14
ORIGINAL RESEARCH ARTICLE A Population Pharmacokinetic and Pharmacodynamic Study of a Peripheral j-Opioid Receptor Agonist CR665 and Oxycodone Anne E. Olesen Kim Kristensen Camilla Staahl Sherron Kell Gilbert Y. Wong Lars Arendt-Nielsen Asbjørn M. Drewes Published online: 5 December 2012 Ó Springer International Publishing Switzerland 2012 Abstract Background Peripherally acting opioids, particularly peripheral j-opioid agonists, may be effective for treating visceral pain by activating receptors expressed on afferent nerves within the gut. Objective The objective of this study was to investigate the pharmacokinetic/pharmacodynamic profile of a novel peripherally selective j-opioid agonist, CR665 (JNJ- 38488502), and compare it to that of oxycodone, a non- selective brain-penetrant opioid. Methods In a randomized, placebo-controlled, double- blind, three-way crossover study, healthy male volunteers were administered CR665 (0.36 mg/kg, intravenous), oxycodone (15 mg, oral) or placebo (intravenous and oral), followed by assessment of visceral pain tolerance thres- holds (VPTT) measured as volume of water (mL) in the bag placed on an oesophageal probe. Plasma drug con- centration data were used to generate pharmacokinetic models, which were then used to fit the VPTT data using NONMEM Ò VI to generate population pharmacokinetic/ pharmacodynamic models. Results CR665 kinetics were optimally fitted with a two-compartment model, while oxycodone kinetics were best described by a one-compartment model with transit compartment absorption feeding directly into the central compartment. For both drugs, the plasma concentration effects on VPTT were best fit by a direct linear model, i.e. without the concentration–analgesia delay character- istic of brain-penetrant opioids. The slope of oxycodone (0.089 mL per ng/mL) was steeper than that of CR665 (0.0035 mL per ng/mL) for the plasma drug concentration acting on the VPTT. Conclusion The results are consistent with the peripheral selectivity of CR665, as well as the possibility that peripheral actions of oxycodone contribute to its visceral analgesic efficacy. 1 Introduction Visceral pain is prevalent in clinical practice and causes challenges in pain management [1]. Currently, centrally acting, non-selective opioid agonists are used in the treat- ment of visceral pain. However, inadequate analgesia and undesirable adverse effects often limit their usage [2]. A. E. Olesen (&) A. M. Drewes Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg Hospital, Aarhus University Hospital, Mølleparkvej 4, 9000 Aalborg, Denmark e-mail: [email protected] K. Kristensen Insulin Pharmacology, Novo Nordisk A/S, Ma ˚løv, Denmark C. Staahl R & D, Gru ¨nenthal GmBH, Aachen, Germany S. Kell Research Development, Impax Pharmaceuticals, Hayward, CA, USA S. Kell ALZA Corporation, Mountain View, CA, USA G. Y. Wong Pfizer, Inc., Worldwide Research and Development, South San Francisco, CA, USA L. Arendt-Nielsen A. M. Drewes Center for Sensory-Motor Interaction, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark Clin Pharmacokinet (2013) 52:125–137 DOI 10.1007/s40262-012-0023-8

Drug Abuse 6

Embed Size (px)

DESCRIPTION

vhvj

Citation preview

Page 1: Drug Abuse 6

ORIGINAL RESEARCH ARTICLE

A Population Pharmacokinetic and Pharmacodynamic Studyof a Peripheral j-Opioid Receptor Agonist CR665 and Oxycodone

Anne E. Olesen • Kim Kristensen • Camilla Staahl • Sherron Kell •

Gilbert Y. Wong • Lars Arendt-Nielsen • Asbjørn M. Drewes

Published online: 5 December 2012

� Springer International Publishing Switzerland 2012

Abstract

Background Peripherally acting opioids, particularly

peripheral j-opioid agonists, may be effective for treating

visceral pain by activating receptors expressed on afferent

nerves within the gut.

Objective The objective of this study was to investigate

the pharmacokinetic/pharmacodynamic profile of a novel

peripherally selective j-opioid agonist, CR665 (JNJ-

38488502), and compare it to that of oxycodone, a non-

selective brain-penetrant opioid.

Methods In a randomized, placebo-controlled, double-

blind, three-way crossover study, healthy male volunteers

were administered CR665 (0.36 mg/kg, intravenous),

oxycodone (15 mg, oral) or placebo (intravenous and oral),

followed by assessment of visceral pain tolerance thres-

holds (VPTT) measured as volume of water (mL) in the

bag placed on an oesophageal probe. Plasma drug con-

centration data were used to generate pharmacokinetic

models, which were then used to fit the VPTT data using

NONMEM� VI to generate population pharmacokinetic/

pharmacodynamic models.

Results CR665 kinetics were optimally fitted with a

two-compartment model, while oxycodone kinetics were

best described by a one-compartment model with transit

compartment absorption feeding directly into the central

compartment. For both drugs, the plasma concentration

effects on VPTT were best fit by a direct linear model,

i.e. without the concentration–analgesia delay character-

istic of brain-penetrant opioids. The slope of oxycodone

(0.089 mL per ng/mL) was steeper than that of CR665

(0.0035 mL per ng/mL) for the plasma drug concentration

acting on the VPTT.

Conclusion The results are consistent with the peripheral

selectivity of CR665, as well as the possibility that

peripheral actions of oxycodone contribute to its visceral

analgesic efficacy.

1 Introduction

Visceral pain is prevalent in clinical practice and causes

challenges in pain management [1]. Currently, centrally

acting, non-selective opioid agonists are used in the treat-

ment of visceral pain. However, inadequate analgesia and

undesirable adverse effects often limit their usage [2].

A. E. Olesen (&) � A. M. Drewes

Mech-Sense, Department of Gastroenterology and Hepatology,

Aalborg Hospital, Aarhus University Hospital, Mølleparkvej 4,

9000 Aalborg, Denmark

e-mail: [email protected]

K. Kristensen

Insulin Pharmacology, Novo Nordisk A/S, Maløv, Denmark

C. Staahl

R & D, Grunenthal GmBH, Aachen, Germany

S. Kell

Research Development, Impax Pharmaceuticals,

Hayward, CA, USA

S. Kell

ALZA Corporation, Mountain View, CA, USA

G. Y. Wong

Pfizer, Inc., Worldwide Research and Development,

South San Francisco, CA, USA

L. Arendt-Nielsen � A. M. Drewes

Center for Sensory-Motor Interaction,

Department of Health Science and Technology,

Aalborg University, Aalborg, Denmark

Clin Pharmacokinet (2013) 52:125–137

DOI 10.1007/s40262-012-0023-8

Page 2: Drug Abuse 6

Because dose-limiting adverse effects such as euphoria,

sedation, respiratory depression and nausea are mediated

by l-opioid receptors in the central nervous system (CNS),

there has been a focus on the development of peripherally

acting opioids (i.e. opioids that do not readily enter the

CNS), as well as receptor subtype-selective opioids (e.g.

j- and d-opioid agonists) that do not interact with the

classical l-opioid receptor [3, 4]. Visceral afferents

expressing j-opioid receptors in the gut are thought to have

an important role in the modulation of visceral pain and

are, therefore, a possible receptor target for novel drugs to

attenuate visceral pain [4, 5]. CR665 (JNJ-38488502) is a

novel peripherally selective j-opioid receptor peptide

agonist that has demonstrated efficacy in rodent models of

visceral pain, as well as an experimental visceral pain

model in humans [6–8]. Moreover, animal studies have

demonstrated analgesic effect of the main metabolite of

CR665 (CR665N-oxide) [unpublished data].

Opioid effects can be difficult to evaluate in a clinical

setting due to the many confounding factors affecting a

patient’s pain experience. Experimental pain models in

healthy volunteers provide less confounding pain mea-

sures, and may be more suitable for establishing pharma-

cokinetic/pharmacodynamic relationships for analgesic

effects [9–11]. In population pharmacokinetic/pharmaco-

dynamic modelling, mathematical models are used to

summarize the information on dose–plasma concentration–

effect relationships. The population method is able to iso-

late and characterize variability between subjects and it

allows characterizing of the unique time course of the pain

behaviour [12]. Thus, this technique offers advantages over

traditional statistical methods in situations when high inter-

and/or intra-subject variability can be expected, e.g. in

research on pain behaviour. Pharmacokinetic/pharmaco-

dynamic modelling has possibilities to enable the under-

standing of the underlying mechanisms of drug action [13].

Additionally, the population method has the potential of

using sparse and randomly collected data in contrast to

traditional analysis [14].

The rationale for the present study was that the phar-

macokinetic/pharmacodynamic profile of CR665 could

support evidence relating to the peripheral mechanism of

action in the management of visceral pain [15–18]. The

pharmacokinetic/pharmacodynamic relationship for orally

administered oxycodone (15 mg) has previously been

successfully described for different dynamic endpoints in

human experimental pain models [9, 11] and, therefore,

oxycodone was used as active comparator.

The aim of this study was to investigate the pharma-

cokinetic/pharmacodynamic profile of single doses of a

novel peripherally selective j-opioid receptor peptide

agonist CR665 and compare it to that of oxycodone in a

human experimental model of visceral pain.

2 Subjects and Methods

2.1 Subjects and Study Design

This pharmacokinetic/pharmacodynamic analysis was based

on data from a previous study investigating the analgesic

efficacy of the peripheral j-opioid receptor agonist CR665

compared to oxycodone in a multi-modal, multi-tissue

experimental human pain model [8]. Healthy male volun-

teers (aged 18–45 years) were recruited at a single site to

participate in this randomized, double-blind, placebo-con-

trolled, three-way crossover study. The study was conducted

in the laboratory at Mech-Sense, Department of Gastroen-

terology, Aalborg Hospital, Aalborg, Denmark. Inclusion

criteria were as follows: healthy male aged 18–45 years,

inclusive; body mass index (BMI) within 18–25 kg/m2,

inclusive, and stable bodyweight; bodyweight B90 kg; good

general health with no clinically relevant abnormalities as

assessed by the investigator; semi-recumbent blood pressure

(after resting for 5 min) in the ranges of 90–139 mmHg

systolic and 50–89 mmHg diastolic, inclusive; negative

urine drug test at screening and at check-in prior to each

treatment period; negative alcohol test at check-in prior to

each treatment period; and no known allergies to any of the

compounds used in the study. Exclusion criteria were as

follows: blood donor within 4 weeks prior to dosing; use of

any medication up until 30 days before study start; drinking

of alcohol-, grapefruit juice-, caffeine- or chinin-containing

products 48 h before study start; smoking; drinking more

than 5 cups of coffee, tea or 2.5 L of cola a day; and drinking

more than 14 units of alcohol per week.

All subjects were informed about the risks of the study,

gave their written informed consent prior to participating,

and were compensated for participating. The study was

conducted in accordance with the Declaration of Helsinki

on biomedical research involving human subjects and

guidelines on Good Clinical Practice were followed. The

study protocol was approved by the Regional Ethics Com-

mittee, Aalborg, Denmark (registration no. VN-20060021)

and the Danish Medicine Agency, Copenhagen, Denmark

(ref. no. 2612-3145).

2.2 Medication

Equal numbers of subjects were randomly assigned to one

of three treatment sequences (ABC, BCA or CAB). Intra-

venous infusions were administered in the right forearm

vein and oral solutions were administered via the medica-

tion port in the oesophageal tube. All subjects received the

following treatments:

• Treatment A: CR665 solution 1.1 mL vials containing

10 mg/mL of active drug, total dose of 0.36 mg/kg

126 A. E. Olesen et al.

Page 3: Drug Abuse 6

mixed with an appropriate volume of isotonic saline

solution for injection to achieve a final volume of

25 mL and administered as an intravenous infusion

over 1 h at a rate of 25 mL/h, and an oral placebo

solution consisting of 15 mL orange juice solution.

• Treatment B: oxycodone 15 mg (1.5 mL of a 10 mg/mL

oral liquid solution of oxycodone hydrochloride in a

pure water vehicle) mixed with a 13.5 mL orange juice

solution used for the oral placebo solution in treatment

A, and a 1-h intravenous infusion of vehicle placebo

solution (i.e. isotonic saline for injection).

• Treatment C: 1-h intravenous infusion of vehicle

placebo solution and oral placebo solution, as used in

treatments A and B.

Subjects were admitted to the unit on the morning of

each dosing after overnight fasting and an oesophageal

tube and intravenous lines were placed prior to dosing.

Subjects were released from the clinic 24 h after the start of

dosing. Each treatment was followed by a 1- to 3-week

washout period. Prior to the pharmacokinetic analysis of

CR665 the individual doses were calculated by multiplying

the dose in mg/kg with individual bodyweights.

2.3 Pain Assessment

The visceral pain assessment parameter was measured

before each treatment, and at 30, 60 and 90 min after drug

administration.

The oesophageal probe has been described previously

[19]. The bag, attached to the probe, was placed 7 cm

proximal to the sphincter.

Water was infused to the bag by infusion channels

connected to a precision infusion–withdrawal pump (Type

111, Ole Dich Instrument Makers, Hvidovre, Denmark).

For distension, the oesophageal bag was filled with 37 �C

water at a constant infusion rate of 10 mL/min. As a pre-

viously validated measure of tolerated distension, the total

volumes of water (mL) in the oesophageal bag at pain

detection threshold and moderate pain were recorded.

Stimulation was stopped when subjects reported moderate

pain.

A single 10-point electronic visual analogue scale was

used to assess painful sensations in response to the dis-

tension of the bag in the oesophagus. A rating of 5 was

defined as the ‘pain detection threshold’ and a rating of 7 as

‘moderate pain’ [20].

2.4 Blood Samples

Blood samples for pharmacokinetic analysis were drawn

from an intravenous line in the left forearm pre-dosing and

then at 5, 15, 30, 60, 75 and 120 min after start of dosing.

Therefore, collection of blood samples was initiated during

the 1-h infusion phase. Blood was collected into pre-chilled

3 mL tripotassium (K3) EDTA-containing glass tubes

(Vacutainer�) and placed on ice for 1–2 h until centrifu-

gation. The samples were centrifuged at 1,0009g (about

2,500–3,000 rpm) for 10 min at approximately 4 �C. For

each sample, the separated plasma was transferred into two

polypropylene tubes and stored immediately at approxi-

mately –20 �C until analysis.

The analysis of the plasma samples for the determina-

tion of the concentrations of CR665 and its N-oxide

metabolite (CR665N-oxide) was carried out at Covance

Laboratories Limited (UK). Plasma concentrations were

determined using liquid chromatography with tandem mass

spectrometric detection (LC-MS/MS) with a lower limit of

quantification of 1 ng/mL.

The analysis of the plasma samples for the determina-

tion of the concentrations of oxycodone was carried out at

CEDRA Corporation (USA). Oxycodone concentrations

were determined by high-pressure liquid chromatography

(HPLC) with LC-MS/MS. The limits of quantification were

0.1 ng/mL for oxycodone. Values that were below the limit

of quantification were considered to be zero. The calibra-

tion range was 0.1–50 ng/mL.

Inter- and intra-day precision (% coefficient of varia-

tion) were below 10.2 and 10.5 %, respectively, across the

entire calibration range for all analyses.

2.5 General Population Modelling Methods

In total, 180 plasma concentration samples of CR665,

including 72 metabolite samples, were available for the

population pharmacokinetic analysis. For pharmacokinetic/

pharmacodynamic modelling an additional 140 pharma-

codynamic measurements were used. For the oxycodone

pharmacokinetic model, 91 plasma concentration samples

and 144 pharmacodynamic measurements were available

for the modelling. Both pharmacokinetic and pharmaco-

dynamic models were fitted to the data using non-linear

mixed–effects modelling software NONMEM� version VI,

level 1.0 (Globomax LLC, Elliott City, MD, USA), with

the first-order conditional estimation with interaction

approach. Unexplained inter-individual variability (IIV) in

structural model parameters was estimated using a log–

normal error model if not otherwise indicated (Eq. 1):

Pj ¼ TVP� egj ð1Þ

where Pj is the individual value of the parameter in the jth

individual, TVP is the typical value of the parameter for the

population and gj is an independent random variable with a

mean of zero and variance of x2.

Residual error was represented by a combined propor-

tional and/or additive error model. This was reduced to

PopPK/PD Study of CR665 and Oxycodone 127

Page 4: Drug Abuse 6

either a proportional or an additive model by omitting

small residual error terms, as indicated.

The main tool for selection between hierarchical models

was the difference in objective function value (OFV)

between models, i.e. the likelihood ratio test. The OFV is

proportional to minus twice the log likelihood and the

difference in OFV for the two models is approximately

chi-square (v2)-distributed. A difference in OFV of greater

than 3.84 is significant at the 5 % level if the models differ

by one parameter. Corresponding values for p = 0.01 and

p = 0.001 are 6.63 and 10.83, respectively.

Goodness of fit was also assessed by visual inspection of

plots of observed and predicted values against time,

residual plots and the distribution of the residual error. The

standard errors of model parameters outputted by NON-

MEM� were compared with 1,000 bootstraps of these final

models from 1,000 models using Perl-speaks-NONMEM�

(PsN) version 3.4.2 [21, 22].

2.5.1 Pharmacokinetic Models

Using all available data, pharmacokinetic modelling of the

plasma drug concentrations was performed to make infer-

ences about the kinetics of intravenously administered

CR665 and orally administered oxycodone in these healthy

volunteers. The pharmacokinetics was then used to provide

input concentrations for separate subsequent pharmacody-

namic models. The CR665 and CR665N-oxide concentra-

tion data were fitted to one- and two-compartment

pharmacokinetic models, with zero-order infusion for

60 min. A zero-order reaction has a rate that is independent

of the concentration of the reactant and increasing the

concentration of the reacting species will not speed up the

rate of the reaction. Rate constants of the CR665 and

metabolite model were used to determine which process in

the model was the rate-limiting step in the pharmacokinetic

model.

The oxycodone concentration data were fitted to one-

and two-compartment pharmacokinetic models, with oral

absorption represented as either a first-order absorption

process with a lag term, or as one or two chains of transit

compartments [23].

2.5.2 Effect Models

Models were also tested for the placebo response encom-

passing any changes in oesophageal distension. These were

models with no systematic change from baseline value,

linear change with time and quadratic change with time

(the latter giving a curved shape to effect versus time).

Details of the equations used and their purpose were

described previously by our group [11].

The combined data for the placebo and CR665 or oxy-

codone response for each subject were modelled together in

this stage of the analysis. The placebo response (if any) was

assumed to be additive with any drug response (that is, the

placebo data established what would have happened if the

drug dose was zero). Linear and maximum concentration–

effect relationships were examined as described in Table 1.

For linear models, the concentration–effect relationship was

described by the term ‘slopeD’ (D for drug). Positive values

indicated concentration-dependent analgesia. The models

were characterized by the maximum effect (Emax) and the

concentration producing 50 % of the maximum effect

(EC50). Pain measures were related to either the measured

venous plasma concentration of drug (C) or concentration of

the drug in the hypothetical effect compartment (Ceff). The

Ceff was delayed relative to the plasma concentration by an

equilibrium rate constant between plasma and the effect

compartment (ke0). If the pain measures were not delayed

relative to the opioid plasma concentration (i.e. the elimi-

nation half-life for effect–site equilibrium [t�,ke0] = 0), the

data would support a pharmacodynamic model without a

separate effect compartment. If the analgesic effects were

delayed relative to the opioid plasma concentration, e.g. due

to a requirement for transport across the blood–brain barrier,

the data would support a pharmacodynamic model with an

effect compartment, with a delay quantified by a positive,

non-zero value of t�,ke0.

2.6 Statistical Analysis

The parameter estimates obtained using NONMEM� were

expressed as the population average value and the popu-

lation parameter variability (its variability across the

population).

2.6.1 Model Evaluation

A predictive model check was used to validate the final

model using PsN version 3.4.2 [21, 22]. The final popula-

tion pharmacokinetic and pharmacokinetic/pharmacody-

namic model, including the final fixed and random effects,

were used to simulate a dataset 1,000 times for the subjects

studied. The 95 % confidence intervals for the simulated

plasma concentration–time profile or effect–time profile

were plotted against the observed data [24, 25].

3 Results

Eighteen healthy, non-smoking, opioid-naıve male volun-

teers (17 Caucasian and 1 Asian; ages 19–43 years, median

25 years; bodyweight 62.4–94.5 kg, median 80.0 kg; BMI

20.5–27.4 kg/m2, median 23.6 kg/m2) completed the study.

128 A. E. Olesen et al.

Page 5: Drug Abuse 6

The mean plasma concentrations of CR665, the

N-oxide metabolite of CR665 and oxycodone for all 18

healthy subjects are shown in Fig. 1. The subjects’

tolerances to pain induced by oesophageal distensions

measured as visceral pain tolerance threshold (VPTT)

versus time following placebo and drug treatment are

shown in Fig. 2.

3.1 Pharmacokinetics

3.1.1 CR665

A two-compartment and one-compartment distribution

model was fitted to CR665 and N-oxide metabolite data,

respectively. Distribution of CR665 was controlled by

rate constants k12 (first-order rate constant of drug

transfer for the central compartment to the peripheral

compartment) and k21 (first-order rate of drug transfer for

the peripheral compartment to the central compartment).

The elimination of CR665 was described by a first-order

formation rate constant (k13) from the central compart-

ment of CR665 to the N-oxide compartment, reflecting

the known conversion of the parent compound to this

active metabolite. Due to lack of information during the

elimination of the metabolite, a first-order elimination

rate constant for this metabolite (k30), equal to the for-

mation rate constant of metabolite (i.e. k30 = k13), best

described the pharmacokinetics of the N-oxide following

intravenous infusion of CR665 (a schematic model is

depicted in Fig. 3).

10 1

Time (h)

20.5 1.5

10

100

a 1,000

Pla

sma

conc

entr

atio

n (n

g/m

L)

0.10 1

Time (h)

20.5 1.5

1

10

b 100

Pla

sma

conc

entr

atio

n (n

g/m

L)

CR665N-oxide metabolite

Fig. 1 Spaghetti plots describing individual time courses of the

plasma concentration of CR665 and its N-oxide metabolite following

intravenous infusion over 1 h of 0.36 mg/kg CR665 (a) and oral

administration of 15 mg oxycodone (b)

Ta

ble

1T

este

dm

od

els

of

the

CR

66

5an

do

xy

cod

on

eco

nce

ntr

atio

n–

effe

ctre

lati

on

ship

s

Model

Des

crip

tion

Equat

ion

Purp

ose

1P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

linea

rphar

mac

odynam

icm

odel

E=

bas

elin

e?

slopeD

9C

Tes

ted

the

assu

mpti

on

that

the

dat

aco

uld

be

des

crib

edby

ali

nea

rm

odel

wit

hno

effe

ct

del

ay

2P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

IIV

inbas

elin

ean

dsl

ope

wit

h

Cfi

xed

atze

ro

E=

bas

elin

e?

slopeD

9C

Tes

ted

the

assu

mpti

on

that

dru

gco

nce

ntr

atio

ns

did

not

infl

uen

ceth

eef

fect

3P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

log-l

inea

rphar

mac

odynam

ic

model

E=

m9

ln(C

?bas

elin

e)T

este

dth

eas

sum

pti

on

that

the

dat

aco

uld

be

des

crib

edby

ali

nea

rm

odel

wit

hno

effe

ct

del

ay

4P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

non-l

inea

rphar

mac

odynam

ic

model

;a

refe

rence

non-l

inea

rm

odel

wit

hno

effe

ctco

mpar

tmen

t

E=

bas

elin

e?

(Em

ax

9C

)/

(EC

50

?C

)

Tes

ted

the

assu

mpti

on

that

the

dat

aco

uld

be

des

crib

edby

anon-l

inea

rm

odel

wit

hno

effe

ctdel

ay;

hig

hE

C50

val

ues

mad

eth

ism

odel

rever

tto

ali

nea

rm

odel

5P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

linea

rphar

mac

odynam

icm

odel

wit

hef

fect

del

ay;

IIV

inbas

elin

e,sl

ope

and

ke0

Ceff0

=k

e0

9(C

-C

eff)

As

for

model

1but

wit

ha

ke0

term

E=

bas

elin

e?

slopeD

9C

eff

6P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

non-l

inea

rphar

mac

odynam

ic

model

wit

hef

fect

del

ay

Ceff0

=k

e0

9(C

-C

eff)

As

for

model

3but

wit

ha

ke0

term

E=

m9

ln(C

eff

?bas

elin

e)

7P

Km

odel

inte

rpola

tion

of

pla

sma

conce

ntr

atio

ns;

non-l

inea

rphar

mac

odynam

ic

model

wit

hef

fect

del

ay

Ceff0

=k

e0

9(C

-C

eff)

As

for

model

4but

wit

ha

ke0

term

E=

bas

elin

e?

(Em

ax

9C

eff)/

(EC

50

?C

eff)

Cpla

sma

dru

gco

nce

ntr

atio

n,

Ceff

obse

rved

conce

ntr

atio

nin

the

effe

ctco

mpar

tmen

t,C

eff0

esti

mat

edco

nce

ntr

atio

nin

the

effe

ctco

mpar

tmen

t,E

tim

eco

urs

eof

the

effe

ctof

the

oxyco

done

dose

,E

C50

pla

sma

dru

g

conce

ntr

atio

nat

whic

hth

ere

was

ahal

f-re

duct

ion

inth

epai

nsc

ore

,E

max

pla

sma

dru

gco

nce

ntr

atio

nat

whic

hth

ere

was

am

axim

um

reduct

ion

inth

epai

nsc

ore

,II

Vin

ter-

indiv

idual

var

iabil

ity,

k e0

equil

ibri

um

rate

const

ant

bet

wee

npla

sma

and

the

effe

ctco

mpar

tmen

t,w

hic

hw

asal

soa

popula

tion

par

amet

er,

msl

ope

of

the

appar

ent

linea

rse

gm

ent

of

the

log-t

ransf

orm

edco

nce

ntr

atio

nver

sus

resp

onse

curv

e,P

Kphar

mac

okin

etic

,

slopeD

conce

ntr

atio

n–ef

fect

rela

tionsh

ip

PopPK/PD Study of CR665 and Oxycodone 129

Page 6: Drug Abuse 6

Covariate bodyweight (WT) was incorporated into the

models according to Holford [26], so the volumes of

distribution were scaled with the quantity (WT/median

bodyweight), and rate constants with the quantity (WT/

median bodyweight)-0.25. IIV was found for the apparent

volume of distribution of the central compartment for the

parent compound (V1) and metabolite (V3) as well as for

k12. Various one- and two-compartment distribution models

with similar and different elimination rate constants were

tested, with IIV in different parameters estimated using an

exponential error structure, and residual error modelled as a

proportional error structure. The model described above

was judged based on convergence of the estimation and

covariance routines, goodness-of-fit plots, precision of the

population pharmacokinetic parameters, and minimum

value of the objective function, a statistical criterion for the

addition of a single parameter, as the model best suited for

further pharmacokinetic/pharmacodynamic modelling of

CR665. The parameters for this model are summarized in

Table 2 and visual predicted check plots are shown in

Fig. 4. The model seems to describe the data of both CR665

and metabolite well as the visual predicted check plots

shows that the 2.5th, 50th and 97.5th percentiles of the

observed data are closely followed by the by the simulated

2.5th, 50th and 97.5th percentiles. In addition, the areas of

95 % prediction intervals around the 2.5th, 50th and 97.5th

percentiles of the simulated data overlap the 2.5th, 50th and

97.5th percentiles of the observed data.

3.1.2 Oxycodone

For the observed oxycodone plasma concentrations, a one-

compartment model with a single transit chain representing

CR665OxycodonePlacebo

b

0

Time (h)

10

20

30

50

40

60

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

c

0

Time (h)

0 1.0 2.00.5 1.5 0 1.0 2.00.5 1.5

10

20

30

50

40

60

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

151.00

Time (h)

2.01.50.5

20

25

a30

CR665Placebo

OxycodonePlacebo

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

Fig. 2 a Oesophageal distension measured as visceral pain tolerance

threshold (mean ± standard error of the mean) vs. nominal time

following CR665, oxycodone and placebo treatment. b, c Spaghetti

plots describing individual time courses of the visceral pain tolerance

threshold after 1 h intravenous infusion of CR665 and placebo (b) and

oral administration of 15 mg oxycodone and placebo (c)

ktr ktr ktr ktr ktrAmountA

Vd/F

CL/F

TransA1

TransAn −1

TransAn

MTT

IV infusion

CR665V1

V2

k12

k21

k13 k30N-oxideV3

a

b

Fig. 3 a Schematic representation of the CR665 pharmacokinetic

model. b Schematic representation of the oxycodone pharmacokinetic

model. A, A1, An–1, An represent the amount of oxycodone in each

compartment, CL/F apparent total oral clearance, IV intravenous, k12

first-order rate constant of drug transfer for the central compartment

to the peripheral compartment, k13 first-order rate constant of

metabolite formation, k30 elimination rate constant of N-oxide (k13

and k30 are equal in the final model), k21 first-order rate constant of

drug transfer for the peripheral compartment to the central compart-

ment, ktr rate constants for oxycodone transfer through transit

compartment, MTT mean transit time for oxycodone movement

through n transit compartments, Trans transit compartment, V1 central

volume of distribution for CR665, V2 peripheral volume of distribu-

tion, V3 central volume of distribution for CR665N-oxide, Vd/Fvolume of distribution after oral administration of the drug

130 A. E. Olesen et al.

Page 7: Drug Abuse 6

gastrointestinal absorption after oral administration pro-

vided the best fit to the data (the model is depicted in

Fig. 3). The parameters for this model are summarized in

Table 3. The absorption process was characterized by a

mean transit time of 0.295 h (17.7 min), with a typical

value of N (number of compartments in the transit

chain) = 4.88. Visual predicted check plots performed for

the oxycodone pharmacokinetic model are shown in Fig. 4.

The model seems to describe the data of oxycodone well as

the visual predicted check plots shows that the 2.5th, 50th

and 97.5th percentiles of the observed data are closely

followed by the simulated 2.5th, 50th and 97.5th percen-

tiles. In addition, the areas of 95 % prediction intervals

around the 2.5th, 50th and 97.5th percentiles of the simu-

lated data overlap the 2.5th, 50th and 97.5th percentiles of

the observed data.

3.2 Pharmacodynamics

3.2.1 Placebo

A baseline model was selected for the placebo measured

effect. As the study was a three-way crossover study,

the same placebo data were used for both the CR665 or

oxycodone pharmacodynamic modelling. In the final CR665

or oxycodone models similar estimates of the population

mean baseline of 21.7 and 21.0 mL as well as similar base-

line IIV of 0.175 and 0.180 mL, respectively, were found

(see Table 4).

3.2.2 Cr665

Due to a large difference in concentrations of parent

compound and metabolite (Fig. 1), parent compound was

considered the driver for the dynamics and only parent

compound was included in the pharmacokinetic/pharma-

codynamic modelling. The model with the best fit of the

data from the CR665 arm of the study was Model 1,

describing a linear relationship between the concentration

and the effect, with IIV in the baseline–effect relationship

and a proportional error model. There was a clear

advantage of Model 1 over the baseline model, Model 2

(OFV difference of [5, see Table 5), demonstrating a

significant analgesic effect of CR665 on the oesophageal

distension VPTT. This finding was supported by the

positive value of slopeD (concentration–effect relation-

ship) for this model (Table 4). Model 3 had a higher OFV

than Model 1. Models 4, 5 and 6 all had a significant

drop in the OFV but EC50 values for Model 4 were very

low (0.05 ng/mL), and the ke0 of Models 5 and 6 was low

and high, respectively. None of Models 4, 5 and 6 passed

the covariance step of NONMEM�. The non-linear model

Emax Model 7 in Table 5 with an effect delay ke0 passed

the covariance step with an OFV difference of [10

compared to Model 1. The estimation of the parameters

were baseline 20.0 mL (95 % CI 14.5–25.5), ke0 3.61 h

(95 % CI 1.14–6.51), Emax 3.65 mL (95 % CI 2.07–5.23)

and EC50 11.2 ng/mL (95% CI –2.52 to 24.9). However,

considering the study design with a relatively sparse

number of pharmacodynamic samples and use of only

one dose level, the maximum effect of CR665 might not

have been obtained and, thus, Model 1 was selected as

the final pharmacokinetic/pharmacodynamic model for

CR665.

3.2.3 Oxycodone

The model with the best fit of the data from the oxycodone

arm of the study was Model 1, describing a linear rela-

tionship between the concentration and the effect, with IIV

Table 2 Parameters of the best pharmacokinetic model for CR665

and its N-oxidea

Parameter Population-

typical value

[%RSE]

Median parameter estimates of

1,000 bootstrap replicates

[2.5–97.5 % range]

V1 (L) 8.08 [8.33] 8.11 [6.93–9.60]

k13 ( = k30) [h-1] 0.501 [15.6] 0.505 [0.367–0.670]

k12 (h-1) 4.85 [9.65] 4.85 [3.79–5.70]

k21 (h-1) 0.0778 [13.1] 0.078 [0.056–0.099]

V3 (L) 488 [21.1] 493 [315–741]

Inter-individual variability (x)

xV1 0.0166 [65.1] 0.0155 [0.0012–0.0621]

xk12 0.0331 [57.4] 0.0314 [0.0041–0.0906]

xV3 0.164 [35.9] 0.145 [0.054–0.274]

Residual variability (r)

Proportional

error—CR665

(%CV)b

0.113 [12.9] 0.109 [0.083–0.143]

Proportional

error—N-oxide

(%CV)b

0.0311 [17.2] 0.030 [0.018–0.041]

CL clearance, CV coefficient of variation, k12 first-order rate constant

of drug transfer from the central compartment to the peripheral

compartment, k13 first-order rate constant of metabolite formation, k21

first-order rate constant of drug transfer from the peripheral com-

partment to the central compartment, k30 first-order rate constant of

metabolite elimination, RSE relative standard error, V1 apparent

volume of the central compartment for the parent compound, V3

apparent volume of the central compartment for the metabolite, WTbodyweighta Pharmacokinetic parameter values for CR665 following adminis-

tration of 0.36 mg/kg as intravenous infusion. All parameters were

bodyweight normalized so the volumes were scaled with WT/median

bodyweight (80 kg), and rate constants with (WT/median bodyweight

[80 kg])-0.25

b The residual errors (proportional) represented the variability in the

data that was unexplained by the model

PopPK/PD Study of CR665 and Oxycodone 131

Page 8: Drug Abuse 6

in the baseline–effect relationship and a mixed structural

error model, i.e. a combined proportional and additive error

model. There was a clear advantage of Model 1 over the

baseline model (Model 2) as the OFV difference was

greater than 20 (see Table 6), thus demonstrating a sig-

nificant analgesic effect of oxycodone on the oesophageal

distension VPTT. This result was also supported by the

positive value of slopeD for this model (Table 4).

Models 3 and 4 also converged and passed the covari-

ance step with a non-significant drop in OFV and a sig-

nificant drop in OFV [6, respectively. The estimation of

the parameters of direct Emax for Model 3 were baseline

21.1 mL (95 % CI 16.8–25.4), Emax 13.2 mL (95 % CI

10.0–16.4) and EC50 166 ng/mL (95 % CI –69.2 to 401).

However, considering the study design with a relatively

sparse number of pharmacodynamic samples and use of

a

Time (h)

CR665

2.00.0 1.00.5 1.5

500

1000

Pla

sma

conc

entr

atio

n (n

g/m

L)b

Time (h)

CR665 N-oxide

5

10

Pla

sma

conc

entr

atio

n (n

g/m

L)

c

Time (h)

Oxycodone

10

20

Pla

sma

conc

entr

atio

n (n

g/m

L)

30

40

50

60

2.00.0 1.00.5 1.5 2.00.0 1.00.5 1.5

Fig. 4 Visual predictive check of pharmacokinetics of CR665 (a),

CR665N-oxide (b) and oxycodone (c) including 95 % prediction

intervals. On each plot: observed data (circles), median (solid red line),

2.5th and 97.5th percentiles (dashed red lines) of the observations. The

2.5th, 50th and 97.5th percentiles of the simulated data are plotted as

dashed black lines, solid black lines and dashed black lines, respec-

tively. The 95 % predicted confidence intervals around 2.5th, 50th and

97.5th percentiles of the simulated data are shown as coloured areas:

the 95 % confidence interval for the median (pink areas) and the 2.5th

and 97.5th percentiles (blue areas) of the simulated data

Table 3 Parameters of the best pharmacokinetic model for oxycodonea

Parameter Population typical value [%RSE] Median parameter estimates

of 1,000 bootstrap replicates [2.5–97.5 % range]

CL/F (L/h) 201 [5.12] 201 [185–221]

V1/F (L) 356 [6.43] 355 [310–401]

MTT (h) 0.295 [11.3] 0.300 [0.240–0.380]

NN -4.06 [9.29] –4.24 [–4.76 to –3.73]

Nb 4.88 [2.61–8.31] NC

Inter-individual variability (x)

xV1/F 0.0527 [42.1] 0.0420 [0.009–0.086]

xMTT 0.192 [31.6] 0.190 [0.07–0.32]

xNNc 0.516 [52.3] 0.490 [0.097–1.235]

Residual variability (r)

Proportional error (%CV)d 0.0638 [41.4] 0.0700 [0.0200–0.1300]

Additive error (nmol/L)d 0.131 [63.8] 0.098 [0.022–0.295]

CL/F apparent clearance after oral administration, CV coefficient of variation, MTT mean transit time for absorption, N number of compartments

in the transit chain (untransformed), NC not calculated, NN number of compartments in the transit chain (logit transformed), RSE relative

standard error, V1/F apparent volume of the central compartment for the parent compound after oral administration of the druga Pharmacokinetic parameter values for immediate-release oxycodone 15 mg, administered orallyb The logit transform constrained N to be between 1 and 200 via the equation N = 199 9 EXP(NN)/[1 ? EXP(NN)] ? 1c Additived The residual errors (additive and proportional) represented the variability in the data that was unexplained by the model. The additive error was

the component of the total error that was the same regardless of the opioid concentration. The proportional error was the component of the total

error that was proportional to the concentration

132 A. E. Olesen et al.

Page 9: Drug Abuse 6

Ta

ble

4E

ssen

tial

feat

ure

so

fth

eb

est

mo

del

so

fth

ep

ain

mea

sure

Co

mpo

un

dan

dst

imu

lus

mo

dal

ity

Bas

elin

ep

ain

val

ue

[95

%C

I]B

asel

ine

IIV

(SE

[95

%C

I])

Conce

ntr

atio

n–ef

fect

rela

tionsh

ip(s

lopeD

)(p

ain

un

its

per

ng

/mL

of

com

po

un

d[9

5%

CI]

)

Slo

peD

IIV

(SE

[95

%C

I])

Res

idual

erro

r(%

[95

%C

I])

CR

665

VP

TT

(mL

)a

Po

pu

lati

on

esti

mat

eb2

1.7

[17

.5–

25

.9]

0.1

75

[0.0

81

5–

0.2

68]

0.0

03

5[0

.00

08

7–

0.0

061

]N

A0

.04

07

[0.0

276

–0

.05

38

]c

Bo

ots

trap

esti

mat

ed2

1.6

[17

.8–

26

.5]

0.1

59

[0.0

79

8–

0.2

67]

0.0

03

5[0

.00

08

4–

0.0

060

]N

A0

.04

00

[0.0

290

–0

.05

39

]

Ox

yco

do

ne

VP

TT

(mL

)a

Po

pu

lati

on

esti

mat

eb2

1.0

[16

.7–

25

.3]

0.1

8[0

.08

75

–0

.27

3]

0.0

79

3[0

.01

34

–0

.14

5]

1.3

4[–

0.0

96

7to

2.7

8]

0.1

51

[0.0

02

67

–0.0

275

]c;

4.5

2[–

1.0

9to

10

.1]e

Bo

ots

trap

esti

mat

ed2

1.3

[17

.6–

25

.8]

0.1

71

[0.0

91

–0

.26

7]

0.0

79

7[0

.02

32

–0

.15

2]

1.2

5[0

.15

9–

6.7

4]

0.0

13

5[0

.00

58

4–

0.0

26

6]c

;4

.53

[0.5

48–

10

.8]e

BIC

Bay

esia

nin

form

atio

ncr

iter

ion

,II

Vin

ter-

ind

ivid

ual

var

iab

ilit

y,

NA

no

tap

pli

cab

le,

SE

stan

dar

der

ror,

slop

eDco

nce

ntr

atio

n–

effe

ctre

lati

on

ship

,V

PT

Tv

isce

ral

pai

nto

lera

nce

thre

sho

lda

Mea

sure

das

the

vo

lum

eo

fw

ater

inth

eb

agp

lace

do

nan

oes

oph

agea

lp

rob

eb

Th

e9

5%

CIs

of

the

par

amet

eres

tim

ates

wer

eb

ased

on

the

asy

mp

toti

cst

and

ard

erro

rfo

rth

ep

aram

eter

retu

rned

by

NO

NM

EM

cP

rop

ort

ion

aler

ror

dM

edia

np

aram

eter

esti

mat

eso

f1

,00

0b

oo

tstr

apre

pli

cate

s[2

.5–

97

.5%

ran

ge]

eA

dd

itiv

eer

ror

(th

ep

aram

eter

was

no

tsi

gn

ifica

nt

ifth

eco

nfi

den

cein

terv

alin

clu

ded

zero

);h

ow

ever

,th

isad

dit

ive

erro

rst

abil

ized

the

mod

elto

con

ver

ge

Ta

ble

5M

od

el-b

uil

din

gst

eps

for

the

CR

66

5p

har

mac

ok

inet

ic/p

har

mac

od

yn

amic

mo

del

and

chan

ges

inth

eo

bje

ctiv

efu

nct

ion

val

ue

Mo

del

Eq

uat

ion

NO

PD

OF

V

1E

=b

asel

ine

?sl

op

eD9

C,

IIV

on

bas

elin

e1

4

2A

sM

od

el1

wit

hsl

op

eDfi

xed

toze

ro1

35

.30

2

3E

=m

9ln

(C?

bas

elin

e),

IIV

on

C0

14

7.4

5

4E

=b

asel

ine

?(E

max

9C

)/(E

C50

?C

),II

Vo

nb

asel

ine

and

Em

ax

16

–8

.15

1

5C

eff0

=k

e0

9(C

-C

eff),

E=

bas

elin

e?

slo

peD

9C

eff

,II

Vo

nb

asel

ine,

ke0

and

Slo

peD

17

–6

.39

4

6C

eff0

=k

e0

9(C

-C

eff),

E=

m9

ln(C

eff

?b

asel

ine)

,II

Vo

nb

asel

ine

and

ma

17

–1

4.1

94

7C

eff0

=k

e0

9(C

-C

eff),

E=

bas

elin

e?

(Em

ax

9C

eff

)/(E

C50

?C

eff

),II

Vo

nb

asel

ine

and

ke0

17

–1

3.6

26

Res

idu

aler

ror

mo

del

su

sed

for

the

ph

arm

aco

dy

nam

icp

art

wer

ep

rop

ort

ion

erro

rm

od

els

un

less

spec

ified

oth

erw

ise

Cp

lasm

ad

rug

con

cen

trat

ion

,C

0in

itia

l(fi

ctiv

e)o

rb

ack

-ex

trap

ola

ted

pla

sma

dru

gco

nce

ntr

atio

nat

tim

eze

rofo

llo

win

gb

olu

sin

trav

eno

us

inje

ctio

n,

Ceff

ob

serv

edco

nce

ntr

atio

nin

the

effe

ct

com

par

tmen

t,C

eff0

esti

mat

edco

nce

ntr

atio

nin

the

effe

ctco

mp

artm

ent,

Eti

me

cou

rse

of

the

effe

cto

fth

eo

xy

cod

on

ed

ose

,E

C50

pla

sma

dru

gco

nce

ntr

atio

nat

wh

ich

ther

ew

asa

hal

f-re

du

ctio

nin

the

pai

nsc

ore

,E

max

pla

sma

dru

gco

nce

ntr

atio

nat

wh

ich

ther

ew

asa

max

imu

mre

du

ctio

nin

the

pai

nsc

ore

,II

Vin

ter-

ind

ivid

ual

var

iab

ilit

y,

k e0

equ

ilib

riu

mra

teco

nst

ant

bet

wee

np

lasm

aan

dth

e

effe

ctco

mp

artm

ent,

wh

ich

was

also

ap

op

ula

tio

np

aram

eter

,m

slo

pe

of

the

app

aren

tli

nea

rse

gm

ent

of

the

log

-tra

nsf

orm

edco

nce

ntr

atio

nv

ersu

sre

spo

nse

curv

e,N

OP

nu

mb

ero

fes

tim

ated

par

amet

ers

inth

em

od

elin

clu

din

gp

har

mac

ok

inet

ics

par

amet

ers,

OF

Vo

bje

ctiv

efu

nct

ion

val

ue,

slo

peD

con

cen

trat

ion

–ef

fect

rela

tio

nsh

ipa

Mix

eder

ror

mo

del

use

d

PopPK/PD Study of CR665 and Oxycodone 133

Page 10: Drug Abuse 6

only one dose level, the maximum effect of oxycodone

might not have been obtained and, thus, Model 1 was

selected as the final pharmacokinetic/pharmacodynamic

model for oxycodone. Model 5 passed the covariance step

of NONMEM�, but with an increase in OFV compared

with Model 1. The drop in OFV was significant for Model

6, but not for the later Model 7 when considering the

increased number of parameters compared with Model 1.

For Model 6 the ke0 was very small (\0.02 h) and, in

addition, neither of Models 6 or 7 passed the covariance

step of NONMEM�. An overview of the best effect model

fits for the various effect measures and the slopes of the

functions is given in Table 4.

Visual predicted check plots of effect observation and

population predictions of the model, as a summary of the

model predictions for the placebo, CR665 and oxycodone

arms of the study, are illustrated in Fig. 5. The models

seem to describe the pharmacodynamic data of placebo,

CR665 and oxycodone well as the visual predicted check

plots show that the 2.5th, 50th and 97.5th percentiles of the

observed data are closely followed by the by the simulated

2.5th, 50th and 97.5th percentiles. In addition, the areas of

95 % prediction intervals around the 2.5th, 50th and 97.5th

percentiles of the simulated data overlap with the 2.5th,

50th and 97.5th percentiles of the observed data.

3.2.4 CR665 Versus Oxycodone

Comparing the CR665 and oxycodone pharmacodynamic

models, the data fitted the same models and the baseline

values were similar, despite the small differences described

in Sects. 3.2.2 and 3.2.3. However, the slopes of the direct

linear pharmacodynamic model were found to be 0.0035

and 0.0793 VPTT mL per ng/mL for CR665 and oxyco-

done, respectively.

4 Discussion

A two-compartment and one-compartment distribution

model described the CR665 and CR665N-oxide metabolite

data, respectively, following a 1-h intravenous infusion.

For the observed oxycodone plasma concentrations, a one-

compartment model with a single transit chain representing

oral absorption provided the best fit for the data. The effect

of plasma concentration on VPTT for both CR665 and

oxycodone was best described by a direct linear model.

4.1 Methodological Considerations

More frequent pharmacodynamic evaluations and a longer

blood sampling period was not feasible in the present study

as the full study was a comprehensive pain study using a

variety of pain and safety measures [8]. This comprehen-

sive set-up in experimental pain studies is highly different

from a study in which the pharmacodynamic endpoint is,

for example, blood pressure, serum glucose, corrected QT

interval, hormone concentrations, temperature or circulat-

ing white cells. However, pharmacokinetic/pharmacody-

namic modelling can be performed sufficiently with

intervals of 30 min between pharmacodynamic evaluations

[9].

4.2 Pharmacokinetics

Together with the number of samples within the timeframe,

a two-compartmental distribution model for CR665 was

superior to a one-compartment model based on the drop in

the NONMEM� OFV of approximately 60. Distribution of

the CR665 and N-oxide model in the final pharmacokinetic

model resulted in different distributions of parent and

metabolite, which might be due to the relatively short

Table 6 Model-building steps for the oxycodone and changes in the objective function value

Model Equation NOP DOFV

1 E = baseline ? slopeD 9 C, IIV on baseline and SlopeD 15

2 As Model 1 with slopeD fixed to zero 13 21.972

3 E = m 9 ln(C ? baseline), IIV on m 15 0.459

4 E = baseline ? (Emax 9 C)/(EC50 ? C), IIV on baseline and EC50 16 –6.498

5 Ceff0 = ke0 9 (C - Ceff), E = baseline ? slopeD 9 Ceff, IIV on baseline and SlopeD 16 4.828

6 Ceff0 = ke0 9 (C - Ceff), E = m 9 ln(Ceff ? baseline), IIV on baseline and m 15 –5.279

7 Ceff0 = ke0 9 (C - Ceff), E = baseline ? (Emax 9 Ceff)/(EC50 ? Ceff), IIV on baseline, ke0, Emax and EC50 19 –6.498

Residual error models used for the pharmacodynamic part were mixed error models

C plasma drug concentration, Ceff observed concentration in the effect compartment, Ceff0 estimated concentration in the effect compartment,

E time course of the effect of the oxycodone dose, EC50 plasma drug concentration at which there was a half-reduction in the pain score, Emax

plasma drug concentration at which there was a maximum reduction in the pain score, IIV inter-individual variability, ke0 equilibrium rate

constant between plasma and the effect compartment, which was also a population parameter, m slope of the apparent linear segment of the

log-transformed concentration versus response curve, NOP number of estimated parameters in the model including pharmacokinetics parameters,

OFV objective function value, slopeD concentration–effect relationship

134 A. E. Olesen et al.

Page 11: Drug Abuse 6

blood sampling collection period of 2 h in which the

N-oxide metabolite only seems to be formed and distri-

bution/elimination of the metabolite has not been captured

(see Fig. 1). This in turn caused the slightly empirical

model for CR665 and the metabolite where k13 = k30

along with the different distribution of the two compounds.

Based on the current data, it might be difficult to conclude

whether the formation rate of the metabolite is rate limiting

for its elimination or if the N-oxide elimination is con-

trolled by its own elimination. Thus, studies including

longer sampling periods are warranted for a more extensive

pharmacokinetic description of CR665 and the N-oxide.

The resulting one-compartment distribution model with

a single chain of transit compartments for oxycodone was

also supported by previous studies with longer sampling

periods [27, 28]. The final pharmacokinetic model and

parameter estimates for oxycodone were also comparable

with previous findings from a study with a short pharmaco-

kinetic sampling period [11]. This earlier study identified a

similar one-compartment model with a single transit chain

a

Time (h)

Placebo

20

40

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

60

80

b

Time (h)

CR665

20

40

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

60

80

c

Time (h)

Placebo

20

40

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

60

80

d

Time (h)

Oxycodone

20

40

Vis

cera

l pai

n to

lera

nce

thre

shol

d (m

L)

60

80

2.00.0 1.00.5 1.5 2.00.0 1.00.5 1.5

2.00.0 1.00.5 1.5 2.00.0 1.00.5 1.5

Fig. 5 Visual predictive check of pharmacodynamics depicted as

effect on visceral pain tolerance threshold vs. time. Placebo (a),

CR665 (b), placebo (c) and oxycodone (d) with 95 % prediction

intervals. On each plot: observed data (circles), median (solid redline), 2.5th and 97.5th percentiles (dashed red lines) of the

observations. The 2.5th, 50th and 97.5th percentiles of the simulated

data are plotted as dashed black lines, solid black lines and dashedblack lines, respectively. The 95 % predicted confidence intervals

around 2.5th, 50th and 97.5th percentiles of the simulated data are

shown as coloured areas: the 95 % confidence interval for the median

(pink areas) and the 2.5th and 97.5th percentiles (blue areas) of the

simulated data

PopPK/PD Study of CR665 and Oxycodone 135

Page 12: Drug Abuse 6

representing the absorption phase, with a mean transit time

of 26 min, which is comparable with the result obtained

from the current study (0.295 h, corresponding to 18 min).

The number of transit compartments of 4.88 (range

2.6–8.3) resulting from the Stirling approximation inherent

in the model [29] seems to be in a comparable range with

that of a previous finding of 2.66 (2.2–7.2) after oral

administration of oxycodone [11].

4.3 Pharmacodynamics

The pharmacokinetic/pharmacodynamic relationship in the

present study revealed identical models for CR665 and

oxycodone with similar baseline model and parameter

estimates for the placebo treatment. A direct linear effect of

the plasma drug concentration acting on the VPTT

described by the slope of the models indicated a steeper

slope of oxycodone of 0.0793 VPTT (mL per ng/mL)

[balloon volume per plasma concentration] than that of

0.0035 (mL per ng/mL) for CR665. This observation could

indicate that oxycodone is more potent than CR665 in

the VPTT model, e.g. due to its additional effects on

l-receptors in the CNS, or simply that oxycodone and

CR665 were not given in equivalent doses relative to their

intrinsic efficacy. Such dose equivalencies can be estab-

lished readily between two compounds if they share an

efficacy biomarker response, e.g. miosis for CNS-acting

l-opioids, but this has not yet been possible for oxycodone

and a peripherally selective j-opioid such as CR665.

4.4 Peripheral Versus Central Effect?

Clinically used opioids are generally believed to exert their

main analgesic effects via l-opioid receptors in the CNS.

Consistent with this view, there is evidence for a delay in

the time course of the pharmacodynamic effect (i.e. relief

of somatic pain) with respect to the time course of the

plasma drug concentration following opioid administration

[9, 30]. This delay in the analgesic response relative to

the pharmacokinetic profile, described as ‘hysteresis’, is

usually considered a consequence of rate-limiting transport

across the blood–brain barrier, in addition to other factors.

In principle, both the efficacy and the immediacy of vis-

ceral pain relief reported here could be explained by the

involvement of peripheral j-opioid receptors on visceral

afferents as rat studies have demonstrated that j-opioids

are more efficient than l-opioids in reducing visceral pain

[31]. However, from the present study, it is not possible to

make a clear evaluation of the effect–delay component and

therefore other explanations for the current observations

cannot at present be excluded, e.g. a very rapid uptake of

oxycodone into a CNS effect compartment.

The development of a peripherally selective j-opioid

remains a powerful approach to creating selective, rationally

targeted visceral pain analgesics. Substantial evidence has

emerged for the significance of peripherally located j-opioid

receptors as a target for treating visceral pain [4]. FE200041

(an earlier analogue of CR665) and CR665 have been

developed with the idea that these peptides should be less

likely to cross the blood–brain barrier and to cause centrally

mediated adverse effects [6, 7]. Evaluation of CR665 dem-

onstrated peripheral j-opioid selectivity and antinociceptive

activity in a wide range of preclinical visceral pain models

[6, 7]. However, in the present study, some CNS effect was

seen in the form of paraesthesia, which was reported as an

adverse event in 61.1 % of the healthy volunteers receiving

CR665 [8]. It cannot be excluded that CR665 also exerted

analgesic effects in the CNS.

Oxycodone metabolites were not included in this phar-

macokinetic/pharmacodynamic analysis, as it has previ-

ously been demonstrated that the pharmacodynamic data

are driven by the plasma concentration of the parent

compound [11]. This was again supported by successful

fitting of the pharmacodynamic data to the plasma con-

centration of the parent compound.

In the future, the integration of population-based phar-

macokinetic/pharmacodynamic models can be used for a

clinical trial simulation where trial design and execution

can be explored relative to performance and outcomes [32].

5 Conclusion

Pharmacokinetic/pharmacodynamic modelling provided

results consistent with a peripheral visceral analgesic effect

of both CR665 and oxycodone, which could be mediated

by opioid receptors on visceral afferent nerves. The mod-

elling showed a direct linear plasma drug concentration–

effect relationship for the plasma drug concentration acting

on the VPTT.

Acknowledgments Stephen Hwang, PhD, and Sarita Khanna, PhD,

are acknowledged for participating in the study design.

Conflicts of interest The study was sponsored by Johnson and

Johnson (Mountain View, CA, USA). Sherron Kell is affiliated with

the sponsor and took part in study design, review and approval of the

manuscript. Gilbert Y. Wong was affiliated with the sponsor at the

time the study was conducted, and took part in study design, review

and approval of the manuscript. Sherron Kell owns stock in Johnson

& Johnson. Asbjørn Mohr Drewes has received unrestricted research

grants from Mundipharma, AstraZeneca, Lundbeck and Pfizer and

served as a Consultant/Advisory Board member for Mundipharma,

AstraZeneca and Shire. Anne Estrup Olesen has received honorarium

from Mundipharma. Camilla Staahl is currently employed by Gru-

nenthal GmBH, but was affiliated with Mech-Sense during conduct of

study and preparation of manuscript.

136 A. E. Olesen et al.

Page 13: Drug Abuse 6

References

1. Konig HH, Bernert S, Angermeyer MC. Health status of the

German population: results of a representative survey using the

EuroQol questionnaire. Gesundheitswesen. 2005;67(3):173–82.

2. Bell TJ, Panchal SJ, Miaskowski C, et al. The prevalence,

severity, and impact of opioid-induced bowel dysfunction: results

of a US and European Patient Survey (PROBE 1). Pain Med.

2009;10(1):35–42.

3. Stein C, Schafer M, Machelska H. Attacking pain at its source:

new perspectives on opioids. Nat Med. 2003;9(8):1003–8.

4. Riviere PJ. Peripheral kappa-opioid agonists for visceral pain. Br

J Pharmacol. 2004;141(8):1331–4.

5. Burton MB, Gebhart GF. Effects of kappa-opioid receptor ago-

nists on responses to colorectal distension in rats with and without

acute colonic inflammation. J Pharmacol Exp Ther. 1998;285(2):

707–15.

6. Binder W, Machelska H, Mousa S, et al. Analgesic and antiin-

flammatory effects of two novel kappa-opioid peptides. Anes-

thesiology. 2001;94(6):1034–44.

7. Vanderah TW, Largent-Milnes T, Lai J, et al. Novel D-amino

acid tetrapeptides produce potent antinociception by selectively

acting at peripheral kappa-opioid receptors. Eur J Pharmacol.

2008;583(1):62–72.

8. Arendt-Nielsen L, Olesen AE, Staahl C, et al. Analgesic efficacy

of peripheral kappa opioid receptor agonist CR665 compared to

oxycodone in a multi-modal, multi-tissue experimental human

pain model: selective effect on visceral pain. Anesthesiology.

2009;111:616–24.

9. Staahl C, Upton R, Foster DJ, et al. Pharmacokinetic-pharma-

codynamic modeling of morphine and oxycodone concentrations

and analgesic effect in a multimodal experimental pain model.

J Clin Pharmacol. 2008;48(5):619–31.

10. Andresen T, Upton RN, Foster DJ, et al. Pharmacokinetic/phar-

macodynamic relationships of transdermal buprenorphine and

fentanyl in experimental human pain models. Basic Clin Phar-

macol Toxicol. 2011;108(4):274–84.

11. Olesen AE, Upton R, Foster DJ, et al. A pharmacokinetic and

pharmacodynamic study of oral oxycodone in a human experi-

mental pain model of hyperalgesia. Clin Pharmacokinet.

2010;49(12):817–27.

12. Shafer SL, Struys MM. Mixed effect modeling in analgesia trials.

Anesth Analg. 2008;107(1):9–10.

13. Csajka C, Verotta D. Pharmacokinetic-pharmacodynamic mod-

elling: history and perspectives. J Pharmacokinet Pharmacodyn.

2006;33(3):227–79.

14. Rosenbaum SE, Carter AA, Dudley MN. Population pharmaco-

kinetics: fundamentals, methods and applications. Drug Dev Ind

Pharm. 1995;21(9):1115–41.

15. Holtman JR, Wala EP. Characterization of the antinociceptive

effect of oxycodone in male and female rats. Pharmacol Biochem

Behav. 2006;83(1):100–8.

16. Nielsen CK, Ross FB, Lotfipour S, et al. Oxycodone and mor-

phine have distinctly different pharmacological profiles:

radioligand binding and behavioural studies in two rat models of

neuropathic pain. Pain. 2007;132(3):289–300.

17. Nozaki C, Saitoh A, Kamei J. Characterization of the antinoci-

ceptive effects of oxycodone in diabetic mice. Eur J Pharmacol.

2006;535(1–3):145–51.

18. Ross FB, Smith MT. The intrinsic antinociceptive effects of

oxycodone appear to be kappa-opioid receptor mediated. Pain.

1997;73(2):151–7.

19. Drewes AM, Gregersen H. Multimodal pain stimulation of the

gastrointestinal tract. World J Gastroenterol. 2006;12(16):

2477–86.

20. Drewes AM, Schipper KP, Dimcevski G, et al. Multimodal

assessment of pain in the esophagus: a new experimental model.

Am J Physiol Gastrointest Liver Physiol. 2002;283(1):G95–103.

21. Lindbom L, Pihlgren P, Jonsson EN. PsN-Toolkit: a collection of

computer intensive statistical methods for non-linear mixed effect

modeling using NONMEM. Comput Methods Programs Biomed.

2005;79(3):241–57.

22. Lindbom L, Ribbing J, Jonsson EN. Perl-speaks-NONMEM

(PsN): a Perl module for NONMEM related programming.

Comput Methods Programs Biomed. 2004;75(2):85–94.

23. Savic RM, Jonker DM, Kerbusch T, et al. Implementation of a

transit compartment model for describing drug absorption in

pharmacokinetic studies. J Pharmacokinet Pharmacodyn. 2007;

34(5):711–26.

24. Fang L, Holford NH, Hinkle G, et al. Population pharmacoki-

netics of humanized monoclonal antibody HuCC49deltaCH2 and

murine antibody CC49 in colorectal cancer patients. J Clin

Pharmacol. 2007;47(2):227–37.

25. Yano Y, Beal SL, Sheiner LB. Evaluating pharmacokinetic/

pharmacodynamic models using the posterior predictive check.

J Pharmacokinet Pharmacodyn. 2001;28(2):171–92.

26. Holford NH. A size standard for pharmacokinetics. Clin Phar-

macokinet. 1996;30(5):329–32.

27. Lalovic B, Kharasch E, Hoffer C, et al. Pharmacokinetics and

pharmacodynamics of oral oxycodone in healthy human subjects:

role of circulating active metabolites. Clin Pharmacol Ther.

2006;79(5):461–79.

28. Mandema JW, Kaiko RF, Oshlack B, et al. Characterization and

validation of a pharmacokinetic model for controlled-release

oxycodone. Br J Clin Pharmacol. 1996;42(6):747–56.

29. Kass R, Raftery A. Bayes factors. J Am Stat Assoc. 1995;90:

773–95.

30. Lotsch J. Pharmacokinetic-pharmacodynamic modeling of opi-

oids. J Pain Symptom Manage. 2005;29(5):S90–103.

31. Ozaki N, Sengupta JN, Gebhart GF. Differential effects of mu-,

delta-, and kappa-opioid receptor agonists on mechanosensitive

gastric vagal afferent fibers in the rat. J Neurophysiol.

2000;83(4):2209–16.

32. Barrett JS, Fossler MJ, Cadieu KD, et al. Pharmacometrics: a

multidisciplinary field to facilitate critical thinking in drug

development and translational research settings. J Clin Pharma-

col. 2008;48(5):632–49.

PopPK/PD Study of CR665 and Oxycodone 137

Page 14: Drug Abuse 6

Reproduced with permission of the copyright owner. Further reproduction prohibited withoutpermission.