Next Article in Journal
A Fungicide, Fludioxonil, Formed the Polyploid Giant Cancer Cells and Induced Metastasis and Stemness in MDA-MB-231 Triple-Negative Breast Cancer Cells
Previous Article in Journal
Relation of T Cell Profile with Vitamin D Receptor and Vitamin D-Binding Protein Gene Polymorphisms in Atopy
Previous Article in Special Issue
Monocytic Differentiation in Acute Myeloid Leukemia Cells: Diagnostic Criteria, Biological Heterogeneity, Mitochondrial Metabolism, Resistance to and Induction by Targeted Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

KMT2A Rearrangements in Leukemias: Molecular Aspects and Therapeutic Perspectives

by
Luca Guarnera
1,2,
Matteo D’Addona
1,
Carlos Bravo-Perez
1,3 and
Valeria Visconte
1,*
1
Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44114, USA
2
Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
3
Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, CIBERER—Instituto de Salud Carlos III, University of Murcia, IMIB-Pascual Parrilla, 30005 Murcia, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 9023; https://doi.org/10.3390/ijms25169023
Submission received: 15 July 2024 / Revised: 14 August 2024 / Accepted: 15 August 2024 / Published: 20 August 2024
(This article belongs to the Special Issue Molecular Mechanism of Leukemia 2.0)

Abstract

:
KMT2A (alias: mixed-lineage leukemia [MLL]) gene mapping on chromosome 11q23 encodes the lysine-specific histone N-methyltransferase 2A and promotes transcription by inducing an open chromatin conformation. Numerous genomic breakpoints within the KMT2A gene have been reported in young children and adults with hematologic disorders and are present in up to 10% of acute leukemias. These rearrangements describe distinct features and worse prognosis depending on the fusion partner, characterized by chemotherapy resistance and high rates of relapse, with a progression-free survival of 30–40% and overall survival below 25%. Less intensive regimens are used in pediatric patients, while new combination therapies and targeted immunotherapeutic agents are being explored in adults. Beneficial therapeutic effects, and even cure, can be reached with hematopoietic stem cell transplantation, mainly in young children with dismal molecular lesions; however, delayed related toxicities represent a concern. Herein, we summarize the translocation partner genes and partial tandem duplications of the KMT2A gene, their molecular impact, clinical aspects, and novel targeted therapies..

1. Introduction

Lysine-specific N-methyltransferase 2A (KMT2A), also known as mixed-lineage leukemia (MLL), is the target of balanced rearrangements. Two breakpoint cluster regions have been identified: a major region between intron 7 and exon 13, and a minor region between intron 20 and exon 24. These rearrangements are very diverse and, due to the heterogeneous nature of the fusion partners, therapeutic decision is difficult. Patients with these genomic lesions are characterized by chemotherapy resistance and high rates of relapse after standard therapies. The genomic sequencing of breakpoints has also been used to monitor measurable residual disease [1]. Typically, these rearrangements are associated with poor prognosis, likely due to the marked epigenetic changes induced by the lesions, with fusions displaying an abnormal increase in H3K79 methylation. The rearrangements usually impact transcriptional regulatory genes, including homeobox A cluster (HOXA), which are linked to leukemia. Recently, acute myeloid leukemia (AML) with rearrangement of the nucleoporin 98 (NUP98) gene has been found to be sensitive to menin inhibitors, which hold promise to play an important role in this adverse disease subgroup (Figure 1).
This review provides an analysis of all KMT2A rearrangements in AML patients, describing their specific prognostic outcomes in different age ranges. We conducted an extensive narrative review of the literature, focusing on clinical approaches among the chemotherapies available, the role of transplant, and new therapeutic strategies using menin inhibitors.

2. Biology of Rearrangements

Rearrangements involving KMT2A and its fusion partner genes are found in precursor B-cell acute lymphoblastic leukemia (B-ALL), T-ALL, AML, myelodysplastic neoplasm (MDS), mixed-lineage (biphenotypic) leukemia (MPAL), and secondary leukemia [1,2,3]. Using the next-generation sequencing (NGS) of primary AML specimens, Lavallée and colleagues identified the first unifying genetic network of KMT2A. Two subgroups of KMT2A-rearranged leukemia have been described: ones characterized by translocations with other genes, and one characterized by partial tandem duplications (PTDs) [4].
The first one accounts for 5−10% of adult AML patients [5], while PTD rearrangements are found in an additional 5–6% of AML patients [6]. Study done by Meyer et al. in 2023 showed data obtained from 3401 acute leukemia patients using LDI-PCR technology and targeted NGS [1]. All the identified rearrangements were characterized by many reciprocal chromosomal translocations and, to a lesser extent, also by spliced fusions, inversions, deletions, insertions, and PTD. AML patients displayed a more heterogenous landscape of partner genes (Table 1). Among them, the most frequent one is MLLT3 (30.4%). About 84.8% of all AML patients are characterized by seven fusion genes: KMT2A::MLLT3, KMT2A::MLLT10, KMT2A-PTD, KMT2A::ELL, KMT2A::AFDN, KMT2A::MLLT1, and KMT2A::MLLT11. To improve the accurate identification of complex KMT2A gene rearrangements, considerations could be given to the incorporation of multiplex ligation probe amplification (MLPA), optical genome mapping, or third-generation long-read sequencing into conventional molecular diagnostic algorithms based on short-read NGS [7,8,9].
Regarding the breakpoint distribution of the KMT2A recombinome, this configuration almost invariably occurs in the major breakpoint cluster region 1 (BCR1) mapping between KMT2A intron 7 and KMT2A exon 13.
In terms of molecular associations, co-mutational analysis reveals a lower mutational rate than AML with a normal karyotype, with an enrichment of mutations in genes of the RAS pathway (e.g., KRAS, NRAS, and PTPN11, up to 30%) and FLT3 (8–14%). KRAS mutations occurred more often in patients with t(6;11) (q27;q23)/KMT2A::AFDN compared with patients with other 11q23/KMT2A subsets [10].
This holds also true for KMT2A::EDC4, a new breakpoint fusion gene spanning exon 7 of KMT2A and exon 5 of EDC4 gene, involving DNA damage response and repair [11]. Along with the involvement of KRAS mutations, some studies have shown that subclonal-activating mutations, for instance FLT3N676K, accelerate KMT2A-MLLT3 leukemia onset [12]. In relation to KMT2A::AFF1 (MLL::AF4) fusions, it has been reported that specific alterations in circRNA expression might be oncogenic or sustain the oncogenic properties of chimeric proteins. However, these findings require further investigation [13].

3. Clinical Aspects

Overall, KMT2A rearrangements are present in up to 10% of acute leukemias. However, the array of disease subtypes embodying this genetic abnormality is remarkable, and distinctive clinical profiles can be identified. According to the age at presentation, KMT2A-rearranged leukemias represent 80% of cases in infants (<1 of age) and 5–15% of cases during childhood and adulthood, including ALL, AML, MPAL, and MDS/secondary AML [14,15,16,17]. As specific diagnostic entities with recurrent genetic abnormalities, B-ALL/lymphoblastic lymphoma and AML with KMT2A rearrangements are categories included in the latest classification of the World Health Organization (5th WHO) and the International Consensus Classification (2022 ICC) [18,19,20]. Nevertheless, regardless of disease subtype, the prognosis of KMT2A-rearranged leukemias is generally poor, with a progression-free survival (PFS) of 30–40% and an overall survival (OS) below 25% [14,15,16,17].

3.1. Incidence, Clinical Features, and Prognosis in Infancy and Childhood

There is a dramatic incidence of KMT2A rearrangements in infants, being present in 80–90% of acute leukemias. Most of them are B-ALL (80–90%), followed by T-ALL, AML (up to 5–10% each), and other disease subtypes (<5%) [21]. Molecularly, the most frequent fusion partners are AFF1, MLLT1, and MLLT3 [1]. Infant-onset B-ALL with KMT2A rearrangement normally exerts a very immature phenotype (pro-B) with the frequent aberrant co-expression of myeloid markers. Patients normally present with hyperleukocytosis and extramedullary disease with central nervous system involvement, and have poor prognosis [22]. Interestingly, another 20% of infant ALL cases are associated with germline KMT2A rearrangements. These post-natal cases, unlike the first ones, are B-ALL with a more mature immunophenotype, and appear to have a better clinical outcome. Genetic analysis has shown an enrichment of germline KMT2A::NUTM1 in these cases, representing a particularly low-risk group with a PFS of 75–95% [23,24,25,26].

3.2. Incidence, Clinical Features, and Prognosis in Adulthood

During the juvenile period/adulthood, the incidence of KMT2A rearrangement significantly decreases, and at 50–60 years of age slightly increases again. Overall, it is present in 5–15% of leukemias. Both in children and adults, 50–60% of KMT2A-rearranged cases are ALL, 40% are AML, and <10% are other disease subtypes. Overall, the top three frequent fusion partners are again AFF1, MLLT1, and MLLT3 [1]. However, they significantly differ between ALL and AML. In KMT2A-rearranged AML, the most prevalent fusion partner is MLLT3, followed by MLLT10, AFDN, ELL, MLLT1, and MLLT11. Additionally, in adults, KMT2A-PTD is a characteristic lesion that is present in up to 10% of cases [1].
Clinically, KMT2A-rearranged AML patients often present with high blast counts and monocytic or myelomonocytic differentiation. Analysis across fusions reveals that KMT2A::MLLT3 and KMT2A::MLLT10 usually have lower blast counts versus the other cases and, particularly in children, megakaryoblastic differentiation. KMT2A-rearranged AML is clearly associated with inferior outcomes in several clinical series, including refractory disease, poor PFS and OS, and higher risks of coagulopathy, bleeding, and early death than AML with a normal karyotype [10,27].
The analysis of large cohorts integrating molecular data suggests that fusion partners are of prognostic value, even though some studies do not confirm differences between them. KMT2A::MLLT3 vs. other fusions seems to be associated with relatively better survival outcomes, both in children (PFS 50% vs. 14–46% in other fusions; OS: 63% vs. 22–47% in other fusions) and in adults (PFS 47% vs. 13% in other fusions; OS: 46% vs. 11% in other fusions) [16,27,28]. KMT2A::AFDN and KMT2A::MLLT10 are among the rearrangements with the poorest prognosis, with an OS of 30% in children and 5–30% in adults. In a large series of pediatric patients, KMT2A::MLLT11 showed a significantly more favorable outcome, with a PFS of 92% and OS of 100% [16]. Based on this, the 2017 European Leukemia Net (2017ELN) stratified KMT2A rearrangements into KMT2A::MLLT3, of intermediate prognosis, and other fusions, of adverse prognosis [29,30]. Similarly, the 2022ICC, within AML and related entities, incorporated different categories of KMT2A-rearranged AML, including AML with KMT2A::MLLT3, AML with other KMT2A rearrangements, and MPAL with KMT2A rearranged [20].
Other conventional factors determining clinical outcomes in KMT2A-rearranged AML are diverse and include the diagnosis of therapy-related disease, primary chemoresistance (adverse factor), age at diagnosis < 60 years, frontline treatment, and allogeneic hematopoietic stem cell transplantation (HSCT) after first remission (favorable factor) [27]. Different studies have explored the role of co-mutations in refining the aggressiveness of the disease without finding a definitive predictive effect, probably because of the heterogeneity of these studies [10,27].
Distinctive clinical associations of KMT2A rearrangements in myeloid neoplasms rather than de novo or relapsed AML also include lymphoid-to-myeloid lineage switching among B-ALL patients treated with B-cell-targeted immunotherapy (up to 60% with KMT2A rearrangements); [29] therapy-related myeloid neoplasms, in particular in patients treated with topoisomerase II inhibitors [6,30]; and MDS/secondary AML, in which KMT2A-PTD has been described in up to 7–10% of cases [30]. All these entities, despite having clinical, genetic, and mechanistic idiosyncrasies, are unified by a dismal prognosis [30]

4. Clinical Management of KMT2A-Rearranged AML and Current Clinical Practice

In recent years, new, promising, chemo-free approaches have been tested, with mixed outcomes in AML and MDS, enlarging therapeutic opportunities and allowing for tailored treatment strategies [31]. In this area, due to the not-negligible incidence and prognostic weight of KMT2A-rearranged AML, several drugs and strategies targeting this entity have been investigated.
Currently there is no specific therapy approved to target KMT2A rearrangements and, in clinical practice, treatment strategies are based on canonical risk stratification [32].

4.1. Chemotherapy

Bill et al. [10] described 172 patients with KMT2A-rearranged AML, classified as intermediate-risk KMT2A::MLLT3 or as having adverse-risk rearrangements different from KMT2A::MLLT3, according to the ELN2017 classification [33]. The patients were similarly treated with Cancer and Leukemia Group B (CALGB)/Alliance for Clinical Trials in Oncology (Alliance) protocols. Overall, 68% of patients obtained complete remission (CR), with a 3-year disease-free survival (DFS) of 29% and an OS of 26%. Patients harboring KMT2A::MLLT3 presented the highest OS (3-year OS 41%) and DFS (3-year DFS 47%) when compared to other KMT2A rearrangements. Pollard et al., in a sub-analysis of the Children’s Oncology Group (COG) Trial AAML0531, investigated the administration of Gemtuzumab Ozogamicin (GO), an anti-CD33 antibody–calicheamicin conjugate, in combination with conventional chemotherapy; 215 pediatric patients with KMT2A-rearranged AML showed an advantage in terms of 5-year OS and 5-year event-free survival (EFS) as a result of receiving GO (63% vs. 53%, and 48% vs. 29%, respectively). Furthermore, patients receiving CR showed lower relapse rates in the GO arm (40% vs. 66%) and improved 5-year DFS (57% vs. 33%). The advantage was consistent across risk classes and GO administration was independently associated with improved EFS, DFS, and reduced relapse rates [34]. However, these results were not confirmed in the adult population by the UK group by Dillon et al., who found no clear benefit of GO in this subset of patients [35].

4.2. Less Intensive Regimens

Ball et al. investigated the impact of venetoclax in combination with hypomethylating agents. The overall response rate was 83% in newly diagnosed AML (12 patients) and 17% in relapsed/refractory AML (12 patients), with a median OS of 11 months in the first group and 6 months in the second one [36].

4.3. Hematopoietic Stem Cell Transplantation

Several authors have explored the role of HSCT in KMT2A-rearranged AML. In 2015, a study by the Acute Leukemia Working Party EBMT showed that 2- and 3-year OS and DFS were 56% and 51%, and 51% and 47%, respectively. Furthermore, KMT2A::MLLT3 rearrangement was confirmed to be the most common and the one with better prognosis [37]. Recently, Jiang et al. reported similar results analyzing a cohort of 42 patients, with a 2-year OS and DFS of 59.1% and 49.6%, respectively [38]. In a pediatric setting, Miyamura et al. observed a 3-year OS and DFS of 52.1% and 46.7%, respectively [39]. Menghrajani et al. analyzed a large population of KMT2A-rearranged AML patients undergoing HSCT and observed rates of OS and EFS similar to those of adverse-risk AML with no significant differences across the rearrangement partners [40]. Tong et al. focused on a small cohort of 22 patients with KMT2A::MLLT3 rearrangement in CR undergoing umbilical-cord HSCT and observed a 3-year OS and DFS of 71.3% and 60.8%, respectively (significantly higher than in patients not undergoing HSCT) [41]. Finally, Antherieu et al. analyzed a cohort of KMT2A-PTD AML patients and found that those receiving HSCT presented a significantly improved outcome. In the multivariable analysis, HSCT and FLT3-ITD status were the only independent variables associated with OS [42].

5. Menin Inhibitors

Menin inhibitors find their biological background in the interaction between KMT2A and menin. From the identification of this pathogenetic mechanism in 2005 by Yokoyama et al. [43], ten years passed to the development of the first menin inhibitors, MI-463 and MI-503, developed by Borkin and colleagues in 2015 and successfully tested in AML mouse models harboring MLL rearrangements, granting a survival benefit without impairing normal hemopoiesis [44]. These results were confirmed in patient-derived xenografts treated with two small molecules (VTP50469 and MI-3454) belonging to the same class [45,46], showing an on-target mechanism of action and strong antileukemic activity, prolonging survival without toxicity.

5.1. Revumenib

The first, recently published, phase I clinical trial (AUGMENT-101 (NCT04065399)) on revumenib (previously known as SNDX-5613), a selective oral inhibitor of the menin–KMT2A interaction, investigated the effect of this drug in R/R AML. In total, 68 heavily pre-treated (median of four previous lines of therapy) patients were enrolled, of whom 46 had KMT2A rearrangements. Grade III or higher treatment-related adverse events were observed in 16% of patients and 16% patients presented differentiation syndrome (all grade II). The trial protocol included an RNA-seq analysis that highlighted a downregulation of the critical leukemogenic targets MEIS1, homeobox A9 (HOXA9), pre-B-cell leukemia transcription factor 3 (PBX3), and cyclin-dependent kinase 6 (CDK6), and an increase in the expression of genes associated with differentiation, such as integrin alpha M (CD11b) and CD14. In the subset of KMT2A-rearranged AML, an overall response rate (ORR) of 59% and a CR [including CR with incomplete hematologic recovery (CRh)] of 33% were achieved. The median OS of all the population was 7 months [47]. Intriguingly, the analysis of patients with acquired resistance to menin inhibition revealed somatic mutations in MEN1 after revumenib/menin treatment. These findings were confirmed in xenograft models and proved that a chromatin-targeting strategy exerts sufficient selection pressure to drive the evolution of escape mutants as a mechanism of therapeutic resistance [48].

5.2. Ziftomenib

Another ongoing phase I-II trial, KOMET-001 (NCT04067366), is investigating another oral menin inhibitor, ziftomenib (KO-539). In the 2022 update, 30 R/R AML patients were enrolled, of whom 24 presented an NPM1 mutation or KMT2A rearrangements. The dose-finding study showed a 600 mg dose as feasible and effective, obtaining an ORR of 42%, a CR/CRh of 25%, and a CR/CR with incomplete hematologic recovery (CRi) of 33.3% [49].

5.3. Other Menin Inhibitors

75276617ALE1001 (NCT04811560) is an ongoing phase 1 trial investigating JNJ-75276617, an oral menin inhibitor. In the 2023 update, 56 patients with R/R AML were enrolled, 57% with KMT2A rearrangements. In the dose-finding study, among the eight patients treated at the highest dose (900 mg × 2/die), four obtained an ORR (two presented KMT2A rearrangements) [50]. In the last ASH annual meeting, the results of a phase I/II trial investigating the menin–MLL inhibitor DSP-5336 in R/R AML patients were presented; out of the six patients enrolled with R/R AML and KMT2A rearrangements, one patient achieved CRi, one patient achieved a morphologic leukemia-free state, and one achieved stable disease with the clearance of peripheral blasts, the recovery of peripheral counts, the resolution of leukemic gingival infiltration, and a reduction in bone marrow blasts from 85% to 31% [51].

6. Experimental Data of Combination Therapy

The rationale of the use of menin inhibitor paired with preclinical studies has shown the importance of potential drug partners.
Fleischmann and colleagues tested revumenib in combination with tamibarotene, an RAR alpha agonist, in cell lines and patient-derived AML cells. The authors observed a significant and synergistic pro-apoptotic effect in MV4-11 cells carrying translocation t(4;11), resulting in KMT2A::AFF1 and OCI-AML3 cells (NPM1-mutated cells). Furthermore, MOLM13 cells carrying a cryptic insertion (11;9), resulting in KMT2A-MLLT3 and OCI-AML3 cells, showed an increased expression of CD11b. Of note is that, in HL-60 cells (acute promyelocytic leukemia, not carrying NPM1 mutations or KMT2Ar), no apoptotic or differentiating effect was observed. Primary AML cells from five patients carrying KMT2Ar were analyzed. Among these samples, revumenib–tamibarotene combination was effective in inducing apoptosis and in increasing CD11b expression in one patient-derived cell line [carrying t(11;19)], and in another one [carrying t(9;11)], CD11b expression increased without obvious apoptotic effects [52].
Ling et al. observed that AML cell lines characterized by KMT2Ar (MV4-11, MOLM13, THP-1) were sensitive to venetoclax, a BCL2-inhibitor. This finding inspired the authors to test venetoclax in combination with MI-503. The combined therapy exerted a synergistic, selective pro-apoptotic effect in KMT2Ar-AML cell lines. The treatment was also effective in reducing the proportion of leukemic cells and in prolonging survival in cell-derived MOLM13 xenografts. Transcriptomic analysis revealed that (a) the histone deacetylases 9 (HDAC9), but not other types of HDACs, was consistently down-regulated in KMT2Ar-AML; (b) HDAC9 inhibition with the HDAC inhibitor TMP-269 impaired AML proliferation; and (c) HDAC9 knockout did not enhance MI-503–venetoclax efficacy. Moreover, RNA expression analysis shed light on the mechanism of action of the combination therapy, which resulted in the inhibition of the hypoxia pathway. Finally, hypoxia-inducing experiments showed that MI-503–venetoclax down-regulated HDAC9 by suppressing HIF-1A [53].
In the same line, Ye and colleagues investigated the synergistic effect of chidamide (HDAC inhibitor) with MI-3, a menin inhibitor, showing a strong synergistic effect suppressing the growth of KMT2Ar cells (MOLM-13, MV4-11). Colony-formation assays and flow cytometry revealed that chidamide interacts synergistically with MI-3 via the generation of reactive oxygen species (ROS) production and mitochondrial damage. The authors then further investigated the mechanism of action of the combination therapy, observing differently regulated genes associated with survival and cytokine signaling pathways in MOLM-13 cells treated with MI-3 and chidamide alone or in a combination regimen. Furthermore, the gene set enrichment analysis suggested that interference with the DNA repair machinery might contribute to the synergistic interaction between chidamide and MI-3. These findings were confirmed in MOLM13 cell xenograft models [54].
The potential of combinations with venetoclax was also explored by Fiskus and colleagues. The authors observed an inhibition of Bcl2 protein levels and Cdk6 following SNDX-50469 (menin inhibitor) treatment. In addition, they investigated the use of respective inhibitors (venetoclax and abemaciclib) in inhibiting the residual levels of these proteins, finding a strong synergistic activity of each drug with SNDX-50469 on both cell lines (MV4-11, MOLM13, OCI-AML3) and patient-derived AML cells expressing KMT2Ar or NPM1 mutation. The venetoclax–SNDX-50469 combination was also effective in xenograft mice bearing KMT2Ar or NPM1 mutation with mutated FLT3 [55].
Aubrey and colleagues, using functional genetic screens, identified a key role of IKZF1/IKAROS, a transcription factor, in maintaining leukemia genes’ expression and repressing tumor suppressor genes in KMT2Ar AML. Thus, the authors successfully tested immunomodulatory drugs (IMiDs), such as thalidomide and newer-generation lenalidomide, pomalidomide, and iberdomide, able to induce IKAROS degradation, in both NPM1-mutated and KMT2Ar cell lines. The lenalidomide–VTP-50469 combination displayed synergy in MV411 and MOLM13 cell lines, with the response correlated to the potency of IKAROS degradation. The combination therapy was also confirmed to be effective in KMT2Ar- and NPM1-mutated AML-patient-derived xenograft models, leading to rapid leukemia cell clearance and the differentiation of myelomonocytic lineage [56].
These findings have recently been paired with the research by Bourgeois et al., who successfully tested, in KMT2Ar- and NPM1-mutated AML models, mezigdomide (cereblon E3 ligase modulator), which was able to degrade IKAROS more efficiently (increased depth, rate, and duration of protein degradation) than lenalidomide and iberdomide. Furthermore, the authors showed that the combination of mezigdomide with VTP-50469 increased survival and overcame the MEN1 mutations responsible for resistance in patients receiving menin inhibitor monotherapy [57].
Thus, based on the promising data of early trials and in vitro and in vivo experiments on therapy combination, several clinical trials are currently investigating the association between menin inhibitors and chemotherapy [AUGMENT-102 (NCT05326516), KOMET-007 (NCT05735184)], as well as venetoclax and/or hypomethylating agents [SAVE (NCT05360160), NCT03013998, KOMET-007 (NCT05735184), NCT04752163]. Intriguingly, the gene expression profiles of KMT2A-PTD AML and of HOXA genes show similarities with NPM1-mutated and KMT2A-rearranged AML, suggesting a role of menin inhibitors in this subset of patients [58]. Table 2 summarizes several studies highlighting outcomes of patients with KMT2A rearrangements after treatment with pharmacologic agents and/or HSCT.

7. Conclusions

The analysis of KMT2A rearrangements in AML patients shows diverse prognostic outcomes based on age range. In the last few years, advances in biology have broadened our knowledge on the rearrangement partners and the molecular mechanisms of leukemogenesis in KMT2A anomalies of AML. The worldwide availability of next-generation sequencing and the introduction of alternative antileukemic treatment strategies (e.g., new drugs and broader indications for HSCT) in clinical practice have led to the identification of an intermediate-risk subtype, KMT2A::MLLT3, and adverse-risk ones [rearrangements different from t(9;11)] which have recently been added to the newest ELN risk stratification.
The promising results obtained by the first phase I trial investigating revumenib have prompted the design of new clinical trials with different molecules and drug combinations, possibly mimicking the path of other successful predecessors among smart drugs, such as all-trans retinoic acid and arsenic trioxide for acute promyelocytic leukemia [59] and FLT3 inhibitors for FLT3 mutant AML [60], leading to a change in the paradigms of the treatment and prognosis of the disease. Novel menin inhibitors are being tested for KMT2A conditions.

Author Contributions

Conceptualization, figure generation, writing—review and editing: L.G., M.D., C.B.-P. and V.V. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank AAMDSIF (to C.B.-P. and V.V.). C.B-P. has a postdoctoral fellowship from Instituto of de Salud Carlos III (JR22/00041).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Meyer, C.; Larghero, P.; Almeida, L.B.; Burmeister, T.; Gröger, D.; Sutton, R. The KMT2A recombinome of acute leukemias in 2023. Leukemia 2023, 37, 988–1005. [Google Scholar] [CrossRef] [PubMed]
  2. Górecki, M.; Kozioł, I.; Kopystecka, A.; Budzyńska, J.; Zawitkowska, J.; Lejman, M. Updates in KMT2A Gene Rearrangement in Pediatric Acute Lymphoblastic Leukemia. Biomedicines 2023, 11, 821. [Google Scholar] [CrossRef]
  3. Larson, J.K.; Hunter-Schlichting, D.N.; Crowgey, E.L.; Mills, L.J.; Druley, T.E.; Marcotte, E.L. KMT2A-D pathogenicity, prevalence, and variation according to a population database. Cancer Med. 2023, 12, 7234–7245. [Google Scholar] [CrossRef] [PubMed]
  4. Lavallée, V.P.; Baccelli, I.; Krosl, J.; Wilhelm, B.; Barabé, F.; Gendron, P. The transcriptomic landscape and directed chemical interrogation of MLL-rearranged acute myeloid leukemias. Nat. Genet. 2015, 47, 1030–1037. [Google Scholar] [CrossRef] [PubMed]
  5. Krivtsov, A.V.; Armstrong, S.A. MLL translocations, histone modifications and leukaemia stem-cell development. Nat. Rev. Cancer 2007, 7, 823–833. [Google Scholar] [CrossRef] [PubMed]
  6. Steudel, C.; Wermke, M.; Schaich, M.; Schäkel, U.; Illmer, T.; Ehninger, G. Comparative analysis of MLL partial tandem duplication and FLT3 internal tandem duplication mutations in 956 adult patients with acute myeloid leukemia. Genes Chromosomes Cancer 2003, 37, 237–251. [Google Scholar] [CrossRef] [PubMed]
  7. Seto, A.; Downs, G.; King, O.; Salehi-Rad, S.; Baptista, A.; Chin, K. Genomic Characterization of Partial Tandem Duplication Involving the KMT2A Gene in Adult Acute Myeloid Leukemia. Cancers 2024, 26, 1693. [Google Scholar] [CrossRef] [PubMed]
  8. Sedlazeck, F.J.; Rescheneder, P.; Smolka, M.; Fang, H.; Nattestad, M.; von Haeseler, A. Accurate detection of complex structural variations using single-molecule sequencing. Nat. Methods 2018, 15, 461–468. [Google Scholar] [CrossRef] [PubMed]
  9. Bravo-Perez, C.; Cifuentes-Riquelme, R.; Padilla, J.; de la Morena-Barrio, M.E.; Ortuño, F.J.; Garrido-Rodríguez, P. The whole is greater than the sum of its parts: Long-read sequencing for solving clinical problems in haematology. J. Cell Mol. Med. 2024, 28, e17961. [Google Scholar] [CrossRef]
  10. Bill, M.; Mrózek, K.; Kohlschmidt, J.; Eisfeld, A.K.; Walker, C.J.; Nicolet, D. Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/KMT2A. Proc. Natl. Acad. Sci. USA 2020, 117, 26340–26346. [Google Scholar] [CrossRef]
  11. Hu, D.Y.; Wang, M.; Shen, K.; Pan, J.L.; Guo, Y.S.; Zhang, Z.B. A new breakpoint fusion gene involving KMT2A::EDC4 rearrangement in de novo acute myeloid leukemia. Int. J. Lab. Hematol. 2023, 45, 596–598. [Google Scholar] [CrossRef]
  12. Hyrenius-Wittsten, A.; Pilheden, M.; Sturesson, H.; Hansson, J.; Walsh, M.P.; Song, G. De novo activating mutations drive clonal evolution and enhance clonal fitness in KMT2A-rearranged leukemia. Nat. Commun. 2018, 9, 1770. [Google Scholar] [CrossRef]
  13. Dal Molin, A.; Tretti Parenzan, C.; Gaffo, E.; Borin, C.; Boldrin, E.; Meyer, L.H.; Te Kronnie, G.; Bresolin, S.; Bortoluzzi, S. Discovery of fusion circular RNAs in leukemia with KMT2A::AFF1 rearrangements by the new software CircFusion. Brief Bioinform. 2023, 24, bbac589. [Google Scholar] [CrossRef]
  14. Pui, C.H.; Gaynon, P.S.; Boyett, J.M.; Chessells, J.M.; Baruchel AKamps, W. Outcome of treatment in childhood acute lymphoblastic leukaemia with rearrangements of the 11q23 chromosomal region. Lancet 2002, 359, 1909–1915. [Google Scholar] [CrossRef]
  15. Pui, C.H.; Chessells, J.M.; Camitta, B.; Baruchel, A.; Biondi, A.; Boyett, J.M. Clinical heterogeneity in childhood acute lymphoblastic leukemia with 11q23 rearrangements. Leukemia 2003, 17, 700–706. [Google Scholar] [CrossRef] [PubMed]
  16. Balgobind, B.V.; Raimondi, S.C.; Harbott, J.; Zimmermann, M.; Alonzo, T.A.; Auvrignon, A. Novel prognostic subgroups in childhood 11q23/MLL-rearranged acute myeloid leukemia: Results of an international retrospective study. Blood 2009, 114, 2489–2496. [Google Scholar] [CrossRef]
  17. Szczepański, T.; Harrison, C.J.; van Dongen, J.J.M. Genetic aberrations in paediatric acute leukaemias and implications for management of patients. Lancet Oncol. 2010, 11, 880–889. [Google Scholar] [CrossRef]
  18. Alaggio, R.; Amador, C.; Anagnostopoulos, I.; Attygalle, A.D.; Araujo, I.B.D.O.; Berti, E.; Bhagat, G.; Borges, A.M.; Boyer, D.; Calaminici, M.; et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022, 36, 1720–1748. [Google Scholar] [CrossRef]
  19. Khoury, J.D.; Solary, E.; Abla, O.; Akkari, Y.; Alaggio, R.; Apperley, J.F. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022, 36, 1703–1719. [Google Scholar] [CrossRef] [PubMed]
  20. Arber, D.A.; Orazi, A.; Hasserjian, R.P.; Borowitz, M.J.; Calvo, K.R.; Kvasnicka, H.M. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: Integrating morphologic, clinical, and genomic data. Blood 2022, 140, 1200–1228. [Google Scholar] [CrossRef] [PubMed]
  21. Teachey, D.T.; Pui, C.H. Comparative features and outcomes between paediatric T-cell and B-cell acute lymphoblastic leukaemia. Lancet Oncol. 2019, 20, e142–e154. [Google Scholar] [CrossRef] [PubMed]
  22. Pieters, R.; De Lorenzo, P.; Ancliffe, P.; Aversa, L.A.; Brethon, B.; Biondi, A. Outcome of Infants Younger than 1 Year with Acute Lymphoblastic Leukemia Treated with the Interfant-06 Protocol: Results from an International Phase III Randomized Study. J. Clin. Oncol. 2019, 37, 2246–2256. [Google Scholar] [CrossRef]
  23. Nagayama, J.; Tomizawa, D.; Koh, K.; Nagatoshi, Y.; Hotta, N.; Kishimoto, T. Infants with acute lymphoblastic leukemia and a germline MLL gene are highly curable with use of chemotherapy alone: Results from the Japan Infant Leukemia Study Group. Blood 2006, 107, 4663–4665. [Google Scholar] [CrossRef] [PubMed]
  24. Fazio, G.; Bardini, M.; De Lorenzo, P.; Grioni, A.; Quadri, M.; Pedace, L. Recurrent genetic fusions redefine MLL germ line acute lymphoblastic leukemia in infants. Blood 2021, 137, 1980–1984. [Google Scholar] [CrossRef]
  25. Boer, J.M.; Valsecchi, M.G.; Hormann, F.M.; Antić, Ž.; Zaliova, M.; Schwab, C. Favorable outcome of NUTM1-rearranged infant and pediatric B cell precursor acute lymphoblastic leukemia in a collaborative international study. Leukemia 2021, 35, 2978–2982. [Google Scholar] [CrossRef] [PubMed]
  26. Guest, E.M.; Kairalla, J.A.; Hilden, J.M.; Dreyer, Z.E.; Carroll, A.J.; Heerema, N.A. Outstanding outcomes in infants with KMT2A-germline acute lymphoblastic leukemia treated with chemotherapy alone: Results of the Children’s Oncology Group AALL0631 trial. Haematologica 2022, 107, 1205–1208. [Google Scholar] [CrossRef]
  27. Issa, G.C.; Zarka, J.; Sasaki, K.; Qiao, W.; Pak, D.; Ning, J. Predictors of outcomes in adults with acute myeloid leukemia and KMT2A rearrangements. Blood Cancer J. 2021, 11, 162. [Google Scholar] [CrossRef] [PubMed]
  28. Nguyen, D.; Kantarjian, H.M.; Short, N.J.; Qiao, W.; Ning, J.; Cuglievan, B. Early mortality in acute myeloid leukemia with KMT2A rearrangement is associated with high risk of bleeding and disseminated intravascular coagulation. Cancer 2023, 129, 1856–1865. [Google Scholar] [CrossRef] [PubMed]
  29. Zhou, T.; Curry, C.V.; Khanlari, M.; Shi, M.; Cui, W.; Peker, D. Genetics and pathologic landscape of lineage switch of acute leukemia during therapy. Blood Cancer J. 2024, 14, 19. [Google Scholar] [CrossRef]
  30. Bacher, U.; Haferlach, T.; Kern, W.; Haferlach, C.; Schnittger, S. A comparative study of molecular mutations in 381 patients with myelodysplastic syndrome and in 4130 patients with acute myeloid leukemia. Haematologica 2007, 92, 744–752. [Google Scholar] [CrossRef]
  31. Guarnera, L.; Bravo-Perez, C.; Visconte, V. Immunotherapy in Acute Myeloid Leukemia: A Literature Review of Emerging Strategies. Bioengineering 2023, 10, 1228. [Google Scholar] [CrossRef]
  32. Döhner, H.; Wei, A.H.; Appelbaum, F.R.; Craddock, C.; DiNardo, C.D.; Dombret, H. 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] [PubMed]
  33. Döhner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Büchner, T. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
  34. Pollard, J.A.; Guest, E.; Alonzo, T.A.; Gerbing, R.B.; Loken, M.R.; Brodersen, L.E. Gemtuzumab Ozogamicin Improves Event-Free Survival and Reduces Relapse in Pediatric KMT2A-Rearranged AML: Results From the Phase III Children’s Oncology Group Trial AAML0531. J. Clin. Oncol. 2021, 39, 3149–3160. [Google Scholar] [CrossRef] [PubMed]
  35. Dillon, R.; Hills, R.K.; Burnett, A.K.; Russell, N.H. Gemtuzumab ozogamicin in (KMT2A)-rearranged adult acute myeloid leukaemia (AML) in the UK Medical Research Council AML15 and AML16 trials. Br. J. Haematol. 2022, 196, e50–2. [Google Scholar] [CrossRef]
  36. Ball, B.J.; Arslan, S.; Koller, P.; Ngo, D.; Afkhami, M.; Salhotra, A. Clinical experience with venetoclax and hypomethylating agents (HMA) in patients with newly diagnosed and relapsed or refractory KMT2A-Rearranged acute myeloid leukemia (AML). Leuk Lymphoma 2022, 63, 3232–3236. [Google Scholar] [CrossRef]
  37. Pigneux, A.; Labopin, M.; Maertens, J.; Cordonnier, C.; Volin, L.; Socié, G. Outcome of allogeneic hematopoietic stem-cell transplantation for adult patients with AML and 11q23/MLL rearrangement (MLL-r AML). Leukemia 2015, 29, 2375–2381. [Google Scholar] [CrossRef]
  38. Jiang, B.; Zhao, Y.; Luo, Y.; Yu, J.; Chen, Y.; Ye, B. Outcomes of Allogeneic Hematopoietic Stem Cell Transplantation in Adult Patients With Acute Myeloid Leukemia Harboring KMT2A Rearrangement and Its Prognostic Factors. Cell Transplant. 2024, 33, 9636897231225821. [Google Scholar] [CrossRef] [PubMed]
  39. Miyamura, T.; Kudo, K.; Tabuchi, K.; Ishida, H.; Tomizawa, D.; Adachi, S. Hematopoietic stem cell transplantation for pediatric acute myeloid leukemia patients with KMT2A rearrangement; A nationwide retrospective analysis in Japan. Leuk. Res. 2019, 87, 106263. [Google Scholar]
  40. Menghrajani, K.; Gomez-Arteaga, A.; Madero-Marroquin, R.; Zhang, M.J.; Bo-Subait, K.; Sanchez, J. Risk classification at diagnosis predicts post-HCT outcomes in intermediate-, adverse-risk, and KMT2A-rearranged AML. Blood Adv. 2022, 6, 828–847. [Google Scholar]
  41. Tong, J.; Zhang, L.; Liu, H.; Xu, X.; Zheng, C.; Yao, W. Umbilical cord blood transplantation can overcome the poor prognosis of KMT2A-MLLT3 acute myeloid leukemia and can lead to good GVHD-free/relapse-free survival. Ann. Hematol. 2021, 100, 1303–1309. [Google Scholar] [CrossRef]
  42. Antherieu, G.; Bidet, A.; Huet, S.; Hayette, S.; Migeon, M.; Boureau, L. Allogenic Stem Cell Transplantation Abrogates Negative Impact on Outcome of AML Patients with KMT2A Partial Tandem Duplication. Cancers 2021, 13, 2272. [Google Scholar] [CrossRef]
  43. Yokoyama, A.; Somervaille, T.C.P.; Smith, K.S.; Rozenblatt-Rosen, O.; Meyerson, M.; Cleary, M.L. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005, 123, 207–218. [Google Scholar] [CrossRef] [PubMed]
  44. Borkin, D.; He, S.; Miao, H.; Kempinska, K.; Pollock, J.; Chase, J. Pharmacologic inhibition of the Menin-MLL interaction blocks progression of MLL leukemia in vivo. Cancer Cell 2015, 27, 589–602. [Google Scholar] [CrossRef] [PubMed]
  45. Krivtsov, A.V.; Evans, K.; Gadrey, J.Y.; Eschle, B.K.; Hatton, C.; Uckelmann, H.J. A Menin-MLL Inhibitor Induces Specific Chromatin Changes and Eradicates Disease in Models of MLL-Rearranged Leukemia. Cancer Cell 2019, 36, 660–673.e11. [Google Scholar] [CrossRef]
  46. Klossowski, S.; Miao, H.; Kempinska, K.; Wu, T.; Purohit, T.; Kim, E. Menin inhibitor MI-3454 induces remission in MLL1-rearranged and NPM1-mutated models of leukemia. J. Clin. Investig. 2020, 130, 981–997. [Google Scholar] [CrossRef] [PubMed]
  47. Issa, G.C.; Aldoss, I.; DiPersio, J.; Cuglievan, B.; Stone, R.; Arellano, M. The menin inhibitor revumenib in KMT2A-rearranged or NPM1-mutant leukaemia. Nature 2023, 615, 920–924. [Google Scholar] [CrossRef] [PubMed]
  48. Perner, F.; Stein, E.M.; Wenge, D.V.; Singh, S.; Kim, J.; Apazidis, A. MEN1 mutations mediate clinical resistance to menin inhibition. Nature 2023, 615, 913–919. [Google Scholar] [CrossRef] [PubMed]
  49. Erba, H.P.; Fathi, A.T.; Issa, G.C. Update on a Phase 1/2 First-in-Human Study of the Menin-KMT2A (MLL) Inhibitor Ziftomenib (KO-539) in Patients with Relapsed or Refractory Acute Myeloid Leukemia. Blood 2022, 140 (Suppl. S1), 153–156. [Google Scholar] [CrossRef]
  50. Jabbour, E.; Searle, E.; Abdul-Hay, M. A First-in-Human Phase 1 Study of the Menin-KMT2A (MLL1) Inhibitor JNJ-75276617 in Adult Patients with Relapsed/Refractory Acute Leukemia Harboring KMT2A or NPM1 Alterations. Blood 2023, 142 (Suppl. S1), 57. [Google Scholar] [CrossRef]
  51. Daver, N.; Zeidner, J.F.; Yuda, J. Phase 1/2 First-in-Human Study of the Menin-MLL Inhibitor DSP-5336 in Patients with Relapsed or Refractory Acute Leukemia. Blood 2023, 142 (Suppl. S1), 2911. [Google Scholar] [CrossRef]
  52. Fleischmann, M.; Bechwar, J.; Voigtländer, D.; Fischer, M.; Schnetzke, U.; Hochhaus, A. Synergistic Effects of the RARalpha Agonist Tamibarotene and the Menin Inhibitor Revumenib in Acute Myeloid Leukemia Cells with KMT2A Rearrangement or NPM1 Mutation. Cancers 2024, 16, 1311. [Google Scholar] [CrossRef] [PubMed]
  53. Ling, Q.; Zhou, Y.; Qian, Y.; Qian, J.; Zhang, Y.; Wang, J. Repressing HIF-1α-induced HDAC9 contributes to the synergistic effect of venetoclax and MENIN inhibitor in KMT2Ar AML. Biomark Res. 2023, 11, 105. [Google Scholar] [CrossRef]
  54. Ye, J.; Zha, J.; Shi, Y.; Li, Y.; Yuan, D.; Chen, Q. Co-inhibition of HDAC and MLL-menin interaction targets MLL-rearranged acute myeloid leukemia cells via disruption of DNA damage checkpoint and DNA repair. Clin. Epigenetics 2019, 11, 137. [Google Scholar] [CrossRef]
  55. Fiskus, W.; Boettcher, S.; Daver, N.; Mill, C.P.; Sasaki, K.; Birdwell, C.E. Effective Menin inhibitor-based combinations against AML with MLL rearrangement or NPM1 mutation (NPM1c). Blood Cancer J. 2022, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  56. Aubrey, B.J.; Cutler, J.A.; Bourgeois, W.; Donovan, K.A.; Gu, S.; Hatton, C. IKAROS and MENIN coordinate therapeutically actionable leukemogenic gene expression in MLL-r acute myeloid leukemia. Nat. Cancer 2022, 3, 595–613. [Google Scholar] [CrossRef] [PubMed]
  57. Bourgeois, W.; Cutler, J.A.; Aubrey, B.J.; Wenge, D.V.; Perner, F.; Martucci, C. Mezigdomide is effective alone and in combination with menin inhibition in preclinical models of KMT2A-r and NPM1c AML. Blood 2024, 143, 1513–1527. [Google Scholar] [CrossRef]
  58. Kubota, Y.; Reynold, M.; Williams, N.D. Genomic Analyses Unveil the Pathogenesis and Inform on Therapeutic Targeting in KMT2A-PTD AML. Blood 2023, 142 (Suppl. S1), 5696. [Google Scholar] [CrossRef]
  59. Lo-Coco, F.; Avvisati, G.; Vignetti, M.; Thiede, C.; Orlando, S.M.; Iacobelli, S. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N. Engl. J. Med. 2013, 369, 111–121. [Google Scholar] [CrossRef]
  60. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of KTM2A gene structure and cofactors. KMT2A gene is located on chromosome 11q23.3. Presence of KMT2A fusion defines some subtypes of leukemia. Fusion proteins recruit transcription cofactors to promote transcription elongation. A potential therapeutic window derives from the inhibition of such cofactors (for instance, menin). Abbreviations: AT-H, AT-hooks; Zn-F, zinc fingers; P, PHD fingers; B, bromodomain; FYRN, FY-rich domain N-terminal; FYRC, FY-rich domain C-terminal.
Figure 1. Schematic representation of KTM2A gene structure and cofactors. KMT2A gene is located on chromosome 11q23.3. Presence of KMT2A fusion defines some subtypes of leukemia. Fusion proteins recruit transcription cofactors to promote transcription elongation. A potential therapeutic window derives from the inhibition of such cofactors (for instance, menin). Abbreviations: AT-H, AT-hooks; Zn-F, zinc fingers; P, PHD fingers; B, bromodomain; FYRN, FY-rich domain N-terminal; FYRC, FY-rich domain C-terminal.
Ijms 25 09023 g001
Table 1. KMT2A partner genes in acute myeloid leukemia.
Table 1. KMT2A partner genes in acute myeloid leukemia.
Partner
Gene
Chromosomal Location Infant
AML
Pediatric
AML
Adult
AML
% Total AML
MLLT3(9p22)28.4%36.5%24.4%30.4%
MLLT10(10p12.3)27.4%23.4%9.9%18.7%
KMT2A-PTD(11q23.3)01.5%25.6%10.7%
ELL(19p13.1)15.2%6.8%11.5%10.1%
AFDN(6q27)1.5%9.03%10.2%8.1%
MLLT1(19p13.3)1%6.02%3.6%4.2%
MLLT11(1q21.3)6.59%2.15%0.9%2.4%
SEPTIN6(Xq24)3.55%2.15%0.45%1.7%
MLLT6(17q12)01.07%2.72%1.6%
EPS15(1p32.3)1.01%1.07%1.58%1.3%
SEPTIN9(17q25)1.01%1.29%1.36%1.3%
CBLDel (11) (q23.3q23.3)000.22%0.89%
AFF1(4q21)1.5%0.2%1.1%0.8%
SEPTIN5(22q11.2)1.01%0.64%0.45%0.71%
ABI1(10p11.2)1.52%1.07%00.71%
Abbreviations: AML, acute myeloid leukemia. Partner genes were extracted from Meyer C. et al., Leukemia. 2023;37(5):988-1005 [1].
Table 2. Treatments in patients with KMT2A rearrangements.
Table 2. Treatments in patients with KMT2A rearrangements.
PopulationDrugsKMT2Ar TypeOutcomeReference #
Adult AML-cytarabine/daunorubicin-based induction chemotherapy ± autologous stem cell transplantation
-decitabine ± bortezomib
t(9;11) (p22;q23)
t(6;11) (q27;q23)
t(11;19) (q23;p13.1)
t(11;19) (q23;p13.3)
t/ins(10;11) (p13;q23)
% CR (42-77)
% 3-year OS (41-5)
[10]
-venetoclax + HMAt(9;11) (p22;q23)
t(11;19) (q23;p13.1)
t(6;11) (q23;q27)
t(10;11) (p11.2;q23)
t(4;11) (q21.3;q23)
t(11;17) (q13;q23)
t(11;11) (q13;q23)
t/ins(10;11) (p13;q23)
% CR 83
median OS 11.05 months
[36]
-allo-HSCTt(6;11) (q27;q23)
t(9;11) (p22:q23)
t(11;19) (q23;p13.1)
t(10;11) (p12;q23)
t(1;11) (q21;q23)
% 3-year OS 59.1[38]
-intensively treatedKMT2A-PTD% CR 64.5
median OS 24.4 months
[42]
Adult R/R AML-venetoclax + HMAt(9;11) (p22;q23)
t(11;19) (q23;p13.1)
t(6;11) (q23;q27)
t(10;11) (p11.2;q23)
t(4;11) (q21.3;q23)
t(11;17) (q13;q23)
t(11;11) (q13;q23)
t/ins(10;11) (p13;q23)
% CR 17
median OS 6.05 months
[36]
Adult R/R AML-ALL-MPAL-revumenib (SNDX-5613)t(9;11) (p22:q23)
t(4;11) (q21.3;q23)
t(11;19) (q23;p13.1)
t(6;11) (q27;q23)
t(11;17) (q13;q23)
Mut. NPM1
% CR 33[47]
Pediatric AML-intensively treated + GOt(6;11) (q23;q27)
t(10;11) (p11.2;q23) t(10;12) (p12;q23)
t(4;11) (q21.3;q23)
t(11;19) (q23;p13.3)
% CR 77
% 5-year OS 50
[34]
-allo-HSCTt(9;11) (p22:q23)
t(11;19) (q23;p13.1)
t(1;11) (q21;q23)
t(6;11) (q27;q23)
t(10;11) (p12;q23)
t(4;11) (q21.3;q23)
t(5;11) (q31;q23)
t(7;11) (q22;q23)
t(8;11) (q24;q23)
% 3-year OS 52.1[39]
Abbreviations: AML, acute myeloid leukemia; MPAL, mixed-phenotype acute leukemia; ALL, acute lymphocytic leukemia; KMT2Ar, KMT2A rearrangement; HSCT, hematopoietic stem cell transplantation; CR, complete response; R/R, refractory/relapsed; PTD, partial tandem duplication; OS, overall survival; GO, gemtuzumab ozogamicin.
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

Guarnera, L.; D’Addona, M.; Bravo-Perez, C.; Visconte, V. KMT2A Rearrangements in Leukemias: Molecular Aspects and Therapeutic Perspectives. Int. J. Mol. Sci. 2024, 25, 9023. https://doi.org/10.3390/ijms25169023

AMA Style

Guarnera L, D’Addona M, Bravo-Perez C, Visconte V. KMT2A Rearrangements in Leukemias: Molecular Aspects and Therapeutic Perspectives. International Journal of Molecular Sciences. 2024; 25(16):9023. https://doi.org/10.3390/ijms25169023

Chicago/Turabian Style

Guarnera, Luca, Matteo D’Addona, Carlos Bravo-Perez, and Valeria Visconte. 2024. "KMT2A Rearrangements in Leukemias: Molecular Aspects and Therapeutic Perspectives" International Journal of Molecular Sciences 25, no. 16: 9023. https://doi.org/10.3390/ijms25169023

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