Zum Hauptinhalt springen

Optimization of the Imaged cIEF Method for Monitoring the Charge Heterogeneity of Antibody-Maytansine Conjugate

Abbood, Ayat
In: Journal of Analytical Methods in Chemistry, 2023
Online unknown

Optimization of the Imaged cIEF Method for Monitoring the Charge Heterogeneity of Antibody-Maytansine Conjugate  1. Introduction

The aim of this study was to develop a whole-column imaging-detection capillary isoelectric focusing (icIEF) method for the analytical characterization of charge heterogeneity of a novel humanized anti-EphA2 antibody conjugated to a maytansine derivative. In addition to focusing time, sample composition was optimized: pH range, percent of carrier ampholytes, conjugated antibody concentration, and urea concentration. A good separation of charge isoforms was obtained with 4% carrier ampholytes of a large (3–10) and narrow pH range (8–10.5) (1 : 1 ratio), conjugated antibody concentration (0.3–1 mg/ml) with a good linearity (R2: 0.9905), 2 M of urea concentration, and 12 minute for focusing. The optimized icIEF method demonstrated a good interday repeatability with RSD values: <1% (pI), <8% (% peak area), and 7% (total peak areas). The optimized icIEF was useful as an analytical characterization tool to assess the charged isoform profile of a discovery batch of the studied maytansinoid-antibody conjugate in comparison to its naked antibody. It exhibited a large pI range (7.5–9.0), while its naked antibody showed a narrow pI range (8.9–9.0). In the discovery batch of maytansinoid-antibody conjugate, 2% of charge isoforms had the same pI as the pI of naked antibody isoforms.

Monoclonal antibodies (mAbs) constitute a very important therapeutic class, with more than 100 mAbs approved by the US Food and Drug Administration (FDA) for the treatment of different diseases [[1]]. Nearly half of approved mAbs (45%) were anticancer.

Naked mAbs are the most common type used as antitumor. Conjugating antibodies with radioisotopes, chemotherapeutic drugs, or toxins were extensively investigated to improve the antitumor efficiency of an antibody [[2]–[10]]. Antibody-drug conjugates (ADCs) can selectively deliver cytotoxic drugs directly to targeted cancer cells.

ADCs may present a considerable heterogeneity resulting from various modifications in the protein structure of antibody itself. Furthermore, linking several drug molecules per antibody decreases the homogeneity of ADC [[11]]. These probable modifications may lead to the presence of different isoforms in crude and final ADC products. Therefore, full characterization of ADCs should be performed including charge heterogeneity, size heterogeneity, and peptide mapping [[13]–[16]].

The main quality attribute of mAb or ADC characterization is the determination of charge variants [[17]]. Monitoring the charge variants of mAb or ADC gives information on protein stability, product purity from batch to batch, the pathways of degradation, and so on [[19]].

The charge heterogeneity of ADC is characterized using several analytical methods as well as liquid chromatography [[20]], ion exchange chromatography (IEC) [[21]–[23]], capillary electrophoresis (CE) [[24]–[26]], capillary isoelectric focusing (cIEF) [[27]–[32]], and imaged capillary isoelectric focusing (icIEF) [[33]–[35]].

Isoelectric focusing (IEF) is widely used for the separation of proteins based on their isoelectric point (pI). Compared to conventional gel IEF, cIEF has several advantages. cIEF offers higher resolution, speed, and quantitative analysis [[36]].

In cIEF, a solution of carrier ampholytes and sample is used to fill the capillary. The two ends of the capillary are placed into acidic and basic solutions. When an electric field is applied, the presence of the ampholytes permits to create a pH gradient through the capillary. Proteins migrate through the capillary until they reach a region of pH equal to their pI, where they become neutral and stop to migrate, resulting in a series of narrow focused zones. In the conventional cIEF, the focalisation of analytes is followed by subsequent mobilization of the focused sample zone to the detection point by different methods (pressure, vacuum, and gravity). Indeed, the mobilization step greatly affects the separation efficiency and reproducibility of the method. Also, the single-point detection cannot permit to monitor the progress of the separation process. Whole-column detection (WCD), for which there is no mobilization step, allows for the simultaneous detection along the entire length of a column and is a better option.

The aim of this study was to separate and determine the pI isoform profile of a novel humanized anti-EphA2 antibody conjugated to a cytotoxic maytansine derivative. To achieve this objective, a whole-column imaging-detection capillary IEF (icIEF) method was optimized and applied to a discovery batch of the studied maytansinoid-antibody conjugate.

2. Materials and Methods

2.1. Materials

Kit ICE280 chemical test, Kit ICE280 electrolytic solution, methyl cellulose, and 1%, 0.5%, and pI markers (6.6, 7.05, 8.18, 9.5, and 10.10) were purchased from Convergent Bioscience. Urea was from Sigma. Pharmalyte solutions (3–10 and 8–10.5) were from GE healthcare.

A monoclonal naked antibody and its maytansinoid conjugate were analyzed. The discovery batch of antibodies was formulated in HGS buffer consisted of histidine 10 mM, glycine 130 mM, and sucrose 5% (w/v).

2.2. Sample Preparation

A protein sample was prepared by diluting to a desired final concentration in 0.35% methyl cellulose, 2% pharmalytes (3–10), and 2% pharmalytes (8–10.5) (1 : 1 ratio) and urea of the desired concentration. pI markers (6.61, 8.81, 7.05, and 9.5) were added to the sample for pI calibration.

An example demonstrating the preparation of an antibody sample (8 mg/ml) (final volume 200 μl and final concentration 1 mg/ml) was reported in Table 1. Each test sample was then vortexed by centrifugation. After centrifugation, the sample was transferred to a glass autosampler vial and centrifuged to remove bubbles before placing in the autosampler carousel for analysis.

Table 1 An example of 200 μl sample preparation of ADC sample.

1 mg/mlVolume μlFinal concentration
Methyl cellulose 1%700.35%
Pharmalytes 3–1042%
Pharmalytes 8–10.542%
Urea (8 M)502 M
ADC 8 mg/ml251 mg/ml
Water Milli-Q45
pI marker 6.611
pI marker 9.51

2.3. icIEF Apparatus

The icIEF analysis was conducted using an iCE280 instrument with a PrinCE autosampler and capillary cartridge from Convergent Bioscience. A transparent capillary column (50 mm, 100 μm ID, and 200 μm OD) is embedded into the glass cartridge, with its inner surface coated with a fluorocarbon to minimize electroosmotic flow. Reservoirs for cathodic (100 mM NaOH and 0.1% methyl cellulose) and anodic solutions (80 mM H3PO4 and 0.1% methyl cellulose) were attached to the glass cartridge and separated from the capillary by the hollow fiber membrane. Protein focusing time ranged from 7 to 15 min at 3000 V, and detection at 280 nm was achieved with a CCD camera.

3. Results and Discussion

3.1. Development of the icIEF Method

To characterize the charge heterogeneity of maytansinoid-antibody, the sample, in the preliminary experiments, composed of ADC, 0.35% methyl cellulose, 4% of ampholyte solution with a pH range from 3 to 10 (pharmalytes 3–10), 2 M urea, and two pI markers (6.61 and 9.50).

In order to optimize the separation of charge variants of maytansinoid-antibody by the icIEF method, several experimental parameters have to be considered and optimized: sample pH range and percent of carrier ampholytes, sample ADC concentration, sample urea concentration, and focusing time. Table 2 summarizes the different experimental conditions for the optimization of charge isoform separation of studied ADC by icIEF.

Table 2 Optimized experimental parameters for ADC analyses by iCE280.

Ampholyte pH rangePharmalytes 8–10.5/pharmalytes 3–10
Sample concentration0.3, 0.5, 0.8, 1, and 1.5 mg/ml
Urea concentration0, 1, 2, and 3 M
Focusing time7, 10, 12, and 15 min
% ampholytes4%, 6%, and 8%

3.1.1. pH Range of Carrier Ampholytes

A solution of carrier ampholytes contains an extensive mixture of zwitterionic compounds of different pI values, which can produce a pH gradient in the capillary. With uniform absorbance along the whole pH range at 280 nm, they are commercially available at different ranges of pH: wide (3–10) and narrow (8–10.5, 5–8). In the preliminary conditions, some charge isoforms were co-migrated with the marker 9.50 using 4% ampholyte solution with a wide pH range (3–10). Therefore, the ampholyte solution with a narrow pH range from 8 to 10.5 (pharmalytes 8–10.5) was added to the pharmalyte solution (3–10) in ratio 1 : 1. Thanks to the addition of ampholytes with narrow pH range, the different charge isoforms of maytansinoid-antibody were separated from the markers 9.5 (Figure 1). As we can see from this figure, the studied maytansinoid-antibody had several isoforms with pI values varied from 7.5 to 9.0.

Graph: Figure 1 Analysis of maytansinoid-antibody by icIEF. Experimental conditions: final concentration 1 mg/mL in 0.35% methyl cellulose, 2% pharmalytes 3–10 and 2% pharmalytes 8–10.5 (1 : 1 ratio), 2 M urea, pI markers: 6.61, 9.50. Focusing time: 10 min at 3000 V. λ = 280 nm.

3.1.2. Maytansinoid-Antibody Concentration

In order to study the effects of the final concentration sample on charge isoform separation, maytansinoid-antibody samples were prepared at different final concentrations: 0.3, 0.5, 0.8, 1, and 1.5 mg/ml (Figure 2).

Graph: Figure 2 Analysis of different concentrations of maytansinoid-antibody by icIEF (0.3, 0.5, 0.8, 1, and 1.5 mg/ml). Other experimental conditions are as mentioned in Figure 1.

The increase of maytansinoid-antibody concentrations led to a shift of pI isoforms values towards higher values. This shift of pI values may be related to the increase of additives with increasing maytansinoid-antibody concentration in the sample matrix (histidine, glycine, and sucrose). These additives can impact the linearity of the ampholyte pH gradient and lead to changes of pI values [[38]]. These additives may also adsorb on the wall of capillary and suppress residual electroosmotic flow, leading to a shift of pI isoforms values.

A saturation of the UV absorbance was observed at a concentration of maytansinoid-antibody of 1.5 mg/ml. A good linearity was demonstrated for the measurement of isoforms over the range of sample concentrations (0.3–1 mg/ml) (Figure 3). For the following studies, a maytansinoid-antibody concentration of 1 mg/ml was chosen.

Graph: Figure 3 Plot of total peak area against maytansinoid-antibody concentration (0.3, 0.5, 0.8, 1, and 1.5 mg/ml).

3.1.3. Urea Concentration

In cIEF, the charge isoform separations can be greatly affected by protein aggregation and precipitation resulted from low protein solubility at or near the pI and at high protein concentrations in the focused bands. Out of a few tested additives known to increase protein solubility, such as nonionic surfactants or urea, the presence of urea had a significant influence on charge isoform separation of ADC. The amount of urea in a sample matrix of maytansinoid-antibody has been varied over the concentration range from 0 to 3 M (Figure 4).

Graph: Figure 4 Effects of urea concentration on the separation of charge isoforms of maytansinoid-antibody by icIEF (0, 1, 2, and 3 M urea). Other experimental conditions are as mentioned in Figure 1.

Clearly, the presence of urea improved the charge profile of the maytansinoid-antibody. At a urea concentration of 0 or 1 M, there were the spikes due to the aggregation of proteins into the capillary. However, the charge isoform separation profile was stable over a urea concentration of 2 M. Therefore, 2 M urea has been chosen as the optimal concentration for charge isoform separation of maytansinoid-antibody for the following experiments.

Urea concentration can affect the separation of protein charge isoforms and the apparent pI values [[38]]. For example, Turner and Schiel studied the effects of urea concentration on the apparent pI of the main mAb charge variants [[38]]. The results showed that the apparent pI of the main mAb charge variant was significantly affected by added urea, probably due to urea-mediated denaturation. In order to clarify the impact of added urea, pI values of main ADC charge variants were plotted against urea concentration (Figure 5). A slight decrease of the pI values was remarked with increasing urea concentration.

Graph: Figure 5 Effect of urea concentration on the pI values of main ADC charge variants.

We should note that urea addition also increased the viscosity of the solution in the capillary and decreased the electrophoretic mobility of the components. Therefore, focusing times need to be optimized for the selected urea concentration.

3.1.4. Focusing Time

Focusing time was varied between 7 and 15 min to ascertain that it was long enough for the maytansinoid-antibody isoforms of high relative molecular masses. The profile of maytansinoid-antibody, as shown in Figure 6 as a function of focusing time, indicated that the resolution between charge isoforms improved by increasing the focusing time from 7 min to 15 min. However, the focusing time of 15 min was too long; the marker of pI (9.5) disappeared from the window of detection. Therefore, the focusing time of 12 min was chosen as the optimal conditions of charge isoforms separation.

Graph: Figure 6 Effects of focusing time on the separation of charge isoforms of maytansinoid-antibody by icIEF. Other experimental conditions are as mentioned in Figure 1.

3.1.5. Ampholyte Percent

In order to optimize the separation of charge isoforms of maytansinoid-antibody, the effects of ampholyte percent of 1 : 1 mixture of a narrow pH range (8–10.5) and a wide pH range (3–10) were studied: 4%, 6%, and 8% (Figure 7). As shown in Figure 7, increasing the ampholyte percent led to a decrease of resolution between maytansinoid-antibody isoforms. These results demonstrated that the 4% ampholytes are a good choice for the separation of maytansinoid-antibody charge isoforms.

Graph: Figure 7 Effects of % pharmalytes on the separation of charge isoforms of maytansinoid-antibody by icIEF. Other experimental conditions are as mentioned in Figure 1.

To develop a method for separation and pI determination of charge isoforms of maytansinoid-antibody by icIEF, different experimental conditions were optimized.

With increasing maytansinoid-antibody concentration in the sample matrix from 0.3 to 1.5 mg/ml, a shift of pI values of charge variants to higher values was observed. In addition, loss of linearity was remarked when increasing maytansinoid-antibody concentration to 1.5 mg/ml as a result of a saturation of UV absorbance. The maytansinoid-antibody concentration was fixed at 1 mg/ml.

The pI markers of 6.61 and 9.50 were the appropriate markers as internal standards since the pI values of charge isoforms of maytansinoid-antibody ranged from 7.5 to 9.0.

The addition of narrow range (8–10.5) to wide range (3–10) pharmalytes led to the improvement of resolution between charge isoforms of maytansinoid-antibody. With increasing the percent of mixture of wide range and narrow range pharmalytes (1 : 1 ratio) from 4% to 8%, the quality of charge isoform separation decreased. Therefore, 4% pharmalytes have been selected to ensure the optimal resolution.

The urea concentration in the sample matrix has been varied from 0 to 3 M. The presence of urea led to the increase of maytansinoid-antibody solubility (the absence of peaks corresponding to antibody aggregates in the icIEF profile) and to improve the resolution of charge isoforms. The urea concentration had slight effects on the charge isoform pI values. The separation of maytansinoid-antibody charge isoforms was stable from 2 to 8 m urea. 2 M urea has been selected to obtain optimal and repeatable charge isoform resolution.

In addition to the different additives in the sample matrix of the maytansinoid-antibody, focusing time is a key parameter to obtain a repeatable and stable icIEF profile. With increasing the focusing time from 7 to 15 min, the resolution of charge isoforms increased. A loss of marker of pI (9.5) was observed at 15 min. Therefore, 12 min at 3000 V was enough time to achieve the optimal separation of charge isoforms.

3.2. Interday Repeatability of the Optimized icIEF Method

To test the interday repeatability of the developed method for maytansinoid-antibody, icIEF profiles were performed within three consecutive days. The statistically obtained results, summarized in Table 3, demonstrated a good interday repeatability.

Table 3 Statistical results of interday repeatability of icIEF profile of maytansinoid-antibody (n = 6 and 3 days).

pI% areaTotal area
MeanSDRSD%MeanSDRSD%MeanSDRSD%
8.40.0170.2110.4404524000366807
8.50.0340.4150.7505
8.60.0170.2130.6505
8.70.0260.3110.7707
8.80.0090.170.2804
8.90.0090.140.1604
90.0180.21.70.1026

3.3. Application of the Developed icIEF Method

The developed icIEF method was applied to characterize the charge heterogeneity profile of the discovery batch of the studied maytansinoid conjugate (Figure 8(a)) in comparison with its naked antibody (Figure 8(b)). The naked antibody showed a narrow pI range (8.9–9.0), while the studied maytansinoid conjugate exhibited a large pI range (7.5–9.0). The level of unconjugated antibody in an ADC formulation is a critical parameter in process control because it can directly affect the efficacy of ADC. From Table 4, the percent of charge isoforms corresponding to naked antibody represented 2% of charge isoforms in the ADC discovery batch. These results demonstrated the success of the conjugating process of a naked antibody with a maytansinoid derivative.

Graph: Figure 8 icIEF profiles of naked antibody (a) and its corresponding maytansinoid conjugate (b). Final concentration of maytansinoid-antibody in sample matrix is 1 mg/ml diluted in 0.35% methyl cellulose, 4% 3–10 pharmalytes/8–10.5 pharmalytes (1 : 1 ratio), and 2 M urea, pI markers: 8. 18, 9.50 (a) and 6.61, 9.50 (b), and focusing time of 12 min at 3000 V. Other experimental conditions are as mentioned in Figure 1.

Graph: (b)

Table 4 Comparison of charge profiles of studied naked antibody and its corresponding ADC, including isoform number, pI range, ΔpI, and pI and % area of major species.

Isoform numberpI rangeΔpIpI and % area of major species
Naked antibody28.9–9.00.1Peak 1: pI 8.9 : 36%, Peak 2: pI 9.0 : 64%
Maytansinoid-antibody127.5–91.5Peak 1: pI 8.4 : 11%, Peak 2: pI 8.5 : 15%, Peak 3: pI 8.6 : 13%, Peak 3: pI 8.7 : 11%, Peak 4: pI 8.8 : 7%, Peak 5: pI 8.9 : 4%, Peak 6: pI 9.0 : 1.7%

This heterogeneity of ADC is related to the covalent linking of the cytotoxic drug to the free amine groups of lysine of mAb. Monoclonal antibodies often have 40–60 lysine residues, and chemical conjugation results in a heterogeneous mixture consisting of unconjugated mAbs and mAbs conjugated with a variable number of cytotoxins in random combinations at different sites on the antibody [[40]]. The charge variants of maytansinoid conjugate differed by the number of amine groups of lysine conjugated to a linker molecule, leading to decrease their pI with increase of the number of modified amino groups (more acidic). The obtained results were in agreement with a study of charge variants of IgG1-Fc and conjugated IgG1-Fc performed by Boylon et al. [[42]]. The study demonstrated that the chemical conjugation of IgG1-Fc to different drugs via the amino acid Lys modified the electrostatic properties of the mAb surface and introduces further complexities. The chemical conjugation led to the decrease in pI upon conjugation.

The developed chromatofocusing is suitable for the analytical characterization of charge heterogeneity of the studied monoclonal antibody and its drug conjugate based on differences in the isoelectric points of the variants presents.

4. Conclusion

Whole-column imaging-detection capillary isoelectric focusing (icIEF) is a promising technique to check the quality of therapeutic proteins. In this work, an icIEF method was developed for monitoring the charge heterogeneity of maytansinoid-monoclonal antibody. The composition of maytansinoid-monoclonal antibody was optimized including pH and percent of carrier ampholytes, conjugated antibody concentration, urea concentration, and focusing time. Under optimized condition, peaks corresponding to charge variants of conjugated antibody were separated with good resolution. Method results showed a good interday repeatability and linearity within the concentration range of 0.3–1 mg/ml. The developed method was applied to assess the quality of discovery batch of maytansinoid-monoclonal antibody [[43]].

Data Availability

The data used to support the findings of this study are available from the corresponding author upon request.

Conflicts of Interest

The author declares that there are no conflicts of interest.

Authors' Contributions

The author alone is responsible for the content and writing of this article.

Acknowledgments

This work was supported by the Tishreen University.

REFERENCES 1 Kaplon H., Reichert J. M. Antibodies to watch in 2021. mAbs. 2021; 13(1), 10.1080/19420862.2020.1860476, 1860476 2 Baah S., Laws M., Rahman K. M. Antibody-drug conjugates-A tutorial review. Molecules. 2021; 26(10): 2943, 10.3390/molecules26102943 3 Khongorzul P., Ling C. J., Khan F. U., Ihsan A. U., Zhang J. Antibody-drug conjugates: a comprehensive review. Molecular Cancer Research. 2020; 18(1): 3-19, 10.1158/1541-7786.mcr-19-0582 4 Sievers E. L., Senter P. D. Antibody-drug conjugates in cancer therapy. Annual Review of Medicine. 2013; 64(1): 15-29, 10.1146/annurev-med-050311-201823, 2-s2.0-84873053339 5 Hafeez U., Parakh S., Gan H. K., Scott A. M. Antibody–drug conjugates for cancer therapy. Molecules. 2020; 25(20): 4764, 10.3390/molecules25204764 6 Dean A. Q., Luo S., Twomey J. D., Zhang B. Targeting cancer with antibody-drug conjugates: promises and challenges. mAbs. 2021; 13(1), 10.1080/19420862.2021.1951427, 1951427 7 Drago J. Z., Modi S., Chandarlapaty S. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nature Reviews Clinical Oncology. 2021; 18(6): 327-344, 10.1038/s41571-021-00470-8 8 Criscitiello C., Morganti S., Curigliano G. Antibody-drug conjugates in solid tumors: a look into novel targets. Journal of Hematology and Oncology. 2021; 14(1): 20, 10.1186/s13045-021-01035-z 9 Wang N., Mei Q., Wang Z., Zhao L., Zhang D., Liao D., Zuo J., Xie H., Jia Y., Kong F. Research progress of antibody-drug conjugate therapy for advanced gastric cancer. Frontiers Oncology. 2022; 12, 10.3389/fonc.2022.889017, 889017 Fu Z., Li S., Han S., Shi C., Zhang Y. Antibody drug conjugate: the "biological missile" for targeted cancer therapy. Signal Transduction and Targeted Therapy. 2022; 7(1): 93, 10.1038/s41392-022-00947-7 Cauchon N. S., Oghamian S., Hassanpour S., Abernathy M. Innovation in chemistry, manufacturing, and controls-A regulatory perspective from industry. Journal of Pharmaceutical Sciences. 2019; 108(7): 2207-2237, 10.1016/j.xphs.2019.02.007, 2-s2.0-85063045253 Torkashvand F., Vaziri B. Main quality attributes of monoclonal antibodies and effect of cell culture components. Iranian Biomedical Journal. 2017; 21(3): 131-141, 10.18869/acadpub.ibj.21.3.131, 2-s2.0-85016239464 Wagh A., Song H., Zeng M., Tao L., Das T. K. Challenges and new frontiers in analytical characterization of antibody-drug conjugates. mAbs. 2018; 10(2): 222-243, 10.1080/19420862.2017.1412025, 2-s2.0-85042491611 Chiu M. L., Goulet D. R., Teplyakov A., Gilliland G. L. Antibody structure and function: the basis for engineering therapeutics. Antibodies. 2019 3; 8(4): 55, 10.3390/antib8040055 Anami Y., Otani Y., Xiong W., Ha S. Y. Y., Yamaguchi A., Rivera-Caraballo K. A., Zhang N., An Z., Kaur B., Tsuchikama K. Homogeneity of antibody-drug conjugates critically impacts the therapeutic efficacy in brain tumors. Cell Reports. 2022; 39(8), 10.1016/j.celrep.2022.110839, 110839 Wakankar A., Chen Y., Gokarn Y., Jacobson F. S. Analytical methods for physicochemical characterization of antibody drug conjugates. mAbs. 2011; 3(2): 161-172, 10.4161/mabs.3.2.14960, 2-s2.0-79952718320 Yüce M., Sert F., Torabfam M., Parlar A., Gürel B., Çakır N., Dağlıkoca D. E., Khan M. A., Çapan Y. Fractionated charge variants of biosimilars: a review of separation methods, structural and functional analysis. Analytica Chimica Acta. 2021; 1152, 10.1016/j.aca.2020.12.064, 238189 Hintersteiner B., Lingg N., Zhang P., Woen S., Hoi K. M., Stranner S., Wiederkum S., Mutschlechner O., Schuster M., Loibner H., Jungbauer A. Charge heterogeneity: basic antibody charge variants with increased binding to Fc receptors. mAbs. 2016; 8(8): 1548-1560, 10.1080/19420862.2016.1225642, 2-s2.0-84991030693 Sule S. V., Fernandez J. E., Mecozzi V. J., Kravets Y., Yang W. C., Feng P., Liu S., Zang L., Capili A. D., Estey T. B., Gupta K. Assessing the impact of charge variants on stability and viscosity of a high concentration antibody formulation. Journal of Pharmaceutical Sciences. 2017; 106(12): 3507-3514, 10.1016/j.xphs.2017.08.016, 2-s2.0-85030750159 Zhu X., Huo S., Xue C., An B., Qu J. Current LC-MS-based strategies for characterization and quantification of antibody-drug conjugates. Journal of Pharmaceutical Analysis. 2020; 10(3): 209-220, 10.1016/j.jpha.2020.05.008 Yan Y., Liu A. P., Wang S., Daly T. J., Li N. Ultrasensitive characterization of charge heterogeneity of therapeutic monoclonal antibodies using strong cation exchange chromatography coupled to native mass spectrometry. Analytical Chemistry. 2018; 90(21): 13013-13020, 10.1021/acs.analchem.8b03773, 2-s2.0-85055702244 Füssl F., Trappe A., Carillo S., Jakes C., Bones J. Comparative elucidation of cetuximab heterogeneity on the intact protein level by cation exchange chromatography and capillary electrophoresis coupled to mass spectrometry. Analytical Chemistry. 2020; 92(7): 5431-5438, 10.1021/acs.analchem.0c00185 Zhang Z., Zhou S., Han L., Zhang Q., Pritts W. A. Impact of linker-drug on ion exchange chromatography separation of antibody-drug conjugates. mAbs. 2019; 11(6): 1113-1121, 10.1080/19420862.2019.1628589, 2-s2.0-85068110645 Lechner A., Giorgetti J., Gahoual R., Beck A., Leize-Wagner E., François Y. N. Insights from capillary electrophoresis approaches for characterization of monoclonal antibodies and antibody drug conjugates in the period 2016-2018. Journal of Chromatography B. 2019; 1122-1123, 1-17, 10.1016/j.jchromb.2019.05.014, 2-s2.0-85065862746 Kaur H., Beckman J., Zhang Y., Li Z. J., Szigeti M., Guttman A. Capillary electrophoresis and the biopharmaceutical industry: therapeutic protein analysis and characterization. TrAC, Trends in Analytical Chemistry. 2021; 144, 10.1016/j.trac.2021.116407, 16407 Chen T., Chen Y., Stella C., Medley C. D., Gruenhagen J. A., Zhang K. Antibody-drug conjugate characterization by chromatographic and electrophoretic techniques. Journal of Chromatography B. 2016; 1032, 39-50, 10.1016/j.jchromb.2016.07.023, 2-s2.0-84979537030 Mack S., Cruzado-Park I., Chapman J., Ratnayake C., Vigh G. A systematic study in CIEF: defining and optimizing experimental parameters critical to method reproducibility and robustness. Electrophoresis. 2009; 30(23): 4049-4058, 10.1002/elps.200800690, 2-s2.0-72849122502 Salas-Solano O., Kennel B., Park S. S., Roby K., Sosic Z., Boumajny B., Free S., Reed-Bogan A., Michels D., McElroy W., Bonasia P., Hong M., He X., Ruesch M., Moffatt F., Kiessig S., Nunnally B. Robustness of iCIEF methodology for the analysis of monoclonal antibodies: an interlaboratory study. Journal of Separation Science. 2012; 35(22): 3124-3129, 10.1002/jssc.201200633, 2-s2.0-84870001724 Suba D., Urbányi Z., Salgó A. Capillary isoelectric focusing method development and validation for investigation of recombinant therapeutic monoclonal antibody. Journal of Pharmaceutical and Biomedical Analysis. 2015; 114, 53-61, 10.1016/j.jpba.2015.04.037, 2-s2.0-84930216206 Cruzado-Park I. D. Optimization of an IgG1 CIEF separation by using narrow-range ampholytes and DMSO as protein solubilizer. Electrophoresis. 2020; 41(15): 1308-1315 Lin J., Tan Q., Wang S. A high-resolution capillary isoelectric focusing method for the determination of therapeutic recombinant monoclonal antibody. Journal of Separation Science. 2011; 34(14): 1696-1702, 10.1002/jssc.201100067, 2-s2.0-79960363638 Lin J., Lazar A. C. Determination of charge heterogeneity and level of unconjugated antibody by imaged cIEF. Methods in Molecular Biology. 2013; 1045, 295-302, 10.1007/978-1-62703-541-5_19, 2-s2.0-84934436480 Kahle J., Wätzig H. Determination of protein charge variants with (imaged) capillary isoelectric focusing and capillary zone electrophoresis. Electrophoresis. 2018; 39(20): 2492-2511, 10.1002/elps.201800079, 2-s2.0-85054745530 Felten C., Salas-Solano O., Michels D. Imaged capillary isoelectric focusing for charge-variant analysis of biopharmaceuticals. BioProcess Int. 2011; 9 Wu J., McElroy W., Pawliszyn J., Heger C. D. Imaged capillary isoelectric focusing: applications in the pharmaceutical industry and recent innovations of the technology. TrAC, Trends in Analytical Chemistry. 2022; 150 Zhang X., Chemmalil L., Ding J., Mussa N., Li Z. Imaged capillary isoelectric focusing in native condition: a novel and successful example. Analytical Biochemistry. 2017; 537, 13-19, 10.1016/j.ab.2017.08.014, 2-s2.0-85028516567 Wu J., Huang T. Peak identification in capillary isoelectric focusing using the concept of relative peak position as determined by two isoelectric point markers. Electrophoresis. 2006; 27(18): 3584-3590, 10.1002/elps.200500889, 2-s2.0-33749480329 Turner A., Schiel J. E. Qualification of NISTmAb charge heterogeneity control assays. Analytical and Bioanalytical Chemistry. 2018; 410(8): 2079-2093, 10.1007/s00216-017-0816-6, 2-s2.0-85041533237 Demirdirek B., Lan W., Qiu D., Ding W., Iyer L. K., Bolgar M. S., Valente J. J. Comparison of imaged capillary isoelectric focusing and cation exchange chromatography for monitoring dextrose-mediated glycation of monoclonal antibodies in infusion solutions. Journal of Chromatography B. 2019; 1105, 156-163, 10.1016/j.jchromb.2018.12.021, 2-s2.0-85059095591 Tsuchikama K., An Z. Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein and cell. 2018; 9(1): 33-46, 10.1007/s13238-016-0323-0, 2-s2.0-84991081605 Harris R. J., Kabakoff B., Macchi F. D., Shen F. J., Kwong M., Andya J. D., Shire S. J., Bjork N., Totpal K., Chen A. B. Identification of multiple sources of charge heterogeneity in a recombinant antibody. Journal of Chromatography B: Biomedical Sciences and Applications. 2001; 752(2): 233-245, 10.1016/s0378-4347(00)00548-x, 2-s2.0-0035836065 Boylan N. J., Zhou W., Proos R. J., Tolbert T. J., Wolfe J. L., Laurence J. S. Conjugation site heterogeneity causes variable electrostatic properties in Fc conjugates. Bioconjugate Chemistry. 2013; 24(6): 1008-1016, 10.1021/bc4000564, 2-s2.0-84879376486 Xu T., Sun L. A mini review on capillary isoelectric focusing-mass spectrometry for top-down proteomics. Frontiers in Chemistry. 2021; 9, 651757

By Ayat Abbood

Reported by Author

Titel:
Optimization of the Imaged cIEF Method for Monitoring the Charge Heterogeneity of Antibody-Maytansine Conjugate
Autor/in / Beteiligte Person: Abbood, Ayat
Link:
Zeitschrift: Journal of Analytical Methods in Chemistry, 2023
Veröffentlichung: Hindawi, 2023
Medientyp: unknown
ISSN: 2090-8865 (print)
DOI: 10.1155/2023/8150143
Sonstiges:
  • Nachgewiesen in: OpenAIRE
  • Sprachen: English
  • File Description: text/xhtml
  • Language: English
  • Rights: OPEN

Klicken Sie ein Format an und speichern Sie dann die Daten oder geben Sie eine Empfänger-Adresse ein und lassen Sie sich per Email zusenden.

oder
oder

Wählen Sie das für Sie passende Zitationsformat und kopieren Sie es dann in die Zwischenablage, lassen es sich per Mail zusenden oder speichern es als PDF-Datei.

oder
oder

Bitte prüfen Sie, ob die Zitation formal korrekt ist, bevor Sie sie in einer Arbeit verwenden. Benutzen Sie gegebenenfalls den "Exportieren"-Dialog, wenn Sie ein Literaturverwaltungsprogramm verwenden und die Zitat-Angaben selbst formatieren wollen.

xs 0 - 576
sm 576 - 768
md 768 - 992
lg 992 - 1200
xl 1200 - 1366
xxl 1366 -