Considerable progress has also been made regarding the development of novel agents, including mutant p53-targeted approaches and immunotherapy.
A. Targeted treatments
Novel targeted therapies incorporated into combination regimens have also been explored in the
TP53mut AML setting. Pevonedistat (PEVO), an inhibitor of the NEDD8-activating enzyme (NAE) seems to exert antiproliferative effects on LCs and preclinical data supports synergistic effects with AZA and VEN [
47,
48,
49]. A phase 1b study of unfit, treatment-naïve AML patients treated with PEVO and AZA showed improved responses with an ORR of 50%, with
TP53mut patients achieving a CR and partial response (PR) rate of 80% [
47]. Based on these results, a phase 2 study consisting of
TP53mut AML patients was conducted, but failed to show enhanced CRR rates and was prematurely terminated [
48]. Intriguingly, a phase 1/2 study evaluating the efficacy of combined PEVO, AZA, and VEN in ND secondary AML reported a CR/CRi rate of 64%, but a dreadful 1-year OS of 0% in
TP53mut patients, contrary to a median OS of 18 months in
TP53wt patients [
49]. Moreover, the DOR differed significantly among these patients [
49]. These conflicting results may be attributable to the different VAF of patients since the second study included only
TP53mut patients with a VAF of >30%. Nevertheless, although these results seem discouraging, data are scarce and derive from small studies, thus PEVO may still have a role to play in this setting.
Ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor has been previously shown to impede the proliferation of human AML blasts in vitro, either alone or combined with cytarabine or daunorubicin [
50]. A randomized phase 2 study evaluated the outcomes of adding ibrutinib to DEC10 versus DEC10 monotherapy in elderly, previously untreated AML patients [
51]. Surprisingly, although the addition of ibrutinib did not yield favorable outcomes,
TP53mut was correlated with higher responses and CR/CRi rates of 56% [
51]. However, these responses were not translated into a superior OS [
51]. Nevertheless, although ibrutinib’s efficacy in
TP53mut AML needs to be further validated, it remains a highly appealing approach.
Finally, bortezomib, a proteasome inhibitor, is widely investigated in the management of AML patients, since it has been associated with potent antiproliferative properties [
52]. A randomized phase 2 trial of AML patients treated with either combined bortezomib and DEC10 or DEC10 alone failed to show a potential advantage of the combination in those with
TP53 mutations [
52]. Moreover, the addition of bortezomib conferred no benefit to the study patients overall [
52]. Conclusively, targeted therapies’ efficacy remains ambiguous and warrants further exploration of these agents in
TP53mut AML through large clinical trials.
B. TP53 targeting agents
Although p53 has traditionally been considered undruggable, efforts have been made to overcome this hurdle and have led to the development of a new, small molecule, called “p53 reactivation and induction of massive apoptosis” (PRIMA-1), that can reverse the mutant conformation of p53, which induces protein unfolding and restores wild-type functions to mutant p53, such as induction of apoptosis and promotion of cell cycle arrest [
53]. Eprenetapopt (EP) or APR-246, a methylated derivative of PRIMA-1 (PRIMA-1
MET), is a first-in-class agent that binds covalently to cysteine residues in mutant p53 protein [
53]. Preclinical studies have demonstrated that EP exerts apoptotic effects on AML cell lines and primary LCs from AML patients in a dose-dependent manner [
54]. Noteworthily, the presence of
TP53 mutations did not significantly affect sensitivity to this agent [
54]. Subsequent studies have shown significant synergistic cytotoxicity of EP and AZA in
TP53 mut primary cells from MDS/AML patients [
55]. Apart from the reported mutant p53 reactivation, preclinical data have also demonstrated that EP results in glutathione depletion and induction of ferroptosis, irrespective of the
TP53 status, thus indicating a different mechanism of action that leads to p53-independent cell death [
56,
57].
Recently, EP’s efficacy in combination with AZA has been evaluated in patients with
TP53 mut MDS and AML in two phase 2 studies, one in the USA and another one in Europe [
58,
59]. EP was administered by an intravenous infusion, at a fixed dose, on days 1-4 of each 28-day cycle and AZA was administered subcutaneously, at the standard dose, for seven days of each 28-day cycle [
58,
59].
TP53 mut AML patients in the US trial have achieved ORR and CR rates of 64% and 36%, respectively, and a median OS of 10.8 months [
58]. However, the sample size was significantly small and only patients with oligoblastic AML (20-30% marrow blasts) were included [
58]. The European trial, additionally including
TP53mut AML patients with more than 30% marrow blasts, has demonstrated an ORR of 33% and a CR rate of 17% [
59]. However, none of the patients with a high blast count achieved a CR [
59]. Median OS in patients with less and more than 30% marrow blasts was 13.9 months and 3.0 months respectively [
59]. Both studies have reported a significant reduction in the
TP53 VAF and p53 expression by immunochemistry in responding patients, with some patients achieving
TP53 negativity (VAF <5%) [
59]. These findings indicate a promising efficacy, since ORR, CR, and OS rates are generally higher than those reported with AZA monotherapy, particularly for patients with oligoblastic AML [
59]. Of note, patients with
TP53mut MDS have also yielded high response rates in both studies, with a CR rate of around 50% [
58,
59]. The doublet of EP and AZA has also been evaluated in a phase 2 trial of
TP53mut AML patients, as post-aSCT maintenance therapy administered for up to 12 cycles, with reported relapse-free survival and median OS being 12.5 and 20.6 months, respectively, which is quite encouraging for this high-risk population [
60]. The triplet combination of EP, AZA, and VEN has also been studied recently in the
TP53mut AML setting. In a phase 1, dose-finding and expansion study, patients with ND
TP53mut AML achieved an ORR, CR, and CR/CRi rate of 64%, 38%, and 56%, respectively, whereas DOR and median OS were 4.2 and 7.3 months, respectively [
61]. Importantly, the blast count did not have an impact on patients’ responses [
61]. Moreover,
TP53 negativity (VAF <5%) by NGS was achieved in 27% [
61]. These results are highly promising, since CR rates are higher than the CR rates of 22% that have been reported in patients with previously untreated
TP53mut AML receiving AZA in combination with VEN [
61]. Collectively, EP has demonstrated promising efficacy in
TP53 mut AML patients and provides the basis for further investigation in randomized clinical trials in the near future.
C. Immunotherapeutic approaches
Increasing interest has also grown regarding the use of immunotherapeutic agents in
TP53mut AML. CD47 or the “don’t eat me signal” is a transmembrane protein that interacts with signal-regulatory protein alpha (SIRPa), which is expressed in macrophages, and impedes macrophage-mediated phagocytosis [
62]. LCs have high levels of CD47, thus escaping immune surveillance [
62]. Increased CD47 expression in AML hematopoietic stem cells (HSCs) has been independently correlated with inferior outcomes, thus making the CD47/SIRPa axis an appealing therapeutic target [
63]. Blockade of CD47 in AML models has resulted in the induction of phagocytosis and elimination of LCs [
63,
64]. Magrolimab (MAG) is a novel, first-in-class, IgG4 monoclonal antibody against CD47 that acts as a macrophage checkpoint inhibitor and has exerted synergistic effects with AZA and VEN in preclinical in vitro and in vivo studies, with the latter agents eliciting “eat me” signals by upregulating calreticulin [
64,
65]. A phase 1b study has evaluated the combination of MAG and AZA in patients with previously untreated AML, ineligible for IC, with the majority of patients (82.8%) having
TP53 mutations [
65]. The CR rate was similar among
TP53mut and
TP53wt patients (31.9% and 32.2%, respectively), whereas OS was 9.8 months and 18.9 months, respectively [
65]. A phase 1/2 study of the triplet AZA, VEN, and MAG in ND elderly AML, high-risk (HR)-AML, and relapsed/refractory (R/R) AML patients has demonstrated an ORR and a CR rate of 74% and 41%, respectively, in ND
TP53mut patients [
66]. Although preliminary results were encouraging, a subsequent phase 3 trial (ENHANCE-2), evaluating MAG and AZA versus physician’s choice of VEN and AZA or IC in
TP53mut AML, was prematurely terminated, since MAG failed to demonstrate a survival benefit compared to standard of care [
67].
Several other agents targeting the disrupted CD47-SIRPa axis are also being explored in MDS/AML. Maplirpacept (MAP) or TTI-622 is a soluble fusion protein with anti-CD47 properties that, unlike other anti-CD47 agents, binds minimally to normal erythrocytes [
68]. In vivo studies of AML xenografts have demonstrated the efficacy of TTI-622 in enhancing macrophage-mediated phagocytosis [
68]. A phase 1a/1b dose-escalation and expansion trial of MAP alone or in combination with other agents in patients with advanced hematologic malignancies, including a cohort of ND
TP53mut AML patients treated with MAP and AZA is currently active (NCT03530683). Lemzoparlimab is another anti-CD47 agent that is currently being investigated in patients with HR-MDS and AML, in combination with AZA and/or VEN (NCT04202003, NCT0491206). A recent phase 1b study has evaluated the efficacy of AK117, an anti-CD47 agent, in combination with AZA as frontline treatment in AML patients and has demonstrated a CR and CR/CRi rate of 45% and 55%, respectively [
69]. Evorpacept (EVO) or ALX148 has been associated with increased LC phagocytosis in
TP53mut AML lines and mouse xenograft models, and its combination with HMA and/or VEN confers better survival [
70]. Hence, EVO entered a phase 1/2 trial, which studied its combination with VEN and AZA in patients with AML (ASPEN-05 trial, NCT04755244). However, ASPEN-05 was terminated, based on data from the ASPEN-02 trial, which was also terminated, reporting failure to achieve superior outcomes in MDS patients treated with EVO and AZA [
71]. Other anti-CD47 agents that are currently being studied in AML, combined with AZA and VEN include DSP107 (NCT04937166) and SL-172154 (NCT05275439), whereas a phase 1b study (NCT04485052) of IB188 (letaplimab) plus AZA in AML was suspended.
T-cell immunoglobulin mucin-3 (TIM-3) is a cell-surface glycoprotein that is constitutively expressed on the surface of certain immune cells, such as the T-cells and acts as a co-inhibitory receptor [
72,
73]. When interacting with one of its ligands, such as galectin-9, TIM-3 prompts the inhibition of T-cell responses [
72,
73]. It has also been demonstrated that TIM-3 is overexpressed in LCs and that TIM-3
+ AML leukemic stem cells (LSCs) secrete galectin-9 in an autocrine loop, that regulates self-renewal of these cells, via enhanced NF-κB and β-catenin signaling [
72,
73]. Hence, antibodies targeting TIM-3 provide a highly appealing therapeutic opportunity. Sabatolimab (SAB) or MBG453, is a humanized, high-affinity IgG4 antibody that targets TIM-3 [
74]. A phase 1b study that has evaluated SAB in combination with HMAs in patients with HR-MDS and ND AML displayed promising preliminary results, with ND AML patients exhibiting ORR and CR rates of 40% and 25%, respectively and a median duration of response of 12.6 months [
74]. Importantly, durable responses have been observed in patients with adverse-risk mutations, including
TP53, indicating that this combination may be effective in the
TP53mut setting [
74]. The addition of VEN is also explored in an ongoing phase 1b trial (NCT03940352), which investigates the combination of SAB and VEN in AML and HR-MDS patients. Furthermore, a phase 2 trial (STIMULUS-AML1, NCT04150029) is currently underway, investigating the combination of SAB, AZA, and VEN in patients with ND AML.
CD123 also serves as an appealing candidate for targeting. CD123 is a component of the interleukin-3 receptor (IL-3R) that plays a multifaceted role in hematopoiesis and immune responses; it stimulates HSC proliferation through activation of the PI3K/MAPK pathway and upregulation of antiapoptotic proteins, while it also participates in the modulation of T-cell responses [
75]. CD123 is widely expressed in blasts of AML patients, and its overexpression has been correlated with poor prognosis [
76]. In vitro and in vivo studies have demonstrated that a novel CD123 x CD3 dual-affinity retargeting (DART) molecule mediates T-cell activation and proliferation, leading to dose-dependent elimination of AML cell lines and primary AML blasts [
75]. Flotetuzumab (FLOT), is a CD123 x CD3 DART antibody that has been evaluated in a phase 1/2 study in R/R AML after primary induction failure or in early relapse, with the reported ORR being 30% [
77]. Remarkably,
TP53mut patients yielded encouraging responses with a CR rate of 47% and a median OS of 10.3 months in responding patients [
78]. Currently, early-phase trials are also exploring FLOT in post-transplant relapsed AML (NCT04582864, NCT05506956). Pivekimab sunirine (PVEK) or IMGN632, is a first-in-class antibody-drug conjugate (ADC) with a high affinity for CD123, which has displayed synergy with AZA and/or VEN in preclinical models [
79]. An ongoing multicenter, phase 1/2 study investigates PVEK as a triplet with AZA and VEN or in combination with MAG, in patients with R/R AML or ND CD123
+ AML [
80]. Preliminary data have shown that treatment with the triplet in R/R AML patients has led to an ORR and a composite CR rate (coCR) rate of 51% and 31%, respectively [
79]. However, VEN-naïve patients yielded significantly higher responses, than those with prior exposure to VEN [
79]. Recent data regarding patients in the ND AML cohort receiving frontline triplet treatment have reported robust responses with a CR and a coCR rate of 52% and 66%, respectively, whereas CR and coCR rates for
TP53mut patients were 13% and 47%, respectively [
81]. Rapid minimal residual disease (MRD) negativity was achieved in 73% of patients achieving coCR [
81]. Exceptionally, high coCR
MRD rates have been demonstrated among adverse risk patients,
TP53mut included [
81]. Triple combination therapy has been also associated with a manageable safety profile [
79,
81]. A phase 1 clinical trial of PVEK in combination with fludarabine, high-dose cytarabine (HiDAC), G-CSF, and idarubicin (FLAG-Ida) for frontline treatment of ND adverse-risk AML is ongoing (NCT06034470).
Tagraxofusp (TAG) is a CD123-targeted immunotoxin and has been evaluated as monotherapy in a phase 1 trial of AML and MDS patients, with reported responses being modest [
82]. However, recent data have supported that AZA, when combined with TAG, overcomes TAG resistance and restores TAG sensitivity, thus providing a rationale for the combination of these two agents [
83]. A phase 1b trial of TAG with AZA and/or VEN in AML and MDS patients is ongoing and preliminary results indicate promising efficacy [
84]. Remarkably,
TP53mut patients have achieved a CR/CRi/morphologic leukemia-free state (MLFS) rate of 54%, with a CR rate of 31% [
84]. Early-phase studies include the use of TAG as maintenance therapy for post-transplant AML patients (NCT05233618), for ND secondary AML after previous exposure to HMA (NCT05442216), and in combination with gemtuzumab ozogamicin for R/R AML (NCT05716009). Vibecotamab or XmAb14045, a CD3-CD123 bispecific T-cell engaging (BiTE) antibody is currently being investigated in the treatment of R/R AML, with preliminary data reporting modest ORR rates of 14% [
85]. Vibecotamab has also been associated with cytokine release syndrome (CRS), which is manageable with premedication [
85]. Other CD123-targeting agents that are in early clinical development include APV0436, MGD024, and CD123 chimeric antigen receptor T-cell (CAR-T) therapy [
86]. In summary, these results suggest that these agents may have a role to play in AML patients,
TP53mut included, and research in this field continues to uncover new insights into potential applications of CD123.
Immune-checkpoint inhibitor-based approaches have also been studied in AML. Ipilimumab, an antibody targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), has yielded a CR rate of 42% in patients with post-aSCT relapsed AML [
87]. Nivolumab (NIVO), a programmed cell death protein 1 (PD-1) inhibitor, has been evaluated as first-line AML therapy, in combination with idarubicin and AraC and has yielded encouraging responses in
TP53mut patients [
88]. NIVO has also been studied in R/R AML patients, in combination with AZA, with a modest ORR of 33% and an ORR of 13% in
TP53mut patients [
89]. A recent phase 2 trial of R/R AML patients receiving pembrolizumab, a PD-1 inhibitor, with HiDAC has demonstrated promising clinical activity in
TP53mut patients, reporting a CR rate of 40% [
90]. A randomized phase 2 trial of AZA with or without durvalumab (DURV), a PD-L1 inhibitor, as first-line treatment for elderly AML patients, has failed to show a potential benefit, since the addition of DURV did not enhance clinical outcomes and recorded ORR and OS were similar among both treatment arms [
91]. Interestingly, responses were similar between
TP53mut and
TP53wt patients (ORR 35% and 34%, respectively) [
91]. Nonetheless, the use of CTLA-4, PD1, and PD-L1 inhibitors in AML necessitates further research for strong conclusions to be drawn.
Leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) is an immune inhibitory receptor which is present on most immune cell subsets and is implicated in immunosuppressive responses [
92]. It has been demonstrated that LAIR-1 is highly expressed in AML blasts and LSCs and is responsible for the inhibition of intracellular downstream survival signals and blast proliferation, while its expression is relatively lower in normal HSCs, thus rendering LAIR-1 an ideal anti-leukemic target [
92]. NC525 is a humanized monoclonal antibody that binds specifically to AML blasts and LSCs, while sparing normal hematopoiesis, and induces apoptosis through a unique signaling pathway, without evidence of immunomodulatory effects on other immune subsets [
92]. Furthermore, it has been shown that NC525 displays synergistic activity when combined with AZA and VEN and results in leukemic cell destruction in patients who are refractory to VEN-AZA [
92]. A phase 1 trial investigating the safety and tolerability of NC525 in patients with advanced HMs, including R/R AML is underway (NCT05787496).
D. Other agents
Murine double minute protein 2 (MDM2) is an E3 ubiquitin ligase that negatively regulates the activity of p53 [
93]. MDM2 interacts with p53 and promotes its degradation via ubiquitination [
93]. Inhibition of MDM2 mediates antileukemic effects in
TP53wt AML through an increase in p53 levels [
93]. A phase 1/1b study has evaluated the use of idasanutlin (IDASA), an oral MDM2 inhibitor (MDM2i), either alone or in combination with AraC, in unfit for IC patients with R/R or ND AML and has demonstrated a coCR rate of 18.9% and 35.6% in patients receiving monotherapy or combination treatment, respectively [
94]. A subsequent randomized, double-blind, phase 3 trial (MIRROS trial), evaluating IDASA combined with AraC or placebo in R/R AML patients, has failed to show an improvement in OS, although the overall remission rate was enhanced by the addition of IDAS [
95]. Although MDM2i require wt-p53 to be effective, hence being unable to act directly in
TP53mut AML, they indirectly induce degradation of MCL-1, which is associated with VEN resistance, thus providing a rationale for the combined use of MDM2i and VEN, even in
TP53mut patients, in order to overcome VEN resistance [
96]. Milademetan, an MDM2i, in combination with LDAC, with or without VEN, has been recently explored in AML with discouraging responses and significant gastrointestinal toxicity [
97]. A phase 1b trial of IDASA and VEN in R/R AML patients has shown modest responses, with
TP53 mutations having been associated with unfavorable outcomes [
98]. A concern regarding the use of MDM2i is whether they select for the outgrowth of
TP53mut clones since studies have reported emergent
TP53 mutations in some patients [
97,
98]. Nevertheless, further studies are needed in order to assess the safety and efficacy of these agents in this setting.
Various agents for
TP53mut treatment are currently in early clinical development. Arsenic trioxide (ATO) has been shown to inactivate
TP53, by inducing proteasomal degradation of mutant p53 and upregulating
TP53wt functions [
99]. Therefore, it can lead to inactivation of proliferation of LCs and apoptosis promotion. Atorvastatin, is a potent destabilizing agent of mutant p53, since it has been shown that it effectively induces degradation for conformational or misfolded p53 mutants, via inhibition of the mevalonate pathway, with minimal effects on wt-p53 and DNA contact mutants [
100]. Collectively, these findings provide insight into exploring arsenic compound-based and statin-based therapies for AML harboring
TP53 mutations. A trial of combined ATO and DEC to treat
TP53mut AML/MDS (PANDA-T0 trial, NCT03855371) and a pilot trial of atorvastatin in
TP53mut and
TP53wt malignancies (NCT03560882) are currently enrolling.