BTK inhibition sensitizes acute lymphoblastic leukemia to asparaginase by suppressing the amino acid response pathway

Miriam Butler, Dorette S van Ingen Schenau, Jiangyan Yu, Silvia Jenni, Maria P Dobay, Rico Hagelaar, Britt M T Vervoort, Trisha M Tee, Fieke W Hoff, Jules P Meijerink, Steven M Kornblau, Beat Bornhauser, Jean-Pierre Bourquin, Roland P Kuiper, Laurens T van der Meer, Frank N van Leeuwen, Miriam Butler, Dorette S van Ingen Schenau, Jiangyan Yu, Silvia Jenni, Maria P Dobay, Rico Hagelaar, Britt M T Vervoort, Trisha M Tee, Fieke W Hoff, Jules P Meijerink, Steven M Kornblau, Beat Bornhauser, Jean-Pierre Bourquin, Roland P Kuiper, Laurens T van der Meer, Frank N van Leeuwen

Abstract

Asparaginase (ASNase) therapy has been a mainstay of acute lymphoblastic leukemia (ALL) protocols for decades and shows promise in the treatment of a variety of other cancers. To improve the efficacy of ASNase treatment, we used a CRISPR/Cas9-based screen to identify actionable signaling intermediates that improve the response to ASNase. Both genetic inactivation of Bruton's tyrosine kinase (BTK) and pharmacological inhibition by the BTK inhibitor ibrutinib strongly synergize with ASNase by inhibiting the amino acid response pathway, a mechanism involving c-Myc-mediated suppression of GCN2 activity. This synthetic lethal interaction was observed in 90% of patient-derived xenografts, regardless of the genomic subtype. Moreover, ibrutinib substantially improved ASNase treatment response in a murine PDX model. Hence, ibrutinib may be used to enhance the clinical efficacy of ASNase in ALL. This trial was registered at www.clinicaltrials.gov as # NCT02884453.

© 2021 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
CRISPR/Cas9-based kinome screen identify modifiers of ASNase sensitivity in BCP-ALL. (A) Schematic representation of our screening strategy. Nalm6 cells were transduced stepwise with a doxycycline-inducible Cas9 and a kinome sgRNA library. Cells were cultured for 2 weeks in the presence of 2 μg/mL of doxycycline to induce Cas9 expression and 1 week in the absence of doxycycline. Then, cells were treated for 2 weeks with 5 IU/mL of ASNase, DNA was isolated and subjected to massively parallel sequencing, and results were analyzed using the MAGeCK algorithm. (B) Gene list of expressed gRNA targets that significantly modulate ASNase response, ranked by P value calculated using the MAGeCK algorithm. (C) Counts of individual gRNAs targeting TRIB3, GCN2, and BTK, respectively, before and after ASNase treatment. (D,F) Immunoblot analysis of TRIB3 or GCN2 protein expression in cells upon CRISPR/Cas9-based targeting of GCN2 or TRIB3, respectively. (E) ASNase-induced cell death as determined by quantification of cells positive for amine-reactive dyes using flow cytometry in Nalm6 WT and Nalm6 GCN2-deleted cells after a 3-day treatment with 1 IU/mL of ASNase. Each bar represents a mean of 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test). (G) ASNase-induced cell death as determined by quantification of cells in subG1 phase using flow cytometry of Hoechst-stained cells. Bars represent mean ± standard error of the mean (SEM) of n = 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test). (H) Cell viability as measured by MTT in Nalm6 WT, and TRIB3del cells after treatment with the indicated dose of ASNase. Bars represent mean ± SEM of n = 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test).
Figure 2.
Figure 2.
Targeted KO of BTK sensitizes BCP-ALL cell lines to ASNase. (A) Immunoblot analysis of BTK protein expression in single cell clones upon CRISPR/Cas9-based targeting of BTK and apoptosis induction as measured by immunoblot analysis of PARP in Nalm6 WT and Nalm6 BTK-deleted cells after a 7-day treatment with 5 IU/mL of ASNase. Representative blot of 3 independent experiments. (B) Schematic overview of experimental procedure. Nalm6 WT and Nalm6 BTK-deleted cells were treated with 5 IU/mL of ASNase for 7 days, followed by direct evaluation of apoptosis or recovery capacity in a clonogenic assay after washout of the ASNase. (C) ASNase-induced cell death as determined by quantification of cells positive for amine-reactive dyes using flow cytometry in Nalm6 WT and Nalm6 BTK-deleted cells after a 7-day treatment with 5 IU/mL of ASNase. Each data point represents a mean of 1 clone of 3 independent experiments. Mean of all clones is indicated by line. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test). (D-E) Clonogenic proliferation assay of Nalm6 WT and Nalm6 BTK-deleted cells. After a 7-day treatment with 5 IU/mL of ASNase 5000 viable (trypan blue–negative) cells from each sample were seeded in soft agar. Recovery capacity was determined by quantification of colonies. Representative of 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test).
Figure 3.
Figure 3.
The BTK inhibitor ibrutinib potentiates ASNase-induced apoptosis. (A) Schematic overview of the experimental procedure. BCP-ALL cell lines were treated with ASNase, ibrutinib, or a combination of both for 7 days, followed by direct evaluation of apoptosis or recovery capacity in a clonogenic assay after washout of the ASNase. (B) Apoptosis induction as measured by immunoblot analysis of PARP in BCP-ALL cell lines. Nalm6, Sem, and Reh cells were treated with indicated doses of ASNase and ibrutinib. Representative of 3 independent experiments. (C-D) Clonogenic proliferation assay of Nalm6 and Sem cells. After 7 days’ treatment with indicated doses of ibrutinib and ASNase 5000 to 10 000 viable (trypan blue–negative) cells from each sample were seeded in soft agar. Recovery capacity was determined by quantification of colonies. Bars represent mean ± SEM of n = 3 replicates of 1 experiment. Representative of 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test).
Figure 4.
Figure 4.
Ibrutinib synergizes with ASNase treatment in a large panel of ALL PDX ex vivo and induces a delay in leukemia development in vivo. (A) Schematic overview representing the workflow used to determine ex vivo drug responses in PDX samples. ALL-PDX samples were seeded on hTERT-immortalized mesenchymal stem cells and treated with ASNase, ibrutinib, or combinations of both. After 3 and 7 days of incubation, cell death was analyzed by automated microscopy using a live cell staining using CyQuant (synergy matrix). (B) Overview of the calculated drug interactions between ASNase and ibrutinib in the PDX samples. (C) Dose-response curves and synergy matrix plots showing δ-scores of 3 representative ALL PDX samples treated with drug matrix of ASNase and ibrutinib (upper panel). (D) ASNase-induced cell death as determined by quantification of cells positive for amine-reactive dyes using flow cytometry in a primary refractory ALL patient sample. Cells were seeded on hTERT-immortalized mesenchymal stem cells and treated with indicated doses of ASNase in the presence or absence of 10 μM of ibrutinib. (E) Schematic overview of the experimental procedure. NSG mice were engrafted with 2 ALL-PDX 2 weeks before start of treatment with vehicle, 300 IU/kg of ASNase (days 1, 4, and 7), 25 mg/kg of ibrutinib (days 1 to 9), or a combination of both. Leukemia development was followed over time by weekly determination of the percentage of human CD10+, CD45+, and CD19+ cells in the blood. Postmortem, histological analysis of organs was executed. (F) Leukemia development as determined by percentage of human CD10 cells detected by flow cytometry in peripheral blood samples of mice treated with ibrutinib, ASNase, or a combination of both. Lines represent percentage of human CD10+ cells in 1 mouse. (G) Survival analysis of mice of different treatment groups. ***P < .001; **P < .01; ***P < .05 (log-rank (Mantel-Cox) test).
Figure 5.
Figure 5.
BTK signaling intersects with the amino acid stress response pathway. (A) Fold induction of fragments per kilobase million (FPKM) values of RNA expression of genes upregulated (upper panel) or downregulated (lower panel) in response to ASNase treatment while suppressed (upper panel) or induced (lower panel) when combined with ibrutinib. RNA expression in Nalm6 and Sem cells treated with ASNase, ibrutinib, or a combination of both was determined by RNA sequencing. (B) Upstream regulators of gene products identified in panel (A) determined by Ingenuity software. (C) Protein expression in Nalm6 and Sem cells treated with ASNase, ibrutinib, or a combination of both as determined by RPPA. Quantified protein expression levels from treated samples were normalized relative to untreated samples and subjected to principal component analysis. The top 50 proteins that contributed most to the difference between the treatments (PC1) were selected, and unsupervised hierarchical clustering with Ward’s linkage was applied. (D) Immunoblot analysis of protein expression. Nalm6 and Sem cells were treated for 72 hours (Sem) or 96 hours (Nalm6) with indicated doses of ASNase and ibrutinib. Representative of 3 independent experiments is shown. (E) Cell death determined by quantification of cells positive for amine-reactive dyes using flow cytometry. Nalm6 BTK KO control cells or cells made to express an ASNS transgene were treated with indicated doses of ASNase. Each bar represents a mean of 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test). (F) Apoptosis induction, measured by immunoblot analysis of PARP in Nalm6 BTK KO control cells or cells with ASNS overexpression. Cells were treated with indicated doses of ASNase. A representative of 3 independent experiments is shown.
Figure 6.
Figure 6.
Loss of BTK potentiates ASNase-induced apoptosis by repression of GCN2 activity mediated by c-Myc. (A) Upstream regulators of genes differentially expressed in Nalm6 and Sem treated with ASNase alone vs treatment with ASNase and ibrutinib, determined by Ingenuity software. (B) Immunoblot analysis of c-Myc protein expression. Nalm6 and Sem cells were treated for 72 hours (Sem) or 96 hours (Nalm6) with indicated doses of ASNase and ibrutinib. (C) Cell death determined by quantification of cells positive for amine-reactive dyes using flow cytometry. Nalm6 and Sem cells were treated with indicated doses of JQ1 and/or ASNase for 3 days (Sem) or 6 days (Nalm6). Each bar represents a mean of 3 independent experiments. ***P < .001; **P < .01; ***P < .05 (2-tailed, unpaired Student t test). (D) Immunoblot analysis of PARP, ATF4, ASNS, and actin. Nalm6 and Sem cells were treated for 72 hours with indicated doses of ASNase and JQ1. (E) Working model explaining the synergistic interaction of BTK inhibition and ASNase treatment. In response to ASNase treatment, cells upregulate the AAR pathway via GCN2 to adapt to nutrient stress. BTK inhibition renders cells incapable of activating c-Myc, thus preventing activation of the GCN2-ATF4 axis. As a consequence, cells cannot mount an appropriate amino acid stress response and eventually die.

References

    1. Pieters R, Hunger SP, Boos J, et al. . L-asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer. 2011;117(2):238-249.
    1. Covini D, Tardito S, Bussolati O, et al. . Expanding targets for a metabolic therapy of cancer: L-asparaginase. Recent Patents Anticancer Drug Discov. 2012;7(1):4-13.
    1. Fernandes HS, Silva Teixeira CS, Fernandes PA, Ramos MJ, Cerqueira NM. Amino acid deprivation using enzymes as a targeted therapy for cancer and viral infections. Expert Opin Ther Pat. 2017;27(3):283-297.
    1. Tong WH, Pieters R, Kaspers GJ, et al. . A prospective study on drug monitoring of PEG-asparaginase and Erwinia asparaginase and asparaginase antibodies in pediatric acute lymphoblastic leukemia. Blood. 2014;123(13):2026-2033.
    1. Henriksen LT, Nersting J, Raja RA, et al. ; Nordic Society of Paediatric Haematology and Oncology (NOPHO) group . Cerebrospinal fluid asparagine depletion during pegylated asparaginase therapy in children with acute lymphoblastic leukaemia. Br J Haematol. 2014;166(2):213-220.
    1. Rizzari C, Lanvers-Kaminsky C, Valsecchi MG, et al. . Asparagine levels in the cerebrospinal fluid of children with acute lymphoblastic leukemia treated with pegylated-asparaginase in the induction phase of the AIEOP-BFM ALL 2009 study. Haematologica. 2019;104(9):1812-1821.
    1. Iwamoto S, Mihara K, Downing JR, Pui CH, Campana D. Mesenchymal cells regulate the response of acute lymphoblastic leukemia cells to asparaginase. J Clin Invest. 2007; 117(4):1049-1057.
    1. Wang T, Wei JJ, Sabatini DM, Lander ES. Genetic screens in human cells using the CRISPR-Cas9 system. Science. 2014; 343(6166):80-84.
    1. Li W, Xu H, Xiao T, et al. . MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 2014;15(12):554.
    1. Schmitz M, Breithaupt P, Scheidegger N, et al. . Xenografts of highly resistant leukemia recapitulate the clonal composition of the leukemogenic compartment. Blood. 2011;118(7):1854-1864.
    1. Frismantas V, Dobay MP, Rinaldi A, et al. . Ex vivo drug response profiling detects recurrent sensitivity patterns in drug-resistant acute lymphoblastic leukemia. Blood. 2017;129(11):e26-e37.
    1. Ianevski A, He L, Aittokallio T, Tang J. SynergyFinder: a web application for analyzing drug combination dose-response matrix data. Bioinformatics. 2017;33(15):2413-2415.
    1. Li J, Zhao W, Akbani R, et al. . Characterization of human cancer cell lines by reverse-phase protein arrays. Cancer Cell. 2017;31(2):225-239.
    1. Tibes R, Qiu Y, Lu Y, et al. . Reverse phase protein array: validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol Cancer Ther. 2006;5(10):2512-2521.
    1. Lin G, Chai J, Yuan S, et al. . VennPainter: a tool for the comparison and identification of candidate genes based on Venn diagrams. PLoS One. 2016;11(4):e0154315.
    1. Szklarczyk D, Gable AL, Lyon D, et al. . STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607-D613.
    1. Krämer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics. 2014;30(4):523-530.
    1. Wortel IMN, van der Meer LT, Kilberg MS, van Leeuwen FN. Surviving stress: modulation of ATF4-mediated stress responses in normal and malignant cells. Trends Endocrinol Metab. 2017;28(11):794-806.
    1. Donnelly N, Gorman AM, Gupta S, Samali A. The eIF2α kinases: their structures and functions. Cell Mol Life Sci. 2013;70(19):3493-3511.
    1. Leprivier G, Remke M, Rotblat B, et al. . The eEF2 kinase confers resistance to nutrient deprivation by blocking translation elongation. Cell. 2013;153(5):1064-1079.
    1. Wang X, Li W, Williams M, Terada N, Alessi DR, Proud CG. Regulation of elongation factor 2 kinase by p90(RSK1) and p70 S6 kinase. EMBO J. 2001;20(16):4370-4379.
    1. Ohoka N, Yoshii S, Hattori T, Onozaki K, Hayashi H. TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. EMBO J. 2005;24(6):1243-1255.
    1. Lien EC, Dibble CC, Toker A. PI3K signaling in cancer: beyond AKT. Curr Opin Cell Biol. 2017;45:62-71.
    1. Campbell R, Chong G, Hawkes EA. Novel indications for Bruton’s tyrosine kinase inhibitors, beyond hematological malignancies. J Clin Med. 2018;7(4):E62.
    1. Kim HO. Development of BTK inhibitors for the treatment of B-cell malignancies. Arch Pharm Res. 2019;42(2):171-181.
    1. Nguyen HA, Su Y, Zhang JY, et al. . A novel l-asparaginase with low l-glutaminase coactivity is highly efficacious against both T- and B-cell acute lymphoblastic leukemias in vivo. Cancer Res. 2018;78(6):1549-1560.
    1. Tameire F, Verginadis II, Leli NM, et al. . ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumour progression [published correction appears in Nat Cell Biol. 2019;21:1052]. Nat Cell Biol. 2019;21(7):889-899.
    1. Yeomans A, Thirdborough SM, Valle-Argos B, et al. . Engagement of the B-cell receptor of chronic lymphocytic leukemia cells drives global and MYC-specific mRNA translation. Blood. 2016;127(4):449-457.
    1. Pede V, Rombout A, Vermeire J, et al. . CLL cells respond to B-cell receptor stimulation with a microRNA/mRNA signature associated with MYC activation and cell cycle progression [published correction appears in PLoS One. 2014;9(1):10.1371]. PLoS One. 2013; 8(4):e60275.
    1. Moyo TK, Wilson CS, Moore DJ, Eischen CM. Myc enhances B-cell receptor signaling in precancerous B cells and confers resistance to BTK inhibition. Oncogene. 2017;36(32):4653-4661.
    1. Lee J, Zhang LL, Wu W, et al. . Activation of MYC, a bona fide client of HSP90, contributes to intrinsic ibrutinib resistance in mantle cell lymphoma. Blood Adv. 2018;2(16):2039-2051.
    1. Chong IY, Aronson L, Bryant H, et al. . Mapping genetic vulnerabilities reveals BTK as a novel therapeutic target in oesophageal cancer. Gut. 2018;67(10):1780-1792.
    1. Davidson M, Chong IY-S, Cunningham D, et al. . iMYC: proof-of-concept study of ibrutinib in c-MYC and HER2 amplified oesophagogastric carcinoma. J Clin Oncol. 2017;35(4 suppl):TPS221.
    1. Delmore JE, Issa GC, Lemieux ME, et al. . BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146(6):904-917.
    1. Nakamura A, Nambu T, Ebara S, et al. . Inhibition of GCN2 sensitizes ASNS-low cancer cells to asparaginase by disrupting the amino acid response. Proc Natl Acad Sci USA. 2018;115(33):E7776-E7785.
    1. Wilson GJ, Bunpo P, Cundiff JK, Wek RC, Anthony TG. The eukaryotic initiation factor 2 kinase GCN2 protects against hepatotoxicity during asparaginase treatment. Am J Physiol Endocrinol Metab. 2013;305(9):E1124-E1133.
    1. Phillipson-Weiner L, Mirek ET, Wang Y, McAuliffe WG, Wek RC, Anthony TG. General control nonderepressible 2 deletion predisposes to asparaginase-associated pancreatitis in mice. Am J Physiol Gastrointest Liver Physiol. 2016;310(11):G1061-G1070.
    1. Kim E, Hurtz C, Koehrer S, et al. . Ibrutinib inhibits pre-BCR+ B-cell acute lymphoblastic leukemia progression by targeting BTK and BLK. Blood. 2017;129(9):1155-1165.
    1. Weber ANR, Bittner Z, Liu X, Dang TM, Radsak MP, Brunner C. Bruton’s tyrosine kinase: an emerging key player in innate immunity. Front Immunol. 2017;8:1454.
    1. Woo MH, Hak LJ, Storm MC, et al. . Cerebrospinal fluid asparagine concentrations after Escherichia coli asparaginase in children with acute lymphoblastic leukemia. J Clin Oncol. 1999;17(5):1568-1573.
    1. Goldwirt L, Beccaria K, Ple A, Sauvageon H, Mourah S. Ibrutinib brain distribution: a preclinical study. Cancer Chemother Pharmacol. 2018;81(4):783-789.
    1. Cabannes-Hamy A, Lemal R, Goldwirt L, et al. . Efficacy of ibrutinib in the treatment of Bing-Neel syndrome. Am J Hematol. 2016;91(3):E17-E19.
    1. Bernard S, Goldwirt L, Amorim S, et al. . Activity of ibrutinib in mantle cell lymphoma patients with central nervous system relapse. Blood. 2015;126(14):1695-1698.
    1. Herman SE, Mustafa RZ, Jones J, Wong DH, Farooqui M, Wiestner A. Treatment with ibrutinib inhibits BTK- and VLA-4-dependent adhesion of chronic lymphocytic leukemia cells in vivo. Clin Cancer Res. 2015;21(20):4642-4651.
    1. Regan JA, Cao Y, Dispenza MC, et al. . Ibrutinib, a Bruton’s tyrosine kinase inhibitor used for treatment of lymphoproliferative disorders, eliminates both aeroallergen skin test and basophil activation test reactivity. J Allergy Clin Immunol. 2017;140(3):875-879.e1.
    1. Eifert C, Wang X, Kokabee L, et al. . A novel isoform of the B cell tyrosine kinase BTK protects breast cancer cells from apoptosis. Genes Chromosomes Cancer. 2013;52(10):961-975.
    1. Li T, Deng Y, Shi Y, et al. . Bruton’s tyrosine kinase potentiates ALK signaling and serves as a potential therapeutic target of neuroblastoma. Oncogene. 2018;37(47):6180-6194.

Source: PubMed

3
Subscribe