Basic Concepts in Population Modeling
Basic Concepts in Population Modeling
Abstract
Go to:
Overview
This tutorial serves as an introduction to model (and simulation)-based approaches for drug development
for novice modelers and for those who, while not being modelers themselves, nevertheless use the
approach and want to increase their understanding of the process. The tutorial provides some history,
describes modeling and simulation (e.g., pharmacometrics) with emphasis on population modeling and
simulation, and discusses some regulatory, project management, and information technology issues. This
is the first in a series of articles aimed at providing basic information on pharmacometrics.
Brief history
Atkinson and Lalonde1 stated that “dose selection and dose regimen design are essential for converting
drugs from poisons to therapeutically useful agents.” Modeling and simulation have emerged as important
tools for integrating data, knowledge, and mechanisms to aid in arriving at rational decisions regarding
drug use and development. Figure 1 presents a brief outline of some areas in which modeling and
simulation are commonly employed during drug development. Appropriate models can provide a
framework for predicting the time course of exposure and response for different dose regimens. Central to
this evolution has been the widespread adoption of population modeling methods that provide a
framework for quantitating and explaining variability in drug exposure and response.
Figure 1
Modeling and simulation during drug development.
All drugs exhibit between-subject variability (BSV) in exposure and response, and many studies
performed during drug development are aimed at identifying and quantifying this variability. A sound
understanding of the influence of factors such as body weight, age, genotype, renal/hepatic function, and
concomitant medications on drug exposure and response is important for refining dosage
recommendations, thereby improving the safety and efficacy of a drug agent by appropriately controlling
variability in drug exposure.
Population modeling is a tool to identify and describe relationships between a subject's physiologic
characteristics and observed drug exposure or response. Population pharmacokinetics (PK) modeling is
not a new concept; it was first introduced in 1972 by Sheiner et al.2 Although this approach was initially
developed to deal with sparse PK data collected during therapeutic drug monitoring, 3 it was soon
expanded to include models linking drug concentration to response (e.g., pharmacodynamics
(PD)).4 Thereafter, modeling has grown to become an important tool in drug development.
Population parameters were originally estimated either by fitting the combined data from all the
individuals, ignoring individual differences (the “naive pooled approach”), or by fitting each individual's
data separately and combining individual parameter estimates to generate mean (population) parameters
(the “two-stage approach”). Both methods have inherent problems, which become worse when
deficiencies such as dosing compliance, missing samples, and other data errors are present, 5 resulting in
biased parameter estimates. The approach developed by Sheiner et al. addressed the problems associated
with both the earlier methods and allowed pooling of sparse data from many subjects to estimate
population mean parameters, BSV, and the covariate effects that quantitate and explain variability in drug
exposure. This approach also allowed a measure of parameter precision by generation of SE.
At first glance, the term “population PK” suggests that the individual patient is ignored; however, the
importance of the individual in population models is highlighted by the description of variability, with
data from each individual contributing to the identification of trends such as changes in drug exposure
with changing age or weight, and the subsequent estimation of the population characteristics.
Pharmacometrics can be used to improve our understanding of mechanisms (e.g., linear or saturable
metabolism), inform the initial selection of doses to test, modify or personalize dosage for subpopulations
of patients, and evaluate the the appropriateness of study designs. 6
Go to:
Types of Models
PK models
PK models describe the relationship between drug concentration(s) and time. The building block of many
PK models is a “compartment”—a region of the body in which the drug is well mixed and kinetically
homogenous (and can therefore be described in terms of a single representative concentration at any time
point10). Compartments have proven to be ubiquitous and fundamental building blocks of PK models,
with differences between models often being defined by the way the compartments are connected.
Mammillary models generally have a central compartment representing plasma with one or two
peripheral compartments linked to the central compartment by rate constants (e.g., k 12 and
k21).11 Compartments in mammillary models can sometimes be real physiologic spaces in the body (such
as the blood or extravascular fluid), but are more typically abstract concepts that do not necessarily
represent any particular region of the body.
In contrast, physiology-based PK models (PBPK) use one or more compartments to represent a defined
organ of the body, with a number of such organ models connected by vascular transport (blood flow) as
determined by anatomic considerations.12 Mammillary PK models can generally be informed by blood or
plasma concentrations alone whereas, with PBPK models, tissue and plasma concentrations are typically
required, or parameters may have to be set according to values mentioned in the literature. This makes it
complicated to apply PBPK models to clinical data; on the other hand, it provides a greater scope to
understand the effect of physiologic perturbations and disease on drug disposition, and often improves the
ability to translate findings from preclinical to clinical settings.
PKPD models
PK/PD (PKPD) models include a measure of drug effect (PD). They have been the focus of considerable
attention because they are vital for linking PK information to measures of activity and clinical
outcomes.13Models describing continuous PD metrics often represent the concentration–effect relationship
as a continuous function (e.g., linear, Emax, or sigmoid Emax). The concentration that “drives” the PD model
can be either the “direct” central compartment (plasma) drug concentration, or an “indirect” effect
wherein the PD response lags behind the plasma drug concentration. Models describing discrete PD
effects (e.g., treatment failure/success, or the grade of an adverse event) often use logistic equations to
convert the effect to a probability within a cohort of subjects. This probability can be related to a PK
model. Exposure–response models are a class of PKPD models wherein the independent variable is not
time, but rather, a metric describing drug exposure at steady-state (e.g., dose, area under the curve (AUC),
or peak plasma concentration (Cmax)).
Disease progression models
Disease progression models were first used in 1992 to describe the time course of a disease metric (e.g.,
ADASC in Alzheimer's disease14). Such models also capture the inter-subject variability in disease
progression, and the manner in which the time course is influenced by covariates or by treatment. 15 They
can be linked to a concurrent PK model and used to determine whether a drug exhibits symptomatic
activity or affects progression.16 Models of disease progress in placebo groups are crucial for
understanding the time course of the disease in treated groups, as well as for predicting the likely
response in a placebo group in a clinical trial.17
This model states the relationship between the independent variable, time (t), and the dependent variable,
concentration (C). The notation C(t) suggests that C depends on t. Dose, clearance (CL), and distribution
volume (V) are parameters (constants); they do not change with different values of t. Note the differences
in the uses of the terms “variable” and “parameter.” The dependent and independent variables are chosen
merely to extract information from the equation. In PK, time is often the independent variable. However,
Equation (1) could be rearranged such that CL is the independent variable and time is a constant (this may
be done for sensitivity analysis for example).
A differential equation describes the rate of change of a variable. In this example, dC/dt is the notation for
the rate of change of concentration with respect to time (sometimes abbreviated as C′). Note that
differential equations require specification of the initial value of the dependent variables. Here, the value
of C at time zero (C0) is Dose/V.
Numerical methods are needed to solve systems of differential equations. Euler's method is a simple
example and can be easily coded. Numerically solving Equation (2) requires approximating the value of
the variable (C2) after an increment in time (t2 – t1) based on the previous value (C1) and the implied rate
of change (–CL/V*C1):
An initial value is needed for this process (to give the first value for C1, see Equation (2)). Computational
errors are minimized by keeping the time increments very small. There has been extensive development
of algorithms to solve differential equations numerically, and in most contexts the difference between an
analytical solution and the approximate numerical solution is inconsequential. However, solving a system
of equations is computationally intensive and, even with automated, rapid processors, there is a time
penalty for using differential equations to describe a model. Generally, algebraic equations and
superposition are exploited unless the model is complex or nonlinear with respect to its parameters (e.g.,
saturable metabolism), in which case differential equations are necessary.
Figure 2
Best fit for a simple population pharmacokinetics model. (a) Goodness-of-fit plots for the model shown
in Table 1. Symbols represent the observed data. The solid blue line denotes individual predicted
concentration (CIPRED). The broken line denotes the ...
Figure 3
Effect of covariates on variability. (a) Plot of concentrations vs. time. The solid green line denotes
population average, the broken black lines denote individual averages, the red symbols represent
concentration values in elderly subjects, and the black ...
Table 1
Equations for a simple population pharmacokinetic model
Population models need to include a description of RUV. RUV is defined by a quantity (EPS) reflecting
the difference between the observed data for an individual and the model's prediction (the residual). EPS
is assumed to be normally distributed and centered around zero, and is summarized by its variance (or
SD), often termed as SIGMA. SIGMA is estimated from the data (e.g., “RUV was estimated to be 18%”).
There are four estimated parameters in the model that have been described as an example: THETA1,
THETA2, OMEGA, and SIGMA. The relationships between parameter values and variables in the
example model are summarized in Table 2.
Table 2
Parameter and variable values for a simple population model
Estimation methods
The concept of estimating the “best parameters” for a model is central to the modeling endeavor. There
are clear analogies to linear regression, wherein the slope and intercept parameters of a line are estimated
from the data. Linear regression is based on “least squares” minimization. The difference between each
pair of observed (e.g., Cobs) and predicted (e.g., “ ”) values for the dependent variables is calculated,
yielding the residual (Cobs – ). The best parameters achieve the lowest value of the sum of the squares of
the residuals (which is used so that positive and negative residuals do not cancel each other out). The
“sum of squares” term can be thought of as an “objective function.” It has a given value for each unique
pair of slope and intercept parameters, and is lowest for the line of best fit.
Most pharmacometric models need some extensions to this least squares concept for estimating the
parameters. The first extension is needed because the least squares objective function is dependent on the
magnitude of the data (i.e., high data points can be given more “weight” than low data points) and,
because there is often a subjective component to the choice of weights, it is best to avoid this situation.
Maximum likelihood estimation is commonly used because it avoids the need for data weighting. For a
given pair of observed and predicted data values, is considered to have a possible range of values
described by a normal distribution, with a mean of and a SD given by the estimate of sigma (see Table
1). The likelihood of the observed data (closely associated with probability) is a metric summarizing the
deviation of the observed data (Cobs) from the center of this distribution. For ease of computation, the
maximum likelihood estimation objective function is usually expressed as the negative sum of the log of
the likelihoods, yielding a single number—the maximum likelihood estimation objective function value
(OFV). The minimum value of the OFV for a particular model and data set is associated with the “best
fit” parameter values, but the absolute value of the OFV is not important. It is used within a model for
comparing parameter values, and is compared between models for ranking them in order of goodness of
fit for the same dataset. The OFV also offers some advantages. It allows simultaneous fitting of random
effects and residual error (crucial to population models) and has a distribution (approximately χ 2) that
facilitates the use of statistical tests to make comparisons between models.
The second extension arises from the fact that, unlike linear models, most PK models are too complex to
solve for the minimum value of the OFV by means of algebraic methods. Optimization approaches are
used, involving searching for combinations of parameter values that produce the lowest value of the OFV.
When two parameters are fitted, it is possible to show the OFV as a three-dimensional surface (Figure
2b). There are many optimization algorithms (“estimation methods”) for finding the minimum value of
this OFV surface. The simplest of these is the “gradient method.” Starting at one point on the surface, the
parameters are evaluated to determine the direction in which the OFV decreases the most. The next set of
parameters is chosen to take a “step” in this direction, and the process is repeated until the minimum OFV
is found. There are some key features of optimization processes, regardless of the actual algorithm used
(the algorithm is usually chosen on the basis of accuracy, robustness, and speed). First is the need to
specify initial parameter values (essentially telling the search algorithm where to start on the OFV
surface). Second is the concept of local minima on the OFV surface. There is a risk that the search
algorithm will find a local minimum rather than the lower global minimum. Local minima arise for some
combinations of models and data when there are two sets of parameter values that, although different,
provide similar fits to the data. Appropriate choice of initial values helps reduce the risk of finding a local
minimum in estimation (for instance, by starting the search nearer the global minimum). Finally, as can
be seen in Figure 2b, the minimum of the OFV sits in a “trough” on the OFV surface. The shape of this
trough provides important information about the uncertainty in the parameter estimates. For a steep sided
trough, there are a limited range of parameter values that can describe the data for this model. In contrast,
a broad, shallow trough implies that a greater range of parameter values can describe the data for this
model (i.e., uncertain/imprecise parameter estimates). This uncertainty in parameter estimates can be
quantified from the shape of the “trough” on the OFV surface, and is usually reported either as SE of the
parameter estimate or as confidence intervals for the parameter. For example, the statement “V was
estimated to be 13.6 l with a SE of 11%” means that, given the model and data, there is a high certainty in
the prediction of V. Precise parameter estimates are a desirable feature of a model, particularly when the
parameter value is crucial in making inferences from the model. Over-parameterized models generally
have one or more parameters with high imprecision (i.e., there is not enough information in the data to
estimate the parameter) and may therefore benefit from simplification.
Simulation methods
Using models to simulate data is an important component of pharmacometric model evaluation and
inference. For the purpose of evaluation, the model may be used to simulate data that are suitable for
direct comparison with the index data. This can be done either by using a subset of the original database
used in deriving the model (internal validation) or a new data set (external validation). For the purpose of
inference, the model is generally used to simulate data other than observed data. Interpolation involves
simulation of nonobserved data that lie within the bounds of the original data (e.g., simulating AUC for a
25 mg dose when the observed data used in building the model was for 20 and 30 mg doses).
Extrapolation involves simulation of nonobserved data that lie outside the bounds of the original data
(e.g., simulating AUC for a 100 mg dose when the observed data was for 20 and 30 mg doses).
Extrapolation requires confidence in the assumptions of the underlying model. In this example, if the
model has been designed with the assumption of dose linearity, and if the drug has saturable metabolism,
the model predictions may be erroneous. Simulations should therefore be interpreted with a clear
understanding of the limitations and assumptions inherent in the model. Nevertheless, using models to
frame mechanisms and hypotheses, and for extrapolating and experimentally testing the model
predictions, is part of the “Learn and Confirm” paradigm of model building.
Simulating from models with fixed-effect and random-effect parameters (i.e., stochastic simulation with
population models) is more complex than non-stochastic simulation from simple fixed-effect models.
Random-effect parameters account for unexplained variability in the data that must be recreated during
simulation. This is done by using a random number generator to sample parameter values from a
distribution, with the mean and SD of the distribution of random effects as found from the estimation
process. Most modeling software has random number generators for a variety of distributions (e.g.,
uniform, normal, log-normal, binomial, etc.) as appropriate for a given model.
For stochastic simulations, the model needs to be simulated repeatedly so that the distribution of the
simulated output can be summarized (e.g., mean values and SD). In theory, more simulation replicates are
better, but the number that are actually performed is often limited by considerations of time and data size.
A common “rule of thumb” is that at least 200 simulations are needed when summarizing simulated data
as mean values, and at least 1,000 are needed when summarizing as confidence intervals. When
simulating stochastic models with more than one random effect parameter, it is important to understand
potential correlations among the parameters, and to account for this factor during simulation so as to
avoid implausible combinations of parameters in individual subjects.
Clinical trial simulation is an important application of the simulation method. It is not a new method; it
involves the application of old technologies to the problem of maximizing the information content
obtained in earlier trials in order to ensure the greatest chance of conducting a new clinical trial with the
desired outcome. Bonate26 has reviewed applications for clinical trial simulation, and reported successful
evaluations by several researchers. Simulation is a useful tool for determining key aspects of study design
such as the appropriate doses for First-in-Humans trials, dose selection for proof-of-concept and pivotal
studies, study design, subject numbers, sample numbers, timing, and other factors. When designing a
clinical trial, it is important to ensure that sufficient information to estimate model parameters is
collected, while also ensuring that the schedule is not onerous. Although it has been referenced in the
Guidance to Industry,27 the collection of a single trough value from each subject is insufficient to estimate
parameters. A process referred to as “D-optimization” uses information from previous models to optimize
the numbers and timing of samples collected from subjects.28 Potential study designs can then be tested
using simulations to ensure appropriateness of the design.
As described by Miller et al.,29 clinical trial simulation is a part of the “Learn and Confirm” cycle of drug
development. Information from previously conducted studies can be used to simulate expected ranges of
responses for upcoming trials. Subsequently, information gathered in the new trial can be used to confirm
the model and potentially augment information provided by the model. With each cycle, the robustness
and suitability of the model becomes better established.
A hypothetical example of clinical trial simulation is provided in Figure 4. In panel a, the effect of weight
on clearance of a drug is provided. Based on the narrow confidence intervals for this trend, the effect is
well estimated and should be robust for simulation. A simulation of expected concentration–time profiles
after a 1-h infusion of a 10 mg/kg dose to neonates, infants, young children, older children, adolescents,
and adults is shown in Figure 4b. With this weight-based dose strategy, neonates and infants show a
concentration–time profile that is substantially lower than expected as compared to adults. When the data
are summarized into AUC values, it can be seen that the overall exposure levels in neonates, infants, and
young children are markedly different from those in adults. In this example, higher doses are required in
pediatric patients in order to obtain exposure levels comparable to those in adults. Therefore, a study that
is designed such that all subjects receive the same weight-based (mg/kg) dose regardless of age would be
unlikely to succeed in younger patients, for whom alternative dose recommendations would have to be
considered. It is common to find that weight-based dosing is an inappropriate dose metric for use in
children. This is because the relationship between weight and clearance is usually not linear, and weight-
based dosing does not take into account the extent of maturation of organs. 30
Figure 4
Simulated exposures for different age groups. (a) Relationship between clearance and weight. The solid
line denotes the mean relationship, and the upper and lower broken lines denote the upper and lower 95%
confidence intervals, respectively. (b) Simulation ...
Go to:
Regulatory Aspects
The US Food and Drug Administration (FDA), through the FDA Modernization Act of 199731 and the
FDA “effectiveness” guidance of 199832 allowed the use of exposure–response information in
combination with a single pivotal clinical trial as sufficient evidence of effectiveness. Although the use of
an exposure–response evaluation to replace a pivotal trial is not common, population PK modeling and
exposure–response evaluations are frequently used to support registration decisions and labeling. This is
because population PK modeling enables the identification of the sources of variability that ultimately
have an impact on both safety and efficacy. In particular, the FDA has acknowledged the use of
population modeling as being informative in extending information from adult indications to pediatric
indications.33In a recent review of the impact of population modeling,34 the authors evaluated 198
submissions from January 2000 through December 2008. The number of submissions wherein
pharmacometrics analyses were included increased sixfold over 9 years (from 45 submissions during the
5 years from 2000 to 2004 to 87 submissions during the 2 years 2007–2008). The impact of these
analyses on labeling decisions has also increased across all sections of the drug label. Among the 198
submissions surveyed, pharmacometrics analyses of 126 submissions (64%) contributed to drug approval
decisions, while those of 133 submissions (67%) contributed to labeling decisions. Modeling and
simulation also play a large role in personalized medicine.
Modeling and simulation also play a large role in personalized medicine. Personalized medicine aims to
provide more accurate predictions of individual responses to therapy based on the characteristics of the
individuals.35 Pharmacogenetics tests allow clinicians to individualize treatment, potentially improving
compliance because the medication and dosage are more likely to be safe and effective. The warfarin drug
label changes made in 2007 and 2010 provide a good example. These changes were based, in part, on
research conducted by the C-Path Institute and others.36 Work by Hamberg et al.37 on warfarin exposure
and response identified the CYP2C9 genotype and age as being predictive of exposure, and the VKORC1
genotype as being predictive of response. The authors showed the importance of CYP2C9 and VKORC1
genotypes and the patient's age for arriving at strategies to improve the success of warfarin therapy.
In a recent commentary on the impact of population modeling on regulatory decision making, Manolis
and Herold38 described three broad classifications of model-based evaluations: (i) those that are generally
well accepted, (ii) those that may be acceptable if justified, and (iii) those that are controversial.
Examples of the first category are:
The use of modeling and simulation for final recommendation of intermediate doses that were not
specifically tested in phase II/III trials or to bridge efficacy data across indications.
Modeling of phase II/III data to support regulatory claims (e.g., absence of suspected drug–drug
interactions, effect of pharmacogenetics on exposure).
Model-based inference as the “sole” evidence of efficacy/safety, or based on simulated data for
efficacy and safety (notwithstanding exceptional scenarios).
Reporting requirements
In 1999, Sun et al. published a detailed description of the general expectations by regulators for
submission of population modeling work.39 In addition, there are guidance documents from FDA27 and the
Committee for Medicinal Products for Human Use (CHMP) of the European Medicines Agency
(EMA)40 which should always be considered when conducting population modeling evaluations. In
general, a prespecified analysis plan is useful and should be included in the final report. Because of the
importance of the quality of the data in determining the modeling results, it is essential to spend the
necessary time to ensure that the data are of good quality, and to describe the methods used for data
merging and evaluation. Both the analysis plan and the report should describe all data editing procedures
that have been used to detect and correct errors, including the criteria used for declaring data unusable
(e.g., missing information on dates or times of doses or measurements). The rationale for declaring a data
point to be an outlier needs to be statistically convincing and should be specified in the analysis plan. The
methods used for handling concentrations below quantitation limits and missing covariate data must also
be specified.
A final report should be sufficiently descriptive so as to allow a reviewer to understand how the
conclusions were reached. The objectives of the analyses, the hypotheses being investigated, and the
assumptions imposed should be clearly stated, both in the analysis plan and in the report. The steps taken
to develop the population model should be clearly described. This can be done through the use of flow
charts or decision trees. The criteria and rationale for the model-building procedures adopted should be
specified. Often, one or more tables showing the models tested and summaries of the results of each
evaluation are also included to provide a clear description of the results and decision-making process.
The reliability and robustness of the results can be supported by generating standard diagnostic plots, key
parameter estimates and associated SE, and other metrics. A model that is appropriate for a specific
purpose (e.g., describing data) may or may not be appropriate for other purposes (such as simulation).
The objective of model qualification is to examine whether the model is suitable for the proposed
applications. For example, if the model is to be used for simulation and dosage recommendation, the
predictive performance of the model should be tested.
Go to:
Table 3
Learning vs. confirming by development stage
In a report in 2004, the FDA addressed the issue of decline in new drug submissions and escalating
development costs.45 The report indicated a need for applied scientific work to create new and better tools
to evaluate the safety and effectiveness of new products, in shorter time frames, with more certainty, and
at lower cost. The FDA has advocated model-based drug development as an approach to improving
knowledge management and decision making relating to drug development (in line with the “Learn and
Confirm” paradigm) and has taken an active role in encouraging model development for various
therapeutic areas.46 A recent review on model-based drug development by Lalonde et al.47 suggested that
prior information is often ignored when analyzing and interpreting results from the most recent clinical
trial. However, modeling allows data from different studies to be combined in a logical manner, based on
an understanding of the drug and the disease. The authors suggested that drug development can be viewed
as a model-building exercise, during which knowledge about a new compound is continuously updated
and used to inform decision making and optimize drug development strategy.
Resources
Modeling and simulation require investments in resources, because input is needed from several areas.
Input from the clinical team is essential for the design of the protocol including implementation and
monitoring, so as to ensure that the necessary data are collected. The creation of population modeling
databases usually involves assistance from either database management staff or statistics staff. Database
preparation calls for special attention because it is important to have the exact times and dates for all
doses and measurements/observations. The results of the evaluation should be available sufficiently early
so that the information can either be used in new clinical trials or included in the filing. It may be helpful
to use preliminary data to meet important timelines, but the risks of using data that are not final should be
weighed and considered.
The generating of a model usually requires the inputs of an analyst, and because the science changes
continually, analysts should have their training updated regularly. Interpretation of the results may require
input from clinical staff. The results of any modeling evaluations should also be discussed within the
project team to ensure that the results are reasonable, understandable, and applicable to development
decisions.
The report must also be checked for accuracy and completeness. Depending on the size of the database
(the number of subjects and the number of observations per subject) and the complexity of the model, the
process of development, qualification, and report generation for a model can take many weeks to
complete.
Go to:
However, the selection and installation of the modeling software are not the only prerequisites for
conducting population modeling. In many cases, a supporting programming language is necessary to run
the modeling package (e.g., NONMEM requires Fortran). Because some modeling packages do not have
user-friendly interfaces, “front-end” software may be needed (for example, there are several of these
available for NONMEM, free of cost or for commercial licensing). Similarly, “back-end” packages for
generating graphical outputs of modeling results, along with supporting languages, may also be
necessary. It should also be noted that the analysts themselves should have appropriate experience,
education and/or training. User-written model codes, subroutines, and scripts should also be provided for
review as part of a regulatory submission.
User training
Modelers have a wide variety of backgrounds, including medicine, pharmacy, pharmacology, biophysics,
engineering, and statistics. Given the complexity of population modeling approaches, user training is as
important as ensuring software functionality. Unfortunately, the method of determining whether a user
has sufficient education, training, and experience to conduct these assessments is not clearly defined.
Many universities have training programs in population modeling, but the curriculum content and hands-
on experience available to students vary substantially. Similarly, there are numerous postgraduate training
courses, but these generally focus on introductory training, and users may require further training or
mentoring before undertaking an analysis. Continuing education through courses, meetings, and other
forums is important to ensure that analysts are familiar with new concepts and approaches.
Go to:
Conclusions
There is no doubt that the use of model-based approaches for drug development and for maximizing the
clinical potential of drugs is a complex and evolving field. The process of gaining knowledge in the area
is continuous for all participants, regardless of their levels of expertise. The inclusion of population
modeling in drug development requires allotment of adequate resources, sufficient training, and clear
communication of expectations and results. For one who is approaching the field for the first time, it can
be intimidating and confusing. A wise approach is to break the task into manageable pieces (“divide and
conquer”). One should try to understand one topic or master one piece of software at a time, seek
literature and training appropriate for one's level and needs and, most importantly seek the advice of
mentors and develop sources for collaboration and support.