You are on page 1of 7

Eng. Life Sci. 2016, 16, 107113 www.els-journal.

com

Tobias Hahn1 Research Article


Thiemo Huuk1
Anna Osberghaus1 Calibration-free inverse modeling of
Katharina Doninger2
Susanne Nath2
ion-exchange chromatography in industrial
Stefan Hepbildikler2 antibody purification
Vincent Heuveline3
Jrgen Hubbuch1 The identification of optimal process parameters for the isolation of a target com-
ponent from multicomponent mixtures is especially challenging in industrial ap-
1
Biomolecular Separation plications. With constantly increasing time-to-market pressure, screening a large
Engineering, Karlsruhe Institute parameter space is not feasible and design-of-experiment approaches with few ex-
of Technology (KIT), Karlsruhe, periments might fail due to dynamic and nonlinear reactions to small parameter
Germany changes. Model-based optimization can determine optimal operating conditions,
2
Pharmaceutical Biotech
once the model has been calibrated to the specific process step. In this work, pa-
Production, Roche Diagnostics rameters for the steric mass action model were estimated for the target protein and
GmbH, Penzberg, Germany three impurities of an industrial antibody cation-exchange purification step using
3
only chromatograms at different wavelengths and additional fraction analyses with
Engineering Mathematics and size exclusion chromatography. Information on the molar or mass concentrations
Computing Lab, Heidelberg
in the feed are not available. The model-based optimization results coincide with
University, Heidelberg, Germany
conventional chromatogram-based optimization.

Keywords: Industrial application / Inverse method / Liquid chromatography / Mechanistic


modeling / Optimization
Received: January 28, 2015; revised: April 13, 2015; accepted: April 29, 2015

DOI: 10.1002/elsc.201400248

1 Introduction time and material-efficient process optimization as long as the


model parameters can be determined reliably with less effort.
Biopharmaceutical products currently constitute one of the For some sorption submodels, such as the steric mass action
fastest growing markets for the pharmaceutical industry [1]. In- (SMA) model for ion-exchange chromatography of proteins [8],
dustrial downstream processing faces the challenge of efficiently model calibration protocols for pure components exist [8], which
purifying a product out of a very heterogeneous mixture. Espe- allow for determining the component-specific parameters in a
cially for mAb, the purification sequence is commonly based on consecutive fashion. With no a priori knowledge about the com-
platform processes that are only slightly adapted to new target ponents behavior within mixtures, the inverse method is a suited
components [2, 3]. This approach ensures fast process develop- option, which alters parameters in a systematic fashion to achieve
ment and reduces the time to market, but it is probable that a match of measured chromatogram and model prediction. In
the reduced exploration of design space leads to suboptimal a comparative study [9], the direct approach and the inverse
processes [4]. Process understanding is hence a sensible and im- method were found to reach equal prediction quality such that
portant addition to the use of platform processes [5]. As the the latter is recommended for fast process development.
integration of modeling tools into process development is also The potential of mathematical modeling and numerical op-
an essential part of the strategy for the implementation of the timization of chromatography has been demonstrated in aca-
quality by design approach [6,7], modeling tools are increasingly demic set-ups for mixtures of small molecules [10, 11], model
gaining the attention of the pharmaceutical industry. It ensures proteins [12], and antibodies [13, 14], as well as for industrial
process steps [15, 16]. All applications have in common, that
sensor calibrations existed for all components, allowing to trans-
Correspondence: Prof. Jurgen Hubbuch (juergen.hubbuch@kit. form UV absorbance values into mass or molar concentrations.
edu), Biomolecular Separation Engineering, Karlsruhe Institute of This paper discusses the challenges when applying mechanistic
Technology (KIT), Engler-Bunte-Ring 3, 76131 Karlsruhe, Germany modeling and numerical optimization to a dataset of industrial
Abbreviations: CDR, convection diffusion reaction; CV, column vol- preparative chromatography without knowledge of feed compo-
ume; CEX, cation-exchange chromatography; FEM, finite element sition in terms of molar concentrations. Additional experiments
method; HMW, high molecular weight; LMW, low molecular weight; and analyses are avoided by employing a modeling approach that
SEC, size exclusion chromatography; SMA, steric mass action estimates the absorption coefficients from the recorded UV data.


C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim 107
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

Table 1. Measured and calculated column parameters.

Parameter Symbol Value Unit Proceeding

Diameter D 10 mm From manufacturer


Length L 246 mm From manufacturer
Bead radius rp 0.025 mm From manufacturer
System dead volume Vd 1.98 mL Acetone pulse injection without column
Retention volume acetone VRetAc 16.94 mL Acetone pulse injection with column
Retention volume dextran VRetDex 10.29 mL Dextran pulse injection with column
SD of dextran Dex 0.161 mL
Akta peak integration
Volume of HCl VHCl 130.2 mL Acid/base titration
Molarity of HCl c HCl 0.01 M Manually controlled
Flow rate u 0.69 mm/s Manually controlled
4d L
1 2
Volume V 19.32 mL
Fluid volume Vf 14.96 mL VRetAc Vd
Interstitial volume Vint 6.43 mL VRetDex Vd
Vf
Total column porosity t 0.77 V
Vint
Interstitial porosity b 0.43 V
Vf Vint
Particle porosity p 0.6 VVint
u
Interstitial flow uint 1.61 mm/s b
Dex 2
2 L u int ( Vint )
1
Axial dispersion D ax 0.124 mm/s
c HCl VHCl
Ionic capacity  0.3 M V(1b )(1p )

The case study bases on a cation-exchange chromatographic solution until an increase in conductivity signal was recorded.
(CEX) process step (Poros 50 HS resin). The antibody mix- From the Na-ion concentration of the titrant and the volume of
ture fed into CEX is a protein A eluate after low pH incubation the applied titrant, the total number of exchangeable ions was
and conditioning. The mixture contains several antibody vari- calculated. All chemicals used were obtained highest quality.
ants differing in size and charge, that all elute in a common peak. With this set of parameters all system-specific parameters
Fraction collection and analysis has to be performed to obtain in- occurring in the mathematical model can be fixed as also given
formation on the location of the impurities. Using size exclusion in Table 1.
chromatography, the targeted monomer and three impurities
can be distinguished, two high molecular weight (HMW) and
one low molecular weight (LMW) species. The found model
parameters were used for in silico optimization and eventu-
2.2 Bind-elute experiments
ally coincide with conventional optimization by chromatogram
As system liquid a 50 mM acetate buffer was used at pH 4.95, for
evaluation.
elution a high-salt buffer of 50 mM acetate and 750 mM NaCl
was employed. The different salt profiles in the following were
2 Materials and methods mixed from these two buffers.
A sample volume of 81.3 mL = 4.2 column volumes (CV) was
Initial experiments were conducted to determine system proper- injected for each experiment. First, the gradient experiment was
ties, thereafter six experiments in bind/elution mode were per- ran from 50 mM (0% high salt buffer) to 550 mM (66% high-
formed. The following sections describe the experimental set-up, salt buffer), with the gradient starting at 8.4 CV and ending at
data processing, and parameter estimation. 18.1 CV. As the elution peak reached its maximum at a total salt
concentration of 210 mM, five step elutions with concentrations
190, 200, 210, 220, and 230 mM were performed for optimiza-
2.1 Column parameter determination tion. Each step was induced at 7 CV and was followed by a wash
with 100% high-salt buffer as soon as the 280 nm UV signal fell
A 20 mL column, with Poros 50 HS (Applied Biosystems, below a threshold of 100 mAU.
Carlsbad, CA, USA) resin was analyzed first with an acetone The elution peaks of all experiments were captured in 3 mL

pulse and second with dextran using an Akta Avant system (GE samples and analyzed by size-exclusion chromatography (SEC)
Healthcare, Little Chalfont, UK) to determine the essential model to determine the relative contribution of the two HMW species,
parameters as described in [17] and presented in Table 1. Acid- the monomer, and the LMW species to the sum signal. In the case
base-titration was carried out to determine the total ionic capac- of the step elutions, the peak occurring during the subsequent
ity: The column was flushed with a 0.5 M HCl solution until a wash was also fractionated and analyzed. The same analysis was
constant UV and conductivity signal was achieved. Afterwards performed for the feed material.
the column was washed with ultrapure water until a constant As the gradient elution resulted in a blunt top (Fig. 2) and
UV and conductivity baseline was reached. After that the col- not a distinct peak, a longer gradient elution was performed to
umn was titrated at a flow of 100 cm/h with a 0.01 M NaOH check for charge variants.

108 
C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

2.3 Mathematical model path length, where the extinction coefficients are unknown for
the impurities and cannot be determined easily as the compo-
The UV absorbance-based model as developed in [18] is used in nents are not available in pure form.
the following. The general rate model [17] models the macro-  i
k
scopic protein transport through the column. The system is 1 q i j + j
kdes,i = keq,i  q j c p ,i c pi,salt q i (7)
of convection diffusion reaction (CDR) type. Equation (1) de- t j =1
a j
scribes the rate of change of a concentration c i (x, t) , measured
in [M] for salt and [mAU] for proteins, in the interstitial volume k
j
of a column with length L to consist of convective mass trans- q salt =  qj (8)
port in space with the average interstitial velocity of the fluid a
j =1 j
u, peak broadening effects that are modeled as dispersion in
The model is chosen because of its capability of simulating the
axial direction with respect to a coefficient Dax , and transition
whole chromatographic process, including elution, by changing
from the interstitial concentration into the particle pore con-
the induced salt concentration at the inlet.
centration c p ,i (x, r, t) that depends on the porosity of the bed
b , the radius of adsorbent particles rp and a component-specific
film transfer coefficient kf ilm,i . The model is complemented with 2.4 Numerical solution
Danckwerts boundary conditions, Eqs. (2) and (3). Equations
(4)(6) model the accumulation of mass in the pore volume c p ,i The numerical simulation is performed using the in-house soft-
and stationary phase q i depending on the particle porosity p ware package ChromX. Following the method of lines, the equa-
and the component-specific pore-diffusion coefficient Dp . tion system is first discretized in space on given nodes, using the
finite element method (FEM). FEM is a highly versatile method
c i c i 2 c i with strong mathematical foundation and well suited for CDR
(x, t) = u (t) (x, t) + D ax 2 (x, t)
t x x equations. The solutions procedure starts with the weak formu-
1 b 3   lation, incorporating the boundary conditions and representing
kf ilm,i c i (x, t) c p ,i (x, t) (1) the variables with basic functions from the respective spaces.
b rp
A Streamline-Upwind-Petrov-Galerkin method was used here
with linear basis and test functions. The discretization in time
c i u (t)  
(0, t) = c i (0, t) c in,i (t) (2) is performed with the fractional step -scheme, a semiimplicit
x D ax

c i Volume [ml]
(L , t) = 0 (3)
150 200 250 300 350
x
4,000 120
c p 1 p q
(x, r, t) = (x, r, t)
t p t
  3,000 90
1 c p

Conductivity [mS/cm]
Absorption [mAU]

+ 2 r2 D p (x, r, t) (4)
r r r

c p   kf ilm    2,000 60
x, r p , t = c (x, t) c p x, r p , t (5)
r p D p

c p 1,000 30
(x, 0, t) = 0 (6)
r
The SMA isotherm [8], modified in [18], is a commonly used
semimechanistic isotherm in ion-exchange chromatography. It 0 0
is capable of reproducing the influence of counter ions on the Close-up
retention behavior of protein species using the proteins charac- 30 Monomer
teristic charges i . In addition, it considers adsorbent properties
such as the total ionic capacity  and steric shielding effects 20
i of the protein i covering an amount of binding sites, greater Conductivity
than the actual number of sites it interacts with. The UV 10
absorbance-based kinetic SMA isotherm is given in Eq. (7), with
q i and c p ,i being the concentration of the protein i adsorbed and 0
160 180 200 220 240
in solution, respectively. c p ,salt is the salt concentration of the
solution. keq,i and kdes,i are the constants of equilibrium and des- Figure 1. Single-component absorbance profiles at UV 280 nm for
orption rate, and ai the absorption coefficient that scales molar the 190 mM salt step elution. The curves are generated by multi-
concentrations to absorbance units, according to LambertBeer plying the chromatogram by component ratios found by fraction
law. The factors ai consist of extinction coefficient and UV cell analysis with SEC.


C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim 109
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

3500 70 UV 300 nm captures approximately four times higher concent-


rations than UV 280 nm (data not shown). The chromatograms
3000 60
were also corrected by the influence of the salt concentration in

Conductivity [mS/cm]
Absorption [mAU]

2500 50 the mobile phase.


A variety of algorithms is available for the solution of this
2000 40
optimization problem. In this work, we employed a heuris-
1500 30 tic method based on a genetic algorithm (GAlib ver. 2.4.7,
http://lancet.mit.edu/ga). Genetic algorithms avoid local min-
1000 20
imums by performing random jumps and hence explore a larger
500 10 area of the search space.
As the UV sensor is quickly saturated at 280 nm, we con-
0 0
150 200 250 300 350 400 sider additional absorbance measurements at 260 and 300 nm
Volume [ml] to capture the complete peak shape. In addition, we include
260 nm 280 nm 300 nm Conductivity SEC analyses of fractions at a common wavelength, such that
the contributions to the UV sum signal can be quantified. This
Figure 2. Gradient elution peak recorded at UV 260, 280, and data enters the least squares problem Eq. (9): m is extended with
300 nm. Only the 300 nm signal shows the entire top. The peak additional entries of the contributions for each fraction and
shape indicates that the monomer consists of differently eluting species and c is extended accordingly with entries only includ-
charge variants. ing those simulated concentrations that belong to the respective
contribution.
The estimation procedure using SEC data relies on pseudo
procedure providing second-order accuracy [19]. Finally, the
absorbance profiles m(tj ) for all components i that are generated
nonlinearity of the equation system introduced by the isotherm
by multiplying the UV signal with the components fraction per-
must be treated with an iterative procedure, here, Picard itera-
centage in the corresponding time interval. Figure 1 shows the
tion. The resulting linear systems are solved by LU factorization.
pseudo profiles for the step elution with lowest salt concentra-
tion. A HMW1 peak is visible during the high-salt step following
2.5 Parameter estimation elution, while LMWs are only visible at the beginning of the
elution peak.
Estimation of an unknown parameter set p op t solves the least
squares optimization problem
 2
      3 Results and discussion
p op t = arg min m tj c i L , tj ; p (9)
p j i1
3.1 Bind-elute experiments
where m(tj ) is the measured chromatogram value at point in
time tj . The measurements might also contain noise, which can The feed analysis by SEC showed 96.79% monomer, 2.95%
be neglected when assuming the noise to be zero-mean Gaussian + 0.17% species with higher molecular weight (HMW 1 and
and isotropic. The concentration ci is simulated in absorbance HMW 2) and 0.09% species with LMW at 280 nm. Further IEC
units. Linear scaling to molar concentrations was checked with analysis revealed that the exact composition of the monomer is
concentrated feed samples. The sensor signal is approximately 15.4% acidic, 63.3% main, and 21.3% basic variant. An example
proportional to the feed concentration up to the saturation limit of a step elution result is presented in Fig. 1. It shows a steep
in UV 280, as suggested by LambertBeer law. Furthermore, front, long tailing and a considerable peak after the final high

100
Target purity [%]

99.5

99

98.5

98
60 65 70 75 80 85 90 95 100
Target yield [%] Figure 3. Monomer purity over yield when collecting
Gradient 210 mM fractions consecutively. Higher steps lead to coelut-
ing impurities and lower purity. Lower steps allow to
190 mM 220 mM
separate impurities but do not collect all of the target
200 mM 230 mM component.

110 
C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

Figure 4. Comparison of measured chromatogram () and simulated sum signal () for the salt elutions (- -) used for model calibration.
0.19 mM step elution, UV 280 nm (A) and UV 300 nm (B), 0.23 mM step elution, UV 280 nm (C) and UV 300 nm (D), and gradient elution,
UV 280 nm (E) and UV 300 nm (F).

salt step. The gradient elution result in Fig. 2 shows a blunt top, defined as average SEC target peak area of involved fractions
indicating that the monomer variants elute differently. divided by total absorbance and yield is similarly defined as ratio
of collected peak area by total area. Because of process perfor-
mance requirements, only step elution scenarios are considered
3.2 Reference optimum that achieve at least 99% purity and 80% yield with fraction size
smaller than 5 column volumes.
When not using model-based optimization in combination with Figure 3 shows the development of monomer purity over yield
reliable feed concentrations or alternative assays, the optimum when starting to collect from the first fraction of the elution peak
has to be defined using only UV absorbance data. Purity is and successively adding the other fractions. Based on this data,


C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim 111
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

100 Salt step [M]


0.23
99.5

Target purity [%]


0.22
99

0.21
98.5

98 0.20

97.5 0.19
60 70 80 90 100
Target yield [%]

Figure 5. Intermediate values of salt step optimization with a genetic algorithm. Step concentrations below 210 mM allow a purity above
99%. Highest yield values are achieved by step concentrations above 200 mM.

Table 2. Estimated model parameters.

Component kf ilm Dp k1
des keq a
[mm/s] [mm/s] [sM ] [-] [-] [-] [mAU/M]

Salt 0.0083 7.00104


LMW 0.0083 3.49106 1.61102 1.70 3.32 65.0 4.4107
Monomer acidic 0.0083 2.10105 9.00105 1.98 3.05 75.3 8.22107
Monomer main 0.0083 5.34106 1.90103 1.42 5.21 75.3 8.22107
Monomer basic 0.0083 1.57104 6.08104 1.91 6.90 75.3 8.22107
HMW 1 0.0083 3.60106 3.50106 2.40 8.23 210 2.14108
HMW 2 0.0083 7.72106 3.90105 5.70 5.20 287 2.10108

the 200 and 210 mM steps perform best. Gradient elutions are The found parameter set in Table 2 is reasonable; the char-
undesired in the final process and were not considered. When acteristic charges ascend with the molecule size and in the
collecting the first 33 fractions ( = 99 mL  5 CV), the 210 mM monomer case with charge variant type. Steric shielding factors
step achieves a purity of 99.3 and 86.5% yield. The 200 mM step and absorption coefficients ascend approximately with molecule
achieves a slightly better purity of 99.5%, but only 80% yield. size as expected. Sorption kinetics and film diffusion are fast, as
The 220 mM step reaches a higher yield with fewer fractions but indicated by the steep elution fronts.
does not attain the desired purity.

3.4 Optimization

3.3 Protein parameter estimation Again, the genetic algorithm was employed to determine the
optimal salt step height together with the fractionation bound-
The estimation algorithm employed is a genetic algorithm, to aries. The optimization of load conditions was performed with
quickly cover a large search space. The inlet absorbance values traditional high-throughput screening beforehand. Hence, com-
are set for all components using the known peak area at 300 nm, parison to a nonmodel-based approach in this particular scale
the scaling factor from 300 to 280 nm and the results from SEC was not possible.
and IEC analysis of the feed at 280 nm. Results of the genetic algorithm are plotted in Fig. 5. Although
From the available data, the gradient and the highest and the model was calibrated only with the gradient and the 190
lowest salt steps were used for estimation, as the peak shape did and 230 mM steps, the results resemble the findings in Section
not change much for the intermediate steps. 3.2 closely: only step concentrations below 210 mM allow a
Absorbance-based modeling is able to determine the non- purity above 99% and only step concentrations below 200 mM
linear parameters i and ai uniquely from single-component achieve high yields above 95%. The yield values are slightly higher
curves [18]. These are available for all impurities, but not for in the model-based optimization results as some parts of the
the monomer charge variants. As the variants are indistinguish- reference elution peaks were not analyzed by SEC and could
able in SEC, we can assume them to have equal steric shielding not be considered when calculating the reference optimum (cf.
coefficients and absorption coefficients. Fig 1).
Curve fitting finished with a very good match of measurement A compromise between yield and purity is found at approxi-
and simulation, considering the complex elution behavior with mately 200 mM, closely followed by 210 mM, just like in the
long tailing step elutions and blunt gradient top. The result is reference analysis. The admissible volume of 100 mL would
shown in Fig. 4. be fully exploited by a 195200 mM step. A 210 mM step is

112 
C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
www.els-journal.com Eng. Life Sci. 2016, 16, 107113 www.biotecvisions.com

predicted to achieve a yield of 91% at the required purity of 99% [4] Nfor, B. K., Ahamed, T., van Dedem, G. W., van der Wielen, L.
using only 60 mL. A. et al., Design strategies for integrated protein purification
processes: challenges, progress and outlook. J. Chem. Technol.
Biotechnol. 2008, 83, 124132.
4 Concluding remarks [5] Helling, C., Borrmann, C., Strube, J., Optimal integration of
directly combined hydrophobic interaction and ion exchange
The benefit of model based optimization compared to the tra- chromatography purification processes. Chem. Eng. Technol.,
ditional approach is foremost given by the fast adaptation to 2012, 35, 17861796.
changes in the feed composition and the possibilities of fur- [6] ICQ Quality Implementation Working Group, ICH-Endorsed
ther optimization. For example, salt profiles can be constructed Guide for ICH Q8/Q9/Q10 Implementation, 2011.
consisting of several steps in combination with a gradient. [7] Chhatre, S., Farid, S. S., Coffman, J., Bird, P. et al., How
The reduction of the number of experiments in this case is not implementation of quality by design and advances in bio-
significant, as conventional optimization was straight forward chemical engineering are enabling efficient bioprocess develop-
and the model-based optimum would have to be verified by an ment and manufacture. J. Chem. Technol. Biotechnol. 2011, 86,
additional experiment. 11251129.
Fractionation itself is a necessity for both approaches, where [8] Brooks, C. A., Cramer, S. M., Steric mass-action ion exchange:
further studies should examine if good results can be obtained displacement profiles and induced salt gradients. AIChE J.
with lower resolution. 1992, 38, 19691978.
[9] Osberghaus, A., Hepbildikler, S., Nath, S., Haindl, M. et al., De-
termination of parameters for the steric mass action modela
Practical application comparison between two approaches. J. Chromatogr. A 2012,
1233, 5465.
This paper demonstrates that model-based optimization [10] Abel, S., Erdem, G., Amanullah, M., Morari, M. et al., Opti-
can be applied to an industrial cation-exchange step, even mizing control of simulated moving bedsexperimental im-
if the molar concentrations of the feed components are plementation. J. Chromatogr. A 2005, 1092, 216.
unknown. Based on a few experiments, which might be [11] Forssen, P., Arnell, R., Fornstedt, T., An improved algorithm for
already available from previous screenings, the model can solving inverse problems in liquid chromatography. Comput.
be calibrated. For differentiating components eluting hid- Chem. Eng. 2006, 30, 13811391.
den in the sum signal, single components elution curves [12] Osberghaus, A., Hepbildikler, S., Nath, S., Haindl, M. et al.,
must be generated for peak fitting. This is accomplished Optimizing a chromatographic three component separation: A
by SEC fraction analyses at the same wavelength as the comparison of mechanistic and empiric modeling approaches.
chromatogram. J. Chromatogr. A 2012, 1237, 8695.
[13] Karlsson, D., Jakobsson, N., Axelsson, A., Nilsson, B.,
Model-based optimization of a preparative ion-exchange
step for antibody purification. J. Chromatogr. A 2004, 1055,
The research leading to these results was partly funded by EURO
2939.
TRANS-BIO (grant agreement no. 0316071B, ECs Seventh Frame-
[14] Degerman, M., Jakobsson, N., Nilsson, B. Constrained opti-
work Programme).
mization of a preparative ion-exchange step for antibody pu-
rification. J. Chromatogr. A 2006, 1113, 92100.
The authors have declared no conflicts of interest. [15] Mollerup, J. M., Hansen, T. B., Kidal, S., Sejergaard, L., Staby,
A., Development, modelling, optimisation and scale-up of
chromatographic purification of a therapeutic protein. Fluid
5 References Phase Equilib. 2007, 261, 133139.
[16] Close, E. J., Salm, J. R., Bracewell, D. G., Sorensen, E., Modelling
[1] Knablein, J., Twenty thousand years of biotechfrom tra- of industrial biopharmaceutical multicomponent chromatog-
ditional to modern biotechnology, in: Knablein, J. (Ed.), raphy. Chem. Eng. Res. Des. 2013, 92, 13041314.
Modern Biopharmaceuticals: Recent Success Stories, Wiley-VCH [17] Michel, M., Epping, A. and Jupke, A., Modeling and determina-
Verlag, Weinheim 2013, pp. 138. tion of model parameters, in: Schmidt-Traub, H. (Ed.), Prepar-
[2] Shukla, A. A., Hubbard, B., Tressel, T., Guhan, S., Low, ative Chromatography: Of Fine Chemicals and Pharmaceutical
D., Downstream processing of monoclonal antibodies Agents, Wiley-VCH Verlag, Weinheim 2005, pp. 215312.
application of platform approaches. J. Chromatogr. B 2007, [18] Hahn, T., Baumann, P., Huuk, T., Heuveline, V., Hubbuch, J.,
848, 2839. UV absorption-based inverse modelling of protein chromatog-
[3] Liu, H. F., Ma, J., Winter, C., Bayer, R., Recovery and purifi- raphy. Eng. Life Sci. 2016, 16, 99106.
cation process development for monoclonal antibody produc- [19] Glowinski, R., Handbook of Numerical Analysis, Vol. IX, North-
tion. mAbs 2010, 2, 480499. Holland, Amsterdam 2003.


C 2015 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim 113

You might also like