Proteasome inhibition targets the KMT2A transcriptional complex in acute lymphoblastic leukemia

Jennifer L Kamens, Stephanie Nance, Cary Koss, Beisi Xu, Anitria Cotton, Jeannie W Lam, Elizabeth A R Garfinkle, Pratima Nallagatla, Amelia M R Smith, Sharnise Mitchell, Jing Ma, Duane Currier, William C Wright, Kanisha Kavdia, Vishwajeeth R Pagala, Wonil Kim, LaShanale M Wallace, Ji-Hoon Cho, Yiping Fan, Aman Seth, Nathaniel Twarog, John K Choi, Esther A Obeng, Mark E Hatley, Monika L Metzger, Hiroto Inaba, Sima Jeha, Jeffrey E Rubnitz, Junmin Peng, Taosheng Chen, Anang A Shelat, R Kiplin Guy, Tanja A Gruber, Jennifer L Kamens, Stephanie Nance, Cary Koss, Beisi Xu, Anitria Cotton, Jeannie W Lam, Elizabeth A R Garfinkle, Pratima Nallagatla, Amelia M R Smith, Sharnise Mitchell, Jing Ma, Duane Currier, William C Wright, Kanisha Kavdia, Vishwajeeth R Pagala, Wonil Kim, LaShanale M Wallace, Ji-Hoon Cho, Yiping Fan, Aman Seth, Nathaniel Twarog, John K Choi, Esther A Obeng, Mark E Hatley, Monika L Metzger, Hiroto Inaba, Sima Jeha, Jeffrey E Rubnitz, Junmin Peng, Taosheng Chen, Anang A Shelat, R Kiplin Guy, Tanja A Gruber

Abstract

Rearrangments in Histone-lysine-N-methyltransferase 2A (KMT2Ar) are associated with pediatric, adult and therapy-induced acute leukemias. Infants with KMT2Ar acute lymphoblastic leukemia (ALL) have a poor prognosis with an event-free-survival of 38%. Herein we evaluate 1116 FDA approved compounds in primary KMT2Ar infant ALL specimens and identify a sensitivity to proteasome inhibition. Upon exposure to this class of agents, cells demonstrate a depletion of histone H2B monoubiquitination (H2Bub1) and histone H3 lysine 79 dimethylation (H3K79me2) at KMT2A target genes in addition to a downregulation of the KMT2A gene expression signature, providing evidence that it targets the KMT2A transcriptional complex and alters the epigenome. A cohort of relapsed/refractory KMT2Ar patients treated with this approach on a compassionate basis had an overall response rate of 90%. In conclusion, we report on a high throughput drug screen in primary pediatric leukemia specimens whose results translate into clinically meaningful responses. This innovative treatment approach is now being evaluated in a multi-institutional upfront trial for infants with newly diagnosed ALL.

Conflict of interest statement

The authors declare no competing interests.

© 2023. The Author(s).

Figures

Fig. 1. High throughput screening of infant…
Fig. 1. High throughput screening of infant ALL.
A Study design. Primary leukemia blasts were thawed and injected into immunodeficient mice. Cells were harvested from moribund mice as described in “Methods” and viably stored in liquid nitrogen. Samples were thawed for drug sensitivity studies at a later time point. Figure created with BioRender.com, academic license. B In vitro activity of FDA-approved compounds. Six passaged patient specimens were thawed and cultured in the presence of compounds at a single concentration of 10 µM in duplicate as described in “Methods”. Normalized activity, defined as percent toxicity, was determined for each compound with each patient sample. Each data point corresponds to a single compound, the size of the data point indicates the number of patient samples with 80% or greater activity for that compound and the color indicates the coefficient of variation within the patient samples treated with that compound. Compounds are grouped according to class (see Supplementary Data 1). The number of compounds for each class with greater than 80% activity in five or more patient samples are shown. Source data are provided as a Source data file.
Fig. 2. Active compounds.
Fig. 2. Active compounds.
A Overview of FDA screening hits. 1116 unique compounds were screened. 172 compounds demonstrated >80% activity across five or more patient samples. Drugs with untolerable side effects or those that lacked systemic preparations were removed. The top 43 compounds underwent a secondary validation with a 10-point dose–response assay in technical triplicates. B Median IC50 values of top 43 compounds. 50% of the top compounds with IC50 values less than 2 µM belonged to one of three classes of agents: proteasome inhibitors, HDAC inhibitors, and anthracyclines.
Fig. 3. Bortezomib depletes H2Bub1 and H3K79me2…
Fig. 3. Bortezomib depletes H2Bub1 and H3K79me2 leading to downregulation of the KMT2A transcriptional program.
A Ten patient samples were treated with 5 nM bortezomib, histones were extracted and blotted for total and ubiquitinated H2B. The ratio of H2Bub1 to total H2B levels was determined by densitometry. Pre-treatment specimens were set to one and relative levels at 2 h are shown with the standard error of the mean. B Four patient samples were exposed to 5 nM bortezomib for 6 h or left untreated. CUT&RUN reaction with an H2Bub1 antibody followed by quantitative PCR for two KMT2A target genes, MEIS1 and CDK6, was done. Fold change over IgG was calculated and untreated specimens were set to one. The average across the four patient specimens with the standard error of the mean is shown. C ChIP-Rx of the KMT2Ar infant ALL cell line SEM at four time points (0, 2, 4, and 6 h following exposure to bortezomib) for H2Bub1 and H3K79me2 was done to quantitatively assess this epigenetic mark over time throughout the genome. Heatmaps of H2Bub1 and H3K79me2 genome-wide are shown. D Profiles at HOXA1-A11, FLT3, and MEIS1 gene loci. E Five patient specimens were grown in the presence or absence of 5 nM bortezomib for 20 h followed by RNA extraction from 2 million live cells and sequencing. Cells were analyzed at 20 h based on the half-life of mRNA which is 10 h. A heatmap of differentially expressed genes is shown. F Log2 fold change of proteasome complex transcripts, KMT2Ar target genes, and the cMYC target genes TP53 and CDKN2A are shown following exposure to bortezomib. The upregulation of proteasome complex genes demonstrates target inhibition. G The KMT2Ar infant ALL cell line (SEM) was grown in the presence or absence of bortezomib. CUT&RUN reactions at 0, 2, 4, and 6 h following exposure to bortezomib was done with an N-Terminal KMT2A antibody and DOT1L. Quantitative PCR for two KMT2A target genes, MEIS1 and CDK6 was determined. Fold change over IgG was calculated and untreated specimens were set to one. The average across four independent experiments are shown with the standard error of the mean. Source data are provided as a Source data file.
Fig. 4. Vorinostat is synergistic with bortezomib…
Fig. 4. Vorinostat is synergistic with bortezomib in KMT2Ar leukemia.
A SEM cells were incubated for 8 h in 3 nM bortezomib, 1 µM vorinostat, or both followed by staining for acetylated tubulin (green) and vimentin (red) to visualize microtubules and aggresomes, respectively. Representative images are shown. 100 cells were scored per group by three individuals independently for the presence of aggresomes, average counts with the standard error of the mean are shown. Unpaired t test comparing bortezomib to bortezomib+vorinostat, two-tailed p value equals 0.0276. Source data are provided as a Source data file. B Cytotoxicity assay of bortezomib in combination with vorinostat. The KMT2Ar infant ALL cell line SEM and two patient samples were grown in the presence of bortezomib and vorinostat at the indicated concentrations for 72 h in technical triplicates. Viability was assessed by cell titer glo. Estimated effect of various levels of vorinostat in combination with bortezomib are shown. Curve shifts to the left indicate potentiation. EC50 values of bortezomib in the presence of varying concentrations of vorinostat are presented in Table 1. See Supplementary Fig. 17 for the best fit BRAID surface for each combination with estimated value of κ and confidence interval.
Fig. 5. Restoration of trilineage hematopoiesis following…
Fig. 5. Restoration of trilineage hematopoiesis following treatment with bortezomib and vorinostat containing chemotherapy regimen in a refractory patient.
A Bone marrow aspirates of case 3b (see Table 2) at the time of relapse post-transplant and post-treatment with bortezomib and vorinostat-containing chemotherapy regimen are shown at ×10 and ×100 magnification. Flow-based MRD and RT-PCR for the KMT2A-MLLT1 oncogene present in this patient were both negative post-treatment. B Model of proteasome and HDAC inhibition in KMT2Ar leukemia. Proteasome inhibitors deplete H2Bub1 leading to downregulation of the KMT2A gene expression program and accumulation of the KMT2A fusion which triggers caspase 812. HDACi prevent aggresome formation and increase in histone acetylation and death gene induction promoting apoptosis.

References

    1. Pieters R, et al. Outcome of infants younger than 1 year with acute lymphoblastic leukemia treated with the interfant-06 protocol: results from an international phase III randomized study. J. Clin. Oncol. 2019;37:2246–2256. doi: 10.1200/JCO.19.00261.
    1. Pui CH, et al. Treating childhood acute lymphoblastic leukemia without cranial irradiation. N. Engl. J. Med. 2009;360:2730–2741. doi: 10.1056/NEJMoa0900386.
    1. Meyer C, et al. The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32:273–284. doi: 10.1038/leu.2017.213.
    1. Andersson AK, et al. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat. Genet. 2015;47:330–337. doi: 10.1038/ng.3230.
    1. Bernt KM, et al. MLL-rearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20:66–78. doi: 10.1016/j.ccr.2011.06.010.
    1. Daigle SR, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20:53–65. doi: 10.1016/j.ccr.2011.06.009.
    1. Stein EMG-MG, et al. The DOT1L inhibitor EPZ-5676: safety and activity in relapsed/refractory patients with MLL-rearranged leukemia. Blood. 2014;124:387. doi: 10.1182/blood.V124.21.387.387.
    1. Pieters R, et al. Relation between age, immunophenotype and in vitro drug resistance in 395 children with acute lymphoblastic leukemia–implications for treatment of infants. Leukemia. 1998;12:1344–1348. doi: 10.1038/sj.leu.2401129.
    1. Ramakers-van Woerden NL, et al. In vitro drug-resistance profile in infant acute lymphoblastic leukemia in relation to age, MLL rearrangements and immunophenotype. Leukemia. 2004;18:521–529. doi: 10.1038/sj.leu.2403253.
    1. Szczepanek J, et al. Differential ex vivo activity of bortezomib in newly diagnosed paediatric acute lymphoblastic and myeloblastic leukaemia. Anticancer Res. 2010;30:2119–2124.
    1. Boccadoro M, Morgan G, Cavenagh J. Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy. Cancer Cell Int. 2005;5:18. doi: 10.1186/1475-2867-5-18.
    1. Liu H, et al. Proteasome inhibitors evoke latent tumor suppression programs in pro-B MLL leukemias through MLL-AF4. Cancer Cell. 2014;25:530–542. doi: 10.1016/j.ccr.2014.03.008.
    1. Prenzel T, et al. Estrogen-dependent gene transcription in human breast cancer cells relies upon proteasome-dependent monoubiquitination of histone H2B. Cancer Res. 2011;71:5739–5753. doi: 10.1158/0008-5472.CAN-11-1896.
    1. Mimnaugh EG, Chen HY, Davie JR, Celis JE, Neckers L. Rapid deubiquitination of nucleosomal histones in human tumor cells caused by proteasome inhibitors and stress response inducers: effects on replication, transcription, translation, and the cellular stress response. Biochemistry. 1997;36:14418–14429. doi: 10.1021/bi970998j.
    1. Lee JS, et al. Histone crosstalk between H2B monoubiquitination and H3 methylation mediated by COMPASS. Cell. 2007;131:1084–1096. doi: 10.1016/j.cell.2007.09.046.
    1. Zhu B, et al. Monoubiquitination of human histone H2B: the factors involved and their roles in HOX gene regulation. Mol. Cell. 2005;20:601–611. doi: 10.1016/j.molcel.2005.09.025.
    1. McGinty RK, Kim J, Chatterjee C, Roeder RG, Muir TW. Chemically ubiquitylated histone H2B stimulates hDot1L-mediated intranucleosomal methylation. Nature. 2008;453:812–816. doi: 10.1038/nature06906.
    1. Wang E, et al. Histone H2B ubiquitin ligase RNF20 is required for MLL-rearranged leukemia. Proc. Natl Acad. Sci. USA. 2013;110:3901–3906. doi: 10.1073/pnas.1301045110.
    1. Stumpel DJ, et al. Connectivity mapping identifies HDAC inhibitors for the treatment of t(4;11)-positive infant acute lymphoblastic leukemia. Leukemia. 2012;26:682–692. doi: 10.1038/leu.2011.278.
    1. Ahmad K, et al. Inhibition of class I HDACs abrogates the dominant effect of MLL-AF4 by activation of wild-type MLL. Oncogenesis. 2014;3:e127. doi: 10.1038/oncsis.2014.39.
    1. Kikuchi J, et al. Histone deacetylases are critical targets of bortezomib-induced cytotoxicity in multiple myeloma. Blood. 2010;116:406–417. doi: 10.1182/blood-2009-07-235663.
    1. Carew JS, Giles FJ, Nawrocki ST. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett. 2008;269:7–17. doi: 10.1016/j.canlet.2008.03.037.
    1. Dimopoulos M, et al. Vorinostat or placebo in combination with bortezomib in patients with multiple myeloma (VANTAGE 088): a multicentre, randomised, double-blind study. Lancet Oncol. 2013;14:1129–1140. doi: 10.1016/S1470-2045(13)70398-X.
    1. Premkumar DR, Jane EP, Agostino NR, DiDomenico JD, Pollack IF. Bortezomib-induced sensitization of malignant human glioma cells to vorinostat-induced apoptosis depends on reactive oxygen species production, mitochondrial dysfunction, Noxa upregulation, Mcl-1 cleavage, and DNA damage. Mol. Carcinog. 2013;52:118–133. doi: 10.1002/mc.21835.
    1. Simms-Waldrip T, et al. The aggresome pathway as a target for therapy in hematologic malignancies. Mol. Genet. Metab. 2008;94:283–286. doi: 10.1016/j.ymgme.2008.03.012.
    1. Catley L, et al. Aggresome induction by proteasome inhibitor bortezomib and alpha-tubulin hyperacetylation by tubulin deacetylase (TDAC) inhibitor LBH589 are synergistic in myeloma cells. Blood. 2006;108:3441–3449. doi: 10.1182/blood-2006-04-016055.
    1. Zhang J, Zhong Q. Histone deacetylase inhibitors and cell death. Cell Mol. Life Sci. 2014;71:3885–3901. doi: 10.1007/s00018-014-1656-6.
    1. Bai B, et al. Deep multilayer brain proteomics identifies molecular networks in Alzheimer’s disease progression. Neuron. 2020;105:975–991 e977. doi: 10.1016/j.neuron.2019.12.015.
    1. Zhang X, et al. Proteome-wide identification of ubiquitin interactions using UbIA-MS. Nat. Protoc. 2018;13:530–550. doi: 10.1038/nprot.2017.147.
    1. Twarog NR, Stewart E, Hammill CV, Shelat AA. BRAID: a unifying paradigm for the analysis of combined drug action. Sci. Rep. 2016;6:25523. doi: 10.1038/srep25523.
    1. Parker C, et al. Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial. Lancet. 2010;376:2009–2017. doi: 10.1016/S0140-6736(10)62002-8.
    1. Messinger Y, et al. Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatr. Blood Cancer. 2010;55:254–259. doi: 10.1002/pbc.22456.
    1. Messinger YH, et al. Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: Therapeutic Advances in Childhood Leukemia & Lymphoma (TACL) Study. Blood. 2012;120:285–290. doi: 10.1182/blood-2012-04-418640.
    1. Kaspers GJ, et al. Improved outcome in pediatric relapsed acute myeloid leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group. J. Clin. Oncol. 2013;31:599–607. doi: 10.1200/JCO.2012.43.7384.
    1. Koss C, et al. Targeted inhibition of the MLL transcriptional complex by proteosome inhibitors elicits a high response rate in relapsed/refractory MLL rearranged leukemia. Blood. 2014;124:972–972. doi: 10.1182/blood.V124.21.972.972.
    1. Cheung LC, et al. Preclinical evaluation of carfilzomib for infant KMT2A-rearranged acute lymphoblastic leukemia. Front Oncol. 2021;11:631594. doi: 10.3389/fonc.2021.631594.
    1. Walters BJ, Zovkic IB. Building up and knocking down: an emerging role for epigenetics and proteasomal degradation in systems consolidation. Neuroscience. 2015;300:39–52. doi: 10.1016/j.neuroscience.2015.05.005.
    1. Harned TM, Gaynon P. Relapsed acute lymphoblastic leukemia: current status and future opportunities. Curr. Oncol. Rep. 2008;10:453–458. doi: 10.1007/s11912-008-0070-3.
    1. Horton TM, et al. Toxicity assessment of molecularly targeted drugs incorporated into multiagent chemotherapy regimens for pediatric acute lymphocytic leukemia (ALL): review from an international consensus conference. Pediatr. Blood Cancer. 2010;54:872–878. doi: 10.1002/pbc.22414.
    1. Ko RH, et al. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J. Clin. Oncol. 2010;28:648–654. doi: 10.1200/JCO.2009.22.2950.
    1. Horton TM, et al. Bortezomib interactions with chemotherapy agents in acute leukemia in vitro. Cancer Chemother. Pharmacol. 2006;58:13–23. doi: 10.1007/s00280-005-0135-z.
    1. Van der Velden VH, et al. Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol. Leukemia. 2009;23:1073–1079. doi: 10.1038/leu.2009.17.
    1. Hadley, W. Ggplot2 (Springer Science+Business Media, LLC, 2016).
    1. Shechter D, Dormann HL, Allis CD, Hake SB. Extraction, purification and analysis of histones. Nat. Protoc. 2007;2:1445–1457. doi: 10.1038/nprot.2007.202.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Andersson AK, et al. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat. Genet. 2015;47:330. doi: 10.1038/ng.3230.
    1. Frank SR, Schroeder M, Fernandez P, Taubert S, Amati B. Binding of c-Myc to chromatin mediates mitogen-induced acetylation of histone H4 and gene activation. Genes Dev. 2001;15:2069–2082. doi: 10.1101/gad.906601.
    1. Orlando DA, et al. Quantitative ChIP-Seq normalization reveals global modulation of the epigenome. Cell Rep. 2014;9:1163–1170. doi: 10.1016/j.celrep.2014.10.018.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Tischler G, Leonard S. biobambam: tools for read pair collation based algorithms on BAM files. Source Code Biol. Med. 2014;9:13. doi: 10.1186/1751-0473-9-13.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Landt SG, et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 2012;22:1813–1831. doi: 10.1101/gr.136184.111.
    1. Yang X, et al. Differentiation of human pluripotent stem cells into neurons or cortical organoids requires transcriptional co-regulation by UTX and 53BP1. Nat. Neurosci. 2019;22:362–373. doi: 10.1038/s41593-018-0328-5.
    1. Kharchenko PV, Tolstorukov MY, Park PJ. Design and analysis of ChIP-seq experiments for DNA-binding proteins. Nat. Biotechnol. 2008;26:1351–1359. doi: 10.1038/nbt.1508.
    1. Ramírez F, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016;44:W160–W165. doi: 10.1093/nar/gkw257.
    1. Guenther MG, et al. Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia. Genes Dev. 2008;22:3403–3408. doi: 10.1101/gad.1741408.
    1. Law CW, Chen Y, Shi W, Smyth GK. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15:R29. doi: 10.1186/gb-2014-15-2-r29.
    1. Bai B, et al. Deep profiling of proteome and phosphoproteome by isobaric labeling, extensive liquid chromatography, and mass spectrometry. Methods Enzymol. 2017;585:377–395. doi: 10.1016/bs.mie.2016.10.007.
    1. Wang X, et al. JUMP: a tag-based database search tool for peptide identification with high sensitivity and accuracy. Mol. Cell Proteom. 2014;13:3663–3673. doi: 10.1074/mcp.O114.039586.
    1. Gong J, et al. The C. elegans taste receptor homolog LITE-1 is a photoreceptor. Cell. 2017;168:325. doi: 10.1016/j.cell.2016.12.040.
    1. Tan H, et al. Integrative proteomics and phosphoproteomics profiling reveals dynamic signaling networks and bioenergetics pathways underlying T cell activation. Immunity. 2017;46:488–503. doi: 10.1016/j.immuni.2017.02.010.
    1. Nesvizhskii AI, Aebersold R. Interpretation of shotgun proteomic data: the protein inference problem. Mol. Cell Proteom. 2005;4:1419–1440. doi: 10.1074/mcp.R500012-MCP200.
    1. Warlick ED, Cao Q, Miller J. Bortezomib and vorinostat in refractory acute myelogenous leukemia and high-risk myelodysplastic syndromes: produces stable disease but at the cost of high toxicity. Leukemia. 2013;27:1789–1791. doi: 10.1038/leu.2013.61.
    1. Garcia-Manero G, et al. SWOG S1203: a randomized phase III study of standard cytarabine plus daunorubicin (7+3) therapy versus idarubicin with high dose cytarabine (IA) with or without vorinostat (IA+V) in younger patients with previously untreated acute myeloid leukemia (AML) Blood. 2016;128:901–901. doi: 10.1182/blood.V128.22.901.901.
    1. Waldschmidt JM, et al. Safety and efficacy of vorinostat, bortezomib, doxorubicin and dexamethasone in a phase I/II study for relapsed or refractory multiple myeloma (VERUMM study: vorinostat in elderly, relapsed and unfit multiple myeloma) Haematologica. 2018;103:e473–e479. doi: 10.3324/haematol.2018.189969.
    1. Keller A, et al. Vorinostat (V), bortezomib (B), doxorubicin (Dox) and dexamethasone (Dex, VBDD) in relapsed or refractory multiple myeloma patients (pts): results of an open, non-comparative, phase I/II investigator initiated trial (IIT) Clin. Lymphoma Myeloma Leuk. 2015;15:e276–e277. doi: 10.1016/j.clml.2015.07.572.
    1. Deutsch, E. W. et al. The ProteomeXchange consortium at 10 years: 2023 update. Nucleic Acids Res.51, D1539–D1548 (2023).
    1. Perez-Riverol Y, et al. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 2022;50:D543–D552. doi: 10.1093/nar/gkab1038.

Source: PubMed

3
Se inscrever