Abstract
Although outcomes for newly diagnosed acute myeloid leukaemia (AML) have been incrementally improved over the last decades, management of relapsed and refractory (R/R) AML remains a medical challenge. A curative intent for R/R AML usually involves chemotherapy (with or without targeted therapy) with subsequent consolidation, including allogeneic haematopoietic stem cell transplantation. Despite this, long‐term survival rates of R/R AML only reach approximately 10% in adults and 40% in children. Given this great unmet clinical need, this review outlines the current and emerging paradigms for preventing and treating R/R AML. Somatic mutations, gene expression, and functional drug testing are important for the selection of small molecule inhibitors of oncogenic signalling pathways (e.g., FLT3), menin inhibitors that disrupt leukemogenic programmes, inhibitors of isocitrate dehydrogenases to restore oncometabolic homoeostasis, and proapoptotic Bcl‐2 homology 3 (BH3) mimetics, such as venetoclax. Targeting the recently identified resistance factor SAMHD1 promises to overcome resistance to cytarabine and fludarabine. Given the growing number of potential combinatorial drug regimens and the genetic heterogeneity of AML, real‐time ex vivo drug response profiling to guide individualized treatment decisions will become an important complement. We argue that better outcomes for R/R AML critically depend on being guided by precision oncology to define the best combination of chemotherapy, targeted therapy, and immunological therapy informed by phenotypic and genotypic patient‐ and disease‐specific parameters.
Keywords: azacitidine, cladribine, cytarabine, drug screening, FLT3, fludarabine, hydroxyurea, IDH1, IDH2, menin, resistance mechanisms, SAMHD1, venetoclax
Introduction
Acute myeloid leukaemia (AML) is caused by the acquisition of distinct genetic aberrations in myeloid progenitors of the bone marrow, leading to maturation block and uncontrolled proliferation of leukemic blasts. The identification and characterization of distinct genetic and epigenetic aberrations have led to a vastly increased understanding of AML pathobiology in recent years. At the same time, a plethora of novel therapeutic agents have become available. So far, these developments have not been convincingly translated into improved outcomes in patients with relapsed or refractory (R/R) disease. This article aims to provide a comprehensive review of state‐of‐the‐art and biology‐informed emerging strategies for R/R AML management. We argue that future decisions in terms of R/R AML treatment must be approached in a multimodal manner, taking into consideration molecular alterations (e.g., oncogenic mutations), mechanisms of chemotherapy resistance, immunological properties and functional ex vivo drug sensitivity profiling.
Epidemiology and definitions
The incidence of AML is around 4 per 100,000 in adults, with 30% of patients developing AML secondary to a prior myeloid malignancy or to prior cancer treatment [1]. Patients receiving intensive chemotherapy achieve complete remission (CR) or CR with incomplete recovery (CRi) in 50%–90% of cases, with ∼70% being measurable residual disease (MRD) negative after two induction courses [2]. Approximately 65% of unfit adults receiving non‐intensive therapy, primarily azacitidine and venetoclax, achieve CR/CRi, and approximately 15% reach MRD negativity [3]. The probability of obtaining a response to treatment depends on the underlying genetics/disease risk category, treatment intensity and age. The 5‐year overall survival (OS) for intensively treated patients depends on patient characteristics and ranges from around 41% to 62% in patients categorized as low‐risk but only around 8%–25% in those with high‐risk [4].
Refractory AML is defined as the persistence of leukemic blasts in the bone marrow (>5%) after two cycles of intensive induction chemotherapy. Relapsed AML is defined by the recurrence of more than 5% bone marrow blasts or extramedullary growth after achieving an initial remission [5]. However, in many previous and ongoing trials, lower levels of residual leukemic cells as measured by flow cytometry of molecular methods are considered a relapse event. Even fit low‐risk patients treated with high‐intensity chemotherapy have a significant risk of relapse of ∼25% [6], and nearly all high‐risk patients would relapse without subsequent allogeneic haematopoietic cell transplantation (allo‐HSCT).
In children, AML constitutes ∼20% of leukaemias and is characterized by age‐specific features in clinical presentation [7], genetic profile [8, 9] and to some extent outcome [10, 11]. In contrast to adults, AML in children infrequently arises as a continuum of previous myelodysplastic syndrome [12] or after cytotoxic therapy [13]. Instead, germline genetic predisposition is more common in children [14].
Children with de novo AML are treated according to collaborative study group intensive chemotherapy protocols leading to CR in approximately 90% of children [15, 16], of whom 70%–80% may obtain MRD negativity [17]. Despite the immediate efficacy of primary AML therapy, 25%–40% of children experience relapse [17], which remains a significant therapeutic challenge and still portends an unsatisfactorily poor outcome with only 35%–50% long‐term OS [18].
Approximately 5%–10% of children with AML respond poorly to primary therapy with refractory disease (RD) [17, 19]. CR may be achieved through additional induction attempts, but prognosis is generally poor in children with RD, with OS rates below 40% [20, 21].
Current concepts to prevent relapse
Despite the high initial response rates to induction therapy, a significant proportion of patients will eventually relapse, particularly adult patients. To reduce the need for salvage treatment, large efforts both in paediatric and adult AML have been made to optimize the primary treatment to achieve deeper responses and to reduce the risk of relapse. Recently, intensified induction in adults with Fludarabine/Cytarabine/Granulocyte Colony‐Stimulating Factor (G‐CSF)/Idarubicin/Gemtuzumab‐ozogamicin (FLAG‐Ida‐GO) has been shown to result in superior outcome in NPM1‐mutated AML [6].
Importantly, genetic risk stratification has improved substantially, and predictive scores for both fit and unfit AML have recently been published for adults (ELN 2022—fit, ELN 2024—unfit) [5, 22]. The risk scores not only predict response to treatment but also identify patients who are at high risk of disease recurrence. The risk of relapse can be reduced by allo‐HSCT, which is generally considered for non‐favourable risk fit AML patients in first remission. An increasing number of AML patients receive allo‐HSCT thanks to expanding international donor registries, improved protocols for using haploidentical donors, co‐morbidity‐adapted conditioning regimens and improved supportive care.
In the paediatric setting, primary induction therapy is usually more intensive, and further intensification is associated with unacceptable treatment‐related mortality and morbidity [23, 24]. Collaborative study groups have reached a consensus on response features, but risk‐classification systems based on high‐risk genomic features in recent clinical trials still vary [25].
Another strategy to reduce the risk of relapse is to administer maintenance therapy after achieving remission, including in some cases maintenance after allo‐HSCT. Successful maintenance strategies that have demonstrated survival benefits in randomized trials in adult AML include the FLT3‐inhibitors midostaurin, quizartinib and gilteritinib [26, 27, 28], the oral hypomethylating agent (HMA) azacitidine [29] and the immune‐stimulatory combination of histamine dihydrochloride and interleukin‐2 [30]. In children with FLT3‐internal tandem duplication (ITD) AML, maintenance with sorafenib has been evaluated [31], and ongoing trials are exploring gilteritinib (NCT04293562) and quizartinib [32]. Several additional maintenance strategies are currently being studied in phase 3 trials in genetic subtypes of AML, including inhibitors of isocitrate dehydrogenase (IDH)1/2 and menin [33].
If a relapse is identified early, at the MRD level, patients are generally in good condition. In this situation, the disease is often easier to treat (absence of cytopenia, coagulopathies, leukostasis and tumour lysis syndrome), and eligible patients can proceed rapidly to allo‐HSCT with or without prior anti‐leukemic treatment. Pre‐emptive treatment of imminent relapse detected by longitudinal molecular MRD monitoring with highly sensitive assays is an area undergoing rapid development and may be an attractive strategy to improve outcome, at least in select AML genotypes [34, 35, 36].
Despite extensive efforts to optimize treatment, R/R AML remains a challenge in many patients. This review focuses on the key aspects of how to achieve a second remission to bridge the patient to a potentially curative allo‐HSCT or to maintain disease control with sustained quality of life.
Challenges in treating R/R AML
Resistance to the first line of intensive therapy may be due to inherent resistance mechanisms in leukemic cells, present either in all AML cells or in a subclone. Drug‐specific resistance mechanisms and strategies to overcome resistance are described below. The importance of disease heterogeneity and clonal evolution of more resistant subclones during the course of the disease has been characterized in depth [37, 38, 39]. As stem cell‐like leukemic cells (leukemic stem cells, LSCs) are less proliferative and possess drug efflux activity [40], they are also less sensitive towards cytotoxic treatments. Accordingly, stem cell disorders are less responsive to chemotherapy [41]. Smouldering MDS‐AML are often true stem cell disorders, where the malignant transformation has occurred near the level of the haematopoietic stem cell. There are several strategies of how to target cells with low proliferation, including apoptosis‐inducers such as the Bcl‐2 homology 3 domain (BH3) mimetic venetoclax or antibodies directed at surface molecules on the LSCs. Another strategy is to use growth factors that increase the proliferation of the LSCs. G‐CSF has been explored greatly, though without much success [42], potentially due to insufficient expression of the G‐CSF receptor on LSCs. Finally, given the low proliferation rate, extended maintenance therapy with HMAs, with or without venetoclax, may gradually wear down the leukemic clones and delay or even prevent relapse [3, 29], a notion most clearly illustrated in acute lymphoblastic leukaemia [43].
Extramedullary AML manifestation (such as skin, lymph nodes, gastrointestinal tract or central nervous system [CNS] [44, 45]) requires special consideration. It is crucial to achieve local disease control because graft‐versus‐leukaemia (GvL) effects seem to be less protective against extramedullary relapse after allo‐HSCT [46]. Depending on the sites and extent of extramedullary involvement, radiotherapy may be useful as an add‐on to systemic treatment to ensure local disease control, particularly in adults. The optimal dose of radiation is unknown, although data suggest that 12 fractions of 2 Gy (24 Gy in total) may be sufficient [47]. Several small case series have indicated that novel AML regimens may be effective also in extramedullary disease, including HMAs, venetoclax and FLT3 inhibitors such as gilteritinib [48, 49]. Some novel drugs are small molecules that penetrate the blood‐brain barrier, which may be important for AML involving the CNS [50]. Additionally, checkpoint inhibitors, such as the anti‐CTLA4 monoclonal antibody ipilimumab, have shown promising efficacy in a small case series [51].
Finally, certain subtypes of AML are exceedingly challenging to cure in the R/R setting, in particular adult patients with double hit TP53 mutations, where a palliative strategy with good supportive care and cytoreductive chemotherapy to control hyperleukocytosis need to be considered [52, 53].
Salvage chemotherapy approaches
Intensive chemotherapy strategies
A prerequisite for long‐term survival is allo‐HSCT, generally after a second CR has been achieved following salvage therapy [18, 20, 54, 55]. Although less intensive regimens based on HMAs with or without the Bcl‐2 inhibitor venetoclax as well as small‐molecule inhibitors of FLT3, IDH1 or IDH2 can achieve CR in a subset of patients [54, 55] (see below), re‐induction with chemotherapy is still mostly used in patients fit‐for‐transplantation (Table 1).
Table 1.
Summary of potential treatment strategies for relapsed and refractory (R/R) acute myeloid leukaemia (AML).
Treatment modality | Treatment specifics | References |
---|---|---|
Intensive treatment |
|
[61] |
Non‐intensive treatment |
|
[55], this review |
Targeted therapy augmentation | Consider adding inhibitors of FLT3 (gilteritinib, quizartinib), IDH1 (ivosidenib), IDH2 (enasidenib), menin (e.g., revumenib) depending on mutations and rearrangements | [31, 66, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84] |
Overcoming SAMHD1‐mediated resistance | Potentially add HU for ara‐C or ara‐C + F‐ara‐A ‐containing regimens | [85, 86, 87, 88, 89] |
Cardioprotection | Consider dexrazoxane when using anthracyclines/anthraquinones | [90, 91] |
Ex vivo drug profiling | May be informative when selecting chemotherapy backbones and targeted inhibitors | [92, 93, 94, 95] |
Cure | Allo‐HSCT | [55, 96] |
Maintenance post allo‐HSCT |
|
[98] [33] [55] [30] |
Abbreviations: Allo‐HSCT, allogeneic haematopoietic cell transplantation; CAV, cladribine, ara‐C, and venetoclax; IDH, isocitrate dehydrogenase; HMA, hypomethylating agent; HU, hydroxyurea.
The attempt of re‐induction with intensive chemotherapy must consider treatment resistance. It is therefore surprising that most established salvage chemotherapy regimens contain the same backbone as the primary treatment: intermediate or high doses of cytarabine (ara‐C), with the rationale to overcome ara‐C resistance by increasing the dose. Historically used as monotherapy, some studies reported CR rates of up to 50% [99]. With the exception of HMAs that have a different mode of action (see below), monotherapy with other cytotoxic nucleoside analogues than ara‐C has been less effective, as demonstrated for the purine analogues cladribine [100] and clofarabine [101, 102, 103]. Of note, addition of an anthracycline to cladribine or clofarabine did not improve outcomes either [104, 105].
Ara‐C is usually combined with a topoisomerase II inhibitor, where daunorubicin is frequently exchanged with another anthracycline (e.g., idarubicin), the anthraquinone mitoxantrone or the non‐anthracycline amsacrine. Shifting the topoisomerase II inhibitor might circumvent daunorubicin resistance [106, 107]. A long‐established two‐drug re‐induction course combines high‐dose ara‐C (considered to be superior to intermediate dose ara‐C [108], see below) with mitoxantrone (HAM) [109]. In a retrospective analysis, the German–Austrian AML Study Group found addition of gemtuzumab‐ozogamicin (together with all‐trans retinoic acid) to the HAM backbone to be more potent in inducing CR/CRi (50% vs. 25%) [110]. Just like with ara‐C, dose‐escalation of anthracyclines has been tested [111, 112]; any potential antileukemic benefit must be balanced against the increased risk of cardiotoxicity. In the primary treatment setting, a large randomized trial did not show a benefit of intensifying daunorubicin doses from 60 to 90 mg/m2 [113] (in contrast to a landmark trial comparing 45 to 90 mg/m2) [114]. Another rationale to overcome resistance to first‐line therapy is adding drugs with a different mode of action, such as the canonical topoisomerase‐II inhibitor etoposide, which is most widely used in combinations with ara‐C and mitoxantrone (MEC) and has shown feasibility as a bridge‐to‐transplant approach with success rates of around 50% [64, 65].
More importantly, overcoming resistance to ara‐C remains the central element for re‐induction/salvage courses aiming to increase exposure to the active metabolite ara‐CTP, with levels of ara‐CTP correlating with enhanced anti‐leukemic effect [115, 116, 117]. Further, based on in vitro experiments showing moderate increases of ara‐CTP when pre‐treating leukemic cells with fludarabine (F‐ara‐A) [118], addition of F‐ara‐A to intermediate and high doses of ara‐C with or without (G‐CSF) as a priming strategy was introduced clinically (FLA(G)) [119, 120]. In the paediatric setting, the addition of liposomal daunorubicin to FLAG resulted in better OS of R/R AML; however, only in the subset of core‐binding factor AML [121]. Idarubicin has been incorporated into FLAG‐Ida protocols [60, 122], which improved remission rates. Perplexingly, addition of F‐ara‐A to ara‐C and idarubicin‐based re‐induction therapy only modestly improved CR rates and did not improve overall or relapse‐free survival in a randomized phase 3 trial for R/R AML [122]. This is, however, consistent with a lack of improved survival reported for upfront addition of F‐ara‐A to ara‐C and daunorubicin [123].
Two additional adenosine analogues have been combined with ara‐C‐based intensive reinduction therapies. Addition of cladribine has been shown to achieve an increase of ara‐CTP in cells from R/R AML patients [124]. The Polish Adult Leukemia Group (PALG) showed that addition of cladribine to ara‐C (together with G‐CSF; CLAG) in an anthracycline‐free regimen resulted in a CR rate of 50% in R/R AML, and responding patients had a 1‐year OS of 65% [125]. Contrary to those findings, a paediatric and young adult phase 2 trial at St. Jude's did not show efficacy of cladribine plus (low‐intermediate dose) ara‐C in R/R AML [126]. Another PALG study evaluated the CLAM regimen in R/R AML, that is, addition of cladribine to HAM. with a CR rate of ∼50% and a 1‐year OS in patients achieving CR of 73% [62]. Similar results were obtained in an American trial [63]. Taken together, also considering that addition of cladribine to ara‐C and daunorubicin has been successful in newly diagnosed AML patients [123], salvage regimens based on high‐dose ara‐C, an anthracycline and cladribine appear efficacious.
Clofarabine in combination with ara‐C and mitoxantrone (CLoAM) resulted in overall response rates of 90% in 52 R/R AML patients in a Hongkong phase 2 trial [127]. Addition of clofarabine to ara‐C and idarubicin in MD Anderson R/R AML trials yielded objective response rates of up to 80% [128, 129]. On the other hand, a phase 1B trial in children with R/R AML combining clofarabine, ara‐C and liposomal daunorubicin produced CR rates of 68% with 2‐year OS of 50% [130].
Inclusion of anthracyclines/mitoxantrone increases the risk of cardiotoxicity. Therefore, salvage treatment using amsacrine instead of idarubicin together with F‐ara‐A and ara‐C (FLAMSA) was evaluated and found to be an effective alternative [61]. Despite these modifications, prognosis for R/R AML remains bleak for most patients, warranting further therapy improvements. A standard of care for intensive salvage therapy remains to be established, and the backbone may need to be tailored to patients with different disease characteristics.
Novel concepts of improving the efficacy and tolerability of intensive chemotherapy
Resistance to ara‐C is a hurdle for effective therapy, and salvage courses that can overcome this resistance are aimed at increasing leukemic exposure to the active metabolite ara‐CTP, as its accumulation is critical for its anti‐leukemic effect [117]. The enzyme SAMHD1 is a deoxynucleoside triphosphate triphosphohydrolase that can metabolize ara‐CTP into inactive precursors, leading to ara‐C resistance in leukemic cells [85, 86]. In line with this, patients with AML who have high levels of SAMHD1 fare worse than their SAMHD1‐low counterparts when treated with ara‐C‐based regimens [87]. Hydroxyurea (HU) is a ribonucleotide reductase inhibitor that can indirectly inhibit SAMHD1 by inducing dNTP pool imbalances, thus reducing the enzymatic activity of SAMHD1 towards ara‐CTP [88]. Addition of HU to ara‐C overcomes SAMHD1‐mediated resistance and improves treatment efficacy in vitro, ex vivo and in vivo [88] and has shown the ability to safely increase leukemic ara‐CTP exposure in a phase 1 clinical trial in adults with newly diagnosed AML [89] (Fig. 1).
Fig. 1.
Resistance mechanisms and targets in relapsed and refractory (R/R) acute myeloid leukaemia (AML), part 1. This figure illustrates three resistance mechanisms and molecular targets for the treatment of R/R AML. (a) SAMHD1 is a triphosphohydrolase that detoxifies leukemic cells from cytarabine and fludarabine's active metabolites ara‐CTP and F‐ara‐ATP; SAMHD1 can be indirectly inhibited by hydroxyurea (HU). (b) Mutations in isocitrate dehydrogenase (IDH)1/2 lead to an increase of the oncometabolite 2‐hydroxyglutarate that in turn inhibits histone and DNA demethylases. This process can be inhibited by IDH inhibitors. Hypermethylation can also be targeted by hypomethylating agents (HMAs) and cladribine (2‐CdA). (c) Upregulation of anti‐apoptotic protein Bcl‐2 confers resistance to conventional chemotherapy. The Bcl‐2 inhibitors can alleviate this process, but Bcl‐xL and MCL‐1 can mediate resistance to Bcl‐2 inhibition.
Similarly, SAMHD1 has been shown to confer resistance to F‐ara‐A in vitro [85]. We have gathered unpublished evidence that resistance to F‐ara‐A‐containing regimens, in particular FLA, against AML is mediated by SAMHD1 and can be alleviated by addition of HU (Lilienthal et al., personal communcation).
While increasing the anthracycline dose has been shown to improve survival in primary treatment [114], dose‐escalation of anthracyclines is limited by the dose‐dependent risk of cardiotoxicity [131]. The cardioprotectant dexrazoxane effectively reduces risk of cardiotoxicity by protecting cardiac topoisomerase 2β from anthracyclines [90]. In osteosarcoma, addition of dexrazoxane allowed a safe increase in doxorubicin cumulative doses from 450 to 600 mg/m2 without compromising treatment efficacy or safety [91]. Hence, adding dexrazoxane appears a feasible strategy to allow for anthracycline‐intensified treatment of R/R AML. A different strategy to increase daunorubicin efficacy while reducing cardiotoxicity is to use liposomal formulations that have been shown to decrease cardiotoxicity in vitro [132]. CPX‐351 is a liposomal ara‐C/daunorubicin formulation that has shown similar efficacy as compared to FLA‐Ida in newly diagnosed AML [133]. In a non‐randomized paediatric R/R AML study, CPX‐351 nevertheless was associated with a significant deterioration of left‐ventricular ejection fraction and cardiac dysfunction [134]. A randomized phase 2 trial in adults with first relapse of AML comparing CPX‐351 with investigator's choice of salvage resulted in a CR rate of ∼40% [135]. In children with de novo AML, CPX‐351 + GO resulted in inferior efficacy as compared to DA + GO [136] (Fig. 2).
Fig. 2.
Resistance mechanisms and targets in relapsed and refractory (R/R) acute myeloid leukaemia (AML), part 2. This figure illustrates three resistance mechanisms and molecular targets for the treatment of R/R AML. (a) Activating mutations in the receptor tyrosine kinase FLT3, in particular internal tandem duplications, can be targeted FLT3 inhibitors. (b) Surface expression of CD33 makes AML vulnerable to the antibody‐drug conjugate Gemtuzumab‐ozogamicin. (c) Interaction of KTM2A (and NUP98) chimera with menin leads to transactivation of HOXA9 and myeloid ecotropic virus insertion site 1 (MEIS1). This interaction can be inhibited by menin inhibitors. They are also effective in NPM1‐mutated and upstream binding transcription factor tandem duplicated (UBTF‐TD) AML.
Targeted therapies
Venetoclax and other BH3 mimetics
BH3‐only proteins constitute an important part of a pro‐apoptotic signalling pathway, resulting in activation of Bax and Bak, which triggers apoptosis via cytochrome c release from mitochondria. BH3‐only proteins are partially sequestered by anti‐apoptotic Bcl‐2 family proteins Bcl‐xL, MCL1 and Bcl‐2‐related protein A1, which often are upregulated in cancer cells, thus inhibiting apoptosis [137].
BH3‐mimetics are a class of drugs that bind the anti‐apoptotic Bcl‐2 family proteins, which unleashes the BH3‐only proteins to trigger apoptosis via activation of Bax and Bak [138]. BH3‐mimetics may also interfere with the oxidative metabolism in LSCs, which starves them to a greater degree than normal haematopoietic stem cells [139]. Importantly, preclinical and clinical data have demonstrated synergy between BH3‐mimetics and numerous other drugs with different modes of action, making them some of the most promising novel targeted drugs in AML.
Venetoclax specifically inhibits Bcl‐2 (Fig. 1) and is at present the only BH3‐mimetic approved for AML by the European Medicines Agency (EMA). The EMA approval is based on phase 2 and phase 3 data demonstrating markedly improved outcomes when combining venetoclax with the HMAs azacitidine or decitabine [3, 140]. Promising results were also observed in newly diagnosed AML when venetoclax was combined with low‐dose cytarabine in a randomized trial; however, OS was not significantly improved, which contributed to the EMA's decision not to approve the combination for this indication [59]. Positive predictors for response to HMA and venetoclax include mutations in IDH2 and NPM1, and negative predictors include mutations in TP53, FLT3‐ITD or K / NRAS [3, 141]. The main side effect is haematotoxicity, and some patients experience extended cytopenias already after the first treatment cycle [3].
Resistance to venetoclax can be mediated via various mechanisms, including activation of tyrosine kinase signalling (e.g., via mutations in genes such as FLT3, NRAS, KRAS), inactivation of TP53, mutations in BCL2 that impair the binding of venetoclax and inactivating mutations in BAX [141, 142, 143, 144, 145]. Moreover, leukemic cells may be dependent on other anti‐apoptotic Bcl‐2‐family proteins, exemplified by a relative dependence on MCL1 in monocytic leukemic progenitors and Bcl‐xL in leukemic cells with erythroid/megakaryocytic features [146, 147]. These resistance factors may be present at diagnosis, resulting in primary resistance, or may be acquired or enriched during therapy, resulting in relapse. Compounds inhibiting MCL1 and Bcl‐xL have also been explored, with and without venetoclax, with promising efficacy. Some safety concerns have been observed, including potential cardiac toxicity when using MCL1 inhibitors, and optimal drug combinations and dosage of these novel BH3‐mimetics remain to be defined [138].
In R/R AML, venetoclax combinations have been extensively explored and will be covered in other sections of this review. Promising combinations include HMAs and venetoclax, particularly in HMA‐naïve patients, and combinations with intensive chemotherapy or kinase inhibitors and other targeted drugs.
Several novel BH3 mimetics are under development in AML, including the next generation Bcl‐2 inhibitor sonrotoclax [148]. Careful assessment of toxicity will be important, and it remains to be seen if they eventually will replace venetoclax in up‐front therapy or be reserved for R/R AML.
Low intensity chemotherapy combinations with venetoclax
Hypomethylating agents and venetoclax
Treatment with HMAs and venetoclax has revolutionized treatment of newly diagnosed AML, whereas the response rate and duration in R/R AML is markedly lower. The probability of response largely depends on the underlying disease biology. Recently, a large retrospective analysis compared patients with R/R AML who had failed intensive chemotherapy and received azacitidine with or without venetoclax. The composite CR, CRc, (CR + Cri + CR with incomplete platelet recovery [CRp]) rate was 55 versus 22%, with and without venetoclax, respectively, and the OS was significantly longer both in patients receiving subsequent allo‐HSCT and in those who did not [66]. Combinations of azacitidine and venetoclax may also induce durable responses in up to 40% of heavily pre‐treated children with R/R AML [67]. Positive predictors for better response to the combination were, as expected, mutations in IDH1, IDH2 or NPM1 [66].
Cladribine, low‐dose cytarabine and venetoclax
A lower intensity regimen for newly diagnosed AML aimed at older or unfit patients, combining cladribine, low‐dose cytarabine and venetoclax (CAV) alternated with azacitidine‐venetoclax, has shown an impressive CR/CRi rate of 93% in phase 2 data [68]. In the relapsed setting, this would be an attractive option in patients failing azacitidine‐venetoclax who are ineligible for intensive treatment or who are lacking druggable targets.
However, in a retrospective analysis of 39 elderly patients (median age 65 years) with R/R AML and intermediate or high‐risk genetics, of which the majority had received azacitidine‐venetoclax as prior therapy, the CAV regimen only resulted in a 28% CR/CRi rate [69]. Similar results were observed in prospective non‐randomized study in R/R AML, including 30 patients who were considerably younger (median age 40 years) and with a large proportion of favourable genetics (47%), with a CR/CRi rate of 27% [70]. However, the ORR was high (70%), and the majority (60%) could be transitioned to allo‐HSCT.
Importantly, monocytic leukaemia appears to respond better to CAV, which is in line with preclinical evidence that the addition of cladribine to azacitidine‐venetoclax targets the monocytic leukemic stem cell [149]. AML with monocytic phenotype is associated with mutations in the mitogen‐activated protein kinase (MAPK) pathway, and MAPK pathway mutations are known to respond less favourably to azacitidine‐venetoclax [149, 150]. In the trials above evaluating CAV in R/R AML, the response rates for patients with RAS mutations were unfortunately inconsistent, with 0 of 8 K/NRAS and 7 of 9 NRAS mutated patients achieving CR/CRi in the two trials, respectively [69, 70].
In summary, cladribine, low‐dose cytarabine and venetoclax is a tolerable option to consider in R/R AML with promising responses in particular in monocytic AML, whereas molecular predictors warrant further exploration.
High‐intensity chemotherapy combination with venetoclax
FLAG‐Ida with venetoclax (FLAVIDA)
Addition of venetoclax to FLAG‐Ida (FLAVIDA) has been attempted in a phase‐2 trial in R/R AML with improved CR rates of 47%, MRD negativity of 74% and 1‐year survival of around 50% [56, 57]. Similar results were obtained from registry data even though during the short follow‐up and with a very small cohort of 13 patients, no differences in outcome were seen as compared to a control cohort of FLAG‐Ida patients (n = 81) [58].
Other intensive regimens with venetoclax
Venetoclax is also being explored together with several other intensive regimens, both in the up‐front and R/R setting. In newly diagnosed AML, the prospective CAVEAT trial—combining venetoclax with intensive chemotherapy—showed that this combination was efficacious and well tolerated even in elderly patients [151]. In R/R AML, a retrospective report demonstrated that venetoclax added to cladribine, high‐dose cytarabine and idarubicin or mitoxantrone resulted in ORR of 77% versus 50% with or without venetoclax, respectively [152]. These intensive combinations need further exploration, with particular focus on toxicity, until they can be considered for broader use.
In early phase trials in children, venetoclax in combination with intensive chemotherapy appears well tolerated, and promising efficacy has been reported [59].
FLT3 inhibitors including combinations
Mutations in FMS‐related tyrosine kinase 3 (FLT3) occurs in 20%–30% of patients with AML and is often associated with rapidly proliferating disease. Two main categories of mutations occur; ITD or activating mutations in the tyrosine kinase domain (TKD). Available FLT3 inhibitors differ in their kinase specificity and in their mode of action. Type 1 inhibitors (midostaurin, gilteritinib, crenolanib) target both ITD and TKD mutations, whereas type 2 inhibitors (sorafenib, quizartinib) target only ITD. Mechanisms of resistance include acquisition of other downstream kinase mutations or mutations in FLT3 that are resistant to the utilized inhibitor, including the known gatekeeper mutation F691L, which confers resistance to all currently approved FLT3 inhibitors [153].
In newly diagnosed AML, midostaurin and quizartinib are approved by the EMA as an add‐on to intensive chemotherapy, based on confirmed survival benefit in randomized trials [26, 27]. Quizartinib in combination with chemotherapy has an acceptable toxicity profile in children and is currently tested by the NOPHO‐DB‐SHIP consortium [32]. Sorafenib lacks formal approval in AML by the EMA but is recommended in several guidelines, including the Swedish National Guidelines for maintenance treatment post allo‐HSCT, also based on results from randomized trials [97]. Sorafenib administered in between courses of primary therapy is well tolerated in children and showed encouraging signals of activity in FLT3‐ITD AML with high allelic ratio [31]. Gilteritinib used as post‐transplantation maintenance improves survival in patients who have detectable MRD for FLT3‐ITD pre‐ or post‐transplantation; however, at present, gilteritinib is not approved in this setting by the EMA [28].
In R/R AML, the only drug currently approved by the EMA is gilteritinib (Fig. 2). Used as a single agent in R/R AML with FLT3‐ITD or TKD, the CRc rate was 54.3% versus 21.8% for conventional salvage therapy, with a significant improvement in median OS of 9.3 versus 5.6 months (HR 0.64, p < 0.001). In children, only limited data on gilteritinib is available. Overall, gilteritinib is well tolerated, with moderate haematotoxicity, although other significant side effects may occur, including differentiation syndrome [75].
Preclinical data indicate a synergy between FLT3 inhibitors and venetoclax [154]. So‐called doublet therapies (FLT3 inhibitor + venetoclax) and triplet therapies (FLT3 inhibitor + HMA + venetoclax) have been explored in phase 1 and 2 trials in R/R AML, with impressive response rates. The mCRc (CRc + MLFS, morphologic leukaemia‐free state) for gilteritinib combined with venetoclax was 75%, and 44% of evaluated patients achieved at least a 3‐log reduction of the FLT3‐ITD MRD level [76]. Corresponding figures for the triplet of gilteritinib, azacitidine and venetoclax were an mCRc rate of 73%, with MRD negativity in 43% of evaluable patients [77]. Somewhat disappointingly, both the gilteritinib doublet and triplet therapies yielded similar median OS as gilteritinib alone (9.3, 10.0, 5.8 months, respectively, for singlet, doublet, triplet therapy). Despite high initial efficacy, relapse remains a significant challenge even in combination trials. Another caveat is that both doublet and triplet therapies are highly myelotoxic, with frequent extended cytopenias in addition to other adverse events.
Novel trials exploring other FLT3 inhibitor combinations aim to optimize dosing and treatment duration of the doublet or triplet drugs during each treatment cycle, to improve both efficacy and tolerance. Moreover, implementing doublet or triplet therapy as an early salvage therapy likely leads to deeper responses and potentially improved long‐term outcome, particularly for patients bridged to allo‐HSCT.
Finally, available FLT3 inhibitors are not fully specific for FLT3, and inhibition of other kinases may play a role in their anti‐leukemic effect. Clinical and pre‐clinical data suggest that FLT3 inhibitors may be efficacious both as single agents and in combination with chemotherapy or venetoclax, even in subsets of AML patients without FLT3 mutations [154]. Intriguingly, a randomized trial with sorafenib as an add‐on to conventional chemotherapy demonstrated an improved event‐free but not OS, indicating an added efficacy but also increased toxicity [155]. Moreover, preliminary results from a randomized trial of standard chemotherapy with or without quizartinib demonstrated and increased OS in FLT3‐negative patients (median not reached vs. 29.3 months, hazard ratio 0.63) [78]. Quizartinib is being further explored in this setting in a recently started prospective randomized phase 3 trial (QuANTUM‐Wild) [156].
IDH inhibitors including combinations
Mutations in IDH 1 or IDH 2 occur in around 20% of patients with AML, with IDH2 being somewhat more frequent. Several inhibitors have been developed, and the most extensively investigated are ivosidenib (IDH1) and enasidenib (IDH2). When the mutant IDH protein is inhibited, the oncometabolite 2‐hydroxyglutarate is formed, which interferes with the cellular metabolism — resulting in DNA hypermethylation by impairing the function of TET2 — and also induces Bcl‐2 dependency to evade apoptosis [157, 158]. These mechanisms of action provide rationales for combining IDH inhibitors with HMAs and Bcl‐2 inhibitors. In line with this, clinical data have demonstrated high activity of venetoclax‐based regimens in patients with IDH mutations [3].
In R/R AML treated with ivosidenib, the CRc rate was 30.4% and the median response duration 8.2 months [79]. In IDH2 mutated R/R AML, enasidenib resulted in a CRc rate of 27.8%, with a median response duration of 5.6 months. IDH inhibitors are well tolerated, and a class effect is that they may result in differentiation syndrome.
To improve efficacy, IDH inhibitors have been combined both with low‐ and high‐intensity chemotherapy. In newly diagnosed AML with IDH1 mutation, a randomized trial of azacitidine with or without ivosidenib demonstrated a marked improvement in OS for the combination, which has led to market approval by tEMA [159]. Moreover, in newly diagnosed AML, ivosidenib or enasidenib have been combined with standard intensive chemotherapy, with promising activity, and the concept is now being explored in a randomized setting [80]. Interestingly, a retrospective analysis of 398 newly diagnosed AML patients treated in a randomized fashion with ara‐C and daunorubicin plus/minus cladribine suggested that IDH2 mutations conferred a better survival in patients receiving cladribine, possibly through a reversal of IDH2‐induced hypermethylation [160] (Fig. 1). IDH inhibition as maintenance therapy after induction and consolidation, as well as after allo‐HSCT, is an important area currently under investigation, where early data are promising [98].
The triplet of ivosidenib, venetoclax and azacitidine has shown promising activity in newly diagnosed AML, whereas the results were less impressive in R/R AML, with a CRc rate of 63% and an OS of 9 months [81]. Moving forward, novel IDH inhibitors may prove to have even higher efficacy, and a key issue will be how best to combine the targeted inhibitors with other available drugs.
Menin inhibitors including combinations
The complex of the nuclear scaffold protein menin and the histone‐lysine N‐methyltransferase 2A (KMT2A) protein constitutes a molecular dependency for pro‐leukemic gene expression in acute leukaemia characterized by enforced transcription of HOX homeobox genes and the co‐factor myeloid ecotropic virus insertion site 1 (MEIS1) [161]. These entities include AML with KMT2A‐rearrangements (KMT2A‐r) [162, 163] as the most prominent example but also AML with rearranged nucleoporin 98 (NUP98‐r) [164], mutated nucleophosmin (NPM1) [165] and tandem duplications in the upstream binding transcription factor (UBTF‐TD) [166]. KMT2A‐r occur in up to 10% of all acute leukaemias in adults and children, are particularly frequent in infants with ALL or AML [8], and are generally associated with high risk of relapse [167]. During the last decade, extensive preclinical research using models of acute leukaemia with aberrant HOX/MEIS1 expression has demonstrated how menin inhibitors selectively block the menin‐KMT2A interaction, leading to restoration of myeloid differentiation, leukaemia cell apoptosis and loss of the leukemic transcription signature [165, 166, 168, 169, 170]. These results established the foundation of recently initiated early‐phase clinical trials of menin inhibitors in the setting of R/R AML (Fig. 2).
The first‐in‐human study with a menin inhibitor was AUGMENT‐101 (NCT04065399), a phase 1/2 trial of revumenib in children and adults with R/R acute leukaemias. The toxicity profile was acceptable, mainly with mild to moderate cytopenias and a moderate risk of differentiation syndrome and QTc prolongation [82]. In the biomarker‐driven phase 2 portion of AUGMENT‐101, NPM1‐mutated patients are still enrolling while efficacy data on 57 patients with KMTA2‐r acute leukaemias showed an overall response rate of 63% of whom 15/22 responders (68%) achieved MRD negativity, and 14 patients (39%) were bridged to allo‐HSCT [83].
In the KOMET‐001 phase 1/2 trial of ziftomenib in adults with R/R AML, the safety profile was manageable (cytopenias and differentiation syndrome). Twelve of 38 patients (32%) with KMT2A‐r or NPM1‐mutated AML receiving ziftomenib at the highest dose level of the dose‐validation phase achieved an overall response [84].
Early phase clinical trials of bleximenib (NCT04811560) and enzomenib (NCT04988555) are currently recruiting adult acute leukaemia patients, and preliminary results indicate comparable safety and efficacy profiles across the different menin inhibitors [171, 172].
Genome‐wide sequencing of bone‐marrow samples from patients with only temporary responses to revumenib in the AUGMENT‐101 identified mutations in the MEN1 gene conferring structural changes of the KMT2A‐binding pocket that prohibit interaction with the inhibitor but without affecting menin‐KMT2A complex formation. Longitudinal tracking by digital PCR demonstrated emergence of these MEN1 mutations in approximately 40% of patients continuously exposed to revumenib [173].
Application of contemporary chemotherapy to menin inhibitors may be an appealing strategy to obtain therapeutic synergy and prevent expansion of MEN1‐mutated leukemic clones. A number of trials, including both intensive and less‐intensive chemotherapy, with or without venetoclax, in combination with menin inhibitors, are currently recruiting patients with newly diagnosed and R/R acute leukaemias. Given the acquisition of resistance to menin inhibitors over the course of treatment reported from the AUGMENT‐101 trial [173], it will be important to monitor resistance in these trials and to evaluate how combination therapies may be used to circumvent resistance.
Targeting the p53 pathway
Up to 20% of patients with newly diagnosed AML harbour TP53 aberrations [174], and TP53‐mutated AML is strongly associated with resistance to traditional therapy options and higher rates of relapse after allo‐HSCT [174, 175, 176]. One study found that 15% of patients with R/R AML had TP53 mutations that were not detectable in their primary disease, suggesting that these mutations can emerge under selective pressure of treatment [177]. Given the inferior outcomes of TP53‐mutated AML with traditional regimens, targeting the p53 pathway and harnessing the immune system (see below) have been evaluated as alternative strategies.
APR‐246 (eprenetapopt) is a p53 activator, albeit with many off‐target effects [178]. Two parallel phase 2 trials in patients with newly diagnosed TP53‐mutated MDS or AML demonstrated that APR‐246 in combination with azacitidine yielded high CR rates (40%–50%) and notably improved OS in patients who were bridged to allo‐HSCT [179, 180]. One trial combining APR‐246 and azacitidine as maintenance therapy following allo‐HSCT in TP53‐mutated MDS/AML showed promising OS [181]. Although trials using APR‐246 in the R/R setting are lacking, results indicate it may be beneficial in bridging TP53‐mutated patients to transplant, and its use in the setting of R/R AML warrants further investigation.
Leukemic cells with wild‐type TP53 can be sensitized to p53‐mediated apoptosis by MDM2 inhibition. One phase 1b trial tested a combination of venetoclax and the MDM2 inhibitor idasanutlin in a cohort of patients with R/R AML with modest results [182]. Another study in the R/R AML setting with a different MDM2 inhibitor (milademetan) showed similar results [183]. This effect was not enhanced when combining milademetan with venetoclax and low‐dose cytarabine in R/R AML [184].
Ex vivo drug screening to guide therapeutic choices
Evaluating laboratory responses to anti‐cancer therapies prior to clinical treatment—inspired by the success of antibiotic sensitivity testing in microbial diseases—has been a longstanding goal in leukaemia and cancer research [185]. Multiple trials have investigated the feasibility of ex vivo drug sensitivity testing for AML in a prospective setting.
The Helsinki Functional Precision Medicine Tumour Board combined clinical, molecular and functional data to guide AML treatment decisions [92]. In 39 of 186 R/R AML patients, recommendations based on drug response data resulted in a 59% response rate for tailored therapies. This trial primarily relied on bulk‐sensitivity assays. The EXALT trial profiled 143 patients with advanced haematologic malignancies using a microscopy‐based single‐cell readout, with 56 participants treated [93]. Among these, 54% achieved clinical benefit, demonstrating more than a 1.3‐fold increase in PFS compared to prior therapies, although data specific to AML patients were limited. Similarly, the SMART trial provided drug‐response profiling reports to 91% of participants within 7 days, linking ex vivo chemotherapy resistance to in vivo treatment failure. However, this study mainly focused on newly diagnosed AML cases [186].
From these studies, it is clear that ex vivo drug sensitivity testing for AML is technically achievable. Due to the nature of these studies, they did not systematically compare ex vivo drug responses with clinical outcomes in a prospective manner.
Translating ex vivo drug testing into clinical care requires careful consideration of methodological factors, including variations in cell culture media and viability testing techniques. Previous studies [187, 188] have shown that evaluating response using bulk bone marrow samples can lead to false predictions of resistance. Flow cytometry analysis demonstrated that distinct cell populations, such as monocytic and granulocytic cells, have different sensitivities to venetoclax, potentially masking blast‐specific responses [94, 188].
The prospective VenEx trial [94, 95] (NCT04267081) evaluated the correlation between ex vivo venetoclax sensitivity and clinical outcomes with venetoclax and azacitidine in patients with AML. To systematically assess the variables, the trial's initial phase, involving 39 participants, tested three cell culture media and two viability methods. The highest correlation with clinical outcomes was achieved using conditioned culture medium and blast‐specific response evaluation by flow cytometry. Final results from VenEx demonstrated the practicality of integrating ex vivo drug sensitivity testing into clinical workflows and highlighted its value in identifying AML patients likely to benefit from venetoclax, particularly in the R/R setting.
Ex vivo venetoclax sensitivity testing was successfully performed in 98% of participants, with results available within 3 days of sampling. In R/R and secondary AML patients, where treatment options are often limited, ex vivo sensitivity corresponded to a 62% CR/CRi rate and a median OS of 9.7 months compared to 3.3 months in resistant patients. In untreated AML, sensitivity was associated with an 85% CR/CRi rate and a median OS of 28.7 months. Additionally, ex vivo sensitivity correlated with MRD negativity and emerged as the most significant predictor of favourable outcomes in univariate and multivariate analyses [94, 95].
These findings underscore the transformative potential of ex vivo drug sensitivity testing for tailoring AML treatments. By addressing technical challenges and enhancing predictive accuracy, this approach holds promise for broader clinical implementation and improved patient outcomes.
In recent years, efforts have been undertaken to establish patient‐derived AML reconstitution in PDX models to study drug sensitivity. Following the initial challenges in engrafting immunodeficient mice with primary AML cells, protocols were developed to allow for such engraftment [189]. Interestingly, in such a model, lower EFS rates in patients were associated with more successful reconstitution of their primary AML cells in PDX animals. Furthermore, enrichment was observed for R/R clones upon serial transfer in PDX animals that mimicked clonal evolution upon R/R disease in corresponding patients [190]. Although several groups have reported the use of PDX models to test specific drugs, the models still lack robustness to effectively engraft AML blasts from all patients; hence, implementation in clinical practice has yet to happen.
Immunotherapy and allo‐HSCT
Allo‐HSCT plays a critical role in the management of R/R AML and is one of the few potentially curative treatment options for these high‐risk patients (Table 1). In cases in which AML is refractory to initial therapy or has relapsed following a period of remission, outcomes with conventional salvage chemotherapy are generally poor. Allo‐HSCT offers the benefit of both intensive cytoreduction and a GvL effect mediated by donor immune cells, which can target residual leukemic cells and reduce the risk of relapse. Patients with R/R AML who achieve a second CR or even proceed to transplantation with residual disease may benefit from transplantation [5]. Novel strategies — such as the use of reduced‐intensity conditioning, haploidentical donors and post‐transplant maintenance therapies — make allo‐HSCT more accessible and may improve outcomes in this challenging patient population [96]. Despite these advances, relapse remains a significant cause of post‐transplant mortality.
The monoclonal antibody drug conjugate GO that has been described as an augmentation to conventional chemotherapy for CD33+ disease above (Fig. 2) does not in itself elicit direct immunotherapeutic effects such as antibody‐dependent cellular cytotoxicity [191]. However, GO simultaneously targets CD33+ myeloid‐derived suppressor cells [192] and has therefore been suggested for a clinical trial in patients with solid tumours (EUDRACT: 2020‐002428‐36).
Immune checkpoint inhibitors have been suggested in particular for patients who do not respond to HMAs, as this was correlated with higher expression of PD‐1/PD‐L1 and therefore suggested to be a resistance mechanism (reviewed in [71]). Accordingly, anti‐PD‐1 (nivolumab or pembrolizumab) antibodies have been evaluated in combination with azacitidine in non‐randomized phase 2 clinical trials for R/R AML with an ORR of 18%–33% [72, 73]. Pembrolizumab has also been given in a phase 2 trial as a maintenance treatment following high‐dose ara‐C with a CR rate of 38% in R/R AML [74].
Targeting immune evasion, CD47‐SIRPα interaction has been explored extensively. CD47 is a cell surface protein expressed on malignant cells often referred to as a ‘don't eat me’‐signal binding to SIRPα on macrophages, inhibiting phagocytosis. Blocking the interaction with anti‐CD47/SIRPα‐antibodies or Fc fusion proteins promote phagocytosis of leukaemia cells by macrophages and other immune cells. The first‐in‐class monoclonal antibody against CD47, magrolimab, showed promising early‐phase efficacy in combination with azacitidine in previously untreated AML [193]. The initially high expectations of the anti‐CD47/SIRPα concept have been dampened when FDA placed all studies of magrolimab in haematologic malignancies on hold due to futility and increased risk of death. However, several other drugs targeting CD47/SIRPα combined with azacitidine ± venetoclax are currently being investigated in phase 1/2 clinical trials in both newly diagnosed and R/R AML [194].
Following the success of anti‐CD19 chimeric antigen receptor (CAR)‐T cells in lymphoid disease, efforts have been made to introduce CAR‐T cell treatment for R/R myeloid disease. CAR‐T cells have been developed to target, for instance, CD33, CD123 or CD135 [195]. A complicating factor in these approaches is that many of these targets are expressed on haematopoietic stem and/or progenitor cells, resulting in severe and prolonged cytopenia. In addition, downregulation of antigens (immune escape) is frequently observed in relapsed AML [196], which hinders antigen recognition by CAR‐T cells. The development of newer generations of CAR‐T cells is aimed at overcoming these challenges.
Finally, a large number of other immunotherapies are being explored that are not covered here due to a paucity of clinical data and space limitations. This field is developing rapidly, as recently reviewed by Bawek et al. [71]. Given the impressive results in other haematological malignancies, we trust that, with further optimization, immunotherapies will play a key role in the future management of R/R AML, in particular for patients who are ineligible for curative allo‐HSCT.
Conclusions and future directions
Despite the rapidly expanding portfolio of active anti‐leukemic agents and the broader availability of genetic and phenotypic diagnostic tools, including MRD monitoring, the treatment of R/R AML remains a clinical challenge, and the long‐term overall outcome is poor. Novel combinations, including targeted drugs, hold promise to improve outcomes in specific subtypes of AML. Conversely, extreme high‐risk diseases, such as double‐hit TP53 mutated AML, require particular efforts and entirely new approaches, ideally directed at overcoming the intrinsic apoptosis resistance.
Conventional clinical trials, even when biomarker‐driven, are hampered by the inherent clinical and biological heterogeneity of the patient population; in children, a limited number of patients, unequal access to new drugs and limited health economy resources are further complications. Multimodal precision oncology approaches appear to be a rational strategy for guiding future practice, but they require prospective evaluation (Fig. 3). It is important to consider the genetic and phenotypic architecture at high resolution (e.g., massive parallel sequencing allowing for detection of minor allele frequencies) as well as the intra‐patient disease heterogeneity (e.g., single‐cell based approaches) in combination with informed ex vivo testing of drugs and drug combinations with flow cytometry‐based readouts. Regarding maintenance therapies, proper ex vivo simulation remains difficult. We believe that a combination of genetic and phenotypic data together with ex vivo drug profiling may allow better individualized treatment decisions. In addition, new methods need to be developed to allow prediction of the efficacy of immunomodulating and immunotherapeutic drugs.
Fig. 3.
Schematic of treatment modalities for treatment of relapsed and refractory (R/R) acute myeloid leukaemia (AML) with intention‐to‐cure. Upper panel: To cure R/R AML, remission induction followed by consolidation and maintenance therapy is advised. Lower panel: Remission induction heavily relies on intensive chemotherapy that might be informed by ex vivo drug testing. Intensive chemotherapy should be complemented by targeted therapy (as outlined in Figs. 1 and 2), and immunotherapy might be considered for individual cases. Consolidation is achieved by allogeneic haematopoietic cell transplantation (allo‐HSCT). Maintenance therapy is usually reserved for targeted agents, but also hypomethylating agents (HMAs) and immunotherapy might be considered (see Table 1).
We trust that ongoing efforts to further understand the complex biology of AML and improve diagnostic tools and predictive capabilities will serve as a foundation to develop novel and more efficient patient‐tailored therapies, including a combination of conventional chemotherapy, targeted drugs and immunotherapeutic approaches.
Conflict of interest statement
M.J. has been part of AdBoards (receiving institutional support) for AbbVie and Cancell Therapeutics. He has also been involved in educational events (receiving institutional support) for AbbVie, Pfizer, Lab Delbert, and Astra Zeneca. M.K. has been part of AdBoards for Faron and Proteina. He has also been an advisor for BMS and Astellas, served as a consultant for Ferring and Faron, and been a member of the speaker's bureau for AbbVie, Servier, Faron and Jazz. The remaining authors declare no conflicts of interest.
Acknowledgements
We would like to thank Anette Langebäck for critical comments on the manuscript. NH received funding from the Swedish Society for Medical Research (SG‐23‐0178‐B), the Swedish Cancer Society (24 3398 Pj; 25 3999 IA JCIA), the Swedish Childhood Cancer Foundation (PR2023‐0031) and the Swedish Research Council (2024‐02941).
Jädersten M, Lilienthal I, Nilsson C, Fredrikson L, Pronk CJ, Juul‐Dam KL, et al. Precision oncology to overcome resistance in R/R AML in children and adults requires combinations of cytotoxic, targeted, and immunological treatments. J Intern Med. 2025;298:297–318.
References
- 1. Nilsson C, Linde F, Hulegårdh E, Garelius H, Lazarevic V, Antunovic P, et al. Characterization of therapy‐related acute myeloid leukemia: increasing incidence and prognostic implications. Haematologica. 2023;108(4):1015–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Short NJ, Zhou S, Fu C, Berry DA., Walter RB, Freeman SD, et al. Association of measurable residual disease with survival outcomes in patients with acute myeloid leukemia: a systematic review and meta‐analysis. JAMA Oncol. 2020;6(12):1890–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Dinardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–629. [DOI] [PubMed] [Google Scholar]
- 4. Rausch C, Rothenberg‐Thurley M, Dufour A, Schneider S, Gittinger H, Sauerland C, et al. Validation and refinement of the 2022 European LeukemiaNet genetic risk stratification of acute myeloid leukemia. Leukemia. 2023;37(6):1234–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Döhner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 2022;140(12):1345–1377. [DOI] [PubMed] [Google Scholar]
- 6. Russell NH, Wilhelm‐Benartzi C, Othman J, Dillon R, Knapper S, Batten LM, et al. Fludarabine, cytarabine, granulocyte colony‐stimulating factor, and idarubicin With gemtuzumab ozogamicin improves event‐free survival in younger patients with newly diagnosed AML and overall survival in patients with NPM1 and FLT3 mutations. J Clin Oncol. 2024;42(10):1158–1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Støve HK, Sandahl JD, Abrahamsson J, et al. Extramedullary leukemia in children with acute myeloid leukemia: a population‐based cohort study from the Nordic Society of Pediatric Hematology and Oncology (NOPHO). Pediatr Blood Cancer. 2017;64(12): e26520. [DOI] [PubMed] [Google Scholar]
- 8. Bolouri H, Farrar JE, Triche T, Ries RE, Lim EL, Alonzo TA, et al. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age‐specific mutational interactions. Nat Med. 2018;24(1):103–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Umeda M, Ma J, Westover T, Ni Y, Song G, Maciaszek JL., et al. A new genomic framework to categorize pediatric acute myeloid leukemia. Nat Genet. 2024;56(2):281–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Løhmann DJ, Abrahamsson J, Ha SY, et al. Effect of age and body weight on toxicity and survival in pediatric acute myeloid leukemia: results from NOPHO‐AML 2004. Haematologica. 2016;101(11):1359–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Sandahl KJ, Juul‐Dam K, Herlin MK, Abrahamsson J, Arad‐Cohen N, Cheuk D, et al. Comparison of the WHO and ICC classifications of cytogenetic abnormalities in childhood acute myeloid leukemia. Blood. 2024;144(Supp1):322–322. [Google Scholar]
- 12. Strahm B, Nöllke P, Zecca M, Korthof ET, Bierings M, Furlan I, et al. Hematopoietic stem cell transplantation for advanced myelodysplastic syndrome in children: results of the EWOG‐MDS 98 study. Leukemia. 2011;25(3):455–462. [DOI] [PubMed] [Google Scholar]
- 13. Brown CA, Youlden DR, Aitken JF, Moore AS. Therapy‐related acute myeloid leukemia following treatment for cancer in childhood: a population‐based registry study. Pediatr Blood Cancer. 2018;65(12):e27410. [DOI] [PubMed] [Google Scholar]
- 14. Samaraweera SE, Wang PPS, Li KL, Casolari DA, Feng J, Pinese M, et al. Childhood acute myeloid leukemia shows a high level of germline predisposition. Blood. 2021;138(22):2293–2298. [DOI] [PubMed] [Google Scholar]
- 15. Pession A, Masetti R, Rizzari C, Putti MC, Casale F, Fagioli F, et al. Results of the AIEOP AML 2002/01 multicenter prospective trial for the treatment of children with acute myeloid leukemia. Blood. 2013;122(2):170–178. [DOI] [PubMed] [Google Scholar]
- 16. Tsukimoto I, Tawa A, Horibe K, Tabuchi K, Kigasawa H, Tsuchida M, et al. Risk‐stratified therapy and the intensive use of cytarabine improves the outcome in childhood acute myeloid leukemia: the AML99 trial from the Japanese Childhood AML Cooperative Study Group. J Clin Oncol. 2009;27(24):4007–4013. [DOI] [PubMed] [Google Scholar]
- 17. Tierens A, Arad‐Cohen N, Cheuk D, De Moerloose B, Fernandez Navarro JM, Hasle H, et al. Mitoxantrone versus liposomal daunorubicin in induction of pediatric AML with risk stratification based on flow cytometry measurement of residual disease. J Clin Oncol. 2024;42(18):2174–2185. [DOI] [PubMed] [Google Scholar]
- 18. Karlsson L, Forestier E, Hasle H, Jahnukainen K, Jónsson ÓG, Lausen B, et al. Outcome after intensive reinduction therapy and allogeneic stem cell transplant in paediatric relapsed acute myeloid leukaemia. Br J Haematol. 2017;178(4):592–602. [DOI] [PubMed] [Google Scholar]
- 19. Rubnitz JE, Lacayo NJ, Inaba H, Heym K, Ribeiro RC, Taub J, et al. Clofarabine can replace anthracyclines and etoposide in remission induction therapy for childhood acute myeloid leukemia: the AML08 multicenter, randomized phase III trial. J Clin Oncol. 2019;37(23):2072–2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Karlsson L, Cheuk D, De Moerloose B, Hasle H, Jahnukainen K, Juul‐Dam KL, et al. Characteristics and outcome of primary resistant disease in paediatric acute myeloid leukaemia. Br J Haematol. 2023;201(4):757–765. [DOI] [PubMed] [Google Scholar]
- 21. Miyamura T, Moritake H, Nakayama H, Tanaka S, Tomizawa D, Shiba N, et al. Clinical and biological features of paediatric acute myeloid leukaemia (AML) with primary induction failure in the Japanese Paediatric Leukaemia/Lymphoma Study Group AML‐05 study. Br J Haematol. 2019;185(2):284–288. [DOI] [PubMed] [Google Scholar]
- 22. Lachowiez CA, Ravikumar VI, Othman J, O'Nions J, Peters DT, Mcmahon C, et al. Refined ELN 2024 risk stratification improves survival prognostication following venetoclax‐based therapy in AML. Blood. 2024;144(26):2788–2792. [DOI] [PubMed] [Google Scholar]
- 23. Klein K, Van Litsenburg RRL, De Haas V, Dors N, Van Den Heuvel‐Eibrink MM, Knops RRG, et al. Causes of early death and treatment‐related death in newly diagnosed pediatric acute myeloid leukemia: recent experiences of the Dutch Childhood Oncology Group. Pediatr Blood Cancer. 2020;67(4):e28099. [DOI] [PubMed] [Google Scholar]
- 24. Bhatt NS, Baassiri MJ, Liu W, Bhakta N, Chemaitilly W, Ehrhardt MJ, et al. Late outcomes in survivors of childhood acute myeloid leukemia: a report from the St. Jude Lifetime Cohort Study. Leukemia. 2021;35(8):2258–2273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Tomizawa D, Tsujimoto SI. Risk‐stratified therapy for pediatric acute myeloid leukemia. Cancers (Basel). 2023;15(16):4171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Stone RM, Mandrekar SJ, Sanford BL, Laumann K, Geyer S, Bloomfield CD, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377(5):454–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Erba HP, Montesinos P, Kim H‐J, Patkowska E, Vrhovac R, Žák P, et al. Quizartinib plus chemotherapy in newly diagnosed patients with FLT3‐internal‐tandem‐duplication‐positive acute myeloid leukaemia (QuANTUM‐first): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet. 2023;401(10388):1571–1583. [DOI] [PubMed] [Google Scholar]
- 28. Levis MJ, Hamadani M, Logan B, Jones RJ, Singh AK, Litzow M, et al. Gilteritinib as post‐transplant maintenance for AML with internal tandem duplication mutation of FLT3. J Clin Oncol. 2024;42(15):1766–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Döhner H, Wei AH, Roboz GJ, et al. Prognostic impact of NPM1 and FLT3 mutations in patients with AML in first remission treated with oral azacitidine. Blood. 2022;140(15):1674–1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Brune M. Improved leukemia‐free survival after postconsolidation immunotherapy with histamine dihydrochloride and interleukin‐2 in acute myeloid leukemia: results of a randomized phase 3 trial. Blood. 2006;108(1):88–96. [DOI] [PubMed] [Google Scholar]
- 31. Pollard JA, Alonzo TA., Gerbing R, Brown P, Fox E, Choi J, et al. Sorafenib in combination with standard chemotherapy for children with high allelic ratio FLT3/ITD+ acute myeloid leukemia: a report from the children's oncology group protocol AAML1031. J Clin Oncol. 2022;40(18):2023–2035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kaspers GJL, Wijnen NE, Koedijk JB, Ishimaru S, Benedictus R, Van Opstal EC, et al. Chip‐AML22 master protocol: an open‐label clinical trial in newly diagnosed pediatric de novo acute myeloid leukemia (AML) patients including a linked phase II trial with quizartinib in FLT3‐ITD/NPM1wt patients–a study by the NOPHO‐DB‐SHIP consortium. Blood. 2023;142(Supp1):1532–1532. [Google Scholar]
- 33. Issa GC, Aldoss I, Thirman MJ, Dipersio J, Arellano M, Blachly JS, et al. Menin inhibition with revumenib for KMT2A‐rearranged relapsed or refractory acute leukemia (AUGMENT‐101). J Clin Oncol. 2025;43(1):75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Jimenez‐Chillon C, Othman J, Taussig D, Jimenez‐Vicente C, Martinez‐Roca A, Tiong IS, et al. Venetoclax‐based low intensity therapy in molecular failure of NPM1‐mutated AML. Blood Adv. 2024;8(2):343–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Platzbecker U, Middeke JM, Sockel K, Herbst R, Wolf D, Baldus CD, et al. Measurable residual disease‐guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open‐label, multicentre, phase 2 trial. Lancet Oncol. 2018;19(12):1668–1679. [DOI] [PubMed] [Google Scholar]
- 36. Potter N, Jovanovic J, Ivey A, Othman J, Thomas A, Gilkes A, et al. Molecular monitoring versus standard clinical care in younger adults with acute myeloid leukaemia: results from the UK NCRI AML17 and AML19 randomised, controlled, phase 3 trials. Lancet Haematol. 2025;12(5):e346–e356. [DOI] [PubMed] [Google Scholar]
- 37. Miles LA, Bowman RL, Merlinsky TR, Csete IS, Ooi AT, Durruthy‐Durruthy R, et al. Single‐cell mutation analysis of clonal evolution in myeloid malignancies. Nature. 2020;587(7834):477–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Schwede M, Jahn K, Kuipers J, Miles LA, Bowman RL, Robinson T, et al. Mutation order in acute myeloid leukemia identifies uncommon patterns of evolution and illuminates phenotypic heterogeneity. Leukemia. 2024;38(7):1501–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Leppä A‐M, Grimes K, Jeong H, Huang FY, Andrades A, Waclawiczek A, et al. Single‐cell multiomics analysis reveals dynamic clonal evolution and targetable phenotypes in acute myeloid leukemia with complex karyotype. Nat Genet. 2024;56(12):2790–2803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Moitra K, Lou H, Dean M. Multidrug efflux pumps and cancer stem cells: insights into multidrug resistance and therapeutic development. Clin Pharmacol Ther. 2011;89(4):491–502. [DOI] [PubMed] [Google Scholar]
- 41. Stelmach P, Trumpp A. Leukemic stem cells and therapy resistance in acute myeloid leukemia. Haematologica. 2023;108(2):353–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Gurion R, Belnik‐Plitman Y, Gafter‐Gvili A, et al. Colony‐stimulating factors for prevention and treatment of infectious complications in patients with acute myelogenous leukemia. Cochrane Database Syst Rev. 2012;2012(6):CD008238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Toksvang LN, Lee SHR, Yang JJ, Schmiegelow K. Maintenance therapy for acute lymphoblastic leukemia: basic science and clinical translations. Leukemia. 2022;36(7):1749–1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kaur V, Swami A, Alapat D, Abdallah AO, Motwani P, Hutchins LF, et al. Clinical characteristics, molecular profile and outcomes of myeloid sarcoma: a single institution experience over 13 years. Hematology. 2018;23(1):17–24. [DOI] [PubMed] [Google Scholar]
- 45. Goyal G, Bartley AC, Patnaik MM, Litzow MR, Al‐Kali A, Go RS. Clinical features and outcomes of extramedullary myeloid sarcoma in the United States: analysis using a national data set. In: Blood Cancer J. 2017;7(8):e592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Harris AC, Kitko CL, Couriel DR, Braun TM, Choi SW, Magenau J, et al. Extramedullary relapse of acute myeloid leukemia following allogeneic hematopoietic stem cell transplantation: incidence, risk factors and outcomes. Haematologica. 2013;98(2):179–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Bakst R, Wolden S, Yahalom J. Radiation therapy for chloroma (granulocytic sarcoma). Int J Radiat Oncol Biol Phys. 2012;82(5):1816–1822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Jian X, Cha J, Lin Z, Xie S, Huang Y, Lin Y, et al. Real‐world experience with venetoclax‐based therapy for patients with myeloid sarcoma. Discov Oncol. 2024;15(1):210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Duminuco A, Maugeri C, Parisi M, Mauro E, Fiumara PF, Randazzo V, et al. Target therapy for extramedullary relapse of FLT3‐ITD acute myeloid leukemia: emerging data from the field. Cancers (Basel). 2022;14(9):2186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Condorelli A, Matteo C, Leotta S, Schininà G, Sciortino R, Piccolo GM, et al. Venetoclax penetrates in cerebrospinal fluid of an acute myeloid leukemia patient with leptomeningeal involvement. Cancer Chemother Pharmacol. 2022;89(2):267–270. [DOI] [PubMed] [Google Scholar]
- 51. Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, et al. Ipilimumab for patients with relapse after allogeneic transplantation. N Engl J Med. 2016;375(2):143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Stengel A, Haferlach T, Baer C, Hutter S, Meggendorfer M, Kern W, et al. Specific subtype distribution with impact on prognosis of TP53 single‐hit and double‐hit events in AML and MDS. Blood Adv. 2023;7(13):2952–2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ferraro F, Gruszczynska A, Ruzinova MB, Miller CA, Percival ME, Uy GL, et al. Decitabine salvage for TP53‐mutated, relapsed/refractory acute myeloid leukemia after cytotoxic induction therapy. Haematologica. 2022;107(7):1709–1713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Thol F, Heuser M. Treatment for relapsed/refractory acute myeloid leukemia. Hemasphere. 2021;5(6):e572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. DeWolf S, Tallman MS. How I treat relapsed or refractory AML. Blood. 2020;136(9):1023–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Dinardo CD, Lachowiez CA, Takahashi K, Loghavi S, Xiao L, Kadia T, et al. Venetoclax combined with FLAG‐IDA induction and consolidation in newly diagnosed and relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2021;39(25):2768–2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Dinardo CD, Jen W‐Y, Takahashi K, Kadia TM, Loghavi S, Daver NG, et al. Long term results of venetoclax combined with FLAG‐IDA induction and consolidation for newly diagnosed and relapsed or refractory acute myeloid leukemia. Leukemia. 2025;39(4):854–863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Shahswar R, Beutel G, Klement P, Rehberg A, Gabdoulline R, Koenecke C, et al. FLA‐IDA salvage chemotherapy combined with a seven‐day course of venetoclax (FLAVIDA) in patients with relapsed/refractory acute leukaemia. Br J Haematol. 2020;188(3):e11–e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Karol SE, Alexander TB, Budhraja A, Pounds SB, Canavera K, Wang L, et al. Venetoclax in combination with cytarabine with or without idarubicin in children with relapsed or refractory acute myeloid leukaemia: a phase 1, dose‐escalation study. Lancet Oncol. 2020;21(4):551–560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Fleischhack G, Hasan C, Graf N, Mann G, Bode U. IDA‐FLAG (idarubicin, fludarabine, cytarabine, G‐CSF), an effective remission‐induction therapy for poor‐prognosis AML of childhood prior to allogeneic or autologous bone marrow transplantation: experiences of a phase II trial. Br J Haematol. 1998;102(3):647–655. [DOI] [PubMed] [Google Scholar]
- 61. Fong CY, Grigoriadis G, Hocking J, Coutsouvelis J, Muirhead J, Campbell P, et al. Fludarabine, cytarabine, granulocyte‐colony stimulating factor and amsacrine: an effective salvage therapy option for acute myeloid leukemia at first relapse. Leuk Lymphoma. 2013;54(2):336–341. [DOI] [PubMed] [Google Scholar]
- 62. Wrzesień‐Kuś A, Robak T, Wierzbowska A, et al. A multicenter, open, noncomparative, phase II study of the combination of cladribine (2‐chlorodeoxyadenosine), cytarabine, granulocyte colony‐stimulating factor and mitoxantrone as induction therapy in refractory acute myeloid leukemia: a report of the Polish Adult Leukemia Group. Ann Hematol. 2005;84(9):557–564. [DOI] [PubMed] [Google Scholar]
- 63. Halpern AB, Othus M, Huebner EM, Scott BL, Hendrie PC, Percival M‐EM, et al. Phase I/II trial of cladribine, high‐dose cytarabine, mitoxantrone, and G‐CSF with dose‐escalated mitoxantrone for relapsed/refractory acute myeloid leukemia and other high‐grade myeloid neoplasms. Haematologica. 2019;104(4):e143–e146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Marconi G, Talami A, Abbenante MC, Sartor C, Parisi S, Nanni J, et al. MEC (mitoxantrone, etoposide, and cytarabine) induces complete remission and is an effective bridge to transplant in acute myeloid leukemia. Eur J Haematol. 2020;105(1):47–55. [DOI] [PubMed] [Google Scholar]
- 65. Zhong S, Kurish H, Walchack R, Li H, Edwards J, Singh A, et al. Efficacy and safety of mitoxantrone, etoposide, and cytarabine for treatment of relapsed or refractory acute myeloid leukemia. Leuk Res. 2024;139:107468. [DOI] [PubMed] [Google Scholar]
- 66. Petit C, Higue J, Acheaibi Z, Gilhodes J, Hospital M‐A, Devillier R, et al. Venetoclax‐azacitidine versus azacitidine for the treatment of primary refractory or first relapsed acute myeloid leukemia. An IPC‐DATAML‐MSKCC retrospective study. Am J Hematol. 2025;100(5):906–908. [DOI] [PubMed] [Google Scholar]
- 67. Niswander LM, Chung P, Diorio C, Tasian SK. Clinical responses in pediatric patients with relapsed/refractory leukemia treated with azacitidine and venetoclax. Haematologica. 2023;108(11):3142–3147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Kadia TM, Reville PK, Wang X, Rausch CR, Borthakur G, Pemmaraju N, et al. Phase II study of venetoclax added to cladribine plus low‐dose cytarabine alternating with 5‐azacitidine in older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2022;40(33):3848–3857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Steinauer N, McCullough K, Al‐Kali A, et al. Cladribine plus cytarabine plus venetoclax in acute myeloid leukemia relapsed or refractory to venetoclax plus hypomethylating agent. Haematologica. 2024;109(8):2706–2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Li Y‐Y, Ge S‐S, Huang Y‐H, Xu M‐Z, Wan C‐L, Tan K‐W, et al. Efficacy and safety of cladribine, low‐dose cytarabine and venetoclax in relapsed/refractory acute myeloid leukemia: results of a pilot study. Blood Cancer J. 2024;14(1):12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Bawek S, Gurusinghe S, Burwinkel M, Przespolewski A. Updates in novel immunotherapeutic strategies for relapsed/refractory AML. Front Oncol. 2024;14:1374963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Daver N, Garcia‐Manero G, Basu S, Boddu PC, Alfayez M, Cortes JE, 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):370–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Gojo I, Stuart RK, Webster J, Blackford A, Varela JC, Morrow J, et al. Multi‐center phase 2 study of pembroluzimab (pembro) and azacitidine (AZA) in patients with relapsed/refractory acute myeloid leukemia (AML) and in newly diagnosed (≥65 Years) AML patients. Blood. 2019;134(Supp1):832–832. [Google Scholar]
- 74. Zeidner JF, Vincent BG, Ivanova A, Moore D, Mckinnon KP, Wilkinson AD, et al. Phase II trial of pembrolizumab after high‐dose cytarabine in relapsed/refractory acute myeloid leukemia. Blood Cancer Discov. 2021;2(6):616–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Perl AE, Martinelli G, Cortes JE, Neubauer A, Berman E, Paolini S, et al. Gilteritinib or chemotherapy for relapsed or refractory. N Engl J Med. 2019;381(18):1728–1740. [DOI] [PubMed] [Google Scholar]
- 76. Daver N, Perl AE, Maly J, Levis M, Ritchie E, Litzow M, et al. Venetoclax plus gilteritinib for FLT3‐mutated relapsed/refractory acute myeloid leukemia. J Clin Oncol. 2022;40(35):4048–4059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Short NJ, Daver N, Dinardo CD, Kadia T, Nasr LF, Macaron W, et al. Azacitidine, venetoclax, and gilteritinib in newly diagnosed and relapsed or refractory. J Clin Oncol. 2024;42(13):1499–1508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Montesinos P, Rodriguez‐Veiga R, Bergua Burgues JM, Algarra JL, Botella C, Rodriguez Arboli E, et al. Final results of Quiwi: a double blinded, randomized pethema trial comparing standard chemotherapy plus quizartinib versus placebo in adult patients with newly diagnosed FLT3‐ITD negative AML. Blood. 2024;144(Supp1):1512–1512. [Google Scholar]
- 79. Dinardo CD, Stein EM, De Botton S, Roboz GJ, Altman JK, Mims AS, et al. Durable remissions with ivosidenib in IDH1‐mutated relapsed or refractory AML. N Engl J Med. 2018;378(25):2386–2398. [DOI] [PubMed] [Google Scholar]
- 80. Stein EM, Dinardo CD, Fathi AT, Mims AS, Pratz KW, Savona MR, et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: a phase 1 study. Blood. 2021;137(13):1792–1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lachowiez CA, Loghavi S, Zeng Z, Tanaka T, Kim YJ, Uryu H, et al. A phase Ib/II study of ivosidenib with venetoclax ± azacitidine in IDH1‐mutated myeloid malignancies. Blood Cancer Discov. 2023;4(4):276–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Issa GC, Aldoss I, Dipersio J, Cuglievan B, Stone R, Arellano M, et al. The menin inhibitor revumenib in KMT2A‐rearranged or NPM1‐mutant leukaemia. Nature. 2023;615(7954):920–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Issa GC, Aldoss I, Thirman MJ, Dipersio J, Arellano M, Blachly JS, et al. Menin Inhibition With Revumenib for KMT2A‐rearranged relapsed or refractory acute leukemia (AUGMENT‐101). J Clin Oncol. 2025;43(1):75–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Wang ES, Issa GC, Erba HP, Altman JK, Montesinos P, Debotton S, et al. Ziftomenib in relapsed or refractory acute myeloid leukaemia (KOMET‐001): a multicentre, open‐label, multi‐cohort, phase 1 trial. Lancet Oncol. 2024;25(10):1310–1324. [DOI] [PubMed] [Google Scholar]
- 85. Herold N, Rudd SG, Sanjiv K, Kutzner J, Bladh J, Paulin CBJ, et al. SAMHD1 protects cancer cells from various nucleoside‐based antimetabolites. Cell Cycle. 2017;16(11):1029–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Herold N, Rudd SG, Ljungblad L, Sanjiv K, Myrberg IH, Paulin CBJ, et al. Targeting SAMHD1 with the Vpx protein to improve cytarabine therapy for hematological malignancies. Nat Med. 2017;23(2):256–263. [DOI] [PubMed] [Google Scholar]
- 87. Rassidakis GZ, Herold N, Myrberg IH, Tsesmetzis N, Rudd SG, Henter J‐I, et al. Low‐level expression of SAMHD1 in acute myeloid leukemia (AML) blasts correlates with improved outcome upon consolidation chemotherapy with high‐dose cytarabine‐based regimens. Blood Cancer J. 2018;8(11):98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Rudd SG, Tsesmetzis N, Sanjiv K, Paulin CBJ, Sandhow L, Kutzner J, et al. Ribonucleotide reductase inhibitors suppress SAMHD1 ara‐CTPase activity enhancing cytarabine efficacy. EMBO Mol Med. 2020;12:e10419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Jädersten M, Lilienthal I, Tsesmetzis N, Lourda M, Bengtzén S, Bohlin A, et al. Targeting SAMHD1 with hydroxyurea in first‐line cytarabine‐based therapy of newly diagnosed acute myeloid leukaemia: results from the HEAT‐AML trial. J Intern Med. 2022;292: 925–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Herold N. Pharmacological strategies to overcome treatment resistance in acute myeloid leukemia: increasing leukemic drug exposure by targeting the resistance factor SAMHD1 and the toxicity factor Top2β. Expert Opin Drug Discov. 2021;16(1):7–11. [DOI] [PubMed] [Google Scholar]
- 91. Schwartz CL, Wexler LH, Krailo MD, Teot LA, Devidas M, Steinherz LJ, et al. Intensified chemotherapy with dexrazoxane cardioprotection in newly diagnosed nonmetastatic osteosarcoma: a report from the Children's Oncology Group. Pediatr Blood Cancer. 2016;63(1):54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Malani D, Kumar A, Brück O, Kontro M, Yadav B, Hellesøy M, et al. Implementing a functional precision medicine tumor board for acute myeloid leukemia. Cancer Discov. 2022;12(2):388–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kornauth C, Pemovska T, Vladimer GI, Bayer G, Bergmann M, Eder S, et al. Functional precision medicine provides clinical benefit in advanced aggressive hematologic cancers and identifies exceptional responders. Cancer Discov. 2022;12(2):372–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Kuusanmäki H, Kytölä S, Vänttinen I, Ruokoranta T, Ranta A, Huuhtanen J, et al. Ex vivo venetoclax sensitivity testing predicts treatment response in acute myeloid leukemia. Haematologica. 2023;108(7):1768–1781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Kytölä S, Vänttinen IM, Ruokoranta T, et al. Ex Vivo venetoclax sensitivity predicts clinical response in acute myeloid leukemia in the prospective venex trial. Blood. 2025;145(4):409–421. [DOI] [PubMed] [Google Scholar]
- 96. Craddock C, Labopin M, Robin M, Finke J, Chevallier P, Yakoub‐Agha I, et al. Clinical activity of azacitidine in patients who relapse after allogeneic stem cell transplantation for acute myeloid leukemia. Haematologica. 2016;101(7):879–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Burchert A, Bug G, Fritz LV, Finke J, Stelljes M, Röllig C, et al. Sorafenib maintenance after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia with. J Clin Oncol. 2020;38(26):2993–3002. [DOI] [PubMed] [Google Scholar]
- 98. Fathi AT, Kim HT, Soiffer RJ, Levis MJ, Li S, Kim AS, et al. Multicenter phase I trial of ivosidenib as maintenance treatment following allogeneic hematopoietic cell transplantation for IDH1‐mutated acute myeloid leukemia. Clin Cancer Res. 2023;29(11):2034–2042. [DOI] [PubMed] [Google Scholar]
- 99. Megías‐Vericat JE, Martínez‐Cuadrón D, Sanz MÁ, Montesinos P. Salvage regimens using conventional chemotherapy agents for relapsed/refractory adult AML patients: a systematic literature review. Ann Hematol. 2018;97(7):1115–1153. [DOI] [PubMed] [Google Scholar]
- 100. Gordon MS, Young ML, Tallman MS, Cripe LD, Bennett JM, Paietta E, et al. Phase II trial of 2‐chlorodeoxyadenosine in patients with relapsed/refractory acute myeloid leukemia: a study of the Eastern Cooperative Oncology Group (ECOG), E5995. Leuk Res. 2000;24(10):871–875. [DOI] [PubMed] [Google Scholar]
- 101. Kantarjian H, Gandhi V, Cortes J, Verstovsek S, Du M, Garcia‐Manero G, et al. Phase 2 clinical and pharmacologic study of clofarabine in patients with refractory or relapsed acute leukemia. Blood. 2003;102(7):2379–2386. [DOI] [PubMed] [Google Scholar]
- 102. Jeha S, Razzouk B, Rytting M, Rheingold S, Albano E, Kadota R, et al. Phase II study of clofarabine in pediatric patients with refractory or relapsed acute myeloid leukemia. J Clin Oncol. 2009;27(26):4392–4397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Roberts DA, Wadleigh M, McDonnell AM, DeAngelo DJ, Stone RM, Steensma DP. Low efficacy and high mortality associated with clofarabine treatment of relapsed/refractory acute myeloid leukemia and myelodysplastic syndromes. Leuk Res. 2015;39(2):204–210. [DOI] [PubMed] [Google Scholar]
- 104. Van Den Neste E, Martiat P, Mineur P, Delannoy A, Doyen C, Zenebergh A, et al. 2‐Chlorodeoxyadenosine with or without daunorubicin in relapsed or refractory acute myeloid leukemia. Ann Hematol. 1998;76(1):19–23. [DOI] [PubMed] [Google Scholar]
- 105. Faderl S, Ferrajoli A, Wierda W, Huang X, Verstovsek S, Ravandi F, et al. Clofarabine combinations as acute myeloid leukemia salvage therapy. Cancer. 2008;113(8):2090–2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Hargrave RM, Davey MW, Davey RA, Kidman AD. Development of drug resistance is reduced with idarubicin relative to other anthracyclines. Anticancer Drugs. 1995;6(3):432–437. [DOI] [PubMed] [Google Scholar]
- 107. Pogliani EM, Carpenedo M, Miccolis I, Belotti D, Corneo GM. P‐glycoprotein expression in acute myeloid leukaemia cells at diagnosis: its relationship to daunorubicin or idarubicin induction therapy and survival; malignancy. Hematology. 2001;5(5):359–367. [PubMed] [Google Scholar]
- 108. Kern W, Aul C, Maschmeyer G, Schönrock‐Nabulsi R, Ludwig WD, Bartholomäus A, et al. Superiority of high‐dose over intermediate‐dose cytosine arabinoside in the treatment of patients with high‐risk acute myeloid leukemia: results of an age‐adjusted prospective randomized comparison. Leukemia. 1998;12(7):1049–1055. [DOI] [PubMed] [Google Scholar]
- 109. Hiddemann W, Kreutzmann H, Straif K, Ludwig WD, Mertelsmann R, Donhuijsen‐ Ant R, et al. High‐dose cytosine arabinoside and mitoxantrone: a highly effective regimen in refractory acute myeloid leukemia. Blood. 1987;69(3):744–749. [PubMed] [Google Scholar]
- 110. Wattad M, Weber D, Döhner K, Krauter J, Gaidzik VI, Paschka P, et al. Impact of salvage regimens on response and overall survival in acute myeloid leukemia with induction failure. Leukemia. 2017;31(6):1306–1313. [DOI] [PubMed] [Google Scholar]
- 111. Thomas X, Cambier N, Taksin AL, Reman O, Vekhoff A, Pautas C, et al. Dose‐escalation study of single dose mitoxantrone in combination with timed sequential chemotherapy in patients with refractory or relapsing acute myelogenous leukemia. Leuk Res. 2000;24(11):957–963. [DOI] [PubMed] [Google Scholar]
- 112. Cortes J, Estey E, O'Brien S, Giles F, Shen Y, Koller C, et al. High‐dose liposomal daunorubicin and high‐dose cytarabine combination in patients with refractory or relapsed acute myelogenous leukemia. Cancer. 2001;92(1):7–14. [DOI] [PubMed] [Google Scholar]
- 113. Burnett AK., Russell NH., Hills RK., Kell J, Cavenagh J, Kjeldsen L, et al. A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients. Blood. 2015;125(25):3878–3885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM, et al. Anthracycline dose intensification in acute myeloid leukemia. N Engl J Med. 2009;361(13):1249–1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Yamauchi T, Kawai Y, Kishi S, Goto N, Urasaki Y, Imamura S, et al. Monitoring of intracellular 1‐beta‐D‐arabinofuranosylcytosine 5'‐triphosphate in 1‐beta‐D‐arabinofuranosylcytosine therapy at low and conventional doses. Jpn J Cancer Res. 2001;92(5):546–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Plunkett W, Iacoboni S, Estey E, Danhauser L, Liliemark JO, Keating MJ. Pharmacologically directed ara‐C therapy for refractory leukemia. Semin Oncol. 1985;12(2 Supp 3):20–30. [PubMed] [Google Scholar]
- 117. Riva‐Lavieille C, Ressayre C, Barra Y, Carcassonne Y, Iliadis A. Calculation of individual dosage regimen of cytosine arabinoside (ara‐C) based on metabolite levels in leukemic cells. Ther Drug Monit. 1994;16(4):375–379. [DOI] [PubMed] [Google Scholar]
- 118. Gandhi V, Estey E, Keating MJ, Plunkett W. Fludarabine potentiates metabolism of cytarabine in patients with acute myelogenous leukemia during therapy. J Clin Oncol. 1993;11(1):116–124. [DOI] [PubMed] [Google Scholar]
- 119. Ossenkoppele GJ. The value of fludarabine in addition to ARA‐C and G‐CSF in the treatment of patients with high‐risk myelodysplastic syndromes and AML in elderly patients. Blood. 2004;103(8):2908–2913. [DOI] [PubMed] [Google Scholar]
- 120. Estey E, Plunkett W, Gandhi V, Rios MB, Kantarjian H, Keating MJ. Fludarabine and arabinosylcytosine therapy of refractory and relapsed acute myelogenous leukemia. Leuk Lymphoma. 1993;9(4–5):343–350. [DOI] [PubMed] [Google Scholar]
- 121. Kaspers GJL, Zimmermann M, Reinhardt D, Gibson BES, Tamminga RYJ, Aleinikova O, et al. Improved outcome in pediatric relapsed acute myeloid leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group. J Clin Oncol. 2013;31(5):599–607. [DOI] [PubMed] [Google Scholar]
- 122. Fiegl M, Unterhalt M, Kern W, Braess J, Spiekermann K, Staib P, et al. Chemomodulation of sequential high‐dose cytarabine by fludarabine in relapsed or refractory acute myeloid leukemia: a randomized trial of the AMLCG. Leukemia. 2014;28(5):1001–1007. [DOI] [PubMed] [Google Scholar]
- 123. Holowiecki J, Grosicki S, Giebel S, Robak T, Kyrcz‐Krzemien S, Kuliczkowski K, et al. Cladribine, but not fludarabine, added to daunorubicin and cytarabine during induction prolongs survival of patients with acute myeloid leukemia: a multicenter, randomized phase III study. J Clin Oncol. 2012;30(20):2441–2448. [DOI] [PubMed] [Google Scholar]
- 124. Kornblau SM, Gandhi V, Andreeff HM, et al. Clinical and laboratory studies of 2‐chlorodeoxyadenosine ± cytosine arabinoside for relapsed or refractory acute myelogenous leukemia in adults. Leukemia. 1996;10(10):1563–1569. [PubMed] [Google Scholar]
- 125. Wrzesień‐Kuś A, Robak T, Lech‐Marańda E, et al. A multicenter, open, non‐comparative, phase II study of the combination of cladribine (2‐chlorodeoxyadenosine), cytarabine, and G‐CSF as induction therapy in refractory acute myeloid leukemia ‐ a report of the Polish Adult Leukemia Group (PALG). Eur J Haematol. 2003;71(3):155–162. [DOI] [PubMed] [Google Scholar]
- 126. Rubnitz JE, Razzouk BI, Srivastava DK, Pui CH, Ribeiro RC, Santana VM. Phase II trial of cladribine and cytarabine in relapsed or refractory myeloid malignancies. Leuk Res. 2004;28(4):349–352. [DOI] [PubMed] [Google Scholar]
- 127. Gill H, Yim R, Pang HH, Lee P, Chan TSY, Hwang Y‐Y, et al. Clofarabine, cytarabine, and mitoxantrone in refractory/relapsed acute myeloid leukemia: high response rates and effective bridge to allogeneic hematopoietic stem cell transplantation. Cancer Med. 2020;9(10):3371–3382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Short NJ, Kantarjian H, Ravandi F, Huang X, Xiao L, Garcia‐Manero G, et al. A phase I/II randomized trial of clofarabine or fludarabine added to idarubicin and cytarabine for adults with relapsed or refractory acute myeloid leukemia. Leuk Lymphoma. 2018;59(4):813–820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Nazha A, Kantarjian H, Ravandi F, Huang X, Choi S, Garcia‐Manero G, et al. Clofarabine, idarubicin, and cytarabine (CIA) as frontline therapy for patients ≤60 years with newly diagnosed acute myeloid leukemia. Am J Hematol. 2013;88(11):961–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Van Eijkelenburg NKA, Rasche M, Ghazaly E, Dworzak MN, Klingebiel T, Rossig C, et al. Clofarabine, high‐dose cytarabine and liposomal daunorubicin in pediatric relapsed/refractory acute myeloid leukemia: a phase IB study. Haematologica. 2018;103(9):1484–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Feijen EAM, Leisenring WM, Stratton KL, Ness KK, Van Der Pal HJH, Van Dalen EC, et al. Derivation of anthracycline and anthraquinone equivalence ratios to doxorubicin for late‐onset cardiotoxicity. JAMA Oncol. 2019;5(6):864–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Fortin MC, LaCroix AS, Grammatopoulos TN, Tan L, Wang Q, Manca D. Lower cardiotoxicity of CPX‐351 relative to daunorubicin plus cytarabine free‐drug combination in hiPSC‐derived cardiomyocytes in vitro. Sci Rep. 2023;13(1):21054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Othman J, Wilhelm‐Benartzi C, Dillon R, Knapper S, Freeman SD, Batten LM, et al. A randomized comparison of CPX‐351 and FLAG‐Ida in adverse karyotype AML and high‐risk MDS: the UK NCRI AML19 trial. Blood Adv. 2023;7(16):4539–4549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Leger KJ, Absalon MJ, Demissei BG, Smith AM, Gerbing RB, Alonzo TA, et al. Cardiotoxicity of CPX‐351 in children and adolescents with relapsed AML: a Children's Oncology Group report. Front Cardiovasc Med. 2024;11:1347547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Cortes JE, Goldberg SL, Feldman EJ, Rizzeri DA, Hogge DE, Larson M, et al. Phase II, multicenter, randomized trial of CPX‐351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121(2):234–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Pollard JA, Alonzo TA, Gerbing R, Kutny MA, Hirsch B, Raca G, et al. A Phase 3 randomized trial for patients with de novo AML comparing standard therapy including gemtuzumab ozogamicin (GO) to CPX‐351 with GO—a report from the Children's Oncology Group. Blood. 2024;144(Supp1):967–967. [Google Scholar]
- 137. Czabotar PE, Garcia‐Saez AJ. Mechanisms of BCL‐2 family proteins in mitochondrial apoptosis. Nat Rev Mol Cell Biol. 2023;24(10):732–748. [DOI] [PubMed] [Google Scholar]
- 138. Wei AH, Roberts AW. BCL2 inhibition: a new paradigm for the treatment of AML and Beyond. Hemasphere. 2023;7(6):e912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Pollyea DA, Stevens BM, Jones CL, Winters A, Pei S, Minhajuddin M, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24(12):1859–1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Wei AH, Montesinos P, Ivanov V, Dinardo CD, Novak J, Laribi K, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo‐controlled trial. Blood. 2020;135(24):2137–2145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Dinardo CD, Tiong IS, Quaglieri A, Macraild S, Loghavi S, Brown FC, et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood. 2020;135(11):791–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Zhang Qi, Riley‐Gillis B, Han L, Jia Y, Lodi A, Zhang H, et al. Activation of RAS/MAPK pathway confers MCL‐1 mediated acquired resistance to BCL‐2 inhibitor venetoclax in acute myeloid leukemia. Signal Transduct Target Ther. 2022;7(1):51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Thijssen R, Diepstraten ST, Moujalled D, Chew E, Flensburg C, Shi MX, et al. Intact TP‐53 function is essential for sustaining durable responses to BH3‐mimetic drugs in leukemias. Blood. 2021;137(20):2721–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Blombery P, Anderson MA, Gong J‐N, Thijssen R, Birkinshaw RW, Thompson ER, et al. Acquisition of the recurrent Gly101Val mutation in BCL2 confers resistance to venetoclax in patients with progressive chronic lymphocytic leukemia. Cancer Discov. 2019;9(3):342–353. [DOI] [PubMed] [Google Scholar]
- 145. Moujalled DM, Brown FC, Chua CC, Dengler MA, Pomilio G, Anstee NS, et al. Acquired mutations in BAX confer resistance to BH3‐mimetic therapy in acute myeloid leukemia. Blood. 2023;141(6):634–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu R, et al. Monocytic subclones confer resistance to venetoclax‐based therapy in patients with acute myeloid leukemia. Cancer Discov. 2020;10(4):536–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Kuusanmäki H, Dufva O, Vähä‐Koskela M, Leppä A‐M, Huuhtanen J, Vänttinen I, et al. Erythroid/megakaryocytic differentiation confers BCL‐XL dependency and venetoclax resistance in acute myeloid leukemia. Blood. 2023;141(13):1610–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Bruzzese A, Martino EA, Labanca C, Mendicino F, Lucia E, Olivito V, et al. Potential of BGB‐11417, a BCL2 inhibitor, in hematological malignancies. Expert Opin Investig Drugs. 2024;33(2):73–77. [DOI] [PubMed] [Google Scholar]
- 149. Pei S, Shelton IT, Gillen AE, Stevens BM, Gasparetto M, Wang Y, et al. A novel type of monocytic leukemia stem cell revealed by the clinical use of venetoclax‐based Therapy. Cancer Discov. 2023;13(9):2032–2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Döhner H, DiNardo CD, Appelbaum FR, et al. Genetic risk classification for adults with AML receiving less‐intensive therapies: the 2024 ELN recommendations. Blood. 2024;144(21):2169–2173. [DOI] [PubMed] [Google Scholar]
- 151. Chua CC, Loo S, Fong CY, Ting SB, Tiong IS, Fleming S, et al. Final analysis of the phase 1b chemotherapy and venetoclax in elderly acute myeloid leukemia trial (CAVEAT). Blood Adv. 2025;9(8):1827–1835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Zhang Y, Yin Z, Yao Z, Xu D, Jiang X, Nie X, et al. Venetoclax added to CLAG regimen might improve the outcome of patients with relapsed/refractory acute myeloid leukemia. Ther Adv Hematol. 2025;16:20406207251319603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Ambinder AJ, Levis M. Potential targeting of FLT3 acute myeloid leukemia. Haematologica. 2021;106(3):671–681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Janssen M, Schmidt C, Bruch P‐M, Blank MF, Rohde C, Waclawiczek A, et al. Venetoclax synergizes with gilteritinib in FLT3 wild‐type high‐risk acute myeloid leukemia by suppressing MCL‐1. Blood. 2022;140(24):2594–2610. [DOI] [PubMed] [Google Scholar]
- 155. Röllig C, Serve H, Hüttmann A, Noppeney R, Müller‐Tidow C, Krug U, et al. Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial. Lancet Oncol. 2015;16(16):1691–1699. [DOI] [PubMed] [Google Scholar]
- 156. Montesinos P, Cheong J‐W, Daver N, et al. Trial in progress: the phase 3, randomized, double‐blind, placebo‐controlled QuANTUM‐wild study of quizartinib in combination with chemotherapy and as single‐agent maintenance in newly diagnosed, FLT3‐ITD‐negative acute myeloid leukemia. Blood. 2024;144(Supp1):15043. [Google Scholar]
- 157. Figueroa ME, Abdel‐Wahab O, Lu C, Ward PS, Patel J, Shih A, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18(6):553–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Chan SM, Thomas D, Corces‐Zimmerman MR, Xavy S, Rastogi S, Hong W‐J, et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL‐2 dependence in acute myeloid leukemia. Nat Med. 2015;21(2):178–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Montesinos P, Recher C, Vives S, Zarzycka E, Wang J, Bertani G, et al. Ivosidenib and azacitidine in. N Engl J Med. 2022;386(16):1519–1531. [DOI] [PubMed] [Google Scholar]
- 160. Libura M, Bialopiotrowicz E, Giebel S, Wierzbowska A, Roboz GJ, Piatkowska‐Jakubas B, et al. IDH2 mutations in patients with normal karyotype AML predict favorable responses to daunorubicin, cytarabine and cladribine regimen. Sci Rep. 2021;11(1):10017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Juul‐Dam KL, Shukla NN, Cooper TM, Cuglievan B, Heidenreich O, Kolb AE, et al. Therapeutic targeting in pediatric acute myeloid leukemia with aberrant HOX/MEIS1 expression. Eur J Med Genet. 2023;66(12):104869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Yokoyama A, Somervaille TC, Smith KS, Rozenblatt‐Rosen O, Meyerson M, Cleary ML. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL‐associated leukemogenesis. Cell. 2005;123(2):207–218. [DOI] [PubMed] [Google Scholar]
- 163. Armstrong SA, Staunton JE, Silverman LB, Pieters R, Den Boer ML, Minden MD, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat Genet. 2002;30(1):41–47. [DOI] [PubMed] [Google Scholar]
- 164. Xu H, Valerio DG, Eisold ME, Sinha A, Koche RP, Hu W, et al. NUP98 fusion proteins interact with the NSL and MLL1 complexes to drive leukemogenesis. Cancer Cell. 2016;30(6):863–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Kühn MW, Song E, Feng Z, Sinha A, Chen CW, Deshpande AJ, et al. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016;6(10):1166–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Barajas JM, Rasouli M, Umeda M, Hiltenbrand R, Abdelhamed S, Mohnani R, et al. Acute myeloid leukemias with UBTF tandem duplications are sensitive to menin inhibitors. Blood. 2024;143(7):619–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Van Weelderen RE, Klein K, Harrison CJ, Jiang Y, Abrahamsson J, Arad‐Cohen N, et al. Measurable residual disease and fusion partner independently predict survival and relapse risk in childhood KMT2A‐rearranged acute myeloid leukemia: a study by the International Berlin‐Frankfurt‐Münster Study Group. J Clin Oncol. 2023;41(16):2963–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Grembecka J, He S, Shi A, Purohit T, Muntean AG, Sorenson RJ, et al. Menin‐MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat Chem Biol. 2012;8(3):277–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Krivtsov AV, Evans K, Gadrey JY, Eschle BK, Hatton C, Uckelmann HJ, et al. A menin‐MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL‐rearranged leukemia. Cancer Cell. 2019;36(6):660–673.e611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Heikamp EB, Henrich JA, Perner F, Wong EM, Hatton C, Wen Y, et al. The menin‐MLL1 interaction is a molecular dependency in NUP98‐rearranged AML. Blood. 2022;139(6):894–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Searle E, Recher C, Abdul‐Hay M, Abedin S, Aldoss I, Pierola AA, et al. Bleximenib dose optimization and determination of RP2D from a phase 1 study in relapsed/refractory acute leukemia patients with KMT2A and NPM1 Alterations. Blood. 2024;144(Supp1):212–212. [Google Scholar]
- 172. Zeidner JF, Yuda J, Watts JM, Levis MJ, Erba HP, Fukushima K, et al. Phase 1 results: first‐in‐human phase 1/2 study of the menin‐MLL inhibitor enzomenib (DSP‐5336) in patients with relapsed or refractory acute leukemia. Blood. 2024;144(Supp1):213–213. [Google Scholar]
- 173. Perner F, Stein EM, Wenge DV, Singh S, Kim J, Apazidis A, et al. MEN1 mutations mediate clinical resistance to menin inhibition. Nature. 2023;615(7954):913–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Rücker FG, Schlenk RF, Bullinger L, Kayser S, Teleanu V, Kett H, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119(9):2114–2121. [DOI] [PubMed] [Google Scholar]
- 175. Bowen D, Groves MJ, Burnett AK, Patel Y, Allen C, Green C, et al. TP53 gene mutation is frequent in patients with acute myeloid leukemia and complex karyotype, and is associated with very poor prognosis. Leukemia. 2009;23(1):203–206. [DOI] [PubMed] [Google Scholar]
- 176. Ciurea SO, Chilkulwar A, Saliba RM, Chen J, Rondon G, Patel KP, et al. Prognostic factors influencing survival after allogeneic transplantation for AML/MDS patients with. Blood. 2018;131(26):2989–2992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Alwash Y, Khoury JD, Tashakori M, Kanagal‐Shamanna R, Daver N, Ravandi F, et al. Development of TP53 mutations over the course of therapy for acute myeloid leukemia. Am J Hematol. 2021;96(11):1420–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Sallman DA. To target the untargetable: elucidation of synergy of APR‐246 and azacitidine in TP53 mutant myelodysplastic syndromes and acute myeloid leukemia. Haematologica. 2020;105(6):1470–1472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Cluzeau T, Sebert M, Rahmé R, Cuzzubbo S, Lehmann‐Che J, Madelaine I, 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):1575–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Sallman DA, Dezern AE, Garcia‐Manero G, Steensma DP, Roboz GJ, Sekeres MA, et al. Eprenetapopt (APR‐246) and azacitidine in TP53‐mutant myelodysplastic syndromes. J Clin Oncol. 2021;39(14):1584–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Mishra A, Tamari R, Dezern AE, Byrne MT, Gooptu M, Chen Y‐B, et al. Eprenetapopt plus azacitidine after allogeneic hematopoietic stem‐cell transplantation for TP53‐mutant acute myeloid leukemia and myelodysplastic syndromes. J Clin Oncol. 2022;40(34):3985–3993. [DOI] [PubMed] [Google Scholar]
- 182. Daver NG, Dail M, Garcia JS, Jonas BA, Yee KWL, Kelly KR, et al. Venetoclax and idasanutlin in relapsed/refractory AML: a nonrandomized, open‐label phase 1b trial. Blood. 2023;141(11):1265–1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Dinardo CD, Rosenthal J, Andreeff M, Zernovak O, Kumar P, Gajee R, et al. Phase 1 dose escalation study of MDM2 inhibitor DS‐3032b in patients with hematological malignancies–preliminary results. Blood. 2016;128(22):593–593. [Google Scholar]
- 184. Senapati J, Muftuoglu M, Ishizawa J, Abbas HA, Loghavi S, Borthakur G, et al. A phase I study of milademetan (DS3032b) in combination with low dose cytarabine with or without venetoclax in acute myeloid leukemia: clinical safety, efficacy, and correlative analysis. Blood Cancer J. 2023;13(1):101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Hourigan CS, Karp JE. Personalized therapy for acute myeloid leukemia. Cancer Discov. 2013;3(12):1336–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Liebers N, Bruch P‐M, Terzer T, Hernandez‐Hernandez M, Paramasivam N, Fitzgerald D, et al. Ex vivo drug response profiling for response and outcome prediction in hematologic malignancies: the prospective non‐interventional SMARTrial. Nat Cancer. 2023;4(12):1648–1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Snijder B, Vladimer GI, Krall N, Miura K, Schmolke A‐S, Kornauth C, et al. Image‐based ex‐vivo drug screening for patients with aggressive haematological malignancies: interim results from a single‐arm, open‐label, pilot study. Lancet Haematol. 2017;4(12):e595–e606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Kuusanmäki H, Leppä A‐M, Pölönen P, Kontro M, Dufva O, Deb D, et al. Phenotype‐based drug screening reveals association between venetoclax response and differentiation stage in acute myeloid leukemia. Haematologica. 2020;105(3):708–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Hassan N, Yang J, Wang JY. An improved protocol for establishment of AML patient‐derived xenograft models. STAR Protoc. 2020;1(3):100156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Kawashima N, Ishikawa Y, Kim JH, Ushijima Y, Akashi A, Yamaguchi Y, et al. Comparison of clonal architecture between primary and immunodeficient mouse‐engrafted acute myeloid leukemia cells. Nat Commun. 2022;13(1):1624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Collados‐Ros A, Muro M, Legaz I. Gemtuzumab ozogamicin in acute myeloid leukemia: efficacy, toxicity, and resistance mechanisms‐A systematic review. Biomedicines. 2024;12(1):208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Fultang L, Panetti S, Ng M, Collins P, Graef S, Rizkalla N, et al. MDSC targeting with gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. eBioMedicine. 2019;47:235–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Daver NG, Vyas P, Kambhampati S, Al Malki MM, Larson RA, Asch AS, et al. Tolerability and efficacy of the anticluster of differentiation 47 antibody magrolimab combined with azacitidine in patients with previously untreated AML: phase Ib results. J Clin Oncol. 2023;41(31):4893–4904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Yang H, Xun Y, You H. The landscape overview of CD47‐based immunotherapy for hematological malignancies. Biomark Res. 2023;11(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Deng H, Wang Q, Tong X, Cui Z, Yang Y, Xiang Y. Recent advances of CAR‐T cells in acute myeloid leukemia. Ther Adv Hematol. 2025;16:20406207251326802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Christopher MJ, Petti AA, Rettig MP, Miller CA, Chendamarai E, Duncavage EJ, et al. Immune escape of relapsed AML cells after allogeneic transplantation. N Engl J Med. 2018;379(24):2330–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]