Indirect Pharmacodynamic Models for Tolerance
Download
Report
Transcript Indirect Pharmacodynamic Models for Tolerance
APPLICATION OF PK/PD MODELING IN
DRUG DEVELOPMENT
Amarnath Sharma, Ph.D.
Pfizer Global R & D
Groton, CT
Objectives of Early Drug Development
Identification
of critical risk factors prior to
investment in full clinical development
selection of better compounds
Provide
critical data to identify safe and
effective dose and dose regimens
more efficient development
New Paradigm in Drug Development
PK/PD in patients &/or in experimental models
in healthy subjects (POM)
PK/PD in dose-ranging proof of
efficacy study in patients (POC)
Confirm PD in the pivotal studies
New Drug Application
Post-marketing comparative
PK/PD in patients
Why Study PK/PD ?
Characterize
time course of pharmacologic response
(therapeutic &/or toxic effects)
Understand complex relationships
– tolerance, sensitization, mechanistic delay
Explain
variability in response
Identify biomarkers and validate surrogate endpoints
Aid dose/dose regimen selection through simulation
Bridge clinical efficacy and safety results across ethnic
populations
Bridge clinical results between adult and pediatric
patients
Requirements to Characterize
PK/PD Relationship
Validated
biomarkers for therapeutic effects & toxicity
– Should be meaningful (relates to MOA), reproducible,
quantitative and allows frequent sampling to characterize the
time course of effect
– Validated Assay (reproducible, high precision….)
– Exposure-response relationship
Understanding
of pharmacologic behavior of the drug
and pathophysiology of the disease
– Pharmacology and pharmacokinetic modeling
Modeling Direct Responses
Pharmacodynamics
1.0
Static Functions Related
to Hill Equation
0.8
E = Eo ± S. CP
Eq 1
E = Eo ± S. ln CP
Eq 2
0.6
0.4
n
max .Cp
E
E = Eo ±
EC50 n + Cp n
0.2
E max .(Cp - CT )
=
±
o
E E
( EC50 - CT ) + (Cp - CT )
0.0
0.001
0.01
0.1
1
10
100
Eq 3
Eq 4
1000
Concentration/EC50
Examples of direct PD effect with equilibration delay: CNS effects of benzodiazepines &
anesthetics; Muscle Relaxants of d-tubocurarine
Complexities in PK/PD Modeling
Equilibration
Mechanistic
delay
delay
Tolerance
Sensitization
Active
Drug
metabolites
interaction
Modeling Indirect Responses
Drug
Examples: Anticoagulants effects of warfarin;
Gene-mediated effects of corticosteroids
kin
Cp
Vc
ke
Ce
Biosignal
R
o
kout
CL
Pharmacokinetics (equilibration delay)
Cp =
n
Ai.e-li.t
i =1
dCe =
keo .(Cp - Ce )
dt
Pharmacodynamics (mechanistic delay)
dR =
kin .H (t ) - kout .R
dt
dR = kin kout .H (t ).R
dt
Ro = kin / kout
n
E max .Ce
H (t ) = 1±(
)
n
n
EC50 + Ce
Dayneka et al., JPB, 1993
Jusko et al., JPB, 1995
Sharma & Jusko, JPB, 1996
Sharma & Jusko, BJCP, 1998
Examples
IL12:
Tolerance in efficacy & safety biomarker response
(IFNg).
CD4
mAbs: Validate a safety biomarker in the preclinical
transgenic mice model.
IL5
mAb: Biomarker (eosinophil) is not a validated
surrogate endpoint.
P38
MAPK: Characterize an experimental model of acute
inflammation for anti-TNF response.
Avitriptan:
Pop
Characterize safety profile (BP and heart rate).
PK/PD approach in Linezolid bridging program.
IL12: An example of complex
PK/PD relationship
IL12
A 70 kDa heterodimer cytokine (35+40 kDa subunits).
Enhances T helper 1-type immunity.
Potentiates secretion of IFNg by, and the cytolytic
activity of, NK cells and CTLs.
IL12-induced secretion of IFNg is required for activity.
mIL12 has potent antitumor& antimetastatic activity in
murine tumor models.
Under development for cancer and infectious diseases.
Phase I Study Design
Open
label dose-escalation study in cancer
patients.
A single dose of rhIL12 followed by cycles of
5 consecutive daily iv injection at the same
dose every 3 weeks.
Days 1
MTD
study
2 weeks washout
15
16
17
18
19
Repeat every 3 weeks
of 500 ng/kg was established in this
Atkins et al,Clin Cancer Res. 1997
Phase II Study Design
Open
label repeat-dose efficacy study in patients
with advanced renal cell carcinoma.
Cycles of 5 consecutive daily iv injection at MTD
(500 ng/kg) dose every 3 weeks.
Days 1 2 3 4
5
3 weeks washout
27 28 29 30
31
Repeat every
3 weeks
Leonard et al., Blood, 1997
Phase II Study Results
Treatment
was associated with unexpected serious
adverse events.
Most of the patients experienced serious AEs after
2nd and 3rd doses.
Two patients died and no one entered the 2nd cycle
due to drug related toxicity such as GI bleeding.
PK profiles for IL12 were comparable to those
observed in Phase I study.
Leonard et al., Blood, 1997
Reason for unexpected toxicity:
A four-fold
higher trough
IFNg concentrations in Phase
II may have caused the serious
toxicity.
Leonard et al., Blood, 1997
Summary
If
IFNg concentrations were used as a safety biomarker,
it would have been possible to avoid serious AEs by
stopping after 2nd dose in Phase II study.
A single dose of IL12 causes tolerance in its ability to
induce IFNg production upon further dosing.
IL12 produces tolerance rapidly (3-4 days) during
multiple dosing which lasts for a relatively long time
period (14 days) in humans.
PK/PD modeling to characterize schedule-dependent
IL12-induced IFNg production is crucial for designing
safe and effective dosing regimens.
Comparative PD of Anti-CD4 mAbs
in Transgenic Mice
Sharma et al., JPET, 2000
Anti-CD4 mAbs
Mediate
their immunomodulatory effects via
indirect response mechanisms:
– removal of CD4+ T cells via effector mechanism;
– down-modulation of cell surface CD4 via internalization or
stripping and/or
– inhibition of CD4-MHC II interactions.
Under
development for autoimmune disorder such
as rheumatoid arthritis.
Anti-CD4 mAbs
Clenoliximab
Keliximab
Cynomolgus Macaque
V-domain
Human C l-domain
VL
VH
VH
VH
VH
CH1 C H1 V H
H
H
CH2 CH2
Human g1 CH-domains
VL
VL
CH3 CH3
• Primate/human chimeric CD4 mAb of
IgG1 isotype.
• Does not mediate complement
dependent cytotoxicity.
• Exhibits efficient binding to human
IgG Fc receptors and can cause
depletion of CD4+ cells.
Pro
Cys 227
Pro
Ser
Cys 230
VH
VH
CH1 CH1
H
H
CH2 CH2
CH3 CH3
V L Cynomolgus Macaque
V-domain
VH
Human Cl-domain
235 Phe
Leu
Gly
Gly
Pro
240 Ser
Glu
Human g4 CH-domains
• IgG4 derivative of Keliximab.
• Does not mediate complement
dependent cytotoxicity.
• Does not exhibit efficient binding to
human IgG Fc receptors.
Reddy et al. J Immun, 2000
FcR and CD4 Mediated Cell Adhesion
mAb Binding to FcR g
(% Adhesion of THP-1 Cells)
25
20
Keliximab
Keliximab
Keliximab + sCD4
20
Clenoliximab
Keliximab F(ab') 2
15
15
10
10
5
5
0
0
0.1
1
10
100
1000
0.1
1
10
100
1000
mAb (ng/mL)
Reddy et al. J Immun, 2000
Study Design
Male
transgenic mice (n=10-13 per group) bearing
human CD4 in place of the mouse CD4.
Three dose levels (5, 25 & 125 mg/kg).
PK: unbound plasma mAb concentrations.
PD: CD4+ T cells; number of CD4 epitopes on the
surface of T cells and CD8+ T cells.
Preclinical Species
Plasma Keliximab Concentration (ug/mL)
Target-mediated Disposition
1000
hCD4+ Transgenic
CD4 knock-out
1 mg/kg
1 mg/kg
10 mg/kg
30 mg/kg
100
10
1
Davis et al., Drug Metab Disp, 1996
0.1
0.01
0
20
40
60
80
Time (hours)
100
120
140
PK Model for Anti-CD4 mAbs
Dose
kPT
Plasma
(CP)
Tissue
(CT)
V max . C P
Km + CP
dC p
dt
V max . C T
Km + CT
= -
V max. C p
Km+ Cp
- k .C p
PT
dC T
V max . C T
= k .CpPT
dt
Km+ CT
IC: Cp = Dose/Vc; CT = 0
Sharma et al., JPET, 2000
Mean Plasma Concentration (ng/mL)
Pharmacokinetics of Anti-CD4 mAbs
Clenoliximab
5mg/kg
25mg/kg
125mg/kg
1e+6
Keliximab
5mg/kg
25mg/kg
125mg/kg
1e+5
Parameter (unit)
Estimate
890
Vmax (mg/mL/h)
1e+4
Km (ng/mL)
1e+3
5249
Vc (mL)
2.5
VT (mL)
25.6
kPT (day-1)
0.15
1e+2
0
100
200
300
TIME (hours)
400
500
Sharma et al., JPET, 2000
PD Model for Anti-CD4 mAbs
koin
kout
CD4+ cell
SC50
Smax
dR
dt
= k in - k out . S(t) . R
S max . Cp
S(t) = 1 +
SC 50 + Cp
At a very high dose:
Smax = (Ro - Rmax) / Rmax
IC: Ro = kin/kout
Sharma et al., JPET, 2000
Sharma & Jusko, Br J Clin Pharmaco, 1998
PK/PD of Anti-CD4 mAbs
60
Keliximab
60
5 mg/kg
25 mg/kg
125 mg/kg
40
40
20
20
Clenoliximab
25 mg/kg
125 mg/kg
0
0
0
400
800
1200
1600
0
400
800
1200
1600
Time (hour)
Sharma et al., JPET, 2000
PD Parameters of Circulating
CD4+ T Cell Number
Treatment
Keliximab
Clenoliximab
0.035 (11%)
0.032
28.2 (18%)
16.2
SC50 (ng/mL)
37500 (54%)
419000
Ro (% lymphocytes)
34.1 (7.9%)
34.1
between-animal variability in R
13% (48%)
13%
proportional residual error
29% (20%)
26%
Kin (% lymphocytes/hr)
Smax
Variances
Summary
Clenoliximab
is less potent and efficient
than keliximab in causing depletion of
circulating CD4+ T cells.
The results of this study are similar to those
from clinical trials at comparable doses.
This study validates the transgenic mice as
an appropriate model for preclinical PK/PD
evaluation of anti-CD4 mAbs.