Although therapy escalation has led to improved 5-year overall survival rates for patients with B-cell acute lymphoblastic leukemia (B-ALL), few effective treatment options are available for relapsed and treatment-resistant disease. This applies particularly to specific subtypes of B-ALL, such as patients harboring TCF3 (formerly E2A) fusions. TCF3, encoding members of the E protein (class I) family of helix-loop-helix transcription factors, is a master regulator of B-cell development and is involved in several chromosomal translocations associated with lymphoid malignancies, such as the translocation t(1;19)(q23;p13.3), resulting in the TCF3::PBX1 fusion (5% of pediatric B-ALL) or the translocation t(17;19)(q22;p13) generating the TCF3::HLF fusion (~0.5% of pediatric B-ALL).2 Omics research for the discovery of novel treatment strategies in hematological cancer is still based largely on transcriptomics, although it is increasingly recognized that this does not translate well into the expression of proteins, which are the main targets of drugs and functional entities of biological processes. In this study, we comprehensively analyzed the proteomic landscapes of TCF3::HLF+ (N=6) and TCF3::PBX1+ (N=5) B-ALL employing primary patient-derived xenografts (PDX), liquid chromatography tandem mass spectrometry and data-dependent acquisition. Approval for the study reported here was granted by the Ethics Committee of the Medical Faculty of the Christian-Albrechts-University, Kiel, Germany (vote D508/13). We detected 6,863 proteins (6,123 without ≥2 missing values; Online Supplementary Table S1), which allowed a clear distinction between TCF3::HLF+ and TCF3::PBX1+ leukemia by unsupervised hierarchical clustering and principal component analysis (Figure 1A, B). Proteomic profiling proved a useful tool for prioritizing drug targets, as only 8.45% of the significantly differentially expressed genes (N=119/1,409; P<0.05 and minimal log2 fold change of ±1) previously detected by RNA sequencing2 showed differential expression on protein level confirmed by our proteomic analysis (Online Supplemenary Figure S1A). In contrast, 34.8% (N=119/342) of differentially regulated proteins detected by proteomics were also differentially expressed on RNA level. As a proof-of-concept, we examined overlap of differentially expressed genes (cutoffs: P<0.05 and minimal log2 fold change of ±1) from RNA sequencing and proteomic analysis obtained from a previously published dataset of ETV6::RUNX1+ (N=9) and high hyperdiploid (N=18) primary ALL patient samples.3 While only 3.63% (N=82/2,262) of differentially expressed genes detected via RNA sequencing showed differential expression on protein level, 92.13% (N=82/89) of differentially regulated proteins were also differentially expressed on RNA level (Online Supplementary Figure S1B).
In order to identify protein classes presenting specific therapeutic vulnerabilities, we performed gene set enrichment analysis (GSEA). We identified several gene sets enriched in either of the two subgroups (Figure 1C). RNA biology, mitochondrial translation and cellular respiration were the most prominent enriched gene sets for TCF3::HLF+ leukemia. In addition, strongly increased MYC expression and enrichment in MYC targets (Figure 1D, E) were detected, consistent with TCF3::HLF-driven activation of a MYC enhancer cluster previously shown using extensive functional genomics.4 For TCF3::PBX1+ leukemia, immune response/ cell cycle, actin cytoskeleton, cell morphogenesis and RTK signaling were among the most prominent enriched gene sets (Figure 1C). We validated therapeutic vulnerabilities indicated by GSEA using high-throughput drug screening. To this end, we tested the sensitivity of leukemic cell lines (TCF3::HLF+: HAL-01; TCF3::PBX1+: 697 and RCH-ACV) and mononuclear cells from peripheral blood of three healthy donors against a drug library of over 600 Food and Drug Administration-approved or clinical trial phase I-IV anti-cancer drugs. TCF3::HLF+ and TCF3::PBX1+ leukemic cells showed a differential response towards 109 drugs based on the area under the curve (AUC) as response parameter (Figure 2A; Online Supplementary Table S2; AUC<0.8 and >1.2 as a cutoff). Compared to our previous screening of bioactive compounds (N=98) employing the PDX samples,2 the cell lines showed similarly increased sensitivity towards compounds, such as BCL2 and mTOR inhibitors for TCF3::HLF+, and aurora kinase and polo-like kinase inhibitors for TCF3::PBX1+ B-ALL (Figure 2A). In addition, we identified novel potential drug targets. These included MDM2 and DNA/RNA synthesis for TCF3::HLF+ and micro-tubule/tubulin and cyclin-dependent kinases (CDK) for TCF3::PBX1+ leukemic cells (Figure 2A). In order to confirm these findings, we chose drugs from those groups, which did not affect normal peripheral blood cells (Figure 2B-E), and treated TCF3::PBX1+ (RCH-ACV) and TCF3::HLF+ (HAL-01) cells with half half-maximal inhibitory concentration (IC50), IC50 and double IC50 concentrations to investigate apoptosis induction. We demonstrated increased caspase 3/7 activity and apoptotic subG1 cells in TCF3::PBX1+ B-ALL in response to the microtubule/tubulin inhibitor ixabepilone and the CDK inhibitor SNS-032. For TCF3::HLF+ leukemic cells, we verified increased apoptotic cell death upon idasanutlin (MDM2 inhibitor) and bleomycin sulfate (DNA/RNA synthesis inhibitor) treatment (Figure 2F-I).
Besides the detection of vulnerabilities to specific drug classes, we aimed to identify novel targets for drug development. In our proteomic analyses, the B-lymphoid tyrosine kinase (BLK) was the most significantly upregulated protein for the TCF3::PBX1+ subtype (Figure 3A; Online Supplementary Figure S1C; Online Supplementary Table S1; minimal log2 fold change of ±1 and significance level of P<0.05 as cutoffs). BLK encodes a non-receptor tyrosine kinase of the src family of proto-oncogenes that plays an important role in precursor (pre) B-cell receptor (BCR) signaling and early B-cell development.5 RNA-sequencing and expression microarray data by us and others supported this finding (Figure 3B-D). We examined human gene expression data derived from four independent data sets of >3,000 leukemia cases6-9 available at the R2: genomics analysis and visualization platform (http://r2.amc.nl). These data indicated a subpopulation of leukemia samples that highly co-expresses BLK and PBX1 (Figure 3B; Online Supplementary Figure S1D-F). In two of the data sets, information on chromosomal translocations was available. There, the BLK and PBX1 co-expressing subpopulation was specifically associated with the TCF3::PBX1 fusion (Figure 3B; Online Supplementary Figure S1D).8,9 In the Microarray Innovations in LEukemia (MILE) study8 all 36 cytogenetically identified TCF3::PBX1+ cases were BLKhigh expressing (N=1,897 other leukemia or myelodysplastic syndrome [MDS], N=71 normal controls). Similarly, in another study9 all six TCF3::PBX1+ cases and none of the other samples (N=185 other B-ALL, N=3 normal controls) were both PBX1 and BLK high expressing. RNA-sequencing data of our cohort showed high RNA expression of BLK in all TCF3::PBX1+ leukemia cases (N=5 at diagnosis, N=8 after transplantation into NSG mice)2 compared to TCF3::HLF+ cases (N=5 at diagnosis, N=22 after transplantation) (Figure 3C, D).
Thus, we hypothesized that interference with BLK signaling might present a potential treatment strategy for TCF3::PBX1-rearranged B-ALL in particular. In order to test this, we treated TCF3::PBX1+ BLKhigh (RCH-ACV) and TCF3::HLF+ BLKlow (HAL-01) cells with a first selective irreversible BLK inhibitor BLK-IN-2.10 BLKhigh cells responded in a dose-dependent manner starting at nanomolar concentrations (IC50=0.2169 μM), whereas BLKlow cells showed little or no response (≥167-fold less, IC50=36.20 μM) (Figure 3E; Online Supplementary Figure S1G-I). We further tested the impact of BLK-IN-2 on other genetic subtypes of B-ALL and noticed preferential sensitivity of only the TCF3::PBX1+ subtype to BLK-IN-2 (Online Supplementary Figure S1I). In order to test if BLK inhibition synergizes with the specific vulnerabilities identified in our proteomic screen, we performed combined treatment with ixabepilone (microtubule/tubulin inhibitor). Indeed, both drugs synergized in TCF3::PBX1+ cell lines, but not in the TCF3::HLF+ cell line HAL-01 (Figure 3F-H). We further tested, if interference with pre-BCR signaling including BTK inhibitors would have the same impact. To this end, we tested the response of TCF3::PBX1+ B-ALL cell lines to ibrutinib and other BTK-targeting drugs (acalabrutinib, spebrutinib, LFM-A13). The response, however, was low and did not differ from cells lacking pre-BCR expression.
As previously reported by Geng et al., BLK is a signature gene of adult TCF3::PBX1+ B-ALL.9 Combining chromatin immoprecipitation sequencing, DNA methylation and expression profiling, the authors identified hypomethylation and overexpression of BLK in adult TCF3::PBX1+ B-ALL. In this study, upregulated genes targeted by TCF3::PBX1 included pre-BCR components and pre-BCR downstream signaling molecules.11 Ligand-independent autonomous tonic preBCR activation via self-aggregation is a main mechanism for pre-BCR activation and leads to constitutive activation of BLK11 (indicated by phosphorylation of the activating tyrosine Y388) observed in several TCF3::PBX1+ cell lines and primary B-ALL.11 Pre-BCR function induces activation of the transcription factor BCL6, which further increases pre-BCR signaling in a self-enforcing positive feedback loop and directly activates BLK transcription. More than 90% of TCF3::PBX1+ leukemia cases are pre-BCR+ and critically rely on pre-BCR-dependent signaling for proliferation.5 Thus, targeting BLK to abrogate pre-BCR downstream signaling presents an attractive approach for therapeutic intervention in TCF3::PBX1+ B-ALL.
Previously, interference with pre-BCR signaling has been suggested as a therapeutic option for TCF3-rearranged ALL12 and the inhibitors ibrutinib, dasatinib and idelalisib to be effective against TCF3::PBX1+ B-ALL.5,12,13 Inhibition of BTK, a downstream signaling kinase of the BCR, by ibrutinib is clinically beneficial in BCR+ B-cell malignancies such as non-Hodgkin lymphomas and multiple myeloma. However, in our analyses, the response of TCF3::PBX1+ B-ALL cell lines to BTK-targeting drugs was low and not corresponding to pre-BCR expression. This is in line with the observation that ibrutinib exerts a cytostatic rather than a cytotoxic effect on pre-BCR+ B-ALL cells5 and is further supported by the lack of in vivo effectivity against TCF3::PBX1+ primogafts.14 However, TCF3::PBX1+ PDX samples responded well to the tyrosine kinase inhibitor dasatinib.2 Still, high doses might be required for targets other than BCR::ABL1 and might be limited in the relapse setting due to toxicity.13
Taken together, proteomic-based profiling is a powerful tool to discover highly specific and sensitive cancer biomarkers and oncogenic pathway activation.15 Here, we uncovered proteomic alterations associated with TCF3::HLF+ or TCF3::PBX1+ B-ALL and revealed potential therapeutic options for these subtypes. These include previously known sensitivities for TCF3::HLF+ (e.g., BCL2 and mTOR) and TCF3::PBX1+ B-ALL (e.g., aurora kinase and polo-like kinase), as well as potential novel drug targets, such as MDM2 and DNA/RNA synthesis for TCF3::HLF+ or microtubule/tubulin and CDK for TCF3::P-BX1+ leukemic cells. Our data suggest that TCF3::PBX1+ B-ALL might be sensitive to treatment with selective BLK inhibitors, especially in combination with microtubule/tubulin targeting drugs, such as ixabepilone. Due to high BLK expression in TCF3::PBX1+ B-ALL cells, such inhibitors could selectively eradicate leukemic cells at doses eliciting less side-effects on normal tissue. A limitation of our study is that this was not tested in mouse models. In future studies, it would be interesting to apply BLK inhibition to suitable mouse models of TCF3::PBX1+ leukemia and to test synergism with other drugs. Larger numbers of patient samples need to be tested to show the applicability for TCF3::PBX1+ leukemia.
Footnotes
- Received July 17, 2023
- Accepted February 15, 2024
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
LB, JB, MS, J-PB, AB, MR, OA and UF planned and directed the study. Patient-derived xenograft models were provided by BB, BM and J-PB. FB, JTD, VM, FD, DL and OA conducted proteomic profiling and analyzed proteomic data. DP provided bioinformatic analyses. LB designed and performed the in vitro experiments, supported by VJ, JS-D and RH. NQ generated dose-response curves. SB provided intellectual contributions to the project and to the interpretation of the results. LB, VJ and UF wrote the manuscript. Figures were designed and drafted by LB, DP, NQ, VJ, JS-D and UF. All authors edited and contributed to the final manuscript.
Funding
Acknowledgments
Frauke-Dorothee Meyer, Sarah Göbbels and Bianca Killing are acknowledged for excellent technical assistance.
References
- Perez-Riverol Y, Bai J, Bandla C. The PRIDE database resources in 2022: a hub for mass spectrometry-based proteomics evidences. Nucleic Acids Res. 2022; 50(D1):D543-D552. Google Scholar
- Fischer U, Forster M, Rinaldi A. Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options. Nat Genet. 2015; 47(9):1020-1029. Google Scholar
- Yang M, Vesterlund M, Siavelis I. Proteogenomics and Hi-C reveal transcriptional dysregulation in high hyperdiploid childhood acute lymphoblastic leukemia. Nat Commun. 2019; 10(1):1519. Google Scholar
- Huang Y, Mouttet B, Warnatz HJ. The Leukemogenic TCF3-HLF complex rewires enhancers driving cellular identity and self-renewal conferring EP300 vulnerability. Cancer Cell. 2019; 36(6):630-644. Google Scholar
- Kim E, Hurtz C, Koehrer S. Ibrutinib inhibits pre-BCR(+) B-cell acute lymphoblastic leukemia progression by targeting BTK and BLK. Blood. 2017; 129(9):1155-1165. Google Scholar
- Polak R, Bierings MB, van der Leije CS. Autophagy inhibition as a potential future targeted therapy for ETV6-RUNX1-driven B-cell precursor acute lymphoblastic leukemia. Haematologica. 2019; 104(4):738-748. Google Scholar
- Loh ML, Zhang J, Harvey RC. Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children’s Oncology Group TARGET Project. Blood. 2013; 121(3):485-488. Google Scholar
- Kohlmann A, Kipps TJ, Rassenti LZ. An international standardization programme towards the application of gene expression profiling in routine leukaemia diagnostics: the Microarray Innovations in LEukemia study prephase. Br J Haematol. 2008; 142(5):802-807. Google Scholar
- Geng H, Brennan S, Milne TA. Integrative epigenomic analysis identifies biomarkers and therapeutic targets in adult B-acute lymphoblastic leukemia. Cancer Discov. 2012; 2(11):1004-1023. Google Scholar
- Fu T, Zuo Y, Zhong Z, Chen X, Pan Z. Discovery of selective irreversible inhibitors of B-Lymphoid tyrosine kinase (BLK). Eur J Med Chem. 2022; 229:114051. Google Scholar
- Geng H, Hurtz C, Lenz KB. Self-enforcing feedback activation between BCL6 and pre-B cell receptor signaling defines a distinct subtype of acute lymphoblastic leukemia. Cancer Cell. 2015; 27(3):409-425. Google Scholar
- van der Veer A, van der Velden VH, Willemse ME. Interference with pre-B-cell receptor signaling offers a therapeutic option for TCF3-rearranged childhood acute lymphoblastic leukemia. Blood Cancer J. 2014; 4(2):e181. Google Scholar
- Eldfors S, Kuusanmaki H, Kontro M. Idelalisib sensitivity and mechanisms of disease progression in relapsed TCF3-PBX1 acute lymphoblastic leukemia. Leukemia. 2017; 31(1):51-57. Google Scholar
- van de Ven C, Boeree A, Stalpers F, Zwaan CM, Den Boer ML. Ibrutinib is not an effective drug in primografts of TCF3-PBX1. Transl Oncol. 2020; 13(10):100817. Google Scholar
- Meissner F, Geddes-McAlister J, Mann M, Bantscheff M. The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov. 2022; 21(9):637-654. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.