Principles of Clinical Pharmacology - PowerPoint PPT Presentation

1 / 53
About This Presentation
Title:

Principles of Clinical Pharmacology

Description:

Principles of Clinical Pharmacology Steven P. Stratton, Ph.D. * * * * 84 Complete Response, 34 Incomplete response * * * * Patients should not be made to feel that ... – PowerPoint PPT presentation

Number of Views:3154
Avg rating:3.0/5.0
Slides: 54
Provided by: vailworks
Category:

less

Transcript and Presenter's Notes

Title: Principles of Clinical Pharmacology


1
Principles ofClinical Pharmacology
  • Steven P. Stratton, Ph.D.

2
Learning Objectives
  • To define Pharmacokinetics Pharmacodynamics
  • To identify PK/PD approaches, terminology, and
    parameters
  • To consider endpoints for PK/PD modeling
  • To identify barriers and opportunities with
    molecularly targeted drugs
  • To see new advances in clinical pharmacology
  • To understand some practical considerations in
    design of PK studies in clinical protocols

3
Potential Therapeutic Outcomes
  • Efficacy without toxicity
  • Palliation
  • Efficacy with toxicity
  • Treatment, potentially curative
  • Toxicity without efficacy
  • Poison
  • Neither toxicity nor efficacy
  • Alternative medicine

4
Pharmacokinetics
5
Pharmacodynamics
6
Practical considerations in designing clinical
drug intervention trials
  • Why this drug?
  • What dose?
  • What schedule?
  • What combination?
  • What about other interactions?

7
Administering Drugs Things to consider
  • Age
  • Renal status
  • Liver function
  • Polymorphisms
  • Cytochrome P450 (genetics, drug interactions)
  • Acetylator status (genetics)
  • Target present?

8
Administering DrugsThings 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?
9
Audience Question 1 Once in the clinic, what
is the primary reason for failure of
experimental drugs to gain FDA approval?
  • Toxicity
  • Efficacy
  • Pharmacokinetic Properties
  • Cost
  • Marketing

10
Reasons for Attrition During Clinical Development
50
40
30
Percentage of New Drugs Failing
20
10
0
Other
Safety
PK
Efficacy
Toxicology
Commercial
Formulation
Cost of Goods
Nature Reviews Drug Discovery 2, 566-580 (2003)
11
PK Terminology
12
Audience Question 2 What is the most
important pharmacokinetic variable?
  • Volume of Distribution (Vd)
  • Bioavailability (F)
  • Clearance (CL)
  • Half-life (t1/2)
  • Area Under the Curve (AUC)

13
Apparent Volume of Distribution (Vd)
14
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 bound 10 free ? 11
free Free drug concentration ? 10
Experimental Drug B 99 bound 1 free ? 2
free Free drug concentration ? 100
15
Clearance (CL)
  • Renal
  • Hepatic
  • Lung

It hurts when I pee.
16
Area Under the Curve (AUC)
  • Integration of Conc. vs. Time
  • Measure of systemic exposure

17
Half-life (t½)
  • Time required to clear 50 of drug
  • Depends on Volume of Distribution (Vd) and
    Clearance (CL)
  • Multi-phasic (if you can capture the distribution
    phase)
  • Rule of Thumb Drug is cleared in 5 half-lives

t½ Vd x ln(2) / CL
18
Other Important Parameters
  • Peak plasma concentration
  • Bioavailability
  • Duration above a threshold concentration
  • Free drug vs. total drug
  • Cumulative dose
  • Bioactivation to active metabolite

19
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 the
    concentration
  • AUC not affected by schedule
  • Example doxorubicin

20
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

21
Audience Question 3 If you failed to abstain
from one of these, but had to be at work and
drug-free in one hour, which would be least
likely to result in your dismissal?
  • 5 mg oxycodone
  • 150 mg erlotinib
  • Top-shelf (Patron) margarita
  • 4-5 bong hits

22
What is Translational Research?
23
Translational Research
  • the interphase between basic research and its
    application in a clinical setting for the
    diagnosis, treatment, or prevention of a
    disease.
  • Dr. William Hait, Past Pres. AACR
  • Observation ? Practice
  • PK/PD is a cornerstone of translational research

24
PK/PD Modeling
25
PK Variability in Ovarian Cancer Patients250
mg/m2, 24 hr infusion, 22-23 hr sample, n 48
Cancer Chemother Pharmacol 3348-52 (1993)
26
PK/PD modeling of Taxol-induced neutropenia
  • Non-linear kinetics
  • Myelosuppression related to duration of threshold
    plasma concentration
  • Taxol 0.05 mM
  • Prediction of disposition and toxicity

Gianni et al J Clin Oncol 13180-190 (1995)
27
PK/PD ModelingEffect of formulation on
paclitaxel PK
  • First-Order Elimination (Abraxane)
  • Rate of elimination is proportional to drug
    concentration
  • Constant fraction of drug eliminated per unit time
  • Zero-Order Elimination (Taxol)
  • Rate of elimination constant regardless of drug
    concentration
  • Constant amount of drug eliminated per unit time

28
  • paclitaxel (Taxol)
  • 6 hr infusion, q 21d
  • Cremaphor formulation
  • Premedication
  • Non-linear kinetics
  • J Clin Oncology 91261-1267 (1991)
  • paclitaxel (ABI_007)
  • nanoparticles
  • 30 min infusion, q 21d
  • No cremaphor
  • No premeds
  • Linear kinetics
  • Clin Cancer Res 81038-1044 (2002)

Stratton Clin Pharm AACR/ASCO Vail 2005
29
PD Modeling Example Pharmacogenetics
Myelotoxicity and UGT genetic polymorphisms
  • Irinotecan
  • 350 mg/m2
  • 90 min infusion, q3w
  • n 66
  • SN-38 metabolism dependent on UGT variant
  • Identification of patients predisposed to severe
    irinotecan toxicity

Innocenti et al. J Clin Oncol 221382-1388 (2004)
30
Molecularly-targeted Drugs
We found a drug. Now go find something for it
to cure.
31
Shift Towards Target-based vs. Compound-based
Development
  • Compound-based (backward)
  • Interesting compound discovered with activity in
    in vitro models
  • Target-based (forward)
  • Protein or gene targets identified on
    carcinogenesis pathway.
  • Drugs designed to interfere with these specific
    targets

32
EGFR as a Molecular Target
  • Member of erbB family of receptor tyrosine
    kinases
  • 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 in angiogenesis,
    proliferation, and inhibition of apoptosis

33
EGFR Mechanism
Courtesy of Genentech
34
EGFR Targeted Therapy
  • Neutralizing monoclonal antibody
  • cetuximab
  • competitive inhibitor
  • prevents dimerization
  • Tyrosine kinase inhibitors
  • erlotinib, gefitinib
  • reversible inhibitors
  • lapatinib
  • duel EGFR/erbB2 irreversible inhibitor

35
Issues with molecularly targeted EGFR inhibitors
  • Mutation in EGFR
  • Activation of redundant pathways
  • Constitutive activation of downstream signaling
    factors
  • Ligand-independent activation of EGFR

36
Altered response to EGFR inhibitors
Mutations in the EGFR gene
  • EGFR mutations have been characterized in
    gliomas, NSCLC, breast, ovarian cancers
  • Activating mutations correlated with increased
    response to gefitinib in NSCLC

37
Resistance to EGFR inhibitors
Activation of redundant pathways
  • Resistance caused by activation of other tyrosine
    kinase receptors that bypass the EGFR pathway

Camp ER et al, Clin Cancer Res 11397-405 (2005)
38
Resistance to EGFR inhibitors
Constitutive activation of pathways downstream of
EGFR
Camp ER et al, Clin Cancer Res 11397-405 (2005)
39
Resistance to EGFR inhibitors
Ligand-independent activation of EGFR
  • EGFR can be activated by integrins
  • cetuximab could not inhibit this pathway

40
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

41
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 mechanism
  • Almost all in vitro mechanisms are convenient to
    believe once the xenograft data is positive
  • A good (valid) biomarker is hard to find

42
How do we improve targeted therapies?
  • Combinations
  • We need better tools to select the best
    patient/therapy combinations

43
Pharmacogenomics
  • How variations in the genome affect the response
    to medications

44
Personalized therapy in ovarian cancer A genomic
approach Dressman et al, JCO 25517 (2007)
  • Primary ovarian tumors collected at surgery from
    119 patients
  • All patients recd platinum-based therapy
  • 85 CR, 34 IR
  • DNA microarray analysis
  • Gene expression signatures used to predict
    oncogenic pathways activated in a tumor
  • Relationship between pathway activation and
    survival was analyzed in CRs and IRs

45
Colors represent predicted probability of pathway
activation
Src or E2F3 pathway activation differentiated
survival in Incomplete Responders
Pathway activation had no effect on survival in
Complete Responders
46
How is this helpful? Is it real?
  • Potential (very cool) application of pathway
    prediction in this patient population

Dressman et al, JCO 25517 (2007)
47
Practical Advice in PK Study Design
48
Practical Advice in PK Study Design
49
Typical Phase 1/PK Study
  • Goal
  • Capture adequate tissue samples to measure
    drug/metabolite levels over time
  • 0, ½, 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?

50
Practical Advice in PK Study Design
  • 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

51
Practical Advice in PK Study Design
  • 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
    frozen plasma ? Disaster
  • Cheap ink, cheap labels, and freezers dont mix

52
(No Transcript)
53
Compound-based vs. Target-based Drug Development
Compound-Based Target-Based Compound
isolated Target identified Compound screened
in cell culture Target validated in vitro
Activity in Animal Models Compounds screened for
target selectivity Mechanism
Toxicology Toxicology performed
Clinical Trials Phase I Phase I, II, III
Clinical Trials in Patients Expressing
Target Phase II Phase III
Write a Comment
User Comments (0)
About PowerShow.com