systems pharmacology; an industrial perspective … · , systems pharmacology of the ngf pathway;...

53
SYSTEMS PHARMACOLOGY; AN INDUSTRIAL PERSPECTIVE (Warwick school of Engineering vacation school) Dr Neil Benson. Director, Xenologiq Ltd. [email protected] www.xenologiq.com Quantitative drug discovery solutions Interested in PKPD modelling, dose prediction, PBPK and systems pharmacology

Upload: others

Post on 19-Oct-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

  • SYSTEMS PHARMACOLOGY; AN INDUSTRIAL

    PERSPECTIVE

    (Warwick school of Engineering vacation school)

    Dr Neil Benson. Director, Xenologiq Ltd.

    [email protected]

    www.xenologiq.com Quantitative drug discovery solutions

    Interested in PKPD modelling, dose prediction, PBPK and systems pharmacology

    mailto:[email protected]

  • Why should drug discovery be interested in systems pharmacology? Attrition

    www.xenologiq.com Quantitative drug discovery solutions

    Attrition – a small proportion of the ideas worked on in drug discovery become drugs -> significant $ spent on ideas that never become products

  • 10 years of evaluation & proposals and arguably nothing has changed

    www.xenologiq.com Quantitative drug discovery solutions

    Kola & Landis, Nature Rev DD (2004); PHII attrition due to lack of efficacy or safety key issue

    Paul et al, Nature reviews, DDT, (2010) – PHII attrition by far largest contributor to productivity

    Hay et al (2014); Nature biotech; The attrition statistics showed a decrease in probability of success compared to previous evaluations

    2004 2010 2014

  • Why drug discovery needs mathematical models •Kola & Landis, Nature Rev DD (2004); Paul et al, Nature reviews, DDT, (2010), Hay et al (2014) – PHII attrition by far largest contributor to productivity

    •Mainly due to lack of efficacy or safety (ie we didn’t understand the biology -> consequences perturbing a complex system)

    Mathematical modelling & simulation proven approach for tackling complexity in many areas of science and engineering

  • What is systems pharmacology ?

    Time

    Effect

    Concen

    tration

    PD

    PK

    PKPD

    Understanding of behaviour of a drug in a disease www.xenologiq.com Quantitative drug discovery solutions

    Requires mathematical modelling approach

  • Example 1 – PKPD & systems pharmacology approach to Toll-like 7 receptor agonists for Hepatitis C virus

    •HCV unmet medical need

    •Hypothesis - TLR7 agonism -> interferon response -> improved anti HCV effect

    www.xenologiq.com Quantitative drug discovery solutions

  • Project questions at preclinical stage

    www.xenologiq.com Quantitative drug discovery solutions

    1. Can we achieve a dose that will deliver efficacy ?

    2. What is the minimum anticipated biological effect level ?

    Benson et al Antimicrobial Agents and Chemotherapy, March 2010, p. 1179-1185, Vol. 54, No. 3

  • Toll-like 7 receptor agonists for Hepatitis C virus; aims

    Challenge:

    No animal model for HCV Link between TLR7 agonism and antiviral effect not established Dose prediction for interferon inducers for hepatitis C ?

    Opportunity:

    IFNα available as translatable biomarker Clinical PK and PKPD data for recombinant IFNα Published math model for antiviral efficacy of IFNα

    Solution:

    Integrated mechanism-based PKPD approach linking exposurebiomarkeroutcome

    www.xenologiq.com Quantitative drug discovery solutions

  • www.xenologiq.com 9

    Very complex – mathematical modelling solution required

    Predicting dose/ antiviral response in man

  • TLR7 – projecting human interferon induction

    • Data generated in the mouse

    • Rapid and robust induction of IFNa observed from zero baseline

    5 15 25

    5 15 25

    TIME

    100

    300

    100

    300

    100

    300

    DV

    AMT: 100 AMT:300

    AMT: 500 AMT1000

    AMT: 5000 AMT:1000.00

    5 15 25

    5 15 25

    TIME

    100

    300

    100

    300

    100

    300

    DV

    AMT: 100 AMT:300

    AMT: 500 AMT1000

    AMT: 5000 AMT:1000.00

    Plasma IFNa (IU/ml)

    www.xenologiq.com Quantitative drug discovery solutions

    How do we use this data in our model ?

  • TLR7 – indirect PKPD model of IFNa production

    .RCSC

    .CS

    dt

    dR

    p50

    pmax

    outk

    Data described using a modified indirect effect eqn

    www.xenologiq.com Quantitative drug discovery solutions

    Where; dR/dt = rate of production IFNa (IUml-1h-1), Smax = max rate of production IFNa ( IUml

    -1h-1) SC50 (ng/ml) = conc drug for ½ max production IFNa kout = elimination rate constant IFNa (h

    -1), Cp = predicted plasma concentration (ng/ml) R = plasma IFNa conc (IUml-1)

    5 15 25

    5 15 25

    TIME

    100

    300

    100

    300

    100

    300

    DV

    AMT: 100 AMT:300

    AMT: 500 AMT1000

    AMT: 5000 AMT:1000.00

    5 15 25

    5 15 25

    TIME

    100

    300

    100

    300

    100

    300

    DV

    AMT: 100 AMT:300

    AMT: 500 AMT1000

    AMT: 5000 AMT:1000.00

    Fit to PKPD data

    Mathematical model of time & dose dependence

    PK

    tktkea

    a ae ee)kV(k

    Dose.kC

    +

    PD

  • TLR7 agonist PKPD fit results

    Parameter Estimate CV(%)

    kout (h-1) 0.958 0.1

    SC50 (ng/ml) 135 24

    Smax (IU/ml/h) 294 8

    IIV Smax (%) 70 26

    Residual error

    (IU/ml)

    65 19

    Individual predicted IFNa in plasma (IU/ml)

    0 200 400 600O

    bse

    rve

    d I

    FN

    a in p

    lasm

    a (

    IU/m

    l)

    0

    200

    400

    600

    www.xenologiq.com Quantitative drug discovery solutions

    0 10 20 30 40 50 60 70 80

    101

    102

    103

    104

    105

    Time (hours)

    Co

    nce

    ntr

    atio

    n (

    dru

    g (

    ng

    /ml IF

    Na

    pg

    /ml)

    PK

    PD

    Example individual

  • TLR7 – linking IFNa to antiviral effect

    VTT ).1(dsdt

    dT IVT ).1(

    dt

    dI

    Parameter estimates from clinical data (Neumann, A.U., et al, Science, 1998. 282(5386): p. 103-7)

    HCV = hepatitis C viral load

    c the viral decay rate constant,

    δ the infected hepatocyte decay

    rate constant.

    ε = inhibition of viral release (p)

    = inhibition of viral infection s&d = Target cell

    production/degradation rates

    II TT

    Infection Rate

    c

    Clearance

    p

    Virions/d

    Target Cell

    Infected

    Cell

    Loss

    cVpI ).1(dt

    dV

    IFNa blocks viral release

    www.xenologiq.com

    Can be represented as a set of equations

    IFNIC

    IIFN

    50

    max

  • Modelling enabled an integrated mechanism-based PKPD approach for predicting antiviral efficacy of TRL7 agonists

    Rat PK

    Mouse IFNα induction

    PKPD

    Human IFNα PKPD Human PK

    Human IFNα induction

    PKPD

    Human antiviral PKPD

    PBPK

    Clinical

    Literature

    Viral load

    PKPD model

    Experimental

    Literature

    In silico

    0 20 40 60

    time (days)

    101102103104105106107

    101102103104105106107

    101102103104105106107

    vir

    al lo

    ad

    (R

    NA

    co

    pie

    s/m

    L)

    dose: 1 dose: 3

    dose: 10 dose: 30

    dose: 100

    Trial simulation

    Human dose

    prediction

    www.xenologiq.com

  • Conclusions

    • Collected & integrated all available data for decision making

    • Allowed conversion of information into knowledge for; 1. Dose prediction – likely to achieve efficacy at a practical dose

    2. Minimum anticipated biological effect level (MABEL) ID

    3. Sensitivity analysis – what is important? (compound potency <

    system properties)

    www.xenologiq.com Quantitative drug discovery solutions

  • Example 2; systems pharmacology & technology feasibility

    Drug molecule – binds the target but has poor pharmacokinetics (the ‘war head’)

    Molecule coupled to the drug; improves the PK whilst retaining the drug effect

    General outline of the approach;

    Acknowledgement;Tomomi Matsuura & Piet van der Graaf, Pfizer

    Gives improved PK

    ‘War head’ binds the target

    www.xenologiq.com Quantitative drug discovery solutions

    +

    Pharmacophore + Linker Antibody CovX- body

  • Technology feasibility

    Project question for target Z

    • The coupled drug exhibits pharmacological action, but...

    • The war head strongly binds human serum albumin (>>90% bound in plasma)

    • Given dose of the new drug is limited by cost & what can be delivered...

    • Can the required receptor occupancy be achieved via this approach in this case?

    www.xenologiq.com Quantitative drug discovery solutions

  • Model of the system

    Serum albumin

    Drug

    R= receptor

    Parameters measured (eg surface plasmon resonance) or derived from pharmacokinetic pharmacodynamic analysis of in vivo experiments

    www.xenologiq.com Quantitative drug discovery solutions

  • Summary

    •FR: free receptor concentration/initial receptor concentration

    •Green is good, red is bad

    •At higher ppb require v low target Kd –project terminated 19

  • Perspective

    • A plausible scenario is that this molecule would have exhibited;

    Good in vitro efficacy

    Clean pre-clinical safety profile (no pharmacology)

    Good PK prediction

    Good safety & PK in Phase 1 trials (no pharmacology)

    Phase II negative (no pharmacology)

    www.xenologiq.com Quantitative drug discovery solutions

    • Model based approach - terminating this early saved $$’s

  • Example 3; NFKB pathway

    Ihekwaba AE, Broomhead DS, Grimley RL, Benson N, Kell DB.(2004). Systems Biology 1(1): 93-103.

    & Science, 306, Oct 2004, 704 - 708.

    • Project question – what are the optimal targets in the pathway and what % effect/inhibition is required?

    • Use published ODE model; 64 parameters & 26 variables

    0

    0.4

    0.8

    1.2

    1.6

    2

    0 100 200 300 400 500 600

    Time (min) after TNFa

    Am

    plitu

    de

    B

    215 350 440 (min)

    0 50 170

    A

    www.xenologiq.com Quantitative drug discovery solutions

    NFKBn

    Acknowledgement; L Cucurull-Sanchez, Karen Spink

  • Further detail

    www.xenologiq.com Quantitative drug discovery solutions

    Cucurull-Sanchez, K.S Spink & S. Moschos, Drug Discovery Today, 17, 13-14, 665-, 2012

  • Project approach – Small interfering RNA (siRNA)

    Challenge: IL-1

    Target: IKK-2

    Biomarker: IL-8

    www.xenologiq.com Quantitative drug discovery solutions

    Max attenuation [IKK] – 80% ?

  • Sensitivity analysis identified IKK as a potential target

    IKK would appear to be a good potential target

    dk

    tdx )( Change in the species (eg [NFKBn])

    Change in variable (eg parameter or reactant conc)

    www.xenologiq.com Quantitative drug discovery solutions

  • But high % inhibition required to fully dampen NFKB oscillations..

    0 1 2 3 4 5 6 7 8 9 10

    x 104

    0

    0.01

    0.02

    0.03

    0.04

    0.05

    0.06

    Time

    Sta

    tes

    States versus Time

    IkBa

    NF_kB

    IkBa_NF_kB

    IkBb

    IkBb_NF_kB

    IkBe

    IkBe_NF_kB

    IKKIkBa

    IKKIkBa_NF_kB

    IKK

    IKKIkBb

    IKKIkBb_NF_kB

    IKKIkBe

    IKKIkBe_NF_kB

    NF_kBn

    IkBan

    IkBan_NF_kBn

    IkBbn

    IkBbn_NF_kBn

    IkBen

    IkBen_NF_kBn

    source

    IkBa_t

    sink

    IkBb_t

    IkBe_t

    Basal 80% knock-down of IKK

    0 1 2 3 4 5 6 7 8 9 10

    x 104

    0

    0.01

    0.02

    0.03

    0.04

    0.05

    0.06

    0.07

    0.08

    0.09

    Time

    Sta

    tes

    States versus Time

    IkBa

    NF_kB

    IkBa_NF_kB

    IkBb

    IkBb_NF_kB

    IkBe

    IkBe_NF_kB

    IKKIkBa

    IKKIkBa_NF_kB

    IKK

    IKKIkBb

    IKKIkBb_NF_kB

    IKKIkBe

    IKKIkBe_NF_kB

    NF_kBn

    IkBan

    IkBan_NF_kBn

    IkBbn

    IkBbn_NF_kBn

    IkBen

    IkBen_NF_kBn

    source

    IkBa_t

    sink

    IkBb_t

    IkBe_t

    Time (s) Time (s)

    [NF-

    B

    (nu

    c)]

    (M

    )

    [NF-

    B

    (nu

    c)]

    (M

    ) •Assuming ~remove oscillation to suppress IL8

    •and given technology typically gives maximum 80% decrease in target

    •Risk that strong attenuation of biomarker may not be achieved

    www.xenologiq.com Quantitative drug discovery solutions

    Maximum achievable inhibition via siRNA

  • Validation of model prediction

    Thanks; Sterghios Moschos Karen Spink for data

    •80% inhibition of IKK enzyme

    •~30% attenuation of IL8 production

    •Project terminated

    www.xenologiq.com Quantitative drug discovery solutions

  • Perspective

    • Laboratory costs to test siRNA hypothesis considerable

    • Failure of outcome could have been predicted before any work was done

    • Model based approach - $$ savings and focus on a less risky target/approach

    www.xenologiq.com Quantitative drug discovery solutions

  • Example 4; systems pharmacology of the nerve growth factor pathway

    •Extracellular Regulated Kinase (ERK) activation controls pain response (eg trka levels, ion channel activity etc)

    •We assume dppERKnuc is proportional to pain

    •Team question – what are the best pain targets ?

    0 20 40 60 80 1000

    0.05

    0.1

    0.15

    0.2

    0.25

    0.3

    0.35

    Time (mins)

    dppE

    RK

    nuc(µ

    M)

    Acknowledgement; Cesar Pichardo, Pinky Dua & Piet van der Graaf (Pfizer Neusentis) and at ISB Oleg Demin)

    www.xenologiq.com Quantitative drug discovery solutions

    Nat Cell Biol

  • Models in literature Fujioka et al, J Biol Chem, 2006

    0 2 4 6 8 10 12 14 16 18 200

    0.05

    0.1

    0.15

    0.2

    0.25

    0.3

    0.35

    Pfizer model Sasagawa et al

    ppnuc

    ERK

    Time (mins)

    Sasagawa et al, Nat Cell Biol, 2005

    Benson & Dua et al, Systems pharmacology of the NGF pathway; Interface Focus, 6 April 2013 vol. 3 no. 2, 20120071.

    www.xenologiq.com Quantitative drug discovery solutions

    • One compartment integrated ‘systems biology’ model constructed (59 molecular species and 233 parameters)

  • 0 10 20 30 40 50 60

    10-2

    100

    102

    104

    106

    Species

    Tim

    e in

    teg

    ral d

    [dp

    pE

    RK

    nu

    c]/d

    [X]

    Finding optimal targets; sensitivity analysis of the model

    www.xenologiq.com Quantitative drug discovery solutions

    dk

    tdx )(Change in the species (eg [dppERKnuc])

    Change in variable (eg parameter or reactant conc)

    Rank

    (dppERKnuc)

    Species Initial

    value

    Unit Description

    1 NGFext 30 pM NGF concentration in extracellular matrix

    2 Grb2_SOS_pShc_pTrkA 0 µM Downstream multi-protein complex

    3 pShc_pTrkA 0 µM Downstream multi-protein complex

    4 Shc_pTrkA 0 µM Downstream multi-protein complex

    5 pTrkA 0 µM Phosphorylated TrkA concentration

    6 L_NGFR 0 µM NGF NGFR complex

    7 FRS2_pTrkA 0 µM Downstream multi-protein complex

    8 pFRS2_pTrkA 0 µM Downstream multi-protein complex

    9 Crk_C3G_pFRS2_pTrkA 0 µM Downstream multi-protein complex

    10 Grb2_SOS_pShc_pTrkA_endo 0 µM Downstream multi-protein complex

    NGF

    By calculating integrals -> rank of importance

  • Finding optimal targets; sensitivity analysis (SA) of the model

    www.xenologiq.com Quantitative drug discovery solutions

    • SA provides a way to rank targets & further triage by druggability criteria -> interesting targets;

    1. NGF (Consistent with NGF mAb efficacy in pain; eg Lane et al, 2010 )

    2. RAS activity (Mutations in NF1 gene associated with a chronic pain phenotype (J Neurophysiol 94: 3659-3660, 2005)

    3. Trka kinase activity (eg Expert Opin Ther Pat. 2009 Mar;19(3):305-19. Trk kinase inhibitors as new treatments for cancer and pain).

  • Systems biology -> systems pharmacology

    www.xenologiq.com Quantitative drug discovery solutions

    Systems biology model; One compartment No physiological data No easy way to compare eg

    mAb’s and small molecules Gives large dose over-

    prediction

    Systems pharmacology model; Two (n) compartments Physiological information (eg

    compartment volume) Separate compartments enable

    comparison of small molecules and mAb’s

    Can be used for dose prediction

  • But what about dose? The NGF Systems pharmacology model

    www.xenologiq.com Quantitative drug discovery solutions

    2 compartments

    1. Extracellular body water (~15L)

    2. Neuronal compartment (~0.001 L)

    •Signal transduction elements in neuronal compartment

    •Enables dose prediction for mAbs and small molecules

    •Model predicted max efficacious dose hypothetical mAb at ~ 1000xKD (a non-intuitively high ratio)

    Benson & Dua et al, Systems pharmacology of the

    NGF pathway; Interface focus, 2013.

  • Model predictions quantitatively concordant with data for prototype NGF mAbs eg tanezumab

    www.xenologiq.com Quantitative drug discovery solutions

    http://www.fda.gov/downloads/AdvisoryCommi

    ttees/CommitteesMeetingMaterials/Drugs/Arthr

    itisAdvisoryCommittee/UCM301305.pdf

    • Cmax ~ 30 nM (=3000 x KD) • -> Model prediction consistent with clinical pain data • -> efficacy & dose could have been predicted from ‘05 model

    http://www.page-

    meeting.org/pdf_asset

    s/7203-

    PageAthensCleton.pdf

    • Model predicts higher than expected drug concentration (>1000 x KD) for maximum efficacy

  • Perspective

    • ‘05 published model predicted efficacy & (non-intuitive) dose for NGF mAb well ahead of PHII data

    • Model provides mechanistic insight into why this is so;

    ie due to a negative feedback loop in the NGF pathway (Interface focus 6 April 2013 vol. 3 no. 2, 20120071.)

    www.xenologiq.com Quantitative drug discovery solutions

  • Example 5; application of systems pharmacology to the clinical development of Fatty acid amide

    hydrolase (FAAH) inhibitors for pain.

    www.xenologiq.com Quantitative drug discovery solutions

    ‘A systems pharmacology perspective on the clinical development of Fatty acid

    amide hydrolase (FAAH) inhibitors for pain’. CPT Pharmacometrics Syst. Pharmacol. (2014) 3, e91; doi:10.1038/psp.2013.72

  • Fatty acid amide hydrolase (FAAH) inhibitors for pain.

    • Cannabinoid receptors CB1&2 target for THC – active ingredient of cannabis

    • Endogenous Anadamide (AEA) is an agonist of CB1&2

    • Wealth of interesting biological data eg increased AEA attenuates nerve firing

    (eg Bacci et al, Nature 431,312–6).

    • Some evidence of efficacy of cannabis preparations in pain

    (eg Nurmikko et al., 2007,Pain, 133, 210-220)

    www.xenologiq.com Quantitative drug discovery solutions

    Acknowledgements; van der Graaf, PV D. Nichols & G. L. Li (Pfizer) & Demin,O (Institute for SB, Moscow).

  • FAAH • AEA degraded by FAAH

    • Project hypothesis;

    • FAAHi PF-04457845 animal model data positive

    • PF-04457845; good pharmacology & PK

    • Appears to be very promising drug discovery target

    www.xenologiq.com Quantitative drug discovery solutions

  • Aim for systems pharmacology approach ‘09

    • Project questions;

    1. will we test the pharmacology fully?

    2. can we predict receptor occupancy and level required for efficacy?

    • Systems pharmacology approach with available internal & external data prior to PII data

    • Collaboration with Demin & Metelkin, ISB in 2009 as PF-4457845 –> phase 1

    www.xenologiq.com Quantitative drug discovery solutions

  • Literature review; main conclusions 1. Some useful data (eg AEA disposition, enzyme kinetics to

    enable model construction)

    2. Endocannabinoid biology complex & not elucidated eg with

    apparent contradictory impacts for eg AEA on CB1 & TRPV1 ( Piscitelli and Di MarzoChem. Neurosci. 2012, 3, 356−363)

    ? Steady state impact

    3. Link between occupancy & nerve firing attenuation – no quantitative data

    Can’t specify what success would be

    in receptor occupancy vs time in man www.xenologiq.com Quantitative drug discovery solutions

    Presynaptic neuron

    Reaction

    or transport

    Activation

    Inhibition

    Reaction

    or transport

    Activation

    Inhibition

    signal

    CB1RCa2+

    Ca2+

    med

    iato

    rs

    Ca2+

    AEA

    NA-PE

    PECa2+

    ?

    AEA

    ?

    AEACOX-2, LOX FAAH

    NAT

    NAPE-PLD

    G proteins

    G proteins

    signal

    2-AG

    DAG

    PI

    ?

    2-AG

    PLC

    DAG lipase

    degradation

    ?

    rece

    pto

    rs

    COX-2, LOX MGL

    degradation

    2-AG

  • Summary of model construction

    1. XEA disposition; inter-

    compartmental equilibration of

    XEA’s, location of enzymes

    involved, tissue binding

    • Large ODE model variables – 39, reactions – 75. 5 main components…

    The model

    FAAH + AEA FAAH_AEA FAAH + P

    +

    XEA

    FAAH_XEA

    FAAH + P

    .XEAK

    KAEAK

    Vmax.Sv

    xeaFAAH,m,

    aeaFAAH,m,

    FAAHm,

    2. Multi-substrate binding of FAAH competitive

    3. 2 step bio-synthesis of XEA’s

    FAAH + I FAAH_I*

    kinh

    4. Irreversible inhibition of FAAH

    & PK;clinical data parameters

    5.Ligand partitioning &

    receptor occupancy

    theoretical model KD,obs=Kp

    -1*KD

  • An additional clearance mechanism?

    400 350 300 250 200 150 100 50 0

    10

    9

    8

    7

    6

    5

    4

    3

    2

    1

    400 350 300 250 200 150 100 50 0

    260

    240

    220

    200

    180

    160

    140

    120

    100

    80

    60

    40

    20

    0

    With unknown enzyme Without unknown enzyme

    Plateau

    No plateau

    Pla

    sm

    a A

    EA

    , n

    M

    Pla

    sm

    a A

    EA

    , nM

    Time, h Time, h

    Modelling results Modelling results

    Enzymes

    • FAAH-2

    • NAAA N-acylethanolamine

    hydrolysing acid amidase

    • COX-2 cycloxygenase-2

    • 12-LOX 12-lipoxygenase

    • 15-LOX 15-lipoxygenase

    • CYP2D6 P450

    • CYP4F2 P450

    • CYP3A4 P450

    Consume

    all XEA’s Incorporating quantitative perspective -> hypothesis NAAA most likely

    Dosage: 10 mg PF

    OEA

    PEA

    LEA

    AEA

    TIME, hours

    FA

    As

    co

    nc

    en

    tra

    tio

    n,n

    M

    350300250200150100500

    50

    45

    40

    35

    30

    25

    20

    15

    10

    5

    0

  • The model simulated observed clinical biomarker

    data

  • 0 50 100 150 200 250 300 350

    Time after dose (h)

    0

    5

    10

    15

    20

    25

    CB

    1 r

    ecepto

    r o

    ccupancy (

    %)

    0.1

    0.3

    1

    3

    10

    20

    40

    PF-04457845 (mg)

    Simulation of CB1 RO

    • FAAH inhibition >> 96%

    •Receptor occupancy limited at ~ 25% (on basis of model hypothesise this is; N-acylethanolamine hydrolysing acid amidase (NAAA))

    •Is 25% enough to give pharmacological effect?

  • Pre phase II clinical feedback 2010

    • Model suggested at doses planned we will test pharmacology as far as possible (FAAH inhibition >96%).

    • But alternative clearance of AEA exists that limits effect – hypothesis NAAA

    • No quantitative data to link receptor occupancy and pain outcome; success criteria open question

    • Model suggests some receptor occupancy, but data to confirm or rule this out desirable; ‘Pillar 2’ (Morgan et al, Drug Discovery Today Volume 17, Issues 9–10, May 2012, Pages 419–424)

    • High risk project & will we learn from the experiment?

  • Epilogue

    www.xenologiq.com Quantitative drug discovery solutions

    •2012 PF-04457845 failed in OA

    •? Confidence in animal model translation

    •? Did we express the pharmacology

    •Systems pharmacology perspective –> build knowledge & technology before any further phase II

  • Perspective

    • Model contains numerous assumptions –> ‘wrong’

    • Value not ‘description’ but as a tool to ask better questions

    • This process highlighted risks that were not apparent via other means –> ‘useful’

    www.xenologiq.com Quantitative drug discovery solutions

  • www.xenologiq.com Quantitative drug discovery solutions

    A vision for the future of drug discovery; systems models central in all projects as a tool to support decision making

  • Conclusions

    • PKPD & Systems pharmacology models can help identify optimal targets & approaches and estimate dose at very early stage

    • Growing body of evidence that systems models can add value in drug discovery (eg Benson et al, Proceedings of the 11th International Conference on Systems Biology Advances in Experimental Medicine and Biology. 2012, Volume 736, Part 5)

    • Can be applied to efficacy and safety toxicity/questions -> make better decisions in drug discovery & tackle attrition

    www.xenologiq.com Quantitative drug discovery solutions

  • Questions/ comments?

    www.xenologiq.com Quantitative drug discovery solutions

  • Backups

    www.xenologiq.com Quantitative drug discovery solutions

  • Initial clinical data; how could AEA exhibit pharmacology?

    Dosage: 10 mg PF

    OEA

    PEA

    LEA

    AEA

    TIME, hours

    FA

    As

    co

    ncen

    tra

    tio

    n,n

    M

    350300250200150100500

    50

    45

    40

    35

    30

    25

    20

    15

    10

    5

    0

    •AEA binds strongly to HSA (fu ~ 0.0001)

    •Primary pharmacology ~300 nM •Peak total AEA ~ 10nM

    •Peak free = 1 pM

  • How could AEA exhibit pharmacology?

    www.xenologiq.com Quantitative drug discovery solutions

    Some evidence AEA binds within membrane (THE JOURNAL OF BIOLOGICAL CHEMISTRY, Vol. 280, 2005).

    ie ~ order of magnitude for the hypothesis that we can drive CBR pharmacology

    Log D7.4 anandamide ~5.67^ KD = 2.1e-6*300nM = ~1pM

    ^brain:plasma rat Kp= 7.2 Thanks Katie Critchell PDM Pfizer.