Compounds. Synthesis of new compounds and their intermediates is described in the Supplementary Note. MZ1 10, MT1 42 and ARV-771 40 were synthesized following previously described procedures, unless stated otherwise.
Cell lines and culture. HEK293, 22Rv1 and MV4;11 cells (ATCC) were grown in DMEM and RPMI (Invitrogen) respectively and supplemented with 10% v/v fetal bovine serum (FBS) (South American origin, Life Science Production) at 37°C and 5% CO2 in a humidified atmosphere. CRISPR HiBiT-BRD2, HiBiT-BRD3, and HiBiT-BRD4 HEK293 cells stably expressing LgBiT were grown in DMEM with 10% v/v FBS and CRISPR cMyc-HiBiT MV4;11 cells were grown in RPMI with 10% v/v FBS. All cells were split 1-2 times per week when 90% confluent and were not used beyond passage 30. Cells were routinely checked for mycoplasma contamination using Mycoalert detection kit (Lonza).
Vector constructs. Wild-type and mutant versions of human proteins BRD2 (P25440), BRD3 (Q15059) and BRD4 (O60885) VHL (UniProt accession number: P40337), ElonginC (Q15369), ElonginB (Q15370), and Ubiquitin UBB (UniProt P0CG47) were used for all protein expression.
pET-His-SUMO TEV BRD4 tandem was produced by cloning truncated BRD4 containing the two bromodomains (residue 1- 463) into parent pET His6 Sumo TEV LIC cloning vector (1S) using ligase independent cloning. pET His6 Sumo TEV LIC cloning vector (1S) was a gift from Scott Gradia (Addgene plasmid # 29659). Quikchange mutagenesis was performed on pET-His-SUMO BRD4 tandem with mutagenic primer following standard procedures, to generate mutant with the conserved Asparagine sitting in the Acetyl-lysine binding pocket substituted with Phenylalanine, i.e. BRD4 N140F and BRD4 N433F. For cellular NanoBRET experiments, the following vectors were generated: N-terminal NanoLuc fusions of full-length human BRD4 wild-type (WT), N140F, or N433F, HaloTag-VHL (Promega), HaloTag-UBB (Promega), and the biosensor BRD4 NanoLuc-BD1-BD2-HaloTag containing the tandem BD1-BD2 domain (AA 44-460) with a wild-type sequence or N433F mutation.
Degradation assays. MV4;11 cells were seeded at 1 x 106 cells/mL of 10 cm dishes for 12-24h before treatment. 22Rv1 and HEK293 cells were seeded at 2.5-6 x 105 cells/well of 6 well plates for 12-24h before treatment. Cells were treated with test compounds with and without inhibitors as indicated or an equivalent volume of DMSO and lysed at the stated time point. For lysis, cells were washed twice in ice cold PBS (Invitrogen) then lysed in 250µL/plate for MV4;11 cells or 80µL/well for 22Rv1 and HEK293 cells of ice cold lysis buffer containing 50mM Tris hydrochloride pH 7.4, 150mM sodium chloride, 1mM EDTA pH 7.4, 1 % v/v Triton X-100, 1x Halt™ Protease Inhibitor Cocktail (ThermoFisher). Lysates were sonicated, cleared by centrifugation at 4°C, at 15800 x g for 10 mins and the supernatants stored at -80°C. Protein concentration was determined by BCA assay (Pierce) and the absorbance at 562nm measured by spectrophotometry (NanoDrop ND1000). Samples were run on SDS-PAGE using NuPAGE Novex 4-12% Bis-Tris gels (Invitrogen) with 40µg total protein/well, transferred to 0.2μm pore nitrocellulose membrane (Amersham) by wet transfer and blocked with 3% w/v BSA (Sigma) in 0.1% TBST. Blots were incubated in anti-BRD2 (1:2000, abcam #ab139690), anti-BRD3 (1:500, abcam #ab50818), anti-BRD4 (1:1000, abcam #ab128874), anti-c-myc (1:1000, abcam #32072), anti-PARP (1:1000, CST #9542S), anti-cleaved PARP (1:1000, BD Pharmingen #51-9000017), anti-caspase-3 (1:1000, CST #9662S), anti-tubulin (1:3,000, Bio-Rad #12004165) or anti-β-actin (1:2500, CTS #4970S) antibody overnight at 4°C with rotation. Blots were then incubated in goat anti-mouse or donkey anti-rabbit IRDye 800CW secondary antibodies (1:10,000, LICOR #925-32210 and #926-32213) for 1 h at room temperature with rotation. Bands were detected using a ChemiDoc MP imaging system (BioRad) and quantified (Image Studio Lite, version 5.2) with normalisation to β-actin and the DMSO control per time point. Data are the average of two biological repeats unless indicated otherwise. Degradation data were plotted and fitted by nonlinear regression using a single-phase exponential decay model in GraphPad Prism.
Cell Viability Assay. MV4;11 cells were incubated with compounds at the desired concentration for 48 h on a clear-bottom 384-well plate. MV4;11 cells were kept in RPMI medium supplemented with 10% FBS, L-glutamine. Initial cell density was 3 × 105 per mL. The cells were treated with various concentrations of compound or 0.05% DMSO in triplicates for each concentration point. After treatment, cell viability was measured with Promega CellTiter-Glo luminescent cell viability assay kit according to the manufacturer instructions. Signal was recorded on a BMG Labtech PHERAstar luminescence plate reader with recommended settings. Data were analyzed with Graphpad Prism software to obtain EC50 values of each test compound.
Kinetic Degradation and Quantitation. HEK293 cells (ATCC) stably expressing LgBiT (Promega) were edited using CRISPR/Cas9 to endogenously HiBiT tag the N-terminal genomic loci of BRD2, BRD3, or BRD4 31. For kinetic degradation assays, cells were plated in quadruplicate in white 96-well tissue culture plates at a density of 2 × 104 cells per well in 100µL of growth medium and incubated overnight at 37°C, 5% CO2. The following day, medium was replaced with CO2-independent medium (Gibco) containing a 1:100 dilution of Endurazine (Promega) and were incubated at 37°C in 5% CO2 for 2.5 h before addition of a 3-fold serial dilution of 1µM (SIM1-SIM3 with BRD4),10nM (SIM1 with BRD2, 3, and 4), or 100nM (ARV-771 (MedChemExpress) with BRD2, 3, and 4) final concentration of compound. Plates were placed with lids into the GloMax Discover Microplate Reader (Promega) set to 37°C and continuous luminescent measurements with readings every 5-15min were carried out over a 24 h period post-compound treatment. Degradation rate and degradation plateau were calculated from above determined kinetic degradation profiles. Briefly, the initial degradation portion of each kinetic concentration curve was fitted to the equation:
where ƛ = degradation rate in units of h–1. The degraded fraction, Dmax, was calculated as 1 – plateau. For each curve, the data points before onset of degradation were excluded from the fits. The Dmax was then plotted against concentration to determine Dmax50 values.
Mass spectrometry proteomics. Sample preparation. MV4;11 cells in RPMI (Invitrogen) were seeded at 5 × 106 cells on a 100mm plate 24 h before treatment. Cells were treated in triplicate by addition of test compound. After 4 h, the cells were centrifuged at 250g for 5 min and washed twice with 12mL of cold PBS. Cells were lyzed in 500µL of 100mM TRIS pH 8.0, 4% (w/v) SDS supplemented with protease inhibitor cocktail (Roche). The lysate was pulse sonicated briefly and then centrifuged at 15,000g for 10 min at 4°C. Samples were quantified using a micro BCA protein assay kit (Thermo Fisher Scientific) and 200µg of each sample was processed and digested using the filter aided sample preparation method followed by alkylation with iodoacetamide and digestion with trypsin as previously described 21. The samples were then desalted using a 7mm, 3mL C18 SPE cartridge column (Empore, 3M) and labeled with TMT 10-plex Isobaric Label Reagent Set (Thermo Fisher Scientific) as per the manufacturer’s instructions. After labeling, the peptides from the nine samples were pooled together in equal proportion. The pooled sample was fractionated using high pH reverse-phase chromatography on an XBridge peptide BEH column (130Å, 3.5µm, 2.1 × 150mm, Waters) on an Ultimate 3000 HPLC system (Thermo Scientific/Dionex). Buffers A (10mM ammonium formate in water, pH 9) and B (10mM ammonium formate in 90% acetonitrile, pH 9) were used over a linear gradient of 5 to 60% buffer B over 60 min at a flow rate of 200µL min−1. Then, 80 fractions were collected before concatenation into 20 fractions on the basis of the ultraviolet signal of each fraction. All the fractions were dried in a Genevac EZ-2 concentrator and resuspended in 1% formic acid for mass spectrometry analysis.
LC–MS/MS analysis. The fractions were analyzed sequentially on a Q Exactive HF Hybrid Quadrupole-Orbitrap Mass Spectrometer (Thermo Scientific) coupled to an UltiMate 3000 RSLCnano ultra HPLC system (Thermo Scientific) and EasySpray column (75µm × 50 cm, PepMap RSLC C18 column, 2µm, 100Å, Thermo Scientific). Buffers A (0.1% formic acid in water) and B (0.08% formic acid in 80% acetonitrile) were used over a linear gradient from 5 to 35% buffer B over 125 min at 300nL min−1. The column temperature was 50°C. The mass spectrometer was operated in data dependent mode with a single mass spectrometry survey scan from 335–1,600 m/z followed by 15 sequential m/z dependent MS2 scans. The 15 most intense precursor ions were sequentially fragmented by higher energy collision dissociation. The MS1 isolation window was set to 0.7 m/z and the resolution set at 120,000. MS2 resolution was set at 60,000. The automatic gain control (AGC) targets for MS1 and MS2 were set at 3 × 106 ions and 1 × 105 ions, respectively. The normalized collision energy was set at 32%. The maximum ion injection times for MS1 and MS2 were set at 50 and 200 ms, respectively.
Peptide and protein identification. The raw mass spectrometry data files for all 20 fractions were merged and searched against the Uniprot-sprot-Human-Canonical database by MaxQuant software v.1.6.0.16 for protein identification and TMT reporter ion quantitation. The MaxQuant parameters were set as follows: enzyme used trypsin/P; maximum number of missed cleavages equal to two; precursor mass tolerance equal to 10 ppm; fragment mass tolerance equal to 20 ppm; variable modifications: oxidation (M), acetyl (N-term), deamidation (NQ), Gln→pyro-Glu (Q N-term); fixed modifications: carbamidomethyl (C). The data was filtered by applying a 1% false discovery rate followed by exclusion of proteins with fewer than two unique peptides. Quantified proteins were filtered if the absolute fold-change difference between the three DMSO replicates was ≥1.5.
Monitoring cMyc Loss and Cell Viability in MV4;11 Cells. CRISPR cMyc-HiBiT MV4;11 cells (Promega) were plated at a density of 5 × 104 cells per well in solid, white 96-well tissue culture plates (Corning Costar #3917). Following an overnight incubation, they were treated with 1-100nM concentration of the indicated compounds and at the plotted time points, cMyc levels were determined using luminescent measurement with NanoGlo HiBiT lytic reagent (Promega). Replicate plates of all compound treatments were prepared and at identical timepoints as the protein level measurements, cell viability was measured using Cell-Titer Glo (Promega). Plates were shaken on an orbital shaker for 10-20 min before reading luminescence on a GloMax Discover Microplate Reader (Promega).
Caspase-Glo® 3/7 assays. 22Rv1 cells were seeded at 10,000 cells/well of white 96 well plates (Corning #3917) 12-24 h before treatment with test compounds with and without inhibitors or an equivalent volume of DMSO for 24 h. 100µL/well of Caspase-Glo 3/7 Reagent (Promega) was added and the plate shaken at 500 rpm for 30 s. The plate was incubated for 2 h and luminescence measured using a PHERAstar FS plate reader (BMG Labtech).
Clonogenic assay. 22Rv1 cells were treated with 10nM SIM1, cis-SIM1, MT1, MZ1 and ARV-711 for 24 h. The next day, cells were trypsinised and counted. 500 cells were re-plated and allowed to grow at 37ºC and 5% CO2 for 20 days. After 20 days incubation, the cells were fixed with ice-cold 100% (v/v) methanol for 30 min at 4ºC. Afterwards, methanol was removed, and the cells were stained with 500µl 0.1% crystal-violet dye (in MeOH) for 30 min at room temperature. Following incubation, the cells were washed with dH2O and left to dry overnight. Plates were scanned on an Epson Perfection V800 Photo scanner. And image analysis was done in ImageJ software. Plating efficiency (PE) was calculated by counting colonies for each treatment condition and dividing the average by number of cells plated. Survival fraction was determined by diving PE of treated cells by PE of untreated cells, multiplied by 100 59. Bar graphs were generated using GraphPad Prism software. Error bars indicate the mean values ± S.D. Two independent experiments were performed.
Protein expression and purification. For expression of BRD4 tandem construct, N-terminally His6-tagged BRD4 (1–463) or similar mutants were expressed in Escherichia coli BL21(DE3) at 18°C for 16 h using 0.4mM isopropyl β-D-1-thiogalactopyranoside (IPTG). E. coli cells were lysed using a pressure cell homogenizer (Stansted Fluid Power) and lysate clarified by centrifugation. His6-tagged VCB was purified on a HisTrap HP affinity column (GE Healthcare) by elution with an imidazole gradient. The His6 tag was removed using TEV protease and the untagged complex dialyzed into low-concentration imidazole buffer. BRD4 was then flowed through the HisTrap HP column a second time, allowing impurities to bind, as the complex eluted without binding. BRD4 was then additionally purified by anion exchange and size-exclusion chromatography using Mono S and Superdex-200 columns (GE Healthcare), respectively. The final purified complex was stored in 20mM HEPES, pH 7.5, 100mM sodium chloride and 1mM TCEP. The VCB complex was expressed and purified as described previously 21. Briefly, N-terminally His6-tagged VHL (54–213), ElonginC (17–112) and ElonginB (1– 104) were co-expressed and the complex was isolated by Ni-affinity chromatography, the His6 tag was removed using TEV protease, and the complex further purified by anion exchange and size-exclusion chromatography. The BET protein BDs were expressed and purified as described previously 21. Briefly, N-terminally His6-tagged BRD2-BD1 (71–194), BRD2-BD2 (344–455), BRD3-BD1 (24–146), BRD3-BD2 (306–416), BRD4-BD1 (44–178) and BRD4-BD2 (333–460) were expressed and isolated by Ni-affinity chromatography and size-exclusion chromatography.
Size exclusion chromatography (SEC). SEC experiments were carried out in a ÄKTA pure system (GE Healthcare) at room temperature. The oligomeric state of the BRD4 BD1-BD2 tandem protein in solution was analyzed by gel filtration in a buffer containing 20mM HEPES (pH 7.5), 100mM NaCl and 1mM TCEP using a Superdex 200 Increase 10/300 GL column (GE Healthcare) calibrated with globular proteins of known molecular weight (GE Healthcare, 28-4038-41/42). BRD4 tandem (25 µM) was incubated with SIM1 (25µM), MZ1 (25µM), MT1 (25µM) or DMSO (0.5 %) for 30 min at room temperature prior to injection. Sample volume for each injection was 200µl, and the flow rate was 0.8 ml/min. Peak elution was monitored using ultraviolet absorbance at 280nm.
ITC. Titrations were performed as reverse titration on an ITC200 micro-calorimeter (Malvern). SIM1 was not soluble enough to be loaded at the required concentrations in the syringe (normal direct titration), therefore reverse titrations were performed. The titrations consisted of 19 injections of 2µl tandem BRD4 BD1-BD2 construct (WT or N140F or N433F) solution in 20mM Bis–Tris propane, 100mM NaCl, 1mM TCEP, 1.6% DMSO, pH 7.5, at a rate of 0.5µl/s at 120 s time intervals. An initial injection of protein (0.4µl) was made and discarded during data analysis. All experiments were performed at 25°C, whilst stirring at 750 r.p.m. SIM1 from 10mM DMSO stock solution and VCB were diluted in buffer containing 20mM Bis-Tris propane, 100mM NaCl, 1mM TCEP, pH 7.5. The final DMSO concentration was 1.6% v/v. BRD4 protein (200µM, in the syringe) was titrated into the SIM1-VCB complex (SIM1 16µM, VCB 32µM, in the cell). Data were fitted to a single-binding site model for each BRD4 mutant to obtain the stoichiometry (n), the dissociation constant (Kd) and the enthalpy of binding (ΔH). Data for WT BRD4 was fitted to a two sets of sites model to account for the reverse titration set-up whereby a two-site protein is titrated into a bivalent ligand (see Malvern MicroCal ITC analysis software using Origin™
User Manual, pg. 102). Data fitting was performed using Microcal LLC ITC200 Origin software provided by the manufacturer.
AlphaLISA assays. Ligands were titrated against 4nM His-tagged BRD4 BD2 and 10nM biotinylated JQ1. All reagents were diluted in 50mM HEPES, 100mM NaCl, 0.1% BSA, 0.02% CHAPS, pH7.5 (final concentration). On VCB premixed condition, the buffer also included 12.5µM VCB. Ligands were tested over an 11-point 3-fold serial dilution in duplicates for each concentration point, starting at 100µM without VCB or starting at 10µM with 20µM VCB, and giving a final DMSO concentration of 1%. Binding was detected using anti-His6 antibody-conjugated AlphaLISA acceptor beads and streptavidin-coated donor beads (PerkinElmer), with a final concentration of 10µg/ml for each bead). Titrations were prepared in white 384-well Alphaplates (PerkinElmer), and read on a Pherastar FS plate reader (BMG) equipped with an AlphaLISA excitation/emission module. Data was analyzed and dose-response curves generated using GraphPad Prism 7. Each assay well had a final volume of 25µl. First 10µl of 2.5X ligand or 2.5X ligand with VCB was mixed with 5µl of a 5X mix of bromodomain and biotinylated JQ1 and incubated for 1 h at room temperature. The assay plate was then moved to a dark room and 5µl of 5X acceptor beads were added and incubated for 1 h. Then (still in darkness) 5µl of 5X donor beads were added, the plate was incubated for 1 h before being read.
Fluorescence polarization assay. Fluorescence polarization (FP) competitive binding assays were run as described previously 33 with a final volume of 15µL, with each well solution containing 15 nM VCB protein, 10nM FAM-labeled HIF-1α peptide (FAM-DEALAHypYIPMDDDFQLRSF, “JC9”) and decreasing concentrations of PROTAC (14-point 2-fold serial dilution starting from 50µM) or PROTAC:BRD4 tandem protein (14-point 2-fold serial dilutions starting from 10µM PROTAC:20µM tandem bromodomain). Assays were prepared in triplicate on 384-well plates (Corning 3575) and all measurements taken using a PHERAstar FS (BMG LABTECH) with fluorescence excitation and emission wavelengths (λ) of 485 and 520nm, respectively. Components were dissolved from stock solutions using 100mM Bis-Tris propane, 100mM NaCl, 1mM TCEP, pH 7.5, and DMSO was added as appropriate to ensure a final concentration of 1%. Control wells containing VCB and JC9 with no compound (zero displacement), or JC9 in the absence of protein (maximum displacement) were also included to allow for normalization. Normalized (%) displacement values were plotted against log[PROTAC] and curves were fitted by nonlinear regression using GraphPad Prism to determine the IC50 values for each titration. Ki values were back-calculated from the Kd for JC9 (~2nM, determined from direct binding) and fitted IC50 values, as described previously 60. Cooperativity (α) values were calculated from the ratio of binary Ki and ternary Ki values determined for JC9 displacement by SIM1 alone or SIM1 + BRD4, respectively.
SPR binding studies. SPR experiments were performed on Biacore T200 instruments (GE Healthcare) as described previously 33. Immobilization of Biotinylated VCB was carried out at 25 °C on a pre-coupled Series S SA chip in running buffer containing 20mM TRIS, 150mM potassium chloride, 2mM magnesium chloride, 2mM TCEP, 0.005% TWEEN 20, 1% DMSO; pH 8.3. Multiple surface densities of biotinylated VHL were used (40, 80 and 120 RU). Biotinylated VCB was prepared as previously described 33. All interaction experiments were performed at 9 °C in running buffer containing 20mM TRIS, 150mM potassium chloride, 2mM magnesium chloride, 2mM TCEP, 0.005% TWEEN 20, 1% DMSO; pH 8.3. SIM1 (10mM in 100% DMSO) were initially prepared at 1µM in running buffer with a concentration of 2% DMSO. This solution was mixed 1:1 with a solution of 50µM of the BRD4 tandem protein in running buffer without DMSO, to prepare a final solution of 500nM SIM1 and 25µM BRD4 tandem protein in running buffer containing 1% DMSO. This complex was then serially diluted in running buffer containing 2µM bromodomain and 1% DMSO (5-point five-fold serial dilution). Solutions were injected sequentially in single-cycle kinetic format without regeneration (three replicate series per experimental repeat, contact time 100 sec, flow rate 100µL/min, dissociation time 800 sec) using a stabilization period of 30 sec and syringe wash (50% DMSO) between injections. High flow rates and multiple surface densities were used to minimise mass transfer effects. Two series of blank injections were performed for all single cycle experiments. Sensorgrams from reference surfaces and blank injections were subtracted from the raw data before data analysis using Biacore insight evaluation software. To calculate the association rate (kon), dissociation rate (koff), and dissociation constant (KD), experiments were fitted using a 1:1 Langmuir interaction model, with a term for mass-transport included.
NanoBRET Ubiquitination, Ternary Complex, and Biosensor Experiments.
For endogenous live cell BET:Ubiquitin and BET:VHL assays, CRISPR HiBiT-BRD2, HiBiT-BRD3, and HiBiT-BRD4 HEK293 cells stably expressing LgBiT were transfected with 2µg of HaloTag-UBB or HaloTag-VHL vectors in 6-well plates using FuGENE HD (Promega). For full transient NanoBRET experiments with NanoLuc-BRD4 WT, N433F, or N140F mutants, HEK293 cells (8 ×105) were co-transfected with 0.02µg NanoLuc-BRD4 and 2µg of HaloTag-VHL vectors. For transient NanoBRET experiments with the BRD4 NL-BD1-BD2-HT biosensor containing either the WT tandem BD1-BD2 domain the N433F mutation, HEK293 cells (8 ×105) were transfected with 0.02µg biosensor plasmid and 2µg carrier DNA. The following day, transfected cells (2 × 104) were replated in quadruplicate into white 96-well tissue culture plates in the presence or absence of HaloTag NanoBRET 618 Ligand (Promega) and incubated overnight at 37°C, 5% CO2. For kinetic experiments, medium was replaced with Opti-MEM (Gibco) containing a 1:100 dilution of Vivazine (Promega), and plates were incubated at 37 °C, 5% CO2, for 1 h before addition of DMSO or 10nM-1µM final concentration of the indicated compounds. Continual BRET measurements were then made every 3 min up to 5 h on a CLARIOstar equipped with an atmospheric control unit (BMG Labtech) set to 37 °C and 5% CO2. For the biosensor experiments, the cells were treated with a 3-fold serial titration of 10µM of the indicated compounds. NanoBRET NanoGlo (Promega) substrate was added and BRET was measured two hours post-compound treatment using a GloMax Discover Microplate Reader (Promega). Dual filtered luminescence was collected with a 460/80nm bandpass filter and a 610nm long pass filter (acceptor, HaloTag NanoBRET ligand) using an integration time of 0.5 s. For all NanoBRET experiments, background subtracted NanoBRET ratios expressed in milliBRET units were calculated from the equation:
Fold increase in BRET was calculated by normalizing mBRET ratios to the average mBRET ratios for DMSO controls.
NanoBRET Target Engagement and Residence Time. For target engagement experiments in live and permeabilized cells, CRISPR HiBiT-BRD4 HEK293 cells stably expressing LgBiT were plated into white 96-well tissue culture plates at a density of 2 × 104 cells/well. Cells were equilibrated for 1 h with energy transfer probes and the indicated test compound prior to NanoBRET measurements. NanoBRET tracers were prepared at a working concentration of 20X in tracer dilution buffer (12.5mM HEPES, 31.25% PEG-400, pH 7.5). NanoBRET BRD Tracer-02 was added to cells at a final concentration of 0.5µM. To measure NanoBRET in live cells, NanoBRET NanoGlo Substrate and Extracellular NanoLuc Inhibitor (Promega) were added according to the manufacturer’s recommended protocol, and filtered luminescence was measured on a GloMax Discover Microplate Reader (Promega) equipped with 450 nm BP filter (donor) and 600 nm LP filter (acceptor) using 0.3 s integration time. To measure NanoBRET in permeabilized cells, digitonin was added to the cells to a final concentration of 50µg/mL and Extracellular NanoLuc inhibitor was omitted during the detection step. For residence time experiments CRISPR HiBiT-BRD4 HEK293 cells stably expressing LgBiT were trypsinized, washed and resuspended to a density of 2 × 105 cells/ml in Opti-MEM and incubated with either 1µM JQ1, 1µM SIM1, 100nM cis-SIM1, 100nM MT1, or 10µM MZ1 representing the representative IC80 values for tracer displacement in live cell format. Cells were incubated in 15mL conical tubes with caps loosened in a tissue culture incubator for 1 h. Following incubation, cells were spun at 300x g for 5 min, washed once with Opti-MEM, spun a second time at 300x g for 5 min, then resuspended with fresh Opti-MEM before plating at 2 × 104 cells/well. NanoBRET BRD Tracer-02 was added at a final concentration of 0.5µM cells and kinetic NanoBRET measurements were collected. NanoBRET ratios were expressed in milliBRET units and calculated according to the equation in the NanoBRET Ubiquitination, Ternary Complex and Biosensor Experiments section.