+ All Categories
Home > Documents > Pharmacometric Models in Anesthesia and Analgesia

Pharmacometric Models in Anesthesia and Analgesia

Date post: 29-Jan-2017
Category:
Upload: vandiep
View: 219 times
Download: 1 times
Share this document with a friend
58
ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2013 Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 173 Pharmacometric Models in Anesthesia and Analgesia MARCUS BJÖRNSSON ISSN 1651-6192 ISBN 978-91-554-8726-3 urn:nbn:se:uu:diva-205580
Transcript
Page 1: Pharmacometric Models in Anesthesia and Analgesia

ACTAUNIVERSITATIS

UPSALIENSISUPPSALA

2013

Digital Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Pharmacy 173

Pharmacometric Models inAnesthesia and Analgesia

MARCUS BJÖRNSSON

ISSN 1651-6192ISBN 978-91-554-8726-3urn:nbn:se:uu:diva-205580

Page 2: Pharmacometric Models in Anesthesia and Analgesia

Dissertation presented at Uppsala University to be publicly examined in B22, UppsalaBiomedicinska Centrum (BMC), Husargatan 3, Uppsala, Friday, October 4, 2013 at 13:15for the degree of Doctor of Philosophy (Faculty of Pharmacy). The examination will beconducted in English.

AbstractBjörnsson, M. 2013. Pharmacometric Models in Anesthesia and Analgesia. Acta UniversitatisUpsaliensis. Digital Comprehensive Summaries of Uppsala Dissertations from the Faculty ofPharmacy 173. 56 pp. Uppsala. ISBN 978-91-554-8726-3.

Modeling is a valuable tool in drug development, to support decision making, improving studydesign, and aid in regulatory approval and labeling. This thesis describes the development ofpharmacometric models for drugs used in anesthesia and analgesia.

Models describing the effects on anesthetic depth, measured by the bispectral index (BIS), fora commonly used anesthetic, propofol, and for a novel anesthetic, AZD3043, were developed.The propofol model consisted of two effect-site compartments, and could describe the effects ofpropofol when the rate of infusion is changed during treatment. AZD3043 had a high clearanceand a low volume of distribution, leading to a short half-life. The distribution to the effectsite was fast, and together with the short plasma half-life leading to a fast onset and offset ofeffects. It was also shown that BIS after AZD3043 treatment is related to the probability ofunconsciousness similar to propofol.

In analgesia studies dropout due to lack of efficacy is common. This dropout is not atrandom and needs to be taken into consideration in order to avoid bias. A model was developeddescribing the PK, pain intensity and dropout hazard for placebo, naproxen and a novel analgesiccompound, naproxcinod, after removal of a wisdom tooth. The model provides an opportunityto describe the effects of other doses or formulations. Visual predictive checks created bysimultaneous simulations of PI and dropout provided a good way of assessing the goodness offit when there is informative dropout.

The performance of non-linear mixed effects models in the presence of informative dropout,with and without including models that describe such informative dropout was investigated bysimulations and re-estimations. When a dropout model was not included there was in generalmore bias. The bias increased with decreasing number of observations per subject, increasingplacebo effect and increasing dropout rate. Bias was relatively unaffected by the number ofsubjects in the study. The bias had, in general, little effect on simulations of the underlyingefficacy score, but a dropout model would still be needed in order to make realistic simulations.

Keywords: Pharmacometrics, Anesthesia, Analgesia, Dropout, NONMEM

Marcus Björnsson, Uppsala University, Department of Pharmaceutical Biosciences, Box 591,SE-751 24 Uppsala, Sweden.

© Marcus Björnsson 2013

ISSN 1651-6192ISBN 978-91-554-8726-3urn:nbn:se:uu:diva-205580 (http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-205580)

Page 3: Pharmacometric Models in Anesthesia and Analgesia

To Life

Page 4: Pharmacometric Models in Anesthesia and Analgesia
Page 5: Pharmacometric Models in Anesthesia and Analgesia

List of Papers

This thesis is based on the following papers, which are referred to in the text by their Roman numerals.

I Björnsson MA, Norberg Å, Kalman S, Karlsson MO, Si-

monsson USH. (2010) A two-compartment effect site model describes the Bispectral Index (BIS) after different rates of propofol infusion. J Pharmacokinet Pharmacodyn, 37:243-255

II Björnsson MA, Norberg Å, Kalman S, Simonsson USH. Population model for pharmacokinetics and bispectral in-dex after intravenous infusion of the sedative and anesthet-ic AZD3043 in healthy volunteers. Submitted

III Björnsson MA, Simonsson USH. (2011) Modelling of pain intensity and informative dropout in a dental pain model after naproxcinod, naproxen and placebo admin-istration. Br J Clin Pharmacol, 71(6):899-906

IV Björnsson MA, Friberg LE, Simonsson USH. Performance of non-linear mixed effects models in the presence of in-formative dropout. Submitted

Reprints were made with permission from the respective publishers.

Page 6: Pharmacometric Models in Anesthesia and Analgesia
Page 7: Pharmacometric Models in Anesthesia and Analgesia

Contents

Introduction ................................................................................................... 11 Pharmacometrics ...................................................................................... 11

Pharmacokinetics ................................................................................. 12 Pharmacodynamics .............................................................................. 12 Non-linear mixed effects models ......................................................... 12

Anesthesia ................................................................................................ 14 BIS ....................................................................................................... 15

Analgesia .................................................................................................. 15 Dental pain model ................................................................................ 16

Dropout .................................................................................................... 16

Aims .............................................................................................................. 19

Material and Methods ................................................................................... 20 Study drugs ............................................................................................... 20 Data .......................................................................................................... 20

Propofol study ...................................................................................... 20 AZD3043 studies ................................................................................. 21 Naproxcinod, naproxen and placebo study .......................................... 21 Simulation study .................................................................................. 21

Software ................................................................................................... 23 Model development .................................................................................. 23

Propofol ............................................................................................... 24 AZD3043 ............................................................................................. 24 Naproxcinod and naproxen .................................................................. 25

Model selection ........................................................................................ 25 Simulations and re-estimations ................................................................ 26

Results ........................................................................................................... 27 Anesthesia ................................................................................................ 27

Propofol PKPD model ......................................................................... 27 AZD3043 PKPD model ....................................................................... 31

Analgesia .................................................................................................. 35 Naproxcinod, naproxen and placebo PKPD model ............................. 35 Simulations and estimations of dropout............................................... 38

Discussion ..................................................................................................... 42

Page 8: Pharmacometric Models in Anesthesia and Analgesia

Anesthesia ................................................................................................ 42 Pharmacokinetics ................................................................................. 42 Pharmacodynamics .............................................................................. 43

Analgesia .................................................................................................. 44 Pharmacokinetics ................................................................................. 44 Pharmacodynamics .............................................................................. 45 Dropout ................................................................................................ 45

Conclusions ................................................................................................... 47

Populärvetenskaplig sammanfattning ........................................................... 48

Acknowledgements ....................................................................................... 49

References ..................................................................................................... 51

Page 9: Pharmacometric Models in Anesthesia and Analgesia

Abbreviations

BIS Bispectral index Bmax Maximum binding capacity Ce Drug concentration at effect site CL Clearance EC50 Concentration giving 50% of maxi-

mum effect EEG Electroencefalogram Emax Maximum effect FDA Food and drug administration FO First Order method FOCE First Order Conditional Estimation

method FOCE-I First Order Conditional Estimation

method with Interaction Frel Relative bioavailability h(t) Hazard at time t he Hazard dependent on effect score h0 Baseline hazard IIV Inter-individual variability iv Intravenous ke0 Rate constant for equilibration be-

tween plasma and effect site. ke12 Rate constant for distribution from

central to peripheral effect site ke21 Rate constant for distribution from

peripheral to central effect site Km Concentration at half of maximum

protein binding kpl Rate constant for onset of placebo

effect MBDD Model-based drug development MOAA/S Modified Observer’s Assessment of

Alertness/Sedation MTT Mean transit time NN Number of transit compartments OFV Objective Function Value

Page 10: Pharmacometric Models in Anesthesia and Analgesia

PC-VPC PRED-corrected visual predictive check

PD Pharmacodynamics PI Pain intensity PK Pharmacokinetics PKPD Pharmacokinetic/pharmacodynamic PLmax Maximum placebo effect PRED Population prediction PsN Perl speaks NONMEM Q Distribution clearance RMSE Root mean square error RSE% Relative standard error in percent SSE Stochastic Simulation and Estimation S(t) Probability of remaining in study at

time t TCI Target controlled infusions VAS Visual analogue scale VPC Visual Predictive Check V Volume of distribution Vss Volume of distribution at steady state ε Difference between observation and

individual prediction ω2 Variance of η σ2 Variance of ε θ Typical value of a parameter η Difference between individual value

and typical value of a parameter

Page 11: Pharmacometric Models in Anesthesia and Analgesia

11

Introduction

Drug development is facing challenges with fewer drugs to the market and escalating costs [1,2]. According to an analysis by Forbes, the average cost for developing a new drug is 4 billion US$ (10 February 2012, www.forbes.com). These issues were highlighted by the US Food and Drug Administration (FDA) in a white paper in 2004, where they identified mod-el-based drug development (MBDD) as an important way of improving drug development [3]. MBDD was defined as pharmaco-statistical models of drug efficacy and safety from preclinical and clinical data to improve drug devel-opment knowledge management and decision-making. The concept has been adopted and developed by pharmaceutical industry [1,2,4-6] and is also ap-preciated by other regulatory agencies [7].

Pharmacometrics, which is an important part of MBDD, has become broadly used within drug development, and were shown in a survey among ten large or mid-sized pharmaceutical companies to positively affect internal decision making [8]. It has also been increasingly used, and has had an in-creased importance, in New Drug Applications to the regulatory authorities to support drug approval, labeling, and trial design decisions [9,10].

Pharmacometrics Pharmacometrics has been defined as “the science of developing and apply-ing mathematical and statistical methods to characterize, understand, and predict a drug’s pharmacokinetic, pharmacodynamic, and biomarker-outcome behavior” [11]. Important tasks in pharmacometrics are to find the optimal dose or dosing regimen, improve decision making, and decrease time and cost of drug development.

Pharmacometric models are useful tools to aid drug development, thera-peutic decisions and regulatory decisions [7,9]. The models can provide a quantitative basis for informed decisions, and they also have the potential for increasing power in clinical studies [12,13]. Pharmacometrics has evolved from pharmacokinetics (PK), to pharmacodynamics (PD), and also involves other aspects of drug treatment, such as disease progression, dropout and placebo response.

Page 12: Pharmacometric Models in Anesthesia and Analgesia

12

Pharmacokinetics PK describes the processes and rates of absorption of a drug, distribution into tissues and the elimination by metabolism or excretion [14]. The phar-macokinetic properties of a drug can usually be investigated by collecting repeated plasma concentrations of the drug after administration. A model including parameters describing the absorption, distribution and elimination processes is then fitted to the data, and conclusions about the PK can be made.

Pharmacodynamics While PK has been described as “what the body does to the drug”, PD can be described as “what the drug does to the body” [14]. PD quantifies the relationship between drug concentrations at the site of action and the desired or undesired effects. When PK is linked to PD, the drug effect over time after drug administration can be described. As PD involves the concentra-tions at the effect site, and PK typically describes concentrations in plasma, there might be a need to find a link between these concentrations. The con-centrations at the effect site can often be difficult to measure, and therefore a model describing a hypothetical effect site can be used. The effect compart-ment model assumes that there is a distribution of drug between plasma and the effect compartment, leading to a delay in effects in relation to the plasma concentrations. This delay is quantified by the rate constant ke0. A large ke0 means that equilibrium between plasma and effect site is fast, and a small ke0 implies that equilibrium is slow. There can also be other reasons for a de-layed effect in relation to plasma concentrations, e.g. indirect effects, when it takes time for the measurable effects to appear after the drug has interacted with the target [15].

In order to describe the drug effects over time it is sometimes of im-portance to describe the progression of the disease, and the effects of place-bo. Sometimes it is difficult to separate the disease progression and the pla-cebo effects, especially when there are existing treatments, and it is unethical to study the disease without treating the patient.

Non-linear mixed effects models Non-linear mixed effects modeling is an important tool in pharmacometrics. It is suitable for biological systems, as biological data are in general best described by non-linear functions, and there is usually a natural variability in biological systems, which can be described by mixed effects models.

Non-linear mixed effects models consist of fixed effects and random ef-fects. The fixed effects describe the structure, or the trend of the data, in the typical individual. The fixed effects parameters are usually denoted θ. The

Page 13: Pharmacometric Models in Anesthesia and Analgesia

13

random effects describe different levels of variability in data, typically vari-ability between subjects as well as residual unexplained variability, for ex-ample measurement error. Other levels of variability can also be estimated, for example variability between different occasions within a subject [16].

The difference between an individual value for a parameter and the popu-lation mean of that parameter is usually denoted η. The variability in the parameter can have any distribution but in parametric models they are typi-cally normally distributed or any transformation of a normal distribution. For a parameter with a normal distribution, the value of parameter P in individu-al i, Pi, can be described as where is normally distributed with mean zero. The variance of , , is estimated in the non-linear mixed effects model. As many physiological parameters tend to be log-normally distributed the parameter can be de-scribed as exp

The residual unexplained variability, usually denoted ε, describes the differ-ence between an observation and the individual model predicted value at that particular time-point. The jth observation in individual i, yij, can be described as ,

where , is the individual prediction described by a function of all parameters and the independent variables xij, which typically are time and dose. ε is normally distributed with mean zero, and the variance , which is estimated in the model. Often measurement errors are proportional to the value of the observed variable, for example a plasma concentration. The residual unexplained variability can be parameterized in different ways, so that for example the magnitude of the residual variability is proportional to the prediction. Combinations of proportional and additive residual errors are also possible [17].

Covariates, such as demographics, genetics, or organ function can be add-ed to the model in order to explain the reasons for variability. For example, a drug that is eliminated through the kidneys is likely to be eliminated slower in patients with poor renal function than in patients with normal renal func-tion. The magnitude of the covariate effects on the model parameters can be estimated in the non-linear mixed effects model. The covariate relationships can be used to individualize the dosing regimen, so that a patient that is like-ly to have a fast elimination, based on its set of covariates, can receive a

Page 14: Pharmacometric Models in Anesthesia and Analgesia

14

higher dose or a shorter interval between doses in order to reach the thera-peutic concentrations of the drug.

As all data from all individuals are analyzed simultaneously, both sparse and rich data, as well as data from unbalanced study designs, can be used [18], which is an advantage when sampling is difficult, e.g. in small children, or not possible to do at the same times in all subjects.

The parameter estimates of a model are found by iteratively searching for the most likely parameter estimates, given the data. This is done by minimiz-ing the objective function (OFV), which represents minus twice the log-likelihood of the data. For hierarchical models, where the more complex model can be collapsed into the simpler one, the difference in OFV is ap-proximately chi-square distributed, which means that a difference of 3.84 in OFV corresponds to a p-value of 0.05 when there is one more parameter in the more complex model. There are several different estimation methods that can be used to minimize the OFV. The first-order (FO) and first-order condi-tional (FOCE) methods use a first-order Taylor-series expansion at η=0 or the conditional value of η [19,20]. The FOCE method can also be applied with interaction between η and ε (FOCE-I). The Laplacian method uses a second-order Taylor-series expansion [20,21], and needs to be used for mod-eling non-continuous data, such as categorical or time-to-event data.

Anesthesia Anesthesia is the blocking of sensations, including pain. In local or regional anesthesia a specific part of the body, e.g. the tooth while at the dentist, is deprived of its sensations, while in general anesthesia all sensations in the whole body are blocked and the patient becomes unconscious. General anes-thesia can be obtained using inhaled anesthetic gasses or by intravenous (iv) administration of anesthetic agents.

In general anesthesia practice, it is desirable to reach anesthesia quickly, to easily adjust the dose to keep the patient at an appropriate depth of anes-thesia, and for the patient to quickly become conscious and recover from the anesthesia after the procedure [22]. In contrast to many other therapeutic areas, where patients are chronically dosed and it may take days or weeks to reach appropriate effects, anesthesia deals with very fast onset and offset of effect, within minutes, and short durations where concentrations and effects are not at steady state.

PK and PD models are widely used in iv anesthesia in order to optimize dosing. Dosing regimens are often based on covariates such as body weight, age, and disease status. To achieve a rapid onset and smooth anesthesia vary-ing infusion rates are often administered, e.g. starting with a short high-rate infusion followed by a slower maintenance infusion. The infusion rate is then further adjusted and titrated to the desired effects. This can also be done

Page 15: Pharmacometric Models in Anesthesia and Analgesia

15

by using target controlled infusions (TCI). In TCI systems a PKPD model, including covariate effects, is driving the infusion pump, and the infusion rate is automatically adjusted in order to achieve and maintain the target effect site concentrations of the drug. If proper effects are not achieved a new target effect site concentration can be entered and the infusion rates are automatically altered to rapidly reach the new target concentration. Com-mercial TCI systems are available in most parts of the world [23], and algo-rithms are available for several drugs, e.g. propofol, fentanyl, remifentanil, alfentanil, sufentanil, midazolam and ketamine [24-30].

In order to give dosing recommendations, not only on the starting dose but also on how to titrate to the desired effects, it is important to be able to describe the effects after different infusion rates so that the PKPD model is as accurate as possible.

To describe the delay between plasma concentration and effect of iv anes-thetics an effect-compartment model is commonly used. For propofol, dif-ferent estimates of ke0 have been reported, depending on the rate and dura-tion of administration. A higher ke0 has been observed for bolus doses and high rate, short infusions compared to longer, slow rate infusions [31]. Therefore, a single effect compartment model is not suitable for modeling or simulations when the rate of infusion changes during the treatment.

BIS Bispectral index (BIS) is a dynamic measure of anesthetic depth derived from the electroencephalogram (EEG) [32]. The EEG signals are trans-formed into a scale ranging from 0 (isoelectric EEG) to 100 (fully awake). At BIS below 60 the probability of consciousness is very low. BIS is de-pendent on the anesthetic agent, and has been shown to reflect the depth of anesthesia well for some anesthetics, e.g. propofol, midazolam and isoflurane [33], but cannot accurately capture the depth of anesthesia for others, e.g. ketamine and nitrous oxide [34,35]. BIS is used in anesthesia practice to monitor the anesthetic depth in real time, but can also be used to document the effects of new anesthetics in drug development, and be used in pharmacokinetic-pharmacodynamic models to optimize dosing regimens.

Analgesia While anesthesia refers to the blocking of all sensations, analgesia is the blocking of the sensation of pain. Pain is a condition that affects most peo-ple, up to 70% of the population in western countries use analgesics regular-ly [36]. Pain is also the most common reason for consulting a physician [36]. There are several different types of pain, e.g. nociceptive pain, in which pe-ripheral specialized sensory nerves, nociceptors, are activated, and neuro-

Page 16: Pharmacometric Models in Anesthesia and Analgesia

16

pathic pain, in which the nerve fibers are damaged or dysfunctional, sending signals although there is no stimulus that normally should be painful. There are many existing treatments, especially for nociceptive pain, but due to variability in the efficacy between patients, and adverse effect, there is still an unmet medical need for better analgesics.

As pain is a personal and subjective experience [37], the measurement of pain need to be done by the patients themselves. One of the more sensitive methods to measure pain is the Visual Analogue Scale (VAS) [38]. On the VAS, the patients rate their pain by marking a line between “No pain” and “Worst pain imaginable”, or similar wordings. The investigator could then measure how many millimeters from the “No pain” side the patient put their mark.

Dental pain model Surgical removal of an impacted wisdom tooth is a painful procedure. The pain is well described and characterized, short-lasting, and responds well to nociceptive pain treatment, such as non-steroidal anti-inflammatory drugs and opioids [39]. This makes it a good model for studying novel drugs tar-geted for the treatment of nociceptive pain.

The impacted wisdom tooth is removed under local anesthesia, using standard surgical procedures. Patients that experience pain when the effect of the local anesthetic vanishes are randomized to study drug, comparator or placebo. The effect on the pain intensity could then be studied over time using a pain scale, such as the VAS.

For ethical reasons, patients that require additional pain relief during the study are given an active analgesic. Once this rescue medication is given no further measurements of pain intensity are performed, as these measure-ments would not only reflect the effects of study drug, but also the effects of the rescue medication. In order to allow for the study drug to be absorbed and start giving an effect, the patients are sometimes asked to refrain from rescue medication during the first hour or two, but they are still allowed to take rescue medication if they cannot wait.

Dropout In longitudinal studies data are collected repeatedly over time in each sub-ject. However, some patients might not be able to participate for the whole planned study duration. The patients are then dropping out of the study, and no further assessments of the study variables are being performed in that patient. Dropout can be due to various reasons, including adverse events or lack of efficacy of the study drug, but also due to other reasons not related to

Page 17: Pharmacometric Models in Anesthesia and Analgesia

17

the disease or drug under investigation [40]. When dropout occurs in a study the interpretation of the results can become more difficult.

Dropout has been classified as missing completely at random, when the dropout does not depend on observed or unobserved values of the dependent variable, missing at random when the dropout is dependent on the observed, but not the unobserved, value of the dependent variable, and missing not at random when the dropout is dependent on the unobserved value of the de-pendent variable [41-44]. Dropout can also be classified as ignorable, when the interpretation of the results is valid even if the dropout is ignored [42]. This is the case when the dropout is completely at random, and not depend-ent on the studied disease, treatment or procedures. When the interpretation of the results is affected by the dropout it is not ignorable, and needs to be taken into account to make valid inferences of the drug effects [42]. Dropout missing not at random is non-ignorable and can also be referred to as in-formative dropout [45]. This could be the case for example when dropout is dependent on the effects of the study drug. The fact that a patient has dropped out of the study could provide information about the treatment ef-fect in that patient. Such informative dropout could be important to handle in non-linear mixed effects modeling in order to avoid bias in the parameter estimates [46-48]. In the dental pain model, the patients with the most pain tend to request rescue medication, i.e. the probability of dropping out is de-pendent on the pain intensity. At the same time the observed pain intensity is dependent on the dropout. At the end of the study period the differences in pain intensity is small between placebo and active treatment, as the patients that remain in the study are those who are no longer in pain, regardless of treatment.

When analyzing dropout the time of dropout is recorded for the subjects that drop out during the study. Other subjects may still be remaining in the study at the time of terminating the study procedures. This is called censor-ing. When it is known that a subject still has not dropped out at a certain time, but it is unknown what happens after that time it is called right censor-ing. Censored observations can be modeled using a survival function, in which the probability of still remaining in the study (not having dropped out) at time t, S(t), can be described as

where h(t) is a function describing the hazard over time. For subjects that drop out, the probability density of dropping out at time t, f(t) is modeled as

Page 18: Pharmacometric Models in Anesthesia and Analgesia

18

If the exact time of dropout is not known, but rather that a subject has dropped out any time between two visits, it is called interval censoring, and the probability of dropping out during that interval could be modeled as the difference between the probability of remaining in the study at the beginning of the interval and the probability of remaining in the study at the end of the interval.

Page 19: Pharmacometric Models in Anesthesia and Analgesia

19

Aims

The aim of this thesis was to improve the understanding of the time course of drug effect in anesthesia and analgesia by using non-linear mixed effects modeling. The specific aims were to:

• Investigate the influence of target site distribution on the onset and

offset of propofol anesthesia • Describe the PK and PD for a novel investigational anesthetic com-

pound, AZD3043 • Assess the validity of BIS as a marker for anesthesia for AZD3043

• Describe the analgesic effects of naproxcinod and naproxen in a

study with a large proportion of informative dropout • Investigate the need of handling informative dropout in non-linear

mixed effects modeling, including factors affecting bias and impre-cision in parameter estimates

Page 20: Pharmacometric Models in Anesthesia and Analgesia

20

Material and Methods

Study drugs This thesis describes the PK and PD of propofol (Paper I), AZD3043 (Paper II), naproxcinod and naproxen (Paper III). Propofol is a widely used anes-thetic, while AZD3043 is a novel compound under development for induc-tion and maintenance of anesthesia [49]. Similarly, naproxen is a widely used non-steroidal anti-inflammatory analgesic, while naproxcinod [50-52] is under the development for analgesia. Naproxcinod is rapidly metabolized to naproxen in the intestines and in blood [53]. No real drugs were described in the simulation study in Paper IV.

Data The data used in Papers I-III were obtained from clinical studies. All studies were performed in accordance with the Declaration of Helsinki and Good Clinical Practice. The studies were approved by an Independent Ethics Committee or Research Review Committee, and written informed consent was obtained from all subjects. In Paper IV only simulated data were used.

Propofol study Data on arterial plasma propofol concentrations and BIS were obtained in a randomized cross-over study in 21 healthy male and female volunteers. The subjects received a 1-minute iv infusion of 2 mg/kg of propofol at one occa-sion, and a 1-minute iv infusion of 2 mg/kg of propofol immediately fol-lowed by a 29-minute iv infusion of 12 mg/kg/hour of propofol at another occasion. Arterial plasma concentrations of propofol were measured repeat-edly up to 4 hours after the infusion. BIS was measured until the subjects were regarded as awake after the anesthesia. As arterial concentrations are more closely related to the concentrations at the effect site, arterial rather than venous concentrations were studied [54-55].

Page 21: Pharmacometric Models in Anesthesia and Analgesia

21

AZD3043 studies Data on arterial AZD3043 plasma concentrations and BIS were obtained in two parallel group dose escalation studies. In the first study AZD3043 was administered iv in 9 panels of 6 healthy male volunteers (53 in total due to one missing subject in one of the panels). Each volunteer received one 30-minute infusion of AZD3043, with the infusion rate ranging from 1 mg/kg/h in the first panel to 81 mg/kg/h in the last panel. The second study was di-vided into two parts. In part A, a total of 40 healthy male or female volun-teers (5 panels of 8 subjects) received a 1-minute infusion of AZD3043, with the dose ranging from 1 to 6 mg/kg (60 to 360 mg/kg/h). In part B a total of 32 healthy male or female volunteers (4 panels of 8 subjects) received a 1-minute infusion (dose ranging from 0.8 to 4 mg/kg, or 48 to 240 mg/kg/h) immediately followed by a 30-minute infusion of AZD3043 (10 to 40 mg/kg/h). Arterial plasma concentrations of AZD3043 were measured re-peatedly up to 2 h after the infusion. BIS was recorded until the subjects were regarded as awake according to clinical signs. Consciousness was as-sessed using the Modified Observer’s Assessment of Alertness/Sedation score (MOAA/S) [56], where a MOAA/S of 2 or less was classified as un-conscious.

Naproxcinod, naproxen and placebo study Data on naproxen plasma concentrations, pain intensity and time of dropout were obtained in a randomized, double-blind, parallel-group study in 242 patients undergoing surgical removal of a wisdom tooth [52]. Patients who experienced pain after the effects of local anesthetics had vanished were randomized to one of four different doses of naproxcinod (375, 750, 1500 or 2250 mg), naproxen (500 mg) or placebo. Total and unbound plasma con-centrations of naproxen were obtained repeatedly in a subset of patients (n=90) up to 8 hours after administration of study drug, and pain intensity was measured in all patients on a visual analogue scale for 8 hours, or until request of rescue medication. Patients were asked not to take rescue medica-tion during the first 1.5 h after administration of study drug, but they were still allowed to take it if they could not wait. The time of request of rescue medication was recorded, and was used for the dropout analysis.

Simulation study Paper IV is based on simulations from a model adapted from the one pre-sented in Paper III. The model simultaneously simulated an efficacy varia-ble, and dropout that was dependent on the efficacy variable. A one-compartment PK model with a constant-rate infusion was used to simulate individual concentration-time profiles, which were used as an input to the

Page 22: Pharmacometric Models in Anesthesia and Analgesia

22

PD-model. The PD-model consisted of an exponential placebo effect model, and an inhibitory Emax-model describing the concentration-effect relation-ship. The dropout hazard was related to the efficacy variable, where the haz-ard increased exponentially with increasing efficacy score. Parameter values used in the simulations are shown in Table 1, and the plasma concentrations, effect variable and probability of dropout for a typical individual for the Base Model are shown in Figure 1.

Table 1. Parameter values used in the simulation of the Base Scenario (Scenario 1).

Parameter Typical value

Inter individual variability (%)a

Explanation

BL 50 30 Efficacy variable at baselinePLmax (%) 20 120 Maximum placebo effectkpl (h

-1) 0.25 44 Rate constant for onset of placebo effect EC50 (units/L) 20 122 Concentration leading to 50 % of maximum

efficacyCL (L/h) 10 30 ClearanceV (L) 10 30 Volume of distributionh0 (h

-1) 0.01 - Baseline hazardhe 0.05 - Parameter for relationship between hazard

and effect variableσ (effect units) 7.5 - Additive residual variabilitya Inter individual variability expressed as coefficient of variation.

Figure 1. Drug concentrations, effect variable and probability of remaining in the study for a typical individual based on the true model parameters from the Base Model. Solid line – placebo, dashed line – low dose, circles – medium dose, dotted line – high dose.

Different scenarios with varying system-specific properties (dropout rate, magnitude of placebo effect), and design specific properties (number of pa-tients, number of observations), were simulated (Table 2). In the simulations the exact time of dropout was recorded and used in the estimations. For the scenarios where number of observations was varied, simulations were also performed without recording the exact time of dropout, but rather recording

Time (h)

Dru

g c

on

cen

tra

tion

0

5

10

15

20

25

0 2 4 6 8

Time (h)

Effe

ct

0

10

20

30

40

50

60

0 2 4 6 8

Time (h)

% r

em

ain

ing

in s

tud

y

0

20

40

60

80

100

0 2 4 6 8

Page 23: Pharmacometric Models in Anesthesia and Analgesia

23

that a subject had dropped out any time between two observations of the efficacy variable (interval censoring).

The simulations, one thousand simulations per scenario, were performed using the stochastic simulation and estimation (SSE) functionality in the software PsN.

Table 2. Parameters and design variables varied in the different simulation scenarios. For each scenario, the grey cells gives the value of the variable that was changed compared to the Base Scenario (Scenario 1).

Scenario N per group

PLmaxa

(%)

Observation interval (h)

Observations per patient

heb Dropoutc (%)

1 (Base) 45 20 1 9 0.05 22-52 2 15 20 1 9 0.05 22-52 3 25 20 1 9 0.05 22-52 4 65 20 1 9 0.05 22-52 5 45 10 1 9 0.05 23-57 6 45 40 1 9 0.05 20-43 7 45 80 1 9 0.05 16-30 8 45 20 0.5 17 0.05 22-52 9 45 20 2 5 0.05 22-52 10 45 20 4 3 0.05 22-52 11 45 20 1 9 0 0 12 45 20 1 9 0.025 13-22 13 45 20 1 9 0.06 27-68 14 45 20 1 9 0.07 34-83

a Maximum placebo effect b Parameter relating the hazard to the effect variable. A high value represents a larger prob-ability of dropout. c The range of probability of dropout for a typical individual between treatment groups, where the lower number represents the highest dose and the highest number represents placebo.

Software All non-linear mixed effects analyses performed in this thesis were per-formed using NONMEM (versions VI and 7) [20.]. The PsN toolkit [57] was used together with NONMEM for automation of estimations and simula-tions. The Xpose [58] package in R (http://www.r-project.org) was used for goodness of fit assessments and production of graphs.

Model development For each study the model building was done sequentially, starting with a model for the PK. After finalizing the PK model, the PD model was devel-oped. In Paper I the individual empirical Bayes estimates of the parameters were used as an input to the PD-model, while in Papers II and III PK and PD data were analyzed simultaneously, with the PK parameter estimates fixed

Page 24: Pharmacometric Models in Anesthesia and Analgesia

24

based on the final PK model [59]. In Paper III a dropout model was then added, and the PD and dropout models were refined simultaneously.

Propofol The pharmacokinetics of propofol has been described in various ways in numerous papers [60-64], and the starting point for the PK modeling was a commonly used three-compartment model. In order to get an as good as possible input to the PD model, further components were added, such as a third disposition compartment, a lag-time to describe the time for the drug to transfer from the site of administration to the site of sampling, and time-dependency in the clearance parameters. Log-normally distributed inter indi-vidual variability was added to the parameters were it was supported by the data. Additive, proportional and combined additive and proportional residual unexplained variability models were explored, as well as a different residual error during the first minutes after start of infusion as the variability was larger during that time. Body weight was investigated as a covariate on clearance and volumes of distribution.

Individual empirical Bayes estimates of the PK parameters were used as an input to the PD-model. As there are several publications modeling the effects of propofol on BIS as an effect compartment model combined with a sigmoid Emax model [31,62,64-68] this was used as the starting point for the modeling. In order to describe the different onset and offset rates after dif-ferent infusion rates, a second effect-site compartment was added, allowing for distribution within the brain. The decrease in BIS was linked to the cen-tral effect site compartment concentrations through a sigmoid Emax model. ke0 as well as the rate constants to and from the peripheral effect site com-partment were estimated.

AZD3043 To describe the arterial PK of AZD3043 two- and three-compartment mod-els were applied. A lag-time was used to describe the time from administra-tion to the time of appearance of drug at the sampling site. Log-normally distributed inter individual variability was added to the parameters were it was supported by the data. Additive, proportional and combined additive and proportional residual unexplained variability models were explored. Body weight was included as a covariate on clearance and volume parameters in an allometric fashion [69]. Dose-dependency in the parameters was investi-gated, as well as the effects of sex, age and esterase activity on CL.

An effect-compartment model with a sigmoid Emax model was used as the starting point for the PD-model. A two-compartment effect-site model was also investigated.

Page 25: Pharmacometric Models in Anesthesia and Analgesia

25

To assess the usefulness of BIS as a measure of depth of anesthesia after AZD3043 administration logistic regression was performed. The logit of the probability of unconsciousness was modeled as a slope-intercept model. Inter-individual variability was investigated in both slope and intercept. The values of BIS where there was a 50 % and a 95 % probability of uncon-sciousness were derived by simulating from the model.

Naproxcinod and naproxen Naproxcinod is rapidly metabolized to naproxen in the intestines and only a small fraction of the dose is present unchanged in plasma [53]. Naproxen is also believed to be responsible for the effects after naproxcinod administra-tion, and therefore only naproxen concentrations were modeled. As naproxen is highly bound to albumin [70] and this binding is saturable at high concen-trations, both total and unbound concentrations of naproxen were modeled. One- and two-compartment disposition models were evaluated for the PK, as well as several different absorption models, including first-order, zero-order, sequential zero- and first order absorption, with and without lag-time, and a transit compartment model [71]. Unbound oral clearance and volume of distribution of naproxen were assumed to be the same regardless of treat-ment, while the absorption properties were different for naproxcinod and naproxen. A binding model was used to describe the relationship between total and unbound naproxen concentrations. Log-normally distributed inter individual variability was added to the parameters were it was supported by the data, and additive, proportional and combined additive and proportional residual unexplained variability models were explored. The predicted un-bound concentrations were used as an input to the PD model.

PK and PD data were then analyzed simultaneously while the PK parame-ters were fixed in the subsequent PD analysis. When analyzing the pain in-tensity, a preliminary model for the PI after placebo administration was first established. Exponential, inverse Bateman and Weibull functions were ex-plored for the placebo model. Thereafter the effects of naproxen were added to the model, as an inhibitory sigmoid Emax model, as well as a model for dropout. Exponential, Weibull and Gompertz models were explored for the dropout hazard, and linear and exponential models for the influence of pain intensity and baseline pain intensity on the hazard were investigated. All parts of the PD and dropout models were then refined simultaneously.

Model selection The selection of a more complex model compared with a simpler model was based on goodness of fit plots, precision in the parameter estimates, scien-tific plausibility and statistically using the OFV.

Page 26: Pharmacometric Models in Anesthesia and Analgesia

26

Visual predictive checks (VPC) were used to assess the simulation prop-erties of the developed models. A VPC is produced by simulating a large number of studies using the model. The means and different percentiles of the simulated data at each time point are then compared with the correspond-ing means and percentiles of the observed data. In cases where dosing is individualized, or where there is a large range of doses with few individuals per dose, PRED-corrected VPC (PC-VPC) were used [72]. In a PC-VPC the data are normalized with the population prediction for each individual before calculating the means and percentiles. All VPC were performed using PsN [57] and Xpose [58].

Simulations and re-estimations In paper IV, the simulated data from each scenario was analyzed using two different models; 1) the same model as used for the simulations, e.g. includ-ing a model describing the dropout, 2) the same model as used for the simu-lations, but ignoring the part of the model describing the dropout. In the cas-es where the dropout was ignored, both the FOCE-I and Laplace estimation methods were used. In addition, for the scenarios where the exact time of dropout was not simulated, interval censoring was used in the estimation of the dropout model.

From the results of the estimations, bias and imprecision in the parameter estimates were calculated. Bias was calculated as 100%

where Est and True are the estimated and true values of the parameter. Im-precision is described as the root mean square error, calculated as 100%

Bias and imprecision were then compared for the different simulated scenar-ios and estimation models.

To illustrate the need of modeling informative dropout in order to make realistic simulations, VPCs were created. An arbitrarily simulated dataset, simulated from the Base Scenario, served as the “observed” data. In addi-tion, a dataset simulated with 40% dropout completely at random was simu-lated to serve as “observations”. Simulations were then performed with or without including a dropout model, and were compared with the “observa-tions”.

Page 27: Pharmacometric Models in Anesthesia and Analgesia

27

Results

Anesthesia

Propofol PKPD model When the commonly used 3-compartment PK model was fitted to the arterial concentrations of propofol, it was shown that it could not adequately be fit-ted to the two dosing regimens simultaneously. Even if analyzed by itself, the dosing regimen that started with a high-rate 1-minute infusion and was followed by a slower rate 29-minute infusion could not be adequately de-scribed with the 3-compartment model. In order to get an as good as possible input to the PD-model, a third disposition compartment, a lag-time between the site of administration and the site of sampling, and a time-dependency in CL, were added to the model (Figure 2).

As with the PK, the BIS could not be accurately described for the two dif-ferent dosing regimens when analyzed together, nor could the primed con-stant infusion by itself, with the previously used model, the effect-site mod-el. This was true both when using the 3-compartment model and the more complicated newly developed PK-model as an input to the PD-model. After the 1-minute infusion a higher ke0 was estimated than for the other regimen, even though both regimens were studied in the same individuals.

Using a two-compartment effect-site model (Figure 2) the fit of the BIS data significantly improved, allowing for different onset and offset rates for the different treatment regimens. This was true both when the standard 3-compartment model and the new PK model were used as an input to the PD-model (Figure 3). The rate constant for distribution within the effect site, ke12 and ke21 were 0.114 min-1 and 0.0214 min-1, implying that the volume of the peripheral part of the effect site was approximately 5 times larger than the central part of the effect compartment. PK and PD parameter estimates are shown in Tables 3 and 4.

Page 28: Pharmacometric Models in Anesthesia and Analgesia

28

Figure 2. Final PK/PD-model for the relation of arterial propofol concentrations and BIS. CL is the clearance from V1, the central compartment, V2, V3 and V4 are volumes of the peripheral compartments, Q2, Q3 and Q4 are distribution clearances between central and peripheral compartments, tlag is the lag-time for dose into cen-tral compartment, Imax is the maximum fractional decrease in elimination and distri-bution clearances, t50 is the time-point for 50% of the maximum fractional decrease in elimination and distribution clearances, ke0 is the rate constant for distribution from effect compartment, Emax is the maximum effect, EC50 is the effect site concen-tration (Ce) needed to reach 50% of Emax, γ is the shape factor, ke12 and ke21 are rate constants for distribution between central and peripheral effect compartments.

V1 (CentralComp.)

EffectSite

ke0 ke0

Periph.EffectComp.

ke21ke12

PK PD

V2 (Periph.Comp.)

V3(Periph.Comp.)

V4(Periph.Comp.)

CL

Q4

Q3

Q2

+⋅−⋅= γγ

γ

e

e

CEC

CEBaselineBIS

50

max1

tlag

DOSE

+⋅−⋅

−=

tt

tI

I

PP ss

t50

max

max

11

V1 (CentralComp.)

EffectSite

ke0 ke0

Periph.EffectComp.

ke21ke12

PK PD

V2 (Periph.Comp.)

V3(Periph.Comp.)

V4(Periph.Comp.)

CL

Q4

Q3

Q2

+⋅−⋅= γγ

γ

e

e

CEC

CEBaselineBIS

50

max1

tlag

DOSE

+⋅−⋅

−=

tt

tI

I

PP ss

t50

max

max

11

Page 29: Pharmacometric Models in Anesthesia and Analgesia

29

Figure 3. Population prediction corrected visual predictive check (PC-VPC) for the standard effect compartment model (left) and the final two-compartment effect site model (right) after the primed constant infusion (top panel) and bolus administration (bottom panel). Open circles represent the population prediction (PRED) corrected observations. The dotted lines represent the median, 2.5th and 97.5th percentile of the PRED-corrected observations. The shaded areas represent the 95 % confidence intervals for the PRED-corrected simulated median and 2.5th and 97.5th percentiles.

Standard effect-compartment model Two-compartment effect site model

Page 30: Pharmacometric Models in Anesthesia and Analgesia

30

Table 3. Parameter estimates of the final 4-compartment propofol PK model with time-dependent elimination and distribution.

Parameter Estimate (RSE%)

IIV (RSE%)

Explanation

CL (L/min) 1.60 (4.1) 8.7 (39) ClearanceV1 (L) 2.88 (9.0) 19 (100) Volume of central compartmentQ2 (L/min) 1.29 (16.1) - Distribution clearance between compartments

1 and 2V2 (L) 4.47 (19.6) 48 (48) Volume of peripheral compartmentQ3 (L/min) 0.680 (12.1) - Distribution clearance between compartments

1 and 3V3 (L) 12.4 (12.6) 23 (50) Volume of peripheral compartmentQ4 (L/min) 0.458 (9.7) - Distribution clearance between compartments

1 and 4V4 (L) 71.2 (10.0) 12 (120) Volume of peripheral compartmenttlag (s) 5.23 (32.2) - Lag-time for appearance of drug at site of

samplingImax (%) 75.7 (4.7) - Maximum fractional decrease in elimination

and distribution clearance t50,bolus (s) 50.5 (26.3) 78 (35) Time-point for 50% of the maximum frac-

tional decrease in elimination and distribution clearances for the bolus dose

t50,PCI (s) 70.2 (34.4) 78 (35) Time-point for 50% of the maximum frac-tional decrease in elimination and distribution clearances for the primed constant infusion

σ (%) 12.9 (3.9) - Proportional residual errorσ2min (%) 21.6 (16.3) - Proportional residual error first 2 minθWT-CL (%/kg) 0.726 (22.2) - Effect of body weight on CLRSE% - relative standard error calculated as the ratio between the standard error and the estimate, and multiplied by 100. IIV - inter-individual variability in % of the parameter esti-mate.

Table 4. Parameter estimates for the final two-compartment effect site BIS model.

Parameter Estimate (RSE%)

IIV (RSE%)

Explanation

ke0 (min-1) 0.159 (5.96) 15 (38) Rate constant for distribution from effect compartment

Baseline BIS 92.5 (0.75) 3.5 (35)Emax (%) 90.4 (2.74) - Maximum decrease in BIS from baseline EC50 (ng/mL) 2550 (4.78) 18 (36) Effect site concentration needed to reach 50%

of Emax

γ 2.93 (4.98) - Shape factorke12 (min-1) 0.114 (13.16) - Rate constant for distribution from central to

peripheral effect compartmentke21 (min-1) 0.0214 (9.44) - Rate constant for distribution from peripheral

to central effect compartmentσ 6.55 (3.51) - Additive residual errorRSE% - relative standard error calculated as the ratio between the standard error and the estimate, and multiplied by 100, IIV - inter-individual variability in % of the parameter esti-mate.

Page 31: Pharmacometric Models in Anesthesia and Analgesia

31

AZD3043 PKPD model The PK of AZD3043 was described by a 3-compartment disposition model, with a lag-time for the drug to appear at the site of sampling. Clearance was high, 2.14 L/min, which is higher than the expected liver blood flow. This is consistent with AZD3043 being metabolized by esterases, both in plasma and in the liver. Despite this, the esterase activity was not found to influence CL. However, only subjects with a normal esterase activity were included in the study. Sex and age did not have a statistically significant influence on CL. Volume of distribution was low but dose-dependent. At the lowest ad-ministered dose, 1 mg/kg/h, Vss (the sum of central and peripheral volumes of distribution) was 12 L, and increased to 35 L at the highest administered dose, 81 mg/kg/h. PK parameters are shown in Table 5, and plasma concen-tration-time plots for the different dose-groups are shown in Figure 4.

Table 5. Parameter estimates of the final PK model for arterial concentrations of AZD3043.

Parameter Estimate (RSE%)

IIV (RSE%)

Description

CL (L/min) 2.14 (1.5) 13 (15) ClearanceV1 (L) 2.52 (10) 73 (27) Volume of central compartment Q2 (L/min) 3.77 (7.5) - Distribution clearance between com-

partments 1 and 2V2 (L) 4.68 (7.3) 68 (18) Volume of peripheral compartment 2 Q3 (L/min) 0.772 (6.2) - Distribution clearance between com-

partments 1 and 3V3 (L) 8.37 (4.6) 22 (25) Volume of peripheral compartment 3 tlag (min) 0.412 (0.56) - Lag-time for appearance of drug at site

of samplingθDose-V2 (%/mg/kg) 0.00122 (16) - Effect of dose on V2θDose-V3 (%/mg/kg) 0.000212 (17) - Effect of dose on V3σadd (μmol/L) 0.0202 (8.6) - Additive residual variabilityσprop (%) 16.9 (5.7) - Proportional residual variabilityRSE% relative standard error calculated as the ratio between the standard error and the esti-mate, and multiplied by 100, IIV inter-individual variability in % of the parameter estimate. Parameter estimates are for a typical individual of median weight (77 kg) and at a dose of 6 mg/kg.

The rate constant for effect-site distribution, ke0, was high, 0.69 min-1, corre-sponding to a half-life of the distribution of 1 minute. A two-compartment effect-site model, as developed for propofol in Paper I, did not improve the fit, suggesting that distribution within the effect site is limited or rapid. EC50 was estimated to 54 µmol/L, with an inter-individual variability of 35%. Plots of BIS over time for the different dose-groups are shown in Figure 5, and final parameter estimates for the BIS model are shown in Table 6.

Page 32: Pharmacometric Models in Anesthesia and Analgesia

32

Figure 4. Individual observed (grey) and predicted typical (black) arterial plasma concentrations of AZD3043 versus time, stratified for different dosing regimens.

Time (min)

Art

eria

l Con

cent

ratio

n (µ

mol

/L)

0.1

1

10

100

1 mg/kg/h 3 mg/kg/h 6 mg/kg/h 12 mg/kg/h

0.1

1

10

100

18 mg/kg/h 27 mg/kg/h 36 mg/kg/h 54 mg/kg/h

0.1

1

10

100

81 mg/kg/h 60 mg/kg/h 90 mg/kg/h 120 mg/kg/h

0.1

1

10

100

240 mg/kg/h 360 mg/kg/h 48 + 10 mg/kg/h 60 + 15 mg/kg/h

0.1

1

10

100

0 30 60 90 120

180 + 30 mg/kg/h

0 30 60 90 120

240 + 40 mg/kg/h

Page 33: Pharmacometric Models in Anesthesia and Analgesia

33

Figure 5. Individual observed (grey) and predicted typical (black) BIS versus time, stratified for different dosing regimens.

The analysis showed that BIS could serve as an indicator of anesthetic depth, as the relationship between BIS and the probability of being unconscious was similar to that for e.g. propofol. The values for BIS where 50% and 95% of the subjects are unconscious were estimated to 65 and 48, respectively. A VPC of the relationship is shown in Figure 6.

Time (min)

BIS

0

20

40

60

80

1001 mg/kg/h 3 mg/kg/h 6 mg/kg/h 12 mg/kg/h

0

20

40

60

80

10018 mg/kg/h 27 mg/kg/h 36 mg/kg/h 54 mg/kg/h

0

20

40

60

80

10081 mg/kg/h 60 mg/kg/h 90 mg/kg/h 120 mg/kg/h

0

20

40

60

80

100240 mg/kg/h 360 mg/kg/h 48 + 10 mg/kg/h 60 + 15 mg/kg/h

0

20

40

60

80

100

0 20 40 60

180 + 30 mg/kg/h

0 20 40 60

240 + 40 mg/kg/h

Page 34: Pharmacometric Models in Anesthesia and Analgesia

34

Table 6. Parameter estimates of the final BIS model for AZD3043.

Parameter Estimate (RSE%)

IIV (RSE%)

Description

Baseline 94.1 (0.31) 3.3 (18) Baseline BISEC50 (µmol/L) 54.3 (6.2) 35 (28) Concentration needed to reach 50% of Emax γ 1.51 (5.7) - Shape factorke0 (min-1) 0.689 (9.1) 78 (42) Rate constant for delay of effectσ 5.07 (3.7) - Additive residual variabilityRSE% relative standard error calculated as the ratio between the standard error and the esti-mate, and multiplied by 100, IIV inter-individual variability in % of the parameter estimate.

The short half-life, limited distribution, and fast equilibration between plas-ma and effect-site provided possibilities for a rapid onset and offset of anes-thesia. The between-subject variability in EC50 and in the relationship be-tween BIS and the probability of being unconscious makes individual titra-tion necessary in order to provide and maintain a proper depth of anesthesia.

Figure 6. Visual predictive check of the relationship between BIS and the probabil-ity of consciousness (MOAA/S>2) after administration of AZD3043. Circles repre-sent the observed proportion of conscious subjects at each value of BIS, and the shaded area represents the 95 % prediction interval based on the simulations. The solid line represents the model simulated probability of consciousness at each value of BIS.

Page 35: Pharmacometric Models in Anesthesia and Analgesia

35

Analgesia

Naproxcinod, naproxen and placebo PKPD model The pharmacokinetics of naproxen, during the first 8 hours after administra-tion, was best described by a one-compartment model with transit compart-ment absorption into the central compartment. Mean transit times for the absorption were 1.8 hours after naproxcinod and 0.5 hours after naproxen administration, respectively. The saturable protein binding was modeled using a binding model, with a Km of 0.55 µmol/L. Final PK parameter esti-mates are shown in Table 7.

Table 7. Parameter estimates for the final PK model of total and unbound naproxen.

Parameter Estimate (RSE%)

IIV (RSE%)

Description

CLu/F (L/h) 515 (12.1) 25 (37) Oral unbound clearanceVu/F (L) 4,290 (13.6) 44 (29) Oral unbound volume of distribution MTTnaproxcinod (h) 1.77 (10.8) 58 (24) Mean transit time for naproxcinodNNnaproxcinod 3.58 (9.9) 58 (26) Number of transit compartments for

naproxcinodMTTnaproxen (h) 0.500 (23.8) 100 (60) Mean transit time for naproxenNNnaproxen 4.23 (24.8) 64 (68) Number of transit compartments for

naproxenBmax (μmol/L) 643 (7.1) 17 (44) Maximum binding of naproxen to plasma

proteinsKm (μmol/L) 0.549 (10.2) - Naproxen concentration at half maximum

bindingFrel (%) 59.7 (14.6) - Relative naproxen bioavailability compared

to naproxen dosingσT,add (μmol/L) 6.19 (22.3) - Additive residual variability for total

naproxen concentrationsσT,prop (%) 8.43 (8.0) - Proportional residual variability for total

naproxen concentrationsσU,prop (%) 18.6 (11.0) - Proportional residual variability for un-

bound naproxen concentrationsCorr. MTT-NN (%)

-52 (38) - Correlation between MTTnaproxcinod and NNnaproxcinod

RSE% - relative standard error calculated as the ratio between the standard error and the estimate, and multiplied by 100, IIV inter-individual variability in % of the parameter esti-mate.

The placebo effect, and/or the natural course of the progression of pain, after removal of an impacted wisdom tooth was described using an exponential model, where the pain intensity declined or increased towards a new level with a rate constant of the equilibration of 0.24 h-1. On average, the placebo effect on pain intensity was a decrease by 20%, but with a large variability, allowing the pain intensity to decrease to 0 or increase to 100 mm on the VAS.

Page 36: Pharmacometric Models in Anesthesia and Analgesia

36

A sigmoid Emax model described the effect of unbound naproxen concen-trations on the pain intensity. EC50 was estimated to 0.135 μmol/L.

A Weibull time-to-event model described the time to request of rescue medication. The Weibull model suggests a higher hazard at early times and a lower hazard at later times. Pain intensity was found to have a large impact on the hazard, with the hazard increasing exponentially with increased pain intensity. The pain intensity at baseline was also found to influence the haz-ard. Subjects entering the study with a high pain intensity had a lower hazard at a given pain intensity than those entering the study with low pain intensi-ty. Parameter estimates for the pain intensity and dropout model are shown in Table 8. Visual predictive checks for the dropout are shown in Figure 7.

Figure 7. Visual predictive check of time to request of rescue medication after ad-ministration of naproxcinod 375 mg, 750 mg, 1500 mg and 2250 mg, naproxen 500 mg or placebo, based on the final PK/PD model. Solid line represents the observed Kaplan-Meier curve and the shaded area represents the 90% prediction interval for the simulated Kaplan-Meier curve.

Placebo

Naproxen 500 mg

Naproxcinod 375 mg

Naproxcinod 2250 mg

Naproxcinod 750 mg

Naproxcinod 1500 mg

Page 37: Pharmacometric Models in Anesthesia and Analgesia

37

Table 8. Parameter estimates for the final pain intensity and dropout model.

Parameter Estimate (RSE%)

IIV (RSE%)

Description

PIbaseline (mm) 52.7 (13.4) 32 (27) Baseline pain intensityPmax (%) 20.2 (12.2) 120 (16) Maximum placebo responsekpl (h

-1) 0.237 (68.8) 43 (39) Rate constant for placebo effectEC50 (μmol/L) 0.135 (10.4) 120 (21) Unbound concentration needed for half

maximum effectγ 1.61 (12.4) - Shape factorσPI (mm) 7.82 (13.3) - Residual variability for pain intensity λ 0.00999 (15.6) - Scale parameter in the Weibull distribu-

tionα 0.729 (9.9) - Shape parameter in the Weibull distribu-

tionθPI 0.0782 (9.2) - Influence of current PI on the hazard θbaseline -0.00261 (19.2) - Influence of baseline PI on the hazard RSE% relative standard error calculated as the ratio between the standard error and the esti-mate, and multiplied by 100, IIV inter-individual variability in % of the parameter estimate.

When using the model for simulations, for example for visual predictive checks of the efficacy variable, it was shown to be necessary to include the dropout model in the simulations in order to produce realistic plots (Figure 8). If the simulations are performed without using the dropout model, higher pain intensity would be simulated at the later time points, as those patients with the higher pain intensity are the ones that tend to drop out.

Figure 8. Visual predictive check of pain intensity (PI) versus time after administra-tion of naproxcinod 375 mg, 750 mg, 1500 mg and 2250 mg, naproxen 500 mg or placebo, based on the final PK/PD model. The top row is simulated without dropout and the bottom row is simulated with dropout. Open circles represent the observa-tions and the lines represent the median, 2.5th and 97.5th percentile of the observa-tions. The shaded areas represent the 95 % confidence intervals for the simulated median and 2.5th and 97.5th percentiles.

Page 38: Pharmacometric Models in Anesthesia and Analgesia

38

Simulations and estimations of dropout In the Base Scenario, average dropout ranged from 22% in the highest dose group, to 52% in the placebo group. When the simulated data were analyzed with the same model as used for the simulations, i.e. including a model for the dropout, bias in the fixed effects parameters was low, less than 5%. When a dropout model was not included in the analysis, bias was larger, but still less than 8% when the Laplace method was used. When FOCE-I was used, bias increased further in the fixed effects parameters, to more than 20% for EC50. The effect of the bias on the underlying efficacy profile was, however, not much affected (Figure 9).

Figure 9. Simulated effect variable for a typical individual given Placebo, Low Dose, Medium Dose and High Dose for a) Scenario 1, b) Scenario14 (increased dropout rate, on average 57%), and c) Scenario 10 (observations every 4th hour on-ly). Simulations are based on true parameters (thick solid line), parameters estimated with dropout model (dotted line, hidden by thick solid line), parameters estimated without dropout model using Laplace (dashed line) or FOCE-I (thin solid line). A lower value of the effect variable indicates a better effect, and hence a higher dose.

Bias in EC50 increased with increasing magnitude of placebo effect, increas-ing dropout rate, and decreasing number of assessments of the efficacy vari-able (Figure 10). Bias in EC50 was relatively unaffected by number of pa-tients per treatment group, although there was a trend towards higher bias with lower number of subjects per group, especially when the FOCE-I meth-od was used.

When the exact time of dropout was not recorded in the simulations, but rather the information that dropout occurred at some time between two measurements of the efficacy variable (interval censoring), bias was higher than when the exact time of dropout was recorded. This was more obvious the longer time between the observations.

a)

Time (h)

Effe

ct

0

10

20

30

40

50

60

0 2 4 6 8

b)

Time (h)

Effe

ct

0

10

20

30

40

50

60

0 2 4 6 8

c)

Time (h)

Effe

ct

0

10

20

30

40

50

60

0 2 4 6 8

Page 39: Pharmacometric Models in Anesthesia and Analgesia

39

Figure 10. Bias in EC50 when estimating with a dropout model (white), without a dropout model, using the Laplace method (grey), and without a dropout model, using the FOCE-I method (black), when varying a) observation interval, b) extent of dropout (relation between hazard and effect score, he), c) maximum placebo re-sponse (PLmax), and d) number of patients per group.

The imprecision in the parameters was affected similar to the bias, with higher imprecision with increased placebo effect, higher dropout rate and fewer observations. In contrast to the bias, imprecision was affected by number of subjects, with a higher imprecision with decreased number of subjects (Figure 11).

VPCs showed that in order to make realistic simulations in the presence of informative dropout a model describing the dropout is necessary. The VPCs created when not simulating with a dropout model showed clear dif-ferences between the “observed” and simulated medians and percentiles (Figure 12a). This was most apparent for the placebo group, where the pa-tients had the highest effect score, and hence the largest dropout rate. The “observed” median was lower than the simulated median as the patients with

c)

PLmax

Bia

s (%

)

-30

-20

-10

0

0.1 0.2 0.4 0.8

d)

Patients per group

Bia

s (%

)

-20

-15

-10

-5

0

15 25 45 65

a)

Observation interval (h)

Bia

s (%

)

-50

-40

-30

-20

-10

0

0.5 1 2 4

b)

he

Bia

s (%

)

-30

-20

-10

0

0.025 0.05 0.6 0.7

Page 40: Pharmacometric Models in Anesthesia and Analgesia

40

the highest effect score had dropped out in the “observed” data. For the ac-tive treatment groups it was apparent that the simulated 97.5th percentile was high, while in the “observed” data most patients with a high effect variable had dropped out. When a dropout model was included in the VPC simula-tions the resulting VPCs showed a good fit and agreement between “ob-served” and simulated medians and percentiles (Figure 12b). When the dropout was completely at random, however, a dropout model was not need-ed in order to produce realistic VPCs (Figure 12c).

Figure 11. Root mean square errors (RMSE) in EC50 when including a dropout model (white), without a dropout model, using the Laplace method (grey), and with-out a dropout model, using the FOCE-I method (black), when varying a) observation interval, b) extent of dropout (relation between hazard and effect score, he), c) max-imum placebo response (PLmax), and d) number of patients per group.

c)

PLmax

RM

SE

(%

)

0

10

20

30

40

0.1 0.2 0.4 0.8

d)

Patients per group

RM

SE

(%

)

0

5

10

15

20

25

30

15 25 45 65

a)

Observation interval (h)

RM

SE

(%

)

0

10

20

30

40

50

0.5 1 2 4

b)

he

RM

SE

(%

)

0

10

20

30

0.025 0.05 0.6 0.7

Page 41: Pharmacometric Models in Anesthesia and Analgesia

41

Figure 12. Visual Predictive Checks, a) without inclusion of dropout model for data containing informative dropout (Scenario 1), b) with dropout model for data contain-ing informative dropout (Scenario 1) and c) without inclusion of dropout model for data with 40% dropout completely at random. Circles represent “observations” in an arbitrarily simulated dataset. Solid and dashed lines are the median, 2.5th and 97.5th percentiles of the “observations”. Shaded areas represent the 95% confidence inter-val of the simulated median, 2.5th and 97.5th percentiles.

a)

b)

c)

Placebo Medium Dose High DoseLow Dose

Page 42: Pharmacometric Models in Anesthesia and Analgesia

42

Discussion

Anesthesia

Pharmacokinetics In anesthesia, a fast onset of effect is often wanted. In order to induce anes-thesia, short-duration, high-rate infusions or bolus doses are often used. This makes it very difficult to capture the PK, partly because it is difficult to col-lect enough blood samples in a very short time-frame, but also because the traditional PK models are not adequate for describing the pharmacokinetics during the first minutes after a short infusion. Henthorn et al. showed, by frequently sampling arterial concentrations of indocyanine green after a bo-lus injection in dogs, that there is a delay before the substance reaches the site of sampling [73]. Thereafter the concentrations oscillate during the first one to two minutes before starting to decrease monotonically. The same pattern was later shown in humans [74]. This behavior is not captured by the traditional compartmental models. Recirculatory PK models have been pro-posed to describe the early oscillating concentrations [75], but they require more frequent sampling than what is possible in a first-time-in-man study, like the studies described in Paper II. In Papers I and II a lag-time was used to describe the delay between site of administration and site of sampling. In the studies used in Papers I and II the drug was not given as a bolus dose, but rather as a constant infusion, even if the duration of some of the infusions were only 1 minute. This may have decreased the fluctuations in concentra-tions during the first minutes after start of administration. Due to the oscilla-tions, also the timing of sampling is of importance during the first two minutes. There could be a large difference in the measured concentrations if a sample happens to be collected at the peak or the trough of the oscillations. In addition, when the concentrations are changing fast, any error in the re-cording of sampling time may have a large influence on the model. In Paper I a higher residual unexplained variability was used during the first two minutes to account for the possible model misspecification and potential errors in sampling time.

The effects of propofol on cardiac output and liver blood flow could po-tentially alter the PK after administration [76,77]. In Paper I it was also not-

Page 43: Pharmacometric Models in Anesthesia and Analgesia

43

ed that with a time-dependency in CL, possibly due to effects on liver blood flow, the model could better describe the PK of propofol.

The purpose of the propofol PK model was to get a good input to the PD model. It was speculated that the differences in ke0 seen after different infu-sion rates of propofol could be due to the use of an inadequate PK model. Therefore additional components were added to the PK model to improve the fit in each patient. However, different ke0 were found with the different infusion rates also with the more complex PK model, suggesting that the difference in ke0 was not due to an inadequate PK model.

In Paper II the peripheral volumes of distribution after AZD3043 admin-istration were estimated to be dependent on the dose. The larger the dose, the larger was the volume of distribution. As AZD3043 is a lipophilic com-pound, and was administered as an emulsion, it is possible that the distribu-tion of the lipids in the emulsion could affect the distribution of AZD3043. The higher the dose, the more lipids were infused, and the higher was the volume of distribution. Administration of lipid emulsions is used as a treat-ment for overdoses or accidental iv administration of local anesthetics, lead-ing to a distribution of the anesthetic to the lipids in the emulsion [78]. It has also been shown that plasma concentrations of bupivacaine decrease and volume of distribution increase after treatment with iv administration of a lipid emulsion [79]. Similar to AZD3043, propofol is also a lipophilic com-pound administered as an emulsion. However, as only two different dosing regimens of propofol were studied in Paper II, and there were other differ-ences between the two treatments, any possible dose-dependent distribution was not noted for propofol.

Pharmacodynamics In contrast to the commonly used single effect-compartment model, where a ke0 that changes with infusion rate would be needed, a two-compartment effect site model could describe the different rates of onset and offset of measured BIS effects between the two dosing regimens of propofol, with a set of parameters that were independent of rate and duration of administra-tion. The two-compartment effect site model allows for distribution within the effect site, without influencing the plasma PK. The shorter the admin-istration, the shorter the time is for distribution, leading to a fast offset rate of effects. With increasing duration of infusion there is more time for distri-bution, and the offset of effects on BIS becomes more and more dependent on re-distribution from the peripheral effect-site compartment.

Upton et al. has suggested that distribution occurs within the brain, both in sheep [80] and in man [81]. In their human model the peripheral brain volume was estimated to 2.2 times larger than the central brain volume, whereas our results in Paper II suggest the peripheral compartment to be approximately 5 times larger. In the model developed by Upton et al., how-

Page 44: Pharmacometric Models in Anesthesia and Analgesia

44

ever, it was assumed that the total rather than the central brain concentration was related to the effect, and that the cerebral blood flow changed during treatment, which might have impacted the parameters. Furthermore, it has also been shown that the rate of distribution of drug to the brain was slower than suggested by the ke0 derived from efficacy data [82-84]. This indicates that distribution to the whole brain is slower than the distribution to the ef-fect site, which supports the rationale of the two-compartment effect site model. The two-compartment effect-site model has subsequently been used successfully in modeling the effects of propofol in morbidly obese patients [85].

The two-compartment effect-site model did not improve the fit of BIS for AZD3043. This suggests that distribution within the brain is limited, or that equilibration within the brain is very fast. This is also in accordance with the low volume of systemic distribution and the high ke0 that was found for AZD3043. The fast equilibration between plasma and effect-site and the limited distribution within the brain, together with the high clearance and low volume of distribution provides a possibility of a fast offset of effect even after longer infusions. However, the dose-dependent volume of distri-bution could potentially lead to a slower offset of effects when the duration of the infusion increases.

The fact that the relationship between BIS and the probability of uncon-sciousness for AZD3043 is similar to that for propofol is not surprising, giv-ing the similar mechanisms of action. It is still of importance to know the relationship if one would use BIS as a marker for efficacy when selecting dosing regimens for future studies.

Analgesia

Pharmacokinetics The pharmacokinetics of naproxen, after both naproxen and naproxcinod dosing, were described using a one-compartment model with saturable pro-tein binding and transit compartment absorption. The systemic parameters, clearance and volume of distribution, were assumed to be the same regard-less of in which form naproxen entered the systemic circulation. The absorp-tion parameters were different, however, after administration of naproxen and naproxcinod. For naproxcinod, the transit compartment parameters de-scribe not only the absorption, but also the metabolism to naproxen. In the study in Paper III, naproxen concentrations were collected for only 8 hours, while the half-life of naproxen is 14 hours [86]. Therefore the PK model might not be suitable for extrapolation to repeated dosing, although the first 8 hours are well described.

Page 45: Pharmacometric Models in Anesthesia and Analgesia

45

Pharmacodynamics In Paper III, plasma samples for analysis of naproxen concentrations were not collected in all patients. The patients without concentration measure-ments could still be included in the development of the PD-model. Keeping the concentration data in the PD-dataset and fixing the PK-parameters pro-vide a way of allowing for the most likely PK profile for those individuals, given the PK model and their PD data [59].

The pain scale used in Paper III, the VAS, is bounded between 0 and 100. In this case the model for the typical effect did not allow values below 0, and observations at 100 were very rare. However, with residual variability the model could still simulate values below 0 and above 100, and in order to avoid this, the simulations were truncated at these values. Different trans-formations of the VAS, such as logit transformation, could be used to avoid truncation, but this makes the scale and the effects difficult to interpret. There are also other pain scales available, for example categorical scales, which have been used in similar studies [87-89]. These scales do not have the same issues with boundaries, and make the dropout modeling easier as there is typically one value for the hazard at each pain score.

Dropout In Paper IV it was shown that bias in the PD-parameters in general were higher when a dropout model was not included in the analysis, but the bias was still in most cases low and did not have a large influence on the simula-tions. This was true also in Paper III, where an analysis of pain intensity only, without including a dropout model, gave very similar parameter esti-mates as in the final model where a dropout model was included. This is probably because the pain intensity data was frequently sampled, the exact time of dropout was collected, and the patients were asked to refrain from rescue medication during the first 90 minutes, which contributed to keep the bias low. In other situations, with larger bias, the development of the PD model and dropout model simultaneously could be needed. Even though bias was small, a dropout model was still necessary to use in order to find a good model for pain intensity, as it was not possible to select a good model based on goodness of fit plots produced without accounting for the dropout.

A larger bias and imprecision was seen with increasing dropout rate, and the more dropout the more important it is to take the dropout into account. This seems obvious, not only because the more dropout there is the more information of the efficacy does the dropout contain, but there is also a de-creased number of efficacy measurements with increasing dropout, making the dropout information more and more important in relation to the efficacy data.

Page 46: Pharmacometric Models in Anesthesia and Analgesia

46

With an increasing placebo effect, the number of dropouts due to lack of efficacy will decrease. Despite this, the bias and imprecision increased with increasing placebo effect. This could be due to the fact that the efficacy score used in the studies presented here is bound to be above 0. The larger the placebo effect, the less room is left for drug effect, and the smaller the drug effect is in relation to the placebo effect, the larger the bias and impre-cision in EC50.

When the number of observations per subject was decreased, bias in the parameters increased. As the dropout hazard was dependent on the predicted efficacy score continuously, and not only the observed efficacy score, the dropout carried information on what happens with the efficacy between ob-servations. As the observation interval becomes longer, the more important is the dropout to describe the efficacy score between measurements, and hence the dropout model becomes more important. As the number of meas-urements decrease, there is less information on the efficacy score, and the imprecision in the parameters increase. If the exact time of dropout is not available, there is less information about the efficacy score between observa-tions, and therefore the bias increase if interval censoring is used.

The bias was relatively unaffected by the number of subjects in the study. By changing the number of subjects the structure of the data is not changed, but only the amount of data. This was reflected in the higher imprecision found when number of subjects was decreased.

Bias in baseline score was low in all scenarios in Paper IV. This could be explained by the fact that baseline scores were collected in all subjects and were not affected by the subsequent dropout.

In the simulation study, the estimation models, both for the efficacy vari-able and for the dropout in the cases where a dropout model was used, was always the same as the model used for the simulations. If modeling had in-stead been performed in a stepwise manner, like modeling analyses normally are, the analysis might have ended in a final model with a different structure.

Page 47: Pharmacometric Models in Anesthesia and Analgesia

47

Conclusions

The distribution to peripheral parts of the effect site can explain why differ-ent ke0 have been observed for different infusion rates of propofol. A two-compartment effect-site model can be used to describe the effects of propofol on BIS after different rates of administration, or when the rate of administration is changed during treatment.

The probability of being unconscious is related to BIS after AZD3043 administration, similar to after propofol administration. AZD3043 has a high clearance, low but dose-dependent distribution, and a fast equilibration with the effect site, providing a rapid onset and offset of anesthesia. Variability makes individual titration of AZD3043 necessary in order to provide an ap-propriate depth of anesthesia.

In the dental pain model, dropout carries information on the efficacy vari-able, pain intensity. Both the current pain intensity and the pain intensity at baseline affect the hazard of requesting rescue medication. High pain intensi-ty increases the hazard exponentially, and subjects with a low baseline pain have a higher dropout hazard than patients with a high baseline, at the same level of pain intensity.

Ignoring informative dropout leads to biased parameter estimates. Using the Laplace method may lead to lower bias than if FOCE-I is used. Bias increases with increasing placebo effect, increasing dropout rate, and de-creasing number of observations per subject, but is relatively unaffected by number of subjects. Knowing the exact time of dropout leads to lower bias than using interval censoring, especially if the interval between observations is long.

Although bias produced by ignoring informative dropout may be small and have a small influence on the underlying predictions, the dropout is im-portant to consider in order to making realistic simulations, such as visual predictive checks or clinical trial simulations.

Page 48: Pharmacometric Models in Anesthesia and Analgesia

48

Populärvetenskaplig sammanfattning

Det har blivit allt dyrare att ta fram nya läkemedel och antalet nya läkemedel som tas fram per år minskar. Ett sätt att göra utvecklingen av läkemedel mer effektiv är att använda sig av matematiska modeller för att beskriva och för-klara resultaten av olika läkemedelsstudier.

Smärta drabbar i stort sett alla människor någon gång och är den vanligas-te orsaken till att man besöker sjukvården. Inom smärtlindring, analgesi, vill man ta bort smärtupplevelsen utan att påverka andra känsloupplevelser. An-estesi, det vill säga narkos, innebär att alla patientens känsloupplevelser, inklusive eventuell smärta, blockeras och patienten blir medvetslös.

Denna avhandling beskriver utvecklingen av olika modeller för hur läke-medel för smärtlindring och anestesi tas upp och sprids i kroppen, bryts ner och utsöndras, samt hur detta är relaterat till de effekter läkemedlet ger upp-hov till.

En av modellerna beskriver hur ett anestesiläkemedel, propofol, sprider sig i hjärnan. Modellen kan förklara varför det tar olika lång tid för läkemed-lets effekt att avta efter olika långa behandlingar. En annan modell beskriver hur ett nytt anestesiläkemedel, AZD3043, sprider sig i kroppen och bryts ner samt hur detta påverkar de effekter man ser av läkemedlet. Modellerna är till stor nytta när man ska bestämma hur man ska dosera läkemedlet för att snabbt få effekt, behålla effekten på rätt nivå, samt öka eller minska effekten efter behov.

Ett vanligt sätt att undersöka effekten hos nya läkemedel mot smärta är att ge dem till patienter som dragit ut en visdomstand. Det går då att studera hur patienternas smärta förändras över tiden efter ingreppet. Om det nya läke-medlet inte ger tillräcklig effekt får patienterna ett befintligt fungerande lä-kemedel och man kan inte längre studera effekten av enbart det nya läke-medlet. Att de patienter som har mest ont i större utsträckning vill ha ytterli-gare smärtlindring än de patienter där smärtan minskat leder till obalans i de data man samlar in. Detta kan hanteras med hjälp av en modell som beskri-ver hur sannolikheten att vilja ha ytterligare smärtlindring är relaterat till den smärta patienten har. Modellen användes för att beskriva effekten av två smärtläkemedel, naproxen och naproxcinod. I avhandlingen studeras också hur systematiska fel uppstår om man inte tar hänsyn till sambandet mellan smärta och sannolikheten att vilja ha ytterligare smärtlindring, samt olika faktorer som kan påverka storleken av det systematiska felet.

Page 49: Pharmacometric Models in Anesthesia and Analgesia

49

Acknowledgements

This thesis work was carried out at Clinical Pharmacology & Pharmacometrics, AstraZeneca R&D, Södertälje, Sweden, in collaboration with the Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Sweden. AstraZeneca is gratefully acknowledged for sponsoring this thesis. I would like to express my sincere gratitude to all who have supported me, encouraged me or contributed to this thesis in one way or another. I especial-ly would like to thank: Ulrika Simonsson, my supervisor, for always being positive and enthusiastic and always having time for discussions. I admire your skill in balancing the use of the stick and the carrot to make me move forward. I have really en-joyed working together with you. Mats Karlsson, my co-supervisor, for all the good advice, excellent courses, and always having a solution to the problems. Your passion for pharmacometrics is outstanding. My co-authors, Åke Norberg for your genuine interest in modeling and for our very interesting discussion about almost anything, Sigridur Kalman for your insightful clinical perspectives, and Lena Friberg for your brilliant ideas and ability to spot every mistake.

Magnus Jansson for the help with layout of the unpublished manuscripts. My managers at AstraZeneca, especially Marie Gårdmark, Marie Sandström, Per Dalén, Eva Bredberg and Jim Lynch, who kindly let me start working on the thesis and gave me the opportunity to continue the work over the years.

Margareta Hammarlund-Udenaes for always welcoming me to the Depart-ment.

Per-Henrik Zingmark for inspiring me to work with pharmacometrics and to start the PhD project. Thank you for all the fun we have had during work, travelling and vacations, and for keeping me fit.

Page 50: Pharmacometric Models in Anesthesia and Analgesia

50

My other past and present pharmacometrics colleagues at AstraZeneca in Södertälje; Anders Viberg, Angelica Quartino, Anja Henningsson, Emma Boström, Eva Hanze, Hung Lam, Jacob Brogren, Kristin Karlsson, Matts Kågedal, Petra Ekerot and Siv Jönsson for good advice, interesting discus-sions and a lot of fun. All other past and present colleagues and friends at AstraZeneca, in Sweden, UK, Japan, and the USA, for making it a pleasure going to work. All past and present colleagues at the Department of Pharmaceutical Biosci-ences, for all your help, excellent courses, and all the fun at different confer-ences and Christmas parties. Anders, Bengt, Carl-Magnus, Henrik, Jesper, Johan and Mattias för alla de roliga och mindre vetenskapliga delarna av den farmaceutiska världen och för att ni låter mig vinna när vi spelar poker. Alla andra vänner som gör att man gärna kopplar bort jobbet ett tag. Bibbi, Ray, Jocke och Sarah för trevligt umgänge och allt barnvaktande när jag reser eller jobbar sent.

Mina bröder, Peter och Tomas, och deras familjer för all omtanke. Ett speci-ellt tack för logi i Uppsala, även om jag fortfarande saknar den där lilla cho-kladbiten på kudden. Mina föräldrar, Karin och Örjan, för att ni alltid stöttar mig och hjälper till på alla sätt och vis. Min familj - Erika, Alva and Edvin. Ni är det bästa som finns. Jag älskar er!

Page 51: Pharmacometric Models in Anesthesia and Analgesia

51

References

1. Lalonde RL, Kowalski KG, Hutmacher MM, Ewy W, Nichols DJ, Milligan PA, Corrigan BW, Lockwood PA, Marshall SA, Benincosa LJ, Tensfeldt TG, Parivar K, Amantea M, Glue P, Koide H, Miller R. Model-based Drug Devel-opment. Clin Pharmacol Ther 2007; 82:21-32

2. Milligan PA, Brown MJ, Marchant B, Martin SW, van der Graaf PH, Benson N, Nucci G, Nichols DJ, Boyd RA, Mandema JW, Krishnaswami S, Zwillich S, Gruben D, Anziano RJ, Stock TC, Lalonde RL. Model-Based Drug Develop-ment: A Rational Approach to Efficiently Accelerate Drug Development. Clin Pharmacol Ther 2013; 93:502-514

3. US Department of Health and Human Services, Food and Drug Administration. Innovation or stagnation? Challenge and opportunity on the critical path to new medical products. 2004. (Accessed 15 August 2012) Available from: http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/CriticalPathInitiative/CriticalPathOpportunitiesReports/ucm113411.pdf

4. Zhang L, Sinha V, Forgue ST, Callies S, Ni L, Peck R, Allerheiligen SRB. Model-Based Drug Development: The Road to Quantitative Pharmacology. J Pharmacokinet Pharmacodyn 2006; 33:369-393

5. Allerheiligen SRB. Next-Generation Model-Based Drug Discovery and Devel-opment: Quantitative and Systems Pharmacology. Clin Pharmacol Ther 2010; 88:135-137

6. Suryawanshi S, Zhang L, Pfister M, Meibohm B. The current role of model-based drug development. Expert Opin Drug Discov 2010; 5:311-321

7. Manolis E, Rohou S, Hemmings R, Salmonson T, Karlsson M, Milligan PA. The Role of Modeling and Simulation in Development and Registration of Me-dicinal Products: Output From the EFPIA/EMA Modeling and Simulation Workshop. CPT: Pharmacomet Syst Pharmacol 2013; 2, e31

8. Stone JA, Banfield C, Pfister M, Tannenbaum S, Allerheiligen S, Wetherington JD, Krishna R, Grasela DM. Model-Based Drug Development Survey Finds Pharmacometrics Impacting Decision Making in the Pharmaceutical Industry. J Clin Pharmacol 2010; 50:20S-30S

9. Lee JY, Garnet CE, Gobburu, JVS, Bhattaram VA, Brar S, Earp JC, Jadhav PR, Krudys K, Lesko LJ, Li F, Liu J, Madabushi R, Marathe A, Mehrotra N, Tornoe C, Wang Y, Zhu H. Impact of Pharmacometric Analyses on New Drug Approv-al and Labelling Decisions: A Review of 198 Submissions Between 2000 and 2008. Clin Pharmacokinet 2011; 50:627-635

10. Jönsson S, Henningsson A, Edholm M, Salmonson T. Role of Modeling and Simulation – A European Regulatory Perspective. Clin Pharmacokinet 2012; 51:69-76

11. Ette E, Williams P. Pharmacometrics: The science of quantitative pharmacolo-gy. John Wiley & Sons, Inc, Hoboken, New Jersey, 2007

12. Jonsson EN, Sheiner LB. More efficient clinical trials through use of scientific model-based statistical tests. Clin Pharmacol Ther 2002; 72:603-614

Page 52: Pharmacometric Models in Anesthesia and Analgesia

52

13. Karlsson KE, Vong C, Bergstrand M, Jonsson EN, Karlsson MO. Comparisons of Analysis Methods for Proof-of-Concept Trials. CPT: Pharmacomet Syst Pharmacol 2013; 2, e23

14. Holford NH, Sheiner LB. Kinetics of pharmacological response. Pharmacol Ther 1982; 16:143-166

15. Holford NH, Sheiner LB. Understanding the Dose-Effect Relationship: Clinical Application of Pharmacokinetic-Pharmacodynamic Models. Clin Pharmacokinet 1981; 6: 429-453

16. Karlsson MO, Sheiner LB. The importance of modeling interoccasion variabil-ity in population pharmacokinetic analyses. J Pharmacokinet Biopharm 1993; 21:735-750

17. Karlsson MO, Beal SL, Sheiner LB. Three new residual error models for popu-lation PK/PD analyses. J Pharmacokinet Biopharm 1995; 23:651-672

18. Sun H, Fadiran EO, Jones CD, Lesko L, Huang SM, Higgins K, Hu C, Machado S, Maldonado S, Williams R, Hossain M, Ette EI. Population pharmacokinetics; A regulatory perspective. Clin Pharmacokinet. 1999; 37:41-58

19. Racine-Poon A, Wakefield J. Statistical methods for population pharmacokinet-ic modeling. Stat Meth Med Res 1998; 7:63-84

20. Beal S, Sheiner LB, Boeckmann A, Bauer RJ (eds). NONMEM users guides (1989-2009). ICON Development Solutions, Ellicott City, MD, USA 2009

21. Wang Y. Derivation of various NONMEM estimation methods. J Pharmacokinet Pharmacodyn 2007; 34:575-593

22. Minto CF and Schnider TW (2008) Contributions of PK/PD modelling to intra-venous anesthesia. Clin Pharmacol Ther 84:27-38

23. Sahinovic MM, Absalom AR, Struys MMRF. Administration and monitoring of intravenous anesthetics. Curr Opin Anesthesiol 2010;23:734-740

24. White M, Kenny GNC. Intravenous propofol anaesthesia using a computerised infusion system. Anaesthesia 1990;45:204–9

25. Schüttler J, Kloos S, Schwilden H, Stoeckel H. Total intravenous anaesthesia with propofol and alfentanil by computer-assisted infusion. Anaesthesia 1988; 43(Supplement): 2–7

26. De Castro V, Godet G, Mencia G, Raux M, Coriat P. Target-controlled infusion for remifentanil in vascular patients improves hemodynamics and decreases remifentanil requirement. Anesth Analg 2003;96:33-38

27. Fiset P, Mathers L, Engstrom R, Fitzgerald D, Brand SC, Hsu F, Shafer SL. Pharmacokinetics of Computer-controlled Alfentanil Administration in Children Undergoing Cardiac Surgery. Anesthesiology 1995;83:944-955

28. Shafer SL, Varvel JR, Aziz N, Scott JC. Pharmacokinetics of fentanyl adminis-tered by computer-controlled infusion. Anesthesiology 1990; 73: 1091–102

29. Mason DG, Swinhoe CF, Linkens DA, Reilly CS. Development of a pharmaco-kinetic model-based infusion system for ketamine analgesia. Int J Clin Monit Comput 1996;13:139-142

30. Jain U, Body SC, Bellows W, Wolman R, Mora Mangano C, Mathew J, Youngs E, Wilson R, Zhang A, Mangano DT. Multicenter Study of Target-Controlled Infusion of Propofol-Sufentanil or Sufentanil-Midazolam for Coronary Artery Bypass Graft Surgery. Anesthesiology 1996; 85:522-535

31. Struys MMRF, Coppens MJ, De Neve N, Mortier EP, Doufas AG, Van Bocxlaer JFP, Shafer SL. Influence of administration rate on propofol plasma-effect site equilibration. Anesthesiology 2007; 107:386-396

32. Sigl JC, Nassib GC. An introduction to bispectral analysis for the electroen-cephalogram. J Clin Monitoring 1994;10:392-404

Page 53: Pharmacometric Models in Anesthesia and Analgesia

53

33. Glass PS, Bloom M, Kearse L, Rosow C, Sebel P, Manberg P. Bispectral Anal-ysis Measures Sedation and Memory Effects of Propofol, Midazolam, Isoflurane, and Alfentanil in Healthy Volunteers. Anesthesiology 1997;86:836-847

34. Vereecke HEM, Struys MMRF, Mortier EP. A comparison of bispectral index and ARX-derived auditory evoked potential index in measuring the clinical in-teraction between ketamine and propofol anaesthesia. Anaesthesia, 2003; 58:957-961

35. Barr G, Jakobsson JG, Öwall A, Andersson RE. Nitrous oxide does not alter bispectral index: study with nitrous oxide as sole agent and as an adjunct to i.v. anaesthesia. Br J Anaesth 1999;82:827-830

36. Abbott FV, Fraser MI. Use and abuse of over-the-counter analgesic agents. J Psychiat Neurosci 1998; 23:13-34

37. Katz J, Melzack R. Measurement of pain. Surg Clin North Am 1999; 79:231-252

38. Huskisson EC. Measurement of pain. Lancet 1974; 2:1127-1131 39. Urquhart E. Analgesic agents and strategies in the dental pain model. J Dent

1994; 22:336-341 40. Heyting A, Tolboom JTBM, Essers JGA. Statistical handling of drop-outs in

longitudinal clinical trials. Statist Med 1992; 11:2043-2061 41. Little RJA, Rubin DB. Statistical Analysis with Missing Data, Wiley, New

York 1987 42. Rubin DB. Inference and missing data. Biometrika 1976; 63:581-592 43. Laird NM. Missing data in longitudinal studies. Stat Med 1988; 7:305-315 44. Siddiqui O, Hung HMJ, O’Neill R. MMRM vs. LOCF: a comprehensive com-

parison based on simulation study and 25 NDA datsets. J Biopharm Stat 2009; 19:227-246

45. Gastonguay MR, French JL, Heitjan DF, Rogers JA, Ahn JE, Ravva P. Missing Data in Model-Based Pharmacometric Applications: Points to Consider. J Clin Pharmacol 2010; 50:63S-74S

46. Gomeni R, Lavergne A, Merlo-Pich E. Modeling placebo response in depres-sion trials using a longitudinal model with informative dropout. Eur J Pharm Sci 2009; 36:4-10

47. Hu C, Sale ME. A joint model for nonlinear longitudinal data with informative dropout. J Pharmacokinet Pharmacodyn 2003; 30:83-103

48. Hu C, Szapary PO, Yeilding N, Zhou H. Informative dropout modeling of lon-gitudinal ordered categorical data and model validation: application to expo-sure-response modeling of physician’s global assessment score for ustekinumab in patients with psoriasis. J Pharmacokinet Pharmacodyn 2011; 38:237-260

49. Egan TD, Obara S, Jenkins TE, Jaw-Tsai SS, Amagasu S, Cook DR, Steffensen SC, Beattie DT. AZD-3043: A Novel Metabolically Labile Sedative-Hypnotic Agent with Rapid and Predictable Emergence from Hypnosis. Anesthesiology 2012;116:1267-1277

50. Schnitzer TJ, Kivitz AJ, Lipetz RS, Sanders N, Hee A. Comparison of the COX-inhibiting nitric oxide donator AZD3582 and rofecoxib in treating the signs and symptoms of osteoarthritis of the knee. Arthritis Rheum 2005; 53:827-837

51. Karlsson J, Pivodic A, Aguirre D, Schnitzer TJ. Efficacy, safety, and tolerability of the cyclooxygenase-inhibiting nitric oxide donator naproxcinod in treating osteoarthritis of the hip or knee. J Rheumatol 2009; 36:1290-1297

Page 54: Pharmacometric Models in Anesthesia and Analgesia

54

52. Hill CM, Sindet-Pederson S, Seymour RA, Hawkesford JE 2nd, Coulthard P, Lamey P-J, Cowan CG, Wickens M, Jeppsson L, Dean ADP, Svensson O. An-algesic efficacy of the cyclooxygenase-inhibiting nitric oxide donor AZD3582 in postoperative dental pain: comparison with naproxen and rofecoxib in two randomized, double-blind, placebo-controlled studies. Clin Ther 2006; 28:1279-1295

53. Fagerholm U, Björnsson MA. Clinical pharmacokinetics of the cyclooxygenase inhibiting nitric oxide donator (CINOD) AZD3582. J Pharm Pharmacol 2005; 57:1539-1554

54. Chiou WL. The Phenomenon and Rationale of Marked Dependence of Drug Concentration on Blood Sampling Site – Implications in Pharmacokinetics, Pharmacodynamics, Toxicology and Therapeutics (Part II). Clin Pharmacokinet 1989; 17:275-290

55. Tuk B, Danhof M, Mandema JW. The Impact of Arteriovenous Concentration Differences on Pharmacodynamic Parameter Estimates. J Pharmacokinet Biopharm 1997; 25:39-62

56. Chernik DA, Gillings D, Laine H, Hendler J, Silver JM, Davidson AB, Schwam EM, Siegel JL. Validity and reliability of the Observer’s Assessment of Alert-ness/Sedation Scale: study with intravenous midazolam. J Clin Psychopharmacol 1990;10:244-251

57. 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;79:241-257

58. Jonsson EN, Karlsson MO. Xpose - an S PLUS population pharmacokinet-ic/pharmacodynamic model building aid for NONMEM. Comput Methods Pro-grams Biomed 1999;58: 51-64

59. Zhang L, Beal SL, Sheiner LB. Simultaneous vs. Sequential Analysis for Popu-lation PK/PD Data I: Best-case Performance. J Pharmacokin Pharmacodyn 2003;30:387-404

60. Gepts E, Camu F, Cockshott ID, Douglas EJ (1987) Disposition of propofol administered as constant rate infusions in humans. Anesth Analg 66:1256-1263

61. Marsh B, White M, Morton N, Kenny GNC (1991) Pharmacokineitc model driven infusion of propofol in children. Br J Anaesthesia 67:41-48

62. Schnider TW, Minto CF, Gambus PL, Andresen C, Goodale DB, Shafer SL, Youngs EJ. The influence of method of administration and covariates on the pharmacokinetics of propofol in adult volunteers. Anesthesiology 1998; 88:1170-1182

63. Schüttler J and Ihmsen H. Population pharmacokinetics of propofol. Anesthesi-ology 2000; 92:727-738

64. Kazama T, Ikeda K, Morita K, Kikura M, Ikeda T, Kurita T, Sato S. Investiga-tion of effective anesthesia induction doses using a wide range of infusion rates with undiluted and diluted propofol. Anesthesiology 2000; 92:1017-1028

65. Billard V, Gambus PL, Chamoun N, Stanski DR, Shafer SL. A comparison of spectral edge, delta power, and bispectral index as EEG measures of alfentanil, propofol, and midazolam drug effect. Clin Pharmacol Ther 1997; 61:45-58

66. Struys MMRF, De Smeet T, Depoorter B, Versichelen LFM, Mortier EP, Dumortier FJE, Shafer SL, Rolly G. Comparison of plasma compartment versus two methods for effect compartment-controlled target-controlled infusion for propofol. Anesthesiology 2000; 92:399-406

67. Doufas AG, Bakhshandeh M, Bjorksten AR, Shafer SL, Sessler DI. Induction speed is not a determinant of propofol pharmacodynamics. Anesthesiology 2004; 101:1112-1121 10

Page 55: Pharmacometric Models in Anesthesia and Analgesia

55

68. Jeleazcov C, Ihmsen H, Schmidt J, Ammon C, Schwilden H, Shüttler J, Fechner J. Pharmacodynamic modelling of the bispectral index response to propofol-based anaesthesia during general surgery in children. Br J Anaesthesia 2008; 100:509-516

69. Holford NHG. A size Standard for Pharmacokinetics. Clin Pharmacokin 1996;30:329-332

70. Runkel R, Forchielli E, Sevelius H, Chaplin M, Segre E. Nonlinear plasma level response to high doses of naproxen. Clin Pharmacol Ther 1974; 15:261-266

71. Savic RM, Jonker DM, Kerbusch T, Karlsson MO. Implementation of a transit compartment model for describing drug absorption in pharmacokinetic studies. J Pharmacokinet Pharmacodyn 2007; 34:711-726

72. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Prediction-Corrected Visual Predictive Checks for Diagnosing Non-Linear Mixed Effects Models. AAPS J 2011;13:143-151

73. Henthorn TK, Avram MJ, Krejcie TC, Shanks CA, Asada A, Kaczynski DA. Minimal compartmental model of circulatory mixing of indocyanine green. Am J Physiol 1992;262:H903-910

74. Avram MJ, Krejcie TC, Henthorn TK, Niemann CU. β-Adrenergic Blockade Affects Initial Drug Distribution Due to Decreased Cardiac Output and Altered Blood Flow Distribution. J Pharmacol Exp Ther 2004; 311:617-624

75. Krejcie TC, Henthorn TK, Nieman CU, Klein C, Gupta DK, Gentry WB, Shanks CA, Avram MJ. Recirculatory Pharmacokinetic Models of Markers of Blood, Extracellular Fluid and Total Body Water Administered Concomitantly. J Pharmacol Exp Ther 1996; 278:1050-1057.

76. Upton RN, Ludbrook GL, Grant C, Martinez AM. Cardiac output is a determi-nant of the initial concentrations of propofol after short-infusion administration. Anesth Analg 1999; 89:545-552

77. Peeters MYM, Aarts LPHJ, Boom FA, Bras LJ, Tibboel D, Danhof M, Knibbe CAJ. Pilot study on the influence of liver blood flow and cardiac output on the clearance of propofol in critically ill patients, Eur J Clin Pharmacol 2008; 64:329-334

78. Ciechanowicz S, Patil V. Lipid Emulsion for Local Anesthetic Systemic Toxici-ty. Anesthesiology Research and Practice. 2012; Article ID 131784

79. Litonius E, Tarkkila P, Neuvonen PJ, Rosenberg PH. Effect of intravenous lipid emulsion on bupivacaine plasma concentration in humans. Anaesthesia 2012;67:600-605

80. Upton RN, Ludbrook GL, Grant C. The cerebral and systemic kinetics of thiopentone and propofol in halothane anaesthetized sheep. Anaesth Intensive Care 2001; 29:117-123

81. Upton RN and Ludbrook G. A Physiologically Based, Recirculatory Model of the kinetics and dynamics of propofol in man. Anesthesiology 2005; 103:344-352

82. Peacock JE, Blackburn A, Sherry KM, Reilly CS. Arterial and jugular venous bulb blood propofol concentrations during induction of anesthesia. Anesth Analg 1995; 80:1002-1006

83. Dutta S, Matsumoto Y, Muramatsu A, Matsumoto M, Fukuoka M, Ebling WF. Steady-state propofol brain:plasma and brain:blood partion coefficients and the effect-site equilibration paradox. Br J Anaesthesia 1998; 81:422-424

84. Ludbrook GL, Visco E, Lam AM. Propofol: relation between brain concentra-tions, electroencephalogram, middle cerebral artery blood flow velocity, and cerebral oxygen extraction during induction of anesthesia. Anesthesiology 2002; 97:1363-1370

Page 56: Pharmacometric Models in Anesthesia and Analgesia

56

85. van Kralingen S, Diepstraten J, Peeters MYM, Deneer VHM, van Ramshorst B, Wiezer RJ, van Dongen EPA, Danhof M, Knibbe CAJ. Population Pharmacoki-netics and Pharmacodynamics of Propofol in Morbidly Obese Patients. Clin Pharmacokinet 2011; 50:739-750

86. Runkel R, Chaplin M, Boost G, Segre E, Forchielli E. Absorption, distribution, metabolism, and excretion of naproxen in various laboratory animals and hu-man subjects. J Pharm Sci 1972; 61:703-708

87. Sheiner LB. A new approach to the analysis of analgesic drug trials, illustrated with bromfenac data. Clin Pharmacol Ther 1994; 56:309-322

88. Mandema JW, Stanski DR. Population pharmacodynamic model for ketorolac analgesia. Clin Pharmacol Ther 1996; 60:619-635

89. Rohatagi S, Kastrissios H, Sasahara K, Truitt K, Moberly JB, Wada R, Salazar DE. Pain relief model for a COX-2 inhibitor in patients with postoperative den-tal pain. Br J Clin Pharmacol 2008; 66:60-70

Page 57: Pharmacometric Models in Anesthesia and Analgesia
Page 58: Pharmacometric Models in Anesthesia and Analgesia

Acta Universitatis UpsaliensisDigital Comprehensive Summaries of Uppsala Dissertationsfrom the Faculty of Pharmacy 173

Editor: The Dean of the Faculty of Pharmacy

A doctoral dissertation from the Faculty of Pharmacy, UppsalaUniversity, is usually a summary of a number of papers. A fewcopies of the complete dissertation are kept at major Swedishresearch libraries, while the summary alone is distributedinternationally through the series Digital ComprehensiveSummaries of Uppsala Dissertations from the Faculty ofPharmacy.

Distribution: publications.uu.seurn:nbn:se:uu:diva-205580

ACTAUNIVERSITATIS

UPSALIENSISUPPSALA

2013


Recommended