Next Article in Journal
Propyl Gallate Attenuates Methylglyoxal-Induced Alzheimer-like Cognitive Deficits and Neuroinflammation in Mice
Previous Article in Journal
Geraniin Mitigates Neuropathic Pain Through Antioxidant, Anti-Inflammatory, and Nitric Oxide Modulation in a Rat Model of Chronic Constriction Injury
Previous Article in Special Issue
NETosis and Neutrophil Activity Quantification in Pediatric Patients with Essential Thrombocythemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations

by
Sarvath Aafreen Sanaullah
1,
Pierre-Alexandre Vidi
1,2 and
Timothy S. Pardee
1,3,*
1
Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
2
Integrity of the Genome and Oncology (InGenO) Laboratory, Institut de Cancérologie de l’Ouest–ICO, 49055 Angers, France
3
Department of Cancer Medicine, Wake Forest University School of Medicine, Winston Salem, NC 27157, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2026, 27(1), 510; https://doi.org/10.3390/ijms27010510
Submission received: 14 November 2025 / Revised: 25 December 2025 / Accepted: 25 December 2025 / Published: 3 January 2026
(This article belongs to the Special Issue Molecular Research in Hematologic Malignancies)

Abstract

Racial and ethnic disparities in acute myeloid leukemia (AML) survival persist despite advances in treatment, with non-Hispanic black (NHB) patients and Hispanic patients often experiencing worse outcomes than Non-Hispanic White (NHW) patients due to a combination of clinical, socioeconomic, and biological factors. This review focuses on these disparities and emphasizes potential contributions of biology, as illustrated by the effects of the nucleophosmin 1 (NPM1) mutation. Mutation landscapes and chromosomal abnormalities strongly influence AML patient outcomes. While AML cases with NPM1 mutations are associated with favorable prognoses for NHW patients, NHB patients with NPM1-mutated AML have adverse outcomes. Thus, treatment algorithms and prognostic systems based on outcomes from a single racial ancestry group are inadequate. Beyond the more traditional socioeconomic determinants of health, addressing disparities in AML to achieve equity in care requires exploring biological factors linked to ancestry that shape treatment response.

1. Introduction

Acute myeloid leukemia (AML) is a heterogeneous disease characterized by a diverse set of genetic mutations and variable clinical outcomes [1]. The origin of AML involves the accumulation of mutations in hematopoietic stem or progenitor cells, leading to the uncontrolled proliferation of malignant myeloid cells. Risk stratification in AML is based on cytogenetic abnormalities and specific gene mutations [2]. Thus, next-generation sequencing and karyotyping is an essential component of AML diagnostic work up [2]. The prognostic value of these genetic changes is derived from clinical trials data examined retrospectively. The standard of care for AML often involves induction chemotherapy, with or without specific targeted treatments, with the goal of achieving a remission. Patients in remission then undergo consolidation therapy, including either repeated cycles of chemotherapy or allogeneic hematopoietic stem cell transplant (HSCT). Importantly, the decision to recommend HSCT in consolidation is based largely on the cytogenetic and molecular risk category of the patient. The incidence of AML varies across different racial groups, with higher incidence rates seen among White individuals compared to Black and Hispanic populations in the United States. Despite the survival advantage seen in certain genetic subtypes, Black and Hispanic patients experience significantly lower overall survival compared to White patients [3,4,5]. This review will examine the data on traditional factors of racial disparities, as well as possible biological differences by racial ancestry.

2. External Factors Contributing to AML Disparities

2.1. Clinical Trials Enrollment

Clinical trials are critical for the validation of new therapies for the treatment of cancer. They also provide data on which prognostic schemas (like the European Leukemia Net (ELN) 2022 risk classification system) are built, with the underlying assumption that outcomes in trial participants will be applicable to all AML patients. Currently, less than five percent of all adult cancer patients enroll in cancer clinical trials [6]. Clinical trial enrollment is impacted by several factors, such as limited awareness, insurance-related issues, distrust toward healthcare providers, especially in minority racial groups, socioeconomic barriers, and environment [7]. Several studies and meta-analyses of clinical trials in the past decade have shown that only 21 out of 90 AML clinical trials reported race or ethnicity of the participants. When race was reported, less than 5% were non-Hispanic black (NHB), and more than 80% of the enrolled participants were non-Hispanic white (NHW) [8,9,10,11,12,13,14]. Statler et al. (2019) investigated clinical trial enrollment at the Cleveland Clinic and found no difference in AML etiology or risk classification between NHB and NHW patients [15]. However, NHB patients were underrepresented in trials [15]. Similarly, another study noted that disparities in clinical trial enrollment, transplant access, and supportive care use contributed to racial differences in outcomes across hematologic malignancies [16]. Because treatment algorithms rely on clinical trial data, current AML care recommendations are largely shaped by outcomes from NHW patients. Worse survival outcomes are seen in minority racial groups compared to NHW patients with AML when treated with traditional cytotoxic regimens [17,18]. In contrast, emerging data suggest NHB patients may do better than NHW patients when treated with hypomethylating agents combined with venetoclax [19]. As a result of low minority clinical trial accrual, current treatment guidelines are biased towards optimizing the outcomes of NHW patients and do not consider the influence of racial ancestry on response and survival.

2.2. Socioeconomic Factors

Structural inequities such as income, insurance status, and geographic access play a critical role in who receives the best care [20,21]. Retrospective cohort studies revealed that NHB patients experience significantly higher in-hospital mortality compared to their NHW counterparts, with disparities influenced by insurance status, hospital type, and patient comorbidities [22]. Byrne et al. (2011) showed that NHB patients, despite being younger at diagnosis, had significantly worse outcomes [23]. Key independent predictors of poor survival included Black race, Medicaid-only coverage, tobacco use, and residence in high-poverty areas [23]. These findings were corroborated by Abraham et al. (2018), who identified race and access to allogeneic transplants as independent predictors of survival in intermediate-risk AML [24]. Additionally, Bradley et al. (2022) found that, while Hispanic patients presented younger and had more adverse-risk cytogenetics (e.g., KMT2A rearrangements), when treated at academic institutions, their overall survival was not significantly different from NHW patients [25]. Hence, equal access to care may help mitigate some disparities. Despite improvements in survival rates, systemic inequities persist. Access to newer therapies such as venetoclax stay uneven, particularly in community settings, where older adults and individuals with higher socioeconomic status are more likely to receive advanced treatments [26]. These findings reflect ongoing challenges in fair access to therapeutic advancements for patients with lower socioeconomic status.

2.3. Environmental Factors

Additionally, environmental exposures and alterable lifestyle factors contribute significantly to the development and progression of AML. A study of 789 adult AML patients from six Chicago academic cancer centers found that NHB and Hispanic individuals experienced significantly higher exposure to air pollutants, including PAHPOM, PM2.5, diesel particulates, and benzene, compared to NHW patients [27]. PAHPOM exposure was independently associated with intermediate- and adverse-risk disease, suggesting a possible role of air pollution in disease biology [27]. Occupational chemical exposure, particularly to benzene, was associated with an increased risk of AML and myelodysplastic syndromes in a Minnesota-based population study [28]. Additionally, short-term exposures to vinyl chloride, creosote, and coal dust were positively correlated with AML risk [28]. In addition to occupational exposures, tobacco use can also contribute to AML risk. A meta-analysis including 7746 AML cases reported a 40% increased risk among current smokers and a dose-dependent relationship between smoking intensity, duration, and AML incidence [29]. A case–control study from Texas found significant associations between AML and solvent exposure, smoking, and obesity, with sex-specific and subtype-specific risk variations. The joint effect of solvent exposure and smoking further elevated risk [30]. Additionally, exposure to cytotoxic agents, electrical power sources, mental disorders, and family history of malignancy were identified as significant risk factors for AML [31]. These findings emphasize the importance of addressing environmental and occupational exposures, particularly in socioeconomically and racially marginalized populations, to reduce AML incidence and improve outcomes.

3. Mutations and Prognosis in AML

The molecular landscape of AML directly impacts its prognosis. The ELN has proposed cytogenetic risk stratification and response criteria. They currently describe three risk groups: favorable, intermediate, and adverse. As discussed above, these risk groups are based on large cohorts and clinical trial data that predominantly feature NHW patients. Likewise, studies validating risk stratification also use datasets that are dominated by NHW patients [32].

3.1. Favorable Risk Group

According to ELN 2022, approximately 30% of AML patients fall under the favorable risk category [33]. Studies show that patients in this risk group respond better to standard chemotherapy [2]. Genetic changes in this group include mutations in nucleophosmin-1 (NPM1) and the transcription factor CEBPA [34]. The five-year overall survival rate for these patients is over 50% [35]. Given this survival, HSCT is not recommended as consolidation therapy for these patients.
NPM1 mutations are among the most common in AML, present in roughly 30% of adult cases and up to 50–60% of cytogenetically normal AML (CN-AML) [36]. These mutations disrupt the nucleolar localization of the NPM1 protein, leading to its aberrant cytoplasmic accumulation, which contributes to leukemogenesis by dysregulating differentiation through the upregulation of homeobox genes (HOX). Cytoplasmic NPM1 blocks the migration of GADD45A to the nucleus, hindering its interaction with p53 to initiate apoptosis [37,38,39,40]. NPM1-mutated AML is categorized as favorable risk when FMS-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD) is absent, according to the 2022 ELN classification [41,42]. However, relapses occur in up to 40% of NPM1-mutant (NPM1c) cases, suggesting the need for refined prognostic models incorporating measurable residual disease (MRD) and co-mutations [43]. Recent advances include the use of menin inhibitors, targeting the NPM1: HOX gene expression axis, which has shown preclinical and clinical promise for NPM1-mutant AML [44]. Revumenib, a menin inhibitor recently approved by the FDA for the treatment of AML with KMT2Ar, has also shown activity in NPM1-mutated AML patients and is now approved by the FDA for these patients as well [45]. As will be further discussed below, this improved prognosis appears to be specific to NHW patients. NHB patients with NPM1 mutations have outcomes that more closely resemble those of the adverse-risk category [3]. Despite these findings, the current standard practice is to not recommend HSCT for NHB patients with NPM1 mutations in first remission and highlights the need to incorporate patient specific outcomes, such as the presence of MRD at the time of remission, known to be highly prognostic in NMP1-mutated AML [46,47]. Guidelines emphasize the importance in obtaining MRD measurements. Therefore, MRD measurements in NPM1-mutated NHB patients are of great value.
CCAAT enhancer binding protein alpha (CEBPα) is a transcription factor regulating granulocytic differentiation. CEBPα has two transactivation domains in the N-terminal and one basic leucine zipper region (bZIP) in the C-terminal. The mutations are found in two hotspots, either insertion or deletions in the N-terminal or C-terminal. Frameshift in the N-terminal results in a dominant negative truncated protein, and the same mutation in the C-terminal is in-frame and results in the disruption of CEBPα to DNA or dimerization [48]. CEBPα mutations affect 5–10% of all AML cases [32,49]. Importantly, only mutations affecting the leucine zipper domain of the protein confer a favorable prognosis, and this is irrespective of whether this is mono- or bi-allelic [50]. Complete remission rates with chemotherapy are high in AML patients with these mutations. NHB patients have a lower frequency of CEBPA mutations [51].
Furthermore, translocations t(8;21) or inv(16)/t(16;16) are associated with the formation of novel fusion genes, leading to effects in the regulation of proliferation, differentiation, and viability of leukemic [52,53]. These translocations are seen in 5–10% of all AML cases and are associated with a favorable prognosis [52]. AML patients with these mutations are often treated with cytarabine-based intensive chemotherapy [53].

3.2. Intermediate-Risk Category

The ELN intermediate-risk category includes karyotypes and mutations not classified as favorable or adverse but does specifically include mutations in the receptor tyrosine kinase FLT3. As per ELN 2022 classification, 15–30% of patients fall into this category, and the five-year overall survival rate is less than 40% [35]. HSCT is recommended as consolidation for eligible patients who achieve a complete remission in induction therapy.
FLT3 mutations, particularly the internal tandem duplication (ITD), are associated with increased relapse and poor survival [54,55]. FLT3 mutations activate constitutive signaling pathways that promote proliferation and block apoptosis. They are often co-mutated with NPM1, worsening risk categorization and affecting approximately 10–20% of all adult AML cases [54]. FLT3 inhibitors have improved outcomes, especially in younger patients when combined with intensive chemotherapy [56]. The first-generation FLT3 inhibitor midostaurin was FDA approved in combination with intensive induction chemotherapy in 2017 [57]. The second-generation FLT3 inhibitor gliteritinib has clinical activity and is approved as a monotherapy for relapsed or refractory (R/R) AML [58,59]. Quizartinib, another second-generation FLT3 inhibitor, was more recently approved for combination therapy with intensive induction chemotherapy [60]. Resistance via secondary mutations and clonal evolution remains a challenge [61]. FLT3 mutations are also more prevalent in certain demographic groups. For instance, FLT3-ITD was detected more frequently in younger and female patients, though racial disparities in mutation prevalence remain inconclusive [62].

3.3. Adverse-Risk Category

AML patients under the adverse-risk category harbor autosomal monosomies involving chromosomes 5 or 7, complex karyotypes, mutations in certain tumor suppressors, epigenetic regulators, or spliceosome factors. As the name implies, adverse risk is associated with a worse overall survival rate (less than 10%) and usually affects older patients [35].
Around 2–8% of AML patients have TP53 mutations and are linked to complex karyotypes, therapy-related AML (t-AML), and poor outcomes. These mutations are common in elderly patients and those with prior exposure to chemotherapy or radiation [63]. TP53 mutations confer resistance to standard chemotherapy, hypomethylating agents, and venetoclax, making them a high-priority target for novel therapies [64]. While agents like eprenetapopt and magrolimab (anti-CD47) showed preliminary efficacy [64], confirmatory studies were disappointing. Eprenetapopt in combination with azacitidine failed to achieve the primary outcome in a phase 3 trial of TP53-mutant myelodysplastic syndrome (MDS), and magrolimab in combination with venetoclax and azacitidine was discontinued in phase 3 trials for both MDS and AML [65,66]. Although studies have been limited, a recent report found that NHB patients with TP53 mutations are younger and have more co-mutations than NHW patients [67]. Outcomes in TP53-mutated AML remain dismal regardless of racial ancestry, and there remains a large unmet medical need.
Other mutations contributing to the adverse-risk category include RNA splicing genes such as SRSF2, U2AF1, SF3B1, and ZRSR2, which causes aberrant splicing, resulting in AML, and is associated with unfavorable prognosis [68,69,70]. Furthermore, mutations in genes of the cohesin complex such as STAG1/2, SMC1/3, and RAD21 affect ~10% of AML patients and contribute to poor and adverse prognosis of the disease [70,71]. Frameshift mutations are mostly common in STAG2 and result in a loss of function causing abnormal differentiation of hematopoietic stem cells [71]. Chromatin modifiers such as EZH2, BCOR, and ASXL1 are mutated in AML and result in worse outcomes [70,72]. For currently unknown reasons, mutations in ASXL1 and BCOR are more common in NHB patients [51]. Currently, for adverse-risk AML, the only therapeutic modality with the ability to cure patients is HSCT, and this is recommended for all eligible patients.

4. Racial Ancestry and AML Biology

While many mutations are evenly distributed across races, trends suggest a higher prevalence of favorable-risk mutations (e.g.; NPM1) in NHW patients and lower rates in NHB and Hispanic populations [3,5] contributing to disparities in survival rates. Furthermore, minority populations continue to be underrepresented in clinical trials, limiting the generalizability of newer targeted therapies and risk models [73]. Despite the same treatment regimens, racial disparities in survival persist even among matched molecular risk patients, suggesting the influence of genetic ancestry [74]. Abraham et al. highlighted that Black patients experienced shorter survival even when treated under the same clinical trial protocols, suggesting that disease biologycontributes to the gap [24].
Several authors have explored how NPM1 mutation frequencies differ across racial groups (Table 1) [24,75]. Furthermore, Yunsuk et al. also noted that, while NPM1 mutations are prognostically favorable in general, their frequency and clinical impact may vary by race (summarized in Table 1) [75]. NPM1 mutations are not as frequently seen in NHB patients as in NHW patients [3,5]. Two independent studies by Bhatnagar et al. and Stiff et al. demonstrated that poor outcomes among NHB patients with AML are contradictorily associated with favorable-risk molecular profiles, including NPM1 mutations and wild-type FLT3 [3,5]. Given that NPM1 mutations are present in approximately one-third of AML cases, its clinical and prognostic significance is enormous. As discussed, treatment recommendations are influenced by risk classification, and currently, HSCT is recommended for intermediate and poor AML patients in first complete remission (CR1) [41,76]. This means that NPM1-mutated NHB patients treated with cytotoxic therapy who may benefit from HSCT in consolidation would not typically be recommended one under current guidelines. Additionally, NHB patients without high-risk mutations treated with a hypomethylating agent and venetoclax-based regimens showed improved survival compared to NHW patients [19]. Recently, the randomized PARADIGM study demonstrated that fit patients with intermediate- or adverse-risk AML had better outcomes when treated with azacitidine and venetoclax [77]. Given the classification of NPM1c as favorable risk, providers caring for NHB NPM1c patients may prioritize cytotoxic chemotherapy, as younger NPM1c patients were specifically excluded from this study. Prioritizing research investigating the molecular biology of NPM1c within different racial models to uncover ancestry-specific disease mechanisms is essential to narrow survival gaps and improve treatment recommendations. Survival rates of other minority racial groups also need to be further explored.

5. Molecular Properties of NPM1

NPM1 is a multifunctional protein localized in the nucleoplasm. It is a primary regulator of nucleolar architecture and function, coordinating ribosome biogenesis, stress responses, chromatin organization, and DNA repair. Each domain has a unique characteristic to allow interactions with molecules of important molecular pathways [78]. The protein’s dynamic localization, multivalent binding ability, and liquid–liquid phase separation (LLPS) highlight its involvement in multiple physiological and pathological processes.

Structure of NPM1

The NPM1 gene is found at ch5q35. The transcript has twelve exons. Varying splicing events result in two isoforms of the protein: NPM1.2 and NPM1.3 [78,79]. The primary transcript translates to a protein of 294 amino acids with a molecular weight of 37 kDa. NPM1 is comprised of three independent domains, an N-terminal hydrophobic domain for oligomerization to form homo-pentamers or heterodimers with a nuclear export signal (NES), a central domain with a nuclear localization signal (NLS), and a C-terminal domain allows for nuclear binding: both DNA and RNA and has a nucleolar localization signal (NoLS) (Figure 1a). NPM1 forms a homo-pentamer that is crucial for its activity [79]. X-ray crystallization of the NPM1 pentamer revealed that each monomer has two four-stranded anti-parallel β-sheets that are counter-aligned (Figure 1b). The β-strands connect in a sequential manner to form a pentamer. The far ends of the N- and C-termini in each monomer link together in a random manner for flexibility, and a loop between β2 and β3 of the core region joins to the first acidic tract. A decamer is formed when two pentamers align face to face and assemble into a closed ring, with each monomer from one pentamer connecting to a corresponding monomer from the other [78,79]. NPM1’s oligomeric feature facilitates its interaction with 130 proteins across various subcellular compartments. Through this feature, NPM1 forms pentamers with NPM1c and is transited to cytoplasm. Additionally, in the nucleolus, LLPS occurs through NPM1 condensates formation by its N-terminal oligomerization domain, hence further aiding in interactions with other proteins [80].

6. Role of NPM1 in Nucleolar Organization

NPM1 is a central organizer of the nucleolus through LLPS. It takes part in the formation of membrane-less compartments essential for ribosome biogenesis. Proteins such as SURF6 and ZNF692, as well as helicases like DDX24, modulate NPM1’s localization and dynamics within the granular part of the nucleolus. Alterations in these interactions affect the nucleolar structure and disrupt ribosome production [81,82]. One of the key regulators of NPM1 function is the long non-coding RNA LETN, recently identified as a primate-specific transcript that facilitates the assembly of NPM1 pentamers and nucleolar condensates. LETN’s scaffolding activity is essential for ribosomal RNA transcription and chromatin organization in both neural development and oncogenesis [83]. NPM1 holds intrinsically disordered regions (IDRs), particularly in the acidic domains and linker segments. These IDRs contribute to their ability to engage in multivalent and transient interactions with RNA, histones, and other proteins. Such interactions enable NPM1 to form a “fuzzy” molecular meshwork, essential for buffering proteotoxic stress and preventing aggregation of misfolded proteins [84]. Recent structural analyses have found specific aggregation-prone regions (APRs) within the NTD that predispose NPM1 to aggregation. These regions become particularly relevant in NPM1c+ mutants, which mislocalize to the cytoplasm in AML. This mislocalization is driven by the acquisition of a NES and loss of nucleolar retention motifs, leading to the formation of cytoplasmic aggregates that disrupt cellular homeostasis (also shown in Figure 1a) [40,85].

7. NPM1 in Genome Stability and DNA Repair

NPM1 interacts with proteins involved in cell cycle regulation, chromatin remodeling, and tumor suppression to regulate protein stability, localization, and function through NoLS and multivalent binding domains. APE1 overexpression co-localizes with nucleolar NPM1 biomolecular condensates that recruit ATR, TopBP1, and ETAA1, thereby triggering cell cycle arrest and transcriptional silencing of rRNA genes [86,87]. In the nucleoplasm, NPM1 directly modulates the activity of enzymes involved in base excision repair (BER), such as APE1, Polβ, LigIII, and FEN1. It enhances APE1 endonuclease activity, ensuring the efficient repair of abasic sites (Figure 2) [87,88,89]. Phosphorylated NPM1 (pNPM1) plays a role in homologous recombination repair (HRR) (Figure 2) [90,91,92]. Cyclin-dependent kinases such as CDK1 and CDK2 phosphorylate NPM1 at T199 in the S phase [93,94]. NPM1 shuttles out of the nucleolus into the nucleoplasm and binds to DNA double-stranded break sites (DSBs) and colocalizes with γH2AX [95]. At the break sites, SUMOylation of NPM1 at K263 by SUMO1 helps in the recruitment of HRR proteins like CtIP, BRCA1, and RAD51 [96]. DeSUMOylation of NPM1 by SENP3 dislodges NPM1 from the break site for the DSB repair by HRR to occur successfully [97]. Furthermore, NPM1 stabilizes Polh (DNA polymerase eta), which is involved in trans-lesion synthesis (TLS), allowing replication to proceed past DNA lesions [87,98,99,100,101]. NPM1 also regulates the activity of DOT1L, a histone methyltransferase responsible for H3K79 methylation. Together, NPM1-DOT1L preserves chromatin architecture around the nucleoli and controls the expression of repetitive DNA sequences. Disruption of this axis results in heterochromatin mis-localization and compromised genome stability [102]. NPM1 participates in the PIDDosome complex, which senses centrosome amplification and triggers caspase-2 activation. This leads to MDM2 cleavage, p53 stabilization, and subsequent cell cycle arrest, ensuring genomic integrity in response to mitotic anomalies (Figure 2) [103]. Therefore, AML patients with aberrant cytoplasmic localization of NPM1 may have compromised nucleolar/nucleoplasm pathways, affecting their genomic integrity and promoting genomic abnormalites.

8. NPM1c in AML: Mutational Landscape and Functional Impacts

Mutations in NPM1, particularly frameshift mutations in exon 12, result in aberrant cytoplasmic localization (NPM1c), which is a defining feature in ~30% of AML cases. These mutations disrupt nucleolar retention and enable NPM1 to function abnormally in the cytoplasm and nucleoplasm, promoting leukemogenesis [85]. In NPM1-haploinsufficient mouse models, overexpression of MEIS1 is sufficient to induce AML, with MEIS1 aberrantly binding to the promoter of SMC4 and activating its transcription. This reprograms gene expressions in hematopoietic progenitors, contributing to transformation [104]. NPM1 mutations often cooccur with mutations in cohesin complex genes (e.g., SMC1A, RAD21, and STAG2). These mutations synergistically dysregulate HOXA/MEIS1 expression, promote enhancer hijacking, and block myeloid differentiation, establishing a transcriptional landscape conducive to leukemia [105,106,107]. After Etoposide treatment in the OCI-AML3 AML cell line (which has NPM1c), nucleolar re-localization of NPM1c is observed. This effect is specific to genotoxic drugs and has not been seen with agents acting via other mechanisms [108]. This indicates that NPM1c may retain the DNA repair roles mentioned above and may be partaking in nuclear/nucleoplasm mechanisms, raising awareness of a critical need for deeper investigation and targeting these pathways as a potential therapeutic strategy.

9. Therapeutic Implications and Strategies

NPM1’s centrality in AML pathogenesis makes it an attractive therapeutic target. NPM1c serves as a reliable biomarker for MRD monitoring and risk stratification [2]. Therapies targeting NPM1 aim to restore its nucleolar localization, disrupt its oligomerization, or harness the immune system against NPM1-derived neoantigens. NSC348884 is a small molecule that disrupts NPM1 oligomerization, leading to apoptosis in AML and non-small cell lung cancer (NSCLC) cells. This suggests a promising strategy to impair NPM1c+ functionality and enhance chemosensitivity [109]. Additionally, targeting LLPS has emerged as a novel strategy [110]. In adenovirus infection models, inhibiting NPM1-mediated LLPS hinders viral genome packaging [111,112]. Similarly, modulating interactions between NPM1 and binding partners like p14ARF or LETN may provide avenues to control proliferation in cancer or developmental conditions [83]. Menin-KMT2A inhibitors are a promising approach for targeting HOX-driven transcriptional programs in NPM1c AML [113]. These therapies may overcome resistance mechanisms and improve outcomes for patients with adverse-risk genetic profiles [114]. Finally, drugs that induce nucleolar stress, such as RNA polymerase I inhibitors, could exploit the vulnerability of cancer cells with hyperactive ribosome biogenesis [115,116]. These agents may synergize with therapies targeting NPM1 to enhance apoptotic responses. The use of Menin-KMT2A inhibitors or actinomycin D, especially in conjunction with MEIS1-SMC4 axis inhibitors, has shown synergy in suppressing AML growth in NPM1c contexts [86]. DNA-damaging agents like cytarabine induce the re-localization of NPM1c from the cytoplasm back to the nucleolus, restoring normal nucleolar functions and sensitizing AML cells to nucleolar stress [108]. How effective these strategies will prove to be in patients of differing racial ancestry is an important question.

10. Models to Investigate NPM1c in AML and Disease Progression

Currently, models for studying AML consist of immortalized cell lines, 3D models, and patient-derived xenografts (PDXs). At present, most AML models are of NHW ancestry, with only one cell line of African descent, and it does not contain NPM1c [117,118]. To investigate the role of NPM1c in AML and disease progression, there are no available models of African ancestry. Although primary patient sample cell cultures have been used to study AML, the genetic and functional heterogeneity among different AML samples make the results inconsistent and unreliable [119,120]. In AML, only 25% of all NHB AML patients harbor NPM1 mutations; thus, acquiring samples for primary cell culture and establishing cell lines is challenging [3]. Therefore, increased efforts must be made to generate pre-clinical models of NPM1c using patient-derived xenografts or NPM1c-induced AML in hematopoietic stem cells (HSCs) of NHB racial ancestry. These models are needed to address the influence of NHB racial ancestry on the mechanisms and pathways of NPM1c-mediated leukemogenesis and response to therapy.

11. Conclusions

Disparities in AML survival across racial and ethnic groups are rooted in clinical, social, and biological factors. While socioeconomic barriers and unequal access to care are known, emerging evidence highlights that disease biology may differ by ancestry. Closing the survival gap in AML will require changes across biomedical research, clinical practice, and public health to create a fairer healthcare system. Considering that AML with NPM1 mutations is associated with favorable prognosis when treated with DNA damaging chemotherapy in NHW patients but not NHB, we should prioritize research on NPM1’s substantial role in DNA repair, particularly through the lens of ancestral background. NPM1, as a driver of AML, is a promising target for therapeutic strategies focusing on inhibiting oligomerization, disrupting its protein–protein interactions, and regulating its role in phase-separated nuclear compartments. The lack of appropriate preclinical models has hindered the investigation of DNA repair mechanisms in AML with mutated NPM1. Future clinical trials must include participants across different ancestral groups with pre-planned subgroup analysis to ensure ancestry-specific responses can be determined when present. The current approach of applying results from a predominantly NHW population and assuming similar efficacy in all groups must be replaced. This will pave the way for the development of next-generation targeted therapies that are both precise and inclusive, improving outcomes for underrepresented populations.

Author Contributions

Conceptualization, S.A.S.; writing—original draft preparation, S.A.S.; writing—review and editing, S.A.S.; P.-A.V. and T.S.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Cancer Institute, grant number 1R21CA283135-01A1.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Schematics were created using Biorender.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
NHBNon-Hispanic Black
NHWNon-Hispanic White
NPM1Nucleophosmin 1
AMLAcute myeloid leukemia
HSCTAllogeneic hematopoietic stem cell transplant
ELNEuropean Leukemia Net
CN-AMLCytogenetically normal AML
HOXHomeobox genes
MRDMeasurable residual disease
CEBPαCCAAT enhancer binding protein alpha
bZIPBasic leucine zipper region
FLT3-ITDFMS-like tyrosine kinase 3 internal tandem duplication
t-AMLTherapy-related AML
MDSMyelodysplastic syndrome
CR1Complete remission
LLPSLiquid–liquid phase separation
NES Nuclear export signal
NLSNuclear localization signal
NoLSNucleolar localization signal
IDRsIntrinsically disordered regions
APRsAggregation-prone regions
BERBase excision repair
pNPM1Phosphorylated NPM1
HRRHomologous recombination repair
DSBsDNA double-stranded break sites
TLS Trans-lesion synthesis
NSCLCNon-small cell lung cancer
NPM1cCytoplasmic NPM1
PDXsPatient-derived xenografts
HSCsHematopoietic stem cells

References

  1. Bullinger, L.; Döhner, K.; Döhner, H. Genomics of Acute Myeloid Leukemia Diagnosis and Pathways. J. Clin. Oncol. 2017, 35, 934–946. [Google Scholar] [CrossRef] [PubMed]
  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. Bhatnagar, B.; Kohlschmidt, J.; Mrózek, K.; Zhao, Q.; Fisher, J.L.; Nicolet, D.; Walker, C.J.; Mims, A.S.; Oakes, C.; Giacopelli, B.; et al. Poor Survival and Differential Impact of Genetic Features of Black Patients with Acute Myeloid Leukemia. Cancer Discov. 2020, 11, 626–637. [Google Scholar] [CrossRef]
  4. SEER*Explorer: An Interactive Website for SEER Cancer Statistics [Internet]. Surveillance Research Program, National Cancer Institute; 2 July 2025. Data Source(s): SEER Incidence Data, November 2024 Submission (1975–2022), SEER 21 Registries (Excluding Illinois). Expected Survival Life Tables by Socio-Economic Standards. Available online: https://seer.cancer.gov/statistics-network/explorer/ (accessed on 11 December 2025).
  5. Stiff, A.; Fornerod, M.; Kain, B.N.; Nicolet, D.; Kelly, B.J.; Miller, K.E.; Mrózek, K.; Boateng, I.; Bollas, A.; Garfinkle, E.A.R. Multiomic Profiling Identifies Predictors of Survival in African American Patients with Acute Myeloid Leukemia. Nat. Genet. 2024, 56, 2434–2446. [Google Scholar] [CrossRef]
  6. Unger, J.M.; Cook, E.; Tai, E.; Bleyer, A. The Role of Clinical Trial Participation in Cancer Research: Barriers, Evidence, and Strategies. In American Society of Clinical Oncology Educational Book; American Society of Clinical Oncology: Alexandria, VA, USA, 2016. [Google Scholar]
  7. Hamel, L.M.; Penner, L.A.; Albrecht, T.L.; Heath, E.; Gwede, C.K.; Eggly. Barriers to Clinical Trial Enrollment in Racial and Ethnic Minority Patients with Cancer. Cancer Control 2016, 23, 327–337. [Google Scholar] [CrossRef]
  8. Goel, S.; Negassa, A.; Ghalib, M.H.; Chaudhary, I.; Desai, K.; Shah, U.; Swami, U.; Cohen, B.; Maitra, R.; Mani, S. Outcomes Among Racial and Ethnic Minority Patients with Advanced Cancers in Phase 1 Trials: A Meta-Analysis. JAMA Netw. Open 2024, 7, e2421485. [Google Scholar] [CrossRef] [PubMed]
  9. Loeb, N.; Katsnelson, O.; Jain, A.; Tahvildar, P.; Teitelbaum, D.; Garcia-Horton, A. Race and age disparities in randomized trials of acute myeloid leukemia: A systematic review and meta-analysis. Blood Neoplasia 2025, 2, 100070. [Google Scholar] [CrossRef]
  10. Grant, S.R.; Lin, T.A.; Miller, A.B.; Mainwaring, W.; Espinoza, A.F.; Jethanandani, A.; Walker, G.V.; Smith, B.D.; Ashleigh Guadagnolo, B.; Jagsi, R.; et al. Racial and Ethnic Disparities Among Participants in US-Based Phase 3 Randomized Cancer Clinical Trials. JNCI Cancer Spectr. 2020, 4, pkaa060. [Google Scholar] [CrossRef]
  11. Duma, N.; Vera Aguilera, J.; Paludo, J.; Haddox, C.L.; Gonzalez Velez, M.; Wang, Y.; Leventakos, K.; Hubbard, J.M.; Mansfield, A.S.; Go, R.S.; et al. Representation of Minorities and Women in Oncology Clinical Trials: Review of the Past 14 Years. J. Oncol. Pract. 2018, 14, e1–e10. [Google Scholar] [CrossRef]
  12. Casey, M.; Odhiambo, L.; Aggarwal, N.; Shoukier, M.; Islam, K.M.; Cortes, J. Representation of the population in need for pivotal clinical trials in lymphomas. Blood 2023, 142, 846–855. [Google Scholar] [CrossRef] [PubMed]
  13. Hantel, A.; Luskin, M.R.; Garcia, J.S.; Stock, W.; DeAngelo, D.J.; Abel, G.A. Racial and ethnic enrollment disparities and demographic reporting requirements in acute leukemia clinical trials. Blood Adv. 2021, 5, 4352–4360. [Google Scholar] [CrossRef]
  14. Miranda-Galvis, M.; Tjioe, K.C.; Balas, E.A.; Agrawal, G.; Cortes, J.E. Disparities in survival of hematologic malignancies in the context of social determinants of health: A systematic review. Blood Adv. 2023, 7, 6466–6491. [Google Scholar] [CrossRef]
  15. Statler, A.; Hobbs, H.; Radivoyevitch, T.; Mukherjee, S.; Bell, K.; Advani, A.; Gerds, A.; Nazha, A.; Patel, B.; Carraway, H.; et al. Are Racial Disparities in Acute Myeloid Leukemia (AML) Clinical Trial Enrollment Associated with Comorbidities and/or Organ Dysfunction? Blood 2019, 134, 381. [Google Scholar] [CrossRef]
  16. Kirtane, K.; Lee, S.J. Racial and Ethnic Disparities in Hematologic Malignancies. Blood 2017, 130, 1699–1705. [Google Scholar] [CrossRef]
  17. Özdemir, B.C.; Dotto, G.P. Racial Differences in Cancer Susceptibility and Survival: More Than the Color of the Skin? Trends Cancer 2017, 3, 181–197. [Google Scholar] [CrossRef]
  18. Esnaola, N.F.; Ford, M.E. Racial differences and disparities in cancer care and outcomes: Where’s the rub? Surg. Oncol. Clin. N. Am. 2012, 21, 417–437. [Google Scholar] [CrossRef] [PubMed]
  19. Wang, X.; Gimotty, P.A.; Matthews, A.H.; Mamtani, R.; Luger, S.M.; Hexner, E.O.; Babushok, D.V.; McCurdy, S.R.; Frey, N.V.; Bruno, X.J.; et al. Evolving racial/ethnic disparities in AML survival in the novel therapy era. Blood Adv. 2025, 9, 533–544. [Google Scholar] [CrossRef]
  20. Ross, J.S.; Bradley, E.H.; Busch, S.H. Use of health care services by lower-income and higher-income uninsured adults. JAMA 2006, 295, 2027–2036. [Google Scholar] [CrossRef] [PubMed]
  21. Jindal, M.; Chaiyachati, K.H.; Fung, V.; Manson, S.M.; Mortensen, K. Eliminating health care inequities through strengthening access to care. Health Serv. Res. 2023, 58, 300–310. [Google Scholar] [CrossRef]
  22. Alam, S. Disparities in Mortality Among Acute Myeloid Leukemia-Related Hospitalizations. Cancer Med. 2022, 12, 3387–3394. [Google Scholar] [CrossRef] [PubMed]
  23. Byrne, M.M.; Halman, L.J.; Koniaris, L.G.; Cassileth, P.A.; Rosenblatt, J.D.; Cheung, M.C. Effects of Poverty and Race on Outcomes in Acute Myeloid Leukemia. Am. J. Clin. Oncol. 2011, 34, 297–304. [Google Scholar] [CrossRef]
  24. Abraham, I.E.; Patel, A.A.; Wang, H.; Galvin, J.P.; Frankfurt, O.; Liu, L.; Khan, I. Impact of Race on Outcomes in intermediate Risk Acute Myeloid Leukemia. Blood 2018, 132, 3603. [Google Scholar] [CrossRef]
  25. Bradley, T.; Kwon, D.; Monge, J.; Sekeres, M.; Chandhok, N.; Thomassen, A.; Swords, R.; Padron, E.; Lancet, J.; Talati, C.; et al. Molecular Characteristics and Outcomes in Hispanic and Non-Hispanic Patients with Acute Myeloid Leukemia. eJHaem 2022, 3, 1231–1240. [Google Scholar] [CrossRef]
  26. Hantel, A. Sociodemographic Associations with Uptake of Novel Therapies for Acute Myeloid Leukemia. Blood Cancer J. 2023, 13, 192. [Google Scholar] [CrossRef]
  27. Palmer, A.; Rauscher, G.; Abraham, I.; Burkart, M.; Dave, A.; Raptis, N.; Aleem, A.; Gannamani, V.; Erra, A.; Monick, S.; et al. Air pollutant impact on disease characteristics and outcomes in patients with acute myeloid leukemia. Blood Adv. 2024, 8, 4647–4650. [Google Scholar] [CrossRef]
  28. Poynter, J.N.; Richardson, M.; Roesler, M.; Blair, C.K.; Hirsch, B.; Nguyen, P.; Cioc, A.; Cerhan, J.R.; Warlick, E. Chemical exposures and risk of acute myeloid leukemia and myelodysplastic syndromes in a population-based study. Int. J. Cancer 2017, 140, 23–33. [Google Scholar] [CrossRef]
  29. Fircanis, S.; Merriam, P.; Khan, N.; Castillo, J.J. The relation between cigarette smoking and risk of acute myeloid leukemia: An updated meta-analysis of epidemiological studies. Am. J. Hematol. 2014, 89, E125–E132. [Google Scholar] [CrossRef]
  30. Strom, S.S.; Oum, R.; Elhor Gbito, K.Y.; Garcia-Manero, G.; Yamamura, Y. De novo acute myeloid leukemia risk factors: A Texas case-control study. Cancer 2012, 118, 4589–4596. [Google Scholar] [CrossRef]
  31. Maleki Behzad, M.; Abbasi, M.; Oliaei, I.; Ghorbani Gholiabad, S.; Rafieemehr, H. Effects of Lifestyle and Environmental Factors on the Risk of Acute Myeloid Leukemia: Result of a Hospital-based Case-Control Study. J. Res. Health Sci. 2021, 21, e00525. [Google Scholar] [CrossRef] [PubMed]
  32. Mrózek, K.; Kohlschmidt, J.; Blachly, J.S.; Nicolet, D.; Carroll, A.J.; Archer, K.J.; Mims, A.S.; Larkin, K.T.; Orwick, S.; Oakes, C.C.; et al. Outcome prediction by the 2022 European LeukemiaNet genetic-risk classification for adults with acute myeloid leukemia: An Alliance study. Leukemia 2023, 37, 788–798. [Google Scholar] [CrossRef] [PubMed]
  33. Sargas, C.; Ayala, R.; Larráyoz, M.J.; Chillón, M.C.; Rodriguez-Arboli, E.; Bilbao, C.; Prados de la Torre, E.; Martínez-Cuadrón, D.; Rodríguez-Veiga, R.; Boluda, B.; et al. Comparison of the 2022 and 2017 European LeukemiaNet risk classifications in a real-life cohort of the PETHEMA group. Blood Cancer J. 2023, 13, 77. [Google Scholar] [CrossRef]
  34. Ruhnke, L.; Bill, M.; Zukunft, S.; Eckardt, J.N.; Schäfer, S.; Stasik, S.; Hanoun, M.; Schroeder, T.; Fransecky, L.; Steffen, B.; et al. Validation of the revised 2022 European LeukemiaNet risk stratification in adult patients with acute myeloid leukemia. Blood Adv. 2025, 9, 1392–1404. [Google Scholar] [CrossRef]
  35. Sasaki, K.; Ravandi, F.; Kadia, T.; DiNardo, C.; Borthakur, G.; Short, N.; Jain, N.; Daver, N.; Jabbour, E.; Garcia-Manero, G.; et al. Prediction of survival with intensive chemotherapy in acute myeloid leukemia. Am. J. Hematol. 2022, 97, 865–876. [Google Scholar] [CrossRef] [PubMed]
  36. Boissel, N.; Renneville, A.; Biggio, V.; Philippe, N.; Thomas, X.; Cayuela, J.M.; Terre, C.; Tigaud, I.; Castaigne, S.; Raffoux, E.; et al. Prevalence, clinical profile, and prognosis of NPM mutations in AML with normal karyotype. Blood 2005, 106, 3618–3620. [Google Scholar] [CrossRef] [PubMed]
  37. Falini, B. NPM1-mutated acute myeloid leukemia: New pathogenetic and therapeutic insights and open questions. Am. J. Hematol. 2023, 98, 1452–1464. [Google Scholar] [CrossRef]
  38. Falini, B.; Brunetti, L.; Sportoletti, P.; Martelli, M.P. NPM1-mutated acute myeloid leukemia: From bench to bedside. Blood 2020, 136, 1707–1721. [Google Scholar] [CrossRef]
  39. Falini, B.; Sorcini, D.; Perriello, V.M.; Sportoletti, P. Functions of the native NPM1 protein and its leukemic mutant. Leukemia 2025, 39, 276–290. [Google Scholar] [CrossRef]
  40. Zarka, J.; Short, N.J.; Kanagal-Shamanna, R.; Issa, G.C. Nucleophosmin 1 Mutations in Acute Myeloid Leukemia. Genes 2020, 11, 649. [Google Scholar] [CrossRef] [PubMed]
  41. Shimony, S.; Stahl, M.; Stone, R.M. Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk-Stratification, and Management. Am. J. Hematol. 2025, 100, 860–891. [Google Scholar] [CrossRef]
  42. Chan, O.; Al Ali, N.; Tashkandi, H.; Ellis, A.; Ball, S.; Grenet, J.; Hana, C.; Deutsch, Y.; Zhang, L.; Hussaini, M.; et al. Mutations highly specific for secondary AML are associated with poor outcomes in ELN favorable risk NPM1-mutated AML. Blood Adv. 2024, 8, 1075–1083. [Google Scholar] [CrossRef]
  43. Bertoli, S.; Tavitian, S.; Bérard, E.; Mansat-De Mas, V.; Largeaud, L.; Gadaud, N.; Rieu, J.B.; Vergez, F.; Luquet, I.; Huguet, F.; et al. More Than 10% of NPM1-Mutated AML Relapses Occur after 5 Years from Complete Remission. Blood 2018, 132, 2802. [Google Scholar] [CrossRef]
  44. Perner, F.; Stein, E.M.; Wenge, D.V.; Singh, S.; Kim, J.; Apazidis, A.; Rahnamoun, H.; Anand, D.; Marinaccio, C.; Hatton, C.; et al. MEN1 mutations mediate clinical resistance to menin inhibition. Nature 2023, 615, 913–919. [Google Scholar] [CrossRef] [PubMed]
  45. Syed, Y.Y. Revumenib: First Approval. Drugs 2025, 85, 577–583. [Google Scholar] [CrossRef] [PubMed]
  46. Shayegi, N.; Kramer, M.; Bornhäuser, M.; Schaich, M.; Schetelig, J.; Platzbecker, U.; Röllig, C.; Heiderich, C.; Landt, O.; Ehninger, G.; et al. The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML. Blood 2013, 122, 83–92. [Google Scholar] [CrossRef] [PubMed]
  47. Balsat, M.; Renneville, A.; Thomas, X.; de Botton, S.; Caillot, D.; Marceau, A.; Lemasle, E.; Marolleau, J.P.; Nibourel, O.; Berthon, C.; et al. Postinduction Minimal Residual Disease Predicts Outcome and Benefit From Allogeneic Stem Cell Transplantation in Acute Myeloid Leukemia with NPM1 Mutation: A Study by the Acute Leukemia French Association Group. J. Clin. Oncol. 2017, 35, 185–193. [Google Scholar] [CrossRef]
  48. Su, L.; Shi, Y.Y.; Liu, Z.Y.; Gao, S.J. Acute Myeloid Leukemia with CEBPA Mutations: Current Progress and Future Directions. Front. Oncol. 2022, 12, 806137. [Google Scholar] [CrossRef]
  49. Tarlock, K.; Lamble, A.J.; Wang, Y.C.; Gerbing, R.B.; Ries, R.E.; Loken, M.R.; Brodersen, L.E.; Pardo, L.; Leonti, A.; Smith, J.L.; et al. CEBPA-bZip mutations are associated with favorable prognosis in de novo AML: A report from the Children’s oncology Group. Blood 2021, 138, 1137–1147, Erratum in Blood 2022, 139, 1601. [Google Scholar] [CrossRef]
  50. Taube, F.; Georgi, J.A.; Kramer, M.; Stasik, S.; Middeke, J.M.; Röllig, C.; Krug, U.; Krämer, A.; Scholl, S.; Hochhaus, A.; et al. CEBPA mutations in 4708 patients with acute myeloid leukemia: Differential impact of bZIP and TAD mutations on outcome. Blood 2022, 139, 87–103. [Google Scholar] [CrossRef]
  51. Larkin, K.T.; Nicolet, D.; Kelly, B.J.; Mrózek, K.; LaHaye, S.; Miller, K.E.; Wijeratne, S.; Wheeler, G.; Kohlschmidt, J.; Blachly, J.S.; et al. High early death rates, treatment resistance, and short survival of Black adolescents and young adults with AML. Blood Adv. 2022, 6, 5570–5581. [Google Scholar] [CrossRef]
  52. Prébet, T.; Boissel, N.; Reutenauer, S.; Thomas, X.; Delaunay, J.; Cahn, J.Y.; Pigneux, A.; Quesnel, B.; Witz, F.; Thépot, S.; et al. Acute Myeloid Leukemia with Translocation (8;21) or Inversion (16) in Elderly Patients Treated with Conventional Chemotherapy: A Collaborative Study of the French CBF-AML Intergroup. J. Clin. Oncol. 2009, 28, 4747–4753. [Google Scholar] [CrossRef]
  53. Han, S.Y.; Mrózek, K.; Voutsinas, J.; Wu, Q.; Morgan, E.A.; Vestergaard, H.; Ohgami, R.; Kluin, P.M.; Kristensen, T.K.; Pullarkat, S.; et al. Secondary cytogenetic abnormalities in core-binding factor AML harboring inv(16) vs t(8;21). Blood Adv. 2021, 5, 2481–2489. [Google Scholar] [CrossRef]
  54. Wang, L.H.; Zhou, C.L.; Zhang, X.W.; Chen, S.; Wang, M.; Wang, J.X. Prevalence and clinical significance of FLT3 internal tandem duplication mutation in acute leukemia. Zhonghua Xue Ye Xue Za Zhi 2004, 25, 393–396. [Google Scholar]
  55. Liu, H.; Yu, H.; Jia, H.Y.; Zhang, W.; Guo, C.J. Detection of FLT3 gene mutation in hematologic malignancies and its clinical significance. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2007, 15, 709–713. [Google Scholar]
  56. Kelvin, J.M.; Jain, J.; Thapa, A.; Qui, M.; Birnbaum, L.A.; Moore, S.G.; Zecca, H.; Summers, R.J.; Switchenko, J.M.; Costanza, E.; et al. Constitutively synergistic multiagent drug formulations targeting MERTK, FLT3, and BCL-2 for treatment of AML. Pharm. Res. 2023, 40, 2133–2146. [Google Scholar] [CrossRef] [PubMed]
  57. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Döhner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
  58. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740, Erratum in N. Engl. J. Med. 2022, 386, 1868. [Google Scholar] [CrossRef]
  59. Kiyoi, H.; Kawashima, N.; Ishikawa, Y. FLT3 mutations in acute myeloid leukemia: Therapeutic paradigm beyond inhibitor development. Cancer Sci. 2020, 111, 312–322. [Google Scholar] [CrossRef]
  60. Cortes, J. Quizartinib: A potent and selective FLT3 inhibitor for the treatment of patients with FLT3-ITD–positive AML. J. Hematol. Oncol. 2024, 17, 111. [Google Scholar] [CrossRef]
  61. Daver, N.; Schlenk, R.F.; Russell, N.H.; Levis, M.J. Targeting FLT3 mutations in AML: Review of current knowledge and evidence. Leukemia 2019, 33, 299–312. [Google Scholar] [CrossRef] [PubMed]
  62. Elyamany, G.; Awad, M.; Fadalla, K.; Albalawi, M.; Al Shahrani, M.; Al Abdulaaly, A. Frequency and Prognostic Relevance of FLT3 Mutations in Saudi Acute Myeloid Leukemia Patients. Adv. Hematol. 2014, 2014, 141360. [Google Scholar] [CrossRef] [PubMed]
  63. Qian, Z.; Joslin, J.M.; Tennant, T.R.; Reshmi, S.C.; Young, D.J.; Stoddart, A.; Larson, R.A.; Le Beau, M.M. Cytogenetic and Genetic Pathways in Therapy-Related Acute Myeloid Leukemia. Chem.-Biol. Interact. 2010, 184, 50–57. [Google Scholar] [CrossRef] [PubMed]
  64. DiNardo, C.D.; Tiong, I.S.; Quaglieri, A.; MacRaild, S.; Loghavi, S.; Brown, F.C.; Thijssen, R.; Pomilio, G.; Ivey, A.; Salmon, J.M.; et al. Molecular Patterns of Response and Treatment Failure After Frontline Venetoclax Combinations in Older Patients with AML. Blood 2020, 135, 791–803. [Google Scholar] [CrossRef] [PubMed]
  65. Gilead Sciences. A Phase 3, Randomized, Double-Blind, Placebo-Controlled Study Evaluating the Safety and Efficacy of Magrolimab Versus Placebo in Combination with Venetoclax and Azacitidine in Newly Diagnosed, Previously Untreated Patients with Acute Myeloid Leukemia Who Are Ineligible for Intensive Chemotherapy. 2021. Available online: https://onderzoekmetmensen.nl/en/node/54256/pdf (accessed on 11 December 2025).
  66. Aprea Therapeutics, Inc. 2021. Available online: https://ir.aprea.com/news-releases/news-release-details/aprea-therapeutics-announces-removal-fda-clinical-hold/#:~:text=About%20Aprea%20therapeutics%2C%20inc.&text=the%20Company’s%20lead%20product%20candidate,disclosure%20obligations%20under%20Regulation%20FD (accessed on 11 December 2025).
  67. Abraham, I.E.; Rauscher, G.H.; Patel, A.A.; Pearse, W.B.; Rajakumar, P.; Burkart, M.; Aleem, A.; Dave, A.; Bharadwaj, S.; Paydary, K.; et al. Structural racism is a mediator of disparities in acute myeloid leukemia outcomes. Blood 2022, 139, 2212–2226. [Google Scholar] [CrossRef]
  68. DiNardo, C.D.; Cortes, J.E. Mutations in AML: Prognostic and therapeutic implications. Hematol. Am. Soc. Hematol. Educ. Program 2016, 2016, 348–355. [Google Scholar] [CrossRef]
  69. Ogawa, S. Splicing factor mutations in AML. Blood 2014, 123, 3216–3217. [Google Scholar] [CrossRef]
  70. Ochi, Y.; Ogawa, S. Chromatin-Spliceosome Mutations in Acute Myeloid Leukemia. Cancers 2021, 13, 1232. [Google Scholar] [CrossRef]
  71. Fisher, J.B.; McNulty, M.; Burke, M.J.; Crispino, J.D.; Rao, S. Cohesin Mutations in Myeloid Malignancies. Trends Cancer 2017, 3, 282–293. [Google Scholar] [CrossRef]
  72. Greenblatt, S.M.; Nimer, S.D. Chromatin modifiers and the promise of epigenetic therapy in acute leukemia. Leukemia 2014, 28, 1396–1406. [Google Scholar] [CrossRef] [PubMed]
  73. Mahmood, O.; Saini, T.; Park, D.; Bekal, S.; Bal, I.; Saini, S.; Altai, T.; Fayed, Y.; Yan, Y.; Abdulhaq, H.; et al. Disparities in Genetic Profiles, Risk Stratification and Outcomes in Adults with Acute Myeloid Leukemia Comparing Patients of Hispanic and Non-Hispanic Ethnicity in Central California. Blood 2023, 142, 7423. [Google Scholar] [CrossRef]
  74. Eisfeld, A.K. Disparities in acute myeloid leukemia treatments and outcomes. Curr. Opin. Hematol. 2024, 31, 58–63. [Google Scholar] [CrossRef]
  75. Choi, Y.; Lee, J.-H.; Lee, J.-H.; Kim, D.-Y.; Lim, S.; Kim, S.-D.; Seol, M.; Seo, E.-J.; Park, S.-J.; Jang, S.; et al. Molecular Characterization of Normal Karyotype Acute Myeloid Leukemia: Role of the NPM1 Mutations. Blood 2009, 114, 4684. [Google Scholar] [CrossRef]
  76. Loke, J.; Buka, R.; Craddock, C. Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia: Who, When, and How? Front. Immunol. 2021, 12, 659595. [Google Scholar] [CrossRef] [PubMed]
  77. Fathi, A.; Perl, A.; Fell, G.; Jonas, B.; Ragon, B.; Mims, A.; Borate, U.; Mannis, G.; Quillen, K.; Stahl, M.; et al. Results from paradigm—A phase 2 randomized multi-center study comparing azacitidine and venetoclax to conventional induction chemotherapy for newly diagnosed fit adults with acute myeloid leukemia. Blood 2025, 146, 6. [Google Scholar] [CrossRef]
  78. Lee, H.H.; Kim, H.S.; Kang, J.Y.; Lee, B.I.; Ha, J.Y.; Yoon, H.J.; Lim, S.O.; Jung, G.; Suh, S.W. Crystal structure of human nucleophosmin-core reveals plasticity of the pentamer–pentamer interface. Proteins Struct. Funct. Bioinform. 2007, 69, 672–678. [Google Scholar] [CrossRef]
  79. Mitrea, D.M.; Grace, C.R.; Buljan, M.; Yun, M.K.; Pytel, N.J.; Satumba, J.; Nourse, A.; Park, C.G.; Madan Babu, M.; White, S.W.; et al. Structural Polymorphism in the N-Terminal Oligomerization Domain of NPM1. Proc. Natl. Acad. Sci. USA 2014, 111, 4466–4471. [Google Scholar] [CrossRef]
  80. Mitrea, D.M.; Cika, J.A.; Stanley, C.B.; Nourse, A.; Onuchic, P.L.; Banerjee, P.R.; Phillips, A.H.; Park, C.G.; Deniz, A.A.; Kriwacki, R.W. Self-interaction of NPM1 modulates multiple mechanisms of liquid-liquid phase separation. Nat. Commun. 2018, 9, 842. [Google Scholar] [CrossRef]
  81. Mitrea, D.M.; Kriwacki, R.W. Phase separation in biology; functional organization of a higher order. Annu. Rev. Biophys. 2018, 47, 285–305. [Google Scholar] [CrossRef] [PubMed]
  82. Zhang, H.; Chen, Q.; Zhang, Q.; Gan, H.; Li, H.; Chen, S.; Shan, H.; Pang, P.; He, H. DDX24 Mutation Alters NPM1 Phase Behavior and Disrupts Nucleolar Homeostasis in Vascular Malformations. Int. J. Biol. Sci. 2023, 19, 4123–4138. [Google Scholar] [CrossRef]
  83. Wang, X.; Hu, X.; Song, W.; Xu, H.; Xiao, Z.; Huang, R.; Bai, Q.; Zhang, F.; Chen, Y.; Liu, Y.; et al. Mutual dependency between lncRNA LETN and protein NPM1 in controlling the nucleolar structure and functions sustaining cell proliferation. Cell Res. 2021, 31, 664–683. [Google Scholar] [CrossRef]
  84. Saluri, M.; Leppert, A.; Gese, G.V.; Sahin, C.; Lama, D.; Kaldmäe, M.; Chen, G.; Elofsson, A.; Allison, T.M.; Arsenian-Henriksson, M.; et al. A grappling hook mechanism drives network assembly and chaperone function of NPM1 in response to stress. Nat. Commun. 2023, 14, 3132. [Google Scholar]
  85. Chen, E.C.; Shimony, S.; Luskin, M.R.; Stone, R.M. Biology and Management of Acute Myeloid Leukemia with Mutated NPM1. Am. J. Hematol. 2025, 100, 652–665. [Google Scholar] [CrossRef]
  86. Li, J.; Yan, S. Molecular mechanisms of nucleolar DNA damage checkpoint response. Trends Cell Biol. 2023, 33, 361–364. [Google Scholar] [CrossRef]
  87. Li, J.; Zhao, H.; McMahon, A.; Yan, S. APE1 assembles biomolecular condensates to promote the ATR-Chk1 DNA damage response in nucleolus. Nucleic Acids Res. 2022, 50, 10503–10525. [Google Scholar] [CrossRef]
  88. Di Matteo, A.; Franceschini, M.; Paiardini, A.; Grottesi, A.; Chiarella, S.; Rocchio, S.; Di Natale, C.; Marasco, D.; Vitagliano, L.; Travaglini-Allocatelli, C.; et al. Structural investigation of nucleophosmin interaction with the tumor suppressor Fbw7gamma. Oncogenesis 2017, 6, e379. [Google Scholar] [CrossRef] [PubMed]
  89. Poletto, M.; Lirussi, L.; Wilson, D.M., 3rd; Tell, G. Nucleophosmin modulates stability, activity, and nucleolar accumulation of base excision repair proteins. Mol. Biol. Cell 2014, 25, 1641–1652. [Google Scholar] [CrossRef] [PubMed]
  90. Taha, M.S.; Ahmadian, M.R. Nucleophosmin: A Nucleolar Phosphoprotein Orchestrating Cellular Stress Responses. Cells 2024, 13, 1266. [Google Scholar] [CrossRef]
  91. Traver, G.; Sekhar, K.R.; Crooks, P.A.; Keeney, D.S.; Freeman, M.L. Targeting NPM1 in irradiated cells inhibits NPM1 binding to RAD51, RAD51 foci formation and radiosensitizes NSCLC. Cancer Lett. 2021, 500, 220–227. [Google Scholar] [CrossRef] [PubMed]
  92. Penthala, N.R.; Crooks, P.A.; Freeman, M.L.; Sekhar, K.R. Development and validation of a novel assay to identify radiosensitizers that target nucleophosmin 1. Bioorg. Med. Chem. 2015, 23, 3681–3686. [Google Scholar] [CrossRef]
  93. Eirín-López, J.M.; Frehlick, L.J.; Ausió, J. Long-term evolution and functional diversification in the members of the nucleophosmin/nucleoplasmin family of nuclear chaperones. Genetics 2006, 173, 1835–1850. [Google Scholar] [CrossRef]
  94. Okuda, M.; Horn, H.F.; Tarapore, P.; Tokuyama, Y.; Smulian, A.G.; Chan, P.K.; Knudsen, E.S.; Hofmann, I.A.; Snyder, J.D.; Bove, K.E.; et al. Nucleophosmin/B23 is a target of CDK2/cyclin E in centrosome duplication. Cell 2000, 103, 127–140. [Google Scholar] [CrossRef]
  95. Lee, S.Y.; Park, J.H.; Kim, S.; Park, E.J.; Yun, Y.; Kwon, J. A proteomics approach for the identification of nucleophosmin and heterogeneous nuclear ribonucleoprotein C1/C2 as chromatin-binding proteins in response to DNA double-strand breaks. Biochem. J. 2005, 388, 7–15. [Google Scholar] [CrossRef]
  96. Liu, X.; Liu, Z.; Jang, S.W.; Ma, Z.; Shinmura, K.; Kang, S.; Dong, S.; Chen, J.; Fukasawa, K.; Ye, K. Sumoylation of nucleophosmin/B23 regulates its subcellular localization, mediating cell proliferation and survival. Proc. Natl. Acad. Sci. USA 2007, 104, 9679–9684. [Google Scholar] [CrossRef] [PubMed]
  97. Xu, R.; Yu, S.; Zhu, D.; Huang, X.; Xu, Y.; Lao, Y.; Tian, Y.; Zhang, J.; Tang, Z.; Zhang, Z.; et al. hCINAP regulates the DNA-damage response and mediates the resistance of acute myelocytic leukemia cells to therapy. Nat. Commun. 2019, 10, 3812. [Google Scholar] [CrossRef]
  98. Vascotto, C.; Fantini, D.; Romanello, M.; Cesaratto, L.; Deganuto, M.; Leonardi, A.; Radicella, J.P.; Kelley, M.R.; D’Ambrosio, C.; Scaloni, A.; et al. APE1/Ref-1 interacts with NPM1 within nucleoli and plays a role in the rRNA quality control process. Mol. Cell. Biol. 2011, 31, 2559–2574. [Google Scholar] [CrossRef]
  99. Komaniecka, N.; Porras, M.; Cairn, L.; Santas, J.A.; Ferreiro, N.; Penedo, J.C.; Bañuelos, S. Conformational Rearrangements Regulating the DNA Repair Protein APE1. Int. J. Mol. Sci. 2022, 23, 8015. [Google Scholar] [CrossRef]
  100. Lopez, D.J.; Rodriguez, J.A.; Banuelos, S. Molecular Mechanisms Regulating the DNA Repair Protein APE1: A Focus on Its Flexible N-Terminal Tail Domain. Int. J. Mol. Sci. 2021, 22, 6308. [Google Scholar] [CrossRef]
  101. López, D.J.; de Blas, A.; Hurtado, M.; García-Alija, M.; Mentxaka, J.; de la Arada, I.; Urbaneja, M.A.; Alonso-Mariño, M.; Bañuelos, S. Nucleophosmin interaction with APE1: Insights into DNA repair regulation. DNA Repair 2020, 88, 102809. [Google Scholar] [CrossRef]
  102. Izzo, A.; Akol, I.; Villarreal, A.; Lebel, S.; Garcia-Miralles, M.; Cheffer, A.; Bovio, P.; Heidrich, S.; Vogel, T. Nucleophosmin 1 cooperates with the methyltransferase DOT1L to preserve peri-nucleolar heterochromatin organization by regulating H3K27me3 levels and DNA repeats expression. Epigenetics Chromatin 2023, 16, 36. [Google Scholar] [CrossRef]
  103. Fava, L.L.; Schuler, F.; Sladky, V.; Haschka, M.D.; Soratroi, C.; Eiterer, L.; Demetz, E.; Weiss, G.; Geley, S.; Nigg, E.A.; et al. The PIDDosome activates p53 in response to centrosome loss. Nat. Cell Biol. 2017, 19, 486–496. [Google Scholar]
  104. Muranyi, A.; Ammer, T.; Kechter, A.; Rawat, V.P.S.; Sinha, A.; Gonzalez-Menendez, I.; Quintanilla-Martinez, L.; Azoitei, A.; Günes, C.; Mupo, A.; et al. Npm1 haploinsufficiency in collaboration with MEIS1 is sufficient to induce AML in mice. Blood Adv. 2023, 7, 351–364. [Google Scholar] [CrossRef] [PubMed]
  105. Simonetti, G.; Mengucci, C.; Padella, A.; Fonzi, E.; Picone, G.; Delpino, C.; Nanni, J.; De Tommaso, R.; Franchini, E.; Papayannidis, C.; et al. Integrated genomic-metabolic classification of acute myeloid leukemia defines a subgroup with NPM1 and cohesin/DNA damage mutations. Leukemia 2021, 35, 2813–2826. [Google Scholar] [CrossRef]
  106. Tyner, J.W.; Tognon, C.E.; Bottomly, D.; Wilmot, B.; Kurtz, S.E.; Savage, S.L.; Long, N.; Schultz, A.R.; Traer, E.; Abel, M.; et al. Genomic landscape of AML. Nature 2021, 584, 273–278. [Google Scholar]
  107. Heimbruch, K.E.; Meyer, A.E.; Agrawal, P.; Viny, A.D.; Rao, S. A cohesive look at leukemogenesis: The cohesin complex and other driving mutations in AML. Neoplasia 2021, 23, 337–347. [Google Scholar] [CrossRef] [PubMed]
  108. Bailey, G.D.; Qutob, H.M.H.; Akhtar, A.; Russell, N.H.; Seedhouse, C.H. DNA damage corrects the aberrant cytoplasmic localisation of nucleophosmin in NPM1 mutated acute myeloid leukaemia. Br. J. Haematol. 2019, 186, 343–347. [Google Scholar] [CrossRef] [PubMed]
  109. Balusu, R.; Fiskus, W.; Rao, R.; Chong, D.G.; Nalluri, S.; Mudunuru, U.; Ma, H.; Chen, L.; Venkannagari, S.; Ha, K.; et al. Targeting levels or oligomerization of nucleophosmin 1 induces differentiation and loss of survival of human AML cells with mutant NPM1. Blood 2011, 118, 3096–3106. [Google Scholar] [CrossRef]
  110. Datar, G.K.; Khabusheva, E.; Anand, A.; Beale, J.; Sadek, M.; Chen, C.W.; Potolitsyna, E.; Alcantara-Contessoto, N.; Liu, G.; De La Fuente, J.; et al. Nuclear Phase Separation Drives NPM1-mutant Acute Myeloid Leukemia. bioRxiv 2025. bioRxiv:2025.05.23.655671. [Google Scholar] [CrossRef]
  111. Genoveso, M.J.; Okuwaki, M.; Kato, K.; Nagata, K.; Kawaguchi, A. Nuclear reorganization by NPM1-mediated phase separation triggered by adenovirus core protein VII. Microbiol. Spectr. 2024, 12, e00416-24. [Google Scholar] [CrossRef]
  112. Genoveso, M.J.; Hisaoka, M.; Komatsu, T.; Wodrich, H.; Nagata, K.; Okuwaki, M. Formation of adenovirus DNA replication compartments and viral DNA accumulation sites by host chromatin regulatory proteins including NPM1. FEBS J. 2020, 287, 205–217. [Google Scholar] [CrossRef] [PubMed]
  113. Issa, G.C.; Aldoss, I.; DiPersio, J.; Cuglievan, B.; Stone, R.; Arellano, M.; Thirman, M.J.; Patel, M.R.; Dickens, D.S.; Shenoy, S.; et al. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature 2023, 615, 920–924. [Google Scholar] [CrossRef]
  114. Uckelmann, H.J. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science 2020, 367, 586–590. [Google Scholar] [CrossRef]
  115. Carotenuto, P.; Pecoraro, A.; Palma, G.; Russo, G.; Russo, A. Therapeutic Approaches Targeting Nucleolus in Cancer. Cells 2019, 8, 1090. [Google Scholar] [CrossRef] [PubMed]
  116. Hein, N.; Cameron, D.P.; Hannan, K.M.; Nguyen, N.N.; Fong, C.Y.; Sornkom, J.; Wall, M.; Pavy, M.; Cullinane, C.; Diesch, J.; et al. Inhibition of Pol I transcription treats murine and human AML by targeting the leukemia-initiating cell population. Blood 2017, 129, 2882–2895. [Google Scholar] [CrossRef]
  117. Dutil, J.; Chen, Z.; Monteiro, A.N.; Teer, J.K.; Eschrich, S.A. An Interactive Resource to Probe Genetic Diversity and Estimated Ancestry in Cancer Cell Lines. Cancer Res. 2019, 79, 1263–1273. [Google Scholar] [CrossRef]
  118. Ghandi, M.; Huang, F.W.; Jané-Valbuena, J.; Kryukov, G.V.; Lo, C.C.; McDonald, E.R., 3rd; Barretina, J.; Gelfand, E.T.; Bielski, C.M.; Li, H.; et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 2019, 569, 503–508. [Google Scholar] [CrossRef]
  119. Salazar-Terreros, M.J.; Vernot, J.P. In Vitro and In Vivo Modeling of Normal and Leukemic Bone Marrow Niches: Cellular Senescence Contribution to Leukemia Induction and Progression. Int. J. Mol. Sci. 2022, 23, 7350. [Google Scholar] [CrossRef] [PubMed]
  120. Cucchi, D.G.J.; Groen, R.W.J.; Janssen, J.J.W.M.; Cloos, J. Ex vivo cultures and drug testing of primary acute myeloid leukemia samples: Current techniques and implications for experimental design and outcome. Drug Resist. Updates 2020, 53, 100730. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure and conformation of NPM1. (a) Gene and domain structure of NPM1 highlighting TCTG insertion, causing the frameshift mutation of the NPM1c mutant. NES, nuclear export signal; NLS, nuclear localization signal; NoLS, nucleolar localization signal. (b) Side view of NPM1 pentamers forming homo-decamers that facilitate molecular interactions.
Figure 1. Structure and conformation of NPM1. (a) Gene and domain structure of NPM1 highlighting TCTG insertion, causing the frameshift mutation of the NPM1c mutant. NES, nuclear export signal; NLS, nuclear localization signal; NoLS, nucleolar localization signal. (b) Side view of NPM1 pentamers forming homo-decamers that facilitate molecular interactions.
Ijms 27 00510 g001
Figure 2. Molecular interactions of NPM1. (a) NPM1 is a DNA repair factor. Phosphorylation of NPM1 (-P) at T199 is mediated by CDK1/2 in the S phase and leads to NPM1 recruitment to DNA double-stranded breaks (DSBs), where the proteins participate in the initiation of repair by homologous recombination (HR): SUMOylation of NPM1 by Arf at K263 helps recruit HR factors, including CTIP, BRCA1, and RAD51. In single-stranded break (SSB) repair, NPM1 interacts with APE1 to facilitate the base excision repair (BER) pathway. (b) NPM1 is abundantly found in nucleoli and interacts with SURF6, DDX4, and ZNF692 for ribosome biogenesis and the formation of nuclear condensates. (c) NPM1 regulates MDM2 and is involved in the PIDDosome complex with caspase 2, resulting in cell cycle arrest and apoptosis. (d) NPM1 monomers and pentamers of wild-type (or mutated) NPM1 are exported to the cytoplasm via XPO1. There, NPM1 interacts with MAPK31 (part of the MAPKKK pathway), thereby impacting proliferation and differentiation.
Figure 2. Molecular interactions of NPM1. (a) NPM1 is a DNA repair factor. Phosphorylation of NPM1 (-P) at T199 is mediated by CDK1/2 in the S phase and leads to NPM1 recruitment to DNA double-stranded breaks (DSBs), where the proteins participate in the initiation of repair by homologous recombination (HR): SUMOylation of NPM1 by Arf at K263 helps recruit HR factors, including CTIP, BRCA1, and RAD51. In single-stranded break (SSB) repair, NPM1 interacts with APE1 to facilitate the base excision repair (BER) pathway. (b) NPM1 is abundantly found in nucleoli and interacts with SURF6, DDX4, and ZNF692 for ribosome biogenesis and the formation of nuclear condensates. (c) NPM1 regulates MDM2 and is involved in the PIDDosome complex with caspase 2, resulting in cell cycle arrest and apoptosis. (d) NPM1 monomers and pentamers of wild-type (or mutated) NPM1 are exported to the cytoplasm via XPO1. There, NPM1 interacts with MAPK31 (part of the MAPKKK pathway), thereby impacting proliferation and differentiation.
Ijms 27 00510 g002
Table 1. Frequency of NPM1c mutations in different ethnicities.
Table 1. Frequency of NPM1c mutations in different ethnicities.
Non-Hispanic
Whites
NPM1c Frequency
Non-Hispanic
Blacks
NPM1c Frequency
Hispanic
NPM1c Frequency
Non-Hispanic
NPM1c Frequency
OutcomesTherapy
Bhatnagar et al. [3]38% (n = 777)25% (n = 72)--Worse OS in NHB patientsIntensive Chemotherapy
Andrew et al. [5]29.3% (n = 323)20% (n = 100)--Worse OS in NHB patientsIntensive Chemotherapy
Mahmood et al. [73]--6.9% (n = 58)10.8% (n = 167)No differences in survival-
Abraham I et al. [24]47% (n = 59)31% (n = 18)25% (n = 17)-Better OS in NHW patientsIntensive or HMA treatment
Wang et al. [19]10% (n = 870)6% (n = 61)13% (n = 45)-Better OS in NHB patientsVenetoclax based low intensity induction therapy
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

Sanaullah, S.A.; Vidi, P.-A.; Pardee, T.S. AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations. Int. J. Mol. Sci. 2026, 27, 510. https://doi.org/10.3390/ijms27010510

AMA Style

Sanaullah SA, Vidi P-A, Pardee TS. AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations. International Journal of Molecular Sciences. 2026; 27(1):510. https://doi.org/10.3390/ijms27010510

Chicago/Turabian Style

Sanaullah, Sarvath Aafreen, Pierre-Alexandre Vidi, and Timothy S. Pardee. 2026. "AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations" International Journal of Molecular Sciences 27, no. 1: 510. https://doi.org/10.3390/ijms27010510

APA Style

Sanaullah, S. A., Vidi, P.-A., & Pardee, T. S. (2026). AML Disparities Across Racial Ancestry Groups: A Spotlight on the NPM1 Mutations. International Journal of Molecular Sciences, 27(1), 510. https://doi.org/10.3390/ijms27010510

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