You are on page 1of 10

Citation: CPT Pharmacometrics Syst. Pharmacol. (2019) 8, 97–106;  doi:10.1002/psp4.

12378

ARTICLE

A General Framework for Assessing In vitro/In vivo


Correlation as a Tool for Maximizing the Benefit-­Risk Ratio of
a Treatment Using a Convolution-­Based Modeling Approach
Roberto Gomeni1,* , Lanyan (Lucy) Fang2, Françoise Bressolle-Gomeni1, Thomas J. Spencer3, Stephen V. Faraone4 and
Andrew Babiskin2

The net benefit of a treatment can be defined by the relationship between clinical improvement and risk of adverse events:
the benefit-­risk ratio. The optimization of the benefit-­risk ratio can be achieved by identifying the most adequate dose (and/
or dosage regimen) jointly with the best-­performing in vivo release properties of a drug. A general in silico tool is presented
for identifying the dose, the in vitro and the in vivo release properties that maximize the benefit-­risk ratio using convolution-­
based modeling, an exposure-­response model, and a surface response analysis. A case study is presented to illustrate how
the benefit-­risk ratio of methylphenidate for the treatment of attention deficit hyperactivity disorder can be maximized
using the proposed strategy. The results of the analysis identified the characteristics of an optimized dose and in vitro/in
vivo release suitable to provide a sustained clinical response with respect to the conventional dosage regimen and
formulations.

Study Highlights

WHAT IS THE CURRENT KNOWLEDGE ON THE TOPIC? WHAT DOES THIS STUDY ADD TO OUR KNOWLEDGE?
✔  The development of a model-­ informed approach is ✔  A general in silico tool is presented for identifying the
currently extensively used for supporting drug develop- dose, the in vitro and the in vivo release properties that
ment. However, no quantitative framework has been pro- maximize the benefit-­risk ratio using convolution-­based
posed for identifying the best performing in vivo delivery modeling, an exposure-­response model, and a surface
rate appropriate for maximizing the benefit-­risk ratio and response analysis.
for facilitating the development of such a formulation HOW MIGHT THIS CHANGE DRUG DISCOVERY,
using in vitro/in vivo correlation. DEVELOPMENT, AND/OR THERAPEUTICS?
WHAT QUESTION DID THIS STUDY ADDRESS? ✔  The proposed model-­informed approach provides the
✔  How a model-­ informed drug development strategy pharmaceutical companies with a methodological frame-
based on surface-­response analysis can be prospectively work for developing drugs with drug delivery and a dose
developed and applied to optimize the drug development selection suitable to produce a clinical benefit prospec-
process by identifying the drug properties associated tively defined by the clinicians and not just a clinical re-
with an optimized benefit-­risk. sponse better than the placebo response.

The clinical benefit (CB) of a treatment is usually defined in The convolution-­ based modeling approach has been
relation to the size of the treatment effect, such as the base- proposed as a tool for optimizing the CB of a pharmaco-
line corrected change from placebo at study end. logical treatment.1 The CB optimization can be achieved by
However, the level of efficacy cannot be considered as identifying the best performing dose and dosage regimen
the unique criterion for determining the CB of a treatment as jointly with the best performing in vivo release properties of
the safety and tolerability information need to be accounted the drug.
for. A better definition of the CB of a pharmacological treat- The key components of this model-­informed approach
ment should be based on the concept of benefit-­risk ratio. are: (i) the assessment of the in vivo/in vitro correlation
Accordingly, CB can be defined as a treatment effect with (IVIVC), (ii) the characterization of the pharmacokinetics
a size sufficiently large to constitute a real clinical improve- (PKs) of the drug, (iii) the model linking circulating drug
ment with a minimal risk of adverse events (AEs). ­exposure and pharmacological response accounting for the

1
Pharmacometrica, La Fouillade, France; 2Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug
Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA; 3Massachusetts General Hospital, Boston, Massachusetts, USA; 4 SUNY
Upstate Medical University, Syracuse, New York, USA. *Correspondence: Roberto Gomeni (roberto.gomeni@pharmacometrica.com)
Received: October 5, 2018; accepted: December 7, 2018; published online on February 05, 2019. doi:10.1002/psp4.12378
General Framework for Assessing IVIVC as a Tool
Gomeni et al.
98

Connect the in vitro dissolution


In-vitro
experiments with the in vivo drug release
IVIVC

In-vivo release
process Describe the drug PK as a
function of the in vivo drug
release and the In vivo
In-vivo
disposition/elimination
disposition
n processes
Pharmacokinetics

Exposure- Describe the PD


response response as a
Pharmacodynamics relationship
function of the PK
Relationship
Optimize the between
response and
Pharmacotherapy benefit-risk ratio clinical Benefit

Figure 1  General framework for maximizing the benefit-­risk ratio of a treatment. PD, pharmacodynamic; PK, pharmacokinetic; IVIVC,
in vitro/in vivo correlation.

placebo effect, and (iv) the optimization tool to determine (iii) provide an in silico tool for identifying dose and in vitro
the optimal dose and in vivo drug release. release properties that maximize the benefit-­risk ratio of a
The IVIVC can be used: (i) to predict human absorption treatment.
and the in vivo plasma concentration profiles based on in A case study is presented to illustrate the implementation
vitro data, (ii) to optimize dosage forms, (iii) to set dissolu- of the proposed strategy for maximizing the benefit-­risk ratio
tion acceptance criteria, and (iv) to establish bioequivalence of methylphenidate hydrochloride (MPH) for the treatment of
based on in vitro data. attention deficit hyperactivity disorder (ADHD).
Many approaches have been proposed for evaluating MPH is a central nervous system stimulant currently con-
IVIVC. These methods can be classified as methods requir- sidered as the first-­line treatment for school-­aged children or
ing deconvolution and methods based on the implemen- adolescents with ADHD. A variety of extended-­release for-
tation of a convolution-­ based approach. Concerns have mulations of MPH have been developed in the attempt to
been raised about the reliability of traditional deconvolution-­ improve symptom control. Among these products, we focus
based methods.2,3 In those approaches, modeling is con- on Ritalin LA (Manufactured for Novartis Pharmaceuticals
ducted on deconvoluted data rather than on raw plasma Corporation East Hanover, New Jersey 07936 By Alkermes
drug concentration-­ time data. This analysis strategy fre- Gainesville LLC Gainesville, GA 30504). This formulation is an
quently leads to unstable and unreliable results.4 In contrast, extended-­release capsule with a bimodal release profile (with
convolution-­based methods provide more consistent and two distinct peaks ~ 4 hours apart) developed using a propri-
reliable results.5 etary Spheroidal Oral Drug Absorption System technology.7
One of the most efficient implementations of convolution-­ The most common AEs reported with MPH included head-
based modeling is based on the translation of a convolution aches, decreased appetite, nausea, irritability, and insom-
integral into a system of differential equations.6 This method nia.8,9 A recent meta-­analysis on the exposure-­response of
is a single-­stage approach as the model utilizes the ob- blood pressure (BP) and heart rate (HR) for MPH in healthy
served data directly without transformation (i.e., through de- adults indicated that the BP and HR changes were directly re-
convolution). One of the benefits of this modeling approach lated and highly dependent on the MPH PK profiles.10 These
is the ability to directly predict the plasma concentration-­ safety issues associated with MPH treatment may compro-
time course resulting from a given in vivo input function. mise the treatment course of ADHD in children and also raise
The objective of this paper is to develop a model-­ parents’ concerns over them. For this reason, it is critical to
informed methodology for optimizing the drug development determine what could be the minimal exposure of MPH asso-
process by identifying drug and formulation properties ciated with a CB that maximizes the benefit-­risk ratio.
­associated with an optimized benefit-­risk ratio (Figure 1). Ritalin LA data extracted from the literature were used to
This was conducted in three steps to: (i) implement IVIVC implement an IVIVC.11 The PK/PD model previously devel-
using a convolution-­ based approach applicable when a oped to establish the exposure-­response of MPH was used
parametric form of dissolution data can be identified and to estimate the changes in Swanson, Kotkin, Agler, M-­Flynn,
when in vivo PK is characterized by any parametric model, and Pelham scale (SKAMP) clinical scores associated with a
(ii) evaluate the impact of dose and of in vivo release prop- single dose of 40 mg of Ritalin LA.12 Finally, the optimization
erties on PK and on pharmacodynamic (PD) outcomes, and algorithm was applied to identify the dose and the in vivo/in

CPT: Pharmacometrics & Systems Pharmacology


General Framework for Assessing IVIVC as a Tool
Gomeni et al.
99

2 1
(t) = f pk(t,
(t) = fvitro(t,

f(t) I(t)
CPref = A/V
pPK = kel, V

Fixed parameters: pdiss Fixed parameters: pPK


Cp = A/V

Estimated parameters: pscal, F

Model validation
4

Figure 2  Schematic of the in vitro/in vivo correlation (IVIVC) analysis: rvitro = fraction of drug dissolved; rvivo = fraction of drug absorbed;
f(t) = in vivo input function; I(t) = unitary impulse response; V = volume of distribution; kel = first order elimination rate; * = convolution
operator; A = amount of drug; Cp = drug concentration; F = relative bioavailability; pdiss = dissolution function; pscal = time scaling
function;  = relative time change for the calculation for numerical approximation of f(t); t” = time scaled; pPK = pharmacokinetic
parameters of the immediate release formulation; Cpref = plasma concentration of the immediate release formulation.

vitro release properties expected to maximize the benefit-­ may be directly superimposable or may be made to be su-
risk ratio of a treatment with MPH.1 perimposable by using a “scaling factor.” A time-­scaling
function was included in the model to account for potential
METHODS time differences in the in vitro and the in vivo processes
Data (for example, when the dissolution is faster than the in vivo
The original study enrolled 17 participants in a four-­ input rate). A general time-­scaling model was applied in the
treatment, four-­period, single-­dose, randomized crossover assessment of the IVIVC:
design study, but only 16 subjects were retained for the
final PK analysis.11 The subjects received four treatments, rvivo (t) = a1 + a2 ⋅ rvitro (tt)
(3)
after at least a 10-­hour fast, 3 Ritalin LA capsule formula- tt = b1 + b2 ⋅ t b3
tions (40 mg) and Ritalin IR tablets (two 20 mg given 4 hours
apart) as the reference. The three Ritalin LA formulations In case of absence of time scaling between rvivo and rvitro:
were selected to provide slow-­ release, medium-­ release, a1 = 0, a2 = 1, b1 = 0, b2 = 1, and b3 = 1. Otherwise, the time
and fast-­release in vitro dissolution rates, respectively. scaling can be defined by estimating the appropriate val-
ues of the parameters a1, a2, b1, b2, and b3. Eq. 3 includes
Convolution-­based model a linear component (intercept of a1 and slope of a2) and a
The plasma concentration, resulting from an arbitrary dose, nonlinear component describing the time-­shifting (b1), time-­
was described by convolution as: scaling (b2), and time-­shaping factor (b3).
The final step in the IVIVC analysis is to validate the model
Cp (t) = f (t) ∗ I(t) (1) by providing quantified evidence of the predictive perfor-
mance of the model. The model validation can be accom-
dr dA dr A plished using data from the formulations used to build the
f (t) = ; = F ⋅ Dose ⋅ − kel ⋅ A; Cp = (2)
dt dt dt V model (internal validation) or using data obtained from a
­different (new) formulation (external validation).
where f(t) is the in vivo input function, I(t) is the unitary The internal validation is implemented using the IVIVC
impulse response (defined, for a one-­
­ compartment pro- model: the relevant exposure parameters (peak plasma con-
cess, by the volume of distribution (V) and the first order centration (Cmax) and area under the curve to infinity (AUC∞))
elimination rate (kel)), * is the convolution operator, r(t) is the are predicted using the model for each formulation and com-
time-­varying fraction of the dose released, A is the amount pared with the observed values. The prediction error (%PE)
of drug, and F is the fraction of the dose absorbed. is calculated for each PK parameter using the equation
n
IVIVC modeling 1 ∑ |Observed value − Predicted value|
%PE = ⋅ 100 (4)
The present analysis is focused on the development of a n 1 Observed value
level A IVIVC by evaluating a point-­to-­point correlation be-
tween the fraction of drug absorbed in vivo (r vivo (t)) and where n is the number of formulations. The criteria for as-
the fraction of drug dissolved (rvitro (t)).13 In the IVIVC as- sessing the level of predictability are for each PK parame-
sessment, the in vitro dissolution and in vivo input curves ter average %PE ≤ 10% with no individual values > 15%. If

www.psp-journal.com
General Framework for Assessing IVIVC as a Tool
Gomeni et al.
100

criteria are not met, the evaluation of external predictability dA A (8)


= f (t) − kel ⋅ A = F ⋅ Dose ⋅ ka ⋅ e−ka ⋅t − kel ⋅ A; Cp =
would be required.13 dt V
In addition to AUC∞ and Cmax, additional metrics based on
In the analysis of MPH:
the concept of partial AUC were considered for the assessment
of %PE. These criteria were based on the March 2015 rec-
ommendations of the US Food and Drug Administration (FDA) • The Ritalin LA multiphase in vitro dissolution data were
for using partial AUC (pAUC) metrics for studies conducted in described by a double Weibull function rvitro(t):
fasting conditions to assess the bioequivalence of generic ex- [ ( (( )ss )
(( )ss1 ) )]
tended release formulations of MPH against Ritalin LA.14 time time
− − td1
rvitro (t) = Dose ⋅ 1 − ff ⋅ e td + (1 − ff) ⋅ e (9)
The following pAUCs were considered:

• pAUC0−3, AUC from 0−3 hours where ff = fraction of the dose released in the first pro-
• pAUC3−7, AUC from 3−7 hours cess, td and td1 =  times to release 63.2% of the dose
• pAUC7−12, AUC from 7−12 hours in the first and in the second process, ss and ss1 =  sig-
moidicity factors for the first and the second process.15
The IVIVC analysis was conducted using a four-­
step • The f(t) function was approximated using a finite differ-
­approach, as illustrated in Figure 2: ence approach
• The Ritalin IR PK was described by a one-compartment
1. Fit the mean PK time-course of the Ritalin IR for- model with first order elimination and an absorption lag-time
mulation (step 1) • The final convolution model was defined by:
2. Individually fit the mean in vitro dissolution data of the
slow, medium, and fast formulations using the release rvivo (t) = a1 + a2 ⋅ rvitro (tt); tt = b1 + b2 ⋅ t b3 (10)
function (r(t)) (step 2)
3. Evaluate IVIVC by jointly applying the convolution drvivo rvivo (t − Δ) − rvivo (t + Δ)
model to the in vivo data of the slow, medium, and fast f (t) = ≅ (11)
dt 2⋅Δ
formulations (step 3) and by:
i. Fixing the in vivo drug release parameters for each dA A
formulation to the values estimated in step 2
= Fi ⋅ Dosei ⋅ fi (t) − kel ⋅ A; Cp = (12)
dt V
ii. Estimating the time-scaling factors common to all
formulations (Eq. 3) where i represents the formulation, F the relative bioavail-
4. Evaluate the internal predictability by comparing the ability, and dose the in vivo dose.
predicted (estimated in step 3) Cmax, AUC∞, pAUC0−3,
pAUC3−7, and pAUC7−12 with the observed values Exposure-­response
(step 4) The assessment of the exposure-­ response relationship
was implemented using the PK/PD model previously de-
As an example, in case of a simple in vitro dissolu- veloped describing the relationship of MPH concentrations
tion and in vivo disposition (i.e., one-­
compartment), the (resulting from the administration of Concerta, Janssen-
convolution-­
based model describing plasma concentra- Cilag Manufacturing, LLC Gurabo, Puerto Rico 00778) with
tions can be written as: the SKAMP scores describing the ADHD impairment.12
The placebo response evaluated with the SKAMP scores
• In vitro dissolution, constant dissolution rate: was described by an indirect response model (P(t)):

dP
rvitro = Dose ⋅ (1 − e−ka ⋅t ) (5) = kin ⋅ (1 + AA ⋅ (e−t⋅P1 − e−t⋅P2 )) − kout P (13)
dt

where ka is the first order absorption rate constant where kin = zero-­order rate constant for production of re-
• PK disposition, one-compartment with first order elimi- sponse (P), kout = first-­order rate constant for the loss of re-
nation rate: sponse; AA = amplitude of the placebo effect, and P1 and
P2 =  rate of onset and offset of placebo effect.
dA The MPH effect was described by a drug-­related change
= −kel ⋅ A (6)
dt from placebo using a maximum effect (Emax) model. The Emax
represented the maximal achievable MPH-­related effect. The
with the assumption that rvivo = rvitro, the in vivo drug de- tolerance effect was included in the model assuming that
livery rate is computed (by numerical approximation or by half-­maximal effective concentration (EC50) increases with
analytical solution) as: time.16 Finally, a Delta parameter accounted for the score
difference at baseline between the placebo and active arms:
drvivo rvitro (t − Δ) − rvitro (t + Δ)
f (t) = ≅ = Dose ⋅ ka ⋅ e−ka ⋅t (7)
dt 2⋅Δ
Emax ⋅ Cp
PD(t) = P(t) + Delta −
EC50 (t) + Cp (14)
• Convolution model:

CPT: Pharmacometrics & Systems Pharmacology


General Framework for Assessing IVIVC as a Tool
Gomeni et al.
101

Benefit/risk parameters Convolution-based


• Dose PK Model
• In-vitro dissolution rate
PK/PD Model Benefit/risk

Benefit/risk
optimizer

Figure 3  Schematic of the benefit-­risk analysis. PD, pharmacodynamic; PK, pharmacokinetic.

A time-­dependent EC50 (EC50(t)) value was used to The analyses were conducted using NONMEM, version
­account for tachyphylaxis: 7.4 (ICON Development Solutions, Hanover, MD, USA).
NONMEM model code is provided in the supplementary
( )
t ga information. Simulations and optimization of CB were con-
EC50 (t) = EC50b 1+ (15)
t50ga + t ga ducted in R using the NLOPTR nonlinear-­optimization l­ ibrary,
version 3.2.5 (R Foundation for Statistical Computing).
where EC50b represents the EC50 value at time 0, t50 is the
time at which half of the tachyphylaxis occurs (the time at RESULTS
which 50% of the maximal change in EC50(t) is reached), and Step 1: ritalin IR analysis
ga is the rate of change of the EC50(t). The disposition and elimination of the Ritalin IR formulation
was characterized by a one-­compartment model with ka,
Optimize the benefit-­risk absorption lag time (Lag), kel, and V. The model was fitted
The optimization of CB as a function of dose and in vivo to data assuming an additive residual model. The model
release (defined by: ff, ss, ss1, td, and td1) was conducted was selected based on the available data and on the prior
using a response surface analysis (Figure 3). The optimal ­models developed for describing the Ritalin IR PK.17
benefit-­risk ratio was defined by the minimal MPH exposure The estimated parameter values are presented in Table S1.
providing a constant and sustained improvement in the The mean MPH concentrations for the Ritalin IR formulation
SKAMP scores during all day. The assumption underlying with the model-­predicted curve are presented in Figure S1.
this definition was that the increase in the risk of AEs and
the risk of elevation of the cardiovascular parameters was Step 2: dissolution data analysis
proportional to the MPH exposure. Details of dissolution testing procedures can be found in
For the analysis, the maintenance of the SKAMP scores Wang et al.11 The Weibull model was individually fitted to
from 8−10 during 12 hours was arbitrarily considered as the the mean in vitro dissolution data of the slow, medium, and
target clinical response. fast formulations.
The optimization algorithm searched for the minimal value The estimated parameter values are presented in Table
of the cumulative time during which the SKAMP scores fail S2. The mean dissolution data for each formulation with the
to comply with the CB definition as a function of the dose, model predictions are presented in Figure 4. These results
and the ff, ss, ss1, td, and td1 parameters. indicated that the dissolution model well characterizes the
The minimization of this criterion is equivalent to maximiz- dissolution data for each formulation.
ing the time during which the SKAMP scores are compliant The time requested to dissolve the first fraction of the
with the CB definition. dose (td) is similar for the three formulations, whereas
the time necessary for dissolving the second fraction of the
Software dose (td1) seems to decrease proportionally from the slow
The data used in the analyses were extracted from the (9.91-­hour) formulation to the fast (5.42-­hour) formulation.
­publication of Wang et al.11 using Plot Digitizer s­ oftware, Similarly, the shape of the dissolution (ss1) decreases pro-
version 2.0 (University of South Alabama, Mobile, AL, USA). portionally from the slow (3.59) to the fast (1.75) formulation.

www.psp-journal.com
General Framework for Assessing IVIVC as a Tool
Gomeni et al.
102

Figure 4  Mean dissolution data for the slow, medium, and fast formulations (dots) with the model-­predicted dissolution curves
(solid lines).

Figure 5  Mean pharmacokinetic observed concentrations (orange dots) with the predicted values by the convolution model (blue
solid lines) by formulation. The shaded areas represent the 80%, 90%, and 95% prediction intervals. CI, confidence interval.

Step 3: IVIVC the absorption was purely driven by the in vitro release
The MPH concentrations of the three formulations were time-­scaled properties. Two analysis scenarios were con-
jointly fitted to estimate the time-­ scaling parameters sidered: in the first one, three distinct F_ parameters were
(a1, a 2, b1, b2, and b3) and the fraction of the dose ad- estimated, and in the second one, the same F_value was
ministered available for the systemic circulation (F_slow, assumed for the three formulations. The comparison of
F_medium, and F_fast). The F_ parameters represent a the change in the objective function value using the log-­
dose-­scaling value with respect to the reference Ritalin likelihood ratio test in the two scenarios indicated that
IR formulation. A value < 1 indicates that a formulation is the assumption of a common F_value was the preferred
expected to have a lower relative bioavailability than the option.
Ritalin IR formulation. The estimated parameter values are presented in Table
The time-­scaling parameters were fixed to a common S3. The mean observed and model-­ predicted MPH in
value for all formulations and estimated as fixed-­ effect vivo concentrations for the three formulations with the
parameters. No random effect was associated with any 80%, 90%, and 95% prediction intervals are presented in
parameter of the convolution model. In this framework, Figure 5.

CPT: Pharmacometrics & Systems Pharmacology


General Framework for Assessing IVIVC as a Tool
Gomeni et al.
103

Table 1  Comparison of the Cmax, AUC∞, pAUC0−3, pAUC3−7, and pAUC7−12 values estimated on the observed MPH PK data with the values estimated using
the convolution model predicted PK data with the assessment of the %PE

Observed values Predicted values Prediction error

%PE %PE
Formulation Cmax (ng/mL) AUC∞ (ng·hour/mL) Cmax (ng/mL) AUC∞ (ng·hour/mL) AUC Cmax

Slow 13.01 126.97 14.12 122.94 3.17 8.56


Medium 14.02 130.56 14.6 123.77 5.2 4.08
Fast 14.67 131.29 14.7 123.47 5.96 0.2
Average 4.78 4.28

pAUC 0−3 pAUC3−7 pAUC7−12 pAUC 0−3 pAUC3−7 pAUC7−12 %PE %PE %PE
Formulation (ng·hour/mL) (ng·hour/mL) (ng·hour/mL) (ng·hour/mL) (ng·hour/mL) (ng·hour/mL) pAUC 0−3 pAUC3−7 pAUC7−12
Slow 25.83 44.69 25.66 25.49 40.98 27.43 1.28 8.3 6.87
Medium 27.54 46.7 25.32 26.95 47.2 23.9 2.13 1.09 5.6
Fast 27.57 53.26 35.15 26.41 53.39 31.22 4.19 0.25 11.19
Average 2.53 3.21 7.89
%PE, prediction error; AUC∞, area under the curve to infinity; Cmax, peak plasma concentration; MPH, methylphenidate hydrochloride; pAUC, partial area
under the curve; PK, pharmacokinetic.

Step 4: Validation in Figure 6b with the simulated MPH PK and SKAMP scores
The in vitro dissolution data and the convolution model at the dose of 10 mg/day of the reference formulation.
were used to predict in vivo PK profiles of the three for- The objective of the optimization was to estimate the MPH
mulations. The convolution model predictions were in close plasma concentration and the shape of the MPH PK time
agreement with the observed concentrations (Figure S2 course associated with a SKAMP response compliant with
and Table S4). The regression analysis on the predicted vs. the target CB. For the purpose of the present analysis, CB
observed values indicated that the intercept was not statis- was arbitrarily defined as sustained and constant SKAMP
tically different from 0 and that the slope was not statisti- score values in the range 8–10 during 12 hours.
cally different from 1 as the 95% confidence limits included The optimization was conducted considering MPH PK
0 for intercept and 1 for slope. and SKAMP scores at the dose of 40 mg of Ritalin LA as
The area under the PK curves was estimated using a log-­ reference. The results indicated that the dose of 10 mg/
linear trapezoidal rule. The %PEs were estimated for Cmax, day with a maximal MPH concentration of 3.38 ng/mL and
AUC∞, pAUC0−3, pAUC3−7, and pAUC7−12 (Table 1). The larg- an improved in vitro/in vivo drug release/absorption profile
est %PE (= 11.19%) was estimated for pAUC7−12 of the fast was sufficient to assure the target CB. The estimated dis-
formulation. The average %PE across the formulations were solution parameters with the in vitro dissolution profiles of
below the 10% reference value. These results support the the optimized and reference formulations are presented in
level A IVIVC. Figure S4. The comparison of the optimized dissolution
values jointly with the comparison of the reference and the
Exposure-­response optimal dissolution profiles presented in Figure S4 indi-
The change from placebo of the SKAMP scores associated cated that the optimal values of the dissolution parameters
with the MPH exposure expected after the treatment with remain in a domain that could be easily accommodated
Ritalin LA at the dose of 40 mg were estimated using the by the currently available pharmaceutical development
model defined in Eqs. 13–15 and the parameter values pre- technologies. One of the benefits of the present model-
viously estimated.12 ing framework is to facilitate the comparison of alterna-
Figure S3 shows the model-­predicted MPH plasma con- tive hypothetical test product performances and to provide
centrations and the associated model-­predicted trajectories quantitative criteria for selecting the best product to move
of the SKAMP scores for the three formulations. Simulations forward.
were conducted to estimate MPH exposure and SKAMP
scores in the range of the recommended doses of Ritalin LA: DISCUSSION
10 mg to 40 mg/day (Figure 6a).
The simulated response showed a similar shape of Although the benefit-­risk analysis is usually conducted by
SKAMP trajectories at different doses with an important jointly assessing both favorable (i.e., efficacy) and unfavor-
drop in the early part of the day followed by a constant loss able effects of a treatment (i.e., AEs), the present analysis
of response during the rest of the day. However, none of the of risk assessment was focused on the optimization of dose
doses seem to provide a constant and sustained response and formulation properties considered as drivers of efficacy
during all day consistent with the target expectation. and tolerability (i.e., BP and HR changes). The aim of the
The results of the joint optimization of the in vitro/in vivo methodology presented is to identify the best performing
drug release/absorption and the dose strength are presented dose and the best performing in vitro release properties of

www.psp-journal.com
General Framework for Assessing IVIVC as a Tool
Gomeni et al.
104

Figure 6  Simulation results: (a) Simulated Swanson, Kotkin, Alger, M-­Flynn, and Pelham (SKAMP) scores and methylphenidate
hydrochloride (MPH) exposure at the recommended doses of Ritalin LA: 10–40 mg/day. (b) Simulated SKAMP score) and MPH
exposure at the optimized in vitro/in vivo drug release/absorption, at the optimized dose of 10 mg/day. The shaded areas represent the
target region of the response: SKAMP score) in the range 8–10 during all day. CI, confidence interval.

a drug providing an improved CB while minimizing potential A convolution-­based modeling approach was proposed
safety issues. for facilitating the development of drug formulations with
The SKAMP scores show almost superimposable trajec- optimal in vivo release properties by using IVIVC. Finally, a
tories for the three formulations with a maximal drop from surface-­response analysis was used to identify the drug-­
baseline at about 1.5 hours postdose and with a constant related properties that could affect the CB of a treatment
deterioration in the response after this time. Wang et al.11 by connecting in vitro and in vivo drug release, in vivo drug
conducted a bioequivalence analysis among the three release to PK, and PK to PD. The overall objective was to
Ritalin LA formulations using primary PK parameters (Cmax, provide a quantitative framework facilitating the develop-
time of maximum plasma concentration (Tmax), AUC0−t, and ment of new medications with an improved benefit-­risk ratio.
AUC0−∞) and secondary PK parameters describing the bi- The evaluation of the benefit-­risk ratio of a new treatment
modal shape of the PK time course (Cmax 0–4, Tmax 0−4, and represents a critical milestone in the regulatory approval
AUC0−4 for the first peak and Cmax 4−8, Tmax 4−8, and AUC4−8 process. This assessment provides rational criteria for char-
for the second peak). Wang et al.11 concluded that the three acterizing the clinical relevance of a new medicine for the
formulations were bioequivalent despite a different shape of benefit of the patients accounting for potential safety risks.
the PK time courses. It should be noted that the criteria used Structured approaches have been proposed to formally
by Wang et al.11 to assess the bioequivalence of MPH for- address benefit-­risk given the multidimensional nature of the
mulations (Ritalin LA) are not the same as the current recom- data required for this assessment.18,19 More recently, quan-
mendations of the FDA.14 The current criteria indicate that titative approaches based on modeling and simulation were
bioequivalence can be assessed when the 90% confidence also proposed to formally assess the benefit-­risk ratio.20
intervals (CIs) of the geometric mean of the test/reference These methods utilize the exposure-­ response relation-
ratios for the metrics (Cmax, AUC0−3, AUC3−7, AUC7−12, and ship, assessed by modeling multiple end points related to
AUC0−∞) fall within the limits of 80−125%. efficacy and safety and trial simulation to assess benefit-­risk

CPT: Pharmacometrics & Systems Pharmacology


General Framework for Assessing IVIVC as a Tool
Gomeni et al.
105

across different dose and dosage regimens. In the context parameters is different between Ritalin IR and ER formula-
of regulatory approval of new drugs, the assessments drug tions and that a linear model (in the concentration range of
benefit-­risk represent a template for product reviews, as well 0–25 ng/mL) described the relationship between MPH and
as a criterion to support regulatory decisions. changes in cardiovascular parameters.
At variance of this approach, where the benefit-­risk ratio Furthermore, it was shown that MPH has the greatest ef-
is utilized to summarize the properties of new medications at fect on HR, followed by systolic blood pressure, and diastolic
the end of the drug development process, we are proposing blood pressure, with 1 ng/mL MPH concentration resulting in
a model-­informed methodology that can be prospectively a percentage increase of 0.33% for systolic blood pressure,
applied to optimize the drug development process by iden- 0.27% for diastolic blood pressure, and 0.98% for HR.
tifying the drug properties associated with an optimized In conclusion, the proposed model-­ informed approach
benefit-­risk ratio. provided an operational framework for the development of
A case study is presented to illustrate how the benefit-­risk formulations with optimized in vitro/in vivo releases and for the
ratio of MPH for the treatment of ADHD can be optimized. selection of the dose that delivers the minimal exposure as-
A large number of MPH formulations have been developed sociated with a CB matching the expectation of the clinicians.
(and are currently in development) with different rates of
drug delivery. The choice of the delivery rate was established Supporting Information. Supplementary information accompa-
on the basis of empirical approaches in the attempt: (i) to nies this paper on the CPT: Pharmacometrics & Systems Pharmacology
mimic the PK of some reference compounds or (ii) to explore website (www.psp-journal.com).
­alternative delivery mechanisms. In any case, none of these
approaches was developed having as a target the maximi- Figure S1. Mean MPH concentrations for the Ritalin IR formulation in
zation of the expected CB of a treatment as defined by a linear and log-­linear scale.
physician and as expected by the parents. On the basis of Figure S2. Regression analysis of the predicted concentration by the
this target, the paper provided a methodological framework convolution model vs. the observed concentrations (open circle) for the
to pharmaceutical companies for the development of formu- slow, medium, and fast formulations.
lations with drug delivery suitable to produce a CB expected Figure S3. Predicted MPH plasma concentrations (left panel) and the
by clinicians and not just a response better than placebo. associated trajectories of the SKAMP scores (right panel) for the slow,
For the present analysis, the target CB was arbitrarily de- medium, and fast formulations.
fined as constant SKAMP scores in the range 8–10 during Figure S4. Comparison of the in vitro dissolution profiles of the opti-
12 hours. The proposed algorithm can easily be generalized mized and the reference formulations.
to account for any alternative target CB. The optimization Table S1. Estimated MPH parameter values for the Ritalin IR formulation.
procedure was applied to identify the in vivo MPH delivery Table S2. Estimated dissolution data parameters for the slow, medium,
appropriate for attending this target using the MPH PK and and fast formulations (RSE).
the SKAMP scores at the dose of 40 mg of Ritalin LA as Table S3. Estimated parameter values in the convolution analysis.
reference. The results of the optimization indicated that the SE is the standard error and RSE is the relative standard error of the
dose of 10 mg/day with a maximal MPH concentration of parameters.
3.38 ng/mL and an improved in vitro/in vivo drug release/ Table S4. Results of the regression analysis of the predicted vs. the
absorption profile was sufficient to deliver the target CB. The observed concentrations.
presence of a tolerance effect for MPH represents a poten- Model code. NONMEM control stream (IVIVC_NONMEM_code.txt) for
tial issue associated with the optimized formulation resulting the IVIVC analysis.
in a low Cmax value. The proposed model accounted for a
time-­dependent loss of efficacy by providing an estimate of Acknowledgments.  Faraone is also supported by the K.G. Jebsen
the optimal MPH PK time course characterized by a dual Centre for Research on Neuropsychiatric Disorders, University of Bergen,
peaks shape with the second peak greater than the first one Bergen, Norway.
for counteracting the loss of efficacy due to a tachyphylaxis
effect (Figure 6b). Funding.  Funding for this article was possible, in part, by the US
These results indicated that a substantial reduction in the Food and Drug Adminstration (FDA) through grant 1U01FD005240-­02.
circulating MPH exposure can be associated with an opti-
mized MPH release and dose values. The dose expected to Conflict of Interest/Disclosures.  Disclosures within the last
deliver a clinically relevant benefit was estimated at 10 mg/ 36  months for Dr Thomas Spencer: Dr Thomas Spencer receives
day based on optimized in vivo delivery rate. At this dose, a ­research support or is a consultant from the following sources: Alcobra,
significant reduction of the maximal exposure is expected Enzymotec Ltd., Heptares, Impax, Ironshore, Lundbeck Shire Laboratories
with respect to the reference formulation: Cmax = 3.73 ng/ Inc., Sunovion, VayaPharma, the FDA, and the Department of Defense.
mL at the dose of 10 mg/day for the optimized formulation Consultant fees are paid to the MGH Clinical Trials Network and not directly
vs. Cmax = 3.96 ng/mL, 7.92 ng/mL, and 15.85 ng/mL at the to Dr Spencer. Dr Thomas Spencer is on an advisory board for the fol-
dose of 10, 20, and 40 mg/day for the reference formula- lowing pharmaceutical companies: Alcobra, he receives research support
tions, respectively. from Royalties and Licensing fees on copyrighted ADHD scales through
These data have to be considered in the context of the MGH Corporate Sponsored Research and Licensing. Dr. Spencer has a
recent meta-­ analysis on the relationship between MPH US Patent Application pending (Provisional Number 61/233,686), through
exposure and BP and HR changes. This meta-­ analysis MGH corporate licensing, on a method to prevent stimulant abuse. In the
showed that the effect of MPH exposure on cardiovascular past year, Dr Faraone received income, potential income, travel expenses,

www.psp-journal.com
General Framework for Assessing IVIVC as a Tool
Gomeni et al.
106

and/or research support from Lundbeck, Rhodes, Arbor, Pfizer, Ironshore, 10. Li, L. et al. Exposure-­response analyses of blood pressure and heart rate changes
for methylphenidate in healthy adults. J. Pharmacokinet. Pharmacodyn. 44, 245–
Shire, Akili Interactive Labs, CogCubed, Alcobra, VAYA Pharma, and
262 (2017).
NeuroLifeSciences. With his institution, he has US patent US20130217707 11. Wang, Y. et al. In vitro dissolution and in vivo oral absorption of methylphenidate
A1 for the use of sodium-­hydrogen exchange inhibitors in the treatment of from a bimodal release formulation in healthy volunteers. Biopharm. Drug Dispos.
ADHD. In the past year, R. Gomeni was consultant for Ironshore, Sunovion, 25, 91–98 (2004).
12. Gomeni, R., Bressolle-Gomeni, F., Spencer, T.J., Faraone, S.V., Fang, L. & Babiskin,
Supernus, and UCB. The other authors declare no conflict of interest. A. Model-­based approach for optimizing study design and clinical drug perfor-
mances of extended-­release formulations of methylphenidate for the treatment of
Author Contributions.  R.G., F.M.M.B.-­G., T.J.S., S.V.F., L.F., and ADHD. Clin. Pharmacol. Ther. 102, 951–960 (2017).
13. US Food and Drug Administration. Guidance for industry: extended release oral dosage
A.B. wrote the manuscript. R.G., F.M.M.B.-­G., T.J.S., S.V.F., and A.B. de- forms: development, evaluation, and application of in vitro/in vivo correlations <https://
signed the research. R.G. and F.M.M.B.-­G. performed the research. R.G. www.fda.gov/downloads/drugs/guidances/ucm070239.pdf > (1997).
and F.M.M.B.-­G. analyzed the data. 14. US Food and Drug Administration. Draft guidance on methylphenidate hydrochlo-
ride. <https://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinfor-
mation/guidances/ucm281454.pdf> (2015).
Disclaimer.  The views expressed in this article do not necessar- 15. Costa, P. & Sousa Lobo, J.M. Modeling and comparison of dissolution profiles. Eur.
ily reflect the official policies of the Department of Health and Human J. Pharm. Sci. 13, 123–133 (2001).
Services, nor does any mention of trade names, commercial practices, or 16. Kimko, H. et al. Population pharmacodynamic modeling of various extended-­
release formulations of methylphenidate in children with attention deficit hyper-
organization imply endorsement by the United States Government. activity disorder via meta-­analysis. J. Pharmacokinet. Pharmacodyn. 39, 161–176
(2012).
17. Adjei, A. et al. Single-­dose pharmacokinetics of methylphenidate extended-­release
1. Gomeni, R., Bressolle-Gomeni, F. & Fava, M. Response surface analysis and multiple layer beads administered as intact capsule or sprinkles versus methylphe-
nonlinear optimization algorithm for maximization of clinical drug performance: nidate immediate-­release tablets (Ritalin(®)) in healthy adult volunteers. J. Child.
application to extended-­release and long-­acting injectable paliperidone. J. Clin. Adolesc. Psychopharmacol. 24, 570–578 (2014).
Pharmacol. 56, 1296–1306 (2016). 18. European Medicines Agency. Benefit-risk methodology project Work package 2 report:
2. Dunne, A., Gaynor, C. & Davis, J. Deconvolution based approach for level A in vivo– applicability of current tools and processes for regulatory benefit-risk assessment
in vitro correlation modeling: statistical considerations. Clin. Res. Regul. Aff. 22, <http://www.ema.europa.eu/docs/en_GB/document_library/Report/2010/10/
1–14 (2005). WC500097750.pdf> (2010).
3. O’Hara, T., Hayes, S., Davis, J., Devane, J., Smart, T. & Dunne, A. In vivo-­in vitro 19. US Food and Drug Administration. Structured approach to benefit-risk assess-
correlation (IVIVC) modeling incorporating a convolution step. J. Pharmacokinet. ment in drug regulatory decision-making - draft PDUFA V Implementation Plan.
Pharmacodyn. 28, 277–298 (2001). <https://www.fda.gov/downloads/forindustry/userfees/prescriptiondruguserfee/
4. Wing, G.M. A Primer on Integral Equations of the First Kind—The Problem of ucm329758.pdf> (2013).
Deconvolution and Unfolding. (Society for Industrial and Applied Mathematics, 20. Bellanti, F., van Wijk, R.C., Danhof, M. & Della Pasqua, O. Integration of PKPD rela-
Philadelphia, PA, 1991). tionships into benefit-­risk analysis. Br. J. Clin. Pharmacol. 80, 979–991 (2015).
5. Gaynor, C., Dunne, A. & Davis, J. A comparison of the prediction accuracy of two
IVIVC modelling techniques. J. Pharm. Sci. 97, 3422–3432 (2008).
6. Buchwald, P. Direct, differential-­equation-­based in-­vitro-­in-­vivo correlation (IVIVC) © 2019 The Authors CPT: Pharmacometrics & Systems
method. J. Pharm. Pharmacol. 55, 495–504 (2003). Pharmacology published by Wiley Periodicals, Inc. on be-
7. Lyseng-Williamson, K.A. & Keating, G.M. Extended-­ release methylphenidate
half of the American Society for Clinical Pharmacology and
(Ritalin1 LA). Drugs 62, 2251–2259 (2002).
8. Buitelaar, J.K. et al. Safety and tolerability of flexible dosages of prolonged-­release Therapeutics. This is an open access article under the terms
OROS methylphenidate in adults with attention-­ deficit/hyperactivity disorder. of the Creative Commons Attribution-NonCommercial
Neuropsychiatr. Dis. Treat. 5, 457–466 (2009). License, which permits use, distribution and reproduction
9. Huss, M. et al. Methylphenidate hydrochloride modified-­release in adults with
attention deficit hyperactivity disorder: a randomized double-­ blind placebo-­ in any medium, provided the original work is properly cited
controlled trial. Adv. Ther. 31, 44–65 (2014). and is not used for commercial purposes.

CPT: Pharmacometrics & Systems Pharmacology

You might also like