Stratton Clinical Pharmacology

Embed Size (px)

Citation preview

  • 8/22/2019 Stratton Clinical Pharmacology

    1/53

    Principles of

    Clinical PharmacologySteven P. Stratton, Ph.D.

  • 8/22/2019 Stratton Clinical Pharmacology

    2/53

    Learning Objectives

    To define Pharmacokinetics & Pharmacodynamics

    To identify PK/PD approaches, terminology, andparameters

    To consider endpoints for PK/PD modeling

    To identify barriers and opportunities withmolecularly targeted drugs

    To see new advances in clinical pharmacology

    To understand some practical considerations indesign of PK studies in clinical protocols

  • 8/22/2019 Stratton Clinical Pharmacology

    3/53

    Potential Therapeutic Outcomes

    Efficacy without toxicity Palliation

    Efficacy with toxicity Treatment, potentially curative

    Toxicity without efficacy

    Poison Neither toxicity nor efficacy

    Alternative medicine

  • 8/22/2019 Stratton Clinical Pharmacology

    4/53

    80 mg/m2

    Time (min)

    0 100 200 300 400 500

    PlasmaConcentrations(g

    /ml)

    0

    2

    4

    6

    8

    10PT 004

    PT 005

    PT 006PT 007

    LOQ

    Pharmacokinetics

  • 8/22/2019 Stratton Clinical Pharmacology

    5/53

    Pharmacodynamics

  • 8/22/2019 Stratton Clinical Pharmacology

    6/53

    Practical considerations in designingclinical drug intervention trials

    Why this drug?

    What dose?

    What schedule?

    What combination?

    What about other interactions?

  • 8/22/2019 Stratton Clinical Pharmacology

    7/53

    Administering Drugs:

    Things to consider Age

    Renal status

    Liver function Polymorphisms

    Cytochrome P450 (genetics, drug interactions)

    Acetylator status (genetics)

    Target present?

  • 8/22/2019 Stratton Clinical Pharmacology

    8/53

    Administering Drugs:

    Things to consider What should I measure?

    How do I measure it?

    Correct sampling schedule

    Validated method available?

    and most importantly

    What do I do with the answer?

  • 8/22/2019 Stratton Clinical Pharmacology

    9/53

    Audience Question #1:Once in the clinic, what is the primary

    reason for failure of experimental drugs togain FDA approval?

    A. Toxicity

    B. Efficacy

    C. Pharmacokinetic Properties

    D. CostE. Marketing

  • 8/22/2019 Stratton Clinical Pharmacology

    10/53

    Reasons for Attrition During Clinical Development

    Perc

    entageofNew

    DrugsFailin

    g

    0

    10

    20

    30

    40

    50

    Nature Reviews Drug Discovery2, 566-580 (2003)

  • 8/22/2019 Stratton Clinical Pharmacology

    11/53

    PK Terminology

  • 8/22/2019 Stratton Clinical Pharmacology

    12/53

    Audience Question #2:

    What is the most important

    pharmacokinetic variable?

    A. Volume of Distribution (Vd)

    B. Bioavailability (F)

    C. Clearance (CL)

    D. Half-life (t1/2)E. Area Under the Curve (AUC)

  • 8/22/2019 Stratton Clinical Pharmacology

    13/53

    Apparent Volume of Distribution (Vd)

    Vd =Concentration

    Amt of Drug (dose)

    Small VdLow tissue binding Large Vd

    Drug tightly bound

    Concentration =Vd

    Amt of Drug (dose)

  • 8/22/2019 Stratton Clinical Pharmacology

    14/53

    Protein Binding

    Large fraction of drug bound to tissue

    Unavailable for drug function

    Easily measured in vitro(% bound)

    Consequences

    What if bound drug is displaced?

    e.g. aspirin, warfarin displaces 1%

    Experimental Drug A: 90% bound10% free 11% freeFree drug concentration 10%

    Experimental Drug B: 99% bound1% free 2% freeFree drug concentration 100%

  • 8/22/2019 Stratton Clinical Pharmacology

    15/53

    Clearance (CL)

    It hurts when I pee.

    CL =Concentration

    Elimination Rate

    RenalHepatic

    Lung

  • 8/22/2019 Stratton Clinical Pharmacology

    16/53

    Area Under the Curve (AUC)

    Integration ofConc. vs. Time

    Measure ofsystemicexposure

    AUC

    Serum concentration

    (mg/mL)

  • 8/22/2019 Stratton Clinical Pharmacology

    17/53

    Half-life (t)

    Time required to clear50% of drug

    Depends on Volume ofDistribution (Vd) andClearance (CL)

    Multi-phasic (if you cancapture the distribution

    phase)

    Rule of Thumb: Drugis cleared in 5 half-lives

    t = Vd x ln(2) / CL

  • 8/22/2019 Stratton Clinical Pharmacology

    18/53

    Other Important Parameters

    Peak plasma concentration

    Bioavailability

    Duration above a threshold concentration

    Free drug vs. total drug

    Cumulative dose Bioactivation to active metabolite

  • 8/22/2019 Stratton Clinical Pharmacology

    19/53

    PK Analysis

    Linear Pharmacokinetics

    First order kinetics

    Covers most drugs Rate of change depends

    only on the current [drug]

    Half-life remains constant

    no matter how high theconcentration

    AUC not affected byschedule

    Example: doxorubicin

    dC

    dt= -kC

  • 8/22/2019 Stratton Clinical Pharmacology

    20/53

    PK Analysis

    Non-Linear Pharmacokinetics (zero order)

    Classic examples: ethanol, phenytoin

    Saturable metabolism Decreased CL at higher doses

    Shortened infusion increased AUC

    Examples: 5-FU, Taxol

    Saturable absorption

    Decreased proportional AUC at higher doses

    Lengthened infusion increased plasma conc.

    Examples: methotrexate, cisplatin

  • 8/22/2019 Stratton Clinical Pharmacology

    21/53

    Audience Question #3:

    If you failed to abstain from one of these,

    but had to be at work and drug-free in onehour, which would be least likely to resultin your dismissal?

    A. 5 mg oxycodone

    B. 150 mg erlotinib

    C. Top-shelf (Patron) margaritaD. 4-5 bong hits

  • 8/22/2019 Stratton Clinical Pharmacology

    22/53

    What is Translational Research?

  • 8/22/2019 Stratton Clinical Pharmacology

    23/53

    Translational Research

    the interphase between basic research andits application in a clinical setting for thediagnosis, treatment, or prevention of adisease.

    Dr. William Hait, Past Pres. AACR

    Observation Practice

    PK/PD is a cornerstone of translationalresearch

  • 8/22/2019 Stratton Clinical Pharmacology

    24/53

    PK/PD Modeling

  • 8/22/2019 Stratton Clinical Pharmacology

    25/53

    Cancer Chemother Pharmacol 33:48-52 (1993)

    PK Variability in Ovarian Cancer Patients250 mg/m2, 24 hr infusion, 22-23 hr sample, n = 48

    MyelosuppressionLoweredefficacy

  • 8/22/2019 Stratton Clinical Pharmacology

    26/53

    PK/PD modeling of Taxol-

    induced neutropenia Non-linear kinetics

    Myelosuppression relatedto duration of thresholdplasma concentration [Taxol] 0.05 M

    Prediction of dispositionand toxicity

    Gianni et al J Clin Oncol13:180-190 (1995)

  • 8/22/2019 Stratton Clinical Pharmacology

    27/53

    PK/PD ModelingEffect of formulation on paclitaxel PK

    First-Order Elimination (Abraxane)

    Rate of elimination is proportionalto drug concentration

    Constant fraction of drugeliminated per unit time

    Zero-Order Elimination (Taxol)

    Rate of elimination constantregardless of drug concentration

    Constant amount of drugeliminated per unit time

  • 8/22/2019 Stratton Clinical Pharmacology

    28/53

    Stratton Clin Pharm AACR/ASCO Vail 2005

    paclitaxel (ABI_007) nanoparticles 30 min infusion, q 21d

    No cremaphor No premeds Linear kinetics

    Clin Cancer Res8:1038-1044 (2002)

    paclitaxel (Taxol) 6 hr infusion, q 21d

    Cremaphor formulation Premedication

    Non-linear kinetics

    J Clin Oncology9:1261-1267 (1991)

    275 mg/m2

    250 mg/m2

    175 mg/m2

  • 8/22/2019 Stratton Clinical Pharmacology

    29/53

    Innocenti et al. J Clin Oncol22:1382-1388 (2004)

    PD Modeling Example: PharmacogeneticsMyelotoxicity and UGT genetic polymorphisms

    Irinotecan

    350 mg/m2

    90 min infusion, q3w

    n = 66

    SN-38 metabolismdependent on UGT variant

    Identification of patientspredisposed to severeirinotecan toxicity

    Grade 4

  • 8/22/2019 Stratton Clinical Pharmacology

    30/53

    We found a drug. Now go find something for it to cure.

    Molecularly-targeted Drugs

  • 8/22/2019 Stratton Clinical Pharmacology

    31/53

    Shift Towards Target-based vs.Compound-based Development

    Compound-based (backward)

    Interesting compound discovered with

    activity in in vitromodels

    Target-based (forward)

    Protein or gene targets identified oncarcinogenesis pathway.

    Drugs designed to interfere with thesespecific targets

  • 8/22/2019 Stratton Clinical Pharmacology

    32/53

    EGFR as a Molecular Target

    Member of erbB family of receptor tyrosinekinases EGFR (ErbB1), HER2/Neu (ErbB2), HER3 (ErbB3)

    and HER4 (ErbB4)

    Overexpressed in various solid tumors Overexpression has been correlated with poor

    prognosis

    EGFR signaling is implicated inangiogenesis, proliferation, and inhibition ofapoptosis

  • 8/22/2019 Stratton Clinical Pharmacology

    33/53

    EGFR Mechanism

    Courtesy of Genentech

  • 8/22/2019 Stratton Clinical Pharmacology

    34/53

    EGFR Targeted Therapy

    Neutralizing monoclonal antibody cetuximab

    competitive inhibitor

    prevents dimerization

    Tyrosine kinase inhibitors

    erlotinib, gefitinib reversible inhibitors

    lapatinib duel EGFR/erbB2 irreversible inhibitor

  • 8/22/2019 Stratton Clinical Pharmacology

    35/53

    Issues with molecularly targetedEGFR inhibitors

    Mutation in EGFR

    Activation of redundant pathways

    Constitutive activation of downstreamsignaling factors

    Ligand-independent activation of EGFR

  • 8/22/2019 Stratton Clinical Pharmacology

    36/53

    Altered response to EGFR inhibitors

    EGFR mutations have been characterized in

    gliomas, NSCLC, breast, ovarian cancers

    Activating mutations correlated withincreased response to gefitinib in NSCLC

    Mutations in the EGFR gene

  • 8/22/2019 Stratton Clinical Pharmacology

    37/53

    Resistance to EGFR inhibitors

    Resistance caused by activation of other tyrosine kinase receptorsthat bypass the EGFR pathway

    Camp ER et al, Clin Cancer Res11:397-405 (2005)

    Activation of redundant pathways

  • 8/22/2019 Stratton Clinical Pharmacology

    38/53

    Resistance to EGFR inhibitors

    Constitutive activation of pathways downstream of EGFR

    Activating mutations ingenes downstream ofEGFR signaling couldbypass the effect ofthe EGFR inhibitor

    Camp ER et al, Clin Cancer Res11:397-405 (2005)

  • 8/22/2019 Stratton Clinical Pharmacology

    39/53

    Resistance to EGFR inhibitors

    EGFR can be activated by integrins

    cetuximab could not inhibit this pathway

    Ligand-independent activation of EGFR

  • 8/22/2019 Stratton Clinical Pharmacology

    40/53

    Concerns with Targeted Therapy

    The Butterfly effect Predicting toxicities of a single target is difficult when the

    target of interest is relatively upstream in a pathway Example: bortezomib (Velcade) myelosuppression, fatigue,

    etc.

    Dosing regimens are difficult to determine High potency difficult detection of drug Cytostatic mechanism low toxicity, MED vs. MTD

    Targeted therapies are not as specific as we think(e.g., imatinib mesylate, sorafenib) Pleiotropism

  • 8/22/2019 Stratton Clinical Pharmacology

    41/53

    Concerns with Targeted Therapy(contd)

    Redundancy Cells that find a way get rewarded and select

    for resistance

    Delivery (chemistry) The drug may not reach the target in vivo (PK)

    Bogus mechanismAlmost all in vitromechanisms are convenient to

    believe once the xenograft data is positive

    A good (valid) biomarker is hard to find

  • 8/22/2019 Stratton Clinical Pharmacology

    42/53

    How do we improve

    targeted therapies?1. Combinations2. We need better tools to select the best

    patient/therapy combinations

    Personalized Medicine

    the future of clinicalpharmacology

  • 8/22/2019 Stratton Clinical Pharmacology

    43/53

    Pharmacogenomics

    How variations in the genome affect theresponse to medications

    P li d th i i

  • 8/22/2019 Stratton Clinical Pharmacology

    44/53

    Personalized therapy in ovarian cancer:A genomic approach

    Dressman et al, JCO 25:517 (2007)

    Primary ovarian tumors collected at surgeryfrom 119 patients

    All patients recd platinum-based therapy 85 CR, 34 IR

    DNA microarray analysis

    Gene expression signatures used to predictoncogenic pathways activated in a tumor

    Relationship between pathway activationand survival was analyzed in CRs and IRs

  • 8/22/2019 Stratton Clinical Pharmacology

    45/53

    Colors representpredicted probability ofpathway activation

    Src or E2F3 pathwayactivation differentiated

    survival in IncompleteResponders

    Pathway activation hadno effect on survival inComplete Responders

  • 8/22/2019 Stratton Clinical Pharmacology

    46/53

    How is this helpful? Is it real?

    Potential (very cool) application of pathwayprediction in this patient population

    Dressman et al,

    JCO 25:517 (2007)

  • 8/22/2019 Stratton Clinical Pharmacology

    47/53

    Practical Advice in PK Study Design

  • 8/22/2019 Stratton Clinical Pharmacology

    48/53

    I was hoping I could choose my own doctor

    Patient Considerations

    Its a baby. Regulations prohibit ourmentioning its race, age, or gender.

    Regulatory Considerations

    Practical Advice in PK Study Design

  • 8/22/2019 Stratton Clinical Pharmacology

    49/53

    Typical Phase 1/PK Study

    Goal

    Capture adequate tissue samples to

    measure drug/metabolite levels over time 0, , 1, 2, 4, 8, 24, 48 hr

    Day 8, Day 15

    Capture 4-5 half-lives if possible May need to collect urine, other fluids?

  • 8/22/2019 Stratton Clinical Pharmacology

    50/53

    Know your analyst Ensure that the analytical technique is available Ensure that the method is available, validated, and reliable Define sample preparation

    Know your sample size The biometrist is your friend visit them early and often

    Be kind to nurses Do you really want that 16 hr PK? Dont require a sample at the end of the infusion- too

    many things at once is trouble

    Practical Advice in PK Study Design

  • 8/22/2019 Stratton Clinical Pharmacology

    51/53

    Consider your patients Dont exsanguinate them

    Extended PK sampling can be exhausting

    Dont sample from the infusion port

    Define and monitor sample handling!! Ensure study personnel are informed and understand

    SOPs

    Shipping whole blood at room temp instead of frozenplasma Disaster

    Cheap ink, cheap labels, and freezers dont mix

    Practical Advice in PK Study Design

  • 8/22/2019 Stratton Clinical Pharmacology

    52/53

  • 8/22/2019 Stratton Clinical Pharmacology

    53/53

    Compound-Based Target-Based

    Compound isolated Target identified

    Compound screened in cell culture Target validated in vitro

    Activity in Animal Models Compounds screened fortarget selectivity

    Mechanism ToxicologyToxicology performed

    Clinical TrialsPhase I Phase I, II, III Clinical Trialsin Patients Expressing Target

    Phase II

    Phase III

    Compound-based vs. Target-basedDrug Development