Next Article in Journal
Underneath Images and Robots, Looking Deeper into the Pneumoperitoneum: A Narrative Review
Next Article in Special Issue
FLAG/FLAG-Ida Regimen in Secondary and Relapsed/Refractory Acute Myeloid Leukemia—Even in the Era of New Treatment Modalities Still a Significant Player
Previous Article in Journal
Do Postural and Walking Stabilities Change over a Decade by Aging? A Longitudinal Study
Previous Article in Special Issue
Approach to Acute Myeloid Leukemia with Increased Eosinophils and Basophils
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia

by
Christos Stafylidis
,
Dimitra Vlachopoulou
,
Christina-Nefeli Kontandreopoulou
and
Panagiotis Τ. Diamantopoulos
*
Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(4), 1082; https://doi.org/10.3390/jcm13041082
Submission received: 27 December 2023 / Revised: 7 February 2024 / Accepted: 9 February 2024 / Published: 14 February 2024

Abstract

:
Acute myeloid leukemia (AML) remains a challenging hematologic malignancy. The presence of TP53 mutations in AML poses a therapeutic challenge, considering that standard treatments face significant setbacks in achieving meaningful responses. There is a pressing need for the development of innovative treatment modalities to overcome resistance to conventional treatments attributable to the unique biology of TP53-mutated (TP53mut) AML. This review underscores the role of TP53 mutations in AML, examines the current landscape of treatment options, and highlights novel therapeutic approaches, including targeted therapies, combination regimens, and emerging immunotherapies, as well as agents being explored in preclinical studies according to their potential to address the unique hurdles posed by TP53mut AML.

1. Introduction

P53 is a tumor suppression protein encoded by the TP53 gene and a vital regulator of genomic stability preservation in response to DNA damage. This regulatory activity occurs through the activation of DNA repair pathways, the triggering of cell-cycle arrest, and the induction of apoptosis [1].
Acute myeloid leukemia (AML) harboring TP53 mutations, which is now classified as a distinct AML subtype according to the International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias [2], presents a redoubtable clinical challenge as it is associated with an adverse prognosis [3,4,5]. These mutations are observed mostly in treatment-related, relapsed, and elderly AML patients, often characterized by remarkable genomic instability [3,4,5]. While the rate of TP53 mutations in de novo AML is 5–10%, it is significantly increased (up to 25%) in older patients with de novo AML, with a median age of 60–67 years [6]. Higher frequency rates, up to 35%, are reported in treatment-related AML (t-AML) [6], whereas the highest rates of up to 70% are observed in patients with a complex karyotype and those with loss of chromosome 17/17p, 5/5q, or 7/7q [4,5,7].
Mutated TP53 induces genomic instability, contributing to leukemogenesis. It also confers unique characteristics to AML and results in the evasion of apoptosis by tumor cells, inherent resistance to conventional chemotherapy, and poor clinical outcomes [1,3,4,5]. Several studies report lower complete response (CR) rates, inferior complete remission duration, and dismal overall survival (OS) among TP53-mutated (TP53mut) AML patients [3,4,5]. Importantly, TP53 mutations have been found to be predictors of adverse outcomes irrespective of age, chemotherapy regimen, or complex karyotype [3,4]. Moreover, AML patients with TP53 mutations are at a higher risk of relapse and death after allogeneic stem cell transplantation (aSCT) [8]. Diagnostic approaches such as fluorescence in situ hybridization (FISH), next-generation sequencing (NGS), and in silico approaches may promptly identify these patients and may hold significant predictive value, thus facilitating decision-making relating to treatment strategies [9,10]. Importantly, loss of TP53, detected by FISH at diagnosis, has been correlated with poor response to chemotherapy [10].
The challenging management of TP53-mutated AML highlights the crucial need for the development of novel therapeutic approaches. In recent years, targeted agents, immunotherapy, and combination strategies have come into the spotlight and have become the subject of intense research in this setting, aiming to overcome the hurdle of the intrinsic treatment resistance caused by TP53 mutations. In this review, we discuss the role of TP53 mutations in AML and outcomes based on current treatment options, as well as examining data on innovative agents that are currently being investigated in the preclinical setting and clinical trials.

1.1. The Role of TP53 in AML

TP53 is a 20-kbp tumor suppressor gene located on chromosome 17p13.1 [6]. It encodes for the transcription factor p53 and functions as the “guardian of the human genome” [6]. The p53 protein is a key transcription factor, playing a pivotal role in tumor suppression through DNA repair, cell cycle arrest, differentiation, senescence, apoptosis, autophagy, metabolism, and chemosensitivity [11,12]. The protein contains five important domains: the N-terminal trans-activation domain, a proline-rich domain, a central DNA-binding domain (DBD), a C-terminal oligomerization domain, and a regulatory domain [6].
Since its first description in 1979, TP53 has been the most frequently mutated gene across all human cancers. It has been previously described that more than 50% of human tumors carry TP53 mutations, and that many others carrying wild-type TP53 alleles exhibit decreased TP53 activity via other mechanisms [13,14]. However, recent data have demonstrated that TP53 mutations are found in 39% of patients with cancer, a frequency lower than that of previous reports [15]. One of the most well-studied functions of TP53 is its role in limiting cellular proliferation in response to aberrant oncogene expression. Therefore, TP53 inactivation through gene deletion or mutation enhances the effect of oncogenes and plays a key role in promoting uncontrolled proliferation of cancer cells. Germline TP53 mutations cause Li-Fraumeni syndrome (LFS), a disorder that predisposes patients to different types of cancer, including sarcomas, breast cancer, leukemias, and lymphomas [16,17].
It has been observed that the vast majority of de novo AML cases have intact, unaltered TP53 alleles [18]. However, the frequency of genomic TP53 alterations is increased in certain patients [4,5,6,7]. In AML, TP53 mutations are mostly missense somatic substitutions, mostly heterozygous, and include those that are observed in the known hotspot sites of the gene [19]. Diverse genetic aberrations in TP53, such as chromosomal alterations leading to allelic gain, loss, or frameshift insertions and deletions have also been described, with the impact ranging from partial to complete loss of function (LOF), mostly in the germline LOF mutations that underlie LFS [19]. Gain of function (GOF) mutations with varied effect sizes are also present in different TP53 mutants and are thought to mostly result from their binding to different proteins, including transcription factors [20,21]. GOF TP53 mutants have also been reported to affiliate with epigenetic pathways, e.g., binding and enhancing transcription of the methyl-transferases MLL1 and MLL2 [22]. Monoallelic TP53 mutations frequently have co-mutations in other genes, mostly TET2, SF3B1, ASXL1, and DNMT3A, and are likely to be subclonal events with varying impacts on outcomes of MDS/AML [23]. On the other hand, multihit TP53mut MDS/AML represents a distinct disorder, with co-mutations occurring in less than 25% of cases [24]. Finally, the mutational burden of TP53 has also arisen as a crucial prognostic factor and determinant of therapy choice in AML cases [25]. Despite being one of the most studied genes, TP53 is still considered “undruggable”, so future studies are needed to ascertain the role of TP53 mutations in myeloid malignancies.

1.2. Current Treatment Options for TP53-Mutated AML

Intensive chemotherapy (IC) with an anthracycline and cytosine arabinoside (AraC) remains the backbone of treatment in patients with newly diagnosed (ND) AML. Eligibility for IC is largely based upon age and comorbidities, hence patients with TP53mut AML, who are frequently elderly, may be unfit for this treatment option. Additionally, the presence of TP53 mutations in AML patients who receive anthracycline- and cytarabine-based induction chemotherapy has been previously associated with inferior outcomes, with reported initial response rates of 20–30% and poor OS of less than a year [4,5]. Baseline TP53 variant allelic frequency (VAF) has been previously shown to be predictive of response to cytarabine-based treatment, with VAF ≤ 40% being associated with a superior CR/CR with incomplete hematologic recovery (CRi) rate of 79% and a median OS of 7.3 months. This is juxtaposed with VAF > 40%, which has been associated with a CR/CRi rate of 35% and a median OS of 4.7 months [25].
Lower-intensity therapies, including low-dose cytarabine (LDAC) monotherapy or in combination with cladribine and hypomethylating agents (HMAs), are also being used in these patients and are an attractive option since they are accompanied by significantly lower toxicity. There is conflicting data available regarding the efficacy of lower-intensity chemotherapy regimens. A single-center study demonstrated superior CR rates in TP53mut AML patients receiving IC as compared to patients treated with lower-intensity regimens (45% vs. 14.3%), but no difference in OS (8.8 months versus 9.4 months respectively) [26]. On the contrary, another study demonstrated lower CR rates among patients with TP53mut AML regardless of regimen intensity, and also showed that the intensity of therapy does not predict improved survival [3].
Azacitidine (AZA) and decitabine (DEC) are HMAs that are currently being used either alone or in combination with other agents in the management of TP53mut AML. Although the efficacy of AZA monotherapy in AML has been previously demonstrated [27], with a reported CR/CRi rate of 28% [27], its efficacy in AML harboring TP53 mutations is not well established. A randomized phase three trial comparing the impact of AZA on the survival of AML patients versus conventional care regimens (CCREs), including IC, LDAC, and best supportive care, has shown that the median OS was prolonged in TP53mut patients treated with AZA compared to those treated with CCREs (7.2 months versus 2.4 months, respectively); however, this result did not reach statistical significance [28].
Correspondingly, data regarding the efficacy of DEC monotherapy is conflicting [29,30]. A retrospective study has shown similar CR rates among TP53mut AML patients treated with either LDAC or a 5-day or 10-day DAC regimen (DEC5 and DEC10, respectively), as well as comparable OS rates among all treatment arms [29]. Accordingly, a study of AML patients treated with DEC has also shown no response or survival benefit in TP53mut patients versus TP53 wild type (TP53wt) ones [30]. Conversely, a single-institution trial evaluated the efficacy of DEC10 in AML patients and demonstrated exceptionally higher responses in TP53mut patients (100% versus 41% in the TP53wt arm) [31]. These responses were accompanied by clearance of TP53mut leukemic clones in most of the cases, but mutation clearance was never complete [31]. Although TP53 VAF predicts response and OS in AML patients treated with IC, no effect has been demonstrated on response rates and OS in those treated with HMAs [25]. Moreover, despite the fact that DEC augments chemotherapy responses in TP53mut AML, with a currently unknown underlying mechanism, these responses are not durable and do not significantly affect subclones bearing TP53 mutations [31]. Nonetheless, this enhanced effect paves the way for the design of more combination strategies in these patients.
Recently, the combination of AZA and Venetoclax (VEN), a selective B-cell lymphoma-2 (BCL-2) inhibitor, has become the cornerstone in the treatment of elderly AML patients who are ineligible for IC [32]. First-line treatment of TP53mut AML with poor-risk cytogenetics using AZA and VEN initially showed promising results, with a study reporting CR and CRi combined rates of 41% in the combination arm versus 17% in the AZA monotherapy arm, exceeding the historical standards of 28% CR rates [5,33]. However, the duration of response (DOR) and median OS were similar among both treatment arms (6.5 versus 6.7 months and 5.2 versus 4.9 months, respectively) [33]. Furthermore, a study evaluating the efficacy of DEC10 and VEN combination in patients with ND AML has shown pronouncedly inferior outcomes in TP53mut patients compared to TP53wt patients, with reported ORR and CR/CRi rates in TP53mut patients of 66% and 57%, versus 89% and 77%, respectively, in the TP53wt group [34]. Importantly, the 60-day mortality rate was higher in TP53mut patients (26% versus 4% in TP53wt), and OS was profoundly lower in these patients (5.2 versus 19.4 months in TP53wt) [34]. It has been previously demonstrated that TP53 mutations disrupt the BAX/BAK pathway and establish an elevated activation threshold in leukemic cells (LCs). Although VEN initially suppresses this effect, LCs finally avoid BCL-2 inhibition due to competitive advantage, thus conferring resistance to VEN [35]. Additionally, adaptive resistance associated with alterations in mitochondrial homeostasis and increased oxidative phosphorylation has also been observed [36,37]. Despite this resistance to VEN, its incorporation in novel combination therapies in TP53mut AML may still be promising. Concurrent inhibition of BCL-2 and myeloid leukemia 1 (MCL-1) can achieve long-term outcomes by increasing the early apoptotic response in TP53-deficient cells, thus making this approach highly promising [36].
ASCT remains the only potentially curative option for high-risk AML patients who are in remission after induction treatment. Prior data have supported that transplanted TP53mut patients have yielded superior outcomes as compared to non-transplanted patients who receive other treatment regimens or palliative care [3,25,38]. However, whether aSCT is beneficial for TP53mut patients is controversial. A single-center study has shown that although transplanted patients have had significantly superior outcomes in comparison to non-transplanted patients, this association was lost in time-dependent and landmark analysis [39]. Previous studies have also shown poor outcomes in TP53mut patients with AML/MDS and AML as compared to TP53wt patients, with an increased risk of relapse and death [8,39,40]. A recent meta-analysis has shown a pooled two-year OS of 30% and a pooled relapse rate of 61% at two years post aSCT [41]. Moreover, only a few patients, irrespective of age or performance status, are able to proceed to aSCT at first remission [25,34]. A large multicenter trial (COMMAND) has demonstrated that only 18% of TP53mut patients have been bridged to aSCT [42]. The COMMAND trial has also reported a median event-free survival (EFS) and median OS of 12.4 and 24.5 months, respectively [42]. Conversely, a 3-year EFS and OS rate of 61% and 66.3% have been recorded by a retrospective study of TP53mut patients post aSCT [43]. Lower TP53 VAF has been associated with improved clinical outcomes in transplanted patients [25,39]. Hence, VAF may be implemented as a future tool for selecting which TP53mut patients may benefit from aSCT. Among other factors, pretransplant minimal residual disease (MRD) and complex karyotype have been associated with inferior outcomes, while the type of pre-aSCT treatment and conditioning regimen have not had a significant impact on clinical outcomes [39,43,44]. Nevertheless, efforts to optimize transplant outcomes by modifying the intensity of conditioning or using novel drug combinations as induction or maintenance treatment are warranted. Notwithstanding the poor outcomes reported, aSCT still remains an appealing choice for achieving long-term survival in TP53mut patients.

1.3. Currently Available Combination Strategies

Recently, several combination strategies for the management of AML patients harboring TP53 mutations have been investigated in clinical trials. A recent cohort study evaluated the efficacy of DEC, LDAC, aclarubicin, and granulocyte colony-stimulating factor (G-CSF) [DCAG regimen] versus standard chemotherapy in TP53mut AML patients [37], based on the previous encouraging results of a multicenter phase 2 trial which reported 82.4% ORR and 64.7% CR rates relating to the DCAG regimen in elderly AML patients [45]. Although differences were not statistically significant, a trend towards higher ORR, CR, and OS rates was observed in the DCAG arm [37]. Importantly, patients with poor cytogenetics in the DCAG arm displayed superior responses with a significantly higher CR rate of 56.3% and a median OS of 7.8 months (versus a CR of 0% and median OS of 3 months in the standard chemotherapy arm) [37].
The combination of LDAC with clofarabine or cladribine alternating with DEC has been evaluated in the management of treatment-naïve elderly AML patients with reported CR and CRi rates of 59% and 7%, respectively, and a median OS of 12.5 months [46,47,48]. Long-term results from these studies have shown that among all patients, those with TP53 mutations yielded the lowest responses, with a composite complete remission (cCR) rate of 44% and a poor median OS of 5.4 months [48]. Addition of AZA prior to treatment with high-dose cytarabine (HiDAC) and mitoxantrone, considering that epigenetic priming induced by AZA before cytotoxic chemotherapy could contribute to enhanced responses has been previously examined in a phase 1 study of high-risk AML patients, resulted in an ORR of 61% [49]. However, patients with TP53 mutations seemed not to benefit from this regimen [49]. A study of TP53mut AML patients demonstrated that the combination of DEC, chidamide, a histone deacetylase inhibitor (HDACi) with a priming regimen consisting of omacetaxine mepesuccinate (an alkaloid herbal derivative), cytarabine, and G-CSF (HAG) yielded potent responses with an ORR of 71.4% and manageable toxicity [50]. The study sample size was small, and thus, definite conclusions cannot be drawn; however, these promising results warrant further investigation in the near future [50].
CPX-351, a liposomal formulation of cytarabine and daunorubicin, constitutes the contemporary treatment of AML with myelodysplasia-related changes (MRC-AML) and t-AML [51]. Real-life data from the French cohort study of CPX-351 indicated that TP53 mutations were the only predictive factor of inferior responses in multivariate analysis, although high-risk molecular prognosis subgroups, including patients with ASXL1 and RUNX1 mutations, displayed higher than expected response rates [51]. In accordance, similar results were reported by another retrospective study, demonstrating inferior responses in TP53mut patients, who achieved lower CR and CRi rates as compared to TP53wt patients (33% versus 62%, respectively) [52]. Consistently, a post hoc analysis of a randomized phase 3 trial showed poor outcomes in TP53mut patients [53]. Opposingly, a German retrospective analysis demonstrated that the presence of TP53 mutations did not impact responses to CPX-351 or survival [54]. Nonetheless, the role of CPX-351 in the management of TP53mut AML needs to be further evaluated.

1.4. Novel Therapeutic Agents

Considerable progress has also been made regarding the development of novel agents, including mutant p53-targeted approaches and immunotherapy.

1.4.1. 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 [55,56,57]. 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% [55]. 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 [56]. 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 [57]. Moreover, the DOR differed significantly among these patients [57]. 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%. although these results seem discouraging, data are scarce and derived from small studies; thus, PEVO may still have a role to play in this setting.
Ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor has been shown to impede the proliferation of human AML blasts in vitro, either alone or combined with cytarabine or daunorubicin [58]. A randomized phase 2 study evaluated the outcomes of adding ibrutinib to DEC10 versus DEC10 monotherapy in elderly, previously untreated AML patients [59]. Surprisingly, although the addition of ibrutinib did not yield favorable outcomes, TP53mut was correlated with higher responses and CR/CRi rates of 56% [59]. However, these responses did not translate into a superior OS [59]. Although ibrutinib’s efficacy in TP53mut AML needs to be further validated, it remains a highly appealing approach.
Finally, bortezomib, a proteasome inhibitor, has been widely investigated for use in the management of AML patients, since it has been associated with potent antiproliferative properties [60]. A randomized phase 2 trial of AML patients treated with either combined bortezomib and DEC10 or DEC10 alone failed to demonstrate any advantage of the combination treatment in those with TP53 mutations [60]. Moreover, the addition of bortezomib conferred no benefit to the study patients overall [60]. Conclusively, the efficacy of targeted therapies remains ambiguous. Further exploration of these agents in TP53mut AML through large clinical trials is warranted.

1.4.2. 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. This molecule induces protein unfolding and restores wild-type functions to mutant p53, such as induction of apoptosis and promotion of cell cycle arrest [61]. Eprenetapopt (EP) or APR-246, a methylated derivative of PRIMA-1 (PRIMA-1MET), is a first-in-class agent that binds covalently to cysteine residues in mutant p53 protein [61]. Preclinical studies have demonstrated that EP exerts apoptotic effects on AML cell lines and primary LCs from AML patients in a dose-dependent manner [62]. Noteworthily, the presence of TP53 mutations did not significantly affect sensitivity to this agent [62]. Subsequent studies have shown significant synergistic cytotoxicity of EP and AZA in TP53mut primary cells from MDS/AML patients [63]. Apart from the reported mutant p53 reactivation, preclinical data have also demonstrated that EP results in glutathione depletion and induction of ferroptosis irrespective of TP53 status, thus indicating a different mechanism of action that leads to p53-independent cell death [64,65].
Recently, EP’s efficacy in combination with AZA has been evaluated in patients with TP53mut MDS and AML in two phase 2 studies, one in the USA and another in Europe [66,67]. EP was administered by 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 [66,67]. TP53mut AML patients in the US trial achieved ORR and CR rates of 64% and 36%, respectively, and a median OS of 10.8 months [66]. However, the sample size was small and only patients with oligoblastic AML (20–30% marrow blasts) were included [66]. 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% [67]. However, none of the patients with a high blast count achieved a CR [67]. the median OS in patients with less and more than 30% marrow blasts was 13.9 months and 3.0 months, respectively [67]. 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%) [67]. 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 [67]. Of note, patients with TP53mut MDS have also yielded high response rates in both studies, with a CR rate of around 50% [66,67]. 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 encouraging for this high-risk population [68]. 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 [69]. Importantly, blast count did not have an impact on patients’ responses [69]. Moreover, TP53 negativity (VAF < 5%) by NGS was achieved in 27% of patients [69]. These results are highly promising, since the observed 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 [69]. Collectively, EP has demonstrated promising efficacy in TP53mut AML patients and provides the basis for further investigation in randomized clinical trials in the near future.

1.4.3. Immunotherapeutic Approaches

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 [70]. LCs have high levels of CD47, thus escaping immune surveillance [70]. 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 [71]. Blockade of CD47 in AML models has resulted in the induction of phagocytosis and elimination of LCs [71,72]. 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 [72,73]. A phase 1b study has evaluated the combination of MAG and AZA in patients with previously untreated AML who were ineligible for IC, with the majority of patients (82.8%) having TP53 mutations [73]. The CR rate was similar among TP53mut and TP53wt patients (31.9% and 32.2%, respectively), whereas the OS was 9.8 months and 18.9 months, respectively [73]. 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 demonstrated an ORR and a CR rate of 74% and 41%, respectively, in ND TP53mut patients [74]. 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 as MAG failed to demonstrate a survival benefit compared to the current standard of care [75].
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 [76]. In vivo studies of AML xenografts have demonstrated the efficacy of TTI-622 in enhancing macrophage-mediated phagocytosis [76]. 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, NCT04912063). A recent phase 1b study has evaluated the efficacy of AK117, an anti-CD47 agent, in combination with AZA as a frontline treatment for AML patients and has demonstrated a CR and CR/CRi rate of 45% and 55%, respectively [77]. 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 [78]. Hence, EVO entered a phase 1/2 trial, which aimed to study 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 [79]. 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. Concisely, the employment of anti-CD47 agents in the treatment of TP53mut has been met with unsatisfactory results.
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 [80,81]. When interacting with one of its ligands, such as galectin-9, TIM-3 prompts the inhibition of T-cell responses [80,81]. 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 [80,81]. 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 [82]. 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 [82]. 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 [82]. 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, and it also participates in the modulation of T-cell responses [83]. CD123 is widely expressed in blasts of AML patients, and its overexpression has been correlated with poor prognosis [84]. 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 [83]. Flotetuzumab (FLOT) is a CD123 × 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% [85]. Remarkably, TP53mut patients yielded encouraging responses with a CR rate of 47% and a median OS of 10.3 months in responding patients [86]. 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 [87]. An ongoing multicenter phase 1/2 study is investigating PVEK as a triplet with AZA and VEN or in combination with MAG in patients with R/R AML or ND CD123+ AML [88]. 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 [87]. However, VEN-naïve patients yielded significantly higher responses than those with prior exposure to VEN [87]. 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 [89]. Rapid MRD negativity was reported in 73% of patients achieving coCR [89]. Exceptionally, high coCRMRD rates were demonstrated among adverse risk patients, TP53mut patients included [89]. Triple combination therapy has been also associated with a manageable safety profile [87,89]. 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 [90]. 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 [91]. A phase 1b trial of TAG with AZA and/or VEN in AML and MDS patients is ongoing and preliminary results indicate promising efficacy [92]. Remarkably, TP53mut patients have achieved a CR/CRi/morphologic leukemia-free state (MLFS) rate of 54%, with a CR rate of 31% [92]. 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% [93]. Vibecotamab has also been associated with cytokine release syndrome (CRS), which is manageable with premedication [93]. Other CD123-targeting agents that are in early clinical development include APV0436, MGD024, and CD123 chimeric antigen receptor T-cell (CAR-T) therapy [94]. In summary, these results suggest that these agents may have a role to play in the treatment of 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 [95]. 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 [96]. 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 [97]. A recent phase 2 trial of R/R AML patients receiving pembrolizumab, a PD-1 inhibitor, with HiDAC demonstrated promising clinical activity in TP53mut patients, reporting a CR rate of 40% [98]. A randomized phase 2 trial of AZA with or without durvalumab (DURV), a PD-L1 inhibitor, as first-line treatment for elderly AML patients failed to show a potential benefit; the addition of DURV did not enhance clinical outcomes, and recorded ORR and OS were similar among both treatment arms [99]. Interestingly, responses were similar between TP53mut and TP53wt patients (ORR 35% and 34%, respectively) [99]. 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 [100]. 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. Its expression is relatively lower in normal HSCs, thus rendering LAIR-1 an ideal anti-leukemic target [100]. NC525 is a humanized monoclonal antibody that binds specifically to AML blasts and LSCs while sparing normal hematopoiesis. It induces apoptosis through a unique signaling pathway, without evidence of immunomodulatory effects on other immune subsets [100]. 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 [100]. A phase 1 trial investigating the safety and tolerability of NC525 in patients with advanced HMs, including R/R AML is underway (NCT05787496).

1.4.4. Other Agents

Murine double minute protein 2 (MDM2) is an E3 ubiquitin ligase that negatively regulates the activity of p53 [101]. MDM2 interacts with p53 and promotes its degradation via ubiquitination [101]. Inhibition of MDM2 mediates antileukemic effects in TP53wt AML through an increase in p53 levels [101]. 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 [102]. 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 [103]. 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 [104]. Milademetan, an MDM2i, in combination with LDAC, with or without VEN, has been recently explored in AML with discouraging responses and significant gastrointestinal toxicity [105]. 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 [106]. 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 [105,106]. 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 [107]. Therefore, it can lead to inactivation of proliferation of LCs and apoptosis promotion. Atorvastatin is a potent destabilizing agent of mutant p53; 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 [108]. 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.

1.4.5. Novel Treatments in TP53mut AML: Does a Promising Future Await?

In brief, targeted treatments, including those targeting mutant p53, along with immunotherapeutic agents, have yielded vastly different response rates in TP53mut AML patients, as seen in Table 1 [53,54,55,56,57,58,64,65,66,67,71,72,80,84,87,90,94,95,96,97,104]. However, these responses have not translated into a survival benefit; the reported median OS was less than a year in the majority of the studies [53,54,55,56,57,58,64,65,66,67,71,72,80,84,87,90,94,95,96,97,104]. Although these results may be discouraging, they derive mostly from small studies; hence, further study is required and these agents may still hold promise for this challenging clinical setting, particularly in combination with HMAs. As seen in Table 2, several ongoing early-phase trials are investigating the use of novel agents in TP53mut AML patients and may offer a promising treatment option for these patients in the near future.

1.5. Novel Agents in Preclinical Studies

Despite growth in the understanding of AML pathobiology, therapeutic progress is still inadequate. The requirement for improvement has yielded development of novel drugs targeting various molecularly defined AML entities, including p53-based therapies. Cells with mutant or deleted TP53 frequently have a defective G1 checkpoint and are more dependent on the G2 checkpoint to repair DNA damage; the G2 checkpoint allows p53-deficient AML cells to repair genetic lesions and continue through the cell cycle. Consistent with this finding, inhibition of kinases involved in the G2 checkpoint, such as aurora kinase A (AURKA) and aurora kinase B (AURKB), has induced mitotic catastrophe and p53-independent cell death in TP53mut cancer cells [109]. TP-0903, a small molecule originally developed as an AXL inhibitor, is a multikinase inhibitor with activity against AURKA/B, Chk1/2, and other cell cycle regulators and has activity in models of drug-resistant AML with both WT and mutated TP53 [109]. Xpo7, a putative nuclear/cytoplasmic transporter, was recently identified as a factor necessary for the survival of Trp53-knockout (KO) AML cells with the performance of genome-wide CRISPR-Cas9 screens using Trp53-KO and WT mouse AML cells, indicating a synthetic lethal relationship between TP53 and XPO7 [110]. TP53mut targeted therapy aims to abolish TP53mut cancer cells or to rescue p53 mutational inactivation. Pharmacological strategies are directed toward regaining p53wt-like conformation and p35mut tumor-suppressive functions, abrogating distinct mechanisms underlying p53mut GOF, and promoting p53mut degradation [111,112,113,114,115,116,117]. On the other hand, dysfunctional p53wt targeted therapy aims to rescue p53wt by addressing various AML-related p53wt inactivating mechanisms. As aforementioned, one such strategy involves MDM2i that disrupts WTp53-MDM2 interactions [101]. Table 3 summarizes the available preclinical studies targeting TP53/p53 in AML in vitro and in vivo models. Finally, agents that can help overcome resistance to currently available therapies have also been investigated. Targeting mitochondrial metabolism with novel antimitochondrial agents, including electron transport chain complex inhibitors, pyruvate dehydrogenase inhibitors, and mitochondrial ClpP protease agonists has led to enhanced sensitivity of leukemic cells to combination treatment with VEN and AraC and substantially delayed relapse [118].

2. Conclusions

In conclusion, the management of TP53mut AML remains a formidable clinical challenge. Current therapeutic approaches yield suboptimal outcomes, thus denoting the urgency for tailored strategies addressing the molecular landscape of TP53 mutations along with the inherent resistance and aggressive nature of the disease. Although the armamentarium of promising approaches keeps expanding, most novel agents have not met with satisfactory efficacy, with survival rates similar to current treatments. However, these data derive mostly from quite small studies, so strong conclusions cannot be drawn. Among novel treatments, immunotherapeutic agents such as pevonedistat, nivolumab, and flotetuzumab have displayed promising efficacy and warrant rigorous investigation through large clinical trials. Preclinically, agents that target TP53/p53 have also yielded encouraging responses, thus necessitating their study in the clinical setting. What is certain is that as we delve deeper into the molecular landscape of AML, the significance of TP53 mutations becomes increasingly apparent, thus requiring a paradigm shift in our clinical strategies, with hopes of fostering a brighter future for patients with TP53mut AML.

Funding

This research received no external funding.

Conflicts of Interest

C.S. reports personal fees from Abbvie, outside of the submitted work. P.D. reports personal fees from Novartis, Amgen, Janssen, Abbvie, BMS, and Roche, outside of the submitted work.

References

  1. Lakin, N.D.; Jackson, S.P. Regulation of p53 in response to DNA damage. Oncogene 1999, 18, 7644–7655. [Google Scholar] [CrossRef]
  2. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Godley, L.A.; Hasserjian, R.P.; et al. Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 2022, 140, 1345–1377. [Google Scholar] [CrossRef]
  3. Kadia, T.M.; Jain, P.; Ravandi, F.; Garcia-Manero, G.; Andreef, M.; Takahashi, K.; Borthakur, G.; Jabbour, E.; Konopleva, M.; Daver, N.G.; et al. TP53 mutations in newly diagnosed acute myeloid leukemia: Clinicomolecular characteristics, response to therapy, and outcomes. Cancer 2016, 122, 3484–3491. [Google Scholar] [CrossRef]
  4. Bowen, D.; Groves, M.J.; Burnett, A.K.; Patel, Y.; Allen, C.; Green, C.; Gale, R.E.; Hills, R.; Linch, D.C. TP53 gene mutation is frequent in patients with acute myeloid leukemia and complex karyotype, and is associated with very poor prognosis. Leukemia 2009, 23, 203–206. [Google Scholar] [CrossRef]
  5. Rücker, F.G.; Schlenk, R.F.; Bullinger, L.; Kayser, S.; Teleanu, V.; Kett, H.; Habdank, M.; Kugler, C.M.; Holzmann, K.; Gaidzik, V.I.; et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood 2012, 119, 2114–2121. [Google Scholar] [CrossRef]
  6. George, B.; Kantarjian, H.; Baran, N.; Krocker, J.D.; Rios, A. TP53 in Acute Myeloid Leukemia: Molecular Aspects and Patterns of Mutation. Int. J. Mol. Sci. 2021, 22, 10782. [Google Scholar] [CrossRef]
  7. Hunter, A.M.; Sallman, D.A. Current status and new treatment approaches in TP53 mutated AML. Best Pract. Res. Clin. Haematol. 2019, 32, 134–144. [Google Scholar] [CrossRef] [PubMed]
  8. Della Porta, M.G.; Gallì, A.; Bacigalupo, A.; Zibellini, S.; Bernardi, M.; Rizzo, E.; Allione, B.; van Lint, M.T.; Pioltelli, P.; Marenco, P.; et al. Clinical Effects of Driver Somatic Mutations on the Outcomes of Patients with Myelodysplastic Syndromes Treated with Allogeneic Hematopoietic Stem-Cell Transplantation. J. Clin. Oncol. 2016, 34, 3627–3637. [Google Scholar] [CrossRef] [PubMed]
  9. Madarász, K.; Mótyán, J.A.; Bedekovics, J.; Miltényi, Z.; Ujfalusi, A.; Méhes, G.; Mokánszki, A. Deep Molecular and In Silico Protein Analysis of p53 Alteration in Myelodysplastic Neoplasia and Acute Myeloid Leukemia. Cells 2022, 11, 3475. [Google Scholar] [CrossRef] [PubMed]
  10. Tavor, S.; Rothman, R.; Golan, T.; Voskoboinik, N.; Katz, B.Z.; Sarid, N.; Shomrat, R.; Orr-Urtreger, A.; Naparstek, E. Predictive value of TP53 fluorescence in situ hybridization in cytogenetic subgroups of acute myeloid leukemia. Leuk. Lymphoma 2011, 52, 642–647. [Google Scholar] [CrossRef] [PubMed]
  11. Levine, A.J. p53: 800 million years of evolution and 40 years of discovery. Nat. Rev. Cancer 2020, 20, 471–480. [Google Scholar] [CrossRef] [PubMed]
  12. Ferreira, C.G.; Tolis, C.; Giaccone, G. p53 and chemosensitivity. Ann. Oncol. 1999, 10, 1011–1021. [Google Scholar] [CrossRef]
  13. Vogelstein, B.; Lane, D.; Levine, A.J. Surfing the p53 network. Nature 2000, 408, 307–310. [Google Scholar] [CrossRef]
  14. Hainaut, P.; Hernandez, T.; Robinson, A.; Rodriguez-Tome, P.; Flores, T.; Hollstein, M.; Harris, C.C.; Montesano, R. IARC database of p53gene mutations in human tumors and cell lines: Updated compilation, revised formats and new visualisation tools. Nucleic Acids Res. 1998, 26, 205–213. [Google Scholar] [CrossRef] [PubMed]
  15. Sosinsky, A.; Ambrose, J.; Cross, W.; Turnbull, C.; Henderson, S.; Jones, L.; Hamblin, A.; Arumugam, P.; Chan, G.; Chubb, D.; et al. Insights for precision oncology from the integration of genomic and clinical data of 13,880 tumors from the 100,000 Genomes Cancer Programme. Nat. Med. 2024, 30, 279–289. [Google Scholar] [CrossRef] [PubMed]
  16. Olivier, M.; Hollstein, M.; Hainaut, P. TP53 mutations in human cancers: Origins, consequences, and clinical use. Cold Spring Harb. Perspect. Biol. 2010, 2, a001008. [Google Scholar] [CrossRef]
  17. Srivastava, S.; Zou, Z.; Pirollo, K.; Blattner, W.; Chang, E.H. Germline transmission of a mutated p53 gene in a cancer-prone family with Li-Fraumeni syndrome. Nature 1990, 348, 747–749. [Google Scholar] [CrossRef]
  18. 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 the cancer genome atlas research network. N. Engl. J. Med. 2013, 368, 2059–2074. [Google Scholar]
  19. Barbosa, K.; Li, S.; Adams, P.D.; Deshpande, A.J. The role of TP53 in acute myeloid leukemia: Challenges and opportunities. Genes Chromosomes Cancer 2019, 58, 875–888. [Google Scholar] [CrossRef]
  20. Brosh, R.; Rotter, V. When mutants gain new powers: News from the mutant p53 field. Nat. Rev. Cancer 2009, 9, 701–713. [Google Scholar] [CrossRef]
  21. Shetzer, Y.; Molchadsky, A.; Rotter, V. Oncogenic mutant p53 gain of function nourishes the vicious cycle of tumor development and cancer stem-cell formation. Cold Spring Harb. Perspect. Med. 2016, 6, a026203. [Google Scholar] [CrossRef]
  22. Zhu, J.; Sammons, M.A.; Donahue, G.; Dou, Z.; Vedadi, M.; Getlik, M.; Barsyte-Lovejoy, D.; Al-awar, R.; Katona, B.W.; Shilatifard, A.; et al. Gain-of-function p53mutants co-opt chromatin pathways to drive cancer growth. Nature 2015, 525, 206–211. [Google Scholar] [CrossRef]
  23. Bernard, E.; Nannya, Y.; Hasserjian, R.P.; Devlin, S.M.; Tuechler, H.; Medina-Martinez, J.S.; Yoshizato, T.; Shiozawa, Y.; Saiki, R.; Malcovati, L.; et al. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat. Med. 2020, 26, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
  24. Sill, H.; Zebisch, A.; Haase, D. Acute myeloid leukemia and myelodysplastic syndromes with TP53 aberrations: A distinct stem cell disorder. Clin. Cancer Res. 2020, 26, 5304–5309. [Google Scholar] [CrossRef]
  25. Short, N.J.; Montalban-Bravo, G.; Hwang, H.; Ning, J.; Franquiz, M.J.; Kanagal-Shamanna, R.; Patel, K.P.; DiNardo, C.D.; Ravandi, F.; Garcia-Manero, G.; et al. Prognostic and therapeutic impacts of mutant TP53 variant allelic frequency in newly diagnosed acute myeloid leukemia. Blood Adv. 2020, 4, 5681–5689. [Google Scholar] [CrossRef] [PubMed]
  26. Bewersdorf, J.P.; Shallis, R.M.; Gowda, L.; Wei, W.; Hager, K.; Isufi, I.; Kim, T.K.; Pillai, M.M.; Seropian, S.; Podoltsev, N.A.; et al. Clinical outcomes and characteristics of patients with TP53-mutated acute myeloid leukemia or myelodysplastic syndromes: A single center experience. Leuk. Lymphoma 2020, 61, 2180–2190. [Google Scholar] [CrossRef] [PubMed]
  27. Dombret, H.; Seymour, J.F.; Butrym, A.; Wierzbowska, A.; Selleslag, D.; Jang, J.H.; Kumar, R.; Cavenagh, J.; Schuh, A.C.; Candoni, A.; et al. International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. Blood 2015, 126, 291–299. [Google Scholar] [CrossRef] [PubMed]
  28. Döhner, H.; Dolnik, A.; Tang, L.; Seymour, J.F.; Minden, M.D.; Stone, R.M.; Del Castillo, T.B.; Al-Ali, H.K.; Santini, V.; Vyas, P.; et al. Cytogenetics and gene mutations influence survival in older patients with acute myeloid leukemia treated with azacitidine or conventional care. Leukemia 2018, 32, 2546–2557. [Google Scholar] [CrossRef] [PubMed]
  29. Boddu, P.; Kantarjian, H.; Ravandi, F.; Garcia-Manero, G.; Borthakur, G.; Andreeff, M.; Jabbour, E.J.; Benton, C.B.; DiNardo, C.D.; Konopleva, M.; et al. Outcomes with lower intensity therapy in TP53-mutated acute myeloid leukemia. Leuk. Lymphoma 2018, 59, 2238–2241. [Google Scholar] [CrossRef] [PubMed]
  30. Middeke, J.M.; Teipel, R.; Röllig, C.; Stasik, S.; Zebisch, A.; Sill, H.; Kramer, M.; Scholl, S.; Hochhaus, A.; Jost, E.; et al. Decitabine treatment in 311 patients with acute myeloid leukemia: Outcome and impact of TP53 mutations—A registry based analysis. Leuk. Lymphoma 2021, 62, 1432–1440. [Google Scholar] [CrossRef]
  31. Welch, J.S.; Petti, A.A.; Miller, C.A.; Fronick, C.C.; O’Laughlin, M.; Fulton, R.S.; Wilson, R.K.; Baty, J.D.; Duncavage, E.J.; Tandon, B.; et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N. Engl. J. Med. 2016, 375, 2023–2036. [Google Scholar] [CrossRef] [PubMed]
  32. 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]
  33. Pollyea, D.A.; Pratz, K.W.; Wei, A.H.; Pullarkat, V.; Jonas, B.A.; Recher, C.; Babu, S.; Schuh, A.C.; Dail, M.; Sun, Y.; et al. Outcomes in Patients with Poor-Risk Cytogenetics with or without TP53 Mutations Treated with Venetoclax and Azacitidine. Clin. Cancer Res. 2022, 28, 5272–5279. [Google Scholar] [CrossRef] [PubMed]
  34. Kim, K.; Maiti, A.; Loghavi, S.; Pourebrahim, R.; Kadia, T.M.; Rausch, C.R.; Furudate, K.; Daver, N.G.; Alvarado, Y.; Ohanian, M.; et al. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer 2021, 127, 3772–3781. [Google Scholar] [CrossRef] [PubMed]
  35. Thijssen, R.; Diepstraten, S.T.; Moujalled, D.; Chew, E.; Flensburg, C.; Shi, M.X.; Dengler, M.A.; Litalien, V.; MacRaild, S.; Chen, M.; et al. Intact TP-53 function is essential for sustaining durable responses to BH3-mimetic drugs in leukemias. Blood 2021, 137, 2721–2735. [Google Scholar] [CrossRef]
  36. Nechiporuk, T.; Kurtz, S.E.; Nikolova, O.; Liu, T.; Jones, C.L.; D’Alessandro, A.; Culp-Hill, R.; d’Almeida, A.; Joshi, S.K.; Rosenberg, M.; et al. The TP53 Apoptotic Network Is a Primary Mediator of Resistance to BCL2 Inhibition in AML Cells. Cancer Discov. 2019, 9, 910–925. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, S.S.; Sun, Q.; Cao, L.; Wu, W.Z.; Xie, Y.; Qiao, C.; Li, J.Y.; Qian, S.X.; Hong, M. Efficacy and safety of decitabine combined with low-dose cytarabine, aclarubicin, and granulocyte colony-stimulating factor compared with standard therapy in acute myeloid leukemia patients with TP53 mutation. Chin. Med. J. 2020, 134, 1477–1479. [Google Scholar] [CrossRef] [PubMed]
  38. Badar, T.; Atallah, E.; Shallis, R.M.; Goldberg, A.D.; Patel, A.; Abaza, Y.; Bewersdorf, J.P.; Saliba, A.N.; Correia, G.S.C.; Murthy, G.; et al. Outcomes of TP53-mutated AML with evolving frontline therapies: Impact of allogeneic stem cell transplantation on survival. Am. J. Hematol. 2022, 97, E232–E235. [Google Scholar] [CrossRef]
  39. Zhao, D.; Zarif, M.; Zhou, Q.; Capo-Chichi, J.M.; Schuh, A.; Minden, M.D.; Atenafu, E.G.; Kumar, R.; Chang, H. TP53 Mutations in AML Patients Are Associated with Dismal Clinical Outcome Irrespective of Frontline Induction Regimen and Allogeneic Hematopoietic Cell Transplantation. Cancers 2023, 15, 3210. [Google Scholar] [CrossRef]
  40. Yoshizato, T.; Nannya, Y.; Atsuta, Y.; Shiozawa, Y.; Iijima-Yamashita, Y.; Yoshida, K.; Shiraishi, Y.; Suzuki, H.; Nagata, Y.; Sato, Y.; et al. Genetic abnormalities in myelodysplasia and secondary acute myeloid leukemia: Impact on outcome of stem cell transplantation. Blood 2017, 129, 2347–2358. [Google Scholar] [CrossRef]
  41. Shahzad, M.; Tariq, E.; Chaudhary, S.G.; Anwar, I.; Iqbal, Q.; Fatima, H.; Abdelhakim, H.; Ahmed, N.; Balusu, R.; Hematti, P.; et al. Outcomes with allogeneic hematopoietic stem cell transplantation in TP53-mutated acute myeloid leukemia: A systematic review and meta-analysis. Leuk. Lymphoma 2022, 63, 3409–3417. [Google Scholar] [CrossRef]
  42. Badar, T.; Atallah, E.; Shallis, R.; Saliba, A.N.; Patel, A.; Bewersdorf, J.P.; Grenet, J.; Stahl, M.; Duvall, A.; Burkart, M.; et al. Survival of TP53-mutated acute myeloid leukemia patients receiving allogeneic stem cell transplantation after first induction or salvage therapy: Results from the Consortium on Myeloid Malignancies and Neoplastic Diseases (COMMAND). Leukemia 2023, 37, 799–806. [Google Scholar] [CrossRef]
  43. Feng, D.; Wang, M.Y.; Liu, J.; Zhang, H.X.; Chen, X.; Zhang, R.L.; Zhai, W.H.; Ma, Q.L.; Pang, A.M.; Yang, D.L.; et al. Survival efficacy of MDS/AML patients with TP53 abnormal received allogeneic hematopoietic stem cell transplantation. Zhonghua Xue Ye Xue Za Zhi 2023, 44, 222–229. [Google Scholar] [CrossRef] [PubMed]
  44. Loke, J.; Labopin, M.; Craddock, C.; Cornelissen, J.J.; Labussière-Wallet, H.; Wagner-Drouet, E.M.; Van Gorkom, G.; Schaap, N.P.M.; Kröger, N.M.; Veelken, J.H.; et al. Additional cytogenetic features determine outcome in patients allografted for TP53 mutant acute myeloid leukemia. Cancer 2022, 128, 2922–2931. [Google Scholar] [CrossRef] [PubMed]
  45. Li, J.; Chen, Y.; Zhu, Y.; Zhou, J.; Xu, Y.; Li, Y.; Yu, K.; Pan, L.; Wang, J.; Ding, J.; et al. Efficacy and safety of decitabine in combination with G-CSF, low-dose cytarabine and aclarubicin in newly diagnosed elderly patients with acute myeloid leukemia. Oncotarget 2015, 6, 6448–6458. [Google Scholar] [CrossRef]
  46. Kadia, T.M.; Faderl, S.; Ravandi, F.; Jabbour, E.; Garcia-Manero, G.; Borthakur, G.; Ferrajoli, A.; Konopleva, M.; Burger, J.; Huang, X.; et al. Final results of a phase 2 trial of clofarabine and low-dose cytarabine alternating with decitabine in older patients with newly diagnosed acute myeloid leukemia. Cancer 2015, 121, 2375–2382. [Google Scholar] [CrossRef] [PubMed]
  47. Kadia, T.M.; Cortes, J.; Ravandi, F.; Jabbour, E.; Konopleva, M.; Benton, C.B.; Burger, J.; Sasaki, K.; Borthakur, G.; DiNardo, C.D.; et al. Cladribine and low-dose cytarabine alternating with decitabine as front-line therapy for elderly patients with acute myeloid leukaemia: A phase 2 single-arm trial. Lancet Haematol. 2018, 5, e411–e421. [Google Scholar] [CrossRef]
  48. Kadia, T.M.; Ravandi, F.; Borthakur, G.; Konopleva, M.; DiNardo, C.D.; Daver, N.; Pemmaraju, N.; Kanagal-Shamanna, R.; Wang, X.; Huang, X.; et al. Long-term results of low-intensity chemotherapy with clofarabine or cladribine combined with low-dose cytarabine alternating with decitabine in older patients with newly diagnosed acute myeloid leukemia. Am. J. Hematol. 2021, 96, 914–924. [Google Scholar] [CrossRef]
  49. Cahill, K.E.; Karimi, Y.H.; Karrison, T.G.; Jain, N.; Green, M.; Weiner, H.; Fulton, N.; Kadri, S.; Godley, L.A.; Artz, A.S.; et al. A phase 1 study of azacitidine with high-dose cytarabine and mitoxantrone in high-risk acute myeloid leukemia. Blood Adv. 2020, 4, 599–606. [Google Scholar] [CrossRef]
  50. Zhang, B.; Pei, Z.; Wang, H.; Wu, H.; Wang, J.; Bai, J.; Song, Q. Chidamide and Decitabine in Combination with a HAG Priming Regimen for Acute Myeloid Leukemia with TP53 Mutation. Acta Med. Okayama 2022, 76, 63–70. [Google Scholar] [CrossRef]
  51. Chiche, E.; Rahmé, R.; Bertoli, S.; Dumas, P.Y.; Micol, J.B.; Hicheri, Y.; Pasquier, F.; Peterlin, P.; Chevallier, P.; Thomas, X.; et al. Real-life experience with CPX-351 and impact on the outcome of high-risk AML patients: A multicentric French cohort. Blood Adv. 2021, 5, 176–184. [Google Scholar] [CrossRef] [PubMed]
  52. Goldberg, A.D.; Talati, C.; Desai, P.; Famulare, M.; Delvin, S.M.; Farnoud, N.; Sallman, D.A.; Lancet, J.E.; Roboz, G.J.; Sweet, K.L.; et al. TP53 Mutations Predict Poorer Responses to CPX-351 in Acute Myeloid Leukemia. Blood 2018, 132 (Suppl. S1), 1433. [Google Scholar] [CrossRef]
  53. Cortes, J.E.; Lin, T.L.; Asubonteng, K.; Faderl, S.; Lancet, J.E.; Prebet, T. Efficacy and safety of CPX-351 versus 7 + 3 chemotherapy by European LeukemiaNet 2017 risk subgroups in older adults with newly diagnosed, high-risk/secondary AML: Post hoc analysis of a randomized, phase 3 trial. J. Hematol. Oncol. 2022, 15, 155. [Google Scholar] [CrossRef] [PubMed]
  54. Rautenberg, C.; Stölzel, F.; Röllig, C.; Stelljes, M.; Gaidzik, V.; Lauseker, M.; Kriege, O.; Verbeek, M.; Unglaub, J.M.; Thol, F.; et al. Real-world experience of CPX-351 as first-line treatment for patients with acute myeloid leukemia. Blood Cancer J. 2021, 11, 164. [Google Scholar] [CrossRef] [PubMed]
  55. Swords, R.T.; Coutre, S.; Maris, M.B.; Zeidner, J.F.; Foran, J.M.; Cruz, J.; Erba, H.P.; Berdeja, J.G.; Tam, W.; Vardhanabhuti, S.; et al. Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, combined with azacitidine in patients with AML. Blood 2018, 131, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
  56. Saliba, A.N.; Kaufmann, S.H.; Stein, E.M.; Patel, P.A.; Baer, M.R.; Stock, W.; Deininger, M.; Blum, W.; Schiller, G.J.; Olin, R.L.; et al. Pevonedistat with azacitidine in older patients with TP53-mutated AML: A phase 2 study with laboratory correlates. Blood Adv. 2023, 7, 2360–2363. [Google Scholar] [CrossRef]
  57. Short, N.J.; Muftuoglu, M.; Ong, F.; Nasr, L.; Macaron, W.; Montalban-Bravo, G.; Alvarado, Y.; Basyal, M.; Daver, N.; Dinardo, C.D.; et al. A phase 1/2 study of azacitidine, venetoclax and pevonedistat in newly diagnosed secondary AML and in MDS or CMML after failure of hypomethylating agents. J. Hematol. Oncol. 2023, 16, 73. [Google Scholar] [CrossRef]
  58. Rushworth, S.A.; Murray, M.Y.; Zaitseva, L.; Bowles, K.M.; MacEwan, D.J. Identification of Bruton’s tyrosine kinase as a therapeutic target in acute myeloid leukemia. Blood 2014, 123, 1229–1238. [Google Scholar] [CrossRef]
  59. Huls, G.; Chitu, D.A.; Pabst, T.; Klein, S.K.; Stussi, G.; Griskevicius, L.; Valk, P.J.M.; Cloos, J.; van de Loosdrecht, A.A.; Breems, D.; et al. Ibrutinib added to 10-day decitabine for older patients with AML and higher risk MDS. Blood Adv. 2020, 4, 4267–4277. [Google Scholar] [CrossRef]
  60. Roboz, G.J.; Mandrekar, S.J.; Desai, P.; Laumann, K.; Walker, A.R.; Wang, E.S.; Kolitz, J.E.; Powell, B.L.; Attar, E.C.; Stock, W.; et al. Randomized trial of 10 days of decitabine ± bortezomib in untreated older patients with AML: CALGB 11002 (Alliance). Blood Adv. 2018, 2, 3608–3617. [Google Scholar] [CrossRef]
  61. Zhang, Q.; Bykov, V.J.N.; Wiman, K.G.; Zawacka-Pankau, J. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis. 2018, 9, 439, Correction in Cell Death Dis. 2019, 10, 769. [Google Scholar] [CrossRef]
  62. Ali, D.; Jönsson-Videsäter, K.; Deneberg, S.; Bengtzén, S.; Nahi, H.; Paul, C.; Lehmann, S. APR-246 exhibits anti-leukemic activity and synergism with conventional chemotherapeutic drugs in acute myeloid leukemia cells. Eur. J. Haematol. 2011, 86, 206–215. [Google Scholar] [CrossRef]
  63. Maslah, N.; Salomao, N.; Drevon, L.; Verger, E.; Partouche, N.; Ly, P.; Aubin, P.; Naoui, N.; Schlageter, M.H.; Bally, C.; et al. Synergistic effects of PRIMA-1Met (APR-246) and 5-azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia. Haematologica 2020, 105, 1539–1551. [Google Scholar] [CrossRef]
  64. Fujihara, K.M.; Zhang, B.Z.; Jackson, T.D.; Ogunkola, M.O.; Nijagal, B.; Milne, J.V.; Sallman, D.A.; Ang, C.S.; Nikolic, I.; Kearney, C.J.; et al. Eprenetapopt triggers ferroptosis, inhibits NFS1 cysteine desulfurase, and synergizes with serine and glycine dietary restriction. Sci. Adv. 2022, 8, eabm9427. [Google Scholar] [CrossRef]
  65. Birsen, R.; Larrue, C.; Decroocq, J.; Johnson, N.; Guiraud, N.; Gotanegre, M.; Cantero-Aguilar, L.; Grignano, E.; Huynh, T.; Fontenay, M.; et al. APR-246 induces early cell death by ferroptosis in acute myeloid leukemia. Haematologica 2022, 107, 403–416. [Google Scholar] [CrossRef] [PubMed]
  66. Sallman, D.A.; DeZern, A.E.; Garcia-Manero, G.; Steensma, D.P.; Roboz, G.J.; Sekeres, M.A.; Cluzeau, T.; Sweet, K.L.; McLemore, A.; McGraw, K.L.; et al. Eprenetapopt (APR-246) and Azacitidine in TP53-Mutant Myelodysplastic Syndromes. J. Clin. Oncol. 2021, 39, 1584–1594. [Google Scholar] [CrossRef] [PubMed]
  67. 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] [PubMed]
  68. Mishra, A.; Tamari, R.; DeZern, A.E.; Byrne, M.T.; Gooptu, M.; Chen, Y.B.; Deeg, H.J.; Sallman, D.; Gallacher, P.; Wennborg, A.; et al. Eprenetapopt Plus Azacitidine After Allogeneic Hematopoietic Stem-Cell Transplantation for TP53-Mutant Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2022, 40, 3985–3993. [Google Scholar] [CrossRef] [PubMed]
  69. Garcia-Manero, G.; Goldberg, A.D.; Winer, E.S.; Altman, J.K.; Fathi, A.T.; Odenike, O.; Roboz, G.J.; Sweet, K.; Miller, C.; Wennborg, A.; et al. Eprenetapopt combined with venetoclax and azacitidine in TP53-mutated acute myeloid leukaemia: A phase 1, dose-finding and expansion study. Lancet Haematol. 2023, 10, e272–e283. [Google Scholar] [CrossRef] [PubMed]
  70. 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] [PubMed]
  71. Majeti, R.; Chao, M.P.; Alizadeh, A.A.; Pang, W.W.; Jaiswal, S.; Gibbs, K.D., Jr.; 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]
  72. 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]
  73. Daver, N.; Vyas, P.; Kambhampati, S.; Al Malki, M.M.; Larson, R.A.; Asch, A.S.; Mannis, G.; Chai-Ho, W.; Tanaka, T.N.; Bradley, T.J.; et al. Tolerability and Efficacy of the Anticluster of Differentiation 47 Antibody Magrolimab Combined with Azacitidine in Patients with Previously Untreated AML: Phase Ib Results. J. Clin. Oncol. 2023, 41, 4893–4904. [Google Scholar] [CrossRef]
  74. Daver, N.; Senapati, J.; Maiti, A.; Longhavi, S.; Kadia, T.M.; DiNardo, C.D.; Pemmaraju, N.; Jabbour, E.; Montalban-Bravo, G.; Tang, G.; et al. Phase I/II Study of Azacitidine (AZA) with Venetoclax (VEN) and Magrolimab (Magro) in Patients (pts) with Newly Diagnosed (ND) Older/Unfit or High-Risk Acute Myeloid Leukemia (AML) and Relapsed/Refractory (R/R) AML. Blood 2022, 140 (Suppl. S1), 141–144. [Google Scholar] [CrossRef]
  75. Gilead Statement on the Discontinuation of Magrolimab Study in AML with TP53 Mutations. News release. 26 September 2023. Available online: https://www.gilead.com/news-and-press/company-statements/gilead-statement-on-the-discontinuation-of-magrolimab-study-in-aml-with-tp53-mutations (accessed on 4 December 2023).
  76. Petrova, P.S.; Viller, N.N.; Wong, M.; Pang, X.; Lin, G.H.; Dodge, K.; Chai, V.; Chen, H.; Lee, V.; House, V.; et al. TTI-621 (SIRPαFc): A CD47-Blocking Innate Immune Checkpoint Inhibitor with Broad Antitumor Activity and Minimal Erythrocyte Binding. Clin. Cancer Res. 2017, 23, 1068–1079. [Google Scholar] [CrossRef] [PubMed]
  77. Miao, M.; Qingliang, T.; Depei, W.; Zhongxing, J.; Songfu, J.; Fei, L.; Yang, L.; Zhongmin, M.W.; Baiyong, L.; Yu, X.; et al. AK117 (anti-CD47 monoclonal antibody) in Combination with Azacitidine for Newly Diagnosed Higher Risk Myelodysplastic Syndrome (HR-MDS): AK117-103 Phase 1b Results. Blood 2023, 142 (Suppl. S1), 1865. [Google Scholar] [CrossRef]
  78. Chen, A.; Harrabi, O.; Fong, A.P.; Ruffner, K.L.; Forgie, A.J.; Sim, J.; Randolph, S.S.; Wan, H.I.; Pons, J.; Kuo, T.C. Alx148 enhances the depth and durability of response to multiple aml therapies. Blood 2020, 136, 15–16. [Google Scholar] [CrossRef]
  79. ALX Oncology Reports Second Quarter 2023 Financial Results and Provides Clinical Program Update. 10 August 2023. Available online: https://ir.alxoncology.com/news-releases/news-release-details/alx-oncology-reports-second-quarter-2023-financial-results-and/ (accessed on 5 December 2023).
  80. Asayama, T.; Tamura, H.; Ishibashi, M.; Kuribayashi-Hamada, Y.; Onodera-Kondo, A.; Okuyama, N.; Yamada, A.; Shimizu, M.; Moriya, K.; Takahashi, H.; et al. Functional expression of Tim-3 on blasts and clinical impact of its ligand galectin-9 in myelodysplastic syndromes. Oncotarget 2017, 8, 88904–88917. [Google Scholar] [CrossRef] [PubMed]
  81. Kikushige, Y.; Miyamoto, T.; Yuda, J.; Jabbarzadeh-Tabrizi, S.; Shima, T.; Takayanagi, S.; Niiro, H.; Yurino, A.; Miyawaki, K.; Takenaka, K.; et al. A TIM-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression. Cell Stem Cell. 2015, 17, 341–352. [Google Scholar] [CrossRef] [PubMed]
  82. Brunner, A.M.; Esteve, J.; Porkka, K.; Knapper, S.; Vey, N.; Scholl, S.; Garcia-Manero, G.; Wermke, M.; Janssen, J.; Traer, E.; et al. Efficacy and safety of sabatolimab (MBG453) in combination with hypomethylating agents (HMAs) in patients (Pts) with very high/high-risk myelodysplastic syndrome (vHR/HR-MDS) and acute myeloid leukemia (AML): Final analysis from a phase Ib study. Blood 2021, 138, 244. [Google Scholar] [CrossRef]
  83. Al-Hussaini, M.; Rettig, M.P.; Ritchey, J.K.; Karpova, D.; Uy, G.L.; Eissenberg, L.G.; Gao, F.; Eades, W.C.; Bonvini, E.; Chichili, G.R.; et al. Targeting CD123 in acute myeloid leukemia using a T-cell-directed dual-affinity retargeting platform. Blood 2016, 127, 122–131. [Google Scholar] [CrossRef] [PubMed]
  84. Testa, U.; Riccioni, R.; Militi, S.; Coccia, E.; Stellacci, E.; Samoggia, P.; Latagliata, R.; Mariani, G.; Rossini, A.; Battistini, A.; et al. Elevated expression of IL-3Ralpha in acute myelogenous leukemia is associated with enhanced blast proliferation, increased cellularity, and poor prognosis. Blood 2002, 100, 2980–2988. [Google Scholar] [CrossRef] [PubMed]
  85. Uy, G.L.; Aldoss, I.; Foster, M.C.; Sayre, P.H.; Wieduwilt, M.J.; Advani, A.S.; Godwin, J.E.; Arellano, M.L.; Sweet, K.L.; Emadi, A.; et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood 2021, 137, 751–762. [Google Scholar] [CrossRef] [PubMed]
  86. Vadakekolathu, J.; Lai, C.; Reeder, S.; Church, S.E.; Hood, T.; Lourdusamy, A.; Rettig, M.P.; Aldoss, I.; Advani, A.S.; Godwin, J.; et al. TP53 abnormalities correlate with immune infiltration and associate with response to flotetuzumab immunotherapy in AML. Blood Adv. 2020, 4, 5011–5024. [Google Scholar] [CrossRef] [PubMed]
  87. Daver, N.; Montesinos, P.; Aribi, A.; Marconi, G.; Altman, J.K.; Wang, E.S.; Roboz, G.J.; Burke, P.W.; Gaidano, G.; Walter, R.B.; et al. Broad Activity for the Pivekimab Sunirine (PVEK, IMGN632), Azacitidine, and Venetoclax Triplet in High-Risk Patients with Relapsed/Refractory Acute Myeloid Leukemia (AML). Blood 2022, 140 (Suppl. S1), 145–149. [Google Scholar] [CrossRef]
  88. Daver, N.; Montesinos, P.; Aribi, A.; Martinelli, G.; Wang, E.S.; Altman, J.K.; Roboz, G.J.; Burke, P.W.; Walter, R.B.; Begna, K.; et al. A phase 1b/2 study of pivekimab sunirine (PVEK, IMGN632) in combination with venetoclax/azacitidine or magrolimab for patients with CD123-positive acute myeloid leukemia (AML). J. Clin. Oncol. 2023, 41 (Suppl. S16), TPS7073. [Google Scholar] [CrossRef]
  89. Daver, N.; Montesinos, P.; Altman, J.; Wang, E.S.; Martinelli, G.; Roboz, G.J.; Begna, K.; Vyas, P.; Lunghi, M.; Platzbecker, U.; et al. Pivekimab Sunirine (PVEK, IMGN632), a CD123-Targeting Antibody-Drug Conjugate, in Combination with Azacitidine and Venetoclax in Patients with Newly Diagnosed Acute Myeloid Leukemia. Blood 2023, 142 (Suppl. S1), 2906. [Google Scholar] [CrossRef]
  90. Frankel, A.; Liu, J.S.; Rizzieri, D.; Hogge, D. Phase I clinical study of diphtheria toxin-interleukin 3 fusion protein in patients with acute myeloid leukemia and myelodysplasia. Leuk. Lymphoma 2008, 49, 543–553. [Google Scholar] [CrossRef]
  91. Togami, K.; Pastika, T.; Stephansky, J.; Ghandi, M.; Christie, A.L.; Jones, K.L.; Johnson, C.A.; Lindsay, R.W.; Brooks, C.L.; Letai, A.; et al. DNA methyltransferase inhibition overcomes diphthamide pathway deficiencies underlying CD123-targeted treatment resistance. J. Clin. Investig. 2019, 129, 5005–5019. [Google Scholar] [CrossRef]
  92. Lane, A.; Garcia, J.; Raulston, E.; Garzon, J.L.; Galinsky, I.; Baxter, E.W.; Leonard, R.; DeAngelo, D.J.; Luskin, M.R.; Reilly, C.R.; et al. Tagraxofusp in Combination with Azacitidine and Venetoclax in Newly Diagnosed CD123+ Acute Myeloid Leukemia, Expansion Cohort of a Phase 1b Multicenter Trial. Blood 2023, 142 (Suppl. S1), 4277. [Google Scholar] [CrossRef]
  93. Ravandi, F.; Bashey, A.; Foran, J.; Stock, W.; Mawad, R.; Short, N.; Yilmaz, M.; Kantarjian, H.; Odenike, O.; Patel, A.; et al. Phase 1 study of vibecotamab identifies an optimized dose for treatment of relapsed/refractory acute myeloid leukemia. Blood Adv. 2023, 7, 6492–6505. [Google Scholar] [CrossRef] [PubMed]
  94. Pelosi, E.; Castelli, G.; Testa, U. CD123 a Therapeutic Target for Acute Myeloid Leukemia and Blastic Plasmocytoid Dendritic Neoplasm. Int. J. Mol. Sci. 2023, 24, 2718. [Google Scholar] [CrossRef] [PubMed]
  95. Davids, M.S.; Kim, H.T.; Bachireddy, P.; Costello, C.; Liguori, R.; Savell, A.; Lukez, A.P.; Avigan, D.; Chen, Y.B.; McSweeney, P.; et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. New Engl. J. Med. 2016, 375, 143–153. [Google Scholar] [CrossRef] [PubMed]
  96. Ravandi, F.; Assi, R.; Daver, N.; Benton, C.B.; Kadia, T.; Thompson, P.A.; Borthakur, G.; Alvarado, Y.; Jabbour, E.J.; Konopleva, M.; et al. Idarubicin, cytarabine, and nivolumab in patients with newly diagnosed acute myeloid leukaemia or high-risk myelodysplastic syndrome: A single-arm, phase 2 study. Lancet Haematol. 2019, 6, e480–e488. [Google Scholar] [CrossRef]
  97. 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] [PubMed]
  98. Zeidner, J.F.; Vincent, B.G.; Ivanova, A.; Moore, D.; McKinnon, K.P.; Wilkinson, A.D.; Mukhopadhyay, R.; Mazziotta, F.; Knaus, H.A.; Foster, M.C.; et al. Phase II Trial of Pembrolizumab after High-Dose Cytarabine in Relapsed/Refractory Acute Myeloid Leukemia. Blood Cancer Discov. 2021, 2, 616–629. [Google Scholar] [CrossRef]
  99. Zeidan, A.M.; Boss, I.; Beach, C.L.; Copeland, W.B.; Thompson, E.; Fox, B.A.; Hasle, V.E.; Hellmann, A.; Taussig, D.C.; Tormo, M.; et al. A randomized phase 2 trial of azacitidine with or without durvalumab as first-line therapy for older patients with AML. Blood Adv. 2022, 6, 2219–2229. [Google Scholar] [CrossRef]
  100. Lovewell, R.R.; Hong, J.; Kundu, S.; Fielder, C.M.; Hu, Q.; Kim, K.W.; Ramsey, H.E.; Gorska, A.E.; Fuller, L.S.; Tian, L.; et al. LAIR-1 agonism as a therapy for acute myeloid leukemia. J. Clin. Investig. 2023, 133, e169519. [Google Scholar] [CrossRef]
  101. Tovar, C.; Rosinski, J.; Filipovic, Z.; Higgins, B.; Kolinsky, K.; Hilton, H.; Zhao, X.; Vu, B.T.; Qing, W.; Packman, K.; et al. Small-molecule MDM2 antagonists reveal aberrant p53 signaling in cancer: Implications for therapy. Proc. Natl. Acad. Sci. USA 2006, 103, 1888–1893. [Google Scholar] [CrossRef]
  102. Yee, K.; Papayannidis, C.; Vey, N.; Dickinson, M.J.; Kelly, K.R.; Assouline, S.; Kasner, M.; Seiter, K.; Drummond, M.W.; Yoon, S.S.; et al. Murine double minute 2 inhibition alone or with cytarabine in acute myeloid leukemia: Results from an idasanutlin phase 1/1b study small star, filled. Leuk. Res. 2021, 100, 106489. [Google Scholar] [CrossRef]
  103. Konopleva, M.Y.; Röllig, C.; Cavenagh, J.; Deeren, D.; Girshova, L.; Krauter, J.; Martinelli, G.; Montesinos, P.; Schäfer, J.A.; Ottmann, O.; et al. Idasanutlin plus cytarabine in relapsed or refractory acute myeloid leukemia: Results of the MIRROS trial. Blood Adv. 2022, 6, 4147–4156. [Google Scholar] [CrossRef]
  104. Pan, R.; Ruvolo, V.; Mu, H.; Leverson, J.D.; Nichols, G.; Reed, J.C.; Konopleva, M.; Andreeff, M. Synthetic Lethality of Combined Bcl-2 Inhibition and p53 Activation in AML: Mechanisms and Superior Antileukemic Efficacy. Cancer Cell. 2017, 32, 748–760.e6. [Google Scholar] [CrossRef]
  105. Senapati, J.; Muftuoglu, M.; Ishizawa, J.; Abbas, H.A.; Loghavi, S.; Borthakur, G.; Yilmaz, M.; Issa, G.C.; Dara, S.I.; Basyal, M.; et al. A Phase I study of Milademetan (DS3032b) in combination with low dose cytarabine with or without venetoclax in acute myeloid leukemia: Clinical safety, efficacy, and correlative analysis. Blood Cancer J. 2023, 13, 101. [Google Scholar] [CrossRef]
  106. Daver, N.; Dail, M.; Garcia, J.S.; Jonas, B.A.; Yee, K.W.L.; Kelly, K.R.; Vey, N.; Assouline, S.; Roboz, G.J.; Paolini, S.; et al. Venetoclax and idasanutlin in relapsed/refractory AML: A nonrandomized, open-label phase 1b trial. Blood 2023, 141, 1265–1276. [Google Scholar] [CrossRef]
  107. Chen, S.; Wu, J.L.; Liang, Y.; Tang, Y.G.; Song, H.X.; Wu, L.L.; Xing, Y.F.; Yan, N.; Li, Y.T.; Wang, Z.Y.; et al. Arsenic Trioxide Rescues Structural p53 Mutations through a Cryptic Allosteric Site. Cancer Cell. 2021, 39, 225–239.e8. [Google Scholar] [CrossRef] [PubMed]
  108. Parrales, A.; Ranjan, A.; Iyer, S.V.; Padhye, S.; Weir, S.J.; Roy, A.; Iwakuma, T. DNAJA1 controls the fate of misfolded mutant p53 through the mevalonate pathway. Nat. Cell Biol. 2016, 18, 1233–1243. [Google Scholar] [CrossRef] [PubMed]
  109. Eisenmann, E.D.; Stromatt, J.C.; Fobare, S.; Huang, K.M.; Buelow, D.R.; Orwick, S.; Jeon, J.Y.; Weber, R.H.; Larsen, B.; Mims, A.S.; et al. TP-0903 Is Active in Preclinical Models of Acute Myeloid Leukemia with TP53 Mutation/Deletion. Cancers 2022, 15, 29. [Google Scholar] [CrossRef] [PubMed]
  110. Semba, Y.; Yamauchi, T.; Nakao, F.; Nogami, J.; Canver, M.C.; Pinello, L.; Bauer, D.E.; Akashi, K.; Maeda, T. CRISPR-Cas9 Screen Identifies XPO7 As a Potential Therapeutic Target for TP53-Mutated AML. Blood 2019, 134 (Suppl. S1), 3784. [Google Scholar] [CrossRef]
  111. Kravchenko, J.E.; Ilyinskaya, G.V.; Komarov, P.G.; Agapova, L.S.; Kochetkov, D.V.; Strom, E.; Frolova, E.I.; Kovriga, I.; Gudkov, A.V.; Feinstein, E.; et al. Small-molecule RETRA suppresses mutant p53-bearing cancer cells through a p73-dependent salvage pathway. Proc. Natl. Acad. Sci. USA 2008, 105, 6302–6307. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, Y.; Chen, C.; Xu, Z.; Scuoppo, C.; Rillahan, C.D.; Gao, J.; Spitzer, B.; Bosbach, B.; Kastenhuber, E.R.; Baslan, T.; et al. Deletions linked to TP53 loss drive cancer through p53-independent mechanisms. Nature 2016, 531, 471–475. [Google Scholar] [CrossRef] [PubMed]
  113. Boeckler, F.M.; Joerger, A.C.; Jaggi, G.; Rutherford, T.J.; Veprintsev, D.B.; Fersht, A.R. Targeted rescue of a destabilized mutant of p53 by an in silico screened drug. Proc. Natl. Acad. Sci. USA 2008, 105, 10360–10365. [Google Scholar] [CrossRef] [PubMed]
  114. Blanden, A.R.; Yu, X.; Loh, S.N.; Levine, A.J.; Carpizo, D.R. Reactivating mutant p53 using small molecules as zinc metallochaperones: Awakening a sleeping giant in cancer. Drug Discov. Today 2015, 20, 1391–1397, Correction in Drug Discov. Today 2016, 10, 1728. [Google Scholar] [CrossRef] [PubMed]
  115. Gao, N.; Budhraja, A.; Cheng, S.; Liu, E.H.; Chen, J.; Yang, Z.; Chen, D.; Zhang, Z.; Shi, X. Phenethyl isothiocyanate exhibits antileukemic activity in vitro and in vivo by inactivation of Akt and activation of JNK pathways. Cell Death Dis. 2011, 2, e140. [Google Scholar] [CrossRef]
  116. Carter, B.Z.; Mak, P.Y.; Muftuoglu, M.; Tao, W.; Ke, B.; Pei, J.; Bedoy, A.D.; Ostermann, L.B.; Nishida, Y.; Isgandarova, S.; et al. Epichaperome inhibition targets TP53-mutant AML and AML stem/progenitor cells. Blood 2023, 142, 1056–1070. [Google Scholar] [CrossRef] [PubMed]
  117. Lindemann, A.; Patel, A.A.; Silver, N.L.; Tang, L.; Liu, Z.; Wang, L.; Tanaka, N.; Rao, X.; Takahashi, H.; Maduka, N.K.; et al. COTI-2, A Novel Thiosemicarbazone Derivative, Exhibits Antitumor Activity in HNSCC through p53-dependent and -independent Mechanisms. Clin. Cancer Res. 2019, 25, 5650–5662. [Google Scholar] [CrossRef]
  118. Bosc, C.; Saland, E.; Bousard, A.; Gadaud, N.; Sabatier, M.; Cognet, G.; Farge, T.; Boet, E.; Gotanègre, M.; Aroua, N.; et al. Mitochondrial inhibitors circumvent adaptive resistance to venetoclax and cytarabine combination therapy in acute myeloid leukemia. Nat. Cancer 2021, 2, 1204–1223. [Google Scholar] [CrossRef]
Table 1. Available current data from studies of novel agents in TP53-mutated acute myeloid leukemia.
Table 1. Available current data from studies of novel agents in TP53-mutated acute myeloid leukemia.
AgentStudy TypeRegimenPopulationTP53mut Patients (n)ResponseOS (Months)Refs
PevonedistatOpen-label, phase 1B, multicenterPEVO + AZAUnfit, untreated AML patients8CR/CRi/PR 75%NR[55]
Open-label, phase 2, multicenterPEVO + AZA≥60 y, untreated, TP53mut AML patients10CR/CRi 0%mOS 6.2 m[56]
Phase 1/2 single-centerPEVO + AZA + VENUnfit ND secondary AML patients, MDS and CMML patients after failure of HMAs11CR/CRi 64%mOS 8.1 m[57]
IbrutinibRandomized, phase 2, multicenterIbrutinib + DEC10 vs. DEC10 monotherapyElderly, unfit, untreated AML patients27CR/CRi 56% in both armsInferior OS compared to TP53wt patients[59]
BortezomibRandomized, phase 2, multicenterBortezomib + DEC10 vs. DEC10 monotherapyElderly, ND AML patients12 in combination arm and 14 in DEC10 armCR 17% in combination arm vs. 21% in DEC10 arm1-year OS 17% in combination vs. 21% in DEC10 arm[60]
Eprenetapopt (APR-246)Open-label, phase 1b/2, multicenterAPR-246 + AZA≥18 y, TP53mut, HMA-naïve MDS, MDS/MPN, CMML, oligoblastic (20–30% blasts) AML patients11ORR 64%
CR 36%
mOS 10.8 m[66]
Open-label, phase 2, multicenterAPR-246 + AZA≥18 y, TP53mut, HMA-naïve MDS, CMML, oligoblastic and >30% blasts AML patients18ORR 33%
CR 17%
CR 27% in oligoblastic AML
CR 0% in AML with >30% blasts
mOS 10.4 m
mOS 13.4 m in oligoblastic AML
mOS 3 m in AML with >30% blasts
[67]
Open-label, phase 2, multicenterAPR-246 + AZA as maintenance treatment after HCT≥18 y, TP53mut, MDS or AML patients post-HCT14NAmRFS 12.5 m
mOS 20.6 m
(for all patients)
[68]
Open-label, phase 1, multicenterAPR-246 + AZA + VEN≥18 y, TP53mut, untreated AML patients43ORR 64%
CR 38%
CR/CRi 56%
mOS 7.3 m[69]
MagrolimabOpen-label, phase 1b, multicenterMAG + AZA≥18 y, unfit, untreated AML patients72ORR 47%
CR 32%
mOS 9.8 m[73]
Open-label, phase 1b/2, multicenterMAG + AZA + VEN≥18 y, unfit, ND or untreated secondary and VEN-naïve or VEN-exposed R/R AML patients27
in the ND and untreated secondary AML cohort
ORR 74%
CR 86%
CR/CRi 63%
1-year OS 53%[74]
Sabatolimab
(MBG453)
Open-label, phase 1b, multicenterSAB + HMAUnfit, ND or R/R HMA-naïve AML, high risk HMA-naïve MDS and CMML patientsNRORR 53.8% in ND AML patients with at least 1 ELN adverse-risk mutation, including TP53NR[82]
FlotetuzumabOpen-label, phase 1/2, multicenterFLOT monotherapyR/R AML/MDS patients15 in the R/R AML cohortORR 60%
CR 47%
mOS 10.3 m [86]
Pivekimab sunirine
(IMGN632)
Open-label, phase 1b/2, multicenterPVEK + AZA + VENND, CD123+ AML patients19CR 13%
coCR 47%
NR[89]
TagraxofuspOpen-label, phase 1b, multicenter TAG + AZA +/− VENUnfit, ND, CD123+ AML patients13CR 31%
CR/CRi/MLFS 54%
mOS 9.5 m
mPFS 5.1 m
[92]
NivolumabOpen-label, phase 1/2, single-centerNIVO + idarubicin + AraCFit for IC, ND AML patients8CR/CRi/CRp 50% for TP53mut patients
CR 67% for patients with poor-risk mutation profile, TP53 included
NR[96]
Open-label, phase 2, single-centerNIVO + AZAR/R AML patients16ORR 28%NR[97]
PembrolizumabOpen-label, phase 2, two-centerPEMBRO + HiDACR/R AML patients5CR 40%NR[98]
DurvalumabRandomized, open-label, phase 2, multicenterDURV + AZA vs. AZA monotherapyElderly, unfit, ND AML patients21 in the combination arm, 17 in the monotherapy arm ORR 34% in TP53mut AML vs. ORR 33% in TP53wt AML for both treatment arms NR[99]
IdasanutlinOpen-label, phase 1, multicenterIDASA + VENUnfit, ND sAML or R/R AML patients10CR/CRi/CRp 20%mOS 3.7 m [106]
PEV: pevonedistat, AZA, azacitidine; AML, acute myeloid leukemia; CR, complete remission; CR, complete remission with incomplete count recovery; PR, partial remission; NR, not reported; mOS, median overall survival; VEN, venetoclax; ND, newly diagnosed; MDS, myelodysplastic syndrome; CMML, chronic myelomonocytic leukemia; HMA, hypomethylating agent; DEC10, 10 days decitabine treatment; MDS/MPN, myelodysplastic syndrome/myeloproliferative neoplasm; ORR, overall response rate; mRFS, median relapse free survival; HCT, hematopoietic cell transplant; NA, not applicable; MAG, Magrolimab; R/R, relapsed/refractory; SAB, sabatolimab; ELN, European Leukemia Net; FLOT, flotetuzumab; PVEK, pivekimab sunirine; coCR, composite complete remission; TAG, tagraxofusp; MLFS, morphologic leukemia-free state; PFS, progression free survival; NIVO, nivolumab; AraC, cytarabine; IC, intensive chemotherapy; CRp, complete remission with incomplete platelet recovery; PEMBRO, pembrolizumab; HiDAC, high dose cytarabine; DURV, durvalumab; IDASA, idasanutlin; sAML, secondary acute myeloid leukemia.
Table 2. Ongoing clinical trials of interest in patients with TP53-mutated acute myeloid leukemia.
Table 2. Ongoing clinical trials of interest in patients with TP53-mutated acute myeloid leukemia.
AgentTargetRegimenPatient PopulationPhaseIdentifier
MaplirpaceptCD47Maplirpacept + AZAND TP53mut AML1a/1bNCT03530683
SL-172154CD47SL-172154 + AZA +/− VENND or secondary AML and MDS (TP53mut included)1a/1bNCT05275439
Sabatolimab (MBG453)TIM-3SAB + AZA + VENND unfit for IC AML (TP53mut included)2NCT04150029
FlotetuzumabCD123FLOT monotherapyPost allo-HCT relapsed CD123+ AML (TP53mut included)1bNCT05506956
2NCT04582864
Pivekimab sunirine (IMGN632)CD123PVEK + FLAG-IdaND, adverse risk, CD123+ AML (TP53mut included)1NCT06034470
TagraxofuspCD123TAG + GOR/R AML (TP53mut included) 1NCT05716009
NivolumabPD-1NIVO + DEC + VENND TP53mut AML1NCT04277442
NC525LAIR-1NC525 monotherapyR/R AML, MDS, CMML (TP53mut included)1NCT05787496
Arsenic trioxidep53ATO + DECTP53mut AML/MDS1NCT03855371
Atorvastatinp53Atorvastatin monotherapyTP53mut AML and solid tumors1NCT03560882
MbIL-21 NK cellsAntitumor effectsMbIL-21 NK cells + DEC + fludarabineR/R AML (TP53mut patients included)1NCT04220684
ASTX727
(oral DEC and cedazirudine)
ASTX727 + entrectinibR/R TP53mut AML1NCT05396859
AZA, azacitidine; ND, newly diagnosed; AML, acute myeloid leukemia; NCT, national clinical trial, VEN, venetoclax; MDS, myelodysplastic syndrome; SAB, sabatolimab; FLOT, flotetuzumab; IC, intensive chemotherapy; allo-HCT, allogeneic-hematopoietic cell transplantation; PVEK, pivekimab sunirine; FLAG-Ida, fludarabine, cytarabine, idarubicin and granulocyte colony-stimulating factor; GO, gemtuzumab ozogamicin; R/R, relapsed/refractory; NIVO, nivolumab; DEC, decitabine; LAIR-1, leukocyte associated immunoglobulin like receptor 1; CMML, chronic myelomonocytic leukemia; ATO, arsenic trioxide; mbIL-21 NK cells, membrane-bound interleukin 21 natural killer cells.
Table 3. TP53/p53 targeting in preclinical studies.
Table 3. TP53/p53 targeting in preclinical studies.
CompoundTargetModelMechanism of ActionCombination with Other TherapyRef
Compounds that restore p53 wildtype function
PK7088Y220CCell linesSelective induction of caspase 3/7 in p53-Y220C cells and restoration of p53wt conformationNA
PhiKan083Y220CIn silicoBAX nuclear export induction to the mitochondria, and restoration of p53 nontranscriptional apoptosisNA[113]
NSC319726 (ZMC1)R175H, R172HIn silicoZinc chelator, providing optimal zinc concentration for mut p53-R175H proper folding; induction of ROS formation
Restoration of p53wt conformation and activity with MDM2-dependent degradation
NA[114]
PEITCR175HCell linesSensitization of p53mut to proteasome-mediated degradation and further restoration of p53wt conformation and transactivationNA[115]
COTI-2R175H, R273HCell linesRestoration of p53wt activity by targeting and binding to misfolded p53 mutantNA[117]
Compounds that induce degradation of mutant p53
PU-H71 (Zelavespib)R248WMolm13 and K562 cells Induction of cell death in TP53wt, TP53-KO, and TP53mut cellsVEN enhanced the killing of both TP53wt and TP53mut cells by PU-H71[116]
Compounds with miscellaneous targets
TP-0903 (Dubermatinib)Multikinase inhibitorCell linesAURKA/B inhibition in TP53mut AML
G2/M arrest and apoptosis in TP53mut AML cells
Chk1/2 inhibition in TP53mut AML cells
DNA damage response through upregulation of pH2AX
Combination of TP-0903 and DEC is active in vitro demonstrating an additive effect
TP-0903/DEC prolongs survival in vivo in a HL-60 xenograft model
[109]
XPO7 Mouse cell linesTrp53-KO cells are vulnerable to XPO7 depletion, while XPO7 functions as a Trp53-dependent tumor suppressor in Trp53wt AML cells
Synthetic lethal relationship between TP53 and XPO7
NA[110]
RETRAmutp53-p73 bindingMouse cell linesIncrease in the expression level of p73, and release of p73 from the blocking complex with p53mut, which produces tumor-suppressor effects similar to the functional reactivation of p53.
RETRA is active against tumor cells expressing a variety of p53 mutants and does not affect normal cells.
NA[111]
p53, protein 53; mut, mutant; BAX, Bcl-2 associated X protein; TP53, tumor protein 53; KO, knockout; CHIP, carboxyl terminus of Hsc70 interacting protein; AML, acute myeloid leukemia; AURKA/B, aurora kinase A/B; CHK1/2, checkpoint kinase ½; pH2AX, phosphor-histone h2AX; HL-60; human leukemia cell line 60; XPO7, exportin 7; RETRA, reactivation of transcriptional reporter activity; p73, protein 73; VEN, venetoclax; DEC, decitabine; NA, not applicable.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stafylidis, C.; Vlachopoulou, D.; Kontandreopoulou, C.-N.; Diamantopoulos, P.Τ. Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia. J. Clin. Med. 2024, 13, 1082. https://doi.org/10.3390/jcm13041082

AMA Style

Stafylidis C, Vlachopoulou D, Kontandreopoulou C-N, Diamantopoulos PΤ. Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia. Journal of Clinical Medicine. 2024; 13(4):1082. https://doi.org/10.3390/jcm13041082

Chicago/Turabian Style

Stafylidis, Christos, Dimitra Vlachopoulou, Christina-Nefeli Kontandreopoulou, and Panagiotis Τ. Diamantopoulos. 2024. "Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia" Journal of Clinical Medicine 13, no. 4: 1082. https://doi.org/10.3390/jcm13041082

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