Вы находитесь на странице: 1из 8

Journal of Chromatography A, 1393 (2015) 5764

Contents lists available at ScienceDirect

Journal of Chromatography A
journal homepage: www.elsevier.com/locate/chroma

Purication process of recombinant monoclonal antibodies with


mixed mode chromatography
Sophie Maria a , Gilles Joucla a , Bertrand Garbay a , Wilfrid Dieryck a ,
Anne-Marie Lomenech b , Xavier Santarelli a , Charlotte Cabanne a,
a
Univ Bordeaux, BPRVS, EA 4135, F-33000, Bordeaux, France, Bordeaux INP, BPRVS, EA 4135, F-33000, Bordeaux, France
b
Univ Bordeaux, Centre de Gnomique Fonctionnelle, Plateforme Protome, Bordeaux F-33000, France

a r t i c l e i n f o a b s t r a c t

Article history: An innovative process to purify mAb from CHO cell culture supernatant was developed. This three-
Received 28 January 2015 step process involved two mixed mode resins and an anion exchange membrane. We used a human IgG
Received in revised form 6 March 2015 mixture to determine the optimal conditions for each purication step. Thereafter, the whole process was
Accepted 7 March 2015
evaluated and improved for the purication of a recombinant mAb produced in the supernatant of CHO
Available online 14 March 2015
cells. Once optimized, yield and purity of 88% and 99.9%, respectively were comparable to those obtained
in a conventional process based on a capture step using protein A. In addition, aggregates, HCPs and DNA
Keywords:
levels in the puried fraction were below regulatory specications. Then we used mass spectrometry to
Mixed mode chromatography
Antibody
identify contaminating proteins in the antibody fraction in order to highlight the behavior of HCPs.
Host cell proteins 2015 Elsevier B.V. All rights reserved.
Purication process

1. Introduction neglected in the protein purication eld. It was not before the
pioneering work of Burton and Harding, who developed a vari-
Industrial processes for the purication of recombinant mono- ant called Hydrophobic Charge Induction Chromatography, that
clonal antibodies (mAbs) produced in CHO cells are usually based interest for mixed mode chromatography to purify proteins was
on three successive chromatography steps. The rst consists in rekindled [8]. They tested numerous ligands having heterocycles
the capture of the recombinant antibody from the culture super- known for their hydrophobicity and demonstrated that the combi-
natant by afnity chromatography using protein A. This technique nation of hydrophobic and ionic interactions led to the emergence
has proved to be very efcient in terms of selectivity and purity, of new selectivity. Since then, mixed-mode chromatography has
and has become the standard in pharmaceutical companies. How- been used to evaluate its performances for mAb purication that
ever, a survey indicated that 60% of biopharmaceutical companies does not involve protein A [914].
would like to replace the protein A step [1]. Indeed, there are The second and third chromatography steps used to purify
several technical reasons for this such as protein A leakage, the recombinant antibodies should provide orthogonal modes of inter-
formation of protein aggregates during the elution step which is action when compared to the capture step to enable effective
performed at acidic pH, and the nancial aspect linked to the high separation of any remaining contaminants (Host Cell Proteins, DNA,
cost of the protein A resins. These problems explain why numerous viruses) or unwanted forms of the antibody (aggregates). While the
studies have attempted to nd alternative technologies to cap- process uses Protein A for capture, the second and third steps are
ture antibodies. Multimodal or mixed-mode chromatography has respectively cation-exchange chromatography (CEX) and anion-
recently emerged as a candidate for innovative methods of anti- exchange chromatography (AEX) operated in a owthrough mode.
body purication. Mixed-mode chromatography was developed in The CEX step eliminates host cell proteins, aggregates and leached
the late 1950s with hydroxyapatite [2,3]. The following genera- Protein A, whereas the AEX owthrough step removes contaminat-
tions of mixed-mode ligands were developed after 1970 and were ing DNA from the producing cells and achieves further reduction
used in numerous applications [47]. In the 80s, these resins were in host cell protein impurities. This sequence of steps has been
widely used for the purication of nucleic acids but were curiously widely adopted as a generic purication scheme for a number of
recombinant mAbs [15,16]. Concerning the AEX step, it is generally
accepted that the owthrough mode is the most effective. Indeed,
Corresponding author. at neutral pH, the positively charged antibody does not bind to the
E-mail address: charlotte.cabanne@bordeaux-inp.fr (C. Cabanne). matrix, whereas negatively charged species do [17,18]. However,

http://dx.doi.org/10.1016/j.chroma.2015.03.018
0021-9673/ 2015 Elsevier B.V. All rights reserved.
58 S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764

owing to the limitations of the classical chromatography column, 2.4. Liquid chromatographic resins and columns
i.e. ow distribution, ow rate limitations and cleaning validation,
membrane chromatography was developed, and anion-exchange We used three types of chromatographic resin: (i) HEA HyperCel
membrane adsorbers are already being used as an alternative to (Pall Life Science, Saint Germain en Laye, France); (ii) Capto MMC
column chromatography in some mAb manufacturing processes ImpRes (GE Healthcare, Uppsala, Sweden) packed in columns Tri-
[19]. In light of this advantage, protocols have been developed for corn GE Healthcare (4 ml, 1 cm 5.5 cm) and (iii) HiScreen Capto
the use of membrane adsorbers for all purication steps [20]. MMC column (4.7 ml, 0.77 cm 10 cm) prepacked (GE Healthcare,
Our goal was to benet from these recent improvements in Uppsala, Sweden). Height Equivalent to a Theoretical Plate (HETP)
order to develop an innovative process to purify recombinant mAbs and asymmetry were assessed by injection of 5% acetone (v/v) at
produced by CHO cells. Our strategy was to evaluate a three-step 1% of the volume of the column in order to validate the column
process based on two successive mixed mode chromatography package.
steps, followed by an anion-exchange membrane step. Preliminary We used two types of membrane chromatography: (i) a Sar-
experiments to validate the three steps individually were rst per- tobind Q 75 and (ii) a Sartobind STIC PA Nano (Sartorius Stedim
formed using a human antibody. Thereafter, the whole process was Biotech, Aubagne, France). A cleaning protocol was performed after
evaluated for the purication of a recombinant mAb produced by each purication step according to the manufacturers instructions.
CHO cells. We paid special attention to the monitoring of con- The SEC-UPLC column ACQUITY UPLC BEH200 SEC was pur-
taminating proteins and DNA. At each step of the process, DNA chased from Waters (Mildford, USA).
concentration was measured, the global HCPs content was quan-
tied using an ELISA test, and the identity of the contaminating
proteins was determined by mass spectrometry analyses. 2.5. Analysis

2.5.1. Antibody aggregates and quantication analysis by


2. Material and methods SEC-UPLC
Antibody concentration in the collected fractions was deter-
2.1. Equipment mined by Size Exclusion Ultra Performance Liquid Chromatography
(SEC-UPLC). Samples of 10 l were injected onto the SEC column
An Akta Explorer 100 workstation monitored with Unicorn 5.0 regulated at 30 C. The mobile phase consisted in 0.1 M sodium
(GE Healthcare, Uppsala, Sweden) was used for chromatographic sulphate in 0.1 M phosphate buffer pH 6.8. Elution was performed
experiments. Protein detection was performed by measuring UV isocratically at 0.3 ml/min with a run time of 10 min. Antibody peak
absorbance at 280 nm. A HANNA HI8820N conductivity meter and a areas were determined by integration of the 215 nm signal and
HANNA HI2210 pH meter (Tanneries, France) were used to prepare compared to a calibration curve generated with diluted samples of
buffers. a known concentration of human polyclonal antibody SLH66 from
An ACQUITY UPLC system (Waters Corporation, Milford, USA) Equitech-Bio (Kerrville, Texas, USA).
was used for analysis.
The buffers and cell culture supernatant were ltered through
0.45 m cellulose membranes (Sartorius Stedim Biotech, Goettin- 2.5.2. Host cell protein determination by ELISA
gen, Germany) before use. HCPs were quantied with the CHO HCP ELISA kit (#CM015)
from Cygnus Technologies (Southport, NC, USA) according to the
2.2. Biological material high sensitivity protocol recommended by the manufacturer.

Two types of samples were used for purication: (i) a human


immunoglobulin G partially puried from Cohn fractions II and 2.5.3. Host cell DNA
III, which was purchased from Sigma (St Louis, MO, USA) and The Quant-iTTM dsDNA HS assay kit with the Qubit apparatus
(ii) a crude cell culture supernatant from an antibody-producing (Invitrogen, Carlsbad, USA) was used to measure DNA concentra-
CHO cell line (CHO-DP12, ATCC CRL-12445). These cells pro- tion.
duce a humanized anti-IL-8 antibody and have been adapted to
suspension culture in serum-free medium CD CHO (GIBCO Invit-
rogen SARL, Cergy-Pontoise, France) supplemented with 8 mM 2.5.4. Sodium dodecyl sulphate polyacrylamide gel
L-glutamine [21]. Cell cultures were performed in a 2 l stirred tank electrophoresis (SDS-PAGE)
bioreactor with a supply and control tower (DCU, DFC4, Biostat Fractions showing absorbance peaks at 280 nm were analyed
B plus, Sartorius Stedim Biotech, Aubagne, France) and a super- by SDS-PAGE using TGX the Stain-Free FastCast Acrylamide Kit.
vision system (MFCS, Sartorius Stedim Biotech, Aubagne, France). Molecular masses were estimated using standard markers (Bio-Rad
Dissolved oxygen was set at 50%, pH value at 7.4 and culture was Precision Plus Protein Standard). SDS-PAGE gels were revealed by
performed at 37 C. Supernatant was harvested from extended cul- Stain-Free technology (Bio-Rad).
tures until viability dropped below 92%. Cell counting and viability
were determined with a hemocytometer (Malassez type) by using
2.5.5. HCP identication
the trypan blue exclusion method at 0.2% (w/v) nal dye concen-
For each sample, the peptide mixture generated from trypsin
tration [22].
digestion was analyzed by online capillary high-performance liq-
uid chromatography (Dionex, Amsterdam, Netherlands) coupled
2.3. Chemicals to a nanospray LTQ-Orbitrap XL mass spectrometer (Thermo Scien-
tic, Illkirch, France). Data were acquired in a data-dependent mode
Buffers and solutions were prepared using chemicals of ana- alternating, a high-resolution mass spectrometry (MS) scan survey
lytical grade from Sigma (St Louis, MO, USA). Tris-HCl, sodium over the range m/z 3001700, and 10 MS/MS scans in an exclusion
phosphate, sodium citrate, sodium acetate and citric acid were dynamic mode. Identication was carried out by database search
prepared at desired pH and conductivity. against the Uniprot entries from Cricetulus griseus.
S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764 59

of arginine, but it conrmed the low yield. Finally, we evaluated the


use of 50 mM sodium citrate buffer at pH 4 and 6 mS/cm as elution
buffer. The chromatographic prole showed that the pH transition
that occurred between the wash and the elution steps was not opti-
mal, leading to a broader peak. The wash step was thus removed to
allow a larger pH jump. After this change, we obtained a chromato-
graphic prole similar to that previously obtained after elution by
acetate buffer. The corresponding chromatogram is presented in
Fig. 2. A. The percentage of aggregates was reduced to around 10%,
and the yield was around 97%. In conclusion, the sodium citrate
buffer was chosen because it did not require the use of an additive
like Tris-HCl or arginine. Moreover, citrate is more kosmotropic
than acetate, hence the lower amount of aggregates [29].
Fig. 1. Ligands used during the purication process.
3.1.2. Second step: intermediate purication
It has been previously demonstrated that the presence of both
3. Results and discussion an aromatic group and an amide bond allow protein binding at high
salt concentration [30,31]. In view of these observations, GE Health-
3.1. Optimization of each step with a polyclonal antibody care has commercialized the Capto MMC ligand with multimodal
functionality. It contains a carboxyl group that confers the charac-
3.1.1. First step: capture teristics of a weak cation exchanger, and a phenyl group that may
To date, several mixed mode resins have been tested for anti- be involved in hydrophobic interactions. In addition, other types
body capture [23,24]. Both PPA and HEA HyperCel resins gave good of interactions such as hydrogen bonds can occur. The diameter of
results in these studies so we examined the following character- the resin beads (75 m) is less than that of the HEA HyperCel resin
istics to choose the most appropriate one: yield, elimination of (90 m), which leads to better resolution. The structure of Capto
HCPs and pH for elution. The pH value needed to obtain a high MMC resin is presented in Fig. 1.
antibody yield was less acidic with HEA HyperCel than with PPA Conditions for binding and elution were tested in microplates
HyperCel resin. This may be advantageous for the stability of mAb, as described previously [23,28]. Optimal conditions were found to
given that low pH may trigger mAb aggregation. Nevertheless, the be 50 mM sodium citrate buffer pH 4 (6 mS/cm) for equilibration
quantity of HCPs coeluted with the antibody with HEA HyperCel and 200 mM sodium phosphate buffer at pH 7.5 and 21 mS/cm for
was slightly higher than that obtained with PPA HyperCel. How- elution. They were tested in columns with the human IgG prepared
ever, even though the total amount was higher, the number of HCP extemporaneously in the equilibration buffer (Fig. 2B). Desorption
species was lower with HEA HyperCel. We thus selected the latter was obtained by increasing the pH from 4 to 7.5 and by moderately
for the capture step. It contains a hexyl group that could be involved increasing the conductivity (from 6 to 21 mS/cm), which resulted in
in hydrophobic interactions and a protonable amine localized in the decreasing ionic interactions. However, care must be taken not to
spacer arm (Fig. 1). increase the conductivity excessively because this could lead to an
Optimal conditions for mAb capture by HEA HyperCel were increase in the hydrophobic attraction at the expense of the ionic
previously dened [23]. Briey, 0.1 M sodium phosphate buffer repulsion. Another resin has been developed with the same ligand
pH 7.3 and 13 mS/cm were used for equilibration and binding, as the Capto MMC: Capto MMC ImpRes. It has smaller bead size
then 0.1 M sodium phosphate pH 6.5 and 15 mS/cm were used as (40 m) and lower ligand density than Capto MMC in order to
intermediate buffer (wash step) to remove any weakly adsorbed improve resolution and selectivity. Other experiments conrmed
contaminants. Finally, a 50 mM sodium acetate buffer at pH 4 and that it reduced aggregates efciently (data not shown). Similar per-
5 mS/cm was used for elution. Using these conditions, we puried a formances were obtained with this resin in our case as the sample
human IgG prepared extemporaneously in the equilibration buffer. contained few aggregates. Capto MMC is better for productivity
As expected, one peak was observed during the elution step (data while Capto MMC ImpRes is better for removing aggregates. The
not shown). SEC-UPLC was then used to determine the yield and formation of aggregates is mAb- and process-dependent, a factor
the presence of any aggregates. The pattern obtained after SEC- that determines which resin should be used.
UPLC showed that there were more aggregates in the elution peak
(around 80%) than in the starting sample (around 15%). Aggregation 3.1.3. Third step: polishing
of antibodies at an acid pH is known to occur during purication and Anion exchange membrane adsorbers are used in the biophar-
must be limited in order to avoid any undesirable immunological maceutical industry in polishing steps in a ow-through mode
responses [25]. We consider that there are three ways to overcome [3234,19,35]. Indeed, anion exchange is used to bind numerous
this problem: (i) buffering of the elution fraction with 1 M Tris-HCl contaminant species: virus particles, host cell proteins and DNA.
pH 8.5 to raise the pH [26], (ii) addition of arginine to the elution Therefore, the goal is to choose conditions where the contaminant
buffer [27], (iii) the use of sodium citrate as an alternative elution species are negatively charged and thus bind onto the membrane,
buffer: [28]. These three possibilities were tested in the column. whereas the positively charged mAb are able to ow through [36].
First, we buffered the elution fraction by using 1/10 (v/v) of 1 M The expected performance depends on the type of ligand. The qua-
Tris-HCl pH 8.5, which resulted in an increase in the pH of the frac- ternary amine-based ligands are strong anion exchangers but they
tion from 4 to 5.4. As expected, the SEC-UPLC pattern indicated are based only on ionic interactions. Consequently, their capacities
that the percentage of aggregates was reduced to around 10%. The are reduced at high ionic strength. On the contrary, the primary
addition of 1 M arginine-HCl to the elution buffer was also tested, amine-based ligands still have high capacities at high ionic strength
but the elution peak was smaller and broader. In addition, a second owing to their secondary interactions. The latter type is termed
peak appeared during the cleaning step with 0.1 M citric acid, so salt-tolerant.
the elution step was not efcient in these conditions and the anti- First, we tested a classical anion exchanger Sartobind Q (Fig. 1).
body was lost in part. The SEC-UPLC pattern indicated a decrease in The optimal buffer for elution of the mAb during the second chro-
aggregates (around 8%), probably owing to the kosmotropic effect matographic step was phosphate buffer at pH 7.5 and 21 ms/cm. At
60 S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764

Fig. 2. The three steps chosen for purication of human IgG. (A) First step on the HEA HyperCel resin. (B) Second step on Capto MMC resin. (C) Third step on Sartobind Q
membrane.

this conductivity, salts could compete with contaminants for bind- was performed by injecting 200 ml of supernatant (Fig. 3 A). The
ing. Therefore, the human IgG was prepared in this buffer diluted chromatographic prole showed two peaks: a owthrough peak
4-fold in water and then was loaded onto the membrane. The chro- containing unbound proteins and an elution peak that contained
matogram obtained after anion exchanger Sartobind Q purication the antibody. The antibody-containing peak was then applied to
showed a peak in the owthrough fraction as expected (Fig. 2C). the Capto MMC column (Fig. 3B). The prole showed a small
Dilution of the sample before loading onto the membrane could owthrough fraction and a single elution peak. SEC-UPLC con-
be avoided by using a salt-tolerant membrane like Sartobind STIC, rmed that no antibody was lost in the owthrough fraction. The
which is based on a polyallylamine ligand. In this case, a multivalent antibody-containing elution peak was then diluted to 1/4 with
buffer such as a phosphate buffer is not recommended because it water before injection onto the Sartobind Q membrane (Fig. 3C).
can bind to the ligand and thus decrease the binding capacity. We The prole showed a owthrough peak and a small elution peak.
chose to condition the antibody directly in a Tris HCl buffer at pH Fractions were analyzed by SEC-UPLC to measure the amount of
7.0 before its purication with the STIC membrane. Quantication antibody and the percentage of aggregates. HCPs and DNA were also
of the antibody in the owthrough with SEC-UPLC analysis showed quantied in each fraction. These data are summarized in Table 2.
that the yield was good. The Sartobind STIC membrane could be Yields of each purication step ranged from 94 to 99%. Purity of
used as an efcient polishing step. However, as explained above, the mAb was greatly improved throughout the process, increasing
the sample should be previously conditioned in a Tris-HCl buffer, from 45% to 99.9%. Concomitantly, HCPs were reduced at each step
which implies that the preceding chromatographic step should also with a concentration in the nal fraction product of 15 ppm, which
be performed using the same buffer. We tested the second step is an acceptable level. Concerning DNA, 10-fold and 100-fold reduc-
with a Tris-HCl buffer for elution, but the chromatographic prole tions were obtained after the rst and third steps, respectively.
was not satisfactory with a broad peak for the eluted fraction. An SDS-PAGE was performed to analyze the protein content during
exchange of buffer could have been foreseen between the second the process (Fig. 4). The same volume of sample was loaded in each
and the third step, but this would have complicated the process. well. The two bands corresponding to the heavy and light chains
We thus decided to use the Sartobind Q for the polishing step, as were predominant in each puried fraction. Some contaminants
dilution was simpler than buffer exchange.

Table 1
3.2. Purication of a recombinant mAb produced by CHO DP12 Steps and conditions of purication.

Binding Elution
3.2.1. Validation of the whole process
HEA HyperCel Na Phosphate (100 mM); Na Citrate (50 mM); pH
The three steps were selected and optimized independently pH 7.313 mS/cm 46 mS/cm
with human IgG. The process was then validated as a whole with Capto MMC Na Citrate (50 mM); pH Na Phosphate (200 mM);
a mAb from CHO cell culture supernatant. The conditions are sum- 46 mS/cm pH 7.521 mS/cm
marized in Table 1. Purication steps were chained according to Sartobind Q Na Phosphate (50 mM); pH
7.55 mS/cm
the conventional purication process. Capture on HEA HyperCel
S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764 61

Fig. 3. Chromatograms of the three steps for purication of recombinant antibody from CHO cell culture supernatant. (A) First step on HEA HyperCel resin. (B) Second step
on Capto MMC resin. (C) Third step on Sartobind Q membrane.

were detected after the rst step but they fell below detection level
after the second step. Owing to the dilution factor required prior
the Sartobind Q membrane purication step, the intensity of the
mAb bands was lower in this fraction than in the others.

3.2.2. Identication of HCPs


The HCP composition of each fraction, i.e. owthrough, elution
and cleaning fractions, was investigated and identied by mass
spectrometry. The corresponding characteristics (pI, MM) were
retrieved using the Uniprot database and bibliography.
Approximately 400 proteins were identied from the super-
natant. At each step of the process, the number of HCPs signicantly
decreased to 200 and 100 proteins after the rst and second steps,
respectively. At the end of the process, around 30 proteins were
detected.
The distribution of the theoretical isoelectric point (pI) and
grand average of hydropathy (GRAVY) were analyzed (Fig. 5 and
Fig. 6). In the supernatant of the CHO cell culture, the majority of
Fig. 4. SDS-PAGE of antibody-containing fractions from the purication of recom-
binant antibody from CHO cell culture supernatant. MM: Precision Plus ProteinTM
proteins (56.7%) had isoelectric points between 5 and 8, which cor-
Unstained Standards (Biorad). S: culture supernatant. 1: Elution fraction of rst step responds to the normal distribution of eukaryotic cellular proteins
on HEA HyperCel resin. 2: Elution fraction of second step on Capto MMC resin. 3: [37]. During the purication process, we observed a progressive
Flow through fraction of third step on Sartobind Q membrane. enrichment in proteins whose pI ranged from 6 to 8, from 31.3%

Table 2
mAb, HCPs, DNA and aggregate values during the purication process of recombinant antibody from CHO-DP12 cell culture supernatant (for 1 liter of culture).

mAb (mg) Yield (%) Purity (%) HCP (ppm) DNA clearance (log) Aggregates (%)

Supernatant (1 l) 40.2 45 24417 NA 6.5%


Capture HEA HyperCel 37.8 94 98.7 270 2 0.5%
Intermediate Capto MMC 35.5 94 99.3 132 0 0.4%
Polishing Sartobind Q 35.2 99 99.9 15 1 0.3%

DNA clearance is expressed in log 10 reduction; NA: not applicable.


62 S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764

Table 3
HCPs identied in mAb fraction at the end of purication (in ppm). Proteins are presented in decreasing order based on their relative amount in the supernatant.

in the supernatant to 71.4%, 83.6%, and 89.2% after the HEA Hyper- (ii) those with an overall charge unfavorable to ionic interaction but
cel, Capto MMC and Sartobind Q steps, respectively In agreement with sufcient hydrophobicity to counterbalance the ionic repul-
with this observation, the isoelectric point of the puried mAb was sion; (iii) those with pI and hydrophobicity allowing both ionic and
around 8. With regard to hydrophobicity, the majority of proteins hydrophobic interactions; and (iv) those which directly interact g
had GRAVY values in the 0.5 to +0.5 range. The calculated GRAVY with the antibody and thus are co-puried.
for the puried mAb was 0.311. There was a slight increase in the For the sake of clarity, we focused on HCPs that were retrieved
proportion of proteins whose GRAVY index was between 0 and 0.5 in the nal mAb fraction. Table 3 shows the evolution of their
after the rst two purication steps, ranging from 68% to 82%. These relative amounts after each purication step. Most of these pro-
proteins are slightly hydrophobic and were able to establish some teins were already identied in our previous study [23]. Such is
hydrophobic interactions with the two mixed mode resins. This the case of procollagen C-endopeptidase enhancer 1, cathepsin
could explain why some HCPs were enriched despite carrying the L1 and peroxiredoxin-1, whereas cathepsin Z was not found as
same charge of the ligand. In addition, it should be remembered that a main contaminant in the antibody-containing fraction. On the
theoretical pI and GRAVY are calculated with the primary sequence, other hand, we identied other proteins such as cathepsin B, com-
so they cannot account for changes that may occur after protein plement C1r-A subcomponent, and legumain. To explain these
folding. Different classes of HCPs can be co-puried with the anti- differences, the change in the elution buffer between these two
body: (i) those with an overall charge allowing ionic interactions; studies, from sodium acetate to sodium citrate, should be taken into
S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764 63

The overall yield of the process (88%) and the nal purity of
the mAb (99.9%) are comparable to those of conventional afn-
ity process. Impurities were reduced to levels that meet purity
specications for therapeutic products. This new process based on
chromatography techniques could now be easily implemented in
industrial facilities.

References

[1] E.S. Langer, Bioplan Associates, INC., Pharma Manufacturing, 2009.


[2] A. Tiselius, S. Hjerten, O. Levin, Protein chromatography on calcium phosphate
columns, Arch. Biochem. Biophys. 65 (1956) 132155.
[3] S. Hjerten, Calcium phosphate chromatography of normal human serum and of
electrophoretically isolated serum proteins, Biochim. Biophys. Acta 31 (1959)
216235.
[4] B.H.J. Hofstee, R.B. Dunlap (Eds.), Immobilized Biochemicals and Afnity Chro-
Fig. 5. Distribution of proteins according to their pI in the supernatant and the
matography, Plenum Publ. Corp., New York, 1974, pp. 4359.
antibody-containing fraction during the purication of recombinant antibody from
[5] R.J. Yon, R.J. Simmonds, Protein chromatography on adsorbents with hydropho-
CHO cell culture supernatant. bic and ionic groups. Purication of human erythrocyte glycophorin, Biochem.
J. 163 (1977) 397400.
[6] I. Sasaki, H. Gotoh, R. Yamamoto, H. Hasegawa, J. Yamashita, T. Horio,
Hydrophobic-ionic chromatography. Its application to purication of porcine
pancreas enzymes, J. Biochem. 86 (5) (1979) 15371548.
[7] W. Kopaciewicz, M.A. Rounds, F.E. Regnier, Stationary phase contributions to
retention in high-performance anion-exchange protein chromatography: lig-
and density and mixed mode effects, J. Chromatogr. 318 (1985) 157172.
[8] S.C. Burton, D.R.K. Harding, Hydrophobic charge induction chromatography:
salt independent protein adsorption and facile elution with aqueous buffers, J.
Chromatogr. A 814 (1998) 7181.
[9] L. Guerrier, I. Flayeux, E. Boschetti, A dual-mode approach to the selective sep-
aration of antibodies and their fragments, J. Chromatogr. B 755 (2001) 3746.
[10] M.C. Mowry, M. Meagher, L. Smith, J. Marks, A. Subramanian, Production and
purication of a chimeric monoclonal antibody against botulinum neurotoxin
serotype A, Protein Expres. Purif. 37 (2004) 399408.
[11] S. Ghose, B. Hubbard, S.M. Cramer, Evaluation and comparison of alternatives
to Protein A chromatography Mimetic and hydrophobic charge induction chro-
matographic stationary phases, J. Chromatogr. A 1122 (2006) 144152.
[12] H. Bak, O.R.T. Thomas, Evaluation of commercial chromatographic adsorbents
Fig. 6. Distribution of proteins according to their GRAVY value in the supernatant for the direct capture of polyclonal rabbit antibodies from claried antiserum,
and the antibody-containing fraction during the purication of recombinant anti- J. Chromatogr. B 848 (2007) 116130.
body from CHO cell culture supernatant. [13] J. Chen, J. Tetrault, Y. Zhang, A. Wasserman, G. Conley, M. DiLeo, E. Haimes, A.E.
Nixon, A. Ley, The distinctive separation attributes of mixed-mode resins and
their application in monoclonal antibody downstream purication process, J.
Chromatogr. A 1217 (2010) 216224.
account. Indeed, buffers can modify protein conformation through [14] E. Boschetti, Antibody separation by hydrophobic charge induction chromatog-
kosmotropic or chaotropic effects [26]. Consequently, this could raphy, Trends Biotechnol. 20 (2002) 333337.
modify protein-ligand interaction. This was veried for cathep- [15] R.L. Fahrner, H.L. Knudsen, C.D. Basey, W. Galan, D. Feuerhelm, M. Vanderlaan,
G.S. Blank, Industrial purication of pharmaceutical antibodies: development,
sin Z, which was eluted with the antibody when using sodium operation, and validation of chromatography processes, Biotechnol. Gen. Eng.
acetate buffer for elution, and in cleaning fractions when sodium Rev. 18 (2001) 301327.
citrate was used. Finally, after the polishing step, cathepsin L1 and [16] A. Shukla, B. Hubbard, T. Tressel, S. Guhan, D. Low, Downstream processing of
monoclonal antibodiesapplication of platform approaches, J. Chromatogr. B
cathepsin B were the most abundant contaminants. They were not
848 (2007) 2839.
quantitatively abundant in the supernatant, but cathepsin L1 and [17] G. Kemp, P. ONeil, Large-Scale Production of Therapeutic Antibodies: Con-
cathepsin B were enriched during the rst and second steps, respec- siderations for Optimizing Product Capture and Purication, vol. 1, Plenum
Publishers, New York, 2004.
tively. Thanks to the third step based solely on ionic interactions,
[18] H. Knudsen, R. Fahrner, Y. Xu, L. Norling, G. Blank, Membrane ion-exchange
their amount fell to acceptable levels. It has been previously shown chromatography for process-scale antibody purication, J. Chromatogr. A 907
that some HCPs can directly interact with mAbs [38,39]. Such is (2001) 145154.
the case for glutathione S-transferase P1, complement C1r-A sub- [19] J.X. Zhou, T. Tressel, Basic concepts in Q membrane chromatography for large-
scale antibody production, Biotechnol. Prog. 22 (2006) 341349.
component, peroxiredoxin-1, lipoprotein lipase and procollagen [20] H. Varadaraju, S. Schneiderman, L. Zhang, H. Fong, T.J. Menkhaus, Process and
C-endopeptidase enhancer 1. In agreement with this nding, we economic evaluation for monoclonal antibody purication using a membrane-
found these proteins in the puried mAbs fraction. However, our only process, Biotechnol. Prog. 27 (2011) 12971305.
[21] T.N. Gonzalez, S.R. Leong, L.G. Presta (Genentech), Nucleic acids encoding
purication process was efcient enough to lower their amounts humanized anti-IL-8 monoclonal antibodies, US 6025158, 2000.
to levels compatible with regulatory recommendations. [22] B.A. Reeb, Dye exclusion test for bone marrow viability, in: E.M. Areman, H.J.
Deeg, R.A. Sacher (Eds.), Bone Marrow and Stem Cell Processing: A Manual of
Current Techniques, F.A. Davis Company, New York, 1992, pp. 403404.
4. Conclusion [23] J. Pezzini, G. Joucla, R. Gantier, M. Toueille, A.-M. Lomenech, C. Le Snchal, B.
Garbay, X. Santarelli, C. Cabanne, Antibody capture by mixed-mode chromatog-
raphy: a comprehensive study from determination of optimal purication
We describe a three-step process for mAb purication using two conditions to identication of contaminating host cell proteins, J. Chromatogr.
mixed mode resins followed by use of an anion exchange mem- A 1218 (2011) 81978208.
brane for polishing. The proposed sequence is efcient in terms [24] M. Toueille, A. Uzel, J.-F. Depoisier, R. Gantier, Designing new monoclonal anti-
body purication processes using mixed-mode chromatography sorbents, J.
of specicity and efciency so it could be used to replace pro- Chromatogr. B 879 (2011) 836843.
cesses based on protein A chromatography. The two mixed mode [25] A.S. Rosenberg, Effects of protein aggregates: an immunologic perspective,
steps allow good selectivity that is specic to each step. Most of AAPS J. 8 (2006) 501507.
[26] S. Ugwu, S. Apte, The effect of buffers on protein conformational stability,
the main contaminants in the supernatant were removed by these Pharm. Technol. (2004) 86113.
steps. Finally the third step provided orthogonal selectivity and [27] H. Hamada, T. Arakawa, K. Shiraki, Effect of additives on protein aggregation,
good polishing of the remaining DNA and HCPs. Curr. Pharm. Biotechnol. 10 (2009) 400407.
64 S. Maria et al. / J. Chromatogr. A 1393 (2015) 5764

[28] G. Joucla, C. Le Snchal, M. Bgorre, B. Garbay, X. Santarelli, C. Cabanne, Cation [34] H. Shirataki, C. Sudoh, T. Eshima, Y. Yokoyama, K. Okuyama, Evaluation of
exchange versus multimodal cation exchange resins for antibody capture from an anion-exchange hollow-ber membrane adsorber containing -ray grafted
CHO supernatants: identication of contaminating Host Cell Proteins by mass glycidyl methacrylate chains, J. Chromatogr. A 1218 (2011) 23812388.
spectrometry, J. Chromatogr. B 942943 (2013) 126133. [35] J.X. Zhou, T. Tressel, X. Yang, T. Seewoester, Implementation of advanced
[29] A.A. Raibekas, E.J. Bures, C.C. Siska, Ta. Kohno, R.F. Latypov, B.A. Kerwin, Anion technologies in commercial monoclonal antibody production, Biotechnol. J. 3
binding and controlled aggregation of human interleukin-1 receptor antago- (2008) 910.
nist, Biochemistry 44 (2005) 98719879. [36] J. Curling, U. Gottschalk, Process chromatography: ve decades of innovation,
[30] B.-L. Johansson, M. Belew, S. Eriksson, G. Glad, O. Lind, J.-L. Maloisel, N. Norrman, BioPharm. Int. 20 (2007) 1019.
Preparation and characterization of prototypes for multi-modal separation [37] M. Jin, N. Szapiel, J. Zhang, J. Hickey, S. Ghose, Proling of host cell proteins
media aimed for capture of negatively charged biomolecules at high salt con- by two-dimensional difference gel electrophoresis (2D-DIGE): implications
ditions, J. Chromatogr. A 1016 (2003) 3549. for downstream process development, Biotechnol. Bioeng. 105 (2) (2010)
[31] T. Yang, G. Malmquist, B.-L. Johansson, J.-L. Maloisel, S. Cramer, Evaluation of 306316.
multi-modal high salt binding ion exchange material, J. Chromatogr. A 1157 [38] N. Aboulaich, W.K. Chung, J.H. Thompson, C. Larkin, D. Robbins, M. Zhu, A novel
(2007) 171177. approach to monitor clearance of host cell proteins associated with monoclonal
[32] C. Boi, Membrane adsorbers as purication tools for monoclonal antibody antibodies, Biotechnol. Prog. 30 (2014) 11141124.
purication, J. Chromatogr. B 848 (2007) 1927. [39] N.E. Levy, K.N. Valente, L.H. Choe, K.H. Lee, A.M. Lenhoff, Identication and char-
[33] H.L. Knudsen, R.L. Fahrner, Y. Xu, L.A. Norling, G.S. Blank, Membrane acterization of host cell protein product-associated impurities in monoclonal
ion-exchange chromatography for process-scale antibody purication, J. Chro- antibody bioprocessing, Biotechnol. Bioeng. 111 (2014) 904912.
matogr. A 907 (2001) 145154.

Вам также может понравиться