SGC-CK2-1 A chemical probe for CK2

This probe is available from TocrisSigma and Cayman

This control is available from Sigma

overview
Probe Negative control

 

SGC-CK2-1

 

SGC-CK2-1N

Based upon a pyrazolopyrimidine scaffold with potent CK2 inhibition exemplified by AstraZeneca, the SGC has developed a high-quality chemical probe and its negative control for protein kinase CK2.1 This compound shows equal potency in binding to both catalytic subunits of CK2: CK2α (CSNK2A1) and  CK2α' (CSNK2A2). CK2 is a serine/threonine kinase that is part of the larger CMGC family, which is named after the initials of its subfamily members including cyclin-dependent kinases (CDK), mitogen-activated protein kinase (MAPK), glycogen synthase kinase (GSK), and CDC-like kinase (CLK). A plethora of biological functions have been ascribed to CK2 ranging from cell survival and proliferation to inflammation. CK2 phosphorylates more than 300 proteins and, via genetic and biochemical studies in a variety of experimental models, has been found to be both constitutively active and ubiquitously expressed.2-5 While CK2 inhibition has been widely studied in the context of cancer, pervasive use of non-selective CK2 inhibitors has confounded interpretation of results in many oncological studies.

Figure 1: Phylogenetic kinase tree with CK2α and CK2α' highlighted with red circles. Illustration is reproduced courtesy of Cell Signaling Technology, Inc. (www.cellsignal.com)

Key profiling data:

  • Enzymatic assays (Eurofins): CK2α IC50 = 4.2 nM; CK2α' IC50 = 2.3 nM at 10µM ATP
  • Cellular data (nanoBRET): CK2α IC50 = 36 nM; CK2α' IC50 = 16 nM
  • Only 11/403 kinases with PoC <35 when screened at 1 μM
properties
Probe

SGC-CK2-1

SMILES:
CC1=CC=C(C=C1NC(CC)=O)NC2=NC3=C(C=NN3C(NC4CC4)=C2)C#N
InChI:
InChI=1S/C20H21N7O/c1-3-19(28)25-16-8-15(5-4-12(16)2)23-17-9-18(24-14-6-7-14)27-20(26-17)13(10-21)11-22-27/h4-5,8-9,11,14,24H,3,6-7H2,1-2H3,(H,23,26)(H,25,28)
InChIKey: YKDZIFFKQUNVHH-UHFFFAOYSA-N

Physical and chemical properties
Molecular weight375.44
Molecular formulaC20 H21 N7 O
IUPAC nameN-(5-((3-cyano-7-(cyclopropylamino)pyrazolo[1,5-a]pyrimidin-5-yl)amino)-2-methylphenyl)propionamide
clogP1.94
PSA104.91
No. of chiral centres0
No. of rotatable bonds7
No. of hydrogen bond acceptors4
No. of hydrogen bond donors3
StorageStable as a solid at room temperature. DMSO stock solutions (up to 10 mM) are stable at -20oC
DissolutionSoluble up to 10mM in DMSO
Negative control

SGC-CK2-1N

SMILES:

CC1=CC=C(C=C1NC(C2CC2)=O)N(C3=NC4=C(C=NN4C(N(C5CC5)C)=C3)C#N)C
InChI:
InChI=1S/C23H25N7O/c1-14-4-7-18(10-19(14)26-23(31)15-5-6-15)28(2)20-11-21(29(3)17-8-9-17)30-22(27-20)16(12-24)13-25-30/h4,7,10-11,13,15,17H,5-6,8-9H2,1-3H3,(H,26,31)
InChIKey: LGMBLPOARZRSKR-UHFFFAOYSA-N

Physical and chemical properties
Molecular weight415.2
Molecular formulaC23 H25 N7 O
IUPAC name4-((3-(cyclopropyl-formylamino)-4-methyl-phenyl)-methyl-amino)-2-(cyclopropyl-methyl-amino)-1,5,9-triaza-bicyclo[4.3.0]nona-2,4,6,8-tetraene-7-carbonitrile
clogP3.67
PSA63.46
No. of chiral centres0
No. of rotatable bonds8
No. of hydrogen bond acceptors5
No. of hydrogen bond donors1
StorageStable as a solid at room temperature. DMSO stock solutions (up to 10 mM) are stable at -20oC
DissolutionSoluble up to 10mM in DMSO
selectivity profile

SGC-CK2-1 was profiled in the KINOMEscan assay against 403 wild-type kinases at 1 μM. Only 11 kinases showed PoC <35 giving an S(35) at 1 μM = 0.027. Potential off-targets were tested in the nanoBRET target engagement assay (DYRK2) and via Eurofins radiometric and LANCE kinase assays. Data corresponding with off-target kinase activity is shown in the table below.

 

Kinase% Control at 1µMEnzymatic IC50 (nM)NanoBRET IC50 (nM)
CK2α'/CSNK2A202.316
DRAK10>10000NT
CK2α/CSNK2A10.54.236
DYRK2144403700
PLK423>10000NT
HIPK2263400NT
MEK5280% AT 1µMNT
HIPK1323700NT
HIPK3348100NT
RIOK232NTNT
SGK334>10000NT

Figure 1: SGC-CK2-1 was profiled in the KINOMEscan assay against 403 wild-type kinases at 1 μM and off-target kinases inhibited PoC <35 were tested in an orthogonal assay.
 

SGC-CK2-1N was also tested in the DiscoverX panel and 1 kinase had a PoC <35. The negative control was sent to Eurofins for testing in enzyme assays for CSNK2A1 and CSNK2A2. The results are in the table below.

Kinase% Control at 1µMEnzymatic IC50 (nM)NanoBRET IC50 (nM)
RIOK123NTNT
RET35NTNT
FLT340NTNT
BRAF55NTNT
IKK-alpha58NTNT
MKNK158NTNT
STK3361NTNT
TNK162NTNT
EPHA263NTNT
CK2α'/CSNK2A265>10000>10000
CK2α/CSNK2A1100>10000NT

Figure 2: SGC-CK2-1N was profiled in the KINOMEscan assay against 403 wild-type kinases at 1 μM and follow-up CK2α and CK2α' enzymatic assays were done to confirm no activity.

in vitro potency
cell based assay data

A NanoBRET assay was utilized to assess the binding affinity of SGC-CK2-1 to CK2α and CK2α'. The negative control shows no binding affinity for CK2α'.

Figure 1: SGC-CK2-1 and SGC-CK2-1N were profiled in the CK2 NanoBRET assays.

references
  1. Wells, C., Drewry, D. H., Pickett, J. E. & Axtman, A. D. SGC-CK2-1: the first selective chemical probe for the pleiotropic kinase CK2. ChemRxiv, 10.26434/chemrxiv.12296180.v12296181 (2020).
  2. Rabalski, A. J., Gyenis, L. & Litchfield, D. W. Molecular Pathways: Emergence of Protein Kinase CK2 (CSNK2) as a Potential Target to Inhibit Survival and DNA Damage Response and Repair Pathways in Cancer Cells. Clinical Cancer Research 22, 2840-2847, doi:10.1158/1078-0432.Ccr-15-1314 (2016).
  3. Meggio, F. & Pinna, L. A. One-thousand-and-one substrates of protein kinase CK2? FASEB journal : official publication of the Federation of American Societies for Experimental Biology 17, 349-368, doi:10.1096/fj.02-0473rev (2003).
  4. Nuñez de Villavicencio-Diaz, T., Rabalski, A. J. & Litchfield, D. W. Protein Kinase CK2: Intricate Relationships within Regulatory Cellular Networks. Pharmaceuticals 10, 27 DOI: 10.3390/ph10010027 (2017).
  5. Ahmed, K., Gerber, D. A. & Cochet, C. Joining the cell survival squad: an emerging role for protein kinase CK2. Trends in Cell Biology 12, 226-230, doi:10.1016/S0962-8924(02)02279-1 (2002).

Related Links: https://www.tocris.com/products/sgc-ck2-1_7450#product-literature

pk properties
co-crystal structures
synthetic schemes
materials and methods

PFI-6 A novel chemical probe for MLLT1/3

The probe PFI-6 is available from Sigma and Cayman Chemicals.

The negative control PFI-6N is available from Sigma.

overview
Probe Negative control

 

PFI-6

 

PFI-6N

Increasing numbers of papers describing YEATS biology are emerging, and there is a growing need for high quality chemical probes that can be used to study YEATS domains. A collaboration between Pfizer and the SGC has resulted in the discovery of PFI-6, a potent inhibitor of MLLT1/3. PFI-6 has a unique chemotype relative to the current MLLT1/3 chemical probe NVS-MLLT-1.  PFI-6N has also been developed as a structurally similar negative control compound.

properties
Probe

PFI-6

Physical and chemical properties for PFI-6
Molecular weight391.43
Molecular formulaC22H21N3O4
IUPAC name(R0-N-(2,3-dihydro-1H-inden-1-yl)-5-(4-(dimethylcarbamoyl)-3-hydroxyphenyl)isoxazole-3-carboxamide
MollogP2.6
PSA91.23
No. of chiral centres0
No. of rotatable bonds6
No. of hydrogen bond acceptors7
No. of hydrogen bond donors2
Storagestore as a dry poweder or as DMSO stock solutions (10mM) at -20 °C
DissolutionSoluble in DMSO up to 10mM
Negative control

PFI-6N

Physical and chemical properties for PFI-6N
Molecular weight357.41
Molecular formulaC19H23N3O4
IUPAC nameN-cyclopentyl-5-(4-(dimethylcarbamoyl)-3-hydroxyphenyl)-N-methylisoxazole-3-carboxamide
MollogP1.86
PSA82.44
No. of chiral centres0
No. of rotatable bonds6
No. of hydrogen bond acceptors7
No. of hydrogen bond donors1
StorageStore as a dry powder or as DMSO stock solutions (10mM) at -20 °C
DissolutionSoluble in DMSO up to 10mM

SMILES:

PFI-6: O=C(C1=C(C=C(C=C1)C2=CC(C(N[C@@H]3CCC4=C3C=CC=C4)=O)=NO2)O)N(C)C
PFI-6N: O=C(C1=C(C=C(C=C1)C2=CC(C(N(C3CCCC3)C)=O)=NO2)O)N(C)C

InChI:

PFI-6: 1S/C22H21N3O4/c1-25(2)22(28)16-9-7-14(11-19(16)26)20-12-18(24-29-20)21(27)23-17-10-8-13-5-3-4-6-15(13)17/h3-7,9,11-12,17,26H,8,10H2,1-2H3,(H,23,27)/t17-/m1/s1
PFI-6-N: 1S/C19H23N3O4/c1-21(2)18(24)14-9-8-12(10-16(14)23)17-11-15(20-26-17)19(25)22(3)13-6-4-5-7-13/h8-11,13,23H,4-7H2,1-3H3

InChIKey:

PFI-6: IXWUILRSNIQHDM-QGZVFWFLSA-N
PFI-6-N:CKEICVFLYGXFOP-UHFFFAOYSA-N

selectivity profile

PFI-6 shows potent activity on MLLT1/3 (Table 1). PFI-6 is universally inactive in the SGC Bromodomain panel, and showed no activity in an Invitrogen panel of 40 kinases (screening conducted at 10µM). Additionally, PFI-6 showed no activity in a panel of 25 PDEs, ion channels and GPCRs >50µM. The negative control, PFI-6N, was not reactive against MLLT1 >30µM, MLLT3 >30µM, YEATS2 >30µM and YEATS4 >30µM.

 

 MLLT1MLLT3YEATS2YEATS4
HTRF IC50 (µM)0.140.16>40>40
BLI Kd (µM)0.110.11n.d.n.d.
ITCKD (µM)0.0820.076n.d.n.d.
Tm Shift (°C)3.615.130.00-0.02

Table 1: Potency Against Target Family 

Figure 1: PFI-6 specfically inhibits MLLT1 and MLLT3.

in vitro potency
cell based assay data

In the NanoBRET cellular target engagement assay, PFI-6 displayed potent inhibition, with an average IC50 value of 0.76 μM (±0.1) (Figure 1). In comparison, PFI-6N had no inhibitory properties (up to 30 μM) (Figure 1). Further in cell validation using a Fluorescence Recovery After Photobleaching (FRAP) assay was used to confirm target inhibition by PFI-6  (Figure 2) . 

Figure 1: A NanoBRET assay was used to determine MLLT3 target engagement in cells.

Figure 2: Confirmation of MLLT1 target engagement in cells by Fluorescence Recovery After Photobleaching analysis.

 

references
pk properties
co-crystal structures

The figure below shows PFI-6 is soaked into the YEATS domain. PDB codes will be added when they become available. 

 

 

synthetic schemes
materials and methods
15.05.2020

The Viral Interruption Medicines Initiative (VIMI): Open Science coalition in Canada to prevent future pandemics

by: SGC

In an opinion piece to Canada's The Hill Times, Drs Richard Gold, Tania Bubela, Aled Edwards and John Bamforth announced the creation of the Viral Interruption Medicines Initiative (VIMI). The partnership will leverage open science to develop anti-viral drugs in anticipation for future pandemics. 

07.04.2020

Open science drug discovery partnership, READDI, aims to invest $125 million to prevent future pandemics

by: SGC

(Chapel Hill, N.C.— April 7, 2020) — Today, the Structural Genomics Consortium (SGC), the University of North Carolina at Chapel Hill and the Eshelman Institute for Innovation, announce the launch of the Rapidly Emerging Antiviral Drug Development Initiative (READDI), a global organization formed to discover and develop drugs to put “on the shelf” for clinical trial testing in anticipation of future viral pandemics.

NVS-MLLT-1 A Potent and Selective inhibitor of YEATS proteins

This probe is available from Cayman Chemical, Sigma and Tocris.

overview
Probe Negative control

 

NVS-MLLT-1

 

NVS-MLLT-C

MLLT1 (also known as ENL, LTG19 and YEATS1) is a fusion gene partner within the MLL oncogene, involved in chromosome translocation [1]. In MLL1 rearranged mixed lineage leukaemia, MLLT1 acts as fusion partner for the transcriptional co-activation of MLL1. This interaction leads to linkage of MLL1 with the super elongation complex, and histone methyl transferase DOT1L, ultimately driving the dysregulation of oncogenes [2-6].
Accumulating evidence suggests that MLLT1 can also be a driver in non-MLLT1 rearranged acute myeloid leukaemia (AML). The MLLT1 epigenetic reader domain (YEATS domain) recognises acylated lysine residues on histone 3 (mainly acetyl lysine) (Figure 1). In AML, the MLLT1 YEATS domain directly links the epigenetic readout to the oncogenic dysregulation of gene expression [7,8]

MLLT1 CRISPR/Cas9 studies have shown significant reductions in cell proliferation, and invasiveness of AML cell lines. Subsequent rescue with ectopically expressed native MLLT1 restores the disease phenotype, while rescue with mutants that are deficient in the binding pocket (where MLLT1 recognises acyl lysine residues) is unable to restore the phenotype [8]. These studies, combined with the knowledge of known epigenetic readers being used as tractable targets for small molecule inhibitors, indicate that inhibition of MLLT1 may be a viable approach for the treatment of AML. The SGC therefore develped SGC-iMLLT, a first in class potent and selective inhibitor of YEATS proteins [9]. Expanding this work, a probe with improved in cell activity was developed with Novartis. Here, we present NVS-MLLT-1 as a potent and selective inhibitor of YEATS proteins. 

Figure 1: Biology of the Targets.

 

Potency Against Target Family
An AlphaScreen competition assay was used to assess the potency of SGC-iMLLT against all four human YEATS domains. NVS-MLLT-1 showed inhibition of both MLLT1 and MLLT3 at 0.15 µM and 0.254 µM respectively (Table 1). Conversely, the negative control molecule NVS-MLLT-C did not show any inhibition in the assay. More information on MLLT1 as a target can be found in our TEPs repository: https://www.thesgc.org/tep/mllt1.

In vitro assaysNVS-MLLT-1 IC50 (µM)NVS-MLLT-C IC50 (µM)
TR-FRET MLLT10.15> 20
TR-FRET MLLT30.254> 20 
TR-FRET YEATS2> 20> 20
TR-FRET YEATS4> 20> 20
KD BLI MLLT10.075-
KD ITC MLLT10.109-

Table 1: Potency Against Target Family and biophysical properties

Selectivity
NVS-MLLT-1 shoes potent activity on MLLT1/3 whilst being selective for YEATS2/4. NVS-MLLT-1 shows selectivity across all bromodomains (CERC2 IC50 > 40 µM) and no activity was shown in a kinase panel. NVS-MLLT-1 has some activity on ACES and H3 binding. 

Dosage
Use between 1 and 10 µM for cellular assays, and 1 µM for screening at a single shot.

In vitro Activity
In the NanoBRET cellular target engagement assay, NVS-MLLT-1 displayed potent inhibition, with an average IC50 value of 0.5 μM (±0.12) (Figure 2). In comparison,NVS-MLLT-C had no inhibitory properties (up to 30 μM). Further in cell validation was used to confirm target inhibition by NVS-MLLT-1.

 
properties
Probe

NVS-MLLT-1

Physical and chemical properties for NVS-MLLT-1
Molecular weight385.47
Molecular formulaC23H23N5O
IUPAC name(S)-N-(2-((-methylpyrrolidin-1-yl)methyl-1H-benzo[d]imidazel-5-yl)isoquinoline-6-carboxamide
MollogP3.6
PSA69.09
No. of chiral centres1
No. of rotatable bonds5
No. of hydrogen bond acceptors5
No. of hydrogen bond donors2
StorageDMSO stock solutions (up to 50mM) are stable at -20 °C
DissolutionSoluble in DMSO up to 50mM
Negative control

NVS-MLLT-C

Physical and chemical properties for NVS-MLLT-C
Molecular weight385.43
Molecular formulaC22H19N5O2
IUPAC nameN-(2-((2-oxopyrrolidin-1-yl)methyl)-1Hbenzo[d]imidazol-5-yl)isoquinoline-6-carboxamide
MollogP 
PSA86.16
No. of chiral centres0
No. of rotatable bonds5
No. of hydrogen bond acceptors6
No. of hydrogen bond donors2
StorageDMSO stock solutions (up to 50mM) are stable at -20 °C
DissolutionSoluble in DMSO up to 50mM

SMILES:

NVS-MLLT-1: C[C@H]1CCCN1CC2=NC3=CC(NC(C4=CC5=CC=NC=C5C=C4)=O)=CC=C3N2
NVS-MLLT-C: O=C(C1=CC2=CC=NC=C2C=C1)NC3=CC=C(N4)C(N=C4CN5C(CCC5)=O)=C3

InChI:

NVS-MLLT-1: 1S/C23H23N5O/c1-15-3-2-10-28(15)14-22-26-20-7-6-19(12-21(20)27-22)25-23(29)17-4-5-18-13-24-9-8-16(18)11-17/h4-9,11-13,15H,2-3,10,14H2,1H3,(H,25,29)(H,26,27)/t15-/m0/s1
NVS-MLLT-C: 1S/C22H19N5O2/c28-21-2-1-9-27(21)13-20-25-18-6-5-17(11-19(18)26-20)24-22(29)15-3-4-16-12-23-8-7-14(16)10-15/h3-8,10-12H,1-2,9,13H2,(H,24,29)(H,25,26)

InChIKey:

NVS-MLLT-1: ZRTFTZCKJUNZIU-HNNXBMFYSA-N
NVS-MLLT-C: GMSVIEOZPBLYCQ-UHFFFAOYSA-N

selectivity profile

The AlphaScreen assays were employed to screen NVS-MLLT-1 and NVS-MLLT-C against a panel of bromodomains. NVS-MLLT-1 shows potent activity on MLLT1/3 whilst being selective for YEATS2/4. NVS-MLLT-1 shows selectivity across all bromodomains (CERC2 IC50 > 40 μM). NVS-MLLT-1 shows no activity on kinases tested, NVS-MLLT-1 has some activity on ACES and E3 binding. NVS-MLLT-C shows no activity in a panel of kinases, and no activties on other targets tested. 

In vitro assaysNVS-MLLT-1 IC50 (µM)NVS-MLLT-C IC50 (µM)
TR-FRET MLLT10.15> 20
TR-FRET MLLT30.254> 20 
TR-FRET YEATS2> 20> 20
TR-FRET YEATS4> 20> 20
KD BLI MLLT10.075-
KD ITC MLLT10.109-

Table 1: Potency Against Target Family and biophysical properties

General Selectivity- NVS-MLLT-1

NVS-MLLT-1 shows no activity on kinases tested. NVS-MLLT-1 has some activity on ACES and H3 binding.

General Selectivity- NVS-MLLT-C

NVS-MLLT-C shows no activity on kinases tested. NVS-MLLT-C shows no activities on other targets tested

in vitro potency
cell based assay data

Cellular target Engagement

In the NanoBRET cellular target engagement assay, NVS-MLLT-1 displayed potent inhibition, with an average IC50 value of 0.5 μM (±0.12) (Figure 1). In comparison, NVS-MLLT-C had no inhibitory properties (up to 30 μM) (Figure 1). Further in cell validation using a Fluorescence Recovery After Photobleaching (FRAP) assay was used to confirm target inhibition by NVS-MLLT-1  (Figure 2) . 

Figure 1: A NanoBRET assay was used in HEK293 cells to determine target engagement in cells. All probes were tested for 24h in the presence of 2.5 μM SAHA (24h). Graph represents Mean±SEM, n=3 independent replicates, with n=4 technical replicates.  mBU: BRET units

Figure 2: Further confirmation of MLLT1 target engagement in cells.  Fluorescence Recovery After Photobleaching analysis shows NVS-MLLT-1 decreases the half-life recovery time of cells. n=3 independent experiments, Mean±SEM, fold change from SAHA treatment alone. One way ANOVA, Dunnett's multiple comparisons.

Materials and Methods

NanoLuciferase Bioluminescent Resonance Energy Transfer (NanoBRET) Assay 
Cellular activity against MLLT3 was assessed using a NanoBRET assay. HEK293 cell (8 x 105) were plated a 6-well plate, after 6h cells were co-transfected with C-terminal HaloTag-Histone 3.3 (NM_002107) and an N-terminal NanoLuciferase fusion of MLLT3 (original MLLT3 WT sequences from Promega HaloTag® human ORF in pFN21A or MLLT3 MUT - Y78A Tyrosine is changed to an Alanine) at a 1:10 (NanoLuc® to HaloTag®) ratio respectively with FuGENE HD transfection reagent. Sixteen hours post-transfection, cells were collected, washed with PBS, and exchanged into media containing phenol red-free DMEM and 4% FBS in the absence (control sample) or the presence (experimental sample) of 100 nM NanoBRET 618 fluorescent ligand (Promega). Cells were then re-plated in a 96-well assay white plate (Corning Costar #3917) at 2x104 cells per well. Compounds were then added directly to the cells (in the presence of SAHA 2.5 µM) at final concentrations 0-30 μM or an equivalent amount of DMSO as a vehicle control, and the plates were incubated for 24 h at 37°C in the presence of 5% CO2. NanoBRET Nano-Glo substrate (Promega) was added to both control and experimental samples at a final concentration of 10 µM. Readings were performed within 10 minutes using a ClarioSTAR (BMG Labtech) equipped with 460 nm and 610 nm filters. A corrected BRET ratio was calculated and is defined as the ratio of the emission at 610 nm/460 nm for experimental samples minus the emission at 610 nm/460 nm for control samples (without NanoBRET fluorescent ligand). BRET ratios are expressed as milliBRET units (mBU), where 1 mBU corresponds to the corrected BRET ratio multiplied by 1000.

Peptide Displacement Assay
Peptide displacement assays were set up with biotinylated peptides (chosen based on ChIPseq data from the literature and purchased from LifeTein) and 6His tagged protein (Table 2).  For detection, two orthogonal technologies were used, i. AlphaScreen® technology from Perkin Elmer and ii. HTRF from Cisbio.  Compounds were dispensed in duplicate at single concentration (100 µM) for the initial screen and as 11-point dose response curves starting from 200 µM for IC50 value determination.

YEATS domainPeptide shorthandPositionPeptide sequence
MLLT1H3K18ac12-30GGKAPR(K-acetyl)QLATKAARKSAPY(K-biotin)
MLLT3H3K9ac2-20ARTKTAR(K-acetyl)STGGKAPRKQLY(K-biotin)
YEATS2H3K27cro15-32biotin-GKPRKQLATAAR(K-crotonyl)SAPAT
YEATS4H3K27cro15-32biotin-GKPRKQLATAAR(K-crotonyl)SAPAT

Table 2: Peptides used for the peptide displacement assays

To determine optimal assay conditions for each protein prep, proteins and peptides were titrated against each other in a 16 by 16 matrix in 1:1 dilutions, starting from 3.2 µM. For the final ratio of protein and peptide to use in the assay, the point representing the EC90 in the two-dimensional titration was chosen.  Typically, final assay concentrations for protein and peptide fell between 25 and 200 nM.  For AlphaScreen®, AlphaScreen Histidine (Nickel Chelate) Detection Kit donor and acceptor beads were used at a 1:2500 dilution from purchased stock; for HTRF, SA-XL665 and anti-6His antibody were used at 1:2000 and 1:10000 dilution from purchased stock, respectively.  Assays were performed on 384 well ProxiPlates (Perkin Elmer) at a final volume of 20 µl and plates were read using a Pherastar FSX plate reader (BMG Labtech).

Isothermal Titration Calorimetry
Isothermal titration calorimetry (ITC) was carried out using a TA NanoITC (standard volume) instrument.  Protein was prepared by dialysis (overnight at 4°C) against a ~1000 times excess of buffer (20 mM Tris at pH 7.5, 500 mM NaCl, 5% (v/v) glycerol, 2 mM DTT) using SnakeSkin® Dialysis Tubing with a 7 kDa MWCO and then concentrated to 300 µM.  The experiment was carried out at 20°C in reverse mode with the compound in the cell at 50 µM and the protein in the syringe at 300 µM due to the solubility of the compound with the first injection at 4 µl and the following 30 at 8 µl. Data was analysed using the NanoAnalyze software package by TA Instruments.

Differential Scanning Fluorimetry
Differential scanning fluorimetry (DSF) to determine the effect of compounds on the thermal stability of proteins (DTm) was carried out on 384 well PCR plates using a LightCycler 480 (Roche).  Protein at 10 µM was buffered in 10 mM HEPES at pH 7.5 and 500 mM NaCl.  The experiment was carried out from 25 to 95°C with three acquisitions per degree.  Compounds were added at 50 µM final concentration and DMSO reference and no-addition controls were also collected.

references

References

1. Doty, R.T., et al., The leukemia-associated gene Mllt1/ENL: characterization of a murine homolog and demonstration of an essential role in embryonic development. Blood Cells Mol Dis, 2002. 28(3): p. 407-17.https://www.ncbi.nlm.nih.gov/pubmed/12367585

2. Mueller, D., et al., A role for the MLL fusion partner ENL in transcriptional elongation and chromatin modification. Blood, 2007. 110(13): p. 4445-54.DOI: 10.1182/blood-2007-05-090514.https://www.ncbi.nlm.nih.gov/pubmed/17855633

3. Mueller, D., et al., Misguided transcriptional elongation causes mixed lineage leukemia. PLoS Biol, 2009. 7(11): p. e1000249.DOI: 10.1371/journal.pbio.1000249.https://www.ncbi.nlm.nih.gov/pubmed/19956800

4. Yokoyama, A., et al., A higher-order complex containing AF4 and ENL family proteins with P-TEFb facilitates oncogenic and physiologic MLL-dependent transcription. Cancer Cell, 2010. 17(2): p. 198-212.DOI: 10.1016/j.ccr.2009.12.040.https://www.ncbi.nlm.nih.gov/pubmed/20153263

5. Biswas, D., et al., Function of leukemogenic mixed lineage leukemia 1 (MLL) fusion proteins through distinct partner protein complexes. Proc Natl Acad Sci U S A, 2011. 108(38): p. 15751-6.DOI: 10.1073/pnas.1111498108.https://www.ncbi.nlm.nih.gov/pubmed/21896721

6. Monroe, S.C., et al., MLL-AF9 and MLL-ENL alter the dynamic association of transcriptional regulators with genes critical for leukemia. Exp Hematol, 2011. 39(1): p. 77-86 e1-5.DOI: 10.1016/j.exphem.2010.09.003.https://www.ncbi.nlm.nih.gov/pubmed/20854876

7. Erb, M.A., et al., Transcription control by the ENL YEATS domain in acute leukaemia. Nature, 2017. 543(7644): p. 270-274.DOI: 10.1038/nature21688.https://www.ncbi.nlm.nih.gov/pubmed/28241139

8. Wan, L., et al., ENL links histone acetylation to oncogenic gene expression in acute myeloid leukaemia. Nature, 2017. 543(7644): p. 265-269.DOI: 10.1038/nature21687.https://www.ncbi.nlm.nih.gov/pubmed/28241141

9. Christott, T. et al., Discovery of a selective inhibitor for the YEATS domains of ENL/AF9. SLAS Discov, 2019. 24(2):133-141. DOI: 10.1177/2472555218809904. https://www.ncbi.nlm.nih.gov/pubmed/30359161

10. Chaikuad, A., S. Knapp, and F. von Delft, Defined PEG smears as an alternative approach to enhance the search for crystallization conditions and crystal-quality improvement in reduced screens. Acta Crystallogr D Biol Crystallogr, 2015. 71(Pt 8): p. 1627-39.DOI: 10.1107/S1399004715007968.https://www.ncbi.nlm.nih.gov/pubmed/26249344

11. Moustakim, M., et al., Discovery of an MLLT1/3 YEATS Domain Chemical Probe. Angew Chem Int Ed Engl, 2018. 57(50): p. 16302-16307.DOI: 10.1002/anie.201810617.https://www.ncbi.nlm.nih.gov/pubmed/30288907

12. Heidenreich, D., et al., Structure-Based Approach toward Identification of Inhibitory Fragments for Eleven-Nineteen-Leukemia Protein (ENL). J Med Chem, 2018. 61(23): p. 10929-10934.DOI: 10.1021/acs.jmedchem.8b01457.https://www.ncbi.nlm.nih.gov/pubmed/30407816

pk properties

In vitro Metabolic stability of NVS-MLLT-1

To establish the in cell half life of NVS-MLLT-1, metabolic stability studies were carried out exposing nominated compounds to samples of primary human hepatocytes. Results from ADME and phys chem experiments are summarised below. 

In vitro Metabolism (Liver microsomes)NVS-MLLT-1
[CL int(µL/min/mg)])
Human 89
Rat 68
Mouse 90

Table 1: In vitro metabolism

Permeability

NVS-MLLT-1
PAMPA:
Log PAMPA (cm/s) -5.1
% FA  49
Low Efflux MDCK:
Papp(A-B)(x10-6 )11
% FA80-100
% Recovery 81

Table 2: PhysChem Properties: Permeability 

Solubility and pKa

NVS-MLLT-1
Equilibrium Solubility 
pH 6.8 (g/L)0.375
pKa14.6
pKa2 2.1

Table 3: PhysChem Properties: Solubility and pKa

Solubility and pKa

NVS-MLLT-1
clogP3.6
logP (octanol)2.8

Table 4: PhysChem Properties: Lipophilicity

co-crystal structures

The figures below shows MLLT1 co-crystal strutcure at 1.7 Å. NVS-MLLT-1 is soaked into the YEATS domain. PDB codes will be added when they become available. 

synthetic schemes
materials and methods
28.02.2020

The Chordoma Foundation and The Mark Foundation for Cancer Research fund open-source research to accelerate drug discovery following recent breakthroughs for chordoma, a rare bone cancer with few treatment options

by: SGC

February 28, 2020 - The Chordoma Foundation (CF) and The Mark Foundation for Cancer Research (MFCR) announced today a two-year, $1.4M partnership with a team of researchers at three institutions to develop new treatments for chordoma, a rare and difficult-to-treat bone cancer. The researchers will focus on creating the first drugs to inhibit a protein known as brachyury.

SGC-CLK-1 A chemical probe for CLK1, CLK2, and CLK4

This probe is available at Cayman Chemicals and Sigma.

Click here to obtain the control.

overview
Probe Negative control

 

SGC-CLK-1

 

SGC-CLK-1N

In collaboration with scientists at Luceome Biotechnologies the SGC has developed a chemical probe for the kinases CLK1, CLK2, and CLK4 and a corresponding closely related negative control compound.

Figure 1: Phylogenetic kinase tree with CLK1, CLK2, CLK3, and CLK4 highlighted with red circles. Illustration is reproduced courtesy of Cell Signaling Technology, Inc. (www.cellsignal.com).

The four members of the CDC Like Kinases (CLKs) family (CLK1, CLK2, CLK3, and CLK4) are found in the CMGC branch of the kinome. They are central to RNA-splicing events in cells. CLKs play a role in pre-mRNA splicing is by regulating serine-arginine-rich (SR) proteins. When the SR proteins are phosphorylated by CLKs they relocate to the spliceosome and interact with pre-mRNA to facilitate exon recognition in the splicing machinery. Cancer can gain a survival advantage when the splicing machinery is “hijacked”, so splicing machinery proteins are often mutated or overexpressed in cancers. CLK1, CLK2, and CLK4 for example are overexpressed in renal cancer, breast cancer, colorectal cancer, liver cancer, and glioblastoma. One report suggests a potential vulnerability to CLK inhibitors in cancers with MYC amplification. These findings suggest that CLK inhibitors may be useful for the treatment of some cancers. CLK chemical probes will also help tease out details of the splicing process.

Biological activity summary:

  • Binding assays (Luceome Biotechniology) CLK1 IC50 = 41 nM; CLK2 IC50 = 36 nM, CLK3 inactive at 10,000 nM
  • Cellular data (nanoBRET): CLK1 IC50 = 165 nM; CLK2 IC50 = 70 nM; CLK4 IC50 = 100 nM
  • Only 6/403 kinases with PoC<35 when screened at 1 µM.
properties
Probe

SGC-CLK-1

Physical and chemical properties for SGC-CLK-1
Molecular weight416.4
Molecular formulaC19H15F3N6O2
IUPAC nameN-(3-methoxy-5-(trifluoromethyl)phenyl)-4-(6-methoxypyrazolo[1,5-b]pyridazin-3-yl)pyrimidin-2-amine
MollogP3.42
PSA86.46
No. of chiral centres0
No. of rotatable bonds6
No. of hydrogen bond acceptors9
No. of hydrogen bond donors1
StorageStable as a solid at room temperature.  DMSO stock solutions (up to 10mM) are stable at -20 °C
DissolutionSoluble in DMSO up to 10 mM. 
Negative control

SGC-CLK-1N

Physical and chemical properties for SGC-CLK-1N
Molecular weight430.4
Molecular formulaC20H17F3N6O2
IUPAC nameN-(3-methoxy-5-(trifluoromethyl)phenyl)-4-(6-methoxypyrazolo[1,5-b]pyridazin-3-yl)-6-methylpyrimidin-2-amine
MollogP3.81
PSA86.46
No. of chiral centres0
No. of rotatable bonds6
No. of hydrogen bond acceptors9
No. of hydrogen bond donors1
StorageStable as a solid at room temperature.  DMSO stock solutions (up to 10mM) are stable at -20 °C
DissolutionSoluble in DMSO

SMILES:
SGC-CLK-1: COC1=NN2N=CC(C3=NC(NC4=CC(C(F)(F)F)=CC(OC)=C4)=NC=C3)=C2C=C1
SGC-CLK-1N: COC1=NN2N=CC(C3=NC(NC4=CC(C(F)(F)F)=CC(OC)=C4)=NC(C)=C3)=C2C=C1

InChI:
SGC-CLK-1: InChI=1S/C19H15F3N6O2/c1-29-13-8-11(19(20,21)22)7-12(9-13)25-18-23-6-5-15(26-18)14-10-24-28-16(14)3-4-17(27-28)30-2/h3-10H,1-2H3,(H,23,25,26)
SGC-CLK-1N: InChI=1S/C20H17F3N6O2/c1-11-6-16(15-10-24-29-17(15)4-5-18(28-29)31-3)27-19(25-11)26-13-7-12(20(21,22)23)8-14(9-13)30-2/h4-10H,1-3H3,(H,25,26,27)

InChIKey:
SGC-CLK-1: GJYVLTPTDBQQCY-UHFFFAOYSA-N
SGC-CLK-1N: BJVQXSHZMUFJBQ-UHFFFAOYSA-N

selectivity profile

SGC-CLK-1 was profiled in the KINOMEscan assay against of 403 wild-type kinases at 1 µM. Only 6 kinases showed PoC < 35 giving an S(35) at 1 µM =0.02. Potential off-targets were tested in the nanoBRET target engagement assay where possible and the data are shown in the table.

Figure 1: SGC-CLK-1 was profiled in the KINOMEscan assay against of 403 wild-type kinases at 1 µM.

SGC-CLK-1N was also tested in the Discoverx panel and no kinases had PoC<45. The negative control was sent to Eurofins for testing in enzyme assays for CLKs and typical CLK inhibitor off targets. These results are in the table below.

Figure 2: Eurofins enzymatic screening results for SGC-CLK-1.
 

in vitro potency
cell based assay data

A Nanobret assay was utilised to assess the binding affinity of SGC-CLK-1 to CLK1, CLK2 and CLK4 (Figure 1). The negative control shows no binding affinity. 

Figure 1: SGC-CLK-1 was profiled in the Nanobret assay.

references

1. Araki, S., et al., Inhibitors of CLK protein kinases suppress cell growth and induce apoptosis by modulating pre-mRNA splicing. PLoS One, 2015. 10(1): p. e0116929.DOI: 10.1371/journal.pone.0116929. https://www.ncbi.nlm.nih.gov/pubmed/25581376

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. Corkery, D.P., et al., Connecting the speckles: Splicing kinases and their role in tumorigenesis and treatment response. Nucleus, 2015. 6(4): p. 279-88.DOI: 10.1080/19491034.2015.1062194. https://www.ncbi.nlm.nih.gov/pubmed/26098145

4. Funnell, T., et al., CLK-dependent exon recognition and conjoined gene formation revealed with a novel small molecule inhibitor. Nat Commun, 2017. 8(1): p. 7.DOI: 10.1038/s41467-016-0008-7. https://www.ncbi.nlm.nih.gov/pubmed/28232751

5. Iwai, K., et al., Anti-tumor efficacy of a novel CLK inhibitor via targeting RNA splicing and MYC-dependent vulnerability. EMBO Mol Med, 2018. 10(6).DOI: 10.15252/emmm.201708289. https://www.ncbi.nlm.nih.gov/pubmed/29769258

6. jain, P., et al., Human CDC2-like kinase 1 (CLK1): a novel target for Alzheimer's disease. Curr Drug Targets, 2014. 15(5): p. 539-50.DOI: 10.2174/1389450115666140226112321. https://www.ncbi.nlm.nih.gov/pubmed/24568585

7.  Koedoot, E., et al., Splicing regulatory factors in breast cancer hallmarks and disease progression. Oncotarget, 2019. 10(57): p. 6021-6037.DOI: 10.18632/oncotarget.27215. https://www.ncbi.nlm.nih.gov/pubmed/31666932

8.  Sako, Y., et al., Development of an orally available inhibitor of CLK1 for skipping a mutated dystrophin exon in Duchenne muscular dystrophy. Sci Rep, 2017. 7: p. 46126.DOI: 10.1038/srep46126. https://www.ncbi.nlm.nih.gov/pubmed/28555643

pk properties
co-crystal structures
synthetic schemes
materials and methods

SGC-STK17B-1 A chemical probe for STK17B/DRAK2 kinase

The probe is available from Cayman and Sigma, and the control is available from Sigma.

overview
Probe Negative control

 

SGC-STK17B-1

 

SGC-STK17B-1N

Chemical structures of SGC-STK17B-1 (thieno[3,2-d]pyrimidine) and the negative control SGC-STK17B-1N (thieno[2,3-d]pyrimidine).

In a collaboration with Pfizer, the SGC has developed a high quality chemical probe and its negative control for protein kinase STK17B/DRAK2, Figure 1, which is a serine/threonine kinase part of the larger Death-associated protein kinase (DAPK) family that also includes DAPK1, DAPK2, DAPK3, and DRAK1. STK17B kinase is predominantly expressed in T-cells and B-cells. Due to this expression it has been identified as a possible therapeutic target for Type 1 diabetes, Multiple sclerosis and graft rejection. STK17B knock out mice are viable and show no abnormalities, but are remarkably resistant to autoimmune challenge. When crossed onto the NOD1 strain, a model of Type 1 diabetes1 or challenged with EAE, a model of Multiple Sclerosis, the STK17B knock out mice are protected from disease progression.2 STK17B is also overexpressed in hepatocellular3 and breast cancer.4 Silencing of STK17B in cells suggests that it may have utility in treatment of these cancers. The starting point in the development of this chemical probe was a series of thienopyrimidines donated by Pfizer, prepared in an effort targeting TPL2 kinase.5 From here, the SGC developed a library of small molecules inhibitors around the thieno[3,2-d]pyrimidine core optimizing for potency, selectivity, and cell activity and from this set, we identified SGC-STK17B-1 as a small molecule chemical probe.

Figure 1. Phylogenetic kinase tree, STK17B/DRAK2 and ST17A/DRAK1 highlighted with red circles. Illustration is reproduced courtesy of Cell Signaling Technology, Inc. (www.cellsignal.com).

Biological activity summary

Selectivity
SGC-STK17B-1 was profiled in the KINOMEscan assay against of 403 wild-type kinases @ 1 mM, followed by Kd determination of potential off-targets at Eurofins. SGC-STK17B-1 shown >30 fold selective (Kd) against the closest off targets STK17A/DRAK1, AURKB and CaMKK2. SGC-STK17B-1 is non-toxic in cells (HEK293 cells) in a time and dose dependent manner and have little to no effect on cell viability.

Cell-based assay
In cellular target engagement assays (NanoBRET) in HEK293 cells, SGC-STK17B-1 showed IC50 of 190 nM against STK17B (n = 9) and >10,000 nM against DRAK1 and AURKB (potential off-targets). The negative control SGC-STK17B-1N shown IC50 > 10,000 nM against STK17B.

Binding Activity
In a binding assay performed at Luceome Biotechnologies SGC-STK17B-1 showed potency of 43 nM against STK17B.

Acknowledgments. NanoBRET cellular assays were performed by Carrow Wells and Julie Pickett at SGC-UNC Chapel Hill.

properties
Probe

SGC-STK17B-1

Physical and chemical properties for SGC-STK17B-1
Molecular weight358.46
Molecular formulaC16H10N2O2S3
IUPAC name2-((6-(benzo[b]thiophen-2-yl)thieno[3,2-d]pyrimidin-4-yl)thio)acetic acid
MollogP4.00
PSA144.86
No. of chiral centres0
No. of rotatable bonds4
No. of hydrogen bond acceptors4
No. of hydrogen bond donors1
StorageStable as a solid at room temperature.  DMSO stock solutions (up to 10mM) are stable at -20 °C
DissolutionSoluble in DMSO up to 10 mM. 
Negative control

SGC-STK17B-1N

Physical and chemical properties for SGC-STK17B-1N
Molecular weight358.46
Molecular formulaC16H10N2O2S3
IUPAC name2-((6-(benzo[b]thiophen-2-yl)thieno[2,3-d]pyrimidin-4-yl)thio)acetic acid
MollogP4.09
PSA144.86
No. of chiral centres0
No. of rotatable bonds4
No. of hydrogen bond acceptors4
No. of hydrogen bond donors1
StorageStable as a solid at room temperature.  DMSO stock solutions (up to 10mM) are stable at -20 °C.
DissolutionSoluble in DMSO up to 10 mM. 

SMILES:
SGC-STK17B-1: O=C(CSC1=C2C(C=C(S2)C3=CC4=C(S3)C=CC=C4)=NC=N1)O
SGC-STK17B-1N: O=C(CSC1=C2C(SC(C3=CC4=C(S3)C=CC=C4)=C2)=NC=N1)O

InChI:
SGC-STK17B-1: 1S/C16H10N2O2S3/c19-14(20)7-21-16-15-10(17-8-18-16)6-13(23-15)12-5-9-3-1-2-4-11(9)22-12/h1-6,8H,7H2,(H,19,20)
SGC-STK17B-1N: 1S/C16H10N2O2S3/c19-14(20)7-21-15-10-6-13(23-16(10)18-8-17-15)12-5-9-3-1-2-4-11(9)22-12/h1-6,8H,7H2,(H,19,20)

InChIKey:
SGC-STK17B-1: BNYXRPMABDQJJR-UHFFFAOYSA-N
SGC-STK17B-1N: RRIRDIKTUQVZCW-UHFFFAOYSA-N

selectivity profile

The KINOMEscan of SGC-STK17B-1 at 1mM showed few off-targets, Figure 1. With cutoff 10% control (90% inhibition), MET, NEK6, PIM2, WEE1, and CAMKK2 are shown in red circles in the TREEspot. This screening was followed by Kd determination and NanoBRET assays for several potential off-targets (Table 1).


Figure 1:  Kinome selectivity and TREEspot analysis of 403 wild-type kinases for SGC-STK17B-1.

Results in Table 1 confirmed the high selectivity of SGC-STK17B-1 towards STK17B/DRAK2.  Non-off targets were found for this chemical probe at concentrations shown.

 

Entry

Kinase

% Control (1 µM)

Enzyme assay         
 Kd (nM)

Cell assay (NanoBRET) IC50 (nM)

1

MET

0

>10,000

n.t

2

NEK6

0

>10,000

n.t

3

PIM2

0

nt

n.t

4

WEE1

0

>10,000

n.t

5

STK17B

0.9

34

190

6

CAMKK2

7.8

>10,000

n.t

7

AURKB

14

8947

>10,000

8

CAMKK1

14

5183

n.t

9

STK38L

29

>10,000

>10,000

10

CDKL1

40

>10,000

n.t

11

HIPK1

41

>10,000

>10,000

12

HIPK4

41

4352

>10,000

13

HIPK2

53

8612

>10,000

14

STK17A

73

4666

>10,000

15

STK16

77

648

n.t

in vitro potency
cell based assay data
references
  1. Farag, A.K. and E.J. Roh, Death-associated protein kinase (DAPK) family modulators: Current and future therapeutic outcomes. Med Res Rev, 2019. 39(1): p. 349-385.DOI: 10.1002/med.21518. https://www.ncbi.nlm.nih.gov/pubmed/29949198
  2. Lan, Y., et al., STK17B promotes carcinogenesis and metastasis via AKT/GSK-3beta/Snail signaling in hepatocellular carcinoma. Cell Death Dis, 2018. 9(2): p. 236.DOI: 10.1038/s41419-018-0262-1. https://www.ncbi.nlm.nih.gov/pubmed/29445189
  3. McGargill, M.A., et al., Drak2 regulates the survival of activated T cells and is required for organ-specific autoimmune disease. J Immunol, 2008. 181(11): p. 7593-605.DOI: 10.4049/jimmunol.181.11.7593. https://www.ncbi.nlm.nih.gov/pubmed/19017948
  4. Ni, Y., et al., Identification and SAR of a new series of thieno[3,2-d]pyrimidines as Tpl2 kinase inhibitors. Bioorg Med Chem Lett, 2011. 21(19): p. 5952-6.DOI: 10.1016/j.bmcl.2011.07.069. https://www.ncbi.nlm.nih.gov/pubmed/21862328
  5. Yang, K.M., et al., DRAK2 participates in a negative feedback loop to control TGF-beta/Smads signaling by binding to type I TGF-beta receptor. Cell Rep, 2012. 2(5): p. 1286-99.DOI: 10.1016/j.celrep.2012.09.028. https://www.ncbi.nlm.nih.gov/pubmed/23122956
  6. Picado et al. A Chemical Probe for Dark Kinase STK17B Derives Its Potency and High Selectivity through a Unique P-Loop Conformation. J Med Chem 2020. 63 (23): p14626. https://doi.org/10.1021/acs.jmedchem.0c01174. https://pubs.acs.org/doi/10.1021/acs.jmedchem.0c01174.
pk properties
co-crystal structures

The co-crystal structure shown an unusual flip of R41 (P-loop) forming salt bridge network with inhibitor COOH group, R41 and E117 in one of the two experimentally observed conformations is a likely mechanism of selectivity. PDB depositions for the chemical probe are 6Y6F and 6Y6H (links will be updated as soon as they become available). 

 

Important features:

  • Type-I binding mode
  • Back pocket interaction of COOH with lysine (K62)
  • Additional ethylene diol fills back pocket

Acknowledgments.  Co-crystal structure was obtained by Apirat Chaikuad and Stefan Knapp at SGC-Frankfurt.

synthetic schemes
materials and methods

SGC-CAMKK2-1 A chemical probe for CAMKK2/CAMKK1

The probe is available from Sigma and Tocris, and the control is available from Sigma.

overview
Probe Negative control

 

SGC-CAMKK2-1

 

SGC-CAMKK2-1N

Chemical structures of furopyridines SGC-CAMKK2-1 (CAMKK2/CAMKK1 chemical probe) and SGC-CAMKK2-1N (CAMKK2/CAMKK1 negative control)

In collaboration with scientists at St. Vincent’s Institute at the University of Melbourne, MD Anderson, Duke University, and Baylor College of Medicine, the SGC has developed a chemical probe for the kinase CAMKK2 and a corresponding closely related negative control compound. Because of the active site similarity of CAMKK1 and CAMKK2, it is likely that both targets are engaged. When we have gathered appropriate CAMKK1 data this page will be updated. Until such data becomes available, we recommend that users of the probe consider both CAMKK1 and CAMKK2 as targets.

CAMKK2 is a serine-threonine protein kinase and a member of the “Other” group of protein kinases (Figure 1). CAMKK2 is primarily expressed in the brain, with lower levels in some peripheral tissues including thymus, spleen, lung, and testis.

Figure 1. Phylogenetic kinase tree, CAMKK1/CAMKK2 highlighted with red circles. Illustration is reproduced courtesy of Cell Signaling Technology, Inc. (www.cellsignal.com).

CAMKK2 is activated by binding to a calcium/calmodulin complex, and activated CAMKK2 can phosphorylate CAMK1, CAMK4, AMPK, and AKT (Figure 2). Activation of these kinases regulates a number of important processes. Because of its involvement in so many critical cellular pathways, CAMKK2 activity has been linked to several diseases. For example, CAMKK2 is upregulated and/or overexpressed in several cancers (Inlcuding gastric, HCC, glioma, and prostate). As such, CAMKK2 inhibitors hold promise as potential therapeutics. STO-609 is used widely as a CAMKK2 inhibitor tool molecule. Although not promiscuous, it has off targets that may confound interpretation of cellular screening results, so a more selective chemical probe or a chemical probe with a different scaffold and different off-target effects will benefit the community.

Figure 2. Overview of the CAMKK2 signaling pathway.

 

Biological activity summary

  • SGC-CAMKK2-1 inhibits purified CAMKK2 with an IC50 = 30 nM.
  • In a kinome wide screen (Discoverx) against 404 kinases at a screening concentration of 1000 nM, only CAMKK2 and CAMKK1 showed binding with PoC <15 (CAMKK2 PoC = 5.4; CAMKK1 PoC = 12) indicating excellent selectivity.
  • SGC-CAMKK2-1 inhibits phosphorylation of AMPK in C4-2 cells (in cell Western) with an IC50 of 1.6 µM.

SGC-CAMKK2-1N is a suitable structurally similar negative control. It has an IC50 = 27 µM on purified CAMKK2, and cellular potency > 10,000 µM.

properties
Probe

SGC-CAMKK2-1

Physical and chemical properties for SGC-CAMKK2-1
Molecular weight397.5
Molecular formulaC26H23NO3
IUPAC name2-cyclopentyl-4-(5-(m-tolyl)furo[2,3-b]pyridin-3-yl)benzoic acid
MollogP5.53
PSA63.3
No. of chiral centres0
No. of rotatable bonds4
No. of hydrogen bond acceptors4
No. of hydrogen bond donors1
StorageStable as a solid in the dark at -20°C. NB making aliquots rather than freeze-thawing is recommended. DMSO stock solutions (up to 10mM) are stable at -20 °C
DissolutionSoluble in DMSO up to 10 mM. 
Negative control

SGC-CAMKK2-1N

Physical and chemical properties for SGC-CAMKK2-1N
Molecular weight349.8
Molecular formulaC20H12ClNO3
IUPAC name2-chloro-4-(5-phenylfuro[2,3-b]pyridin-3-yl)benzoic acid
MollogP4.37
PSA63.33
No. of chiral centres0
No. of rotatable bonds3
No. of hydrogen bond acceptors4
No. of hydrogen bond donors1
StorageStable as a solid at room temperature.  DMSO stock solutions (up to 10mM) are stable at -20 °C.
DissolutionSoluble in DMSO up to 10 mM. 

SMILES:
SGC-CAMKK2-1: CC1=CC=CC(C2=CC3=C(N=C2)OC=C3C4=CC(C5CCCC5)=C(C=C4)C(O)=O)=C1
SGC-CAMKK2-1N: ClC1=C(C=CC(C2=COC3=C2C=C(C=N3)C4=CC=CC=C4)=C1)C(O)=O

InChI:
SGC-CAMKK2-1:1S/C26H23NO3/c1-16-5-4-8-18(11-16)20-13-23-24(15-30-25(23)27-14-20)19-9-10-21(26(28)29)22(12-19)17-6-2-3-7-17/h4-5,8-15,17H,2-3,6-7H2,1H3,(H,28,29)
SGC-CAMKK2-1N: 1S/C20H12ClNO3/c21-18-9-13(6-7-15(18)20(23)24)17-11-25-19-16(17)8-14(10-22-19)12-4-2-1-3-5-12/h1-11H,(H,23,24)

InChIKey:
SGC-CAMKK2-1: TXIYVFVMXNFNRX-UHFFFAOYSA-N
SGC-CAMKK2-1N: KHPDNSBTNDXSBL-UHFFFAOYSA-N

selectivity profile

SGC-CAMKK2-1 was profiled in the KINOMEscan assay against 403 wild-type kinases at 1 µM. Only two kinases demonstrated percentage of control (PoC) values of less than 15% (CAMKK2 PoC = 5.4, CAMKK1 PoC = 12), indicating excellent kinome wide selectivity.


Figure 1:  Kinome wide selectivity of SGC-CAMKK2-1. Only CAMKK2 and CAMKK1 bound to the inhibitor with Poc <15 (green dots).
For all other kinases PoC > 15 (yellow dots).

in vitro potency
cell based assay data

SGC-CAMKK2-1 (also known as YL-36) was tested from 0 – 10 µM in C4-2 cells (a prostate cancer cell line). Expression levels of CaMKK2, p-AMPK and AMPK were measured (Figure 1). The in cell Western IC50 = 1.6 µM.


Figure 1: SGC-CAMKK2-1 (also known as YL-36) was tested from 0 – 10 µM in C4-2 cells (a prostate cancer cell line) in western blot assays. Levels of P-AMPK were measured.
This work was performed by members of the Dan Frigo lab at MD Anderson (Dan Frigo, Thomas Pulliam, Chenchu Li, Dominik Awad)  

references
pk properties
co-crystal structures

Co-structure of CAMKK2 with YL-10 (resolution 1.7 Å). PDB ID  5UY6. YL-10 is very similar to the probe SGC-CAMKK2-1, only missing a methyl group on the phenyl ring that projects towards solvent.

synthetic schemes
materials and methods
02.12.2019

U of T Researchers Uncover Versatility of an Ancient DNA Repair Factor

by: SGC

If a bone breaks or a tendon snaps, you know to seek treatment immediately.

But your most fragile and precious cellular commodity, chromosomal DNA, breaks with astounding frequency—some estimate as many as 10,000 times a day per cell — usually without consequence.