Next Article in Journal
Improving the Effect of Cancer Cells Irradiation with X-rays and High-Energy Protons Using Bimetallic Palladium-Platinum Nanoparticles with Various Nanostructures
Previous Article in Journal
The Study of the Extracellular Matrix in Chronic Inflammation: A Way to Prevent Cancer Initiation?
Previous Article in Special Issue
Clinical Impact of Measurable Residual Disease in Acute Myeloid Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia

by
Andrew H. Matthews
1,
Keith W. Pratz
1 and
Martin P. Carroll
2,*
1
Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
2
Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 715 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(23), 5906; https://doi.org/10.3390/cancers14235906
Submission received: 30 August 2022 / Revised: 9 November 2022 / Accepted: 9 November 2022 / Published: 29 November 2022
(This article belongs to the Special Issue New Therapeutic Strategies for Acute Myeloid Leukemia)

Abstract

:

Simple Summary

Despite recent, rapid drug development success for patients with acute myeloid leukemia, distinct molecular and genetic aberrations still confer a poor prognosis. In this review, we explore the preclinical and early clinical development of two promising approaches: disrupting menin signaling leading to cell differentiation or blocking CD47 to unlock the innate immune system. These two approaches may improve treatment for patients with high unmet needs today.

Abstract

After forty years of essentially unchanged treatment in acute myeloid leukemia (AML), innovation over the past five years has been rapid, with nine drug approvals from 2016 to 2021. Increased understanding of the molecular changes and genetic ontology of disease have led to targeting mutations in isocitrate dehydrogenase, FMS-like tyrosine kinase 3 (FLT3), B-cell lymphoma 2 and hedgehog pathways. Yet outcomes remain variable; especially in defined molecular and genetic subgroups such as NPM1 (Nucleophosmin 1) mutations, 11q23/KMT2A rearranged and TP53 mutations. Emerging therapies seek to address these unmet needs, and all three of these subgroups have promising new therapeutic approaches. Here, we will discuss the normal biological roles of menin in acute leukemia, notably in KMT2A translocations and NPM1 mutation, as well as current drug development. We will also explore how CD47 inhibition may move immunotherapy into front-line settings and unlock new treatment strategies in TP53 mutated disease. We will then consider how these new therapeutic advances may change the management of AML overall.

1. Introduction: Continued Need for New Therapies

Every year there are 20,000 new cases of AML diagnosed in the United States [1] and 119,000–352,000 diagnosed worldwide [2]. Induction therapy achieves complete remission (CR) in 60–80% of cases but median survival remains short (8.5 months), and 2-year and 5-year overall survival (OS) rates are only 32% and 24% [3]. OS varies widely by age, performance status, karyotype and molecular changes. While new drug approvals exist for an ever-increasing number of patient subsets, large groups remain with poor prognosis and limited therapy options.
Molecular subsets such as patients with FLT3 and IDH mutations have therapeutic agents approved for use in AML, but many patients lack these lesions and could benefit from development of novel therapeutic approaches. Several subsets, ranging from the common NPM1 mutation (~33% of AML) to the rarer lysine methyltransferase 2A (KMT2A) (5–10% of AML) rearrangements (formerly known as mixed lineage leukemia, MLL), may have a common sensitivity to disruption of the protein menin as these two subsets share a common, targetable final pathway. NPM1 mutated and KMT2A rearrangements are often mutually exclusive, models of both demonstrate common gene expression patterns, preclinical testing shows activity in both settings and clinical trials are underway to see if both groups may benefit. As reviewed elsewhere in this series, [4] TP53 mutant AML remains a largely incurable disease. Although TP53 modulating agents are being studied, [5] new approaches including targeting of CD47, an immunologic “don’t eat me” signal, may provide a way forward for these subtypes of disease. In this review, we will focus on two of the types of therapy not addressed in other articles in this series, Menin inhibition and targeting of CD47 using antibodies.

2. Menin Biology: A Short Introduction

Extensive work has been done characterizing the KMT2A epigenetic complex and its large group of interacting proteins [6] and we will only briefly introduce this complex topic. Menin inhibition has emerged as a promising target for patients with KMT2A fusion proteins as well as NPM1 mutations. 11q23/KMT2A rearrangements occur in 5–10% of patients, [7] are associated with decreased survival [8,9] and may share a common final pathway with NPM1 mutated AML [10]. NPM1 mutation occurs in approximately 35% of all cases and up to 60% of patients with normal karyotype AML [7,11] and prognostic implications are mediated by presence of other mutations including FLT3-internal tandem duplication (FLT3-ITD). NPM1 and FLT3 mutations are now routinely screened at diagnosis with NPM1 mutation and wildtype FLT3 carrying a favorable prognosis [12] and NPM1 mutation with FLT-3 ITD carrying a worse prognosis. Patients with both a FLT3-ITD and NPM1 mutation have poorer prognosis and may undergo transplantation in first remission when possible [13]. Development of menin inhibitors has focused on all of these key subsets, with early clinical trial results expected in the near-term.

3. Menin Inhibition

Menin, a nuclear protein comprised of 610 amino acid residues, is ubiquitously expressed in various organs. Menin helps regulate tissue-specific gene expression via interaction with chromatin regulators and transcription factors. Chromatin immunoprecipitation studies show menin interacts with thousands of human gene promoters. By linking transcription-factor function to histone-modification pathways, menin can act as a global regulator of transcription with different functions in different tissues [14,15]. Intriguingly, menin suppresses tumorigenesis in endocrine organs, with its loss resulting in both the hereditary MEN1 syndrome and sporadic parathyroid adenomas, insulinomas, gastrinomas and lung carcinoid tumors [16]. However, menin can also be an essential leukemogenic cofactor for KMT2A fusion proteins [17]. Endogenous KMT2A, is the human homolog of the drosophila gene trithorax which is essential for murine mid-gestational development [18]. The wildtype KMT2A protein assembles into a protein complex, that in conjunction with transcription factors, marks gene promoters and regulates gene expression. KMT2A protein is a member of a large multiprotein complex that interacts with chromatin and normally acts as a histone H3 lysine 4 (H3K4) methyltransferase, a function critical for epigenetic modification and transcription regulation [19,20]. Both wild type [21,22,23] and mutant forms of the KMT2A protein [24,25] play a role in regulating stem cell phenotype through regulation of homeobox (HOX) genes. Chromosomal translocations frequently disrupt transcription factors and lead to leukemogenesis. These translocations can form fusion proteins with novel functions such as new binding partners or changes in interactions with prior binding partners. These changes impact epigenetic regulation and affect hematopoietic differentiation. Most KMT2A rearrangements are balanced translocations resulting in the production of in-frame gain-of-function fusion oncoproteins. Importantly, these chromosomal rearrangements alone may be sufficient to generate a full leukemic phenotype, [26] as seen with KMT2A fusion proteins in murine models [27,28,29]. KMT2A chromosomal alterations have been extensively studied in mixed lineage leukemia for more than twenty years [30]. KMT2A encoded wildtype (wt)-KMT2A requires interaction with menin to maintain HOX expression and self-renewal properties of hematopoietic progenitors [31,32,33,34]. Mechanistic mouse models show KMT2A fusion proteins induce leukemic transformation from hematopoietic stem cells in a dose-dependent manner [35,36,37]. As epigenetic regulation becomes dysfunctional a stereotypical pattern of novel target gene expression emerges including overexpression of FLT3, [38,39] PBX3, [40] HOXA9 and MEIS1 [21,41]. Transcription of the latter two genes promotes self-renewal through stem-cell like transcription programs [29,37,42] and is critically dependent on the KMT2A-fusion protein binding to menin [17,43].
Balanced translocations that initiate AML, like KMT2Ar—or CBFB-MYH11, RUNX1T1 and PML-RARA—are often mutually exclusive with NPM1 mutations [44,45]. Patients with NPM1 mutated AML commonly have co-mutated DNMT3A and/or FLT3-ITD and are associated with a characteristic gene expression profile including downregulation of CD34 and overexpression of HOXA cluster genes [46]. As noted above this same pattern of HOXA 7/9/10 and MEIS1 gene overexpression is also seen in KMT2A rearrangements. The Armstrong lab demonstrated that NPM1 mutant leukemia is dependent on menin binding to wild type KMT2A and loss of KMT2A-menin abrogates the leukemogenic effect of NPM1 mutations [47]. These observations raise the possibility of disrupting KMT2A protein interactions in both patients with KMT2A fusion proteins and those with NPM1 mutations. There may be a common, shared, sufficient AML pathway vulnerable to menin disruption.

4. Targeting Menin in Acute Leukemia

Small molecules disrupting KMT2A and menin interactions show substantial antileukemic efficacy in cellular and murine models of KMT2A-rearranged (KMT2Ar) AML [17,48]. The KMT2A fusion complex is a large multi-protein complex and disruption of the menin-KMT2A fusion protein complex restores histone methylation, gene expression and permits leukemic cell differentiation. Inhibitors thus lead to improved survival in genetically engineered mouse and xenotransplantation models [48,49,50]. Small molecule inhibitors produce the same effects as deleting menin in KMT2Ar models using a CRISPR/cas9 model system [47]. Menin inhibitors also appear to lead to decreased expression of HOXa/b targets, including MEIS1, and lead to differentiation in NPM1/DNMT3a mutant knock-in mice and leukemia cell lines [51,52]. Based on these promising preclinical data, development has moved into clinical trials focusing on KMT2Ar and NPM1 mutated leukemias.
Although many of the details of the KMT2A complex have been understood for some time, developing small molecule inhibitors of this epigenetic complex has progressed slowly [53,54,55]. The focus has now turned to disrupting menin binding to either KMT2Ar fusion proteins, the wildtype complex or both (Figure 1). After years of optimization and drug selection, four drugs that inhibit MLL-menin binding have recently entered clinical testing as single agents for AML (K0539 (NCT04067336); SNDX-5613 (NCT04065399); JNJ-75276617 (NCT04811560); DS-1594b (NCT04752163); BMF-219 (NCT05153330). After safety and dose optimization are established, efficacy testing will follow in patients with KMT2A-rearrangements or NPM1 mutations.
Preliminary data from the phase I/II KOMET-001 trial (NCT04067336) has been presented. Among patients in four dosing cohorts, median age was 67 years-old, a median of 3 prior lines of therapy had been received and 2 of 12 achieved a complete response (one patient had SET2D/RUNX1 and the other NPM1 and KMT2D mutations). There were no discontinuations due to adverse events and grade 3 or higher adverse events included elevated lipase, pancreatitis, neutropenia, tumor lysis syndrome and deep vein thrombosis each occurring in 1 patient each (8.3%) while grade 1 or 2 adverse events included nausea, rash and diarrhea [56,57]. The phase I/II AUGMENT-101 trial of SNDX-5613 has preliminary results presented in abstract form from 45 evaluable patients showed an objective response rate of 44%. In the 35 patient subgroup with MLL rearrangement, the ORR was 49% and in the 10 patients with NPM1 mutation the ORR was 30% (of those with a CR, 70% achieved minimal residual disease negative state). Patients lacking either KMT2Ar or NPM1 mutations did not respond [58,59]. There were no treatment discontinuations due to adverse events, however 13% (n = 7) of patients experienced grade 3 QTc prolongation. These trials and others are ongoing (Table 1) but the preliminary conclusion is that these drugs will be tolerated, although QTc monitoring will be important, and they potentially have single agent activity in patients with MLL-rearranged or NPM1 mutant AML.

5. Future Steps in Development

Depending on monotherapy activity and tolerability, combination therapy is likely to follow. Early preclinical data favor combinations targeting FLT3-ITD. Recent data suggests NPM1 and KMT2A translocations might share significant cooperativity with FLT3 in leukemogenesis. In particular MEIS1 is a critical upstream regulator of FLT3, [60] therefore disrupting menin may decrease MEIS1 protein expression and FLT3 protein activity. In fact menin inhibition led to a profound downregulation of FLT3 expression in AML cell lines, [47] opening up the possibility of combination therapy in NPM1 mutated, FLT3-ITD mutated AML. Additionally menin complex inhibitors downregulated and dephosphorylated FLT3, FLT3-ITD inhibitor and menin inhibitor combination therapy led to a left shift of the 50% inhibitory concentration for combination components over the individual drugs, and a murine model of KMT2A-rearranged FLT3-ITD mutated AML showed improved survival [61,62]. Other preclinical evidence has demonstrated potential synergy by disrupting menin and inhibiting BCL-2 or CDK6 with venetoclax or abemaciclib, respectively, in both AML cell lines as well as patient derived AML cell xenografts [63].
Competing agents are also targeting the KMT2A machinery and downstream pathways. Numerous protein complexes act as binding partners for KMT2Ar associated fusion proteins and several including PAFc, DOT1L and SEC have all been therapeutic targets [64]. Success has yet to be realized. In recent years, pinometostat, an inhibitor of DOT1L failed to show more than modest activity with two complete responses among 51 patients [65]. Still other groups are targeting the presumed shared downstream pathway components, such as spleen tyrosine kinase (SYK) [66]. Entospletinib, an oral SYK inhibitor, has completed phase Ib/II trial (NCT02343939) and showed favorable response rates when combined with induction chemotherapy in AML patients with KMT2A translocations, NPM1 mutations and FLT-3 ITD mutations all with a shared HOXA9/MEIS1 signature [67]. The planned phase 3 trial will be targeting patients with newly diagnosed NPM1 mutated AML and may pursue accelerated approval on the basis of measurable residual disease negative complete response as the primary endpoint. Similar to menin inhibitors, SYK inhibitors are being examined in combination with FLT3-ITD inhibitors as well as venetoclax and azacitidine in patients with newly diagnosed AML with NPM1 and FLT3 mutations. Significant new work has defined the various KMT2Ar fusion protein containing complexes and their discrete interacting proteins; preclinical work suggests that combinatorial inhibition of menin interactions with, for example, ENL inhibition, may have cooperative or synergistic effects on leukemic cell differentiation. This exciting area of research has been recently reviewed [68].
Overall, menin inhibitors represent a promising target for KMT2A translocated and NPM1 mutated AML, perhaps especially patients with NPM1 mutated FLT3-ITD high AML. These subgroups represent an ongoing unmet need despite several new agents for AML. The near-term results from phase I/II monotherapy trials will elucidate future development. Combination therapy with either traditional chemotherapy, FLT3 inhibitors or other epigenetic modifiers may be an exciting path forward as our understanding of the basic biology of leukemic transformation improves.

6. Anti-CD47 Antibodies

Immunotherapy has changed the clinical landscape of solid oncology and is reshaping lymphoma and multiple myeloma treatment. Unfortunately, PD-1 and PD-L1 inhibitor immunotherapies have not been transportable to relapsed/refractory AML [69,70]. Numerous groups are trying to advance therapy earlier in the disease process or combine immunotherapy with hypomethylating agents although early studies have been mixed [71]. While other immunotherapies beyond checkpoint inhibitors have garnered success in lymphoma, myeloma, and even acute lymphoblastic leukemia, progress has stalled in AML. Monoclonal antibodies, T cell engager antibodies, alloreactive NK and CART cell therapy, immune checkpoint blockade beyond PD-1/PD-L1 or CTLA4 such as TIM3 are still actively being investigated [72]. Cellular immunotherapies like autologous NK and CART cells, have thus far been of limited relevance to AML. Even when a target exists, all immunotherapy approaches seem to face hurdles of collateral hematopoietic damage or inactivity. While standard immune checkpoint inhibitors or cell therapies have not yet been successful in AML, strategies to unlock the innate immune response and may represent a much needed new strategy against poor risk AML such as those with TP53 mutations.
Blocking cluster of differentiation (CD) 47 may provide a path forward. CD47 is a nearly ubiquitous expressed cell surface protein belonging to the immunoglobulin family. It is a heavily glycosylated 50Kd cell surface protein with an extracellular N-terminal IgV domain, five transmembrane domains, and a short C-terminal cytoplasmic tail. CD47 is the ligand for signal-regulatory protein (SIRP)α, an inhibitory receptor expressed on myeloid cells. SIRPα is a transmembrane protein whose cytoplasmic domain contains immunoreceptor tyrosine-based inhibition motifs which when phosphorylated recruit phosphatases and inhibits the activity of non-muscle myosin IIa. This decreases the ability of prophagocytic signals on tumor cells to trigger phagocytosis by macrophages [73]. As a cell surface ligand CD47 is present in low levels on nearly all cells [74]. For example, it is a marker of self on red blood cells [75] and expression decreases with age leading to splenic clearance of senescent red blood cells [76]. By interacting with SIRPα on macrophages and dendritic cells it sends a potent “don’t eat me” anti-phagocytosis message signal. This signal serves a role in normal immune tolerance, autoimmunity and both solid [77,78,79,80] and hematological malignancy [81,82,83,84]. It also acts as immune checkpoint with overexpression of CD47 usually contributing to innate and adaptive immune escape [85,86,87,88]. Typically upon cell damage or stress, cells will increase expression of prophagocytic signals, such as calreticulin, that leads to cell removal [89,90]. These signals counteract the phagocytosis checkpoint from CD47-SIRPα axis.
Preclinical studies have shown that CD47 blockade is not sufficient for phagocytosis of tumor cells [91]. For anti-tumor activity, another co-stimulatory signal is needed. Tumor cells may need to express additional pro-phagocytic signals such as SLAMF7 with Mac-1 [92] and calreticulin induced by azacitidine, [93] or additional activation of macrophages via Fcγ receptor activation by the Fc region of a monoclonal antibody targeting CD47 or by a second agent targeting leukemia cells (e.g., CD33). Preclinical findings have also shown that F(ab’)2 fragments of a monoclonal antibody against CD47 showed no apoptotic effect on CD34+ hematopoietic progenitor/stem or human endothelial cells in leukemia xenograft mouse models [94]. Thus the selectivity of CD47 antibodies relies on the balance of CD47 as well as prophagocytic signals on normal cells and leukemia cells (Figure 2).

7. Targeting CD47 in Acute Myeloid Leukemia

CD47 is highly expressed in AML cells including leukemic stem cells [95] with approximately 25–30% of patients across multiple AML cohorts having high levels of CD47 mRNA expression in peripheral blood blasts [96]. Interestingly, CD47 mRNA expression varies across cytogenic and molecular subgroups with lower expression in cases with t(8;21), a favorable risk translocation, and higher expression in cases harboring FLT3-ITD mutations. High CD47 expression has been shown to be an independent prognostic factor for poor overall survival in AML patient cohorts [96]. Thus CD47 expression is thought to have both prognostic and therapeutic implications in AML.
Blocking CD47 may be most effective in certain subsets of AML, where the balance of pro-phagocytic and anti-phagocytic signals is altered. For example, TP53 mutant patients have been shown to have an immune anergic profile, immunosuppressive tumor microenvironment and PD-L1 is upregulated on leukemic stem cells in TP53 mutant AML and MDS [97,98]. Furthermore, p53-null tumors appear able to reeducate myeloid cells such that they attenuate T-cell responses with decreased CD4+ and CD8+ T cell proliferation [99]. TP53 mutational status may also be critical within the microenvironment itself. In p53 null mice, the size of tumor xenografts are greatly increased and correlate with the presence of myeloid derived suppressor cells, regulatory T cells, and a loss of effector function [100,101]. CD47 blockade may therefore be able to exploit how TP53 mutations reshape the local innate and adaptive immune response.
Multiple groups established monoclonal antibodies against CD47 that enabled phagocytosis of AML leukemic stem cells in vitro and inhibited their growth in leukemia cell lines and mice models [95,96]. Thereafter, a humanized IgG4 anti-CD47 antibody (magrolimab) demonstrated leukemic eradication and long term survival in xenografted mice [102]. Agents quickly moved into clinical trial settings in multiple malignancies. The first agent targeting CD47 entered clinical trials in 2014, there are now over 20 agents in active clinical development in over 30 trials spanning multiple indications and cancer types (Table 2 lists clinical trial agents in AML). Groups have also begun to target CD47 with recombinant fusion proteins (e.g., TTI-662 a fully human recombinant SIRPαFc fusion protein) [103] and bispecific antibodies [104].
Magrolimab, as the first-in-class clinical trial candidate, has provided early lessons. In a Phase 1 monotherapy trial, magrolimab failed to show any CR or CR with incomplete count recovery (CRi) in patients with relapsed and/or refractory AML [105]. Another early lead agent CC-90002 showed a similar low single agent activity, 36% of patients developed anti-drug antibodies and adverse events included disseminated intravascular coagulation, purpura, heart failure and acute respiratory failure presumably from hemagglutination [107]. The focus has shifted to combination therapies earlier in the treatment course and selecting candidates with low preclinical signals of hemagglutination or hemolysis.
Magrolimab has since shown promising overall response rates in combination with azacitidine for AML and MDS. In a phase Ib trial of untreated AML, among 34 evaluable patients overall response rate was 65% (44% CR, 12% CRi, 3% partial response and 6% morphological leukemia free state, 32% with stable disease. In an interim analysis of 72 patient with TP53 mutated AML CR was 33% and CRi was 8% with median time to CR of 3 months. 45% (14/31) of evaluable patients with any level of response achieved negative MRD by flow cytometry [108]. As the response of TP53 AML to conventional treatment is poor, these clinical trial results have lead some investigators to focus anti-CD47 trials on patients with TP53 mutant AML. Whether TP53 mutation status will be a robust biomarker remains to be determined. Magrolimab has since moved into phase III trials. Other CD47 directed agents are not as far along in clinical development but reflect strategies to either enhance immune activity by coupling CD47 blockade with an activating signal or decrease the risk of hemolysis and hemagglutination.
Toxicity of these agents may continue to present challenges but we are learning more about managing such toxicity. Adverse effects could include hemagglutination and off-target myelosuppression, given the role of CD47 in preventing programmed red cell removal [75]. In the phase 1b magrolimab trial, although anemia was seen in 38% of patients, the anemia predominantly occurred in cycle 1 with a return to baseline by cycle 2 and improvement over time associated with hematologic response. There was a decline in Hb levels in all patients (median Hb change, −1.0 g/dL); eighteen developed a newly positive direct antiglobulin tests without evidence of hemolysis and there were increased transfusion requirements among nineteen patients with relapsed/refractory AML [109]. This has led to so called RBC pruning strategies [110] which has been implemented in AML trials with a magrolimab priming dose-escalation regimen (escalating 1–30 mg/kg given intravenously weekly followed by 30 mg/kg every 2-week dosing in cycle 3 and beyond) to try to mitigate on-target anemia [105].

8. Future Development

Assuming hemolysis and hemagglutination are not limiting, numerous potential combinations could further boost efficacy. Although the PD-(L)1 pathway has been disappointing in late-stage AML perhaps combination therapy could be feasible. Preclinical evidence is strongest for the combination of azacitidine and anti-CD-47 antibodies, with the combination leading to higher phagocytosis of AML cells in vitro, induction of proposed pro-phagocytic signals including increased calreticulin, long-term remission in AML xenograft mice models [111]. There are also compelling theoretical combinations to further activate the immune system, such as TIM-3 or CD200 targeting combinations, or to increase the longevity of cellular therapy. While there may be strong mechanistic rationale for other preclinical combinations, such as combing PD-(L)1 axis therapeutics or chimeric antigen T cell receptor therapies, they have yet to be robustly investigated and reported.
Clinical results may very well start to return before robust preclinical evidence. Given TP53-mutated AML’s altered immune profile, spanning both innate and adaptive immunity, a trial of atezolizumab, an anti-PD-L1 antibody, and magrolimab will assess activity in relapsed or refractory AML patients (NCT03922477). Additionally CD-47 targeting antibodies could be combined with other monoclonal antibodies such as gemtuzumab ozogamicin. Alternatively, combination with targeted therapy might be feasible if toxicities are not overlapping. Although VIALE-A showed no statistically significant survival benefit for venetoclax and azacitadine versus azacitidine in TP53 mutated patients, [112] synergism of CD47 blockade and Bcl-2 targeting therapy may be possible as seen in in chronic lymphocytic leukemia [113].
CD47 blockade remains a promising immune checkpoint in AML, and therapies may offer new approaches for hard-to-treat patient populations like those with TP53 mutations. Magrolimab in combination with azacitidine has provided proof of concept, it remains to be seen if mutational profiling will be sufficient to identify patients who will respond to therapy or if immunological biomarkers will have to be incorporated into trials. Additionally, novel combinations have entered clinical trials and may further increase efficacy. Ongoing large trials will determine if early signals of hemolysis and hemagglutination can be overcome with dose titration or if instead earlier pipeline agents will be needed to overcome this hurdle.

9. Conclusions

As noted above and in other articles in this series, AML therapy has advanced significantly in the last few years but remains challenging. Ongoing basic studies continue to both elucidate the basic biology of leukemic transformation and the immunology of the immune escape of AML cells leading to more targeted and less toxic approaches to AML therapy. The days of high dose induction or no therapy are thankfully behind us. Targeting CD47 and menin inhibitors, if successful clinically, would have a major impact on subsets of AML patients with poor outcomes. Menin disruption represents more than twenty years of preclinical work and appears to have the strongest rationale in KMT2A rearranged AML but may hold additional potential in NPM1 mutated AML and even FLT3-ITD patients. Early clinical trials have confirmed an efficacy signal even as a monotherapy and beyond QTc prolongation no other major safety signals have yet emerged. On the other hand, CD47 blockade may finally bring immunotherapy to the AML treatment landscape. To date, this strategy appears to be meeting the major challenge of targeting a marker restricted to leukemia cells. Larger trials will attempt to confirm the promise of improved combination therapy efficacy while sparing hematopoietic cells and erythrocytes. These two treatment approaches may reshape management of intermediate and high-risk AML.

Author Contributions

Conceptualization, A.H.M., K.W.P. and M.P.C.; resources, M.P.C.; writing—original draft preparation, A.H.M.; writing—review and editing, K.W.P. and M.P.C.; visualization, A.H.M.; supervision, M.P.C.; project administration, M.P.C.; funding acquisition, M.P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

K.W.P. has received research funding from AbbVie, Agios, Daiichi Sankyo, Millennium. He has been involved on advisory committees for AbbVie, Astellas, Boston BioMedical, BMS, Celgene, Novartis, Jazz Pharmaceuticals, and Servier. M.P.C. has been involved as a consultant for Janssen Pharmaceuticals and on advisory committee for Cartogrpahy Biosciences. A.H.M. declares no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2021. CA A Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Yi, M.; Li, A.; Zhou, L.; Chu, Q.; Song, Y.; Wu, K. The global burden and attributable risk factor analysis of acute myeloid leukemia in 195 countries and territories from 1990 to 2017: Estimates based on the global burden of disease study 2017. J. Hematol. Oncol. 2020, 13, 72. [Google Scholar] [CrossRef] [PubMed]
  3. Shallis, R.M.; Wang, R.; Davidoff, A.; Ma, X.; Zeidan, A.M. Epidemiology of acute myeloid leukemia: Recent progress and enduring challenges. Blood Rev. 2019, 36, 70–87. [Google Scholar] [CrossRef] [PubMed]
  4. Shallis, R.M.; Bewersdorf, J.P.; Stahl, M.F.; Halene, S.; Zeidan, A.M. Are we moving the needle for patients with TP53-mutated acute myeloid leukemia? Cancers 2022, 14, 2434. [Google Scholar] [CrossRef]
  5. Cluzeau, T.; Sebert, M.; Rahmé, R.; Cuzzubbo, S.; Lehmann-Che, J.; Madelaine, I.; Peterlin, P.; Bève, B.; Attalah, H.; Chermat, F.; 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. [Google Scholar] [CrossRef]
  6. Uckelmann, H.J.; Armstrong, S.A. Chromatin complexes maintain self-renewal of myeloid progenitors in AML: Opportunities for therapeutic intervention. Stem Cell Rep. 2020, 15, 6–12. [Google Scholar] [CrossRef] [PubMed]
  7. Ley, T.J.; Miller, C.; Ding, L.; Raphael, B.J.; Mungall, A.J.; Robertson, A.; Hoadley, K.; Triche, T.J., Jr.; Laird, P.W.; Baty, J.D.; et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar]
  8. Szczepański, T.; Harrison, C.J.; van Dongen, J.J.M. Genetic aberrations in paediatric acute leukaemias and implications for management of patients. Lancet Oncol. 2010, 11, 880–889. [Google Scholar] [CrossRef]
  9. Balgobind, B.V.; Raimondi, S.C.; Harbott, J.; Zimmermann, M.; Alonzo, T.A.; Auvrignon, A.; Beverloo, H.B.; Chang, M.; Creutzig, U.; Dworzak, M.N.; et al. Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: Results of an international retrospective study. Blood 2009, 114, 2489–2496. [Google Scholar] [CrossRef] [Green Version]
  10. Muñoz, L.; Nomdedéu, J.F.; Villamor, N.; Guardia, R.; Colomer, D.; Ribera, J.M.; Torres, J.P.; Berlanga, J.J.; Fernández, C.; Llorente, A.; et al. Acute myeloid leukemia with MLL rearrangements: Clinicobiological features, prognostic impact and value of flow cytometry in the detection of residual leukemic cells. Leukemia 2003, 17, 76–82. [Google Scholar] [CrossRef] [Green Version]
  11. Thiede, C.; Koch, S.; Creutzig, E.; Steudel, C.; Illmer, T.; Schaich, M.; Ehninger, G.; Dsil, F.T.D.S.L. Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML). Blood 2006, 107, 4011–4020. [Google Scholar] [CrossRef]
  12. Liu, Y.; He, P.; Liu, F.; Shi, L.; Zhu, H.; Zhao, J.; Wang, Y.; Cheng, X.; Zhang, M. Prognostic significance of NPM1 mutations in acute myeloid leukemia: A meta-analysis. Mol. Clin. Oncol. 2014, 2, 275–281. [Google Scholar] [CrossRef] [Green Version]
  13. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [Green Version]
  14. Scacheri, P.C.; Davis, S.; Odom, D.T.; Crawford, G.E.; Perkins, S.; Halawi, M.J.; Agarwal, S.K.; Marx, S.J.; Spiegel, A.M.; Meltzer, P.S.; et al. Genome-wide analysis of menin binding provides insights into MEN1 tumorigenesis. PLOS Genet. 2006, 2, e51. [Google Scholar] [CrossRef] [Green Version]
  15. Dreijerink, K.M.; Höppener, J.W.; Timmers, H.M.; Lips, C.J. Mechanisms of disease: Multiple endocrine neoplasia type 1-relation to chromatin modifications and transcription regulation. Nat. Clin. Pract. Endocrinol. Metab. 2006, 2, 562–570. [Google Scholar] [CrossRef]
  16. Marx, S.J.; Agarwal, S.K.; Kester, M.B.; Heppner, C.; Kim, Y.S.; Skarulis, M.C.; James, L.A.; Goldsmith, P.K.; Saggar, S.K.; Park, S.Y.; et al. Multiple endocrine neoplasia type 1: Clinical and genetic features of the hereditary endocrine neoplasias. Recent Prog. Horm. Res. 1999, 54, 397–438. [Google Scholar]
  17. Yokoyama, A.; Somervaille, T.C.P.; Smith, K.S.; Rozenblatt-Rosen, O.; Meyerson, M.; Cleary, M.L. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005, 123, 207–218. [Google Scholar] [CrossRef]
  18. Mbangkollo, D.; Burnett, R.; McCabe, N.; Thirman, M.; Gill, H.; Yu, H.; Rowley, J.D.; Diaz, M.O. The human MLL gene: Nucleotide sequence, homology to the Drosophila trx zinc-finger domain, and alternative splicing. DNA Cell Biol. 1995, 14, 475–483. [Google Scholar] [CrossRef]
  19. Miller, T.; Krogan, N.J.; Dover, J.; Erdjument-Bromage, H.; Tempst, P.; Johnston, M.; Greenblatt, J.F.; Shilatifard, A. COMPASS: A complex of proteins associated with a trithorax-related SET domain protein. Proc. Natl. Acad. Sci. USA 2001, 98, 12902–12907. [Google Scholar] [CrossRef] [Green Version]
  20. Rowley, J.D. The critical role of chromosome translocations in human leukemias. Annu. Rev. Genet. 1998, 32, 495–519. [Google Scholar] [CrossRef]
  21. Jude, C.D.; Climer, L.; Xu, D.; Artinger, E.; Fisher, J.K.; Ernst, P. Unique and independent roles for MLL in adult hematopoietic stem cells and progenitors. Cell Stem Cell 2007, 1, 324–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Ernst, P.; Fisher, J.K.; Avery, W.; Wade, S.; Foy, D.; Korsmeyer, S.J. Definitive hematopoiesis requires the mixed-lineage leukemia gene. Dev. Cell 2004, 6, 437–443. [Google Scholar] [CrossRef] [PubMed]
  23. Alharbi, R.A.; Pettengell, R.; Pandha, H.S.; Morgan, R. The role of HOX genes in normal hematopoiesis and acute leukemia. Leukemia 2013, 27, 1000–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Yagi, H.; Deguchi, K.; Aono, A.; Tani, Y.; Kishimoto, T.; Komori, T. Growth disturbance in fetal liver hematopoiesis of Mll-mutant mice. Blood 1998, 92, 108–117. [Google Scholar] [CrossRef] [PubMed]
  25. Hess, J.L.; Yu, B.D.; Li, B.; Hanson, R.; Korsmeyer, S.J. Defects in yolk sac hematopoiesis in Mll-null embryos. Blood 1997, 90, 1799–1806. [Google Scholar] [CrossRef] [Green Version]
  26. Aplan, P.D. Chromosomal translocations involving the MLL gene: Molecular mechanisms. DNA Repair 2006, 5, 1265–1272. [Google Scholar] [CrossRef] [Green Version]
  27. Martin, M.E.; Milne, T.A.; Bloyer, S.; Galoian, K.; Shen, W.; Gibbs, D.; Brock, H.W.; Slany, R.; Hess, J.L. Dimerization of MLL fusion proteins immortalizes hematopoietic cells. Cancer Cell 2003, 4, 197–207. [Google Scholar] [CrossRef] [Green Version]
  28. Cozzio, A.; Passegué, E.; Ayton, P.M.; Karsunky, H.; Cleary, M.L.; Weissman, I.L. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003, 17, 3029–3035. [Google Scholar] [CrossRef] [Green Version]
  29. Krivtsov, A.V.; Twomey, D.; Feng, Z.; Stubbs, M.C.; Wang, Y.; Faber, J.; Levine, J.E.; Wang, J.; Hahn, W.C.; Gilliland, D.G.; et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 2006, 442, 818–822. [Google Scholar] [CrossRef]
  30. Ayton, P.M.; Cleary, M.L. Molecular mechanisms of leukemogenesis mediated by MLL fusion proteins. Oncogene 2001, 20, 5695–5707. [Google Scholar] [CrossRef] [Green Version]
  31. Brunetti, L.; Gundry, M.C.; Sorcini, D.; Guzman, A.G.; Huang, Y.-H.; Ramabadran, R.; Gionfriddo, I.; Mezzasoma, F.; Milano, F.; Nabet, B.; et al. Mutant NPM1 maintains the leukemic state through HOX expression. Cancer Cell 2018, 34, 499–512. [Google Scholar] [CrossRef] [Green Version]
  32. Milne, T.A.; Hughes, C.M.; Lloyd, R.; Yang, Z.; Rozenblatt-Rosen, O.; Dou, Y.; Schnepp, R.W.; Krankel, C.; LiVolsi, V.A.; Gibbs, D.; et al. Menin and MLL cooperatively regulate expression of cyclin-dependent kinase inhibitors. Proc. Natl. Acad. Sci. USA 2005, 102, 749–754. [Google Scholar] [CrossRef]
  33. Novotny, E.; Compton, S.; Liu, P.P.; Collins, F.S.; Chandrasekharappa, S.C. In vitro hematopoietic differentiation of mouse embryonic stem cells requires the tumor suppressor menin and is mediated by Hoxa9. Mech. Dev. 2009, 126, 517–522. [Google Scholar] [CrossRef]
  34. Hughes, C.M.; Rozenblatt-Rosen, O.; Milne, T.A.; Copeland, T.D.; Levine, S.S.; Lee, J.C.; Hayes, D.N.; Shanmugam, K.S.; Bhattacharjee, A.; Biondi, C.A.; et al. Menin Associates with a trithorax family histone methyltransferase complex and with the Hoxc8 locus. Mol. Cell 2004, 13, 587–597. [Google Scholar] [CrossRef]
  35. Schwaller, J. Learning from mouse models of MLL fusion gene-driven acute leukemia. Biochim. Biophys. Acta Gene Regul. Mech. 2020, 1863, 194550. [Google Scholar] [CrossRef]
  36. Corral, J.; Lavenir, I.; Impey, H.; Warren, A.J.; Forster, A.; Larson, T.A.; Bell, S.; McKenzie, A.N.; King, G.; Rabbitts, T. An Mll–AF9 fusion gene made by homologous recombination causes acute leukemia in chimeric mice: A method to create fusion oncogenes. Cell 1996, 85, 853–861. [Google Scholar] [CrossRef] [Green Version]
  37. Chen, W.; Li, Q.; Hudson, W.A.; Kumar, A.; Kirchhof, N.; Kersey, J.H. A murine Mll-AF4 knock-in model results in lymphoid and myeloid deregulation and hematologic malignancy. Blood 2006, 108, 669–677. [Google Scholar] [CrossRef] [Green Version]
  38. Kohlmann, A.; Schoch, C.; Dugas, M.; Schnittger, S.; Hiddemann, W.; Kern, W.; Haferlach, T. New insights into MLL gene rearranged acute leukemias using gene expression profiling: Shared pathways, lineage commitment, and partner genes. Leukemia 2005, 19, 953–964. [Google Scholar] [CrossRef]
  39. Armstrong, S.A.; Staunton, J.E.; Silverman, L.B.; Pieters, R.; Boer, M.D.; Minden, M.D.; Sallan, S.E.; Lander, E.S.; Golub, T.R.; Korsmeyer, S.J. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nat. Genet. 2002, 30, 41–47. [Google Scholar] [CrossRef]
  40. Li, Z.; Chen, P.; Su, R.; Hu, C.; Li, Y.; Elkahloun, A.G.; Zuo, Z.; Gurbuxani, S.; Arnovitz, S.; Weng, H.; et al. PBX3 and MEIS1 cooperate in hematopoietic cells to drive acute myeloid leukemias characterized by a core transcriptome of the MLL—Rearranged DISEASE. Cancer Res. 2016, 76, 619–629. [Google Scholar] [CrossRef] [Green Version]
  41. Schuettengruber, B.; Martinez, A.-M.; Iovino, N.; Cavalli, G. Trithorax group proteins: Switching genes on and keeping them active. Nat. Rev. Mol. Cell Biol. 2011, 12, 799–814. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, W.; Kumar, A.R.; Hudson, W.A.; Li, Q.; Wu, B.; Staggs, R.A.; Lund, E.A.; Sam, T.N.; Kersey, J.H. Malignant transformation initiated by Mll-AF9: Gene dosage and critical target cells. Cancer Cell 2008, 13, 432–440. [Google Scholar] [CrossRef] [PubMed]
  43. Yokoyama, A.; Cleary, M.L. Menin critically links MLL proteins with LEDGF on cancer-associated target genes. Cancer Cell 2008, 14, 36–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Falini, B.; Mecucci, C.; Tiacci, E.; Alcalay, M.; Rosati, R.; Pasqualucci, L.; La Starza, R.; Diverio, D.; Colombo, E.; Santucci, A.; et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N. Engl. J. Med. 2005, 352, 254–266. [Google Scholar] [CrossRef] [PubMed]
  45. Brunangelo, F.; Cristina, M.; Giuseppe, S.; Francesco Lo, C.; Daniela, D.; Patrick, B.; Fabrizio, P.; Marco, M.; Maria Paola, M.; Stefano, P.; et al. NPM1 mutations and cytoplasmic nucleophosmin are mutually exclusive of recurrent genetic abnormalities: A comparative analysis of 2562 patients with acute myeloid leukemia. Haematologica 2008, 93, 439–442. [Google Scholar]
  46. Collins, C.T.; Hess, J.L. Deregulation of the HOXA9/MEIS1 axis in acute leukemia. Curr. Opin. Hematol. 2016, 23, 354–361. [Google Scholar] [CrossRef]
  47. Kühn, M.W.M.; Song, E.; Feng, Z.; Sinha, A.; Chen, C.-W.; Deshpande, A.J.; Cusan, M.; Farnoud, N.; Mupo, A.; Grove, C.; et al. Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia. Cancer Discov. 2016, 6, 1166–1181. [Google Scholar] [CrossRef] [Green Version]
  48. Borkin, D.; He, S.; Miao, H.; Kempinska, K.; Pollock, J.; Chase, J.; Purohit, T.; Malik, B.; Zhao, T.; Wang, J.; et al. Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell 2015, 27, 589–602. [Google Scholar] [CrossRef] [Green Version]
  49. Krivtsov, A.V.; Evans, K.; Gadrey, J.Y.; Eschle, B.K.; Hatton, C.; Uckelmann, H.J.; Ross, K.N.; Perner, F.; Olsen, S.N.; Pritchard, T.; et al. A menin-MLL inhibitor induces specific chromatin changes and eradicates disease in models of MLL-rearranged leukemia. Cancer Cell 2019, 36, 660–673. [Google Scholar] [CrossRef]
  50. Numata, M.; Shiroishi, M.; Yoshikawa, K.; Haginoya, N.; Hirata, T.; Takata, Y.; Nagase, R.; Takashima, K.; Kurimoto, A.; Tanzawa, F.; et al. Development and characterization of a novel orally bioavailable menin-MLL inhibitor for treatment of acute leukemia patients with MLL-rearrangement or NPM1 mutation. AACR Annu. Meet. 2021, 2021, 1132. [Google Scholar]
  51. Uckelmann, H.J.; Kim, S.M.; Wong, E.M.; Hatton, C.; Giovinazzo, H.; Gadrey, J.Y.; Krivtsov, A.V.; Rücker, F.G.; Döhner, K.; McGeehan, G.M.; et al. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science 2020, 367, 586–590. [Google Scholar] [CrossRef]
  52. Klossowski, S.; Miao, H.; Kempinska, K.; Wu, T.; Purohit, T.; Kim, E.; Linhares, B.M.; Chen, D.; Jih, G.; Perkey, E.; et al. Menin inhibitor MI-3454 induces remission in MLL1-rearranged and NPM1-mutated models of leukemia. J. Clin. Investig. 2020, 130, 981–997. [Google Scholar] [CrossRef] [Green Version]
  53. Shi, A.; Murai, M.J.; He, S.; Lund, G.; Hartley, T.; Purohit, T.; Reddy, G.; Chruszcz, M.; Grembecka, J.; Cierpicki, T. Structural insights into inhibition of the bivalent menin-MLL interaction by small molecules in leukemia. Blood 2012, 120, 4461–4469. [Google Scholar] [CrossRef] [Green Version]
  54. Grembecka, J.; He, S.; Shi, A.; Purohit, T.; Muntean, A.G.; Sorenson, R.J.; Showalter, H.D.; Murai, M.J.; Belcher, A.M.; Hartley, T.; et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol. 2012, 8, 277–284. [Google Scholar] [CrossRef] [Green Version]
  55. Grembecka, J.; Belcher, A.M.; Hartley, T.; Cierpicki, T. Molecular basis of the mixed lineage leukemia-menin interaction: Implications for targeting mixed lineage leukemias. J. Biol. Chem. 2010, 285, 40690–40698. [Google Scholar] [CrossRef] [Green Version]
  56. Wang, E.; Altman, J.K.; Issa, G.C.; Pettit, K.; DeBotten, S.; Walter, R.; Fenaux, P.; Ades, L.; Burrows, F.; Tomkinson, B.; et al. Phase 1/2 First in Human Study of the Menin-KMT2A (MLL) Inhibitor KO-539 in Patients with Relapsed or Refractory Acute Myeloid Leukemia; European Hematology Association: The Hague, The Netherlands, 2021; Abstract PB1408. [Google Scholar]
  57. Wang, E.; Altman, J.K.; Pettit, K.; DeBotten, S.; Walter, R.; Fenaux, P.; Burrows, F.; Tomkinson, B.; Martell, B.; Fathi, A. Preliminary data on a phase 1/2A first in human study of the menin-KMT2A (MLL) inhibitor KO-539 in patients with relapsed or refractory acute myeloid leukemia. In Proceedings of the 62nd Annual American Society of Hematology Annual Meeting and Exposition, Virtual Conference, 5–8 December 2020. [Google Scholar]
  58. Syndax Pharmaceuticals. Syndax Announces Positive Interim Data Demonstrating Robust Clinical Activity in Phase 1 Portion of the AUGMENT-101 Trial of SNDX-5613 in Patients with Genetically-Defined Acute Leukemias; Syndax Pharmaceuticals, Inc.: Waltham, MA, USA, 2021. [Google Scholar]
  59. Stein, E.M.; Aldoss, I.; DiPersio, J.F.; Stone, R.M.; Arellano, M.L.; Rosen, G.; Meyers, M.L.; Huang, Y.; Smith, S.; Bagley, R.G.; et al. Safety and efficacy of menin inhibition in patients (Pts) with MLL-rearranged and NPM1 mutant acute leukemia: A phase (Ph) 1, first-in-human study of SNDX-5613 (AUGMENT 101). Blood 2021, 138, 699. [Google Scholar] [CrossRef]
  60. Wang, G.G.; Pasillas, M.P.; Kamps, M.P. Meis1 programs transcription of FLT3 and cancer stem cell character, using a mechanism that requires interaction with Pbx and a novel function of the Meis1 C-terminus. Blood 2005, 106, 254–264. [Google Scholar] [CrossRef] [Green Version]
  61. Dzama, M.M.; Steiner, M.; Rausch, J.; Sasca, D.; Schönfeld, J.; Kunz, K.; Taubert, M.C.; McGeehan, G.M.; Chen, C.-W.; Mupo, A.; et al. Synergistic targeting of FLT3 mutations in AML via combined menin-MLL and FLT3 inhibition. Blood 2020, 136, 2442–2456. [Google Scholar] [CrossRef]
  62. Miao, H.; Kim, E.; Chen, D.; Purohit, T.; Kempinska, K.; Ropa, J.; Klossowski, S.; Trotman, W.E.; Danet-Desnoyers, G.-A.; Cierpicki, T.; et al. Combinatorial treatment with menin and FLT3 inhibitors induces complete remission in AML models with activating FLT3 mutations. Blood 2020, 136, 2958. [Google Scholar] [CrossRef]
  63. Fiskus, W.; Boettcher, S.; Daver, N.; Mill, C.P.; Sasaki, K.; Birdwell, C.E.; Davis, J.A.; Takahashi, K.; Kadia, T.M.; DiNardo, C.D.; et al. Effective Menin inhibitor-based combinations against AML with MLL rearrangement or NPM1 mutation (NPM1c). Blood Cancer J. 2022, 12, 5. [Google Scholar] [CrossRef]
  64. Brzezinka, K.; Nevedomskaya, E.; Lesche, R.; Steckel, M.; Eheim, A.L.; Haegebarth, A.; Stresemann, C. Functional diversity of inhibitors tackling the differentiation blockage of MLL-rearranged leukemia. J. Hematol. Oncol. 2019, 12, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Stein, E.M.; Garcia-Manero, G.; Rizzieri, D.A.; Tibes, R.; Berdeja, J.G.; Savona, M.R.; Jongen-Lavrenic, M.; Altman, J.K.; Thomson, B.; Blakemore, S.J.; et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 2018, 131, 2661–2669. [Google Scholar] [CrossRef] [PubMed]
  66. Mohr, S.; Doebele, C.; Comoglio, F.; Berg, T.; Beck, J.; Bohnenberger, H.; Alexe, G.; Corso, J.; Ströbel, P.; Wachter, A.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Walker, A.R.; Byrd, J.C.; Blachly, J.S.; Bhatnagar, B.; Mims, A.S.; Orwick, S.; Lin, T.L.; Crosswell, H.E.; Zhang, D.; Minden, M.D.; 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. [Google Scholar] [CrossRef]
  68. Li, X.; Song, Y. Structure, function and inhibition of critical protein-protein interactions involving mixed lineage leukemia 1 and its fusion oncoproteins. J. Hematol. Oncol. 2021, 14, 56. [Google Scholar] [CrossRef]
  69. Garcia-Manero, G.; Daver, N.G.; Montalban-Bravo, G.; Jabbour, E.J.; DiNardo, C.D.; Kornblau, S.M.; Bose, P.; Alvarado, Y.; Ohanian, M.; Borthakur, G.; et al. A phase II study evaluating the combination of nivolumab (Nivo) or ipilimumab (Ipi) with azacitidine in Pts with previously treated or untreated myelodysplastic syndromes (MDS). Blood 2016, 128, 344. [Google Scholar] [CrossRef]
  70. Bewersdorf, J.P.; Zeidan, A.M. Randomized trials with checkpoint inhibitors in acute myeloid leukaemia and myelodysplastic syndromes: What have we learned so far and where are we heading? Best Pract. Res. Clin. Haematol. 2020, 33, 101222. [Google Scholar] [CrossRef]
  71. Daver, N.; Garcia-Manero, G.; Basu, S.; Boddu, P.C.; Alfayez, M.; Cortes, J.E.; Konopleva, M.; Ravandi-Kashani, F.; Jabbour, E.; Kadia, T.; 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. [Google Scholar] [CrossRef] [Green Version]
  72. Isidori, A.; Cerchione, C.; Daver, N.; DiNardo, C.; Garcia-Manero, G.; Konopleva, M.; Jabbour, E.; Ravandi, F.; Kadia, T.; Burguera, A.D.L.F.; et al. Immunotherapy in acute myeloid leukemia: Where we stand. Front. Oncol. 2021, 11, 656218. [Google Scholar] [CrossRef]
  73. Veillette, A.; Chen, J. SIRPα-CD47 immune checkpoint blockade in anticancer therapy. Trends Immunol. 2018, 39, 173–184. [Google Scholar] [CrossRef]
  74. Brown, E.; Hooper, L.; Ho, T.; Gresham, H. Integrin-associated protein: A 50-kD plasma membrane antigen physically and functionally associated with integrins. J. Cell Biol. 1990, 111, 2785–2794. [Google Scholar] [CrossRef]
  75. Oldenborg, P.A.; Zheleznyak, A.; Fang, Y.F.; Lagenaur, C.F.; Gresham, H.D.; Lindberg, F.P. Role of CD47 as a marker of self on red blood cells. Science 2000, 288, 2051–2054. [Google Scholar] [CrossRef]
  76. Khandelwal, S.; Van Rooijen, N.; Saxena, R.K. Reduced expression of CD47 during murine red blood cell (RBC) senescence and its role in RBC clearance from the circulation. Transfusion 2007, 47, 1725–1732. [Google Scholar] [CrossRef]
  77. Weiskopf, K.; Jahchan, N.S.; Schnorr, P.J.; Cristea, S.; Ring, A.; Maute, R.L.; Volkmer, A.K.; Volkmer, J.-P.; Liu, J.; Lim, J.S.; et al. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer. J. Clin. Investig. 2016, 126, 2610–2620. [Google Scholar] [CrossRef]
  78. Willingham, S.B.; Volkmer, J.P.; Gentles, A.J.; Sahoo, D.; Dalerba, P.; Mitra, S.S.; Wang, J.; Contreras-Trujillo, H.; Martin, R.; Cohen, J.D.; et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 6662–6667. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, Y.; Xu, Z.; Guo, S.; Zhang, L.; Sharma, A.; Robertson, G.P.; Huang, L. Intravenous delivery of siRNA targeting CD47 effectively inhibits melanoma tumor growth and lung metastasis. Mol. Ther. 2013, 21, 1919–1929. [Google Scholar] [CrossRef] [Green Version]
  80. Xiao, Z.; Chung, H.; Banan, B.; Manning, P.T.; Ott, K.C.; Lin, S.; Capoccia, B.J.; Subramanian, V.; Hiebsch, R.R.; Upadhya, G.A.; et al. Antibody mediated therapy targeting CD47 inhibits tumor progression of hepatocellular carcinoma. Cancer Lett. 2015, 360, 302–309. [Google Scholar] [CrossRef] [Green Version]
  81. Chao, M.P.; Alizadeh, A.A.; Tang, C.; Myklebust, J.H.; Varghese, B.; Gill, S.; Jan, M.; Cha, A.C.; Chan, C.K.; Tan, B.T.; et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 2010, 142, 699–713. [Google Scholar] [CrossRef] [Green Version]
  82. Kim, D.; Wang, J.; Willingham, S.B.; Martin, R.; Wernig, G.; Weissman, I.L. Anti-CD47 antibodies promote phagocytosis and inhibit the growth of human myeloma cells. Leukemia 2012, 26, 2538–2545. [Google Scholar] [CrossRef] [Green Version]
  83. Chao, M.P.; Tang, C.; Pachynski, R.K.; Chin, R.; Majeti, R.; Weissman, I.L. Extranodal dissemination of non-Hodgkin lymphoma requires CD47 and is inhibited by anti-CD47 antibody therapy. Blood 2011, 118, 4890–4901. [Google Scholar] [CrossRef]
  84. Chao, M.P.; Alizadeh, A.A.; Tang, C.; Jan, M.; Weissman-Tsukamoto, R.; Zhao, F.; Park, C.Y.; Weissman, I.L.; Majeti, R. Therapeutic antibody targeting of CD47 eliminates human acute lymphoblastic leukemia. Cancer Res. 2011, 71, 1374–1384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Brown, E.J.; Frazier, W.A. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 2001, 11, 130–135. [Google Scholar] [CrossRef] [PubMed]
  86. Tseng, D.; Volkmer, J.-P.; Willingham, S.B.; Contreras-Trujillo, H.; Fathman, J.W.; Fernhoff, N.B.; Seita, J.; Inlay, M.A.; Weiskopf, K.; Miyanishi, M.; et al. Anti-CD47 antibody–mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response. Proc. Natl. Acad. Sci. USA 2013, 110, 11103–11108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPα signaling axis as an innate immune checkpoint in cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef] [PubMed]
  88. Liu, X.; Pu, Y.; Cron, K.; Deng, L.; Kline, J.; Frazier, W.A.; Xu, H.; Peng, H.; Fu, Y.-X.; Xu, M.M. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 2015, 21, 1209–1215. [Google Scholar] [CrossRef] [Green Version]
  89. Gardai, S.J.; McPhillips, K.A.; Frasch, S.C.; Janssen, W.J.; Starefeldt, A.; Murphy-Ullrich, J.E.; Bratton, D.L.; Oldenborg, P.-A.; Michalak, M.; Henson, P.M. Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte. Cell 2005, 123, 321–334. [Google Scholar] [CrossRef] [Green Version]
  90. Chao, M.P.; Jaiswal, S.; Weissman-Tsukamoto, R.; Alizadeh, A.A.; Gentles, A.J.; Volkmer, J.; Weiskopf, K.; Willingham, S.B.; Raveh, T.; Park, C.Y.; et al. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47. Sci. Transl. Med. 2010, 2, 63ra94. [Google Scholar] [CrossRef] [Green Version]
  91. Weiskopf, K.; Ring, A.M.; Ho, C.C.; Volkmer, J.-P.; Levin, A.M.; Volkmer, A.K.; Özkan, E.; Fernhoff, N.B.; van de Rijn, M.; Weissman, I.L.; et al. Engineered SIRPα variants as immunotherapeutic adjuvants to anticancer antibodies. Science 2013, 341, 88–91. [Google Scholar] [CrossRef] [Green Version]
  92. Chen, J.; Zhong, M.C.; Guo, H.; Davidson, D.; Mishel, S.; Lu, Y.; Rhee, I.; Pérez-Quintero, L.-A.; Zhang, S.; Cruz-Munoz, M.-E.; et al. SLAMF7 is critical for phagocytosis of haematopoietic tumour cells via Mac-1 integrin. Nature 2017, 544, 493–497. [Google Scholar] [CrossRef] [Green Version]
  93. Boasman, K.; Simmonds, M.; Rinaldi, C. CALR and CD47: An insight into their roles in the disease progression of MDS and MPN. J. Blood Disord. Transfus. 2018, 10, 1–5. [Google Scholar] [CrossRef]
  94. Uno, S.; Kinoshita, Y.; Azuma, Y.; Tsunenari, T.; Yoshimura, Y.; Iida, S.; Kikuchi, Y.; Yamada-Okabe, H.; Fukushima, N. Antitumor activity of a monoclonal antibody against CD47 in xenograft models of human leukemia. Oncol. Rep. 2007, 17, 1189–1194. [Google Scholar] [CrossRef]
  95. Jaiswal, S.; Jamieson, C.H.; Pang, W.W.; Park, C.Y.; Chao, M.P.; Majeti, R.; Traver, D.; van Rooijen, N.; Weissman, I.L. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 2009, 138, 271–285. [Google Scholar] [CrossRef] [Green Version]
  96. Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D.; Van Rooijen, N.; Weissman, I.L. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell 2009, 138, 286–299. [Google Scholar] [CrossRef] [Green Version]
  97. Sallman, D.A.; McLemore, A.F.; Aldrich, A.L.; Komrokji, R.S.; McGraw, K.L.; Dhawan, A.; Geyer, S.; Hou, H.-A.; Eksioglu, E.A.; Sullivan, A.; et al. TP53 mutations in myelodysplastic syndromes and secondary AML confer an immunosuppressive phenotype. Blood J. Am. Soc. Hematol. 2020, 136, 2812–2823. [Google Scholar] [CrossRef]
  98. Williams, P.; Basu, S.; Garcia-Manero, G.; Hourigan, C.S.; Oetjen, K.A.; Cortes, J.E.; Ravandi, F.; Jabbour, E.J.; Al-Hamal, Z.; Konopleva, M.; et al. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2019, 125, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
  99. Blagih, J.; Zani, F.; Chakravarty, P.; Hennequart, M.; Pilley, S.; Hobor, S.; Hock, A.K.; Walton, J.B.; Morton, J.P.; Gronroos, E.; et al. Cancer-specific loss of p53 leads to a modulation of myeloid and T cell responses. Cell Rep. 2020, 30, 481–496. [Google Scholar] [CrossRef]
  100. Wang, B.; Lim, C.B.; Yan, J.; Li, L.; Wang, J.; Little, J.B.; Yuan, Z.-M. MDMX phosphorylation-dependent p53 downregulation contributes to an immunosuppressive tumor microenvironment. J. Mol. Cell Biol. 2020, 12, 713–722. [Google Scholar] [CrossRef]
  101. Guo, G.; Marrero, L.; Rodriguez, P.; Del Valle, L.; Ochoa, A.; Cui, Y. Trp53 inactivation in the tumor microenvironment promotes tumor progression by expanding the immunosuppressive lymphoid-like stromal network. Cancer Res. 2013, 73, 1668–1675. [Google Scholar] [CrossRef] [Green Version]
  102. Liu, J.; Wang, L.; Zhao, F.; Tseng, S.; Narayanan, C.; Shura, L.; Willingham, S.; Howard, M.; Prohaska, S.; Volkmer, J.; et al. Pre-Clinical development of a humanized anti-CD47 antibody with anti-cancer therapeutic potential. PLoS ONE 2015, 10, e0137345. [Google Scholar] [CrossRef] [Green Version]
  103. Lin, G.H.Y.; Chai, V.; Lee, V.; Dodge, K.; Truong, T.; Wong, M.; Johnson, L.; Linderoth, E.; Pang, X.; Winston, J.; et al. TTI-621 (SIRPαFc), a CD47-blocking cancer immunotherapeutic, triggers phagocytosis of lymphoma cells by multiple polarized macrophage subsets. PLoS ONE 2017, 12, e0187262. [Google Scholar] [CrossRef] [Green Version]
  104. Johnson, Z.; Papaioannou, A.; Bernard, L.; Cosimo, E.; Daubeuf, B.; Richard, F.; Chauchet, X.; Moine, V.; Broyer, L.; Shang, L.; et al. Bispecific antibody targeting of CD47/CD19 to promote enhanced phagocytosis of patient B lymphoma cells. J. Clin. Oncol. 2015, 33 (Suppl. 15), e14016. [Google Scholar] [CrossRef]
  105. Sallman, D.A.; Donnellan, W.; Asch, A.; Lee, D.; Al Malki, M.; Pollyea, D.; Kambhampati, S.; Komrokji, R.S.; Van Elk, J.; Lin, M.; et al. The First-in-Class Anti-CD47 Antibody HU5F9-G4 is Active and Well Tolerated Alone or in Combination with Azacitidine in AML and MDS Patients; Initial Phase 1B Results; European Hematology Association: The Hague, The Netherlands, 2019; Volume Abstract: S878. [Google Scholar]
  106. Vyas, P.; Knapper, S.; Kelly, R.; Salim, R.; Lubowiecki, M.; Royston, D.; Johnson, H.; Roberts, C.; Chen, J.Y.; Agoram, B.; et al. Initial Phase 1 Results of the First-In-Class Anti-CD47 Antibody HU5F9-G4 in Relapsed/Refractory Acute Myeloid Leukemia Patients; European Hematology Association: The Hague, The Netherlands, 2018; Volume PF232. [Google Scholar]
  107. Zeidan, A.M.; DeAngelo, D.J.; Palmer, J.M.; Seet, C.S.; Tallman, M.S.; Wei, X.; Li, Y.F.; Hock, R.N.; Burgess, M.R.; Hege, K.; et al. A phase I study of CC-90002, a monoclonal antibody targeting CD47, in patients with relapsed and/or refractory (R/R) Acute Myeloid Leukemia (AML) and high-risk myelodysplastic syndromes (MDS): Final results. Blood 2019, 134 (Suppl. 1), 1320. [Google Scholar] [CrossRef]
  108. Daver, N.G.; Vyas, P.; Kambhampati, S.; Malki, M.M.A.; Larson, R.A.; Asch, A.S.; Mannis, G.N.; Chai-Ho, W.; Tanaka, T.N.; Bradley, T.J.; et al. Tolerability and efficacy of the first-in-class anti-CD47 antibody magrolimab combined with azacitidine in frontline TP53m AML patients: Phase 1b results. J. Clin. Oncol. 2022, 40 (Suppl. 16), 7020. [Google Scholar] [CrossRef]
  109. Brierley, C.K.; Staves, J.; Roberts, C.; Johnson, H.; Vyas, P.; Goodnough, L.; Murphy, M. The effects of monoclonal anti-CD47 on RBCs, compatibility testing, and transfusion requirements in refractory acute myeloid leukemia. Transfusion 2019, 59, 2248–2254. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, J.Y.; McKenna, K.M.; Choi, T.S.; Duan, M.J.; Brown, M.L.; Stewart, J.J.; Sompalli, M.K.; Vyas, P.; Schrier, S.; Majeti, R.; et al. RBC-specific CD47 pruning confers protection and underlies the transient anemia in patients treated with anti-CD47 antibody 5F9. Blood 2018, 132, 2327. [Google Scholar] [CrossRef]
  111. Boasman, K.; Bridle, C.; Simmonds, M.; Rinaldi, C. Role of pro-phagocytic calreticulin and anti-phagocytic CD47 in MDS and MPN models treated with azacytidine or ruxolitinib. In Haematolgica; Ferrata Storti Foundation: Pavia, Italy, 2017; p. 763. [Google Scholar]
  112. DiNardo, C.D.; Jonas, B.A.; Pullarkat, V.; Thirman, M.J.; Garcia, J.S.; Wei, A.H.; Konopleva, M.; Döhner, H.; Letai, A.; Fenaux, P.; et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N. Engl. J. Med. 2020, 383, 617–629. [Google Scholar] [CrossRef] [PubMed]
  113. Valentin, R.; Peluso, M.O.; Lehmberg, T.Z.; Adam, A.; Zhang, L.; Armet, C.M.; Guerriero, J.L.; Lee, B.H.; Palombella, V.J.; Holland, P.M.; et al. The fully human anti-CD47 antibody SRF231 has dual-mechanism antitumor activity against chronic lymphocytic leukemia (CLL) cells and increases the activity of both rituximab and venetoclax. Blood 2018, 132, 4393. [Google Scholar] [CrossRef]
Figure 1. Proposed Mechanisms of Inhibiting Menin. KMT2Ar fusion proteins bind menin and lead to a stereotyped gene profile expression which includes HOX family genes and MEIS1 leading to a leukemia stem cell like state. The menin inhibitors disrupt binding to menin reducing this gene expression and leading to cell differentiation. FLT3 expression is regulated by upstream factors including MEIS1. Mutated NPM1 also up-regulates the expression of HOXA9 and MEIS1 possibly by cytoplasmic sequestration of transcriptional regulators (such as HEXIM1) which normally inhibit components of KMT2Ar fusion protein complexes.
Figure 1. Proposed Mechanisms of Inhibiting Menin. KMT2Ar fusion proteins bind menin and lead to a stereotyped gene profile expression which includes HOX family genes and MEIS1 leading to a leukemia stem cell like state. The menin inhibitors disrupt binding to menin reducing this gene expression and leading to cell differentiation. FLT3 expression is regulated by upstream factors including MEIS1. Mutated NPM1 also up-regulates the expression of HOXA9 and MEIS1 possibly by cytoplasmic sequestration of transcriptional regulators (such as HEXIM1) which normally inhibit components of KMT2Ar fusion protein complexes.
Cancers 14 05906 g001
Figure 2. Proposed Mechanisms of Targeting CD47. CD47 blockade decreases the anti-phagocytic “don’t-eat-me” signal on leukemia cells. Azacitidine may upregulate pro-phagocytic signals, such as calreticulin, increasing the likelihood of phagocytosis of leukemia cells. This may be further increased if CD47 binding to SIRPα is blocked or an antibody targeting a leukemic antigen (e.g., CD33) is present to bind a macrophage Fcγ receptor. Phagocytosis may lead to tumor peptide presentation to the adoptive immune system, multiple targets exist to increase T-cell activation such as antibodies to PD-L1 or to PD-1.
Figure 2. Proposed Mechanisms of Targeting CD47. CD47 blockade decreases the anti-phagocytic “don’t-eat-me” signal on leukemia cells. Azacitidine may upregulate pro-phagocytic signals, such as calreticulin, increasing the likelihood of phagocytosis of leukemia cells. This may be further increased if CD47 binding to SIRPα is blocked or an antibody targeting a leukemic antigen (e.g., CD33) is present to bind a macrophage Fcγ receptor. Phagocytosis may lead to tumor peptide presentation to the adoptive immune system, multiple targets exist to increase T-cell activation such as antibodies to PD-L1 or to PD-1.
Cancers 14 05906 g002
Table 1. Menin Inhibitors in Clinical Trials. Note: Aza, azacitidine; KMT2Ar histone-lysine-N-methyltransferase 2 rearranged; mini-HCV lower intensity cyclophosphamide, vincristine, doxorubicin and dexamethasone; R/R, relapsed or refractory; ven, venetoclax.
Table 1. Menin Inhibitors in Clinical Trials. Note: Aza, azacitidine; KMT2Ar histone-lysine-N-methyltransferase 2 rearranged; mini-HCV lower intensity cyclophosphamide, vincristine, doxorubicin and dexamethasone; R/R, relapsed or refractory; ven, venetoclax.
MoleculePhaseEnrollmentStatusRegimenNCTPopulation
SNDX-5613I/II186RecruitingMonotherapyNCT04065399Relapsed/refractory (R/R) MPAL, KMT2Ar AML, NPM1m AML
KO-539I/II100RecruitingMonotherapyNCT04067336Relapsed/refractory all patients
expansion: KMT2Ar and NPM1m R/R AML [57]
JNJ-75276617I110RecruitingMonotherapyNCT04811560Relapsed/refractory: KMT2Ar, NPM1m AML
BMF-219I/II100RecruitingMonotherapyNCT05153330Relapsed/refractory all patients
(with and without strong CYP3A4 inhibitors)
DS-1594bI/II122RecruitingCombination
(ven/aza or Mini-HCV)
NCT04752163Monotherapy in relapsed/Refractory KMT2Ar AML
Monotherapy in relapsed/Refractory NPM1m AML
Aza/Ven Combo in R/R NPM1 or KMT2Ar AML
Mini-HCVD in R/R ALL with KMT2Ar
Table 2. Anti-CD47 Agents in Clinical Development. Abbreviations: AML, acute myeloid leukemia; aza, azacitidine; CMML, chronic myelomonocytic leukemia; F, fit for intensive chemotherapy; MDS, myelodysplastic syndrome; R/R relapsed or refractory; TN, treatment naïve; U, unfit for intensive chemotherapy; Ven, venetoclax. Note: * SIRPa targeting agent, all others target CD47. The following agents are in clinical development but AML patients not included in study: TG-1801, A0-176, TG1801 (anti-CD19 CD47 bispecific), IMC-002, STI-6643, SL-172154, ZL-1201, IMM01, & SRF231 (no longer in active development); PF-07257876 and IBI322 (anti-CD47 and PD-L1); IMM-0306 CD47xCD20 bispecific. SRF231 showed 90% receptor occupancy no complete or partial responses in solid tumor and lymphoma cohorts, development has been suspended. The following agents target SIRPa but AML patients are not included in study: GS-0189, CC-95251, BI765063/OSE-172.
Table 2. Anti-CD47 Agents in Clinical Development. Abbreviations: AML, acute myeloid leukemia; aza, azacitidine; CMML, chronic myelomonocytic leukemia; F, fit for intensive chemotherapy; MDS, myelodysplastic syndrome; R/R relapsed or refractory; TN, treatment naïve; U, unfit for intensive chemotherapy; Ven, venetoclax. Note: * SIRPa targeting agent, all others target CD47. The following agents are in clinical development but AML patients not included in study: TG-1801, A0-176, TG1801 (anti-CD19 CD47 bispecific), IMC-002, STI-6643, SL-172154, ZL-1201, IMM01, & SRF231 (no longer in active development); PF-07257876 and IBI322 (anti-CD47 and PD-L1); IMM-0306 CD47xCD20 bispecific. SRF231 showed 90% receptor occupancy no complete or partial responses in solid tumor and lymphoma cohorts, development has been suspended. The following agents target SIRPa but AML patients are not included in study: GS-0189, CC-95251, BI765063/OSE-172.
AgentPhaseEnrollmentStatusRegimenNCTPopulation
Magrolimab (Hu5F9-G4)III520RecruitingCombination with azaNCT04313881Treatment naïve (TN) higher risk MDS
Magrolimab (Hu5F9-G4)III346RecruitingCombination with azaNCT04778397TP53 mutated TN unfit (TNU) AML;
TP53 mutated TN fit AML
Magrolimab (Hu5F9-G4)Ib/II98RecruitingCombination with ven/azaNCT04435691Ib: Relapsed/Refractory (R/R) AML;
II: TNU AML or TN < 75 years old
with high risk cytogenetics +/− TP53
Magrolimab (Hu5F9-G4)Ib287Recruiting [105]Combination with azaNCT03248479R/R AML, TNU AML, RBC
transfusion dependent low risk MDS
Magrolimab (Hu5F9-G4)Ib13CompletedCombination with AtezolizumabNCT03922477R/R AML
Magrolimab (Hu5F9-G4)Ia20Completed [106]MonotherapyNCT02678338R/R AML; R/R higher risk MDS
TJ011133 (TJC4, Lemzoparlimab)IIA80RecruitingCombination with azaNCT04202003TNU AML or TN higher risk MDS
TJ011133 (TJC4, Lemzoparlimab)Ib120RecruitingCombination with aza or ven/azaNCT04912063TNU AML; high risk MDS
ALX148 (Evorpacept *)I/II97RecruitingCombination with ven/azaNCT04755244R/R AML or new AML in patients
ineligible for standard induction
AK117Ib/II160RecruitingCombination with azaNCT04980885AML, newly diagnosed or R/R
DSP107 *Ib/II36RecruitingCombination with azaNCT04937166TNU AML, TN MDS, R/R MDS/CMML
TTI-621Ia/Ib260RecruitingMonotherapy NCT02663518R/R AML
TTI-662Ia/Ib150RecruitingIa: single agent
1b: combination with ven/aza
NCT03530683TN TP53-mutated AML aza+TTI-662;
TNU AML TTI-662 + aza/ven
IBI188Ia12RecruitingCombination with azaNCT04485065TN higher risk MDS
IBI188Ib126RecruitingCombination with azaNCT04485052R/R AML, TNU AML
CC-90002I28Terminated [107]MonotherapyNCT02641002R/R AML or high risk MDS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Matthews, A.H.; Pratz, K.W.; Carroll, M.P. Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia. Cancers 2022, 14, 5906. https://doi.org/10.3390/cancers14235906

AMA Style

Matthews AH, Pratz KW, Carroll MP. Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia. Cancers. 2022; 14(23):5906. https://doi.org/10.3390/cancers14235906

Chicago/Turabian Style

Matthews, Andrew H., Keith W. Pratz, and Martin P. Carroll. 2022. "Targeting Menin and CD47 to Address Unmet Needs in Acute Myeloid Leukemia" Cancers 14, no. 23: 5906. https://doi.org/10.3390/cancers14235906

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop