+ All Categories
Home > Documents > ATP mediated kinome selectivity – The missing link in understanding the contribution of ...

ATP mediated kinome selectivity – The missing link in understanding the contribution of ...

Date post: 03-Jun-2018
Category:
Upload: fabienvincent
View: 217 times
Download: 0 times
Share this document with a friend

of 15

Transcript
  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    1/15

    ATP mediated kinome selectivity The missing link in understanding the contribution of

    individual JAK kinases isoforms to cellular signaling.

    Atli Thorarensen*, Mary Ellen Banker, Andrew Fensome, Jean-Baptiste Telliez, Brian Juba, Fabien

    Vincent, Robert M Czerwinski, Agustin Casimiro-Garcia

    Pfizer Worldwide Research, 200 Cambridgepark Drive, Cambridge, Massachusetts 02140, USA.

    [email protected]

    Abstract: Kinases constitute an important class of therapeutic targets being explored by both academia

    and the pharmaceutical industry. The major focus of this effort has been directed towards the

    identification of ATP-competitive inhibitors. Although it has long been recognized that the intracellular

    concentration of ATP is very different from the concentrations utilized in biochemical enzyme assays,

    little thought has been devoted to incorporating this discrepancy into our understanding of translation

    from enzyme inhibition to cellular function. Significant work has been dedicated to the discovery of JAK

    kinase inhibitors; however, a disconnect between enzyme and cellular function is prominently displayed

    in the literature for this class of inhibitors. Herein we demonstrate utilizing the four JAK family members

    that the difference in the ATP KMof each individual kinase has a significant impact on the enzyme to cell

    inhibition translation. We evaluated a large number of JAK inhibitors in enzymatic assays utilizing either

    1 mM ATP or KMATP for the four isoforms as well as in primary cell assays. This dataset provided the

    opportunity to examine individual kinase contribution to the heterodimeric kinase complexes mediating

    cellular signaling. In contrast to a recent study, we demonstrate that for IL-15 cytokine signaling it is

    sufficient to inhibit either JAK1 or JAK3 to fully inhibit downstream STAT5 phosphorylation. This

    additional data thus provides a critical piece of information explaining why JAK1 has incorrectly been

    thought as having a dominant role over JAK3. Beyond enabling a deeper understanding of JAK signaling,

    conducting similar analyses for other kinases by taking into account potency at high ATP rather than KM

    ATP may provide crucial insights into a compounds activity and selectivity in cellular contexts.

    Kinases constitute a large family within the human genome with individual members playing key

    roles in numerous cellular signaling pathways. To date, it is estimated that only a fraction of the kinome

    has been mined as a source of therapeutic targets.(1) Multiple kinases inhibitors have been approved by

    the FDA In recent years. While a large majority of these were directed at oncology indications, recent

    approval of Xeljanz for rheumatoid arthritis illustrates the potential importance of this class of targets

    for other disease indications.(2) These successes led to explosive growth in the field of kinase drug

    discovery and significant effort has been devoted towards the identification, optimization andpharmacological characterization of novel inhibitors. While historically a majority of the work has

    focused on the design of ATP competitive inhibitors (type I), kinase inhibitor design started to include

    the discovery of alternative inhibition mechanisms (type II-IV) as the field matured.(3) Significant effort

    is being devoted to understanding the structural requirements that place inhibitors in the appropriate

    mechanistic class. While structural elements likely to result in type I or II inhibitors are known, the other

    classes are less well understood.(4,5) Selectivity is another key component in kinase inhibitor design

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    2/15

    and great attention has been devoted to kinase selection for inclusion in selectivity panels.(6)

    Substantial selectivity datasets containing either a large number of compounds tested against a select

    panel of kinases(7) or comprehensive selectivity assessments across the kinome for a more limited

    number of compounds have been published in recent years.(8,9) This work resulted in the need to

    quantify selectivity to allow comparisons between compounds and prompted the development of

    methods such as Gini or thermodynamic partition index.(10,11)

    Janus kinases (JAK) are non-receptor tyrosine kinases required for signaling through type I/II

    cytokine receptors.(12) There are four JAK family members, JAK1, JAK2, JAK3 and TYK2. JAK3 and TYK2

    are primarily involved in immune related functions while JAK1 and JAK2 also play critical roles related to

    hematopoiesis, growth and neuronal functions amongst others. A notable feature of JAK signaling is the

    requirement for a dimer of JAK kinases at the cytokine receptor complex level. Cytokine receptors

    signaling through JAK1/JAK3, JAK1/JAK2, JAK1/TYK2, JAK2/TYK2 and JAK2/JAK2 have been described.

    The importance of the various individual JAKs in cytokine signaling has resulted in significant

    effort to identify selective inhibitors for each family member.(13,14) Inhibitor design has been guided

    by the availability of crystal structures for each isoform.(15) These inhibitors have served an important

    role in elucidating the role of JAKs in cell signaling.(16,17) The selectivity characterization of these

    inhibitors has exclusively utilized enzymatic assays performed with an ATP concentration equal to

    enzyme KMproducing IC50,KMvalues or by describing the intrinsic affinity of the inhibitors as a KD. This

    data was then supplemented by various cellular assays to confirm the selectivity profile.(18,19)

    Discrepancies between enzyme and cellular data for JAK kinases has been recognized for some time but

    only recently have potential explanations supported by experimentation been published to address

    these inconsistencies.(20) It has long been recognized that the cellular concentration of ATP is in the 1

    to 5 mM range. Therefore, neither a KDnor an IC50measurement at ATP KMcan be a good descriptor of

    cellular activity for ATP competitive inhibitors. The relationship of activity at various substrateconcentrations was described by Cheng-Prusoff in the early 1970s (eq 1) for competitive

    compounds.(21) The importance of that relationship did not escape the drug discovery community and

    it has been invoked to explain disconnects between biochemical and cellular compound activity,(22,23)

    as presented in a seminal article by Shokat et al.(24) It should be noted that these concerns are only

    relevant in the context of ATP competitive, type I kinase inhibitors. Nonetheless, the literature is very

    sparse in case studies where this concern is clearly articulated or addressed experimentally. An example

    of incorporating this thinking in our understanding of kinase function was described for the

    identification of inhibitors for interleukin-2 inducible T cell kinase (ITK).(25) In recent years the

    importance of JAK3 in the JAK1/JAK3 heterodimeric pair has been the source of significant discussion

    and conflicting conclusions.(26, 27, 28) There is a challenge for a dual JAK3/1 compound, to determinecorrelations utilizing enzyme data to cell data and the individual kinase contribution to cellular signaling.

    In this report we describe the relationships of enzyme inhibition measured at of both KM and

    physiological ATP concentrations and how these relate to cellular activity. We find these considerations

    to be critical in understanding the role of individual JAK kinases in their heterodimeric signaling

    complexes.

    IC50= Ki*(1+[ATP]/[KM,ATP]) (eq. 1)

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    3/15

    Results and discussion

    Starting from the Cheng-Prusoff relationship (eq.1), the predicted potency shift for a compound

    tested at two different ATP concentrations (for example [A] and [B]) is described by eq.2. This equation

    can be further simplified to eq.3 when one of the IC50values is determined at [ATP]=KM.

    Shift = IC50A/IC50B= (1+[ATPA]/[KM,ATP])/ (1+[ATPB]/[KM,ATP]) (eq. 2)

    Shift = IC50A/IC50KM= (1+[ATPA]/[KM,ATP])/2 (eq. 3)

    Experimentally, we measured the enzyme activity of the catalytic domains of the four JAK

    isoforms, determining both KMas well as the fraction of active protein through active site titration (Table

    1). The assays were also optimized to measure enzyme inhibition in the presence of 1 mM ATP with all

    isoforms producing linear correlations between IC50values obtained in assays run at [ATP]=KMand 1 mM

    ATP. In order to understand in greater detail the interplay of the various JAK heterodimers on cellular

    function, we mined the Pfizer database for compounds having been evaluated in both enzymatic and

    cellular JAK assays. We then evaluated these compounds against the four JAK isoforms utilizing a 1 mM

    ATP concentration. The enzyme concentration coupled with its active fraction determines the lower

    limit of accuracy for enzymatic assays. Consequently, IC50values above 3 nM were considered relevant

    for this discussion. Similarly, while the compound concentrations used allowed for IC50measurements of

    up to 10 M, we chose to restrict the range of IC50values used in this analysis to those below 1 M due

    to the lack of solubility at 10M of some compounds in this broad set. The entire range of determined

    IC50for all compounds is nonetheless illustrated in each graph. The measured shift was largest for JAK3

    (approximately 100 fold, Figure 1), while being smallest for JAK1 (approximately 10 fold, figure S1). In

    the case of TYK2 and JAK2 the shift was around 25 and 50 fold, respectively (data not shown). Whencompared to the shifts predicted by eq.3 there was good agreement between measured and theoretical

    shifts.

    Table 1. Characterization of the four JAK isoforms.

    Enzyme% Active

    Enzyme

    ATP KMAssays1mM ATP

    AssaysTheoretical

    Shift

    Experimental

    ShiftATP

    (M)Total

    Enzyme

    (nM)

    Total

    Enzyme

    (nM)

    JAK1 12 40 20 30 12 ~10

    JAK2 60 4 1 2 125 ~50

    JAK3 40 4 1 2 125 ~100

    TYK2 40 12 1 2 42 ~25

    Figure 1. Correlation of JAK3 compound potency at [ATP]=KMvs 1mM

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    4/15

    Six cytokines (IL-2, -4, -7, -9, -15, -21) signal through the JAK1/JAK3 heterodimeric pair. In this

    study we elected to study IL-15 and IL-21 signaling through JAK1/JAK3 by measuring STAT5

    phosphorylation in peripheral blood mononuclear cells (PBMC) and in Kit225 cells. When we compared

    enzyme IC50values obtained at [ATP]=KMfor JAK1 and JAK3 isoforms and cellular activities, JAK3 enzyme

    potency displayed little correlation with Kit225 cellular activity, even when cellular permeability was

    taken into account (Figure S2a-b). Following this finding, we focused our analysis on data generated with

    human PBMCs to avoid any artificial bias introduced by using immortalized cells. Using this more

    physiologically relevant cellular system, a different picture emerges irrespective of whether the

    correlation is being performed with JAK3 enzyme data obtained at KMor 1mM ATP (Figure 2a-b). In both

    instances compounds which are permeable (green) and have high selectivity at 1mM ATP (JAK1

    IC50/JAK3 IC50>50, triangles) display a good correlation between enzymatic and cellular activities.

    Figure 2. Relationship between IL-15 stimulated STAT5 phosphorylation and JAK3 enzymatic activity. a)

    Enzyme assay with ATP concentration at KM, b) enzyme assay with ATP concentration at 1 mM.

    2a) 2b)

    Legend color by binned RRCK passive permeability AB red 10 (high), gray data not available. Shape by x = JAK1/JAK3 binned selectivity using 1mM ATP assay IC50

    100x

    100x

    10x

    10x

    JAK3 CaliperIC50 (4uM ATP, uM)

    0.005

    0.01

    0.05

    0.1

    0.5

    1

    5

    10

    0.00... 0 .00... 0.001 0.005 0.01 0.05 0.1 0.5 1 5 10

    JAK3 Caliper IC50 (1mM ATP, uM)

    0.01

    0.05

    0.1

    0.5

    1

    5

    10

    20

    0.005 0.01 0.05 0.1 0.5 1 5 10

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    5/15

    value. Square: x

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    6/15

    3a) 3b)

    Legend Shape by x = JAK1/JAK3 binned selectivity using 1 mM ATP assay IC50 values. Square: x

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    7/15

    Legend color by binned IL-15 cellular IC50, pink: 0.25

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    8/15

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    9/15

    concentration rather than at ATP KMin order to properly assess compound selectivity and better predict

    cellular function.

    Acknowledgements

    We would like to thank the numerous members of Pfizers JAK research project team who designed,

    prepared and evaluated JAK compounds thus enabling the data mining and additional evaluation of

    these compounds.

    Materials and Methods

    JAK Enzymes

    GST-tagged recombinant human kinase domains of JAK1, JAK2 and JAK3 were purchased from

    Invitrogen. His-tagged recombinant human TYK2 kinase domain was expressed in SF21/Baculovirus and

    purified using a 2-step affinity (Ni-NTA) and size exclusion (SEC S200) purification method. Seesupplementary information for commercial and sequence information.

    JAK Caliper Assays

    The human Janus Kinase (JAK) activity was determined by using a microfluidic assay to monitor

    phosphorylation of a synthetic peptide by the recombinant human kinase domain of each of the four

    members of the JAK family, JAK1, JAK2, JAK3 and TYK2. Reaction mixtures contained 1 M of a

    fluorescently labeled synthetic peptide, and ATP at either a level equal to the apparent KMfor ATP or at

    1 mM ATP. Each assay condition was optimized for enzyme concentration and room temperature

    incubation time to obtain a conversion rate of 20% - 30% phosphorylated peptide product. Reactions

    were terminated by the addition of stop buffer containing EDTA. Utilizing the LabChip 3000 mobility

    shift technology (Caliper Life Science), each assay reaction was sampled to determine the level of

    phosphorylation. This technology is separation-based, allowing direct detection of fluorescently labeled

    substrates and products with separations controlled by a combination of vacuum pressure and electric

    field strength optimized for the peptide substrate.

    Determination of apparent KMfor ATP using the Caliper JAK Enzyme Assays

    Kinase assays were carried out at room temperature in a 384-well polypropylene plate in 80L of

    reaction buffer containing 20 mM HEPES, pH 7.4, 10 mM magnesium chloride, 0.01% bovine serum

    albumin (BSA), 0.0005% Tween 20, 1mM DTT and 2% DMSO. Reaction mixtures contained 1M of a

    fluorescently labeled synthetic peptide (5FAM-KKSRGDYMTMQID for JAK1 and TYK2 and FITC-

    KGGEEEEYFELVKK for JAK2 and JAK3) and various concentrations of ATP. The kinase reactions wereinitiated by the addition of JAK enzymes and were sampled at various time points to determine the level

    of peptide phosphorylation. The percent product converted was determined for each sample based on

    peak height (percent product = product/(product+substrate)). The enzyme velocities were determined

    for each concentration of ATP and a KMfor ATP was determined using the Michaelis-Menten model,

    Y = Vmax*X/(KM+ X). Vmaxis the maximum enzyme velocity and KM, the Michaelis-Menten constant, is the

    substrate concentration needed to achieve a half-maximum enzyme velocity.

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    10/15

    Caliper JAK Enzyme Endpoint IC50Assays

    Test compounds were solubilized in dimethyl sulfoxide (DMSO) to a stock concentration of 30 mM.

    Compounds were diluted in DMSO to create an 11-point half log dilution series with a top concentration

    of 600 M. The test compound plate also contained positive control wells with a known inhibitor to

    define 100% inhibition and negative control wells with DMSO to define no inhibition. The compound

    plates were diluted 1 to 60 in the assay, resulting in a final assay compound concentration range of

    10 M to 100 pM and a final assay concentration of 1.7% DMSO. 250 nL of test compounds and controls

    solubilized in 100% DMSO were added to a 384 well polypropylene plate (Matrical) using an non contact

    acoustic dispenser. Kinase assays were carried out at room temperature in a 15L reaction buffer

    containing 20 mM HEPES, pH 7.4, 10 mM magnesium chloride, 0.01% bovine serum albumin (BSA),

    0.0005% Tween 20 and 1mM DTT. Reaction mixtures contained 1M of a fluorescently labeled

    synthetic peptide, a concentration less than the apparent KM (5FAM-KKSRGDYMTMQID for JAK1 and

    TYK2 and FITC-KGGEEEEYFELVKK for JAK2 and JAK3). Reaction mixtures contained ATP at either a level

    equal to the apparent KMfor ATP (40 M for JAK1, 4 M for JAK2, 4 M for JAK3 and 12 M for TYK2) or

    at 1 mM ATP. The assays were stopped with 15L of a buffer containing 180 mM HEPES, pH=7.4, 20 mM

    EDTA, 0.2% Coating Reagent, resulting in a final concentration of 10 mM EDTA, 0.1% Coating Reagent

    and 100 mM HEPES, pH=7.4. Each assay reaction was then sampled to determine the level of

    phosphorylation. The data output used for calculations was percent product converted and was was

    determined for each sample and control well based on peak height (percent product =

    product/(product+substrate)). The percent effect at each concentration of test compound was

    calculated based on the positive and negative control well contained within each assay plate using the

    formula % effect = 100*((sample well negative control)/(positive control-negative control)). The

    percent effect was plotted against the compound concentration compound. An unconstrained sigmoid

    curve was fitted using a 4 parameter logistic model and the concentration of test compound required

    for 50% inhibition (IC50) was determined for each test compound.Preparation of peripheral blood mononuclear cells (PBMC)

    Cryopreserved human PBMCs (Catalog No. PB005F), which were used in the IL-2, IL-10, IL-15, and IFN

    assays, were purchased from Allcells (Emeryville, CA). Frozen PBMCs were thawed in a water bath

    (37C), and washed once with RPMI1640 medium (Catalog No. 72400, Invitrogen, Grand Island, NY).

    Cells were resuspended in RPMI medium containing 10% fetal bovine serum (FBS) and incubated at 37C

    for 1.5 hours for resting.

    Cytokine Stimulation and FACS Sample Preparation

    Serum was removed from resting PBMCs by washing once with D-PBS. PBMCs were resuspended in

    RPMI1640 medium. Ninety (90) L of resuspended PBMCs (5 million cells/mL) were aliquoted in 96-

    well, deep well, V-bottom plates (Catalog No. 82007-292; VWR, Radnor, PA) and incubated at 37C for

    75 minutes. Cells were treated with compound (5L/well) at various concentrations (0.2% DMSO final)

    at 37C for 60 minutes, followed by the challenge with cytokine (5L/well; final concentration of 41

    ng/mL IL-2, 41 ng/mL IL-15, 5000 U/mL IFN, 15 ng/mL IL-10) for 15 minutes. Cells were treated with

    warm Lyse/Fix buffer (700 L/well; Catalog No. 558049; BD Biosciences) to terminate activation and

    further incubated at 37C for 15 minutes. Plates were centrifuged at 300 x g for 5 minutes, supernatant

    was aspirated, and cells were washed with 800L per well of staining buffer (0.5% heat inactivated FBS

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    11/15

    and 0.001% sodium azide in D-PBS). The washed cell pellets were resuspended with 350L per well of

    cold 90% methanol (-20C), and incubated on ice for 30 minutes. After the removal of 90% methanol,

    cells were washed once with staining buffer (800L/well). Cell pellets were resuspended in staining

    buffer containing anti-phospho-STAT-AlexaFluor647 conjugated antibodies (1 to 150 dilution, 150

    L/well), and incubated at room temperature in the dark overnight. Anti-phospho STAT3-AlexaFluor647

    (Catalog No. 557815; BD Biosciences) was used for IL-10 and IFN stimulated cells, and anti-phospho

    STAT5-AlexaFluor647 (Catalog No. 612599; BD Biosciences) was used for Il-2 and IL-15 stimulated cells.

    Flow Cytometry

    Samples were transferred to 96-well U-bottom plates and flow cytometric analysis wasperformed on a

    FACSCalibur, FACSCanto or LSRFortessa equipped with a HTS plate loader(BD Biosciences). Lymphocyte

    population was gated for the pSTAT3 or pSTAT5 (APC channel) histogram analysis. Background

    fluorescence was defined using unstimulated cells and a gate (M1) was placed at the foot of the peak to

    include ~0.5% gated population. The histogram statistical analysis was performed using CellQuestPro

    version 5.2.1 (BD Biosciences), FACSDiva version 6.2 (BD Biosciences) or FlowJo version 7.6.1 (Ashland,OR) software. Relative fluorescence unit (RFU), which measures the level of pSTAT, was calculated by

    multiplying the percent positive population (M1) and its mean fluorescence. IC50 values were

    determined using the Prism version 5 software (GraphPad, La Jolla, CA).

    Supporting information

    Figure S1. Correlation of JAK1 enzyme activity at KMvs 1mM [ATP]

    Figure S2. Correlation of KMenzyme activity to inhibition of IL-15 stimulated STAT5 phosphorylation inKit225 cells in relationship to JAK1/JAK3 selectivity derived from enzyme assay at KM. a) JAK3 enzyme

    activity at KM. b) JAK3 enzyme activity at 1 mM ATP.

    JAK1 Caliper IC50 (40 uM ATP, uM)

    0.0005

    0.001

    0.005

    0.01

    0.05

    0.1

    0.5

    1

    5

    10

    0.0005 0.001 0.005 0.01 0.05 0.1 0.5 1 5 10

    100x

    100x

    10x

    10x

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    12/15

    a)

    b)

    Legend color by binned RRCK passive permeability AB red 10 (high). Shape by x = JAK1/JAK3 binned selectivity using KMassay value. Square: x

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    13/15

    841 ptqfeerhlk flqqlgkgnf gsvemcrydp lqdntgevva vkklqhstee hlrdfereie

    901 ilkslqhdni vkykgvcysa grrnlklime ylpygslrdy lqkhkeridh ikllqytsqi

    961 ckgmeylgtk ryihrdlatr nilvenenrv kigdfgltkv lpqdkeyykv kepgespifw

    1021 yapesltesk fsvasdvwsf gvvlyelfty ieksksppae fmrmigndkq gqmivfhlie

    1081 llknngrlpr pdgcpdeiym imtecwnnnv nqrpsfrdla lrvdqirdnm ag

    Jak3 kinase domainInvitrogen part Number PV4080

    Recombinant Human Protein, Catalytic Domain, GST-tagged, expressed in insect cells

    GenBank Accesion Number NP_000206

    Amino Acids 781 1124

    781 issdyellsd ptpgalaprd glwngaqlya cqdptifeer hlkyisqlgk gnfgsvelcr

    841 ydplgdntga lvavkqlqhs gpdqqrdfqr eiqilkalhs dfivkyrgvs ygpgrqslrl

    901 vmeylpsgcl rdflqrhrar ldasrlllys sqickgmeyl gsrrcvhrdl aarnilvese

    961 ahvkiadfgl akllpldkdy yvvrepgqsp ifwyapesls dnifsrqsdv wsfgvvlyel

    1021 ftycdkscsp saeflrmmgc erdvpalcrl lelleegqrl pappacpaev helmklcwap

    1081 spqdrpsfsa lgpqldmlws gsrgcethaf tahpegkhhs lsfs

    Tyk2 kinase domainPurification: in house

    Recombinant Human Protein, Catalytic Domain, His-tagged, expressed in insect cells

    Amino Acid sequence of construct: human His-TEV-tagged TYK2 Wild Type

    1 MAHHHHHHHH HHGALEVLFQ GPGDPTVFHK RYLKKIRDLG EGHFGKVSLY

    51 CYDPTNDGTG EMVAVKALKA DAGPQHRSGW KQEIDILRTL YHEHIIKYKG

    101 CCEDAGAASL QLVMEYVPLG SLRDYLPRHS IGLAQLLLFA QQICEGMAYL

    151 HSQHYIHRDL AARNVLLDND RLVKIGDFGL AKAVPEGHEY YRVREDGDSP

    201 VFWYAPECLK EYKFYYASDV WSFGVTLYEL LTHCDSSQSP PTKFLELIGI

    251 AQGQMTVLRL TELLERGERL PRPDKCPAEV YHLMKNCWET EASFRPTFEN

    301 LIPILKTVHE KYQGQAPS

    References

    (1) Knapp, S.; Arruda, P.; Blagg, J.; Burley, S.; Drewry, D.H.; Edwards, A.; Fabbro, D.; Gillespie, P.; Gray, N.S.; Kuster,

    B.; Lackey, K.E.; Mazzafera, P.; Tomkinson, N.C.; Willson, T.M.; Workman, P.; Zuercher, W.J. A public-private

    partnership to unlock the untargeted kinome. Nat Chem Biol. 2013, 9, 3-6.

    (2) Cohen, P.; Alessi, D.R. Kinase Drug Discovery - What's Next in the Field? ACS Chem Biol. 2013, 18, 96-104.

    (3) Dar, A. C; Shokat, K. M The evolution of protein kinase inhibitors from antagonist to agonist of cellular

    signaling Ann. Rev. Biochem. 2011, 80, 769-795.

    (4) Backes, A.C.; Zech, B.; Felber, B.; Klebl, B.; Mller, G.; Small-molecular inhibitors binding to protein kinases.

    Part I: exceptions from the traditional pharmacophore approach of type I inhibition. Expert Opin. Drug Discov.

    2008, 3, 1409-1425.

  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    14/15

    (5) Backes, A.C.; Zech, B.; Felber, B.; Klebl, B.; Mller, G.; Small-molecular inhibitors binding to protein kinases.

    Part II: the novel pharmacophore approach of type II and type III inhibition. Expert Opin. Drug Discov. 2008, 3,

    1427-1449.

    (6) Brandt, P.; Jensen, A. J.; Nilsson, J. Small kinase assay panels can provide a measure of selectivity Bioorg.

    Med. Chem. Lett. 2009, 19, 5861-3.

    (7) Posy, S. L.; Hermsmeier, M. A.; Vaccaro, W.; Ott, K-H.; Todderud, G.; Lippy, J. S.; Trainor, G. L.; Loughney, D. A.;

    Johnson, S. R. Trends in kinase selectivity: insights for target class-focused library screening. J. Med. Chem. Lett.

    2011, 54, 54-66.

    (8) Anastassiadis, T.; Deacon, S. W.; Devarajan, K.; Ma, H.; Peterson, J. R. Comprehensive assay of kinase catalytic

    activity reveals features of kinase inhibitor selectivity Nat. Biotechnol. 2011, 29, 1039-1046.

    (9) Davis, M. I.; Hunt, J. P.; Herrgard, S.; Ciceri, P.; Wodicka, L. M.; Pallares, G.; Hocker, M.; Treiber, D. K.; Zarrnkar,

    P. P. Comprehensive analysis of kinase inhibitor selectivity Nat. Biotechnol. 2011, 29, 1046-1052

    (10) Graczyk, P. P. Gini coefficient: A new way to express selectivity of kinase inhibitors against a family of

    kinases J. Med. Chem. 2007, 5773-5779.

    (11) Cheng, A. C.; Eksterowicz, J.; Geuns-Meyer, S.; Sun, Y. Analysis of kinase inhibitor selectivity using a

    thermodynamics-based partition index. J. Med. Chem. 2010, 53, 4502-4510.

    (12) Laurence, A.; Pesu, M.; Silvennoinen, O.; O'Shea J. JAK Kinases in Health and Disease: An Update. Open

    Rheumatol J. 2012, 6, 232-44.

    (13) Norman, P. Selective JAK1 inhibitor and selective TYK2 inhibitor patents Expert Opin. Ther. Patents 2012, 22,

    1233-1249.

    (14)Dymock, B.W.; See, C.S. Inhibitors of JAK2 and JAK3: an update on the patent literature 2010 2012 Expert

    Opin. Ther. Pat. 2013, 23, 449-501

    (15) Alicea-Velzquez, N. L.; Boggon, T. J. The use of structural biology in Janus kinase targeted drug discovery

    Current Drug Targets, 2011, 12, 546-555.

    (16) Lee, J. E.; Lee, A. S.; Kim, D. H.; Jung, Y. J.; Lee, S.; Park, B-H.; Lee, S. H.; Park, S. K.; Kim, W.; Kang, K. P.; Janex-

    1, a JAK3 inhibitor, ameliorates tumor necrosis factor--induced expression of cell adhesion molecules and

    improves myocardial vascular permeability in endotoxemic mice Int. J. Mol. Med. 2012, 29, 864-870.

    (17) Cetkovic-Cvrlje M, Olson M, Ghate K. Targeting Janus tyrosine kinase 3 (JAK3) with an inhibitor induces

    secretion of TGF- by CD4+ T cells. Cell Mol. Immunol. 2012,9, 350-60.

    (18) Zak, M.; Mendonca, R.; Balazs, M.; Barrett, K.; Bergeron, P.; Blair, W. S.; Chang, C.; Deshmukh, G.; DeVoss, J.;

    Dragovich, P. S.; Eigenbrot, C.; Ghilardi, N.; Gibbons, P.; Gradl, S.; Hamman, C.; Hanan, E. J.; Harsstad, E.; Hewitt, P.

    R.; Hurley, C. A.; Jin, T.; Johnson, A.; Johnson, T.; Kenny, J. R.; Koehler, M. F. T.; Kohli, P. B.; Kulagowski, J. J.;

    Labadie, S.; Liao, J.; Liimatta, M.; Lin, Z.; Lupardus, P. J.; Maxey, R. J.; Murray, J. M.; Pulk, R.; Rodriguez, M.; Savage,

    S.; Hia, S.; Steffek, M.; Ubhayakar, S.; Ultsch, M.; Abbema, A. V.; Ward, S. I.; Xiao, L.; Xiao, Y. Discovery and

    http://informahealthcare.com.proxy1.athensams.net/action/doSearch?action=runSearch&type=advanced&result=true&prevSearch=%2Bauthorsfield%3A%28See%2C+C+S%29http://informahealthcare.com.proxy1.athensams.net/action/doSearch?action=runSearch&type=advanced&result=true&prevSearch=%2Bauthorsfield%3A%28See%2C+C+S%29http://informahealthcare.com.proxy1.athensams.net/action/doSearch?action=runSearch&type=advanced&result=true&prevSearch=%2Bauthorsfield%3A%28See%2C+C+S%29
  • 8/12/2019 ATP mediated kinome selectivity The missing link in understanding the contribution of individual JAK kinases isof

    15/15

    optimization of C-2 methyl imidazopyrrolopyridines as potent and orally bioavailable JAK1 inhibitors with

    selectivity over JAK2. J. Med. Chem. 2012, 55, 6176-6193.

    (19) Soth, M.; Hermann, J. C.; Yee, C.; Alam, M.; Barnett, J. W.; Berry, P.; Browner, M. F.; Frank, K.; Frauchiger, S.;

    Harris, S.; He, Y.; Hekmat-Nejad, M.; Hendricks, T.; Henningsen, R.; Hilgenkamp, R.; Ho, H.; Hoffman, A.; Hsu, P-Y.;

    Hu, D-Q.; Itano, A.; Jaime-Figueroa, S.; Jahangir, A.; Jin, S.; Kuglstatter, A.; Kutach, A. K.; Liao, C.; Lynch, S.; Menke,

    J.; Niu, L.; Patel, V.; Railkar, A.; Roy, D.; Shao, A.; Shaw, D.; Steiner, S.; Sun, Y.; Tan, S-L.; Wang, S.; Vu, M. D. 3-

    Amido pyrrolopyrazine JAK kinase inhibitors: development of a JAK3 vs JAK1 selective inhibitor and evaluation in

    cellular and in in vivo models. J. Med. Chem. 2013, 56, 345-356.

    (20) Yu, V.; Pistillo, J.; Archibeque, I.; Lee, J.; Sun, B-C.; Schenkel, L.B.; Geuns-Meyer, S.; Liu, L.; Emkey, R.

    Differential Selectivity of JAK2 Inhibitors in Enzymatic and Cellular Settings. Exp. Hematol. 2013, doi:

    10.1016/j.exphem.2013.01.005.

    (21) Cheng, Y.; Prusoff, W.H. Relationship between the inhibition constant (K1) and the concentration of inhibitor

    which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 1973, 22, 3099-108.

    (22) Frantz, B.; Klatt, T.; Pang, M.; Parsons, J.; Rolando, A.; Williams, H.; Toci, M. J.; OKeefe, S. J.; ONeill, E. A. The

    activation state of p38 mitogen-activated protein kinase determines the efficiency of ATP competition for

    pyridinylimidazole inhibitor binding. Biochemistry 1998, 37, 13846-13853.

    (23) Anderson, D. R.; Meyers, M. J.; Vernier, W. F.; Mahoney, M. W.; Kurumbail, R. G.; Caspers, N.; Poda, G. I.;

    Schindler, J. F.; Reitz, D. B.; Mourey, R. J. Pyrrolopyridine inhibitors of mitogen-activated protein kinase-activated

    protein kinase 2 (MK-2). J. Med. Chem. 2007, 50, 2647-2654.

    (24) Knight, Z. A.; Shokat, K. M.; Features of selective kinase inhibitors Chemistry & Biology 2005, 12, 621-637.

    (25) Zapf, C. W.; Gerstenberger, B. S.; Xing, L.; Limburg, D. C.; Anderson, D. R.; Caspers, N.; Han, S.; Aulabaugh, A.;

    Kurumbail, R.; Shakya, S.; Li, X.; Spaulding, V.; Czerwinski, R. M.; Seth, N.; Medley, Q. G. Covalent inhibitors ofinterleukin-2 inducible T cell kinase (Itk) with nanomolar potency in a whole-blood assay. J. Med Chem. 2012, 55,

    10047-10063.

    (26) Thoma, G.; Nuninger, F.; Falchetto, R.; Hermes, E.; Tavares, G.A.; Vangrevelinghe, E.; Zerwes, H.G.

    Identification of a potent Janus kinase 3 inhibitor with high selectivity within the Janus kinase family. J. Med.

    Chem. 2011, 54, 284-8.

    (27) Lin, T.H.; Hegen, M.; Quadros, E.; Nickerson-Nutter, C.L.; Appell, K.C.; Cole, A.G.; Shao, Y.; Tam, S.; Ohlmeyer,

    M.; Wang, B.; Goodwin, D.G.; Kimble, E.F.; Quintero, J.; Gao, M.; Symanowicz, P.; Wrocklage, C.; Lussier, J.;

    Schelling, S.H.; Hewet, A.G.; Xuan, D.; Krykbaev, R.; Togias, J.; Xu, X.; Harrison, R.; Mansour, T.; Collins, M.; Clark,

    J.D.; Webb, M.L.; Seidl, K.J. Selective functional inhibition of JAK-3 is sufficient for efficacy in collagen-induced

    arthritis in mice. Arthritis Rheum. 2010, 62, 2283-93.

    (28) Haan C, Rolvering C, Raulf F, Kapp M, Drckes P, Thoma G, Behrmann I, Zerwes HG. Jak1 has a dominant role

    over Jak3 in signal transduction through c-containing cytokine receptors. Chem Biol. 201125, 314-23.


Recommended