Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Biology of Human Tumors Clinical Cancer Research Biological Activities of RUNX1 Mutants Predict Secondary Acute Transformation from Chronic Myelomonocytic Leukemia and Myelodysplastic Syndromes Shu-Chun Tsai1, Lee-Yung Shih1,2, Sung-Tzu Liang3, Ying-Jung Huang1, Ming-Chung Kuo1,2, Chein-Fuang Huang1, Yu-Shu Shih1, Tung-Huei Lin1, Ming-Chun Chiu1, and Der-Cherng Liang3

Abstract

Purpose: factor RUNX1 is essential for normal Results: We observed that most RUNX1 mutants had hematopoiesis. High frequencies of RUNX1 in reduced abilities in DNA binding, CBFb heterodimerization, chronic myelomonocytic leukemia (CMML) and myelodysplastic and C-FMS gene induction. The relative biologic activities of syndromes (MDS) have been described, whereas the biologic RUNX1 mutants were grouped into high- and low-activity significances of the were not investigated. Here, we mutations. Correlation of the activities of RUNX1 mutants aimed to correlate the biologic activities of the RUNX1 mutants with the clinical outcomes revealed that patients harboring with the clinical outcomes of patients. lower activities of RUNX1 mutants had a higher risk and Experimental Design: We examined the mutational status of shorter time to secondary transfor- RUNX1 in 143 MDS and 84 CMML patients. Then, we studied the mation in MDS and CMML. In multivariate analysis, low DNA and CBFb binding abilities of all the RUNX1 mutants RUNX1 activity remained an independent predictor for sec- identified by using electrophoretic mobility shift assay and co- ondary acute myeloid leukemia–free survival in MDS immunoprecipitation assay, and also determined their activities patients. on target C-FMS gene induction by Western blotting and lucifer- Conclusions: The biologic activity rather than the mutation- ase reporter assay. Using luciferase reporter assay, the relative al status of RUNX1 might be an indicator in predicting out- biologic activities of each RUNX1 mutant could be quantified come of patients with MDS and CMML. Clin Cancer Res; 1–11. and correlated with the patient outcomes by statistical analyses. 2015 AACR.

Introduction mutants has not been investigated. In the present study, we sought to study the biologic function of the RUNX1 mutants to further Chronic myelomonocytic leukemia (CMML) and myelodys- determine the prognostic impact of RUNX1 mutation. plastic syndromes (MDS) are clonal hematopoietic malignancies. RUNX1 is an essential for normal fetal liver Both are characterized by dysplasia of one or more myeloid hematopoiesis (4). Knockout of RUNX1 caused severe hemor- lineages and with high possibilities of transformation to second- rhage within the brain and vertebral canal of the mouse embryo ary acute myeloid leukemia (sAML). Cytogenetic and genetic and lethality at E12.5 (4). In adult mice, RUNX1 was expressed in alterations in patients with CMML and MDS have been a subject multiple hematopoietic lineage cells, mainly in myeloid, lym- of investigation over the last decade. Previously, we and others phoid, and megakaryocytic, and less in erythroid lineage cells have described a high mutation rate of RUNX1 in CMML or MDS (5, 6). RUNX1 was first identified in patients with translocation (1–3). However, the clinical relevance of dysfunction of RUNX1 involving t(8;21) AML. In addition to the translocation, intragenic mutations of RUNX1 were also frequently detected in de novo 1Division of Hematology-Oncology, Department of Internal Medicine, MDS (2–3, 7, 8), dominant familial platelet disorder (9), therapy- Chang Gung Memorial Hospital, Taipei, Taiwan. 2College of Medicine, related MDS/AML (10), CMML (1), and de novo AML (11), 3 Chang Gung University, Taoyuan, Taiwan. Division of Pediatric especially AML M0 subtype (12, 13). Hematology-Oncology, Department of Pediatrics, Mackay Memorial Hospital, Taipei, Taiwan. Two distinct promoters generate 3 RUNX1 isoforms, includ- ing RUNX1a and RUNX1b transcribed from the P2 proximal Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). , and RUNX1c from the distal P1 promoter (14). RUNX1b and RUNX1c are different at their first pro- S.-C. Tsai and L.-Y. Shih contributed equally to this article. moter-specific , but their remaining 448 amino acids are Corresponding Author: Lee-Yung Shih, Chang Gung Memorial Hospital, 199 identical and exert similar functions. Structurally, RUNX1 pro- Tung Hwa North Road, Taipei 105, Taiwan. Phone: 886-3-3281200, ext. 2524; tein is composed of an N-terminal Runt homology domain Fax: 886-3-3286697; E-mail: [email protected] (RHD) and a C-terminal transactivation domain (TAD) which doi: 10.1158/1078-0432.CCR-14-2203 mediates DNA binding and gene transactivation, respectively 2015 American Association for Cancer Research. (15, 16). However, RUNX1a derived from alternative

www.aacrjournals.org OF1

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Tsai et al.

examined. Our patients were classified based on the 2008 WHO Translational Relevance classification system. All the CMML patients had persistent mono- In the present study, we systematically analyzed the biologic cytosis during a long-time follow-up. BM cells of the patients were activities of RUNX1 mutations identified from patients with fractionated with Ficoll–Hypaque density gradient centrifugation chronic myelomonocytic leukemia or myelodysplastic syn- to isolate the mononuclear cells. DNA and RNA of the BM dromes by in vitro functional assays. Apart from the mutational mononuclear cells were extracted and stored at 80C. The study status of RUNX1 gene, we demonstrated that the biologic was approved by the Institutional Board of Chang Gung Memo- activity of mutant RUNX1 was a more important predictor rial Hospital. of disease progression. Patients with the lower activities of RUNX1 mutants had a higher risk and shorter time to second- Mutational analysis ary acute myelogenous leukemia transformation. Our study Mutational analysis of RUNX1 was performed by direct was the first one to directly correlate the biologic functions sequencing of all RT-PCR or genomic DNA (gDNA)-PCR products with clinical outcome and showed their prognostic relevance. amplified with different primer pairs covering the entire coding Our results suggest that mutant RUNX1 activity would be a sequences from exons 3 to 8 of RUNX1b gene as described useful indicator in predicting the progression of the diseases, previously (1). The mutational status of 57 patients with CMML and potentially helpful in identifying patients for early thera- was reported before (1), and the results have been updated in the peutic intervention, even in case of low-risk patients. present study. The gDNA-PCR or RT-PCR assays followed by direct sequencing for the detection of FLT3-ITD, FLT3-TKD, C-KIT, and C-FMS mutations, point mutations at codons 12, 13, and 61 in exons 1 and 2 of N-RAS gene and K-RAS gene, PTPN11, TP53, exons 1 to 3 and exons 7 to 9 of WT-1, JAK2V617F, CEBPa, NPM1, – splicing of the proximal promoter transcribed transcript, TET2, IDH1, IDH2, DNMT3A, ASXL1, and EZH2 mutations were which retains the RHD but lacks the TAD, has been considered carried out as previously described (13). as an antagonist for RUNX1b (17). The RHD spans amino acids 50–177, and TAD is located at Cell culture, DNA transfection, and lentivirus production amino acids 291–371 of RUNX1b protein. In addition to DNA HEK293T cells were cultured in DMEM supplemented with binding, the RHD of RUNX1 dimerizes with CBFb, which does not penicillin/streptomycin, 1 mmol/L L-glutamine, and 10% FBS. harbor any transcriptional activity, but stabilizes RUNX1 and Leukemia and lymphoma cell lines, including KG1a, HL60, NB4, DNA interaction. Tahirov and colleagues (18) have demonstrated U937, THP-1, MV4-11, RS4-11, Kasumi, K562, KU812, REH, the crystal structures of RUNX1(RHD)-CBFb(core domain)-C/ BJAB, Raji, and Jurkat cells, were all cultured in RPMI-1640 medi- EBPb-DNA, and precisely map the regions at which RUNX1 um supplemented with penicillin/streptomycin, 1 mmol/L directly interacted with DNA and CBFb. Three amino acids Arg80, L-glutamine, and 10% FBS. All the cell cultures were kept in a 37 C Arg174, and Arg177 of RUNX1b forming bipartite hydrogen humidified incubator supplemented with 5% CO . Transfection of bonds with DNA are especially critical for DNA binding. The 2 DNA into cells was performed using TrasIT-LT1 Transfection other 3 residues Lys83, Arg142, and Asp171 of RUNX1b involved Reagent (Mirus) according to the manufacturer's instructions. The in DNA recognition are also important for DNA binding. The methods for production and infection of lentiviruses were per- interacting interfaces between RUNX1 and CBFb are hydrophilic formed as our previous description (22). Briefly, 0.5 mgp8.91,1mg and away from the DNA binding site. The interfaces are located at pMD2.G, and 1.5 mg pCMSCV vector or pCMSCV-FLAG-RUNX1b two areas of RUNX1b, area I (amino acids 102–116 and 147–152) 6 were cotransfected into 1.2 10 HEK293T cells. After 60 to 72 and area II (amino acids 64–70 and 152–162; ref. 18). hours, the supernatants containing infectious lentiviruses were Several specific for hematopoiesis were directly regulated collected, filtered through a 0.45 mm filter, and incubated with by RUNX1, including IL3 (19), GM-CSF (20), C-FMS (21), etc. target cells for transduction. Only the regulatory region of RUNX1 on C-FMS promoter was well defined (21); therefore, we chose C-FMS as an indicator to Electrophoretic mobility shift assay determine the biologic activities of RUNX1 mutants. We exam- HEK293T cells were seeded in 6-well plates at the density of ined DNA binding affinities of each RUNX1 mutant on C-FMS 7.5 105 cells/well. After 24 hours, the cells were transfected with promoter, and their activities in the induction of C-FMS protein 0.5 mg pCR3-CBFb combined with 0.5 mg expression plasmids and C-FMS promoter. Here, we reported that the biologic activity of either wild-type (wt)-RUNX1b, its mutants, or control vector, of RUNX1 mutants had impact on the disease progression of pcDNA, respectively. The nuclear extracts of the cells were har- MDS/CMML. vested as previously described (22) and applied for electropho- retic mobility shift assay (EMSA) by using the LightShift Chemi- Materials and Methods luminescent EMSA Kit (Thermo scientific) according to the man- Additional information can be found in Supplementary ufacturer's instructions. Briefly, both forward and reverse strands 0 Methods. of the oligonucleotides were labeled with biotin at the 5 ends and annealed together to form a double strand probe. The 0 Patients and specimen collection probe sequences consisting of C-FMS promoter are 5 - 0 Bone marrow (BM) samples from 143 patients with de novo CAAACTCTGTGGTTGCCTTG-3 . The underlined nucleotides are MDS at initial diagnosis [54 refractory anemia with excess blasts-I recognized by RUNX1, and the mutant (mt) probe was substi- (RAEB-I), 60 RAEB-II, and 29 other subtypes] and 84 patients with tuted to TGTTAG. The binding reaction was carried out in a total CMML (55 CMML1 and 29 CMML2) consecutively diagnosed at volume of 20 mL containing 4 mg nuclear extracts, 1 ng biotin- Chang Gung Memorial Hospital between 1991 and 2011 were labeled probe, 1 mg poly(dI-dC), 2.5% glycerol, and 0.05%

OF2 Clin Cancer Res; 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

RUNX1 Activity Predicts sAML from MDS/CMML

Nonidet-P40 (NP40) in 1X binding buffer. For competition netic regulator genes being the most frequently involved in both EMSA, 200 ng unlabeled probes were added to the reactions. For CMML and MDS patients. antibody supershift EMSA, 2 mg anti-FLAG antibody (Sigma) or normal mouse IgG (Millipore) was added to the reactions. All Abilities of RUNX1 mutants to bind DNA and heterodimerize reaction mixtures were incubated at room temperature for 30 with CBFb minutes, electrophoresed in 6% native polyacrylamide gels with RUNX1 is a transcription factor. It directly binds to DNA but Tris-borate buffer (90 mmol/L Tris, 90 mmol/L boric acid, 2 requires dimerization with CBFb to stabilize its interaction with mmol/L EDTA), and then transferred to Hybond-N membrane DNA. To assess biologic impacts of mutant RUNX1 proteins, we (Amersham Biosciences). The membrane was UV-crosslinked, examined the abilities of DNA binding and heterodimerization blocked, and the signals were revealed with the Chemilumines- with CBFb of the RUNX1 mutants identified from our cent Nucleic Acid Detection Module (Pierce) according to the patients. Figure 1A is the result of EMSA, in which the top manufacturer's instructions. panel shows the RUNX1b mutants identified from CMML, and the bottom panel shows the RUNX1b mutants identified from Co-immunoprecipitation assay MDS. All the RUNX1b expression constructs were in-frame FLAG-wt-RUNX1b or FLAG-RUNX1b mutants were co- fused with a FLAG tag at their N-termini. The wt-RUNX1b expressed with CBFb in HEK293T cells. Cell lysates were extracted formed two clear complexes with the DNA probe comprised with RIPA buffer and incubated with anti-FLAG M2 affinity gel at with the C-FMS promoter region. Anti-FLAG antibody (F) 4C for 2 hours. The gel was washed 4 times with RIPA buffer and caused supershift of the RUNX1–DNA complexes, whereas subjected to SDS-PAGE and Western blot analysis. equal amount of control mouse IgG (m) did not, indicating the presence of RUNX1b protein in the complexes. Cold probes Luciferase reporter assay are composed of the same DNA sequences (w) or mutant K562 cells were seeded at a density of 1 105 cells per well in a sequences (m) with the probe, but lacking biotin label at their 0 24-well plate and then transfected with 0.3 mg C-FMS promoter 5 termini, so they would not show any signal on the gel. Excess luciferase reporter plasmid, pMCSF-R(mB)-luc (Addgene; ref. 23), cold probes were served as competitors to compete the binding combined with 0.1 mg pcDNA or pcDNA-RUNX1b mutant plas- of RUNX1b protein with biotin-labeled probe. Two hundred– mids, and 0.1 mg pEGFP-C1 (Promega) as internal control using fold excess of wild-type (w) cold probes disrupted the 2 com- TransIT-2020 transfection reagent (Mirus). After 72 hours, cells plexes on the gels, but mutant probes (m) did not, suggesting were harvested, lysed with Passive lysis buffer (Promega), and the binding of RUNX1b with the probe is sequence specific. The mixed with Bright-Glo Luciferase assay reagent (Promega) for truncation mutants (Ile150Hisfs 36 and Arg139 )withpartial- detection of luciferase activities and GFP fluorescent intensity. The ly destructive RHD completely lost DNA binding abilities. relative activity of each RUNX1b mutant was derived by normal- However, all the truncation mutants retaining the intact RHD ization of each luciferase intensity with its GFP intensity and then (Ser195Profs 15, Arg293 , Gln186Argfs 24, Thr219Hisfs 15, divided by the value of wt-RUNX1b. Gln245 ,Met283 and Ser383 ) exhibited comparable DNA binding ability with wt-RUNX1b, supporting the integrity of RHD is a determinant factor for DNA binding. The four Statistical analysis missense mutants, Arg80Cys, Asn109Lys, Gly141Val, and Fisher exact test, c2 analysis, and the unpaired t test were used to Arg174Gln, completely abolished their DNA binding, confirm- make comparison between groups. The Cox proportional hazards ing that they may affect the important residues for DNA regression model was used to estimate HRs of sAML-free survival recognition. The other mutations located within RHD showed and overall survival, and both in a univariate and multivariate various degrees of impaired DNA binding abilities, which is setting. Confidence intervals (CI) at 95% for HRs were calculated. related to the polarity and charge of the mutant amino acids, The optimal cutoff point was estimated by ROC curve. The Kaplan– and their distance to DNA binding amino acids. Cys72Trp, Meier estimate was used to plot the time to sAML transformation His78Gln, and Trp79Arg showed a decreasing affinity with according to the activities of the RUNX1 mutants on C-FMS DNA as they are closer to Arg80 (Fig. 1A, top left blot). induction. The log-rank test was used to assess the probabilities Although Arg139Gly and Arg139Gln mutated at the same of sAML transformation. All the statistical analyses were performed residue, the chemical properties of glycine and glutamine are with an SPSS software version 8.0 for Windows (SPSS Inc.). very different, which might be attributed to their different DNA binding abilities (Fig. 1A, bottom right blot). All of the C-ter- Results minal frameshift mutants retained DNA binding ability, but Frequencies and patterns of RUNX1 mutations in patients with their patterns of DNA complexes were different from wt- CMML and MDS RUNX1b, suggesting different protein complexes were formed. RUNX1 mutations were detected in 27 of 84 (32.1%) patients The protein expression of RUNX1 mutants in the nuclear with CMML and 20 of 143 (14.0%) patients with MDS at initial extracts subjected to EMSA was confirmed by Western blot diagnosis. The mutation patterns of RUNX1 of the patients with analysis as shown in Fig. 1B. CMML and MDS are listed in Table 1. Of the patients carrying Figure 2 showed the results of coimmunoprecipitation with RUNX1 mutations at diagnosis, additional 19 genes related to CBFb of the RUNX1 mutants. Six mutations, Arg139 , myeloid neoplasms were also analyzed. The frequencies and Ile150Hisfs 36, Asn109Lys, Ser114Leu, Glu111_Ser114 dup, distribution of co-operating mutated genes in RUNX1 muta- and Ala160Thr, located within area I or area II, lost or dimin- tion–positive patients are shown in Supplementary Fig. S1. The ished their interactions with CBFb. Area I and area II are coexisting mutations included CEBPa, TET2, DNMT3A, IDH2, hydrophilic regions. Cys72Trp and Ser73Phe are both aromatic ASXL1, EZH2, FLT3, NRAS, PTPN11, CBL, and TP53, with epige- and hydrophobic residues. They may change the physical

www.aacrjournals.org Clin Cancer Res; 2015 OF3

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Tsai et al.

Table 1. Mutation patterns of RUNX1 with their effects on C-FMS and risk of sAML transformation in patients with CMML and MDS RUNX1 mutation Patient no. Nucleotidea Amino acid DNA binding CBFb dimerization C-FMS activation sAML transformation CMML1 181G>T Glu61 CMML2 184_185insC Leu62Profs49 þ CMML3 211del Leu71Serfs24 þ CMML4-5 237G>A Trp79 CMML6 254_257dup Ile87Alafs25 CMML7 297_298insGGAC Thr101Argfs11 þ CMML8 314_317del Val105Glyfs12 CMML9 447_448insC Ile150Hisfs36 þ CMML10 583del Ser195Profs15 þþ þ CMML11-13 877C>T Arg293 þþ þ CMML14 216C>G Cys72Trp þ þ CMML15 234C>A His78Gln þþ CMML16 235T>C Trp79Arg þþ þ CMML17 238C>Tþ251C>A Arg80CysþThr84Asn þ CMML18 327T>A Asn109Lys þ CMML19 341C>T Ser114Leu þ þ CMML20 415C>G Arg139Gly þþ þ CMML21 1193C>T Pro398Leu þþ þ CMML22 332_343dup Glu111_Ser114dup þ CMML23 780_798dup Ser268Glyfs311 þþ þ CMML24 969_988del Gly324Aspfs242 þþ CMML25 990_993dup Pro332Aspfs242 þþ þ CMML26 1021_1028del Met341Hisfs229 þþ þ CMML27 1103_1104insC Phe369Valfs204 þþ þ MDS1 51_52insA Thr18Asnfs93 þ MDS2 270_271insGTGTTTTAG Val92Phefs6 þ MDS3 415C>T Arg139 þ MDS4 556del Gln186Argfs24 þþ þ MDS5 654_655insC Thr219Hisfs15 þþ þ MDS6 733C>T Gln245 þþ þ MDS7 847del Met283 þþ þ MDS8 877C>T Arg293 þþ þ MDS9 1148C>A Ser383 þþ þ MDS10 218C>T Ser73Phe þ þ MDS11-12 232C>Tþ478G>A His78Tyrþ Ala160Thr þ þ MDS13 404G>A Arg135Lys þþ þ MDS14 415C>G Arg139Gly þþ þ MDS15-16 416G>A Arg139Gln þ þ MDS17 422G>T Gly141Val þ þ MDS18 521G>A Arg174Gln þ þ MDS19 1274T>G Val425Glyb þþ þ MDS20-21 1315A>T Met439Leub þþ þ þ MDS22 983_984insC Ser329Leufs244 þþ þ MDS23 1270_1271insG Asp424Glyfs149 þþ þ aAccession number: NM_001001890. bSingle nucleotide variants also present in complete remission samples in AML (11).

properties of area II by introducing hydrophobic amino acids or RUNX1c protein was detected in KG1a, THP-1, MV4-11, near area II, therefore abrogated the interaction with CBFb. Kasumi, K562, KU812, RS4-11, REH, and Jurkat cells, and none Taken together, our results provided direct molecular evidences of the cells had detectable RUNX1a protein. We then trans- of structural lesions of the mutants correlated well with DNA duced RUNX1b protein to RUNX1-negative HL60 and U937 and CBFb binding changes. cells to examine its effect on the induction of C-FMS. As shown in Fig. 3B and C, forced expression of wt-RUNX1b in HL60 and Downregulation of target by RUNX1 mutants U937 cells could induce C-FMS protein and mRNA expression, After examining DNA binding ability of each RUNX1 mutant respectively. Therefore, all the RUNX1 mutants were transduced on C-FMS promoter, we further checked whether RUNX1 to HL60 and U937 cells to compare their biologic activities mutants affect C-FMS protein expression. RUNX1 gene encodes with wt-RUNX1b. Figure 3D shows a representative result in 3 isoforms RUNX1a, RUNX1b, and RUNX1c. In order to rule U937 cells. Compared with wt-RUNX1b, C-FMS protein level out the effects of endogenous RUNX1 protein, several leukemia was decreased in most RUNX1 mutant–expressing cells. Seven and lymphoma cell lines were examined to select candidate cell mutants, Cys72Trp, Ser114Leu, Pro398Leu, Val425Gly, lines for the following assays. Expression plasmids of C-FMS, Met439Leu, Asp424Glyfs149, and Ala160Thr, showed - PU.1, RUNX1a, RUNX1b, and RUNX1c were transfected into tively higher levels of C-FMS protein expression compared with 293T cells, and these transfected cell lysates were taken as other mutants. To more accurately estimate the activity of each positive controls in Western blot. As shown in Fig. 3A, RUNX1b RUNX1 mutant, luciferase reporter assay of C-FMS promoter

OF4 Clin Cancer Res; 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

RUNX1 Activity Predicts sAML from MDS/CMML

www.aacrjournals.org Clin Cancer Res; 2015 OF5

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Tsai et al.

Figure 2. Heterodimerization ability of RUNX1 mutants with CBFb. CBFb combined with either wt-RUNX1b, its mutants, or control vector, pcDNA, was co-expressed in HEK293T cells. After 72 hours, the cell lysates were collected with RIPA buffer, immunoprecipitated with anti-FLAG M2 affinity gel, and analyzed by immunoblotting (IB) using anti-CBFb Ab and anti-FLAG Ab. Protein lysates (20 mg) of each transfectant were taken as input control and analyzed by Western blotting using anti-CBFb and b-ACTIN antibodies to confirm the expression of CBFb and the equal loading of protein lysates.

was adapted and the results are shown in Fig. 3E. C-FMS is the cutoff point was estimated by ROC curve. A cutoff point at most well-characterized promoter regulated by RUNX1 and is 0.520 of the relative activity on C-FMS promoter showed the also the only commercially available one to estimate RUNX1 best sensitivity (0.875) and specificity (1.0). The RUNX1 muta- activity. The trend of C-FMS induction in Fig. 3D (C-FMS tions with reporter activities higher than 0.520 were considered protein) and Fig. 3E (C-FMS promoter activity) is similar. To as high-activity mutations, and those with reporter activities determine the activities of the RUNX1 mutants, the optimal lower than 0.520 belonged to low-activity mutations. The

Figure 1. DNA binding ability of the RUNX1 mutants on C-FMS promoter. The RUNX1 mutations identified from CMML and MDS were separately assayed in independent experiments. All the RUNX1b expression constructs were in-frame fused with a FLAG tag at their N-termini. CBFb combined with either wild-type (wt)-RUNX1b, its mutants, or control vector, pcDNA, was co-expressed in HEK293T cells. Nuclear extracts and protein lysates of the cells were harvested at 72 hours after transfection, and applied for EMSA experiment (A) and Western blotting (B). A, top, the RUNX1b mutants identified from CMML; bottom, the RUNX1b mutants identified from MDS.Wt-RUNX1b protein formed 2 complexes with the biotin-labeled probe, and the complexes were supershifted by adding of anti-FLAG antibody (F), but not control mouse IgG (m). Two hundred–fold excess unlabeled wild-type (w) or mutant (m) probes were added in the reaction as cold competitors of the RUNX1–DNA complex. Each RUNX1 mutant protein formed a unique binding pattern with the probe. B, protein lysates (15 mg) of each transfectant were analyzed by Western blotting to confirm the expression of mutant RUNX1 proteins (anti-FLAG Ab); b-ACTIN served as a loading control.

OF6 Clin Cancer Res; 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

RUNX1 Activity Predicts sAML from MDS/CMML

Figure 3. Aberrant regulation of C-FMS by RUNX1 mutants. A, cell lysates (20 mg) of various leukemia and lymphoma cell lines were detected for endogenous RUNX1 and C-FMS protein expression by Western blotting. HEK293T cells transfected with pMSCV-C-FMS, pCR3-PU.1, or pcDNA-RUNX1a, b, or c, respectively, served as positive control. B–C, HL60 and U937 cells were infected with lentiviruses carrying RUNX1b expression plasmid or its control vector, pCMSCV, at MOI ¼ 2. Four days after infection, the cells were selected with 2 mg/mL puromycin for 2 weeks and then harvested with RIPA buffer and Trizol reagent. Protein lysates (20 mg) were applied for Western blot analysis and probed with anti-C-FMS, RUNX1, and b-ACTIN antibodies (B). RNA (2 mg) of each cell was reverse transcribed and detected for C-FMS mRNA level by Q-PCR (C). D, U937 cells were infected with pCMSCV or various pCMSCV-RUNX1 mutant lentiviruses (MOI ¼ 2). Four days after infection, the cells were selected with 2 mg/mL puromycin for 2 weeks and then harvested with RIPA buffer. The protein lysates (20 mg) were applied for Western blot analysis and probed with anti–C-FMS, FLAG, and GAPDH antibodies. E, K562 cells were cotransfected with luciferase reporter plasmid of C-FMS promoter with each RUNX1 mutant plasmid. Seventy-two hours later, luciferase intensity of each transfectant was measured and normalized with the intensity of wt-RUNX1b.

www.aacrjournals.org Clin Cancer Res; 2015 OF7

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Tsai et al.

mutation types and mutant RUNX1 activities of the patients are progressed to sAML compared with 69 of 124 (55.6%) in the summarized in Table 1. The seven mutants which induced high-activity group (P < 0.0001). Likewise, MDS patients with comparable levels of C-FMS protein in Fig. 3D were set as low activities had a shorter time to sAML (12.9 3.1 months high-activity mutations. vs. 21.7 4.7 months, P ¼ 0.008; Fig. 4B). There were no significant differences in overall survival between the 2 groups The correlation between mutant RUNX1 activities and in both CMML (median, 12.8 2.5 months vs. 8.7 1.9 clinicohematological characteristics months, P ¼ 0.238; Fig. 4C) and MDS patients (median, 18.2 To further correlate the role of RUNX1 mutation in disease 2.1 months vs. 16.0 1.4 months, P ¼ 0.246; Fig. 4D). progression, clinicohematological characteristics and outcome We also analyzed the correlation of cytogenetics and muta- were analyzed. The patients without RUNX1 mutation were tional status of the coexisting mutated genes with the risk of sAML counted as high activity. A comparison of clinicohematological transformation (Supplementary Table S2). Cytogenetics did not features between the 2 groups of high and low activities in MDS predict sAML transformation in both MDS and CMML patients. and CMML is shown in Supplementary Table S1. No differences Mutational status of RUNX1 and its mutant activity were the were observed with respect to gender, hemoglobin level, white most significant predictors of sAML transformation. Of the addi- blood cell (WBC) count, percentage of blasts in BM, WHO tional coexisting mutations in both MDS and CMML, only IDH2 subtype, cytogenetic risk group, or revised International Prog- mutation was associated with the risk of sAML transformation in nostic Scoring System (IPSS-R) between mutation-positive and CMML. Among the coexisting mutations, NPM1 and TP53 muta- -negative patients in both MDS and CMML. Patients with low tions predicted inferior sAML-free survival in MDS patients, and activity were significantly older in MDS (P ¼ 0.0002) and CEBPa mutation was associated with rapid sAML progression younger in CMML (P ¼ 0.027). Platelet counts were signifi- in CMML patients. Multivariate analyses showed that mutant cantly lower in the low-activity group of CMML (P ¼ 0.002). RUNX1 activity remained an independent predictor for sAML-free Fifteen of 24 (62.5%) patients with CMML in the low-activity survival (P ¼ 0.010) in MDS, but not for CMML (P ¼ 0.26). groupprogressedtosAMLcomparedwith19of60(31.7%) Although NPM1 mutation (n ¼ 6) in MDS and CEBPa mutation patients with high RUNX1 activity (P ¼ 0.014). CMML patients (n ¼ 5) in CMML had a higher HR for sAML-free survival, the with low activities transformed to sAML more rapidly (23.3 numbers of patients carrying these mutations were very small. 12.6 months vs. median not reached, P ¼ 0.011; Fig. 4A). For Taken together, RUNX1 mutation status with its mutant activity patients with MDS, all of the patients in the low-activity group had the most important impact on sAML transformation.

Figure 4. Activities of the RUNX1 mutants on C-FMS induction predict sAML transformation. Kaplan–Meier estimates of sAML transformation in patients with CMML (A) and MDS (B), and overall survival for CMML (C) and MDS (D) according to RUNX1 activities on the induction of C-FMS.

OF8 Clin Cancer Res; 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

RUNX1 Activity Predicts sAML from MDS/CMML

Discussion by Harada and colleagues previously (2, 10), and our data were consistent with their observation. Recently, Bresciani and collea- We systematically analyzed all RUNX1 mutants by in vitro gues reported that RUNX1 can drive the emergence of nascent functional assays to unveil the possible underlying mechanisms hematopoietic stem cells without its heterodimeric partner CBFb of sAML transformation. Our study provided clear molecular (24), suggesting parts of RUNX1's biologic functions are inde- evidences to verify DNA binding and heterodimerization activi- pendent of CBFb. In addition, lots of cellular proteins, Myb, Ets, ties of mutant RUNX1. We found that DNA binding ability of ALY, TLE/Groucho, p300, CBP, YAP, SIN3A, MOZ, PML, PRMT1, RUNX1 was more important than heterodimerization with CBFb TAL1, MLL, Shp2, SWI/SNF, etc., have been described to interact on its target gene regulation. Here, we chose C-FMS as an indicator with RUNX1 (25). Of note, RUNX1 interacted with several tran- for measuring the remaining biologic activities of mutant RUNX1, scriptional factors through a dynamic combinatorial mechanism because the regulatory region of RUNX1 on C-FMS promoter is to orchestrate hematopoietic differentiation (26, 27). The inter- much well-defined compared with other RUNX1 target genes acting partners that determine turn-on or turn-off of the RUNX1- (21, 23). We adapted the luciferase reporter assay to determine targeting genes and downstream signal cascades with respect to mutant RUNX1 activities quantitatively, and also provided clear the cell fate might be more important than mutant RUNX1 per se. evidences to prove that transcriptional activities of mutant Therefore, to more accurately evaluate the biologic activity of RUNX1 well reflecting their biologic activities on target protein RUNX1 mutants, in vivo cellular functional tests would be much induction in myeloid cells. RUNX1 could also regulate the pro- more representative, which would take all the complicated part- moters of IL3 and GM-CSF, and we predicted that the regulation nerships of mutant RUNX1 into account. RUNX1a protein, which trend of mutant RUNX1 on IL3 or GM-CSF activation might be retains the RHD but lacks the TAD, is considered as an antagonist similar, but the relative cutoff activity on IL3 or GM-CSF might be for RUNX1b and may inhibit RUNX1b function (17). In Fig. 3A, different from that (0.52) of C-FMS activity. Because we correlated none of the cell lines had detectable RUNX1a protein. We had also the reporter assay data of C-FMS promoter with the clinical performed RT-Q-PCR to examine the RUNX1a mRNA level in outcomes and determined the cutoff value (0.52), the bias of these cell lines. Consistent with the Western blot results, none of only using C-FMS promoter activity to represent RUNX activity as the cells were detectable of RUNX1a mRNA (data not shown). It the experimental readout might be limited. has been reported that RUNX1a is preferentially expressed in Based on our data, DNA and CBFb binding abilities of RUNX1 immature hematopoietic cell compartments in cord blood and mutants could be easily predicted according to their locations and plays a role in stem/progenitor cells (28, 29). All the cell lines we patterns. However, the ambiguous mutations, such as single point tested are lineage committed and differentiated cells, hence mutation within RHD or TAD, still need functional assays to RUNX1a expression might be shut down in these cell lines. determine their binding activities. Moreover, the DNA and CBFb Val 425Gly and Met439Leu variants of RUNX1 were described binding assays could not necessarily reflect the biologic activities in de novo AML patients previously but were also detected in of RUNX1 mutants on the induction of C-FMS. The Ser114Leu complete remission samples (11), indicating both mutations are mutant could not bind well to C-FMS promoter and CBFb in vitro, not related to AML pathogenesis. In our functional assays, the 2 but could induce C-FMS expression with comparable level as wt- mutants always displayed equivalent biologic activities as those RUNX1 in U937 cells. The C-terminal frameshift mutants all for wt-RUNX1, supporting they are single nucleotide variants displayed strong interaction with DNA and CBFb, but only weak without functional changes. As these 2 variants were not leuke- activities on C-FMS induction, except the Asp424Glyfs 149 mia-associated, we thus did not count them as mutations in the mutant which does not affect the TAD. The dominant-negative outcome analyses. Figure 5 provides a brief summary of all the effects of these C-terminal frameshift mutants were demonstrated results of our functional assays.

Figure 5. A cartoon of RUNX1b mutants in regulating C-FMS activation. Mutation in RUNX1 may affect its dimerization with CBFb, DNA binding activity, or downstream C-FMS activation. Effects of individual mutants were listed according to the results from current study. Dimerization, DNA binding activity, and C-FMS induction activity were detected by co-IP assay, EMSA analysis, and luciferase reporter assay, respectively.

www.aacrjournals.org Clin Cancer Res; 2015 OF9

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Tsai et al.

According to the results of luciferase reporter assay and Western examined the impacts of additional 19 coexisting mutations blot, we can postulate general rules to predict the biologic activ- relatedtomyeloidneoplasms.Wefoundthatthetwomutated ities of RUNX1 mutants: (i) the mutations with nucleotide inser- genes with higher HR, i.e., NPM1 and CEBPa,werepresentin tion or that resulted in frameshift or destructing the inte- very few patients. Our results demonstrated that the activities grity of RHD or TAD are all low-activity mutations; (ii) missense on C-FMS of RUNX1 mutants had the most important impact mutations that directly affect DNA interaction (Arg80, Arg174, on sAML transformation among all the coexisting mutations and Arg177) or DNA recognition (Lys83, Arg142, and Asp171) analyzed in the present study. are low-activity mutations; (iii) missense mutations located In summary, we demonstrated the prognostic relevance of the outside RHD or TAD are possibly high-activity mutations or biologic activities of RUNX1 mutants in patients with CMML and SNPs; (iv) point mutations that are located within RHD or TAD MDS. Patients with lower activities of RUNX1 mutants exhibit a and nonsense mutations terminated behind TAD could be deter- higher risk and shorter time to sAML transformation. In these mined for their activities by luciferase reporter assay. patients, our results showed that mutant RUNX1 activity was a We have previously reported a high frequency of RUNX1 useful indicator to predict the outcome of the diseases, which in mutations in CMML and the C-terminal mutations had a sig- turn had clinical implications of identifying patients for early nificant higher risk of sAML transformation (1). However, the therapeutic intervention with a more effective agent even in underlying mechanisms of sAML transformation related to patients who did not have high risk IPSS-R. RUNX1 mutation were poorly understood. Herein, we provided molecular evidences to link mutant RUNX1 activities with the Disclosure of Potential Conflicts of Interest expression level of its target gene, C-FMS. Moreover, we showed No potential conflicts of interest were disclosed. that the activities of RUNX1 mutants had prognostic impact on sAML transformation. Apart from the mutational status of Authors' Contributions RUNX1 gene (1), we demonstrated that the biologic activity of Conception and design: S.-C. Tsai, L.-Y. Shih, S.-T. Liang, D.-C. Liang mutant RUNX1 was a more important predictor of disease Development of methodology: S.-C. Tsai, L.-Y. Shih, S.-T. Liang, C.-F. Huang progression. Patients with lower activities of RUNX1 mutants Acquisition of data (provided animals, acquired and managed patients, had a higher risk and shorter time to sAML transformation. Not provided facilities, etc.): S.-C. Tsai, L.-Y. Shih, M.-C. Kuo, Y.-S. Shih, M.-C. Chiu only in CMML, the similar impact of RUNX1 mutations was also Analysis and interpretation of data (e.g., statistical analysis, biostatistics, observed in MDS. Although RUNX1 mutations and their func- computational analysis): S.-C. Tsai, L.-Y. Shih, Y.-J. Huang, T.-H. Lin Writing, review, and/or revision of the manuscript: S.-C. Tsai, L.-Y. Shih, tional studies have been reported in several studies of MDS, S.-T. Liang, Y.-J. Huang, D.-C. Liang CMML, and AML (2, 11), the present study was the first one to Administrative, technical, or material support (i.e., reporting or organizing directly correlate the biologic functions with clinical outcome data, constructing databases): L.-Y. Shih, C.-F. Huang, D.-C. Liang and showed their prognostic relevance. Most importantly, the Study supervision: L.-Y. Shih remaining activities of mutant RUNX1 could be a predictor of disease progression. The luciferase reporter construct of C-FMS Acknowledgments promoter and reagents are commercially available. If wt- and The authors thank Drs. Po-Nan Wang, Jin-Hou Wu, Po Dunn, and Tung- Liang Lin for providing clinical data of their patients and Ting-Yu Huang for mutant RUNX1 expression plasmids are available, the results are secretarial assistance. easily reproduced and the processes could be standardized for potential clinical use. Grant Support Familial platelet disorder (FPD) is an autosomal dominant – This study was supported by grant NHRI-EX103-10003NI from National disorder caused by germline heterozygous inherited mutation of Health Research Institute (to L.-Y. Shih), grants OMRPG380031 and RUNX1 gene (30). Patients with FPD have propensity to develop OMRPG3C0021 from Chang Gung Memorial Hospital (to L.-Y. Shih), grant myeloid malignancies with a rate ranging between 20% and MMH-E-101-09 from the Mackay Memorial Hospital (to D.-C. Liang), grant 65%. RUNX1 mutations identified in FPD/AML patients were MOST103-2321-B-182-015 from Ministry of Science and Technology, Taiwan mainly located at N-terminus (9, 30–32). Those mutants would (to L.-Y. Shih), and grant MOHW103-TD-B-111-09 from Ministry of Health express low-activity RUNX1 according to our above postulation, and Welfare, Taiwan (to L.-Y. Shih). fi The costs of publication of this article were defrayed in part by the payment which further supports our ndings that RUNX1 mutant activity of page charges. This article must therefore be hereby marked advertisement is a predictor of sAML transformation. in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. As additional gene mutations and chromosomal abnormal- ities might affect outcomes and thereby the correlation with Received August 24, 2014; revised March 11, 2015; accepted March 30, 2015; functional attributes of RUNX1 might be affected. We thus also published OnlineFirst April 3, 2015.

References 1. Kuo MC, Liang DC, Huang CF, Shih YS, Wu JH, Lin TL, et al. RUNX1 plastic syndrome, but not in myelofibrosis with myeloid metaplasia. Eur J mutations are frequent in chronic myelomonocytic leukemia and muta- Haematol 2005;74:47–53. tions at the C-terminal region might predict acute myeloid leukemia 4. Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. AML1, the transformation. Leukemia 2009;23:1426–31. target of multiple chromosomal translocations in human leukemia, is 2. Harada H, Harada Y, Niimi H, Kyo T, Kimura A, Inaba T. High incidence of essential for normal fetal liver hematopoiesis. Cell 1996;84:321–30. somatic mutations in the AML1/RUNX1 gene in myelodysplastic syn- 5. Lorsbach RB, Moore J, Ang SO, Sun W, Lenny N, Downing JR. Role of drome and low blast percentage myeloid leukemia with myelodysplasia. RUNX1 in adult hematopoiesis: analysis of RUNX1-IRES-GFP knock-in Blood 2004;103:2316–24. mice reveals differential lineage expression. Blood 2004;103:2522–9. 3. Steensma DP, Gibbons RJ, Mesa RA, Tefferi A, Higgs DR. Somatic point 6. Ichikawa M, Asai T, Saito T, Seo S, Yamazaki I, Yamagata T, et al. AML-1 is mutations in RUNX1/CBFA2/AML1 are common in high-risk myelodys- required for megakaryocytic maturation and lymphocytic differentiation,

OF10 Clin Cancer Res; 2015 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

RUNX1 Activity Predicts sAML from MDS/CMML

but not for maintenance of hematopoietic stem cells in adult hematopoi- 19. Nimer S, Zhang J, Avraham H, Miyazaki Y. Transcriptional regula- esis. Nat Med 2004;10:299–304. tion of interleukin-3 expression in megakaryocytes. Blood 1996;88: 7. Imai Y, Kurokawa M, Izutsu K, Hangaishi A, Takeuchi K, Maki K, et al. 66–74. Mutations of the AML1 gene in and their 20. Cockerill PN, Osborne CS, Bert AG, Grotto RJ. Regulation of GM-CSF functional implications in leukemogenesis. Blood 2000;96:3154–60. gene transcription by core-binding factor. Cell Growth Differ 1996; 8. Chen CY, Lin LI, Tang JL, Ko BS, Tsay W, Chou WC, et al. RUNX1 gene 7:917–22. mutation in primary myelodysplastic syndrome–the mutation can be 21. Zhang DE, Fujioka K, Hetherington CJ, Shapiro LH, Chen HM, Look AT, detected early at diagnosis or acquired during disease progression and is et al. Identification of a region which directs the monocytic activity of associated with poor outcome. Br J Haematol 2007;139:405–14. the colony-stimulating factor 1 (macrophage colony-stimulating factor) 9. Michaud J, Wu F, Osato M, Cottles GM, Yanagida M, Asou N, et al. In vitro promoter and binds PEBP2/CBF (AML1). Mol Cell Biol analyses of known and novel RUNX1/AML1 mutations in dominant 1994;14:8085–95. familial platelet disorder with predisposition to acute myelogenous leu- 22.TsaiSC,LinSJ,ChenPW,LuoWY,YehTH,WangHW,etal.EBVZta kemia: implications for mechanisms of pathogenesis. Blood 2002;99: protein induces the expression of interleukin-13, promoting the 1364–72. proliferation of EBV-infected B cells and lymphoblastoid cell lines. 10. Harada H, Harada Y, Tanaka H, Kimura A, Inaba T. Implications of somatic Blood 2009;114:109–18. mutations in the AML1 gene in radiation-associated and therapy-related 23. Zhang DE, Hetherington CJ, Meyers S, Rhoades KL, Larson CJ, Chen HM, myelodysplastic syndrome/acute myeloid leukemia. Blood 2003;101: et al. CCAAT -binding protein (C/EBP) and AML1 (CBF alpha2) 673–80. synergistically activate the macrophage colony-stimulating factor receptor 11. Tang JL, Hou HA, Chen CY, Liu CY, Chou WC, Tseng MH, et al. AML1/ promoter. Mol Cell Biol 1996;16:1231–40. RUNX1 mutations in 470 adult patients with de novo acute myeloid 24. Bresciani E, Carrington B, Wincovitch S, Jones M, Gore AV, Weinstein BM, leukemia: prognostic implication and interaction with other gene altera- et al. CBFbeta and RUNX1 are required at 2 different steps during the tions. Blood 2009;114:5352–61. development of hematopoietic stem cells in zebrafish. Blood 2014;124: 12. Preudhomme C, Warot-Loze D, Roumier C, Grardel-Duflos N, Garand 70–8. R, Lai JL, et al. High incidence of biallelic point mutations in the 25. Chuang LS, Ito K, Ito Y. RUNX family: Regulation and diversification of Runt domain of the AML1/PEBP2 alpha B gene in Mo acute myeloid roles through interacting proteins. Int J Cancer 2013;132:1260–71. leukemia and in myeloid malignancies with acquired 21. 26. Pencovich N, Jaschek R, Tanay A, Groner Y. Dynamic combinatorial Blood 2000;96:2862–9. interactions of RUNX1 and cooperating partners regulates megakaryocytic 13. Kao HW, Liang DC, Wu JH, Kuo MC, Wang PN, Yang CP, et al. Gene differentiation in cell line models. Blood 2011;117:e1–14. mutation patterns in patients with minimally differentiated acute myeloid 27. Wilson NK, Foster SD, Wang X, Knezevic K, Schutte J, Kaimakis P, et al. leukemia. Neoplasia 2014;16:481–8. Combinatorial transcriptional control in blood stem/progenitor cells: 14. Ghozi MC, Bernstein Y, Negreanu V, Levanon D, Groner Y. Expression of genome-wide analysis of ten major transcriptional regulators. Cell Stem the human acute myeloid leukemia gene AML1 is regulated by two Cell 2010;7:532–44. promoter regions. Proc Natl Acad Sci U S A 1996;93:1935–40. 28. Tsuzuki S, Hong D, Gupta R, Matsuo K, Seto M, Enver T. Isoform-specific 15. Ogawa E, Maruyama M, Kagoshima H, Inuzuka M, Lu J, Satake M, et al. potentiation of stem and progenitor cell engraftment by AML1/RUNX1. PEBP2/PEA2 represents a family of transcription factors homologous to PLoS Med 2007;4:e172. the products of the Drosophila runt gene and the human AML1 gene. Proc 29. Tsuzuki S, Seto M. Expansion of functionally defined mouse hematopoie- Natl Acad Sci U S A 1993;90:6859–63. tic stem and progenitor cells by a short isoform of RUNX1/AML1. 16. Kanno T, Kanno Y, Chen LF, Ogawa E, Kim WY, Ito Y. Intrinsic transcrip- Blood 2012;119:727–35. tional activation-inhibition domains of the polyomavirus enhancer bind- 30. Song WJ, Sullivan MG, Legare RD, Hutchings S, Tan X, Kufrin D, et al. ing protein 2/ alpha subunit revealed in the presence of Haploinsufficiency of CBFA2 causes familial thrombocytopenia with the beta subunit. Mol Cell Biol 1998;18:2444–54. propensity to develop acute myelogenous leukaemia. Nat Genet 1999;23: 17. Tanaka T, Tanaka K, Ogawa S, Kurokawa M, Mitani K, Nishida J, et al. An 166–75. acute myeloid leukemia gene, AML1, regulates hemopoietic myeloid cell 31. Owen CJ, Toze CL, Koochin A, Forrest DL, Smith CA, Stevens JM, et al. differentiation and transcriptional activation antagonistically by two alter- Five new pedigrees with inherited RUNX1 mutations causing familial native spliced forms. EMBO J 1995;14:341–50. platelet disorder with propensity to myeloid malignancy. Blood 2008; 18. Tahirov TH, Inoue-Bungo T, Morii H, Fujikawa A, Sasaki M, Kimura K, et al. 112:4639–45. Structural analyses of DNA recognition by the AML1/Runx-1 Runt domain 32. Osato M. Point mutations in the RUNX1/AML1 gene: another actor in and its allosteric control by CBFbeta. Cell 2001;104:755–67. RUNX leukemia. Oncogene 2004;23:4284–96.

www.aacrjournals.org Clin Cancer Res; 2015 OF11

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst April 3, 2015; DOI: 10.1158/1078-0432.CCR-14-2203

Biological Activities of RUNX1 Mutants Predict Secondary Acute Leukemia Transformation from Chronic Myelomonocytic Leukemia and Myelodysplastic Syndromes

Shu-Chun Tsai, Lee-Yung Shih, Sung-Tzu Liang, et al.

Clin Cancer Res Published OnlineFirst April 3, 2015.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-14-2203

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2015/04/04/1078-0432.CCR-14-2203.DC1

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2015/06/02/1078-0432.CCR-14-2203. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on October 6, 2021. © 2015 American Association for Cancer Research.