You are on page 1of 18

Mathematical Models

of Hemostasis 35
Keith B. Neeves and Karin Leiderman

In this chapter, we present mathematical


Introduction ­ odels of thrombin generation, thrombus forma-
m
tion, and fibrin polymerization. Mathematical
There is a vast amount of information on the models have been used to generate new hypoth-
mechanisms of coagulation and its regulation by eses and make novel predictions about the thresh-
chemical, cellular, and biophysical processes. old behavior of thrombin generation, some of
The complex and nonlinear nature of the coagu- which have been confirmed experimentally.
lation pathway makes it difficult to intuit how More recently, some models have been used to
changes in plasma protein levels affect, for identify risk factors for bleeding and thrombosis
instance, thrombin generation. Moreover, bio- in retrospective studies. Based on the success
chemical kinetics alone is not enough to charac- of these studies, mathematical models have the
terize in vivo coagulation because its regulation potential to guide therapeutic strategies and
is intimately tied to platelet and endothelial cell develop novel approaches to reduce cardiovascu-
activation, transport processes, and the local lar disease. Here, we highlight those models that
hemodynamics. Fortunately, coagulation is one have made, in our opinion, important contribu-
of the most studied biochemical networks in tions not only to revealing the fundamental
terms of network topology (which components mechanisms that regulate coagulation, but also
interact with each other) and kinetic rate con- that have shown the potential to contribute to
stants (how fast they work). It is these features clinical science.
and complexities that make coagulation particu- As of this writing, the use of mathematical
larly well-suited for mathematical modeling. models in the field of hemostasis is limited to a
handful of research groups. This is due, in part, to
the complexity and sophistication of the models,
but also to the fact that most of them are still in
their nascent stages. Only a small number of
K.B. Neeves (*) models have been used beyond a few narrow
Chemical and Biological Engineering Department, studies and experimental validations have been
Colorado School of Mines, 1500 Illinois St.,
Golden, CO 80401, USA performed only under very limited conditions. In
our opinion, another major impediment is the
Department of Pediatrics, University of Colorado
Denver, Aurora, CO USA translation from the developers (primarily math-
e-mail: kneeves@mines.edu ematicians, engineers, and physicists) to the
K. Leiderman broader biomedical community. Part of the prob-
University of California Merced, Merced, CA, USA lem lies with developers, who have yet to build

© Springer International Publishing Switzerland 2016 567


E. Gonzalez et al. (eds.), Trauma Induced Coagulopathy, DOI 10.1007/978-3-319-28308-1_35
568 K.B. Neeves and K. Leiderman

interfaces to these models that are accessible to work under the assumption that quantities are
the non-expert. Another problem lies with the well-mixed, that is, changes in these quantities
fact that these two groups speak different lan- are tracked only in time and there are no spatial
guages. Specifically, the basis of most mathemat- variations. Partial differential equation (PDE)
ical models of coagulation are differential models have the ability to track quantities in both
equations, a subject that many biomedical rese­ space and time so that complex spatial informa-
archers and clinicians either do not have expo- tion can be extracted from them, for example, the
sure to, or, would rather not revisit. While it is not heterogeneous composition of a clot. In this sec-
entirely necessary to understand the nuts and tion, we give a brief mathematical primer on the
bolts of a computational model to use it, it can be derivation and meaning of the ODEs and PDEs
instructive at least have an idea of how it works. found in many coagulation models. The aim of
In the section “Primer on Differential this section is to give the reader a qualitative
Equations” we provide a primer on differential understanding of how differential equations are
equations for the non-expert. Our hope is that this formulated and used in coagulation modeling,
section will give the reader a qualitative under- without getting into the methods for solving them.
standing of how differential equations are formu-
lated and used in coagulation modeling, without
details about the numerical methods with which  patially Homogeneous Ordinary
S
they are solved. In section “Mathematical Models Differential Equation Models
of Coagulation and Fibrin Formation,” we pres-
ent models of coagulation that consist of systems One type of mathematical model of coagulation
of ordinary and partial differential equations, par- simulates well-mixed biochemical reactions in
ticularly highlighting those that contain features time and not space. To better understand the well-
of blood flow and platelet-mediated coagulation. mixed assumption, one can think of a test tube
The coverage in this section is not intended to be filled with clotting factors in which the concen-
exhaustive nor do we go into the mathematical tration of each factor is the same at every spatial
details of any of the models. The goal here is to location, as if the contents were instantaneously
give the reader a flavor of the different types of and perfectly mixed. For the model, the initial
computational models and their salient features. factor concentrations and chemical reaction
In section “Predictions and Applications of Mathe­ schemes that describe the factor interactions are
matical Models,” we discuss a few case studies of known. The model results are in the form of con-
where mathematical models have made notable centrations tracked in time, for every species in
contributions to the field. In section “Future the reaction schemes. To “track” these concentra-
Challenges,” we discuss challenges and futures tions, mathematical equations are formulated
research needs. using the reaction schemes and the law of mass
action (LMA). For example, consider the follow-
ing reaction schemes:
Primer on Differential Equations
k+
Many computational models of hemostasis are A + B  C, (35.1)
based on systems of equations that describe k-

dynamic processes; these processes could be
k
biochemical, biophysical, biomechanical, or a F G. (35.2)
combination thereof. The equations track changes ®
in certain desired quantities (e.g., thrombin con- The reversible reaction in Scheme (35.1) states
centration, platelet density, blood flow velocity) that two reactants, A and B, come together to
from their prescribed, or, known initial values. form a single product C at the rate k+, and that C
Ordinary differential equation (ODE) models dissociates back into A and B at the rate k−.
35  Mathematical Models of Hemostasis 569

Scheme (35.2) is an irreversible reaction where a and initial concentrations for each variable are
single reactant F is converted into a single prod- known. Since mass is conserved we can reduce
uct G at a rate k. The LMA states that the rate of the number of equations that need to be solved
reaction is often found to be proportional to the simultaneously from three to two. That is, A
product of the reactant concentrations. Therefore, exists as either itself, A, or as a component of the
the rate of the forward reaction for Scheme (35.1), product, C. So, if the initial concentration of A is
i.e., the rate of accumulation of the product C, is denoted by A0, then the sum of A and C remains
k+[A][B]. The square brackets are used to denote constant in time:
concentration, but for simplicity, when writing
A + C = A0 . (35.6)
rates from this point forward, we will drop the
brackets so that k + AB = k + [ A][ B] . The rate of With this, C can be replaced in Eqs. (35.3), (35.4),
the backward reaction is simply the dissociation and (35.5) by A0 − A so that one only needs to
of the product, which happens at the rate k−C. solve the two equations for A and B. Once A is
The rate of the reaction in Scheme (35.2) is k F. known, C is calculated by Eq. (35.6).
The proportionality constants, k + , k - , and k, are Now consider the Scheme (35.2). The total
called rate constants and depend on the interac- rates of change of F and G are
tion thermodynamics between reactants and their dF
shape and size. = - kF , (35.7)
dt
We can now translate this information into
mathematical equations. First we will consider dG
tracking species C in reaction (35.1). Here the total = kF . (35.8)
dt
rate of change of C is equal to the rate of the accu-
mulation of C minus the rate of dissociation of C. Here, the concentration of species F decays
Using the definition of these rates from the previous exponentially and whatever is lost is converted
paragraph, we can express the total rate of change into G. Using conservation, F + G = F0 , where
of C with the following mathematical equation: F0 is the initial concentration of F so that one
only needs to solve the ODE for F and then
dC G = F0 - F . Example solutions to both systems
= k + AB - k - C. (35.3)
dt of ODEs are shown in Fig. 35.1.
Although the above examples are of elemen-
Equation (35.3) is called an ODE. It is a differen-
tary reactions, the same basic principles can
tial equation because it relates the unknown, C,
be applied to the two-step enzyme reactions of
to its derivative, or how it changes, in time, dC∕dt.
coagulation. The most general reaction is one
It is ordinary because the dependent variable, C,
that includes a substrate, S, enzyme, E, complex,
depends on only one independent variable, t.
E: S, and product, P:
Using the LMA, A and B have similar ODEs:
k+
dA k cat
= - k + AB + k - C , (35.4) E+S  E:S E + P.
dt - ®
k
dB
= - k + AB + k - C. (35.5)
dt Using the LMA, the ODE for the complex E:S is
Together, Eqs. (35.3), (35.4), and (35.5) com-
dE : S
prise a coupled (each equation depends on all of = k + ES - (k - + k cat ) E : S .
(35.9)
the others) system of three ODEs. The three cou- dt
pled equations must be solved simultaneously Similar equations can be formulated for the sub-
and numerically given that the rate constants strate, enzyme, and product.
570 K.B. Neeves and K. Leiderman

1 1
A F
B G
0.8 C 0.8 F+G
Concentration (nM) A+C

Concentration (nM)
0.6 0.6

0.4 0.4

0.2 0.2

0 0
0 0.2 0.4 0.6 0.8 1 0 0.2 0.4 0.6 0.8 1
Time (s) Time (s)

Fig. 35.1  Example solutions to systems of coupled ODE. k = 0.5  s , and k = 6  s . Left: species A and B
-

Nume­ rical solutions to Eqs.  (35.3)–(35.5) (left) and  are depleted as the product C is formed. The sum A + C
(35.7)–(35.8) (right). Initial concentrations are remains constant. Right: species F is converted to species
A0 = 0.5 nM , B0 = 1 nM , C0 = 0, F0 = 1 nM , and G and the sum F + G remains constant
+
G0 = 0. Rate constants set to k = 5  ( nM × s ) ,

 patially Heterogeneous Partial


S Let us return to species F from Scheme (35.2).
Differential Equation Models Imagine a thin cylindrical tube with length L and
constant cross-sectional area. Assume that the
Tracking the reaction and motion of clotting fac- number of molecules of F inside this tube is
tors in space and time is often desirable and changing due to (1) being carried along the length
requires the use of PDE models. These are differ- of the tube with velocity u by advection, (2) being
ent from ODEs in that the unknown dependent spread out along the length of the tube by diffu-
variable is now related to its derivatives taken in sion characterized by a coefficient D that depends
more than one independent variable. For e­ xample, on temperature, size and shape of the molecules,
partial derivatives in space (one dimension) and and (3) decay according to Scheme (35.2). For
time look like ¶F ( x, t )  ¶x and ¶F ( x, t )  ¶t , simplicity the motion is restricted to one spatial
respectively. dimension. Letting F(x, t) represent the concen-
So what kind of motion is modeled and how is tration of F at location x along the length of the
it represented mathematically? The most com- tube and time t, the rate of change of F is
mon types of motion found in clotting models are
¶F ¶F
advection and diffusion. Consider the motion of = - u
¶T
  ¶X
factor X (FX) in plasma. Advection is the process rate of change transport by advection
by which the FX molecules are carried by the (35.10)
¶ æ ¶F ö
bulk flow of the plasma; think of a leaf being car- + çD ÷ - kF
.
¶X ¶ Xø decay
è

ried in a stream. Diffusion describes changes in
transport by diffusion
FX concentration due to random motion from
thermal fluctuations. It causes the net flux of FX As with an ODE, obtaining the solution requires
molecules to move from areas of high concentra- an initial condition, i.e., the initial distribution of
tion to low concentration until no gradients exist. molecules. Further, since a PDE involves space,
Here one can think of a single drop of blue food conditions at the boundaries must also be specified.
coloring in a cup of water spreading out until all For example, one might want to specify a fixed
the water in the cup is blue. concentration of molecules at a boundary, allow
35  Mathematical Models of Hemostasis 571

molecules to enter/leave a boundary at a specified


rate, or require that they remain inside the domain.
This advection–diffusion–reaction PDE can be
derived from physical principles using conserva-
tion laws. With initial and boundary conditions, it
can be solved analytically using standard mathe-
matical techniques. Derivations and basic solu-
tion techniques are found in most PDE textbooks
[27, 59]. Excellent references on mathematical
modeling in biology and differential equations in
the biological setting are [15, 55].
In higher dimensions, the PDE takes the form:
¶F
=- ×ÑF
u + Ñ × ( DÑF )
¶t 
 transport by advection
transport by diffusion (35.11)
rate of change

- kF

rate of decay

where some short-hand notations are used; the
bold u represents the velocity vector (a vector has
information about both magnitude and direction),
the ∇ represents the derivative of the function it
operates on (F) in each spatial dimension.
Figure  35.2 shows a sample solution to
Eq. (35.11) that represents the time evolution of a
bolus of a chemical species in a fluid-filled chan-
nel. The chemical is advected with the fluid, dif-
fuses in the fluid and is decaying at a rate
determined by k. The velocity, u, flows from left
to right and is set to be parabolic, i.e., highest
velocity in the center of the channel and zero on
the top and bottom walls. This is the shape of
pressure driven flow through a tube as in a blood
vessel. The plot on the top shows the initial bolus Fig. 35.2  Example solution of Eq. (35.11). Time evolu-
and the plot on the bottom shows the concentra- tion (top to bottom) of a bolus of a chemical species in a
fluid-filled channel undergoing an irreversible reactions.
tion after a short period of time. What the reader The chemical is dispersed by the flow that runs from left
should notice is that (1) the flow carries the spe- to right and diffusion, which causes the species to spread
cies from left to right (at a higher velocity in out in all directions. The species decays by an irreversible
­center of the tube), (2) the species spreads due to reaction described by Scheme (35.2)
diffusion, and (3) the species decays by means of
Scheme (35.2). These are precisely the three
terms on the right-hand side of Eq. (35.11).
how it affects the transport of chemical species.
The simplest approach is to treat transport as
Treatment of Blood Flow ­simply another rate process in the kinetic ODE
that describes the consumption and generation of
When considering blood flow in coagulation a component by biochemical reactions. This is
models it is necessary to either solve for the done by calculating a mass transfer coefficient,
velocity vector u explicitly, or make some sim- which combines the influence of advection and
plifying assumptions about the flow field and diffusion on solute transport to a solid boundary
572 K.B. Neeves and K. Leiderman

Table 35.1  ODE models of coagulation


Model Initiation pathway Static/flow Phospholipids
Beltrami–Jesty [3] Extrinsic Static Saturating
Hockin et al. [29] Extrinsic Static Saturating
Chatterjee et al. [11] Extrinsic, Intrinsic Static Saturating
Bungay et al. [6, 7] Extrinsic Static Nonspecific lipid binding sites
Fogelson and Extrinsic Flow Explicit platelet binding sites
co-workers [21, 22, 36]

into a single rate constant. The mass transfer of blood flow and platelet-mediated coagulation.
coefficient approach is valid for relatively small Models of platelet adhesion, activation and aggre­
injuries where the well-mixed assumption can gation in the absence of coagulation can be found
still be used and for cases where the velocity near in recent review articles [18, 20, 61].
the boundary does not change much during the
formation of a clot. A slightly more complicated
approach is to assume a certain flow profile, as ODE Models of Coagulation
we have done in Fig. 35.2, and insert the velocity
vector into the PDE solute conservation equation. Table 35.1 summarizes five of the seminal ODE
This approach is valid when the velocity vector models of coagulation developed over the last
either does not change with time or changes in a 20 years. These models are categorized based on
known manner. The most rigorous, but difficult, extrinsic (tissue factor; TF) and intrinsic path-
approach is to couple the conservation of solute ways (factor XII; FXII), treatment of binding
equation (Eq. (35.11)) to the conservation of reactions to lipid surfaces and the inclusion of
momentum equation. The conservation of blood flow. Thrombin generation models of the
momentum equation is a PDE that describes how extrinsic pathway under static conditions are the
fluid forces evolve in time and space. This most numerous and mature models. These mod-
approach is useful when the formation of the els mimic ex vivo thrombin generation in a test
thrombus affects the blood flow, as is the case for tube or well plate initiated by TF in the presence
arterial thrombosis. of lipid membranes (synthetic or platelet derived).
ODE models have been central to elucidating
the mechanisms of the threshold behavior of
Mathematical Models coagulation. Coagulation shows an activation
of Coagulation and Fibrin threshold whereby a sub-threshold stimuli results
Formation in thrombin levels that are quickly neutralized by
endogenous inhibitors and a threshold or higher
Mathematical models of coagulation and anti­ stimulus results in a burst in thrombin generation
coagulation pathways, fibrin polymerization, that overcomes the inhibitors. This behavior
fibrinolysis, platelet adhesion, activation and serves a physiological function in that coagula-
aggregation have been developed that range from tion is not propagated by weak stimuli such as
length scales of individual receptors to entire small changes in circulating procoagulant mole-
thrombi. These models can be categorized in cules (e.g., TF), and thus preventing systemic
terms of the biochemical phenomena they model clotting. But, when sufficient stimuli is present
and their coupling to biophysical processes. such as at the site of a vascular injury or by ele-
Here, we focus on ODE and PDE models of vated levels of procoagulant molecules following
coagulation, including those that contain features trauma, thrombin is produced quickly and at
35  Mathematical Models of Hemostasis 573

l­evels that can activate platelets and catalyze with initial levels of TF and FVII/FVIIa where in
fibrin polymerization. the purified systems, TF:VIIa complexes were
Beltrami and Jesty show that threshold activa- preformed. The activation of FX by FIXa and
tion in coagulation is a characteristic of enzyme APC was later incorporated into the model [5, 9].
systems with feedback loops and inhibitors [3]. The Hockin–Mann model and its derivatives
In the case of coagulation, thrombin provides have served as the basis for other numerical anal-
positive feedback on its own production by cata- yses of coagulation including points of robust-
lyzing the formation of FVa and FVIIIa, which ness and fragility [41] and sensitivity analyses of
must overcome inhibition of thrombin and FXa kinetic rate constants and plasma composi-
by ATIII and FVa and FVIIIa by activated protein tion [13] (Fig. 35.3). A limitation of these models
C (APC). Enzyme networks are initiated by is the assumption that lipid surfaces are in excess,
low levels of enzymes such as small amounts of and therefore, does not address the regulatory
FVIIIa:FIXa or FXa. Their analysis on generic role of the lipid surface. Moreover, validation of
enzyme system predicts that sub-threshold stim- the models against empirical results has only
uli can be greatly amplified by feedback, for been shown under very specific conditions and/or
example, of thrombin on FVIIIa, and in the case by fitting parameters of the model to the results.
of threshold stimuli, feedback greatly increases Chatterjee, Diamond, and colleagues built a
total thrombin generation. A nice review of the “platelet-plasma” ODE model to describe the
structures of the various feedback mechanisms of ­initiation of coagulation in the absence of initial
coagulation, the mechanisms by which they can levels of TF in diluted whole blood (Fig. 35.3) [11].
produce threshold behavior, and the possible role The model, made up of 76 ODEs, includes the
of thresholds in clotting system regulation can be extrinsic pathway based on the Hockin–Mann
found in [30]. model, with the addition of FXIa and FXIIa
The earliest ODE models of coagulation con- dependent thrombin generation, activation-­
sidered individual reactions or a small number of dependent platelet phosphatidylserine, and fibrin
reactions, for example, to investigate the func- formation. It also describes the kinetics of the
tional properties of the prothrombinase complex fluorogenic thrombin substrate boc-VPR-AMC,
and the effects of hirudin and APC on thrombin which can act as a competitive substrate from
generation [45, 65]. Jones and Mann were the thrombin, slowing down positive feedback. This
first to describe the extrinsic pathway in signifi- explicit treatment of the thrombin substrate also
cant detail including activation of FIX, FX, FV, provides a more faithful comparison to experi-
and FVIII and the assembly of the FVIIIa:FIXa ments that use boc-VPR-AMC as a thrombin
and FVa:FXa complexes [31]. This model inclu­ reporter. The inclusion of the intrinsic pathway
des 18 ODEs and is based on the reactions in a was necessary to predict experimental observa-
purified system of zymogens and enzymes [38]. tions of coagulation in the absence of TF, even
Their model results are in good agreement with when treated with the FXIIa inhibitor corn tryp-
their own experimental measurements of throm- sin inhibitor (CTI). Importantly, the model
bin generation in a purified system. The Jones– ­predicts, and thrombin generation experiments
Mann model was extended by Hockin et al. [29] confirm, that FXIIa can leak past high concentra-
to include the inhibitors TFPI, ATIII, and a more tions of CTI and initiate coagulation. Phospho­
detailed description of certain enzymatic and lipids are provided by platelets in this model and
binding reactions. The extended model includes it was shown that platelet activation by the GPVI
34 ODEs and thrombin generation curves showed agonist convulxin dramatically reduces the initia-
good agreement with those in a purified system tion time for coagulation compared to unstimu-
with TFPI and ATIII for varying initial levels of lated platelets.
prothrombin [8]. Variances in other initial con- The initiation, amplification, and propagation
centrations were not compared. In both Jones– of coagulation require phospholipid surfaces that
Mann and Hockin–Mann, coagulation is triggered support the assembly of the enzyme complexes
574 K.B. Neeves and K. Leiderman

Fig. 35.3  ODE models of coagulation. Left: wiring dia- hypothetical population defined by normal range variation
gram of the Chatterjee–Diamond platelet-plasma in factor levels based on the Hockin–Mann model [13]
model [10]. Right: thrombin generation phenotypes in an

TF:FVIIa, FVIIIa:FIXa, and FVa:FXa. The models but is limited at low lipid concentrations by low
discussed above assume lipids are in excess and levels of FVIIa and FXa production (the enzymes
thus do not explicitly account for the binding responsible for TFPI activation) and at high lipid
reactions and/or finite number of binding sites. concentrations because most FXa is bound to lip-
Bungay and Gentry developed a model in which ids. The effectiveness of APC in inhibiting FVa
association and dissociation to a finite concentra- and FVIIIa also shows a strong dependence on
tion of lipids are explicitly modeled in a system lipid concentration. The Bungay–Gentry model
of 31 reactions and 73 ODEs [7]. Although more was later extended to study thrombin production
than a few reactions in the Bungay–Gentry models in human ovarian follicular fluid [6]. Results
differ from those in the Hockin–Mann model, show that the amount of thrombin produced in
results from both models seem to be in qualita- ovarian follicular fluid is much lower than that in
tive agreement with results from the same empir- plasma, supporting the hypothesis that in follicu-
ical study [8]. lar fluid thrombin functions to initiate cellular
Simulations of the Bungay–Gentry model activities via intracellular signaling receptors.
showed that the lag time, peak thrombin concen- The models discussed thus far all simulate
tration, and duration of thrombin generation are coagulation under static conditions. These are
strongly influenced by lipid concentration up to a appropriate models for specific clinical clotting
saturating concentration of 300 nM. No thrombin assays and perhaps even clotting under condi-
is produced for less than 10 nM lipids, thus pro- tions of stasis such as extravascular bleeding or
viding another threshold that is necessary for deep vein thrombosis. However, most hemostatic
thrombin generation. While it is not surprising and thrombotic clots form under conditions of
that thrombin generation is dependent on lipid blood flow and where TF is localized at the vessel
concentration, the model does make nonintuitive wall. Furthermore, only the Chatterjee–Diamond
predictions with respect to the effectiveness of model accounts for the role of platelets in media­
different inhibitors. For example, their results ting coagulation. In the cell-based model of
show that TFPI almost completely inhibits TF at thrombus formation [43], activated platelets with
an intermediate lipid concentration of 150 nM, exposed phosphatidylserine (PS) are the primary
35  Mathematical Models of Hemostasis 575

surface for the propagation of coagulation. Thus, these surfaces as described by the Bungay–
a model without platelets is unlikely to capture Gentry model. An ODE model of coagulation
the dynamics of coagulation in vivo. under flow requires additional terms to describe
Kuharsky and Fogelson developed an ODE the rate of transport of solutes and platelets to and
model that combined coagulation, blood flow, from an injury site as described by the Kuharsky–
and platelet function [36]. The model considers Fogelson model. A unifying theme and outcome
blood flowing over a 10 μm patch of TF where, from all of these models is the concept a of
because of the small size of the injury, the well-­ threshold stimuli needed to induce robust throm-
mixed assumption is justified. The transport of bin generation. These models all use the “well-­
zymogens, co-factors, inhibitors, enzymes, and mixed” assumption such that time is the only
platelets to and from the injury site are modeled independent variable. This is a good assumption
with mass transfer coefficients, as discussed in when a system is homogenous, as in a well-plate
section “Treatment of Blood Flow.” Platelets are assay, or when an injury is small. However, when
treated like chemical solutes and are allowed to this assumption begins to break down, for
accumulate with kinetics based on experimental ­example, in large injuries, PDEs are necessary to
measurements of platelet adhesion and are acti- account for variations in space.
vated by both surface bound (e.g., collagen) and
soluble agonists (ADP or thrombin). Platelets can
physically block access to surface bound enzyme PDE Models of Coagulation
complexes such as TF:FVIIa as they adhere and
aggregate. The model shows that thrombin gen- Table  35.2 summarizes four PDE models of
eration has a threshold-dependence on surface coagulation that investigate the spatial depen-
concentration of TF. Below the threshold concen- dence of thrombin generation, fibrin formation,
tration of TF, little to no thrombin is generated, and thrombus growth. All models consider
above it thrombin generation becomes almost thrombin generation initiated by TF immobilized
saturated. Interestingly, the model also shows on a surface. However, they each couple the
that dilution by flow and blocking access to wall- ­biochemical reactions pathways of coagulation
bound TF by adherent platelets are the primary with transport mechanisms (Eq. (35.10)) includ-
mechanisms that inhibit coagulation and are more ing surface diffusion within a phospholipid mem-
potent than TFPI and APC pathways, at least for brane [4], diffusion away from a TF-rich wall [14,
small injuries [21]. The model has been extended 34, 52], and thrombus formation on a TF-rich
to include activation of FXI by thrombin and FIX wall under flow [39, 40, 68, 70]. These models
by FXI [22]. Simulation results predict that the reveal important biophysical mechanisms that
effect of a severe FXI deficiency depends on the regulate coagulation.
platelet count, and that FXI becomes more impor- Beltrami and Jesty extended their solution
tant in coagulation at high platelet counts. phase ODE model [3] described in section “ODE
In summary, existing ODE models of throm- Models of Coagulation” to investigate model
bin generation have shown qualitative agreement enzyme feedback networks on the surface of
with empirical results under limited conditions in membranes [4]. Here, a PDE is used to describe
purified, closed systems. If phospholipid surfaces the evolution of enzyme generation on a phos-
are in excess, then a qualitative description of pholipid membrane where enzymes and zymo-
thrombin generation is described by ODE rate gens are transported by flow in the solution phase
equations of the extrinsic pathway, intrinsic path- and by diffusion in the membrane phase. Similar
way, and inhibitors as described in the Hockin– to the predictions from the Kuharsky–Fogelson
Mann and Chatterjee–Diamond models. If the ODE model, flow is a profound inhibitor of
amount of phospholipid surface is limited and ­coagulation, and at sufficiently high flow rates,
can be saturated, then additional ODEs are more potent than the biochemical inhibition. This
required that describe the binding kinetics to model also provided important predictions about
576 K.B. Neeves and K. Leiderman

Table 35.2  PDE models of coagulation


Spatial dimensions,
Model Pathway Type of PDE biophysical phenomena
Beltrami–Jesty [4] TF Diffusion–reaction 1, surface diffusion
Ataullakhanov and TF Diffusion–reaction 1, excitable wave
co-workers [14, 34, 52]
Leiderman–Fogelson [39, 40] TF Diffusion–reaction–advection 2, flow in a vessel
Xu et al. [68, 70] TF Diffusion–reaction–advection 2, flow in a vessel

what is the minimum injury size that can induce Leiderman and Fogelson developed a two-­
significant enzyme production. For the base case dimensional model of thrombus formation under
parameters in this model, the membrane patch flow that accounts for coagulation biochemistry,
must be greater than 5 μm, but that size depends including surface-dependent reactions, as well as
on the kinetics of the enzyme and inhibition reac- platelet activation and deposition [39]. The model
tions, as well as the flow rate and binding site extends the Kuharsky–Fogelson model to treat
density. A simplified analytical model predicts spatial variations in concentrations discussed in
that a minimum threshold patch size is needed section “ODE Models of Coagulation,” heteroge-
when the diffusivity of bound proteins is neities within the thrombus, and flow distur-
­considered. This prediction was later confirmed bances caused by the growing thrombus. Platelets
experimentally for both static and flow condi- are treated as solutes, rather than as discrete par-
tions [33, 57]. ticles. The thrombus is treated as a porous mate-
Ataullakhanov and colleagues developed a rial which allows for analysis of intra-thrombus
series of mathematical and experimental models fluid and solute transport, and reveals that diffu-
of the spatial-temporal dynamics of thrombin sive solute transport aids upstream thrombus
generation and fibrin formation initiated on a growth. Results also show that thrombus growth
procoagulant wall in the absence of flow [25]. is strongly influenced by wall shear rate and the
Here, coagulation acts like an excitable media, near-wall excess of platelets and that thrombus
where once initiated by TF bearing cells or lipid permeability strongly impacts growth and struc-
on a surface, travels like a wave away from the ture. Motivated by experiments that suggest an
surface [49, 51]. Different factors play unique important role for intra-thrombus transport [58],
roles at different times and distances from the the Leiderman–Fogelson model was extended to
surface. Initiation is controlled primarily by the further explore the effect of hindered transport of
concentration and distribution of TF, and is ins­ proteins within the thrombus [40]. Results show
ensitive to FVIII, FIX, or FXI levels [2, 49]. that early growth is promoted because platelets
However, propagation away from the TF-rich shelter enzymes from the flow, and that later
surface is controlled by thrombin feedback growth is attenuated because enzymes are con-
mechanisms that produce FVa, FVIIIa, and fined to the interior core of the thrombus and the
FXIa [14, 50]. Interestingly, thrombin generation supply of substrates to them is reduced (Fig. 35.4).
will continue to grow for hours over several cen- With the transport hindrance, the final thrombus
timeters even in the presence of ATIII and TFPI. that emerges from the model is smaller, has a
Only by the addition of thrombomodulin (TM) dense core and a less dense shell similar to
does the thrombin wave arrest [14, 52]. These thrombi described in the laser injury model [58].
reaction–diffusion models may be particularly Xu and colleagues developed a model of
apt for describing extravascular clot formation thrombus formation that includes several sub-
where blood flow is low or absent. models that exchange information with each
35  Mathematical Models of Hemostasis 577

Platelet Density Prothrombin-Prothrombinase Complex


Hindered Unhindered
Transport Transport

Fig. 35.4  Simulations of thrombus growth in a model of reflect their macromolecular size. Left: hindered protein
platelet deposition and coagulation chemistry under flow transport produced smaller thrombi (bottom) with a uni-
in the Leiderman–Fogelson PDE model [39, 40]. A formly highly dense (dark red) core of platelets compared
thrombus is modeled as a porous medium that allow for to the unhindered case. Right: hindering protein transport
interstitial fluid and solute transport. The rate of intersti- severely limits the ability of fluid-phase prothrombin
tial fluid flow decreases as the density of bound platelets to penetrate thrombus to reach platelet-bound proth­
increases. For “hindered transport” simulations, the diffu- rom­binase (bottom) and so greatly reduces thrombin
sion and advection of proteins was further hindered to production

other to simulate the interplay between platelet Fibrin Polymerization and Deposition


function, coagulation, and flow [68–70]. The
coagulation submodel is based on the Hockin– ODE and PDE models of fibrin polymerization
Mann ODE model of coagulation, and binding have been reported under both static and flow
reactions and binding site density are based on conditions, but have yet to be integrated into full
the treatment by Kuharsky–Fogelson. In contrast coagulation models as described above. An ODE
to the Leiderman–Fogelson models, platelets are model of fibrin polymerization kinetics includes
modeled as discrete particles, rather than as a sol- cleavage of fibrinopeptide A by thrombin, forma-
ute. Such particle-based simulations provide a tion of protofibrils by fibrinogen monomers and
direct coupling of platelet motion, adhesion, and oligomers, and lateral aggregation of protofibrils
aggregation to the forces imparted on them by the into fibers [63]. The model is in good agreement
flow. It also allows the model to track the history with turbidity and electron microscopy data. The
and activation state of each platelet. This feature effects of fibrinopeptide B cleavage, thrombos-
allows for identification of potential structures pondin, and ionic strength can be simulated
and conditions that result in embolism. Porosity by varying kinetic rate constants. This is a good
and permeability properties of thrombi were example of how a simple model can capture a
based on intravital imaging data in a laser injury wide range of dynamics even if the details of all
model. The model predicted that low levels of known mechanisms are not included in the model.
FVII in blood result in a significant delay in early There is a well-established relationship bet­
stage thrombin production in thrombi formed ween thrombin concentration and gel structure,
under venous conditions in agreement with where higher thrombin concentration yields gels
in vivo data in mice expressing low levels of with thinner fibers and higher degrees of branch-
FVII [70]. This type of model is considered ing that are less susceptible to fibrinolysis [62].
“multiscale” because the different submodels The branch point density determines, in part, the
operate at different length scales. Microscale mechanical properties of a fibrin clot, but the
phenomena like coagulation reactions and plate- branching process itself is poorly understood.
let adhesion are tied to macroscale phenomena Fogelson and Keener developed a kinetic fibrin
such as blood flow. gelation model that considers how two types of
578 K.B. Neeves and K. Leiderman

reactions—bimolecular linear protofibril extension orders-of-magnitude depending on the fiber


and trimolecular branch formation—can form ­density  [66]. The predicted gel heights from this
different gel structure based on the supply rate of model are in agreement with fibrin gels formed
fibrin monomers [19]. The kinetic gelation model under flow on TF-coated microparticles [48].
tracks the molecular weight of polymer chains
with a single PDE and gelation is defined by
when the average cluster size approaches infinity.  redictions and Applications
P
Higher supply rates of fibrin monomers, which of Mathematical Models
would correlate to higher thrombin concentra-
tion, lead to faster gelation, higher branching, Many of the models in section “Mathematical
and shorter fibers. These results agree qualita- Models of Coagulation and Fibrin Formation”
tively with experimental results. have been partially validated against ­experimental
The morphology and density of fibrin formed data. However, validation is just the first step in
under flow conditions is dramatically different testing the utility of a model. The ultimate test of
than fibrin gels formed under static conditions. these models is whether they can make predic-
Instead of a voluminous hydrogel that is mostly tions, assess risk, and guide therapeutic ­decisions.
water, fibers form a dense mat of fibers emanat- A few examples are highlighted in this section
ing from activated platelets or procoagulant par- that lead to either mechanistic insight or clini-
ticles [48, 54]. There is an inverse relationship cally relevant predictions.
between shear rate and fibrin deposition in vitro The computational power of a desktop com-
flow assays [64]. Neeves et al. described the puter or standard laptop can solve hundreds of
transport limitations of fibrin formation in a ODEs in a matter of minutes and, in some cases,
­purified system where thrombin emanates from a seconds. As such, ODE models are ideal for
wall at a known rate into a flowing stream of exploring how varying ranges of parameters
fibrinogen [44]. A PDE model of fibrinogen and affect model results. In other words, thousands of
thrombin concentration profiles shows that at computational experiments can be conducted in a
shear rates exceeding ∼ 10 s−1, fibrin deposition is very short amount of time. One such experiment
limited to very thin boundary layer near wall and is a sensitivity analysis, which is an extensive
that at shear rates of > 100 s−1 dilution of fibrin study of the sensitivity of model outputs to model
monomers is faster than the rate of gelation. Guy inputs. Analyses of this type would be either pro-
et al. used a kinetic gelation model, in combina- hibitively expensive or impossible in the labora-
tion with PDEs to describe the transport of pro- tory. For example, Danforth et al. examined the
thrombin, thrombin, and fibrinogen, to predict sensitivity of thrombin generation to the input
the height of fibrin gels formed under flow on a values of the rate constants in a modified version
wall capable of converting prothrombin to throm- of the Hockin–Mann model [12]. In this study,
bin [26]. At low shear rates, thrombin concentra- each of the 44 rate constants were varied by
tion limits gel growth such that with increasing one order-of-magnitude up and down from their
shear rate the dilution of thrombin reduces fibrin reported literature value. Results showed throm-
deposition. At high shear rates, fibrin deposition bin generation to be most sensitive to reactions
is limited by the ability of prothrombin to pene- involving the regulation of the formation and
trate through the existing fibrin gel to the surface. function of the TF:FVIIa complex. This identifi-
The penetration of solutes into and out of the gel cation points to where improvements in measure-
is a function of a physical property called the per- ment accuracy might be most important if the
meability, which is essentially a measure of the model is to have reliable predictive capabilities.
pore size of the gel. Gels with low permeability The same group used their ODE model to predict
have small pores that hinder the transport of how changes in plasma composition could affect
­solutes by both convection and diffusion. The thrombin generation [13]; this means they varied
permeability of fibrin gels varies over several initial concentrations (initial conditions to the
35  Mathematical Models of Hemostasis 579

2
10
a b
100

Initiation Time (min)


1
10 100
Thrombin (nM)

500 1500
0
10

-1
10
10

-2
10
Control
TF Titration
-3
10
0 10 20 30 40 50 60 0.1 fM 1 fM 10 fM 100 fM 1 pM 10 pM 100 pM

Tissue Factor fmol/cm2 Tissue Factor (pM)


c
1.40E+09 1.40E+09 1.40E+09
500 s-1
Fibrinogen (FI)

1.20E+09 100 s-1 1000 s-1


1.20E+09
EC50 = 145 pM EC50 = 336 pM 1.20E+09
EC50 = 409 pM
1.00E+09 3.63 TF molec/µm2 1.00E+09 8.40 TF molec/µm2 1.00E+09 2
10.2 TF molec/µm
8.00E+08 8.00E+08 8.00E+08
6.00E+08 6.00E+08 6.00E+08
4.00E+08 4.00E+08 4.00E+08
2.00E+08 2.00E+08 2.00E+08
0.00E+00 0.00E+00 0.00E+00
1.2 1.8 2.4 3 1.2 1.8 2.4 3 1.2 1.8 2.4 3

Tissue Factor (fM)


Fig. 35.5 Importance of biophysical mechanism. c­onfirmed in experiments performed with platelets and
Kuharsky–Fogelson [36] predicted that thrombin prod­ flow [46] (c) but not without [10] (b)
uction showed a threshold dependence (a) that was

ODEs) rather than the kinetic rate constants. As was able to provide kinetic explanations for
expected, most variation in the normal ranges did reduced thrombin production with hemophilia A
not significantly perturb thrombin dynamics, but and B and proposed the idea that platelets ‘pave
a few factors such as TFPI and prothrombin lev- over’ the subendothelium, covering enzyme
els had a significant influence. This analysis also complexes and physically inhibiting their activ-
identified pairs of factors with significant influ- ity. This mechanism, also confirmed experimen-
ence over thrombin generation. TFPI and anti- tally [28] implies and highlights the idea that
thrombin together were shown to be particularly platelets have an important anticoagulant role,
potent at changing clot time. Using patient not just their well-recognized procoagulant ones.
plasma levels as inputs into these models could This balance is the foundation of many of the
be a way to identify thrombotic risk. In a cohort models behaviors including the aforementioned
of 65 patients on warfarin, the three patients with TF threshold. Even more importantly, the TF
highest predicted thrombin generation rate had threshold was seen in experiments performed
thrombotic events. The model was also applied to with platelets and flow [46], but not without
cohort of patients anticoagulated with warfarin them [11] (see Fig. 35.5). These data emphasize
following atrial fibrillation and identified which the importance of biophysical factors on the
patients may be at potential risk for a thrombotic coagulation system and further promote the
event [23]. inclusion of them in mathematical models.
With their ODE model, Kuharsky and The Leiderman–Fogelson model was used in
Fogelson predicted that thrombin production will combination with a microfluidic vascular injury
show threshold dependence on the amount of model in a cohort of FVIII deficient patients [47].
exposed TF. This prediction was later confirmed Here, the model was critical in interpreting
in experiments with whole blood perfused of results, specifically the relative FXa generation
micropatterned TF [46]. In addition, the model by intrinsic and extrinsic Xase, that could not be
580 K.B. Neeves and K. Leiderman

K-L Model Pre-treatment


200 60
Post-treatment Post-treatment
Pre-treatment Pre-treatment
Average Thrombin (nM)

50
Control Control

Fbn Density (RFU)


150
40
50 um
100 30

20
Post-treatment
50
10

0 0
0 100 200 300 0 100 200 300 50 um
time (sec) time (sec)

Fig. 35.6  Modeling bypass therapy in hemophilia. A deposition pre- and post-treatment in comparison to a
patient with severe FVIII deficiency that had developed healthy control in a microfluidic vascular injury model.
inhibitors was treated with 90 μg/kg recombinant FVIIa Right: overlay of platelet (blue) and fibrin (green)
(rFVIIa). Left: predicted average thrombin concentration ­accumulation pre- and post-treatment on a collagen-TF
in the growing thrombus as a function of rFVIIa and FVIII surface following perfusion for 5 min at a wall shear rate
levels in the Leiderman–Fogelson model. Center: fibrin of 150 s−1

measured experimentally. The model was also hemostasis and thrombosis. Models in both areas
able to explain why platelet aggregation, but not have potential to answer different but important
fibrin deposition, was different between patients questions. Static models with lipids could be
with severe and moderate FVIII deficiencies used to probe the coagulation network and look
based on predicted thrombin concentrations. for synergy of factors when considering thera-
A decrease in platelet aggregate size corresponds peutics. An area of future research could be the
with the predicted drop in local thrombin concen- addition of a more sophisticated treatment of
trations that approach sub-threshold concentra- platelet function into a static model which would
tions of thrombin ( ∼ 1 nM) for activating PAR1. improve our understanding of the role and func-
Two patients received bypass therapy (rFVIIa), tion of platelet binding sites. Dynamic models
which resulted in a decreased lag time and show us that the biochemical, cellular, and fluid
increased cumulative fibrin deposition in the mechanical processes are truly and intricately
microfluidic model. The computational model intertwined. Because these models are more
predicts faster assembly of the TF:FVIIa complex closely related to the in vivo situation, they could
owing to a higher flux of FVIIa being delivered to potentially be used to improve the development
the surface compared to endogenous FVIIa levels of intravascular devices, therapeutic strategies
(Fig. 35.6). Both microfluidic and computational related to both the growth and dissolution
models suggest that treatment of FVIII deficiency of thrombi, and patient-specific therapies in
with rFVIIa could lead to prothrombotic risks in conjunction with imaging data and clinical
­
agreement with clinical observations [1]. phenotyping.
There are challenges and room for impro­
vement with both static and flow models. As
Future Challenges mentioned above, the ODE models of static
coagulation that mimic thrombin generation
There are two major research areas of mathemati- assays have only been validated under limited
cal models of hemostasis: static coagulation as in conditions. For example, in the Hockin–Mann
thrombin generation assays and in vivo events study, their model was shown to agree with
involving blood flow in the context of bleeding, empirical results where prothrombin levels were
35  Mathematical Models of Hemostasis 581

varied and the Bungay–Gentry model was shown receptor mutation. These are two examples of
to agree qualitatively with data from the same how multiscale models may have a future in
empirical study. However, the two models differ identifying phenotypes and response to therapies.
greatly. For example, the Bungay–Gentry model The geometry and fluid mechanics of most
includes (1) the positive feedback reaction where hemostasis models consider only the simplest
Xa activates TF:VII and generates more Xa and cases, specifically thrombus formation on the
(2) FIX activation by FXIa and FXI activation wall of a straight, flat, and rigid channel under a
by thrombin. The Hockin–Mann model does not constant, laminar flow. The in vivo situations
include any of these reactions, yet provides simi- are significantly more complicated since vessels
lar results. A useful and predictive model should have complex geometries, are elastic, and flow
be robust; it should be tested and validated under can be pulsatile or even turbulent [35]. There are
many differing conditions such as varying all ini- extensive modeling efforts of blood flow in large
tial coagulation factor concentrations including arteries, heart valves, stenoses, and other com-
those that mimic hemophilic conditions, and also plex geometries [42, 56, 60], however, these
can predict the effect of anticoagulant drugs. models have yet to be coupled to detailed coagu-
A major challenge in developing in vivo mod- lation or platelet models. Models presented here
els of coagulation and thrombus formation is are two-dimensional, and only recently have
integrating phenomena that occur over multiple numerical methods and computational power
length and time scales. Such “multiscale” prob- allowed for simulation of vessel scale, three-­
lems are common in biological systems and pose dimensional blood flow [16, 24].
their own unique computational challenges. For The models reviewed in this chapter all
example, a detailed model of thrombus formation describe a thrombus as a rigid body, that is, they
would include receptor-ligands kinetics, outside- do not deform with force. However, the primary
­in and inside-out signaling pathways of platelets, components of thrombi (fibrin, platelets, red blood
coagulation reactions, platelet shape change and cells) are elastic materials that have complex
retraction, fibrin polymerization, fibrinolysis, and stress–strain behavior [37, 67]. Moreover, plate-
the influence of forces and flows on each of these lets feel and response to their surrounds [53], but
processes. These phenomena span length scales there are no models that explicitly treat platelet
of molecules to tissue, and times scales of nano- retraction. The temporal evolution of thrombus
seconds to days. No one model can possibly cap- composition and mechanics over time scales rele-
ture the dynamics of all these processes, so one vant for fibrin formation and fibrinolysis has been
challenge is the passing of information between reported [32], but mechanical properties have yet
various submodels. In coagulation modeling, this to be incorporated into models with detailed treat-
has been done by Xu and colleagues as described ment of coagulation.
in section “Mathematical Models of Coagulation In summary, mathematical models of coagula-
and Fibrin Formation” using submodels for coag- tion have already made important contributions
ulation reactions, platelet function, blood flow, to the field of hemostasis in terms of revealing
and the interface between platelets and flow [68]. new mechanisms that regulate this complex, non-
Here, dynamics of blood flow are solved at the linear process. Some of these models are begin-
vessel scale and the interactions of platelets with ning to show promise in making clinical relevant
the vessel wall are solved at the cell scale. In predictions, but there is much work to be done in
platelet modeling, Flamm et al. used a neural terms of validating existing models against larger
­network to train a patient-specific intracellular data sets and experimental conditions. Moreover
calcium model coupled to stochastic and deter- it is unclear which mechanisms, and at what level
ministic models of fluid flow and solute trans- of detail, must be included in these models to
port [17]. Interestingly, the model identified a capture the essential dynamics of a system, be it
blood donor with indomethacin resistance that ex vivo or in vivo coagulation. Nevertheless,
turned out to have an unreported thromboxane multidisciplinary teams that include inputs from
582 K.B. Neeves and K. Leiderman

mathematicians, engineers, physical scientists, responses to combinatorial stimuli. Nat Biotechnol.


biologists, and clinicians are likely to generate 2010;28(7):727–32.
12. Danforth CM, Orfeo T, Mann KG, Brummel-Ziedins
models that yield outputs that can guide the KE, Everse SJ. The impact of uncertainty in a blood
development of therapeutic interventions and coagulation model. Math Med Biol. 2009;26(4):
cater these interventions to individual patients. 323–36.
13. Danforth CM, Orfeo T, Everse SJ, Mann KG,

Brummel-Ziedins KE. Defining the boundaries of
Acknowledgements This work was supported by NSF normal thrombin generation: investigations into
CAREER (CBET-1351672), American Heart Associa­ hemostasis. PLoS ONE 2012;7(2):e30,385.
tion (14GRNT20410094), and NIH (RO1HL120728, 14. Dashkevich NM, Ovanesov MV, Balandina AN,

R21NS082933). Karamzin SS, Shestakov PI, Soshitova NP, Tokarev
AA, Panteleev MA, Ataullakhanov FI. Thrombin
activity propagates in space during blood coagulation
as an excitation wave. Biophys J. 2012;103(10):
References 2233–40.
15. Edelstein-Keshet L. Mathematical models in biology.
1. Aledort LM. Comparative thrombotic event incidence New York: Random House; 1988.
after infusion of recombinant factor VIIa versus factor 16. Fedosov DA, Dao M, Karniadakis GE, Suresh S.

VIII inhibitor bypass activity. J Thromb Haemost: Computational biorheology of human blood flow in
JTH 2004;2(10):1700–8. health and disease. Ann Biomed Eng. 2013;42(2):
2. Balandina AN, Shibeko AM, Kireev DA, Novikova 368–87.
AA, Shmirev II, Panteleev MA, Ataullakhanov FI. 17. Flamm MH, Colace TV, Chatterjee MS, Jing H, Zhou
Positive feedback loops for factor V and factor VII S, Jaeger D, Brass LF, Sinno T, Diamond SL.
activation supply sensitivity to local surface tissue Multiscale prediction of patient-specific platelet func-
factor density during blood coagulation. Biophys tion under flow. Blood 2012;120(1):190–8.
J. 2011;101(8):1816–24. 18. Flamm MH, Diamond SL. Multiscale systems biol-
3. Beltrami E, Jesty J. Mathematical analysis of acti­ ogy and physics of thrombosis under flow. Ann
vation thresholds in enzyme-catalyzed positive Biomed Eng. 2012;40(11):2355–64.
feedbacks: application to the feedbacks of blood
­ 19. Fogelson AL, Keener JP. Toward an understanding of
coagulation. Proc Natl Acad Sci USA. 1995;92(19): fibrin branching structure. Phys Rev E 2010;81(5):
8744–8. 051,922.
4. Beltrami E, Jesty J. The role of membrane patch size 20. Fogelson AL, Neeves KB. Fluid mechanics of blood
and flow in regulating a proteolytic feedback thresh- clot formation. Annu Rev Fluid Mech. 2015;47(1):
old on a membrane: possible application in blood 377–403.
coagulation. Math Biosci. 2001;172(1):1–13. 21. Fogelson AL, Tania N. Coagulation under flow: the
5. Brummel-Ziedins KE, Orfeo T, Callas PW, Gissel M, influence of flow-mediated transport on the initiation
Mann KG, Bovill EG. The prothrombotic phenotypes and inhibition of coagulation. Pathophysiol Haemost
in familial protein c deficiency are differentiated by Thromb. 2005;34(2–3):91–108.
computational modeling of thrombin generation. 22. Fogelson AL, Hussain YH, Leiderman K. Blood clot
PLoS ONE 2012;7(9):e44,378. formation under flow: the importance of factor xi
6. Bungay S, Gentry P, Gentry R. Modelling thrombin depends strongly on platelet count. Biophys
generation in human ovarian follicular fluid. Bull J. 2012;102(1):10–18.
Math Biol. 2006;68(8):2283–2302. doi:10.1007/ 23. Foley JH, Orfeo T, Undas A, McLean KC, Bernstein
s11538-006-9115-7. http://dx.doi.org/10.1007/s11538- IM, Rivard GE, Mann KG, Everse SJ, Brummel-­
006-9115-7. Ziedins KE. From principle to practice: bridging the
7. Bungay SD, Gentry PA, Gentry RD. A mathematical gap in patient profiling. PLoS ONE 2013;8(1):e54,728.
model of lipid-mediated thrombin generation. Math 24.
Freund JB. Numerical simulation of flowing
Med Biol: J IMA 2003;20(1):105–29. blood cells. Annu Rev Fluid Mech. 2013;46(1):
8. Butenas S, van’t Veer C, Mann KG. “Normal” throm- 130812142306,005.
bin generation. Blood 1999;94(7):2169–78. 25. Guria K, Guria GT. Spatial aspects of blood coagula-
9. Butenas S, Orfeo T, Gissel MT, Brummel KE, Mann tion: two decades of research on the self-sustained
KG. The significance of circulating factor IXa in traveling wave of thrombin. Thromb Res. 2015;
blood. J Biol Chem. 2004;279(22):22,875–82. 135(3):423–33.
10. Chatterjee MS, Denney WS, Jing H, Diamond
26. Guy RD, Fogelson AL, Keener JP. Fibrin gel forma-
SL. Systems biology of coagulation initiation: kinet- tion in a shear flow. Math Med Biol. 2006;24(1):
ics of thrombin generation in resting and activated 111–30.
human blood. PLoS Comput Biol. 2010;6(9):e1000950. 27. Haberman R. Applied partial differential equations
11. Chatterjee MS, Purvis JE, Brass LF, Diamond SL. with Fourier series and boundary value problems, 5th
Pairwise agonist scanning predicts cellular signaling ed. Upper Saddle River: Pearson Education; 2013.
35  Mathematical Models of Hemostasis 583

28. Hathcock J, Nemerson Y. Platelet deposition inhibits of prothrombinase. J Biol Chem. 1984;259(3):
tissue factor activity: in vitro clots are impermeable to 1447–53.
factor Xa. Blood 2004;104(1):123–127. 46. Okorie UM, Denney WS, Chatterjee MS, Neeves KB,
29. Hockin M, Jones K, Everse S, Mann K. A model for Diamond SL. Determination of surface tissue factor
the stoichiometric regulation of blood coagulation. thresholds that trigger coagulation at venous and arte-
J Biol Chem. 2002;277(21):18,322. rial shear rates: amplification of 100 fM circulating
30. Jesty J. Positive feedbacks of coagulation: their role in tissue factor requires flow. Blood 2008;111(7):
threshold regulation. Arterioscler Thromb Vasc Biol. 3507–13.
2005;25(12):2463–9. 47. Onasoga-Jarvis AA, Leiderman K, Fogelson AL,
31. Jones KC, Mann KG. A model for the tissue factor Wang M, Manco-Johnson MJ, Di Paola JA, Neeves
pathway to thrombin. II. A mathematical simulation. KB. The effect of factor VIII deficiencies and replace-
J Biol Chem. 1994;269(37):23,367–73. ment and bypass therapies on thrombus formation
32. Karsaj I, Humphrey JD. A mathematical model of under venous flow conditions in microfluidic and com-
evolving mechanical properties of intraluminal putational models. PLoS ONE 2013;8(11):e78,732.
thrombus. Biorheology 2009;46(6):509–27. 48. Onasoga-Jarvis AA, Puls TJ, O’Brien SK, Kuang L,
33. Kastrup C, Shen F, Runyon M, Ismagilov R. Charac­ Liang HJ, Neeves KB. Thrombin generation and
terization of the threshold response of initiation of fibrin formation under flow on biomimetic tissue
blood clotting to stimulus patch size. Biophys factor-­rich surfaces. J Thromb Haemost: JTH 2014;
J. 2007;93(8):2969–77. 12(3):373–82.
34. Krasotkina YV, Sinauridze EI, Ataullakhanov FI.
49.
Ovanesov MV, Krasotkina JV, Ul’yanova LI,
Spatiotemporal dynamics of fibrin formation and Abushinova KV, Plyushch OP, Domogatskii SP,
spreading of active thrombin entering non-recalcified Vorob’ev AI, Ataullakhanov FI. Hemophilia A and B
plasma by diffusion. Biochim Biophys Acta 2000; are associated with abnormal spatial dynamics of clot
1474(3):337–45. growth. Biochim Biophys Acta 2002;1572(1):45–57.
35. Ku D. Blood flow in arteries. Annu Rev Fluid Mech. 50. Ovanesov MV, Lopatina EG, Saenko EL, Ananyeva
1997;29(1):399–434. NM, Ul’yanova LI, Plyushch OP, Butilin AA,
36. Kuharsky A, Fogelson A. Surface-mediated control of Ataullakhanov FI. Effect of factor VIII on tissue
blood coagulation: the role of binding site densities factor-­initiated spatial clot growth. Thromb Haemost.
and platelet deposition. Biophys J. 2001;80(3): 2003;89(2):235–42.
1050–74. 51. Ovanesov MV, Ananyeva NM, Panteleev MA,

37. Lam WA, Chaudhuri O, Crow A, Webster KD, Li TD, Ataullakhanov FI, Saenko EL. Initiation and propaga-
Kita A, Huang J, Fletcher DA. Mechanics and con- tion of coagulation from tissue factor-bearing cell
traction dynamics of single platelets and implications monolayers to plasma: initiator cells do not regulate
for clot stiffening. Nat. Mater. 2011;10(1): spatial growth rate*. J Thromb Haemost: JTH 2005;
61–6. 3(2):321–31.
38. Lawson JH, Kalafatis M, Stram S, Mann KG.
52. Panteleev MA, Ovanesov MV, Kireev DA, Shibeko
A model for the tissue factor pathway to thrombin. I. AM, Sinauridze EI, Ananyeva NM, Butylin AA,
An empirical study. J Biol Chem. 1994;269(37): Saenko EL, Ataullakhanov FI. Spatial propagation
23,357–66. and localization of blood coagulation are regulated by
39. Leiderman K, Fogelson AL. Grow with the flow: a intrinsic and protein c pathways, respectively. Biophys
spatial-temporal model of platelet deposition and J. 2006;90(5):1489–1500.
blood coagulation under flow. Math Med Biol. 53. Qiu Y, Brown AC, Myers DR, Sakurai Y, Mannino
2011;28(1):47–84. RG, Tran R, Ahn B, Hardy ET, Kee MF, Kumar S,
40. Leiderman K, Fogelson AL. The influence of hin- Bao G, Barker TH, Lam WA. Platelet mechanosens-
dered transport on the development of platelet thrombi ing of substrate stiffness during clot formation medi-
under flow. Bull Math Biol. 2013;75:1255–83. ates adhesion, spreading, and activation. Proc Natl
41. Luan D, Zai M, Varner JD. Computationally derived Acad Sci. 2014;111(40):14,430–35.
points of fragility of a human cascade are consistent 54. Reininger AJ, Bernlochner I, Penz SM, Ravanat C,
with current therapeutic strategies. PLoS Comput Smethurst P, Farndale RW, Gachet C, Brandl R, Siess
Biol. 2007;3(7):e142. W. A 2-step mechanism of arterial thrombus forma-
42. Marsden AL. Optimization in cardiovascular model- tion induced by human atherosclerotic plaques. J Am
ing. Annu Rev Fluid Mech. 2014;46(1):519–46. Coll Cardiol. 2010;55(11):1147–58.
43. Monroe D, Hoffman M. What does it take to make the 55. Segel LA, Edelstein-Keshet L. A primer on mathe-
perfect clot? Arterioscler Thromb Vasc Biol. 2006; matical models in biology. Society for Industrial and
26(1):41. Applied Mathematics. Philadelphia, PA (2013).
44. Neeves KB, Illing DAR, Diamond SL. Thrombin flux 56. Shadden SC, Taylor CA. Characterization of coherent
and wall shear rate regulate fibrin fiber deposition structures in the cardiovascular system. Ann Biomed
state during polymerization under flow. Biophys Eng. 2008;36(7):1152–62.
J. 2010;98(7):1344–52. 57. Shen F, Kastrup CJ, Liu Y, Ismagilov RF. Threshold
45. Nesheim ME, Tracy RP, Mann KG. “Clotspeed,” a response of initiation of blood coagulation by tissue
mathematical simulation of the functional properties factor in patterned microfluidic capillaries is
584 K.B. Neeves and K. Leiderman

c­ontrolled by shear rate-supplement. Arterioscler quantitative and qualitative platelet defects. J Clin
Thromb Vasc Biol. 2008;28(11):2035–41. Invest. 1986;78(4):1072–82.
58. Stalker TJ, Traxler EA, Wu J, Wannemacher KM, 65. Willems GM, Lindhout T, Hermens WT, Hemker

Cermignano SL, Voronov R, Diamond SL, Brass LF. HC. Simulation model for thrombin generation in
Hierarchical organization in the hemostatic response plasma. Haemostasis 1991;21:197–207.
and its relationship to the platelet-signaling network. 66. Wufsus AR, Macera NE, Neeves KB. The hydraulic
Blood 2013;121(10):1875–85. permeability of blood clots as a function of fibrin and
59. Strauss WA. Partial differential equations: an intro- platelet density. Biophys J. 2013;104(8):1812–23.
duction, 2nd ed. Wiley; 2008 Hoboken, NJ. 67. Wufsus AR, Rana K, Brown A, Dorgan JR, Liberatore
60. Taylor CA, Draney MT. Experimental and computa- MW, Neeves KB. Elastic behavior and platelet retrac-
tional methods in cardiovascular fluid mechanics. tion in low- and high-density fibrin gels. Biophys
Annu Rev Fluid Mech. 2004;36(1):197–231. J. 2015;108(1):173–83.
61. Wang W, King MR. Multiscale modeling of platelet 68. Xu Z, Chen N, Kamocka MM, Rosen ED, Alber M. A
adhesion and thrombus growth. Ann Biomed Eng. multiscale model of thrombus development. J R Soc
2012;40(11):2345–54. Interface 2008;5(24):705–22.
62. Weisel J. Fibrinogen and fibrin. Adv Protein Chem. 69. Xu Z, Chen N, Shadden SC, Marsden JE, Kamocka
2005;70:248–299. MM, Rosen ED, Alber M. Study of blood flow impact
63. Weisel JW, Nagaswami C. Computer modeling of on growth of thrombi using a multiscale model. Soft
fibrin polymerization kinetics correlated with elec- Matter 2009;5(4):769–79.
tron microscope and …. Biophys J. 1992;63: 70. Xu Z, Lioi J, Mu J, Kamocka MM, Liu X, Chen DZ,
111–28. Rosen ED, Alber M. A multiscale model of venous
64. Weiss HJ, Turitto VT, Baumgartner HR. Role of shear thrombus formation with surface-mediated control of
rate and platelets in promoting fibrin formation on blood coagulation cascade. Biophys J. 2010;98(9):
rabbit subendothelium. Studies utilizing patients with 1723–32.

You might also like