute myeloid leukemia (AML) not eligible for intensive chemotherapy (IC). Blood 138(Suppl. 1), 1244 (2021).
    • 64. Dohner H, Dolnik A, Tang L et al. Cytogenetics and gene mutations influence survival in older patients with acute myeloid leukemia treated with azacitidine or conventional care. Leukemia 32(12), 2546–2557 (2018).
    • 65. He J, Xiu L, De Porre P, Dass R, Thomas X. Decitabine reduces transfusion dependence in older patients with acute myeloid leukemia: results from a post hoc analysis of a randomized phase III study. Leuk. Lymphoma 56(4), 1033–1042 (2015).
    • 66. Heuser M, Robak T, Montesinos P et al. Glasdegib (GLAS) plus low-dose cytarabine (LDAC) in AML or MDS: bright AML 1003 final report and 4-year overall survival (OS) follow-up. Hemasphere 4(Suppl. 1), 225 (2020).
    • 67. Heuser M, Smith BD, Fiedler W et al. Clinical benefit of glasdegib plus low-dose cytarabine in patients with de novo and secondary acute myeloid leukemia: long-term analysis of a phase II randomized trial. Ann. Hematol. 100(5), 1181–1194 (2021).
    • 68. Issa J-P, Gobbi M, Kropf PL et al. Progression free survival (PFS), and event free survival (EFS) from a global randomized phase 3 study of guadecitabine (G) vs treatment choice (TC) in 815 patients with treatment naive (TN) AML unfit for intensive chemotherapy (IC): ASTRAL-1 study. Blood 134(Suppl. 1), 4235 (2019).
    • 69. Lin S, Shaik N, Chan G, Cortes JE, Ruiz-Garcia A. An evaluation of overall survival in patients with newly diagnosed acute myeloid leukemia and the relationship with glasdegib treatment and exposure. Cancer Chemother. Pharmacol. 86(4), 451–459 (2020).
    • 70. Luebbert M, Grishina O, Schmoor C et al. Activity of decitabine (DAC) combined with all-trans retinoic acid (ATRA) in oligoblastic AML: subgroup analysis of a randomized 2 × 2 phase II trial. Blood 136(Suppl. 1), 9–10 (2020).
    • 71. Mayer J, Arthur C, Delaunay J et al. Multivariate and subgroup analyses of a randomized, multinational, phase 3 trial of decitabine vs treatment choice of supportive care or cytarabine in older patients with newly diagnosed acute myeloid leukemia and poor- or intermediate-risk cytogenetics. BMC Cancer 14, 69 (2014).
    • 72. Papayannidis C, Smith BD, Heuser M et al. Low-dose cytarabine with or without glasdegib in newly diagnosed patients with acute myeloid leukemia: long-term analysis of a phase 2 randomized trial. Clin. Lymphoma Myeloma Leuk. 19(Suppl. 1), S228–S229 (2019).
    • 73. Pratz KW, Dinardo CD, Selleslag D et al. Cytopenia management in patients with newly diagnosed acute myeloid leukemia treated with venetoclax plus azacitidine in the VIALE-A study. Blood 136(Suppl. 1), 51–53 (2020).
    • 74. Seymour JF, Dohner H, Butrym A et al. Azacitidine improves clinical outcomes in older patients with acute myeloid leukemia with myelodysplasia-related changes compared with conventional care regimens. BMC Cancer 17(1), 852 (2017).
    • 75. Smith BD, Papayannidis C, Heuser M et al. Low-dose cytarabine with or without glasdegib in newly diagnosed patients with acute myeloid leukemia: long-term analysis of a phase 2 randomized trial. J. Clin. Oncol. 37(Suppl. 15), 7010 (2019).
    • 76. Solem CT, Bell TJ, Kwon Y et al. A quality-adjusted survival time without symptoms or toxicities analysis of glasdegib plus low-dose cytarabine versus low-dose cytarabine as initial therapy for acute myeloid leukemia in patients who are not considered candidates for intensive chemotherapy. Cancer 126(19), 4315–4321 (2020).
    • 77. Thomas XG, Arthur C, Delaunay J, Jones M, Berrak E, Kantarjian HM. A post hoc sensitivity analysis of survival probabilities in a multinational phase III trial of decitabine in older patients with newly diagnosed acute myeloid leukemia. Clin. Lymphoma Myeloma Leuk. 14(1), 68–72 (2014).
    • 78. Tomeczkowski J, Lange A, Guntert A et al. Converging or crossing curves: untie the gordian knot or cut it? Appropriate statistics for non-proportional hazards in decitabine DACO-016 study (AML). Adv. Ther. 32(9), 854–862 (2015).
    • 79. Wang ES, Heuser M, Montesinos P et al. Glasdegib with LDAC in newly diagnosed patients with acute myeloid leukemia (AML) unsuitable for intensive chemotherapy: effects on transfusions and marrow recovery vs LDAC alone. Hemasphere 3(Suppl. 1), 87–88 (2019).
    • 80. Wang ES, Heuser M, Sekeres MA et al. Effect of early blood counts on response and overall survival following glasdegib plus LDAC in newly diagnosed AML: Bright AML 1003 post hoc analysis. Hemasphere 4(Suppl. 1), 273 (2020).
    • 81. Wei AH, Panayiotidis P, Montesinos P et al. Cytopenia management in the phase 3 Viale-C study of venetoclax plus low-dose cytarabine for patients with newly diagnosed acute myeloid leukemia ineligible for intensive chemotherapy. Hemasphere 5(Suppl. 2), 179 (2021).
    • 82. Wei AH, Panayiotidis P, Montesinos P et al. 6-month follow-up of VIALE-C demonstrates improved and durable efficacy in patients with untreated AML ineligible for intensive chemotherapy (141/150). Blood Cancer J. 11(10), 163 (2021).
    • 83. Wierzbowska A, Wawrzyniak E, Pluta A et al. Decitabine improves response rate and prolongs progression-free survival in older patients with newly diagnosed acute myeloid leukemia and with monosomal karyotype: a subgroup analysis of the DACO-016 trial. Am. J. Hematol. 93(5), E125–E127 (2018).
    • 84. Yamauchi T, Yoshida C, Usuki K et al. Venetoclax plus low-dose cytarabine in Japanese patients with untreated acute myeloid leukemia ineligible for intensive chemotherapy. Jpn J. Clin. Oncol. 51(9), 1372–1382 (2021).
    • 85. Zeidan AM, Fenaux P, Gobb M et al. Comparative results of azacitidine and decitabine from a large prospective phase 3 study in treatment naive patients with acute myeloid leukemia not eligible for intensive chemotherapy. 62nd ASH Annual Meeting and Exposition (2020).
    • 86. Zeidan AM, Schuster MW, Krauter J et al. Clinical benefit of glasdegib in combination with azacitidine or low-dose cytarabine in patients with acute myeloid leukemia. Blood 134(Suppl. 1), 3916 (2019).
    • 87. Jonas BA, Dinardo CD, Fracchiolla N et al. CYP3A inhibitors and impact of these agents on outcomes in patients with acute myeloid leukemia treated with venetoclax plus azacitidine on the VIALE-A study. Blood 136(Suppl. 1), 50–52 (2020).
    • 88. EU Clinical Trials Register. A randomized study of tipifarnib versus best supportive care (including hydroxyurea) in the treatment of newly diagnosed acute myeloid leukemia (AML) in subjects 70 years or older. www.clinicaltrialsregister.eu/ctr-search/trial/2004-001719-77/results (Accessed 7 September 2022).
    • 89. Heuser M, Fiedler W, Sekeres MA et al. Clinical benefit of glasdegib plus low-dose cytarabine in patients with de novo and secondary acute myeloid leukemia: long-term analysis of a phase 2 randomized trial. The 24th European Hematology Association (EHA) Meeting doi:10.1016/j.clml.2019.07.116 (2019).
    • 90. ClinicalTrials.gov. An efficacy and safety study of decitabine (DACOGEN) plus talacotuzumab (JNJ-56022473; Anti CD123) versus decitabine (DACOGEN) alone in participants with acute myeloid Leukemia (AML) ineligible for intensive chemotherapy. https://clinicaltrials.gov/ct2/show/study/NCT02472145?term=An+Efficacy+and+Safety+Study+of+Decitabine+%28DACOGEN%29+Plus+Talacotuzumab+%28JNJ-56022473%3B+Anti+CD123%29+Versus+Decitabine+%28DACOGEN%29+Alone+in+Participants+With+Acute+Myeloid&draw=2&rank=1 (Accessed 23 September 2022).
    • 91. Montesinos P, Recher C, Vives S et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N. Engl. J. Med. 386(16), 1519–1531 (2022). •• A phase III clinical trial describing the efficacy and safety of ivosidenib plus azacitidine compared to azacitidine plus placebo in patients with IDH1m AML, showing an increase in mOS and EFS with combination therapy compared to the control group.
    • 92. Sterne JA, Savovic J, Page MJ et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 366, l4898 (2019).
    • 93. Kadia TM, Cortes J, Ravandi F et al. Cladribine and low-dose cytarabine alternating with decitabine as front-line therapy for elderly patients with acute myeloid leukemia: a phase 2 single-arm trial. Lancet Haematol. 5(9), e411–e421 (2018).
    • 94. Horowitz MM, Gale RP, Sondel PM et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood 75(3), 555–562 (1990).
    • 95. Sweeney C, Vyas P. The graft-versus-leukemia effect in AML. Front. Oncol. 9, 1217 (2019).
    • 96. Weiden PL, Flournoy N, Thomas ED et al. Antileukemic effect of graft-versus-host disease in human recipients of allogeneic-marrow grafts. N. Engl. J. Med. 300(19), 1068–1073 (1979).
    • 97. Pollyea DA, Dinardo CD, Arellano ML et al. Impact of venetoclax and azacitidine in treatment-naive patients with acute myeloid leukemia and IDH1/2 mutations. Clin. Cancer Res. 28(13), 2753–2761 (2022).
    • 98. Chaturvedi A, Gupta C, Gabdoulline R et al. Synergistic activity of IDH1 inhibitor BAY1436032 with azacitidine in IDH1 mutant acute myeloid leukemia. Haematologica 106(2), 565–573 (2021).
    • 99. Chou WC, Lei WC, Ko BS et al. The prognostic impact and stability of Isocitrate dehydrogenase 2 mutation in adult patients with acute myeloid leukemia. Leukemia 25(2), 246–253 (2011).
    • 100. Patel JP, Gonen M, Figueroa ME et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 366(12), 1079–1089 (2012).
    • 101. Balducci L. Transfusion independence in patients with myelodysplastic syndromes: impact on outcomes and quality of life. Cancer 106(10), 2087–2094 (2006).
    • 102. Dinardo CD, Stein EM, De Botton S et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N. Engl. J. Med. 378(25), 2386–2398 (2018).
    • 103. Roboz GJ, Dinardo CD, Stein EM et al. Ivosidenib induces deep durable remissions in patients with newly diagnosed IDH1-mutant acute myeloid leukemia. Blood 135(7), 463–471 (2020).
    • 104. Ma J, Ge Z. Recent advances of targeted therapy in relapsed/refractory acute myeloid leukemia. Bosn. J. Basic Med. Sci. 21(4), 409–421 (2021).
    • 105. Mamolo CM, Cappelleri JC, Hoang CJ et al. A real-world, cross-sectional, community survey of symptoms and health-related quality of life of adults with acute myeloid leukemia. Future Oncol. 15(16), 1895–1909 (2019).
    • 106. Forsythe A, Kwon CS, Bell T, Smith TA, Arondekar B. Health-related quality of life in acute myeloid leukemia patients not eligible for intensive chemotherapy: results of a systematic literature review. Clinicoecon. Outcomes Res. 11, 87–98 (2019).
    • 107. Urbino I, Secreto C, Olivi M et al. Evolving therapeutic approaches for older patients with acute myeloid leukemia in 2021. Cancers 13(20), 5075 (2021).