scispace - formally typeset
Search or ask a question
Posted ContentDOI

A 3D brain unit model to further improve prediction of local drug distribution within the brain

01 Jul 2019-bioRxiv (Cold Spring Harbor Laboratory)-pp 688648
TL;DR: A 3D brain unit model is developed, which builds on the previous proof-of-concept 2Dbrain unit model, to understand the factors that govern local unbound and bound drug PK within the brain, and shows that all the above mentioned factors affect brain ECF PK in an interdependent manner.
Abstract: The development of drugs targeting the brain still faces a high failure rate. One of the reasons is a lack of quantitative understanding of the complex processes that govern the pharmacokinetics (PK) of a drug within the brain. While a number of models on drug distribution into and within the brain is available, none of these addresses the combination of factors that affect local drug concentrations in brain extracellular fluid (brain ECF). Here, we develop a 3D brain unit model, which builds on our previous proof-of-concept 2D brain unit model, to understand the factors that govern local unbound and bound drug PK within the brain. The 3D brain unit is a cube, in which the brain capillaries surround the brain ECF. Drug concentration-time profiles are described in both a blood-plasma-domain and a brain-ECF-domain by a set of differential equations. The model includes descriptions of blood plasma PK, transport through the blood-brain barrier (BBB), by passive transport via paracellular and trancellular routes, and by active transport, and drug binding kinetics. The impact of all these factors on ultimate local brain ECF unbound and bound drug concentrations is assessed. In this article we show that all the above mentioned factors affect brain ECF PK in an interdependent manner. This indicates that for a quantitative understanding of local drug concentrations within the brain ECF, interdependencies of all transport and binding processes should be understood. To that end, the 3D brain unit model is an excellent tool, and can be used to build a larger network of 3D brain units, in which the properties for each unit can be defined independently to reflect local differences in characteristics of the brain. Author summary Insights on how a drug distributes within the brain over both time and space are still limited. Here, we develop a ‘3D brain unit model’ in order to understand the factors that control drug concentrations within a small piece of brain tissue, the 3D brain unit. In one 3D brain unit, the brain capillaries, which are the smallest blood vessels of the brain, surround the brain extracellular fluid (ECF). The blood-brain barrier (BBB) is located between the brain capillaries and the brain ECF. The model describes the impact of brain capillary blood flow, transport across the BBB, diffusion, flow and drug binding on the distribution of a drug within the brain ECF. We distinguish between free (unbound) drug and drug that is bound to binding sites within the brain. We show that all of the above mentioned factors affect drug concentrations within brain ECF in an interdependent manner. The 3D brain unit model that we have developed is an excellent tool to increase our understanding of how local drug concentrations within the brain ECF are affected by brain transport and binding processes.

Summary (3 min read)

1 Introduction

  • The brain capillary bed is the major site of drug exchange between the blood and the brain.
  • The brain ECF bulk flow results from the generation of brain ECF by the BBB and drainage into the cerebrospinal fluid (CSF).
  • Here, the authors developed a 3D brain unit model, in which local brain drug distribution is explicitly taken into account.
  • Thereafter, drug distribution within the brain ECF is affected by diffusion, bulk flow and binding.

2 The 3D brain unit

  • The 3D brain unit represents the smallest piece of brain tissue that contains all physiological elements of the brain.
  • The segments of red rectangular boxes protruding from the vertices from the 3D brain unit are parts of brain capillaries from neigh- bouring units.
  • (vii) Drug within the blood plasma does not bind to blood plasma proteins.
  • Within the brain ECF, the authors formulate: Assumptions 2. (i) Drug within the brain ECF is transported by diffusion and brain ECF bulk flow.

2.1 Formulation of the 3D brain unit

  • There, xr, yr and zr are constants that represent the length of one unit and are defined as dcap+2r, with dcap the distance between the brain capillaries and r the brain capillary radius.
  • In one brain unit, the brain capillaries, the BBB and the brain ECF are represented by the subsets Upl�U, UBBB�U and UECF�U, respectively, such that U = Upl [ UBBB [ UECF.
  • Within Upl, the authors define Uin as the domain where the blood plasma, containing drug, enters the 3D brain unit from a feeding arteriole.
  • The authors focus on single oral administration but can also study other choices.

2.4 Boundary conditions

  • The authors formulate boundary conditions that describe the change in concentration of drug at the boundary between the blood-plasma-domain (Uok) and the brain-ECF-domain (UECF), hence at UBB as well as at the boundaries of the 3D brain unit (Upl\@ U, UECF\@U).
  • 4.1 Drug exchange between Upl and UECF.
  • The authors describe diffusive transport by the difference in drug concentrations in CECF and Cpl, multiplied by the BBB permeability, P. In PLOS ONE | https://doi.org/10.1371/journal.pone.0238397.
  • September 23, 2020 7 / 24 addition, the authors model active transport into and out of the brain ECF with Michaelis-Menten kinetics, as they are well established and match with most available data on parameters related to BBB active transport, similar to the approach of [6].
  • The authors use additional boundary con- ditions to describe the drug concentrations at the sides of the domain.

2.5 Model parameter values and units

  • The dimensions of the 3D brain unit are based on the properties of the rat brain.
  • The model is suitable for data from human or other species as well, but the authors have chosen for the rat as for this species most data is available.
  • In their model, the authors use Eqs (1)–(5) to describe drug concentration within the blood plasma, with boundary conditions described in Eqs (11)–(15).
  • The range of values the authors use for the parameters in the model as well as their units are given in Table 1 below.
  • The literature does not provide values on the kinetic parameters related to non-specific binding kinetics (B2max, k2on and k2off).

3 Model results

  • The authors study the distribution of a drug within the 3D brain unit by plotting its concentration- time profiles within the brain ECF (brain ECF PK).
  • In addition, the authors study the distribution of the drug within the 3D brain unit.
  • The authors first nondimensionalise the system of equations and boundary conditions by scaling all variables by a characteristic scale.
  • The model can easily be used to study a specific drug by choosing the parameter values that are specific for this drug, provided that parameter values for this drug are known.

3.1 The effect of the brain capillary blood flow velocity on brain ECF PK within the 3D brain unit

  • The impact of the brain capillary blood flow velocity, vblood, on brain ECF PK within the 3D brain unit is evaluated.
  • The total passive permeability, P, includes both transcellular and paracellular permeability.
  • When vblood = 0.5 (left), there are clear differences between Cpl in Uin (Distance = 0) and Cpl in the opposite corner (Distance = 150) at the time-points shown.

3.2 The effect of active transport on the drug concentrations within the brain ECF

  • Active transport kinetics are regulated by the maximal transport rate (Tm) and the concentration of drug needed to reach half of the maximal transport rate (Km), see section 2.4.1.
  • Fig 5 (top) reveals that an increased value of Tm-in correlates with increased concentrations of CECF.
  • The non-specific binding sites within the brain ECF become saturated with drug when Tm-in is sufficiently high (Tm-in = 100�10-7 μmol s-1).
  • Fig 6 reveals that Tm-out affects the time during which specific binding sites are saturated: the time at which B1 attains 90% max(B1) is smaller for a high value of Tm-out.

3.3 The effect of the brain capillary blood flow velocity in the presence of active transport

  • In section 3.1 the authors have shown that both the passive BBB permeability, P, and the brain capillary blood flow velocity, vblood, affect dug brain ECF PK in the absence of active transport.
  • If P is high, drug can easily flow across the BBB back into the brain ECF, following the concentration gradient between the blood plasma and the brain ECF, thereby countering the effect of Tm-out.
  • Values of CECF are given in the table in Fig 10c in order to show the differences within the 3D brain unit more clearly.
  • The table again (as in Figs 7, 8 and 9) shows that vblood and P affect the impact of Tm-in and Tm-out on CECF.

4 Discussion

  • This new model provides an important step towards more realis- tic features of the brain.
  • This enables us to more realistically predict the impact of the interplay of cerebral blood flow, BBB characteristics, brain ECF diffusion, brain ECF bulk flow and brain binding on drug distribution within the brain.
  • Assumption 1(vii) is not violated for drugs that do not bind plasma pro- teins.
  • To ensure the quality of a mathematical model, the model predictions are ideally compared to experimental data.

Did you find this useful? Give us your feedback

Figures (12)

Content maybe subject to copyright    Report

RESEARCH ARTICLE
A 3D brain unit model to further improve
prediction of local drug distribution within the
brain
Esme
´
e Vendel
ID
1
, Vivi Rottscha
¨
fer
1
*, Elizabeth C. M. de Lange
2
*
1 Mathematical Institute, Leiden University, Leiden, The Netherlands, 2 Leiden Academic Centre for Drug
Research, Leiden University, Leiden, The Netherlands
* vivi@math.leidenuniv.nl (VR); ecmdelange@lacdr.leidenuniv.nl (EL)
Abstract
The development of drugs targeting the brain still faces a high failure rate. One of the rea-
sons is a lack of quantitative understanding of the complex processes that govern the phar-
macokinetics (PK) of a drug within the brain. While a number of models on drug distribution
into and within the brain is available, none of these addresses the combination of factors
that affect local drug concentrations in brain extracellular fluid (brain ECF). Here, we develop
a 3D brain unit model, which builds on our previous proof-of-concept 2D brain unit model, to
understand the factors that govern local unbound and bound drug PK within the brain. The
3D brain unit is a cube, in which the brain capillaries surround the brain ECF. Drug concen-
tration-time profiles are described in both a blood-plasma-domain and a brain-ECF-domain
by a set of differential equations. The model includes descriptions of blood plasma PK,
transport through the blood-brain barrier (BBB), by passive transport via paracellular and
transcellular routes, and by active transport, and drug binding kinetics. The impact of all
these factors on ultimate local brain ECF unbound and bound drug concentrations is
assessed. In this article we show that all the above mentioned factors affect brain ECF PK in
an interdependent manner. This indicates that for a quantitative understanding of local drug
concentrations within the brain ECF, interdependencies of all transport and binding pro-
cesses should be understood. To that end, the 3D brain unit model is an excellent tool, and
can be used to build a larger network of 3D brain units, in which the properties for each unit
can be defined independently to reflect local differences in characteristics of the brain.
1 Introduction
The brain capillary bed is the major site of drug exchange between the blood and the brain.
Blood flows from the general blood circulation into the brain capillary bed by a feeding arteri-
ole and back by a draining venule. The rate at which drug molecules within the blood are
exposed to the brain is determined by the brain capillary blood flow rate. Drug exchange
between the blood plasma in the brain capillaries and the brain extracellular fluid (ECF) is con-
trolled by the blood-brain barrier (BBB).
PLOS ONE
PLOS ONE | https://doi.org/10.1371/journal.pone.0238397 September 23, 2020 1 / 24
a1111111111
a1111111111
a1111111111
a1111111111
a1111111111
OPEN ACCESS
Citation: Vendel E, Rottscha¨fer V, de Lange ECM
(2020) A 3D brain unit model to further improve
prediction of local drug distribution within the
brain. PLoS ONE 15(9): e0238397. https://doi.org/
10.1371/journal.pone.0238397
Editor: Stefan Liebner, Institute of Neurology
(Edinger-Institute), GERMANY
Received: March 20, 2020
Accepted: August 15, 2020
Published: September 23, 2020
Copyright: © 2020 Vendel et al. This is an open
access article distributed under the terms of the
Creative Commons Attribution License, which
permits unrestricted use, distribution, and
reproduction in any medium, provided the original
author and source are credited.
Data Availability Statement: All relevant data are
within the manuscript.
Funding: The authors received no specific funding
for this work.
Competing interests: The authors have declared
that no competing interests exist.

Drug distribution into and within the brain has been extensively summarized in a recent
review [1]. In short, the BBB has great impact on the relationship between the concentration-
time profiles of unbound drug in the blood plasma (blood plasma pharmacokinetics (PK)) and
in the brain ECF (brain ECF PK). The BBB consists of brain endothelial cells that are held
closely together by tight junctions. Unbound drug may cross the BBB by passive and/or active
transport [210]. Passive transport is bidirectional and occurs by diffusion through the BBB
endothelial cells (transcellular transport) and through the BBB tight junctions between the
endothelial cells (paracellular transport). Passive transport is quantified by the BBB permeabil-
ity, which is the speed by which a compound passively crosses the BBB, and depends on the
properties of both the drug and the brain. Active transporters located at the BBB move com-
pounds either inward (in the direction of the brain ECF, active efflux) or outward (in the direc-
tion of the blood plasma, active efflux). Once having crossed the BBB, drug distributes within
the brain ECF by diffusion. Diffusion within the brain ECF is hindered by the brain cells [11,
12]. This hindrance is described by the so-called tortuosity and leads to an effective diffusion
that is smaller than normal (in a medium without obstacles). Moreover, a fluid flow, the brain
ECF bulk flow, is present. The brain ECF bulk flow results from the generation of brain ECF
by the BBB and drainage into the cerebrospinal fluid (CSF). Both diffusion and brain ECF
bulk flow are important for the distribution of a drug to its target site, which is the site where a
drug exerts its effect. In order to do induce an effect, a drug needs to bind to specific binding
sites (targets). Only unbound drug, i.e. drug that is not bound to any components of the brain,
can interact with its target [13, 14]. This is a dynamic process of association and dissociation,
the so-called drug binding kinetics. These association and dissociation rates may affect the
concentration of unbound drug at the target site [15, 16]. While the drug dissociation rate has
been thought of as the most important determinant of the duration of interactions between
a drug and its binding site [17], a more recent study shows that the drug association rate is
equally important [16].
A number of models integrating several of the discussed processes of drug distribution into
and within the brain is available, see for example [11, 12, 1825] and [26]. The most recent and
comprehensive brain drug distribution model is the physiologically-based pharmacokinetic
model for the rat and for human [27, 28]. This model takes multiple compartments of the
central nervous system (CNS) into account, including plasma PK, passive paracellular and
transcellular BBB transport, active BBB transport, and distribution between the brain ECF,
intracellular spaces, and multiple CSF sites, on the basis of CNS-specific and drug-specific
parameters. However, it does not take into account distribution within brain tissue (brain
ECF).
Much is still unknown on the spatial distribution of a drug within the brain and quantita-
tive data on the processes governing brain spatial-temporal drug transport are lacking. The
purpose of the present study is therefore to gain insight into the processes governing spatial
drug distribution within the brain. Here, we developed a 3D brain unit model, in which local
brain drug distribution is explicitly taken into account. The 3D brain unit model encompasses
blood plasma PK, the BBB, brain ECF, brain ECF bulk flow, diffusion, and binding to specific
and non-specific binding sites [29, 30] within the brain. This 3D piece of brain tissue can be
considered the smallest physiological unit of the brain in terms of drug transport. Within the
3D brain unit, drug is carried along with the blood plasma by the brain capillary blood flow
and as such presented to the brain ECF. Drug distributes between the blood plasma and the
brain ECF by transport across the BBB. Thereafter, drug distribution within the brain ECF is
affected by diffusion, bulk flow and binding. We describe the distribution of drug within the
brain ECF by a partial differential equation (PDE) and couple this to two ordinary differential
equations (ODEs) to account for specific and non-specific drug binding.
PLOS ONE
A 3D brain unit model to further improve prediction of local drug distribution within the brain
PLOS ONE | https://doi.org/10.1371/journal.pone.0238397 September 23, 2020 2 / 24

The model builds on a proof-of-concept 2D brain unit model [31]. The 2D model is a basic
model covering many essential aspects of drug distribution within the brain, including passive
BBB transport, diffusion, brain ECF bulk flow, specific binding of a drug at its target site and
non-specific binding of a drug to components of the brain. Here, brain cells are implicitly
implemented by describing the hindrance the cells impose on the transport of a drug within
the brain ECF in a tortuosity term, λ. There, λ is defined as
ffiffiffi
D
D
p
, with D being the normal dif-
fusion coefficient and D
the effective diffusion coefficient [12]. The 2D brain unit model has
enabled the study of the effect of drug properties and brain tissue characteristics on the distri-
bution of a drug within the brain ECF and on its specific and non-specific binding behaviour
of the drug.
The current 3D brain unit model further improves the prediction of drug distribution
within the brain. The third dimension improves the realistic features of the model as the brain
is also 3D. Moreover, the third dimension allows the brain ECF to be not entirely surrounded
by capillaries, such that the brain ECF is a continuous medium, like in reality. Then, the brain
capillary blood flow and active transport across the BBB, which are both important mecha-
nisms of drug transport into the brain, are included. Here, we focus on one single brain unit.
This allows for a thorough characterisation of drug distribution within one 3D brain unit
before expanding to a larger scale.
In the remainder of this article, the mathematical representation of the characteristics of the
3D brain unit is introduced (section 2). There, we formulate the model (section 2.1) and the
mathematical descriptions of the drug distribution within the blood plasma of the brain
capillaries (section 2.2) and within the brain (section 2.3). In section 2.4 we formulate the
model boundary conditions that describe drug exchange between the blood plasma and the
brain ECF by passive and active BBB transport, as well as drug transport at the boundaries of
the unit. In section 3, we study the effect of several factors on drug distribution within the
brain ECF. In section 3.1, we evaluate the effect of the brain capillary blood flow velocity on
local brain ECF PK in the 3D brain unit. Next, we evaluate the effect of active influx and efflux
on local brain ECF PK (section 3.2). Then, in section 3.3 we show how the interplay between
the brain capillary blood flow velocity, passive BBB permeability and active transport affects
drug concentrations within the 3D brain unit. Finally, in section 4 we conclude our work and
discuss future perspectives.
2 The 3D brain unit
The 3D brain unit represents the smallest piece of brain tissue that contains all physiological
elements of the brain. The 3D brain unit is part of a larger network of 3D brain units, but here
we focus on just one 3D brain unit that is fed by an arteriole and drained by a venule (Fig 1,
left). The 3D brain unit is a cube in which the brain capillaries (represented by red rectangular
boxes on the ribs) surround the brain ECF (Fig 1, left). The segments of red rectangular boxes
protruding from the vertices from the 3D brain unit are parts of brain capillaries from neigh-
bouring units. As such, each vertex connects three incoming brain capillaries to three outgoing
brain capillaries, with the exception of the vertex connected to the arteriole and the vertex con-
nected to the venule. These connect the arteriole to three outgoing brain capillaries and three
incoming brain capillaries to the venule, respectively.
A single 3D brain unit (Fig 1, middle) has a blood-plasma-domain (red) consisting of multi-
ple sub-domains. These include the brain capillary domain where drug enters the unit (indi-
cated by U
in
in Fig 1), the domains representing the x-directed, y-directed and z-directed
brain capillaries (indicated by U
x1x4
, U
y1y4
and U
z1z4
in Fig 1) and the brain capillary
domain where drug leaves the unit (indicated by U
out
in Fig 1). Drug within the blood plasma
PLOS ONE
A 3D brain unit model to further improve prediction of local drug distribution within the brain
PLOS ONE | https://doi.org/10.1371/journal.pone.0238397 September 23, 2020 3 / 24

is transported by the brain capillary blood flow. The brain capillary blood flow splits at the ver-
tices of the unit, where brain capillary branching occurs (Fig 1, right).
In developing the model, we make the following assumptions about drug distribution
within the brain capillaries:
Assumptions 1.
(i) The drug concentration within the blood plasma changes as a function of time depending
on dose, bioavailability, the rate of absorption (in case of oral administration), distribution vol-
ume and elimination into and from the blood plasma.
(ii) The blood carrying the drug flows into 3D brain unit by a feeding arteriole and leaves via
a draining venule ( Fig 1, left).
(iii) The drugs enters the brain unit in the domain U
in
( Fig 1, middle), and drug concentra-
tions in U
in
are unaffected by the brain capillary blood flow.
(iv) The brain capillary blood flow is directed away from U
in
( Fig 1, right).
(v) In the blood plasma, drug transport by diffusion is negligible compared to drug transport
by the brain capillary blood flow.
(vi) The brain capillary blood flow velocity is by default equal in all brain capillaries.
(vii) Drug within the blood plasma does not bind to blood plasma proteins. All drug within
the blood plasma is in an unbound state and is able to cross the BBB.
Drug within the blood plasma of the brain capillaries crosses the BBB to exchange with the
brain ECF. The BBB is located at the border between the brain capillaries (red) and the brain
ECF (blue), see Fig 1. Drug exchange between the blood plasma and the brain ECF is described
by passive and active transport across the BBB in both directions. Here, we assume that active
influx transporters move a compound from the blood plasma directly into the brain ECF and
that active efflux transporters move a compound from the brain ECF directly into the blood
plasma.
Within the brain ECF, we formulate:
Assumptions 2.
(i) Drug within the brain ECF is transported by diffusion and brain ECF bulk flow.
(ii) Cells are not explicitly considered, but only by taking the tortuosity (hindrance on diffu-
sion imposed by the cells) into account.
Fig 1. Sketch of the 3D model brain unit. Left: The structure represented by the 3D brain unit. An arteriole carries
blood plasma (containing drug) into a brain capillary bed, that is connected to a venule that drains the blood plasma.
The brain capillaries (red) surround the brain ECF (blue). Middle: the 3D brain unit and its sub-domains. The unit
consists of a brain-ECF-domain (blue) and a blood-plasma-domain (red). The blood-plasma-domain is divided into
several subdomains: U
in
is the domain where the dose of absorbed drug enters the 3D brain unit, U
x1-x4
, U
y1-y4
and
U
z1-z4
are the domains representing the x-directed, y-directed and z-directed capillaries, respectively. Right: Directions
of transport in the model. The drug enters the brain capillaries in U
in
. From there, it is transported through the brain
capillaries by the brain capillary blood flow in the direction indicated by the small arrows. Drug in the brain capillary
blood plasma exchanges with the brain ECF by crossing the BBB. Drug within the brain ECF is, next to diffusion,
transported along with brain ECF bulk flow (indicated by the bold arrow).
https://doi.org/10.1371/journal.pone.0238397.g001
PLOS ONE
A 3D brain unit model to further improve prediction of local drug distribution within the brain
PLOS ONE | https://doi.org/10.1371/journal.pone.0238397 September 23, 2020 4 / 24

(iii) The brain ECF bulk flow is unidirectional. It is pointed in the x-direction, see the bold
arrow in Fig 1 (right).
(iv) All drug distributes within the brain ECF and we only have extracellular binding sites.
(v) The total concentration of specific and non-specific binding sites is constant.
(vi) The specific and non-specific binding sites are evenly distributed over the 3D brain unit
and do not change position.
(vii) The specific and non-specific binding sites lie on the outside of cells and the drug does not
have to cross cell membranes in order to bind to binding sites.
(viii) Drug binding is reversible and drugs associate and dissociate from their binding sites.
2.1 Formulation of the 3D brain unit
The 3D brain unit is a cubic domain, U, that represents a piece of brain tissue. We define U =
{(x,y,z) 2 R
3
j 0x x
r
^ 0yy
r
^ 0zz
r
}. There, x
r
, y
r
and z
r
are constants that represent
the length of one unit and are defined as d
cap
+2r, with d
cap
the distance between the brain
capillaries and r the brain capillary radius. In one brain unit, the brain capillaries, the BBB and
the brain ECF are represented by the subsets U
pl
U, U
BBB
U and U
ECF
U, respectively, such
that U = U
pl
[ U
BBB
[ U
ECF
.
Within U
pl
, we define U
in
as the domain where the blood plasma, containing drug, enters
the 3D brain unit from a feeding arteriole. We define U
out
as the domain where the blood
plasma, containing drug, leaves the 3D brain unit to a draining venule. Additionally, we define
the x-directed, y-directed and z-capillaries as the sets {U
xi
,i = 1,,4}, {U
yi
,i = 1,,4} and {U
zi
,
i = 1,,4}. The brain capillaries are divided by the lines x = y (or y = z or x = z) and x+y = y
r
(or
y+z = z
r
or x+z = z
r
), for which an example is shown in Fig 2. The only exceptions for this are
the brain capillaries adjacent to U
in
and U
out
, see below.
The definitions of the regions are as follows:
U
x1
= {(x,y,z) 2 U j rx<x
r
-y, rx<x
r
-z ^0y<r ^ 0z<r}
U
x2
= {(x,y,z) 2 U j y
r
-y<xy ^ zx<x
r
-z ^y
r
y>y
r
-r ^ 0z<r}
U
x3
= {(x,y,z) 2 U j yx<x
r
-y ^ z
r
-z<xz ^ 0y<r ^ z
r
z>z
r
-r}
U
x4
= {(x,y,z) 2 U j y
r
-y<xy ^ z
r
-z<xz ^ y
r
y>y
r
-r ^ z
r
z>z
r
-r}
U
y1
= {(x,y,z) 2 U j ry<y
r
-z ^ryy
r
x ^0x<r ^ 0z<r}
U
y2
= {(x,y,z) 2 U j zy<y
r
-z ^x
r
-xy<x ^ x
r
x>x
r
-r ^ 0z<r}
U
y3
= {(x,y,z) 2 U j z
r
-z<yz ^ x<yy
r
-x ^ 0x<r ^ z
r
z>z
r
-r}
U
y4
= {(x,y,z) 2 U j z
r
-zy<z ^ x
r
-x<yx ^ x
r
x>x
r
-r ^ z
r
z>z
r
-r}
U
z1
= {(x,y,z) 2 U j rzz
r
-x ^ rzz
r
-y ^ 0x<r ^ 0y<r}
U
z2
= {(x,y,z) 2 U j x<zz
r
-x ^y
r
-yz<y ^ 0x<r ^ y
r
y>y
r
-r}
U
z3
= {(x,y,z) 2 U j x
r
-xz<x ^ y<zz
r
-y ^x
r
x>x
r
-r ^ 0y<r}
U
z4
= {(x,y,z) 2 U j x
r
-xz<x ^ y
r
-yz<y ^ x
r
x>x
r
-r ^ y
r
y>y
r
-r}
U
in
= {(x,y,z) 2 U j 0x<r ^ 0y<r ^ 0z<r}
U
out
= {(x,y,z) 2 U j x
r
-rx<x
r
^ y
r
-ry<y
r
^ z
r
-rz<z
r
}.
The BBB is represented by a subset U
BBB
U, such that U
BBB
= @ U
pl
\@ U. This denotes the
border between the blood plasma and the brain ECF, located at distance r from the edges of
the 3D brain unit.
The brain ECF is represented by a subset U
ECF
U, such that U
ECF
= U\(U
pl
[U
BBB
).
Within U we define the following quantities describing drug concentration:
C
pl
(x,y,z,t): U
pl
x R
þ
! R
þ
;
C
ECF
(x,y,z,t): U
ECF
x R
þ
! R
þ
;
B
1
(x,y,z,t): U
ECF
x R
þ
! R
þ
;
B
2
(x,y,z,t): U
ECF
x R
þ
! R
þ
:
PLOS ONE
A 3D brain unit model to further improve prediction of local drug distribution within the brain
PLOS ONE | https://doi.org/10.1371/journal.pone.0238397 September 23, 2020 5 / 24

Citations
More filters
01 May 2012
TL;DR: In this article, the authors examined the dependence of efficacy on drug dose and elution duration of sirolimus-eluting stent implant and found that the persistence time of receptor saturation and effect is more sensitive to duration of elution than to eluted amount.
Abstract: Drug eluting stent designs abound and yet the dependence of efficacy on drug dose and elution duration remains unclear. We examined these issues within a mathematical framework of arterial drug distribution and receptor binding following stent elution. Model predictions that tissue content linearly tracks stent elution rate were validated in porcine coronary artery sirolimus-eluting stents implants. Arterial content varied for stent types, progressively declining from its Day 1 peak and tracking with rate-limiting drug elution--near zero-order release was three-fold more efficient at depositing drug in the stented lesion than near first-order release. In vivo data were consistent with an overabundance of non-specific sirolimus-binding sites relative to the specific receptors and to the delivered dose. The implication is that the persistence time of receptor saturation and effect is more sensitive to duration of elution than to eluted amount. Consequently, the eluted amount should be sufficiently high to saturate receptors at the target lesion, but dose escalation alone is an inefficient strategy for prolonging the duration of sirolimus deposition. Moreover, receptor saturating drug doses are predicted to be most efficacious when eluted from stents in a constant zero order fashion as this maximizes the duration of elution and receptor saturation.

5 citations

Journal ArticleDOI
TL;DR: This report summarizes the state of IVIVE-PBPK-linked models of brain and discusses shortcomings and areas of further research for better prediction of CNS drug disposition.
Abstract: Drug development for the central nervous system (CNS) is a complex endeavour with low success rates, as the structural complexity of the brain and specifically the blood-brain barrier (BBB) poses tremendous challenges. Several in vitro brain systems have been evaluated, but the ultimate use of these data in terms of translation to human brain concentration profiles remains to be fully developed. Thus, linking up in vitro-to-in vivo extrapolation (IVIVE) strategies to physiologically based pharmacokinetic (PBPK) models of brain is a useful effort that allows better prediction of drug concentrations in CNS components. Such models may overcome some known aspects of inter-species differences in CNS drug disposition. Required physiological (i.e. systems) parameters in the model are derived from quantitative values in each organ. However, due to the inability to directly measure brain concentrations in humans, compound-specific (drug) parameters are often obtained from in silico or in vitro studies. Such data are translated through IVIVE which could be also applied to preclinical in vivo observations. In such exercises, the limitations of the assays and inter-species differences should be adequately understood in order to verify these predictions with the observed concentration data. This report summarizes the state of IVIVE-PBPK-linked models and discusses shortcomings and areas of further research for better prediction of CNS drug disposition. Graphical abstract

4 citations

Journal ArticleDOI
TL;DR: In this paper , a Physiologically-Based Kinetic (PBK) model was developed for Tebufenpyrad in order to better evaluate uncertainties related to the development of such a model, harvest the performed work to support further improvements and potentially seek guidance for harmonization.
Abstract: Tebufenpyrad is a pesticide active substance used as insecticide, for which the mechanism of action is an inhibition Complex I of the Mitochondrial Electron Transport Chain (ETC). In vitro experimental data demonstrated that Tebufenpyrad-induced inhibition of ETC's complex I leads to oxidative damage, reduction of oxygen consumption rates with increasing in mitochondrial stress level and dysfunction in dopaminergic neuronal cells (Charli et al. 2016; Chen et al. 2017; Delp et al. 2021). This inhibition of Complex I has been included as a molecular initiating event (MIE) in an adverse outcome pathway (AOP) for Parkinsonian motor deficits. A set of in vitro mechanistic data are available along the AOP (Part 2 of this project) and their interpretation with regards to potential neurotoxicity need to estimate brain exposure to Tebufenpyrad in human, considering its intended uses as plant protection product. To this end, ANSES developed a Physiologically-Based Kinetic (PBK) model supporting several objectives. First objective explores the feasibility of PBK development for Tebufenpyrad when this is accomplished by means of NAMs only. Learnings from this case study will serve as ways to improve active substance dossiers for internal exposure assessment. Second objective relates to the possibility of interpreting the aforementioned AOP when in vitro data are produced on mitochondrial ETC inhibition and whether this inhibition may or may not occur in human too and therefore whether it is possible to discard neurological effects of Tebufenpyrad. This second objective is also intended to support Tebufenpyrad toxicological evaluation during its renewal process. The third objective is to better evaluate uncertainties related to the development of such PBK model, harvest the performed work to support further improvements and potentially seek guidance for harmonization.

1 citations

Journal ArticleDOI
TL;DR: In this article , the authors provide a comprehensive summary of the brain transport mechanisms and the currently available in vivo methods and mathematic models in studying the molecule delivery process at the BBB interface, including in vivo brain uptake measurement methods, in vitro BBB models, and mathematic brain vascular models.
Abstract: The blood-brain barrier (BBB) is a dynamic regulatory barrier at the interface of blood circulation and the brain parenchyma, which plays a critical role in protecting homeostasis in the central nervous system. However, it also significantly impedes drug delivery to the brain. Understanding the transport across BBB and brain distribution will facilitate the prediction of drug delivery efficiency and the development of new therapies. To date, various methods and models have been developed to study drug transport at the BBB interface, including in vivo brain uptake measurement methods, in vitro BBB models, and mathematic brain vascular models. Since the in vitro BBB models have been extensively reviewed elsewhere, we provide a comprehensive summary of the brain transport mechanisms and the currently available in vivo methods and mathematic models in studying the molecule delivery process at the BBB interface. In particular, we reviewed the emerging in vivo imaging techniques in observing drug transport across the BBB. We discussed the advantages and disadvantages associated with each model to serve as a guide for model selection in studying drug transport across the BBB. In summary, we envision future directions to improve the accuracy of mathematical models, establish noninvasive in vivo measurement techniques, and bridge the preclinical studies with clinical translation by taking the altered BBB physiological conditions into consideration. We believe these are critical in guiding new drug development and precise drug administration in brain disease treatment.
References
More filters
Journal ArticleDOI
TL;DR: The present results demonstrate that compounds often considered to be Pgp substrates in rodents with poor passive permeability may still exhibit reasonable CNS penetration in vivo; i.e., where the unbound fractions and nonspecific tissue binding act as a compensating force.
Abstract: This work examines the inter-relationship between the unbound drug fractions in blood and brain homogenate, passive membrane permeability, P-glycoprotein (Pgp) efflux ratio, and log octanol/water partition coefficients (cLogP) in determining the extent of central nervous system (CNS) penetration observed in vivo. The present results demonstrate that compounds often considered to be Pgp substrates in rodents (efflux ratio greater than 5 in multidrug resistant Madin-Darby canine kidney cells) with poor passive permeability may still exhibit reasonable CNS penetration in vivo; i.e., where the unbound fractions and nonspecific tissue binding act as a compensating force. In these instances, the efflux ratio and in vitro blood-brain partition ratio may be used to predict the in vivo blood-brain ratio. This relationship may be extended to account for the differences in CNS penetration observed in vivo between mdr1a/b wild type and knockout mice. In some instances, cross-species differences that might initially seem to be related to differing transporter expression can be rationalized from knowledge of unbound fractions alone. The results presented in this article suggest that the information exists to provide a coherent picture of the nature of CNS penetration in the drug discovery setting, allowing the focus to be shifted away from understanding CNS penetration toward the more important aspect of understanding CNS efficacy.

175 citations


"A 3D brain unit model to further im..." refers background in this paper

  • ...Unbound drug 12 may cross the BBB by passive and/or active transport [2–10]....

    [...]

Journal ArticleDOI
TL;DR: Intravital microfilming by means of a dark-field contact epiobjective was used for measuring capillary blood flow velocity in the brain and skeletal muscles of the rat to indicate that this range of velocities is optimal to adequately supply neurons with oxygen.

174 citations


"A 3D brain unit model to further im..." refers background in this paper

  • ...eu and [57] 5 [58–62], [63] This is the surface area of the BBB that separates one side of a brain capillary within the 3D brain unit from the brain ECF....

    [...]

Journal ArticleDOI
TL;DR: Investigation of the influence of different rates of transport into and out of the brain, including passive and active transport, on unbound brain concentrations and profile in relation to the blood concentration profile found non-passive transport seems to be the case for almost all drugs studies with MD so far.
Abstract: Purpose. The purpose of the study was to investigate the influence of different rates of transport into and out of the brain, including passive and active transport, on unbound brain concentrations and profile in relation to the blood concentration profile. Special emphasis is put on hydrophilic drugs. Methods. Simulations were performed with a model including one body compartment and one brain compartment, with linear or saturable transport into and out of the brain. Comparisons were made with experimental results from microdialysis (MD) studies. Results. Three features were evident when combining the MD results: 1) equilibration across the blood-brain barrier (BBB) is rapid, 2) half-life is similar in brain and blood for most drugs, and 3) unbound brain concentrations seldom reach the level of unbound blood concentrations. A low concentration ratio brain:blood is not mainly caused by a low influx, but rather by different influx and efflux clearances. Active transport out of the brain can explain the results, but also active transport into the brain under certain conditions. A small volume of distribution in brain vs. that in the rest of the body contributes to a rapid equilibration and similar half-lives. Conclusions. Assumptions of slow equilibration of hydrophilic drugs and similar unbound concentrations across the BBB at steady state are contradicted. The results are more in line with recent findings on the presence of P-glycoprotein and other transport mechanisms at the BBB. Non-passive transport across the BBB seems to be the case for almost all drugs studies with MD so far.

169 citations


"A 3D brain unit model to further im..." refers methods in this paper

  • ...In addition, we model active transport into 219 and out of the brain ECF with Michaelis-Menten kinetics, similar to the approach of [6]....

    [...]

Journal ArticleDOI
16 Jun 2014
TL;DR: This review summarizes important concepts of the BBB in normal versus pathophysiology and how this physical, enzymatic, and efflux barrier provides necessary protection to the CNS during drug delivery, and consequently treatment challenging.
Abstract: The treatment of central nervous system (CNS) disease has long been difficult due to the ineffectiveness of drug delivery across the blood-brain barrier (BBB). This review summarizes important concepts of the BBB in normal versus pathophysiology and how this physical, enzymatic, and efflux barrier provides necessary protection to the CNS during drug delivery, and consequently treatment challenging. Small molecules account for the vast majority of available CNS drugs primarily due to their ability to penetrate the phospholipid membrane of the BBB by passive or carrier-mediated mechanisms. Physiochemical and biological factors relevant for designing small molecules with optimal capabilities for BBB permeability are discussed, as well as the most promising classes of transporters suitable for small-molecule drug delivery. Clinically translatable imaging methodologies for detecting and quantifying drug uptake and targeting in the brain are discussed as a means of further understanding and refining delivery parameters for both drugs and imaging probes in preclinical and clinical domains. This information can be used as a guide to design drugs with preserved drug action and better delivery profiles for improved treatment outcomes over existing therapeutic approaches.

153 citations

01 Jan 2010
TL;DR: Concepts and Applications of Clinical Pharmacokinetics: Concepts and Applications, 4/e al precio 64,00 € de Rowland and Tozer.
Abstract: Tienda online donde Comprar Clinical Pharmacokinetics: Concepts and Applications, 4/e al precio 64,00 € de Rowland and Tozer, tienda de Libros de Medicina, Libros de Farmacologia Clinica - Farmacologia general

150 citations

Frequently Asked Questions (2)
Q1. What contributions have the authors mentioned in the paper "A 3d brain unit model to further improve prediction of local drug distribution within the brain" ?

In this article the authors show that all the above mentioned factors affect brain ECF PK in an interdependent manner. 

However, for drugs that do bind plasma proteins, the assumption is likely violated with an impact to be investigated in future work. In similar fashion, assumptions 2 ( ii ), 2 ( iv ) and 2 ( vii ) are not violated for drugs that do not cross cells, but it is likely that for drugs that do, they are violated with an impact to be investigated in future work. With the establishment of the current 3D brain unit model, the authors are now ready to incorporate intra-extracellular exchange and drug binding to intracellular binding sites in future modelling work. It is anticipated that in certain cases, like those of high drug-target binding or active transport, these differences may also exist on a larger time-scale, but this requires further investigation.