Skip to main content
HemaSphere logoLink to HemaSphere
. 2023 May 26;7(6):e914. doi: 10.1097/HS9.0000000000000914

An Overview of Targeted Therapies in Acute Myeloid Leukemia

Sven Turkalj 1,2, Felix A Radtke 1,2,3, Paresh Vyas 1,2,4,
PMCID: PMC10256410  PMID: 37304938

Abstract

Acute myeloid leukemia (AML) is the most aggressive adult leukemia, characterized by clonal differentiation arrest of progenitor or precursor hematopoietic cells. Intense preclinical and clinical research has led to regulatory approval of several targeted therapeutics, administered either as single agents or as combination therapies. However, the majority of patients still face a poor prognosis and disease relapse frequently occurs due to selection of therapy-resistant clones. Hence, more effective novel therapies, most likely as innovative, rational combination therapies, are urgently needed. Chromosomal aberrations, gene mutations, and epigenetic alterations drive AML pathogenesis but concurrently provide vulnerabilities to specifically target leukemic cells. Other molecules, either aberrantly active and/or overexpressed in leukemic stem cells, may also be leveraged for therapeutic benefit. This concise review of targeted therapies for AML treatment, which are either approved or are being actively investigated in clinical trials or recent preclinical studies, provides a flavor of the direction of travel, but also highlights the current challenges in AML treatment.


Acute myeloid leukemia (AML) is the most aggressive adult acute leukemia, which still has a high unmet clinical need. Although treatment outcomes have improved, and multiple new drugs have been licensed over the last few decades, most patients still face a dismal prognosis. Approximately 70% of AML patients are above the age of 65 years and are mostly treated with nonintensive chemotherapy. The current standard of care for this patient group is a combination of the BCL2 inhibitor venetoclax and the hypomethylating agent (HMA) azacitidine. The median survival is 14.5 months, and patients are usually not cured. The remaining 30% of patients, usually aged <60 years, are treated with >1 course of intensive combination chemotherapy, potentially combined with targeted therapies. Patients at high risk of treatment failure receive allogeneic stem cell transplantation (allo-SCT). Although the cure rates for this group are ~30%–70%, the majority of intensively treated patients still die of their disease.

A significant advance in AML biology and treatment has been the definition of the genetic and cytogenetic landscape of the disease.1,2 This has provided genetic markers for diagnostic and prognostic purposes, as well as markers used to follow the depth of therapy response (also known as measurement of residual disease). In some cases, the discovery of genetic markers has led to the development of specific therapies targeting mutant proteins essential for AML propagation.

Therapeutic targets in AML can be broadly classified into 6 categories (Table 1; Figure 1).

Table 1.

Therapeutic Targets in AML

Type of Target Targets
Genetic Targets Fusion Oncoproteins, FLT3, IDH1/2, p53, KIT
Targets involved in apoptosis BCL2, MCL-1, BCL-XL, MDM2
Signaling molecules or nuclear receptors RAS pathway, SYK, RARα
Epigenetic regulators Menin, DOT1L
Surface proteins and/or molecules involved in immune signaling Smoothened, CD33, CD123, CD47, SIRPα, AXL, TIM-3, CLL-1, PD-1/PD-L1, and CTLA-4 axes
Transcription factors and other molecules subject to targeted protein degradation EZH2, GSPT1

Figure 1.

Figure 1.

Overview of therapeutic agents and targeted pathways in AML. Schematic overview of targeted molecules found either on the AML cell surface or within the cytoplasm, nucleus, or mitochondria of AML cells. The molecules used as therapeutic agents are labeled in red. Targeted molecules are labeled in black. Cellular mechanisms involved or targeted molecule expression patterns are labeled in green. AML = acute myeloid leukemia.

FUSION PROTEINS: THE PARADIGM FOR TARGETED THERAPY IN MYELOID MALIGNANCIES

Recurrent chromosomal aberrations can generate fusion proteins, which cause differentiation block and sustain malignant transformation, but may also create a critical therapeutic vulnerability of leukemic cells.3 The fusion protein stemming from the Philadelphia chromosome in chronic myelogenous leukemia (CML),4 which generates the BCR-ABL1 oncogene with constitutive tyrosine kinase activity, is the exemplar of a targetable fusion protein in myeloid malignancy. Imatinib,5 a specific inhibitor of the BCR-ABL1 kinase domain,6 transformed the care of CML patients and introduced the concept of targeting products of somatic alteration in cancer. Targeted therapies have also revolutionized the treatment of acute promyelocytic leukemia (APL), caused by the promyelocytic leukemia-retinoic acid receptor alpha (PML-RARA) fusion oncoprotein,7,8 which blocks granulocytic differentiation.9 Patients now routinely receive all-trans retinoic acid (ATRA), which reverses transcriptional repression imposed by PML-RARA, thus promoting granulocyte differentiation. ATRA is combined with arsenic trioxide (ATO), which degrades PML-RARA, removing the oncogenic protein that sustains APL. A chemotherapy-free regimen combining ATRA and ATO has led to >90% overall survival (OS).10,11 This approach highlighted the potential for differentiation therapy in leukemia12 and demonstrated the importance of combining targeted therapies with synergistic mechanisms of action.13

The therapeutic success in CML and APL has been harder to replicate for other oncogenic fusion proteins, especially those involving transcription factors. One approach to developing targeted therapy for AML driven by transcription factor fusion proteins is to target pathways downstream of these transcription factors. An example is the inhibition of Menin,14 a required coactivator for MLL (KMT2A) fusion proteins. Menin and oncogenic MLL fusion proteins coordinately activate the transcription of HOX family members and MEIS1.15,16 These observations, coupled with preclinical data showing promising activity of Menin inhibition,1719 prompted first-in-human clinical trials of multiple Menin inhibitors, namely revumenib,20,21 ziftomenib,22 and JNJ-6617 (NCT04811560), in MLL-rearranged and NPM1-mutated leukemia. As one may expect, second-site mutations in the MEN1 gene, encoding Menin, confer resistance to revumenib.21 MLL fusions also interact with the histone methyltransferase DOT1L, required for aberrant activation of target genes.23 In a phase I study, the DOT1L inhibitor pinometostat showed limited single-agent activity in MLL-rearranged adult AML.24 Pinometostat is now being evaluated in combination with azacitidine.25

GENETIC TARGETS RESULTING FROM SOMATIC MUTATION AND OTHER ABERRANTLY ACTIVE TARGETS

Driver somatic mutations, which produce mutated proteins imparting a fitness advantage to AML cells, also provide an opportunity to develop targeted therapies (Table 2). For example, gain-of-function mutations in the FLT3 gene, encoding a transmembrane receptor tyrosine kinase (RTK), are found in ~30% of younger AML patients and at lower frequency in older patients. Most FLT3 mutations occur as internal tandem duplications (FLT3-ITD) within the gene locus encoding the juxtamembrane domain or as point mutations within the locus encoding the tyrosine kinase domain activation loop (FLT3-TK). Mutant FLT3 proteins transmit a constitutively active ligand-independent signal to aberrantly activate prosurvival and proproliferative RAS/MAPK, PI3K/AKT, and JAK/STAT pathways.26 Type I FLT3 inhibitors, such as midostaurin, gilteritinib, and crenolanib, bind the kinase active conformation, inhibiting both FLT3-ITD and FLT3-TK proteins. Type II FLT3 inhibitors, including quizartinib and sorafenib, bind the kinase-inactive conformation and are only active against FLT3-ITD. These inhibitors decrease FLT3 autophosphorylation, reducing FLT3 signaling.

Table 2.

Overview of Targeted Therapies in Acute Myeloid Leukemia, Which are Either Approved or Are Being Investigated in Clinical Trials

Target Therapeutic(s) Mechanism of Action
PML-RARA All-trans retinoic acid + arsenic trioxide PML-RARA configuration change, which results in coregulator exchange; sumoylation and degradation of PML-RARA
Menin Revumenib Menin-MLL interaction inhibition
Ziftomenib Menin-MLL interaction inhibition
JNJ-6617 Menin-MLL interaction inhibition
DOT1L Pinometostat Specific DOT1L inhibition via conformational change
FLT3 Type I (targeting FLT3-ITD and FLT3-TK)
Midostaurin
Crenolanib
Gilterinib
FLT3 inhibition, binds FLT3 receptor in active conformation
Type II (targeting FLT3-ITD)
Quizartinib
Sorafenib
FLT3 inhibition, binds FLT3 receptor in inactive conformation
Several kinases (KIT, SRC family, BCR-ABL, others) Dasatinib ATP-competitive protein tyrosine kinase inhibition
Mutated IDH1 Ivosidenib Selective allosteric inhibition of mutant IDH1
Mutated IDH2 Enasidenib Selective allosteric inhibition of mutant IDH2
Mutant p53 Eprenetapopt Covalent p53 core domain binding, partial restoration of WT function
XPO1 Selinexor Selective XPO1 inhibition
Eltanexor Selective XPO1 inhibition
SYK Entospletinib Selective SYK inhibition
RARα Tamibarotene RARα agonism
BCL2 Venetoclax BCL2 sequestration (BH3-mimetic)
MCL-1 Murizatoclax Selective MCL-1 inhibition
MEK Cobimetinib Selective MEK1 inhibition
MDM2 Idasanutlin p53-MDM2 interaction inhibition
Siremadlin p53-MDM2 interaction inhibition
Smoothened Glasdegib Inhibition of the translocation of Smoothened to the primary cilium
CD33 Gemtuzumab ozogamicin Gets internalized, subsequent calicheamicin causes DNA double-strand breaks
CD123 Tagraxofusp Anti-CD123 antibody fused to diphtheria toxin, gets internalized and blocks protein synthesis via eEF2 inhibition
Pivekimab sunirine Anti-CD123 antibody fused to indolinobenzodiazepine pseudodimer payload, leading to DNA alkylation and single-strand breaks
CD3/CD123 Flotetuzumab, UCART123v1.2 Anti-CD3/CD123 bispecific antibody; recruitment of T cells to CD123-expressing cells
CLL-1 CAR-T cells Recruitment of T cells to CLL-1-expressing cells
TIM-3 Sabatolimab Anti-TIM-3 monoclonal antibody—blockage of ligand binding
CD47/SIRPα Magrolimab, TTI-621 Inhibition of the CD47-SIRPα axis
AXL Bemcentinib Selective AXL inhibition
CTLA-4 Ipilimumab Anti-CTLA-4 monoclonal antibody—immune checkpoint inhibition
PD-1 Nivolumab, pembrolizumab Anti-PD-1 monoclonal antibody—immune checkpoint inhibition
GSPT1 CC-90009 Linkage of GSPT1 to an E3 ubiquitin ligase—promotes GSPT1 degradation

Following successful phase III clinical trials, midostaurin27 was approved in combination with chemotherapy in frontline FLT3-mutated AML, while gilteritinib was approved as single-agent in FLT3-mutated relapsed or refractory AML (R/R AML).28 In addition, results of a phase III trial of quizartinib combined with intensive chemotherapy showed superior OS compared with intensive chemotherapy alone,29 and a decision on regulatory approval is awaited. A common mechanism of resistance to FLT3 inhibitors arises from secondary mutations in the ATP-binding region within the FLT3 kinase domain,30,31 which seem to be inhibitor-specific.31 Alternatively, activating mutations in genes encoding members of downstream RAS/MAPK or JAK/STAT pathways, or persistent activation of pathways downstream of FLT3 due to stimulation from microenvironmental factors, can be selected by treatment and mediate resistance.3236 Accordingly, preclinical data indicate that combining FLT3 and MEK inhibition could be more efficient in causing leukemic cell death.34,37 FLT3 inhibitors are actively being evaluated in combination with venetoclax,38 venetoclax/azacitidine,39 venetoclax/ASTX727,40 and as single agents post allo-SCT41,42 (NCT02997202) or postchemotherapy43 (NCT02668653 for quizartinib with chemotherapy or as continuation therapy).

Activating mutations in KIT, encoding another RTK upstream of PI3K/AKT, RAS/MAPK, and JAK/STAT pathways, occur in a subset of core-binding factor (CBF) AML patients. Notably, KIT D816 mutations have been associated with poorer outcomes in chemotherapy-treated patients.44 Dasatinib is a multikinase inhibitor active against KIT, SRC family, BCR-ABL, and other kinases. Phase II clinical trials of dasatinib with intensive chemotherapy reported favorable event-free survival and cumulative incidence of relapse in newly diagnosed CBF AML patients.45,46 Notably, survival outcomes were compared favorably with a historical control group treated with chemotherapy alone.45 Ongoing adequately powered phase III trials will better elucidate dasatinib’s role in this setting (NCT02013648).

Approximately 20% of AMLs harbor hotspot gain-of-function mutations in IDH1 and IDH2, encoding isocitrate dehydrogenase (IDH) enzymes.47 Wild-type IDH catalyzes the conversion of isocitrate to α-ketoglutarate (α-KG) in the tricarboxylic acid (TCA) cycle. Neomorphic mutant IDH catalyzes the production of the oncometabolite 2-hydroxyglutarate (2-HG)48 from α-KG, resulting in competitive inhibition of α-KG-dependent enzymes, including ten-eleven translocation (TET) family of 5-methylcytosine hydroxylases49 and Jumonji-C domain histone demethylases.50,51 This leads to DNA hypermethylation and impaired hematopoietic differentiation.52 The small molecules ivosidenib and enasidenib, allosteric inhibitors of mutant IDH1 and IDH2, respectively, reverse DNA hypermethylation and promote differentiation.47 Both agents were approved by the FDA for the treatment of IDH-mutated AML.5355 Notably, cooccurring RUNX1 and NRAS mutations, as well as a more stem-like transcriptional state of leukemic cells, correlated with poorer responses to mutant IDH inhibition and may mediate primary resistance.56,57 Unsurprisingly, when mutant IDH inhibitors are used as monotherapy, disease relapse occurs by selecting therapy-resistant clones that have acquired multiple genetic resistance mechanisms.56,58 These include TET2, BCOR, FLT3, and RAS pathway mutations, as well as mutations in transcription factors involved in hematopoietic differentiation, which may re-establish differentiation block.56,58 Restoration of 2-HG through either secondary IDH mutations,59,60 which compromise inhibitor binding, or via selection of mutations on the gene encoding the second IDH isoform (isoform switching),58,59,61 have also been reported. Consequently, both ivosidenib and enasidenib are being tested in combination with chemotherapy, HMAs, and venetoclax.53,6264

TP53 mutations occur in 8%–12% of AML patients, a significant proportion of which also have complex or monosomal karyotypes.65 These genetic features correlate with universally poor prognosis.2,66 The small molecule eprenetapopt covalently binds to the core domain of p53,67 which mediates the cooperative DNA binding of p53. In some cases, this leads to appropriate refolding and restoration of p53 function. Eprenetapopt demonstrated encouraging activity in early-phase trials when combined with azacitidine68,69 and when administered post allo-SCT,70 but this activity was not validated in a phase III trial in TP53-mutated higher-risk MDS, which failed to meet its primary end point of a higher complete remission (CR) rate (NCT03745716).

Approximately, 30% of AML patients harbor hotspot mutations in exon 12 of the NPM1 gene,2,71 causing a frameshift that forms a novel C-terminal nuclear export signal (NES) and causes loss of a nucleolar localization signal. This results in the NPM1c neomorphic oncoprotein with predominantly cytoplasmic localization.72,73 Due to the novel C-terminal NES, NPM1c is more efficiently exported from the nucleus and binds with higher affinity to the nuclear exporter XPO1 (CRM1).74 It has been proposed that increased nuclear export of NPM1c hijacks several proteins from the nucleus, including PU.175 and CTCF,76 thus disrupting their normal function.74 Furthermore, NPM1c interferes with the p53 pathway by destabilizing the Arf tumor suppressor.77 Notably, NPM1-mutated AML typically displays a high expression of HOXA/HOXB cluster and MEIS1 genes,78 which depends on NPM1c.79 In this regard, 2 recent studies have demonstrated that the nuclear portion of NPM1c directly regulates the transcription of several target genes, including HOX family and MEIS1.80,81 In fact, NPM1c binds chromatin through the interaction with XPO1 and cooperates with MLL/Menin activity, likely underscoring favorable activity of Menin and DOT1L inhibitors in NPM1-mutated AML models.82,83 Notably, combined inhibition of XPO1 and Menin showed a synergistic decrease in HOX and MEIS1 expression, decreased colony-forming capacity of AML blasts, and induction of differentiation in model systems.80,81 The XPO1 inhibitor selinexor has shown promise in early trials on patients with hematologic malignancies, including AML,84 while the next-generation XPO1 inhibitor eltanexor demonstrated reduced toxicity compared with selinexor in preclinical models.85 Hence, there is a sound rationale for testing the combination of XPO1 and Menin inhibitors in NPM1-mutated AML patients.

Other molecules, which are aberrantly expressed and/or active, rather than mutant, have also been targeted therapeutically. One exemplar is the non-RTK SYK that has been shown to cooperate both with FLT3-ITD86 and HOXA9/MEIS1.87 In a phase Ib/II study in previously untreated AML patients, the SYK inhibitor entospletinib combined with induction chemotherapy led to composite CR of 70%, with potentially improved OS in patients with high basal HOXA9 and MEIS1 expression.88 A phase II trial of entospletinib combined with induction and consolidation chemotherapy in newly diagnosed NPM1-mutated/FLT3-ITD-wild-type AML patients reported a composite CR rate of 78% and a median duration of response of 15.7 months.89 A phase III trial of entospletinib with chemotherapy in previously untreated NPM1-mutated AML patients is ongoing (NCT05020665).

Interestingly, the RARA gene is overexpressed in a significant proportion of non-APL AML and MDS patients. This has provided the impetus for the development of the RARα agonist tamibarotene. In preclinical models of RARα-high AML, tamibarotene induced AML differentiation, reminiscent of ATRA activity in APL.90 A phase II clinical trial combining tamibarotene and azacitidine showed that 61% of evaluable RARα-high patients achieved CR or CR with incomplete hematological recovery (CRi).91 Ongoing trials are testing tamibarotene with venetoclax/azacitidine in newly diagnosed AML or with azacitidine in higher-risk myelodysplastic syndrome (MDS) (NCT04905407 and NCT04797780).

TARGETS INVOLVED IN APOPTOSIS

Regulation of apoptosis is often aberrant in cancer.92 A significant proportion of AML cells are highly dependent on the antiapoptotic BCL2 protein for their survival.93,94 Furthermore, BCL2 is overexpressed in functionally defined leukemia stem cells (LSCs).95 Concordantly, high levels of BCL2 correlate with chemoresistance in AML.96 The BH3-mimetic venetoclax is a selective BCL2 inhibitor that sequesters BCL2, thus precluding its binding to proapoptotic proteins. Venetoclax demonstrated efficacy when combined with azacitidine or low-dose cytarabine (LDAC) in treatment-naïve AML patients.97,98 These combinations are now the standard of care for AML patients unfit for intensive chemotherapy.

Importantly, response to venetoclax, or venetoclax combinations, varies with the genetic subtype of AML. IDH1/2-mutated AML has high response rates and more durable remissions to venetoclax.99 These clinical observations are concordant with preclinical data showing that IDH-mutated AML cells are highly dependent on BCL2 for survival due to the inhibition of cytochrome c oxidase by 2-HG, which decreases the threshold for apoptosis.100 Of note, remission rates for IDH2-mutated patients treated with venetoclax/azacitidine were particularly high, even when compared with IDH1-mutated patients.101 For IDH1-mutated AML, venetoclax has also recently been combined with ivosidenib and ivosidenib/azacitidine, resulting in encouraging preliminary response rates.63 Poorer responses to venetoclax/azacitidine have been noted with FLT3− (in the absence of NPM1 mutation), TP53−, and RAS-mutated AML.102,103 Interestingly, venetoclax combined with the protein translation inhibitor omacetaxine showed encouraging preclinical activity in RUNX1-mutated AML models.104

Despite these promising results, multiple mechanisms of resistance to venetoclax have been described in AML.105 For example, the selection of the monocytic differentiation program106 is associated with increased expression and function of the antiapoptotic protein MCL-1. Consistent with MCL-1 mediating venetoclax resistance, several preclinical studies have shown that inhibition of BCL2 and MCL-1 synergize in AML models.107109 Clinical trials with MCL-1 inhibitors have been ongoing for some time but have been complicated by on-target cardiac toxicity (NCT03465540). An additional mechanism of venetoclax resistance includes the selection of clones with mutations in FLT3, TP53, and genes mediating RAS/MAPK signalling.105 Hence, to potentially mitigate resistance, rational combinations of venetoclax or venetoclax/azacitidine with inhibitors of FLT3,38,39,105,110 MEK,111,112 or MDM2113,114 are being actively studied in preclinical and clinical settings. By combining clonal tracking and RNA-Seq at single-cell resolution, we are currently investigating the mechanisms of therapy response and resistance to venetoclax and ivosidenib +/− azacitidine.63

Besides induction of apoptosis, BCL2 inhibition also decreases levels of oxidative phosphorylation (OxPhos) in functionally defined LSCs.95 Moreover, the venetoclax/azacitidine treatment disrupts amino acid uptake115 and the TCA cycle,116 with consequent decrease in OxPhos and energy metabolism in these cells. Consistently, resistance to venetoclax/azacitidine arises from upregulation of fatty acid oxidation, which replenishes energy production,117 and from increased nicotinamide levels,118 which promote both fatty acid oxidation and amino acid metabolism, thus driving OxPhos. Hence, variations in central energy metabolism represent an important resistance mechanism to venetoclax/azacitidine and correlate with mutations in TP53119 and genes mediating RAS/MAPK signaling.117 Importantly, MCL-1 inhibition decreased fatty acid oxidation and engraftment of venetoclax/azacitidine-resistant LSCs.117 These studies not only further highlight the potential benefits of MCL-1 inhibition in venetoclax/azacitidine-resistant AML but also encourage clinical investigation of agents targeting intermediate metabolism, with key considerations regarding treatment specificity.

SURFACE PROTEIN TARGETING

Several therapeutic strategies target proteins expressed on the AML cell surface (Table 2). One target is the smoothened transmembrane protein, which mediates Hedgehog signaling by activating GLI transcription factors in proximity to the cellular structure known as primary cilium. Glasdegib inhibits the translocation of smoothened to the primary cilium, thus blunting GLI activation.120 Although there is limited knowledge about the exact role of this pathway in AML, encouraging data from a phase II clinical trial showed that the addition of glasdegib to LDAC improved median OS compared with LDAC alone (8.8 months versus 4.9 months), in older or unfit AML and high-risk MDS patients.121 This led to the licensing of a smoothened inhibitor glasdegib combined with LDAC in the USA (2018) and in the EU (2020). A current phase III trial is evaluating glasdegib in combination with intensive chemotherapy or azacitidine in previously untreated AML patients (NCT03416179).

CD33 and CD123 molecules are generally found on subsets of hematopoietic cells but are highly expressed on the AML cell surface in most patients.122 Gemtuzumab ozogamicin is a humanized anti-CD33 monoclonal antibody conjugated to the cytotoxic drug calicheamicin. Upon binding to CD33, gemtuzumab ozogamicin is internalized, and calicheamicin is released, causing DNA double-strand breaks, which trigger cell death. Gemtuzumab ozogamicin has been approved in combination with intensive chemotherapy for newly diagnosed AML.123,124 Interestingly, gemtuzumab ozogamicin, in combination with intensive chemotherapy, has maximal benefit in good prognosis CBF AML, but the reasons for this are still not entirely clear.

Tagraxofusp is a recombinant anti-CD123 (IL-3 receptor alpha chain) antibody fused to diphtheria toxin. When bound to CD123, tagraxofusp is internalized and blocks protein synthesis via eEF2 inhibition. Tagraxofusp has shown clinical benefit in blastic plasmacytoid dendritic cell neoplasm, a specific form of myeloid malignancy with high CD123 expression.125 Tagraxofusp has also been combined with azacitidine or venetoclax/azacitidine in CD123-positive AML and MDS patients. Eight of the 9 previously untreated AML patients and 3 of 4 of previously untreated MDS patients achieved CR/CRi.126 Of note, 2 responding AML patients and 3 responding MDS patients were TP53-mutated. Another anti-CD123 antibody-drug conjugate, pivekimab sunirine, which causes DNA alkylation,127 led to objective response rates (ORR) of 51% when combined with venetoclax/azacitidine in CD123-positive R/R AML, with auspicious activity in the FLT3-ITD subgroup, where ORR was 82%.128

Bispecific antibodies, typically bispecific T-cell engagers, recruit T cells to tumors via an anti-CD3 moiety by binding to a tumor-specific antigen. An exemplar is blinatumomab, used to treat B-cell acute lymphoblastic leukemia. The success of blinatumomab has triggered enormous activity in this therapeutic area with variations in the mechanisms of action. One example is the concept of dual affinity retargeting antibodies such as flotetuzumab, an anti-CD3/CD123 bispecific antibody.129 In phase 1 of 2 studies, 26.6% R/R AML patients who did not respond to induction therapy or who experienced early relapse postinduction therapy achieved CR/CRh with a median OS of 10.2 months. Interestingly, a 10-gene signature derived from diagnostic bone marrow was predictive of CR.

Chimeric antigen receptor (CAR) T-cell therapy has been transformative in lymphoid malignancy. CD19-directed CAR-T led the initial wave of therapies,130132 which was recognized by the American Society of Clinical Oncology133 as the advance of the year in 2018. Between 2018 and 2023, 5 CAR-T cell therapies targeting CD19 or B-cell maturation antigen were authorized for sale. However, finding a suitable antigen target for AML that is not present on normal myeloid cells has been a challenge. CD123-directed CAR-T cells showed promising antileukemic activity in vitro and in vivo, with limited toxicity toward healthy hematopoietic cells.134 A phase I trial evaluated UCART123v1.2, an allogeneic anti-CD123 T-cell product, in 16 CD123-positive R/R AML patients.135 Four patients showed evidence of activity, including 1 long-term CR. Another approach has been universal CAR-T therapy with inducible activity of the CD123-targeting module. In a phase I trial of 14 CD123-positive R/R AML, 3 patients achieved CR/CRi, with 4 partial responses.136 Anti-CD123 CAR-T cells have also been tested in pediatric R/R AML patients137 in a phase I study. CAR-T cells against the surface molecule CLL-1 have shown some promise, with 7 of 10 R/R AML patients achieving CR/CRi in a phase I trial.138

TIM-3 is a surface receptor preferentially expressed on AML cells with LSC function compared with normal HSCs.139 Moreover, TIM-3 marks dysfunctional and/or exhausted subsets of T cells in viral infections and cancer.140,141 Concordantly, xenograft data in nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice showed a decrease in AML reconstitution and LSC function following antibody targeting of TIM-3.142 Sabatolimab, an anti-TIM-3 monoclonal antibody, combined with HMAs, achieved a CR/CRi rate of 30% in the Revised International Prognostic Scoring System for high-/very high-risk MDS and newly diagnosed AML patients unfit for intensive chemotherapy.143 More extensive phase II/III clinical trials are currently evaluating the efficacy of sabatolimab in multiple regimens.144

An alternative targeted strategy harnesses autologous innate immunity to specifically kill leukemic cells. CD47 (integrin-associated protein) is a surface molecule mediating don’t eat me signals. This occurs via CD47-SIRPα interactions, which inhibit macrophage-mediated phagocytosis.145 Notably, CD47 is upregulated both on LSC-enriched and nonenriched AML cells, compared with normal hematopoietic cells, predicting poor outcome.146,147 Targeting the CD47-SIRPα axis has thus led to multiple drugs in preclinical and clinical development, including monoclonal antibodies, fusion proteins, bispecific antibodies, small molecules, RNA interference, and CD47-directed CAR-T cells. Importantly, anti-CD47 monoclonal antibodies stimulated AML cell phagocytosis in vivo while sparing normal HSCs.146 This encouraged the development of magrolimab, a humanized anti-CD47 antibody, which was well-tolerated in R/R AML patients in the CAMELLIA phase 1 trial.148 Results from a subsequent phase IB trial combining magrolimab with azacitidine in 95 untreated intermediate/high- and very high-risk MDS patients have just been reported. Sixty-two percent of patients had poor-risk cytogenetics, and 26% had a TP53 mutation.149 The combination was well-tolerated, the CR rate was 33%, and the duration of the overall response was 9.8 months. Median OS was not reached with a median 17.1-month follow-up. About 40% of TP53-mutated patients achieved CR and had a median OS of 16.1 months. Results from a phase Ib trial of magrolimab+azacitidine in 87 previously untreated AML patients, 72 of which TP53-mutated (median variant allele frequency of 61%), have also just been reported (Daver et al, invited revision resubmitted at Journal of Clinical Oncology). About 79.2% of TP53-mutated patients had adverse-risk cytogenetics according to the European LeukemiaNet (ELN) 2017 classification. Totally 28 (32.2%) patients achieved CR, including 23 (31.9%) patients with TP53 mutations. Median OS in TP53-mutated patients was 9.8 months and 18.9 months in TP53-wild-type patients. Magrolimab has also been combined with venetoclax/azacitidine in a phase I/II study, displaying an ORR of 80% in newly diagnosed AML and 75% in venetoclax-naïve R/R AML.150 Of note, 63% of newly diagnosed TP53-mutant patients achieved CR/CRi. Three phase III trials of magrolimab combinations in previously untreated higher-risk MDS and AML patients are in progress and will determine its clinical role (NCT04313881, NCT05079230, and NCT04778397). Other approaches targeting the CD47-SIRPα axis include the fusion protein TTI-621 (SIRPα-IgG1 Fc), which inhibited the growth of AML xenografts,151 leading to its evaluation in early-phase clinical trials for hematological malignancies.152 In addition, the bispecific molecule PD1xCD47 BsAb HX009 has very recently shown antileukemic activity in a patient-derived xenograft model of AML.153

MicroRNAs repress gene expression by selective binding to the 3′ untranslated regions of target genes, which induces mRNA degradation and inhibition of translation.154 In a preclinical study, miR-708 was shown to negatively regulate CD47 expression and to inhibit T-ALL engraftment.155 These alternative approaches might thus hold promise in reactivating antileukemic autologous innate immunity.

AXL is a member of the TAM family of RTK exerting pleiotropic effects in cancer, including the potentiation of migratory capabilities.156 Axl overexpression correlates with poor prognosis in AML,157 and Axl expression is an independent prognostic factor in de novo AML.158 Bemcentinib is a selective small molecule AXL inhibitor, which is well-tolerated as single agent by AML patients who relapsed after treatment with HMAs.159 In an ongoing phase II trial, bemcentinib is being investigated in AML patients either as single agent or in combination with LDAC or decitabine.160

While immune checkpoint inhibition has shown great promise in several solid tumors, results in AML have been disappointing after some initial encouraging data with ipilimumab (CTLA-4 inhibition) in relapsed patients post allo-SCT.161 Since then, CTLA-4 (ipilimumab) and PD-1 (nivolumab and pembrolizumab) inhibitors have shown modest efficacy in phase I and II trials161169 and when combined with azacitidine in R/R AML.170 Furthermore, PD-1 inhibition combined with chemotherapy showed some efficacy in newly diagnosed AML,167 and PD-1 inhibition combined with venetoclax-azacitidine is being evaluated (NCT04284787).

TARGETED PROTEIN DEGRADATION

The cellular protein degradation machinery can be therapeutically leveraged to degrade proteins that cancer cells depend on (Table 2). The ubiquitin/proteasome system, composed of E3 ubiquitin ligase complexes, ubiquitinates damaged and unwanted proteins, directing them toward proteasomal degradation. In this context, the therapeutic agent acts as molecular glue, linking E3 ubiquitin ligases to target proteins (termed neosubstrates). For example, the small molecules thalidomide, lenalidomide, and pomalidomide link neosubstrates to cereblon, a member of the CRL4 E3 ubiquitin ligase complex. This leads to the degradation of proteins essential for multiple myeloma or MDS cell survival.171 Subsequent development of Cereblon E3 ligase modulating drugs (CELMoDs) allowed the targeting of new neosubstrates by linking them to cereblon-containing E3 ligases. One CELMoD being investigated in AML is CC-90009172: This molecule promotes the degradation of GSPT1, which coregulates the termination of protein translation. In preclinical studies, GSPT1 degradation inhibited AML cell survival and proliferation.173 Of note, CC-90009 efficacy may depend on AML differentiation stage.174 CC-90009, as a single agent and in combination with other targeted approaches, is being evaluated in phase I trials in AML patients (NCT02848001 and NCT04336982). We are currently investigating clonal and molecular determinants of response and resistance to this treatment.

One limitation of the above approaches is the requirement for neosubstrates to contain a common degron peptide recognized by cereblon. Proteolysis-targeting chimeras (PROTACs) bind both to the target protein (via a PROTAC warhead) and to an E3 ubiquitin ligase enzyme via a linker domain connected to the E3 ligand.3 This allows degrading proteins without the requirement for a common degron peptide. PROTACs are being investigated in hematological malignancies, for example, to target BCL2 in chronic lymphocytic leukemia.175 The PROTAC technology has recently been designed to specifically degrade EZH2, the core member of the PRC2 complex with histone methyltransferase activity.176 Mounting evidence suggests that EZH2 inhibits differentiation and promotes AML maintenance.177179 A recent study further identified a noncatalytic role of EZH2 in MLL-rearranged AML caused by the direct binding of EZH2 to cMYC and p300, which promotes malignant cell growth.176 The novel PROTAC MS177, which promotes EZH2 degradation via cereblon, disrupted both the PRC2-dependent and cMYC-dependent EZH2 activity, inhibiting leukemic cell growth in vitro and in vivo.176 Hence, this study exemplifies how PROTACs may be particularly effective in abolishing additional noncatalytic functions of targeted proteins. PROTACs might prove effective against transcription factors with oncogenic function in AML, such as WT1, MEIS1, or HOX family members.

CHALLENGES AHEAD: HOW TO OVERCOME THEM?

Novel targeted therapies are revolutionizing AML therapy, but fundamental challenges persist. The selection of resistant clones under therapy pressure causes treatment failure and relapse. An integrated characterization of genetic and molecular landscapes in longitudinal patient samples is needed to better understand putative resistance mechanisms. Emerging single-cell approaches coupling somatic genotypes with cellular readouts are potent tools to describe clonal complexity.180 Complementing these technologies, our group recently integrated clonal readout with chromatin accessibility profiling at single-cell resolution, linking genetic and epigenetic disease evolution.181 Going forward, this approach will elucidate clone-specific epigenetic mechanisms underlying disease relapse.

An obstacle to cure is the failure to eradicate quiescent LSCs, the seed of malignant growth most frequently residing in leukemic progenitor and/or precursor populations.182,183 Hence, a better understanding of mechanisms driving aberrant self-renewal of LSCs is necessary and might lead to novel therapeutic targets.

In the context of disease prevention, the study of preleukemic clonal outgrowths, occurring years before leukemia onset, is critical. We and others are actively exploring molecular landscapes underlying human clonal hematopoiesis,184186 to elucidate pathways leading to early clonal dominance predating myeloid malignancy.

Going forward, better coordination between basic, translational, and clinical research is critical to maximize the value of technological advances. This will not only boost research through improved sample availability but also will lead to the rapid implementation of new technologies in clinical settings. Larger multicenter trials are warranted for the efficient delivery of promising therapeutic agents. Moreover, the process of drug discovery and clinical evaluation will benefit from increased collaboration between governments, industries, trial centers, and academic research. Finally, huge steps are needed to improve science communication between patients, their families, researchers, clinicians, and the public, to keep the primary focus on improving patients’ lives.

ACKNOWLEDGMENTS

Figure 1 was created using BioRender. The authors thank Dr. Maria Chiara Barbanti and Dr. Bing Tseu for their insightful comments and discussions.

AUTHOR CONTRIBUTIONS

ST and FAR did writing and original draft preparation. PV did writing, review, and editing. All authors have read and agreed to the final version of the article.

DISCLOSURES

The authors have no conflicts of interest to disclose.

SOURCES OF FUNDING

PV acknowledges funding from the Medical Research Council Molecular Hematology Unit Programme Grant (MC_UU_00029/8), Blood Cancer UK Programme Continuity Grant 13008, NIHR Senior Fellowship, and the Oxford BRC Hematology Theme. ST is supported by a Scatcherd European Scholarship in partnership with The Medical Research Council/Radcliffe Department of Medicine and The Clarendon Fund. FAR is supported by the Oxford – Sir David Weatherall Scholarship from Green Templeton College and the WIMM Prize Studentship from the MRC Weatherall Institute of Molecular Medicine.

Footnotes

ST and FAR have contributed equally to this work.

REFERENCES

  • 1.The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult De Novo acute myeloid leukemia. N Engl J Med. 2013;368:2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209–2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pikman Y, Stegmaier K. Targeted therapy for fusion-driven high-risk acute leukemia. Blood. 2018;132:1241–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rowley JD. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and giemsa staining. Nature. 1973;243:290–293. [DOI] [PubMed] [Google Scholar]
  • 5.Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037. [DOI] [PubMed] [Google Scholar]
  • 6.Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr–Abl positive cells. Nat Med. 1996;2:561–566. [DOI] [PubMed] [Google Scholar]
  • 7.de Thé H, Chomienne C, Lanotte M, et al. The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor α gene to a novel transcribed locus. Nature. 1990;347:558–561. [DOI] [PubMed] [Google Scholar]
  • 8.Kakizuka A, Miller WH, Jr, Umesono K, et al. Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML. Cell. 1991;66:663–674. [DOI] [PubMed] [Google Scholar]
  • 9.Wang Z-Y, Chen Z. Acute promyelocytic leukemia: from highly fatal to highly curable. Blood. 2008;111:2505–2515. [DOI] [PubMed] [Google Scholar]
  • 10.Lo-Coco F, Avvisati G, Vignetti M, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–121. [DOI] [PubMed] [Google Scholar]
  • 11.Hu J, Liu Y-F, Wu C-F, et al. Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia. Proc Natl Acad Sci USA. 2009;106:3342–3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Meng-er H, Yu-chen Y, Shu-rong C, et al. Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. Blood. 1988;72:567–572. [PubMed] [Google Scholar]
  • 13.Lallemand-Breitenbach V, Guillemin M-C, Janin A, et al. Retinoic acid and arsenic synergize to eradicate leukemic cells in a mouse model of acute promyelocytic leukemia. J Exp Med. 1999;189:1043–1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yokoyama A, Somervaille TCP, Smith KS, et al. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell. 2005;123:207–218. [DOI] [PubMed] [Google Scholar]
  • 15.Smith E, Lin C, Shilatifard A. The super elongation complex (SEC) and MLL in development and disease. Genes Dev. 2011;25:661–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Soto-Feliciano YM, Sanchez-Rivera FJ, Perner F, et al. A molecular switch between mammalian MLL complexes dictates response to menin-MLL inhibition. Cancer Discov. 2023;13:146–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Borkin D, He S, Miao H, et al. Pharmacologic inhibition of the menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell. 2015;27:589–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fiskus W, Daver N, Boettcher S, et al. Activity of menin inhibitor ziftomenib (KO-539) as monotherapy or in combinations against AML cells with MLL1 rearrangement or mutant NPM1. Leukemia. 2022;36:2729–2733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fiskus W, Boettcher S, Daver N, et al. Effective Menin inhibitor-based combinations against AML with MLL rearrangement or NPM1 mutation (NPM1c). Blood Cancer J. 2022;12:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Issa GC, Aldoss I, DiPersio J, et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature. 2023;615:920–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Perner F, Stein EM, Wenge DV, et al. MEN1 mutations mediate clinical resistance to menin inhibition. Nature. 2023;615:913–919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Erba HP, Fathi AT, Issa GC, et al. Update on a phase 1/2 first-in-human study of the menin-KMT2A (MLL) inhibitor ziftomenib (KO-539) in patients with relapsed or refractory acute myeloid leukemia. Blood. 2022;140(Supplement 1):153–156. [Google Scholar]
  • 23.Bernt Kathrin M, Zhu N, Sinha Amit U, et al. MLL-Rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20:66–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood. 2018;131:2661–2669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Menghrajani K, Cai SF, Devlin SM, et al. A phase Ib/II study of the histone methyltransferase inhibitor pinometostat in combination with azacitidine in patients with 11q23-rearranged acute myeloid leukemia. Blood. 2019;134(Supplement_1):2655–2655. [Google Scholar]
  • 26.Daver N, Schlenk RF, Russell NH, et al. Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. 2019;33:299–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377:454–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381:1728–1740. [DOI] [PubMed] [Google Scholar]
  • 29.Erba H, Montesinos P, Vrhovac R, et al. S100: quizartinib prolonged survival vs placebo plus intensive induction and consolidation therapy followed by single-agent continuation in patients aged 18-75 years with newly diagnosed FLT3-ITD+ AML. HemaSphere. 2022;6:1–2. [Google Scholar]
  • 30.Heidel F, Solem FK, Breitenbuecher F, et al. Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood. 2006;107:293–300. [DOI] [PubMed] [Google Scholar]
  • 31.von Bubnoff N, Engh RA, Åberg E, et al. FMS-like tyrosine kinase 3–internal tandem duplication tyrosine kinase inhibitors display a nonoverlapping profile of resistance mutations in vitro. Cancer Res. 2009;69:3032–3041. [DOI] [PubMed] [Google Scholar]
  • 32.Piloto O, Wright M, Brown P, et al. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood. 2006;109:1643–1652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Alotaibi AS, Yilmaz M, Kanagal-Shamanna R, et al. Patterns of resistance differ in patients with acute myeloid leukemia treated with type I versus type II FLT3 inhibitors. Blood Cancer Discov. 2021;2:125–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.McMahon CM, Ferng T, Canaani J, et al. Clonal selection with RAS pathway activation mediates secondary clinical resistance to selective FLT3 inhibition in acute myeloid leukemia. Cancer Discov. 2019;9:1050–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Traer E, Martinez J, Javidi-Sharifi N, et al. FGF2 from marrow microenvironment promotes resistance to FLT3 inhibitors in acute myeloid leukemia. Cancer Res. 2016;76:6471–6482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rummelt C, Gorantla SP, Meggendorfer M, et al. Activating JAK-mutations confer resistance to FLT3 kinase inhibitors in FLT3-ITD positive AML in vitro and in vivo. Leukemia. 2021;35:2017–2029. [DOI] [PubMed] [Google Scholar]
  • 37.Joshi SK, Nechiporuk T, Bottomly D, et al. The AML microenvironment catalyzes a stepwise evolution to gilteritinib resistance. Cancer Cell. 2021;39:999–1014.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Daver N, Perl AE, Maly J, et al. Venetoclax plus gilteritinib for FLT3-mutated relapsed/refractory acute myeloid leukemia. J Clin Oncol. 2022;40:4048–4059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Short N, DiNardo CD, Daver N, et al. Updated results from a phase I/II study of the triplet combination of azacitidine, venetoclax and gilteritinib for patients with FLT3-mutated acute myeloid leukemia. Blood. 2022;140(Supplement 1):2007–2009. [Google Scholar]
  • 40.Ong F, Short N, Daver N, et al. A phase I/II study of combination of ASTX727, gilteritinib and venetoclax in patients with relapsed/refractory FLT3 mutated acute myeloid leukemia (AML) and frontline FLT3 mutated AML patients unfit for chemotherapy. Blood. 2022;140(Supplement 1):9029–9031. [Google Scholar]
  • 41.Xuan L, Wang Y, Huang F, et al. Sorafenib maintenance in patients with FLT3-ITD acute myeloid leukaemia undergoing allogeneic haematopoietic stem-cell transplantation: an open-label, multicentre, randomised phase 3 trial. Lancet Oncol. 2020;21:1201–1212. [DOI] [PubMed] [Google Scholar]
  • 42.Perl AE, Larson RA, Podoltsev NA, et al. Follow-up of patients with R/R FLT3-mutation–positive AML treated with gilteritinib in the phase 3 ADMIRAL trial. Blood. 2022;139:3366–3375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schlenk RF, Weber D, Fiedler W, et al. Midostaurin added to chemotherapy and continued single-agent maintenance therapy in acute myeloid leukemia with FLT3-ITD. Blood. 2019;133:840–851. [DOI] [PubMed] [Google Scholar]
  • 44.Cairoli R, Beghini A, Grillo G, et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: an Italian retrospective study. Blood. 2006;107:3463–3468. [DOI] [PubMed] [Google Scholar]
  • 45.Paschka P, Schlenk RF, Weber D, et al. Adding dasatinib to intensive treatment in core-binding factor acute myeloid leukemia—results of the AMLSG 11-08 trial. Leukemia. 2018;32:1621–1630. [DOI] [PubMed] [Google Scholar]
  • 46.Marcucci G, Geyer S, Laumann K, et al. Combination of dasatinib with chemotherapy in previously untreated core binding factor acute myeloid leukemia: CALGB 10801. Blood Adv. 2020;4:696–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Issa GC, DiNardo CD. Acute myeloid leukemia with IDH1 and IDH2 mutations: 2021 treatment algorithm. Blood Cancer J. 2021;11:107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739–744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Xu W, Yang H, Liu Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell. 2011;19:17–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chowdhury R, Yeoh KK, Tian Y-M, et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 2011;12:463–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18:553–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kantarjian H, Short NJ, DiNardo C, et al. Harnessing the benefits of available targeted therapies in acute myeloid leukaemia. Lancet Haematol. 2021;8:e922–e933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378:2386–2398. [DOI] [PubMed] [Google Scholar]
  • 55.Stein EM, DiNardo C, Altman JK, et al. Safety and efficacy of AG-221, a Potent inhibitor of mutant IDH2 that promotes differentiation of myeloid cells in patients with advanced hematologic malignancies: results of a phase 1/2 trial. Blood. 2015;126:323–323. [Google Scholar]
  • 56.Wang F, Morita K, DiNardo CD, et al. Leukemia stemness and co-occurring mutations drive resistance to IDH inhibitors in acute myeloid leukemia. Nat Commun. 2021;12:2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Amatangelo MD, Quek L, Shih A, et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood. 2017;130:732–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Quek L, David MD, Kennedy A, et al. Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib. Nat Med. 2018;24:1167–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Choe S, Wang H, DiNardo CD, et al. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML. Blood Adv. 2020;4:1894–1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Intlekofer AM, Shih AH, Wang B, et al. Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations. Nature. 2018;559:125–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Harding JJ, Lowery MA, Shih AH, et al. Isoform switching as a mechanism of acquired resistance to mutant isocitrate dehydrogenase inhibition. Cancer Discov. 2018;8:1540–1547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.DiNardo CD, Stein AS, Stein EM, et al. Mutant isocitrate dehydrogenase 1 inhibitor ivosidenib in combination with azacitidine for newly diagnosed acute myeloid leukemia. J Clin Oncol. 2021;39:57–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lachowiez CA, Loghavi S, Zeng Z, et al. A phase Ib/II study of ivosidenib with venetoclax +/- azacitidine in IDH1-mutated myeloid malignancies. Blood Cancer Discov. 2023. April 27. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Stein EM, DiNardo CD, Fathi AT, et al. Ivosidenib or enasidenib combined with intensive chemotherapy in patients with newly diagnosed AML: a phase 1 study. Blood. 2021;137:1792–1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bullinger L, Döhner K, Döhner H. Genomics of acute myeloid leukemia diagnosis and pathways. J Clin Oncol. 2017;35:934–946. [DOI] [PubMed] [Google Scholar]
  • 66.Rücker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119:2114–2121. [DOI] [PubMed] [Google Scholar]
  • 67.Lambert JMR, Gorzov P, Veprintsev DB, et al. PRIMA-1 reactivates mutant p53 by covalent binding to the core domain. Cancer Cell. 2009;15:376–388. [DOI] [PubMed] [Google Scholar]
  • 68.Cluzeau T, Sebert M, Rahmé R, 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;39:1575–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J Clin Oncol. 2021;39:1584–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mishra A, Tamari R, DeZern AE, et al. Eprenetapopt plus azacitidine after allogeneic hematopoietic stem-cell transplantation for TP53-mutant acute myeloid leukemia and myelodysplastic syndromes. J Clin Oncol. 2022;40:3985–3993. [DOI] [PubMed] [Google Scholar]
  • 71.Falini B, Nicoletti I, Martelli MF, et al. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood. 2006;109:874–885. [DOI] [PubMed] [Google Scholar]
  • 72.Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005;352:254–266. [DOI] [PubMed] [Google Scholar]
  • 73.Falini B, Bolli N, Shan J, et al. Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML. Blood. 2006;107:4514–4523. [DOI] [PubMed] [Google Scholar]
  • 74.Falini B, Brunetti L, Sportoletti P, et al. NPM1-mutated acute myeloid leukemia: from bench to bedside. Blood. 2020;136:1707–1721. [DOI] [PubMed] [Google Scholar]
  • 75.Gu X, Ebrahem Q, Mahfouz RZ, et al. Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates. J Clin Invest. 2018;128:4260–4279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wang AJ, Han Y, Jia N, et al. NPM1c impedes CTCF functions through cytoplasmic mislocalization in acute myeloid leukemia. Leukemia. 2020;34:1278–1290. [DOI] [PubMed] [Google Scholar]
  • 77.Colombo E, Martinelli P, Zamponi R, et al. Delocalization and destabilization of the arf tumor suppressor by the leukemia-associated NPM mutant. Cancer Res. 2006;66:3044–3050. [DOI] [PubMed] [Google Scholar]
  • 78.Alcalay M, Tiacci E, Bergomas R, et al. Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance. Blood. 2005;106:899–902. [DOI] [PubMed] [Google Scholar]
  • 79.Brunetti L, Gundry MC, Sorcini D, et al. Mutant NPM1 maintains the leukemic state through HOX expression. Cancer Cell. 2018;34:499–512.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Uckelmann HJ, Haarer EL, Takeda R, et al. Mutant NPM1 directly regulates oncogenic transcription in acute myeloid leukemia. Cancer Discov. 2023;13:746–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wang XQD, Fan D, Han Q, et al. Mutant NPM1 hijacks transcriptional hubs to maintain pathogenic gene programs in acute myeloid leukemia. Cancer Discov. 2023;13:724–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kühn MWM, Song E, Feng Z, et al. Targeting chromatin regulators inhibits leukemogenic gene expression in npm1 mutant leukemia. Cancer Discov. 2016;6:1166–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Uckelmann HJ, Kim SM, Wong EM, et al. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science. 2020;367:586–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Azmi AS, Uddin MH, Mohammad RM. The nuclear export protein XPO1 — from biology to targeted therapy. Nat Rev Clin Oncol. 2021;18:152–169. [DOI] [PubMed] [Google Scholar]
  • 85.Etchin J, Berezovskaya A, Conway AS, et al. KPT-8602, a second-generation inhibitor of XPO1-mediated nuclear export, is well tolerated and highly active against AML blasts and leukemia-initiating cells. Leukemia. 2017;31:143–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Puissant A, Fenouille N, Alexe G, et al. SYK is a critical regulator of FLT3 in acute myeloid leukemia. Cancer Cell. 2014;25:226–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Mohr S, Doebele C, Comoglio F, et al. Hoxa9 and meis1 cooperatively induce addiction to syk signaling by suppressing mir-146a in acute myeloid leukemia. Cancer Cell. 2017;31:549–562.e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Walker AR, Byrd JC, Blachly JS, et al. Entospletinib in combination with induction chemotherapy in previously untreated acute myeloid leukemia: response and predictive significance of HOXA9 and MEIS1 expression. Clin Cancer Res. 2020;26:5852–5859. [DOI] [PubMed] [Google Scholar]
  • 89.Borate U, Li R, Huang Y, et al. Entospletinib (ENTO) in combination with cytarabine (Ara-C) and daunorubicin (DNR) in newly diagnosed (ND) adult patients with NPM1-mutated and FLT3-ITD wild-type acute myeloid leukemia (AML) is associated with good response and survival: a phase 2 sub-study of the beat AML master trial. Blood. 2022;140(Supplement 1):6200–6203. [Google Scholar]
  • 90.McKeown MR, Corces MR, Eaton ML, et al. Superenhancer analysis defines novel epigenomic subtypes of Non-APL AML, including an RARα dependency targetable by SY-1425, a potent and selective RARα agonist. Cancer Discov. 2017;7:1136–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.de Botton S, Cluzeau T, Vigil CE, et al. Targeting RARA overexpression with tamibarotene, a potent and selective RARα agonist, is a novel approach in AML. Blood Adv. 2023;7:1858–1870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat Rev Mol Cell Biol. 2019;20:175–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Konopleva M, Contractor R, Tsao T, et al. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell. 2006;10:375–388. [DOI] [PubMed] [Google Scholar]
  • 94.Vo T-T, Ryan J, Carrasco R, et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell. 2012;151:344–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lagadinou Eleni D, Sach A, Callahan K, et al. BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells. Cell Stem Cell. 2013;12:329–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Campos L, Rouault J-P, Sabido O, et al. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood. 1993;81:3091–3096. [PubMed] [Google Scholar]
  • 97.DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383:617–629. [DOI] [PubMed] [Google Scholar]
  • 98.Wei AH, Strickland SA, Jr, Hou J-Z, et al. Venetoclax combined with low-dose cytarabine for previously untreated patients with acute myeloid leukemia: results from a phase Ib/II study. J Clin Oncol. 2019;37:1277–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6:1106–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chan SM, Thomas D, Corces-Zimmerman MR, et al. Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia. Nat Med. 2015;21:178–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pollyea DA, DiNardo CD, Arellano ML, et al. Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022;28:2753–2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.DiNardo CD, Tiong IS, Quaglieri A, et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood. 2020;135:791–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Konopleva M, Thirman MJ, Pratz KW, et al. Impact of FLT3 mutation on outcomes after venetoclax and azacitidine for patients with treatment-naive acute myeloid leukemia. Clin Cancer Res. 2022;28:2744–2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mill CP, Fiskus W, DiNardo CD, et al. Effective therapy for AML with RUNX1 mutation by cotreatment with inhibitors of protein translation and BCL2. Blood. 2022;139:907–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ong F, Kim K, Konopleva MY. Venetoclax resistance: mechanistic insights and future strategies. Cancer Drug Resist. 2022;5:380–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Pei S, Pollyea DA, Gustafson A, et al. Monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia. Cancer Discovery. 2020;10:536–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ramsey HE, Fischer MA, Lee T, et al. A novel MCL1 inhibitor combined with venetoclax rescues venetoclax-resistant acute myelogenous leukemia. Cancer Discovery. 2018;8:1566–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Moujalled DM, Pomilio G, Ghiurau C, et al. Combining BH3-mimetics to target both BCL-2 and MCL1 has potent activity in pre-clinical models of acute myeloid leukemia. Leukemia. 2019;33:905–917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tron AE, Belmonte MA, Adam A, et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat Commun. 2018;9:5341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Yilmaz M, Kantarjian H, Short NJ, et al. Hypomethylating agent and venetoclax with FLT3 inhibitor “triplet” therapy in older/unfit patients with FLT3 mutated AML. Blood Cancer J. 2022;12:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Desikan SP, Ravandi F, Pemmaraju N, et al. A Phase II study of azacitidine, venetoclax and trametinib in relapsed/refractory AML harboring a ras pathway-activating mutation. Blood. 2021;138:4436–4436. [DOI] [PubMed] [Google Scholar]
  • 112.Padua RA, Sarda-Mantel L, Chiquet M, et al. BCL-2 inhibitor venetoclax (ABT-199) and MEK inhibitor GDC-0973 synergise to target AML progenitors and overcome drug resistance with the use of PET scanning in a mouse model of HR-MDS to monitor response to treatment. Blood. 2018;132(Supplement 1):5497–5497. [Google Scholar]
  • 113.Daver NG, Dail M, Garcia JS, et al. Venetoclax and idasanutlin in relapsed/refractory AML: a nonrandomized, open-label phase 1b trial. Blood. 2023;141:1265–1276. [DOI] [PubMed] [Google Scholar]
  • 114.Wei AH, Breccia M, Ooi M, et al. Preliminary results from a phase Ib study exploring MDM2 inhibitor siremadlin (HDM201) in combination with B-Cell Lymphoma-2 (BCL-2) inhibitor venetoclax in patients with acute myeloid leukemia (AML) or high-risk myelodysplastic syndrome (HR-MDS). Blood. 2021;138(Supplement 1):1283–1283.34617998 [Google Scholar]
  • 115.Jones CL, Stevens BM, D’Alessandro A, et al. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell. 2018;34:724–740.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Pollyea DA, Stevens BM, Jones CL, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24:1859–1866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Stevens BM, Jones CL, Pollyea DA, et al. Fatty acid metabolism underlies venetoclax resistance in acute myeloid leukemia stem cells. Nature Cancer. 2020;1:1176–1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Jones CL, Stevens BM, Pollyea DA, et al. Nicotinamide metabolism mediates resistance to venetoclax in relapsed acute myeloid leukemia stem cells. Cell Stem Cell. 2020;27:748–764.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Nechiporuk T, Kurtz SE, Nikolova O, et al. The TP53 apoptotic network is a primary mediator of resistance to BCL2 inhibition in AML cells. Cancer Discovery. 2019;9:910–925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Chaudhry P, Singh M, Triche TJ, et al. GLI3 repressor determines Hedgehog pathway activation and is required for response to SMO antagonist glasdegib in AML. Blood. 2017;129:3465–3475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Cortes JE, Heidel FH, Hellmann A, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33:379–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ehninger A, Kramer M, Röllig C, et al. Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J. 2014;4:e218–e218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15:986–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Castaigne S, Pautas C, Terre C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379:1508–1516. [DOI] [PubMed] [Google Scholar]
  • 125.Pemmaraju N, Lane AA, Sweet KL, et al. Tagraxofusp in blastic plasmacytoid dendritic-cell neoplasm. N Engl J Med. 2019;380:1628–1637. [DOI] [PubMed] [Google Scholar]
  • 126.Lane AA, Stein AS, Garcia JS, et al. Safety and efficacy of combining tagraxofusp (SL-401) with azacitidine or azacitidine and venetoclax in a phase 1b study for CD123 positive AML, MDS, or BPDCN. Blood. 2021;138(Supplement 1):2346–2346. [Google Scholar]
  • 127.Kovtun Y, Jones GE, Adams S, et al. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv. 2018;2:848–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Daver N, Montesinos P, Aribi A, et al. Broad activity for the pivekimab sunirine (PVEK, IMGN632), azacitidine, and venetoclax triplet in high-risk patients with relapsed/refractory acute myeloid leukemia (AML). Blood. 2022;140(Supplement 1):145–149. [Google Scholar]
  • 129.Uy GL, Aldoss I, Foster MC, et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood. 2021;137:751–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377:2531–2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Schuster SJ, Svoboda J, Chong EA, et al. Chimeric antigen receptor T cells in refractory B-Cell lymphomas. N Engl J Med. 2017;377:2545–2554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Heymach J, Krilov L, Alberg A, et al. Clinical cancer advances 2018: annual report on progress against cancer from the American Society of Clinical Oncology. J Clin Oncol. 2018;36:1020–1044. [DOI] [PubMed] [Google Scholar]
  • 134.Sugita M, Galetto R, Zong H, et al. Allogeneic TCRαβ deficient CAR T-cells targeting CD123 in acute myeloid leukemia. Nat Commun. 2022;13:2227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Sallman DA, DeAngelo DJ, Pemmaraju N, et al. Ameli-01: a phase I trial of UCART123v1.2, an Anti-CD123 allogeneic CAR-T cell product, in adult patients with relapsed or refractory (R/R) CD123+ acute myeloid leukemia (AML). Blood. 2022;140(Supplement 1):2371–2373.36054916 [Google Scholar]
  • 136.Ehninger G, Kraus S, Sala E, et al. Phase 1 dose escalation study of the rapidly switchable universal CAR-T therapy unicar-T-CD123 in relapsed/refractory AML. Blood. 2022;140(Supplement 1):2367–2368. [Google Scholar]
  • 137.Naik S, Madden RM, Lipsitt A, et al. Safety and anti-leukemic activity of CD123-CAR T cells in pediatric patients with AML: preliminary results from a phase 1 trial. Blood. 2022;140(Supplement 1):4584–4585. [Google Scholar]
  • 138.Jin X, Zhang M, Sun R, et al. First-in-human phase I study of CLL-1 CAR-T cells in adults with relapsed/refractory acute myeloid leukemia. J Hematol Oncol. 2022;15:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Jan M, Chao MP, Cha AC, et al. Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker. Proc Natl Acad Sci USA. 2011;108:5009–5014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Jin H-T, Anderson AC, Tan WG, et al. Cooperation of Tim-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc Natl Acad Sci USA. 2010;107:14733–14738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sakuishi K, Apetoh L, Sullivan JM, et al. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J Exp Med. 2010;207:2187–2194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kikushige Y, Shima T, Takayanagi S-i, et al. TIM-3 Is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010;7:708–717. [DOI] [PubMed] [Google Scholar]
  • 143.Brunner AM, Esteve J, Porkka K, et al. Efficacy and Safety of Sabatolimab (MBG453) in Combination with Hypomethylating Agents (HMAs) in Patients (Pts) with Very High/High-Risk Myelodysplastic Syndrome (vHR/HR-MDS) and Acute Myeloid Leukemia (AML): Final Analysis from a Phase Ib Study. Blood. 2021;138(Supplement 1):244–244. [Google Scholar]
  • 144.Zeidan AM, Westermann J, Kovacsovics T, et al. P582: first results of a phase II study (STIMULUS-AML1) Investigating Sabatolimab + Azacitidine + Venetoclax In Patients With Newly Diagnosed Acute Myeloid Leukemia. HemaSphere. 2022;6:481–482. [Google Scholar]
  • 145.Tsai RK, Discher DE. Inhibition of “self” engulfment through deactivation of myosin-II at the phagocytic synapse between human cells. J Cell Biol. 2008;180:989–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Majeti R, Chao MP, Alizadeh AA, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Jaiswal S, Jamieson CHM, Pang WW, et al. CD47 Is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Brierley CK, Staves J, Roberts C, et al. The effects of monoclonal anti-CD47 on RBCs, compatibility testing, and transfusion requirements in refractory acute myeloid leukemia. Transfusion. 2019;59:2248–2254. [DOI] [PubMed] [Google Scholar]
  • 149.Sallman DA, Al Malki MM, Asch AS, et al. Magrolimab in combination with azacitidine in patients with higher-risk myelodysplastic syndromes: final results of a phase Ib study. J Clin Oncol. 2023;41:2815–2826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Daver N, Senapati J, Maiti A, et al. Phase I/II Study of Azacitidine (AZA) with Venetoclax (VEN) and Magrolimab (Magro) in Patients (pts) with Newly Diagnosed (ND) Older/Unfit or High-Risk Acute Myeloid Leukemia (AML) and Relapsed/Refractory (R/R) AML. Blood. 2022;140(Supplement 1):141–144. [Google Scholar]
  • 151.Petrova PS, Viller NN, Wong M, et al. TTI-621 (SIRPalphaFc): a CD47-blocking innate immune checkpoint inhibitor with broad antitumor activity and minimal erythrocyte binding. Clin Cancer Res. 2017;23:1068–1079. [DOI] [PubMed] [Google Scholar]
  • 152.Ansell SM, Maris MB, Lesokhin AM, et al. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin Cancer Res. 2021;27:2190–2199. [DOI] [PubMed] [Google Scholar]
  • 153.An X, Liu J, Ke H, et al. Abstract 2980: HX009, a first-in-class PD1xCD47 BsAb, demonstrated anti-AML activity in PDX models. Cancer Res. 2023;83(7_Supplement):2980–2980. [Google Scholar]
  • 154.Lau NC, Lim LP, Weinstein EG, et al. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294:858–862. [DOI] [PubMed] [Google Scholar]
  • 155.Huang W, Wang WT, Fang K, et al. MIR-708 promotes phagocytosis to eradicate T-ALL cells by targeting CD47. Mol Cancer. 2018;17:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Abu-Thuraia A, Goyette MA, Boulais J, et al. AXL confers cell migration and invasion by hijacking a PEAK1-regulated focal adhesion protein network. Nat Commun. 2020;11:3586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Rochlitz C, Lohri A, Bacchi M, et al. Axl expression is associated with adverse prognosis and with expression of Bcl-2 and CD34 in de novo acute myeloid leukemia (AML): results from a multicenter trial of the Swiss Group for Clinical Cancer Research (SAKK). Leukemia. 1999;13:1352–1358. [DOI] [PubMed] [Google Scholar]
  • 158.Ben-Batalla I, Schultze A, Wroblewski M, et al. Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma. Blood. 2013;122:2443–2452. [DOI] [PubMed] [Google Scholar]
  • 159.Kubasch AS, Peterlin P, Cluzeau T, et al. Efficacy and Safety of Bemcentinib in Patients with Myelodysplastic Syndromes or Acute Myeloid Leukemia Failing Hypomethylating Agents. Blood. 2020;136(Supplement 1):37–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Loges S, Sutamtewagul G, Heuser M, et al. Bemcentinib (Oral AXL Inhibitor) in Combination with Low-Dose Cytarabine Is Well Tolerated and Efficacious in Older Relapsed AML Patients.Updates from the Ongoing Phase II Trial (NCT02488408) and Preliminary Translational Results Indicating Bemcentinib Elicits Anti-AML Immune Responses. Blood. 2021;138(Supplement 1):3410–3410. [Google Scholar]
  • 161.Davids MS, Kim HT, Bachireddy P, et al. Ipilimumab for patients with relapse after allogeneic transplantation. N Engl J Med. 2016;375:143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zeidan AM, Knaus HA, Robinson TM, et al. A Multi-center phase I trial of ipilimumab in patients with myelodysplastic syndromes following hypomethylating agent failure. Clin Cancer Res. 2018;24:3519–3527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Goswami M, Gui G, Dillon LW, et al. Pembrolizumab and decitabine for refractory or relapsed acute myeloid leukemia. J ImmunoTher Cancer. 2022;10:e003392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Zeidner JF, Vincent BG, Ivanova A, et al. Phase II trial of pembrolizumab after high-dose cytarabine in relapsed/refractory acute myeloid leukemia. Blood Cancer Discov. 2021;2:616–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Tschernia NP, Kumar V, Moore DT, et al. Safety and efficacy of pembrolizumab prior to allogeneic stem cell transplantation for acute myelogenous leukemia. Transplant Cell Ther. 2021;27:1021 e1–1021 e5. [DOI] [PubMed] [Google Scholar]
  • 166.Daver N, Garcia-Manero G, Basu S, 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;9:370–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ravandi F, Assi R, Daver N, et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: a single-arm, phase 2 study. Lancet Haematol. 2019;6:e480–e488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Reville PK, Kantarjian HM, Ravandi F, et al. Nivolumab maintenance in high-risk acute myeloid leukemia patients: a single-arm, open-label, phase II study. Blood Cancer J. 2021;11:60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Daver NG, Garcia-Manero G, Konopleva MY, et al. Azacitidine (AZA) with Nivolumab (Nivo), and AZA with Nivo + Ipilimumab (Ipi) in Relapsed/Refractory Acute Myeloid Leukemia: A Non-Randomized, Prospective, Phase 2 Study. Blood. 2019;134(Supplement_1):830–830. [Google Scholar]
  • 170.Daver N, Basu S, Garcia-Manero G, et al. Azacitidine (AZA) with Nivolumab (Nivo), and AZA with Nivo + Ipilimumab (Ipi) in Relapsed/Refractory (R/R) Acute Myeloid Leukemia: Clinical and Immune Biomarkers of Response. Blood. 2020;136(Supplement 1):43–45. [Google Scholar]
  • 171.Fink EC, Ebert BL. The novel mechanism of lenalidomide activity. Blood. 2015;126:2366–2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Surka C, Jin L, Mbong N, et al. CC-90009, a novel cereblon E3 ligase modulator, targets acute myeloid leukemia blasts and leukemia stem cells. Blood. 2021;137:661–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Matyskiela ME, Lu G, Ito T, et al. A novel cereblon modulator recruits GSPT1 to the CRL4CRBN ubiquitin ligase. Nature. 2016;535:252–257. [DOI] [PubMed] [Google Scholar]
  • 174.Zeng AGX, Bansal S, Jin L, et al. A cellular hierarchy framework for understanding heterogeneity and predicting drug response in acute myeloid leukemia. Nat Med. 2022;28:1212–1223. [DOI] [PubMed] [Google Scholar]
  • 175.Diaz Rohena D, Slawin B, Ravikrishnan J, et al. Targeting venetoclax resistant CLL using a protac-based BCL-2/BCL-XL degrader. Blood. 2022;140(Supplement 1):497–498. [Google Scholar]
  • 176.Wang J, Yu X, Gong W, et al. EZH2 noncanonically binds cMyc and p300 through a cryptic transactivation domain to mediate gene activation and promote oncogenesis. Nat Cell Biol. 2022;24:384–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Basheer F, Giotopoulos G, Meduri E, et al. Contrasting requirements during disease evolution identify EZH2 as a therapeutic target in AML. J Exp Med. 2019;216:966–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Sung PJ, Sidoli S, Riedel SS, et al. FLT3 inhibition downregulates EZH2 in AML and promotes myeloid differentiation. Blood. 2021;138(Supplement 1):785–785.33822002 [Google Scholar]
  • 179.Tanaka S, Miyagi S, Sashida G, et al. Ezh2 augments leukemogenicity by reinforcing differentiation blockage in acute myeloid leukemia. Blood. 2012;120:1107–1117. [DOI] [PubMed] [Google Scholar]
  • 180.Nam AS, Chaligne R, Landau DA. Integrating genetic and non-genetic determinants of cancer evolution by single-cell multi-omics. Nature Rev Genet. 2021;22:3–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Turkalj S, Jakobsen NA, Groom A, et al. GTAC enables parallel genotyping of multiple genomic loci with chromatin accessibility profiling in single cells. Cell Stem Cell. 2023;30:722–740. [DOI] [PubMed] [Google Scholar]
  • 182.Goardon N, Marchi E, Atzberger A, et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell. 2011;19:138–152. [DOI] [PubMed] [Google Scholar]
  • 183.Quek L, Otto GW, Garnett C, et al. Genetically distinct leukemic stem cells in human CD34− acute myeloid leukemia are arrested at a hemopoietic precursor-like stage. J Exp Med. 2016;213:1513–1535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Jakobsen NA, Turkalj S, Stoilova B, et al. Single-cell analysis of human clonal hematopoiesis identifies distinct impact of DNMT3A and TET2 mutations on hematopoietic differentiation. Blood. 2022;140(Supplement 1):2227–2228. [Google Scholar]
  • 185.Nakagawa MM, Inagaki R, Kuroda Y, et al. Distinct pathogenesis of clonal hematopoiesis revealed by single cell RNA sequencing integrated with highly sensitive genotyping method. Blood. 2022;140(Supplement 1):2881–2882. [Google Scholar]
  • 186.Nam AS, Dusaj N, Izzo F, et al. Single-cell multi-omics of human clonal hematopoiesis reveals that DNMT3A R882 mutations perturb early progenitor states through selective hypomethylation. Nat Genet. 2022;54:1514–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from HemaSphere are provided here courtesy of Wiley

RESOURCES