rapid development and scale-up of biosimilar trastuzumab · medicines agency (ema), the us food and...

7
30 BioProcess International 13(4) APRIL 2015 B IO P ROCES S TECHNICAL Rapid Development and Scale-Up of Biosimilar Trastuzumab A Case Study of Integrated Cell Line and Process Development Kumar Dhanasekharan, Claudia Berdugo-Davis, Xiaoming Liu, Carl Richey, Zaneer Segu, David Calabrese, Valerie LeFourn, Pierre-Alain Girod, and Victor Vinci PRODUCT FOCUS: BIOSIMILARS (MONOCLONAL ANTIBODIES) PROCESS FOCUS: PRODUCTION AND DOWNSTREAM PROCESSING WHO SHOULD READ: PROCESS AND PRODUCT DEVELOPMENT, MANUFACTURING, AND ANALYTICAL PERSONNEL KEYWORDS: GENETIC ENGINEERING, DOE, CELL LINE AND PROCESS OPTIMIZATION, SCALE-UP, AND PROCESS CONTROL LEVEL: INTERMEDIATE C ompared with that for new drugs, biosimilar development faces significantly condensed timelines from cell line to first-in-human (FIH) trials. A biosimilar development program needs to accelerate quickly toward preclinical and phase 1 studies; phase 2 studies typically are not required because dose response and other patient-treatment concepts are already established by the original, comparator medicine. Phase 3 studies typically are limited to fewer patients, which ultimately shortens overall timelines and costs. The key challenge remains: demonstrating comparability and high similarity based on in-depth analytics as outlined in a number of guidance documents from the European Medicines Agency (EMA), the US Food and Drug Administration (FDA), and the World Health Organization (WHO) ( 1–3). This often requires multiple iterations during early phase development compared to that for an originator product ( 4). An integrated approach to cell line development and early stage screening and process development is therefore imperative for making appropriate decisions in pursuing the right candidate for preclinical and clinical manufacturing. Here, we present such an approach for a biosimilar trastuzumab, from cell-line through early phase development with manufacturing-ready titers and clinically relevant product quality attributes. Our platform begins with development of a high-producing Chinese hamster ovary (CHO) cell line expressing the desired target protein. Although recombinant technologies are extremely powerful tools for expression of biotherapeutics, manufacturing success depends on identifying cell lines that stably produce high protein titers. A critical part of biopharmaceutical development is choosing the right cells for meeting the objective of confirming cell productivities, cell- line stability, and final-product quality. The most common gene expression systems are based on dihydrofolate reductase (DHFR) and glutamine synthetase (GS). Figure 1A: Cell-line development process used herein, with clone selection in 96-well and expansion to 6-well plates performed manually. Vector construction Transfection Limiting dilution, 96-well plating Screening 1 of clones, then expansion Retransfection of lead clones Limiting dilution and screening 2 of clones, then expansion Fed-batch evaluation in shake flasks Cryo- preservation ELISA and expansion to 24-well plates ELISA and expansion to 6-well plates ELISA, SDS-PAGE, and expansion to shake flask Figure 1B: Analysis of isolated clones in 96-well plates Ranked Clones Titer (mg/L) 140 120 100 80 60 40 20 0 1 21 41 61 81 Screening 1 Screening 2 Author Insights — Online Exclusive http://bit.ly/BPI-author-abs

Upload: others

Post on 12-Jun-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

30 BioProcess International 13(4) April 2015

B i o P r o c e s s TECHNICAL

Rapid Development and Scale-Up of Biosimilar TrastuzumabA Case Study of Integrated Cell Line and Process Development

Kumar Dhanasekharan, Claudia Berdugo-Davis, Xiaoming Liu, Carl Richey,

Zaneer Segu, David Calabrese, Valerie LeFourn, Pierre-Alain Girod, and Victor Vinci

Product Focus: Biosimilars (monoclonal antiBodies)

Process Focus: Production and downstream Processing

Who should read: Process and Product develoPment, manufacturing, and analytical Personnel

KeyWords: genetic engineering, doe, cell line and Process oPtimization, scale-uP, and Process control

level: intermediate

C ompared with that for new drugs, biosimilar development faces significantly condensed timelines from cell line to

first-in-human (FIH) trials. A biosimilar development program needs to accelerate quickly toward preclinical and phase 1 studies; phase 2 studies typically are not required because dose response and other patient-treatment concepts are already established by the original, comparator medicine. Phase 3 studies typically are limited to fewer patients, which ultimately shortens overall timelines and costs.

The key challenge remains: demonstrating comparability and high similarity based on in-depth analytics as outlined in a number of guidance documents from the European Medicines Agency (EMA), the US Food and Drug Administration

(FDA), and the World Health Organization (WHO) (1–3). This often requires multiple iterations during early phase development compared to that for an originator product (4). An integrated approach to cell line development and early stage screening and process development is therefore imperative for making appropriate decisions in pursuing the right candidate for preclinical and clinical manufacturing. Here, we present such an approach for a biosimilar trastuzumab, from cell-line through early phase development with manufacturing-ready titers and clinically relevant product quality attributes.

Our platform begins with development of a high-producing Chinese hamster ovary (CHO) cell line expressing the desired target protein. Although recombinant technologies are extremely powerful tools for expression of biotherapeutics,

manufacturing success depends on identifying cell lines that stably produce high protein titers. A critical part of biopharmaceutical development is choosing the right cells for meeting the objective of confirming cell productivities, cell-line stability, and final-product quality. The most common gene expression systems are based on dihydrofolate reductase (DHFR) and glutamine synthetase (GS).

Figure 1a: Cell-line development process used herein, with clone selection in 96-well and expansion to 6-well plates performed manually.

Vectorconstruction Transfection

Limitingdilution,

96-well plating

Screening 1of clones,

then expansion

Retransfectionof lead clones

Limiting dilutionand screening 2of clones, then

expansion

Fed-batchevaluation inshake �asks

Cryo-preservation

ELISA and expansion to 24-well plates

ELISA and expansion to6-well plates

ELISA, SDS-PAGE, and expansionto shake �ask

Figure 1B: Analysis of isolated clones in 96-well plates

Ranked Clones

Tite

r (m

g/L)

140

120

100

80

60

40

20

01 21 41 61 81

Screening 1 Screening 2

Author Insights — Online Exclusiveh t t p : / / b i t . l y / B P I - a u t h o r - a b s

Page 2: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

32 BioProcess International 13(4) April 2015

However, identifying suitable cell lines is traditionally a time-consuming, labor-intensive process because their productivity and stability can vary enormously. Several thousands of clones must be screened to find those with both the highest yield and the best product quality traits. To improve cell-line selection and reduce timelines, we developed a stable cell line using the SUREtechnology Platform system from Selexis SA. It is based on DNA-based elements (Selexis Genetic Elements, SGEs) that control the dynamic organization of chromatin within all mammalian cells. They allow for unwinding a transgene’s genomic environment — a prerequisite for efficient gene expression. The SGE approach has been shown to prevent transgene silencing and provide high transcription rates for all copies of the transgene, thus enabling higher and more stable expression of recombinant proteins.

Subsequently, we performed design-of-experiment (DoE) studies using shake f lasks to identify base media and feed media strategies, confirming the results at 2-L scale. We further optimized the 2-L process to meet desired product titers and quality. A three-column downstream process was developed to yield high monomer content. We successfully scaled up our process to a pilot-scale setup for both upstream and downstream operations, with comparable process and product attributes. Every step in this process was supported by detailed analyses: e.g., N-glycan analysis to demonstrate comparability and quality, and size-

exclusion chromatography (SEC) and capillary isoelectric focusing (cIEF) to confirm quality based on size variants and charge variants. Finally, we have developed a process ready for transfer to manufacturing using chemically defined medium under an aggressive timeline, with a product titer in the range of 3–4 g/L, while meeting product quality attributes comparable to those of the innovator molecule.

Materials and Methods

We adapted a current good manufacturing practice (CGMP) compliant CHO-K1 ATCC derivative host cell line to grow in BalanCD Growth A media, a commercial chemically defined (CD) product from Irvine Scientific. To keep the environment of the cells unchanged throughout the whole procedure, we used the same medium as a basal medium for transfection, single-cell cloning, and production. We incorporated genes encoding the Hc

and Lc and selection markers into SUREtech vectors containing SGEs to maintain transcription at the maximum level.

Figure 1a is a schematic of the development process we used to establish the cell line for use in this study. DNA was delivered by electroporation to improve transfection efficiencies. We applied an antibiotic selection to create the stable pools, picking about 100 clones for manual expansion from 96-well to 24- and six-well plates. Based on immunoglobulin G (IgG) titers, we transferred selected clones to shake f lasks, then further examined the best performers during a 14-day fed-batch process.

We accomplished those multiple steps of expansion without any antibiotic selection, carefully characterizing and selecting clones for attributes that would be compatible with our production requirements. Finally, we banked and tested 10

Figure 2: Cell culture process design for 2-L and 20-L studies

P0 P1 P2

20 L

2 L

20 L

2 L

25mL 3 L

100mL 10 L

Figure 3: Downstream process design for 2-L and 20-L studies

Protein A withMAbSelect

Sure

Low-pHviral

inactivation

CEX withPOROS XS

Polishing AEXwith POROS HQ

Viral�ltration

UF/DF

Figure 4: Media screening with temperature shift (left); media screening with no temperature shift (right)

Viability (%

)

Time (days)

Via

ble

Cell

Den

sity

(107 c

ells

/mL) 100

9080706050403020100

3.0

2.5

2.0

1.5

1.0

0.5

0.00 2 4 6 8 10 12 14 16 18 20 22 24 26 28

Flask 14

Flask 12

Flask 13

Flask 9

Flask 10

Flask 8Flask 11

Viability (%

)

Time (days)

Via

ble

Cell

Den

sity

(107 c

ells

/mL) 100

9080706050403020100

3.0

2.5

2.0

1.5

1.0

0.5

0.00 2 4 6 8 10 12 14 16 18 20 22 24 26 28

Flask 1

Flask 2Flask 7

Flask 4

Flask 5

Flask 6

Flask 3

Page 3: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

April 2015 13(4) BioProcess International 33

qualified clones for cell identity and for endogenous and adventitious microbial contaminants (bacteria, fungi, and mycoplasma). To analyze their expression constructs at the nucleic-acid level (genetic stability), we assessed the constructs by quantitative polymerase chain reaction (qPCR). Finally, we selected a single clone for further process development.

Process develoPMent and scale-uP

Basal Media Screening and Feed Strategy Development: To evaluate a set of commercially available cell culture media with different feed options, we performed a basal media screening study. To evaluate the media types for both basal and feed media, total feed amount, feed duration, and temperature shift, we performed 14 shake-flask studies with 100-mL working volume (WV). And we subsequently inoculated an additional

12 shake f lasks to further optimize feeding strategy.

To monitor those shake f lasks for cell growth and viability, we used a Vi-Cell counter from Beckman Coulter; to monitor for metabolites, we used a BioProfile Analyzer instrument from Nova Biomedical. We took titer samples on day 3 and then daily after day 5, finally harvesting the f lasks when the cultures reached ~60% viability.

Cell Culture Process Development: To further evaluate and confirm process parameters such as temperature shift, pH, and glucose levels, we ran two sets of four 2-L scale bioreactors, evaluating two different temperature shift conditions (34 °C and 32 °C). Glucose was fed up to 4 g/L when it fell below 3 g/L; pH was controlled using a sodium bicarbonate–carbon dioxide system. The system controlled dissolved oxygen (DO) at a set point of 40% with a fixed air-flow rate and O2

on demand. We defined our feed strategy, total feed amount, and feed duration based on results from our shake-flask studies.

For the seed train, we thawed and passaged cells about every three days. For the last seed-train passage (n – 1) before bioreactor inoculation, we expanded the cells in a Wave bioreactor from GE Healthcare equipped with temperature control. Figure 2 shows this process.

We sampled the culture daily to measure cell density, viability, and metabolite levels. After day 3, we sampled daily for analytical purposes (including titer and quality attributes). Bioreactors were harvested when cultures reached ~60% viability. Using a sterile process, we collected the cell

analytical aBBreviations

CD: Circular dichroism

CE: Capillary electrophoresis

CE-SDS: Sodium dodecyl sulfate capillary electrophoresis

cIEF: Capillary isoelectric focusing

DSC: Differential scanning calorimetry

ELISA: Enzyme-linked immunosorbent assay

FL-HPLC: High-performance liquid chromatography with fluorescence detection

GC: Gas chromatography

HPLC: High-performance liquid chromatography

iCE: Immunoaffinity capillary electrophoresis

ICP-MS: Inductively coupled plasma mass spectrometry

IEX-HPLC: Ion-exchange high-performance liquid chromatography

IgG: Immunoglobulin G

LAL: Limulus amebocyte lysate

LC-MS: Liquid chromatography–mass spectrometry

PCR: Polymerase chain reaction

RP-HPLC: Reverse-phase high-performance liquid chromatography

SDS-PAGE: Sodium dodecyl sulfate polyacrylamide gel electrophoresis

SEC-HPLC: Size-exclusion high-performance liquid chromatography

UV-A280: 280-nm absorbance ultraviolet spectroscopy

Table 1: Typical analytical control strategy for early stage development (Char. = characterization)

Critical Quality Attribute Analytical MethodAnalytical Control Strategy

In-Process Release Stability Char.IgG Protein A HPLC × ×Protein concentration UV-A280 × × ×Protein identification LC-MS peptide

mapping× ×

Physicochemical characterization

LC-MS peptide mapping

× ×

Biophysical analysis Far-UV CD × ×DSC thermal analysis

× ×

Differential fluorimetry

× ×

Functional analysis Binding ELISA ×Cell-based assays ×

Residual DNA PCR ×Residual protein A Protein A ELISA × ×Residual host-cell protein (HCP) HCP ELISA × ×Bioburden Plate-count

methods× ×

Endotoxin LAL assay × ×Charge variants (deamidation, N-terminal pyroglutamates, sialylation)

IEX-HPLC × × × ×cIEF or iCE × × ×

Size variants (aggregation, dimers, fragmentation)

SEC-HPLC × × × ×SDS-PAGE × × × ×CE-SDS × × × ×

Process impurities (Triton X-100, residual solvents, phosphenes, formulation components)

RP-HPLC × ×GC × ×ICP-MS × ×

Glycan profile, intact protein mass

FL-HPLC, LC-MS × ×

Nonglycosylated product CE-SDS × ×

Page 4: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

34 BioProcess International 13(4) April 2015

culture contents and centrifuged them before filtering the supernatant through a 0.2-µm Pall Acropak 500 filter. Filtrate was collected and stored in a –80 °C freezer for downstream processing.

Cell Culture Process Scale-Up to Pilot Scale: To demonstrate scalability, we scaled the 2-L process up to 20 L. We performed several 20-L bioreactor runs using a Sartorius-Stedim BB30 stainless-steel bioreactor. The system maintained scale-independent parameters (pH, pCO2, feed strategy, temperature shift, and so on) as defined in the 2-L bioreactor cell cultures. Scale-dependent parameters were scaled appropriately. We designed the air f low rate to a constant volume per volume per minute (vvm) that translated to 0.15 standard liters per minute (SLPM) with a ring sparger. Agitation rate scale-up was based on constant power per unit volume of f luid (P/V), which translated to 100 rpm.

Downstream Development and Scale-Up: We developed and scaled up a three-column purification process to meet the requirements for our 20-L scale bioreactor run. The downstream process starts with harvest clarification using a POD depth filtration system from EMD Millipore, followed by column operations on a GE Healthcare ÄKTA Pilot system. The protein A capture step used MabSelect SuRe resin from GE Healthcare with phosphate-buffered saline (PBS) equilibration followed by

Figure 5: Effect of feeding timing and frequency on titer

Tite

r (g/

L)4.0

3.5

3.0

2.5

2.0

1.5

1.0

0.5

0.0

Time (days)0 2 4 6 8 10 12 14 16 18 20 22 24

Flask 10

Flask 7Flask 6

Flask 1

Flask 2Flask 4

Flask 3Flask 11

Flask 13Flask 5

Flask 9Flask 8

Figure 6: Comparing the effects of temperature shift (TS) to 32 °C and 34 °C from 37 °C on viable cell density, cell viability, and expression titer

Viability (%

)

Time (days)

Via

ble

Cell

Den

sity

(107 c

ells

/mL)

100

90

80

70

60

50

40

30

20

10

0

3.0

2.5

2.0

1.5

1.0

0.5

0.00 2 4 6 8 10 12 14 16 18 20 22 24 26

2-L TS 32 °C (1)

2-L TS 34 °C (1)

2-L TS 32 °C (2)

2-L TS 34 °C (2)

2-L Baseline

Figure 7: Cell culture process scale-up; (a) viable cell density and viability, (b) titer, (c) glucose profile, and (d) lactate profile

Viability (%

)

Time (days)

Via

ble

Cell

Den

sity

(107 c

ells

/mL)

100

90

80

70

60

50

40

30

20

10

0

2.5

2.0

1.5

1.0

0.5

0.00 2 4 6 8 10 12 14 16 18 20

20-L (2), 1% feed D3−D12, 2% feed D13−18

20-L (1), 1% feed D3−D12, 2% feed D13−18

2-L golden batch

20-L (1), 1% feed D3−D18

20-L (2), 1% feed D3−D18

0.0

0.5

1.0

1.5

2.0

2.5

3.0

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Tite

r (g/

L)

Time (Days)

20-L (1), 1% feed D3−D18

20-L (1), 2% feed D3−D12, 1% feed D13−D18

20-L (2), 2% feed D3−D12, 1% feed D13−D18

20-L (2), 1% feed D3−D18

2-L golden batch

0

1

2

3

4

5

6

7

8

9

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Glu

cose

(g/L

)

Time (Days)

20-L (1), 1% feed D3−D18

20-L (1), 2% feed D3−D12, 1% feed D13−D1820-L (2), 2% feed D3−D12, 1% feed D13−D1820-L (2), 1% feed D3−D18

2-L golden batch

0

1

2

3

4

5

6

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Lact

ate

(g/L

)

Time (Days)

20-L (1), 1% feed D3−D18

20-L (1), 2% feed D3−D12, 1% feed D13−D18

20-L (2), 2% feed D3−D12, 1% feed D13−D18

20-L (1), 1% feed D3−D18

2-L golden batch

A

B

C

D

Page 5: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

36 BioProcess International 13(4) April 2015

loading and washing with PBS. After a secondary wash with PBS and 1 M sodium chloride, we ran a final wash with PBS before eluting the protein with 50 mM sodium acetate at pH 3.4.

After a viral inactivation step, we titrated the pool to pH 5.0. Next, cation exchange (CEX) chromatography in bind–elute mode used POROS XS media from Life Technologies, with equilibration using 50 mM sodium acetate followed by a 4-CV wash at pH 5.0. We used a gradient elution with sodium chloride from 75 to 270 mM over 20 CV. Then a polishing anion-exchange (AEX) chromatography used POROS HQ resin (also from Life Technologies) in f low-through mode, with a 3-CV chase at pH 7.8–8.0. Finally, we processed the eluate through a

Virosart CPV virus filter followed by an ultrafiltration/diafiltration (UF/DF) operation in an ÄKTA Crossflow system from GE Healthcare.

Analytical Development: Table 1 shows a typical analytical strategy during early development. For the current proof-of-concept study, we considered a subset of those methods as presented below.

results

We made research cell banks (RCBs) from the single clone we had selected. Figure 4 shows our initial shake-flask media screening results. Those results helped us identify basal media (Media A) and feed media (Feed B) with a temperature shift (TS) to provide the best performance in

Figure 8: Cell culture process optimized scale-up; (a) VCD and viability, (b) titer, (c) glucose profile, and (d) lactate profile

0

10

20

30

40

50

60

70

80

90

100

0.0

5.0

1.0

1.5

2.0

2.5

0 2 4 6 8 10 12 14 16 18 20 22

Viab

ility

(%)

Viab

le C

ell D

ensi

ty (×

107 c

ells

/mL)

Time (Days)

20-L (2), 1% D3−182-L golden batch

2-L optimized: slow feed2-L optimized: bolus feed

20-L optimized, slow feed20-L optimized, bolus feed

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

0 2 4 6 8 10 12 14 16 18 20 22 24

Tite

r (g/

L)

Time (Days)

2-L golden batch

20L (2), 1% D3−18

2-L optimized: slow feed

2-L optimized: bolus feed

20L-optimized: slow feed

20-L optimized: bolus feed

0.0

1.0

2.0

3.0

4.0

5.0

6.0

7.0

8.0

0 2 4 6 8 10 12 14 16 18 20 22

Glu

cose

(g/L

)

Time (Days)

20L-optimized, slow feed

20L-optimized, bolus feed

2L-optimized: slow feed

2L-optimized: bolus feed

20L-02 1% D3-18

2L golden batch

0

1

2

3

4

5

6

0 2 4 6 8 10 12 14 16 18 20 22

Lact

ate

(g/L

)

Time (Days)

20-L optimized: slow feed

20-L optimized: bolus feed2L optimized: slow feed

2-L optimized: bolus feed

20-L (2) 1% feed D3−18

2-L golden batch

A B

C D

Figure 9: Purity measured by size-exclusion high-performance liquid chromatography (SEC-HPLC) and charge variants measured by capillary isoelectric focusing (cIEF) from Cook Pharmica’s 20-L run compared with the innovator molecule

Time (minutes)

Wav

elen

gth

(AU

)

140

120

100

80

60

40

20

0

−208 9 10 11 12 13 14 15 16 17 18

MonomerRS-Innovator2-L Run

Time (minutes)

Wav

elen

gth

(AU

)

140

120

100

80

60

40

20

0

−20

8 9 10 11 12 13 14 15 16 17 18

Main Peak

Innovator

20-L Run

AcidicPeaks

BasicPeaks

Page 6: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

38 BioProcess International 13(4) April 2015

terms of viable cell density (VCD) and viability over the duration of the runs.

Our feed-strategy optimization results (Figure 5) indicate a nonlinear relationship between total feed percentage and titer. Higher total feed percentages seem to be beneficial up to 16–20%; beyond that point, however, additional feed did not increase titer significantly. Based on statistical analysis using JMP software from SAS Institute, daily feeds from days 3 to 18 gave the best titer results.

A temperature-shift study using 2-L bioreactors showed that the 34 °C condition helped cultures reach higher VCDs, but that viability declines more sharply and earlier than it does at 32 °C. Results are consistent between duplicates, although a shift in growth rate was observed in one run (Figure 6). Based

on protein A HPLC titer measurements, cell culture performance was slightly higher for a TS to 32 °C than it was for a TS to 34 °C.

We studied the combined effects of pH dead-band and glucose control to establish a baseline 2-L process that we labeled as the “golden batch.” Maintaining pH above 6.8 led to higher cell growth rates and higher peak VCDs, but viability dropped to 60% or less sooner. By contrast, maintaining pH >6.6 results in cell cultures with lower peak VCD but extended longevity of viability >60%. To achieve higher cell densities while ensuring extended longevity, we set the pH dead band to 6.8–7.2 and based our TS criteria on reaching a cell density of 1 × 106 cells/mL. And to minimize base consumption for pH control, we increased the post-TS pH dead band to 6.6–7.2.

Cultures controlled at lower glucose levels (around 1 g/L) had lower VCDs than those topped at 4 g/L under the same pH conditions. So we decided to maintain the glucose levels at 4 g/L.

Process Scale-Up Results: As Figure 7 shows, metabolic profiles for the process at 20-L scale compare favorably with those at 2-L scale, although 20-L expression titers f latten out at about 2 g/L whereas viability of the 2-L process declines slower, so titer continues to go beyond 2.5 g/L.

We hypothesized that increased mixing speed (P/V ) and slower additions of feed media and glucose might be required to reduce variability in control of pH, feed, and glucose control. To test our hypothesis, we tried slow and bolus feed media and glucose additions coupled with increased agitation rate. As part of scale-up confirmation, we ran additional 2-L cultures in parallel with the same increased agitation to correspond to a constant P/V and similar feed addition schemes.

As the data show in Figure 8, in all cases the peak cell density was higher than it had been in previous runs, and titer in one of the 20-L runs reached 3.35 g/L. Although the second 20-L run continued on a similar track, a viability drop led us to harvest the reactor early in that case. One key noticeable feature in these runs is the dramatic inf luence on lactate profile of the controlled-feeding strategy in later stages of culture, in which the metabolism shifted to lactate consumption.

We evaluated product quality using size-exclusion high-performance liquid chromatography (SEC-HPLC) for size variants, capillary isoelectric focusing (cIEF) for charge variants, and N-glycan analysis for glycosylation profiles. Although purity was 99.8% after three column steps, at the end of the UF/DF step an overall purity of 99.1% monomer and host-cell protein (HCP) <0.05 ng/mg were reached with an overall product yield of 77%.

As measured by SEC-HPLC, the biosimilar purity is comparable with

Figure 10: Comparing N-glycans released from the innovator product (black) and Cook Pharmica’s 20-L run (blue)

Fucose Mannose Galactose N-acetyl-D-glucosamine Sialic acid

Time (minutes)8 9 10 11 12 13 14 15 16 17 18

Innovator

Mea

sure

men

t? (u

nits

?)25.0

22.5

20.0

17.5

15.0

12.5

10.0

7.5

5.0

2.5

0.0

20-L Run G0-

Glc

NA

c

G0

G0F

M5

G0F

-Glc

NA

cG

1

G1F

a

G

1Fb

G2

G

2f

G2f

+SA

G2f

+2SA

Figure 11: Relative glycoform content from two different 20-L bioreactor runs; afucosylated and fucosylated glycoforms are comparable for both innovator and 20-L bioreactor runs.

54.52

34.86

5.65

88.79

8.33

77.8

14.52

1.57

89.91

7.27

75.88

15.93

1.76

89.73

7.22

G0 G1 G2 Fucosylated Afucosylated

Innovator

20 L (1)

20-L (2)

Page 7: Rapid Development and Scale-Up of Biosimilar Trastuzumab · Medicines Agency (EMA), the US Food and Drug Administration ... development of a high-producing Chinese hamster ovary (CHO)

that of the innovator molecule. Additional product attributes — such as the cIEF charge-variant profiles — are also comparable (Figure 9).

Because our monoclonal antibody (MAb) molecule is a biosimilar product, its N-glycan profile is an important critical quality attribute (CQA) for the design space according to the quality by design (QbD) paradigm. Figure 10 shows the results of our comparison of N-glycans released from the innovator product (black) and Cook Pharmica’s 20-L run product. Clearly, the glycoforms present in the innovator’s molecule are also present in the material we produced. That glycosylation profile is reproducible and consistent across multiple runs.

Relative afucosylation content in both products is also comparable (Figure 11). This is another important CQA for a MAb such as trastuzumab, which has bioactivity based on its binding affinity toward Fcγ receptors on effector cells and clearance of target cells through antibody-dependent cell-mediated cytotoxicity (ADCC) activity. However, fewer end-capped galactosylated glycoforms appear to be present in the biosimilar, and G0 appears to be relatively more abundant than it is in the innovator. Further iterative developments are ongoing to improve comparability and similarity through the use of different culture media supplements. Additional characterization of the innovator molecule is also required to help us understand lot-to-lot variability of our biosimilar MAb.

a successFul PlatForM

We have demonstrated an integrated cell-line development effort with process development and scale-up as a successful model for expediting early phase development for biosimilar antibodies. Our approach did not compromise on the scientifically systematic DoE-based approach, but it took a targeted set of experimental studies to meet the requirements of product quality as well as process scale-up needs. This

approach was successfully demonstrated using a biosimilar Trastuzumab as a model molecule, with scale-up from 2-L bench scale through a 20-L pilot scale process that will be scalable to larger GMP operations for clinical production.

reFerences1 CBER/CDER. Guidance for Industry:

Scientific Considerations in Demonstrating Biosimilarity to a Reference Product. US Food and Drug Administration: Rockville, MD, February 2012; www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/UCM291128.pdf.

2 EMEA/CHMP/BMWP/42832/2005. Guideline on Similar Biological Medicinal Products Containing Biotechnology-Derived Proteins As Active Substance: Non-clinical and Clinical Issues. European Medicines Agency: London, UK, 2006; www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003920.pdf.

3 Guidelines on Evaluation of Similar Biotherapeutic Products. World Health Organization: Geneva, Switzerland, October 2009; www.who.int/biologicals/publications/

trs/areas/biological_therapeutics/BS2110Dft_guidelines_Final_HK_IK_29July_09.pdf.

4 Zelenetz AD, et al. NCCN Biosimilars White Paper: Regulatory, Scientif ic, and Patient Safety Perspectives. J. Natl. Compr. Canc. Netw. 9, 2011: S1–S22. •

Corresponding author Kumar Dhanasekharan ([email protected]) is director of process development; Claudia Berdugo-Davis is a senior scientist, and Xiaoming Liu is an associate scientist in cell culture; Carl Richey and Zaneer Segu are senior scientists in biologics process development and innovation; and Victor Vinci is chief scientific officer and vice president at Cook Pharmica, 1300 South Patterson Drive, Bloomington, IN 47402. David Calabrese is director of cell line services, Valerie LeFourn is head of cell culture, and Pierre-Alain Girod is chief scientific officer at Selexis SA, 18 Chemin des Aulx, 1228 Plan-les-Ouates, Switzerland.

Let ABER take the strain out of your cell counti ng and save valuable ti me while gathering criti cal process

data via our versati le FUTURA Soft ware. Our industry standard ABER probe combined with

our frequency scanning soft ware will help you determine your viable cell concentrati on in real ti me,

without the need for tedious manual counti ng.

ABER Instruments Ltd, Science Park, Aberystwyth, SY23 3AH, UK

Telephone +44 (0) 1970 636 [email protected]

www.aberinstruments.comMEASUR LE...

TIRED OF COUNTING?

BPI_Mar_Aber.indd 1 2/13/15 6:16 PM