Skip to main content
Erschienen in: Experimental Hematology & Oncology 1/2023

Open Access 01.12.2023 | Research

Modelling acquired resistance to DOT1L inhibition exhibits the adaptive potential of KMT2A-rearranged acute lymphoblastic leukemia

verfasst von: Pauline Schneider, Nicholas T. Crump, Susan T.C.J.M. Arentsen-Peters, Alastair L. Smith, Rico Hagelaar, Fabienne R.S. Adriaanse, Romy S. Bos, Anja de Jong, Stefan Nierkens, Bianca Koopmans, Thomas A. Milne, Rob Pieters, Ronald W. Stam

Erschienen in: Experimental Hematology & Oncology | Ausgabe 1/2023

Abstract

In KMT2A-rearranged acute lymphoblastic leukemia (ALL), an aggressive malignancy, oncogenic KMT2A-fusion proteins inappropriately recruit DOT1L to promote leukemogenesis, highlighting DOT1L as an attractive therapeutic target. Unfortunately, treatment with the first-in-class DOT1L inhibitor pinometostat eventually leads to non-responsiveness. To understand this we established acquired pinometostat resistance in pediatric KMT2A::AFF1+ B-ALL cells. Interestingly, these cells became mostly independent of DOT1L-mediated H3K79 methylation, but still relied on the physical presence of DOT1L, HOXA9 and the KMT2A::AFF1 fusion. Moreover, these cells selectively lost the epigenetic regulation and expression of various KMT2A-fusion target genes such as PROM1/CD133, while other KMT2A::AFF1 target genes, including HOXA9 and CDK6 remained unaffected. Concomitantly, these pinometostat-resistant cells showed upregulation of several myeloid-associated genes, including CD33 and LILRB4/CD85k. Taken together, this model comprehensively shows the adaptive potential of KMT2A-rearranged ALL cells upon losing dependency on one of its main oncogenic properties.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s40164-023-00445-8.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Background

Chromosomal translocations involving the KMT2A (MLL) gene constitute the cytogenetic hallmark of acute lymphoblastic leukemia (ALL) diagnosed in infants (< 1 year of age), giving rise to an aggressive malignancy with high relapse rates and low event-free survival (EFS) chances of 30–40% [1, 2]. Hence, currently available treatment regimens for KMT2A-rearranged infant ALL are inadequate and require more effective therapeutic options to improve clinical outcome.
KMT2A translocations result in the fusion of KMT2A to one of its many translocation partner genes, [3] generating chimeric transcripts encoding highly oncogenic KMT2A fusion proteins. Among infant ALL patients, KMT2A is most recurrently fused to either AFF1 (AF4), MLLT1 (ENL), or MLLT3 (AF9) [3]. Functionally, wild-type KMT2A plays an essential role in definitive hematopoiesis [4] regulating gene expression through histone 3 lysine 4 (H3K4) methyltransferase activity, mediated by its Su(Var)3–9, Enhancer-of-zeste, Trithorax (SET) domain [5]. In contrast, KMT2A fusion proteins lose their SET domain, but instead recruit the histone 3 lysine 79 (H3K79) methyltransferase DOT1L through binding motifs encoded by the translocation partner genes [68]. Binding of DOT1L to KMT2A fusion proteins causes inappropriate H3K79 hypermethylation at KMT2A target genes, leading to an altered transcriptomic landscape that strongly favors leukemia development [69].
Interestingly, the mutational landscape of KMT2A-rearranged infant ALL is remarkably silent, with only 1.3–2.5 leukemia-specific, non-silent mutations in the dominant clone per patient, [10, 11] suggesting that the KMT2A translocation may well be the sole oncogenic lesion driving this aggressive type of leukemia [12]. Therefore, targeting DOT1L, through which KMT2A fusion proteins exert their oncogenic effects, represents an attractive therapeutic strategy. Accordingly, the development of the DOT1L inhibitor EPZ004777 and its successor EPZ5676 (pinometostat) [13, 14] were expected to become key to successful treatment of KMT2A-rearranged acute leukemias. However, despite promising preclinical results, subsequent clinical trials revealed that good initial responses in patients treated with pinometostat readily led to non-responsiveness due to acquired resistance and poor pharmacokinetics (PK) [15, 16]. Previously, it was reported that acquired resistance to pinometostat in KMT2A-rearranged acute leukemic cell lines is associated with enhanced drug efflux mediated by the elevated expression of the multidrug resistance transporters ABCB1 and ABCG2 [17].
Despite this, targeting DOT1L remains a promising avenue for treating KMT2A rearranged leukemias, and novel small-molecule DOT1L inhibitors with improved PK profiles have already been identified [18, 19]. For future drug development, more needs to be understood about exactly how DOT1L contributes to leukemogenesis, the role of its enzymatic methyltransferase activity, and how leukemias might develop resistance. Understanding these issues could not only impact the development of novel DOT1L inhibitors but could also be essential for better understanding the activity of novel compounds designed to target similar pathways. Therefore, following up on the study by Campbell and colleagues, [17] we here established and extensively characterized a model of acquired resistance to DOT1L inhibition in KMT2A-rearranged ALL cells.

Methods

Cell line models

The KMT2A::AFF1+ B-cell precursor ALL cell lines used are SEM, (DSMZ, cat.nr. ACC 546), and RS4;11, (ATCC; cat.nr. CRL-1873). Culture conditions are described in detail in the supplemental methods.

Establishment of acquired pinometostat resistance in SEM and RS4;11 cells

SEM and RS4;11 cells were cultured in the presence of gradually increasing concentrations (ranging from 1 to 100 µM) of the DOT1L inhibitor pinometostat (EPZ5676, Selleckchem), for 14 weeks. For assessment of the in vitro response to pinometostat, cells were cultured in the absence of pinometostat for a few passages before exposing the cells to six concentrations (ranging from 0 to 100 μm), of pinometostat for 14 days. Trypan blue exclusion counts were used to calculate the inhibitory pinometostat concentration to 50% of the leukemic cells (i.e., IC50 value). p values were determined by ratio paired t-test using four biological replicates, mean with range.

Immunoblotting

The presence of histone modifications and the levels of protein expression were determined by immunoblot analysis, as described in the supplemental methods.

RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq)

RNA- and ChIP-sequencing was performed on a NextSeq® 500 System (Illumina®). Experimental procedures and analyses are described in the supplemental methods.

Assay for transposase-accessible chromatin sequencing (ATAC-seq)

ATAC sequencing was outsourced to Active Motif (ATACseq Service: https://​www.​activemotif.​com/​catalog/​1233/​atac-seq-service) to identify regions that have open or accessible chromatin states, as described in the supplemental methods.

Flow cytometry (FACS) analysis

Details of FACS analysis are described in the supplemental methods.

RNA interference

To transiently induce mRNA knockdowns, leukemic cells were electroporated in the presence of 500 nM of small-interfering RNAs (siRNAs) directed against the mRNA of selected target genes, as described previously [20], and as described in the supplemental methods.

Quantitative reverse-transcription PCR analysis

RNA, isolated using the RNeasy Mini Kit (QIAGEN), was reverse transcribed and the obtained cDNA was used for quantitative reverse-transcription PCR (qRT-PCR) analysis as described previously [21], and as described in the supplemental methods.

Cell viability assays and high-throughput drug screening

Cell viability assays were performed using flow cytometry and 7-AAD viability dye (BioLegend) to discriminate between viable and dead cells. Further details on the cell viability assays and high-throughput drug screens are described in the supplemental methods.

Statistical analysis

Statistical significance of independent experimental replicates in graphs were determined by two-sided Student’s t-tests. All statistical analyses were conducted using GraphPad Prism8, version 8.3.4. p < 0.05 was considered statistically significant.

Results

Establishment of acquired resistance to DOT1L inhibition in KMT2A-rearranged ALL cells

To induce acquired resistance to DOT1L inhibition, the KMT2A::AFF1+ ALL cell line SEM [22] was exposed to increasing concentrations of the first-in-class DOT1L inhibitor pinometostat for 14 weeks (Fig. 1A). Next, cells were cultured in the absence of drug before evaluating potential changes in pinometostat-induced cytotoxicity. Exposure of maternal SEM cells to 50 µM pinometostat for 1 week reduced the percentage of viable cells to ~ 35%.
In contrast the viability of SEM cells that underwent prolonged exposure to increasing pinometostat concentrations was hardly affected (Fig. 1B). This pinometostat-resistant daughter line, designated as SEMPINO_RES, revealed a 34-fold higher 14-day-IC50 value as compared to maternal SEM cells (Fig. 1C), indicating that SEMPINO_RES became highly resistant. An additional model of intermediate resistance was established in SEM cells (i.e., SEMPINO_INT) by prolonged exposure to 4.5 µM pinometostat for 7 weeks, leading to a 16-fold higher IC50 (Fig. 1C).
Similar to SEMPINO_RES, we also induced pinometostat resistance in the KMT2A::AFF1+ ALL cell line RS4;11. With a mean 14-day-IC50 value of ~ 10 µM, representing pinometostat concentrations well above maximum achievable plasma levels in pinometostat-treated patients, [15, 16, 23] maternal RS4;11 is more resistant than SEM (Fig. 1C). Two emerging pinometostat-resistant RS4;11 daughter lines, i.e., RS4;11PINO_RES#1 and RS4;11PINO_RES#2, showed IC50 values of 33–45 µM (Fig. 1C).
To assess the inhibitory effects of pinometostat on DOT1L-mediated methyltransferase activity, the levels of H3K79 di-methylation (H3K79me2) were determined by immunoblot analysis. Interestingly, the levels of H3K79me2 in SEM and SEMPINO_RES were comparable and pinometostat was able to substantially reduce the levels of H3K79me2 equally in both cell lines (Fig. 1D-E). Hence, despite persistent inhibition of DOT1L-mediated H3K79 methylation, cell viability in SEMPINO_RES is no longer affected, suggesting that these cells became largely independent of H3K79 methylation induced by DOT1L.
Next, we assessed whether changes in global histone modifications had occurred between SEM and SEMPINO_RES. For this we used Mod Spec®, a mass spectrometry-based measurement for the relative abundance of over 80 distinct histone marks. This analysis confirmed no differences in the levels of H3K79 mono-, di-, and tri-methylation (i.e., H3K79me1, H3K79me2, and H3K79me3, respectively) between SEM and SEMPINO_RES, and showed equal reduction of these histone marks upon pinometostat exposure (Figure S1). Moreover, these data demonstrated that the global landscape of histone modifications between SEM cells and SEMPINO_RES largely remained similar. The only histone modification that is downregulated in response to pinometostat exposure appeared to be H3K79 methylation, demonstrating the specificity of this agent.
In KMT2A-rearranged acute leukemias, the KMT2A fusion protein is considered to be the main oncogenic driver and loss of DOT1L was shown to specifically decrease KMT2A fusion-driven transcriptional programs, including the expression of HOXA9. [7] Therefore, we asked whether acquired resistance to DOT1L inhibition was accompanied by an altered dependency on DOT1L, KMT2A::AFF1 and/or HOXA9. Therefore SEM and SEMPINO_RES cells were subjected to siRNA-mediated knockdown of these genes, resulting in significant reductions in mRNA expression of ~ 75–80% for DOT1L and KMT2A::AFF1, and ~ 50–65% for HOXA9, relative to non-silencing controls (NSCs) (Fig. 1F). Validation on the protein level confirmed a reduction of DOT1L of ~ 90% in SEM and of > 90% in SEMPINO_RES (Fig. 1H,I). Knockdown of DOT1L was accompanied by a reduction of H3K79me2 of ~ 90% in SEM, and of ~ 40% SEMPINO_RES (Fig. 1H, J). Surprisingly, knockdown of DOT1L and KMT2A::AFF1 resulted in similar reductions in cell viability in both SEM and SEMPINO_RES. For HOXA9 suppression the effects on cell viability in SEMPINO_RES appeared to be somewhat delayed (Fig. 1G). This suggests that although SEMPINO_RES cells became less sensitive to inhibition of H3K79 methylation in terms of leukemic cell survival, these cells remained dependent on the physical presence of proteins known to be important in KMT2A-mediated leukemogenesis, including DOT1L.

Acquired resistance to DOT1L inhibition leads to selective loss of KMT2A-fusion driven gene expression

Next, we performed RNA- and ChIP-seq for KMT2A, AFF1, H3K4me3, H3K79me2, and H3K27ac, as well as ATAC-seq on SEM and SEMPINO_RES cells cultured in both the absence and presence of 50 µM pinometostat for 7 days. Interestingly, as assessed by ChIP-seq, there are very few observable differences in the global profiles of KMT2A, AFF1 or H3K79me2 as well as ATACseq profiles between SEM or SEMPINO_RES, suggesting that acquired pinometostat resistance does not lead to obvious global changes in open chromatin (Fig. 2A). Upon analyzing gene expression profiles, it became apparent that in the absence of pinometostat, there was a noteworthy decrease in the expression of 760 out of the 13,371 genes expressed (5.7%), while 588 genes (4.4%) exhibited an increase in expression in SEMPINO_RES cells in comparison to SEM cells (Fig. 2B, Table S1). The differences in gene expression patterns triggered by pinometostat were relatively less prominent between the two cell line models (Figure S2, Table S1). In the presence of pinometostat in the original SEM cells, 670 genes (5.0%) revealed a significant decrease in expression, and 596 genes (4.5%) demonstrated a notable increase in expression, when compared to untreated SEM cells. Conversely, in the presence of pinometostat in SEMPINO_RES cells, 208 genes (1.6%) were significantly downregulated, while 388 genes (2.9%) were significantly upregulated compared to untreated SEMPINO_RES cells. Interestingly, a considerable number of genes reported to represent potential target genes of KMT2A fusion proteins [9, 20, 24, 25] were significantly downregulated in SEMPINO_RES cells (Fig. 2C, Table S1). Approximately half of the KMT2A::AFF1 target genes identified by Guenther et al. [24] were downregulated in SEMPINO_RES (Fig. 2D, Table S1), as well as a quarter of the top 50 genes associated with H3K79 methylation in KMT2A-rearranged acute leukemia patient samples as identified by Krivtsov et al. [9].
Likewise, ~ 25% of the genes we previously reported to be differentially expressed in response to siRNA-mediated repression of KMT2A::AFF1 and KMT2A:MLLT1 in KMT2A-rearranged ALL cells, [20] as well as a fourth of the genes reported to display binding of KMT2A:AFF1 that spreads beyond the gene promoter and well into the gene body as recently identified in SEM cells by Kerry et al., [25] were downregulated in SEMPINO_RES (Fig. 2D, Table S1). Thus, acquired resistance to DOT1L inhibition leads to selective (or partial) loss of KMT2A-fusion driven gene expression. To explore biological pathways potentially affected by acquired resistance to DOT1L inhibition, we performed Gene Set Enrichment Analysis (GSEA) on all RNA-seq data, and identified various hallmark gene sets to be significantly (nominal p-value < 0.05) modulated in SEMPINO_RES. These included the upregulated gene sets ‘MYC targets v1 and v2’ and ‘Oxidative Phosphorylation’, as well as downregulated gene sets such as ‘p53 pathway’ (i.e., DNA damage response genes), ‘epithelial-to-mesenchymal transition (EMT)’ and ‘hypoxia’ (Fig. 2E).
Examination of the genes most prominently enriched (n = 50) and the genes most notably under-represented (n = 50) in our GSEA data revealed PROM1 to be the most downregulated gene (GSEA score of -4,83) and CCNA1 the most positively enriched gene (GSEA score of 3,66) in SEMPINO_RES cells (Fig. 2F). Both genes represent putative KMT2A fusion targets epigenetically marked by H3K79 methylation and have shown to be highly and specifically expressed in KMT2A-rearranged ALL [9]. PROM1 encodes a transmembrane glycoprotein (i.e., CD133) commonly regarded as a cancer stem cell marker [2628] and reported to be an important target of KMT2A::AFF1 [24, 29, 30]. PROM1 is robustly expressed in SEM cells but readily downregulated during pinometostat exposure, whereas PROM1 expression was nearly absent in SEMPINO_RES (Fig. 3A). Analysis at protein level by immunoblot and FACS confirmed the complete loss of PROM1/CD133 in SEMPINO_RES, while in SEMPINO_INT PROM1/CD133 was still present in in ~ 88% of the cells (Fig. 3B-E). This indicates that PROM1/CD133 expression is gradually lost from the population after prolonged pinometostat exposure. Similarly, RS4;11 cells firmly express PROM1/CD133, which was markedly reduced in both RS4;11PINO_RES#1 and RS4;11PINO_RES#2 (Fig. 3B-E).
ChIP-sequencing data for KMT2A, AFF1, H3K4me3, H3K79me2, and H3K27ac showd that upon pinometostat exposure, SEM cells display a clear reduction of KMT2A binding in PROM1, which was accompanied by strong reductions of the levels of H3K79me2, H3K4me3, and H3K27ac at the PROM1 gene as well as at its enhancer TAPT1 [29] (Fig. 3F). In untreated SEMPINO−RES cells the PROM1 locus is completely devoid of KMT2A, AFF1, H3K79me2, H3K4me3, and H3K27ac, suggesting that this gene is no longer being regulated by KMT2A::AFF1 and subsequent DOT1L-mediated H3K79 methylation. Also, ATAC-sequencing clearly revealed decreased chromatin accessibility at the promoter and enhancer of PROM1 in SEMPINO−RES (Fig. 3F). Interestingly, at the TAPT1 locus in these same SEMPINO−RES cells KMT2A and AFF1 binding as well as the levels of H3K4me3, H3K79me2, and H3K27ac to some extent remained intact (Fig. 3F).
In addition to PROM1, the expression of other putative KMT2A::AFF1 target genes, including RUNX2, PRSS12, ZC3H12, SERPINB1, GNAQ and BANK1 were severely downregulated in SEMPINO−RES with a logFC of > 3-fold (Fig. 2F and Figure S3A) and exhibited similar patterns of RNA-, ChIP-, and ATAC-seq as observed for PROM1 (Figure S3B), indicating their dependence on KMT2A::AFF1-mediated epigenetic control. In contrast, at SERPINB1 only moderate levels of H3K79me2 were observed, accompanied by rather weak binding of KMT2A and absence of AFF1, suggesting that this gene may not necessarily be regulated KMT2A fusion proteins and DOT1L (Figure S3A, B),).
Collectively, these data demonstrate that a selection of known KMT2A::AFF1 target genes that are responsive to pinometostat-mediated DOT1L inhibition in SEM cells are relieved from the epigenetic control of KMT2A::AFF1 and become transcriptionally silenced in SEMPINO_RES cells.
Following previous evidence on the role of DOT1L in HOXA gene expression in KMT2A::AFF1+ ALL cells, [9, 24] we examined the HOXA locus and found that HOXA9 and HOXA10 were expression at comparable levels in both SEM and SEMPINO_RES (Fig. 4A). Inhibition of DOT1L-mediated H3K79 methylation by pinometostat resulted in moderately decreased expression of HOXA9, HOXA7, and HOXA10, while the levels of H3K27ac, H3K4me3, KMT2A, and AFF1 remained unchanged in both cell lines (Fig. 4A,B). Similar patterns were found for other KMT2A-fusion target genes, including CDK6, involved in cell proliferation in KMT2A rearranged ALL [20] (Fig. 4A,B and S4A-C). The expression of MEIS1, which encodes a required co-factor of HOXA9-driven leukemogenesis, [9, 24, 31, 32] remained unaltered upon pinometostat exposure despite reductions in the levels of H3K79me2 in both SEM and SEMPINO_RES (Fig. 4A,B and S4A,B,C). Collectively, this indicates that a subset of KMT2A-fusion target genes continued to be regulated by DOT1L in pinometostat-resistant ALL cells, while a separate group of genes showed no transcriptional response to the inhibition of DOT1L-mediated H3K79 methylation.
Intriguingly, we also found the expression of some putative KMT2A-fusion target genes to be upregulated in SEMPINO_RES in the absence of pinometostat (Figure S4D), including HOXA7, NLGN4X, CCNA1, FCRLA, IL7R, LYN and FUT4 (Fig. 4A,B and Figure S4D,E).

Upregulation of myeloid-associated gene expression in KMT2A-rearranged ALL cells upon acquired resistance to DOT1L inhibition

Apart from differential gene expression of putative KMT2A::AFF1 target genes, our data also revealed changes in expression of genes not associated with KMT2A fusions and/or H3K79 methylation (Fig. 2F). One of the most enriched and upregulated genes upon acquired pinometostat-resistance according to our GSEA is LILRB4 (Figs. 2F and 5A), encoding the monocytic differentiation marker CD85k [3335]. LILRB4 is hardly expressed in SEM cells but is moderately upregulated during pinometostat exposure and substantially expressed in SEMPINO_RES cells (Fig. 5A). In SEM cells, pinometostat induced an increase of chromatin accessibility as well as an increase in the levels of H3K27ac and KMT2A binding at the LILRB4 locus, yet no H3K79me2 or binding of AFF1 was detected, suggesting that upregulated of LILRB4 expression is not dependent on DOT1L or KMT2A::AFF1 (Fig. 5B).
FACS analysis confirmed an increased population of ~ 60% in LILRB4/CD85k positive cells in SEMPINO_RES compared to only ~ 7% in SEM (Fig. 5C,E). Counterintuitively, instead of an expected moderate increase in LILRB4/CD85k-positive cells, we found SEMPINO_INT to have lost LILRB4/CD85k expression almost completely (Fig. 5C,E). In RS4;11 already 25% of the cells were positive for LILRB4/CD85k, which tremendously increased to approximately 90% of the cells in both RS4;11PINO_RES daughter lines (Fig. 5C,E). Interestingly, apart from LILRB4/CD85k, we found additional myeloid-associated genes to be upregulated in SEMPINO_RES, including CD33, CCL5, LIMK1, and MPEG1, revealing similar patterns of RNA-, ChIP- and ATAC-seq as LILRB4, although less prominent (Fig. 2F, Figures S5A, S5B). CD33, commonly expressed in a subpopulation in KMT2A-rearranged infant ALL [3638], serves as an important immunophenotypic marker for the characterization of pediatric acute myeloid leukemia (AML) by EuroFlow [3942] and has been exploited as a therapeutic target for AML. In SEM a subpopulation of 20% of CD33-positive cells was identified, which was increased in SEMPINO_RES to about 40%, yet CD33-positive cells again were largely absent in SEMPINO−INT (Fig. 5D,F). Similarly, in RS4;11 a CD33-positive subpopulation of 9% was increased upon pinometostat resistance to 14% in RS4;11PINO_RES#1 and up to 50% in RS4;11PINO_RES#2 (Fig. 5D,F).
Together these data indicate that under prolonged pressure of DOT1L inhibition, KMT2A-rearranged ALL cells seem to initiate a reprogramming process that involves the acquisition (or selection) of myeloid-like characteristics.

Drug screens reveal minimal cross resistance, and sensitization towards venetoclax after acquired pinometostat resistance

Finally, acquired pinometostat resistance led to the upregulation of the multidrug efflux pump ABCB1 (Fig. 2F and Figure S6A,B), associated with multidrug resistance and previously reported as the mechanism of pinometostat resistance in KMT2A-rearranged acute leukemia cell lines [17]. However, our data challenges the concept that elevated ABCB1 expression alone is the mechanism of resistance to DOT1L inhibition as reported previously [17]. Despite significant ABCB1 upregulation, we still observe comparable inhibition of H3K79 methylation in SEM and SEMPINO_RES (Fig. 1D and E), indicating that the amount of pinometostat and/or its retention in SEMPINO_RES cells is sufficient to effectively reduce H3K79me2 levels, overriding the impact of ABCB1 upregulation.
Since multidrug efflux pumps are associated with multidrug resistance [43, 44], we assessed whether SEMPINO_RES cells had become more resistant to current chemotherapeutics for KMT2A-rearranged infant ALL [1, 2] and whether we could identify agents to which SEMPINO_RES cells had become more sensitive by performing drug library screens (Table S2, Fig. 6A,B).
This revealed an increased resistance to the glucocorticoids dexamethasone and prednisolone (the liver-activated form of prednisone), vincristine, daunorubicin, and L-asparaginase, and increased sensitivity to cytarabine and for instance to the BCL-2 inhibitor venetoclax (Fig. 6A-E). Interestingly, cytarabine typically represents a drug commonly used in the treatment of myeloid leukemias, and the combination of venetoclax and cytarabine has successfully been tested in AML patients [4547].

Discussion

The currently accepted dogma of KMT2A-fusion driven leukemogenesis dictates the requirement of DOT1L-mediated activation of KMT2A target genes through H3K79 methylation [6, 9, 48, 49]. Therefore, targeting DOT1L [13, 18, 19] represents an attractive therapeutic option for patients diagnosed with KMT2A-rearranged acute leukemia, despite the first-in-class DOT1L inhibitor pinometostat showing dissatisfying results in adult patients [15]. While next generation DOT1L inhibitors with improved pharmacokinetic profiles are in development, [18, 19] we reasoned that the mechanisms by which KMT2A-rearranged acute leukemia cells evade DOT1L inhibition may provide novel insights into the biology of these unique malignancies. Therefore, and in a similar fashion as published by Campbell and colleagues [17], we efficiently induced acquired pinometostat resistance in various cell line models, demonstrating how readily KMT2A-rearranged acute leukemia cells become resistant to DOT1L inhibition. The study of Campbell et al., mainly focused on examples of possible mechanisms of pinometostat resistance including increased expression of drug efflux transporters and activation of the PI3K/AKT and RAS/RAF/MEK/ERK pathways [17]. In contrast, we here specifically focused on the behavior and epigenetic regulation of DOT1L-associated KMT2A fusion-driven target genes and on how the transcriptomic landscape changes in KMT2A-rearranged ALL cells that are able to evade leukemic cell death during prolonged inhibition of DOT1L-mediated H3K79 methylation.
In concordance with the previous finding by Campbell et al., we found increased expression of the multi-drug efflux pump ABCB1 in our pinometostat-resistant SEMPINO_RES cells. However, despite the elevated levels of ABCB1 expression, pinometostat continued to inhibit H3K79 methylation in SEMPINO_RES. This strongly indicates that the increased levels of ABCB1 are insufficient to prevent pinometostat from exerting its inhibitory effects, and therefore cannot be the sole mechanism of acquired pinometostat resistance.
Interestingly, while cell viability of SEMPINO_RES cells was no longer affected by pinometostat-induced inhibition of DOT1L-mediated H3K79 methylation, these cells remained dependent on the physical presence of DOT1L protein. This may indicate that recently described biological functions of DOT1L that are independent of H3K79 methylation [5052] are also important for KMT2A-rearranged acute leukemia cells. Thus, in addition to its enzymatic methylatransferase activity, DOT1L clearly has a scaffold function in assembling transcriptionally competent complexes. Therefore, therapeutic degradation of DOT1L instead of solely inhibiting its catalytic activities might be beneficial in the treatment of KMT2A-rearranged acute leukemia.
Another intriguing aspect of our model of acquired pinometostat resistance is the observation that SEMPINO_RES cells remained vulnerable to knockdown of the KMT2A::AFF1 fusion gene. This may suggest that inappropriate recruitment of DOT1L to loci otherwise not associated with H3K79me2 may not represent the sole KMT2A fusion-mediated attribute driving leukemogenesis and/or leukemia maintenance. If so, the identification of such DOT1L-independent oncogenic properties may well uncover important therapeutic targets and more effective treatment options for KMT2A-rearranged acute leukemias.
As shown, acquired resistance to pinometostat led to marked transcriptional downregulation of putative KMT2A-fusion target genes, which was accompanied by reductions in H3K79me2, as well as loss of binding of KMT2A and AFF1, and chromatin condensation at the corresponding loci. The complete loss of PROM1/CD133, which was shown to be transcriptionally regulated via KMT2A::AFF1-mediated H3K79me2/3 enhancer–promoter interactions, [29] in SEMPINO_RES is highly remarkable, since the expression of PROM1/CD133 was reported to be essential for leukemic cell growth in KMT2A-rearranged ALL [29, 30]. Consequently, targeting PROM1/CD133-positive cells has been proposed as a therapeutic option for KMT2A-rearranged ALL, although the expression of PROM1/CD133 on both fetal and adult hematopoietic stem cells (HSCs) may compromise the specificity of such an approach [29, 30, 53, 54]. Moreover, PROM1/CD133 is expressed in most, but not all, KMT2A-rearranged acute leukemia patients, and its presence seems to reflect the immunophenotype and/or cell of origin of the leukemia, as HSCs and early progenitors typically express PROM1/CD133, while more differentiated B-cell progenitors do not [29, 54, 55]. As acquiring resistance to DOT1L inhibition was accompanied by a complete loss of PROM1/CD133 expression, this may suggest that prolonged exposure to pinometostat triggered the differentiation towards (or selection of) a more mature immunophenotype. On the other hand, our data revealed that prolonged exposure of KMT2A-rearranged ALL cells to pinometostat seems to initiate a reprogramming process that involves the acquirement (or selection) of myeloid-like characteristics. Co-expression of myeloid CD markers, including CD33, [36] represents a familiar phenomenon in KMT2A-rearranged infant ALL with prognostic relevance [37, 38]. Moreover, a recent single-cell multiomics study by Chen and co-workers revealed the presence of pre-existing lymphomyeloid primed progenitors and myeloid blasts in diagnostic samples derived from KMT2A-rearranged B-ALL patients [56]. From this perspective, prolonged inhibition of DOT1L seems to favor KMT2A-rearranged leukemia cells that completely lack PROM1/CD133 but do display LILRB4/CD85k and CD33 expression. Interestingly, both LILRB4/CD85k and CD33 are therapeutic targets in AML [5763] and have shown potential as therapeutic vulnerabilities in KMT2A-rearranged ALL. Targeting LILRB4/CD85k with antibody-conjugates [57] or anti-LILRB4 CAR-T cells [64, 65] and/or CD33 with gemtuzumab ozogamicin, could prevent resistance to DOT1L inhibitors in KMT2A-rearranged ALL. Moreover, combining BCL-2 inhibition by venetoclax with agents targeting DOT1L, LILRB4/CD85k, and/or CD33 may enhance the efficacy of these drug combinations. Venetoclax was found to synergize with DOT1L inhibitors [25, 66] and is being evaluated in clinical trials for pediatric KMT2A-rearranged leukemias [67, 68].
Taken together, we present an in vitro model of acquired resistance to DOT1L inhibition in KMT2A-rearranged ALL, revealing selective loss of epigenetic regulation and gene expression of KMT2A-fusion target genes, accompanied by upregulation of myeloid-like characteristics. This study may not only impact the development of novel DOT1L inhibitors, but also reveal key characteristics of KMT2A-rearranged ALL cells that are able to evade therapy, providing therapeutic targets to prevent that.

Acknowledgements

We thank the members of the Stam group, as well as Jules P.P. Meijerink at the Princess Máxima Center for advice and discussions. We thank Utrecht Sequencing Facility for providing sequencing service and data. Utrecht Sequencing Facility is subsidized by the University Medical Center Utrecht, Hubrecht Institute, Utrecht University and The Netherlands X-omics Initiative (NWO project 184.034.019).

Declarations

Conflict of interest

T.A.M. and N.T.C. are paid consultants for and shareholders in Dark Blue Therapeutics Ltd. The other authors declare no competing financial interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Pieters R, De Lorenzo P, Ancliffe P, et al. 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(25):2246–56.PubMed Pieters R, De Lorenzo P, Ancliffe P, et al. 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(25):2246–56.PubMed
2.
Zurück zum Zitat Pieters R, Schrappe M, De Lorenzo P, et al. A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial. Lancet. 2007;370(9583):240–50.PubMed Pieters R, Schrappe M, De Lorenzo P, et al. A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial. Lancet. 2007;370(9583):240–50.PubMed
3.
Zurück zum Zitat Meyer C, Burmeister T, Groger D, et al. The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32(2):273–84.PubMed Meyer C, Burmeister T, Groger D, et al. The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32(2):273–84.PubMed
4.
Zurück zum Zitat Ernst P, Wang J, Korsmeyer SJ. The role of MLL in hematopoiesis and leukemia. Curr Opin Hematol. 2002;9(4):282–7.PubMed Ernst P, Wang J, Korsmeyer SJ. The role of MLL in hematopoiesis and leukemia. Curr Opin Hematol. 2002;9(4):282–7.PubMed
5.
Zurück zum Zitat Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7(11):823–33.PubMed Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7(11):823–33.PubMed
6.
Zurück zum Zitat Bernt KM, Armstrong SA. A role for DOT1L in MLL-rearranged leukemias. Epigenomics. 2011;3(6):667–70.PubMed Bernt KM, Armstrong SA. A role for DOT1L in MLL-rearranged leukemias. Epigenomics. 2011;3(6):667–70.PubMed
7.
Zurück zum Zitat Bernt KM, Zhu N, Sinha AU, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20(1):66–78.PubMedPubMedCentral Bernt KM, Zhu N, Sinha AU, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20(1):66–78.PubMedPubMedCentral
8.
Zurück zum Zitat Okada Y, Feng Q, Lin Y, et al. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121(2):167–78.PubMed Okada Y, Feng Q, Lin Y, et al. hDOT1L links histone methylation to leukemogenesis. Cell. 2005;121(2):167–78.PubMed
9.
Zurück zum Zitat Krivtsov AV, Feng Z, Lemieux ME, et al. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14(5):355–68.PubMedPubMedCentral Krivtsov AV, Feng Z, Lemieux ME, et al. H3K79 methylation profiles define murine and human MLL-AF4 leukemias. Cancer Cell. 2008;14(5):355–68.PubMedPubMedCentral
10.
Zurück zum Zitat Agraz-Doblas A, Bueno C, Bashford-Rogers R, et al. Unraveling the cellular origin and clinical prognostic markers of infant B-cell acute lymphoblastic leukemia using genome-wide analysis. Haematologica. 2019;104(6):1176–88.PubMedPubMedCentral Agraz-Doblas A, Bueno C, Bashford-Rogers R, et al. Unraveling the cellular origin and clinical prognostic markers of infant B-cell acute lymphoblastic leukemia using genome-wide analysis. Haematologica. 2019;104(6):1176–88.PubMedPubMedCentral
11.
Zurück zum Zitat Andersson AK, Ma J, Wang J, et al. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat Genet. 2015;47(4):330–7.PubMedPubMedCentral Andersson AK, Ma J, Wang J, et al. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat Genet. 2015;47(4):330–7.PubMedPubMedCentral
12.
Zurück zum Zitat Sanjuan-Pla A, Bueno C, Prieto C, et al. Revisiting the biology of infant t(4;11)/MLL-AF4 + B-cell acute lymphoblastic leukemia. Blood. 2015;126(25):2676–85.PubMedPubMedCentral Sanjuan-Pla A, Bueno C, Prieto C, et al. Revisiting the biology of infant t(4;11)/MLL-AF4 + B-cell acute lymphoblastic leukemia. Blood. 2015;126(25):2676–85.PubMedPubMedCentral
13.
Zurück zum Zitat Daigle SR, Olhava EJ, Therkelsen CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53–65.PubMedPubMedCentral Daigle SR, Olhava EJ, Therkelsen CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53–65.PubMedPubMedCentral
14.
Zurück zum Zitat Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122(6):1017–25.PubMedPubMedCentral Daigle SR, Olhava EJ, Therkelsen CA, et al. Potent inhibition of DOT1L as treatment of MLL-fusion leukemia. Blood. 2013;122(6):1017–25.PubMedPubMedCentral
15.
Zurück zum Zitat Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood. 2018;131(24):2661–9.PubMedPubMedCentral Stein EM, Garcia-Manero G, Rizzieri DA, et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood. 2018;131(24):2661–9.PubMedPubMedCentral
16.
Zurück zum Zitat Stein EM, Tallman MS. Mixed lineage rearranged leukaemia: pathogenesis and targeting DOT1L. Curr Opin Hematol. 2015;22(2):92–6.PubMed Stein EM, Tallman MS. Mixed lineage rearranged leukaemia: pathogenesis and targeting DOT1L. Curr Opin Hematol. 2015;22(2):92–6.PubMed
17.
Zurück zum Zitat Campbell CT, Haladyna JN, Drubin DA, et al. Mechanisms of Pinometostat (EPZ-5676) treatment-emergent resistance in MLL-Rearranged leukemia. Mol Cancer Ther. 2017;16(8):1669–79.PubMed Campbell CT, Haladyna JN, Drubin DA, et al. Mechanisms of Pinometostat (EPZ-5676) treatment-emergent resistance in MLL-Rearranged leukemia. Mol Cancer Ther. 2017;16(8):1669–79.PubMed
18.
Zurück zum Zitat Perner F, Gadrey JY, Xiong Y, et al. Novel inhibitors of the histone methyltransferase DOT1L show potent antileukemic activity in patient-derived xenografts. Blood. 2020;136(17):1983–8.PubMedPubMedCentral Perner F, Gadrey JY, Xiong Y, et al. Novel inhibitors of the histone methyltransferase DOT1L show potent antileukemic activity in patient-derived xenografts. Blood. 2020;136(17):1983–8.PubMedPubMedCentral
19.
Zurück zum Zitat Chen C, Zhu H, Stauffer F, et al. Discovery of Novel Dot1L inhibitors through a structure-based Fragmentation Approach. ACS Med Chem Lett. 2016;7(8):735–40.PubMedPubMedCentral Chen C, Zhu H, Stauffer F, et al. Discovery of Novel Dot1L inhibitors through a structure-based Fragmentation Approach. ACS Med Chem Lett. 2016;7(8):735–40.PubMedPubMedCentral
20.
Zurück zum Zitat van der Linden MH, Willekes M, van Roon E, et al. MLL fusion-driven activation of CDK6 potentiates proliferation in MLL-rearranged infant ALL. Cell Cycle. 2014;13(5):834–44.PubMedPubMedCentral van der Linden MH, Willekes M, van Roon E, et al. MLL fusion-driven activation of CDK6 potentiates proliferation in MLL-rearranged infant ALL. Cell Cycle. 2014;13(5):834–44.PubMedPubMedCentral
21.
Zurück zum Zitat Spijkers-Hagelstein JA, Pinhancos SS, Schneider P, Pieters R, Stam RW. Chemical genomic screening identifies LY294002 as a modulator of glucocorticoid resistance in MLL-rearranged infant ALL. Leukemia. 2014;28(4):761–9.PubMed Spijkers-Hagelstein JA, Pinhancos SS, Schneider P, Pieters R, Stam RW. Chemical genomic screening identifies LY294002 as a modulator of glucocorticoid resistance in MLL-rearranged infant ALL. Leukemia. 2014;28(4):761–9.PubMed
22.
Zurück zum Zitat Greil J, Gramatzki M, Burger R, et al. The acute lymphoblastic leukaemia cell line SEM with t(4;11) chromosomal rearrangement is biphenotypic and responsive to interleukin-7. Br J Haematol. 1994;86(2):275–83.PubMed Greil J, Gramatzki M, Burger R, et al. The acute lymphoblastic leukaemia cell line SEM with t(4;11) chromosomal rearrangement is biphenotypic and responsive to interleukin-7. Br J Haematol. 1994;86(2):275–83.PubMed
23.
Zurück zum Zitat Waters NJ. Preclinical pharmacokinetics and pharmacodynamics of Pinometostat (EPZ-5676), a first-in-Class, small molecule S-Adenosyl methionine competitive inhibitor of DOT1L. Eur J Drug Metab Pharmacokinet. 2017;42(6):891–901.PubMed Waters NJ. Preclinical pharmacokinetics and pharmacodynamics of Pinometostat (EPZ-5676), a first-in-Class, small molecule S-Adenosyl methionine competitive inhibitor of DOT1L. Eur J Drug Metab Pharmacokinet. 2017;42(6):891–901.PubMed
24.
Zurück zum Zitat Guenther MG, Lawton LN, Rozovskaia T, et al. Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia. Genes Dev. 2008;22(24):3403–8.PubMedPubMedCentral Guenther MG, Lawton LN, Rozovskaia T, et al. Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia. Genes Dev. 2008;22(24):3403–8.PubMedPubMedCentral
25.
Zurück zum Zitat Kerry J, Godfrey L, Repapi E, et al. MLL-AF4 spreading identifies binding Sites that are distinct from super-enhancers and that govern sensitivity to DOT1L inhibition in Leukemia. Cell Rep. 2017;18(2):482–95.PubMedPubMedCentral Kerry J, Godfrey L, Repapi E, et al. MLL-AF4 spreading identifies binding Sites that are distinct from super-enhancers and that govern sensitivity to DOT1L inhibition in Leukemia. Cell Rep. 2017;18(2):482–95.PubMedPubMedCentral
26.
Zurück zum Zitat Barzegar Behrooz A, Syahir A, Ahmad S. CD133: beyond a cancer stem cell biomarker. J Drug Target. 2019;27(3):257–69.PubMed Barzegar Behrooz A, Syahir A, Ahmad S. CD133: beyond a cancer stem cell biomarker. J Drug Target. 2019;27(3):257–69.PubMed
27.
Zurück zum Zitat Wu Y, Wu PY. CD133 as a marker for cancer stem cells: progresses and concerns. Stem Cells Dev. 2009;18(8):1127–34.PubMed Wu Y, Wu PY. CD133 as a marker for cancer stem cells: progresses and concerns. Stem Cells Dev. 2009;18(8):1127–34.PubMed
28.
Zurück zum Zitat Zhang Q, Shi S, Yen Y, Brown J, Ta JQ, Le AD. A subpopulation of CD133(+) cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett. 2010;289(2):151–60.PubMed Zhang Q, Shi S, Yen Y, Brown J, Ta JQ, Le AD. A subpopulation of CD133(+) cancer stem-like cells characterized in human oral squamous cell carcinoma confer resistance to chemotherapy. Cancer Lett. 2010;289(2):151–60.PubMed
29.
Zurück zum Zitat Godfrey L, Crump NT, O’Byrne S, et al. H3K79me2/3 controls enhancer-promoter interactions and activation of the pan-cancer stem cell marker PROM1/CD133 in MLL-AF4 leukemia cells. Leukemia. 2021;35(1):90–106.PubMed Godfrey L, Crump NT, O’Byrne S, et al. H3K79me2/3 controls enhancer-promoter interactions and activation of the pan-cancer stem cell marker PROM1/CD133 in MLL-AF4 leukemia cells. Leukemia. 2021;35(1):90–106.PubMed
30.
Zurück zum Zitat Mak AB, Nixon AM, Moffat J. The mixed lineage leukemia (MLL) fusion-associated gene AF4 promotes CD133 transcription. Cancer Res. 2012;72(8):1929–34.PubMed Mak AB, Nixon AM, Moffat J. The mixed lineage leukemia (MLL) fusion-associated gene AF4 promotes CD133 transcription. Cancer Res. 2012;72(8):1929–34.PubMed
31.
Zurück zum Zitat Adamaki M, Lambrou GI, Athanasiadou A, Vlahopoulos S, Papavassiliou AG, Moschovi M. HOXA9 and MEIS1 gene overexpression in the diagnosis of childhood acute leukemias: significant correlation with relapse and overall survival. Leuk Res. 2015;39(8):874–82.PubMed Adamaki M, Lambrou GI, Athanasiadou A, Vlahopoulos S, Papavassiliou AG, Moschovi M. HOXA9 and MEIS1 gene overexpression in the diagnosis of childhood acute leukemias: significant correlation with relapse and overall survival. Leuk Res. 2015;39(8):874–82.PubMed
32.
Zurück zum Zitat Hu YL, Fong S, Ferrell C, Largman C, Shen WF. HOXA9 modulates its oncogenic partner Meis1 to influence normal hematopoiesis. Mol Cell Biol. 2009;29(18):5181–92.PubMedPubMedCentral Hu YL, Fong S, Ferrell C, Largman C, Shen WF. HOXA9 modulates its oncogenic partner Meis1 to influence normal hematopoiesis. Mol Cell Biol. 2009;29(18):5181–92.PubMedPubMedCentral
33.
Zurück zum Zitat Dobrowolska H, Gill KZ, Serban G, et al. Expression of immune inhibitory receptor ILT3 in acute myeloid leukemia with monocytic differentiation. Cytometry B Clin Cytom. 2013;84(1):21–9.PubMed Dobrowolska H, Gill KZ, Serban G, et al. Expression of immune inhibitory receptor ILT3 in acute myeloid leukemia with monocytic differentiation. Cytometry B Clin Cytom. 2013;84(1):21–9.PubMed
34.
Zurück zum Zitat Costa AFO, Menezes DL, Pinheiro LHS, et al. Role of new immunophenotypic markers on prognostic and overall survival of Acute myeloid leukemia: a systematic review and Meta-analysis. Sci Rep. 2017;7(1):4138.PubMedPubMedCentral Costa AFO, Menezes DL, Pinheiro LHS, et al. Role of new immunophenotypic markers on prognostic and overall survival of Acute myeloid leukemia: a systematic review and Meta-analysis. Sci Rep. 2017;7(1):4138.PubMedPubMedCentral
35.
Zurück zum Zitat Churchill HRO, Fuda FS, Xu J, et al. Leukocyte immunoglobulin-like receptor B1 and B4 (LILRB1 and LILRB4): highly sensitive and specific markers of acute myeloid leukemia with monocytic differentiation. Cytometry B Clin Cytom. 2021;100(4):476–87.PubMed Churchill HRO, Fuda FS, Xu J, et al. Leukocyte immunoglobulin-like receptor B1 and B4 (LILRB1 and LILRB4): highly sensitive and specific markers of acute myeloid leukemia with monocytic differentiation. Cytometry B Clin Cytom. 2021;100(4):476–87.PubMed
36.
Zurück zum Zitat Hara J, Hosoi G, Okamura T, et al. CD33 + B-cell precursor acute lymphoblastic leukemia in children: a distinct subgroup of B-cell precursor acute lymphoblastic leukemia. Int J Hematol. 1995;61(2):77–84.PubMed Hara J, Hosoi G, Okamura T, et al. CD33 + B-cell precursor acute lymphoblastic leukemia in children: a distinct subgroup of B-cell precursor acute lymphoblastic leukemia. Int J Hematol. 1995;61(2):77–84.PubMed
37.
Zurück zum Zitat Stutterheim J, de Lorenzo P, van der Sluis IM, et al. Minimal residual disease and outcome characteristics in infant KMT2A-germline acute lymphoblastic leukaemia treated on the Interfant-06 protocol. Eur J Cancer. 2022;160:72–9.PubMed Stutterheim J, de Lorenzo P, van der Sluis IM, et al. Minimal residual disease and outcome characteristics in infant KMT2A-germline acute lymphoblastic leukaemia treated on the Interfant-06 protocol. Eur J Cancer. 2022;160:72–9.PubMed
38.
Zurück zum Zitat Stutterheim J, van der Sluis IM, de Lorenzo P, et al. Clinical implications of minimal residual disease detection in infants with KMT2A-Rearranged Acute Lymphoblastic Leukemia treated on the Interfant-06 protocol. J Clin Oncol. 2021;39(6):652–62.PubMedPubMedCentral Stutterheim J, van der Sluis IM, de Lorenzo P, et al. Clinical implications of minimal residual disease detection in infants with KMT2A-Rearranged Acute Lymphoblastic Leukemia treated on the Interfant-06 protocol. J Clin Oncol. 2021;39(6):652–62.PubMedPubMedCentral
39.
Zurück zum Zitat Laszlo GS, Estey EH, Walter RB. The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev. 2014;28(4):143–53.PubMed Laszlo GS, Estey EH, Walter RB. The past and future of CD33 as therapeutic target in acute myeloid leukemia. Blood Rev. 2014;28(4):143–53.PubMed
40.
Zurück zum Zitat van Dongen JJ, Orfao A, EuroFlow C, EuroFlow. Resetting leukemia and lymphoma immunophenotyping. Basis for companion diagnostics and personalized medicine. Leukemia. 2012;26(9):1899–907.PubMedPubMedCentral van Dongen JJ, Orfao A, EuroFlow C, EuroFlow. Resetting leukemia and lymphoma immunophenotyping. Basis for companion diagnostics and personalized medicine. Leukemia. 2012;26(9):1899–907.PubMedPubMedCentral
41.
Zurück zum Zitat van Dongen JJ, Lhermitte L, Bottcher S, et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia. 2012;26(9):1908–75.PubMedPubMedCentral van Dongen JJ, Lhermitte L, Bottcher S, et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia. 2012;26(9):1908–75.PubMedPubMedCentral
42.
Zurück zum Zitat Kalina T, Flores-Montero J, van der Velden VH, et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia. 2012;26(9):1986–2010.PubMedPubMedCentral Kalina T, Flores-Montero J, van der Velden VH, et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia. 2012;26(9):1986–2010.PubMedPubMedCentral
43.
Zurück zum Zitat Baguley BC. Multiple drug resistance mechanisms in cancer. Mol Biotechnol. 2010;46(3):308–16.PubMed Baguley BC. Multiple drug resistance mechanisms in cancer. Mol Biotechnol. 2010;46(3):308–16.PubMed
44.
Zurück zum Zitat Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5(3):219–34.PubMed Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov. 2006;5(3):219–34.PubMed
45.
Zurück zum Zitat Dillon R, Maycock S, Jackson A, et al. Venetoclax combined with low dose cytarabine compared to standard of care intensive chemotherapy for the treatment of favourable risk adult acute myeloid leukaemia (VICTOR): study protocol for an international, open-label, multicentre, molecularly-guided randomised, phase II trial. BMC Cancer. 2022;22(1):1174.PubMedPubMedCentral Dillon R, Maycock S, Jackson A, et al. Venetoclax combined with low dose cytarabine compared to standard of care intensive chemotherapy for the treatment of favourable risk adult acute myeloid leukaemia (VICTOR): study protocol for an international, open-label, multicentre, molecularly-guided randomised, phase II trial. BMC Cancer. 2022;22(1):1174.PubMedPubMedCentral
46.
Zurück zum Zitat Kadia TM, Reville PK, Wang X, et al. Phase II study of Venetoclax added to Cladribine Plus Low-Dose Cytarabine Alternating with 5-Azacitidine in older patients with newly diagnosed Acute myeloid leukemia. J Clin Oncol. 2022;40(33):3848–57.PubMed Kadia TM, Reville PK, Wang X, et al. Phase II study of Venetoclax added to Cladribine Plus Low-Dose Cytarabine Alternating with 5-Azacitidine in older patients with newly diagnosed Acute myeloid leukemia. J Clin Oncol. 2022;40(33):3848–57.PubMed
47.
Zurück zum Zitat Wei AH, Strickland SA Jr, Hou JZ, et al. Venetoclax Combined with Low-Dose Cytarabine for previously untreated patients with Acute myeloid leukemia: results from a phase Ib/II study. J Clin Oncol. 2019;37(15):1277–84.PubMedPubMedCentral Wei AH, Strickland SA Jr, Hou JZ, et al. Venetoclax Combined with Low-Dose Cytarabine for previously untreated patients with Acute myeloid leukemia: results from a phase Ib/II study. J Clin Oncol. 2019;37(15):1277–84.PubMedPubMedCentral
48.
Zurück zum Zitat Nguyen AT, He J, Taranova O, Zhang Y. Essential role of DOT1L in maintaining normal adult hematopoiesis. Cell Res. 2011;21(9):1370–3.PubMedPubMedCentral Nguyen AT, He J, Taranova O, Zhang Y. Essential role of DOT1L in maintaining normal adult hematopoiesis. Cell Res. 2011;21(9):1370–3.PubMedPubMedCentral
49.
Zurück zum Zitat Kuntimaddi A, Achille NJ, Thorpe J, et al. Degree of recruitment of DOT1L to MLL-AF9 defines level of H3K79 Di- and tri-methylation on target genes and transformation potential. Cell Rep. 2015;11(5):808–20.PubMedPubMedCentral Kuntimaddi A, Achille NJ, Thorpe J, et al. Degree of recruitment of DOT1L to MLL-AF9 defines level of H3K79 Di- and tri-methylation on target genes and transformation potential. Cell Rep. 2015;11(5):808–20.PubMedPubMedCentral
50.
Zurück zum Zitat Cao K, Ugarenko M, Ozark PA, et al. DOT1L-controlled cell-fate determination and transcription elongation are independent of H3K79 methylation. Proc Natl Acad Sci U S A. 2020;117(44):27365–73.PubMedPubMedCentral Cao K, Ugarenko M, Ozark PA, et al. DOT1L-controlled cell-fate determination and transcription elongation are independent of H3K79 methylation. Proc Natl Acad Sci U S A. 2020;117(44):27365–73.PubMedPubMedCentral
51.
Zurück zum Zitat Wu A, Zhi J, Tian T, et al. DOT1L complex regulates transcriptional initiation in human erythroleukemic cells. Proc Natl Acad Sci U S A. 2021;118:27. Wu A, Zhi J, Tian T, et al. DOT1L complex regulates transcriptional initiation in human erythroleukemic cells. Proc Natl Acad Sci U S A. 2021;118:27.
52.
Zurück zum Zitat Yi Y, Ge S. Targeting the histone H3 lysine 79 methyltransferase DOT1L in MLL-rearranged leukemias. J Hematol Oncol. 2022;15(1):35.PubMedPubMedCentral Yi Y, Ge S. Targeting the histone H3 lysine 79 methyltransferase DOT1L in MLL-rearranged leukemias. J Hematol Oncol. 2022;15(1):35.PubMedPubMedCentral
53.
Zurück zum Zitat Li D, Hu Y, Jin Z, et al. TanCAR T cells targeting CD19 and CD133 efficiently eliminate MLL leukemic cells. Leukemia. 2018;32(9):2012–6.PubMed Li D, Hu Y, Jin Z, et al. TanCAR T cells targeting CD19 and CD133 efficiently eliminate MLL leukemic cells. Leukemia. 2018;32(9):2012–6.PubMed
54.
Zurück zum Zitat Bueno C, Velasco-Hernandez T, Gutierrez-Aguera F, et al. CD133-directed CAR T-cells for MLL leukemia: on-target, off-tumor myeloablative toxicity. Leukemia. 2019;33(8):2090–125.PubMedPubMedCentral Bueno C, Velasco-Hernandez T, Gutierrez-Aguera F, et al. CD133-directed CAR T-cells for MLL leukemia: on-target, off-tumor myeloablative toxicity. Leukemia. 2019;33(8):2090–125.PubMedPubMedCentral
55.
Zurück zum Zitat O’Byrne S, Elliott N, Rice S, et al. Discovery of a CD10-negative B-progenitor in human fetal life identifies unique ontogeny-related developmental programs. Blood. 2019;134(13):1059–71.PubMed O’Byrne S, Elliott N, Rice S, et al. Discovery of a CD10-negative B-progenitor in human fetal life identifies unique ontogeny-related developmental programs. Blood. 2019;134(13):1059–71.PubMed
56.
Zurück zum Zitat Chen C, Yu W, Alikarami F, et al. Single-cell multiomics reveals increased plasticity, resistant populations, and stem-cell-like blasts in KMT2A-rearranged leukemia. Blood. 2022;139(14):2198–211.PubMedPubMedCentral Chen C, Yu W, Alikarami F, et al. Single-cell multiomics reveals increased plasticity, resistant populations, and stem-cell-like blasts in KMT2A-rearranged leukemia. Blood. 2022;139(14):2198–211.PubMedPubMedCentral
57.
Zurück zum Zitat Anami Y, Deng M, Gui X, et al. LILRB4-targeting antibody-drug conjugates for the treatment of Acute myeloid leukemia. Mol Cancer Ther. 2020;19(11):2330–9.PubMedPubMedCentral Anami Y, Deng M, Gui X, et al. LILRB4-targeting antibody-drug conjugates for the treatment of Acute myeloid leukemia. Mol Cancer Ther. 2020;19(11):2330–9.PubMedPubMedCentral
58.
Zurück zum Zitat Gui X, Deng M, Song H, et al. Disrupting LILRB4/APOE Interaction by an efficacious humanized antibody reverses T-cell suppression and blocks AML Development. Cancer Immunol Res. 2019;7(8):1244–57.PubMedPubMedCentral Gui X, Deng M, Song H, et al. Disrupting LILRB4/APOE Interaction by an efficacious humanized antibody reverses T-cell suppression and blocks AML Development. Cancer Immunol Res. 2019;7(8):1244–57.PubMedPubMedCentral
59.
Zurück zum Zitat Deng M, Chen H, Liu X, et al. Leukocyte immunoglobulin-like receptor subfamily B: therapeutic targets in cancer. Antib Ther. 2021;4(1):16–33.PubMedPubMedCentral Deng M, Chen H, Liu X, et al. Leukocyte immunoglobulin-like receptor subfamily B: therapeutic targets in cancer. Antib Ther. 2021;4(1):16–33.PubMedPubMedCentral
60.
Zurück zum Zitat Dhunputh C, Strullu M, Petit A et al. Single-dose (4.5 mg/m(2)) gemtuzumab ozogamicin in combination with fludarabine, cytarabine and anthracycline as reinduction therapy in relapsed or refractory paediatric acute myeloid leukaemia. Br J Haematol. 2022. Dhunputh C, Strullu M, Petit A et al. Single-dose (4.5 mg/m(2)) gemtuzumab ozogamicin in combination with fludarabine, cytarabine and anthracycline as reinduction therapy in relapsed or refractory paediatric acute myeloid leukaemia. Br J Haematol. 2022.
61.
Zurück zum Zitat de Rooij JD, Zwaan CM, van den Heuvel-Eibrink M, Pediatric AML. From Biology to Clinical Management. J Clin Med. 2015;4(1):127–49.PubMedPubMedCentral de Rooij JD, Zwaan CM, van den Heuvel-Eibrink M, Pediatric AML. From Biology to Clinical Management. J Clin Med. 2015;4(1):127–49.PubMedPubMedCentral
62.
Zurück zum Zitat Abuasab T, Rowe J, Tvito A. Emerging monoclonal antibody therapy for the treatment of Acute Lymphoblastic Leukemia. Biologics. 2021;15:419–31.PubMedPubMedCentral Abuasab T, Rowe J, Tvito A. Emerging monoclonal antibody therapy for the treatment of Acute Lymphoblastic Leukemia. Biologics. 2021;15:419–31.PubMedPubMedCentral
63.
Zurück zum Zitat Percival MM, Estey EH. Current treatment strategies for measurable residual disease in patients with acute myeloid leukemia. Cancer. 2019;125(18):3121–30.PubMed Percival MM, Estey EH. Current treatment strategies for measurable residual disease in patients with acute myeloid leukemia. Cancer. 2019;125(18):3121–30.PubMed
64.
Zurück zum Zitat John S, Chen H, Deng M, et al. A novel Anti-LILRB4 CAR-T cell for the treatment of monocytic AML. Mol Ther. 2018;26(10):2487–95.PubMedPubMedCentral John S, Chen H, Deng M, et al. A novel Anti-LILRB4 CAR-T cell for the treatment of monocytic AML. Mol Ther. 2018;26(10):2487–95.PubMedPubMedCentral
65.
Zurück zum Zitat Smith CHR, Xie J, Liu X, He Y, Ludwig K, Klesse L, Zhang C, John S. LILRB4 is a Novel Target for KMT2A rearranged Acute Leukemia. Blood; 2022. Smith CHR, Xie J, Liu X, He Y, Ludwig K, Klesse L, Zhang C, John S. LILRB4 is a Novel Target for KMT2A rearranged Acute Leukemia. Blood; 2022.
66.
Zurück zum Zitat Benito JM, Godfrey L, Kojima K, et al. MLL-Rearranged Acute Lymphoblastic Leukemias activate BCL-2 through H3K79 methylation and are sensitive to the BCL-2-Specific antagonist ABT-199. Cell Rep. 2015;13(12):2715–27.PubMedPubMedCentral Benito JM, Godfrey L, Kojima K, et al. MLL-Rearranged Acute Lymphoblastic Leukemias activate BCL-2 through H3K79 methylation and are sensitive to the BCL-2-Specific antagonist ABT-199. Cell Rep. 2015;13(12):2715–27.PubMedPubMedCentral
67.
Zurück zum Zitat Gibson A, Trabal A, McCall D et al. Venetoclax for Children and Adolescents with Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. Cancers (Basel). 2021;14(1). Gibson A, Trabal A, McCall D et al. Venetoclax for Children and Adolescents with Acute Lymphoblastic Leukemia and Lymphoblastic Lymphoma. Cancers (Basel). 2021;14(1).
68.
Zurück zum Zitat Winters AC, Maloney KW, Treece AL, Gore L, Franklin AK. Single-center pediatric experience with venetoclax and azacitidine as treatment for myelodysplastic syndrome and acute myeloid leukemia. Pediatr Blood Cancer. 2020;67(10):e28398.PubMed Winters AC, Maloney KW, Treece AL, Gore L, Franklin AK. Single-center pediatric experience with venetoclax and azacitidine as treatment for myelodysplastic syndrome and acute myeloid leukemia. Pediatr Blood Cancer. 2020;67(10):e28398.PubMed
Metadaten
Titel
Modelling acquired resistance to DOT1L inhibition exhibits the adaptive potential of KMT2A-rearranged acute lymphoblastic leukemia
verfasst von
Pauline Schneider
Nicholas T. Crump
Susan T.C.J.M. Arentsen-Peters
Alastair L. Smith
Rico Hagelaar
Fabienne R.S. Adriaanse
Romy S. Bos
Anja de Jong
Stefan Nierkens
Bianca Koopmans
Thomas A. Milne
Rob Pieters
Ronald W. Stam
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Experimental Hematology & Oncology / Ausgabe 1/2023
Elektronische ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-023-00445-8

Weitere Artikel der Ausgabe 1/2023

Experimental Hematology & Oncology 1/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.