You are on page 1of 31

BIOCHEMICAL SEPARATIONS

Adsorption and Chromatographic separations Dr John Hubble


Introduction
This course will consider the recovery of proteins from dilute solutions by partition
from a liquid to a solid (adsorbent) phase. Many of the concepts described will also
apply to the recovery and separation of smaller molecules which is covered in more
detail in the Chemical Separations module. The course will start by considering the
types of partition phenomena which can be employed in protein separations. This will
lead on to a quantitative assessment of adsorption where loading and elution are
operated as sequential process steps and will include descriptions of batch separations
in stirred tanks and packed bed adsorption. Finally, chromatographic separations will
be discussed where the partition conditions cause adsorption/desorption to occur
simultaneously such that materials progress through a packed bed at different rates.
It should be stressed that the complexity of biochemical separations dictates a largely
empirical approach. Design equations serve a valuable role in the design of pilot scale
evaluations, but cannot, as yet, be expected to fully describe the performance of a
industrial separation.
Background
Basis of separation
Most protein recovery processes will require that a specific protein product be
selectively removed and concentrated from a complex mixture usually containing a
number of unwanted proteinaceous by products. Therefore techniques are required
which are capable of separating broadly similar compounds on the basis of the
chemical and physical properties. The properties providing the basis for existing
processes are listed below:
Chemical properties
Charge
Hydrophobicity
Biological function
Surface chemistry
Physical properties
Size
Separations which are based on these characteristics vary in cost capacity and
selectivity, factors which determine their role and position in a separation sequence.
Support conformation
1
Adsorption and chromatographic separations are based on the partition of material
between a mobile and a stationary phase. In this section of the course the focus will be
on the use of solid stationary phases. Choice of support conformation results from a
compromise between kinetics and capacity. The use of non porous supports leads to
rapid kinetics, however, surface area to volume considerations dictate small particle
diameters if reasonable capacity is to be achieved. This in turn leads to high pressure
drops in packed bed operation, this may be acceptable for analytical and small scale
separations (HPLC and FPLC) but is usually unsuitable for large sale processes.
Porous materials offer much higher capacities, but at the expense of slower binding
kinetics. In addition the open structure of many porous materials further limit their
mechanical stability applying additional constraints on the pressure drop achievable in
packed bed operation. Despite these constraints porous bead supports are the most
widely used in protein separations.
Support materials
In practice the physical requirements for the stationary phase will be similar for all of
the separation options detailed above. Thus a single base resin type can be used for
any separation, all that is required is that it is derivatised to provide the appropriate
chemical environment for the separation to be used. This implies that the base resin
should be inert with respect to interactions with proteins. i.e. it should be hydrophilic,
it should not be charged, and it should not contain aromatic groups which could lead
to unwanted (i.e. non specific) hydrophobic interactions.
In addition to these chemical requirements there are a number of physical requirements
which are important, these include pore structure, particle size and mechanical
stability. In practice no single material will be optimal in all respects and so choice of
resin will be a compromise between a number of materials offering specific
advantages/disadvantages.
The following materials have been used:
Agarose Agarose is a galactose containing polysaccharide derived from sea weed
it has a high gel strength and can be cast into beads of different sizes
most common are the preparations from Phamacia i.e. Sepharose 4B,
6B (4 & 6 denote the agarose content of the beads (% w/v)). These
beads have an open structure that allow most proteins free access to the
internal pores. Their mechanical stability can be improved by chemical
cross linking.
Dextran Dextran is a linear polymer of glucose produced by certain bacteria. It
can be cast into beads and crosslinked using epichlorhydrin. The beads
have a less open structure than agarose and can restrict access of larger
proteins. The most common form are Sephadex again from Pharmacia,
which are used for size based separations.
Cellulose Another polymer of glucose. This is not widely used for proteins as its
pore structure is ill defined and it has poor flow properties in packed
beds.
Trisacryl These are purely synthetic gels. They have good biological stability, are
hydrophilic and can be cast in a range of pore sizes
2
Polyacrylamide
Another synthetic gel they have good biological stability but are
mechanically weak. They are produced by Bio Rad for size based
separations.
Hydroxylalkyl Methacrylate
Synthetic gels with good chemical and physical properties. Produced
under the trade name Spheron by Realco Chemical co.
Applications
Applications will be considered under the categories of charge, hydrophobicity,
biological function and surface chemistry and size.
Ion Exchange (charge)
The basis of ion exchange adsorption is provided by the ability of two substances to
dissociate in solution and to exchange ions. If one of these substances is insoluble and
one of the ions which is exchanged is the product this allows adsorption.
In its simplest form ion exchange can be described as follows:
M X N Y N X M Y
+ + + +
+ + + +
This represents the interaction between two dissociable salts in solution. If X were to
represent an insoluble support then the ions exchanged are M and N, both of which
carry a positive charge (positive charges migrate to the negative electrode (cathode)
hence are known as cations). Therefore this would be a cation exchange support. If
negative ions are exchanged the support would be an anion exchanger.
While the situation with simple salts in dilute solution is straight forward in that both
are likely to be fully dissociated the interaction between ion exchange groups
supported on a beaded resin interacting with a protein is more complex. The extent of
dissociation and the net charge carried by the protein will be a function of pH relative
to the isoelectric point of the protein. Hence by changing the pH we can cause a
protein to exchange with ions on an exchange resin to induce both adsorption and
desorption.
Three ion exchange groups are commonly used in protein separations and can be
chemically substituted onto all of the beaded resin materials described above:
CM carboxymethyl COOH
DEAE diethylaminoethyl (CH
2
)
2
N(CH
2
H
5
)
2
SP Sulphur propyl C(CH
3
)
2
CH
2
SO
3
H
These are characterised in terms of the ion exchanged, and the strength of acid or base
required to prevent their dissociation, hence there are strong and weak examples
of both anion and cation exchangers. In practice the pH stability of proteins precludes
the use of most strong ion exchangers.
The nature of the ion exchange interaction means that there are three ways of eluting a
bound protein.
3
(i) Change the pH
(ii) Increase the salt concentration to swamp the charge (can be at the same pH)
(iii) Include a competing ion having the same charge but which has a higher affinity
for the exchanger.
Hydrophobic interaction
Proteins hydrophobicity can vary significantly depending on the amino acid
composition and structural conformation. In practice most proteins are only
marginally soluble in aqueous solution such that relatively small changes in salt pH or
solvent concentration can cause precipitation (and provide the basis for a number of
separation protocols). However a potentially attractive alternative to precipitation is
to use differences in hydrophobicity as a basis for an adsorption based separation.
Supports for hydrophobic interaction chromatography have usually been based on the
Sepharose beaded agarose supports to take advantage of their extremely low level of
intrinsic hydrophobic sites. These base materials are substituted with a number of
groups of varying hydrophobicity. These are usually based on aliphatic chains of
varying lengths with different terminal substitutions but also include a phenyl
derivative. Thus the hydrophobicity can be tailored to the protein target by variation
of aliphatic chain length allowing a high level of control.
Unlike ion exchange, hydrophobic interactions are enhanced by elevated salt
concentrations. This allows a separation protocol based on binding at high salt
concentration followed by elution by a reduction in buffer ionic strength. As the level
of protein hydrophobicity is influenced by its conformation any agent which influences
conformation can potentially be used as eluents. These include salt, pH, solvents, and
partial denaturants.
Affinity interactions
Affinity interactions are based on biological recognition and the extreme selectivity
displayed by these phenomena, which include:
Antibody/antigen
Enzyme/substrate
Lectin/sugar
Neurotransmitter/receptor
Specific binding proteins.
These interactions are widely explained in terms of the lock and key hypothesis
where there is a highly specific binding site which recognises the 3 dimensional
conformation of the molecule to be bound and the presence and orientation of key
chemical groups within this region.
Of all the adsorption phenomena discussed here, these are the most highly selective,
and in many cases offer the tightest binding i.e. lowest dissociation constants. In
addition our ability to manipulate the mammalian immune system means that we can
now tailor adsorbing species to recognise any given protein and a wide range of
smaller compounds.
4
It is normal to describe the small molecule as the ligand in affinity interactions. Hence
appropriate ligands would normally be attached to a suitable support (e.g. Sepharose)
to form an affinity support. However, in some cases (e.g. antibodies and lectins) the
molecule to be attached to the support will also be a protein.
The conditions for the affinity binding will be dependent on the properties of the
biological interaction, with options for elution conditions being similar to those use to
displace hydrophobic interactions. However, in the case of biospecific interactions
there is the additional possibility to use competing free ligands to displace bound
protein.
Immobilised metal ion adsorption (IMAC)
In some cases a highly selective adsorption can be generated via a metal ion mediated
coordination complex between protein and support. The support is produced by
attaching iminodiacetic acid to the support such that there are two dissociable
carboxylic groups capable of forming complexes with metal ions (usually divalent)
including Cu, Ni, Zn, Co, Ca and Mg. These form complexes via specific amino acids
on the protein surface e.g. histidine and cysteine. While perhaps not as widely
applicable as affinity adsorption this techniques is again capable of combining high
selectivity based on differences in amino acid availability with tight binding which can
be controlled by choice of appropriate metal ion.
Size exclusion chromatography (gel filtration)
Unlike the previous methods size exclusion chromatography is based solely on the
physical dimensions rather than the chemistry of the molecules to be separated. The
technique is based on the use of a porous hydrophilic inert support, such that the
potential for chemical interactions between protein and support are minimised. The
most common supports are based on chemically cross-linked dextrans whose synthesis
can be controlled to give a tightly defined pore size distribution. Separation in this
case must be conducted in a column or packed bed configuration (unlike adsorption
which can also be carried out in stirred tanks) as it is based on differences in the
partition coefficients between molecules of different size between the liquid and the gel
phase. The sample is applied to the column as a discrete band and is washed through
the column using a suitable buffer as the protein mixture moves down the column the
smaller molecules are able to access a larger fraction of the gel volume and move more
slowly than material which is completely excluded. This means that the degree of
separation obtained is in part a function of column length.
Adsorption isotherms
An adsorption isotherm describes the relationship between liquid phase concentration
and resin concentration of adsorbate at equilibrium. A wide range of isotherms have
been proposed to describe adsorption but the most common are the Freundlich and
Langmuir isotherms.
Favourable/Unfavourable isotherms
Both Langmuir and Freundlich are examples of favourable isotherms i.e. a plot of q
*

vs c
*
has a convex shape such that q
*
>>c
*
at low concentrations. This allows effective
5
recovery from dilute streams and minimises the unadsorbed losses in an adsorption
process. An unfavourable isotherm is concave in shape i.e. q
*
>>c
*
only at high liquid
phase concentrations such that effective adsorption from dilute streams is not possible.
The Freundlich isotherm
The Freundlich isotherm is a purely empirical relationship used for systems that are not
well described by the Langmuir equation. The Freundlich takes the form
q Kc
m * *


where K & m are empirical parameters determined from experimental data obtained
from the system of interest.
q
*
the occupied resin capacity at equilibrium (g/l of resin)
c
*
adsorbate concentration in solution at equilibrium (g/l)

The relationship between q
*
and c
*
is not linear even at low concentrations.
Langmuir isotherms
The Langmuir isotherm was originally derived for gas adsorption and described the
progressive coverage of an adsorption surface by gas molecules up to the limiting point
of a monolayer coverage.
Although the concept of monolayer coverage is not applicable to protein adsorption an
analogous derivation can be made in terms of a maximum binding capacity instead of a
limiting monolayer value.
A mass balance for adsorbent capacity can be written:
q q q
m u
+
* *
The dissociation constant for the interaction is given by:
K
q c
q
d
u

* *
*
Substitution and rearrangement gives:
q
q c
K c
m
d
*
*
*

+
where q
m
maximum resin capacity (g/l of resin)
q
u
*
the unoccupied resin capacity at equilibrium i.e. q
m
-q
*
(g/l of resin)
K
d
the dissociation constant given by the ratio of off/on constants )g/l)
k
1
formation or on constant (l g
-1
s
-1
)
k
-1
breakdown or of constant (s
-1
)
6
This shows saturation as q
m
is approached, but at low solution adsorbate
concentrations (i.e. c
*
<<K
d
) the Langmuir isotherm simplifies to a linear form (i.e.
Henrys Law):
q
q
K
c
m
d
* *

Although the linear form allows significant simplification of the mathematical


description of column operation, in practice feed concentrations are likely to exceed K
d
for most preparative applications,
Langmuir isotherms in Ion Exchange
The description of ion exchange is some what more complex than the other forms of
adsorption in that the counter ion concentration necessary to maintain electroneutrality
must be accounted for. In the simplest case of two monovalent ions:
A S A S
ads ads
+ +
where A is the feed component to be adsorbed
S is the salt ion to be exchanged
ads
denotes which species is adsorbed
7
Using the nomenclature above c
A
is the liquid phase concentration of A and q
A
is the
stationary phase concentration. Electroneutrality requires that:
q q
S A
+
where is the total concentration of binding sites (fixed charges) on the ion
exchanger.
This derivation has been based on the replacement of activities by concentrations and
assumes that the binding process results from a stoichiometric exchange of counter
ions. In this analysis q
s
is effectively equivalent to the unoccupied capacity (q
m
-q) in an
adsorption system where exchange does not take place.
It also assumes that the equilibrium constant for ion exchange is constant:
K
q c
c q
a
S A
S A

Substitution into the balance equation gives:


q
c
K c c
A
A
A S A

A more realistic situation with proteins is where a single protein molecule caries a
number of charges and hence displaces a number of monovalent ions from the ion
exchanger. If the valency is

then the exchange can be described as follows:


A S A S
ad ads
+ +
The electroneutrality condition is
q q
S A
+
and the equilibrium constant is given by
( )
K
c q
q c
a
A A
A s

It is not possible to derive an explicit isotherm equation in this case and so solutions
have to be obtained numerically.
Multicomponent adsorption
While single component adsorption can be used for adsorbent characterisation studies
in most separation processes of interest there will be components other than the
compound of interest which will adsorb. Characterisation of extent to which these
different species are bound will determine the extent to which separation is achievable
in a chromatographic system, and will determine the extent to which the concentration
of contaminants limit the effective capacity of adsorption systems.
8
The multicomponent Langmuir isotherm can be derived by a simple extension to the
single component approach. Taking a two component system (A & B)the unused resin
capacity is now given by the difference between the maximum capacity and the sum of
the capacities occupied by the two components:
q q q q
m A B u
+ +
* * *
The dissociation constants are:
K
q c
q
A
u A
A

* *
*
K
q c
q
B
u B
b

* *
*
Using these definitions to eliminate q
B
*
and q
u
*
from the balance equation gives:
q
q
K c
c K
K
c
q c
c K
c
K
A
m
A B
A B
A
A
m A
A A
B
B
*
*
* *
*
*
*

+ +

+ +

_
,

1
1
As the two components effectively compete for sites on the adsorbent surface it is
not surprising that this equation mirrors the equation for competitive enzyme
inhibition.
The multicomponent isotherm can be expanded to the case of n adsorbing species to
allow prediction of the bound concentration of the ith component:
q
q c
c K
c
K
i
m i
A A
j
j j
n
*
*
*
*

+ +

_
,

1
1
In most preparative adsorption applications it would be usual to consider two
components, namely product and contaminants. Thus an empirical value for the
apparent contaminant dissociation constant can be used for design purposes.
Gel partition
Unlike the adsorption isotherms described above gel partition (the basis for size
exclusion chromatography) is a linear process. The porous gel particles can be
assumed to be essentially water (Sepharose 4B 4% w/v agarose) such that a
9
suspension of hydrated beads in water can be considered to consist of two immiscible
aqueous phases. The ability of a protein to partition between these two phases is
dependent on the relationship between the hydrodynamic radius of the protein and the
pore size in the beads. The partition coefficient is given by:
K
c
c
p
g
l

thus
c K c
g p l

which is analogous to a linear adsorption isotherm.


Process theory
Adsorption
Adsorption is the central unit operation in the downstream processing of protein
products. It offers the possibility of both purification and concentration in a single
step. Adsorption can be based on a number of chemical properties as described above
and for effective separations needs a number of discrete process steps:
i) Loading - to load the adsorbent by adsorbing the required product(s).
ii) Washing - to wash the adsorbent to remove, or at least reduce the quantity of, non-
adsorbed contaminants.
iii) Elution - to change conditions such that the binding interaction is displaced and
the product can be washed from the adsorbent.
iv) Regeneration - in the case of ion-exchange supports an acid or base treatment
to ensure that the charge groups have the correct counter ion prior to the next
loading cycle.
The requirements and constraints imposed vary between these process steps and it is
important that the choice of a resin for a given application is not based solely on
loading conditions.
The most common mode of application is to used a packed bed of adsorbent.
However, in some cases stirred contactors are used and this provides a logical starting
point for the consideration of adsorption theory. In fact many modelling approaches
for adsorption column design are based on constants determined from data obtained in
stirred tank experiments.
Stirred tank (well mixed contactors)
Equilibrium considerations
10
The mass balance for a well mixed vessel containing a fixed adsorbate charge with a
fixed mass of suspended adsorbate can be formulated to give the equilibrium
concentration of adsorbate remaining in solution:
c c
mq
V
o *
*

where c
o
is the initial or total adsorbate concentration in the tank
m is the mass of adsorbate used
V is the total tank volume (liquid + adsorbent)
The design equation for the adsorber is given by combining the mass balance with the
appropriate binding isotherm. Using the Langmuir form this gives:
( )
c c
m
q c
K c
V
o
m
*
*
*

+
solving for c
*
gives a quadratic with the physically meaningful root.
( )
c
b b c K
o
*

+ +
2
4
2
where b K
m
V
q c
m
o
+

_
,

A similar relation ship can be derived for q


*
such that both the occupied adsorbent
capacity and the adsorbate concentration remaining in solution at equilibrium can be
determined. It should be stressed that an adsorption isotherm is the relationship
between q
*
and c
*
, not q
*
and c
o
.
A graphical solution of this problem can also be constructed. Solving the mass balance
equation for q*:
( ) q q
V
m
c c
o o * *
+
where q
o
is the adsorbate capacity which is occupied prior to a change of conditions
(normally zero for loading)
This represents the operating line which will be linear. The intersection of the
operating line with the equilibrium line gives the equilibrium conditions which will
result from the initial conditions chosen.
_____________________________________________________________________
Worked example 1
Determination of adsorption isotherms (Langmuir form)
Results of a stirred tank adsorbent characterisation were as follows:
11
c
o
c* (g/l)
1.00000000 0.11
2.00000000 0.4
3.00000000 1.02
4.00000000 1.86
5.00000000 2.80
6.00000000 3.76
The solution volume (V) was 50 cm
3
The resin volume (v) was 1 cm
3
Determine q
max
and K
d
Mass balance for the tank
c c
vq
v V
o
+
+
*
*
so
( )
( )
q
v V c c
v
o
*
*

+
Thus
c* q*
0 0
0.114626 45.15408
0.402099 81.49294
1.017342 101.1155
1.864643 108.9032
2.79674 112.3663
3.760232 114.2282
0
20
40
60
80
100
120
0 1 2 3 4
c*
q
*
This data can be plotted in double reciprocal form to allow determination of binding
constants (q
max
=120 g/l, K
d
=0.19 g/l)
_____________________________________________________________________
Worked Example 2
12
Using the data from the previous example the percentage recovery which would be
obtained if 50 l of adsorbent were added to 0.5m
3
of solution containing 10g/l of
protein.
Graphical solution
The equilibrium line is given in the graph for example 1. The operating line is
( ) q c
* *

550
50
10
0
20
40
60
80
100
120
0 1 2 3 4
c*
q*
Operating line
Equilibrium line
So the equilibrium concentration is .95 g/l
Percentage recovery =
( ) 10 95
10
100
.
.
13
Numerical solution
From combing isotherm with mass balance
( )
c
b b c K
o
*

+ +
2
4
2
b K
m
V
q c
m
o
+

_
,

b +

_
,
.19
50
550
120 10 =1.1
( )
c
*
. . .

+ + 11 11 4 10 019
2
2
= 0.934
Percentage recovery =
( ) 10 934
10
100
.
.
_____________________________________________________________________
These relationships provide the equilibrium design equations for the loading stage
where the adsorbent capacity would normally be assumed to be totally available. With
a little modification the can also give an indication of the effects of washing on product
loss.
The total adsorbate mass in the tank is given by:
T c V q m +
* *
So if the tank is drained once loading has reached equilibrium and the liquid phase is
replaced with an equal volume of buffer the total adsorbate mass at the start of
washing will be determined by the equilibrium bound concentration on the resin and
the mass of resin. If we denote this as q
o
for the washing stage we can calculate the
new equilibrium which will form as some of the product dissociates:
mq Vc mq
o
+
* *
Substituting for q* in terms of the isotherm equation gives.
m
V
q c
m
V
q c
K c
o m
+
+
*
*
*
This can be solved for c
*
to give the concentration of product which will be lost during
washing as a result of equilibrium displacement.
c
b b
m
V
q K
o
*

+ +

_
,

2
4
2
where
14
( ) b K
m
V
q q
m
o
+

_
,

Again both numerical and graphical solutions are possible


The same equations can be used to determine the extent of elution, however, elution is
based on a change of conditions to reduce the binding interaction thus the values of K
and q
m
would be different.
_____________________________________________________________________
Worked Example 3
Taking the loading achieved in example 2 calculate the fractional loss of adsorbed
material that would result if the resin was drained and washed in washed in 0.5 m3 of
fresh buffer.
Solution
At equilibrium c*=0.95g/l
Therefore from the mass balance for loading
( ) q
V
m
c c
o * *

( ) q
*
.
550
50
10 95 = 99.5 (This could also be read directly from the graphical
solution)
This value becomes q
o
for the washing step. For washing c
o
is zero. The equation for
the operating line is.
( ) q q
V
m
c c
o o * *
+
( ) q c
* *
. + 995
550
50
0
0
20
40
60
80
100
120
0 1 2 3 4
c*
q*
Operating line
Equilibrium line
Numerical solution
15
( ) b +

_
,
. . 19
50
550
120 995 =2.054
c
*
. . . .

+ +

_
,
2 054 2 054 4
50
550
995 019
2
2
= 0.64
Amount lost = 0.64 x 500
Amount bound = 99.5 x 50
Percentage loss = 6.4
_____________________________________________________________________
Kinetic considerations
While equilibrium calculations are useful in providing preliminary estimates of
adsorbent capacities and comparative assessments of resin suitability, any realistic
design calculations must take account of process kinetics. Conceptually the simplest
system to describe kinetically is the affinity interaction of a ligand with a binding
protein. The differential equation for the interaction can be written in terms of a
mutual depletion model:
dq
dt
k q q c k q
m

1 1
( )
at equilibrium the forward and reverse rates are equal and the equation simplifies to the
Langmuir form as derived earlier. As written, this equation cannot be integrated with
respect to time as both c and q are time dependent variables. To allow integration c
can be eliminated using the mass balance equation.
( )
dq
dt
k q q co q k q
m

1 1
( )
where

= m/V.
This equation can now be integrated to allow calculation of q (and c) as a function of
time using the boundary conditions t=0 q=q
o
and t=t q=q
t
.
( ) ( ) ( ) ( )
( )
( )
( ) ( )
( )
q
b x k q b x b x k q b x e
k e k q b x k q b x
t
o o
t x
t x
o o

+ + + +
+ + +
2 2
2 2 2
1 1
1 1 1


where ( ) x b k c q
o
m

2
1
2
4 and ( ) b k c k q k
o
m
+ +
1 1 2

Substitution of q
t
into the balance equation also allows determination of c
t
. These
expressions not only represent the design equations for a well mixed adsorber, they
also provide a mathematical description of the interaction of two soluble components.
In this case both c and q would have units of concentration and

=1. In fact for


some affinity separations it is possible to measure the binding kinetics to ligands in free
solution, as a soluble macromolecular conjugate, attached to a non porous particle and
16
attached to a porous beads. Data we have obtained for the enzyme LDH interacting
with the ligand Cibacron Blue suggest the following times are required for binding to
reach equilibrium:
Free dye <1 ms
Dye dextran 100ms
Non porous bead (0.05mm) 1 minute
Non porous tentacle 5 minutes
Porous bead 20-30 minutes
Clearly the rate constants will not be the same even if the same equations can be used
for all data sets. The difference in the time constants for the interaction result from
differences in mass transfer coefficients. The free dye conditions represent the free
ligand case where the on constant is largely determined by the molecular diffusivity
of the ligand (mw <1000). The dye dextran is also a soluble system where dye-dextran
is the ligand. However in this case the molecular weight is higher (>50,000) resulting
in a lower diffusivity. Once the ligand has been coupled to a nonporous support the
system becomes heterogeneous and will be limited by external film diffusional
limitations. Finally with porous beads the rate will be limited by both internal and
external diffusional resistances.
So the relationships derived above are only rigorously applicable to the soluble ligand
(homogeneous) case. However, we can lump the effects of kinetic and mass transfer
limitations to give apparent kinetic constants such that the simple mutual depletion
model can be used for scale up predictions provided the limitations are recognised.
This means that care must be taken to ensure that the conditions used to obtain
empirical values for the constants match those to be used on the larger scale.
Chromatography
Chromatography is a separation process which relies on differential partition of a
solute between a mobile and a stationary phase. The properties of the stationary
phases and the physical bases for partition are as described above. Chromatographic
separations are normally divided into two types. The first, frontal chromatography,
represents a column based adsorption in that the resin in the column is loaded to
saturation, prior to separate washing and elution steps. In the second case, elution or
zonal chromatography, the sample is applied to a column as a discrete slug.
Conditions are chosen such that differences in the partition coefficients of the species
of interest are such that they progress through the column at different rates. In elution
chromatography there is no separate wash and elution stages, although the column
may need regeneration between batches.
Frontal chromatography
Frontal chromatography is the most widely used mode for large scale separations. It
allows far more extensive use of resin capacity and hence allows smaller column
dimensions. Depending on the specificity characteristics of the resin, the aim can
either be to selectively adsorb the protein required, wash to remove impurities and then
elute (Affinity chromatography). Alternatively some or all of the protein species can
be adsorbed, and the adsorbed proteins induced to desorb sequentially by changing the
composition of the feed stream (Ion exchange and hydrophobic interaction
chromatography).
17
Rate theory of column operation
Rate theories are based on sets of material balance equations together with appropriate
boundary conditions. The continuity equation for the mobile phase has been
formulated as:
D
c
x
u
c
x
q
t
c
t
x

2
2
1

_
,

where D
x
is the axial dispersion coefficient
u is the linear velocity
x is an axial element of the column
This expression is further complicated by the term dictating the rate of removal from
solution. Which in its simplest form for a non linear isotherm would be the mutual
deletion expression we used to describe stirred tank adsorption.
i.e.
dq
dt
k q q c k q
m

1 1
( )
The complexity of the composite expression increases if we attempt to explicitly
account for diffusional limitations. Even at the simplest level simplifications are
needed if an analytical solution is to be obtained. For numerical solutions the extent
to which simplifications are made is dependent on the computational demands, and the
feasibility of determining values for all of the parameters required.
Analytical solution
An analytical solution for a column with a mutual depletion adsorption was first
derived by Thomas for Ion exchange and adapted by Chase for Affinity adsorption.
This is based on the assumption that the axial dispersion coefficient (i.e. D
x
) is zero:
( ) ( )
[ ]
c
c
J
n
r
nT
J
n
r
nT J n
nT
r
r n nT
o

_
,

_
,
+

_
,

1
]
1

,
, , exp 1 1
1
where
r c K
o
+ 1 /
n q k
h
u
m

1
T ut K c q h
o m
+ ( )
h = column height
t = time
18
( ) J ,
is an integral over a Bessel function which can be approximated by the
following expression when
&
are large:
( )
[ ]
( )
[ ]
( )
J erf ( , )
exp



+

+

1
]
1
1
2
1
1
2
2
1
2
1
4
1
2
These relationships allow the elution profile of a bed to be predicted from values of q
m

K and k
1
determined from small scale stirred tank experiments.
Tanks in series solution
In reaction engineering it is common practice to relate a plug flow reactor to a series of
well mixed vessels. As an adsorption is analogous to a chemical reaction a similar
approach can be used for an adsorption column. The column can be divided in to n
reaction vessels such that the residence time in each vessel can be calculated from
vessel height voidage and linear velocity. The feed is treated incrementally as
elements equivalent to the flow velocity multiplied by the tank residence time. Using
the batch adsorption relationship derived earlier it is possible to calculate the bound
adsorbate concentration for the stage residence time.
( )( ) ( ) ( )
( )
( )
( ) ( ) ( ) x b q k x b q k e k
e x b q k x b x b q k x b
q
o o x t
x t o o
t
+ + +
+ + + +



1 1 1
1 1
2 2 2
2 2
where ( ) x b k c q
o
m

2
1
2
4 and ( ) b k c k q k
o
m
+ +
1 1 2

In this case q
o
will be the adsorbed concentration from the previous loading step while
c
o
will be the liquid phase adsorbate concentration passing from the previous column
stage (or tank).
Using j as loading step index and i as the column stage index the mass balance for
stage i at loading step j can be written as follows:
( ) ( ) ( )
c c q
j i
o
j i j i , , ,
+
1 1 1

Thus a flow sheet for the calculation procedure can be written as follows:
19
Set model parameters
Repeat for new time step
Calculate initial stage values for c
& q
Solve rate equation for stage
residence time
Update stage values for c & q and
store
Proceed to next stage until outlet
is reached
Repeat for number of stages
Store outlet concentration & time
Reapeat until c/cf >= 0.99
This allows prediction of the column elution profile and is again dependent on the
assumption that the axial dispersion term is zero. In practice the number of stages has
little effect once n>15 and so computation times on a modern PC are very short. The
advantage of this approach is that it can be easily modified to incorporate more
complex models of the interaction process which take account of mass transfer
limitations. If necessary the rate expressions which result can be numerically
integrated within the same flow sheet structure.
In addition to simulation of the loading stage the model can also be used to predict the
effects of washing and elution. In these cases the initial conditions represent the pre-
saturated column and the feed concentration (c
f
) is set to zero. For washing the kinetic
constants remain unchanged while for elution the constants are revised to reflect the
effects of the eluting agents. This modelling approach is used in the ADCOL program
which can be used to investigate the performance of an integrated adsorption cycle.
Elution or Zonal chromatography
The major area of application for elution chromatography is in analysis where the
simultaneous detection and quantification of a number of components in a given
sample is desired. The limitation for a process application is that only a small fraction
of the adsorbent capacity will be occupied by the product during the separation. Also
as separation is a function of column length elution chromatography requires
significantly longer columns than are required for frontal methods.
The major use of process scale elution chromatography is for size based separations
i.e. SEC. The capacity constraints imposed by elution methods means that SEC tends
to be used for final polishing or for desalting of product prior to packaging.
Plate models
20
The traditional and still widely used method of quantifying elution column performance
is to relate the separation to a number of theoretical plates - the equivalent of an
equilibrium stage in distillation. The resolving power of the column will be a function
of the number of theoretical plates and the packing efficiency can be quantified in
terms of the height equivalent to a theoretical plate (HETP).
The HETP is defined as the thickness of the layer such that the concentration of the
solution leaving it is at equilibrium with the resin, assuming that there is no diffusion
between plates, that the equilibrium is linear in form and unaffected by the presence of
other solutes.
If we consider the solute balance on plate i:
accumulation + accumulation = solute - solute
in liquid in adsorbent flow in flow out
( ) ( ) V
dc
dt
V
dq
dt
Q c c
s
i
s
i
i i
+

1
1
Where Vs is the plate volume
Q is the volumetric flow rate
The linear isotherm or partition is
q Kc
i i

Therefore
( ) ( ) ( )
[ ]
dc
dt
Q c c V K
i
i i s
+
1
1
This equation must be solved for every plate in the column given that originally none
of the plates contain solute.
Taking n plates and a feed plate which contains the feed concentration (c
f
) at time zero
this gives.
( )
c c
e
i
i f
i

_
,

1
1 !

is the dimensionless time which is given by:


( ) ( )
[ ]

+
Qt
V K
s
1
The plate volume can be expressed in terms of the bed volume and the number of
plates V
V
n
s
b

21
( ) ( )
[ ]

+
nQt
V K
b
1
For a size exclusion column elution should be complete within Qt=Vb
therefore in this case.
( ) ( )

+
n f
K 1
where f is the fraction of the bed volume which has passed
Thus the elution profile can be constructed for a given column height provided the bed
length is specified and the HETP and K values are known.
This analysis is based on the Nobel prize winning work of Martin and Synge in 1942
who first described the performance of chromatographic columns in terms of
equilibrium stage processes. In practice the constraints on its application imposed by
the assumptions of a linear isotherm and local equilibrium limit is rigorous application.
However, plate theories still enjoy considerable popularity as the concept of the HETP
provides a convenient measure of a columns performance. In practice is would be
usual to determine HETP values from calibration data obtained with the column to be
used. This not only reflects the limitations of the theory but is based on the fact that
the HETP will, in part, be determined by the uniformity of the column packing which
will vary every time a column is repacked.
Practical considerations
The bed volume comprises of the adsorbent volume and the void volume such that
V V V
t l g
+
The retention volume for a compound on the column is the volume which must be
passed through the column before the elution peak is reached
V t Q
r r

where t
r
is the retention time i.e. the time between injection and the eluted pulse
reaching its maximum value.
The capacity factor for the component is expressed with respect to the void volume of
the column
( )

V V
V
r o
o
Alternatively the retention ratio is used
R
V
V
f
o
r

22
Size exclusion chromatography is a special case where the partition is purely physical
therefore even the smallest molecules should elute in a volume less than or equal to V
t
.
In this case the elution volume is related to the partition coefficient such that
V V K V
e l p g
+
or in terms of voidage
( ) ( ) V V K
e t
+ 1
Thus the elution volume for a given component can be determined from the column
volume, the partition coefficient and the voidage.
Clearly for an effective separation the elution volumes for two components must be
different and an important aspect of process design is defining resolution i.e. the extent
to which two components are separated. In addition to the elution volumes this
depends on the peak widths of the two components. The relationship can be written
as follows:
( )
R
V V
W W
s
e e
b b


+
2 1
1
2
1 2
where R
s
= resolution
W
b
= peak width at the base
suffixes 1&2 denote the two components.
The peak widths are determined by the number of plates (i.e. column length/HETP) a
number of forms have been proposed which relates number of plates to various peak
dimensions. Probably the most convenient relates n to W
b
.
n
V
W
r
b

_
,
16
2
This allows n to be determined from tracer studies on the column. Once n is known
the relationship can be rearranged to allow determination of W
b
for the components to
be separated and hence the resolution can be determined.
Another useful expression relates the pooled peak concentration to the feed
concentration using the number of plates.
c
c
n
av
f

2
So if the concentration of the pooled peak is measured and the feed concentration is
known the number of plates can be determined.
Worked Example 4
23
A serum albumin protein ii to be desalted in a Sephadex column. In a pilot study 1
gram of protein was eluted from a 7 litre column. It was found that the maximum
effluent concentration was reached after 6 litres had been passed through the column.
The pooled peak concentration was 2% of the feed concentration. Given that the
voidage was found to be 0.38 estimate:
a) The partition coefficient
b) The number of plates
Solution
a )
( ) ( ) V V K
e t
+ 1 so
( ) ( ) 6 7 38 1 38 + . . K
therefore K = 0.77
b) As the pooled peak concentration is given
c
c
n
av
f

2
so
c
c n
av
f

1
2
solving for n gives 398
_____________________________________________________________________
Worked Example 5
A size exclusion column be used to separate serum albumin from a globulin
contaminant. The partition coefficient for the albumin is 0.75 and for globulin is 0.9.
The column packing gives a voidage of 0.38 and an HETP value of 1e-4 m. Its
physical properties are such that the maximum allowable bed height is 0.75m.
What column length is required to give a baseline separation of the two components?
Solution
For a baseline separation the peaks must not overlap at the base. Therefore the
difference in elution volume for the two components must be greater than half the
combined base widths of their peaks.
Expressed in terms of length rather than volume the elution distance for each
component is given by
( )
( )
l K l
r p
+ 038 1 38 . .
The number of plates = l/HETP
The base width is given by
W
l
n
b
r

16
24
Denoting the components A and B
Plots of ( ) l l
r
A
r
B
and ( ) 05 . W W
b
A
b
B
+ against column length (constrained to between
0-0.75m) will intersect with each other at the minimum column length required for
base line separation.
0
0.01
0.02
0.03
0.04
0.05
0.06
0.07
0.1 0.2 0.3 0.4 0.5 0.6 0.7
column length
The number of plates is also a function of feed velocity with the relationship between
plate height and velocity defined by the van Deemter equation in terms of a summation
of factors which determine plate height:
HETP H H H
p d m
+ +
H d
p p
2
- the effects of band broadening resulting from differing flow velocities
through the column ( is constant close to 1,
d
p is the particle diameter).
H
D
u
d
g
2 - the effect of band broadening resulting from axial diffusion (D
g
is the
adsorbate diffusivity in the gel phase).
H
d
D
u
m
p
m

2
- described the effect of band broadening resulting from mass transfer
limitations (D
m
is the adsorbate diffusivity in the mobile phase,

is a constant which
reflect the mass transfer limitations imposed by the gel structure).
Written in this way the van Deemter equation is useful in that it provides an insight
into the mechanisms which determine column performance, but the requirements for
constants which must be determined experimentally means that it cannot be used for
purely theoretical predictions. Given this constraint it is usual to simplify the form of
the equation for the purposes of data interpretation:
HETP A
B
u
Cu + +
A, B and C are now lumped constants which can be determined from experimental
data.
25
Kinetic models
The limitations of plate theories is the need for a linear isotherm and the requirements
for plate equilibrium. In SEC both assumptions are reasonable. The partition is linear
and at the concentrations allowable there is unlikely to be interactions between solutes.
The compressible nature of the gels used in SEC imposes severe restrictions on the
flow rates used such that local equilibrium will often be reached.
In situations where these assumptions do not hold more detailed kinetic models are
needed. The tanks in series model used for adsorption can also be conveniently used
for this purpose with the only major difference being that the sample is loaded for an
appropriate number of stage volumes to represent the sample volume. As the
interaction expressions can be solved numerically the model can cope with solute
interactions and non-linear isotherms.
SCALE UP AND PROCESS LIMITATIONS
Scale up
Models of adsorption and chromatographic separations provide the first step in the
design process providing a framework for assessing capacity and kinetics which
provides a basis for scale-up. In most bio-separation processes these fundamental
considerations can be used to estimate the column length required for effective
separation while it is the scale of operation which dictates the volume of resin and
hence the column diameter required.
A further constraint is imposed by the mechanical stability of the hydrophilic
adsorbents normally employed for protein adsorption. Thus while kinetic limitations
may play an important role in determining feed rate, this must be compatible with the
pressure drop characteristics of the support.
As column diameter is increased it becomes more difficult to ensure a uniform packing
distribution. This is most significant in elution chromatography where poor packing
leads to an increase in the HETP value observed which results in reduced resolution.
Further complications arising from scale up include a reduction in wall effects which
can lead to a reduction in resin support and an increase in compression. Thus lower
feed velocities are often needed in larger columns.
A further difficulty encountered in scale up is distributor design. The models of
column based adsorption and chromatography discussed above all make the implicit
assumption that the sample is applied to the column in plug flow i.e. there is no radial
concentration gradient at the top of the column and that at the point of loading the
feed concentration switches from zero to c
f
as a square wave. In a small column a
good approximation of this situation can be achieved as the radial distance is small. As
column diameter is increased design becomes more difficult. The basic aim is to
maintain a fluid layer across the top of the column which is as thin as possible to
minimise mixing effects. On the top of the adsorbent there would normally be a
porous sheet having a significant flow resistance to ensure that radial pressure drop is
constant. In larger column the feed stream is usually split and fed into the column at a
number of points to minimise localised velocity variations which might otherwise
occur.
26
Process limitations
Feed concentration
While equilibrium considerations usually determine the product concentration to be
applied to a column a further constraint is imposed by the total feed concentration and
the types of contaminant which are present. As most adsorption steps are intended to
give both a significant purification factor and a significant degree of concentration this
suggests that the concentration of product molecule will be very much lower than the
concentration of contaminants (often the difference will be several orders of
magnitude). This suggests that for most feeds a considerable volume would be needed
to saturate column capacity. Given the mechanical constraints on feed velocities pre-
concentration prior to loading appears attractive, however, the effects of concentration
on feed viscosity must be considered. In addition to the effect of viscosity on pressure
drop, the effect of viscosity on solute distribution within the column has to be
considered. High viscosity can significantly increase diffusional mass transfer
limitations such that adsorption efficiency is dramatically reduced. In practice this
constrains most protein feed concentrations to values below 50g/l, if polysaccharides
are present the values would be lower again. Given the heterogeneous nature of
natural protein mixtures this represents a guide figure only.
Feed volume
In frontal operation the column is usually loaded until a predetermined fraction of the
resin capacity is utilised. This is determined by the presence of product in the column
effluent. Typically the column would be operated until breakthrough occurs with the
value of c/cf=0.1 being commonly used as the point where loading is terminated.
Termination of loading is dictated by a number of factors. The first is a simple
compromise between fractional product recovery and maximal use of resin capacity
(see figure). If loading is stopped at point of breakthrough product loss is small but a
significant fraction of resin remains unused conversely if resin is fully used then
product losses are high.
-0.1
0
0.1
0.2
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1
0 200 400 600 800
time s
c/cf
The magnitude of the difference is determined by the adsorption isotherm. The second
problem is one of detection, the product will be only a small fraction of the total
27
Unused capacity
Lost product
protein in the column effluent hence non specific detection methods (e.g. UV
absorbance) will be swamped by the signal from the contaminants. In practice off line
analysis may be required for protein determination. Once a process has been well
characterised the cycles are usually switched on a time basis.
In elution chromatography where samples are applied as a discrete slug the peak width
will be a function of the sample size. The plate theory allows prediction of the peak
with which results from a sample equivalent to a single plate volume which reflects
column performance. When the sample is equivalent to a number of plate volumes this
is equivalent to a generating a series of off-set output peaks using the plate model
resulting in a broadened composite peak. This effect is particularly severe in SEC
where separation occurs in one bed volume and constrains the sample size to <5% of
the bed volume for most separations.
Non specific interactions & fouling
Adsorbent supports are deliberately chosen to be inert and hydrophilic to minimise
background interactions between protein and support. However, once these materials
are derivatised with the required active groups there is potential for non specific
interactions. For example, Cibacron Blue the dye used in affinity chromatography
carries a number of strongly acidic groups which will be ionised at neutral pH hence
Cibacron Blue can act as an ion exchange adsorbent as well as an affinity support. So
while the affinity interaction is highly selective, are must be taken with operating
conditions or the purification will be compromised by non specific ion exchange
adsorption of contaminants. A range of other non specific interactions can arise from
secondary properties of adsorbent groups and provides a strong argument for
maximising effective ligand densities. It has been shown that with highly substituted
Cibacron blue based adsorbents less than 20% may be available for affinity
interactions. However, they all contribute towards ion exchange effects.
Fouling essentially represents an extension of non specific interactions, in that some
material which is non specifically bound may be impossible to elute using conditions
compatible with resin stability. This material accumulates cycle by cycle leading to a
progressive reduction in capacity and potential problems with column hygiene.
Ultimately fouling will limit the resin life.
Dispersion of packing
As stressed earlier homogeneity of packing is critical for elution protocols and packing
irregularities can lead to significant degradation of separation performance. However,
even where original column packing is good the packing quality can degrade during
use. An obvious problem is the introduction of gas bubbles to the column. The
problem of air bubbles entering via leaks is relatively easy t avoid by in situ bubble
formation resulting from changes in dissolved gas solubility with temperature are more
difficult to control and may require feed solutions to be degassed. Other changes
which are impossible to avoid are the formations of voids in the packing resulting from
changes in levels of resin swelling as the feed composition changes between load,
wash, elute, and regeneration cycles. This is to some extent resin dependent and may
influence resin choice, but in many cases the column may need to be repacked several
times before the resin life is exceeded.
28
Ligand leakage
Probably the majority of the protein products of biotechnology are intended for
therapeutic applications. Thus their purity has to be precisely defined and the
production process rigorously validated. Part of this validation concerns the
properties and stability of the materials used in the separation process. Of particular
concern is product contamination by material leached from adsorbents. Thus any new
material has to be extensively characterised with respect to its stability and the rate of
leakage of active groups. The levels which have to be measured in this assessment are
such that ELISAs are commonly used.
Novel adsorbents
The compromises which have to be made between capacity and kinetics when
traditional beaded supports are used have lead to the development of a number of new
materials.
Perfusive adsorbents
These are beaded materials with a much more open pore structure than is usual for
porous beads. The size of the largest pores is such that it is possible to general a
convective flow through the beads themselves thus access to binding sites within the
beads is not solely dependent on molecular diffusion and hence binding kinetics are
improved.
Tentacle resins
From the point of view of mass transfer limitations and mechanical stability the ideal
adsorbent would be based on rigid non porous beads. However the particle diameters
required to allow effective feed velocities means that the bead surface area limits the
volume averaged ligand density which can be achieved. This limitation can be
partially overcome by attaching long polymers (e.g. dextran) to the non porous beads
and then attaching the ligand to the polymer. The bead is now covered with
tentacles each of which can contain many ligands, but the diffusion paths remain
short.
Affinity membranes
This is an example of what are described as combined field separations i.e. two
separation principles are employed in a single process step. A membrane based
adsorbed can be regarded as the extreme example of a short fat column the flow
resistance will be low so the feed velocity can be high subject to binding conditions.
The sieving properties of the membranes can be chosen such that many of the
contaminants can be excluded from the bulk of the adsorbent. The problems are that
breakthrough will tend to be very fast and must be steep given the short bed length,
and that in most schemes the product must be transmissible through the membrane
which will be compromised by fouling effects.
Novel operating protocols
Expanded beds
29
A problem with packed bed adsorbers is that they rapidly become blocked if the feed
contains particulates. This means that after cells have been homogenised a significant
degree of pre-purification is required before the resultant material can be applied to the
column simply to prevent physical blockage. The expanded bed approach is aimed at
avoiding this problem by using the column under up flow conditions such that it is
partially fluidised or expanded.
In an expanded state the bed will allow transmission of particulates without blockage
such that crude homogenates can be applies (subject to chemical stability constraints).
The problems to be overcome result from the balance between the superficial velocities
needed to give bed expansion and the binding kinetics which must allow the use of
shorter column residence times.
Deviations from standard isotherms
Multivalent interactions
Many biologically important proteins have more than one active site per molecule.
This can allow multiple interactions between protein and support and effectively
enhances binding. Under most practical conditions these effects will not be significant
for protein separations as ligand spacing on the support will restrict the potential for
multiple interactions.
Ligand capping effects
Given the size of protein based adsorbents the adsorption of a protein molecule within
a resin pore can significantly reduce the effective pore diameter. The adsorption of a
number of molecules at the entrance to a pore can effectively cap the pore
preventing protein access to unoccupied ligands deeper within the pore. Thus the
maximum binding capacity is reduced. However, as the extent to which capping
occurs will be a function of adsorbate feed concentration this can lead to abnormal
binding isotherms.
Reading List
Books and monograph chapters
Affinity chromatography: a practical approach (eds Dean, Johnson and Middle) IRL
Press 1985.
Bioseparations: Downstream processing for Biotechnology, Belter, Cussler and Hu,
Wiley.
Modelling equilibrium and kinetics in chromatographic processes Costa in
Chromatographic and membrane processes in biotechnology (eds Costa and Cabral)
Kluwer 1991, 3-24. (other chapters also relavent).
Sorption Processes, Roig in Recovery processes for biological materials (eds
Kennedy and Cabral) Wiley 1993, p316-414.
30
Chromatography, Garcia and Pires in Recovery processes for biological materials
(eds Kennedy and Cabral) Wiley 1993, p415-452.
The chemistry and engineering of affinity chromatography, Liapis and Unger in
Highly SelectiveSeparations in Biotechnology (ed. G. Street) Blackie 1994, p121-162.
Papers and Reviews
Chase, H.A. (1984) Affinity separations using immobilised monoclonal antibodies - a
new tool for the biochemical engineer Chemical Engineering Science 39, 1099-1125.
Velayudhan, A. and Horvath, C. (1994) Adsorption and ion-exchange isotherms in
preparative chromatography J. Chromatogr. 663, 1-10.
Mao, Q.M. and Hearn, M.T.W. (1996) Optimization of Affinity and Ion-Exchange
chromatographic processes for the purification of proteins Biotechnol. Bioeng. 52,
204-222.
31

You might also like