The nucleophosmin (NPM1) gene encodes for a multifunctional protein with prominent nucleolar localization that shuttles between nucleus and cytoplasm. NPM1 mutations represent the most common genetic lesion in adult acute myeloid leukemia (AML; about one third of cases), and they act deterministically to cause the aberrant cytoplasmic delocalization of NPM1 mutants. Because of its unique features, NPM1-mutated AML is recognized as a distinct entity in the 2017 World Health Organization (WHO) classification of hematopoietic neoplasms. Here, we focus on recently identified functions of wild-type NPM1 in the nucleolus and address new biological and clinical issues related to NPM1-mutated AML. The relevance of the cooperation between NPM1 and other mutations in driving AML with different outcomes is presented. We also discuss the importance of eradicating NPM1-mutated clones to achieve AML cure and the impact of preleukemic clonal hematopoiesis persistence in predisposing to second AML. The contribution of HOX genes’ expression to the development of NPM1-mutated AML is also highlighted. Clinically, yet unsolved diagnostic issues in the 2017 WHO classification of myeloid neoplasms and the importance of NPM1 mutations in defining the framework of European LeukemiaNet genetic-based risk stratification are discussed. Finally, we address the value and limits of NPM1-based measurable residual disease assessment for treatment guidance and present the results of promising preclinical studies with XPO1 and menin-MLL inhibitors.

The nucleophosmin (NPM1) gene encodes for a ubiquitous multifunctional shuttling protein1  with predominant nucleolar localization. NPM1 is the most commonly mutated gene in adult acute myeloid leukemia (AML; ∼30% of cases).2  The most distinguishing feature of NPM1 mutants is their aberrant cytoplasmic localization,3  which led to the discovery of NPM1 mutations by immunohistochemistry (IHC), prior to the next-generation sequencing (NGS) era.2  It was a long journey before NPM1-mutated AML was recognized as a distinct entity in the 2017 World Health Organization (WHO) classification of hematopoietic neoplasms.4,5  Together with double-CEBPA–mutated AML, NPM1-mutated AML is 1 of the 2 WHO leukemia entities defined by a single-gene mutation. Here, we focus on new insights about NPM1 wild-type (NPM1wt) protein and NPM1-mutated AML.

NPM1wt is a nucleolar chaperone protein, active as an oligomer (pentamer/decamer). The structure and putative functions of NPM1wt have been reviewed extensively6-9  and are summarized in Figure 1. Here, we focus on newly described nucleolar functions of NPM1wt.

Figure 1.

Structure and mutliple functions of NPM1wt. (A) Structure of NPM1wt and mutant NPM1 (NPM1mut) for comparison. NPM1wt and NPM1mut share the same structure, only differing in the very last portion of the C terminus. The hydrophobic N terminus contains 2 weak leucine-rich nuclear export signals (NESs) and a self-oligomerization core responsible for the formation of NPM1 pentamers. The N terminus also acts as a chaperone by preventing protein misfolding in the nucleolus. The central portion of NPM1wt is highly disordered and acidic because it contains two 10- to 20-aa stretches of aspartic and glutamic acid residues whose negative charge is involved in binding of NPM1wt to histones. The central region also contains a bipartite NLS driving the protein from the cytoplasm to the nucleoplasm. The C-terminal basic domain of NPM1wt mediates binding to nucleic acids and TP53. The very last portion of the NPM1wt C terminus, with its highly conserved W288 and W290, forms a globular structure consisting of a 3-helix bundle, which is responsible for its nucleolar localization signal (NoLS). Compared with NPM1wt, the C terminus of NPM1mut loses W288 and W290 (or W290 alone) and gains a new NES. (B) Summary of the multiple functions of NPM1wt. NPM1wt inhibits centrosome duplication (1). During the cell cycle, NPM1wt is released from the centrosome to allow its duplication and the formation of the mitotic spindle. With its predominant nucleolar localization and the ability to bind rRNA and ribosomal proteins, NPM1wt cooperates in ribosome biogenesis (2). NPM1wt is also a histone chaperone (3) with the ability to bind core and linker histones and to regulate the formation of perinucleolar heterochromatin. It is also involved in DNA repair (4); NPM1wt binds to TP53, enhancing its stability and transcriptional activity, as well as to APE1, regulating its endonuclease activity depending on the type of DNA damage. All of the above functions have been reviewed extensively.6-9  More details regarding the involvement of NPM1wt in other recently reported functions, such as 2′-O-methylation of rRNA (5) and LLPS in the nucleolus (6) are provided in the text. (C) Schematic representation of NPM1wt behavior upon nucleolar stress. Nucleolar stress through the inhibition of RNA polymerase I results in the spread of NPM1wt from the nucleolus to the nucleoplasm. There, NPM1wt can bind and inhibit HDM2, a protein with E3 ligase activity that promotes TP53 degradation through the proteasome. HDM2 inhibition results in increased levels of TP53, cell cycle arrest, and apoptosis. FBL, fibrillarin; NIPBL, nipped-B-like protein; snoRNA, small nucleolar RNA.

Figure 1.

Structure and mutliple functions of NPM1wt. (A) Structure of NPM1wt and mutant NPM1 (NPM1mut) for comparison. NPM1wt and NPM1mut share the same structure, only differing in the very last portion of the C terminus. The hydrophobic N terminus contains 2 weak leucine-rich nuclear export signals (NESs) and a self-oligomerization core responsible for the formation of NPM1 pentamers. The N terminus also acts as a chaperone by preventing protein misfolding in the nucleolus. The central portion of NPM1wt is highly disordered and acidic because it contains two 10- to 20-aa stretches of aspartic and glutamic acid residues whose negative charge is involved in binding of NPM1wt to histones. The central region also contains a bipartite NLS driving the protein from the cytoplasm to the nucleoplasm. The C-terminal basic domain of NPM1wt mediates binding to nucleic acids and TP53. The very last portion of the NPM1wt C terminus, with its highly conserved W288 and W290, forms a globular structure consisting of a 3-helix bundle, which is responsible for its nucleolar localization signal (NoLS). Compared with NPM1wt, the C terminus of NPM1mut loses W288 and W290 (or W290 alone) and gains a new NES. (B) Summary of the multiple functions of NPM1wt. NPM1wt inhibits centrosome duplication (1). During the cell cycle, NPM1wt is released from the centrosome to allow its duplication and the formation of the mitotic spindle. With its predominant nucleolar localization and the ability to bind rRNA and ribosomal proteins, NPM1wt cooperates in ribosome biogenesis (2). NPM1wt is also a histone chaperone (3) with the ability to bind core and linker histones and to regulate the formation of perinucleolar heterochromatin. It is also involved in DNA repair (4); NPM1wt binds to TP53, enhancing its stability and transcriptional activity, as well as to APE1, regulating its endonuclease activity depending on the type of DNA damage. All of the above functions have been reviewed extensively.6-9  More details regarding the involvement of NPM1wt in other recently reported functions, such as 2′-O-methylation of rRNA (5) and LLPS in the nucleolus (6) are provided in the text. (C) Schematic representation of NPM1wt behavior upon nucleolar stress. Nucleolar stress through the inhibition of RNA polymerase I results in the spread of NPM1wt from the nucleolus to the nucleoplasm. There, NPM1wt can bind and inhibit HDM2, a protein with E3 ligase activity that promotes TP53 degradation through the proteasome. HDM2 inhibition results in increased levels of TP53, cell cycle arrest, and apoptosis. FBL, fibrillarin; NIPBL, nipped-B-like protein; snoRNA, small nucleolar RNA.

Close modal

New functions of NPM1wt in the nucleolus

The nucleolus, a membrane-less organelle, is formed through a “liquid-liquid” phase separation (LLPS) process10-12  that causes the segregation of NPM1wt and other nucleolar components from the surrounding nucleoplasm. This process is similar to how oil and water separate from each other when mixed. LLPS is facilitated by the interaction between pentameric NPM1wt, proteins containing R-motifs (ie, multivalent arginine-rich linear motifs [sharing features with nucleolar localization signals (NoLSs)] and nascent ribosomal RNA [rRNA]).10,13  These heterotypic interactions are critical for nucleolar phase separation and for maintaining the directionality of ribosome biogenesis through the different subcellular compartments (ie, from the phase-separated nucleolus to the nonphase-separated nucleoplasm).13  Examples include the interactions of NPM1wt with p14Arf R-motifs14  or with SURF6 and rRNA.13 

The nucleolus is made-up of a fibrillar core, surrounded by the dense fibrillar component and the granular component (GC). The multilayered structure of the nucleolus is determined by LLPS between nucleolar proteins, primarily NPM1wt and fibrillarin that segregate in the GC and dense fibrillar component, respectively11  (Figure 1B). Within the GC phase, NPM1wt serves as a scaffolding protein for nascent rRNA13  and helps prevent irreversible aggregation of cytoplasmic misfolded proteins migrating to the nucleolus following stress.15 

NPM1wt also participates in rRNA 2′-O-methylation16  (Figure 1B). Specifically, NPM1wt binds directly to several C/D box small nucleolar RNAs16  and to the methyltransferase fibrillarin. The fibrillarin–NPM1–small nucleolar RNA complex actively methylates rRNA, and NPM1wt loss results in altered rRNA 2′-O-methylation and deregulated translation.16  Accordingly, germline loss-of-function mutations in the NPM1 acidic region may contribute to the pathogenesis of dyskeratosis congenita,16  a well-known ribosomopathy. However, the role of altered translation in NPM1-mutated AML remains to be established.

Intracellular shuttling of NPM1wt

The nucleus-cytoplasm shuttling of NPM1wt is regulated by specific signal motifs3  (Figures 1A and 2A). A bipartite nuclear localization signal (NLS), recognized by importin α/β,17  drives NPM1wt from the cytoplasm to the nucleoplasm. NPM1wt then localizes to the nucleolus through an aromatic NoLS, a 3-helix structure located at the C terminus.18,19  NoLS contains 2 tryptophans (W288 and W290) that stabilize the hydrophobic core of triple helix, enabling its proper folding. This is critical for localizing NPM1wt to the nucleolus18  through interaction with G-quadruplexes of ribosomal DNA.20 

Figure 2.

Nucleo-cytoplasmic shuttling of NPM1wt and NPM1 mutants. (A) Physiological NPM1wt shuttling. The bipartite NLS drives the nuclear import of NPM1wt. The C-terminal 3-helix NoLS dictates NPM1wt positioning within the nucleolus, whereas the 2 weak nuclear export signals (NESs) at the N terminus are responsible for its export from the nucleus to the cytoplasm (dashed arrow). Eventually, the nuclear import predominates over export so that almost all NPM1wt resides in the nucleolus. (B) Abnormal traffic of NPM1 mutants. Changes in tryptophans (W) 288 and 290 (or 290 alone), with consequent unfolding of the C-terminal triple-helix, abrogate the capability of the mutant to reside within the nucleolus. Furthermore, the insertion of a third NES motif at the C terminus markedly increases the export of the protein (thick arrow). Thus, the nuclear export predominates over the import, and the NPM1 mutant is delocalized in the cytoplasm. The 3-dimensional reconstruction of confocal images shown in the circles has been adapted from Falini et al.7  Nuclei are stained with 4′,6-diamidino-2-phenylindole, while the nucleoli are double-stained for NPM1 and nucleolin. Boxes show OCI-AML3 cells engineered to express either endogenous NPM1wt or NPM1 mutant fused to GFP. Nuclei are stained with Hoechst 33342.

Figure 2.

Nucleo-cytoplasmic shuttling of NPM1wt and NPM1 mutants. (A) Physiological NPM1wt shuttling. The bipartite NLS drives the nuclear import of NPM1wt. The C-terminal 3-helix NoLS dictates NPM1wt positioning within the nucleolus, whereas the 2 weak nuclear export signals (NESs) at the N terminus are responsible for its export from the nucleus to the cytoplasm (dashed arrow). Eventually, the nuclear import predominates over export so that almost all NPM1wt resides in the nucleolus. (B) Abnormal traffic of NPM1 mutants. Changes in tryptophans (W) 288 and 290 (or 290 alone), with consequent unfolding of the C-terminal triple-helix, abrogate the capability of the mutant to reside within the nucleolus. Furthermore, the insertion of a third NES motif at the C terminus markedly increases the export of the protein (thick arrow). Thus, the nuclear export predominates over the import, and the NPM1 mutant is delocalized in the cytoplasm. The 3-dimensional reconstruction of confocal images shown in the circles has been adapted from Falini et al.7  Nuclei are stained with 4′,6-diamidino-2-phenylindole, while the nucleoli are double-stained for NPM1 and nucleolin. Boxes show OCI-AML3 cells engineered to express either endogenous NPM1wt or NPM1 mutant fused to GFP. Nuclei are stained with Hoechst 33342.

Close modal

The nuclear export of NPM1wt is mediated by the interaction of 2 N-terminal nuclear export signals (NESs) with the nuclear exporter XPO1 (also named CRM1).7  Binding of XPO1 to some of its cargo occurs within the nucleolus.21  We hypothesize that localization of NPM1wt to the GC, the external component of the nucleolus,11  may facilitate the access of XPO1 to this area and its interaction with NPM1wt. However, NPM1wt is not exported efficiently, because the N-terminal NESs have low affinity for XPO1.17  We believe that nuclear export may also be weak because it is opposed by multiple forces that anchor NPM1wt to the nucleolus, including binding of NoLS to G-quadruplexes and interactions of NPM1wt with nucleolar proteins and rRNA.10  Ultimately, the import and anchoring signals exceed the export signals3,17  so that, at steady-state, NPM1wt resides in the nucleolus, whereas only a minimal fraction (although functionally relevant) shuttles between cell compartments.

Posttranslational modifications influence NPM1wt localization and functions

Nucleolar localization of NPM1wt depends upon its oligomerization status, because pentamers usually reside in the nucleolus, whereas monomers are nucleoplasmic.22  Phosphorylation of S48, S88, T95, and S125 favors the permanence of NPM1wt in a monomeric state, promoting its nucleoplasmic localization.23  Phosphorylation of T199, T219, T234, and T237 by a cell cycle–dependent kinase during mitosis hinders the interaction between NPM1wt and rRNA.24  We hypothesize that altering this interaction results in loss of NPM1wt phase separation, facilitating its distribution through the cell during mitosis. Acetylation of K229, K230, K257, and K267 seems to favor its nucleoplasmic localization and binding to RNA polymerase II,25  likely facilitating DNA transcription. Finally, sumoylation and ubiquitination at specific sites may influence NPM1 localization and stability.19 

Structural consequences of NPM1 mutations

The main features of NPM1 mutations are summarized in Table 1. NPM1 mutations are always heterozygous and primarily restricted to exon 12 (<1% involving other exons).7  They probably arise from replication errors primed by an illegitimate terminal deoxynucleotidyl transferase activity.26 NPM1 mutations usually consist of 4-bp insertions that cause a frameshift in the last few C-terminal amino acids of the protein, leading to loss of W288 and W290 (or W290 alone) and generation of a new C-terminal NES27  (Figures 1A and 2B). Both changes are required for cytoplasmic localization of NPM1 mutants.3,27 

Table 1.

Genetic features of NPM1 mutations

Heterozygous mutations detected in about one third of adult AML (50-60% of AML with normal cytogenetic). Less frequent in children (∼7%). 
Mostly restricted to exon 12 (<1% other exons). Mutation A (duplication TCTG) is the most common mutation (75-80% of cases), followed by mutations B and D (∼5% each). 
Probably arising from replication errors primed by illegitimate terminal deoxynucleotidyl transferase activity. 
Found in the whole leukemic population (as proven by IHC) and stable over time (being detected at relapse*). 
Not detected in individuals with clonal hematopoiesis. Driver mutations acting as “gatekeepers” for AML, mostly de novo. 
Frequently co-occur with mutations of FLT3, DNMT3A, and IDH1/2 genes. Prognosis may vary according to the associated mutations. 
Mutually exclusive with AML entities defined by recurrent genetic abnormalities in the 2017 WHO classification of hematopoietic tumors. 
Close association with normal karyotype (∼85% of cases). About 15% of cases carry chromosome aberrations, especially +8, del(9q), +4. 
Associated with distinct gene expression profile characterized by upregulation of HOX genes (HOXA, HOXB) and low expression of CD34 and CD133. 
Close association with distinct microRNA and long noncoding RNA profiles. 
Heterozygous mutations detected in about one third of adult AML (50-60% of AML with normal cytogenetic). Less frequent in children (∼7%). 
Mostly restricted to exon 12 (<1% other exons). Mutation A (duplication TCTG) is the most common mutation (75-80% of cases), followed by mutations B and D (∼5% each). 
Probably arising from replication errors primed by illegitimate terminal deoxynucleotidyl transferase activity. 
Found in the whole leukemic population (as proven by IHC) and stable over time (being detected at relapse*). 
Not detected in individuals with clonal hematopoiesis. Driver mutations acting as “gatekeepers” for AML, mostly de novo. 
Frequently co-occur with mutations of FLT3, DNMT3A, and IDH1/2 genes. Prognosis may vary according to the associated mutations. 
Mutually exclusive with AML entities defined by recurrent genetic abnormalities in the 2017 WHO classification of hematopoietic tumors. 
Close association with normal karyotype (∼85% of cases). About 15% of cases carry chromosome aberrations, especially +8, del(9q), +4. 
Associated with distinct gene expression profile characterized by upregulation of HOX genes (HOXA, HOXB) and low expression of CD34 and CD133. 
Close association with distinct microRNA and long noncoding RNA profiles. 
*

With the exception of rare cases of second AML occurring in the context of clonal hematopoiesis (see text).

Loss of tryptophan(s) causes unfolding of the C-terminal triple helix,18  impairing NPM1’s ability to reside within the nucleolus20  (Figure 2B). The new C-terminal NES27  reinforces the export activity of the N-terminal NESs,28  increasing the probability of NPM1 mutants wandering in the nucleoplasm to be exported to the cytoplasm. Importantly, XPO1 recognizes the C-terminal NES more efficiently than the N-terminal NESs, likely as a result of unfolding of the C-terminal triple helix.17  Eventually, the export exceeds the import and anchoring signals, and NPM1 mutants are delocalized to the cytoplasm of AML cells (Figure 2B).

NPM1 mutants act dominantly over NPM1wt, causing its cytoplasmic delocalization through the formation of NPM1 mutant/NPM1wt heterodimers.3,27  However, the exact proportion of NPM1wt localized to the cytoplasm of NPM1-mutated cells has never been quantified, and its biological relevance remains unclear.

NPM1 mutants are “born to be exported”

All NPM1 mutations, including those rarely occurring outside of exon 12 (ie, exon 5,29  exon 9,3  and exon 113 ) generate cytoplasmic leukemic mutants (Figure 3A). This is also a feature of rare NPM1 fusion proteins, including NPM1-HAUS1,30  and those generated by novel translocations [ie, t(5;10)(q35;q23) NPM1-RPP30, t(5;18)(q34;q12) NPM1-SETBP1, and t(5;6)(q35;q23) NPM1-CCDC28A].31  These fusion proteins lose the NPM1wt C-terminus (including W288 and W290) and acquire a C-terminal NES that is either newly created or already present in the fusion partner.

Figure 3.

NPM1 mutants are "born to be exported." (A) A new NPM1 mutation at exon 5. IHC highlights leukemic blasts with cytoplasmic NPM1 (arrows) and normal erythroid cells with a nucleus-restricted expression of NPM1 (asterisk) (bone marrow biopsy, immune-alkaline phosphatase staining using anti–N terminus antibody, clone 376; original magnification ×400) (upper left panel). Sequence of exon 12 of NPM1 was wild-type (data not shown) (lower left panel). Targeted sequencing of the other NPM1 gene exons revealed a new mutation at exon 5, consisting of an in-frame 27-nucleotide insertion and leading to a mutant protein that is 9 aa (shown in red) longer than the wild-type (NPM1wt). Analysis of the new protein sequence predicted a newly acquired NES. Schematic representations of the new mutant protein (NPM1mut-Ex5) compared with wild-type (NPM1wt) (upper right panel). Both are recognized by the anti–N terminus monoclonal antibody (clone 376). The C terminus of the new mutant is unchanged. Cloning of the new mutant NPM1 gene sequence into a GFP-containing vector and expression in NIH3T3 cells confirmed the cytoplasmic localization of the NPM1 mutant protein (GFP-NPM1mut-Ex5), similarly to NPM1 mutant A (GFP-NPM1mutA) (lower right panel). Nucleolar localization of NPM1wt is shown as control (GFP-NPM1wt) (fluorescence microscopy, original magnification ×400). (B) Tuning of NES motif strength. When the capability of NPM1 mutant to bind the nucleolus is completely abrogated (loss of W288 and W290), a weaker C-terminal NES is inserted. When the capability of NPM1 mutant to bind the nucleolus is partially retained (loss of W290 alone), a stronger C-terminal NES motif is inserted. The 3-dimensional reconstruction in the upper left panel is from Falini et al.7 

Figure 3.

NPM1 mutants are "born to be exported." (A) A new NPM1 mutation at exon 5. IHC highlights leukemic blasts with cytoplasmic NPM1 (arrows) and normal erythroid cells with a nucleus-restricted expression of NPM1 (asterisk) (bone marrow biopsy, immune-alkaline phosphatase staining using anti–N terminus antibody, clone 376; original magnification ×400) (upper left panel). Sequence of exon 12 of NPM1 was wild-type (data not shown) (lower left panel). Targeted sequencing of the other NPM1 gene exons revealed a new mutation at exon 5, consisting of an in-frame 27-nucleotide insertion and leading to a mutant protein that is 9 aa (shown in red) longer than the wild-type (NPM1wt). Analysis of the new protein sequence predicted a newly acquired NES. Schematic representations of the new mutant protein (NPM1mut-Ex5) compared with wild-type (NPM1wt) (upper right panel). Both are recognized by the anti–N terminus monoclonal antibody (clone 376). The C terminus of the new mutant is unchanged. Cloning of the new mutant NPM1 gene sequence into a GFP-containing vector and expression in NIH3T3 cells confirmed the cytoplasmic localization of the NPM1 mutant protein (GFP-NPM1mut-Ex5), similarly to NPM1 mutant A (GFP-NPM1mutA) (lower right panel). Nucleolar localization of NPM1wt is shown as control (GFP-NPM1wt) (fluorescence microscopy, original magnification ×400). (B) Tuning of NES motif strength. When the capability of NPM1 mutant to bind the nucleolus is completely abrogated (loss of W288 and W290), a weaker C-terminal NES is inserted. When the capability of NPM1 mutant to bind the nucleolus is partially retained (loss of W290 alone), a stronger C-terminal NES motif is inserted. The 3-dimensional reconstruction in the upper left panel is from Falini et al.7 

Close modal

Nuclear export of NPM1 mutants is also fine-tuned by the strength of the C-terminal NES, depending on the loss of W288 and W290 (insertion of a weaker NES) or W290 only (insertion of a stronger NES)32  (Figure 3B). Rarely (eg, in exon 5 NPM1 mutations), the new NES may be strong enough to cause nuclear export of leukemic mutants without losing any of the C-terminal tryptophans (“super-NES”). Thus, all NPM1 mutations act to maximize the export of NPM1 mutants, pointing to cytoplasmic delocalization as being critical for leukemogenesis.3,32 

NPM1 mutations and clonal hematopoiesis

Clonal hematopoiesis represents the expansion of a clonal population of hematopoietic precursors carrying ≥1 somatic mutation, without evidence of hematologic malignancy.33-35  Unlike mutations of genes involved in chromatin remodeling (eg, DNMT3A, TET2, ASXL1) and RNA splicing (eg, SF3B1, SRSF2), NPM1 mutations have not been detected in individuals with clonal hematopoiesis.35  Rather they act as “gatekeepers” for AML.35  A widely accepted model of clonal evolution, also supported at the single-cell level,36  sees NPM1-mutated AML developing from preexisting clonal hematopoiesis (eg, DNMT3A or IDH2 driven)33,34,37-39  (Figure 4A). Accordingly, 1 patient with IDH2-mutated clonal hematopoiesis developed AML shortly after the detection of NPM1 mutation.40  Moreover, AML patients carrying both NPM1 and DNMT3A mutations at diagnosis, who respond optimally to standard therapy, show persistence of DNMT3A mutations, despite achieving the eradication of NPM1 clone (as assessed by quantitation of NPM1-mutant transcripts) and experiencing long-term complete remission (CR) and cure.41-43  However, the return to the status of clonal hematopoiesis may predispose these patients to the development of a second hematologic neoplasm. Indeed, although most relapses in NPM1-mutated AML are due to the recurrence of the original NPM1-mutated clone, ∼5% to 10% are characterized by the absence of NPM1 mutations.44  These cases are thought to develop a second AML that is favored by the persistence of clonal hematopoiesis following the eradication of the original NPM1-mutated clone.45,46  This is supported by the long latency between CR and relapse with NPM1 wild-type AML44-46  (giving enough time for the second genetic hit to occur) and the different mutational patterns detected at diagnosis and relapse. NPM1-mutated AML may also relapse with NPM1 mutation switch (eg, from type D to A),47,48  likely representing another example of second AML arising from persistent clonal hematopoiesis. Rarely, clonal hematopoiesis may predispose to develop 2 hematological neoplasms, as we reported in a patient with angioimmunoblastic T-cell lymphoma and NPM1-mutated AML49  (Figure 4B). Thus, NPM1-mutated AML patients in CR with persistent clonal hematopoiesis should undergo long-term surveillance for the development of a second malignancy. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only therapy that has demonstrated the capability to eradicate clonal hematopoiesis.50 

Figure 4.

NPM1 and other mutations cooperate to promote AML. (A) AML with the triple-mutated (DNMT3A/NPM1/FLT3-ITD) genotype is observed in patients and is associated with a particularly poor outcome. DNMT3A mutations are early genetic events that are responsible for the generation of a clonal hematopoiesis. NPM1 mutations are disease-defining genetic lesions that are gatekeepers for AML, whereas FLT3-ITD mutations are late events. Courtesy of Timothy Ley. (B) Hypothetical steps in the sequential development of angioimmunoblastic T-cell lymphoma (AITL) (RHOA mutation; orange) followed by NPM1-mutated AML (NPM1 mutation; pink) in a 45-year-old man. The 2 neoplasms arose from high-risk clonal hematopoiesis in the bone marrow promoted by multiple TET2 and ASXL1 mutations.49  The TET2 Q1654* mutation (black) was present in virtually all bone marrow cells (variant allele frequency, 49.3%). The other mutations (TET2 E1089Kfs, red; TET2 Q888, gray; and ASXL1 E635Rfs, green) showing lower allele frequencies likely occurred later as subclonal events.

Figure 4.

NPM1 and other mutations cooperate to promote AML. (A) AML with the triple-mutated (DNMT3A/NPM1/FLT3-ITD) genotype is observed in patients and is associated with a particularly poor outcome. DNMT3A mutations are early genetic events that are responsible for the generation of a clonal hematopoiesis. NPM1 mutations are disease-defining genetic lesions that are gatekeepers for AML, whereas FLT3-ITD mutations are late events. Courtesy of Timothy Ley. (B) Hypothetical steps in the sequential development of angioimmunoblastic T-cell lymphoma (AITL) (RHOA mutation; orange) followed by NPM1-mutated AML (NPM1 mutation; pink) in a 45-year-old man. The 2 neoplasms arose from high-risk clonal hematopoiesis in the bone marrow promoted by multiple TET2 and ASXL1 mutations.49  The TET2 Q1654* mutation (black) was present in virtually all bone marrow cells (variant allele frequency, 49.3%). The other mutations (TET2 E1089Kfs, red; TET2 Q888, gray; and ASXL1 E635Rfs, green) showing lower allele frequencies likely occurred later as subclonal events.

Close modal

NPM1 and other gene mutations cooperate to promote AML

NPM1 and other mutations, especially those affecting FLT32  and DNMT3A,51  frequently co-occur in AML patients, implying molecular synergisms promoting AML development.52,53  Cooperation mechanisms have been investigated extensively in mouse models. Npm1/Dnmt3a double-mutated mice develop AML but only after a long latency and following transplants,38  suggesting that other mutations are necessary for AML development. Conversely, Npm1/Flt3–internal tandem duplication (ITD) double-mutated mice generate a fully penetrant and short-latency AML54,55  that is due, at least in part, to GATA1 epigenetic deregulation.56 Npm1/Flt3–tyrosine kinase domain (TKD) double-mutated mice also develop AML.57  In this model, the Npm1 mutant relocates mutated Flt3 from the cell surface to the endoplasmic reticulum, leading to Stat5 activation.57 Npm1/Flt3-ITD/Dnmt3a triple-mutated mice die of a particularly aggressive AML that is chemoresistant, likely due to impaired nucleosome remodeling.58  Accordingly, patients with this AML genotype show a very poor prognosis.53,59  These mutations synergize to drive overexpression of hepatic leukemia factor, which leads to increased self-renewal and the aberrant immunophenotype (GPR56high/CD34low) of triple-mutated cells.60,61 

About 20% of NPM1-mutated AMLs carry NRAS or KRAS mutations.53 Npm1/Nras double-mutated mice develop AML with granulocytic differentiation and a less aggressive behavior than Npm1/Flt3-ITD AML, which instead shows monocytic differentiation and short latency.62,63  Similarly, patients with NPM1/NRAS double-mutated AML exhibit a relatively good prognosis.53  All of the above mouse models may serve as a platform for testing novel treatment strategies.

Does NPM1 haploinsufficiency contribute to leukemogenesis?

NPM1 mutations are always heterozygous,2  and complete loss of NPM1wt is embryonic lethal,64  implying that 1 copy of NPM1wt is necessary for cell survival. Npm1 heterozygous knock-out mice develop a myelodysplastic-like disorder that is due to uncontrolled centrosome duplication and aneuploidy,64  demonstrating that NPM1wt is an haploinsufficient tumor suppressor. However, depletion of NPM1wt in NPM1-mutated AML cells is unlikely to drive leukemia through this mechanism. Indeed, NPM1-mutated AML is closely associated with normal karyotype,7  likely because both NPM1wt and NPM1 mutants can properly regulate centrosome duplication.

Role of cytoplasmic delocalization of NPM1 mutants in AML development

NPM1 mutants induce cytoplasmic delocalization of several nuclear proteins involved in apoptosis, DNA repair and differentiation, including ARF, HEXIM1, FBW7, MIZ1, APE1, and caspases 6 and 8 (see also Falini et al3  and Heath et al9 ). Recently, altered localization of the transcription factors CTCF65  and PU.166  has been also reported (Figure 5). Overall, the degree to which the function of these proteins is disrupted by the altered localization remains to be determined because they have been mostly studied in vitro or in nonhematopoietic cells,3  and none of these NPM1 partners was confirmed to be delocalized to the cytoplasm of primary NPM1-mutated AML cells.3,67 

Figure 5.

Proposed model of the pathogenesis of NPM1-mutated AML. Mutant NPM1 promotes leukemogenesis by interacting with chromatin-bound XPO1 at HOX loci to facilitate their expression (A) and binding and exporting several nuclear proteins, including tumor suppressors and transcription factors, to the cytoplasm, likely inhibiting their normal functions (B). import, importin α/β.

Figure 5.

Proposed model of the pathogenesis of NPM1-mutated AML. Mutant NPM1 promotes leukemogenesis by interacting with chromatin-bound XPO1 at HOX loci to facilitate their expression (A) and binding and exporting several nuclear proteins, including tumor suppressors and transcription factors, to the cytoplasm, likely inhibiting their normal functions (B). import, importin α/β.

Close modal

Physiologically, NPM1wt binds to a small nuclear pool of the transcriptional regulator BRD4, exerting an inhibitory effect on its transcriptional activity.68  NPM1 mutants alter this equilibrium, because a significant proportion of NPM1 is delocalized into the cytoplasm without BRD4, which is then free to drive the transcription of its target genes,68  especially BCL2 and MYC. How this mechanism contributes to AML development remains unclear.

NPM1 mutants maintain the leukemic state through HOX overexpression

HOXA and HOXB gene clusters are highly expressed in normal adult hematopoietic stem and progenitor cells (HSPCs) but are physiologically silenced in mature blood cells, suggesting a role in self-renewal.69  Moreover, ectopic HOX expression in normal HSPCs leads to leukemic transformation.69  We provided the first evidence that NPM1-mutated AML exhibits higher HOX levels (specifically HOXA and HOXB)70  as compared to NPM1wt AML. However, the HOXA and HOXB expression signature of NPM1-mutated AML is nearly identical to that of normal HSPCs, suggesting persistent expression rather than upregulation of HOX genes71  and pointing to common mechanisms regulating HOX expression in normal HSPCs and NPM1-mutated AML.71 

The histone modifier MLL1 contributes by regulating HOX expression in NPM1-mutated AML, and this function depends on the interaction between MLL1 and its cofactor menin.72,73  More recently, we showed that, in NPM1-mutated AML cells, HOX expression is directly dependent on NPM1 mutants.74  Accordingly, nuclear relocalization or targeted degradation of mutated NPM1 led to immediate loss of HOX expression, followed by differentiation and growth arrest.74  Thus, NPM1 mutants act in a gain-of-function manner upstream of HOX to maintain the undifferentiated state of leukemic cells.74 

The link between NPM1 mutants and HOX expression remains unclear. NPM1 could directly bind and displace putative factors from the nucleus that are required for proper differentiation and physiological HOX downregulation (eg, the myeloid transcription factor PU.166  or the architectural protein CTCF,65  although its role in HOX regulation in the context of NPM1-mutated AML has recently been questioned75 ). In this model, loss of NPM1 mutants from the cytoplasm would restore the normal activity of these factors. Alternatively, HOX expression could be maintained by recruiting NPM1 mutants to HOX loci through their interaction with chromatin-bound XPO1,76  and cytoplasmic delocalization of NPM1 would merely represent an epiphenomenon. We propose a model in which NPM1 mutants promote leukemia by acting at the chromatin level and by delocalizing NPM1-interacting partners to the cytoplasm, “killing 2 birds with 1 stone” (Figure 5).

Challenges in the diagnosis of NPM1-mutated AML

NPM1-mutated AML with multilineage dysplasia77,78  (Figure 6, top panels) should be distinguished from AML with myelodysplasia-related changes (AML-MRC). According to the 2017 WHO classfication of hematopoietic tumors,5  when NPM1 mutations and multilineage dysplasia coexist, the genetic abnormality supersedes morphology, and the case is diagnosed as NPM1-mutated AML.5  Conversely, AML-MRC should be diagnosed if typical cytogenetic alterations and/or a previous history of myelodysplasia are documented, even in the presence of NPM1 mutations.5 

Figure 6.

IHC in NPM1-mutated AML and myeloid sarcoma.NPM1-mutated AML with multilineage dysplasia showing infiltration by blasts (double arrows) and a monolobated megakaryocyte (single arrow) (bone marrow biopsy, immune-alkaline phosphatase staining, original magnification ×400) (top left panel). Reprinted from Pasqualucci et al.77  Another case of NPM1-mutated AML with multilineage dysplasia double stained for NPM1 (blue) and GATA1 (brown) (top right panel). Numerous erythroid leukemic cells (GATA1 nuclear brown/NPM1 cytoplasmic blue) are present. The arrow indicates a residual normal erythroid cell (original magnification ×400; double staining with immune-alkaline phosphatase/immunoperoxidase). Skin biopsy, showing dermal infiltration by leukemic cells (original magnification ×400; hematoxylin and eosin stain) (middle left panel). Leukemic cells from the same patient show aberrant cytoplasmic expression of NPM1, confirming the diagnosis of NPM1-mutated skin myeloid sarcoma (middle right panel). Normal cells of the overlying epidermis show nucleus-restricted expression of NPM1 (arrow) (original magnification ×400; immune-alkaline phosphatase staining). Testicular biopsy showing massive infiltration by leukemic cells and a residual seminiferous tubule (*) (original magnification ×400; hematoxylin and eosin stain) (bottom left panel). Leukemic cells from the same patient show aberrant cytoplasmic expression of NPM1, confirming a diagnosis of testicular myeloid sarcoma (bottom right panel). Cells of a normal residual seminiferous tubule (*) show nuclear expression of NPM1 (arrow) (original magnification ×400; immune-alkaline phosphatase staining). E, epidermis.

Figure 6.

IHC in NPM1-mutated AML and myeloid sarcoma.NPM1-mutated AML with multilineage dysplasia showing infiltration by blasts (double arrows) and a monolobated megakaryocyte (single arrow) (bone marrow biopsy, immune-alkaline phosphatase staining, original magnification ×400) (top left panel). Reprinted from Pasqualucci et al.77  Another case of NPM1-mutated AML with multilineage dysplasia double stained for NPM1 (blue) and GATA1 (brown) (top right panel). Numerous erythroid leukemic cells (GATA1 nuclear brown/NPM1 cytoplasmic blue) are present. The arrow indicates a residual normal erythroid cell (original magnification ×400; double staining with immune-alkaline phosphatase/immunoperoxidase). Skin biopsy, showing dermal infiltration by leukemic cells (original magnification ×400; hematoxylin and eosin stain) (middle left panel). Leukemic cells from the same patient show aberrant cytoplasmic expression of NPM1, confirming the diagnosis of NPM1-mutated skin myeloid sarcoma (middle right panel). Normal cells of the overlying epidermis show nucleus-restricted expression of NPM1 (arrow) (original magnification ×400; immune-alkaline phosphatase staining). Testicular biopsy showing massive infiltration by leukemic cells and a residual seminiferous tubule (*) (original magnification ×400; hematoxylin and eosin stain) (bottom left panel). Leukemic cells from the same patient show aberrant cytoplasmic expression of NPM1, confirming a diagnosis of testicular myeloid sarcoma (bottom right panel). Cells of a normal residual seminiferous tubule (*) show nuclear expression of NPM1 (arrow) (original magnification ×400; immune-alkaline phosphatase staining). E, epidermis.

Close modal

Unlike AML with recurrent genetic abnormalities, such as t(8;21), inv(16), t(16;16) or PML/RARα, ≥20% bone marrow (BM) blasts are required to diagnose NPM1-mutated AML.5 NPM1-mutated myeloid neoplasms showing ≤20% BM blasts are very rare and have been reported as myelodysplastic syndrome (MDS) or chronic myelomonocytic leukemia (CMML).79-81  However, NPM1-mutated MDS usually showed normal karyotype, CD34 negativity, and good response to chemotherapy, thus resembling AML.80,81  Similarly, NPM1-mutated CMML cases81-83  carried normal karyotype, evolved rapidly to AML,82  especially those with higher NPM1 mutation allelic burden,83  and responded better to chemotherapy than to hypomethylating agents.81  Moreover, in our experience, BM biopsies from NPM1-mutated MDS and CMML often show clusters of NPM1 cytoplasmic-positive blasts, suggestive of early AML. Whether all of the above neoplasms should be classified as NPM1-mutated AML, without regard to blast cell count,84  should be a topic for the next revision of the WHO classification.

Immunohistochemical detection of cytoplasmic NPM1 (a surrogate for NPM1 mutations85 ) may help in defining myeloid sarcoma86  (Figure 6, middle and bottom panels), which is common in NPM1-mutated AML87  and difficult to diagnose by molecular assays because of the frequent limited availability of pathological tissue. IHC also predicts NPM1 mutations outside of exon 12 that may be missed with conventional molecular techniques (Figure 3A).

Prognostic impact of NPM1 mutations

According to the 2017 European LeukemiaNet (ELN) recommendations,88 NPM1 mutations convey a relatively favorable prognosis only if FLT3-ITD is absent or shows a low allelic ratio (<0.5, FLT3-ITDlow). Three recent studies based on chemotherapy alone89-91  did not confirm the favorable prognosis of NPM1-mutated/FLT3-ITDlow AML, whereas, in the RATIFY trial, this genotype showed 73% overall survival at 5 years following chemotherapy plus the FLT3 inhibitor.92  Thus, risk stratification according to FLT3-ITD allelic ratio requires further validation depending on the treatment setting (eg, FLT3 inhibitors or allo-HSCT). The favorable impact of the NPM1-mutated/FLT3-ITD genotype appears to be less evident in elderly patients,89  especially those older than 65 years.93  In general, adverse-risk cytogenetic abnormalities confer a poor prognosis to patients with the favorable NPM1-mutated/FLT3-ITD genotype.94  However, these cases should be classified as AML-MRC, according to the 2017 WHO classification of hematopoietic tumors.5 

NPM1-mutated AML with FLT3-ITD high allelic ratio (>0.5, FLT3-ITDhigh) and is assigned to the intermediate-risk group.88  Recently, it has been proposed that the 2017 ELN classification should be refined by moving all FLT3-ITDhigh patients, regardless of their NPM1 mutation status, from the intermediate-risk category to the adverse-risk category95 ; however, further studies are required to support this proposal. Finally, the prognostic impact of NPM1 mutant allele burden96-98  at diagnosis remains controversial.

Although the 2017 ELN classification is of practical value, genomic classification of AML has shown that the prognosis of NPM1-mutated AML may depend on a variety of accompanying mutations other than FLT3.53  For example, patients with NPM1/N-RAS, NPM1/RAD21,53  or NPM1/FLT3-TKD99  genotypes seem to have a relatively good prognosis. Conversely, cases with NPM1/WT1 double-mutated95  or NPM1/FLT3-ITD/DNMT3A triple-mutated59  genotypes show a particularly adverse outcome. A search for DNMT3A mutations has been recommended in a recent proposal to update the 2017 ELN classification.95  A limitation to the wide application of genomic classification53  is that NPM1-mutated AML is fragmented into many small molecular subsets whose prognostic impact may be difficult to study in clinical trials. Measurable residual disease (MRD) assessment in NPM1-mutated AML patients may help to overcome this limitation.

MRD assessment in NPM1-mutated AML

NPM1 mutations are ideal targets for MRD monitoring because they are frequent, stable at relapse, and not found in subjects with clonal hematopoiesis. We established a real-time quantitative polymerase chain reaction (RQ-PCR) assay to measure NPM1-mutant transcripts to predict hematological relapse and long-term survival.100  RQ-PCR remains the standard method for MRD monitoring in NPM1-mutated AML, although digital droplet PCR101,102  and NGS103,104  are emerging as alternative options. Digital droplet PCR has the advantage of a high sensitivity, and NGS can detect all types of NPM1 mutants, including those not recognized by the currently used kits for RQ-PCR. At present, both techniques are more time consuming and less cost effective than RQ-PCR, but we envision that they will become more widely used in the future.

MRD negativity measured by RQ-PCR in peripheral blood (PB)50  or BM105  and deep reduction in NPM1 mutant transcripts (>3 log in BM,106  to <1% in BM,107  or >4 log in PB108 ) at multiple time points after chemotherapy predict a low risk for leukemia relapse and good survival. Notably, MRD assessment in PB after 2 chemotherapy cycles appears to be even more accurate than genetic-based prognostication in predicting relapse risk.50 

Patients in first CR who remain MRD positive after consolidation therapy benefit from allo-HSCT.108-110  However, this benefit may be only partial, because pretransplant MRD positivity predicts a poor outcome.111,112  Future studies should assess whether converting pretransplant NPM1 MRD positivity to negativity is feasible and clinically meaningful. In our experience, the risk of posttransplant leukemia relapse is reduced by using myeloablative HLA-haploidentical transplantation with regulatory and conventional T-cell–adoptive therapy.113 

In general, any confirmed increase in NPM1 transcripts in BM and/or PB reliably predicts hematological relapse (usually occurring within 3 months).105,114,115  According to ELN recommendations,115  patients undergoing standard treatment should be tested by RQ-PCR at least at diagnosis, after 2 cycles of therapy, and at the end of treatment. MRD should be measured every 3 months for the first 2 years of follow-up. Then, timing of MRD monitoring should be personalized according to relapse risk. These recommendations are also applicable for the follow-up of patients undergoing allo-HSCT.

Patients in molecular relapse could benefit from preemptive therapy, including 5-azacitidine,116  venetoclax-based regimens,117  or immunotherapy.118  However, the value of preemptive treatment remains unclear.

Current therapeutic approaches

Fit adult (≤60 years) patients with NPM1-mutated AML are treated with intensive chemotherapy. NPM1-mutated AML cells usually express high levels of CD33 that is treatable with gemtuzumab ozogamicin. Adding this agent to chemotherapy improved event-free survival and overall survival in 1 study,119  but it resulted in lower relapse incidence with no survival advantage in another study.120  In addition to chemotherapy, NPM1-mutated AML patients should receive inhibitors of FLT3, if this gene is mutated.92,121  Allo-HSCT is usually recommended for NPM1-mutated AML with FLT3-ITDhigh.88,122  Conversely, it is generally accepted that patients with NPM1-mutated AML without FLT3-ITD or with FLT3-ITDlow should not undergo allo-HSCT.92,123-125  It remains to be investigated whether the small subset of patients with low transplant–related risk, HLA-identical donor,109  and suboptimal reduction in NPM1 MRD after 2 chemotherapy cycles benefit from allo-HSCT in first CR.

Older fit patients (>60 years) with NPM1-mutated AML treated with intensive chemotherapy experience 15% to 20% long-term survival.126,127  However, these results may be ameliorated using venetoclax combined with hypomethylating agents.128  Eligible patients with NPM1-mutated AML also benefit from postremission reduced-intensity conditioning129  or nonmyeloablative130  allo-HSCT. Older unfit patients with NPM1-mutated AML rarely show long-term response to single hypomethylating agents.131  Combining venetoclax with hypomethylating agents or low-dose cytarabine is emerging as the standard of care in this setting, inducing CR in ∼90% of cases.132,133 

Given the good CR rates, the National Cancer Research Institute recommends134  that, during the COVID-19 pandemic, venetoclax-based regimens should be considered as an alternative to induction chemotherapy in NPM1-mutated AML patients, particularly those aged > 50 years with comorbidities.128  Stringent molecular monitoring for NPM1 mutant transcripts is mandatory if such a strategy is adopted. This approach could help to bridge patients through the pandemic with the objective to deliver definitive therapy, including allo-HSCT, later on.

Because of the poor prognosis,81  we tend to treat patients with NPM1-mutated MDS and CMML following the same recommendations for NPM1-mutated AML; however, this issue remains controversial.

Targeting the abnormal transport of NPM1 mutants

Nuclear export of NPM1 mutants is XPO1 dependent.27  Thus, XPO1 inhibitors can redirect NPM1 mutants to the nucleus.66,74,135  However, XPO1 inhibitors are not NPM1 specific, because they also inhibit the nuclear export of other NES-containing proteins (eg, TP53 and p21).136,137  Consequently, their antileukemic activity may occur through more complex mechanisms. The selective XPO1 inhibitor selinexor shows low efficacy in AML patients,138  probably because it can be administered only once or twice a week due to its toxicity.138  This schedule does not ensure the stable XPO1 inhibition required to induce differentiation and growth arrest of NPM1-mutated AML cells139  (L.B., unpublished data, December 2019). Because of its lower blood-brain barrier penetration, the new XPO1 inhibitor eltanexor140  is better tolerated than selinexor and is currently administered up to 5 times per week in phase 1/2 studies (NCT02649790), holding promise for therapeutic success.

Targeting HOX expression

A functional HOX network is necessary to maintain the undifferentiated state of NPM1-mutated AML.62,72,74  Therefore, targeting HOX expression may prove effective in this AML entity. The histone methyltransferase MLL1 and its cofactor menin induce the trimethylation of lysine 4 on histone 3, a histone mark that correlates with active transcription of HOX genes and their cofactor MEIS1 (HOX/MEIS). Reducing HOX/MEIS expression by inhibiting menin-MLL interaction is feasible and effective in NPM1-mutated AML.72  Recently, 2 novel menin-MLL inhibitors (MI3454 and VTP-50469) exhibited striking antileukemic activity against NPM1-mutated AML in mice.73,141  Intriguingly, both inhibitors preferentially downregulated the HOX cofactor MEIS1 without impairing the expression levels of HOX genes. We believe that loss of MEIS1 may interfere with the function of multiple HOX transcription factors, leading to differentiation and growth arrest.142  Menin-MLL inhibition prevented the transformation of Npm1-mutated mouse hematopoietic progenitors into leukemic cells, implying that it could also be effective in Npm1-mutated preleukemia.73  However, we believe that menin-MLL inhibition is more likely to benefit patients with frank NPM1-mutated AML, because NPM1 mutations do not drive preleukemic clonal hematopoiesis.

Because pharmacologic XPO1 inhibition also potently downregulates HOX expression in NPM1-mutated AML,74  we envision synchronized inhibition of menin-MLL and XPO1 as an appealing therapeutic option to block HOX/MEIS expression in NPM1-mutated AML.

Targeting the nucleolus

NPM1wt is a nucleolar stress sensor,143  and the nucleolus of NPM1-mutated AML cells may be particularly vulnerable to stress because it is NPM1 depleted as a result of haploinsufficiency and cytoplasmic delocalization.7  Therefore, inducing nucleolar stress could be therapeutically effective in NPM1-mutated AML. Actinomycin D, which triggers nucleolar stress by inhibiting RNA polymerase I, induces CR in NPM1-mutated AML,144  although the mechanism of action remains unclear. Other drugs causing nucleolar stress through inhibition of ribosome biogenesis145  should be investigated in this setting. Blocking NPM1 oligomerization146  represents another potential therapeutic approach, because pentameric/decameric NPM1wt is required for proper nucleolus formation.

Mutant NPM1 as target for immunotherapy

We proposed for the first time that the unique C-terminal sequences of NPM1 mutants represent a potential immunotherapeutic target147  and that cytoplasmic localization of NPM1 mutant may favor its processing by a class I degradation pathway, leading to HLA presentation.147  This concept was further expanded demonstrating that specific autologous T-cell responses against NPM1 mutant peptides can be elicited.148-151  Immune responses were associated with molecular remission in 1 AML patient,152  and they may explain the relatively favorable outcome of NPM1-mutated AML.153  Recently, the T-cell receptor specific for 1 of these peptides bound to HLA-02*01 was isolated; following its retroviral transduction into CD4+ and CD8+ T cells, it elicited an antileukemic effect against NPM1-mutated cells.149  Thus, using engineered T cells against NPM1-mutated peptides bound to HLA represents a promising therapeutic approach.

NPM1 mutations are AML-driving events occurring in the setting of preleukemic clonal hematopoiesis. Evidence that eradication of NPM1-mutated clones results in AML cure41-43  (although persistent clonal hematopoiesis may require close monitoring) should encourage the development of targeted treatments for this AML entity.

Endogenous tagging of wild-type and mutated NPM174  may help us to better understand the functions of these proteins, and mass spectrometry may assist in unraveling their interactome in the cytoplasm. Clarifying how NPM1 mutants deregulate the HOX program and how HOX transcription factors control their downstream targets may also lead to new therapeutic avenues. Finally, functional genomic studies may highlight the sensitivity of combinatorial mutational events to specific drugs (eg, unexpected in vitro sensitivity to ibrutinib154  of NPM1-mutated AML cells carrying FLT3 and/or DNMT3A mutations).

Clinically, it should be clarified whether diagnosis of NPM1-mutated AML is justified, irrespective of blast percentage. Standardization of NPM1 MRD measurement, better definition of its impact at different time points, and its inclusion in the design of future clinical trials in young and elderly patients are warranted. Combining venetoclax with other drugs (eg, arsenic155  or XPO1 inhibitors)156  should be investigated further in NPM1-mutated AML.

The authors thank Ildo Nicoletti for providing the 3-dimensional reconstruction of the confocal images shown in Figure 3B and Timothy Ley (Washington University, St Louis, MO) for critically reading the manuscript and for providing Figure 4A. The authors apologize to those investigators whose papers could not be cited due to space limitations.

This work was supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC IG 2019 number 23604 and AIRC Start-Up 2019 number 22895), the European Research Council (Advanced Grant 2016 number 740230 and Consolidator Grant 2016 number 725725), and the ARC Foundation for Cancer Research (Leopold Griffuel Prize to B.F.).

This review is dedicated to the memory of David Grimwade, who greatly contributed to establishing the clinical impact of MRD monitoring in NPM1-mutated AML.

Contribution: B.F. coordinated the preparation of the review, and all authors performed experiments related to the review and wrote the manuscript.

Conflict-of-interest disclosure: B.F. holds a patent on NPM1 mutants (number 102004901256449). The remaining authors declare no competing financial interests.

Correspondence: Brunangelo Falini, Institute of Hematology and Centro Ricerche Emato-Oncologiche, University of Perugia, Piazzale Menghini, 9, Perugia 06131, Italy; e-mail: brunangelo.falini@unipg.it.

1.
Borer
RA
,
Lehner
CF
,
Eppenberger
HM
,
Nigg
EA
.
Major nucleolar proteins shuttle between nucleus and cytoplasm
.
Cell
.
1989
;
56
(
3
):
379
-
390
.
2.
Falini
B
,
Mecucci
C
,
Tiacci
E
, et al;
GIMEMA Acute Leukemia Working Party
.
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype
.
N Engl J Med
.
2005
;
352
(
3
):
254
-
266
.
3.
Falini
B
,
Bolli
N
,
Liso
A
, et al
.
Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: molecular basis and clinical implications
.
Leukemia
.
2009
;
23
(
10
):
1731
-
1743
.
4.
Falini
B
,
Martelli
MP
,
Bolli
N
, et al
.
Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity?
Blood
.
2011
;
117
(
4
):
1109
-
1120
.
5.
Arber
DA
,
Brunning
RD
,
Le Beau
MM
, et al
. Acute myeloid leukaemia with recurrent genetic abnormalities. In:
Swerdlow
SH
,
Campo
E
,
Harris
NL
, eds., et al.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
, Revised 4th ed.
Lyon, France
:
International Agency for Research on Cancer
;
2017
:
130
-
149
.
6.
Grisendi
S
,
Mecucci
C
,
Falini
B
,
Pandolfi
PP
.
Nucleophosmin and cancer
.
Nat Rev Cancer
.
2006
;
6
(
7
):
493
-
505
.
7.
Falini
B
,
Nicoletti
I
,
Martelli
MF
,
Mecucci
C
.
Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features
.
Blood
.
2007
;
109
(
3
):
874
-
885
.
8.
Scott
DD
,
Oeffinger
M
.
Nucleolin and nucleophosmin: nucleolar proteins with multiple functions in DNA repair
.
Biochem Cell Biol
.
2016
;
94
(
5
):
419
-
432
.
9.
Heath
EM
,
Chan
SM
,
Minden
MD
,
Murphy
T
,
Shlush
LI
,
Schimmer
AD
.
Biological and clinical consequences of NPM1 mutations in AML
.
Leukemia
.
2017
;
31
(
4
):
798
-
807
.
10.
Mitrea
DM
,
Cika
JA
,
Guy
CS
, et al
.
Nucleophosmin integrates within the nucleolus via multi-modal interactions with proteins displaying R-rich linear motifs and rRNA
.
eLife
.
2016
;
5
:
e13571
.
11.
Feric
M
,
Vaidya
N
,
Harmon
TS
, et al
.
Coexisting liquid phases underlie nucleolar subcompartments
.
Cell
.
2016
;
165
(
7
):
1686
-
1697
.
12.
Mitrea
DM
,
Cika
JA
,
Stanley
CB
, et al
.
Self-interaction of NPM1 modulates multiple mechanisms of liquid-liquid phase separation
.
Nat Commun
.
2018
;
9
(
1
):
842
.
13.
Riback
JA
,
Zhu
L
,
Ferrolino
MC
, et al
.
Composition-dependent thermodynamics of intracellular phase separation
.
Nature
.
2020
;
581
(
7807
):
209
-
214
.
14.
Mitrea
DM
,
Kriwacki
RW
.
On the relationship status for Arf and NPM1 - it’s complicated
.
FEBS J
.
2018
;
285
(
5
):
828
-
831
.
15.
Frottin
F
,
Schueder
F
,
Tiwary
S
, et al
.
The nucleolus functions as a phase-separated protein quality control compartment
.
Science
.
2019
;
365
(
6451
):
342
-
347
.
16.
Nachmani
D
,
Bothmer
AH
,
Grisendi
S
, et al
.
Germline NPM1 mutations lead to altered rRNA 2′-O-methylation and cause dyskeratosis congenita
.
Nat Genet
.
2019
;
51
(
10
):
1518
-
1529
.
17.
Arregi
I
,
Falces
J
,
Olazabal-Herrero
A
, et al
.
Leukemia-associated mutations in nucleophosmin alter recognition by CRM1: molecular basis of aberrant transport
.
PLoS One
.
2015
;
10
(
6
):
e0130610
.
18.
Grummitt
CG
,
Townsley
FM
,
Johnson
CM
,
Warren
AJ
,
Bycroft
M
.
Structural consequences of nucleophosmin mutations in acute myeloid leukemia
.
J Biol Chem
.
2008
;
283
(
34
):
23326
-
23332
.
19.
Federici
L
,
Falini
B
.
Nucleophosmin mutations in acute myeloid leukemia: a tale of protein unfolding and mislocalization
.
Protein Sci
.
2013
;
22
(
5
):
545
-
556
.
20.
Chiarella
S
,
De Cola
A
,
Scaglione
GL
, et al
.
Nucleophosmin mutations alter its nucleolar localization by impairing G-quadruplex binding at ribosomal DNA
.
Nucleic Acids Res
.
2013
;
41
(
5
):
3228
-
3239
.
21.
Daelemans
D
,
Costes
SV
,
Lockett
S
,
Pavlakis
GN
.
Kinetic and molecular analysis of nuclear export factor CRM1 association with its cargo in vivo
.
Mol Cell Biol
.
2005
;
25
(
2
):
728
-
739
.
22.
Jian
Y
,
Gao
Z
,
Sun
J
, et al
.
RNA aptamers interfering with nucleophosmin oligomerization induce apoptosis of cancer cells
.
Oncogene
.
2009
;
28
(
47
):
4201
-
4211
.
23.
Mitrea
DM
,
Grace
CR
,
Buljan
M
, et al
.
Structural polymorphism in the N-terminal oligomerization domain of NPM1
.
Proc Natl Acad Sci USA
.
2014
;
111
(
12
):
4466
-
4471
.
24.
Okuwaki
M
,
Tsujimoto
M
,
Nagata
K
.
The RNA binding activity of a ribosome biogenesis factor, nucleophosmin/B23, is modulated by phosphorylation with a cell cycle-dependent kinase and by association with its subtype
.
Mol Biol Cell
.
2002
;
13
(
6
):
2016
-
2030
.
25.
Shandilya
J
,
Swaminathan
V
,
Gadad
SS
,
Choudhari
R
,
Kodaganur
GS
,
Kundu
TK
.
Acetylated NPM1 localizes in the nucleoplasm and regulates transcriptional activation of genes implicated in oral cancer manifestation
.
Mol Cell Biol
.
2009
;
29
(
18
):
5115
-
5127
.
26.
Borrow
J
,
Dyer
SA
,
Akiki
S
,
Griffiths
MJ
.
Molecular roulette: nucleophosmin mutations in AML are orchestrated through N-nucleotide addition by TdT
.
Blood
.
2019
;
134
(
25
):
2291
-
2303
.
27.
Falini
B
,
Bolli
N
,
Shan
J
, et al
.
Both carboxy-terminus NES motif and mutated tryptophan(s) are crucial for aberrant nuclear export of nucleophosmin leukemic mutants in NPMc+ AML
.
Blood
.
2006
;
107
(
11
):
4514
-
4523
.
28.
Wang
W
,
Budhu
A
,
Forgues
M
,
Wang
XW
.
Temporal and spatial control of nucleophosmin by the Ran-Crm1 complex in centrosome duplication
.
Nat Cell Biol
.
2005
;
7
(
8
):
823
-
830
.
29.
Martelli
MP
,
Rossi
R
,
Varasano
E
, et al
.
Identification and characterization of novel rare nucleophosmin (NPM1) gene mutations in acute myeloid leukemia (AML) by a combinatorial approach of immunohistochemistry and molecular analyses [abstract]
.
Blood
.
2016
;
128
(
22
). Abstract
1717
.
30.
Campregher
PV
,
de Oliveira Pereira
W
,
Lisboa
B
, et al
.
A novel mechanism of NPM1 cytoplasmic localization in acute myeloid leukemia: the recurrent gene fusion NPM1-HAUS1
.
Haematologica
.
2016
;
101
(
7
):
e287
-
e290
.
31.
Martelli
MP
,
Rossi
R
,
Venanzi
A
, et al
. Novel mutations and translocations involving Nucleophosmin (NPM1) gene in acute myeloid leukemia (AML) and leading to aberrant cytoplasmic NPM1. Paper presented at the 23rd Congress of the European Hematology Association. 15 June 2018. Stockholm, Sweden.
32.
Bolli
N
,
Nicoletti
I
,
De Marco
MF
, et al
.
Born to be exported: COOH-terminal nuclear export signals of different strength ensure cytoplasmic accumulation of nucleophosmin leukemic mutants
.
Cancer Res
.
2007
;
67
(
13
):
6230
-
6237
.
33.
Shlush
LI
,
Zandi
S
,
Mitchell
A
, et al;
HALT Pan-Leukemia Gene Panel Consortium
.
Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia [published correction appears in Nature 2014;508(7496):420]
.
Nature
.
2014
;
506
(
7488
):
328
-
333
.
34.
Corces-Zimmerman
MR
,
Hong
WJ
,
Weissman
IL
,
Medeiros
BC
,
Majeti
R
.
Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission
.
Proc Natl Acad Sci USA
.
2014
;
111
(
7
):
2548
-
2553
.
35.
McKerrell
T
,
Park
N
,
Moreno
T
, et al;
Understanding Society Scientific Group
.
Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis
.
Cell Rep
.
2015
;
10
(
8
):
1239
-
1245
.
36.
Potter
N
,
Miraki-Moud
F
,
Ermini
L
, et al
.
Single cell analysis of clonal architecture in acute myeloid leukaemia
.
Leukemia
.
2019
;
33
(
5
):
1113
-
1123
.
37.
Abelson
S
,
Collord
G
,
Ng
SWK
, et al
.
Prediction of acute myeloid leukaemia risk in healthy individuals
.
Nature
.
2018
;
559
(
7714
):
400
-
404
.
38.
Loberg
MA
,
Bell
RK
,
Goodwin
LO
, et al
.
Sequentially inducible mouse models reveal that Npm1 mutation causes malignant transformation of Dnmt3a-mutant clonal hematopoiesis
.
Leukemia
.
2019
;
33
(
7
):
1635
-
1649
.
39.
Herudkova
Z
,
Culen
M
,
Folta
A
, et al
.
Clonal hierarchy of main molecular lesions in acute myeloid leukaemia [published online ahead of print 10 Dec 2019]
.
Br J Haematol
. doi:10.111/bjh.16341.
40.
Desai
P
,
Mencia-Trinchant
N
,
Savenkov
O
, et al
.
Somatic mutations precede acute myeloid leukemia years before diagnosis
.
Nat Med
.
2018
;
24
(
7
):
1015
-
1023
.
41.
Pløen
GG
,
Nederby
L
,
Guldberg
P
, et al
.
Persistence of DNMT3A mutations at long-term remission in adult patients with AML
.
Br J Haematol
.
2014
;
167
(
4
):
478
-
486
.
42.
Bhatnagar
B
,
Eisfeld
AK
,
Nicolet
D
, et al
.
Persistence of DNMT3A R882 mutations during remission does not adversely affect outcomes of patients with acute myeloid leukaemia
.
Br J Haematol
.
2016
;
175
(
2
):
226
-
236
.
43.
Gaidzik
VI
,
Weber
D
,
Paschka
P
, et al;
German-Austrian Acute Myeloid Leukemia Study Group (AMLSG)
.
DNMT3A mutant transcript levels persist in remission and do not predict outcome in patients with acute myeloid leukemia
.
Leukemia
.
2018
;
32
(
1
):
30
-
37
.
44.
Krönke
J
,
Bullinger
L
,
Teleanu
V
, et al
.
Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia
.
Blood
.
2013
;
122
(
1
):
100
-
108
.
45.
Cocciardi
S
,
Dolnik
A
,
Kapp-Schwoerer
S
, et al
.
Clonal evolution patterns in acute myeloid leukemia with NPM1 mutation
.
Nat Commun
.
2019
;
10
(
1
):
2031
.
46.
Höllein
A
,
Meggendorfer
M
,
Dicker
F
, et al
.
NPM1 mutated AML can relapse with wild-type NPM1: persistent clonal hematopoiesis can drive relapse
.
Blood Adv
.
2018
;
2
(
22
):
3118
-
3125
.
47.
Webersinke
G
,
Kranewitter
W
,
Deutschbauer
S
, et al
.
Switch of the mutation type of the NPM1 gene in acute myeloid leukemia (AML): relapse or secondary AML?
Blood Cancer J
.
2014
;
4
(
6
):
e221
.
48.
Bacher
U
,
Porret
N
,
Joncourt
R
, et al
.
Pitfalls in the molecular follow up of NPM1 mutant acute myeloid leukemia
.
Haematologica
.
2018
;
103
(
10
):
e486
-
e488
.
49.
Tiacci
E
,
Venanzi
A
,
Ascani
S
, et al
.
High-risk clonal hematopoiesis as the origin of AITL and NPM1-mutated AML
.
N Engl J Med
.
2018
;
379
(
10
):
981
-
984
.
50.
Ivey
A
,
Hills
RK
,
Simpson
MA
, et al;
UK National Cancer Research Institute AML Working Group
.
Assessment of minimal residual disease in standard-risk AML
.
N Engl J Med
.
2016
;
374
(
5
):
422
-
433
.
51.
Ley
TJ
,
Ding
L
,
Walter
MJ
, et al
.
DNMT3A mutations in acute myeloid leukemia
.
N Engl J Med
.
2010
;
363
(
25
):
2424
-
2433
.
52.
Ley
TJ
,
Miller
C
,
Ding
L
, et al;
Cancer Genome Atlas Research Network;
.
Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia
.
N Engl J Med
.
2013
;
368
(
22
):
2059
-
2074
.
53.
Papaemmanuil
E
,
Gerstung
M
,
Bullinger
L
, et al
.
Genomic classification and prognosis in acute myeloid leukemia
.
N Engl J Med
.
2016
;
374
(
23
):
2209
-
2221
.
54.
Mupo
A
,
Celani
L
,
Dovey
O
, et al
.
A powerful molecular synergy between mutant Nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice
.
Leukemia
.
2013
;
27
(
9
):
1917
-
1920
.
55.
Mallardo
M
,
Caronno
A
,
Pruneri
G
, et al
.
NPMc+ and FLT3_ITD mutations cooperate in inducing acute leukaemia in a novel mouse model
.
Leukemia
.
2013
;
27
(
11
):
2248
-
2251
.
56.
Sportoletti
P
,
Celani
L
,
Varasano
E
, et al
.
GATA1 epigenetic deregulation contributes to the development of AML with NPM1 and FLT3-ITD cooperating mutations
.
Leukemia
.
2019
;
33
(
7
):
1827
-
1832
.
57.
Rudorf
A
,
Müller
TA
,
Klingeberg
C
, et al
.
NPM1c alters FLT3-D835Y localization and signaling in acute myeloid leukemia
.
Blood
.
2019
;
134
(
4
):
383
-
388
.
58.
Guryanova
OA
,
Shank
K
,
Spitzer
B
, et al
.
DNMT3A mutations promote anthracycline resistance in acute myeloid leukemia via impaired nucleosome remodeling
.
Nat Med
.
2016
;
22
(
12
):
1488
-
1495
.
59.
Bezerra
MF
,
Lima
AS
,
Piqué-Borràs
MR
, et al
.
Co-occurrence of DNMT3A, NPM1, FLT3 mutations identifies a subset of acute myeloid leukemia with adverse prognosis
.
Blood
.
2020
;
135
(
11
):
870
-
875
.
60.
Garg
S
,
Reyes-Palomares
A
,
He
L
, et al
.
Hepatic leukemia factor is a novel leukemic stem cell regulator in DNMT3A, NPM1, and FLT3-ITD triple-mutated AML
.
Blood
.
2019
;
134
(
3
):
263
-
276
.
61.
Vassiliou
GS
.
Triple-mutant AML: too clever by HLF?
Blood
.
2019
;
134
(
3
):
222
-
224
.
62.
Dovey
OM
,
Cooper
JL
,
Mupo
A
, et al
.
Molecular synergy underlies the co-occurrence patterns and phenotype of NPM1-mutant acute myeloid leukemia
.
Blood
.
2017
;
130
(
17
):
1911
-
1922
.
63.
Falini
B
,
Sportoletti
P
.
A scale of “bad” co-mutations in NPM1-driven AML
.
Blood
.
2017
;
130
(
17
):
1877
-
1879
.
64.
Grisendi
S
,
Bernardi
R
,
Rossi
M
, et al
.
Role of nucleophosmin in embryonic development and tumorigenesis
.
Nature
.
2005
;
437
(
7055
):
147
-
153
.
65.
Wang
AJ
,
Han
Y
,
Jia
N
,
Chen
P
,
Minden
MD
.
NPM1c impedes CTCF functions through cytoplasmic mislocalization in acute myeloid leukemia
.
Leukemia
.
2020
;
34
(
5
):
1278
-
1290
.
66.
Gu
X
,
Ebrahem
Q
,
Mahfouz
RZ
, et al
.
Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates
.
J Clin Invest
.
2018
;
128
(
10
):
4260
-
4279
.
67.
Pianigiani
G
,
Betti
C
,
Bigerna
B
,
Rossi
R
,
Brunetti
L
.
PU.1 subcellular localization in acute myeloid leukaemia with mutated NPM1
.
Br J Haematol
.
2020
;
188
(
1
):
184
-
187
.
68.
Dawson
MA
,
Gudgin
EJ
,
Horton
SJ
, et al
.
Recurrent mutations, including NPM1c, activate a BRD4-dependent core transcriptional program in acute myeloid leukemia
.
Leukemia
.
2014
;
28
(
2
):
311
-
320
.
69.
Argiropoulos
B
,
Humphries
RK
.
Hox genes in hematopoiesis and leukemogenesis
.
Oncogene
.
2007
;
26
(
47
):
6766
-
6776
.
70.
Alcalay
M
,
Tiacci
E
,
Bergomas
R
, et al
.
Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance
.
Blood
.
2005
;
106
(
3
):
899
-
902
.
71.
Spencer
DH
,
Young
MA
,
Lamprecht
TL
, et al
.
Epigenomic analysis of the HOX gene loci reveals mechanisms that may control canonical expression patterns in AML and normal hematopoietic cells
.
Leukemia
.
2015
;
29
(
6
):
1279
-
1289
.
72.
Kühn
MW
,
Song
E
,
Feng
Z
, et al
.
Targeting chromatin regulators inhibits leukemogenic gene expression in NPM1 mutant leukemia
.
Cancer Discov
.
2016
;
6
(
10
):
1166
-
1181
.
73.
Uckelmann
HJ
,
Kim
SM
,
Wong
EM
, et al
.
Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia
.
Science
.
2020
;
367
(
6477
):
586
-
590
.
74.
Brunetti
L
,
Gundry
MC
,
Sorcini
D
, et al
.
Mutant NPM1 maintains the leukemic state through HOX expression
.
Cancer Cell
.
2018
;
34
(
3
):
499
-
512.e9
.
75.
Ghasemi
R
,
Struthers
H
,
Wilson
ER
,
Spencer
DH
.
Contribution of CTCF binding to transcriptional activity at the HOXA locus in NPM1-mutant AML cells [published online ahead of print 12 May 2020]
.
Leukemia
. doi:10.1038/s41375-020-0856-3.
76.
Oka
M
,
Mura
S
,
Otani
M
, et al
.
Chromatin-bound CRM1 recruits SET-Nup214 and NPM1c onto HOX clusters causing aberrant HOX expression in leukemia cells
.
eLife
.
2019
;
8
:
e46667
.
77.
Pasqualucci
L
,
Liso
A
,
Martelli
MP
, et al
.
Mutated nucleophosmin detects clonal multilineage involvement in acute myeloid leukemia: Impact on WHO classification
.
Blood
.
2006
;
108
(
13
):
4146
-
4155
.
78.
Falini
B
,
Macijewski
K
,
Weiss
T
, et al
.
Multilineage dysplasia has no impact on biologic, clinicopathologic, and prognostic features of AML with mutated nucleophosmin (NPM1)
.
Blood
.
2010
;
115
(
18
):
3776
-
3786
.
79.
Schnittger
S
,
Bacher
U
,
Haferlach
C
, et al
.
Characterization of NPM1-mutated AML with a history of myelodysplastic syndromes or myeloproliferative neoplasms
.
Leukemia
.
2011
;
25
(
4
):
615
-
621
.
80.
Montalban-Bravo
G
,
Kanagal-Shamanna
R
,
Sasaki
K
, et al
.
NPM1 mutations define a specific subgroup of MDS and MDS/MPN patients with favorable outcomes with intensive chemotherapy
.
Blood Adv
.
2019
;
3
(
6
):
922
-
933
.
81.
Patel
SS
,
Ho
C
,
Ptashkin
RN
, et al
.
Clinicopathologic and genetic characterization of nonacute NPM1-mutated myeloid neoplasms
.
Blood Adv
.
2019
;
3
(
9
):
1540
-
1545
.
82.
Vallapureddy
R
,
Lasho
TL
,
Hoversten
K
, et al
.
Nucleophosmin 1 (NPM1) mutations in chronic myelomonocytic leukemia and their prognostic relevance
.
Am J Hematol
.
2017
;
92
(
10
):
E614
-
E618
.
83.
Peng
J
,
Zuo
Z
,
Fu
B
, et al
.
Chronic myelomonocytic leukemia with nucleophosmin (NPM1) mutation
.
Eur J Haematol
.
2016
;
96
(
1
):
65
-
71
.
84.
Forghieri
F
,
Paolini
A
,
Morselli
M
, et al
.
NPM1 mutations may reveal acute myeloid leukemia in cases otherwise morphologically diagnosed as myelodysplastic syndromes or myelodysplastic/myeloproliferative neoplasms
.
Leuk Lymphoma
.
2015
;
56
(
11
):
3222
-
3226
.
85.
Falini
B
,
Martelli
MP
,
Bolli
N
, et al
.
Immunohistochemistry predicts nucleophosmin (NPM) mutations in acute myeloid leukemia
.
Blood
.
2006
;
108
(
6
):
1999
-
2005
.
86.
Falini
B
,
Lenze
D
,
Hasserjian
R
, et al
.
Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas
.
Leukemia
.
2007
;
21
(
7
):
1566
-
1570
.
87.
Luskin
MR
,
Huen
AO
,
Brooks
SA
, et al
.
NPM1 mutation is associated with leukemia cutis in acute myeloid leukemia with monocytic features
.
Haematologica
.
2015
;
100
(
10
):
e412
-
e414
.
88.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al
.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
89.
Straube
J
,
Ling
VY
,
Hill
GR
,
Lane
SW
.
The impact of age, NPM1mut, and FLT3ITD allelic ratio in patients with acute myeloid leukemia
.
Blood
.
2018
;
131
(
10
):
1148
-
1153
.
90.
Sakaguchi
M
,
Yamaguchi
H
,
Najima
Y
, et al
.
Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia
.
Blood Adv
.
2018
;
2
(
20
):
2744
-
2754
.
91.
Boddu
PC
,
Kadia
TM
,
Garcia-Manero
G
, et al
.
Validation of the 2017 European LeukemiaNet classification for acute myeloid leukemia with NPM1 and FLT3-internal tandem duplication genotypes
.
Cancer
.
2019
;
125
(
7
):
1091
-
1100
.
92.
Dohner
K
,
Thiede
C
,
Jahn
N
, et al
.
Impact of NPM1/FLT3-ITD genotypes defined by the 2017 European LeukemiaNet in patients with acute myeloid leukemia
.
Blood
.
2020
;
135
(
5
):
371
-
380
.
93.
Ostronoff
F
,
Othus
M
,
Lazenby
M
, et al
.
Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council report
.
J Clin Oncol
.
2015
;
33
(
10
):
1157
-
1164
.
94.
Angenendt
L
,
Röllig
C
,
Montesinos
P
, et al
.
Chromosomal abnormalities and prognosis in NPM1-mutated acute myeloid leukemia: a pooled analysis of individual patient data from nine international cohorts
.
J Clin Oncol
.
2019
;
37
(
29
):
2632
-
2642
.
95.
Eisfeld
AK
,
Kohlschmidt
J
,
Mims
A
, et al
.
Additional gene mutations may refine the 2017 European LeukemiaNet classification in adult patients with de novo acute myeloid leukemia aged <60 years [published online ahead of print 27 May 2020]
.
Leukemia
. doi:10.1038/s41375-020-0872-3.
96.
Patel
SS
,
Pinkus
GS
,
Ritterhouse
LL
, et al
.
High NPM1 mutant allele burden at diagnosis correlates with minimal residual disease at first remission in de novo acute myeloid leukemia
.
Am J Hematol
.
2019
;
94
(
8
):
921
-
928
.
97.
Abbas
HA
,
Ravandi
F
,
Loghavi
S
, et al
.
NPM1 mutant variant allele frequency correlates with leukemia burden but does not provide prognostic information in NPM1-mutated acute myeloid leukemia
.
Am J Hematol
.
2019
;
94
(
6
):
E158
-
E160
.
98.
Linch
DC
,
Hills
RK
,
Burnett
AK
,
Russell
N
,
Gale
RE
.
Analysis of the clinical impact of NPM1 mutant allele burden in a large cohort of younger adult patients with acute myeloid leukaemia
.
Br J Haematol
.
2020
;
188
(
6
):
852
-
859
.
99.
Boddu
P
,
Kantarjian
H
,
Borthakur
G
, et al
.
Co-occurrence of FLT3-TKD and NPM1 mutations defines a highly favorable prognostic AML group
.
Blood Adv
.
2017
;
1
(
19
):
1546
-
1550
.
100.
Gorello
P
,
Cazzaniga
G
,
Alberti
F
, et al
.
Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations
.
Leukemia
.
2006
;
20
(
6
):
1103
-
1108
.
101.
Bacher
U
,
Dicker
F
,
Haferlach
C
, et al
.
Quantification of rare NPM1 mutation subtypes by digital PCR
.
Br J Haematol
.
2014
;
167
(
5
):
710
-
714
.
102.
Mencia-Trinchant
N
,
Hu
Y
,
Alas
MA
, et al
.
Minimal residual disease monitoring of acute myeloid leukemia by massively multiplex digital PCR in patients with NPM1 mutations
.
J Mol Diagn
.
2017
;
19
(
4
):
537
-
548
.
103.
Jongen-Lavrencic
M
,
Grob
T
,
Hanekamp
D
, et al
.
Molecular minimal residual disease in acute myeloid leukemia
.
N Engl J Med
.
2018
;
378
(
13
):
1189
-
1199
.
104.
Ritterhouse
LL
,
Parilla
M
,
Zhen
CJ
, et al
.
Clinical validation and implementation of a measurable residual disease assay for NPM1 in acute myeloid leukemia by error-corrected next-generation sequencing
.
Mol Diagn Ther
.
2019
;
23
(
6
):
791
-
802
.
105.
Krönke
J
,
Schlenk
RF
,
Jensen
KO
, et al
.
Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia: a study from the German-Austrian acute myeloid leukemia study group
.
J Clin Oncol
.
2011
;
29
(
19
):
2709
-
2716
.
106.
Schnittger
S
,
Kern
W
,
Tschulik
C
, et al
.
Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML
.
Blood
.
2009
;
114
(
11
):
2220
-
2231
.
107.
Shayegi
N
,
Kramer
M
,
Bornhäuser
M
, et al;
Study Alliance Leukemia (SAL)
.
The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML
.
Blood
.
2013
;
122
(
1
):
83
-
92
.
108.
Balsat
M
,
Renneville
A
,
Thomas
X
, 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
(
2
):
185
-
193
.
109.
Röllig
C
,
Bornhäuser
M
,
Kramer
M
, et al
.
Allogeneic stem-cell transplantation in patients with NPM1-mutated acute myeloid leukemia: results from a prospective donor versus no-donor analysis of patients after upfront HLA typing within the SAL-AML 2003 trial
.
J Clin Oncol
.
2015
;
33
(
5
):
403
-
410
.
110.
Lussana
F
,
Caprioli
C
,
Stefanoni
P
, et al
.
Molecular detection of minimal residual disease before allogeneic stem cell transplantation predicts a high incidence of early relapse in adult patients with npm1 positive acute myeloid leukemia
.
Cancers (Basel)
.
2019
;
11
(
10
):
E1455
.
111.
Kayser
S
,
Benner
A
,
Thiede
C
, et al
.
Pretransplant NPM1 MRD levels predict outcome after allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia
.
Blood Cancer J
.
2016
;
6
(
7
):
e449
.
112.
Dillon
R
,
Hills
R
,
Freeman
S
, et al
.
Molecular MRD status and outcome after transplantation in NPM1-mutated AML
.
Blood
.
2020
;
135
(
9
):
680
-
688
.
113.
Martelli
MF
,
Di Ianni
M
,
Ruggeri
L
, et al
.
HLA-haploidentical transplantation with regulatory and conventional T-cell adoptive immunotherapy prevents acute leukemia relapse
.
Blood
.
2014
;
124
(
4
):
638
-
644
.
114.
Xue
E
,
Tresoldi
C
,
Sala
E
, et al
.
Longitudinal qPCR monitoring of nucleophosmin 1 mutations after allogeneic hematopoietic stem cell transplantation to predict AML relapse
.
Bone Marrow Transplant
.
2016
;
51
(
3
):
466
-
469
.
115.
Schuurhuis
GJ
,
Heuser
M
,
Freeman
S
, et al
.
Minimal/measurable residual disease in AML: a consensus document from the European LeukemiaNet MRD Working Party
.
Blood
.
2018
;
131
(
12
):
1275
-
1291
.
116.
Platzbecker
U
,
Middeke
JM
,
Sockel
K
, et al
.
Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial
.
Lancet Oncol
.
2018
;
19
(
12
):
1668
-
1679
.
117.
Tiong
IS
,
Dillon
R
,
Ivey
A
, et al
.
Venetoclax induces rapid elimination of NPM1 mutant measurable residual disease in combination with low-intensity chemotherapy in acute myeloid leukaemia [published online ahead of print 27 May 2020]
.
Br J Haematol
. doi:10.1111/bjh.16722.
118.
Greiner
J
,
Hofmann
S
,
Schmitt
M
, et al
.
Acute myeloid leukemia with mutated nucleophosmin 1: an immunogenic acute myeloid leukemia subtype and potential candidate for immune checkpoint inhibition
.
Haematologica
.
2017
;
102
(
12
):
e499
-
e501
.
119.
Castaigne
S
,
Pautas
C
,
Terré
C
, et al;
Acute Leukemia French Association
.
Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study
.
Lancet
.
2012
;
379
(
9825
):
1508
-
1516
.
120.
Schlenk
RF
,
Paschka
P
,
Krzykalla
J
, et al
.
Gemtuzumab ozogamicin in NPM1-mutated acute myeloid leukemia: early results from the prospective randomized AMLSG 09-09 phase III study
.
J Clin Oncol
.
2020
;
38
(
6
):
623
-
632
.
121.
Stone
RM
,
Mandrekar
SJ
,
Sanford
BL
, et al
.
Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation
.
N Engl J Med
.
2017
;
377
(
5
):
454
-
464
.
122.
Ho
AD
,
Schetelig
J
,
Bochtler
T
, et al;
Study Alliance Leukemia
.
Allogeneic stem cell transplantation improves survival in patients with acute myeloid leukemia characterized by a high allelic ratio of mutant FLT3-ITD
.
Biol Blood Marrow Transplant
.
2016
;
22
(
3
):
462
-
469
.
123.
Schlenk
RF
,
Döhner
K
,
Krauter
J
, et al;
German-Austrian Acute Myeloid Leukemia Study Group
.
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia
.
N Engl J Med
.
2008
;
358
(
18
):
1909
-
1918
.
124.
Bazarbachi
A
,
Labopin
M
,
Kharfan-Dabaja
MA
, et al
.
Allogeneic hematopoietic cell transplantation in acute myeloid leukemia with normal karyotype and isolated nucleophosmin-1 (NPM1) mutation: outcome strongly correlates with disease status
.
Haematologica
.
2016
;
101
(
1
):
e34
-
e37
.
125.
Poiré
X
,
Labopin
M
,
Polge
E
, et al
.
Hematopoietic stem cell transplantation for adult patients with isolated NPM1 mutated acute myeloid leukemia in first remission
.
Am J Hematol
.
2019
;
94
(
2
):
231
-
239
.
126.
Becker
H
,
Marcucci
G
,
Maharry
K
, et al
.
Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a Cancer and Leukemia Group B study
.
J Clin Oncol
.
2010
;
28
(
4
):
596
-
604
.
127.
Scholl
S
,
Theuer
C
,
Scheble
V
, et al
.
Clinical impact of nucleophosmin mutations and Flt3 internal tandem duplications in patients older than 60 yr with acute myeloid leukaemia
.
Eur J Haematol
.
2008
;
80
(
3
):
208
-
215
.
128.
Lachowiez
CA
,
Loghavi
S
,
Kadia
TM
, et al
.
Outcomes of older patients with NPM1-mutated AML: current treatments and the promise of venetoclax-based regimens
.
Blood Adv
.
2020
;
4
(
7
):
1311
-
1320
.
129.
Aldoss
I
,
Nakamura
R
,
Yang
D
, et al
.
Favorable outcomes for allogeneic hematopoietic cell transplantation in elderly patients with NPM1-mutated and FLT3-ITD-negative acute myeloid leukemia
.
Bone Marrow Transplant
.
2020
;
55
(
2
):
473
-
475
.
130.
Jentzsch
M
,
Grimm
J
,
Bill
M
, et al
.
Outcomes of older patients with NPM1 mutated and FLT3-ITD negative acute myeloid leukemia receiving allogeneic transplantation
.
HemaSphere
.
2020
;
4
(
1
):
e326
.
131.
Prata
PH
,
Bally
C
,
Prebet
T
, et al
.
NPM1 mutation is not associated with prolonged complete remission in acute myeloid leukemia patients treated with hypomethylating agents
.
Haematologica
.
2018
;
103
(
10
):
e455
-
e457
.
132.
DiNardo
CD
,
Pratz
K
,
Pullarkat
V
, et al
.
Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia
.
Blood
.
2019
;
133
(
1
):
7
-
17
.
133.
DiNardo
CD
,
Tiong
IS
,
Quaglieri
A
, et al
.
Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML
.
Blood
.
2020
;
135
(
11
):
791
-
803
.
134.
National Cancer Research Institute
. Recommendations for the management of patients with AML during the COVID19 outbreak: a statement from the NCRI AML Working Party. https://www.rcpathorg/uploads/assets/d23030b6-7379-4f80-9ed9178c5f864343/Recommendations-for-the-management-of-patients-with-acute-myeloid-leukaemia-AML-during-the-COVID19-outbreakpdf. Accessed 10 April 2020.
135.
Falini
B
,
Gionfriddo
I
,
Cecchetti
F
,
Ballanti
S
,
Pettirossi
V
,
Martelli
MP
.
Acute myeloid leukemia with mutated nucleophosmin (NPM1): any hope for a targeted therapy?
Blood Rev
.
2011
;
25
(
6
):
247
-
254
.
136.
Kırlı
K
,
Karaca
S
,
Dehne
HJ
, et al
.
A deep proteomics perspective on CRM1-mediated nuclear export and nucleocytoplasmic partitioning
.
eLife
.
2015
;
4
:
e11466
.
137.
Uckelmann
HJ
,
Armstrong
SA
,
Stone
RM
.
Location, location, location: mutant NPM1c cytoplasmic localization is required to maintain stem cell genes in AML
.
Cancer Cell
.
2018
;
34
(
3
):
355
-
357
.
138.
Garzon
R
,
Savona
M
,
Baz
R
, et al
.
A phase 1 clinical trial of single-agent selinexor in acute myeloid leukemia
.
Blood
.
2017
;
129
(
24
):
3165
-
3174
.
139.
Brunetti
L
,
Gundry
MC
,
Goodell
MA
.
New insights into the biology of acute myeloid leukemia with mutated NPM1
.
Int J Hematol
.
2019
;
110
(
2
):
150
-
160
.
140.
Etchin
J
,
Berezovskaya
A
,
Conway
AS
, et al
.
KPT-8602, a second-generation inhibitor of XPO1-mediated nuclear export, is well tolerated and highly active against AML blasts and leukemia-initiating cells
.
Leukemia
.
2017
;
31
(
1
):
143
-
150
.
141.
Klossowski
S
,
Miao
H
,
Kempinska
K
, et al
.
Menin inhibitor MI-3454 induces remission in MLL1-rearranged and NPM1-mutated models of leukemia
.
J Clin Invest
.
2020
;
130
(
2
):
981
-
997
.
142.
Gundry
MC
,
Goodell
MA
,
Brunetti
L
.
It’s all about MEis: menin-MLL inhibition eradicates NPM1-mutated and MLL-rearranged acute leukemias in mice
.
Cancer Cell
.
2020
;
37
(
3
):
267
-
269
.
143.
Yang
K
,
Wang
M
,
Zhao
Y
, et al
.
A redox mechanism underlying nucleolar stress sensing by nucleophosmin
.
Nat Commun
.
2016
;
7
(
1
):
13599
.
144.
Falini
B
,
Brunetti
L
,
Martelli
MP
.
Dactinomycin in NPM1-mutated acute myeloid leukemia
.
N Engl J Med
.
2015
;
373
(
12
):
1180
-
1182
.
145.
Burger
K
,
Mühl
B
,
Harasim
T
, et al
.
Chemotherapeutic drugs inhibit ribosome biogenesis at various levels
.
J Biol Chem
.
2010
;
285
(
16
):
12416
-
12425
.
146.
Balusu
R
,
Fiskus
W
,
Rao
R
, 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
(
11
):
3096
-
3106
.
147.
Liso
A
,
Colau
D
,
Benmaamar
R
, et al
.
Nucleophosmin leukaemic mutants contain C-terminus peptides that bind HLA class I molecules
.
Leukemia
.
2008
;
22
(
2
):
424
-
426
.
148.
Narayan
R
,
Olsson
N
,
Wagar
LE
, et al
.
Acute myeloid leukemia immunopeptidome reveals HLA presentation of mutated nucleophosmin
.
PLoS One
.
2019
;
14
(
7
):
e0219547
.
149.
van der Lee
DI
,
Reijmers
RM
,
Honders
MW
, et al
.
Mutated nucleophosmin 1 as immunotherapy target in acute myeloid leukemia
.
J Clin Invest
.
2019
;
129
(
2
):
774
-
785
.
150.
Greiner
J
,
Hofmann
S
,
Schneider
V
, et al
.
Mutated regions of nucleophosmin 1 (NPM1) elicit both CD4+ and CD8+ T cell responses in patients with acute myeloid leukemia (AML)
.
Onkologie
.
2012
;
35
:
58
.
151.
Kuželová
K
,
Brodská
B
,
Fuchs
O
,
Dobrovolná
M
,
Soukup
P
,
Cetkovský
P
.
Altered HLA class I profile associated with type A/D nucleophosmin mutation points to possible anti-nucleophosmin immune response in acute myeloid leukemia
.
PLoS One
.
2015
;
10
(
5
):
e0127637
.
152.
Hofmann
S
,
Götz
M
,
Schneider
V
, et al
.
Donor lymphocyte infusion induces polyspecific CD8(+) T-cell responses with concurrent molecular remission in acute myeloid leukemia with NPM1 mutation
.
J Clin Oncol
.
2013
;
31
(
3
):
e44
-
e47
.
153.
Greiner
J
,
Schneider
V
,
Schmitt
M
, et al
.
Immune responses against the mutated region of cytoplasmatic NPM1 might contribute to the favorable clinical outcome of AML patients with NPM1 mutations (NPM1mut)
.
Blood
.
2013
;
122
(
6
):
1087
-
1088
.
154.
Tyner
JW
,
Tognon
CE
,
Bottomly
D
, et al
.
Functional genomic landscape of acute myeloid leukaemia
.
Nature
.
2018
;
562
(
7728
):
526
-
531
.
155.
Zhu
HH
,
Qian
JJ
,
Sun
WJ
, et al
.
Venetoclax and arsenic showed synergistic anti-leukemia activity in vitro and in vivo for acute myeloid leukemia with the NPM1 mutation
.
Am J Hematol
.
2020
;
95
(
3
):
E55
-
E57
.
156.
Fischer
MA
,
Friedlander
SY
,
Arrate
MP
, et al
.
Venetoclax response is enhanced by selective inhibitor of nuclear export compounds in hematologic malignancies
.
Blood Adv
.
2020
;
4
(
3
):
586
-
598
.
Sign in via your Institution