The redeployed drug combination of bezafibrate and medroxyprogesterone acetate (designated BaP) has potent in vivo anticancer activity in acute myelogenous leukemia (AML) and endemic Burkitt lymphoma (eBL) patients; however, its mechanism-of-action is unclear. Given that elevated fatty acid biosynthesis is a hallmark of many cancers and that these drugs can affect lipid metabolism, we hypothesized that BaP exerts anticancer effects by disrupting lipogenesis. We applied mass spectrometry–based lipidomics and gene and protein expression measurements of key lipogenic enzymes [acetyl CoA carboxylase 1 (ACC1), fatty acid synthase (FASN), and stearoyl CoA desaturase 1 (SCD1)] to AML and eBL cell lines treated with BaP. BaP treatment decreased fatty acid and phospholipid biosynthesis from 13C D-glucose. The proportion of phospholipid species with saturated and monounsaturated acyl chains was also decreased after treatment, whereas those with polyunsaturated chains increased. BaP decreased SCD1 protein levels in each cell line (0.46- to 0.62-fold; P < 0.023) and decreased FASN protein levels across all cell lines (0.87-fold decrease; P = 1.7 × 10−4). Changes to ACC1 protein levels were mostly insignificant. Supplementation with the SCD1 enzymatic product, oleate, rescued AML and e-BL cells from BaP cell killing and decreased levels of BaP-induced reactive oxygen species, whereas supplementation with the SCD1 substrate (and FASN product), palmitate, did not rescue cells. In conclusion, these data suggest that the critical anticancer actions of BaP are decreases in SCD1 levels and monounsaturated fatty acid synthesis. To our knowledge, this is the first time that clinically available antileukemic and antilymphoma drugs targeting SCD1 have been reported. Cancer Res; 75(12); 2530–40. ©2015 AACR.

Acute myelogenous leukemia (AML) and endemic Burkitt lymphoma (eBL) are two aggressive and biologically very distinct blood cancers that kill patients quickly if untreated. AML mainly affects elderly patients, who often cannot tolerate routine high-grade cytotoxic chemotherapy treatment (1, 2). eBL is a B-cell non-Hodgkin lymphoma predominantly affecting children in equatorial Africa. It responds well to high-grade chemotherapy; however, the high costs of treatment and supportive care often preclude their use in the endemic setting (3). Thus, in elderly AML patients in high-income countries and eBL patients in low-income countries, there is an urgent need for inexpensive treatments that selectively kill tumor cells with low systemic toxicity. One approach is drug redeployment where existing drugs with low toxicity are screened for efficacy against cancers (4). This approach has already been successful in reducing mortality in hematologic malignancies (5, 6). Using this method, we previously identified that the combination of the lipid lowering drug, bezafibrate, and the contraceptive, medroxyprogesterone acetate (MPA), has potent and cancer cell selective in vitro anticancer effects against AML, eBL, chronic lymphocytic leukemia, and B-cell non-Hodgkin lymphoma cells (7–10). The potency of this combination (denoted BaP) extended to the clinic demonstrating efficacy with low toxicity in clinical trials in AML (trial registration number ISRCTN50635541; ref. 11) and eBL patients (ISRCTN34303497; ref. 12). In vitro, BaP induces intracellular reactive oxygen species (ROS) in tumor cells, which is thought to be important for cell killing (8–10); however, as yet, we do not have a complete mechanism that fully explains the action of BaP against these diverse hematologic malignancies.

Both bezafibrate and MPA individually alter lipid metabolism (13, 14); however, nothing is known about their combinatorial effect on lipogenesis. We hypothesized that regulation of lipid metabolism could be an important component of BaP anticancer activity given that abnormally rapid lipogenesis is considered to be a hallmark of most cancers (15, 16). Elevated lipogenesis in tumor cells is at least partly caused by high rates of fatty acid biosynthesis (15–18), a phenomenon also observed in hematologic malignancies (19, 20). Enzymes that synthesize saturated fatty acids [acetyl CoA carboxylase 1 (ACC1) and fatty acid synthase (FASN)] and monounsaturated fatty acids [stearoyl CoA desaturase 1 (SCD1)] are often overexpressed in malignant cells, and selective inhibition of these enzymes exerts an anticancer effect in vitro (18, 20–24). Accelerated fatty acid synthesis may permit faster growth of tumor cells. Furthermore, synthesis of monounsaturated fatty acids may have additional benefits as it has been shown that oleate can protect cells against saturated fatty acid toxicity and cellular stress (25, 26). FASN, ACC1, and SCD1 have therefore been identified as plausible targets for cancer therapy, but agents shown to decrease their activities in the laboratory have yet to be translated to the clinic.

Here, we have investigated the effect of BaP on lipid metabolism in AML- and eBL-derived cell lines by measuring changes to the lipidome and lipid synthesis from 13C-labeled D-glucose using direct-infusion Fourier-transform ion cyclotron resonance (FT-ICR) mass spectrometry. We also measured mRNA and protein levels of key lipogenic enzymes. We show that BaP slows de novo fatty acid and phospholipid biosynthesis, which appears to be achieved by downregulation of lipogenic enzymes, particularly SCD1, at the gene and protein levels. External provision of SCD1 product oleate rescued both AML and eBL cell lines from killing by BaP. This suggests that the dysregulation of lipogenesis is a key component of the anticancer action of BaP.

Cell culture

AML (K562 and HL60; European Collection of Cell Cultures) and Burkitt lymphoma (BL31 and Glor; gift from A.B. Rickinson, University of Birmingham, Birmingham, United Kingdom) cell lines were maintained in exponential growth (between 0.25 and 1.5 × 106 cells/mL) at 37°C with 5% CO2 in RPMI-1640 medium (containing 2 g/L D-glucose) supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin, and 100 μg/mL of streptomycin (Gibco-Invitrogen).

Drug treatments

Unless specified, cells were seeded at 2.5 × 105 cells/mL (AML) or 5 × 105 cells/mL (BL) in flasks (Falcon) and treated for 24 hours with either: BaP (0.5 mmol/L bezafibrate and 5 μmol/L MPA; Sigma-Aldrich); 0.5 mmol/L bezafibrate; 5 μmol/L MPA; or solvent control (ethanol and DMSO). DMSO and ethanol was at a final concentration of 1 in 1,000 in all classes. The 13C D-glucose tracer study was set in glucose-free RPMI-1640 (Gibco-Invitrogen) supplemented with 2 g/L 1-13C d-glucose (Sigma-Aldrich). Where indicated, HL60 cells were treated with a commercial SCD1 inhibitor (4-(2-Chlorophenoxy)-N-[3-[(methylamino)carconyl]phenyl]-1-piperidinecarboxamide; ab142089; Abcam; 10 nmol/L).

Metabolomics/lipidomics cell quenching and extraction

Cell quenching (to rapidly halt metabolic activity) was done as described (27) with minor modifications. Cell suspensions were centrifuged (250 × g, 5 minutes, 20°C) and supernatant was discarded. Cell pellets were resuspended in residual media, of which 200 μL was added to 1.6-mL 60% methanol on dry ice (−40°C) and then centrifuged (2,500 × g, 5 minutes, −9°C). Supernatant was discarded and cell pellet weighed and transferred to a −80°C freezer until extraction. Polar metabolites and lipids were extracted from cell pellets using a modified Bligh-Dyer procedure (28, 29) using 8-μL methanol/mg biomass, 8-μL chloroform/mg biomass, and 7.2-μL water/mg biomass. The polar phase (100 μL) was dried in a centrifugal concentrator and the nonpolar phase (100 μL) was dried under nitrogen. Both were kept at −80°C until analysis.

Mass spectrometry analysis and data processing

Dried samples were resuspended in either: 50 μL 80:20 methanol:water with 20 mmol/L ammonium acetate (polar extracts), or 200 μL 2:1 methanol:chloroform with 5 mmol/L ammonium acetate (nonpolar extracts). All analyses were conducted by direct infusion mass spectrometry (DIMS), using a Triversa nanoelectrospray system (Advion Biosciences; +1.7 kV/−1.7 kV—positive/negative ion mode, 0.3-psi backing pressure) and a hybrid linear ion trap FT-ICR mass spectrometer (LTQ FT Ultra; Thermo Fisher Scientific) using resolutions of 100,000 to 400,000 (at 400 m/z). Each sample was collected as transient data in triplicate using the selected ion monitoring (SIM) stitch approach (Supplementary Data; ref. 30). To measure phosphatidylcholine and phosphatidylserine phospholipids, a novel DIMS approach, termed tandem mass spectrometry (MSMS)-stitch, was used comprising collision-induced dissociation (CID; 80-eV collision energy) fragmentation of phospholipid ions, resulting in characteristic neutral losses from phosphatidylcholine and phosphatidylserine lipids and thereby aiding their identification (see Supplementary Data and Supplementary Fig. S1). SIM stitch transient data were averaged, Fourier transformed, and internally calibrated (Supplementary Tables S1–S4) in MATLAB (Mathworks; ref. 30). For each sample, peaks were retained if they were present in at least two of the triplicate analyses. When collating samples into a data matrix, peaks were retained if they were present in at least 50% of all samples (31). Data matrices were probabilistic quotient normalized (32). Prior to principal components analysis (PCA), a generalized log transformation (λ = 7.3 × 10−11) was applied (33).

Mass spectral peak annotation

Peaks were putatively annotated using KEGG (http://www.genome.jp/kegg/) and LIPID MAPS (http://www.lipidmaps.org) databases in conjunction with MI-Pack software (34). Annotation m/z tolerance was ±1 ppm (for peaks < 500 m/z) and ±3 ppm (500–1,000 m/z). For the DIMS approach, fatty acids, phospholipids, and lysophospholipids were identified as [M−H], except phosphatidycholine and lysophosphatidycholine that ionized as an acetate adduct ([M+OAc]). Diacylglycerol (DAG) and triacylglycerol (TAG) were identified as [M+NH4]+. Polar metabolites were identified as [M−H], [M+OAc], or [M+NaOAc-H]. For the MSMS-stitch DIMS approach, phosphatidylcholine was identified as [M−CH3] and phosphatidylserine was identified as [M−C3H5NO2-H]. The identities of phospholipids, lysophospholipids, and glycerol 3-phosphate (G3P) were confirmed by CID MSn on the LTQ FT Ultra (35 eV; Supplementary Fig. S2).

13C d-glucose tracer study data analysis

The 13C isotopes of fatty acids and phospholipids were assigned by looking for multiple additions of 1.003355 m/z to the 12C parent peak. To establish the amount of each compound that came from 13C D-glucose, the intensity of all the 13C isotope peaks was summed minus the naturally occurring 13C intensity from the 12C peak. Naturally occurring 13C (1.1% abundance) was calculated: (12C intensity × 1.1 × number of carbon atoms present in 12C identification)/100. This was calculated for 13C1, 13C213Cn peaks.

cDNA preparation and quantitative real-time PCR

RNA was extracted from 4 × 106 cells using the RNeasy Mini Kit (Qiagen) and quantified on a NanoDrop (Thermo Scientific). RNA quality was assessed using A260/280 nm and A260/230 nm ratios measured by the NanoDrop. cDNA was synthesized using SuperScript II reverse transcriptase according to the manufacturer's instructions (Invitrogen). Quantitative real-time PCR (qRT-PCR) was carried out using preoptimized QuantiTect Primer Assays (Qiagen) and a SensiMix SYBR Low-ROX Kit (Bioline) on an Agilent Mx3005P quantitative PCR (qPCR) cycler. 18S rRNA was used as an internal standard for cDNA. The qPCR cycle protocol included an initial heating at 95°C for 10 minutes, then 44 cycles of 95°C for 15 seconds, 55°C for 15 seconds and 72°C for 20 seconds, then a final cycle of 95°C for 1 minutes, 55°C for 30 seconds and 95°C for 30 seconds. Delta cycle threshold (ΔCt) values were calculated by subtracting 18S Ct from test gene Ct. Relative mRNA levels were determined by subtraction of control ΔCt values from treatment ΔCt values to give a ΔΔCt value. This was inverse logged (2−ΔΔCt) to generate relative fold changes.

Western blotting

Cells treated with drugs for 24 hours were lysed in radioimmunoprecipitation assay (RIPA) buffer. Thirty micrograms of protein was combined with Laemmli 4× loading dye (Bio-Rad) and 10% β-mercaptoethanol (Sigma) and heated at 70°C (5 minutes), and then separated on SDS-PAGE precast gradient gels (4%–15%; Bio-Rad). Proteins were transferred to Immobilon-P membrane (Millipore Corp.) and probed with 1/1,000 dilution of anti-SCD1 (ab19862; Abcam), anti-ACC1 (4190; New England Biolabs), anti-phospho-ACC Ser79 (11818; New England Biolabs), and anti-FASN (ab22759; Abcam). Detection was by horseradish peroxidase (HRP)–conjugated anti-rabbit/mouse diluted at 1/1,000 and ECL using Supersignal West Pico Chemiluminescent substrate (Pierce). Loading controls used anti-β-actin antibody (Sigma) diluted at 1/25,000 and anti-mouse-HRP secondary at 1/25,000. Densitometry was performed using the ImageJ software (http://rsb.info.nih.gov/ij/) and protein expression normalized to β-actin.

Palmitate and oleate supplementation and measurement of cell viability

To generate soluble fatty acid stocks, palmitic acid (7.5 mmol/L) was complexed to bovine serum albumin (BSA; 1.5 mmol/L; Sigma) as described (35) and oleic acid (3 mmol/L) was purchased precomplexed to BSA (1.5 mmol/L; Sigma). Vehicle control stock of 1.5-mmol/L BSA was created. Cells were set in 100-μL cultures in 96-well flat-bottomed plates, with starting cell counts of 5,000 (AML) or 10,000 (Burkitt lymphoma) cells per well. Cells were treated with BaP and escalating concentrations of palmitic or oleate. After 48 hours, a further 100 μL of media was added, including reapplication of BaP and the fatty acids. After 96 hours, the 200-μL volume of cells was removed into a flow cytometry tube, centrifuged (250 × g) and supernatant discarded. FACS fix (200 μL; 1% formaldehyde, 2% FBS) and 20-μL (20,000 beads) CytoCount beads (Dako) were added, the sample vortexed (10 seconds), and analyzed by flow cytometry (FACSCalibur flow cytometer; Becton Dickenson). A minimum of 1,000 bead events were collected and ratio of the beads to the number viable cells in the forward/side scatter plots was proportional to the cell density. Cell death (after 96 hours) was measured by FACSCalibur flow cytometry (Becton Dickenson) using the Annexin V/propidium iodide kit (Becton Dickenson) according to the manufacturer's instructions.

Measurement of ROS

After drug treatment, 200 μL of cell suspension (approximately 1 × 105 cells) was incubated at 37°C with 2′,7′-dichlorofluorescin diacetate (DCFDA; Sigma) for 1 hour. The oxidized product 2′,7′-dichlorofluorescein (DCF) was measured according to the manufacturer's instructions using a FACSCalibur flow cytometer and CellQuest software (Becton Dickenson). Relative ROS levels were calculated as DCF fluorescence intensity relative to untreated control cells.

Statistical analysis

PCA was conducted using PLS_Toolbox (Eigenvector) in MATLAB. Statistical outliers were defined as samples that fell outside the 95% confidence limit on the PCA Q residuals and/or Hotelling axes. Univariate statistics composed of two-tailed t tests or one-way ANOVA with Dunnett post hoc testing where more than two sample classes were tested (in Minitab 17). Where multiple tests were applied in the metabolomics/lipidomics datasets, a false discovery rate (FDR) of <10% was used to correct for multiple hypothesis testing (36). Intensity changes between treatments are expressed as average fold changes. All bar charts represent the mean ± the standard error of the mean (SEM) unless indicated.

BaP treatment of AML cell lines perturbs the phospholipidome

Negative ion DIMS ([−]DIMS) analysis of lipid extracts from HL60 and K562 AML cells revealed that the majority of the abundant peaks were between 600 and 1000 m/z and attributable to intact phospholipids (Fig. 1A and Supplementary Fig. S2). Lipid extracts from HL60 and K562 cells treated for 24 hours with either BaP (combination of 0.5-mmol/L bezafibrate and 5-μmol/L MPA) or solvent control were analyzed by [−]DIMS. As reported previously, at 24 hours, BaP had little effect on cell viability (9), so any changes observed in the lipidome reflect changes upstream of cell differentiation or death. The PCA scores plot of the [−]DIMS data demonstrated the major segregation to be between HL60 and K562 cells along principal component 1 (Fig. 1B). This is not surprising, given that the cell lines were derived from different individuals with distinct forms of AML (HL60 are de novo AML M2 cells and K562 are AML cells derived from a chronic myeloid leukemia patient undergoing blast crisis; ref. 37, 38). Despite these baseline differences, both cell lines displayed a uniform response to BaP along principal component 2 (Fig. 1B). The PCA scores plot of 24-hour treatment of HL60 cells with 0.5-mmol/L bezafibrate, 5-μmol/L MPA, or the combination demonstrated that bezafibrate contributed most to the BaP effect (Fig. 1C). However, MPA-treated samples were significantly different to control samples along principal component 1 (P = 0.004), as were bezafibrate and BaP samples (P = 0.013), indicating that MPA is required to generate the lipid perturbation profile observed with BaP treatment (Fig. 1C).

Figure 1.

BaP drug treatment alters phospholipid composition in AML cells. [−]DIMS analysis, and subsequent statistics, of HL60 and K562 AML cell line lipid extracts after 24-hour drug treatments. A, representative [−]DIMS spectra of untreated cell lipid extracts. B and C, PCA scores plots of [−]DIMS lipid profiles from K562 and HL60 cells (B) after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA] treatment (control, n = 8; BaP, n = 7) and HL60 cells (C) after 24-hour BaP, bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments (n = 4). Drugs and their metabolites were removed from the data matrix prior to PCA. D, changes to the total levels of phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidylserine (PS), lysophosphatidylcholine (LPC), and lysophosphatidylethanolamine (LPE) after BaP treatment (see Supplementary Table S5; identities confirmed by MS/MS Supplementary Fig. S2). E, heatmaps showing the fold change of each individual phosphatidylcholine species after 24-hour BaP treatment (Supplementary Table S5). Filled blocks represent a phosphatidylcholine species that was detected, and the position on the plot reflects the total number of carbons and double bonds in both acyl chains of that species.

Figure 1.

BaP drug treatment alters phospholipid composition in AML cells. [−]DIMS analysis, and subsequent statistics, of HL60 and K562 AML cell line lipid extracts after 24-hour drug treatments. A, representative [−]DIMS spectra of untreated cell lipid extracts. B and C, PCA scores plots of [−]DIMS lipid profiles from K562 and HL60 cells (B) after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA] treatment (control, n = 8; BaP, n = 7) and HL60 cells (C) after 24-hour BaP, bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments (n = 4). Drugs and their metabolites were removed from the data matrix prior to PCA. D, changes to the total levels of phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidylserine (PS), lysophosphatidylcholine (LPC), and lysophosphatidylethanolamine (LPE) after BaP treatment (see Supplementary Table S5; identities confirmed by MS/MS Supplementary Fig. S2). E, heatmaps showing the fold change of each individual phosphatidylcholine species after 24-hour BaP treatment (Supplementary Table S5). Filled blocks represent a phosphatidylcholine species that was detected, and the position on the plot reflects the total number of carbons and double bonds in both acyl chains of that species.

Close modal

The quantities of the four major diacyl-phospholipid classes (phosphatidylethanolamine, phosphatidylcholine, phosphatidylinositol, and phosphatidylserine) were relatively unchanged after 24-hour BaP treatment of HL60 or K562 cells (Fig. 1D and Supplementary Table S5). Both cell lines were significantly depleted of lysophosphatidylethanolamine and lysophosphatidylcholine after BaP treatment (Fig. 1D and Supplementary Table S5). Lysophosphatidylethanolamine and lysophosphatidylcholine are major breakdown products of phosphatidylethanolamine and phosphatidylcholine, respectively, and a decrease in their steady-state concentrations might indicate a decrease in the rate of phospholipid turnover.

In addition, BaP treatment induced a consistent perturbation of the phospholipid acyl chain composition in HL60 and K562 cells. This is illustrated in Fig. 1E (and Supplementary Table S5) that shows the relative fold changes of individual phosphatidylcholine species in BaP-treated cells compared with control cells. The relative proportions of phosphatidylcholine species with saturated and monounsaturated chains were decreased and those with polyunsaturated chains were increased (Fig. 1E and Supplementary Table S5). This suggested that in the presence of BaP, HL60 and K562 cells had lower de novo synthesis of saturated and monounsaturated fatty acids.

BaP treatment decreases fatty acid and phospholipid synthesis and increases triacylglycerol levels

Glycerol 3-phosphate (G3P) and diacylglycerol (DAG) are intermediates in the de novo phospholipid biosynthetic pathway: two fatty acyl chains are added to G3P-generating phosphatidate, which can then be dephosphorylated to generate DAG. [−]DIMS analysis of polar extracts of HL60 and K562 cells treated with BaP for 24 hours showed elevated steady-state levels of G3P and positive ion DIMS ([+]DIMS) analysis of lipid extracts identified depleted DAG levels (Fig. 2A and Supplementary Table S6). This suggested phospholipid synthesis may be disrupted at the acyl chain addition stage. In contrast, the concentrations of triacylglycerol (TAG) were substantially increased in BaP-treated cells (Fig. 2A and Supplementary Table S6).

Figure 2.

BaP drug treatment disrupts fatty acid and phospholipid synthesis in AML cells. DIMS data from AML cell extracts after 24-hour treatments with BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L). A, the effect of BaP on steady-state levels of G3P (n = 8), DAG (n = 4), and TAG (n = 4). B–D, HL60 cells (n = 4) were drug treated in the presence of 13C d-glucose. Shown is the effect of BaP on the steady-state levels of total levels of free fatty acids (sum of 12C and 13C intensities; B); 13C incorporation into free fatty acids (C); and 13C incorporation into diacyl-phospholipids (D). PC, phosphatidylcholine; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PS, phosphatidylserine.

Figure 2.

BaP drug treatment disrupts fatty acid and phospholipid synthesis in AML cells. DIMS data from AML cell extracts after 24-hour treatments with BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L). A, the effect of BaP on steady-state levels of G3P (n = 8), DAG (n = 4), and TAG (n = 4). B–D, HL60 cells (n = 4) were drug treated in the presence of 13C d-glucose. Shown is the effect of BaP on the steady-state levels of total levels of free fatty acids (sum of 12C and 13C intensities; B); 13C incorporation into free fatty acids (C); and 13C incorporation into diacyl-phospholipids (D). PC, phosphatidylcholine; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PS, phosphatidylserine.

Close modal

C18:1 was the most abundant of the four observed free fatty acids in HL60 cells (Fig. 2B), and BaP treatment led to an approximately 15% decrease in its steady-state concentration (P = 0.005), whereas C16:0, C16:1, and C18:0 levels were unchanged (Fig. 2B). To determine the effects of BaP treatment on fatty acid and phospholipid biosynthesis from glucose, HL60 cells were grown in the presence of 13C d-glucose for 24 hours with and without BaP, and 13C incorporation into lipids was measured by [−]DIMS. BaP treatment decreased the incorporation of labeled carbon from 13C D-glucose into some free fatty acids (C16:1 0.66-fold, P = 0.008; C18:0 0.68-fold, P < 0.001; C18:1 0.25-fold, P < 0.001) and phosphatidylcholine (0.71-fold; P = 0.001), phosphatidylethanolamine (0.6-fold; P < 0.001), and phosphatidylinositol (0.59-fold; P = 0.025) phospholipids but did not significantly alter 13C D-glucose incorporation into C16:0 fatty acid (P = 0.63) or phosphatidylserine (P = 0.22). Bezafibrate was the major contributor to the effects of BaP; however, given that the changes associated with BaP were more substantial and/or more significant than bezafibrate treatment alone, MPA also appears to play a role (Fig. 2C and D and Supplementary Table S7).

The absolute intensities of each newly synthesized 13C phosphatidylcholine (PC) species from 13C D-glucose in HL60 cells are shown in Supplementary Table S8 and Supplementary Fig. S3A. The five most highly produced species [PC(32:1), PC(34:1), PC(34:2), PC(36:1), and PC(36:2): accounting for >80% of all synthesized phosphatidylcholine] contained at least one monounsaturated fatty acyl chain and C18:1 was present in all but one of the species (Supplementary Table S8). The incorporation of 13C from 13C D-glucose into these five phosphatidylcholine species was significantly decreased after BaP treatment (0.65- to 0.73-fold decrease; P < 0.01; Supplementary Table S8), which is consistent with the observed BaP-induced decrease in de novo C18:1 synthesis, as described above. Phosphatidylcholine species that had not been newly synthesized from 13C D-glucose (12C isotope peaks; Supplementary Fig. S3B) were generally increased after BaP treatment, particularly the long-chain polyunsaturated species, suggesting that cells were obtaining these from sources other than glucose, including exogenous import. Phosphatidylcholine species distribution heatmaps for each of the 12C and 13C datasets (Supplementary Fig. S4) showed that BaP decreased the de novo synthesis of phosphatidylcholine species with 0 to 2 double bonds and increased the abundance of polyunsaturated species mainly via a non-glucose, and most likely exogenous source. This is consistent with the earlier findings and gives a rationale for the observed pattern seen in Fig. 1E.

BaP treatment modifies the expression of key enzymes involved in lipogenesis

The decreases in fatty acid and phospholipid synthesis led us to ask whether 24-hour BaP treatment was altering expression levels of the enzymes responsible for fatty acid synthesis and desaturation. These were investigated in the two AML cell lines (HL60 and K562) plus two Burkitt lymphoma cell lines (BL31 and Glor).

Synthesis of malonyl-CoA by ACC1 is the first committed step in de novo fatty acid biosynthesis. There were decreases in ACC1 mRNA (HL60: 0.73-fold, P = 0.016; BL31: 0.78-fold, P = 0.008; Glor: 0.8-fold, P = 0.029) following 24-hour BaP treatment in HL60, BL31, and Glor cells, but not in K562 cells (Fig. 3A), whereas ACC1 protein levels remained relatively unchanged across the four cell lines (Fig. 3B). ACC1 can be posttranslationally inhibited by phosphorylation at serine 79 (denoted pACC). Immunoassay of pACC showed increased ACC1 phosphorylation in K562 cells (2.8-fold; P = 0.0001), but no change in the other three cell lines (Fig. 3B). FASN catalyzes the synthesis of palmitate (C16:0) from acetyl-CoA and malonyl-CoA. BaP treatment decreased mRNA expression of FASN in three of the four cell lines (HL60: 0.41-fold, P < 0.001; BL31: 0.62-fold, P = 0.006; Glor: 0.67-fold, P = 0.036), but not in K562 cells (Fig. 3C), and FASN protein levels were decreased in BaP-treated cells across all cell lines (0.87-fold, P = 0.009; Fig. 3D).

Figure 3.

BaP drug treatment induces minor perturbations in the expression of ACC1 and FASN enzymes. qRT-PCR and Western blotting measurements of ACC1 and FASN in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments. Shown are qRT-PCR measurements of ACC1 (A) and FASN (C); and Western blotting measurements of ACC1 (B) and FASN (D). mRNA levels were normalized to 18S and protein levels were normalized to β-actin, and values shown are relative to solvent treated control cells (all n ≥ 4 ± SEM).

Figure 3.

BaP drug treatment induces minor perturbations in the expression of ACC1 and FASN enzymes. qRT-PCR and Western blotting measurements of ACC1 and FASN in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments. Shown are qRT-PCR measurements of ACC1 (A) and FASN (C); and Western blotting measurements of ACC1 (B) and FASN (D). mRNA levels were normalized to 18S and protein levels were normalized to β-actin, and values shown are relative to solvent treated control cells (all n ≥ 4 ± SEM).

Close modal

The clearest effects of BaP treatment were on SCD1, which mainly converts stearate (C18:0) to oleate (C18:1) and is the rate-limiting step in the synthesis of monounsaturated fatty acids. SCD1 mRNA levels were decreased in HL60, BL31, and Glor cells (HL60: 0.63-fold, P = 0.005; BL31: 0.8-fold, P = 0.007; Glor: 0.83-fold, P = 0.042), though not in K562 cells (Fig. 4A). However, BaP treatment caused a significant decrease in SCD1 protein concentration in all four cell lines (HL60: 0.6-fold, P = 0.016; K562: 0.62-fold, P = 0.017; BL31: 0.52-fold, P = 0.023; Glor: 0.46-fold, P < 0.001; Fig. 4B), with both bezafibrate and MPA both contributing to this effect.

Figure 4.

SCD1 protein levels are deceased after BaP drug treatment. A and B, qRT-PCR (A) and Western blotting measurements (B) of SCD1 mRNA and protein in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments (all n ≥ 4 ± SEM). mRNA levels were normalized to 18S and protein levels were normalized to β-actin, and values shown are relative to solvent treated control cells (all n ≥ 4 ± SEM).

Figure 4.

SCD1 protein levels are deceased after BaP drug treatment. A and B, qRT-PCR (A) and Western blotting measurements (B) of SCD1 mRNA and protein in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) treatments (all n ≥ 4 ± SEM). mRNA levels were normalized to 18S and protein levels were normalized to β-actin, and values shown are relative to solvent treated control cells (all n ≥ 4 ± SEM).

Close modal

BaP-treated cells can be rescued by oleate (C18:1) but not palmitate (C16:0)

The BaP-induced depletion of C18:1 free fatty acid levels and decrease in SCD1 concentration led us to consider whether provision of exogenous oleate (C18:1) might rescue the leukemia and lymphoma cell lines from killing by BaP during 96-hour culture. Supplementation with oleate, which was added as a complex with BSA, increased the viable cell count of BaP-treated cells in a dose-dependent manner in all four cell lines (between 1.4- and 2.1-fold increase with 300-μmol/L oleate compared with no oleate supplementation; P < 1 × 10−6; Fig. 5A). Analysis of the effect on bezafibrate and MPA as single treatments showed that 300-μmol/L oleate rescued bezafibrate-treated cells almost back to untreated control levels in all four cell lines, whereas there was only a small rescue effect in MPA-treated K562 and BL31 cells (Fig. 5B). In contrast, supplementation of BaP-treated cells with palmitate–BSA complex (C16:0), the product of FASN, did not rescue the cells from the effects of BaP, and concentrations of palmitate above 100-μmol/L reduced viable cell counts in both BaP-treated and untreated cells (Fig. 5C).

Figure 5.

Oleate rescues AML and Burkitt lymphoma cells from killing by BaP drug treatment. The effect of fatty acid supplementation on survival of AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines treated with BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) for 96 hours. Viable cell counts (measured by flow cytometry) of drug-treated cells in the presence of escalating amounts of oleate (A) or 300 μmol/L oleate (B). C, viable cell counts of drug-treated cells in the presence of escalating amounts of palmitate. D, apoptosis was measured by Annexin V staining and flow cytometry after cells were treated with drug in the presence of 300 μmol/L oleate. Data, n ≥ 4 ± SEM.

Figure 5.

Oleate rescues AML and Burkitt lymphoma cells from killing by BaP drug treatment. The effect of fatty acid supplementation on survival of AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines treated with BaP [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA], bezafibrate (0.5 mmol/L), and MPA (5 μmol/L) for 96 hours. Viable cell counts (measured by flow cytometry) of drug-treated cells in the presence of escalating amounts of oleate (A) or 300 μmol/L oleate (B). C, viable cell counts of drug-treated cells in the presence of escalating amounts of palmitate. D, apoptosis was measured by Annexin V staining and flow cytometry after cells were treated with drug in the presence of 300 μmol/L oleate. Data, n ≥ 4 ± SEM.

Close modal

Our earlier observations demonstrated that AML cell killing by BaP requires a >96-hour exposure (9), whereas killing of eBL cells is more rapid (7). Consistent with this, oleate induced a strong antiapoptotic effect in the Burkitt cell lines, as measured by decreased Annexin V staining, which was not observed in AML cells (Fig. 5D).

A commercial SCD1 inhibitor can partially recapitulate the effect of BaP on lipogenesis.

As SCD1 appeared to be a target of BaP, we treated cells with a commercial SCD1 inhibitor (4-(2-Chlorophenoxy)-N-[3-[(methylamino)carconyl]phenyl]-1-piperidinecarboxamide [ab142089]) in the presence of 13C-d-glucose to identify if it could generate a similar lipid profile to that seen in BaP-treated cells. With respect to fatty acid synthesis from 13C D-glucose, ab142089 decreased C18:1 (0.70-fold; P = 6 × 10−4) and C16:1 (0.83-fold; P = 0.005) production, increased C18:0 production (1.2-fold, P = 0.03) and had little effect on C16:0 production (P = 0.13; Supplementary Fig. S5A). The levels of synthesis of C16:0, C16:1, and C18:1 after ab142089 treatment were consistent with those observed after BaP treatment (Fig. 2C); however, the effect on C18:0 synthesis was inconsistent: this was decreased by BaP treatment and increased by ab142089 treatment (Fig. 2C and Supplementary Fig. S5A). When looking at individual classes of fatty acid, ab142089 decreased synthesis of monounsaturated fatty acids from 13C D-glucose (0.73-fold; P = 4 × 10−4), but did not significantly alter saturated fatty acid synthesis (P = 0.07; Supplementary Fig. S5B). Considering phosphatidylcholine synthesis from 13C D-glucose, ab142089 decreased production of PC(36:2) (0.77-fold; P = 0.005) and increased production of PC(36:1) (1.37-fold; P = 0.008; Supplementary Fig. S6A), whereas BaP decreased the synthesis of many phosphatidylcholine species, including PC(36:1) and PC(36:2) (Supplementary Fig. S3A). These findings suggest that BaP decreases SCD1 activity but also negatively regulates other lipogenic processes, including FASN (see above).

Oleate decreases the levels of BaP-induced ROS

BaP induces ROS in all cell types that are sensitive to this drug combination (8–10). We therefore tested whether the rescue of cells mediated by oleate was associated with diminished ROS production. As shown in Fig. 6, oleate (300 μmol/L) decreased ROS levels in both AML and eBL control cells (between 0.51- and 0.83-fold) and in BaP-treated cells (between 0.57- and 0.82-fold; Fig. 6). In BaP-treated K562, BL31, and Glor cells, oleate treatment brought ROS down to levels similar to those found in the untreated control cells. Given that ROS production is indicated as a major reason for the killing action of BaP, the ROS decrease is noteworthy and suggests that oleate is cytoprotective to BaP-treated cells by lowering ROS to tolerable levels. The rescue of AML and Burkitt lymphoma cells by oleate was associated with the accumulation of cytoplasmic lipid droplets in both BaP-treated and untreated cells (Supplementary Fig. S7). These are likely to represent the safe storage of fatty acids as TAG (25, 26).

Figure 6.

Oleate decreases ROS production in BaP-treated AML and Burkitt lymphoma cells. The effect of 300 μmol/L oleate supplementation on the levels of ROS in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP treatment [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA] measured by flow cytometry. A, representative flow cytometry histogram showing ROS levels after control and BaP treatments in K562 cells. B, summary of the flow cytometry ROS data from all four cell lines. Data, n = 4 ± SEM; treatments are relative to the controls.

Figure 6.

Oleate decreases ROS production in BaP-treated AML and Burkitt lymphoma cells. The effect of 300 μmol/L oleate supplementation on the levels of ROS in AML (K562 and HL60) and Burkitt lymphoma (BL31 and Glor) cell lines after 24-hour BaP treatment [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA] measured by flow cytometry. A, representative flow cytometry histogram showing ROS levels after control and BaP treatments in K562 cells. B, summary of the flow cytometry ROS data from all four cell lines. Data, n = 4 ± SEM; treatments are relative to the controls.

Close modal

There is a need for new low-toxicity therapies that selectively target cancer cells. Enhanced lipogenesis, arising from increased activities of fatty acid biosynthetic enzymes (including ACC1, FASN, and stearoyl CoA desaturase (SCD1)—see the pathways map in Fig. 7), is a metabolic hallmark of many cancer cells (15–17). Inhibition of these enzymes has been demonstrated to be selectively toxic to some cancer cells in vitro (18, 20–24), suggesting that this pathway might represent a good target for anticancer drugs. In particular, cancer cells appear to have a high dependence on monounsaturated fatty acids (39), which are key components of phospholipids. Monounsaturated fatty acids can also protect cells against saturated fatty acid toxicity by enhancing safe storage of saturates into TAGs (25, 26). In cancer cells, SCD1 inhibition leads to endoplasmic reticulum stress and apoptosis (40), symptoms that are similar to palmitate-induced lipotoxicity (41), and in cardiac myocytes, SCD1 activity protects cells against fatty acid–induced apoptosis (42). Given their rapid saturated fatty acid synthesis by ACC1 and FASN (15), cancer cells may be heavily reliant on SCD1 activity, making SCD1 a potential target for anticancer therapy. However, there are currently no routine therapies targeting SCD1 in the clinic.

Figure 7.

A schematic depicting the effect the BaP drug treatment has on fatty acid and phospholipid synthesis. The weights of the arrows represent the proportion of activity along the pathways observed in AML and Burkitt lymphoma cell lines after BaP treatment [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA]. Lipid/metabolite key: PA, phosphatidic acid; PC, phosphatidylcholine; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PS, phosphatidylserine; LPC, lysophosphatidylcholine; LPE, lysophosphatidylethanolamine.

Figure 7.

A schematic depicting the effect the BaP drug treatment has on fatty acid and phospholipid synthesis. The weights of the arrows represent the proportion of activity along the pathways observed in AML and Burkitt lymphoma cell lines after BaP treatment [0.5 mmol/L bezafibrate (BEZ) and 5 μmol/L MPA]. Lipid/metabolite key: PA, phosphatidic acid; PC, phosphatidylcholine; PE, phosphatidylethanolamine; PI, phosphatidylinositol; PS, phosphatidylserine; LPC, lysophosphatidylcholine; LPE, lysophosphatidylethanolamine.

Close modal

Here, we describe an investigation, at the lipid, gene, and protein levels, of the effects of the antileukemia and antilymphoma therapy BaP (combined 0.5-mmol/L bezafibrate and 5-μmol/L MPA) on lipogenesis in AML and eBL cells. Our clearest observations on the effects of BaP treatment on lipogenesis in these two disease settings are the following: (i) phospholipid species with saturated and monounsaturated acyl chains were decreased, whereas phospholipids with polyunsaturated chains were increased, (ii) BaP treatment decreased the incorporation of 13C from 13C D-glucose into a variety of cellular lipids, especially into monounsaturated free fatty acids and phospholipids with monounsaturated acyl chains, (iii) BaP caused a decrease in the concentration, and probably the activity, of SCD1, and (iv) supplementation of BaP-treated cells with oleate (the product of SCD1 activity), but not with palmitate, protected AML and eBL cells from killing by BaP. Both bezafibrate and MPA appear to have antilipogenic action individually but the effect against AML and eBL cell lines was enhanced when the two drugs were combined.

At first sight, two of these observations appear contradictory: the rate of introduction of new double bonds into fatty acids was decreased, but phospholipid acyl chains became more unsaturated. However, de novo fatty acid biosynthesis in human cells is only capable of producing saturated or omega-7/omega-9 monounsaturated fatty acids (via SCD1). Fatty acids with other double bond structures are classed as essential and have to be imported from exogenous sources (43). We demonstrate that phosphatidylcholine species with saturated and monounsaturated acyl chains represent the overwhelming majority of de novo synthesized PCs in cells used in this study, and the synthesis of these species from 13C D-glucose was strongly decreased by BaP. Conversely, phosphatidylcholine species that were not synthesized de novo from 13C D-glucose increased after BaP treatment, especially those with polyunsaturated acyl chains, which suggested that the cells were taking up exogenous lipids to compensate for the reduction in de novo synthesis. This phenomenon has been observed in prostate cancer cells after ACC1 inhibition that led to a doubling of the quantity of essential polyunsaturated fatty acids being incorporated into phospholipids (44). The increase in unsaturation of phospholipids in ACC1-inhibited prostate cancer cells caused them to become more sensitive to ROS-induced lipid peroxidation and apoptosis and this could be a useful therapeutic consequence of the treatment (44). Therefore, our observation of the change to phospholipid saturation status could be particularly relevant in the case of BaP, which is a strong inducer of intracellular ROS and also lipid peroxidation (9).

The changes to phospholipid synthesis and acyl chain composition can be explained by lowered fatty acid biosynthesis due to a BaP-induced decrease in FASN and SCD1 protein levels. SCD1 disruption appeared to be the more dominant factor as its protein levels were more strongly decreased in all four cell lines compared with FASN. Also the synthesis of the SCD1 products (C18:1 and C16:1 fatty acids) from 13C D-glucose was strongly decreased by BaP, whereas the synthesis of the FASN product (16:0 fatty acid) was unchanged, which was consistent with the actions of a commercial SCD1 inhibitor (ab142089). BaP had a broader negative effect on lipid synthesis than ab142089, including more widespread decrease in the synthesis of phosphatidylcholine species, which can be explained to some extent by the BaP-induced decreases of FASN protein levels and FASN and ACC1 gene expression. However, the critical anticancer component of BaP appeared to be the decrease in SCD1 protein levels because oleate (C18:1, SCD1 product), but not palmitate (C16:0, ACC1 and FASN product), supplementation could rescue both AML and eBL cells from BaP treatment.

Monounsaturated fatty acids have been shown to be important in cancer (39, 45), which, in part, is due to their requirement for cellular growth and proliferation. SCD1 is suggested be a major driver of fatty acid synthesis from glucose (46) and has also been shown to help negotiate cellular stress, with SCD1 inhibition leading to saturated fatty acid accumulation, toxicity, and endoplasmic reticulum stress (40, 47, 48). This is likely to be of particular importance in the cancer phenotype where levels of saturated fatty acid synthesis are often high (15, 16). BaP treatment led to increases in ROS and TAG levels, which are consistent with elevated saturated fatty acid levels as ROS is a symptom of endoplasmic reticulum stress and saturated fatty acid lipotoxicity (49, 50), whereas TAG represent a safe storage of excess saturated fatty acids in the cell (25, 26). In support of the BaP-induced ROS being related to saturated fatty acid build-up, supplementation of BaP-treated cells with oleate, which can rescue cells from palmitate toxicity (25), decreased ROS levels. This was accompanied with an increase in cytoplasmic lipid droplets, a similar phenomenon to that seen during oleate rescue of cells from saturated fatty acid–induced lipotoxicity (25).

To conclude, we show that the redeployed drug combination of bezafibrate and MPA kills AML and eBL cells, in part, by decreasing SCD1 protein levels and slowing monounsaturated fatty acid synthesis. This study is relevant to the wider context of cancer therapeutics where significant effort is being invested into developing therapeutics that can disrupt fatty acid biosynthesis by interfering with SCD1 due to its apparent anticancer activity (21–23). To our knowledge, this is the first study to describe clinically available drugs with demonstrable antileukemic and antilymphoma activity in clinical trials (11, 12) that target this pathway.

C.M. Bunce is a Research Director at Leukaemia & Lymphoma Research. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.D. Southam, F.L. Khanim, M.T. Drayson, C.M. Bunce

Development of methodology: A.D. Southam, M.R. Viant

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.D. Southam, R.E. Hayden, J.K. Constantinou, K.M. Koczula

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.D. Southam, F.L. Khanim, R.H. Michell, C.M. Bunce

Writing, review, and/or revision of the manuscript: A.D. Southam, F.L. Khanim, R.E. Hayden, R.H. Michell, M.R. Viant, M.T. Drayson, C.M. Bunce

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.D. Southam

Study supervision: F.L. Khanim, C.M. Bunce

The authors thank Ulrich Gunther (METAFLUX, Europe Marie Curie ITN) for supplying 13C d-glucose. They also thank Anshul Radotra for assistance with the Western blotting. The FT-ICR MS used here was obtained through Birmingham Science City Translational Medicine: Experimental Medicine Network of Excellence project, with Advantage West Midlands support.

This study was financially supported by Leukaemia & Lymphoma Research.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Buechner
T
,
Berdel
WE
,
Haferlach
C
,
Haferlach
T
,
Schnittger
S
,
Mueller-Tidow
C
, et al
Age-related risk profile and chemotherapy dose response in acute myeloid leukemia: a study by the German Acute Myeloid Leukemia Cooperative Group
.
J Clin Oncol
2009
;
27
:
61
9
.
2.
Burnett
AK
,
Hills
RK
,
Milligan
DW
,
Goldstone
AH
,
Prentice
AG
,
McMullin
M-F
, et al
Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial
.
J Clin Oncol
2010
;
28
:
586
95
.
3.
Molyneux
EM
,
Rochford
R
,
Griffin
B
,
Newton
R
,
Jackson
G
,
Menon
G
, et al
Burkitt's lymphoma
.
Lancet
2012
;
379
:
1234
44
.
4.
Ashburn
TT
,
Thor
KB
. 
Drug repositioning: identifying and developing new uses for existing drugs
.
Nat Rev Drug Discov
2004
;
3
:
673
83
.
5.
Palumbo
A
,
Facon
T
,
Sonneveld
P
,
Blade
J
,
Offidani
M
,
Gay
F
, et al
Thalidomide for treatment of multiple myeloma: 10 years later
.
Blood
2008
;
111
:
3968
77
.
6.
Wang
ZY
,
Chen
Z
. 
Acute promyelocytic leukemia: from highly fatal to highly curable
.
Blood
2008
;
111
:
2505
15
.
7.
Fenton
SL
,
Luong
QT
,
Sarafeim
A
,
Mustard
KJW
,
Pound
J
,
Desmond
JC
, et al
Fibrates and medroxyprogesterone acetate induce apoptosis of primary Burkitt's lymphoma cells and cell lines: potential for applying old drugs to a new disease
.
Leukemia
2003
;
17
:
568
75
.
8.
Hayden
RE
,
Pratt
G
,
Davies
NJ
,
Khanim
FL
,
Birtwistle
J
,
Delgado
J
, et al
Treatment of primary CLL cells with bezafibrate and medroxyprogesterone acetate induces apoptosis and represses the pro-proliferative signal of CD40-ligand, in part through increased 15d Delta(12,14,)PGJ(2)
.
Leukemia
2009
;
23
:
292
304
.
9.
Khanim
FL
,
Hayden
RE
,
Birtwistle
J
,
Lodi
A
,
Tiziani
S
,
Davies
NJ
, et al
Combined bezafibrate and medroxyprogesterone acetate: potential novel therapy for acute myeloid leukaemia
.
PLoS ONE
2009
;
4
:
14
.
10.
Hayden
RE
,
Kussaibati
R
,
Cronin
LM
,
Pratt
G
,
Roberts
C
,
Drayson
MT
, et al
Bezafibrate and medroxyprogesterone acetate target resting and CD40L-stimulated primary marginal zone lymphoma and show promise in indolent B-cell non-Hodgkin lymphomas
.
Leuk Lymphoma
2015
;
56
:
1079
87
.
11.
Murray
JA
,
Khanim
FL
,
Hayden
RE
,
Craddock
CF
,
Holyoake
TL
,
Jackson
N
, et al
Combined bezafibrate and medroxyprogesterone acetate have efficacy without haematological toxicity in elderly and relapsed acute myeloid leukaemia (AML)
.
Br J Haematol
2010
;
149
:
65
9
.
12.
Molyneux
E
,
Merrick
B
,
Khanim
FL
,
Banda
K
,
Dunn
JA
,
Iqbal
G
, et al
Bezafibrate and medroxyprogesterone acetate in resistant and relapsed endemic Burkitt lymphoma in Malawi; an open-label, single-arm, phase 2 study (ISRCTN34303497)
.
Br J Haematol
2014
;
164
:
888
90
.
13.
Willson
TM
,
Brown
PJ
,
Sternbach
DD
,
Henke
BR
. 
The PPARs: from orphan receptors to drug discovery
.
J Med Chem
2000
;
43
:
527
50
.
14.
Yadav
BK
,
Gupta
RK
,
Gyawali
P
,
Shrestha
R
,
Poudel
B
,
Sigdel
M
, et al
Effects of long-term use of depo-medroxyprogesterone acetate on lipid metabolism in Nepalese women
.
Korean J Lab Med
2011
;
31
:
95
7
.
15.
Menendez
JA
,
Lupu
R
. 
Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis
.
Nat Rev Cancer
2007
;
7
:
763
77
.
16.
Swinnen
JV
,
Brusselmans
K
,
Verhoeven
G
. 
Increased lipogenesis in cancer cells: new players, novel targets
.
Curr Opin Clin Nutr Metab Care
2006
;
9
:
358
65
.
17.
Igal
RA
. 
Stearoyl-CoA desaturase-1: a novel key player in the mechanisms of cell proliferation, programmed cell death and transformation to cancer
.
Carcinogenesis
2010
;
31
:
1509
15
.
18.
Noto
A
,
Raffa
S
,
De Vitis
C
,
Roscilli
G
,
Malpicci
D
,
Coluccia
P
, et al
Stearoyl-CoA desaturase-1 is a key factor for lung cancer-initiating cells
.
Cell Death Dis
2013
;
4
:
e947
.
19.
Bhatt
AP
,
Jacobs
SR
,
Freemerman
AJ
,
Makowski
L
,
Rathmell
JC
,
Dittmer
DP
, et al
Dysregulation of fatty acid synthesis and glycolysis in non-Hodgkin lymphoma
.
Proc Natl Acad Sci U S A
2012
;
109
:
11818
23
.
20.
Pizer
ES
,
Wood
FD
,
Pasternack
GR
,
Kuhajda
FP
. 
Fatty acid synthase (FAS): a target for cytotoxic antimetabolites in HL60 promyelocytic leukemia cells
.
Cancer Res
1996
;
56
:
745
51
.
21.
Beckers
A
,
Organe
S
,
Tinunermans
L
,
Scheys
K
,
Peeters
A
,
Brusselmans
K
, et al
Chemical inhibition of Acetyl-CoA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells
.
Cancer Res
2007
;
67
:
8180
7
.
22.
Fritz
V
,
Benfodda
Z
,
Rodier
G
,
Henriquet
C
,
Iborra
F
,
Avances
C
, et al
Abrogation of de novo lipogenesis by stearoyl-CoA desaturase 1 inhibition interferes with oncogenic signaling and blocks prostate cancer progression in mice
.
Mol Cancer Ther
2010
;
9
:
1740
54
.
23.
Kant
S
,
Kumar
A
,
Singh
SM
. 
Fatty acid synthase inhibitor orlistat induces apoptosis in T cell lymphoma: role of cell survival regulatory molecules
.
Biochim Biophys Acta
2012
;
1820
:
1764
73
.
24.
Wang
C
,
Xu
CX
,
Sun
MW
,
Luo
DX
,
Liao
DF
,
Cao
DL
. 
Acetyl-CoA carboxylase-alpha inhibitor TOFA induces human cancer cell apoptosis
.
Biochem Biophys Res Commun
2009
;
385
:
302
6
.
25.
Listenberger
LL
,
Han
XL
,
Lewis
SE
,
Cases
S
,
Farese
RV
,
Ory
DS
, et al
Triglyceride accumulation protects against fatty acid-induced lipotoxicity
.
Proc Natl Acad Sci U S A
2003
;
100
:
3077
82
.
26.
Nolan
CJ
,
Larter
CZ
. 
Lipotoxicity: why do saturated fatty acids cause and monounsaturates protect against it?
J Gastroenterol Hepatol
2009
;
24
:
703
6
.
27.
Sellick
CA
,
Hansen
R
,
Maqsood
AR
,
Dunn
WB
,
Stephens
GM
,
Goodacre
R
, et al
Effective quenching processes for physiologically valid metabolite profiling of suspension cultured mammalian cells
.
Anal Chem
2009
;
81
:
174
83
.
28.
Bligh
EG
,
Dyer
WJ
. 
A rapid method of total lipid extraction and purification
.
Can J Biochem Physiol
1959
;
37
:
911
7
.
29.
Wu
H
,
Southam
AD
,
Hines
A
,
Viant
MR
. 
High-throughput tissue extraction protocol for NMR- and MS-based metabolomics
.
Anal Biochem
2008
;
372
:
204
12
.
30.
Southam
AD
,
Payne
TG
,
Cooper
HJ
,
Arvanitis
TN
,
Viant
MR
. 
Dynamic range and mass accuracy of wide-scan direct infusion nanoelectrospray Fourier transform ion cyclotron resonance mass spectrometry-based metabolomics increased by the spectral stitching method
.
Anal Chem
2007
;
79
:
4595
602
.
31.
Payne
TG
,
Southam
AD
,
Arvanitis
TN
,
Viant
MR
. 
A signal filtering method for improved quantification and noise discrimination in fourier transform ion cyclotron resonance mass spectrometry-based metabolomics data
.
J Am Soc Mass Spectrom
2009
;
20
:
1087
95
.
32.
Dieterle
F
,
Ross
A
,
Schlotterbeck
G
,
Senn
H
. 
Probabilistic quotient normalization as robust method to account for dilution of complex biological mixtures. Application in H-1 NMR metabonomics
.
Anal Chem
2006
;
78
:
4281
90
.
33.
Parsons
HM
,
Ludwig
C
,
Gunther
UL
,
Viant
MR
. 
Improved classification accuracy in 1-and 2-dimensional NMR metabolomics data using the variance stabilising generalised logarithm transformation
.
BMC Bioinform
2007
;
8
:
16
.
34.
Weber
RJM
,
Viant
MR
. 
MI-Pack: increased confidence of metabolite identification in mass spectra by integrating accurate masses and metabolic pathways
.
Chemometrics Intell Lab Syst
2010
;
104
:
75
82
.
35.
Briaud
I
,
Harmon
JS
,
Kelpe
CL
,
Segu
VBG
,
Poitout
V
. 
Lipotoxicity of the pancreatic beta-cell is associated with glucose-dependent esterification of fatty acids into neutral lipids
.
Diabetes
2001
;
50
:
315
21
.
36.
Benjamini
Y
,
Hochberg
Y
. 
Controlling the false discovery rate—a practical and powerful approach to multiple testing
.
J R Stat Soc Ser B Methodol
1995
;
57
:
289
300
.
37.
Lozzio
CB
,
Lozzio
BB
. 
Human chronic myelogenous leukemia cell-line with positive philadelphia chromosome
.
Blood
1975
;
45
:
321
34
.
38.
Collins
SJ
,
Gallo
RC
,
Gallagher
RE
. 
Continuous growth and differentiation of human myeloid leukemic-cells in suspension culture
.
Nature
1977
;
270
:
347
9
.
39.
Mason
P
,
Liang
B
,
Li
L
,
Fremgen
T
,
Murphy
E
,
Quinn
A
, et al
SCD1 inhibition causes cancer cell death by depleting mono-unsaturated fatty acids
.
PLoS ONE
2012
;
7
:
e33823
.
40.
Roongta
UV
,
Pabalan
JG
,
Wang
X
,
Ryseck
R-P
,
Fargnoli
J
,
Henley
BJ
, et al
Cancer cell dependence on unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy
.
Mol Cancer Res
2011
;
9
:
1551
61
.
41.
Wei
Y
,
Wang
D
,
Topczewski
F
,
Pagliassotti
MJ
. 
Saturated fatty acids induce endoplasmic reticulum stress and apoptosis independently of ceramide in liver cells
.
Am J Physiol Endocrinol Metab
2006
;
291
:
E275
E81
.
42.
Matsui
H
,
Yokoyama
T
,
Sekiguchi
K
,
Iijima
D
,
Sunaga
H
,
Maniwa
M
, et al
Stearoyl-CoA desaturase-1 (SCD1) augments saturated fatty acid-induced lipid accumulation and inhibits apoptosis in cardiac myocytes
.
PLoS ONE
2012
;
7
:
13
.
43.
Cunnane
SC
. 
The conditional nature of the dietary need for polyunsaturates: a proposal to reclassify ‘essential fatty acids’ as ‘conditionally-indispensable’ or conditionally-dispensable' fatty acids
.
Br J Nutr
2000
;
84
:
803
12
.
44.
Rysman
E
,
Brusselmans
K
,
Scheys
K
,
Timmermans
L
,
Derua
R
,
Munck
S
, et al
De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation
.
Cancer Res
2010
;
70
:
8117
26
.
45.
Hess
D
,
Chisholm
JW
,
Igal
RA
. 
Inhibition of stearoylCoA desaturase activity blocks cell cycle progression and induces programmed cell death in lung cancer cells
.
PLoS ONE
2010
;
5
:
8
.
46.
Scaglia
N
,
Chisholm
JW
,
Igal
RA
. 
Inhibition of stearoylCoA desaturase-1 inactivates acetyl-CoA carboxylase and impairs proliferation in cancer cells: role of AMPK
.
PLoS ONE
2009
;
4
:
e6812
.
47.
von Roemeling
CA
,
Marlow
LA
,
Wei
JJ
,
Cooper
SJ
,
Caulfield
TR
,
Wu
K
, et al
Stearoyl-CoA desaturase 1 is a novel molecular therapeutic target for clear cell renal cell carcinoma
.
Clin Cancer Res
2013
;
19
:
2368
80
.
48.
Williams
KJ
,
Argus
JP
,
Zhu
Y
,
Wilks
MQ
,
Marbois
BN
,
York
AG
, et al
An essential requirement for the SCAP/SREBP signaling axis to protect cancer cells from lipotoxicity
.
Cancer Res
2013
;
73
:
2850
62
.
49.
Listenberger
LL
,
Ory
DS
,
Schaffer
JE
. 
Palmitate-induced apoptosis can occur through a ceramide-independent pathway
.
J Biol Chem
2001
;
276
:
14890
5
.
50.
Malhotra
JD
,
Miao
H
,
Zhang
K
,
Wolfson
A
,
Pennathur
S
,
Pipe
SW
, et al
Antioxidants reduce endoplasmic reticulum stress and improve protein secretion
.
Proc Natl Acad Sci U S A
2008
;
105
:
18525
30
.

Supplementary data