real-time detection of monoclonal antibodies in protein a

1
Real-Time Detection of Monoclonal Antibodies in Protein A Breakthrough and in Process Samples Atul Goyal 1 , Ujwal Patil 2 , Vijay Maranholkar 2 , Binh Vu 1 , Katerina Kourentzi 1 , and Richard C. Willson 1,2,3 1 Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX, USA; 2 Department of Biology & Biochemistry, University of Houston, Houston, TX, USA; 3 Escuela de Medicina y Ciencias de la Salud ITESM Monterrey, MX Acknowledgements References 1. Najera, M. (2017), Genetic Engineering & Biotechnology News, 37(1), 20-21. 2. Chmielowski, R. A., et al. (2017), J. Chromatogr. A 1526, 58–69. 3. Steinebach, F. et al. (2016), Biotechnol. J. 11, 1126–1141. 4. Skoglar, H., et al. (2015), Bioprocess Int. 13, 1–8. 5. Ghose, S., et al. (2007), Biotechnology and bioengineering, 96(4), 768-779. 6. Rüdt, M., et al. (2017), J. Chromatogr. A, 1490, 2-9. 7. Nilsson, B., et al. (1987), Protein Engineering, Design and Selection 1.2: 107-113. 8. DeLano, W. L., et al. (2000), Science, 287(5456), 1279-1283. 9. Patil, U., et al. (2020), Biosens. Bioelectron., 165, 112327. Figure 1. (A) Fluorescence polarization. Fluors are attached to Fc-binding ligands. When unbound, these reporters rotate faster and emit depolarized light. Larger molecules, such as antibodies, rotate slowly. The binding of the low molecular weight ligands to antibodies results in high fluorescence polarization. (B) Fluorescence intensity. Excitation (black) and emission (red) spectra of fluorescein. The typical fluorescence intensity of unbound affinity reporter is indicated by free and enhanced fluorescence after affinity reporter binds to IgG is indicated by bound. Figure 3. The FITC-labeled Fc-binding ligands (50 nM) were tested against increasing IgG concentrations. (A) Shift in fluorescence polarization was observed for all the Fc-binding ligands. (B) Interestingly, a significant increase in fluorescence intensity was observed for Z3 ( ), Z5 ( ) and peptide ( ). Figure 2. Affinity ligands used in this study. (A) Z domain derived from staphylococcal protein A (Ref 7), trimer (Z3) and pentamer (Z5) are polymerized forms of the Z domain. (B) IgG- binding domain from streptococcal protein G (PDB: 2IGG). (C) 13-mer Fc-binding cyclic peptide (Fc-III, Ref 8). Figure 5. Human IgGs were spiked in Murine-Ab-depleted hybridoma culture fluid (IMDM with 10% FBS). The concentration of each Fc-reporter was 50 nM. Phenol red in the culture medium increases the level of background fluorescence. Presence of cell culture fluid does not significantly affect the human IgG-induced shift in fluorescence polarization (A) and fluorescence intensity (B). Figure 7. Protein A process control using real-time IgG monitoring. (A) Schematic of set-up and communication in the process control scheme. (B) FI (red) is monitored in real-time throughout the loading phase, and baseline is determined from the initial 20 - 40 min with no IgG in the effluent stream. A FI value of 90 intensity units is set as the column switching threshold (black arrow) in the Python script. As the IgG starts to break through, it binds to Fc-reporter resulting in FI increase. When the FI value meets the preset triggering criterion, the Python script sends a valve-switch command to purification unit through Python OpenOPC interface. A dip in UV280 (blue) and FI signal is observed as the equilibration buffer in the column is displaced by incoming feed. C domain of protein A PDB:4NPD (Staphylococcus aureus) IgG PDB:1IGT Figure 4. IgG complex with Fc-binding ligands. (A) IgG structure with protein A binding site, Fc (C H 2-C H 3). (B) The B-domain from protein A is a three-helix bundle (PDB: 5U4Y; 59 AAs); Z-domain is variant of the B-domain with Ala/Val at the N-terminus and a Gly/Ala substitution in Helix 2. (C) Fc-III peptide in complex with the Fc-region at the C H 2-C H 3 domain interface (PDB: 1DN2; 13 AAs). X-ray structure of the peptide bound to Fc suggests that the FITC attached to the N-terminus of the peptide by an aminocaproic acid linker is in proximity to His 433, which we speculate could affect its emission intensity in the bound state. Special thanks to Dr. Glen Bolton (Amgen), Dr. David Robbins (AstraZeneca), Dr. David Roush (Merck Research Labs), and Dr. Kent Göklen (GlaxoSmithKline) for valuable discussions. We would like to thank the Carbonell group and BTEC, NCSU for donating CHO-CCF. This project was a finalist for the MilliporeSigma 2018 Life Science Award in Bioseparations. Fluorescence Polarization Unbound reporter has higher mobility and lower polarization Bound reporter has lower mobility and higher polarization Z domain (derived from staphylococcal protein A) Streptococcal protein G Fc-binding cyclic peptide Fc-binding Reporters Fluorescein isothiocyanate (FITC) Z3 Z5 Protein G Peptide Fluorescence Intensity IgG in PBS (1 g/L) PBS Pump B UV Reagent pump FITC-labeled Z3 (100 nM) PBS , No IgG IgG in PBS PBS , No IgG IgG in PBS A) B) A) A) Fluorescence Polarization Z3 Z5 B) C) A) Fluorescence Polarization B) Fluorescence Intensity F C C) C H 2 C H 3 C L V L C H 1 V H A) IgG concentration 380 430 480 530 580 Wavelength (nm) Excitation Emission bound free D) Introduction Measurement of antibody concentrations is ubiquitous in biopharmaceutical process development and manufacturing. Purification of therapeutic monoclonal antibodies (mAbs) usually involves a protein A affinity capture step. Because column breakthrough of antibody in complex, UV-absorbing culture fluid cannot be readily detected in real time, processes are conservatively designed and column capacity often is underutilized, wasting adsorbent and reducing productivity. Fluorescence Reporting Batch IgG Detection Process Control Binding Site on IgG-Fc Figure 6. Change in the fluorescence intensity and polarization detected upon mixing with human IgG in continuous flow. UV280 absorbance measured by AKTA Explorer. In the absence of IgG in the flow (region before the dashed arrow), the fluorescence intensity (red) and polarization (green) remain low. Fluorescence intensity and polarization increase as the IgG-rich stream flows through the system. The polarization detector is situated downstream of the intensity detector. The system can detect 50 mg/L IgG in a flow of 1 mL/min. The dashed arrow indicates the start of IgG-rich stream. Flow rate: 1 mL/min Flow rate: 1 mL/min IgG Detection in Flow: Soluble reporter We have developed a fluorescence-based monitoring technology which allows mix-and-read mAb detection in cell culture fluid (Ref 9), which may be useful in at-line assays and in clone and culture development, and here report the use of reporters immobilized on agarose monolith supports for continuous detection of IgG in column breakthrough. The agarose monolith structure was optimized using residence time distribution measurement across a range of cooling rates. Column effluent was continuously contacted with immobilized fluorescein-labeled Fc-binding ligands to produce an immediately-detectable shift in fluorescence intensity. The technology allows rapid and reliable monitoring of IgG in a flowing stream, without prior sample preparation. We observed significant shifts in fluorescence intensity at 0.05 g/L human IgG, sufficient to detect 5% breakthrough of a 1 g/L load within 4 minutes or 8 CV of the monolith at a flow rate of 0.5 mL/min. The fluorescence intensity response at different load concentrations was used to calibrate fluorescence intensity with IgG concentration. B) A) Fluorescence Intensity B) Fluorescence Polarization IgG (1 g/L) in CHO-CCF Protein A columns A1 & A2 Flow velocity = 80 cm/h Fluorescein-labeled Z3 reporter (50 nM) Mixer Pump A Pump B Mixer Reagent pump UV AKTA Unit 1 2 3 4 2 A) A1 A2 B) Z3-reporter Immobilized Reporters for Continuous IgG Monitoring Figure 8. Monolith preparation and activation, and immobilization of FITC-labeled protein A ligand on agarose monolith. In step 1, equal volumes of preheated 6% (w/v) agarose in DI water and cyclohexane with 5.7% Tween 80 (v/v) were mixed. In step 2, the emulsion was created by vigorous mixing. In step 3, the emulsion was poured into the glass column. In step 4, agarose rods were solidified at 8 o C. After 5 minutes, the glass column was fitted with a flow distributor sieve, adaptor, and tubing. In step 5, the organic phase was removed by pumping water, ethanol-water (1:1, v/v), and finally degassed water through the column. In step 6, the monolith surface was activated with sodium periodate, followed by water and 1x PBS washes. In step 7, the activated surface was conjugated with FITC-labeled protein A and the Schiff base was reduced with sodium cyanoborohydride. Figure 9. Visualization of reporter loading. Immobilization of FITC-labeled protein A on agarose monoliths was visualized by UV excitation and imaging under the ethidium bromide emission filter. 1. control, no loading of reporter, 2. batch mode, and 3. frontal loading. In the batch mode, the reporter is introduced in the monolith and Schiff base reaction was allowed to happen overnight. In the frontal loading, the reporter was continuously introduced in the monolith at a very slow flow rate till breakthrough was complete. Cooling temperature Residence time (sec) Number of theoretical plates/m Asymmetry factor 8 o C (faster cooling) 19.1 153.3 1.5 13.4 164.4 1.8 8.9 215.0 1.8 Room temperature 19.1 4.9 5.3 13.4 0.4 4.8 8.9 5.0 4.1 Table 1. Effect of agarose cooling rate (step 4, Figure 8) on NaCl residence time distribution and asymmetry. Agarose cooling was carried out at room temperature and at 8 o C. 1 2 3 IgG Detection in Flow: Immobilized reporter Fluorescence polarization and intensity approaches can be used to detect process- relevant levels of targets in column breakthrough and in batch samples. Fluorescence intensity is especially attractive due to the availability of inexpensive commercial detectors, and simpler automation. Fluorescence polarization is very generally applicable, including in the absence of intensity-reporting ligands, and may also be applicable to viruses, such as lentivirus - levels in cell culture fluid. Fluorescence intensity shows significant increase (7% from the baseline value) after loading 5% breakthrough of 1 g/L IgG concentration. Further work needs to be devoted to reducing the fluorescence baseline variability and to monolith regeneration. Figure 10. Detection of 5% breakthrough of 1 g/L IgG concentration using fluorescence intensity. Time course of the fluorescent intensity of the FITC- labeled protein A immobilized on the agarose monolith. IgG concentration of 0.05 g/L in 1x PBS introduced into the monolith starting at 28 min. Elution was done with glycine (100 mM, pH 3.5) and washing was done with 1x PBS. Conclusions B) Fluorescence Intensity Fluorescence polarization/ intensity detector Mixer Mixer Fc-binding ligands Pump A Fluorescence intensity monitor 0 10000 20000 30000 40000 50000 60000 70000 80000 90000 0 50 100 150 Fluorescence intensity (a.u.) Time (min) t = 28 min; 0.05 g/L IgG added Wash Elution Wash IgG Detection in Cell Culture Fluid IgG in PBS IgG in PBS Peptide Z3 Z3 Peptide

Upload: others

Post on 12-May-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Real-Time Detection of Monoclonal Antibodies in Protein A

Real-Time Detection of Monoclonal Antibodies in Protein A Breakthrough and in Process SamplesAtul Goyal1, Ujwal Patil2, Vijay Maranholkar2, Binh Vu1, Katerina Kourentzi1, and Richard C. Willson1,2,3

1Department of Chemical & Biomolecular Engineering, University of Houston, Houston, TX, USA; 2Department of Biology & Biochemistry, University of Houston, Houston, TX, USA;3Escuela de Medicina y Ciencias de la Salud ITESM Monterrey, MX

Acknowledgements

References1. Najera, M. (2017), Genetic Engineering & Biotechnology News, 37(1), 20-21.2. Chmielowski, R. A., et al. (2017), J. Chromatogr. A 1526, 58–69.3. Steinebach, F. et al. (2016), Biotechnol. J. 11, 1126–1141.4. Skoglar, H., et al. (2015), Bioprocess Int. 13, 1–8.5. Ghose, S., et al. (2007), Biotechnology and bioengineering, 96(4), 768-779.6. Rüdt, M., et al. (2017), J. Chromatogr. A, 1490, 2-9.7. Nilsson, B., et al. (1987), Protein Engineering, Design and Selection 1.2: 107-113. 8. DeLano, W. L., et al. (2000), Science, 287(5456), 1279-1283.9. Patil, U., et al. (2020), Biosens. Bioelectron., 165, 112327.

Figure 1. (A) Fluorescence polarization. Fluors are attached to Fc-binding ligands. Whenunbound, these reporters rotate faster and emit depolarized light. Larger molecules, such asantibodies, rotate slowly. The binding of the low molecular weight ligands to antibodiesresults in high fluorescence polarization. (B) Fluorescence intensity. Excitation (black) andemission (red) spectra of fluorescein. The typical fluorescence intensity of unbound affinityreporter is indicated by free and enhanced fluorescence after affinity reporter binds to IgG isindicated by bound.

Figure 3. The FITC-labeled Fc-binding ligands (50 nM) were tested against increasing IgGconcentrations. (A) Shift in fluorescence polarization was observed for all the Fc-bindingligands. (B) Interestingly, a significant increase in fluorescence intensity was observed for Z3( ), Z5 ( ) and peptide ( ).

Figure 2. Affinity ligandsused in this study. (A) Zdomain derived fromstaphylococcal protein A (Ref7), trimer (Z3) and pentamer(Z5) are polymerized forms ofthe Z domain. (B) IgG-binding domain fromstreptococcal protein G (PDB:2IGG). (C) 13-mer Fc-bindingcyclic peptide (Fc-III, Ref 8).

Figure 5. Human IgGs were spiked in Murine-Ab-depleted hybridoma culture fluid (IMDMwith 10% FBS). The concentration of each Fc-reporter was 50 nM. Phenol red in the culturemedium increases the level of background fluorescence. Presence of cell culture fluid doesnot significantly affect the human IgG-induced shift in fluorescence polarization (A) andfluorescence intensity (B).

Figure 7. Protein A process control using real-time IgG monitoring.(A) Schematic of set-up and communication in the process controlscheme. (B) FI (red) is monitored in real-time throughout the loadingphase, and baseline is determined from the initial 20 - 40 min with noIgG in the effluent stream. A FI value of 90 intensity units is set as thecolumn switching threshold (black arrow) in the Python script. As theIgG starts to break through, it binds to Fc-reporter resulting in FIincrease. When the FI value meets the preset triggering criterion, thePython script sends a valve-switch command to purification unitthrough Python OpenOPC interface. A dip in UV280 (blue) and FIsignal is observed as the equilibration buffer in the column is displacedby incoming feed.

C domain of protein A PDB:4NPD

(Staphylococcus aureus) IgG

PDB:1IGT

Figure 4. IgG complex with Fc-binding ligands. (A) IgG structure with protein A binding site,Fc (CH2-CH3). (B) The B-domain from protein A is a three-helix bundle (PDB: 5U4Y; 59AAs); Z-domain is variant of the B-domain with Ala/Val at the N-terminus and a Gly/Alasubstitution in Helix 2. (C) Fc-III peptide in complex with the Fc-region at the CH2-CH3domain interface (PDB: 1DN2; 13 AAs). X-ray structure of the peptide bound to Fc suggeststhat the FITC attached to the N-terminus of the peptide by an aminocaproic acid linker is inproximity to His 433, which we speculate could affect its emission intensity in the bound state.

Special thanks to Dr. Glen Bolton (Amgen), Dr. David Robbins (AstraZeneca), Dr. David Roush (MerckResearch Labs), and Dr. Kent Göklen (GlaxoSmithKline) for valuable discussions. We would like to thank theCarbonell group and BTEC, NCSU for donating CHO-CCF. This project was a finalist for the MilliporeSigma2018 Life Science Award in Bioseparations.

Fluo

resc

ence

Pol

ariz

atio

n

Unbound reporter has higher mobility and lower polarization

Bound reporter haslower mobility andhigher polarization

Z domain (derived from staphylococcal protein A)

Streptococcalprotein G

Fc-binding cyclic peptide

Fc-binding Reporters

Fluorescein isothiocyanate (FITC)

Z3

Z5

Protein G

Peptide

Fluo

resc

ence

Inte

nsity

IgG in PBS (1 g/L)

PBS

Pump B

UV

Reagent pump

FITC-labeled Z3 (100 nM)

PBS , No IgG

IgG in PBS

PBS , No IgG

IgG in PBS

A) B)

A)

A) Fluorescence Polarization

Z3

Z5

B)

C)

A) Fluorescence Polarization B) Fluorescence Intensity

FC

C)CH2

CH3

CL

VLCH1

VH

A)

IgG concentration380 430 480 530 580

Wavelength (nm)

Excitation Emission

bound

free

D)

IntroductionMeasurement of antibody concentrations is ubiquitous in biopharmaceutical processdevelopment and manufacturing. Purification of therapeutic monoclonal antibodies (mAbs)usually involves a protein A affinity capture step. Because column breakthrough of antibody incomplex, UV-absorbing culture fluid cannot be readily detected in real time, processes areconservatively designed and column capacity often is underutilized, wasting adsorbent andreducing productivity.

Fluorescence Reporting

Batch IgG Detection

Process Control

Binding Site on IgG-Fc

Figure 6. Change in the fluorescence intensity and polarization detected upon mixing withhuman IgG in continuous flow. UV280 absorbance measured by AKTA Explorer. In theabsence of IgG in the flow (region before the dashed arrow), the fluorescence intensity (red)and polarization (green) remain low. Fluorescence intensity and polarization increase as theIgG-rich stream flows through the system. The polarization detector is situated downstream ofthe intensity detector. The system can detect 50 mg/L IgG in a flow of 1 mL/min. The dashedarrow indicates the start of IgG-rich stream.

Flow rate: 1 mL/min

Flow rate: 1 mL/min

IgG Detection in Flow: Soluble reporter

We have developed a fluorescence-based monitoring technologywhich allows mix-and-read mAb detection in cell culture fluid (Ref9), which may be useful in at-line assays and in clone and culturedevelopment, and here report the use of reporters immobilized onagarose monolith supports for continuous detection of IgG incolumn breakthrough. The agarose monolith structure wasoptimized using residence time distribution measurement across arange of cooling rates. Column effluent was continuously contactedwith immobilized fluorescein-labeled Fc-binding ligands toproduce an immediately-detectable shift in fluorescence

intensity. The technology allows rapid and reliable monitoring of IgG in a flowing stream,without prior sample preparation. We observed significant shifts in fluorescence intensity at0.05 g/L human IgG, sufficient to detect 5% breakthrough of a 1 g/L load within 4 minutesor 8 CV of the monolith at a flow rate of 0.5 mL/min. The fluorescence intensity response atdifferent load concentrations was used to calibrate fluorescence intensity with IgGconcentration.

B)

A) Fluorescence Intensity B) Fluorescence Polarization

• IgG (1 g/L) in CHO-CCF

• Protein A columns A1 & A2

• Flow velocity = 80 cm/h

• Fluorescein-labeled Z3 reporter (50 nM)

MixerPump A

Pump B

MixerReagent pump

UV

AKTA Unit1

2

3

42A)

A1 A2

B)

Z3-reporter

Immobilized Reporters for Continuous IgG Monitoring

Figure 8. Monolith preparation and activation, and immobilization of FITC-labeled protein Aligand on agarose monolith. In step 1, equal volumes of preheated 6% (w/v) agarose in DIwater and cyclohexane with 5.7% Tween 80 (v/v) were mixed. In step 2, the emulsion wascreated by vigorous mixing. In step 3, the emulsion was poured into the glass column. In step4, agarose rods were solidified at 8 oC. After 5 minutes, the glass column was fitted with aflow distributor sieve, adaptor, and tubing. In step 5, the organic phase was removed bypumping water, ethanol-water (1:1, v/v), and finally degassed water through the column. Instep 6, the monolith surface was activated with sodium periodate, followed by water and 1xPBS washes. In step 7, the activated surface was conjugated with FITC-labeled protein A andthe Schiff base was reduced with sodium cyanoborohydride.

Figure 9. Visualization of reporter loading.Immobilization of FITC-labeled protein A onagarose monoliths was visualized by UVexcitation and imaging under the ethidiumbromide emission filter. 1. control, no loading ofreporter, 2. batch mode, and 3. frontal loading. Inthe batch mode, the reporter is introduced in themonolith and Schiff base reaction was allowed tohappen overnight. In the frontal loading, thereporter was continuously introduced in themonolith at a very slow flow rate tillbreakthrough was complete.

Cooling temperature Residence time (sec) Number of theoreticalplates/m Asymmetry factor

8 oC (faster cooling)

19.1 153.3 1.5

13.4 164.4 1.8

8.9 215.0 1.8

Room temperature

19.1 4.9 5.3

13.4 0.4 4.8

8.9 5.0 4.1Table 1. Effect of agarose cooling rate (step 4, Figure 8) on NaCl residence time distributionand asymmetry. Agarose cooling was carried out at room temperature and at 8 oC.

1 2 3

IgG Detection in Flow: Immobilized reporter

• Fluorescence polarization and intensity approaches can be used to detect process-relevant levels of targets in column breakthrough and in batch samples.

• Fluorescence intensity is especially attractive due to the availability of inexpensivecommercial detectors, and simpler automation.

• Fluorescence polarization is very generally applicable, including in the absence ofintensity-reporting ligands, and may also be applicable to viruses, such as lentivirus -levels in cell culture fluid.

• Fluorescence intensity shows significant increase (7% from the baseline value) afterloading 5% breakthrough of 1 g/L IgG concentration. Further work needs to be devotedto reducing the fluorescence baseline variability and to monolith regeneration.

Figure 10. Detection of 5%breakthrough of 1 g/L IgGconcentration using fluorescenceintensity. Time course of thefluorescent intensity of the FITC-labeled protein A immobilized on theagarose monolith. IgG concentrationof 0.05 g/L in 1x PBS introduced intothe monolith starting at 28 min.Elution was done with glycine (100mM, pH 3.5) and washing was donewith 1x PBS.

Conclusions

B) Fluorescence Intensity

Fluorescence polarization/ intensity detector

MixerMixer

Fc-binding ligands

Pump A

Fluorescence intensity monitor

0100002000030000400005000060000700008000090000

0 50 100 150

Fluo

resc

ence

inte

nsity

(a.u

.)

Time (min)

t = 28 min; 0.05 g/L IgG added

Wash Elution

Wash

IgG Detection in Cell Culture Fluid

IgG in PBS

IgG in PBSPeptide

Z3

Z3

Peptide