Abstract
Acute myeloid leukemia (AML) is associated with poor outcomes in older adults. A major goal of treatment is to balance quality of life and functional independence with disease control. With the approval of new, more tolerable regimens, more older adults are able to receive AML-directed therapy. Among these options are hypomethylating agents (HMAs), specifically azacitidine and decitabine. HMAs have become an integral part of AML therapy over the last two decades. These agents are used either as monotherapy or nowadays more commonly in combination with other agents such as the Bcl-2 inhibitor venetoclax. Biological AML characteristics, such as molecular and cytogenetic risk factors, play crucial roles in guiding treatment decisions. In patients with high-risk AML, HMAs are increasingly used rather than intensive chemotherapy, although further trials based on a risk-adapted approach using patient-and disease-related factors are needed. Here, we review trials and evidence for the use of HMA monotherapy and combination therapy in the management of older adults with AML. Furthermore, we discuss the use of HMAs and HMA combination therapies in AML, mechanisms of action, their incorporation into hematopoietic stem cell transplantation strategies, and their use in patients with comorbidities and reduced organ function.
Keywords: Hypomethylating agents, azacitidine, decitabine, guadecitabine, older adults, acute myeloid leukemia
Introduction
The incidence of acute myeloid leukemia (AML) increases with age; the incidence is 17.6 per 100,000 in those aged ≥65 years compared to 1.8/100,000 in those aged <65 years in the US [1]. Despite improvements in recent years, overall survival (OS) in older adults remains poor; median OS (mOS) is 6–11 months in studies up to 2021 [2–5]. Therefore, consideration of patient preferences is essential when choosing treatments, as older patients may prioritize functional independence and quality of life (QoL) over survival duration [6, 7].
Hypomethylating agents (HMAs) such as azacitidine (AZA) and decitabine (DEC) are lower intensity treatments and have been used increasingly for the treatment of AML in the last two decades. With the approval of the Bcl-2 inhibitor venetoclax (VEN), its combination with HMAs has become the standard of care for many older adults with AML, especially those who are unfit for intensive chemotherapy (IC). As a result, outcomes in older adults have likely improved, although the disease is still largely incurable. For the purpose of this review, “older” is defined as ≥60 years, although it is important to acknowledge that physiologic age is more important than chronological age.
In this narrative review, we provide an overview of HMAs and discuss their use alone and in combination therapies in AML, their mechanisms of action, and their incorporation in hematopoietic stem cell transplantation (HSCT) strategies. We also review the use of HMAs in patients with impaired kidney function.
Mechanism of action of HMAs
HMAs are pan-DNA methyltransferase (DNMT) inhibitors. AZA is a ribonucleoside and DEC, a deoxyribonucleoside [8]. HMAs interfere with cancer cell survival by reactivating transcriptionally silenced tumor suppressor genes, reversing the acquired DNA methylation of cancer cells, and targeting specific gain-of-function mutations in epigenetic enzymes [9]. In addition to the demethylating effects, incorporation of AZA, but not DEC, in RNA blocks transfer RNA methylation [10] and decreases protein synthesis [11] which promotes apoptosis (Figure 1). Furthermore, HMAs reactivate endogenous retroviral elements, thereby inducing an anti-tumor immune response via interferons [12]. HMAs also increase the expression of tumor-associated antigens which potentially triggers anti-tumoral T-cell-mediated immune responses [13]. In a small series of patients with myelodysplastic neoplasia (MDN), HMA treatment is associated with demethylation and up-regulation of the oncogene SALL4 which is associated with an inferior survival [14], although the clinical implication of this finding requires further study.
Figure 1: Mechanisms of action of hypomethylating agents.
Hypomethylating agents are unstable in plasma after absorption due to spontaneous hydrolysis and deamination by cytidine deaminase (CDA) [115]. The cellular uptake is mediated by different nucleoside transporters (hCNT for AZA and hENT for DEC). The enzymes catalyzing the first limiting phosphorylation step are uridine-cytidine kinase (UCK) for AZA and deoxycytidine kinase (DCK) for DEC, respectively. Following their cellular uptake, which is dependent on nucleoside transporters, they are three times successively phosphorylated by intracellular kinases. The active tri-phosphorylated metabolite of 5-aza-dCTP (DEC) is directly incorporated into DNA during cell cycle. For AZA, only 10–20% of AZA follows the DNA incorporation during replication; the majority of 5-aza-CTP is incorporated in RNA. DAC incorporated into DNA binds DNMT1 and leads to its degradation, promoting a progressive DNA hypomethylation after several rounds of replication [17]. This has been postulated to lead to an activation of repressed tumor suppressor genes [116], inducing senescence and apoptosis. The figure was created with BioRender.com.
Abbreviations: AZA, azacitidine; DEC, decitabine; hCNT human concentrative nucleoside transporter; hENT, human equilibrative nucleoside transporter; NDPK, nucleoside diphosphate kinase; NMPK, nucleoside monophosphate kinase; RNR, ribonucleotide reductase; SASP, Senescence-associated secretory phenotype.
AZA and DEC are administered intravenously (AZA and DEC) or via subcutaneous (AZA) injection. AZA is also available orally (CC-486), but oral AZA is not bioequivalent to the injectable form [15] and therefore should not be substituted for injectable versions. When taken continuously, oral AZA prolongs drug exposure and correlates with the extent of hypomethylation [15]. Oral AZA is approved for use in the maintenance setting for AML. DEC is available as a fixed oral combination with cedazuridine, a cytidine deaminase inhibitor that increases the oral bioavailability of DEC. DEC/cedazuridine is approved in the US for use in MDN and chronic myelomonocytic leukemia (CMML), but not in AML [16]. A newer HMA, guadecitabine, a prodrug of DEC, is resistant to deamination by cytidine deaminases, resulting in a prolonged half-life of the active metabolite [17]. As guadecitabine failed its primary endpoint in the ASTRAL study [18], its use is not further explored in clinical trials. Although AZA and DEC are generally seen as equivalent, no randomized trials have compared AZA with DEC. Two large meta-analyses included published clinical trials that had tested either AZA and DEC [19, 20]. One found no difference in all outcomes [20], and the second suggested that mortality may be lower with AZA compared to DEC, although with low certainty [19]. There were no differences in treatment-related toxicities [19]. These results were supported by the ASTRAL-1 study that compared guadecitabine with AZA/DEC/low-dose cytarabine (LDAC) as first-line treatment for older adults with AML. In a separate post-hoc analysis of the standard-of-care arm, no differences between AZA and DEC in mOS or in common treatment-related toxicities were observed [21].
Pharmacokinetics of HMAs under specific considerations: Impaired renal function
Chronic kidney disease (CKD) is common in older adults [22, 23]. AZA and DEC are metabolized via hydrolysis and deamination by cytidine deaminase, which is primarily located in the liver. Metabolism is independent of cytochrome p450 enzymes. AZA and DEC are primarily eliminated in the urine [8]. Because patients with a glomerular filtration rate (GFR) of <40–60ml/min were excluded from many trials, data on HMA use in this population are scarce. A pharmacokinetic study in patients with GFR<30 ml/min demonstrated higher mean concentrations after AZA without drug accumulation [24] that would justify dose adjustment. In a case series of 28 patients with MDN and a GFR of 30–60 ml/min, no increased toxicity was demonstrated compared to individuals with normal kidney function [25]. Another single center study reported similar findings, but patients with GFR<30 ml/min did experience higher rates of toxicities [26]. An additional single center observational study using DEC [27] included 48 patients with GFR<60 ml/min and 63 patients with GRF>60 ml/min; those patients with GFR <60 ml/min demonstrated an increased incidence of grade ≥3 respiratory toxicities measured using the NCI Common Terminology Criteria for Adverse Events (CTCAE) (40% vs. 14%; p=0.0037). All patients who developed grade ≥3 heart failure, myocardial infarction, and new-onset of atrial fibrillation had impaired kidney function [27]. Patients with impaired kidney function in this study were older (73 vs. 68 years; p<0.001), and the presence of CKD in itself is a risk factor for cardiac events. Recently, two case reports suggested that AZA use in patients undergoing hemodialysis is safe [28, 29]. Because AZA is removed during hemodialysis [29], it should be administered after treatment. In summary, severe renal impairment does not preclude HMA therapy. However, patients with a GFR<30 ml/min require close monitoring for side effects [8].
HMA monotherapy
Several clinical trials have investigated HMA monotherapy in the frontline setting of older adults with AML. Pivotal studies are summarized below and in Table 1:
Table 1:
Summary of data on HMAs as monotherapy in AML
| Study | Design and setting | Trial arms | N and age (years) | Key findings |
|---|---|---|---|---|
| Phase III trials | ||||
| AZA-AML-001 (NCT01074047) [30] | Phase III Newly diagnosed AML>30% blasts | AZA 75 mg/m2 d1–8, q4 wks vs. CCRs (18.2% BSC, 64% LDAC, and 17.8% IC) | N=488; median age: 75 y (range: 64–91 y) | mOS: AZA 10.4 m (95% CI, 8.0–12.7 m) vs. CCRs 6.5 m (95% CI, 5.0–8.6 m), p=0.1009 |
| DAC0–016 [31] | Phase III Newly diagnosed AML | DEC 20 mg/m2 d1–5, q4 wks vs. TC (BSC/LDAC) | N=485; median age: 73 y (range: 64–91 y) | mOS: DEC 7.7 m (95% CI, 6.2–9.2 m) vs. TC 5.0 m (95% CI, 4.3–6.3 m), p=0.108 CR/CRp: DEC 17.8% vs. TC 7.8% |
| ASTRAL-1 (NCT02920008) [18] | Phase III Newly diagnosed AML | GUA 60 mg/m2, d1–5, q4 wks vs. TC (AZA, DEC, LDAC) | N=815; median age: 76 y (range: 56–94 y); PA75: ~62% | mOS: GUA 7.1 m vs. TC 8.47 m, p=0.73 mOS (if received ≥4 cycles): GUA 15.6 m vs. TC 13.6 m, p=0.02 mOS (if received ≥6 cycles): GUA 19.5 m vs. TC 14.0 m, p=0.002 |
| FLUGAZA (NCT02319135) [32] | Phase III Newly diagnosed AML | FLUGA q4 wks (fludarabine 25 mg/m2, d2–6, cytarabine 75 mg/m2, d2–6, and filgrastim 5μg/kg, d1–3) vs. AZA 75 mg/m2, d1–7, q4 wks | N= 283; median age: 75 y (range: 65–90 y) | mOS: FLUGA 4.1 m (95% CI, 2.7–5.5 m) vs. AZA 9.8 m (95% CI, 5.6–14 m), p=0.005) CR after 3 cycles: FLUGA 18% vs. AZA 9%, p=0.04 CR/CRi after 9 m: FLUGA 33% vs. AZA 29%, p=0.41 |
| Phase II trials | ||||
| NCT01786343 [118] | Phase II Newly diagnosed AML | DEC-5 q4 wks (DEC 20 mg/m2, d1–5,) vs. DEC-10 q4 wks (DEC 20 mg/m2, d1–10) | N=78; median age: 77 y | 1y-OS: 25% in both arms; no differences in CR, CRi, OS |
| Retrospective data | ||||
| Austrian Azacitidine Registry (NCT01595295) [37] | Newly diagnosed AML (46%) | AZA monotherapy (various regimens) | N=302; median age: 73 y (range: 30–93 y); PA60: n.f.s.; PA75: 43%. | mOS: 9.6 m (95 % CI, 8.53–10.7m) |
| [4] | Pooled European registry data Newly diagnosed AML ≥70 y | HMAs (various monotherapy regimens) vs. IC (various regimens) | N=3,700 (HMAs: N=1,073; IC: N=1,199); median age: 75 y (range: 72.5–79 y) | mOS: IC 10.9 m (95% CI, 9.7–11.6 m) vs. HMA 9.2 m (95% CI, 8.3–10.2 m) CR/CRi: IC 56.1% vs. HMA 19.7%, p <0.0001 |
| [119] | Pooled European registry data | AZA monotherapy | N=710; median age: 75 y (range: 60–93 y) | mOS: 9.0 m (95% CI, 8.8–11 m) |
| [120] | Retrospective data (three centers) Newly diagnosed AML and MDS | AZA, standard regimen in 69%, 5-day regimen in 31% | N=115; N (AML)=63; N (MDS)=53; median age: 82 y (range: 80–92 y) | mOS (AML): 9.7 m |
| [121] | Retrospective data (Apulian Hematological Network) Newly diagnosed AML and R/R AML | DEC 20 mg/m2, d1–5, q4 wks | N=199; newly diagnosed AML: N=94; median age: 75.4 y (range: 61–91 y) | mOS: 8.7 m (95% CI, 7.4–10.3 m) |
| [122] | Single center retrospective data (Moffitt Cancer Center). Newly diagnosed AML | HMAs vs. IC (cytarabine/anthracycli ne or equivalent) | N=980 (HMA: N=255, IC: N=360); mean age (HMA): 76.5 y (range: 70.1–95.2 y), median age (IC): 73.9 y (range: 70.5–90.4 y) | mOS: HMA 14.4 m vs. IC 10.8 m (HR, 1.35; 95% CI, 1.10–1.65, p=0.004) |
Abbreviations: AML, acute myeloid leukemia; AZA, azacitidine; BSC, best supportive care; CCRs, conventional care regimens; CI, confidence interval; CMML, chronic myelomonocytic leukemia; CR, complete response; CRi, complete response with incomplete count recovery; DED, decitabine; GUA, guadecitabine; HMA, hypomethylating agent; HR, hazard ratio; IC, intensive chemotherapy; LDAC, low-dose cytarabine; MDS, myelodysplastic syndrome; m, months; n.f.s., not further specified; ORR, overall response rate; OS, overall survival; mOS, median overall survival; PA60, percentage above 60 years; PA75, percentage above 75 years; R/R, relapsed or refractory; TC, treatment of choice; y, years
AZA-AML-001
The AZA-AML-001 trial demonstrated the efficacy of AZA in comparison to conventional care regimens (CCR) in adults aged ≥65 years with AML and led to the approval of AZA as monotherapy for adults with AML deemed unfit for intensive chemotherapy (IC) approaches [30]. In this randomized phase III trial, patients were preselected for a defined treatment intensity [(IC, LDAC, or best supportive care (BSC)] before they were randomized to AZA versus CCR. Median OS was in favor of patients who received AZA, although this difference was not statistically significant (Table 1). In a post-hoc multivariate analysis, AZA reduced the risk of death by 31% [hazard ratio (HR), 0.69; 95% confidence interval (CI), 0.54–0.88; p=0.0027)] in comparison to CCR. Interestingly, patients who were preselected for BSC and randomized to receive AZA demonstrated a mOS of 5.8 vs. 3.7 months (p=0.0288) in comparison to those who received BSC.
DACO-016 trial
Based on the DACO-016 trial that demonstrated the efficacy of DEC as monotherapy in comparison to BSC/LDAC in adults aged ≥65 years with AML, DEC was approved for treatment of AML in this population. Similar to the AZA-AML-001 trial, DEC given as a five-day regimen was compared with BSC/LDAC regimens in the randomized phase III DACO-016 trial [31]. Median OS was similar in both groups (DEC: 7.7 months; 95% CI, 6.2–9.2 months; BSC/LDAC: 5.0 months; 95% CI, 4.3–6.3 months) with significantly higher response rates in patients randomized to DEC (17.8% vs. 7.8%; p=0.001) [31].
FLUGAZA Trial
The FLUGAZA trial supported the efficacy of AZA as monotherapy in comparison to an induction chemotherapy with moderate intensity in adults ≥65 years with AML. In this randomized phase III trial, AZA monotherapy was compared with FLUGA, a moderately intensive induction therapy regimen consisting of cytarabine (75mg/m2 subcutaneously/intravenously, day 2–6), fludarabine (25 mg/m2 intravenously/40 mg/m2 orally, days 2–6), and G-CSF priming (filgrastim 5μg/kg, days 1–3). Treatment was continued until patients achieved measurable residual disease (MRD) negativity. Although early remission rates were significantly higher in the FLUGA arm with similar early mortality rates, mOS was superior in the AZA arm (9.8 vs. 4.1 months, p=0.005)[32].
Registry-based data
Several registry analyses support the findings from randomized trials demonstrating efficacy of HMAs as monotherapies in real-world settings. An analysis of the Austrian Azacitidine Registry compared patients treated in the real-world setting (n=95) who met the inclusion criteria of the AZA-AML-001 trial and demonstrated similar OS (Table 1) [33]. Pooled European registry data including >3000 patients aged ≥70 years treated with IC or HMAs from 2007 to 2018 [4] demonstrated no significant difference in mOS between the treatment groups, but patients who received IC had higher rates of response (Table 1). A time-dependent effect of the treatments on OS was demonstrated: HMAs were associated with a lower risk for death within the first 1.5 months of treatment, whereas IC was associated with a significantly improved OS after four months of treatment [4].
Benefits of HMA beyond remission
A meta-analysis of 26 trials (20 of which involved HMAs) [34] that included patients with newly diagnosed AML demonstrated a significant correlation between complete remission (CR)/CR with incomplete count recovery (CRi) and mOS. However, failure to achieve a CR does not mean that patients fail to benefit from treatment. In the AZA-AML-001 study, patients in the AZA group had an increased OS compared with CCR even among those patients who failed to achieve a CR (6.9 vs. 4.2 months, respectively; HR, 0.77; 95% CI, 0.62–0.95; p=0.017) [30]. In the DACO-016 study, OS improved in patients who achieved transfusion independence, even among those who did not achieve CR [35]. In a single center study of 56 patients aged 59–91 years with newly diagnosed AML treated with an HMA, stable disease for ≥6 cycles led to an OS benefit (18 vs. 4 months; p=0.0002) [36]. In an analysis of 302 patients from the Austrian Azacitidine Registry, disease stabilization and/or a hematological response resulted in a clinically relevant OS benefit (mOS among those with stable disease: 8.1 months; mOS among those with hematological response: 9.7 months; and mOS among those who achieved both: 18.9 months; mOS (without stable disease/hematological response): 3.2 months] [37]. Other possible benefits included reduction in transfusion frequency and improved QoL [30, 35, 38].
HMA combination therapies: Approved options
Given the benefits of HMA monotherapies, several HMA combination therapies have been investigated and approved. Some of these combinations specifically target potential mechanisms of treatment resistance [39]. In this section, we summarize the key studies.
HMA and venetoclax
Preclinical data demonstrated that HMAs plus VEN provides synergies in inducing apoptosis through several mechanisms (e.g., AZA upregulates NOXA, a key player within the mitochondrial apoptosis pathway, thereby increasing the sensitivity of leukemic blasts to the BCL-2 inhibition by VEN [40].) Based on the results of the VIALE-A study that compared AZA/VEN with AZA monotherapy in adults ≥75 years or with comorbidities precluding IC, AZA/VEN was approved for this patient population and has become the standard of care for many older adults with AML.
After the successful Phase Ib study of HMA/VEN [41], the subsequent phase III VIALE-A study on AZA/VEN versus AZA alone included patients ineligible for IC based on their age (≥75 years) and/or coexisting conditions. Approximately 60% of patients were ≥75 years. The study demonstrated improved mOS in the combined arm (14.7 vs. 9.6 months; HR, 0.66; p<0.0001), and serious AE rates were similar (83% in the AZA/VEN vs. 73% in the AZA arms) [42]. In a subgroup analysis by age, the OS benefit extended to patients ≥75 years (HR, 0.54; 95% CI, 0.39–0.73). In addition, more patients achieved transfusion independence for PRBCs and platelets in the AZA/VEN arm. The median time to first response was 1.0 month (range: 0.6–14.3) in the AZA/VEN arm compared to 2.6 months in the AZA alone arm (range: 0.8–13.2). Although the percentage of patients who discontinued treatment due to AEs and early mortality was similar in both groups, several AEs were more frequent in the AZA/VEN arm [e.g., neutropenia (AZA/VEN: 19%; AZA: 10%), febrile neutropenia (AZA/VEN: 20%; AZA: 4%)] [42].
Response rates for HMA/VEN in patients with NPM1- and IDH1/2-mutated AML were >65% [42–44], which corresponded to reasonable median durations of response (IDH1/2: 29.5 months, NPM1: not reached; Table 3) [45]. These results will likely encourage the comparison of HMA/VEN and IC in fit older adults with NPM1- and IDH1/2-mutated AML. The high CR and OS rates in patients with IDH1/2-mutated AML are due to the strong dependence of these subtypes on BCL-2 due to the oncometabolite (R)-2-hydroxyglutarate. This oncometabolite inhibits the activity of cytochrome c oxidase (COX) in the mitochondrial electron transport chain. Reduced COX activity lowers the mitochondrial threshold to trigger apoptosis during BCL-2 inhibition [46]. In contrast, the ORR in patients with TP53 mutations was ~53–55% [45, 47] with a median duration of response of 6.5 months [45]. Although these response rates were superior to those reported in many different trials in patients treated with IC, the OS was not different from that achieved with AZA alone [48], and the OS in patients with TP53 mutations remained inferior compared to other molecular subtypes (Table 3), as TP53 mutations contribute to primary and acquired AZA/VEN resistance [49].
Table 3.
Composite remission rates according to molecular aberrations
| Study | Therapy regimen | NPM1 mut | IDH1/2 mut | FLT3-ITD | TP53mut | Cytogenetics | Comment |
|---|---|---|---|---|---|---|---|
| NCT02993523 [42] | AZA/VEN vs. AZA | HMA/VEN 66.7% (95% CI, 46.0–83.5) vs. HMA 23.5% (95% CI, 6.8–49.9), p=0.012 | HMA/VEN 75.4% (95% CI, 62.7–85.5) vs. HMA 10.7% (95% CI, 2.3–28.2), p<0.001 | HMA/VEN 72.4% (95% CI, 52.8–87.3) vs. HMA 36.4% (95% CI, 17.2–59.3), p=0.02 | HMA/VEN 55.3% (95% CI, 38.3–71.4) vs. HMA 0%, p<0.001 | Only small subgroups | |
| NCT02993523+ NCT02203773 [138] | AZA/V EN vs. AZA | HMA/V EN 67% vs. HMA 36% | Pooled data, post-hoc analysis | ||||
| NCT02993523+ NCT02203773 [43] | AZA/V EN vs. AZA | HMA/V EN 79% vs. HMA 11% | Pooled data, post-hoc analysis | ||||
| NCT03404193 [139] | DEC-10/VEN | Intermedi ate CGR 89% vs. adverse CGR 66% | Treatment-naïve patients | ||||
| NCT03404193 [140] | DEC10-VEN | Mut 66% vs. wt 89%, p=0.002 | Post-hoc analysis, small numbers in subgroups | ||||
| Retrospective single-center data [44] | HMA/V EN 96%, HMA 36%, IC 89%; age>65 y: HMA + VEN 88%, HMA 28%, IC 56%; HMA/VEN vs. HMA, p<0.001; HMA/V EN vs. IC, p=0.01 | Retrospective single-center data, small numbers |
Abbreviations: AZA, azacitidine; CI, confidence interval; CGR, cytogenetic risk (acc. To ELN classification); DEC, decitabine; DEC-10, decitabine regimen for 10 days; HMA, hypomethylating agent; IC, intensive chemotherapy; mut, mutated; VEN, venetoclax; wt, wild-type.
Most clinical trials on HMA/VEN have excluded patients with core binding factor (CBF-) leukemia [30, 31, 41, 42] based on its sensitivity to high-dose cytarabine. Although CBF leukemia occurs less frequently in older than in younger patients with AML, older patients are often less likely to be able to receive IC with high-dose cytarabine. One multicenter retrospective case series demonstrated a response rate of 80% in patients with newly diagnosed or relapsed/refractory (R/R) CBF leukemia who received HMA/VEN. Of those, 70% were >65 years old [50]. Further data on the efficacy of HMA/VEN-based treatments in CBF leukemia are required before its standard use can be recommended.
HMA and ivosidenib, an isocitrate dehydrogenase (IDH) 1 inhibitor
The IDH1 inhibitor ivosidenib (IVO) is approved by the US Food and Drug Administration (FDA) for R/R AML with IDH1 mutation. Although IVO has shown an ORR of ~40% as monotherapy in R/R AML, the duration of response was limited [51, 52]. IDH1/2 inhibition induces a differentiation of leukemic blasts rather than eradication [53]. This fact underlines the need for combination therapy that synergizes with IDH inhibitors to promote apoptosis. Based on the results of the AGILE trial [54] that compared AZA/IVO with AZA monotherapy, the FDA recently approved this combination for adults with newly diagnosed, IDH1-mutated AML who are unfit for IC.
The phase III AGILE trial compared AZA/IVO with AZA/placebo and included patients with newly diagnosed, IDH1-mutated AML ineligible for IC due to age ≥75 years and/or a coexisting condition (severe cardiac or pulmonary disorder, renal impairment, or elevated bilirubin). The median age was ~76 years. The study demonstrated a mOS benefit for AZA/IVO of 24.0 vs. 7.9 months (HR for death, 0.44; 95% CI, 0.27–0.73; p=0.001) [54]. Median time to CR was not significantly different between arms. Transfusion independence was higher with AZA/IVO (46% vs. 18%; p=0.006) [54].
In summary, assessment of patient-related (through geriatric assessment) and disease-related factors (Table 3) as well as patient preferences is essential when choosing treatments for older adults with newly diagnosed AML. Information on GA domains and tools for their evaluation have been previously described [55, 56]. A proposed approach is shown in Figure 2. In addition to disease responses, important patient-centered outcomes such as time spent at home and quality of life should be considered. For example, time spent at home following HMA treatments is longer than that following IC (205 vs. 187 days), with similar times for AZA/VEN and AZA monotherapy [57, 58].
Figure 2: How to guide therapy-decisions in older adults with AML.
Future therapy-decision making in older adults with AML will most likely be guided by an integrative approach of GA- und biology-driven factors. This figure depicts possible pathways in decision making that are not based on solid evidence due to lack of data but rather biological rationales and practise-guided concepts. GA impairments predictive of worse outcomes with IC are short physical performance battery (SPPB) sum score <9 and/or mini mental state examination (MMSE) <24 [117]. Among comorbidities that preclude from intensive chemotherapy are symptomatic heart failure, advanced chronic kidney failure, and/or other life-threatening comorbidities. For evaluation of patient priorities, possible factors for discussion are the time spend at home, chance of cure or long-term control versus palliative options, treatment-related side-effects, transfusion requirements, and quality of life.
Abbreviations: IC, intensive chemotherapy, comprises “7+3”-like therapies +/− Gemtuzumab ozogamizin and Midostaurin.
In the authors’ opinion, if patients are deemed unfit to receive IC, HMA/VEN should be the first-line treatment. For patients with IDH1-mutated AML, HMA/IVO can also be considered. The median time-to-response (CR/CRi) was 1.3 months (range, 0.6 to 9.9) for AZA/VEN [59] and 4.0 months (range, 1.7–8.6) for AZA/IVO [54]. AZA/VEN may be considered for patients with highly proliferative disease and for those in urgent need of achievement of response. Among patients who are frail but who wish to pursue treatment, monotherapy with HMAs or IDH inhibitors (the latter for among those with IDH-1 mutated AML) can be considered.
HMA combination therapies: Experimental combinations
Several HMA combinations are currently being explored in clinical trials. We summarize synergistic mechanisms and provide an overview of the preliminary results below and in Table 2. Studies generally included heterogenous groups of patients of all ages with newly diagnosed AML and relapsed/refractory (R/R) AML. When available, patient ages are included in Table 2.
Table 2:
Summary of data from combination trials of treatment for AML
| Study | Design and setting | Trial arms | N and age (years) | Key findings |
|---|---|---|---|---|
| HMA plus venetoclax | ||||
| NCT02203773 [41] | Phase Ib Newly diagnosed AML | VEN 400, 800, or 1200 mg/d in combination with either DEC 20 mg/m2, d1–5, or AZA 75 mg/m2, d1–7 | N=145; median age: 74 y (range: 65–86 y); PA75: 36% | CR/CRi: 67% Patients ≥75y: CR/CRi: 65%. CR/CRi median duration: 11.3 m mOS: 17.5 m Median duration of response, EFS, and OS (AZA/VEN; CR/CRi with MRD− vs MRD+): not reached [96] |
| VIALE-A NCT02993523 [42] | Phase III Newly diagnosed AML | AZA 75 mg/m2 subQ or IV, d1-7, q4 wks/VEN (400mg PO, d1–28) vs. AZA 75 mg/m2, d1–7/placebo, q4 wks | N=431; median age: 76 y (range: 49–91 y); PA60: n.f.s.; PA75: 61% | mOS: AZA/VEN 14.7m vs. AZA/placebo 9.6 m (HR for death, 0.66, p<0.001) Composite CR: AZA/VEN 66.4% vs. AZA/placebo 28.3%, p<0.001 |
| NCT03404193 [123] | Phase II, single-center Newly diagnosed and R/R AML | Induction: DEC 20 mg/m2 d1-10/VEN 400mg PO, d1–28; consolidation: DEC 20 mg/m2, d1–5/VEN 400 mg PO, d1–28 | N=70 (newly diagnosed AML); median age: 72 y (range: 70–78 y) | mOS: 18.1 m (95% CI, 10.0 m–not reached) ORR: 89% |
| HMA plus sorafenib | ||||
| NCT02196857 and NCT01254890 [124] | Phase I/II Newly diagnosed AML | AZA 75 mg/m2, d1–7 sorafenib 400 mg BID, d1–28; q4 wks | N=27; median age: 74 y (range: 61–86 y) | mOS: 8.3 m CR: 26% |
| HMA plus FLT3 inhibitor | ||||
| NCT01892371 [63] | Phase I/II Newly diagnosed and R/R AML or high-risk MDS | Arm 1: AZA 75 mg/m2, d1-7/quizartinib 600 mg/d PO Arm 2: LDAC 20 mg BID, d1-10/quizartinib 600mg PO, d1-28, q4 wks | N=74; median age: 72 y (range: 58–82 y); median age (frontline AZA/quizartinib) : 75 y (range: 64–82 y); PA60: n.f.s.; PA75: n.f.s. | Frontline setting* mOS: AZA/quizartinib 12.8 m vs. LDAC/quizartinib 4 m ORR: AZA/quizartinib 87% vs. LDAC/quizartinib 74% |
| LACEWING [62] | Phase III Newly diagnosed AML, FLT3+ | AZA 75 mg/m2, d1–7/GIL 120 mg/d PO, d1–28, q4 wks | N=74 (AZA/GIL), N=49 (AZA); median age: 78 y (AZA/GIL), 75 y (AZA); range: 59–75 | mOS: AZA/GIL 9.82 m vs. AZA 8.87 m (HR 0.916, p=0.753) CR: AZA/GIL 16.2 vs. AZA 14.3 %, p=0.762 |
| HMA plus IDH1–2 inhibitors | ||||
| NCT02677922 [66] | Phase Ib/II Newly diagnosed AML | AZA 75 mg/m2, d1–7/ENA 100 mg/d, d1–28, q4 wks | Phase Ib: N=6; median age: 68 y (range: n.f.s.) Phase II: N=101; median age: 75 y (range: n.f.s.); PA60: n.f.s. | mOS: AZA/ENA 22 m vs. AZA alone 22.3 m (HR, 0.99, p=0.97) CR: AZA/ENA 54% vs. AZA only 12%, p<0.0001 |
| NCT02677922 [67] | Phase Ib Newly diagnosed AML Phase III Newly diagnosed IDH1-mutated AML | AZA 75 mg/m2, d1–7/IVO 500 mg/d, d1–28, q4 wks | N=23; median age: 76 y (range: 61–88 y); PA75: 52.2% | CR: 60.9% CRi/CRp: 8.7% |
| AGILE (NCT03173248) [54] | AZA 75 mg/m2, d1–7/IVO 500 mg/d, d1–28, q4 wks vs. AZA 75 mg/m2, d1-7/placebo, q4 wks | N=146; median age (AZA/IVO): 76.0 y (range: 58–84 y); median age (AZA/placebo): 75.5 y (range: 45–94 y); PA60: n.f.s.; PA75: n.f.s. | mOS: AZA/IVO 24.0 m vs. AZA/placebo 7.9 m (HR for death, 0.44; 95% CI, 0.27–0.73, p=0.001) CR: AZA/IVO 47% (95% CI, 35–59) vs. AZA/placebo 15% (95% CI, 8–25, p<0.001) | |
| HMA plus pevonedistat | ||||
| NCT01814826 [79] | Phase Ib Treatment-unrelated AML | AZA 75 mg/m2, d1–5, d8, d9/pevonedistat 20 or 30 mg/m2, d1, d3, d5, q4 wks | N=64; median age: 75 y (range: 61–89 y); PA75: n.f.s. | mOS: 7 m CR: 31.3% |
| NCT02610777 [80] | Phase II LB-AML and higher risk MDS/CMML | AZA 75 mg/m2, d1-7/pevonedistat 20 mg/m2, d1, d3, d5, q4 wks vs. AZA 75 mg/m2, d1–7, q4 wks alone | N=120; median age: 72 y (range: n.f.s.); PA75: 35% | mOS: AZA/pevonedist at 21.8 m vs. AZA alone 19 m** Median EFS: AZA/pevonedist at 21 m vs. AZA alone 16.6 m |
| HMA plus checkpoint inhibitor | ||||
| NCT02397720 [125] | Phase II R/R AML | AZA 75mg/m2, d1–7, Nivolumab 3mg/kg, d1, d14, q4–6 wks | N=77; median age: 70 y (range: 22–90 y); PA60 (AZA/Nivoluma b): 80% | mOS: 6.3 m CR/CRi: 22% |
| NCT02996474 [126] | Feasibility trial R/R AML | DEC 20mg/m2, d8–12, d15–19/pembrolizum ab 200 mg/m2, d1 q3 wks | N=10; median age: 62 y (range: 30–81 y); PB60: n.f.s. | stable disease (or better): 6/10 patients |
| NCT03390296 [127] | Phase Ib/II R/R AML | AZA 75 mg/m2, d1–7/avelumab 3 mg/kg to the first 7 patients enrolled and 10 mg/kg for the remaining 12 patients | N=19; median age: 66 y (range: 22–83 y); PA60: 74% | mOS: 4.8 m ORR: 10.5% |
| HMA plus magrolimab | ||||
| NCT03248479 Abstract only [73] | Phase Ib Newly diagnosed and R/R high-risk MDS and unfit AML | AZA 75mg/m2, d1–7/Magrolimab 1–30 mg/kg dose escalation, d1, d4, d8, d15; q4 wks | N (total cohort)=68; N (AML)=29; median age: 72 y (range: n.f.s.); PA60: n.f.s. | CR AML patients only: 40% CRi AML patients only: 4% Transfusion independence AML patients only: 64% 6m OS AML patients only: 91% |
| HMA plus ibrutinib | ||||
| NL5751 (NTR6017) [128] | Phase II Unfit AML or high-risk MDS | DEC 20 mg/m2, d1–5/ibrutinib 560 mg PO d1–28 vs. decitabine 20 mg/m2, d1–5 alone | N=144; median age: 75 y (range: 66–89 y); PA75: n.f.s. | CR/CRi: DEC/ibrutinib 40% vs. DEC alone 31% mOS: DEC/ibrutinib 11.4 m vs. DEC alone 11 m |
Abbreviations: AML, acute myeloid leukemia; AZA, azacitidine; BSC, best supportive care; CMML, chronic myelomonocytic leukemia; CR, complete response;; CRi, complete response with incomplete count recovery; CRp, complete response with incomplete platelet recovery; d, days; DED, decitabine; EFS, event free survival; ENA, enasidenib; GIL, gilteritinib; HMA, hypomethylating agent; HR, hazard ratio; IVO, Ivosidinib; LB-AML, low-blast AML; LDAC, low-dose cytarabine; MDS, myelodysplastic syndrome; mOS, median overall survival; m, months; MRD, measurable residual disease; n.f.s., not further specified; ORR, overall response rate; OS, overall survival; PA60, percentage above 60 years; PA75, percentage above 75 years; R/R, relapsed or refractory; VEN, venetoclax; wks, weeks; y, years
Results include patients with AML and MDS
Results include patients with AML, MDS, and CMML
HMA and fms-like tyrosine kinase 3 (FLT3) inhibitors
Synergistic anti-leukemia activity in FLT3-mutated AML was demonstrated in preclinical models for HMA/FLT3-inhibitor combinations compared with either agent alone [9, 60]. These combinations were subsequently studied clinically in FLT3-mutated AML using gilteritinib [61, 62] and in both FLT3-mutated and FLT3-WT AML using quizartinib [63]. Overall response rates (ORR) varied from 67% in the AZA/gilteritinib study to 87% in the AZA/quizartinib study (Table 2) [62–64]. Recently, the combination of DEC as a 10-day regimen plus VEN plus a FLT3-inhibitor of choice (sorafenib, gilteritinib, or midostaurin) showed a promising ORR of 92% with MRD negativity in 56% of newly diagnosed AML patients aged ≥ 60 years (N=12) [65]. The randomized phase III trial (LACEWING) that compared AZA monotherapy with AZA and gilteritinib in patients ineligible for IC was prematurely closed, as interim analysis failed the boundary for futility and the OS did not differ between arms, although the composite CR rate of the combination was superior [61]. Whether the combination of HMA with a FLT3 inhibitor provides added benefits needs to be further evaluated, possibly in the context of triplet therapies with VEN.
HMA and enasidenib, an IDH2 inhibitor
IDH2 inhibition has the same mechanistic rationale as IDH1 inhibition. The IDH2 inhibitor enasidenib is approved by the FDA for R/R AML with IDH2 mutation with an ORR of ~40% [51]. Early phase studies combining HMA and enasidenib demonstrated promising CR rates of 54% in older adults with newly diagnosed AML [66, 67]. Further randomized trials of this combination are pending.
HMA and checkpoint inhibitors
In the last decade, immune checkpoint inhibitors have revolutionized the treatment of several solid cancers. The rationale for their use in AML was the expression of PD-L1 on leukemic blasts, especially in TP53-mutated disease [68]. Early single-arm trials with the combination of HMAs and checkpoint inhibitors in R/R AML patients of all ages provided some encouraging evidence. The results are summarized in Tables 2 and 3. However, the Intergroup LEAP (Less Intense AML Therapy Platform) trial (S1612, NCT03092674), a randomized phase II/III study comparing AZA plus nivolumab with AZA as monotherapy in adults ≥ 60 years with newly diagnosed AML or high-risk MDN deemed unfit for IC [69] was prematurely closed due to an increased rate of therapy-related mortality in the combination arm [70].
HMA and magrolimab
Magrolimab is an anti-CD47 antibody that disrupts the major macrophage immune checkpoint (“do not eat me signal”), thereby enabling a robust anti-leukemic immune response [71]. In preclinical studies, upregulation of CD47 by AZA and synergistic activity of AZA plus magrolimab was demonstrated [72]. A phase Ib trial of AZA/magrolimab demonstrated that the treatment was well tolerated in unfit patients with AML or high-risk MDN who had not received treatment [73]. Remarkably, among participants with TP53-mutated AML, mOS was 12.9 months with 10/21 patients (48%) achieving a CR and 4/21 (19%) a CRi [74]. Thus, the combination of AZA/magrolimab could potentially fill the gap for patients with TP53-mutated AML unfit for IC, although additional studies are needed. The ENHANCE-2 trial, a phase III trial comparing AZA/VEN/magrolimab with AZA/VEN/placebo, is currently ongoing for patients with AML unfit for IC.
Other HMA combinations
Other HMA combinations are currently being tested in clinical trials. Among these are the histone deacetylase inhibitors romidepsin [75] and vorinostat [76], the exportin-1 inhibitor selinexor [77], the CD33-antibody-drug conjugate vadastuximab talirine [78], the inhibitor of neural precursor cell expressed, developmentally downregulated 8 (NEDD8) activating enzyme (NAE) pevonedistat [79, 80], the CD33 antibody-drug conjugate gemtuzumab ozogamicin [81], the proteasome inhibitor bortezomib [82], and the TP53 reactivator eprenetapopt (APR-246) [83].
Future directions in the upfront setting
Many questions remain about the use of HMA-based therapies in the upfront setting. First, should AZA/VEN or AZA/IVO be used in patients with newly diagnosed IDH1-mutated AML. Second, because HMA/VEN is associated with prolonged neutropenia, studies to guide its dose adjustment are needed. AZA/VEN is given continuously. Whether it can be stopped or interrupted after achieving MRD negativity is unclear. Drug interruption could lead to better QoL. Third, sequential therapies are attractive areas of investigation to improve treatment tolerability (e.g., switch from AZA/VEN to AZA/IVO after achieving a CR to potentially avoid prolonged neutropenia; switch from HMA monotherapy to HMA/VEN due to concern over the use of combination therapies in the upfront setting; switch from HMA/VEN to HMA monotherapy due to adverse events). Finally, triplet therapies may have the potential to overcome therapy resistance by targeting molecular vulnerabilities of the respective AML subtypes.
HMAs as maintenance therapy
Oral AZA (CC-486) is approved for maintenance therapy following IC in older adults with AML who are not candidates for HSCT or choose not to move forward to HSCT based on the results of the QUAZAR AML-001 study [84]. Other HMAs are being studied for maintenance therapy as summarized below but are not approved for this indication.
QUAZAR AML-001 confirmed an OS benefit for oral AZA (CC-486; 300 mg daily, d1–14, q4 wks) versus placebo after IC. A total of 472 patients aged ≥55 years with AML in CR/CRi ineligible for HSCT were included. Patients randomized to receive CC-486 AZA had improved mOS (24.7 vs. 14.8 months; p<0.01) and relapse-free survival (RFS) (10.2 vs. 4.8 months; p<0.01) [84]. Neutropenia was more common than in the AZA arm (41% vs, 25%). Patients received a median of 12 cycles (range: 1–90). Common AEs included low-grade gastrointestinal events such as nausea which lasted a median of 10 days and grade 3–4 hematologic events which rarely required dose modifications. Only 13% of AEs resulted in permanent discontinuation of the drug [84]. CC-486 did not worsen patient-reported fatigue and QoL [85]. Treatment-related cytopenias typically resolved by cycle 6. Among patients who relapsed, blood counts generally decreased after approximately 9 cycles [86]. CC-486 resulted in a higher rate of conversion to MRD negativity in comparison to placebo (37% vs. 19%). Conversion was also associated with a better OS and PFS [87].
The randomized phase II ECOG-ACRIN E2906 trial was the first HMA maintenance trial performed in older patients with AML after IC and consolidation therapy (N=120; median age 69 years; range 60–85; 74% had intermediate cytogenetic risk). Patients were randomized 1:1 to receive maintenance DEC (20 mg/m2, d1–3, q4 wks) or observation alone. At a median follow up of 49.8 months and a median of 6 cycles of DEC, disease-free survival (DFS) (15.3 vs. 9.2 months; HR, 0.77; 95% CI, 0.50–1.19; p=0.12) and OS (25.8 vs. 19.5 months; HR, 0.69; 95% CI, 0.43–1.09; p=0.06) showed non-significant trends in favor of DEC [88].
The randomized phase III HOVON97 study demonstrated that AZA (50 mg/m2 subcutaneous injection, d1–5, q4 wks) administered until relapse for a maximum of 12 cycles as post-remission treatment after at least 2 cycles of IC and achievement of CR/CRi resulted in a significantly better DFS compared to observation (N=60) in adults ≥60 years (DFS: 64% vs 42% at 12 months; p=0.04) [89]. The benefit remained significant even after adjustment for poor-risk features (HR, 0.62; 95% CI, 0.41–0.95; p=0.026), but no OS benefit was demonstrated (84% vs. 70% at 12 months; p=0.69) [89].
Based on data from case series, HMAs appear to be effective maintenance therapies in patients with CBF AML who were MRD-positive after completion of IC [90, 91]. These results are not specific to older adults and need to be confirmed in randomized trials before HMA maintenance can be recommended in patients with CBF leukemia.
In summary, based on the QUAZAR AML-001 study, maintenance therapy with CC-486 is recommended in older adults after IC who achieved first CR and chose not to receive HSCT.
HMAs and HSCT
HSCT is a curative treatment approach for patients with intermediate- or high-risk AML. Although HSCT is generally reserved for people who are fit (and usually those aged ≤70–80 years), the percentage of patients >70 years who receive HSCT is steadily increasing [92]. Few randomized trials exist in the HSCT setting, especially among older adults. This paucity of results needs to be taken into account when considering data on HMA use in the HSCT setting.
Bridging to HSCT
HMAs have been used successfully as a bridge to HSCT. In the R/R setting, a retrospective study of 655 patients who received HMAs as salvage therapy found that 6% (mean age=56 years) underwent HSCT [93]. Median OS after HSCT was fifteen months. When HMA/VEN was used as salvage treatment in a case series, six out of nine patients were able to undergo HSCT, although only one was aged >60 years [94]. In another retrospective study of 33 patients (median age=62 years) with R/R AML treated with DEC/VEN, only three patients underwent HSCT [95]. A post-hoc analysis of 31 patients (median age 69 years; 10% of the trial population) who received HSCT after upfront treatment with HMA/VEN or LDAC/VEN in two pivotal clinical trials (NCT02287233 and NCT02203773) found a one-year post-HSCT OS of 68%, and 55% of patients remained in remission for at least one year post-HSCT [96]. Similar survival rates after HSCT following treatment with HMA/VEN in either the front-line or R/R setting have been demonstrated in several observational studies [97–102].
HMA as part of conditioning for HSCT
A few small studies (N=23–65) have investigated the addition of HMAs to the conditioning regimen prior to HSCT, although this is not routinely done in clinical practice [103–105]. A phase I study (N=23) combined high-dose DEC with busulfan and cyclophosphamide conditioning and found a high response rate (40% DFS, median= 3.3 years) without increased treatment-related toxicities in the early phase after HSCT [103]. However, given the myeloablative intensity of the regimen, only patients aged <55 years of age were included in the study. The combination of a non-myeloablative conditioning regimen (fludarabine/2 Gy total-body irradiation) with DEC was tested in a prospective Dutch study of 30 patients (median age=59 years) who underwent HSCT for myeloid malignancies (seventeen patients had AML) [104]. The addition of DEC was found to enhance the induction of a tumor-associated antigens-reactive CD8+ T cell response in vivo, and the authors suggested that DEC may contribute to disease control post-HSCT. A third retrospective study reported outcomes after combining DEC with standard myeloablative regimens in 65 patients with R/R AML (median age=37 years) and found a higher OS at three years when compared with a historical cohort (50% vs. 18.5%; p<0.05) [105]. Whether integration of HMAs into conditioning regimens might provide acceptable tolerability and effectiveness needs to be determined in future clinical trials.
Maintenance after HSCT
Several clinical trials have investigated the use of HMAs as post-HSCT maintenance therapy [106]. A meta-analysis of ten studies found two-year OS and RFS estimates of 66% and 62%, respectively [107]. A randomized, open-label phase II study of granulocyte colony-stimulating factor (G-CSF) combined with DEC prophylaxis vs. control in patients with high-risk MRD-negative AML after HSCT (all aged ≤62 years) found that patients in the intervention arm had decreased relapse rates and improved OS at two years (86% vs. 70% in the control arm; p=0.01) [108]. In contrast, a randomized, open-label phase III trial of AZA maintenance post-HSCT vs. control in patients with AML or MDS (median age=57 years) had inconclusive results for OS (HR for OS, 0.84; p=0.43) [109].
Relapse after HSCT
HMAs have also been investigated for pre-emptive or salvage treatment of relapse post-HSCT [110]. Pre-emptive use, often in conjunction with donor lymphocyte infusion (DLI) in the case of decreasing chimerism and/or molecular relapse is promising [110]. The RELAZA trial evaluated the efficacy of AZA monotherapy in the case of decreasing donor chimerism (as a surrogate for an imminent molecular relapse) after HSCT and demonstrated a stabilization or increase in chimerism in 80% of patients [111]. The median age was 58 years. No randomized or prospective observational studies for the use of HMA with DLIs are currently available, but the efficacy of this combination was shown in several case series (summarized in [112]). The immunological rationales for this strategy comprise the upregulation of cancer antigens and/or endogenous retroviruses together with changes in cell-mediated immune responses. For example, AZA was shown to increase the number of CD4+CD25+FOXP3+CD127–regulatory T-cells after HSCT, thereby promoting a graft-versus-leukemia effect without increasing the risk for graft-versus-host disease [110]. Despite these results, treatment schedules (sequence of AZA/DLIs, duration, and dosages) for this approach are not standardized.
In summary, HMAs could facilitate bridging to HSCT, especially in the R/R setting. Their pre-emptive application in the case of a molecular relapse after HSCT is widely employed, particularly in combination with DLIs. Post-HSCT HMA maintenance is not generally used due to mixed results, and further studies are needed to identify which patients might benefit the most from such an approach.
HMAs for relapsed disease
Relapsed AML has a dismal prognosis and no standardized treatment approach. Although studies, primarily retrospective, have demonstrated that HMA monotherapy or combination therapies may improve outcomes, no prospective randomized trials comparing HMA with IC salvage have been conducted. Therefore, the choice of therapy must be individualized. Early clinical trials and retrospective data are summarized in Table 4. Weak evidence has been reported for HMA/VEN effectiveness, even in patients who had been treated with or progressed during HMA monotherapy [113, 114].
Table 4:
Summary of data from trials on HMA/HMA combinations for treatment of R/R AML
| Study | Design and setting | Trial arms | N and age (years) | Key findings |
|---|---|---|---|---|
| HMA monotherapy | ||||
| NCT01261312 [129] | Phase II dose-expansion R/R AML | GUA-5 60/90 mg/m2, d1–5, q4 wks vs. GUA-10 60 mg/m2, d1–10, q4 wks | N=103; median age: 60 y (range: 22–82 y); PA60: n.f.s. | 1-y OS: 28% ORR: 23.3% (95% CI, 15.5%–32.7%) Median time to CR: GUA-5 236 d (range: 64987 d) vs. GUA-10 77 d (range: 38–172 d, p ≤0.04) |
| NCT02197676 [130] | Phase II R/R AML (low-blast count) and high-risk MDS after prior AZA exposure | GUA 60mg/m2, d1–5, q4 wks | N=56; median age: 75 y (range: 70–79 y) | mOS: 7.1 m mOS (responders): 17.9 m (OS>2y in 3 patients) ORR: 14.3% no response in TP53-mutated AML |
| [131] | Retrospective data, 9 centers R/R AML, 20% post-HSCT | AZA/DEC monotherap y (various regimens; +DLI post-HSCT) | N=100; median age: 64.3 y (range: 25–82 y); PA60: n.f.s. | mOS: 6.5 m mOS (SD) 10.6 m mOS (ORR): 13 m mOS (no response): 3.3 m ORR: 24% SD: 26% |
| [132] | Retrospective data, international multicenter R/R AML | AZA/DEC monotherap y (various regimens, incl. DEC-10, and combination therapies used in 22.4%) | N=655; median age: 65 y (range: 16–92 y); PA60: n.f.s. | CR/CRi:16.5 % mOS: 6.7 m (95% CI, 6.1–7.3 m) mOS (CR/CRi): 21m |
| HMA plus venetoclax | ||||
| [133] | Retrospective data R/R AML prior HMA monotherapy included | AZA/VEN (various regimens) | N=77; median age: 64 y (range: 22–85 y); PA60: n.f.s. | mOS: 13.1 m (95% CI, 9.2–15.1 m) PFS: 12 m (95% CI, 8.2 –15.4 m) median duration of response: 8.9 m (95% CI, 5.7–13.9 m) ORR: 78% |
| NCT03404193 [123] | Phase II Newly diagnosed AML and R/R AML | Induction: DEC 20 mg/m2, d1–10/VEN 400 mg/d, d1–28 consolidatio n: DEC 2 0mg/m2, d1–5/VEN 400 mg, d1–28 | N (R/R)=55; median age: 62 y (53–73 y); PA60: n.f.s. | mOS: 7.8 m (95% CI, 5.4–13.3 m) ORR: 62% (95% CI, 49–74) |
| [134] | Retrospective data R/R AML | DEC/VEN (various regimens | N=22; median age: 47.5 y (range: 12–84 y); PA60: n.f.s. | mOS: 6 m (95% CI, 1–9 m) 1-y OS rate: 31.8% 1-y OS (responders) 59.1% 1-y OS (no responders): 10.4% CR rate after cycle 1: 40.9% |
| [135] | Retrospective analysis R/R AML 23% post-HSCT | HMA/VEN (72%, various regimens), LDAC/VEN (28%, various regimens) | N=47; N (≥ 60 y)=42% (20/47); N (≥70 y)=10% (5/47); median age: 56 y (range: 33–74 y); PA60: 48% | mOS: 10.7 m DFS (CR+): 10.6 m ORR: 55% MRD negativity: 16/47 patients |
| [114] | Retrospective data R/R AML after/during HMA | HMA/VEN (various regimens; LDAC/VEN, N=3) | N=23; median age: 76 y (range: 41–92 y); PA60: n.f.s. | mOS (CR/CRi+): 10.8 m CR/CRi: 43% |
| [136] | Retrospective data R/R AML | DEC-10/VEN (N=65) vs. IC (various regimens, N=130) | N=195; median age (HMA/VEN): 64 y; median age (IC): 58 y; PA60 (HMA/VEN): 60%; PA60 (IC): 53% | mOS: HMA/VEN 6.8 m vs. IC 4.7 m (HR, 0.56; 95% CI, 0.37–0.86 m, p=0.008) |
| [137] | Retrospective data 40% with prior HMA therapy | AZA or DEC/VEN (various regimens) | N=42; median age: 64.5 y (range: 18–79 y); PA60: 90.9% | mOS: 5 m (95% CI, 3–9 m) mOS (CR/CRi+): 15 m (95% CI, 5-not reached) CR/CRi: 14 (33.3%) 60-d mortality rate: 33.3% CR/CRi after prior HMA: AZA 75% vs. DEC 34.3% |
| HMA plus checkpoint inhibitor | ||||
| NCT02397720 [125] | Phase II R/R AML | AZA 75mg/m2, d1–7, Nivolumab 3mg/kg, d1, d14, q4–6 | N=77; median age: 70 y (range: 22–90 y); PA60 (AZA/Nivoluma b): 80% | mOS: 6.3 m CR/CRi: 22% |
| NCT03390296 [127] | Phase Ib/II R/R AML | AZA 75 mg/m2, d1–7/avelumab 3 mg/kg to the first 7 patients enrolled and 10 mg/kg for the remaining 12 patients | N=19; median age: 66 y (range: 22–83 y); PA60: 74% | mOS: 4.8 m ORR: 10.5% |
Abbreviations: AML, acute myeloid leukemia; AZA, azacitidine; CR, complete response; CRc, composite response; CRi, complete response with incomplete count recovery; CRp, complete response with incomplete platelet recovery; d, days; DEC, decitabine; DEC-5, DEC as a 5 days regimen; DEC-10, DEC as a 10 days regimen; DLI, donor lymphocyte infusion; EFS, event free survival; GUA, guadecitabine; GUA-5, GUA as a 5 days regimen; GUA-10, GUA as a 10 days regimen; HMA, hypomethylating agent; HR, hazard ratio; HSCT, hematopoietic stem-cell transplantation; mOS, overall survival; MDS, myelodysplastic syndrome; mOS, median overall survival; m, months; n.f.s., not further specified; ORR, overall response rate; OS, overall survival; PA60, percentage above 60 years; PFS, progression-free survival; R/R, relapsed or refractory, SD, stable disease; wks, weeks; y, years.
Conclusion
HMA combination therapies are effective in older adults with AML and reasonably tolerated in selected patients. Nonetheless, prognosis remains poor in this population. Further research is needed to improve outcomes, especially among those with adverse risk factors such as high-risk cytogenetics and TP53 mutations.
Acknowledgements
We wish to acknowledge Susan Rosenthal, MD, for her editorial assistance. We thank Jessica Krok-Schoen from the Young SIOG Governance and Stuart Lichtman from the SIOG Publication Committee for their thoughtful reviews and insightful comments on this paper. The figures were created with BioRender.com. This work was supported by Deutsche Forschungsgemeinschaft (DFG) in the framework of the DFG University Medicine Essen Clinician Scientist Academy (UMEA), FU 356/12-1 to NRN. KPL is supported by the National Cancer Institute at the National Institutes of Health (R00CA237744) and the Wilmot Research Fellowship Award.
NRN has received honoraria and travel support by Janssen-Cilag, Medac, Novartis, Pfizer, Abbvie, and Jazz Pharmaceutical. KPL has served as a consultant to Pfizer and Seattle Genetics and has received honoraria from Pfizer.
Footnotes
Declaration of Competing Interest
JL, NG, VT, SS, SM, LKG, MR, DA, YYS, YZ, and NM declared no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Mikkael AS, Treatment of older adults with acute myeloid leukemia: state of the art and current perspectives. Haematologica, 2008. 93(12): p. 1769–1772. [DOI] [PubMed] [Google Scholar]
- 2.Oran B. and Weisdorf DJ, Survival for older patients with acute myeloid leukemia: a population-based study. Haematologica, 2012. 97(12): p. 1916–1924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zeidan AM, et al. , Clinical outcomes of older patients with AML receiving hypomethylating agents: a large population-based study in the United States. Blood advances, 2020. 4(10): p. 2192–2201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Récher C, et al. , Long-term survival after intensive chemotherapy or hypomethylating agents in AML patients aged 70 years and older: a large patient data set study from European registries. Leukemia, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ihlow J, et al. , Clinical outcome of older adults with acute myeloid Leukemia: An analysis of a large tertiary referral Center over two decades. J Geriatr Oncol, 2021. 12(4): p. 540–549. [DOI] [PubMed] [Google Scholar]
- 6.Loh KP, et al. , Health-Related Quality of Life and Treatment of Older Adults with Acute Myeloid Leukemia: a Young International Society of Geriatric Oncology Review Paper. Curr Hematol Malig Rep, 2019. 14(6): p. 523–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Loh KP, et al. , Beliefs About Advanced Cancer Curability in Older Patients, Their Caregivers, and Oncologists. Oncologist, 2019. 24(6): p. e292–e302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Derissen EJ, Beijnen JH, and Schellens JH, Concise drug review: azacitidine and decitabine. Oncologist, 2013. 18(5): p. 619–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Williams CB, et al. , Preclinical and phase I results of decitabine in combination with midostaurin (PKC412) for newly diagnosed elderly or relapsed/refractory adult patients with acute myeloid leukemia. Pharmacotherapy, 2013. 33(12): p. 1341–52. [DOI] [PubMed] [Google Scholar]
- 10.Schaefer M, et al. , Azacytidine inhibits RNA methylation at DNMT2 target sites in human cancer cell lines. Cancer Res, 2009. 69(20): p. 8127–32. [DOI] [PubMed] [Google Scholar]
- 11.Hollenbach PW, et al. , A comparison of azacitidine and decitabine activities in acute myeloid leukemia cell lines. PLoS One, 2010. 5(2): p. e9001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wolff F, et al. , The double-edged sword of (re)expression of genes by hypomethylating agents: from viral mimicry to exploitation as priming agents for targeted immune checkpoint modulation. Cell Commun Signal, 2017. 15(1): p. 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Li H, et al. , Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget, 2014. 5(3): p. 587–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Liu Y-C, et al. , Demethylation and Up-Regulation of an Oncogene after Hypomethylating Therapy. New England Journal of Medicine, 2022. 386(21): p. 1998–2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Garcia-Manero G, et al. , Oral Azacitidine (CC-486) for the Treatment of Myeloid Malignancies. Clin Lymphoma Myeloma Leuk, 2021. [DOI] [PubMed] [Google Scholar]
- 16.Dhillon S, Decitabine/Cedazuridine: First Approval. Drugs, 2020. 80(13): p. 1373–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Duchmann M. and Itzykson R, Clinical update on hypomethylating agents. Int J Hematol, 2019. 110(2): p. 161–169. [DOI] [PubMed] [Google Scholar]
- 18.Fenaux P, 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): S879. HemaSphere, 2019. 3: p. 394–395. [Google Scholar]
- 19.Wen B, et al. , Indirect comparison of azacitidine and decitabine for the therapy of elderly patients with acute myeloid leukemia: a systematic review and network meta-analysis. Exp Hematol Oncol, 2020. 9: p. 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Saiz-Rodríguez M, et al. , Use of Azacitidine or Decitabine for the Up-Front Setting in Acute Myeloid Leukaemia: A Systematic Review and Meta-Analysis. Cancers (Basel), 2021. 13(22). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zeidan AM, et al. , Prospective comparison of outcomes with azacitidine and decitabine in patients with AML ineligible for intensive chemotherapy. Blood, 2022. 140(3): p. 285–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Coresh J, et al. , Prevalence of chronic kidney disease and decreased kidney function in the adult US population: Third National Health and Nutrition Examination Survey. Am J Kidney Dis, 2003. 41(1): p. 1–12. [DOI] [PubMed] [Google Scholar]
- 23.Mills KT, et al. , A systematic analysis of worldwide population-based data on the global burden of chronic kidney disease in 2010. Kidney Int, 2015. 88(5): p. 950–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Laille E, et al. , A phase I study in patients with solid or hematologic malignancies of the dose proportionality of subcutaneous Azacitidine and its pharmacokinetics in patients with severe renal impairment. Pharmacotherapy, 2014. 34(5): p. 440–51. [DOI] [PubMed] [Google Scholar]
- 25.Douvali E, et al. , Safety and efficacy of 5-azacytidine treatment in myelodysplastic syndrome patients with moderate and mild renal impairment. Leuk Res, 2013. 37(8): p. 889–93. [DOI] [PubMed] [Google Scholar]
- 26.Batty GN, et al. , Feasibility of therapy with hypomethylating agents in patients with renal insufficiency. Clin Lymphoma Myeloma Leuk, 2010. 10(3): p. 205–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Levine LB, et al. , A comparison of toxicities in acute myeloid leukemia patients with and without renal impairment treated with decitabine. J Oncol Pharm Pract, 2018. 24(4): p. 290298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ham JC, Hoogendijk-van den Akker JM, and Verdonck LF, A hemodialysis patient with higher-risk myelodysplastic syndrome treated with standard-dose azacitidine. Leuk Lymphoma, 2012. 53(12): p. 2521–2. [DOI] [PubMed] [Google Scholar]
- 29.Beer AGE, et al. , Azacitidine is removed effectively by hemodialysis. Leuk Lymphoma, 2021. 62(3): p. 743–745. [DOI] [PubMed] [Google Scholar]
- 30.Dombret H, et al. , International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood, 2015. 126(3): p. 291–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kantarjian HM, 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, 2012. 30(21): p. 2670–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vives S, 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, 2021. 127(12): p. 2003–2014. [DOI] [PubMed] [Google Scholar]
- 33.Pleyer L, et al. , Azacitidine for Front-Line Therapy of Patients with AML: Reproducible Efficacy Established by Direct Comparison of International Phase 3 Trial Data with Registry Data from the Austrian Azacitidine Registry of the AGMT Study Group. Int J Mol Sci, 2017. 18(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Agarwal SK, et al. , Response Rates as Predictors of Overall Survival: A Meta-Analysis of Acute Myeloid Leukemia Trials. J Cancer, 2017. 8(9): p. 1562–1567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.He J, et al. , 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, 2015. 56(4): p. 1033–42. [DOI] [PubMed] [Google Scholar]
- 36.Williams S, et al. , Deficiency of Current Acute Myeloid Leukemia (AML) Response Criteria to Predict Response to Hypomethylating Agent Therapy: The Value of Long-Lasting Stable Disease. Blood, 2016. 128(22): p. 2799–2799. [Google Scholar]
- 37.Pleyer L, et al. , Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-Study Group. Ann Hematol, 2014. 93(11): p. 1825–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Döhner H, et al. , Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood, 2017. 129(4): p. 424–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Stomper J, et al. , Hypomethylating agents (HMA) for the treatment of acute myeloid leukemia and myelodysplastic syndromes: mechanisms of resistance and novel HMA-based therapies. Leukemia, 2021. 35(7): p. 1873–1889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Jin S, et al. , 5-Azacitidine Induces NOXA to Prime AML Cells for Venetoclax-Mediated Apoptosis. Clin Cancer Res, 2020. 26(13): p. 3371–3383. [DOI] [PubMed] [Google Scholar]
- 41.DiNardo CD, et al. , Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood, 2019. 133(1): p. 7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.DiNardo CD, et al. , Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N Engl J Med, 2020. 383(7): p. 617–629. [DOI] [PubMed] [Google Scholar]
- 43.Pollyea DA, et al. , Impact of Venetoclax and Azacitidine in Treatment-Naïve Patients with Acute Myeloid Leukemia and IDH1/2 Mutations. Clin Cancer Res, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lachowiez CA, et al. , Outcomes of older patients with NPM1-mutated AML: current treatments and the promise of venetoclax-based regimens. Blood Adv, 2020. 4(7): p. 13111320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pollyea DA, et al. , Venetoclax with azacitidine or decitabine in patients with newly diagnosed acute myeloid leukemia: Long term follow-up from a phase 1b study. Am J Hematol, 2021. 96(2): p. 208–217. [DOI] [PubMed] [Google Scholar]
- 46.Chan SM, et al. , Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nature Medicine, 2015. 21(2): p. 178–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Venugopal S, et al. , Outcomes in patients with newly diagnosed TP53-mutated acute myeloid leukemia with or without venetoclax-based therapy. Cancer, 2021. 127(19): p. 35413551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pollyea DA, et al. , Outcomes in Patients With Poor-risk Cytogenetics With or Without TP53 Mutations Treated With Venetoclax and Azacitidine. Clin Cancer Res, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.DiNardo CD, et al. , Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood, 2020. 135(11): p. 791–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Arslan S, et al. , Outcomes of therapy with venetoclax combined with a hypomethylating agent in favorable-risk acute myeloid leukemia. American Journal of Hematology, 2021. 96(3): p. E59–E63. [DOI] [PubMed] [Google Scholar]
- 51.Stein EM, et al. , Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood, 2017. 130(6): p. 722–731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.DiNardo CD, et al. , Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. New England Journal of Medicine, 2018. 378(25): p. 2386–2398. [DOI] [PubMed] [Google Scholar]
- 53.Raimondi V, et al. , 2-Hydroxyglutarate in Acute Myeloid Leukemia: A Journey from Pathogenesis to Therapies. Biomedicines, 2022. 10(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Montesinos P, et al. , Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. New England Journal of Medicine, 2022. 386(16): p. 1519–1531. [DOI] [PubMed] [Google Scholar]
- 55.Ritchie EK, et al. , Geriatric assessment for older adults receiving less-intensive therapy for acute myeloid leukemia: report of CALGB 361101. Blood Adv, 2022. 6(12): p. 3812–3820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Klepin HD, Toward consensus on geriatric assessment in AML. Blood, 2022. 139(11): p. 1605–1606. [DOI] [PubMed] [Google Scholar]
- 57.Richardson DR, et al. , Time at home among older adults with acute myeloid leukemia based on treatment intensity: A SEER-Medicare analysis. Journal of Clinical Oncology, 2022. 40(16_suppl): p. 6586–6586. [Google Scholar]
- 58.Jensen C, et al. , Time Spent at Home Among Older Adults with AML Treated with Hypomethylating Agents and Venetoclax. Blood, 2021. 138: p. 281-281.33443556 [Google Scholar]
- 59.DiNardo CD, et al. , Azacitidine and Venetoclax in Previously Untreated Acute Myeloid Leukemia. N Engl J Med, 2020. 383(7): p. 617–629. [DOI] [PubMed] [Google Scholar]
- 60.Chang E, et al. , The combination of FLT3 and DNA methyltransferase inhibition is synergistically cytotoxic to FLT3/ITD acute myeloid leukemia cells. Leukemia, 2016. 30(5): p. 1025–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wang ES, et al. , Phase 3 Trial of Gilteritinib Plus Azacitidine Vs Azacitidine for Newly Diagnosed FLT3mut+ AML Ineligible for Intensive Chemotherapy. Blood, 2022. [DOI] [PubMed] [Google Scholar]
- 62.Wang E, 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, 2021. 138: p. 700–700. [Google Scholar]
- 63.Swaminathan M, et al. , A phase I/II study of the combination of quizartinib with azacitidine or low-dose cytarabine for the treatment of patients with acute myeloid leukemia and myelodysplastic syndrome. Haematologica, 2021. 106(8): p. 2121–2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Tomlinson BK, et al. , A Phase II Study of Midostaurin and 5-Azacitidine for Untreated Elderly and Unfit Patients With FLT3 Wild-type Acute Myelogenous Leukemia. Clin Lymphoma Myeloma Leuk, 2020. 20(4): p. 226–233.e1. [DOI] [PubMed] [Google Scholar]
- 65.Maiti A, et al. , Triplet therapy with venetoclax, FLT3 inhibitor and decitabine for FLT3-mutated acute myeloid leukemia. Blood Cancer Journal, 2021. 11(2): p. 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.DiNardo CD, et al. , Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol, 2021. 22(11): p. 1597–1608. [DOI] [PubMed] [Google Scholar]
- 67.DiNardo CD, et al. , Mutant Isocitrate Dehydrogenase 1 Inhibitor Ivosidenib in Combination With Azacitidine for Newly Diagnosed Acute Myeloid Leukemia. J Clin Oncol, 2021. 39(1): p. 57–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Abaza Y. and Zeidan AM, Immune Checkpoint Inhibition in Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cells, 2022. 11(14). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Walter RB, et al. , Intergroup LEAP trial (S1612): A randomized phase 2/3 platform trial to test novel therapeutics in medically less fit older adults with acute myeloid leukemia. Am J Hematol, 2018. 93(2): p. E49–e52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hay AE, et al. , Accrual Barriers and Detection of Early Toxicity Signal in Older Less-Fit Patients Treated with Azacitidine and Nivolumab for Newly Diagnosed Acute Myeloid Leukemia (AML) or High-Risk Myelodysplastic Syndrome (MDS) in the SWOG 1612 Platform Randomized Phase II/III Clinical Trial. Blood, 2019. 134(Supplement_1): p. 3905-3905. [Google Scholar]
- 71.Chao MP, et al. , Therapeutic Targeting of the Macrophage Immune Checkpoint CD47 in Myeloid Malignancies. Front Oncol, 2019. 9: p. 1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Feng D, et al. , Combination Treatment with 5F9 and Azacitidine Enhances Phagocytic Elimination of Acute Myeloid Leukemia. Blood, 2018. 132: p. 2729. [Google Scholar]
- 73.Sallman DA, et al. , Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in MDS and AML patients: Phase Ib results. Journal of Clinical Oncology, 2020. 38(15_suppl): p. 7507–7507. [Google Scholar]
- 74.Sallman D, et al. , AML-196: The first-in-class anti-CD47 antibody magrolimab in combination with azacitidine is well tolerated and effective in AML patients: Phase 1b results. Clinical Lymphoma Myeloma and Leukemia, 2021. 21: p. S290. [Google Scholar]
- 75.Loke J, et al. , Combination romidepsin and azacitidine therapy is well tolerated and clinically active in adults with high-risk acute myeloid leukaemia ineligible for intensive chemotherapy. Br J Haematol, 2022. 196(2): p. 368–373. [DOI] [PubMed] [Google Scholar]
- 76.Craddock CF, et al. , Outcome of Azacitidine Therapy in Acute Myeloid Leukemia Is not Improved by Concurrent Vorinostat Therapy but Is Predicted by a Diagnostic Molecular Signature. Clin Cancer Res, 2017. 23(21): p. 6430–6440. [DOI] [PubMed] [Google Scholar]
- 77.Bhatnagar B, et al. , Selinexor in combination with decitabine in patients with acute myeloid leukemia: results from a phase 1 study. Leuk Lymphoma, 2020. 61(2): p. 387–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Fathi AT, et al. , A phase 1 trial of vadastuximab talirine combined with hypomethylating agents in patients with CD33-positive AML. Blood, 2018. 132(11): p. 1125–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Swords RT, et al. , Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. Blood, 2018. 131(13): p. 1415–1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sekeres MA, et al. , Randomized phase 2 trial of pevonedistat plus azacitidine versus azacitidine for higher-risk MDS/CMML or low-blast AML. Leukemia, 2021. 35(7): p. 2119–2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Medeiros BC, et al. , A Phase I/II Trial of the Combination of Azacitidine and Gemtuzumab Ozogamicin for Treatment of Relapsed Acute Myeloid Leukemia. Clin Lymphoma Myeloma Leuk, 2018. 18(5): p. 346–352.e5. [DOI] [PubMed] [Google Scholar]
- 82.Roboz GJ, et al. , Randomized trial of 10 days of decitabine ± bortezomib in untreated older patients with AML: CALGB 11002 (Alliance). Blood Adv, 2018. 2(24): p. 3608–3617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cluzeau T, et al. , Eprenetapopt Plus Azacitidine in TP53-Mutated Myelodysplastic Syndromes and Acute Myeloid Leukemia: A Phase II Study by the Groupe Francophone des Myélodysplasies (GFM). J Clin Oncol, 2021. 39(14): p. 1575–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wei AH, et al. , Oral Azacitidine Maintenance Therapy for Acute Myeloid Leukemia in First Remission. N Engl J Med, 2020. 383(26): p. 2526–2537. [DOI] [PubMed] [Google Scholar]
- 85.Roboz GJ, et al. , Oral azacitidine preserves favorable level of fatigue and health-related quality of life for patients with acute myeloid leukemia in remission: results from the phase 3, placebo-controlled QUAZAR AML-001 trial. Haematologica, 2021. 106(12): p. 3240–3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ravandi F, et al. , Management of adverse events in patients with acute myeloid leukemia in remission receiving oral azacitidine: experience from the phase 3 randomized QUAZAR AML-001 trial. J Hematol Oncol, 2021. 14(1): p. 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Roboz GJ, et al. , Oral azacitidine prolongs survival of patients with AML in remission independent of measurable residual disease status. Blood, 2022. [DOI] [PubMed] [Google Scholar]
- 88.Foran JM, et al. , Maintenance Decitabine (DAC) Improves Disease-Free (DFS) and Overall Survival (OS) after Intensive Therapy for Acute Myeloid Leukemia (AML) in Older Adults, Particularly in FLT3-ITD-Negative Patients: ECOG-ACRIN (E-A) E2906 Randomized Study. Blood, 2019. 134(Supplement_1): p. 115–115. [Google Scholar]
- 89.Huls G, et al. , Azacitidine maintenance after intensive chemotherapy improves DFS in older AML patients. Blood, 2019. 133(13): p. 1457–1464. [DOI] [PubMed] [Google Scholar]
- 90.Ragon BK, et al. , Minimal residual disease eradication with epigenetic therapy in core binding factor acute myeloid leukemia. Am J Hematol, 2017. 92(9): p. 845–850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Senapati J, et al. , Activity of Decitabine as Maintenance Therapy in Core Binding Factor Acute Myeloid Leukemia. Am J Hematol, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Muffly L, et al. , Increasing use of allogeneic hematopoietic cell transplantation in patients aged 70 years and older in the United States. Blood, 2017. 130(9): p. 1156–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Stahl M, et al. , Allogeneic Hematopoietic Stem Cell Transplantation Following the Use of Hypomethylating Agents among Patients with Relapsed or Refractory AML: Findings from an International Retrospective Study. Biol Blood Marrow Transplant, 2018. 24(8): p. 1754–1758. [DOI] [PubMed] [Google Scholar]
- 94.Zappasodi P, et al. , Venetoclax and azacytidine combination is an effective bridge to transplant strategy in relapsed/refractory acute myeloid leukemia patients. Annals of Hematology, 2020. 100(4): p. 1111–1113. [DOI] [PubMed] [Google Scholar]
- 95.Aldoss I, et al. , Efficacy of the combination of venetoclax and hypomethylating agents in relapsed/refractory acute myeloid leukemia. Haematologica, 2018. 103(9): p. e404–e407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Pratz KW, et al. , Outcomes after Stem Cell Transplant in Older Patients with Acute Myeloid Leukemia Treated with Venetoclax-Based Therapies. Blood, 2019. 134(Supplement_1): p. 264-264. [Google Scholar]
- 97.Salhotra A, et al. , Venetoclax and hypomethylating agents yield high response rates and favourable transplant outcomes in patients with newly diagnosed acute myeloid leukaemia. Br J Haematol, 2021. n/a(n/a). [DOI] [PubMed] [Google Scholar]
- 98.Sandhu KS, et al. , Outcome of Allogeneic Hematopoietic Cell Transplantation after Venetoclax and Hypomethylating Agent Therapy for Acute Myelogenous Leukemia. Biol Blood Marrow Transplant, 2020. 26(12): p. e322–e327. [DOI] [PubMed] [Google Scholar]
- 99.Pollyea DA, et al. , Venetoclax and azacitidine followed by allogeneic transplant results in excellent outcomes and may improve outcomes versus maintenance therapy among newly diagnosed AML patients older than 60. Bone Marrow Transplant, 2021. [DOI] [PubMed] [Google Scholar]
- 100.Pasvolsky O, et al. , Allogeneic hematopoietic cell transplantation for acute myeloid leukemia in first complete remission after 5-azacitidine and venetoclax: a multicenter retrospective study. Ann Hematol, 2022. 101(2): p. 379–387. [DOI] [PubMed] [Google Scholar]
- 101.Winters AC, et al. , Outcomes Are Similar Following Allogeneic Hematopoietic Stem Cell Transplant for Newly Diagnosed Patients Who Received Venetoclax + Azacitidine Versus Intensive Chemotherapy. Blood, 2021. 138(Supplement 1): p. 3957-3957. [Google Scholar]
- 102.Kennedy VE, et al. , Hypomethylating Agents in Combination with Venetoclax As a Bridge to Allogeneic Transplant in Acute Myeloid Leukemia. Blood, 2020. 136(Supplement 1): p. 32–33. [Google Scholar]
- 103.de Lima M, et al. , Long-term follow-up of a phase I study of high-dose decitabine, busulfan, and cyclophosphamide plus allogeneic transplantation for the treatment of patients with leukemias. Cancer, 2003. 97(5): p. 1242–7. [DOI] [PubMed] [Google Scholar]
- 104.Cruijsen M, et al. , Addition of 10-Day Decitabine to Fludarabine/Total Body Irradiation Conditioning is Feasible and Induces Tumor-Associated Antigen-Specific T Cell Responses. Biol Blood Marrow Transplant, 2016. 22(6): p. 1000–1008. [DOI] [PubMed] [Google Scholar]
- 105.Li Y, et al. , A Retrospective Observation of Treatment Outcomes Using Decitabine-Combined Standard Conditioning Regimens Before Transplantation in Patients With Relapsed or Refractory Acute Myeloid Leukemia. Front Oncol, 2021. 11: p. 702239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Bewersdorf JP, Prebet T, and Gowda L, Azacitidine maintenance in AML post induction and posttransplant. Curr Opin Hematol, 2022. [DOI] [PubMed] [Google Scholar]
- 107.Bewersdorf JP, et al. , Hypomethylating Agents and FLT3 Inhibitors As Maintenance Treatment for Acute Myeloid Leukemia and Myelodysplastic Syndrome After Allogeneic Hematopoietic Stem Cell Transplantation-A Systematic Review and Meta-Analysis. Transplant Cell Ther, 2021. 27(12): p. 997 e1–997 e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gao L, et al. , Effect of rhG-CSF Combined With Decitabine Prophylaxis on Relapse of Patients With High-Risk MRD-Negative AML After HSCT: An Open-Label, Multicenter, Randomized Controlled Trial. J Clin Oncol, 2020. 38(36): p. 4249–4259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Oran B, et al. , A phase 3 randomized study of 5-azacitidine maintenance vs observation after transplant in high-risk AML and MDS patients. Blood Adv, 2020. 4(21): p. 5580–5588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ciotti G, Marconi G, and Martinelli G, Hypomethylating Agent-Based Combination Therapies to Treat Post-Hematopoietic Stem Cell Transplant Relapse of Acute Myeloid Leukemia. Front Oncol, 2021. 11: p. 810387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Platzbecker U, et al. , Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia, 2012. 26(3): p. 381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Webster JA, Luznik L, and Gojo I, Treatment of AML Relapse After Allo-HCT. Front Oncol, 2021. 11: p. 812207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Othman TA, et al. , Retreatment with venetoclax and hypomethylating agents among AML patients who have relapsed after initial response and subsequent interruption of therapy. Leuk Lymphoma, 2020. 61(14): p. 3532–3533. [DOI] [PubMed] [Google Scholar]
- 114.Ram R, et al. , Venetoclax in patients with acute myeloid leukemia refractory to hypomethylating agents-a multicenter historical prospective study. Ann Hematol, 2019. 98(8): p. 1927–1932. [DOI] [PubMed] [Google Scholar]
- 115.Chabner BA, Drake JC, and Johns DG, Deamination of 5-azacytidine by a human leukemia cell cytidine deaminase. Biochem Pharmacol, 1973. 22(21): p. 2763–5. [DOI] [PubMed] [Google Scholar]
- 116.Gonzalez-Zulueta M, et al. , Methylation of the 5’ CpG island of the p16/CDKN2 tumor suppressor gene in normal and transformed human tissues correlates with gene silencing. Cancer Res, 1995. 55(20): p. 4531–5. [PubMed] [Google Scholar]
- 117.Min G-J, et al. , Geriatric assessment predicts nonfatal toxicities and survival for intensively treated older adults with AML. Blood, 2022. 139(11): p. 1646–1658. [DOI] [PubMed] [Google Scholar]
- 118.Short NJ, et al. , Treatment with a 5-day versus a 10-day schedule of decitabine in older patients with newly diagnosed acute myeloid leukaemia: a randomised phase 2 trial. Lancet Haematol, 2019. 6(1): p. e29–e37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Falantes J, et al. , Real life experience with frontline azacitidine in a large series of older adults with acute myeloid leukemia stratified by MRC/LRF score: results from the expanded international E-ALMA series (E-ALMA+). Leuk Lymphoma, 2018. 59(5): p. 1113–1120. [DOI] [PubMed] [Google Scholar]
- 120.Peterlin P, et al. , Azacitidine in patients older than 80 years with acute myeloid leukaemia or myelodysplastic syndromes: a report on 115 patients. Br J Haematol, 2020. 190(3): p. 461464. [DOI] [PubMed] [Google Scholar]
- 121.Dargenio M, et al. , Long-Term Follow-Up of Elderly Patients with Acute Myeloid Leukemia Treated with Decitabine: A Real-World Study of the Apulian Hematological Network. Cancers (Basel), 2022. 14(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Chetasi T, et al. , Comparisons of commonly used front-line regimens on survival outcomes in patients aged 70 years and older with acute myeloid leukemia. Haematologica, 2020. 105(2): p. 398–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.DiNardo CD, et al. , 10-day decitabine with venetoclax for newly diagnosed intensive chemotherapy ineligible, and relapsed or refractory acute myeloid leukaemia: a single-centre, phase 2 trial. Lancet Haematol, 2020. 7(10): p. e724–e736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ohanian M, et al. , Sorafenib Combined with 5-azacytidine in Older Patients with Untreated FLT3-ITD Mutated Acute Myeloid Leukemia. Am J Hematol, 2018. 93(9): p. 1136–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Daver N, et al. , Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Nonrandomized, Open-Label, Phase II Study. Cancer Discov, 2019. 9(3): p. 370–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Goswami M, et al. , Pembrolizumab and decitabine for refractory or relapsed acute myeloid leukemia. J Immunother Cancer, 2022. 10(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Saxena K, et al. , A phase 1b/2 study of azacitidine with PD-L1 antibody avelumab in relapsed/refractory acute myeloid leukemia. Cancer, 2021. 127(20): p. 3761–3771. [DOI] [PubMed] [Google Scholar]
- 128.Huls G, et al. , Ibrutinib added to 10-day decitabine for older patients with AML and higher risk MDS. Blood Adv, 2020. 4(18): p. 4267–4277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Roboz GJ, et al. , Dose, schedule, safety, and efficacy of guadecitabine in relapsed or refractory acute myeloid leukemia. Cancer, 2018. 124(2): p. 325–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Sébert M, et al. , A phase II study of guadecitabine in higher-risk myelodysplastic syndrome and low blast count acute myeloid leukemia after azacitidine failure. Haematologica, 2019. 104(8): p. 1565–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lessi F, et al. , Hypomethylating Agents (HMAs) as Salvage Therapy in Relapsed or Refractory AML: An Italian Multicentric Retrospective Study. Biomedicines, 2021. 9(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Stahl M, et al. , Hypomethylating agents in relapsed and refractory AML: outcomes and their predictors in a large international patient cohort. Blood Adv, 2018. 2(8): p. 923–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Graveno ME, et al. , Venetoclax in combination with hypomethylating agents or low dose cytarabine for relapsed and refractory acute myeloid leukemia. Leuk Lymphoma, 2022: p. 1–6. [DOI] [PubMed] [Google Scholar]
- 134.Tong J, et al. , Efficacy of Venetoclax Combined with Decitabine-Based Treatment for Heavily Pre-Treated Relapsed or Refractory AML Patients in a Real-World Setting. Cancer Manag Res, 2021. 13: p. 5613–5621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Piccini M, et al. , Venetoclax-Based Regimens for Relapsed/Refractory Acute Myeloid Leukemia in a Real-Life Setting: A Retrospective Single-Center Experience. J Clin Med, 2021. 10(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Maiti A, et al. , Ten-day decitabine with venetoclax versus intensive chemotherapy in relapsed or refractory acute myeloid leukemia: A propensity score-matched analysis. Cancer, 2021. 127(22): p. 4213–4220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Morsia E, et al. , Venetoclax and hypomethylating agents in acute myeloid leukemia: Mayo Clinic series on 86 patients. Am J Hematol, 2020. 95(12): p. 1511–1521. [DOI] [PubMed] [Google Scholar]
- 138.Konopleva M, et al. , Impact of F LT3 Mutation on Outcomes after Venetoclax and Azacitidine for Patients with Treatment-Naïve Acute Myeloid Leukemia. Clin Cancer Res, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Venugopal S, et al. , Prognostic impact of conventional cytogenetics in acute myeloid leukemia treated with venetoclax and decitabine. Leuk Lymphoma, 2021. 62(14): p. 35013505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Kim K, et al. , Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer, 2021. 127(20): p. 3772–3781. [DOI] [PMC free article] [PubMed] [Google Scholar]


