01.10.2019

National Institute on Aging awards $73 million for new Alzheimer’s drug discovery centers

by: SGC

 SGC and collaborating institutions will lead development of openly distributed tools to test the efficacy of Alzheimer’s therapies and rapidly share results

The National Institute on Aging (NIA), part of the National Institutes of Health, has awarded a grant expected to total $37.5 million over five years to establish the Open-AD Drug Discovery Center. Led by Emory University, the Center includes investigators at Sage Bionetworks, Structural Genomics Consortium (SGC), Stanford University, Oxford University, and University of North Carolina.

PFI-5 A potent inhibitor of SMYD2 with mixed MOA

Click here to obtain this probe.

overview
Probe Negative control

 

PFI-5

 

PFI-5N

A collaboration between Pfizer and the SGC has resulted in the discovery of PFI-5, a potent inhibitor of SMYD2 with mixed MOA. PFI-5 has a unique chemotype relative to the current SMYD2 chemical probes LLY-507 and BAY-598. PFI-5 inhibits in vitro methylation of p53K370 with IC50 = 9 nM and has more than 100-fold selectivity over other histone methyltransferases and other non-epigenetic targets. PFI-5 inhibits the methylation of p53K370 in cells with IC50 = 0.9 µM.  A control compound, PFI-5N, has also been developed which inhibits the in vitro methylation of p53K370 with IC50 > 10 micromolar.

properties
Probe Negative control

 

PFI-5

 

PFI-5N

Physical and chemical properties for PFI-5
Molecular weight561.3
Molecular formulaC29H39N7O.C2H4O2
IUPAC name(3-(2-amino-4-methyl-3,7,9-triaza-bicyclo[4.3.0]nona-1(6),2,4,8-tetraen-7-yl)-3-methyl-azetidin-1-yl)-(1-((1-(cyclohept-2-enyl)-piperidin-4-yl)-methyl)-1H-pyrrol-3-yl)-methanone;acetic acid
MollogP2.93
PSA93.76
No. of chiral centres1
No. of rotatable bonds6
No. of hydrogen bond acceptors8
No. of hydrogen bond donors3
Physical and chemical properties for PFI-5N (Negative Control)
Molecular weight575.4
Molecular formulaC30H41N7O.C2H4O2
IUPAC name(3-(2-amino-4-methyl-3,7,9-triaza-bicyclo[4.3.0]nona-1(6),2,4,8-tetraen-7-yl)-3-methyl-azetidin-1-yl)-(1-((1-(cyclohept-2-enyl)-piperidin-4-yl)-methyl)-2-methyl-1H-pyrrol-3-yl)-methanone;acetic acid
MollogP3.191
PSA63.57
No. of chiral centres1
No. of rotatable bonds6
No. of hydrogen bond acceptors8
No. of hydrogen bond donors3
  • SMILES:
  • PFI-5: [H][C@@]1(N2CCC(CN3C=CC(C(N4CC(C4)(N5C=NC6=C5C=C(C)N=C6N)C)=O)=C3)CC2)CCCCC=C1
  • PFI-5N: [H][C@]1(N2CCC(CN3C=CC(C(N4CC(C4)(N5C=NC6=C5C=C(C)N=C6N)C)=O)=C3C)CC2)CCCCC=C1
  • InChI:
  • PFI-5: InChI=1S/C29H39N7O.C2H4O2/c1-21-15-25-26(27(30)32-21)31-20-36(25)29(2)18-35(19-29)28(37)23-11-12-33(17-23)16-22-9-13-34(14-10-22)24-7-5-3-4-6-8-24;1-2(3)4/h5,7,11-12,15,17,20,22,24H,3-4,6,8-10,13-14,16,18-19H2,1-2H3,(H2,30,32);1H3,(H,3,4)/t24-;/m0./s1
  • PFI-5N: InChI=1S/C30H41N7O.C2H4O2/c1-21-16-26-27(28(31)33-21)32-20-37(26)30(3)18-36(19-30)29(38)25-12-15-35(22(25)2)17-23-10-13-34(14-11-23)24-8-6-4-5-7-9-24;1-2(3)4/h6,8,12,15-16,20,23-24H,4-5,7,9-11,13-14,17-19H2,1-3H3,(H2,31,33);1H3,(H,3,4)/t24-;/m1./s1
  • InChIKey:
  • PFI-5: AVKAVKZKOFSSMY-DEOSSOPVSA-N
  • PFI-5N: RSNWUCXHNLZHPY-XMMPIXPASA-N
selectivity profile
in vitro potency
cell based assay data
references
pk properties
co-crystal structures

Main features

  • SMYD2 structure with peptide substrate and SAM
  • Structures of BAY-598 and AZ-505 showing these bind in the substrate pocket
  • PFI-5 occupies both the SAM and peptide pockets
  • Overall view of PFI-5 binding to SMYD2
  • Key interactions of PFI-5 with SMYD2
synthetic schemes
materials and methods

UNC6934 A chemical probe for NSD2-PWWP1

The probe UNC6934 is available from Tocris, Cayman Chemical and Sigma.

The negative control UNC7145 is available from Cayman Chemical and Sigma.

overview
Probe Negative control

 

UNC6934

 

UNC7145

A collaboration between SGC and the Centre for Integrative Chemical Biology and Drug Discovery (CICBDD) at the University of North Carolina has resulted in UNC6934, a chemical probe for NSD2-PWWP1. UNC6934 is a potent antagonist of NSD2-PWWP1 with a Kd (SPR) of 80 ± 18 nM and is selective for NSD2-PWWP1 over 14 other PWWP domains including NSD3-PWWP1, the closest on the phylogenetic tree.

UNC6934 disrupts the NSD2-PWWP1 interaction with H3K36me2 nucleosomes in U2OS cells as measured by a NanoBret assay with an IC50 of 1.09 ± 0.23 microM. UNC7145, a closely-related control compound, with an iso-propyl group replacing a cyclo-propyl group, is inactive by SPR and NanoBret assays.

Data relating to the discovery of this probe is being prepared for publication. In the meantime, in order to facilitate research by the community we are making this compound available through this website.

properties
selectivity profile
in vitro potency
cell based assay data
references
pk properties
co-crystal structures
synthetic schemes
materials and methods

SGC6870 A chemical probe for PRMT6

The probe SGC6870 is available from Tocris, Cayman Chemical and Sigma.

The negative control SGC6870N is available from Tocris and Sigma.

overview
Probe Negative control

 

SGC6870

 

SGC6870N

A collaboration between the SGC, Eli Lilly, the Icahn School of Medicine at Mount Sinai, and the Ontario Institute for Cancer Research (OICR) has resulted in the discovery of SGC6870, a potent, selective, and cell-active allosteric inhibitor of PRMT6. SGC6870 inhibits PRMT6 with IC50 of 77 ± 6 nM and is selective over all other PRMTs and 23 methyltransferases.

SGC6870 inhibits the asymmetric dimethylation of H3R2 by overexpressed PRMT6 in HEK293T cells with an IC50 of 0.8 ± 0.2 microM. SGC6870N is the (S)-enantiomer of SGC6870 and is inactive on PRMT6 and thus an excellent control compound.

Data relating to the discovery of this probe is being prepared for publication. In the meantime, in order to facilitate research by the community we are making this compound available through this website.

properties
selectivity profile
in vitro potency
cell based assay data
references
pk properties
co-crystal structures
synthetic schemes
materials and methods

MU1210 A chemical probe for CLK kinases

The probe MU1210 is available from Cayman Chemical and Sigma.

The negative control MU140 is available from Sigma.

overview
Probe Negative control

 

MU1210

 

MU140

The Cdc2-like kinases (CLK) are evolutionary highly conserved dual specificity protein kinases. The CLK family consists of four members; CLK1, 2, 3, and 4 (Figure 1) (1). CLKs have a highly conserved domain structure at the C-terminus, containing a signature amino acid motif EHLAMMERILG (termed ‘LAMMER kinases’) (2). CLKs auto-phosphorylate on serine/threonine and tyrosine residues and phosphorylate exogenous substrates on serine/threonine residues (3). They play an important role in the regulation of RNA splicing through phosphorylation of members of the serine and arginine-rich family of splicing factors (SRSF). Phosphorylation of SRSF proteins on numerous serine residues is a prerequisite for entry of SRSF proteins into the nucleus, and for the assembly of the spliceosome (4). Being part of the splicing machinery, CLKs are often associated with the development of many pathologies, including cancer and neurodegenerative disorders. However, to date CLK-dependent RNA processing events remains poorly defined. Therefore, a chemical probe for CLK1/2/4, MU1210, and a negative control compound, MU140, have been developed in collaboration with the laboratory of Kamil Paruch at Masaryk University, Brno (5).


Figure 1: Phylogenetic kinase tree, CLK family highlighted with blue circles. Illustration is reproduced courtesy of Cell Signaling Technology, Inc. (www.cellsignal.com).

Potency Against Target Family
MU1210 is a potent probe for CLK1/2/4 with IC50 values of 8/20/12 nM, respectively with corresponding good cellular potency in NanoBRET assays (84/91/23 nM), respectively. CLK3 was not inhibited by MU1210 at concentrations up to 3 µM.

Selectivity
MU1210 is selective in a panel of 210 kinases in an activity-based assay (Invitrogen) at 1 µM. The closest off-target is HIPK2 (IC50 of 23 nM; >5x), however this was not inhibited in the cellular NanoBRET assays at 10 µM.

Dosage
Due to the relatively low solubility of the compound, concentrations higher than 10 µM should be avoided. It is recommended to use MU1210 at 1 µM and to make use of the additional CLK chemical probes T3-CLK and UNC-CAF-170 in the experiment.

Cellular Activity
In NanoBRET assays, MU1210 shows also a potency of 1.7 µM against the splicing kinase DYRK2. Western blots analyses showed that MU1210 inhibits the phosphorylation of SRSF proteins in a dose-depended manner and induces alternative splicing of Mdm4 at 10 µM in HeLa cells.
 

properties
Probe

MU1210

Physical and chemical properties for MU1210
Molecular weight352.39
Molecular formulaC22H16N4O
IUPAC name5-(1-methylpyrazol-4-yl)-3-[3-(4-pyridyl)phenyl]furo[3,2-b]pyridine
MollogP3.89
PSA56.74
No. of chiral centres0
No. of rotatable bonds3
No. of hydrogen bond acceptors3
No. of hydrogen bond donors0
StorageStable as a solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended.
DissolutionSoluble in DMSO up to 50 mM. 
Negative control

MU140

Physical and chemical properties for MU140
Molecular weight351.4
Molecular formulaC23H17N3O
IUPAC name5-(1-methylpyrazol-4-yl)-3-(2-phenylphenyl)furo[3,2-b]pyridine
MollogP5.05
PSA43.85
No. of chiral centres0
No. of rotatable bonds3
No. of hydrogen bond acceptors2
No. of hydrogen bond donors0
StorageStable as a solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended.
DissolutionSoluble in DMSO up to 50 mM. 

SMILES:
MU1210: CN1C=C(C2=NC3=C(OC=C3C4=CC=CC(C5=CC=NC=C5)=C4)C=C2)C=N1
MU140: CN1C=C(C2=NC3=C(OC=C3C4=C(C5=CC=CC=C5)C=CC=C4)C=C2)C=N1

InChI:
MU1210: InChI=1S/C22H16N4O/c1-26-13-18(12-24-26)20-5-6-21-22(25-20)19(14-27-21)17-4-2-3-16(11-17)15-7-9-23-10-8-15/h2-14H,1H3
MU140:  InChI=1S/C23H17N3O/c1-26-14-17(13-24-26)21-11-12-22-23(25-21)20(15-27-22)19-10-6-5-9-18(19)16-7-3-2-4-8-16/h2-15H,1H3

InChIKey:
MU1210: HEAGNKNMQVIVMM-UHFFFAOYSA-N
MU140: QSBNKGGRYOLZND-UHFFFAOYSA-N

selectivity profile

Selectivity screening of MU1210 was determined against 210 kinases in an activity-based assay (Invitrogen) at 1 µM (Figure 1). Top hits of the screen are indicated in Table 1.


Figure 1:  Visual representation of the top hits from MU1210 screened against a panel of 210 kinases.

Target 
MU1210
(% Activity at 1 µM)
CLK1 (h)1
CLK2 (h) 1
CLK4 (h) 1
HIPK2 (h) 1
HIPK3 (h) 6
GSK3α (h)10
PIM1 (h)16
PIM3 (h) 19
Haspin (h) 26
GSK3β (h)38
IRAK4 (h)40
TrkA (h)42
CaMKlly (h) 43

Table 1: Potency Against Target Family. Please note DYRKs are not in this panel.

MU1210 is selective in a panel of 210 kinases in an activity-based assay (Invitrogen) at 1 µM. The closest off-target in the activity based screen HIPK2 was not inhibited in the cellular NanoBRET assays at 10 µM (Table 2).

Kinase Ki MU1210 (µM) 
Ki MU140 (µM)
CLK20.091>10
HIPK2 >01NA
HIPK45.41NA
DYRK1A6.58>10 
DYRK1B>10>10
DYRK21.7>10

Table 2: Confirmation of hit CLK2 in the cellular NanoBRET assay.

Hits from the kinase panel were assessed in a NanoBRET assay. MU1210 was much less active against HIPK2 and GSK3A/B in cells than in vitro (Figure 2)Kd values of Staurosporine are shown for comparison (Table 3) (6, 7).



Figure 2: In vitro DSF assay of MU1210 and Staurosporine on potential off-targets.

CompoundCLK1CLK4PIM1GSG2 
GSK3β
ΔTm MU1210 (oC)9.310.3-0.12.96.2
ΔTm Staurosporine (oC)13.212.412.47.79.1
Kd Staurosporine (nM)329.63.2NA69

Table 3: In vitro DSF assay of MU1210 and Staurosporine on potential off-targets.

Materials and Methods

In vitro phophorylation assays
In vitro phosphorylation assays are described in (5).

NanoBRET assay
N-terminal NanoLuc and C-terminal NanoLuc/ kinase fusions, encoded in pFC32K expression vectors (Promega), were used. For cellular BRET target engagement experiments, HEK-293T were transfected with NLuc/target fusion constructs using FuGENE HD (Promega) according to the manufacturer’s protocol. Briefly, Nluc/target fusion constructs were diluted into Transfection Carrier DNA (Promega) at a mass ratio of 1:10 (mass/mass), diluted with OptiMEM media to a twentieth part of the volume of the HEK cells. FuGENE HD was added at a ratio of 1:3 (μg DNA: μL FuGENE HD). One part (vol) of FuGENE HD complexes was combined with 20 parts (vol) of HEK-293 cells suspended at a density of 2 x 105 cells/ml and afterwards the mixture was incubated in a humidified, 37°C/5% CO2 incubator for 24 h. After this, the cells were washed and resuspendend in OptiMEM medium. For Target engagement assays 4 x 103 cells/well were plated out in a 384-well plate (Greiner). For all experiments the recommended energy transfer probes (Promega) were used if possible at a final concentration of the Kd of the tracer on the target. In some cases higher tracer concentrations were used for better signal-to-noise ratios. Compounds and the energy transfer probe were added to the cells and incubated for 2h in humidified, 37°C/5% CO2 incubator. The chemical inhibitors were prepared as concentrated stock solutions in DMSO (Sigma-Aldrich) and diluted with OptiMEM for this experiment. Straight before the measurement NanoGlo Substrate and Extracellular NanoLuc Inhibitor (Promega) were mixed carefully with the supernatant. Luminescence was measured on a BMP PheraStar with 450 nm (donor) and 600 nm filters (acceptor) using 0.5 s integration time. Milli-BRET units (mBU) are calculated by multiplying the raw BRET values by 1000. Tracer and DMSO controls were used to calculate a normalized signal.

Inhibitory constants were calculated by using the sigmoidal dose-response (four parameters) equation in GraphPad Prism.

For a better comparison in between assays of different kinases Ki values were also calculated using the Cheng-Prusoff equation with the corresponding tracer Kds and the used tracer concentrations.

DSF assay
Purified, recombinant proteins were measured as described in (8).

in vitro potency
cell based assay data

In the NanoBRET assay MU1210 the following IC50 values were observed:


Figure 1: IC50 values from screening MU1210 and the negative control MU140 in the NanoBRET assay. 

The Ki values as determined by NanoBRET assays in HEK293T cells were:

Kinase Ki MU1210 (µM) 
Ki MU140 (µM)
CLK10.084>10
CLK20.091>10
CLK40.023>10
DYRK1A6.58>10 
DYRK1B>10>10
DYRK21.7>10

Table 1: Ki vales from screening MU1210 and the negative control MU140 in the NanoBRET assay.

After treatment of HeLa cells with MU1210 (VN339) for 3 hours the phosphorylation state of SR proteins was altered (Figure 2). MU1210 inhibited the phosphorylation of SRSF proteins in a dose-dependend manner, while the negative control MU140 had no effect on SRSF-phosphorylation.


Figure 2: Western blot analysis of phosphorylation state of SR proteins following treatment with MU1210 and its negative control MU140. 

Treatment with MU1210 at 10 µM affected the alternative splicing of Mdm4 in MCF7 cells leading to an accumulation of the shorter Mdm4 form (Mdm4-S) (Figure 3). No changes were observed compared to the DMSO control, as well as the negative control MU140.


Figure 3: PCR showing alternative splicing following treatment with MU1210 (10 µM).

In vitro toxicity was assessed using a MTT assay. MU1210 was not toxic in cells at >1 µM after 24 hours. The negative control MU140 showed no significant toxicity after 24 hours, up to 35 µM (5). MU1210 toxicity was further assessed following 72 hours of treatment (Table 2).  

Cell lineToxicity (µM)
MDA-MB-2311.3
MCF-71.2
MCF-10a1.5

Table 2: Cellular toxicity in selected cell lines. Cells were treated with MU1210 for 72 hours. Cellular toxicity was assessed using a MTT assay.

MU1210 induced severe impairment of cell proliferation at >1µM over a longer time period (Figure 4). The high initial concentration of 5 µM in MEF cells caused precipitations of the probe in the wells due to the limited solubility of the compound.


Figure 4: Cell proliferation following treatment with MU1210 in selected cell lines. 

 

Materials and Methods

Western blot analysis
For the dose dependence change of phosphor-SRSF proteins in Hela cells, 200,000 cells in 3 ml DMEM containing 10% FBS and Penicillin/Streptomycin were seeded in 6 well plates for 24h. The in DMSO diluted compounds were added and incubated for the stated time. Afterwards the cells were lysed mixed with SDS loading buffer, briefly heated and loaded onto a 12% SDS gel. After running the gel and blotting onto a nitrocellulose membrane, anti-phospho-SR antibody (Merck-Millipore, MABE50) was used to analyse the level of phosphorylated SRSF proteins. After washing steps, the membrane was incubated with anti-mouse antibodies coupled with horseradish peroxidase and analysed. Tubulin acted as a loading control.

Splicing analysis
The Mdm4 splicing in MCF7 cells is described in (5).

Cellular proliferation
For the proliferation assay cells were seeded between 100-200 cells/well in a 384 clear bottom plate (Nunc) in DMEM containing 10 % FBS and Penicillin/Streptomycin. After 24h compounds were added by an acoustic dispenser (ECHO) directly in each well. Proliferation was measured by determining the confluency over time in an Incucyte S3 automated microscope.

references

Work on this probe has been published in Furo[3,2-b]pyridine: A Privileged Scaffold for Highly Selective Kinase Inhibitors and Effective Modulators of the Hedgehog Pathway

  1. Aubol, B.E., et al., Release of SR Proteins from CLK1 by SRPK1: A Symbiotic Kinase System for Phosphorylation Control of Pre-mRNA Splicing. Mol Cell, 2016. 63(2): p. 218-228.DOI: 10.1016/j.molcel.2016.05.034. https://www.ncbi.nlm.nih.gov/pubmed/27397683
  2. Bullock, A.N., et al., Kinase domain insertions define distinct roles of CLK kinases in SR protein phosphorylation. Structure, 2009. 17(3): p. 352-62.DOI: 10.1016/j.str.2008.12.023. https://www.ncbi.nlm.nih.gov/pubmed/19278650
  3. Davis, M.I., et al., Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol, 2011. 29(11): p. 1046-51.DOI: 10.1038/nbt.1990. https://www.ncbi.nlm.nih.gov/pubmed/22037378
  4.  Fedorov, O., F.H. Niesen, and S. Knapp, Kinase inhibitor selectivity profiling using differential scanning fluorimetry. Methods Mol Biol, 2012. 795: p. 109-18.DOI: 10.1007/978-1-61779-337-0_7. https://www.ncbi.nlm.nih.gov/pubmed/21960218
  5. Manning, G., et al., The protein kinase complement of the human genome. Science, 2002. 298(5600): p. 1912-34.DOI: 10.1126/science.1075762. https://www.ncbi.nlm.nih.gov/pubmed/12471243
  6. Nayler, O., S. Stamm, and A. Ullrich, Characterization and comparison of four serine- and arginine-rich (SR) protein kinases. Biochem J, 1997. 326 ( Pt 3): p. 693-700.DOI: 10.1042/bj3260693. https://www.ncbi.nlm.nih.gov/pubmed/9307018
  7. Nemec, V., et al., Furo[3,2-b]pyridine: A Privileged Scaffold for Highly Selective Kinase Inhibitors and Effective Modulators of the Hedgehog Pathway. Angew Chem Int Ed Engl, 2019. 58(4): p. 1062-1066.DOI: 10.1002/anie.201810312. https://www.ncbi.nlm.nih.gov/pubmed/3056960
  8. Wodicka, L.M., et al., Activation state-dependent binding of small molecule kinase inhibitors: structural insights from biochemistry. Chem Biol, 2010. 17(11): p. 1241-9.DOI: 10.1016/j.chembiol.2010.09.010. https://www.ncbi.nlm.nih.gov/pubmed/21095574
pk properties
co-crystal structures

Co-structures of CLK1 with closely related compounds (PDB IDs: 6I5I, 6I5L, 6I5K, 6I58), are published in (5).

 

synthetic schemes
materials and methods
13.02.2019

FOP Friends and FOP France renewed partnership with SGC Oxford

by: SGC

SGC at the University of Oxford is pleased to announce that two patient foundations, FOP Friends and FOP France, have renewed their long standing partnership with SGC Oxford to support research into the rare congenital syndrome fibrodysplasia ossificans progressiva (FOP). The research led by Dr Alex Bullock aims to better define the molecular mechanisms that cause FOP and to develop new treatments.

NVS-BPTF-1 A chemical probe for BPTF

The probe NVS-BPTF-1 is available from Cayman Chemical.

overview
Probe Negative control

 

NVS-BPTF-1

 

NVS-BPTF-C

BPTF (Bromodomain PHD-finer Transcription Factor, also known as FALZ) is a multi-domain protein containing multiple histone lysine recognition domains. BPTF binds to acetylated H3 and H4 histone tails via its Bromodomain and H3K4me3 via the second PHD domain (1). BPTF is part of the NURF chromatin remodelling complex and involved in maintaining chromatin accessibility (2).

BPTF has been shown to be essential for normal embryogenesis (3) with studies indicating key roles in controlling embryonic stem cell differentiation (4).  Recently activation of BPTF expression by MITF (Microphthalmia-associated Transcription Factor) has been linked to transducing key pro-survival signals in melanoma (5).  Small molecule inhibition of BPTF may therefore be a viable approach for the treatment of melanoma and other malignancies.

Novartis in partnership with the SGC has developed NVS-BPTF-1 as a potent, selective and cell active chemical probe for BPTF.  NVS-BPTF-1 and its structurally very similar control NVS-BPTF-C will assist scientists in further dissecting the key role that BPTF1 plays in multiple signaling pathways for cell proliferation and survival.

Potency Against Target Family

NVS-BPTF-1 has on target in vitro binding affinity of 71nM with BPTF. The negative control NVS-BPTF-C shows a binding affinity of 1.67µM with BPTF.

Cellular Activity

In HEK293 cells, NVS-BPTF-1 exhibits an on-target IC50 of 16nM (nanoBRET) whilst the negative control NVS-BPTF-C shows no activity.

Note that NVS-BPTF-1 does not have adequate ADME properties for in vivo work.

Dosage

NVS-BPTF-1 is poorly soluble except in the HCl salt form. High cellular potency enables low concentrations of the compound to be used in cellular applications. It is recommended that the compounds are used at concentrations below 1µM.

In vitro Activity

NVS-BPTF-1 shows an IC50 of 56nM against BPTF in an alphascreen assay and a KD of 71nM in a BLI assay.

properties
Probe

NVS-BPTF-1

 
Physical and chemical properties for NVS-BPTF-1
Molecular weight537.19
Molecular formulaC26H28FN7O3S
IUPAC Name9-(1-cyclopropyl-1H-pyrazol-4-yl)-4-(2-fluoro-4-(4-methyl-piperazin-1-ylsulfonyl)-phenylamino)-3-methyl-1,5-diaza-bicyclo[4.4.0]deca-3,5,7,9-tetraen-2-one
MollogP2.9
PSA82.9
No. of chiral centres0
No. of rotatable bonds6
No. of hydrogen bond acceptors10
No. of hydrogen bond donors1
Permeability of HCl SaltPAMPA:
Log PAMPA (cm/s): -4.8
Calculate %FA: 69
Class: medium
MDCK low efflux:
20% of recovery
SolubilityEquilibrium solubility:
pH 4(g/L): >0.538
pH 6.8 (g/L): <0.002
Thermodynamic solubility:
pH 1 (g/L): 0.074
pH 6.8 (g/L): 0.0008
StorageStore at -20°C
DissolutionUp to 50 mM in DMSO
Negative control

NVS-BPTF-C

Physical and chemical properties for NVS-BPTF-C
Molecular weight552.21
Molecular formulaC26H29FN8O3S
IUPAC Name9-(1-cyclopropyl-1H-pyrazol-4-ylamino)-4-(2-fluoro-4-(4-methyl-piperazin-1-ylsulfonyl)-phenylamino)-3-methyl-1,5-diaza-bicyclo[4.4.0]deca-3,5,7,9-tetraen-2-one
MollogP2.43
PSA92.7
No. of chiral centres0
No. of rotatable bonds7
No. of hydrogen bond acceptors10
No. of hydrogen bond donors2
StorageStore at -20°C
DissolutionUp to 50 mM in DMSO

SMILES:
NVS-BPTF-1: CC1=C(N=C2C=CC(C3=CN(C4CC4)N=C3)=CN2C1=O)NC5=C(F)C=C(S(N6CCN(CC6)C)(=O)=O)C=C5
NVS-BPTF-C: CC1=C(N=C2C=CC(NC3=CN(C4CC4)N=C3)=CN2C1=O)NC5=C(F)C=C(S(N6CCN(CC6)C)(=O)=O)C=C5

InChI:
NVS-BPTF-1: InChI=1S/C26H28FN7O3S/c1-17-25(29-23-7-6-21(13-22(23)27)38(36,37)32-11-9-31(2)10-12-32)30-24-8-3-18(15-33(24)26(17)35)19-14-28-34(16-19)20-4-5-20/h3,6-8,13-16,20,29H,4-5,9-12H2,1-2H3
NVS-BPTF-C: InChI=1S/C26H29FN8O3S/c1-17-25(30-23-7-6-21(13-22(23)27)39(37,38)33-11-9-32(2)10-12-33)31-24-8-3-18(15-34(24)26(17)36)29-19-14-28-35(16-19)20-4-5-20/h3,6-8,13-16,20,29-30H,4-5,9-12H2,1-2H3

InChIKey:
NVS-BPTF-1: JYTISQGEFSHUIR-UHFFFAOYSA-N
NVS-BPTF-C: BFSKPRUNBBMNCF-UHFFFAOYSA-N

selectivity profile

A DSF screen against Human bromodomains reveals no significant off-targets (Table 1). A BROMOscan with NVS-BPTF-1 similarly showed good selectivity. BPTF exhibited a KD of 3nM, BRPF 37nM, CECR2 66nM, GCN5L2 62nM and PCAF 74nM in BROMOscan (Table 2)

Table 1: DSF screen Table 2: BromoScan 

NVS-BPTF-1 was also tested against the NIBR principal panel which showed no binding to 12 GPCRs, 3 nuclear receptors, 3 transporters and 7 other enzymes with an IC50<10µM.

The control compound, NVS-BPTF-C showed no binding to 14 GPCRs, 3 nuclear receptors, 3 transporters and 5 enzymes with an IC50<10µM in the NIBR principal panel.

Against the NIBR kinase panel, NVS-BPTF-1 showed no binding against 48 kinases with an IC50<30µM.

Against the NIBR kinase panel, NVS-BPTF-C showed no binding against 59 kinases with an IC50<30µM.

Materials and Methods

Thermal Stability Assay

Thermal melting experiments were carried out using an Mx3005p Real Time PCR machine (Agilent). Proteins were prepared in 10 mM HEPES pH 7.5, 500 mM NaCl and assayed in a 96-well plate at a final concentration of 2 μM in 20 μL volume. Compounds final concentration is 10 μM.

AlphaScreen

All bromodomain proteins were prepared according to the published procedures (Filippakopoulos at al, 2012). Assay was performed as described previously (Philpott et al, 2011). All reagents were pre-diluted in 25 mM HEPES, 100 mM NaCl, 0.1 % BSA, pH 7.4 and 0.05 % CHAPS and allowed to equilibrate to room temperature prior to addition to plates.  Plates filled with 5 uL of the assay buffer followed by 7 µL of biotinylated peptide [H-YSGRGKacGGKacGLGKacGGAKacRHRK(Biotin)-OH and His-tagged protein to achieve final assay concentrations of 25 nM. Plates were sealed and incubated for a further 60 minutes, before the addition of 8 μl of the mixture of streptavidin-coated donor beads (12.5 μg/ml) and nickel chelate acceptor beads (12.5 μg/ml) under low light conditions. Plates were foil-sealed to protect from light, incubated at room temperature for 60 minutes and read on a PHERAstar FS plate reader (BMG Labtech, Germany) using an AlphaScreen 680 excitation/570 emission filter set.

BioLayer Interferometry

BioLayer Interferometry (BLI) experiments were performed on a 16-channel ForteBio Octet RED384 instrument at 25 °C in 25mM HEPES, pH 7.5, and 100mM NaCl buffer. Proteins were biotinylated in vivo using the  construct with C-terminal AviTag. Proteins were immobilized on SSA sensors. Measurements were performed using 240 sec association step followed by 240 sec dissociation step on black tilted low volume 384-well plate (ForteBio). Baseline was stabilized for 120 sec prior to association step. Signal from the reference sensors was subtracted prior to Kd calculations using Analysis software (ForteBio).

in vitro potency
cell based assay data

To determine cellular target engagement, a NanoBRET assay based on a bespoke tracer (5961) was developed to measure the binding of the chemical probe and negative control to the NanoLuc-tagged BPTF bromodomain.

An EC50 of 16 nM was observed in this assay for NVS-BPTF-1 whereas the negative control NVS-BPTF-C was significantly less potent.

The putative cellular off-targets CECR2, PCAF and GCN5L2 were similarly tested in the NanoBRET assay with no activity detected.

Materials and Methods

NanoBRET

Dose-response experiments were conducted in 384 well format using HEK293T cells expressing NanoLuc fused to the N-terminus of the BPTF bromodomain, using the 5961 tracer at a final concentration of 1 µM.

references

1. Wysocka, J., Swigut, T., Xiao, H., Milne, T. A., Kwon, S. Y., Landry, J., Kauer, M., Tackett, A. J., Chait, B. T., Badenhorst, P., Wu, C., and Allis, C. D. (2006) A PHD finger of NURF couples histone H3 lysine 4 trimethylation with chromatin remodelling. Nature 442, 86-90
2. Goller, T., Vauti, F., Ramasamy, S., and Arnold, H. H. (2008) Transcriptional regulator BPTF/FAC1 is essential for trophoblast differentiation during early mouse development. Molecular and cellular biology 28, 6819-6827
3. Landry, J., Sharov, A. A., Piao, Y., Sharova, L. V., Xiao, H., Southon, E., Matta, J., Tessarollo, L., Zhang, Y. E., Ko, M. S., Kuehn, M. R., Yamaguchi, T. P., and Wu, C. (2008) Essential role of chromatin remodeling protein Bptf in early mouse embryos and embryonic stem cells. PLoS genetics 4, e1000241
4. Ngeow, K. C., Friedrichsen, H. J., Li, L., Zeng, Z., Andrews, S., Volpon, L., Brunsdon, H., Berridge, G., Picaud, S., Fischer, R., Lisle, R., Knapp, S., Filippakopoulos, P., Knowles, H., Steingrimsson, E., Borden, K. L. B., Patton, E. E., and Goding, C. R. (2018) BRAF/MAPK and GSK3 signaling converges to control MITF nuclear export. Proceedings of the National Academy of Sciences of the United States of America 115, E8668-e8677
5. Dar, A. A.; Majid, S.; Bezrookove, V.; Phan, B.; Ursu, S.; Nosrati, M.; De Semir, D.; Sagebiel, R. W.; Miller, J. R.; Debs, R.; Cleaver, J. E.; Kashani-Sabet, M., BPTF transduces MITF-driven prosurvival signals in melanoma cells. Proc. Natl. Acad. Sci. U. S. A., 2016, 113 (22), 6254–6258.

 

pk properties
co-crystal structures

Novartis have solved the structure of NVS-BPTF-1 in complex with BPTF at a resolution of 1.76Å. This structure is not currently deposited in the PDB.

Black dotted-line: H-Bond

Orange dotted-line: pi-pi interaction

Red sphere: water molecule

synthetic schemes
materials and methods
29.01.2019

New potential treatment for leukemia discovered by OICR scientists draws major industry investment

by: SGC

TORONTO (January 29, 2019) – A first-of-its-kind therapy for leukemia discovered by researchers in the Ontario Institute for Cancer Research’s (OICR) Drug Discovery Program, and under preclinical development, has attracted investment from Celgene Corporation that could exceed US$1 billion – which would make it the largest transaction to date for a preclinical asset discovered in Canada.

18.12.2018

Interactome Rewiring Following Pharmacological Targeting of BET Bromodomains

by: SGC

TP-238 A chemical probe for CECR2/BPTF bromodomains

This probe (hydrochloride) is available from Cayman Chemical, Sigma and Tocris

overview
Probe Negative control

 

TP-238

 

TP-422

CECR2 (cat eye syndrome chromosome region, candidate 2) gene is predominantly expressed in the nervous system and involved in neurulation. It is located in the segment of chromosome 22q11.2. Multiplication of this segment lead to rare genetic disorder called cat eye syndrome characterized by multiple congenital defects (1). CECR2 has also been implicated in regulation of DNA damage response (2)

Bromodomain PHD finger transcription factor BPTF/FALZ is a core component of the conserved, multi-subunit nucleosome remodelling factor (NURF) complex. BPTF is essential for neural development and haematopoiesis (3). BPTF is involved in c-MYC chromatin recruitment and transcriptional activity in hematopetic and also cancer stem cells (4).

In a collaborative effort Takeda and the SGC have identified and characterised TP-238 as a CECR2/BPTF chemical probe.

Potency Against Target Family

TP-238 has on target biochemical activity of 10-30 nM with CECR2 and 100-350 nM with BPTF. Negative control TP-422 is completely inactive against BPTF and CECR2.

Selectivity

The closest off-target bromodomain inhibition is BRD9 with IC50 of 1.4 µM. TP-238 has been profiled against the panel of 338 kinases and showed no activity at 1 μM.

Dosage

We recommend that TP-238 and TP-422 be used at no more than 2 µM concentration in cells.

Cellular Activity

Cell-based NanoBRETTM experiments measured the target engagement with both BPTF and CECR2 with EC50 in the 200-300 nM range.

In vitro Activity

TP-238 shows an IC50 of 30nM against CECR2 and 350nM against BPTF in an alphascreen assay. ITC shows TP-238 with a KD of 10nM for CECR2 and 120nM for BPTF.

properties
TP-238

Click here to download the SDF file.

SMILES:
CN(CCCOC1=CC=C(C2=NC(S(C)(=O)=O)=NC(NCCCN3C=CC=N3)=C2)C=C1)C
InChI:
InChI=1S/C22H30N6O3S/c1-27(2)13-6-16-31-19-9-7-18(8-10-19)20-17-21(26-22(25-20)32(3,29)30)23-11-4-14-28-15-5-12-24-28/h5,7-10,12,15,17H,4,6,11,13-14,16H2,1-3H3,(H,23,25,26)
InChIKey:
MSIJJXOWLFOYIN-UHFFFAOYSA-N

Physical and chemical properties
Molecular weight458.21
Molecular formulaC22 H30 N6 O3 S
IUPAC name1-(3-(6-(4-(3-dimethylamino-propoxy)-phenyl)-2-methylsulfonyl-pyrimidin-4-ylamino)-propyl)-1H-pyrazole
clogP2.4
PSA84.5
No. of chiral centres0
No. of rotatable bonds12
No. of hydrogen bond acceptors9
No. of hydrogen bond donors1
PAMPA (nm/sec, pH=7.4)28
Aqueous solubility (µM, pH= 6.8)>222
StorageStore at -20oC
DissolutionUp to 50mM in DMSO
TP-422

Click here to download the SDF file.

SMILES:
CN(CCCOC1=CC=C(C2=NC(OC)=NC(OCCCN3C=CC=N3)=C2)C=C1)C
InChI:
InChI=1S/C22H29N5O3/c1-26(2)12-5-15-29-19-9-7-18(8-10-19)20-17-21(25-22(24-20)28-3)30-16-6-14-27-13-4-11-23-27/h4,7-11,13,17H,5-6,12,14-16H2,1-3H3
InChIKey:
CVRFBLOQBLKUBC-UHFFFAOYSA-N

Physical and chemical properties
Molecular weight411.22
Molecular formulaC22 H29 N5 O3
IUPAC name1-(3-(6-(4-(3-dimethylamino-propoxy)-phenyl)-2-methoxy-pyrimidin-4-yloxy)-propyl)-1H-pyrazole
clogP3.4
PSA59.0
No. of chiral centres0
No. of rotatable bonds12
No. of hydrogen bond acceptors7
No. of hydrogen bond donors0
PAMPA (nm/sec, pH=7.4)283
Aqueous solubility (µM, pH= 6.8)>267
StorageStore at -20oC
DissolutionUp to 50mM in DMSO
selectivity profile

A DSF screen against Human bromodomains reveals only one significant off-target; BRD9:

ITC showed good potency and sufficient selectivity against BRD9 to meet SGC probe criteria:

Materials and Methods

Thermal stability assay

Thermal melting experiments were carried out using an Mx3005p Real Time PCR machine (Stratagene). Proteins were buffered in 10 mM HEPES pH 7.5, 500 mM NaCl and assayed in a 96-well plate at a final concentration of 2 μM in 20 μL volume. Compounds were added at a final concentration of 10 μM. SYPRO Orange (Molecular Probes) was added as a fluorescence probe at a dilution of 1:1000. Excitation and emission filters for the SYPRO-Orange dye were set to 465 nm and 590 nm, respectively. The temperature was raised with a step of 3 °C per minute from 25 °C to 96 °C and fluorescence readings were taken at each interval.

AlphaScreen

All bromodomain proteins were prepared according to the published procedures (Filippakopoulos at al, 2012). Assay was performed as described previously (Philpott et al, 2011). All reagents were pre-diluted in 25 mM HEPES, 100 mM NaCl, 0.1 % BSA, pH 7.4 and 0.05 % CHAPS and allowed to equilibrate to room temperature prior to addition to plates.  Plates filled with 5 uL of the assay buffer followed by 7 uL of biotinylated peptide [H-YSGRGKacGGKacGLGKacGGAKacRHRK(Biotin)-OH and His-tagged protein to achieve final assay concentrations of 25 nM. Plates were sealed and incubated for a further 60 minutes, before the addition of 8 μl of the mixture of streptavidin-coated donor beads (12.5 μg/ml) and nickel chelate acceptor beads (12.5 μg/ml) under low light conditions. Plates were foil-sealed to protect from light, incubated at room temperature for 60 minutes and read on a PHERAstar FS plate reader (BMG Labtech, Germany) using an AlphaScreen 680 excitation/570 emission filter set.

Isothermal Titration Calorimetry (ITC)

Experiments were carried out on a VP-ITC microcalorimeter (MicroCal™). All experiments were performed at 15 °C in 25 mM HEPES pH 7.4, 150 mM NaCl, 500 μM TCEP. 50 mM stocks of compound was thawed and diluted in 2 mL of buffer to a final concentration of 10 µM in the ITC cell. The protein titrations were conducted using an initial injection of 2 µl followed by 30 identical injections of 6 µl. The dilution heats were measured on separate experiments and were subtracted from the titration data. Thermodynamic parameters were calculated using ∆G = ∆H - T∆S = -RTlnKB, where ∆G, ∆H and ∆S are the changes in free energy, enthalpy and entropy of binding respectively. In all cases a single binding site model was employed.

in vitro potency
cell based assay data

TP-238 and its negative control were tested for target engagement in HEK cells using NanoBRETTM. Significant inhibition by TP-238 against BPTF (n = 3, EC50 = 228 nM) and CECR2 (n = 3, EC50 = 289 nM) whereas the negative control TP-422 exhibited no significant activity at the tested concentrations:

 

Further, no significant inhibition against BRD9 was found in a NanoBRETTM assay in HEK293 cells:

 

FRAP assays were also performed which showed significant inhibition by TP-238 against BPTF and CECR2:

FL-BPTF FRAP:

FL-CECR2 FRAP:

Materials and Methods

HEK cells were reverse transfected with NL-FL CECR2 or NL-BPTF BD. Cells were treated with a concentration range of TP-238, TP-422 (negative control), with BD070434a tracer (1 µM) for BPTF or BRD-02 tracer (0.5 µM) for CECR2 for 3 hrs before BRET measurements were taken.

For BRD9, the full length protein was used. HEK2983 cells were transfected with NL-BRD9 and treated with tracer (2 µM) and the respective compounds, followed by addition of Nano-Glo substrate and extracellular inhibitor. BRET was determined at 450 and 610nM.

FRAP assays were performed using U2OS cells reverse transfected with GFP-FL FALZ WT or #PHE or GFP-FL CECR2 WT or #ALA for 6-8 hrs. Transfection reagents were replaced with media or 2.5 µM SAHA + incubated O/N. Cells were treated with 1 µM test cpds for 1 hr before imaging. 6 repeat assays were performed for each protein.

references
  1. Footz, TK., Brinkman-Mills, P et al.  Analysis of the cat eye syndrome critical region in humans and the region of conserved synteny in mice: a search for candidate genes at or near the human chromosome 22 pericentromere. Genome Res, 2001,11: 1053–1070. 
  2. Lee SK, Park EJ et al. Genome-wide screen of human bromodomain-containing proteins identifies Cecr2 as a novel DNA damage response protein. Mol Cells, 2012, 34(1):85-91
  3. Wu B, Wang Y, Wang C, Wang GG, Wu J, Wan YY. BPTF Is Essential for T Cell Homeostasis and Function. J Immunol. 2016, 197(11):4325-4333.
  4. Richart L, Real FX, Sanchez-Arevalo Lobo VJ., c-MYC partners with BPTF in human cancer. Mol Cell Oncol. 2016, 3(3)
pk properties
co-crystal structures

A close homologue to TP-248 was co-crystallised to investigate the possible binding mode of the probe:

synthetic schemes
materials and methods