BTK acts as a modulator of the response to imatinib in chronic myeloid leukemia

  • Authors:
    • Lena Schmidlechner
    • Inga Nagel
    • Inga Vater
    • Ingolf Cascorbi
    • Meike Kaehler
  • View Affiliations

  • Published online on: July 4, 2024     https://doi.org/10.3892/ol.2024.14557
  • Article Number: 424
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The use of tyrosine kinase inhibitors, such as imatinib, against the chronic myeloid leukemia (CML)‑causing kinase BCR::ABL1 has become the model for successful targeted therapy. Nevertheless, drug resistance remains a clinical problem. Analysis of genome‑wide expression and genetic aberrations of an in vitro imatinib‑resistant CML cell line revealed downregulation of Bruton's tyrosine kinase (BTK), predominantly associated with B cell malignancies, and a novel BTK kinase domain variant in imatinib resistance. This raised the question of the role of BTK in imatinib‑resistant CML. In the present study, BTK downregulation and the presence of the BTK variant c.1699_1700delinsAG p.(Glu567Arg) were confirmed in imatinib resistance in vitro. Similarly, BTK inhibition or small interfering RNA‑mediated BTK knockdown reduced imatinib susceptibility by 84 and 71%, respectively. BTK overexpression was detrimental to CML cells, as proliferation was significantly reduced by 20.5% under imatinib treatment. In addition, BTK rescue in imatinib‑resistant cells restored imatinib sensitivity. The presence of the BTK p.(Glu567Arg) variant increased cell numbers (57%) and proliferation (37%) under imatinib exposure. These data demonstrate that BTK is important for the development of imatinib resistance in CML: Its presence increased drug response, while its absence promotes imatinib resistance. Moreover, the BTK p.(Glu567Arg) variant abrogates imatinib sensitivity. These findings demonstrate a context‑dependent role for BTK as an oncogene in B cell malignancies, but as a tumor suppressor in other neoplasms.

Introduction

The use of tyrosine kinase inhibitors (TKIs), i.e., imatinib, in the hematological malignancy chronic myeloid leukemia (CML) is the most prominent model for successful targeted therapy. This drug targets the disease-causing, constitutively active BCR::ABL1 (breakpoint cluster region/Abelson tyrosine kinase 1) kinase, which results from the reciprocal t(9;22)(q34;q11) translocation forming the so-called Philadelphia chromosome and the BCR::ABL1 fusion gene (13). With a five-year survival rate of 83%, TKI treatment of CML is tremendously successful, however, the handling of CML has become more complex due to the development of TKI resistance (4,5). About half of the TKI-resistant CML patients acquire sequence variants in BCR::ABL1 that prevent TKI binding and sufficient inhibition of downstream target phosphorylation (69). The most prominent pathogenic variant is the so-called gatekeeper mutation p.(Thr315Ile), which impairs the ATP binding pocket of BCR::ABL1 leading to therapy failure for first and second generation TKIs (1012). In addition, the P-loop mutation p.(Gly250Glu) or the activation loop mutation p.(His396Arg) also lead to treatment failure (13). For the other half of TKI-resistant CML patients, a variety of resistance mechanisms are discussed, e.g., drug-drug interactions via CYP3A4, pharmacogenetic polymorphisms, drug efflux transporter, in particular overexpression of the drug efflux transporters of the ATP binding cassette (ABC) family, epigenetics, microRNA deregulation and activation of alternative signaling pathways, in particular JAK/STAT, MAP kinases or PI3K/Akt signaling (12,14). To analyze drug resistance mechanisms, genome-wide gene expression and exome sequencing data of an in vitro-TKI-resistance CML model were performed in a previous study, which identified downregulation of the non-receptor kinase Bruton's Tyrosine Kinase (BTK) expression and an acquired novel potentially pathogenic dinucleotide BTK variant in imatinib resistance [GSE227347 (15), PRJEB60564 (16)].

First identified in loss-of-function mutations causing X-linked agammaglobulinemia (XLA)/Bruton syndrome, BTK is central for B lymphocyte development (1719). BTK is known to play a crucial role in Toll-like receptor (TLR), chemokine receptor, as well as B cell receptor (BCR) signaling (20). Besides its essential role in the pathogenesis of XLA, there are also several other malignancies based on BTK dysregulation: Waldenström's macroglobulinemia, e.g., is an indolent B cell lymphoma caused by myeloid differentiation primary response 88-(MYD88)-dependent BTK activation (21). In chronic lymphocytic leukemia (CLL), a malignant neoplasm resulting in overproduction of mature CD5+ B lymphocytes by a constitutively activated BCR pathway, BTK is an essential component of signal transmission (22). Another malignancy also linked to the BTK-dependent BCR signaling pathway is mantle cell lymphoma (MCL), an aggressive mature B cell non-Hodgkin lymphoma characterized by t(11;14)(q13;q32) translocation, leading to an upregulation of cyclin D1 and BCR activation (23). For all malignancies, in which BTK is a crucial component of pathogenesis, BTK inhibition represents a possible treatment option.

Consequently, BTK became a potential druggable candidate for targeted therapy, leading to the introduction of the selective and FDA-approved BTK inhibitor (BTKi) ibrutinib, which is used as a single agent or combinational treatment in various cancers (17). Similar to other tyrosine kinase inhibitors, relapses and treatment failure can be observed during the treatment with imatinib. This problem, as well as the occurrence of adverse events, led to the development of second generation BTKIs, e.g., acalabrutinib, zanubrutinib and tirabrutinib, with the goal to improve the tolerability by higher selectivity to BTK and to overcome ibrutinib resistance (2325).

The aim of the current study was to investigate the role of BTK and the previously discovered novel dinucleotide BTK variant in imatinib resistance in chronic myeloid leukemia cells.

Materials and methods

Reagents and compounds

All chemicals and reagents not indicated differently, were purchased from Sigma-Aldrich or Carl Roth. Imatinib and ibrutinib were obtained from Sigma-Aldrich; Merck KGaA. Imatinib was diluted in 10 mM aqueous stock solutions, ibrutinib in 100 mM stock solutions in DMSO, both stored at −20°C.

Cell culture

K-562 cells (RRID: CVCL 0004), retrieved from the pleural effusion of a 53-year old woman (26), originate from the German Collection of Microorganisms and Cell Cultures (DSMZ). Cells were cultivated in RPMI-1640 (Thermo Fisher Scientific, Inc.) supplemented with 10% v/v FCS (Bio&Sell), 1% v/v Penicillin/Streptomycin (Carl Roth) and 1% v/v L-Alanyl-L-Glutamin. Imatinib-resistant cell lines were generated as previously described (15). Authenticity of treatment-naïve as well as cell lines resistant against 0.5 µM and 2 µM imatinib was confirmed by short tandem repeats (STR) analysis using the GenePrint 10 system (Promega). BCR::ABL1 mutations were analyzed as described elsewhere (15). The cell lines showed no BCR::ABL1 mutations.

Site-directed mutagenesis

Mutagenesis of an BTK-encoding plasmid (Gene ID: 2335, ABIN3996197, antibodies-online, Aachen) was performed to insert the BTK variants (NM_000061.3,) c.1699G>A p.(Glu567Lys), c.1700A>G p.(GluE567Gly) and c1699_1700delinsAG p.(GluE567Arg) using the primers BTK_G1699A_F 5′-GTCCCCACCGAAAGTCCTGAT-3′, BTK_G1699A_R 5′-CACCGGACTGGAAATTTGG-3′, BTK_A1700G_F 5′-TCCCCACCGGGAGTCCTGATG-3′, BTK_A1700G_R 5′-CCACCGGACTGGAAATTTGG-3′, BTK_GA1699-1700AG_F 5′-GTCCCCACCGAGAGTCCTGATG-3′ and BTK_GA1699-1700AG_R 5′-CACCGGACTGGAAATTTG-3′ obtained from Sigma-Aldrich. 25 ng of template DNA, an annealing temperature of 67°C and the Q5 Site-Directed Mutagenesis Kit (New England Biolabs) were used according to the manufacturer's protocol. Sequence identification was confirmed using Sanger sequencing.

Isolation of nucleic acids

Total RNA isolation was performed using 1×106 K-562 cells and PeqGOLD TriFast (VWR) according to the manufacturer's recommendation. Cell line DNA was purified using Gentra Puregene Cell Kit (Qiagen) according to the manufacturer's protocol.

Amplicon sequencing (MiSeq, Illumina)

Generation of PCR products was performed using MyTaq Polymerase (Bioline), DNA of treatment-naïve, 0.5 µM and 2 µM imatinib-resistant cells, and the primers BTK_Intron_F 5′-TGACACTCTTGTGACCGTGC-3′ and BTK_Intron_R 5′-ACAGTAAGCACTCCCCAAGG-3′ with annealing at 54°C. PCR products were purified using the GeneJET PCR Purification Kit (Thermo Fisher Scientific, Inc.). After PCR amplicon preparation with the Nextera XT Sequencing Kit (Illumina), Next Generation Sequencing (NGS) SBS technology with Illumina MiSeq was performed as described elsewhere (16,27).

RT-qPCR

According to the manufacturer's protocol, 1 µg total RNA was reversely transcribed using random hexamer primers and the High Capacity cDNA Synthesis Kit (Thermo Fisher Scientific, Inc.) for 10 min at 25°C, 120 min at 37°C and 5 min at 85°C. cDNA was stored at −80°C prior further usage. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) of target genes was performed in triplicates using the TaqMan assays (Thermo Fischer Scientific, Inc.) HPRT1 (Hs02800695_m1, Chr.X:134460145-134500668), TBP (Hs00427620_m1, Chr.6:170554333-170572870) and GAPDH (Hs0266705_g1, Chr.12:6534405-653837) as internal controls, as well as BTK (Hs00975865_m1, Chr.X:101349447-101390796), with Universal Master Mix II without UNG (Thermo Fisher Scientific, Inc.) on the QuantStudio 7 device (Thermo Fisher Scientific, Inc.) with default cycling conditions. Relative gene expression was calculated using the 2−ΔΔCt method (28).

Immunoblotting

Whole cell lysates and immunoblotting were performed as described elsewhere (2931). 20 µg of protein were loaded onto the membranes and blots were probed with the following antibodies, obtained from Santa Cruz, Cell Signaling Technology or LiCOR (Bad Homburg): BTK: Cat# sc-28387, RRID: AB_626770, 1:1,000; p-BTK: Cat# 5082, RRID: AB_10561017, 1:1,000; GAPDH: Cat# sc-47724, RRID: AB_627678, 1:1,000; anti-mouse: Cat# 926-68070, RRID: AB_10956588, all 1:10,000; anti-rabbit: Cat# 926-32211, RRID: AB_621843, all 1:10,000. Primary antibodies were diluted in Intercept/TBS blocking solution (LiCOR), supplemented with 0.2% v/v Tween-20. Secondary antibodies were diluted in TBS, supplemented with 0.1% v/v Tween-20.

Plasmid and siRNA transfection

Plasmid and siRNA transfection were performed using the Amaxa nucleofector Kit V and nucleofector 2 b device (Lonza). For plasmid transfection, 2×106 cells were transfected with 10 µg of the respective plasmid. 24 h after transfection, cells were seeded onto cell plates for subsequent experiments. For siRNA transfection, K-562 cells were transfected with 200 nM negative control stealth (1295300) and stealth RNAi Pre-designed BTK-siRNA 5′-GGAGUCAGGCUGAGCAACUGCUAAA-3′ (HSS101131, both Thermo Fisher Scientific, Inc.). After transfection, cells were seeded onto respective cell culture plates and exposed to 2 µM imatinib for 48 h. Stable transfection was performed exposing the cells to 800 mg/ml G-418 for 4 weeks.

Cellular fitness assays

Cell numbers were obtained by trypan blue staining and performed after 48 h imatinib incubation as described elsewhere (27). WST-1 assay (Sigma-Aldrich; Merck KGaA) was performed after 48 h with 5×104 cells as previously described (29). Proliferation was analyzed using human MKI67 ELISA Kit (MyBioSource) on 1×106 cells according to the manufacturer's recommendation with 50 µg protein/well. Data were analyzed normalizing imatinib-treated to non-treated samples.

Pathogenicity prediction and statistical analysis

Pathogenicity prediction of BTK variants was performed using the CADD score [CADD v1.6, (32)]. Statistical analysis was performed using unpaired student's t-test or one-way ANOVA with subsequent Dunnett's test using GraphPad prism software (Version 9.0 for Windows).

Results

Reduction of BTK expression and gain of a BTK variant in imatinib-resistant CML cells

In a previous study, an in vitro-TKI resistance model of K-562 CML cells was obtained by stepwise exposure to increasing imatinib concentrations to obtain cells resistant against 0.5 and 2 µM imatinib (15). Analysis of genome-wide expression in this model revealed a BTK downregulation in one of the replicate cell lines of imatinib resistance [-3.7-fold, P=7.08×10−7, (15)]. To generate insights into the role of BTK in the pathogenesis of CML, BTK expression in treatment-naïve and imatinib-resistant cell lines was examined on mRNA and protein level. BTK was significantly downregulated on mRNA (0.5 µM: −33%; 2 µM: −87%, both P<0.001) and protein level in 2 µM imatinib-resistant cells confirming the BTK downregulation in imatinib resistance (Fig. 1A and B). To investigate, whether BTK downregulation occurs in response to imatinib treatment, BTK mRNA expression was measured after short-term exposure to imatinib of 3–24 h. Hence, imatinib treatment led to a significant reduction in BTK mRNA levels by 33 to 62% (3 h: P=0.002, 6 h: P=0.005, 12 h: P=0.03, 24 h: P=0.001, Fig. 1C).

As previously reported, the novel BTK dinucleotide variant [NM_000061.3, c.1699_1700delinsAG p.(Glu567Arg)] was detected in imatinib-resistant cells [PRJEB60564 (16)]. This variant leads to an amino acid exchange from glutamic acid to arginine in the BTK kinase domain, which is likely to have a detrimental effect on protein function according to prediction tools (CADD: 25.2). In depth-sequencing confirmed the presence of this variant in 0.5 (14%) and 2 µM imatinib-resistant cells (34%, considering the K-562 triploidy, Fig. 1D). Overall, our findings show a downregulation of BTK expression and a gain of a novel dinucleotide BTK variant in imatinib resistance.

Loss of BTK activity or its expression promotes imatinib resistance

Since a putative pathogenic BTK dinucleotide variant p.(Glu567Arg) was detected in imatinib resistance, the question arose whether deterioration of the kinase function through inhibition could also influence the imatinib susceptibility of CML cells. Thus, treatment-naïve cells were exposed to the BTK-specific inhibitor ibrutinib and compared to single treatment with imatinib. Treatment with imatinib significantly decreased cell viability of treatment-naïve, but not imatinib-resistant K-562 cells by 73 to 89% (300–2,000 nM: P<0.001), while ibrutinib did not affect the cells (Fig. 2A and B). However, in a combined treatment with low dose (100 nM) imatinib, the presence of ibrutinib dose-dependently increased cell viability by 11 to 37% (ibrutinib (nM): 100: P=0.05, 500: P=0.02, 1,000: P=0.002, Fig. 2C), while this was not observed in combination with high dose (2 µM) imatinib (Fig. 2D).

To investigate whether loss of BTK expression also affects K-562 cells, a siRNA-mediated BTK knockdown was performed in treatment-naïve CML cells. Successful BTK downregulation (P<0.001, Fig. 3A) led to a significant increase in cell number (97.2%, P<0.001) and proliferation rate of CML cells compared to negative control-transfected cells (27.5%, P=0.01, Fig. 2B). After subsequent treatment with imatinib, BTK knockdown cells also showed increased cell numbers (70.6%, P=0.04), while proliferation was not significantly altered (Fig. 2C). Overall, these findings indicate that both, inhibition and the loss of BTK expression, are beneficial for CML cells and reduce imatinib susceptibility.

BTK overexpression and its rescue reinstate imatinib susceptibility

Vice versa, to analyze whether high BTK expression is detrimental for CML cells under imatinib exposure, treatment-naïve cells were stably transfected with a BTK-encoding plasmid to induce BTK overexpression (Fig. 4A) and challenged with 2 µM imatinib. In response to imatinib, the presence of BTK resulted in a reduction in cell number (−25.9%, P=0.03), proliferation (−20.5%, P=0.04), and cell viability compared to negative control-transfected cells (−25.7%, P<0.001, Fig. 4B). These data indicate that the presence of BTK augments the response to imatinib treatment.

Since BTK was significantly downregulated in imatinib-resistant cells, restoration of BTK expression in these cells was performed by transfection experiments followed by imatinib exposure. After successful restoration of BTK expression, cell number (−28.4%, P=0.002), proliferation (−26.7%, P=0.04) and cell viability (−12.4%, P=0.003) were decreased compared to negative control-transfected cells indicating an improved response to imatinib (Fig. 4C and D). Therefore, our findings demonstrate that the restoration of BTK in imatinib-resistant cells reinstates imatinib susceptibility.

Influence of BTK variants on imatinib susceptibility

As the BTK variant p.(Glu567Arg) was acquired in imatinib resistance, the question arose whether this mutation affects CML cells and could be involved in the development of resistance. Stable transfection of BTK wild-type and the p.(Glu567Arg) variant in treatment-naïve K-562 cells was performed. For comparison, the pathogenic kinase-dead BTK p.(Glu567Lys) (33) and benign p.(Glu567Gly) variants were also transfected. BTK mRNA expression was significantly increased in all stable transfected cell lines indicating successful overexpression (P<0.001, Fig. 5A). On protein level, however, in p.(Glu567Arg) and p.(Glu567Lys)-expressing cells, a reduction in BTK protein level was observed, while p.(Glu567Gly) had the highest BTK levels pointing to decreased protein stability in the presence of lysine or arginine at this residue (Fig. 5A). To investigate if the cells harboring BTK variants are still able to respond to ibrutinib treatment, the cells were exposed to ibrutinib (Figs. 2B, S1). The presence of 100 nM ibrutinib resulted in increased cell viability for p.(Glu567Arg) (22.6%, P=0.008), similarly to p.(Glu567Lys) (45.6%, P=0.006), but not for p.(Glu567Gly) compared to cells overexpressing BTK wild-type (Fig. 5B). After exposure to 2 µM imatinib, the presence of the p.(Glu567Arg) variant led to an increase in cell number (57.2%, P=0.003), proliferation (36.5%, P<0.001) and cell viability (60.1%, P<0.001) during imatinib treatment compared to wild-type BTK (Fig. 5C). A similar effect was also observed for BTK p.(Glu567Lys), as increased cell numbers (35.4%, P=0.04), proliferation (61.2%, P<0.001) and cell viability were observed under imatinib exposure (29.5%, P<0.001, Fig. 5B). The benign variant p.(Glu567Gly), however, did not affect the response to imatinib treatment. These findings show that p.(Glu567Arg), as well as p.(Glu567Lys), result in diminished imatinib susceptibility. Overall, our data show that the acquired BTK p.(Glu567Arg) variant appears to be relevant for the development of imatinib resistance.

Discussion

The Bruton's tyrosine kinase (BTK) was initially associated with X-linked agammaglobulinemia (XLA), a primary immunodeficiency disorder characterized by a severe blockade of B cell development in the bone marrow resulting in a lack of B cells and serum antibodies due to (18). Since its discovery, BTK was found to be crucial in oncogenic signaling being important for B cell development and mature B cell function. Accordingly, BTK is deregulated in several B cell malignancies, including CLL, small lymphocytic leukemia (SLL), MCL and follicular lymphoma, as well as in various other tumors, such as pancreatic, lung, breast, ovarian, prostate and colorectal cancer (17,34). In addition, BTK has been shown to be an essential player in chronic graft-vs.-host disease or Waldenström macroglobulinemia (17,35). In the present study, we analyzed the role of BTK in the myeloproliferative disease CML. A downregulation of BTK expression, as well as the acquired BTK p.(Glu567Arg) variant were detected in imatinib resistance. Subsequent transfection experiments demonstrated that the presence of BTK affects the response to imatinib in CML cells. Further, imatinib susceptibility was restored after rescue of BTK expression in imatinib-resistant cells. In addition, the BTK p.(Glu567Arg) variant seems to be pathogenic to the protein function resulting in a loss-of-function promoting imatinib resistance.

BTK is a 77 kDa protein composed of 659 amino acids and five domains: A plectrin homology (PH), a Tec homology (TH), an SH3 and SH2 domain, and the C-terminal kinase domain (36). The SH2 domain mutation p.(Thr316Ala) and the kinase domain mutations p.(Thr474Ile/Ser), p.(Cys481Ser/Ala/Phe/Gly/Arg/Tyr) and p.(Leu528Trp) with p.(Cys481Ser) are the main BTK mutations, associated with BTKI resistance in B cell malignancies (36). Clinical trials of secondary drug resistant CLL have shown the presence of the BTK p.(Cys481Ser) mutation in approximately 80% of patients resulting in a loss of drug binding while maintaining kinase activity (36,37). This mutation has also been detected in drug resistant or relapsed MCL and diffuse large B cell lymphoma (DLBCL) leading to a sustained BTK signaling and, consequently, to tumor progression (38). In the present study, the BTK p.(Glu567Arg) variant, which is located at the C-terminal end of the kinase domain, was acquired in imatinib resistance (16,39). This variant showed similar effects on imatinib susceptibility as the published p.(Glu567Lys) variant, which was shown to suppress BTK-mediated NLRP3 inflammasome activation in brain ischemia (33). Loss-of-function variants at this amino residue, such as the exchange to glutamine p.(Glu567Gln), have previously been associated with XLA (40). The p.Glu567 residue was found to form an ionic bond to Arg641, which stabilizes the kinase structure (41). Therefore, mutations at this residue are likely to affect BTK stability and may also explain the reduction in BTK expression in the presence of p.(Glu567Arg) in imatinib resistance observed here. Further, stable transfection of p.(Glu567Arg) was shown to result in increased levels of BTK mRNA, but not protein indicating increased protein turnover or reduced translation. Interestingly, both, BTK p.(Glu567Arg) and p.(Glu567Lys) variants also resulted in an increased cell fitness in the presence of ibrutinib, which could be a results of decreased binding of ibrutinib to the variant proteins.

BCR::ABL1 TKIs are tremendously successful in CML treatment. However, therapy resistance remains a clinical problem with a need for TKI alternatives for 20–25% of CML patients within five years after therapy onset (4). In B cell malignancies, the TKI ibrutinib also shows significant clinical impact with overall response rates ranging from 90 and 97%, e.g., in CLL (42,43). Ibrutinib is a covalent kinase inhibitor, which irreversibly binds to the BTK cysteine 481 in the catalytic site preventing autophosphorylation at tyrosine 223 and thereby, downstream signaling (39). Interestingly, it was shown that BTK is also a target of the second generation BCR::ABL1 inhibitor dasatinib (44). In the present study, we found that inhibiting BTK by ibrutinib or knocking down its expression prevented CML cells from imatinib resulting in increased cell proliferation rates. These findings contradict a study on primary CML stem cells, where BTK inhibition with ibrutinib was demonstrated as a potential therapy approach in combination with BCR::ABL1 TKI therapy to eradicate leukemic stem cells mediated by inhibition of the Fc gamma receptor IIb (FcγRIIb, CD32B) and a case report from concomitant use of imatinib and ibrutinib in a patient with combined CML and CLL (45,46). However, a study on Ba/F3 cells expressing BCR::ABL1 wild-type or p.(Thr315Ile) mutation found that BTK depletion did not affect viability or proliferation indicating that BTK is not essential for leukemogenesis (47). Consistent with our findings of a BTK downregulation at 0.5 and 2 µM imatinib, concentrations that reflect the observed imatinib plasma levels in patients undergoing therapy (48), decreased BTK levels were also shown in imatinib-resistant CML patients (49). In general, BTK appears to be relevant for TKI resistance and CML progression only in cases of a persistent CML, as present in our analyzed K-562 cell line model, but not in the development of the disease. Thus, the BTK downregulation in the imatinib-resistant cell line as presented here, may result in a deregulation of BTK-dependent signaling pathways that are likely involved in the development of imatinib resistance, such as Ras-Map-signaling, to circumvent the imatinib-mediated BCR::ABL1 inhibition. However, the BTK deregulation, but also the acquisition of BTK mutations in CML patients should be further analyzed in a clinical study to understand the influence on resistance.

Due to its function as a kinase and its role as an oncogene, it is generally assumed that BTK would be upregulated or gain-of-function mutations would be present in drug resistance. Accordingly, in previous studies on CLL and MCL, an upregulated BTK expression relative to non-malignant cells have been observed (20,50). However, we found that BTK downregulation is associated with CML progression and imatinib resistance. Our results contradict the common assumption that upregulated kinases, such as BTK, enhance tumor progression and their downregulation suppresses it. However, this discrepancy is consistent with the description of BTK as a context-dependent oncogene or tumor suppressor gene capable to either increase proliferation and cell survival or induce apoptosis and senescence, particularly by affecting p53-signaling (51). Therefore, BTK should be considered as a pleiotropic gene with opposing effects in cancer, acting as an oncogene especially in B cell malignancies, but also as a tumor suppressor in other neoplasms.

In conclusion, the present data show that BTK is involved in the development of imatinib resistance and progression of chronic myeloid leukemia in an in vitro-model. Further, the gain of the BTK c.1699_1700delinsAG p.(Glu567Arg) variant in imatinib-resistant cells further highlights the significance of BTK in imatinib resistance. These findings demonstrate that BTK is not only relevant in B cell malignancies, but may also be involved in other hematopoietic disorders, either as an oncogene or tumor suppressor gene, as well as a factor modulating treatment efficacy. Clinical investigations should confirm whether BTK inhibitors should be considered prior to their usage in imatinib-resistant chronic myeloid leukemia.

Supplementary Material

Supporting Data

Acknowledgements

The authors would like to thank Ms. Irina Naujoks, Ms. Kerstin Viertmann and Ms. Anna Jürgensen (Institute of Experimental and Clinical Pharmacology, Kiel, Germany) and Ms. Claudia Becher (Institute of Human Genetics, Kiel, Germany), for their technical assistance. The authors would like to also thank Dr. D. Langfeldt, Ms. Manuela Pendziwiat and Dr. B. Löscher (Institute of Clinical Molecular Biology, Kiel Germany) for their technical support. The authors would like to acknowledge the Institute of Clinical Molecular Biology in Kiel (Kiel, Germany) for providing Sanger sequencing as supported in part by the DFG Clusters of Excellence ‘Precision Medicine in Chronic Inflammation’ and ‘ROOTS’.

Funding

This study was funded by the Medical Faculty of the University of Kiel.

Availability of data and materials

The data generated in the present study may be requested re from the corresponding author. The exome sequencing and gene expression data in the present study may be found in the European Nucleotide Archive (ENA) repository under the accession number (PRJEB60565; http://www.ebi.ac.uk/ena/) and the GEO dataset accession number (GSE227347; http://www.ncbi.nlm.nih.gov/gds).

Authors' contributions

MK and IN conceptualized the study. MK designed the research. LS performed the experiments. LS, MK and IV analyzed the data. LS, IC, MK interpreted the data. LS and MK wrote the original draft. All authors read and approved the final version of the manuscript. MK and LS confirmed the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

Authors' information

Dr Inga Nagel, https://orcid.org/0000-0001-5174-4454; Professor Ingolf Cascorbi, https://orcid.org/0000-0002-2182-9534; Dr Meike Kaehler, https://orcid.org/0000-0002-2401-6037.

References

1 

Druker BJ, Tamura S, Buchdunger E, Ohno S, Segal GM, Fanning S, Zimmermann J and Lydon NB: Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med. 2:561–566. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Deininger MW, Goldman JM and Melo JV: The molecular biology of chronic myeloid leukemia. Blood. 96:3343–3356. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Quintás-Cardama A and Cortes J: Molecular biology of bcr-abl1-positive chronic myeloid leukemia. Blood. 113:1619–1630. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Milojkovic D and Apperley J: Mechanisms of resistance to imatinib and second-generation tyrosine inhibitors in chronic myeloid leukemia. Clin Cancer Res. 15:7519–7527. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Hochhaus A, Baccarani M, Silver RT, Schiffer C, Apperley JF, Cervantes F, Clark RE, Cortes JE, Deininger MW, Guilhot F, et al: European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 34:966–984. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Cortes J, Jabbour E, Kantarjian H, Yin CC, Shan J, O'Brien S, Garcia-Manero G, Giles F, Breeden M, Reeves N, et al: Dynamics of BCR-ABL kinase domain mutations in chronic myeloid leukemia after sequential treatment with multiple tyrosine kinase inhibitors. Blood. 110:4005–4011. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN and Sawyers CL: Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 293:876–880. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Jabbour E, Parikh SA, Kantarjian H and Cortes J: Chronic myeloid leukemia: Mechanisms of resistance and treatment. Hematol Oncol Clin North Am. 25:981–995. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Baccarani M, Deininger MW, Rosti G, Hochhaus A, Soverini S, Apperley JF, Cervantes F, Clark RE, Cortes JE, Guilhot F, et al: European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 122:872–884. 2013. View Article : Google Scholar : PubMed/NCBI

10 

O'Hare T, Zabriskie MS, Eiring AM and Deininger MW: Pushing the limits of targeted therapy in chronic myeloid leukaemia. Nat Rev Cancer. 12:513–526. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Braun TP, Eide CA and Druker BJ: Response and resistance to BCR-ABL1-targeted therapies. Cancer Cell. 37:530–542. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Kaehler M and Cascorbi I: Pharmacogenomics of impaired tyrosine kinase inhibitor response: Lessons learned from chronic myelogenous leukemia. Front Pharmacol. 12:6969602021. View Article : Google Scholar : PubMed/NCBI

13 

de Lavallade H and Kizilors A: The Importance of mutational analyses in chronic myeloid leukaemia for treatment choice. Eur Med J Oncol. 4:86–95. 2016.

14 

Cilloni D and Saglio G: Molecular pathways: BCR-ABL. Clin Cancer Res. 18:930–937. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Kaehler M, Litterst M, Kolarova J, Böhm R, Bruckmueller H, Ammerpohl O, Cascorbi I and Nagel I: Genome-wide expression and methylation analyses reveal aberrant cell adhesion signaling in tyrosine kinase inhibitor-resistant CML cells. Oncol Rep. 48:1442022. View Article : Google Scholar : PubMed/NCBI

16 

Kaehler M, Osteresch P, Künstner A, Vieth S, Esser D, Möller M, Busch H, Vater I, Spielmann M, Cascorbi I and Nagel I: Clonal evolution in tyrosine kinase inhibitor-resistance: Lessons from in vitro-models. Front Oncol. 13:12008972023. View Article : Google Scholar : PubMed/NCBI

17 

Wen T, Wang J, Shi Y, Qian H and Liu P: Inhibitors targeting Bruton's tyrosine kinase in cancers: Drug development advances. Leukemia. 35:312–332. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Pal Singh S, Dammeijer F and Hendriks RW: Role of Bruton's tyrosine kinase in B cells and malignancies. Mol Cancer. 17:572018. View Article : Google Scholar : PubMed/NCBI

19 

Mattsson PT, Vihinen M and Smith CI: X-linked agammaglobulinemia (XLA): A genetic tyrosine kinase (Btk) disease. Bioessays. 18:825–834. 1996. View Article : Google Scholar : PubMed/NCBI

20 

Hendriks RW, Yuvaraj S and Kil LP: Targeting Bruton's tyrosine kinase in B cell malignancies. Nat Rev Cancer. 14:219–232. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Ntanasis-Stathopoulos I, Gavriatopoulou M, Fotiou D and Dimopoulos MA: Current and novel BTK inhibitors in Waldenstrom's macroglobulinemia. Ther Adv Hematol. 12:20406207219895862021. View Article : Google Scholar : PubMed/NCBI

22 

Palma M, Mulder TA and Österborg A: BTK inhibitors in chronic lymphocytic leukemia: Biological activity and immune effects. Front Immunol. 12:6867682021. View Article : Google Scholar : PubMed/NCBI

23 

Bond DA, Alinari L and Maddocks K: Bruton tyrosine kinase inhibitors for the treatment of mantle cell lymphoma: Review of current evidence and future directions. Clin Adv Hematol Oncol. 17:223–233. 2019.PubMed/NCBI

24 

Byrd JC, Hillmen P, Ghia P, Kater AP, Chanan-Khan A, Furman RR, O'Brien S, Yenerel MN, Illés A, Kay N, et al: Acalabrutinib versus ibrutinib in previously treated chronic lymphocytic leukemia: Results of the first randomized phase III trial. J Clin Oncol. 39:3441–3452. 2021. View Article : Google Scholar : PubMed/NCBI

25 

Hillmen P, Eichhorst B, Brown JR, Lamanna N, O'Brien SM, Tam CS, Qiu L, Kazmierczak M, Zhou K, Šimkovič M, et al: Zanubrutinib versus ibrutinib in relapsed/refractory chronic lymphocytic leukemia and small lymphocytic lymphoma: Interim analysis of a randomized phase III trial. J Clin Oncol. 41:1035–1045. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Lozzio CB and Lozzio BB: Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood. 45:321–334. 1975. View Article : Google Scholar : PubMed/NCBI

27 

Kaehler M, Dworschak M, Rodin JP, Ruemenapp J, Vater I, Penas EMM, Liu C, Cascorbi I and Nagel I: ZFP36L1 plays an ambiguous role in the regulation of cell expansion and negatively regulates CDKN1A in chronic myeloid leukemia cells. Exp Hematol. 99:54–64.e7. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Kaehler M, Ruemenapp J, Gonnermann D, Nagel I, Bruhn O, Haenisch S, Ammerpohl O, Wesch D, Cascorbi I and Bruckmueller H: MicroRNA-212/ABCG2-axis contributes to development of imatinib-resistance in leukemic cells. Oncotarget. 8:92018–92031. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Waetzig V, Haeusgen W, Andres C, Frehse S, Reinecke K, Bruckmueller H, Boehm R, Herdegen T and Cascorbi I: Retinoic acid-induced survival effects in SH-SY5Y neuroblastoma cells. J Cell Biochem. 120:5974–5986. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Bruhn O, Lindsay M, Wiebel F, Kaehler M, Nagel I, Böhm R, Röder C and Cascorbi I: Alternative polyadenylation of ABC transporters of the C-family (ABCC1, ABCC2, ABCC3) and implications on posttranscriptional Micro-RNA regulation. Mol Pharmacol. 97:112–122. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Rentzsch P, Schubach M, Shendure J and Kircher M: CADD-Splice-improving genome-wide variant effect prediction using deep learning-derived splice scores. Genome Med. 13:312021. View Article : Google Scholar : PubMed/NCBI

33 

Ito M, Shichita T, Okada M, Komine R, Noguchi Y, Yoshimura A and Morita R: Bruton's tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun. 6:73602015. View Article : Google Scholar : PubMed/NCBI

34 

Uckun FM and Venkatachalam T: Targeting solid tumors with BTK inhibitors. Front Cell Dev Biol. 9:6504142021. View Article : Google Scholar : PubMed/NCBI

35 

Treon SP, Xu L, Guerrera ML, Jimenez C, Hunter ZR, Liu X, Demos M, Gustine J, Chan G, Munshi M, et al: Genomic landscape of waldenstrom macroglobulinemia and its impact on treatment strategies. J Clin Oncol. 38:1198–1208. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Smith CIE and Burger JA: Resistance mutations to BTK inhibitors originate from the NF-κB but not from the PI3K-RAS-MAPK Arm of the B cell receptor signaling pathway. Front Immunol. 12:6894722021. View Article : Google Scholar : PubMed/NCBI

37 

Ahn IE and Brown JR: Targeting Bruton's tyrosine kinase in CLL. Front Immunol. 12:6874582021. View Article : Google Scholar : PubMed/NCBI

38 

Chiron D, Di Liberto M, Martin P, Huang X, Sharman J, Blecua P, Mathew S, Vijay P, Eng K, Ali S, et al: Cell-cycle reprogramming for PI3K inhibition overrides a relapse-specific C481S BTK mutation revealed by longitudinal functional genomics in mantle cell lymphoma. Cancer Discov. 4:1022–1035. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Gu D, Tang H, Wu J, Li J and Miao Y: Targeting Bruton tyrosine kinase using non-covalent inhibitors in B cell malignancies. J Hematol Oncol. 14:402021. View Article : Google Scholar : PubMed/NCBI

40 

Väliaho J, Faisal I, Ortutay C, Smith CI and Vihinen M: Characterization of all possible single-nucleotide change caused amino acid substitutions in the kinase domain of Bruton tyrosine kinase. Hum Mutat. 36:638–647. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Speletas M, Kanariou M, Kanakoudi-Tsakalidou F, Papadopoulou Alataki E, Arvanitidis K, Pardali E, Constantopoulos A, Kartalis G, Vihinen M, Sideras P and Ritis K: Analysis of Btk mutations in patients with X-linked agammaglobulinaemia (XLA) and determination of carrier status in normal female relatives: A nationwide study of Btk deficiency in Greece. Scand J Immunol. 54:321–327. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Burger JA, Barr PM, Robak T, Owen C, Ghia P, Tedeschi A, Bairey O, Hillmen P, Coutre SE, Devereux S, et al: Long-term efficacy and safety of first-line ibrutinib treatment for patients with CLL/SLL: 5 years of follow-up from the phase 3 RESONATE-2 study. Leukemia. 34:787–798. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Broccoli A, Argnani L, Morigi A, Nanni L, Casadei B, Pellegrini C, Stefoni V and Zinzani PL: Long-term efficacy and safety of ibrutinib in the treatment of CLL patients: A real life experience. J Clin Med. 10:58452021. View Article : Google Scholar : PubMed/NCBI

44 

Hantschel O, Rix U, Schmidt U, Bürckstümmer T, Kneidinger M, Schütze G, Colinge J, Bennett KL, Ellmeier W, Valent P and Superti-Furga G: The Btk tyrosine kinase is a major target of the Bcr-Abl inhibitor dasatinib. Proc Natl Acad Sci USA. 104:13283–13288. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Parting O, Langer S, Kuepper MK, Wessling C, Li S, Braunschweig T, Chatain N, Maié T, Costa IG, Crysandt M, et al: Therapeutic inhibition of FcγRIIb signaling targets leukemic stem cells in chronic myeloid leukemia. Leukemia. 34:2635–2647. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Shea LK, Mikhail FM, Forero-Torres A and Davis RS: Concomitant imatinib and ibrutinib in a patient with chronic myelogenous leukemia and chronic lymphocytic leukemia. Clin Case Rep. 5:899–901. 2017. View Article : Google Scholar : PubMed/NCBI

47 

MacPartlin M, Smith AM, Druker BJ, Honigberg LA and Deininger MW: Bruton's tyrosine kinase is not essential for Bcr-Abl-mediated transformation of lymphoid or myeloid cells. Leukemia. 22:1354–1360. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Peng B, Lloyd P and Schran H: Clinical pharmacokinetics of imatinib. Clinl Pharmacokinet. 44:879–894. 2005. View Article : Google Scholar

49 

Villuendas R, Steegmann JL, Pollán M, Tracey L, Granda A, Fernández-Ruiz E, Casado LF, Martínez J, Martínez P, Lombardía L, et al: Identification of genes involved in imatinib resistance in CML: A gene-expression profiling approach. Leukemia. 20:1047–1054. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Zhu S, Gokhale S, Jung J, Spirollari E, Tsai J, Arceo J, Wu BW, Victor E and Xie P: Multifaceted immunomodulatory effects of the BTK inhibitors ibrutinib and acalabrutinib on different immune cell subsets-beyond B lymphocytes. Front Cell Dev Biol. 9:7275312021. View Article : Google Scholar : PubMed/NCBI

51 

Rada M, Barlev N and Macip S: BTK: A two-faced effector in cancer and tumour suppression. Cell Death Dis. 9:10642018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 28 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Schmidlechner L, Nagel I, Vater I, Cascorbi I and Kaehler M: BTK acts as a modulator of the response to imatinib in chronic myeloid leukemia. Oncol Lett 28: 424, 2024
APA
Schmidlechner, L., Nagel, I., Vater, I., Cascorbi, I., & Kaehler, M. (2024). BTK acts as a modulator of the response to imatinib in chronic myeloid leukemia. Oncology Letters, 28, 424. https://doi.org/10.3892/ol.2024.14557
MLA
Schmidlechner, L., Nagel, I., Vater, I., Cascorbi, I., Kaehler, M."BTK acts as a modulator of the response to imatinib in chronic myeloid leukemia". Oncology Letters 28.3 (2024): 424.
Chicago
Schmidlechner, L., Nagel, I., Vater, I., Cascorbi, I., Kaehler, M."BTK acts as a modulator of the response to imatinib in chronic myeloid leukemia". Oncology Letters 28, no. 3 (2024): 424. https://doi.org/10.3892/ol.2024.14557