mab Titer Analysis with the Agilent Bio-Monolith Protein A Column

Similar documents
Cell Clone Selection Using the Agilent Bio-Monolith Protein A Column and LC/MS

Cell Culture Optimization Using an Agilent Bio-Monolith Protein A Column and LC/MS

Characterize mab Charge Variants by Cation-Exchange Chromatography

Agilent Biocolumns. Titer Determination. Application Compendium

Characterize Fab and Fc Fragments by Cation-Exchange Chromatography

Charge Heterogeneity Analysis of Rituximab Innovator and Biosimilar mabs

Application Note. Author. Abstract. Biopharmaceuticals. Verified for Agilent 1260 Infinity II LC Bio-inert System. Sonja Schneider

Bio-Monolith Protein G Column - More Options for mab Titer Determination

High-resolution Analysis of Charge Heterogeneity in Monoclonal Antibodies Using ph-gradient Cation Exchange Chromatography

Reducing Cycle Time for Charge Variant Analysis of Monoclonal Antibodies

Separate and Quantify Rituximab Aggregates and Fragments with High-Resolution SEC

High-throughput and Sensitive Size Exclusion Chromatography (SEC) of Biologics Using Agilent AdvanceBio SEC Columns

Reversed-phase Separation of Intact Monoclonal Antibodies Using Agilent ZORBAX Rapid Resolution High Definition 300SB-C8 1.

Analysis of Intact and C-terminal Digested IgG1 on an Agilent Bio MAb 5 µm Column

Separation of Recombinant Human Erythropoietin (repo) Using Agilent Bio SEC-3

Disulfide Linkage Analysis of IgG1 using an Agilent 1260 Infinity Bio inert LC System with an Agilent ZORBAX RRHD Diphenyl sub 2 µm Column

Application Note. Authors. Abstract. Biopharmaceuticals

Rapid UHPLC Analysis of Reduced Monoclonal Antibodies using an Agilent ZORBAX Rapid Resolution High Definition (RRHD) 300SB-C8 Column

Seamless Method Transfer from an Agilent 1260 Infinity Bio-inert LC to an Agilent 1260 Infinity II Bio-inert LC

Application Note. Biopharma. Authors. Abstract. James Martosella, Phu Duong Agilent Technologies, Inc Centreville Rd Wilmington, DE 19808

Clone Selection Using the Agilent 1290 Infinity Online 2D-LC/MS Solution

Size Exclusion Chromatography of Biosimilar and Innovator Insulin Using the Agilent AdvanceBio SEC column

Protein Separation with ph Gradients Using Composite Buffer Systems Calculated by the Agilent Buffer Advisor Software

A Comprehensive Workflow to Optimize and Execute Protein Aggregate Studies

Faster Separations Using Agilent Weak Cation Exchange Columns

AdvanceBio HIC: a Hydrophobic HPLC Column for Monoclonal Antibody (mab) Variant Analysis

NISTmAb characterization with a high-performance RP chromatography column

Application of Agilent AdvanceBio Desalting-RP Cartridges for LC/MS Analysis of mabs A One- and Two-dimensional LC/MS Study

Multiple Detector Approaches to Protein Aggregation by SEC

Lifetime stability of size exclusion chromatography columns for protein aggregate analysis

Optimizing Protein Separations with Agilent Weak Cation-Exchange Columns

Monoclonal Antibody Analysis on a Reversed-Phase C4 Polymer Monolith Column

Molecular Characterization of Biotherapeutics The Agilent 1260 Infi nity Multi-Detector Bio-SEC Solution with Advanced Light Scattering Detection

Agilent 1290 Infinity Binary LC System with ISET Emulation of a Waters Alliance 2695 LC Applying Concave, Convex, and Linear Gradients

Agilent OpenLAB CDS MatchCompare for the Comparison of Peptide Mapping Samples

Mobile Phase Optimization in SEC Method Development

Agilent AdvanceBio SEC Columns for Aggregate Analysis: Instrument Compatibility

Chromatography column for therapeutic protein analysis

ph gradient analysis of IgG1 therapeutic monoclonal antibodies using a 5 µm WCX column

ION EXCHANGE KIT FOR MAB SEPARATIONS

Validation of a Dual Wavelength Size Exclusion HPLC Method with Improved Sensitivity to Detect Aggregates of a Monoclonal Antibody Biotherapeutic

Fast and High-Resolution Reversed-Phase Separation of Synthetic Oligonucleotides

Characterization of mab aggregation using a Cary 60 UV-Vis Spectrophotometer and the Agilent 1260 Infinity LC system

TSK-GEL BioAssist Series Ion Exchange Columns

Characterization of IgG monomers & their aggregates

The Agilent 1260 Infinity BioInert Quaternary Pump. Scope of a low-pressure mixing UHPLC pump with Bio-Inert Capabilities

2012 Waters Corporation 1

Fast mass transfer Fast separations High throughput and improved productivity Long column lifetime Outstanding reproducibility Low carryover

A universal chromatography method for aggregate analysis of monoclonal antibodies

ProPac Elite WCX, 5 μm Particle, for Fast, High Resolution Protein and mab Analysis. July 2018

High Throughput Sub-4 Minute Separation of Antibodies using Size Exclusion Chromatography

Choosing the Right Calibration for the Agilent Bio SEC-3 Column

Performance characteristics of the 1260 Infinity Quaternary LC system

Analysis of biomolecules by SEC and Ion-Exchange UPLC

Columns for Biomolecules BioLC Column Lines

Polishing of monoclonal antibodies using Capto S ImpAct

Performance Characteristics of the Agilent 1220 Infinity Gradient LC system

Peptide Mapping: A Quality by Design (QbD) Approach

Separation of Monoclonal Antibodies Using TSKgel HPLC Columns

Quantification of Host Cell Protein Impurities Using the Agilent 1290 Infinity II LC Coupled with the 6495B Triple Quadrupole LC/MS System

Agilent 1290 Infinity II 2D-LC Solution Biopharmaceutical Polymer Analysis. WCBP Jan 2017 Washington, DC

Application Note. Author. Abstract. Biotherapeutics and Biologics. Sonja Schneider Agilent Technologies, Inc. Waldbronn, Germany

Fraction Analysis of Cysteine Linked Antibody-Drug Conjugates Using Hydrophobic Interaction. chromatography. Agilent 1260 Infinity II Bio-Inert System

Size Exclusion BioHPLC columns Ion Exchange BioHPLC columns

Fast and High Resolution Analysis of Intact and Reduced Therapeutic Monoclonal Antibodies (mabs)

Improved Sensitivity with the Agilent 1290 Infinity Evaporative Light Scattering Detector (ELSD)

Reproducible LC/MS Peptide Separation Using Agilent AdvanceBio Peptide Plus Columns

Tools and solutions for separation of charged mab variants

Fast Analysis of Environmental Phenols with Agilent Poroshell 120 EC-C18 Columns

Application Note. Author. Abstract. Pharmaceuticals. A.G.Huesgen Agilent Technologies, Inc. Waldbronn, Germany

Improving Retention Time Precision and Chromatography of Early Eluting Peptides with Acetonitrile/Water Blends as Solvent B

Analysis of Suspected Flavor and Fragrance Allergens in Cosmetics Using the 7890A GC and Capillary Column Backflush Application

Size Exclusion BioHPLC Columns

Application Note. Author. Abstract. Pharmaceuticals. Detlef Wilhelm ANATOX GmbH & Co. KG. Fuerstenwalde, Germany mau

A Fast and Robust Linear ph Gradient Separation Platform for Monoclonal Antibody (MAb) Charge Variant Analysis

Mass Spectrometry Analysis of Liquid Chromatography Fractions using Ettan LC MS System

Physical Stability of a Silica- Based Size Exclusion Column for Antibody Analysis

HyperCel STAR AX Ion Exchange Sorbent

Characterization of Glycosylation in the Fc Region of Therapeutic Recombinant Monoclonal Antibody

3. Close the bottom end of the column and apply the packing device on top. Pump water through the upper adaptor to remove air.

Fast and Efficient Peptide Mapping of a Monoclonal Antibody (mab): UHPLC Performance with Superficially Porous Particles

ADVANCE ACCURACY AND PRODUCTIVITY FOR FASTER ANALYSIS

TOYOPEARL GigaCap Series

Agilent 1290 Infinity Quaternary LC Support of Columns with 2.1 to 4.6 mm ID to 1200 bar

R R Innovation Way P/N SECKIT-7830 Newark, DE 19711, USA Tel: Fax: Website: Published in November 2013

HIC as a Complementary, Confirmatory Tool to SEC for the Analysis of mab Aggregates

Developing Robust and Efficient IEX Methods for Charge Variant Analysis of Biotherapeutics Using ACQUITY UPLC H-Class System and Auto Blend Plus

High-Throughput LC/MS Purification of Pharmaceutical Impurities

Table 1, Performance characteristics of Cellufine SPA-HC

Emulation of the Agilent 1100 Series LC Through Waters Empower Software Analysis of an Analgesic Mixture

Capacity and performance of MabSelect PrismA protein A chromatography resin

Analysis of Monoclonal Antibody Digests with the Agilent 1290 Infinity 2D-LC Solution

High-Resolution Charge Variant Analysis for Top-Selling Monoclonal Antibody Therapeutics Using a Linear ph Gradient Separation Platform

EAG.COM MATERIALS SCIENCES APPLICATION NOTE. By J. Tompkins 1, T. Spurgeon 1, R. Tobias 1, J. Anders 1, E. Butler-Roberts 2, and M.

ADVANCE ACCURACY AND PRODUCTIVITY FOR FASTER ANALYSIS

Rhinophase -AB, Zirconia-based Monoclonal Antibody Purification System

HyperCel STAR AX Ion Exchange Sorbent

Automated Scouting of Stationary and Mobile Phases Using the Agilent 1290 Infinity II Method Development Solution

Transcription:

mab Titer Analysis with the Agilent Bio-Monolith Protein A Column Application Note Biopharmaceuticals and Biosimilars Authors Emmie Dumont, Isabel Vandenheede, Pat Sandra, and Koen Sandra Research Institute for Chromatography (RIC) President Kennedypark 26 B-85 Kortrijk Belgium James Martosella, Phu Duong, and Maureen Joseph Agilent Technologies, Inc. Abstract Monoclonal antibodies (mabs) are becog increasingly important in the treatment of various diseases. During development of recombinant mabs, protein titer and yield from various cell culture supernatants must be monitored. This application note describes how the Agilent Bio-Monolith Protein A column was successfully applied in the deteration of mab concentrations. Introduction Protein A from Staphylococcus aureus has a very strong affinity for the Fc domain of immunoglobulins (IgG), allowing its capture from complex matrixes such as cell-culture supernatants. Affinity chromatography making use of Protein A is the gold standard in therapeutic monoclonal antibody (mab) purification, and typically represents the first chromatographic step in downstream processing. Protein A chromatography finds applications beyond this large-scale purification. At the analytical scale, it is used early in the development of mabs for the high-throughput deteration of mab titer and yield directly from cell culture supernatants, and to purify µg amounts of material for further measurements, for example, by mass spectrometry (MS), ion-exchange (IEX), size exclusion chromatography (SEC), or hydrophobic interaction chromatography (HIC).

This application note describes the use of the Agilent Bio-Monolith Protein A column in mab titer analysis. This HPLC column (Figure 1) has a 5.2-mm id, a column length of 4.95 mm, and is composed of a highly cross-linked poly(glycidyl methacrylate-co-ethylene dimethacrylate) monolithic disk coated with native Protein A from S. aureus. Its monolithic nature, characterized by well-defined channels of 1,2 to 1,5 nm, and by the absence of pores and voids, delivers fast and efficient separations with negligible carryover and excellent robustness. These are features typically expected from a column for mab titer analysis, to successfully guide clone selection and cell-culture optimization. We present the best practice for use of the column in the deteration of absolute mab concentrations in Chinese hamster ovary (CHO) cell-culture supernatants. Data from a trastuzumab biosimilar project are used for illustration purposes. Trastuzumab, marketed as Herceptin since 1998, is used in the treatment of HER2 positive breast cancer, and comes out of patent in 214 in Europe, and 218 in the United States. Experimental Materials Water was obtained from Biosolve (Valkenswaard, The Netherlands). Citric acid, acetic acid, NaH 2 PO 4, and Na 2 HPO 4 were purchased from Sigma-Aldrich, Corp. (St. Louis, MO, USA). Humanized monoclonal antibody trastuzumab, marketed as Herceptin, was obtained from Roche (Basel, Switzerland). Trastuzumab biosimilar CHO cell culture supernatants were obtained from a local biotechnology company. Sample preparation Herceptin stock solution present at 21 mg/ml was diluted in mobile phase A prior to injection. Cell supernatants were diluted 1:1 in 5 mm Na 2 HPO 4. Supernatants were centrifuged at 5, g for 5 utes prior to injection. Instrumentation Agilent Bio-Monolith Protein A measurements were performed on: Agilent 11 Series Quaternary Pump (G1311A) Agilent 11 Series Autosampler (G1313A) Agilent 11 Series Diode Array Detector (G1315A) Software Agilent Technologies OpenLAB CDS ChemStation revision C1.5 (35) Conditions Column: Agilent Bio-Monolith Protein A (p/n 569-3639) Mobile phase: A) 5 mm phosphate, ph 7.4; B) 1 mm citric acid, ph 2.8 mm acetic acid, ph 2.6 Gradient: Time () %B to.5 (binding).6 to 1.7 1 (elution) 1.8 to 3.5 (regeneration) Flow rate: 1 ml/ Injection volume: Variable (5 µl, optimized for CHO cell culture supernatants) Detection: UV at 28 nm A B C Figure 1. Agilent Bio-Monolith Protein A column (A), column housing and monolithic disk (B) and microscopic view on the monolith packing (C). 2

Results and Discussion Buffer selection Figure 2 shows a typical chromatogram from the Protein A column. The example chromatogram is one injection of the supernatant of a specific trastuzumab-producing CHO clone. The unbound material eluted in the flow-through while the mab was retained at neutral ph (binding) and was only released (elution) after lowering the ph upon applying a step gradient. In this case, 5 mm Na-phosphate at ph 7.4 was used for binding/loading, and 1 mm citric acid at ph 2.8 for elution. This represents a good starting condition for any application. When developing a new method for a Protein A column, both binding and elution buffers should be optimized. For binding buffers, 5 mm Na phosphate, ph 7.4, is a good starting point, and can be optimized between ph 7 and 8. For elution buffers, the 1 mm citric acid used here is a good starting point. Other possible elution buffers are 5 mm acetic acid, ph 2.6, 1 mm glycine HCl, ph 2.8, and 12 mm HCl, ph 1.9. Figure 3 compares the elution of a Herceptin originator with acetic acid and citric acid. Very similar peak shape and area were observed, although peaks were slightly sharper using citric acid. In the case of this Herceptin originator, no material was seen in the flow-through, which was not surprising since this represented the marketed product and was devoid of host-cell proteins. In the chromatograms shown, the flow rate was set at 1 ml/. The monolithic nature of the support, characterized by convective instead of diffusive mass transfer, allowed for near flow-rate independence and, hence, high-throughput separations. This is highly desirable in mab titer deteration, which typically requires the processing of a wide range of samples. The maximum flow rate that can be applied on the column is 2 ml/, which allows fast, sub-2-ute separations. 3, 2,5 2, 1,5 1, 5 Binding Elution Regeneration.5 1 1.5 2 2.5 3 Figure 2. UV 28 nm Protein A chromatogram showing the supernatant of a trastuzumab-producing CHO clone. Injection volume was 5 µl. Peak width at half height was.1 utes for the unbound material and.6 utes for the retained mab. A 1, 8 6 4 2.5 1 1.5 2 2.5 3 8 6 4 2 B.5 1 1.5 2 2.5 3 Figure 3. UV 28 nm Protein A chromatogram of Herceptin originator diluted in 5 mm Na-phosphate, ph 7.4, to.5 mg/ml (5-µL injection, 25-µg column load). Elution was achieved using citric acid (A) and acetic acid (B). Peak width at half height is.57 and.67 utes for citric acid and acetic acid, respectively. 3

Precision, linearity, carryover, and injection size Precision is critically important in the deteration of the mab titer. Table 1 shows the peak area and retention time repeatability that can typically be expected upon injecting a Herceptin originator 1 times. Chromatograms are shown in Figure 4. More than acceptable relative standard deviation (RSD) values were obtained for both citric acid and acetic acid as elution buffers. Carryover was simultaneously assessed by injecting a buffer blank after the mab injection sequence (Figure 5). At a 1-fold column load of 5 µg, carryover appeared to be nonexistent, which can again be attributed to the use of a monolithic support. Carryover at 1% levels became apparent upon a single load of 5 µg of mab onto the column. This represents the maximum column load and is one typically not encountered in real-life experiments. It is worth noting that carryover was eliated after the injection of a second buffer blank. Table 1. Retention time and peak area RSD values obtained for the 1-fold analysis of a Herceptin originator at.5 mg/ml (5-µL injection volume). Acetic acid Citric acid Peak area RT () Peak area RT () 1 361 1.669 383 1.666 2 362 1.668 372 1.666 3 373 1.668 365 1.665 4 365 1.669 389 1.667 5 37 1.669 383 1.666 6 373 1.669 378 1.666 7 367 1.671 379 1.668 8 365 1.668 377 1.666 9 366 1.67 376 1.667 1 36 1.67 377 1.667 Mean 366 1.669 378 1.667 S 4.64.1 6.52.1 % RSD 1.27.6 1.73.6 8 6 4 2.5 1 1.5 2 2.5 3 Figure 4. Replicate (n = 1) UV 28 nm Protein A chromatograms of a Herceptin originator diluted in 5 mm Na phosphate, ph 7.4, to.5 mg/ml (injection volume 5 µl). Elution was achieved using acetic acid. 4

8 6 4 2.5 1 1.5 2 2.5 3 Figure 5. UV 28 nm Protein A chromatograms of a Herceptin originator diluted in 5 mm Na phosphate, ph 7.4, to.5 mg/ml, and a blank buffer analyzed after a sequence of 1 Herceptin injections. Elution was achieved using acetic acid, and injection volumes were 5 µl. The limit of detection (LOD) was around column loads of.5 µg. This put some demands on injection volume. If samples have low mab levels, high volume injections are required. Figure 6 shows the linearity obtained when increasing the injection volume from 5 to 5 µl for a 1 mg/ml Herceptin originator. With the knowledge that 5 µl injections are perfectly feasible and that the lowest detectable amount on-column is.5 µg, samples with mab concentrations at 1 µg/ml are within reach. Area 5, 4,5 4, 3,5 3, 2,5 2, 1,5 1, 5 R 2 =.9999 5 1 15 2 25 3 35 4 45 5 Injection volume (µl) Figure 6. Linearity obtained by increasing the injection volume of a Herceptin originator (.5 mg/ml) from 5 to 5 µl. 5

In mab titer deteration, it is important to be able to assess absolute mab concentrations. These can be found by linking the peak areas measured in cell-culture supernatants to an external calibration curve constructed by diluting a mab standard. For the Herceptin biosimilar project, this standard was found in the originator product, which was accurately formulated at 21 mg/ml. The calibration curves of a dilution series of Herceptin originators using citric acid and acetic acid as elution buffers are shown in Figure 7. The corresponding chromatograms are shown in Figure 8. In both cases, linearity was excellent, between.2 mg/ml and 2 mg/ml, which is the typical mab titer range in CHO cells. Area 9, A 1, B 8, R 2 =.9999 9, 7, 8, 7, 6, 6, 5, 5, 4, 4, 3, 3, 2, 1,.2.4.6.8 1 1.2 1.4 1.6 1.8 2 Concentration (mg/ml) Area 2, 1, R 2 =.9996.2.4.6.8 1 1.2 1.4 1.6 1.8 2 Concentration (mg/ml) Figure 7. Herceptin Protein A calibration curve (.2 to 2 mg/ml) using citric acid (A) and acetic acid (B) as elution buffer. A 2,5 2, 1,5 1, 5.5 1 1.5 2 2.5 3 3.5 4 2 mg/ml 1 mg/ml.5 mg/ml.2 mg/ml.1 mg/ml.5 mg/ml.2 mg/ml 2,5 B 2, 1,5 1, 5.5 1 1.5 2 2.5 3 3.5 4 2 mg/ml 1 mg/ml.5 mg/ml.2 mg/ml.1 mg/ml.5 mg/ml.2 mg/ml Figure 8. Overlaid UV 28 nm Protein A chromatograms of Herceptin calibration points using citric acid (A) and acetic acid (B) as elution buffer. 6

Application in mab titer deteration The method possesses all the characteristics for the deteration of mab titer in cell-culture supernatants. It is fast, precise, and linear in the expected mab concentration range and does not suffer from carryover. To illustrate this, nine trastuzumab-producing clones, generated in the framework of a Herceptin biosimilar development program, were analyzed using the Bio-Monolith Protein A column to detere absolute mab concentrations. Chromatograms are displayed in Figure 9, and Table 2 reports the obtained mab titers using both citric acid and acetic acid as elution buffer. Very consistent data were generated using both elution buffers. These results allow clear decisions to be made early in the development of mabs. High-producing clones can be readily selected and subjected to further development. Table 2. Comparison between the absolute mab concentrations in the different trastuzumab CHO clones obtained using either citric acid or acetic acid as elution buffer. CHO clone Concentration (mg/ml) acetic acid 3.251.21 8.296.256 9.492.494 1.728.757 24.256.262 25.133.98 26.126.9 28.24.173 32.178.144 Concentration (mg/ml) citric acid 45 4 35 3 25 2 15 1 5 4 35 3 25 2 15 1 5 A.5 1 1.5 2 2.5 3 B.5 1 1.5 2 2.5 3 Figure 9. Overlaid UV 28 nm Protein A chromatograms of nine trastuzumab-producing CHO clones using citric acid (A) and acetic acid (B) as elution buffer. 7

Maximizing column lifetime Column regeneration A major benefit of using a monolithic disk is that the presence of channels instead of pores decreases the likelihood of column clogging when injecting cell-culture samples. This increases robustness and reduces cleaning efforts. Column contaation can be reduced by running a blank gradient injection after every 3 to 5 samples. If column deterioration is observed (tailing or broad peaks), the following cleaning procedure is recommended. Column regeneration is the first step. If performance is still suboptimal, the clean-in-place procedure can be used, which will reduce the amount of Protein A available. Column regeneration 1. Wash with 2 ml (2 column volumes (CV)) of 1 mm phosphate buffer + 1 M NaCl, ph 7 to 8, at.5 to 1. ml/. 2. Wash with 2 ml (2 CV) of low-ph solution (such as elution buffer). 3. Re-equilibrate with binding buffer. Conclusions The Agilent Bio-Monolith Protein A column is an ideal tool for the deteration of mab titer in cell-culture supernatants. Several method parameters can be optimized for the best method on the Protein A column, including binding and elution buffers and injection size (volume and concentration). As demonstrated here, the column delivers high-throughput, precise, accurate, linear, and robust separations in the concentration range encountered in real-life situations, and allows clear decisions to be made towards clone selection and cell-culture optimization. For More Information These data represent typical results. For more information on our products and services, visit our Web site at www.agilent.com/chem. Clean-in-place 1. Wash with 1 to 2 ml (1 to 2 CV) of.1 M NaOH (reverse flow direction) at.2 to.5 ml/. 2. Wash with 1 to 2 ml (1 to 2 CV) of DI water at.5 to 1. ml/. 3. Wash with 1 to 2 ml (1 to 2 CV) of concentrated buffer (.1 to.5 M) to restore normal ph (7. to 7.4). 4. Re-equilibrate with 5 ml (5 CV) of binding buffer. www.agilent.com/chem For Research Use Only. Not for use in diagnostic procedures. Agilent shall not be liable for errors contained herein or for incidental or consequential damages in connection with the furnishing, performance, or use of this material. Information, descriptions, and specifications in this publication are subject to change without notice. Agilent Technologies, Inc., 214, 217 Printed in the USA November 6, 217 5991-5135EN