Leukemia https://doi.org/10.1038/s41375-018-0327-2

ARTICLE

Acute myeloid leukemia Dasatinib and navitoclax act synergistically to target NUP98-NSD1 +/FLT3-ITD+ acute myeloid leukemia

1 2 1 3,4 3 Jarno L. Kivioja ● Angeliki Thanasopoulou ● Ashwini Kumar ● Mika Kontro ● Bhagwan Yadav ● 1 1 1 2 3,4 Muntasir M. Majumder ● Komal K. Javarappa ● Samuli Eldfors ● Juerg Schwaller ● Kimmo Porkka ● Caroline A. Heckman1

Received: 28 June 2018 / Revised: 21 September 2018 / Accepted: 10 October 2018 © Springer Nature Limited 2018

Abstract Acute myeloid leukemia (AML) with co-occurring NUP98-NSD1 and FLT3-ITD is associated with unfavorable prognosis and represents a particularly challenging treatment group. To identify novel effective therapies for this AML subtype, we screened patient cells and engineered models with over 300 compounds. We found that mouse hematopoietic progenitors co-expressing NUP98-NSD1 and FLT3-ITD had significantly increased sensitivity to FLT3 and MEK-inhibitors compared to cells expressing either aberration alone (P<0.001). The cells expressing NUP98-NSD1 alone had significantly P = NUP98-NSD1+/FLT3- +

1234567890();,: 1234567890();,: increased sensitivity to BCL2-inhibitors ( 0.029). Furthermore, ITD patient cells were also very sensitive to BCL2-inhibitor navitoclax, although the highest select sensitivity was found to SRC/ABL-inhibitor dasatinib + (mean IC50 = 2.2 nM). Topoisomerase inhibitor mitoxantrone was the least effective drug against NUP98-NSD1 /FLT3-ITD + AML cells. Of the 25 significant hits, four remained significant also compared to NUP98-NSD1-/FLT3-ITD+ AML patients. We found that SRC/ABL-inhibitor dasatinib is highly synergistic with BCL2-inhibitor navitoclax in NUP98-NSD1 +/FLT3-ITD+ cells. analysis supported the potential relevance of dasatinib and navitoclax by revealing significantly higher expression of BCL2A1, FGR, and LCK in NUP98-NSD1+/FLT3-ITD+ patients compared to healthy CD34+ cells. Our data suggest that dasatinib–navitoclax combination may offer a clinically relevant treatment strategy for AML with NUP98-NSD1 and concomitant FLT3-ITD.

Introduction recurrent fusion gene involving nucleoporin-98-kDa (NUP98) at 11p15.5 was first described more than two Acute myeloid leukemia (AML) is a complex disease decades ago and since then at least 32 distinct NUP98 characterized by various chromosomal alterations [1]. A fusion partner genes have been identified [2–6]. Regardless of the fusion partner, disruption of NUP98 leads to changes in the stoichiometry and nucleo-cytoplasmic transport functions of the nuclear pore complex [7]. While NUP98 Supplementary information The online version of this article (https:// rearrangements form the second largest gene fusion network doi.org/10.1038/s41375-018-0327-2) contains supplementary fi material, which is available to authorized users. in AML, they also de ne a biologically distinct subgroup of patients with poor prognosis [8–10]. In this study, we * Caroline A. Heckman focused on the t(5;11)(q35;p15.4) translocation [11, 12], fi caroline.heckman@helsinki. which is the most common NUP98 rearrangement in cyto- 1 Institute for Molecular Medicine Finland, Helsinki Institute of Life genetically normal AML. According to Hollink et al. [13], Science, University of Helsinki, Helsinki, Finland the resulting NUP98-NSD1 is present in 16% of pediatric 2 Department of Biomedicine, University Children’s Hospital, and 2% of adult cytogenetically normal AML patients with University of Basel, Basel, Switzerland over 80% also carrying activating FLT3-ITD (internal tan- NUP98- 3 Hematology Research Unit Helsinki, University of Helsinki, dem duplication) mutations. Co-occurrence of Helsinki, Finland NSD1 and FLT3-ITD is associated with especially low – 4 Department of Hematology, Helsinki University Hospital complete remission and survival rates in AML [13 17]. Comprehensive Cancer Center, Helsinki, Finland During a three-year follow-up, the event-free survival rate J. L. Kivioja et al. among NUP98-NSD1+/FLT3-ITD+ patients was shown to epigenetic modifier genes IDH1/2, NPM1,orMLL-rear- be 14% [14]. Although, allogeneic hematopoietic stem cell rangements. BM mononuclear cells (MNCs) were enriched transplantation may improve survival [14–16], no evidence by gradient centrifugation (Ficoll-Paque Premium™,GE regarding the efficacy of specific therapeutic agents exists Healthcare, Little Chalfont, United Kingdom) and either for these patients. cryopreserved until analysis or immediately suspended in The main objective of the present study was to discover Mononuclear Cell Medium (MCM; PromoCell, Heidelberg, novel candidate drugs and drug combinations for NUP98- Germany) supplemented with 0.5 µg/ml gentamicin and 2.5 NSD1+/FLT3-ITD+ AML patients and to understand the µg/ml amphotericin B. From AML patients with multiple contribution of NUP98-NSD1 to AML drug response. We samples, sample with the highest leukemic burden was utilized a standardized high-throughput drug sensitivity and included. Germline controls (skin biopsies) were collected resistance testing assay [18–20] for analyzing hematopoietic from two of the NUP98-NSD1+/FLT3-ITD+ patients for mouse cell lines transduced with NUP98-NSD1 and FLT3- exome sequencing. Table 1 describes clinical and pheno- ITD retroviruses independently, or co-transduced with both. typic features of the NUP98-NSD1+/FLT3-ITD+ study Results from the experimental cell lines were supported by patients (data lock 7 September 2018). similar drug sensitivity assessment of bone marrow (BM) cells from NUP98-NSD1+/FLT3-ITD+ and NUP98-NSD1-/ Mouse BM-derived cells FLT3-ITD+ AML patients. RNA sequencing was per- formed on experimental mouse cells and patient cells to Lineage marker-depleted BM cells from BALB/c mice were understand gene expression changes induced by NUP98- transduced with empty vector, NUP98-NSD1, and FLT3- NSD1 and FLT3-ITD that may associate with the drug ITD encoding retroviruses alone or co-transduced with responses. NUP98-NSD1 and FLT3-ITD as previously described [17]. We found that NUP98-NSD1+/FLT3-ITD+ patient cells The (“preleukemic”) cells were expanded in RPMI-1640 exhibit significantly high sensitivity to multi-target (Lonza, Basel, Switzerland) complete medium (10% fetal inhibitor (TKI) dasatinib and pan-BCL2 inhibitor bovine serum, 1% L-glutamine, and 1% penicillin- navitoclax. Genes encoding dasatinib target LCK streptomycin (10,000 U/ml each) supplemented with and FGR, and navitoclax target BCL2A1 were significantly interleukin-6 (10 ng/ml), mouse interleukin-3 (6 ng/ml), and upregulated in NUP98-NSD1+/FLT3-ITD+ AML cells mouse stem cell factor (10 ng/ml) (Peprotech, Rocky Hill, relative to healthy CD34+ cells. By screening experimental NJ, USA), or propagated in vivo (“leukemic”)byBM mouse cell lines, we observed that co-expression of NUP98- reconstitution in irradiated syngeneic recipient mice as NSD1 and FLT3-ITD induces a significant increase in previously described [17]. Additional cell lines included Ba/ selective FLT3 and MEK inhibitor responses compared to F3 cells (ATCC CRL-12015) transduced with lentiviruses cells expressing FLT3-ITD alone. We also found that carrying either empty backbone vector (pLeGO-iCer2) or dasatinib and navitoclax are highly synergistic in leukemic pLeGO-iCer2-NUP98-NSD1 (Supplementary Figure 1). NUP98-NSD1+/FLT3-ITD+ human and mouse cells, sug- The lentiviral packaging, transduction, and cell selection gesting targeted therapies may be available for this clini- methods are described in the Supplementary information cally challenging subtype of AML. file.

Compound collection Materials and methods The compound collection included 306 drugs including Patient material approved drugs (U.S. Food and Drug Administration or European Medicines Agency), investigational compounds The study was approved by the local institutional review and probes (Supplementary Table 1). The compounds were board at the Helsinki University Hospital (permit numbers reformatted in 100% dimethyl sulfoxide or water and plated 239/13/03/00/2010, 303/13/03/01/2011, Helsinki Uni- in five concentrations over a 10,000-fold range (e.g., 1– versity Hospital Ethics Committee). The clinical samples 10,000 nM) on 384-well flat clear bottom white were collected after informed consent and in accordance polystyrene tissue culture treated microplates (Corning, with the Declaration of Helsinki. BM aspirates were New York, NY, USA) using an Echo 550 acoustic collected from all available Finnish NUP98-NSD1+/FLT3- dispenser (Labcyte Inc, Sunnyvale, CA, USA). The ITD+ AML patients. Control samples were collected from microplates were kept in pressurized StoragePods® (Roylan ten healthy donors and nine NUP98-NSD1-/FLT3-ITD+ Developments Ltd., Fetcham, United Kingdom) under inert AML patients, which lacked concurrent mutations in nitrogen gas until use. Dasatinib and navitoclax act synergistically to target. . .

Drug sensitivity and resistance testing , + The plated compounds were resuspended by adding 5 µl of fl

weak (36%), culture medium to each well and brie y shaking the plates

+ with a Titramax 1000 platform shaker (Heidolph, Schwa- bach, Germany). Afterwards, 20 µl of cell suspension – , CD19-, CD7-, CD9-, (2000 10,000 cells/well) was added to the wells with a , CD15; a proportion of cells + (70%), CD4 + Multidrop™ Combi Reagent Dispenser (Thermo Fisher + ] , (weak 24%), CD3-, CD15 + 58 fi

, MPO Scienti c, Carlsbad, CA, USA). The plates were incubated , CD4 + +

, CD25 for 72 h in a humidified incubator at +37°C. Cell viabilities + , CD4-, CD3-, CD13- (26%), CD41a, CD42a&61-, CD42b-, TdT + , CD135 were measured before and after the incubation using Cell- + + , CD64

+ Titer-Glo® reagent according to the manufacturer’s instructions (Promega, Madison, WI, USA). The lumines- , CD117 (21%) (26%), HLA-DR , CD14 ® + +

+ cence from each well was captured with a PHERAstar FS + , CD19-, CD7 + , CD133-, CD2 microplate reader (BMG Labtech, Offenburg, Germany) + , CD22 , CD65 and normalized to negative (dimethyl sulfoxide) (n = 16) , + + , MPO , HLA-DR + , CD56-, CD203c, CD123 World Health Organization [ n = + + and positive (100 µmol/L benzethonium chloride) ( 8) , CD36 + in gDNA calculated by comparing peak areas generated with +

] ’

, CD15 control wells. The threshold for plate-wise Z values was + 57 WHO

, HLA-DR – FLT3 , CD11b weak (16%), CD11b 0.5 1.0 [21]. The dose-response curves were generated in + , HLA-DR + + (28%), MPO sex,

+ Dotmatics Browser v4.8 (Dotmatics Ltd., Herts, United + S Kingdom) and fitted through a four parametric logistic fit , CD13 , CD13 , CD11b + + +

, CD117 function that considers top and bottom asymptote, slope, + and the inflection point. To improve the interpretation of weak (66%), CD135 , CD38 , CD79-, TdT , CD38 , CD13 + + + +

+ drug-sample interactions, dose-response curves were con- -ITD and wild type

overall survival, verted to a drug sensitivity score (DSS) as described [22]. , IREM2-, CD14-, CD64-, CD71 (48%), CD33 , CD38 , CD33 , CD65 , CD33 + +

FLT3 DSS is a normalized area under the curve, calculated rela- OS + + + + + tive to total area between 10 and 100% inhibition. The (16%), CD96 CD105 + CD34 BM immunophenotype CD177 CD36 cyCD79a-, NG2-, cyCD3-, CD16-, CD117 male, selective drug sensitivity score (sDSS) was calculated for

M each drug by subtracting the median DSS of control sample a = 5.4 8.1 CD33 from the DSS of experimental sample (sDSS DSSsample- (months) 53.3* CD34 12.1 CD34

female, DSScontrol). Pilot combinatorial screens were performed by F treating experimental cell lines with 70 drugs ± dasatinib

. Allelic Ratio between (100 nM). The comprehensive follow-up screening was patients A.R

+ performed using 8 × 8 or 6 × 6 drug concentration matrices. The zero-interaction potency (ZIP) synergy scores (δ) were calculated in SynergyFinder [23, 24]. Allo-HSCT (planned) Therapy OS inhibitor allo-HSCT, azacitidine, DLI allo-HSCT, re-induction

NUP98-NSD1 Gene expression profiling donor leukocyte infusion, FLT3- ITD A. R.

DLI Paired-end RNA sequencing (RNA-Seq) was performed on total RNA (1–3 µg) from experimental BALB/c cell lines, BM MNCs of three AML patients with NUP98-NSD1 +/FLT3- + NUP98 NSD1-/FLT3- +

FLT3-ITD length (bp) ITD , nine - ITD AML patients, and BM CD34+ cells from four healthy donors. The CD34+ cells were enriched with the EasySep® Human + 70 57 0.346 Induction, consolidation, BM blast count (%) 75 57 0.182 Induction x2, TKI, MTOR 40 213 0.310 Induction, consolidation, 95 57 0.403 Induction, consolidation, patients belong to adverse cytogenetic risk group based on European LeukemiaNet (2017) criteria [ CD34 Selection Kit (StemCell Technologies, Vancouver,

+ Canada). RNA extractions were done using the RNeasy

allogeneic hematopoietic stem cell transplantation, Mini Kit or miRNeasy Kit (Qiagen, Hilden, Germany), cation fi while RNA quality was assessed with the 2100 Bioanalyzer AML with maturation WHO classi Acute monocytic leukemia AML with maturation Acute monocytic leukemia using the Agilent RNA 6000 or Pico Kit (Agilent Tech- NUP98-NSD1 Clinical and phenotypic characteristics of the 32 A 54 39 58 allo-HSCT nologies, Santa Clara, CA, USA). RNA concentrations were measured with the Qubit fluorometer (Thermo Fisher Sci- age,

Patient remains in complete remission at data lock fi 7499 F/ fragment analysis (supplemental information), A Table 1 Patient S/ All four a 600 M/ 3600 F/ 3660 M/ enti c). Subsequently, RNA was depleted of ribosomal J. L. Kivioja et al.

RNAs (Ribo-Zero rRNA Removal Kit; Epicentre, Madison, NSD1+ BALB/c and Ba/F3 cells (mean sDSS = 17.5). WI, USA), purified (RNeasy® MinElute® Cleanup Kit, Interestingly, the leukemic NUP98-NSD1+/FLT3-ITD+ Qiagen), and reverse transcribed to double-stranded cDNA cells were resistant (sDSS < 0) to BCL2 inhibitors (navito- with the Superscript® Double-Stranded cDNA Synthesis Kit clax; sDSS = −7.9, obatoclax; sDSS = −6.0). Although the (Thermo Fisher Scientific) using random hexamers (New mechanism of resistance remains unclear, it may arise from England BioLabs, Ipswich, MA, USA). RNA-Seq libraries the ability of FLT3-ITD to up-regulate MCL1 through its were prepared with Nextera (Epicentre), and ScriptSeq STAT5-docking domains [29]. In contrast to BCL2 inhi- technologies (Illumina, San Diego, CA, USA), bar-coded, bitor resistance, the dual positive cells had significantly (P and enriched by ligation PCR according to the manu- < 0.01) increased selective sensitivity to FLT3 and MEK facturer’s instructions (Illumina). PCR fragments were size inhibitors (Fig. 1b), with quizartinib (mean sDSS = 22.8) selected (350–700 bp) from a 2% agarose gel, purified with and pimasertib (mean sDSS = 13.2) being the most effec- the QIAquick Gel Extraction Kit (Qiagen), and sequenced tive FLT3 and MEK inhibitors (Fig. 1c). High MEK inhi- with an Illumina HiSeq™2000 or HiSeq™2500 instrument. bitor sensitivity in the NUP98-NSD1+/FLT3-ITD+ cells Sequenced reads were aligned against human and mouse may result from co-activation of STAT5 and MAPK path- (BALB/c) reference genomes. The spliced alignment of the way downstream of FLT3 signaling as reported [30]. processed paired-end reads from human samples was per- Consistent with earlier studies and drug testing results, our formed against the reference genome (Ensembl GRCh38) phospho-flow analysis showed increased with guidance of the Ensembl reference gene models of STAT5 and ERK1/2 in NUP98-NSD1+/FLT3-ITD+ (Ensembl v82) with the gap-aware STAR aligner [25]. Raw BALB/c BM cells compared to cells expressing either of the read counts were normalized by trimmed mean of M values two aberrations alone (Supplementary Figure 2). The drug scaling normalization method and log2 CPM (counts per screening data of all experimental mouse cell lines are million) values calculated with the edgeR R package shown in Supplementary Table 2 and Supplementary (3.18.1) [26]. The RNA-Seq data preprocessing and ana- Figure 3. lysis pipeline have been previously described [27, 28]. Dasatinib sensitivity is highly specific for NUP98- Statistical analysis NSD1+/FLT3-ITD+ AML cells

Statistical analyses were performed in GraphPad Prism In addition to the mouse cell lines, we evaluated drug v6.01 (GraphPad Software, La Jolla, CA, USA). DSS sensitivity profiles of BM MNCs from four NUP98-NSD1 comparisons were computed with the Mann–Whitney U test +/FLT3-ITD+ AML patients and ten healthy donors (Fig. 2 and correlation analyses with the Spearman’s rank correla- and Supplementary Table 3). Of the 171 drugs tested in all tion coefficient test. Gene expression values were compared samples, 99 (57.9%) had a higher median DSS in the using the unpaired two-sample t tests. All statistical tests NUP98-NSD1+/FLT3-ITD+ BM MNCs compared to heal- applied in this study were two-tailed and P values below thy BM MNCs indicating select targeting of the leukemic 0.05 were considered significant. cell, 51 (30%) had no response (DSS = 0) in either group, and 21 (12.9%) had a higher median DSS in the healthy BM MNCs (Supplementary Figure 4). We identified 25 statisti- Results cally significant differences between the NUP98-NSD1 +/FLT3-ITD+ AML patients and healthy controls (Table 2). Molecular driven responses to BCL2, FLT3, and MEK The most selective drugs included kinase inhibitors, BCL2 inhibitors in experimental mouse cell lines inhibitor navitoclax, four HDAC inhibitors, and three heat shock 90 (HSP90) inhibitors. Of the top candidate To discover small-molecule inhibitors with activity against drugs, SRC-family inhibitor dasatinib had the NUP98-NSD1, we tested lineage marker-depleted BALB/c lowest mean half maximal inhibitory concentration (IC50 = BM-derived cells expressing NUP98-NSD1 and FLT3-ITD 2.2 nM, 95% CI -1.4–5.8 nM). The topoisomerase II inhi- alone or both together as well as NUP98-NSD1+ Ba/F3 bitor mitoxantrone (P < 0.01) was the least effective drug cells with 306 drugs (Fig. 1a). By comparing the selective against the NUP98-NSD1+/FLT3-ITD+ AML patient cells drug sensitivities (sDSS) between cell lines, we discovered (Table 2). Two of the patients (600 and 3660) had pre- that NUP98-NSD1+ BALB/c BM cells have significantly viously been exposed to a combination of cytarabine and (P = 0.029) higher select sensitivity to BCL2 inhibitors idarubicin, thus the tested cells were likely representative of compared to leukemic cells co-expressing NUP98-NSD1 a drug resistant clone that had been selected for by that and FLT3-ITD (Fig. 1b). Navitoclax was identified as the treatment. Nevertheless, treatment naïve cells from patient most selective BCL2 inhibitor (n = 4) towards NUP98- 3600 and 7499 showed comparable resistance to Dasatinib and navitoclax act synergistically to target. . .

Fig. 1 Drug testing results from experimental mouse cell lines. At the inhibitors in each experimental mouse cell line. Significant differences hit identification stage, each experimental cell line was tested once between the cell lines are denoted with an asterisk (*P = 0.029; ****P with a library of 306 compounds. a The histograms show the 25 most < 0.0001). c The lower panel shows dose-response curves of the most effective drugs in Ba/F3 and BALB/c BM-derived cells expressing selective BCL2 inhibitor (navitoclax) in the NUP98-NSD1+ BALB/c NUP98-NSD1 [1, 2], FLT3-ITD [3], or both aberrations together cells, and the most selective FLT3 (quizartinib) and MEK inhibitor [4, 5]. The preleukemic cells were cultured in vitro while the leukemic (pimasertib) in the dual positive BALB/c cells. Drug concentrations cells were propagated in vivo. b The box plots show the individual are shown on the x-axis and inhibition percentage on the y-axis data points and mean sDSS of several BCL2, FLT3, and MEK topoisomerase II inhibitors suggesting that NUP98-NSD1 tivozanib (P = 0.03) had significantly higher DSS in +/FLT3-ITD+ AML cells may be intrinsically resistant to NUP98-NSD1+/FLT3-ITD+ cells compared to NUP98- this class of drugs. Additionally, cells from the index patient NSD1-/FLT3-ITD+ cells, while mitoxantrone (P = 0.007) (600) showed high resistance to nucleoside analogues such had significantly lower DSS (Fig. 3). This provides addi- as clofarabine, cladribine, and fludarabine. tional support that topoisomerase II inhibitor resistance may To evaluate the differential responses further, we com- be specific for NUP98-NSD1+/FLT3-ITD+ AML. pared drug sensitivities between NUP98-NSD1+/FLT3-ITD As NUP98/11p15 translocations were recently shown to + and NUP98-NSD1-/FLT3-ITD+ AML patients. Of the 25 affect CD34+ hematopoietic precursor cells in myeloid drugs, belinostat (P = 0.02), dasatinib (P = 0.03), and neoplasms [31], we also assessed the impact of BM blast J. L. Kivioja et al.

Fig. 2 Drug testing results from patient cells. The waterfall plots show patient. BM aspirates from the patients were collected at relapse (R) or the 20 most and least selective drug responses in BM MNCs of four at diagnosis (D). The index patient’s (600) BM MNCs were tested NUP98-NSD1+/FLT3-ITD+ AML patients in comparison to healthy with 171 drugs, whereas the cells from patient 3600, 3660, and 7499 donor BM MNCs. The sDSS was calculated for each drug by sub- were tested with 306 drugs. Drugs denoted with an asterisk were not tracting the median DSS of ten healthy donors from the DSS of a tested in the index case count on DSS. A trend towards positive correlation was navitoclax (patient 600) (56% inhibition) or 30 nM navito- found for SRC/ABL inhibitor dasatinib (R = 0.800) indi- clax (patient 7499) (61.8% inhibition). Maximum inhibition cating that blast content may explain some of the variation (100%) was achieved at an area of ≥3 nM dasatinib com- in DSS between the NUP98-NSD1+/FLT3-ITD+ patients. bined with ≥1000 nM of navitoclax. In leukemic mouse In NUP98-NSD1-/FLT3-ITD+ BM MNCs the correlation cells, the highest synergy was achieved at 3 nM dasatinib coefficient was 0.25 (Supplementary Figure 5). combined with 300 nM navitoclax and maximum inhibition (74%) achieved at 10 nM dasatinib combined with 10,000 Dasatinib–navitoclax combination is synergistic in nM navitoclax. leukemic cells co-expressing NUP98-NSD1 and FLT3- ITD NUP98-NSD1+/FLT3-ITD+ BM MNCs show increased expression of LCK, FGR, and BCL2A1 compared to To identify drugs that could synergize with dasatinib for healthy CD34+ cells greater efficacy, we performed a pilot combinatorial screen with the mouse cell lines using 70 drugs ± dasatinib (100 NUP98-NSD1 has been reported to enforce the nM) (Fig. 4a). Interestingly, dasatinib had the strongest constitutive expression of distinct HOX-AB cluster genes impact on DSS in NUP98-NSD1+/FLT3-ITD+ mouse cells [9, 13, 16, 32, 33]. Considering this, we initially analyzed as indicated by the slope of linear regression line (Supple- the expression of several HOX-AB genes in patient cells and mentary Figure 6). In the dual positive cells, dasatinib experimental cell lines to validate our samples. Consistent significantly increased sensitivity to FLT3 and MEK inhi- with earlier studies, we observed high expression of HOX- bitors. Moreover, a modest increase in BCL2 inhibitor A9, HOX-A10, HOX-B3, -B4, -B5, -B6, and -B7 genes in the sensitivities was observed in the cells expressing NUP98- NUP98-NSD1+/FLT3-ITD+ BM MNCs compared to heal- NSD1 alone (Fig. 4 and Supplementary Table 4). To follow- thy CD34 + cells (Fig. 6a). In BALB/c BM-derived cells, up on these responses, we selected eleven drug pairs for NUP98-NSD1 alone induced expression of several HOX-AB more comprehensive evaluation using dose-response genes, which remained upregulated in the dual positive cells matrices (Supplementary Table 5-6). The combination of (Fig. 6b). dasatinib and navitoclax had the highest synergistic activity Subsequently, we analyzed the expression of dasatinib in patient cells (patient 600: δ = 14.0; patient 7499: δ = 6.1) and navitoclax target genes in primary patient cells and and leukemic BALB/c BM-derived cells (δ = 9.4) co- experimental cell lines (Fig. 6c–f). Intriguingly, we expressing NUP98-NSD1 and FLT3-ITD (Fig. 5). In observed a significant increase in expression of the dasatinib patient cells, the highest synergy was achieved at an area targets LCK (P = 0.005) and FGR (P < 0.001) and the where 0.1 nM dasatinib was combined with 300 nM navitoclax target BCL2A1 (P = 0.028) in NUP98-NSD1 Dasatinib and navitoclax act synergistically to target. . .

Table 2 Differential analysis of Drug name Drug target Phase of DSS median 95%CI DSS 95% CI DSS P value drug sensitivities in BM MNCs clinical difference (patients) (healthy) of NUP98-NSD1+/FLT3-ITD+ study patients (n = 4) compared to healthy donors (n = 10) (P < Sensitive AZD8055 mTOR Phase 1 20.54 (4.1, 38.9) (1.1, 5.6) 0.004 0.01) Refametinib MEK1/2 Phase 1/2 19.60 (8.3, 26.8) (0.0, 3.0) 0.002 Tivozanib FLT3, VEGFR1-3, Phase 2 18.66 (11.5, 28.9) (−0.3, 6.4) 0.001 c-Kit, PDGFR Idelalisib PI3K, p110 δ- Phase 3 17.35 (4.4, 32.5) (1.1, 3.9) 0.004 selective Pimasertib MEK1/2 Phase 2 17.12 (6.2, 24.7) (0.3, 3.2) 0.002 Dasatinib ABL1, c-KIT, SRC, Approved 16.27 (11.9, 23.2) (−0.2, 7.1) 0.003 LCK BIIB021 HSP90 Phase 2 16.22 (21.5, 27.8) (7.0, 9.8) 0.002 Navitoclax BCL2, BCL-xL, Phase 2 12.37 (12.1, 41.1) (9.6, 12.8) 0.002 BCL-w, BCL2A1, MCL1 Lestaurtinib FLT3, JAK2, TrkA- Phase 3 11.56 (9.6, 20.4) (3.0, 6.6) 0.002 C Doramapimod pan-p38 MAPK, c- Phase 1/2 11.26 (4.9, 19.9) (0.0, 3.6) 0.001 RAF, FYN, LCK Foretinib MET, VEGFR2 Phase 2 10.83 (3.1, 25.8) (2.9, 5.0) 0.004 AZD7762 Chk1 Probe 10.27 (7.5, 24.3) (5.3, 8.3) 0.008 Luminespib HSP90 Phase 2 10.19 (21.4, 26.7) (11.5, 15.8) 0.002 OSI-027 mTOR Phase 1 9.94 (6.7, 18.7) (0.6, 4.2) 0.002 Linsitinib IGF1R, IR Phase 2 9.47 (3.2, 17.0) (0.6, 3.3) 0.004 Tanespimycin HSP90 Phase 2 9.19 (12.6, 24.3) (7.0, 9.6) 0.002 Everolimus mTOR Approved 8.23 (2.8, 14.0) (−0.5, 2.6) 0.002 Belinostat HDAC Phase 2 7.24 (20.9, 29.3) (15.3, 18.3) 0.002 Axitinib VEGFR, PDGFR, Approved 5.21 (2.6, 14.9) (1.6, 4.3) 0.004 KIT Danusertib Aurora kinase A-C, Phase 2 4.94 (10.6, 13.5) (4.7, 8.1) 0.002 Abl, c-RET, LCK Entinostat HDAC Phase 2 4.93 (2.5, 20.4) (3.9, 5.8) 0.004 Pictilisib PI3Kα/δ Phase 2 4.47 (8.9, 16.7) (6.5, 8.5) 0.002 Panobinostat HDAC Phase 3 4.20 (22.9, 30.3) (19.4, 22.6) 0.002 Vorinostat HDAC Approved 4.19 (9.0, 15.4) (5.8, 9.0) 0.002 Resistant Mitoxantrone Topoisomerase II Approved −6.27 (−2.0, 5.8) (5.9, 8.6) 0.008

+/FLT3-ITD+ BM MNCs compared to healthy CD34+ Discussion cells. Moreover, in the BALB/c BM cells expressing NUP98-NSD1 alone, BCL2A1 expression (5.4) was higher In this study, our objective was to identify novel candidate compared to BALB/c BM cells transduced with mock (0.0) drugs and drug combinations for the treatment of AML or FLT3-ITD alone (2.5). The BCL2 inhibitor resistant patients with concomitant NUP98-NSD1 and FLT3-ITD NUP98-NSD1+/FLT3-ITD+ BALB/c cells had the highest alterations by high-throughput drug screening and mole- MCL1 expression and the lowest BCL2 expression com- cular profiling. NUP98-NSD1+/FLT3-ITD+ AML patients pared to BCL2 inhibitor sensitive NUP98-NSD1+ BALB/c are a high-risk patient group that typically have reduced cells. This finding may suggest that co-expression of event-free survival, often develop chemo refractory disease, NUP98-NSD1 and FLT3-ITD leads to BCL2 inhibitor and are in need of novel treatment options. Here, we pro- resistance via upregulation of MCL1 and repression of vide evidence that NUP98-NSD1+/FLT3-ITD+ BM MNCs BCL2. Additionally, to investigate protein level expression have significantly increased select sensitivity to multi- of SRC-family in NUP98-NSD1+/FLT3-ITD+ cells, kinase inhibitor dasatinib and BCL2 inhibitor navitoclax we analyzed phosphokinase antibody array results pre- and that these cells are resistant to topoisomerase II inhi- viously performed on the index patient (600) [18]. We bitors. We demonstrate that combined treatment of these found that FYN and LCK had increased phosphorylation cells with dasatinib and navitoclax is highly synergistic. Our compared to healthy controls in all three samples collected data suggest that increased expression of LCK, FGR, and from the index patient at different stages of disease pro- BCL2A1 may play important roles in sensitizing NUP98- gression (Supplementary Figure 7). NSD1+/FLT3-ITD+ AML cells to SRC/ABL inhibitor J. L. Kivioja et al.

identification of histone methyltransferase and chromatin co-factor inhibitors [33, 36, 37]. Deshpande et al. [36] found that DOT1L inhibitor EPZ004777 leads to increased differentiation and apoptosis of NUP98-NSD1 transformed murine hematopoietic cells, while another study reported that histone methyltransferase inhibitor suramin likely inhibits NSD1 [38]. The potential benefits of incorporating CREB-binding protein (CBP)/p300, FLT3 and HDAC inhibitors into the treatment algorithm for NUP98-NSD1+ patients have also been discussed [9, 13, 14, 39]. Our compound collection included only one CBP/p300 inhibitor (C646), however, only experimental cell lines were tested with this compound and no marked effects were observed. As shown in Table 2, out of the six HDAC and six FLT3 inhibitors tested in all patient samples, four HDAC (beli- nostat, entinostat, vorinostat, panobinostat) and two FLT3 inhibitors (lestaurtinib, tivozanib) were among the top candidate drugs. Based on these findings, our data suggest that future research should explore the potential efficacy of HDAC and FLT3 inhibitors in NUP98-NSD1+/FLT3-ITD+ AML cells. Earlier studies showed that NUP98-NSD1 defines a relatively rare, yet clinically significant subset of AML patients with unfavorable prognosis [13, 14, 40]. Due to infrequency of these patients, sample size was a major limiting factor affecting our study. To compensate for this limitation and achieve high accuracy, two control groups NUP98-NSD1 Fig. 3 Comparison of drug responses between NUP98-NSD1+/− (healthy donors and negative AML patients) AML patients with FLT3-ITD. a The floating bars show the range and and experimental mouse cell lines were used. Another median DSS of the 25 most differentially sensitive drugs tested in BM limitation was that our three-day, cell viability-based, drug MNCs of four NUP98-NSD1+/FLT3-ITD+ AML patients (orange), - + screening assay was suboptimal for capturing histone nine NUP98-NSD1 /FLT3-ITD AML patients (dark blue), and ten fi healthy donors (light blue). Drug names are shown on the y-axis and methyltransferase inhibitor ef cacies, which often require DSS on the x-axis. Significant differences between the two AML 7–14 days of treatment for pronounced effects on cell dif- patient groups are denoted with an asterisk (*P < 0.05; **P < 0.01). ferentiation or proliferation [41, 42]. Graphs b, c show the dose-response curves of dasatinib and navitoclax Based on the frequent co-occurrence of NUP98-NSD1 in the three groups with mean and standard deviation for each FLT3- concentration and ITD [14, 17], we assessed how their co- expression impacts drug response. Although only one mouse model (retroviral expression in BM cells of BALB/c dasatinib and BCL2 inhibitor navitoclax induced cell death. mice) was used, our data shows that co-occurrence of the The potential relevance of LCK is strengthened by the two lesions drastically changes the drug sensitivity land- observation that three of the top candidate drugs (3/24), scape in BALB/c BM cells (Supplementary Figure 3A-E). namely dasatinib, doramapimod, and danusertib, exert We found that FLT3 and MEK inhibitor sensitivities are potency against LCK (Table 2). Consistent with our find- significantly higher in cells co-expressing NUP98-NSD1 ings, a previous study has shown that LCK is a primary and FLT3-ITD compared to cells expressing either of the target of dasatinib and a critical signaling molecule in two aberrations alone. We also observed that the dual NUP214-ABL1+ T-cell acute lymphoblastic leukemia with positive cells are resistant to BCL2 inhibitors in contrast to potential to induce rapid complete cytogenetic remission as the cells expressing NUP98-NSD1 that had significantly monotherapy [34, 35]. increased sensitivity to BCL2 inhibitors. The finding sug- The present study is the first cancer pharmacopeia-wide gests that NUP98-NSD1 may induce the expression of anti- drug screening study systematically assessing novel small- apoptotic BCL2 family proteins such as BCL2A1. molecule inhibitors for NUP98-NSD1+/FLT3-ITD+ AML Interestingly, BCL2 inhibitor venetoclax has shown patients. Due to involvement of histone methyltransferase promising results in clinical trials and in vitro studies among NSD1, earlier studies on this topic have merely focused on patients with mixed-lineage leukemia (MLL) fusions Dasatinib and navitoclax act synergistically to target. . .

Fig. 4 Pilot combinatorial drug screening. a The heatmap shows DSS classes described below the figure. Graphs b–d illustrate FLT3, MEK, of 70 drugs tested with and without additional dasatinib (100 nM) in and BCL2 inhibitor responses (DSS) in the presence and absence of experimental mouse cells. Numbers 1–7 on top of the graph refer to dasatinib. The floating bars show median DSS with range. Significant cell lines, while annotation colors on the y-axis indicate different drug differences are denoted with an asterisk (*P = 0.023; **P < 0.01) overexpressing homeobox A and B (HOX-AB) cluster genes explore whether aberrant histone methylation plays a role in [20, 43, 44]. Considering that MLL and NSD1 genes share sensitizing NUP98-NSD1+ BM cells to BCL2 inhibitors several common functional domains and that over- such as navitoclax. It also remains to be studied whether the expression of HOX-AB cluster genes is well-established results presented here may be applicable to patients with among NUP98-NSD1+ AML patients [9, 13, 16, 32, 33], functionally similar NUP98 rearrangements such as high BCL2 inhibitor sensitivity in NUP98-NSD1+ cells may NUP98-MLL or NUP98-KDM5A. result via a common mechanism. In MLL-AF4+acute lym- In clinical trials, BCL2 inhibitors and TKIs have pro- phoblastic leukemia, venetoclax sensitivity was shown to duced only moderate effects as single agents with responses result from the ability of MLL-AF4 to activate BCL2 usually being incomplete and transient [44, 47, 48]. To through aberrant H3K79 methylation activities [45], which address this challenge, we systematically evaluated pair- is also a critical factor for NUP98-NSD1-driven leukemic wise drug combinations for NUP98-NSD1+/FLT3-ITD+ transformation [36]. A recent study has shown that NUP98- AML patients using dose-response matrices [24]. In patient NSD1 physically interacts with MLL1 and the NSL histone cells and leukemic mouse cells co-expressing NUP98- modifying complexes [46]. Additional studies should NSD1 and FLT3-ITD, the dasatinib–navitoclax combination J. L. Kivioja et al.

Fig. 5 Dose-response synergy screen. The two-dimensional contour expressing NUP98-NSD1 or FLT3-ITD alone (e, f). The synergy δ- plots show areas of synergy (red) and antagonism (green) for eight scores were calculated based on % inhibition values using the ZIP- concentrations of dasatinib (y-axis) and navitoclax (x-axis) in the index method in SynergyFinder [23, 24]. The most synergistic areas are patient (a), patient 7499 (b), leukemic and preleukemic BALB/c BM indicated with a square inside of each plot cells co-expressing NUP98-NSD1 and FLT3-ITD (c, d), and in cells had the highest synergy of the tested combinations, while survival BCL2 family members upregulated by FLT3-ITD high synergies were also observed for a navitoclax–qui- [29, 53, 54]. zartinib combination (Supplementary Table 6). Synergistic In contrast to the BCL2 inhibitor sensitive patient cells, interactions between BCL2 inhibitors (ABT-737, veneto- the leukemic NUP98-NSD1+/FLT3-ITD+ mouse cells were clax, sabutoclax) and TKIs (dasatinib and sorafenib) have resistant (sDSS < 0) to BCL2 inhibitors suggesting that our been reported in myeloid and lymphoid malignancies con- mouse model does not fully recapitulate the molecular firming the potential importance of our findings [49–53]. In complexity of patient cells. At least two of the patients lymphoid neoplasms, the synergy between BCL2 inhibitors (patients 600 and 3660) carried hotspot mutations in the and SRC/ABL inhibitor dasatinib has been linked to the DNA-binding domain of Wilms’ tumor suppressor gene ability of dasatinib to increase the NOXA/MCL-1 ratio and WT1 (Supplementary Table 7-8), which may explain dis- decrease BCL2A1 levels [50], and to concomitant induction crepancies in BCL2 inhibitor sensitivities between the of BIM expression and inhibition of MCL-1 upregulation experimental NUP98-NSD1+/FLT3-ITD+ mouse cells and [51]. In chronic myeloid leukemia, BCL2 inhibitor sabu- the patient cells. Interestingly, wild type WT1 has been toclax was shown to sensitize quiescent blast crisis leu- shown to function as a direct transcriptional regulator of kemic stem cells to dasatinib mediated cell death by BCL2 [55] and loss of its ability to activate pro-apoptotic abrogating BM niche-dependent dasatinib resistance [52]. Bak due to a mutation has been associated with the devel- Furthermore, in hematopoietic cell lines ABT-737 has been opment of Wilms’ tumor [56]. More recent work has shown shown to restore FLT3 inhibitor sensitivity by targeting pro- that WT1 mutations associate with increased ex vivo Dasatinib and navitoclax act synergistically to target. . .

Fig. 6 Gene expression analysis. The heatmaps show unsupervised package pheatmap (1.0.8). c, d The box plots illustrate the range and hierarchical clustering of RNA sequenced human (a) and experimental mean expression of dasatinib and navitoclax target genes in the pri- mouse cells (b) based on the mean centered expression (log 2 CPM) of mary human cells, while histograms show the expression of corre- HOX-AB genes. The clustering was done using Euclidean distance sponding genes in experimental BALB/c BM cells (e, f). Significant with complete linkage method. The genes are listed on the right side of differences are denoted with an asterisk (*P = 0.028; **P = 0.006; the heatmap and samples below. The heatmaps were generated by R ***P = 0.0006) sensitivity to BCL2 inhibitor venetoclax in AML [20]. NUP98-NSD1 and FLT3-ITD. Our data suggest that While up to 45% of NUP98-NSD1+ AML patients carry NUP98-NSD1+/FLT3-ITD+ AML patients may not benefit WT1 mutations [13], future research should explore how from the use of topoisomerase II inhibitors as part of front- these mutations impact BCL2 inhibitor responses and line induction therapy. It remains to be studied whether BCL2-family gene expression in cells expressing NUP98- replacement of topoisomerase II inhibitors with a targeted NSD1 alone or in conjunction with FLT3-ITD. agent such as dasatinib could improve outcomes of NUP98- In summary, the experimental evidence presented here NSD1+ AML patients if it was added to the usual induction provides new insights into treatment optimization for therapy. NUP98-NSD1+ AML as well as proof-of-concept studies with results supporting clinical investigations of targeted Acknowledgements We thank the patients and healthy donors who therapies for high-risk AML patients with concomitant participated in this study. We would also like to thank staff at the J. L. Kivioja et al.

FIMM High-Throughput Biomedicine Unit for their expert technical 12. Brown J, Jawad M, Twigg SR, Saracoglu K, Sauerbrey A, Tho- assistance with the drug screening experiments and the FIMM mas AE, et al. A cryptic t(5;11)(q35; p15.5) in 2 children with Sequencing Unit for their assistance with sequence analysis. We are acute myeloid leukemia with apparently normal karyotypes, grateful to laboratory technicians Alun Parsons, Minna Suvela, and Siv identified by a multiplex fluorescence in situ hybridization telo- Knaappila for sample processing. We acknowledge personnel at the mere assay. Blood. 2002;99:2526–31. Biomedicum Helsinki FACS core and Functional Genomics Unit for 13. Hollink IH, van den Heuvel-Eibrink MM, Arentsen-Peters ST, their help with flow cytometry and RCV-tests. This work was sup- Pratcorona M, Abbas S, Kuipers JE, et al. NUP98/NSD1 char- ported by grants from Finnish Funding Agency for Technology and acterizes a novel poor prognostic group in acute myeloid leukemia Innovation (grant number 40336/09). Personal grant support was with a distinct HOX gene expression pattern. Blood. received from the Väre Foundation for Pediatric Cancer Research, Ida 2011;118:3645–56. Montin Foundation, the Cancer Society of Finland, and Finnish 14. Ostronoff F, Othus M, Gerbing RB, Loken MR, Raimondi SC, Hematology Association (JK). JS and AT were supported by grants Hirsch BA, et al. NUP98/NSD1 and FLT3/ITD coexpression is from the Swiss Cancer League (KFS-3487-08-2014). more prevalent in younger AML patients and leads to induction failure: a COG and SWOG report. Blood. 2014;124:2400–7. Compliance with ethical standards 15. Akiki S, Dyer SA, Grimwade D, Ivey A, Abou-Zeid N, Borrow J, et al. NUP98-NSD1 fusion in association with FLT3-ITD muta- tion identifies a prognostically relevant subgroup of pediatric fl Con ict of interest KP has received honoraria and research funding acute myeloid leukemia patients suitable for monitoring by real fi from Celgene, Novartis, and P zer. CAH has received research time quantitative PCR. Genes Cancer. funding from Celgene, Novartis, Orion, and Innovative Medicines 2013;52:1053–64. Initiatives 2 project HARMONY. The remaining authors declare that 16. Shiba N, Ichikawa H, Taki T, Park MJ, Jo A, Mitani S, et al. fl they have no con ict of interest. NUP98-NSD1 gene fusion and its related gene expression sig- nature are strongly associated with a poor prognosis in pediatric References acute myeloid leukemia. Genes Chromosomes Cancer. 2013;52:683–93. 17. Thanasopoulou A, Tzankov A, Schwaller J. Potent co-operation 1. Papaemmanuil E, Dohner H, Campbell PJ. Genomic classification between the NUP98-NSD1 fusion and the FLT3-ITD mutation in in acute myeloid leukemia. N Engl J Med. 2016;375:900–1. acute myeloid leukemia induction. Haematologica. 2. Gough SM, Slape CI, Aplan PD. NUP98 gene fusions and 2014;99:1465–71. hematopoietic malignancies: common themes and new biologic 18. Pemovska T, Kontro M, Yadav B, Edgren H, Eldfors S, Szwajda insights. Blood. 2011;118:6247–57. A, et al. Individualized systems medicine strategy to tailor treat- 3. Soler G, Kaltenbach S, Dobbelstein S, Broccardo C, Radford I, ments for patients with chemorefractory acute myeloid leukemia. Mozziconacci MJ, et al. Identification of GSX2 and AF10 as Cancer Discov. 2013;3:1416–29. NUP98 partner genes in myeloid malignancies. Blood Cancer J. 19. Pemovska T, Johnson E, Kontro M, Repasky GA, Chen J, Wells 2013;3:e124. P, et al. Axitinib effectively inhibits BCR-ABL1(T315I) with a 4. Lim HH, An GD, Woo KS, Kim KH, Kim JM, Kim SH, et al. distinct binding conformation. Nature. 2015;519:102–5. NUP98 Rearrangement in Acute Myelomonocytic Leukemia with 20. Kontro M, Kumar A, Majumder MM, Eldfors S, Parsons A, t(11;19)(p15; p12): The First Case Report Worldwide. Ann Lab Pemovska T, et al. HOX gene expression predicts response to Med. 2017;37:285–7. BCL-2 inhibition in acute myeloid leukemia. Leukemia. 5. Nakamura T, Largaespada DA, Lee MP, Johnson LA, Ohyashiki 2017;31:301–9. K, Toyama K, et al. Fusion of the nucleoporin gene NUP98 to 21. Zhang JH, Chung TD, Oldenburg KR. A simple statistical para- HOXA9 by the translocation t(7;11)(p15; p15) in meter for use in evaluation and validation of high throughput human myeloid leukaemia. Nat Genet. 1996;12:154–8. screening assays. J Biomol Screen. 1999;4:67–73. 6. Saw J, Curtis DJ, Hussey DJ, Dobrovic A, Aplan PD, Slape CI. 22. Yadav B, Pemovska T, Szwajda A, Kulesskiy E, Kontro M, The fusion partner specifies the oncogenic potential of NUP98 Karjalainen R, et al. Quantitative scoring of differential drug fusion proteins. Leuk Res. 2013;37:1668–73. sensitivity for individually optimized anticancer therapies. Sci 7. Wu X, Kasper LH, Mantcheva RT, Mantchev GT, Springett MJ, Rep. 2014;4:5193. van Deursen JM. Disruption of the FG nucleoporin NUP98 causes 23. Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for selective changes in nuclear pore complex stoichiometry and drug synergy in complex dose-response landscapes using an function. Proc Natl Acad Sci USA. 2001;98:3191–6. interaction potency model. Comput Struct Biotechnol J. 8. Struski S, Lagarde S, Bories P, Puiseux C, Prade N, Cuccuini W, 2015;13:504–13. et al. NUP98 is rearranged in 3.8% of pediatric AML forming a 24. Ianevski A, He L, Aittokallio T, Tang J. SynergyFinder: a web clinical and molecular homogenous group with a poor prognosis. application for analyzing drug combination dose-response matrix Leukemia. 2017;31:565–72. data. Bioinformatics. 2017;33:2413–5. 9. Bisio V, Zampini M, Tregnago C, Manara E, Salsi V, Di Meglio 25. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, A, et al. NUP98-fusion transcripts characterize different biological et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. entities within acute myeloid leukemia: a report from the AIEOP- 2013;29:15–21. AML group. Leukemia. 2017;31:974–7. 26. Robinson MD, Oshlack A. A scaling normalization method for 10. Mertens F, Johansson B, Fioretos T, Mitelman F. The emerging differential expression analysis of RNA-seq data. Genome Biol. complexity of gene fusions in cancer. Nat Rev Cancer. 2010;11:R25. 2015;15:371–81. 27. Kumar A, Kankainen M, Parsons A, Kallioniemi O, Mattila P, 11. Jaju RJ, Fidler C, Haas OA, Strickson AJ, Watkins F, Clark K, Heckman CA. The impact of RNA sequence library construction et al. A novel gene, NSD1, is fused to NUP98 in the t (5; 11)(q35; protocols on transcriptomic profiling of leukemia. BMC Genom. p15. 5) in de novo childhood acute myeloid leukemia. Blood. 2017;18:629. 2001;98:1264–7. Dasatinib and navitoclax act synergistically to target. . .

28. Kivioja JL, Lopez Marti JM, Kumar A, Kontro M, Edgren H, the Bcl-2-selective inhibitor ABT-199. Leukemia. 2014;28:1557– Parsons A, et al. Chimeric NUP98-NSD1 transcripts from the 60. cryptic t(5;11)(q35.2; p15.4) in adult de novo acute myeloid 44. Konopleva M, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman leukemia. Leuk Lymphoma. 2018;59:725–32. T, et al. Efficacy and Biological Correlates of Response in a Phase 29. Yoshimoto G, Miyamoto T, Jabbarzadeh-Tabrizi S, Iino T, Roc- II Study of Venetoclax Monotherapy in Patients with Acute nik JL, Kikushige Y, et al. FLT3-ITD up-regulates MCL-1 to Myelogenous Leukemia. Cancer Discov. 2016;6:1106–17. promote survival of stem cells in acute myeloid leukemia via 45. Benito JM, Godfrey L, Kojima K, Hogdal L, Wunderlich M, Geng FLT3-ITD-specific STAT5 activation. Blood. 2009;114:5034–43. H, et al. MLL-Rearranged Acute Lymphoblastic Leukemias 30. Hayakawa F, Towatari M, Kiyoi H, Tanimoto M, Kitamura T, Activate BCL-2 through H3K79 Methylation and Are Sensitive to Saito H, et al. Tandem-duplicated Flt3 constitutively activates the BCL-2-Specific Antagonist ABT-199. Cell Rep. STAT5 and MAP kinase and introduces autonomous cell growth 2015;13:2715–27. in IL-3-dependent cell lines. Oncogene. 2000;19:624–31. 46. Xu H, Valerio DG, Eisold ME, Sinha A, Koche RP, Hu W, et al. 31. Crescenzi B, Nofrini V, Barba G, Matteucci C, Di Giacomo D, NUP98 Fusion Proteins Interact with the NSL and MLL1 Com- Gorello P, et al. NUP98/11p15 translocations affect CD34+ cells plexes to Drive Leukemogenesis. Cancer Cell. 2016;30:863–78. in myeloid and T lymphoid leukemias. Leuk Res. 2015;39:769– 47. Lindauer M, Hochhaus A. Dasatinib. Recent Results Cancer Res. 72. 2014;201:27–65. 32. Rayasam GV, Wendling O, Angrand PO, Mark M, Niederreither 48. Larrosa-Garcia M, Baer MR. FLT3 Inhibitors in acute myeloid K, Song L, et al. NSD1 is essential for early post-implantation leukemia: current status and future directions. Mol Cancer Ther. development and has a catalytically active SET domain. EMBO J. 2017;16:991–1001. 2003;22:3153–63. 49. Kurtz SE, Eide CA, Kaempf A, Khanna V, Savage SL, Rofelty A, 33. Wang GG, Cai L, Pasillas MP, Kamps MP. NUP98-NSD1 links et al. Molecularly targeted drug combinations demonstrate selec- H3K36 methylation to Hox-A gene activation and leukaemogen- tive effectiveness for myeloid- and lymphoid-derived hematologic esis. Nat Cell Biol. 2007;9:804–12. malignancies. Proc Natl Acad Sci USA. 2017;114: 34. Laukkanen S, Gronroos T, Polonen P, Kuusanmaki H, Mehtonen E7554–63. J, Cloos J, et al. In silico and preclinical drug screening identifies 50. Tromp JM, Geest CR, Breij EC, Elias JA, van Laar J, Luijks DM, dasatinib as a targeted therapy for T-ALL. Blood. Cancer J. et al. Tipping the Noxa/Mcl-1 balance overcomes ABT-737 2017;7:e604. resistance in chronic lymphocytic leukemia. Clin Cancer Res. 35. Deenik W, Beverloo HB, van der Poel-van de Luytgaarde SC, 2012;18:487–98. Wattel MM, van Esser JW, Valk PJ, et al. Rapid complete cyto- 51. Leonard JT, Rowley JS, Eide CA, Traer E, Hayes-Lattin B, genetic remission after upfront dasatinib monotherapy in a patient Loriaux M, et al. Targeting BCL-2 and ABL/LYN in Philadelphia with a NUP214-ABL1-positive T-cell acute lymphoblastic leu- chromosome-positive acute lymphoblastic leukemia. Sci Transl kemia. Leukemia. 2009;23:627–9. Med. 2016;8:354ra114. 36. Deshpande AJ, Deshpande A, Sinha AU, Chen L, Chang J, Cihan 52. Goff DJ, Court Recart A, Sadarangani A, Chun HJ, Barrett CL, A, et al. AF10 regulates progressive H3K79 methylation and Krajewska M, et al. A Pan-BCL2 inhibitor renders bone-marrow- HOX gene expression in diverse AML subtypes. Cancer Cell. resident human leukemia stem cells sensitive to 2014;26:896–908. inhibition. Cell Stem Cell. 2013;12:316–28. 37. Franks TM, McCloskey A, Shokirev MN, Benner C, Rathore A, 53. Kohl TM, Hellinger C, Ahmed F, Buske C, Hiddemann W, Hetzer MW. Nup98 recruits the Wdr82-Set1A/COMPASS com- Bohlander SK, et al. BH3 mimetic ABT-737 neutralizes resistance plex to promoters to regulate H3K4 trimethylation in hemato- to FLT3 inhibitor treatment mediated by FLT3-independent poietic progenitor cells. Genes Dev. 2017;31:2222–34. expression of BCL2 in primary AML blasts. Leukemia. 38. Drake KM, Watson VG, Kisielewski A, Glynn R, Napper AD. A 2007;21:1763–72. sensitive luminescent assay for the histone methyltransferase 54. Minami Y, Yamamoto K, Kiyoi H, Ueda R, Saito H, Naoe T. NSD1 and other SAM-dependent . Assay Drug Dev Different antiapoptotic pathways between wild-type and mutated Technol. 2014;12:258–71. FLT3: insights into therapeutic targets in leukemia. Blood. 39. Ofran Y. Concealed dagger in FLT3/ITD+AML. Blood. 2003;102:2969–75. 2014;124:2317–9. 55. Hewitt SM, Hamada S, McDonnell TJ, Rauscher FJ 3rd, Saunders 40. Fasan A, Haferlach C, Alpermann T, Kern W, Haferlach T, GF. Regulation of the proto-oncogenes bcl-2 and c-myc by the Schnittger S. A rare but specific subset of adult AML patients can Wilms’ tumor suppressor gene WT1. Cancer Res. 1995;55:5386– be defined by the cytogenetically cryptic NUP98-NSD1 fusion 9. gene. Leukemia. 2013;27:245–8. 56. Morrison DJ, English MA, Licht JD. WT1 induces apoptosis 41. Borkin D, He S, Miao H, Kempinska K, Pollock J, Chase J, et al. through transcriptional regulation of the proapoptotic Bcl-2 family Pharmacologic inhibition of the Menin-MLL interaction blocks member Bak. Cancer Res. 2005;65:8174–82. progression of MLL leukemia in vivo. Cancer Cell. 2015;27:589– 57. Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, 602. Buchner T, et al. Diagnosis and management of AML in adults: 42. Song Y, Wu F, Wu J. Targeting histone methylation for cancer 2017 ELN recommendations from an international expert panel. therapy: enzymes, inhibitors, biological activity and perspectives. Blood. 2017;129:424–47. J Hematol Oncol. 2016;9:49. 58. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau 43. Niu X, Wang G, Wang Y, Caldwell JT, Edwards H, Xie C, et al. MM, et al. The 2016 revision to the World Health Organization Acute myeloid leukemia cells harboring MLL fusion genes or classification of myeloid neoplasms and acute leukemia. Blood. with the acute promyelocytic leukemia phenotype are sensitive to 2016;127:2391–405.