1. Introduction
The expression of the BCR–ABL1 fusion protein is a hallmark of Ph
+ ALL, which affects around one-third of adult ALL cases and ~3–5% of pediatric ALL cases [
1]. The oncogenic fusion of the breakpoint cluster region (BCR) at chromosome 22 and the tyrosine-protein kinase ABL1 at chromosome 9 results from a reciprocal chromosomal translocation t(9;22)(q34;q11), which leads to constitutive tyrosine kinase activation [
2,
3]. The prognosis of the disease was very poor until the advent of tyrosine kinase inhibitors (TKIs) such as Imatinib mesylate [
4,
5,
6]. The current treatment regimen for Ph
+ ALL generally involves chemotherapy in combination with TKI, followed by allogeneic hematopoietic stem cell transplantation (allo-HCT) [
7]. In some cases, the combination of TKI with chemotherapy can achieve long-term remission without allo-HCT. Although the inclusion of TKIs in frontline therapy has revolutionized the outcome of the disease, the emergence of TKI resistance through acquired mutation(s) in the ABL kinase domain poses a significant threat to the prognosis of the disease. Some of these mutations, e.g., T315I, can now be targeted by the advanced third-generation TKI inhibitor Ponatinib [
8]. However, the toxicity associated with some of the advanced therapeutics as well as their limited access in many of the clinics worldwide make it challenging for clinicians, especially those in resource-constrained situations [
7]. This therefore necessitates the continuous search for new, improved inhibitors as single or combinatorial therapeutics [
9].
Previously, our laboratory showed that the G-protein coupled chemokine receptor CXCR4 promotes the survival of BCR–ABL1-transformed mouse B cells [
10]. Deletion of CXCR4 results in rapid cell death and the complete absence of colony formation in vitro by these cells. Mechanistically, CXCR4 associates with the interleukin 7 receptor (IL7R) on the surface of BCR–ABL1 cells. This leads to the recruitment of IL7R-associated proteins such as Janus kinase 3 (JAK3) in close proximity to CXCR4, thereby activating the JAK-STAT pathway and stimulating cell survival. The hyper-phosphorylation of JAK1-3 is observed in BCR–ABL1-transformed cells, which is reduced by the inducible deletion of CXCR4 in these cells. Thus, the association between these two receptors is a prerequirement for BCR–ABL- induced cell transformation [
10].
The above observation raises the question as to whether, similar to deletion, the inhibition of CXCR4 also exerts an inhibitory effect on BCR–ABL1-transformed cells. Several small-molecule and peptide inhibitors are available for CXCR4, of which only AMD3100 (Plerixafor) and BL-8040 are FDA-approved drugs, used in stem cell transplantation [
11,
12]. Owing to the unfavorable side effects of some of these drugs, there is an urgent need to develop novel CXCR4 antagonists. The endogenous peptide inhibitor of CXCR4 (EPI-X4), a 16 mer peptide derived from human serum albumin, exhibits specific binding to CXCR4 and the subsequent blocking of CXCL12-mediated CXCR4 activation [
13]. To improve its efficacy and plasma stability, several derivatives of EPI-X4 have been designed using quantitative structure–activity relationship (QSAR) studies [
14,
15]. For instance, activity-improved derivative EPI-X4 JM#21 has shown therapeutic efficacies in mouse models of inflammatory diseases [
14] as well as in oncologic applications [
16]. To further improve the stability of optimized EPI-X4 derivatives, they have been conjugated to long-chain fatty acids, leading to increased serum albumin affinity and, thus, to an improved circulation half-life [
17]. Previous work from our laboratory have shown that most of these advanced EPI-X4 derivatives potentially block CXCL12-mediated intracellular calcium release in BCR–ABL1-transformed cells in the nanomolar range [
14,
17]. However, the effect of these advanced derivatives on the survival of BCR–ABL1 cells has not yet been investigated.
Thus, in the current study, we tested an array of advanced EPI-X4 derivatives for their efficacy in inhibiting BCR–ABL1 cell growth and compared the effect with those of the small-molecule CXCR4 antagonist AMD3100 and the ABL1 TKI Imatinib. We also explored the signaling cascade alterations induced by these inhibitors and tested if the combination of the EPI-X4 derivative with Imatinib exerted a superior cell killing effect. Furthermore, we tested whether the inhibitors would be effective in human ALL cell lines and primary xenograft cells to evaluate their therapeutic potential.
3. Discussion
The seven-transmembrane G-protein-coupled chemokine receptor CXCR4 plays an essential role in the homing and retention of tumor cells in the protective bone marrow niches, thereby preventing their culmination by therapeutic agents and increasing the risk of disease relapses [
21]. The elevated expression of CXCR4 in hematological malignancies as well as in solid tumors is associated with poor prognosis. Consequently, targeting CXCR4 activation through antagonists leads to the egress of hiding cancer cells from bone marrow to the periphery, a process known as chemosensitization, which therefore possesses great therapeutic potential. Several small molecules, peptides, neutralizing antibodies, as well as antibody–drug conjugates are being tested for their efficacy in antagonizing CXCR4 function in different cancers. For instance, the small-molecule inhibitor AMD3100, the peptide-based inhibitors BL-8040 and LY2510924, and anti-CXCR4 monoclonal antibodies have been used in several clinical trials for patients with acute myeloid leukemia (AML) and ALL, both as monotherapies and in combination with chemotherapy (as reviewed in [
22]). Although the trials have demonstrated that targeting the CXCR4 axis in vivo is feasible and safe, the clinical outcome in many cases does not prove to be beneficial compared with that of chemotherapy alone. This therefore warrants new improved CXCR4 antagonists.
Ph
+ B-ALL represents a group of ALL cases that expresses the oncogenic BCR–ABL1 fusion protein. Although the invention of Imatinib, a tyrosine kinase blocker of BCR–ABL1, greatly improved the outcome of the disease, resistance to Imatinib is a major problem that can lead to relapse. It is therefore crucial to identify other contributing pathways that can be additionally targeted in this disease. A previous study from our laboratory identified both CXCR4 and IL7R pathways as contributing to the pathology of BCR–ABL1 [
10]. The complete deletion of CXCR4 was found to be sufficient to block BCR–ABL1 cell growth in vitro. Therefore, the aim of the current work was to study the effect of CXCR4 inhibition on BCR–ABL1-mediated transformation.
EPI-X4, a 16-amino-acid peptide derived from the proteolysis of serum albumin, is a naturally occurring CXCR4 antagonist that prevents CXCR4-tropic HIV-1 virus entry [
13]. Using a molecular docking study to predict the EPI-X4 binding sites to CXCR4 and QSAR to identify modifications that might enhance the binding and functional efficacy of EPI-X4, several optimized EPI-X4 derivatives were generated [
14]. Many of these derivatives effectively block the CXCL12-induced calcium mobilization in BCR–ABL1-transformed mouse B cells, showing much higher efficacy than parental EPI-X4, indicating that they could inhibit CXCR4 signaling in these cells [
14,
17].
In the current manuscript, we presented data that the inhibition of CXCR4 by a lipid-conjugated truncated version of EPI-X4, namely JM#170, induced rapid and strong cell death in BCR–ABL1-transformed malignant mouse cells, whereas it remained inactive against the WT B cells. Live cell imaging of BCR–ABL1 cells in the presence of different inhibitors clearly indicated that JM#170 potently blocked cellular growth within 24 h of treatment, while AMD3100 showed an effect at a much later time point (~60 h post treatment,
Figure 1A–C). Notably, for cells treated with JM#170, a slight increase in GFP
+ cell accumulation was observed 72 and 96 h post treatment (
Figure 1A). This could be explained by the relatively lower stability of peptide-based inhibitors in serum containing cell culture media. Although the optimized derivatives are significantly more stable in blood plasma than the mother peptide EPI-X4 [
17], 72–96 h could still be long. The cells that remained resistant to JM#170 until ~48 h may have started proliferating after the degradation of the inhibitor, which in turn increased the GFP
+ cell density. Interestingly, neither JM#170 nor AMD3100 had any growth inhibitory effect on untransformed bone-marrow-derived B cells (
Figure 2 and
Supplementary Figure S3C). This may have arisen from the relatively high rate of cell proliferation observed in the transformed cells compared to the WT cells, which was clearly evident from the growth curve of the two cell types in live cell imaging (
Figure 1B black line vs.
Figure 2B black line). As the requirement for CXCR4 signaling for survival and proliferation of transformed cells is higher than that of normal cells, the effect of CXCR4 inhibition is also stronger on these cells than on normal cells. This selectivity is a very important aspect for JM#170 to be potentially used as an anticancer peptide drug.
Although both JM#170 and AMD3100 blocked BCR–ABL1 cell growth (
Figure 1A–C), as well as CXCL12-induced AKT and ERK1/2 phosphorylation (
Figure 3B,C), the end effect was different for the two inhibitors. While JM#170 effectively killed BCR–ABL1 cells, AMD3100 halted the proliferation, with no cell killing effect observed within the experimental timeline (
Figure 1E–G). This clearly indicates that JM#170 has a different mechanism of action other than blocking ligand-induced CXCR4 activation in BCR–ABL1-transformed cells. Indeed, we observed that JM#170 spontaneously induced transient JNK1/2 phosphorylation (
Figure 4A,B), leading to the enhanced expression of JNK-downstream genes such as
cJun,
Bax, and
Bim (
Figure 4E,F) and triggered the intrinsic apoptotic pathway in BCR–ABL1-expressing cells (
Figure 4C,D). JNK, also known as stress-activated protein kinase (SAPK), is activated upon various kinds of cellular stress, including treatment with drugs or signaling pathway inhibitors; depending on the context and specific cell types, it can act as both a pro-survival as well as a pro-apoptotic signaling cascade [
20]. In this case, treatment with the inhibitor JM#170 exerted cellular stress, thereby activating the JNK-mediated pro-apoptotic pathway.
The strong cytotoxic effect exerted by JM#170 on BCR–ABL1 cells prompted us to check if it could be used in combination with Imatinib, specifically with a lower dose of Imatinib. This would be particularly interesting in terms of reducing the side effects of Imatinib. Co-treatment of JM#170 at 10 μM with Imatinib at 100 nM (10-fold lower than usual) induced a complete block of BCR–ABL1 cell growth, which was even stronger than that achieved with JM#170 alone (
Figure 5A,C,D). Another 10-fold reduction in Imatinib concentration (10 nM) did not impart any effect on BCR–ABL1 cell growth; the co-treatment yielded an effect similar in magnitude to that of JM#170 alone (
Figure 5A,B). This suggests that the combinatorial treatment of JM#170 and Imatinib could be very effective against BCR–ABL1-induced transformation, which not only allows the lowering of Imatinib doses but might also be helpful for Imatinib-resistant cases.
In addition to its effect on BCR–ABL1-transformed mouse B cells, JM#170 was also active against human BCR–ABL1
+ ALL cell line SupB15. Although JM#170 blocked the CXCL12-induced calcium flux in both SupB15 and 697 cell lines (human BCR–ABL1
− ALL cell line,
Figure 6A–D), the killing effect was only observed in BCR–ABL1
+ SupB15 cells (
Figure 6E,
Supplementary Figure S5). This suggests that the presence of BCR–ABL1 is essential for the inhibitory action of JM#170. It is important to mention that the level of CXCR4 expression on the surface of 697 cells was very high compared to that on other cells, such as SupB15 or healthy human-blood-derived B cells (
Supplementary Figure S4A). It is therefore possible that the maximum concentration of JM#170 used here (10 μM), though sufficient to block ligand-induced calcium mobilization, was not enough to impart an ultimate effect on the survival of these cells. Additionally, as pointed out before, the magnitude of the inhibitory effect observed in the SupB15 cell line was lower compared to that of BCR–ABL1-transformed mouse cells. This indicates the robust nature of the ALL cell lines: in addition to BCR–ABL1 transformation, there might be additional growth-promoting and death-evading mechanisms making these cell lines difficult to target. The mouse cells, on the other hand, with a single transformation event, could be highly susceptible, as observed in this study.
Our experiment with the xenografted primary Ph+ ALL cells also indicated an important aspect of the inhibitory effect of JM#170 (
Supplementary Figure S6A). The relative growth inhibition of the xenografted cells by JM#170 was much milder compared to its effect on BCR–ABL1-transformed cells or the human ALL cell line. This, in our view, is due to the lack of proliferation ability of these cells under in vitro conditions. As revealed from their growth curve, the xenografted cells could survive in culture but never proliferated (
Supplementary Figure S6A). Since CXCR4 is more involved in proliferating cells than in resting ones, the effect of JM#170 may be milder in resting cells, leading to subtle growth inhibition.
In conclusion, we provide evidence that JM#170, a lipid-modified EPI-X4-derived CXCR4 antagonist, is a potent inhibitor of BCR–ABL1-positive ALL cell growth in vitro. An interesting question would be why, among all the derivatives, JM#170 was found to be the most active. Among the derivatives tested in this study, JM#170 has the lowest molecular weight (
Table 1). It could be possible that this size is optimum for fitting into the binding pocket of CXCR4, thereby preventing ligand-induced activation. In addition, such tight interaction might trigger non-classical signal pathway activation such as JNK as observed here. Further structural studies might shed light on this finding. As the next step for therapeutic application, the effect of JM#170 should be investigated using in vivo ALL models.
4. Materials and Method
4.1. Reagents and Optimized EPI-X4 Derivatives
Optimized EPI-X4 peptide derivatives (
Table 1) were synthesized as described previously [
17]. Briefly, the peptides were synthesized via standard Fmoc solid-phase peptide synthesis using a Liberty Blue microwave synthesizer (CEM Corporation, Matthews, NC, USA) and then purified using reversed-phase high-performance liquid chromatography (Waters, Milford, MA, USA), employing an acetonitrile/water gradient under acidic conditions on a Phenomenex C18 Luna column (particle size 5 µm, pore size 100 Å). Purified peptides were lyophilized on a freeze-dryer (Labconco, Kansas City, MI, USA), and the molecular mass was verified by liquid chromatography–mass spectrometry (LC-MS; Waters, Milford, MA, USA). The peptides were dissolved in dimethyl sulfoxide (DMSO, Sigma-Aldrich, Hamburg, Germany) at a stock concentration of 3 mM and further diluted in phosphate-buffered saline (PBS) /complete cell culture media before usage. AMD3100 octahydrochloride hydrate (#A5602) and Imatinib mesylate (#SML1027) were purchased from Sigma-Aldrich, Hamburg, Germany and dissolved in H
2O and DMSO respectively to create a 10 mM stock. Human and mouse CXCL12 were purchased from Peprotech, Hamburg, Germany (#300-28A, #250-20A) and dissolved at 100 μg/mL in H
2O.
4.2. Cell Culture
BCR–ABL1-transformed mouse pre-B cells were generated in-house, as described previously [
10]. Briefly, wild-type (WT) mouse bone marrow B cells were cultured in Iscove’s modified Dulbecco’s media (Sigma- Aldrich, Hamburg, Germany) supplemented with 10% heat-inactivated fetal bovine serum (FBS, PAN Biotech, Aidenbach, Germany), 2 mM L-glutamine (Gibco, Dreieich, Germany), 100 units/mL of penicillin/streptomycin (Gibco), 50 μM beta-marcaptoethanol (Gibco), and 1 ng/mL of recombinant mouse interleukin 7 (IL7, Immunotools, Friesoythe, Germany) for 4–7 days at 37 °C in a 7.5% CO
2 incubator. The cells were then retrovirally transduced with a BCR–ABL1 construct cloned in pMIG vector containing an EGFP marker. Two days after transduction, cells were tested for GFP expression via FACS, and positive cells were selected via IL7 withdrawal from culture medium. BCR–ABL1 transduction transforms the cell, leading to uncontrolled growth and eventually enriching the culture.
Rag2−/− λ5−/− Spl65−/− TKO mouse pro-B cells were cultured in similar IL7 containing Iscove’s media at 37 °C in a 7.5% CO
2 incubator and transduced with either BCR–ABL1 construct (TKO-BCR–ABL1) or empty pMIG vector containing an EGFP marker (TKO-EV). Selection and enrichment of TKO-BCR–ABL1 cells were performed as stated before. Human ALL cell lines SupB15 (ACC 389), Tom1 (ACC 578), SD1 (ACC 366), and 697 (ACC 42) were purchased from DSMZ. SupB15 cells were maintained in McCoy’s 5A (Biowest, Nuaillé, France) and the rest of the cells in RPMI media (Gibco), supplemented with 20% heat-inactivated FBS, 2 mM L-glutamine, and 100 units/mL of penicillin/streptomycin at 37 °C in a 5% CO
2 incubator.
4.3. Isolation of Ph+ ALL Cells Form Xenografted NSG Mice
Xenografting of immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice with an Imatinib-resistant ALL sample from a patient was described in [
10]. Animals showing clinical symptoms of leukemia or with >75% leukemic blasts in blood were sacrificed, and total splenic cells were isolated and frozen for future use.
4.4. Real-Time Cell Growth Analysis by IncuCyte
A flat-bottom 96-well plate was coated with 0.01% poly-L ornithine (#P4957, Sigma-Aldrich, Hamburg, Germany) for 1 h at room temperature (RT). BCR–ABL1, SupB15, and 697 cells were counted as 0.5 × 105 cells/well, while TKO-BCR–ABL1 and TKO-EV cells as 1 × 105 cells/well, which were labeled with IncuCyte Cytotox Red dye (#4632, Sartorius, Ulm, Germany) as per the manufacturer’s protocol. The cells were plated in 100 μL complete medium. The solvent control DMSO, peptide inhibitors, AMD3100 or Imatinib were diluted in complete medium at 2X concentration and mixed with the cells in a 1:1 ratio. Plate was prewarmed at 37 °C for 30 min prior to placing in the IncuCyte chamber. Scanning was conducted every two hours for total of 4 days. Images were acquired using a 20X objective in phase contrast and fluorescence mode at 4 images/well and analyzed using the cell-by-cell analysis module of IncuCyte software version 2020B (Essen Bioscience, Ann Arbor, MI, USA).
For Ph+ ALL cells isolated from the xenografted NSG mice, frozen cells were thawed and cultured in MEM-alpha (Gibco) media supplemented with 20% FBS and 100 units/mL of penicillin/streptomycin for 5–16 h in a 5% CO2 incubator. Next, the cells were overlaid on 10 mL Ficoll-Paque™PLUS (#17144003, Cytiva, Freiburg, Germany) in 50 mL tubes and centrifuged at 400 g without break for 10 min at RT. The cells were collected from the interface of Ficoll and media and washed once with PBS. Viable cells were counted and plated into previously coated 48-well plates at 4.25 × 105 cells/well in 250 μL of media with or without the inhibitors. Scanning was conducted every two hours for total of 6 days in phase contrast mode and analyzed using the cell-by-cell analysis module of IncuCyte software version 2020B (Essen Bioscience).
4.5. Flow Cytometry
Cells treated with either solvent control DMSO or different concentrations of inhibitors for different time points were collected and washed with ice-cold PBS. Cells were then stained with efluor 450-labeled fixable viability dye (FVD, #65-0863-14, eBioscience, San Diego, CA, USA) at 1:1000 dilution at 4 °C or using 50 μg/mL propidium iodide (PI, #P3566, Thermo Fisher, Dreieich, Germany) solution at RT for 15 min in the dark. After washing, labeled cells were resuspended in 100 μL of PBS and acquired using LSR-Fortessa (BD Bioscience, Heidelberg, Germany). FACS data were analyzed with FlowJo software version 10.0.
To determine the expression of CXCR4 on the cell surfaces, 0.5 × 106 BCR–ABL1 and TKO-EV cells were collected, washed with FACS buffer (PBS containing 3% FBS), and stained with a 1:100 dilution of BV421-labeled anti-mouse CXCR4 antibody (#146511, Biolegend, San Deago, CA, USA). Human ALL cell lines were similarly collected and stained with a 1:100 dilution of Alexa flour 488-labeled antihuman CD19 (#53-0199-42, Invitrogen, Dreieich, Germany) and APC-Cy7-labeled anti-human CXCR4 (#306527, Biolegend) antibodies at 4 °C for 15 min in the dark. Cells were then washed with FACS buffer and analyzed using LSR-Fortessa (BD Bioscience).
Human peripheral blood mononuclear cells (PBMCs) were isolated using Ficoll-Paque™PLUS density gradient centrifugation as per the manufacturer’s instructions. Mononuclear cells were collected from the interface of plasma and Ficoll–Paque media, then washed several times with PBS. Cells were then counted and either used directly for experimentation or frozen in 10% DMSO-containing media for future use.
4.6. Cell Proliferation Assay
BCR–ABL1 cells were collected at 0.5 × 105 cells/well of a 96-well plate and labeled with cell trace far red cell proliferation dye (#C34564, Invitrogen, Dreieich, Germany), according to the manufacturer’s instructions. Labeled cells were plated in 100 μL of complete media and were mixed with equal volume of 2X DMSO or inhibitor-containing media. A small part of the labeled cells was measured in FACS to determine the fluorescence signal on day 0. After specific time periods (48 and 72 h), cells were collected, washed with FACS buffer, and analyzed using LSR-Fortessa (BD Bioscience).
4.7. Anti-CXCR4 Antibody Competition Assay
An anti-CXCR4 competition assay was conducted as previously described [
14,
17]. Briefly, 1 × 10
5 BCR–ABL1-transformed mouse B cells were incubated with serially diluted (10 μM to 1 nM) JM#170, AMD3100, and Imatinib in the presence of a fixed concentration of BV421-labeled antimouse CXCR4 antibody (clone L276F12, #146511, Biolegend) for 2 h at 4 °C. Afterward, the cells were washed to remove unbound inhibitors and antibody, and measured via FACS for the fluorescence signal of the bound antibody. The fluorescence signal corresponding to only antibody (no inhibitor) was considered to be 100% antibody binding. For SupB15 cells, a similar experiment was carried out using APC-Cy7-labeled anti-human CXCR4 antibody (clone 12G5, #306527, Biolegend).
4.8. Calcium Flux Analysis
Ca
2+ signaling was analyzed as described previously [
14]. Briefly, SupB15 and 697 cells were collected at 1 × 10
6 cells/treatment and loaded with calcium-sensitive dye Indo-1 AM (#I1223, Invitrogen) and 0.5 mg/mL of pluronic F-127 (#P3000MP, Invitrogen) in their respective media supplemented with 1% FBS at 37 °C for 45 min. Cells were then washed and treated with the inhibitors for 10 min at 37 °C. Baseline signal for calcium was measured for 30 s via flow cytometry, followed by stimulation with 100 ng/mL human CXCL12. The area under the curve (AUC) of each calcium flux plot was determined using FlowJo. The AUC of the water control (solvent for CXCL12) was subtracted from each treatment to obtain the correct estimation of the calcium signal upon CXCL12 stimulation.
4.9. Western Blotting
BCR–ABL1 cells were collected as 5 × 106 cells/500 μL complete media and treated either with DMSO or different inhibitors in the absence and presence of 100 ng/mL mouse CXCL12 for 5, 15, and 30 min at 37 °C. Cells were then immediately transferred onto ice to stop the reaction and washed with ice-cold PBS. Cell extract was prepared by resuspending the cell pellet in 100 μL of radio-immunoprecipitation assay (RIPA) buffer supplemented with 1X protease inhibitor cocktail (#78429, Thermo Fisher), 1 mM sodium orthovanadate (Sigma-Aldrich), and 10 mM beta-glycerophosphate (Sigma-Aldrich). For each treatment, 15 μL of cell extract was mixed with 5 μL of 4X Laemmli buffer (reducing), boiled at 95 °C for 10 min, and run on 10% SDS-PAGE. Gels were wet-transferred to PVDF membranes (Millipore, Burlington, MA, USA) and stained with 0.1% ponceau-S solution to check for efficient blotting. The membranes were blocked with 3% BSA solution and incubated with primary antibodies at 4 °C overnight. The next day, primary antibodies were washed, and membranes were probed with horseradish peroxidase (HRP)-conjugated secondary antibodies at RT for 2 h. After washing, the blots were developed using Immobilon ECL Ultra Western HRP Substrate (#WBULS0500, Millipore), and images were acquired with a Fusion SL gel imaging system (Vilber Lourmat, Eberhardzell, Germany). The following primary antibodies from Cell Signaling were used: anti-Phospho-Akt (Ser473, #4060), anti-AKT (#9272), anti-Phospho-p44/42 MAPK (Thr202/Tyr204, #4370), anti-p44/42 MAPK (137F5, #4695), anti-Phospho-SAPK/JNK (Thr183/Tyr185) (81E11, #4668), anti-SAPK/JNK (#9252), and anti-GAPDH (14C10, #2118) antibody. Anti-rabbit IgG, HRP-linked (#7074, Cell Signaling technology, Danvers, MA, USA) was used as secondary antibody.
4.10. Caspase 3/7 and Caspase 8 Activation Assay
BCR–ABL1 cells were plated at 0.5 × 105 cells/well of a 96-well plate in 100 μL of complete media and treated with DMSO or various inhibitors. At specific time points (24, 48 and 72 h), activation of caspases was measured with a Caspase-Glo® 3/7 Assay (#G8090) and Caspase-Glo® 8 Assay (#G8200, Promega, Madison, WI, USA) following the manufacturer’s instructions.
4.11. RNA Isolation and Real-Time Gene Expression Analysis
BCR–ABL1 cells were plated at 1 × 106 cells/well of a 12-well plate in 1 mL of complete media and treated with DMSO or various inhibitors. After specific time points (24 and 48 h), RNA was isolated from cells using a ReliaPrep Cell RNA miniprep kit (#Z6011, Promega). Fifty nanograms of total RNA was reverse-transcribed using a high-capacity RNA to cDNA kit (#4387406, Thermo Fisher), and gene expression was measured via Taqman assay using Taqman Gene expression master mix (#4369016, Thermo Fisher) following ghd manufacturer’s protocol. The following Taqman probes were used: Jun (Mm07296811_s1), Bax (Mm00432051_m1), Bim (Mm00437796_m1), Bcl2 (Mm00477631_m1), Bcl-xl (00437783_m1), FasL (Mm00438864_m1), and beta actin (04394036_g1).
4.12. Statistical Analysis
Statistical analysis was performed using GraphPad Prism 6.0 software. Specific statistical tests are mentioned in the figure legends. Unless otherwise mentioned, all experiments were independently conducted at least three times.