Abstract
BCL-2 family proteins are frequently aberrantly expressed in mantle cell lymphoma (MCL). Recently, the BCL-2-specific inhibitor venetoclax has been approved by the US Food and Drug Administration for chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). In MCL, venetoclax has shown promising efficacy in early clinical trials; however, a significant subset of patients is resistant. By conducting a kinome-centered CRISPR-Cas9 knockout sensitizer screen, we identified casein kinase 2 (CK2) as a major regulator of venetoclax resistance in MCL. Interestingly, CK2 is over-expressed in MCL and high CK2 expression is associated with poor patient survival. Targeting of CK2, either by inducible short hairpin RNA (shRNA)-mediated knockdown of CK2 or by the CK2-inhibitor silmitasertib, did not affect cell viability by itself, but strongly synergized with venetoclax in both MCL cell lines and primary samples, also if combined with ibrutinib. Furthermore, targeting of CK2 reduced MCL-1 levels, which involved impaired MCL-1 translation by inhibition of eIF4F complex assembly, without affecting BCL-2 and BCL-XL expression. Combined, this results in enhanced BCL-2 dependence and, consequently, venetoclax sensitization. In cocultures, targeting of CK2 overcame stroma-mediated venetoclax resistance of MCL cells. Taken together, our findings indicate that targeting of CK2 sensitizes MCL cells to venetoclax through downregulation of MCL-1. These novel insights provide a strong rationale for combining venetoclax with CK2 inhibition as therapeutic strategy for MCL patients.
Introduction
Mantle cell lymphoma (MCL) is an aggressive incurable B-cell non-Hodgkin lymphoma (NHL), representing 6-8% of all NHL. Currently, first-line therapy for MCL consists of cytarabine-based chemo-immunotherapy regimens followed by autologous stem cell transplantation in eligible patients. Elderly and frail patients are usually treated with adriamycin and cyclophosphamide-based regimens.1 However, virtually all patients relapse and these patients receive other/non-cross-resistant chemo-immunotherapy regimens or targeted therapies such as the Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib. In addition, anti-CD19 chimeric antigen receptor (CAR) T-cell therapy is now approved for relapsed and refractory (R/R) disease, showing complete responses in approximately 65% of patients.1 When these therapies fail, treatment options are limited, and therefore there is a high need for novel treatment strategies. MCL is characterized by the chromosomal translocation t(11;14), resulting in the overexpression of cyclin D1, which facilitates cellular transformation by deregulating the cell cycle.2 For malignant transformation, additional aberrations are required, including alterations in the mitochondrial apoptotic pathway, such as upregulation of BCL-2, BCL-XL and Bcl-W and deletion of BIM.3 Hence, targeting anti-apoptotic BCL-2 family proteins is an attractive option for the treatment of MCL.
Recently, the BCL-2 inhibitor venetoclax was approved by the US Food and Drug Administration for chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML).4 In MCL, venetoclax has also shown promising efficacy in early clinical trials, with complete response (CR) rates of 18-21% and overall response rates (ORR) of 53-75%, depending on the number and the type of prior treatments.5-7 Combining venetoclax with ibrutinib therapy improved efficacy compared with venetoclax monotherapy, with a CR of 62-71%.8,9 However, a significant subset of MCL patients is primary refractory or develops resistance during the course of the disease. Therefore, novel combinations need to be identified to enhance the efficacy of venetoclax in MCL.
Here, we conducted a functional genomic CRISPR-Cas9 knockout sensitizer screen, with a lentiviral guide RNA (gRNA) library that represents all human kinases,10 and identified casein kinase 2 (CK2) as a major determinant of venetoclax sensitivity in MCL cells. We showed that reduced CK2 expression/activity by RNA interference or by treatment with the CK2 inhibitor silmitasertib synergistically increased the effectiveness of venetoclax due to the reduction in MCL-1 protein levels. These findings indicate that the combination of silmitasertib and venetoclax could represent a promising treatment strategy for MCL.
Methods
For detailed information please see the Online Supplementary Appendix.
Cell culture and treatment
The human MCL cell lines Granta-519, Jeko-1, Maver-1, Mino, and Z138 were obtained from DSMZ (Braunschweig, Germany) and cultured in Iscove’s modified Dulbecco’s medium (IMDM; Invitrogen Life Technologies, Carlsbad, CA, USA). The stromal cell line HS-27a and the kidney cell line HEK-293T/17 were obtained from ATCC (Manassas, VA, USA), and cultured in Dulbecco’s Modified Eagle’s Medium (DMEM; Invitrogen).
For co-culture assays, bone-marrow stromal cells were seeded in 96-well plates 4 hours (h) prior to the addition of MCL cells, to allow cell attachment. MCL cells were pre-incubated for 2 h with stromal cells, followed by a 3-day co-culture in the presence of the indicated drug concentrations. Cell viability and specific cell death were measured by flow cytometry.
The following small-molecule inhibitors were used: venetoclax/ABT-199, Q-VD-OPh, IPTG (MedChemExpress, Princeton, NJ, USA), silmitasertib/CX-4945, S63845, MK2206 (Selleckchem, Houston, TX, USA), cycloheximide, puromycin (Sigma-Aldrich, St. Louis, MO, USA), and blasticidin (Thermo Fisher Scientific, Waltham, MA, USA).
Patient samples
Patient samples and peripheral blood naïve B cells were obtained as reported previously.11 This study was approved by the AMC Medical Committee on Human Experimentation. Informed consent was obtained in accordance with the revised Declaration of Helsinki 2008.
Cloning, transfection, and transduction
shRNA targeting CSNK2A1 (#1: ATTACCTGCAGGTGGAATAT T, #2: TGGACAAACTGCTGCGATAT G ) and a control construct (CCTAAGGTTAAGTCGCCCTCG) were inserted into the
Acc65I(Asp718I)/EcoRI site of the pLKO-mCherry-IPTG3xLacO vector (a gift from Dr. Noam Zelcer and Dr. Jessica Nelson, Department of Medical Biochemistry, Amsterdam UMC, The Netherlands; Sigma #shc334). Lentiviral particles were produced as described previously.12 Three days after transduction, cells were FACS-sorted using a Sony SH800S cell sorter (Sony Biotechnology, San Jose, CA, USA). Cells transduced with Lenti-Cas9-Blast (Addgene #52962) were selected for 7 days with blasticidin, 24 h after transduction. Cas9 activity was analyzed by flow cytometry of Lenti-Cas9-reporter-transduced cells (Addgene #67980) using the FACSCanto II flow cytometry system (BD Bio-sciences, San Jose, CA, USA).
Synthetic lethality screen
The screening procedure was performed as outlined in Figure 1C. (See Online Supplementary Methods for full details.)
Real-time-quantitative polymerase chain reaction
Real-time-quantitative polymerase chain reaction (RT-qPCR) was essentially performed as described previously.11
Flow cytometry
Cell viability and (specific) apoptosis were analyzed by flow cytometry using 7-aminoactinomycin-D (7-AAD; Invitrogen) staining. Early apoptosis was determined using Annexin V staining and cell cycle distribution was analyzed using bromodeoxyuridine (BrdU) incorporation performed as previously described.11
Immunobloting
Immunoblotting was essentially performed as described previously.11,12
Gene expression profile analysis and gene set enrichment analysis
The gene expression profiling data of B cells from 165 MCL patients (GSE93291, GSE132929)13,14 and 10 healthy donors (GSE28491, E-MTAB-1771), publicly available and deposited in the NIH Gene Expression Omnibus and the EMBL European Bioinformatics Institute databases, respectively, were analyzed using the R2 Genomics Analysis and Visualization Platform (http://r2.amc.nl). The GSE93291 dataset was analyzed for gene set enrichment using the gene set enrichment analysis (GSEA) application (version 4.0.3).15
Statistical analysis
Statistical analyses were performed using log-rank tests for Kaplan-Meier analysis, 2-tailed Student t tests for comparisons between 2 groups, and two-way analysis of variance (ANOVA) with Tukey, Šidák or Dunnett’s multiple comparison post-hoc tests as indicated in the figure legends for comparisons between more than 2 groups.
Results
A kinome-centered CRISPR screen identifies CK2 as potential target for venetoclax sensitization of MCL cells
To assess their sensitivity to venetoclax, five MCL cell lines (Online Supplementary Table S1) were treated with increasing concentrations of venetoclax, and their viability was analyzed after 3 days. In line with previous reports,16-18 two cell lines, Jeko-1 and Z138, were relatively resistant to venetoclax, with IC50 values approximately 100 times higher compared to the IC50 of the other three cell lines, Granta-519, Maver-1 and Mino (Figure 1A). These IC50 largely correspond to the protein expression of BCL-2 (Figure 1B): the venetoclax-resistant cell lines Jeko-1 and Z138 express relatively low BCL-2 protein levels. Next, we performed an unbiased functional genomic CRISPR knockout screen with the Brunello kinome-centered library to identify kinases that, given their druggability, may serve as targets to sensitize MCL cells to venetoclax.10 For this CRISPR screen, the Z138 cell line was selected, being a venetoclax-insensitive cell line with high Cas9 activity after transduction with a Cas9 construct (Online Supplementary Figure S1A). The abundance of the different gRNA was assessed before initiating drug treatment (T0) and after passaging the cells for 10 population doublings in the presence of either DMSO (T1 DMSO) or the IC30 of venetoclax (500 nM, T1 Ven). Then, enrichment or depletion was determined using the DESeq2 and MAGeCK pipelines (Figure 1C).12 The performed screen was of high quality, given the strong correlation between the three independent replicates (Online Supplementary Figure S1B) and the selective dropout of the gRNA against reference essential genes compared to non-essential genes in the control cells (T1 DMSO) versus T0 samples (Online Supplementary Figure S1C).12
Comparison of the two treated experimental arms (T1 Ven vs. T1 DMSO) identified genes that preferentially affected viability and cell growth in venetoclax-treated cells over DMSO-treated cells (Figure 1D, E). The highest ranked gene was CSNK2A1, with 7 out of 8 guides significantly depleted and with a median fold change of 0.58. In addition, the gRNA targeting CSNK2A2 and CSNK2B were also significantly depleted (Figure 1D, E). These genes encode the catalytic a- and a’-subunits, and the regulatory β-subunits of CK2, respectively. CK2 is a pleiotropic serine/threonine protein kinase comprising two β-subunits and two of the a-subunits, resulting in either a2β2, aa’β2, or a’2β2 configurations.19,20 It is involved in a broad array of cellular processes, including cell cycle progression, survival and differentiation, and is a regulator and/or mediator of several oncogenic pathways, such as Wnt-, PI3K/Akt- and NF-κB signaling.19,20
CK2 is aberrantly expressed in MCL and high expression of CK2 is associated with inferior prognosis for MCL patients
CK2 overexpression has been reported in several hematologic malignancies, including MCL, CLL, diffuse large B-cell lymphoma (DLBCL), and multiple myeloma (MM).20-23 In line with this, comparison of primary MCL versus naïve B cells, either from publically available microarray datasets or RT-qPCR, revealed a significant upregulation of CSNK2A1 and CSNK2B RNA expression in MCL cells (Figure 2A, B; Online Supplementary Figure S2A). Moreover, CK2 expression measured in excisional lymph node biopsy material (tumor content ≥60%) of 122 MCL patients that were all treated with rituximab plus cyclophosphamide, dox-orubicin, vincristine, and prednisone (R-CHOP) (GSE93291) revealed a 5-year survival of 0% versus 72% in the patients with the highest CSNK2B expression and lowest CSNK2B expression, respectively (Figure 2C).13 CSNK2A1 expression in the same cohort showed a similar trend, although this was not significant (Online Supplementary Figure S2B). Taken together, our results indicate that CK2 is aberrantly expressed and associated with an aggressive disease course.
Targeting of CK2 sensitizes MCL cells to venetoclax
To evaluate CSNK2A1 as a hit from the CRISPR screen, the venetoclax-insensitive cell lines Z138 and Jeko-1, which both express high levels of CK2a (Online Supplementary Figure S3A, B), were transduced with different IPTG-inducible shRNA targeting CSNK2A1 (Figure 3A). These shRNA induced efficient knockdown of CK2a protein levels in both cell lines, without affecting the highly homologous CK2a’ protein encoded by the CSNK2A2 gene. Viability was only modestly affected (Figure 3B), supporting our screening results, in which CSNK2A1 KO did not affect cell growth in the absence of venetoclax (Online Supplementary Figure S2C). However, both Z138 and Jeko-1 cells became significantly sensitized to venetoclax upon CSNK2A1 silencing, most prominently in Jeko-1, resulting in a 60% increased cell death compared to control cells (Figure 3C).
We next investigated the effects of silmitasertib (CX-4945), a specific and selective CK2 inhibitor with proven safety and tolerability profiles in several phase I and II clinical trials.24-27 Upon silmitasertib treatment, phosphorylation of Akt S129, which is exclusively phosphorylated by CK2, was effectively reduced in the MCL cell lines (Figure 4A).28,29 However, cell viability was only decreased at concentrations above 3 mM, with IC50 values as high as 8-10 mM (Figure 4B). Silmitasertib also decreased cell viability of primary samples only at concentrations in the micromolar range. The silmitasertib-sensitivity of the primary MCL samples roughly corresponded to CSNK2 RNA expression (Online Supplementary Figure S4A).
To assess potential synergy between venetoclax and silmitasertib in MCL, cells were treated with increasing doses of either venetoclax, silmitasertib, or a combination of both, and cell survival was determined (Figure 4C). Synergy was assessed using the Bliss independence model: silmitasertib and venetoclax were highly synergistic in all MCL cell lines tested, with DBliss scores up to 55 in Jeko-1 (Figure 4C), indicating 55% more cell death in the cells treated with the combination of drugs than expected based upon the effects of single treatment of the drug.30 In agreement, treatment of 6 primary MCL samples with the combination of venetoclax and silmitasertib also resulted in more cell death than predicted, with ΔBliss scores ranging from 6 to 18 (Figure 4D, E). As the IC50 of silmitasertib was only reached in the high micromolar range, indicating off-target effects, assessment of synergy using the Chou Talalay method was not considered appropriate.31 The zero interaction potency (ZIP) model32 revealed synergy scores similar to those of the Bliss independence model (Online Supplementary Figure S4B). Since clinical trials frequently evaluate venetoclax in combination with ibrutinib,8,9,33 we also determined whether targeting CK2 can sensitize MCL cells to venetoclax in the presence of ibrutinib (Online Supplementary Figure S4C, D). Indeed, both CK2 knockdown (KD) and inhibition also potentiated the activity of venetoclax combined with ibrutinib. Interestingly, in Jeko-1, this potentiation by silmitasertib was even stronger for the combination compared to venetoclax alone. Please note that ibrutinib treatment neither affected cell viability by itself nor sensitized the cell lines to venetoclax. This is in line with the observation that, also in patients, ibrutinib does not directly target cell viability, but rather mobilizes the malignant cells from the protective lymphoid organs to the peripheral blood.34,35
To assess whether the venetoclax-sensitizing effect of CK2 silencing or inhibition is a cytostatic or cytotoxic effect, apoptosis and cell cycle were analyzed using Annexin V staining and BrdU labeling, respectively. Treatment with venetoclax significantly increased the percentage of Annexin V-positive cells in both CSNK2A1 KD cells and silmitasertib-treated cells, whereas the cell cycle was only modestly affected (Online Supplementary Figure S5A, B).
CK2 inhibition down-regulates MCL-1 levels
To investigate the mechanism(s) through which loss of CK2 activity sensitizes MCL cells to venetoclax, we evaluated changes in protein levels of BCL-2, MCL-1, and BCL-XL. The ratio of expression of BCL-2 over MCL-1 and BCL-XL combined has been described as an important determinant of venetoclax sensitivity in MCL,3,16,17 and also corresponds with venetoclax sensitivity in the cell lines used in this study (Figure 1B). Both CK2 silencing and inhibition resulted in markedly reduced MCL-1 levels in MCL cells, wheras BCL-2 and BCL-XL protein levels were unaffected (Figure 5A, B; Online Supplementary Figure S6A). Also in primary MCL samples, silmitasertib treatment decreased MCL-1 protein levels without altering BCL-XL levels (Figure 5C).
To assess whether the reduction in MCL-1 levels was sufficient to sensitize MCL cells to venetoclax, we applied a BH3-mimetic specific to MCL-1, S63845, in combination with venetoclax to the MCL cells, and cell survival was assessed. Indeed, upon MCL-1 inhibition, MCL cells became significantly sensitized to venetoclax, as observed by the shift in the response curve (Figure 5D; Online Supplementary Figure S6B). These results confirm that MCL-1 is a major regulator of venetoclax resistance and that CK2-mediated MCL-1 regulation affects venetoclax sensitivity.
CK2 regulates MCL-1 translation
Next, we investigated the mechanism of CK2-mediated MCL-1 protein expression. MCL-1 expression is tightly regulated at multiple levels, involving transcriptional, trans lational, and post-translational processes.36 First, we monitored MCL1 mRNA levels by RT-qPCR, and in none of the MCL cell lines were alterations in the expression levels of MCL1 observed upon CK2 targeting, arguing against transcriptional regulation (Figure 6A, B). Next, we assessed whether CK2 inhibition decreased protein stability of MCL-1, by blocking translation with cycloheximide (CHX) treatment. The MCL-1 protein half-life was unaffected by CK2 inhibition in both Z138 and Jeko-1 (Figure 6C), suggesting that CK2 regulates MCL-1 protein levels by controlling translation. In agreement, GSEA of an MCL patient cohort of 122 patients (GSE93291) revealed that the expression of genes involved in protein translation was strongly enriched in patients with high versus low CSNK2A1 expression (Online Supplementary Figure S7A). Taken together, these data indicate that CK2 is not involved in MCL1 expression or MCL-1 protein stability and proteasomal degradation, but rather affects MCL-1 translation.
The rate of MCL-1 translation is, amongst others, dependent upon the eukaryotic translation initiation factor 4F (eIF4F) complex.37-41 Association of this complex with mRNA depends on the cap-binding factor eIF4E, which is inhibited by eIF4E-binding protein (4EBP). The mTOR-dependent phosphorylation of 4EBP releases eIF4E to increase translation initiation, and p38- and extracellular signal-regulated kinase (ERK)-dependent phosphorylation of eIF4E further enhances formation of the eIF4F complex.39 Furthermore, the phosphorylation of ribosomal protein S6 has been implicated in translation initiation. In line with this, both CK2 silencing and inhibition clearly reduced activation of the Akt/mTOR-, p38- and/or the ERK-pathways and the subsequent phosphorylation/activation of S6 and the eIF4 complex (Figure 6D; Online Supplementary Figure S7B), indicating their possible role in CK2-controlled MCL-1 translation. The importance of the Akt pathway is further supported by the synergy observed between the Akt inhibitor MK2206 and venetoclax in Jeko-1 and Mino (Online Supplementary Figure S7C). However, Akt inhibition did not result in clear synergy with venetoclax in any of the tested cell lines, indicating that, for example, the p38 and ERK pathways are also involved in the synergy observed between CK2 inhibition and venetoclax. In summary, these data indicate that CK2 sensitizes MCL cells to venetoclax by downregulation of MCL-1 levels via interference with the assembly of the eIF4F complex and thus the translation of MCL-1 (Figure 6E).
CK2 inhibition overcomes microenvironmental venetoclax resistance
Microenvironment-mediated drug resistance is commonly observed in MCL, also for venetoclax.3,16 Direct contact with other cells in the lymph node and bone marrow (cell adhesion-mediated drug resistance), as well as paracrine soluble factors such as cytokines and chemokines result in reduced drug efficacy. To study microenvironment-mediated venetoclax resistance, we co-cultured MCL cell lines with the bone marrow stromal cell line HS-27a in the presence of venetoclax. Indeed, the stromal cells protected the MCL cells from venetoclax-induced cytotoxicity (Figure 7A). Importantly, when venetoclax was combined with CK2 silencing or inhibition, this protective effect of the stromal cells was largely abolished, without compromising the viability of the stromal cells (Figure 7B, C; Online Supplementary Figure S8). This further enhances the therapeutic potential of this combination. To investigate the mechanism underlying the stromal cell-mediated venetoclax resistance and the ability of CK2 inhibition to overcome this, we determined the BCL-2 family protein levels in MCL cells co-cultured with stromal cells (Figure 7D). Interestingly, co-culture with stromal cells up-regulated MCL-1 levels in MCL cells without affecting BCL-2 and BCL-XL levels, and this MCL-1 upregulation was counteracted by CK2 inhibition. Taken together, our data indicate that repression of MCL-1 translation by CK2 inhibition prevents stroma-mediated upregulation of MCL-1, thereby sensitizing MCL cells to venetoclax.
Discussion
Whereas venetoclax shows promising anti-tumor activity in MCL patients, a substantial proportion of patients resists the treatment. Our kinome-centered CRISPR-Cas9 venetoclax-sensitizer screen identified the genes encoding the subunits of CK2 as prominent determinants of venetoclax sensitivity in MCL. We observed a strong synergistic effect between venetoclax and CK2 knockdown or inhibition in both venetoclax-sensitive and venetoclax-insensitive MCL cell lines, also in the presence of ibrutinib, and this synergistic effect can be attributed to CK2-mediated regulation of MCL-1 translation. Importantly, combining CK2 inhibition with venetoclax could also overcome microenvironmental protection to venetoclax, providing a clear rationale for combining venetoclax with a CK2 inhibitor in MCL patients.
CK2 is a ubiquitously expressed and constitutively active kinase that is involved in a broad array of cellular processes, including cell cycle progression, survival and differentiation.19,20 CK2 is an attractive kinase to target in cancer, as cancer cells have been shown to be more sensitive to CK2 inhibition than their non-malignant counterparts.20 CK2 is an attractive target also in MCL, as we and others have shown that the CK2 subunits are frequently over-expressed in MCL cells compared to naïve B cells from healthy donors (Figure 2A, B; Online Supplementary Figure S2A).21,22 Furthermore, we show that CSNK2B expression is strongly associated with poor patient survival (Figure 2C), suggesting a possible role for CK2 in the aggressive disease course of MCL.
Using CK2 knockdown, and supported by the use of a specific CK2 inhibitor, our study demonstrates that CK2 is an important regulator of BCL-2 family proteins and thereby venetoclax sensitivity in MCL. This finding may have broader implications, for example, for treatment of other B-cell malignancies. In line with this, Lazaro-Navarro et al.42 recently reported synergy between venetoclax and silmitasertib in B-cell precursor acute lymphoblastic leukemia (BCP-ALL) cells. Furthermore, based on the use of rather non-specific CK2 inhibitors such as TBB, apigenin and quercetin, several prior studies have suggested a possible role for CK2 in the expression of BCL-2 family proteins in various other cell types.23,43-48 For example,
Russo et al.47 have shown that quercetin synergizes with the dual BCL-2 and BCL-XL inhibitor ABT-737 in primary CLL cells, involving quercetin-mediated downregulation of MCL-1. However, quercetin, apart from being an antioxidant, is a broad spectrum kinase inhibitor that targets, amongst others, PI3K, AMPK and several MAPK, besides CK2,48 whereas for silmitasertib several studies have shown that off-target effects are negligible.26,27 The observed synergistic effect between venetoclax and CK2 knockdown or inhibition is best explained by the specific reduction in MCL-1 protein levels after CK2 inhibition or knockdown; the levels of BCL-XL and BCL-2 were not altered (Figure 5A-C, Figure 8). Elevated MCL-1, and BCL-XL, levels have frequently been described as a prominent determinant of primary venetoclax sensitivity in MCL, and, moreover, upregulation of these proteins has been reported as a mechanism of resistance to prolonged venetoclax treatment.3 The up-regulated MCL-1 and BCL-XL proteins serve as a buffer for the released BH3-only proteins upon venetoclax treatment, resulting in reduced BAK/BAX oligomerization and thus reduced apoptosis (Figure 8). Accordingly, venetoclax combined with MCL-1 inhibitors demonstrated strong synergy in several pre-clinical MCL models (Figure 5D; Online Supplementary Figure S6B).3,16,17 However, in vivo toxicities such as cardiotoxicity have thus far hampered the development and clinical approval of MCL-1 therapy.3 Indirect inhibition of MCL-1 by targeting a tumor-specific molecule such as CK2 might prevent these toxicities, especially since silmitasertib showed a safe tolerability profile in clinical trials (NCT03904862, NCT03897036, NCT03571438, NCT04668209, NCT04663737).24,25 In other hematologic malignancies, MCL-1 is also an important determinant in venetoclax resistance and, moreover, CK2 was frequently found to be over-expressed.3,19-21 Combined with the recently reported synergy between silmitasertib and venetoclax in BCP-ALL cells,42 this highlights the possibilities for a combination therapy of CK2 inhibition and venetoclax in other hematologic malignancies as well, and might expand the application of venetoclax to MCL-1-dependent malignancies such as multiple myeloma. Our data indicate that CK2 regulates MCL-1 levels in MCL via its translation machinery in an mTOR-mediated fashion. Targeting CK2 did not result in diminished MCL-1 RNA levels or protein stability (Figure 6B, C), whereas phosphorylation of translation-regulatory proteins, such as S6, eIF4E and 4EBP, was affected (Figure 6D; Online Supplementary Figure S7C). This is considered to specifically affect mRNA translation of, amongst others, MCL-1; elevated eIF4E phosphorylation correlates with high MCL-1 protein levels and overexpression of a phosphomimetic S209D eIF4E variant increases translation of only a limited number of proteins, among which MCL-1.37,38 Furthermore, we have previously shown that macrophages affect MCL-1 translation in CLL cells via the eIF4E-axis40 and others have reported that inhibition of eIF4F assembly in DLBCL cells diminishes MCL-1 levels and synergizes with venetoclax.41 In the pre-B ALL cell line NALM6, silmitasertib treatment was reported to enhance proteasomal degradation of MCL-1.42 However, similar to our findings in MCL, in HEK cells Gandin et al.49 also observed that CK2 controls 4EBP phosphorylation and eIF4F complex assembly. In addition, the expression of genes involved in protein translation correlates with CSNK2A1 expression in MCL patients (Online Supplementary Figure S7A). Notably, in several other cell types CK2 has been shown to phosphorylate components of other translation initiation complexes, thereby also affecting global translation.49-53 Taken together, our data indicate that CK2-mediated regulation of MCL-1 protein levels in MCL is due to the altered activity of translation machinery.
In conclusion, we have shown that aberrant expression of some CK2 subunits in MCL correlates with inferior patient prognosis and that CK2 inhibition strongly sensitizes MCL cells to venetoclax, also in a lymphoid organ-mimetic coculture setting (Figure 8). These pre-clinical findings strongly support further clinical investigation of combination therapy of silmitasertib and venetoclax in MCL patients. Our data indicate that MCL-1 downregulation is the main mechanism underlying the observed synergy, which is in accordance with previous studies showing that MCL-1 downregulation results in sensitization to venetoclax. CK2 inhibition might provide a less toxic therapeutic strategy to sensitize MCL cells to venetoclax as compared to MCL-1 inhibitors, making this a promising combination therapy for patients with MCL, and possibly other hematologic malignancies.
Footnotes
- Received June 28, 2022
- Accepted September 27, 2022
Correspondence
Disclosures
No conflicts of interest to disclose.
Contributions
YJT designed the research, performed experiments, analyzed the data, and wrote the manuscript; MFMdR designed the research and performed bioinformatics; NS performed experiments; RLB and MJK provided materials; JG, EE and STP analyzed the data and revised the manuscript; APK supervised the study, analyzed data and revised the manuscript; MS designed and supervised the study, analyzed data and wrote the manuscript. All authors edited the manuscript.
Data-sharing statement
The Perl, R, and Python scripts used for analyzing the CRISPR screen are available in the public GitHub repository (https://github.com/MFMdeRooij/CRISPRscreen). Raw data and the list of genes for which their targeting gRNAs were depleted in the venetoclax-treated arm are provided in Online Supplementary Table S3. Expression profile data analyzed in this study were obtained from Gene Expression Omnibus (GEO) at GSE93291, GSE132929, and GSE28491 and from the EMBL European Bioinformatics Institute database at E-MTAB-1771.
Funding
This research was supported by grant UVA 2015-7873 from the Dutch Cancer Society (KWF) to MS and APK, and by a grant from Lymph&Co to MS.
References
- Eyre TA, Cheah CY, Wang ML. Therapeutic options for relapsed/refractory mantle cell lymphoma. Blood. 2022; 139(5):666-677. https://doi.org/10.1182/blood.2021013326PubMedPubMed CentralGoogle Scholar
- Jares P, Colomer D, Campo E. Molecular pathogenesis of mantle cell lymphoma. J Clin Invest. 2012; 122(10):3416-3423. https://doi.org/10.1172/JCI61272PubMedPubMed CentralGoogle Scholar
- Thus YJ, Eldering E, Kater AP, Spaargaren M. Tipping the balance: toward rational combination therapies to overcome venetoclax resistance in mantle cell lymphoma. Leukemia. 2022; 36(9):2165-2176. https://doi.org/10.1038/s41375-022-01627-9PubMedPubMed CentralGoogle Scholar
- Kater AP, Wu JQ, Kipps T. Venetoclax plus rituximab in relapsed chronic lymphocytic leukemia: 4-year results and evaluation of impact of genomic complexity and gene mutations from the MURANO phase III study. J Clin Oncol. 2020; 38(34):4042-4054. https://doi.org/10.1200/JCO.20.00948PubMedPubMed CentralGoogle Scholar
- Davids MS, Roberts AW, Kenkre VP. Long-term follow-up of patients with relapsed or refractory Non-Hodgkin lymphoma treated with Venetoclax in a phase 1, first-in-human study. Clin Cancer Res. 2021; 27(17):4690-4695. https://doi.org/10.1158/1078-0432.CCR-20-4842PubMedPubMed CentralGoogle Scholar
- Eyre TA, Walter HS, Iyengar S. Efficacy of venetoclax monotherapy in patients with relapsed, refractory mantle cell lymphoma after Bruton tyrosine kinase inhibitor therapy. Haematologica. 2019; 104(2):e68-e71. https://doi.org/10.3324/haematol.2018.198812PubMedPubMed CentralGoogle Scholar
- Zhao S, Kanagal-Shamanna R, Navsaria L. Efficacy of venetoclax in high risk relapsed mantle cell lymphoma (MCL) -outcomes and mutation profile from venetoclax resistant MCL patients. Am J Hematol. 2020; 95(6):623-629. https://doi.org/10.1002/ajh.25796PubMedGoogle Scholar
- Tam CS, Anderson MA, Pott C. Ibrutinib plus venetoclax for the treatment of mantle cell lymphoma. N Engl J Med. 2018; 378(13):1211-1223. https://doi.org/10.1056/NEJMoa1715519PubMedGoogle Scholar
- Wang M, Ramchandren R, Chen R. Concurrent ibrutinib plus venetoclax in relapsed/refractory mantle cell lymphoma: the safety run-in of the phase 3 SYMPATICO study. J Hematol Oncol. 2021; 14(1):179. https://doi.org/10.1186/s13045-021-01188-xPubMedPubMed CentralGoogle Scholar
- Doench JG, Fusi N, Sullender M. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat Biotechnol. 2016; 34(2):184-191. https://doi.org/10.1038/nbt.3437PubMedPubMed CentralGoogle Scholar
- Lantermans HC, Minderman M, Kuil A, Kersten MJ, Pals ST, Spaargaren M. Identification of the SRC-family tyrosine kinase HCK as a therapeutic target in mantle cell lymphoma. Leukemia. 2021; 35(3):881-886. https://doi.org/10.1038/s41375-020-0934-6PubMedPubMed CentralGoogle Scholar
- de Rooij MFM, Thus YJ, Swier N, Beijersbergen RL, Pals ST, Spaargaren M. A loss-of-adhesion CRISPR-Cas9 screening platform to identify cell adhesion-regulatory proteins and signaling pathways. Nat Commun. 2022; 13(1):2136. https://doi.org/10.1038/s41467-022-29835-yPubMedPubMed CentralGoogle Scholar
- Scott DW, Abrisqueta P, Wright GW. New molecular assay for the proliferation signature in mantle cell lymphoma applicable to formalin-fixed paraffin-embedded biopsies. J Clin Oncol. 2017; 35(15):1668-1677. https://doi.org/10.1200/JCO.2016.70.7901PubMedPubMed CentralGoogle Scholar
- Ma MCJ, Tadros S, Bouska A. Subtype-specific and cooccurring genetic alterations in B-cell non-Hodgkin lymphoma. Haematologica. 2022; 107(3):690-701. https://doi.org/10.3324/haematol.2020.274258PubMedPubMed CentralGoogle Scholar
- Subramanian A, Tamayo P, Mootha VK. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005; 102(43):15545-15550. https://doi.org/10.1073/pnas.0506580102PubMedPubMed CentralGoogle Scholar
- Chiron D, Dousset C, Brosseau C. Biological rational for sequential targeting of Bruton tyrosine kinase and Bcl-2 to overcome CD40-induced ABT-199 resistance in mantle cell lymphoma. Oncotarget. 2015; 6(11):8750-8759. https://doi.org/10.18632/oncotarget.3275PubMedPubMed CentralGoogle Scholar
- Prukova D, Andera L, Nahacka Z. Cotargeting of BCL2 with venetoclax and MCL1 with S63845 is synthetically lethal in vivo in relapsed mantle cell lymphoma. Clin Cancer Res. 2019; 25(14):4455-4465. https://doi.org/10.1158/1078-0432.CCR-18-3275PubMedGoogle Scholar
- Jiang H, Lwin T, Zhao X. Venetoclax as a single agent and in combination with PI3K-MTOR1/2 kinase inhibitors against ibrutinib sensitive and resistant mantle cell lymphoma. Br J Haematol. 2019; 184(2):298-302. https://doi.org/10.1111/bjh.15079PubMedPubMed CentralGoogle Scholar
- Roffey SE, Litchfield DW. CK2 regulation: perspectives in 2021. Biomedicines. 2021; 9(10):1361. https://doi.org/10.3390/biomedicines9101361PubMedPubMed CentralGoogle Scholar
- Borgo C, D’Amore C, Sarno S, Salvi M, Ruzzene M. Protein kinase CK2: a potential therapeutic target for diverse human diseases. Signal Transduct Target Ther. 2021; 6(1):183. https://doi.org/10.1038/s41392-021-00567-7PubMedPubMed CentralGoogle Scholar
- Chua MMJ, Lee M, Dominguez I. Cancer-type dependent expression of CK2 transcripts. PLoS One. 2017; 12(12):e0188854. https://doi.org/10.1371/journal.pone.0188854PubMedPubMed CentralGoogle Scholar
- Manni S, Brancalion A, Mandato E. Protein kinase CK2 inhibition down modulates the NF-κB and STAT3 survival pathways, enhances the cellular proteotoxic stress and synergistically boosts the cytotoxic effect of bortezomib on multiple myeloma and mantle cell lymphoma cells. PLoS One. 2013; 8(9):e75280. https://doi.org/10.1371/journal.pone.0075280PubMedPubMed CentralGoogle Scholar
- Kim JS, Eom JI, Cheong JW. Protein kinase CK2alpha as an unfavorable prognostic marker and novel therapeutic target in acute myeloid leukemia. Clin Cancer Res. 2007; 13(3):1019-1028. https://doi.org/10.1158/1078-0432.CCR-06-1602PubMedGoogle Scholar
- Marschke RF, Borad MJ, McFarland RW. Findings from the phase I clinical trials of CX-4945, an orally available inhibitor of CK2. J Clin Oncol. 2011; 29(Suppl):3087. https://doi.org/10.1200/jco.2011.29.15_suppl.3087Google Scholar
- Borad MJ, Bai LY, Chen MH. Silmitasertib (CX-4945) in combination with gemcitabine and cisplatin as first-line treatment for patients with locally advanced or metastatic cholangiocarcinoma: a phase Ib/II study. J Clin Oncol. 2021; 39:3. https://doi.org/10.1200/JCO.2021.39.3_suppl.312Google Scholar
- Borgo C, Cesaro L, Hirota T. Comparing the efficacy and selectivity of Ck2 inhibitors. A phosphoproteomics approach. Eur J Med Chem. 2021; 214:113217. https://doi.org/10.1016/j.ejmech.2021.113217PubMedGoogle Scholar
- Rosales M, Rodríguez-Ulloa A, Besada V. Phosphoproteomic landscape of AML cells treated with the ATP-competitive CK2 inhibitor CX-4945. Cells. 2021; 10(2):338. https://doi.org/10.3390/cells10020338PubMedPubMed CentralGoogle Scholar
- Di Maira G, Salvi M, Arrigoni G. Protein kinase CK2 phosphorylates and upregulates Akt/PKB. Cell Death Differ. 2005; 12(6):668-677. https://doi.org/10.1038/sj.cdd.4401604PubMedGoogle Scholar
- Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017; 169(3):381-405. https://doi.org/10.1016/j.cell.2017.04.001PubMedPubMed CentralGoogle Scholar
- Bliss CI. The toxicity of poisons applied jointly. Ann Appl Biol. 1939; 26(3):585-615. https://doi.org/10.1111/j.1744-7348.1939.tb06990.xGoogle Scholar
- Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzyme Regul. 1984; 22:27-55. https://doi.org/10.1016/0065-2571(84)90007-4PubMedGoogle Scholar
- Yadav B, Wennerberg K, Aittokallio T, Tang J. Searching for drug synergy in complex dose–response landscapes using an interaction potency model. Comput Struct Biotechnol J. 2015; 13:504-513. https://doi.org/10.1016/j.csbj.2015.09.001PubMedPubMed CentralGoogle Scholar
- Le Gouill S, Morschhauser F, Chiron D. Ibrutinib, obinutuzumab, and venetoclax in relapsed and untreated patients with mantle cell lymphoma: a phase 1/2 trial. Blood. 2021; 137(7):877-887. https://doi.org/10.1182/blood.2020008727PubMedGoogle Scholar
- De Rooij MFM, Kuil A, Geest CR. The clinically active BTK inhibitor PCI-32765 targets B-cell receptor- and chemokine-controlled adhesion and migration in chronic lymphocytic leukemia. Blood. 2012; 119(11):2590-2594. https://doi.org/10.1182/blood-2011-11-390989PubMedGoogle Scholar
- Chang BY, Francesco M, De Rooij MFM. Egress of CD19+CD5+ cells into peripheral blood following treatment with the Bruton tyrosine kinase inhibitor ibrutinib in mantle cell lymphoma patients. Blood. 2013; 122(14):2412-2424. https://doi.org/10.1182/blood-2013-02-482125PubMedPubMed CentralGoogle Scholar
- Thomas LW, Lam C, Edwards SW. Mcl-1; the molecular regulation of protein function. FEBS Lett. 2010; 584(14):2981-2989. https://doi.org/10.1016/j.febslet.2010.05.061PubMedGoogle Scholar
- Wendel HG, Silva RLA, Malina A. Dissecting eIF4E action in tumorigenesis. Genes Dev. 2007; 21(24):3232-3237. https://doi.org/10.1101/gad.1604407PubMedPubMed CentralGoogle Scholar
- Martínez A, Sesé M, Losa JH. Phosphorylation of eIF4E confers resistance to cellular stress and DNA-damaging agents through an interaction with 4E-T: a rationale for novel therapeutic approaches. PLoS One. 2015; 10(4):e0123352. https://doi.org/10.1371/journal.pone.0123352PubMedPubMed CentralGoogle Scholar
- Bhat M, Robichaud N, Hulea L, Sonenberg N, Pelletier J, Topisirovic I. Targeting the translation machinery in cancer. Nat Rev Drug Discov. 2015; 14(4):261-278. https://doi.org/10.1038/nrd4505PubMedGoogle Scholar
- van Attekum M, Terpstra S, Slinger E. Macrophages confer survival signals via CCR1-dependent translational MCL-1 induction in chronic lymphocytic leukemia. Oncogene. 2017; 36(26):3651-3660. https://doi.org/10.1038/onc.2016.515PubMedPubMed CentralGoogle Scholar
- Herzog LO, Walters B, Buono R. Targeting eIF4F translation initiation complex with SBI-756 sensitises B lymphoma cells to venetoclax. Br J Cancer. 2021; 124(6):1098-1109. https://doi.org/10.1038/s41416-020-01205-9PubMedPubMed CentralGoogle Scholar
- Lazaro-Navarro J, Pimentel-Gutiérrez HJ, Gauert A. Inhibiting casein kinase 2 sensitizes acute lymphoblastic leukemia cells to venetoclax via MCL1 degradation. Blood Adv. 2021; 5(24):5501-5506. https://doi.org/10.1182/bloodadvances.2021004513PubMedPubMed CentralGoogle Scholar
- Pagano MA, Bain J, Kazimierczuk Z. The selectivity of inhibitors of protein kinase CK2: an update. Biochem J. 2008; 415(3):353-365. https://doi.org/10.1042/BJ20080309PubMedGoogle Scholar
- Spagnuolo C, Cerella C, Russo M, Chateauvieux S, Diederich M, Russo GL. Quercetin downregulates Mcl-1 by acting on mRNA stability and protein degradation. Br J Cancer. 2011; 105(2):221-230. https://doi.org/10.1038/bjc.2011.229PubMedPubMed CentralGoogle Scholar
- Zhao M, Ma J, Zhu HY. Apigenin inhibits proliferation and induces apoptosis in human multiple myeloma cells through targeting the trinity of CK2, Cdc37 and Hsp90. Mol Cancer. 2011; 10:104. https://doi.org/10.1186/1476-4598-10-104PubMedPubMed CentralGoogle Scholar
- Jacquemin G, Granci V, Gallouet AS. Quercetin-mediated Mcl-1 and survivin downregulation restores TRAIL-induced apoptosis in non-Hodgkin’s lymphoma B cells. Haematologica. 2012; 97(1):38-46. https://doi.org/10.3324/haematol.2011.046466PubMedPubMed CentralGoogle Scholar
- Russo M, Spagnuolo C, Volpe S, Tedesco I, Bilotto S, Russo GL. ABT-737 resistance in B-cells isolated from chronic lymphocytic leukemia patients and leukemia cell lines is overcome by the pleiotropic kinase inhibitor quercetin through Mcl-1 down-regulation. Biochem Pharmacol. 2013; 85(7):927-936. https://doi.org/10.1016/j.bcp.2013.01.011PubMedGoogle Scholar
- Rather RA, Bhagat M. Quercetin as an innovative therapeutic tool for cancer chemoprevention: molecular mechanisms and implications in human health. Cancer Med. 2020; 9(24):9181-9192. https://doi.org/10.1002/cam4.1411PubMedPubMed CentralGoogle Scholar
- Gandin V, Masvidal L, Cargnello M. mTORC1 and CK2 coordinate ternary and eIF4F complex assembly. Nat Commun. 2016; 7:11127. https://doi.org/10.1038/ncomms11127PubMedPubMed CentralGoogle Scholar
- Wang X, Paulin FEM, Campbell LE. Eukaryotic initiation factor 2B: identification of multiple phosphorylation sites in the ϵ-subunit and their functions in vivo. EMBO J. 2001; 20(16):4349-4359. https://doi.org/10.1093/emboj/20.16.4349PubMedPubMed CentralGoogle Scholar
- Homma MK, Wada I, Suzuki T, Yamaki J, Krebs EG, Homma Y. CK2 phosphorylation of eukaryotic translation initiation factor 5 potentiates cell cycle progression. Proc Natl Acad Sci U S A. 2005; 102(43):15688-15693. https://doi.org/10.1073/pnas.0506791102PubMedPubMed CentralGoogle Scholar
- Llorens F, Duarri A, Sarró E, Roher N, Plana M, Itarte E. The N-terminal domain of the human eIF2β subunit and the CK2 phosphorylation sites are required for its function. Biochem J. 2006; 394(1):227-236. https://doi.org/10.1042/BJ20050605PubMedPubMed CentralGoogle Scholar
- Borgo C, Franchin C, Salizzato V. Protein kinase CK2 potentiates translation efficiency by phosphorylating eIF3j at Ser127. Biochim Biophys Acta - Mol Cell Res. 2015; 1853(7):1693-1701. https://doi.org/10.1016/j.bbamcr.2015.04.004PubMedGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.