Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Publication year range
1.
Expert Rev Proteomics ; 21(5-6): 259-270, 2024.
Article in English | MEDLINE | ID: mdl-38934922

ABSTRACT

INTRODUCTION: The pharmaceutical industry continues to expand its search for innovative biotherapeutics. The comprehensive characterization of such therapeutics requires many analytical techniques to fully evaluate critical quality attributes, making analysis a bottleneck in discovery and development timelines. While thorough characterization is crucial for ensuring the safety and efficacy of biotherapeutics, there is a need to further streamline analytical characterization and expedite the overall timeline from discovery to market. AREAS COVERED: This review focuses on recent developments in liquid-phase separations coupled with ion mobility-mass spectrometry (IM-MS) for the development and characterization of biotherapeutics. We cover uses of IM-MS to improve the characterization of monoclonal antibodies, antibody-drug conjugates, host cell proteins, glycans, and nucleic acids. This discussion is based on an extensive literature search using Web of Science, Google Scholar, and SciFinder. EXPERT OPINION: IM-MS has the potential to enhance the depth and efficiency of biotherapeutic characterization by providing additional insights into conformational changes, post-translational modifications, and impurity profiles. The rapid timescale of IM-MS positions it well to enhance the information content of existing assays through its facile integration with standard liquid-phase separation techniques that are commonly used for biopharmaceutical analysis.


Subject(s)
Ion Mobility Spectrometry , Mass Spectrometry , Humans , Ion Mobility Spectrometry/methods , Mass Spectrometry/methods , Antibodies, Monoclonal/chemistry , Biological Products/chemistry , Protein Processing, Post-Translational , Immunoconjugates/chemistry , Immunoconjugates/analysis , Polysaccharides/chemistry , Polysaccharides/analysis , Phase Separation
2.
Rapid Commun Mass Spectrom ; 38(15): e9774, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38812280

ABSTRACT

RATIONALE: A common strategy for antibody-drug conjugate (ADC) quantitation from in vivo study samples involves measurement of total antibody, conjugated ADC, and free payload concentrations using multiple reaction monitoring (MRM) mass spectrometry. This not only provides a limited picture of biotransformation but can also involve lengthy method development. Quantitation of ADCs directly at the intact protein level in native conditions using high-resolution mass spectrometers presents the advantage of measuring exposure readout as well as monitoring the change in average drug-to-antibody ratio (DAR) and in vivo stability of new linker payloads with minimal method development. Furthermore, site-specific cysteine-conjugated ADCs often rely on non-covalent association to retain their quaternary structure, which highlights the unique capabilities of native mass spectrometry (nMS) for intact ADC quantitation. METHODS: We developed an intact quantitation workflow involving three stages: automated affinity purification, nMS analysis, and data processing in batch fashion. The sample preparation method was modified to include only volatile ion-pairing reagents in the buffer systems. A capillary size-exclusion chromatography (SEC) column was coupled to a quadrupole time-of-flight high-resolution mass spectrometer for high-throughput nMS analysis. Samples from two mouse pharmacokinetic (PK) studies were analyzed using both intact quantitation workflow and the conventional MRM-based approach. RESULTS: A linear dynamic range of 5-100 µg/mL was achieved using 20 µL of serum sample volume. The results of mouse in vivo PK measurement using the intact quantitation workflow and the MRM-based approach were compared, revealing excellent method agreement. CONCLUSIONS: We demonstrated the feasibility of utilizing nMS for the quantitation of ADCs at the intact protein level in preclinical PK studies. Our results indicate that this intact quantitation workflow can serve as an alternative generic method for high-throughput analysis, enabling an in-depth understanding of ADC stability and safety in vivo.


Subject(s)
Cysteine , Immunoconjugates , Mass Spectrometry , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/blood , Immunoconjugates/analysis , Cysteine/chemistry , Cysteine/blood , Animals , Mice , Mass Spectrometry/methods , Chromatography, Gel/methods
3.
Anal Chem ; 93(40): 13505-13512, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34585915

ABSTRACT

Monoclonal antibodies (mAbs) and related products undergo a wide range of modifications, many of which can often be directly associated to culture conditions during upstream processing. Ideally, such conditions should be monitored and fine-tuned based on real-time or close to real-time information obtained by the assessment of the product quality attribute (PQA) profile of the biopharmaceutical produced, which is the fundamental idea of process analytical technology. Therefore, methods that are simple, quick and robust, but sufficiently powerful, to allow for the generation of a comprehensive picture of the PQA profile of the protein of interest are required. A major obstacle for the analysis of proteins directly from cultures is the presence of impurities such as cell debris, host cell DNA, proteins and small-molecule compounds, which usually requires a series of capture and polishing steps using affinity and ion-exchange chromatography before characterization can be attempted. In the current study, we demonstrate direct coupling of protein A affinity chromatography with native mass spectrometry (ProA-MS) for development of a robust method that can be used to generate information on the PQA profile of mAbs and related products in as little as 5 min. The developed method was applied to several samples ranging in complexity and stability, such as simple and more complex monoclonal antibodies, as well as cysteine-conjugated antibody-drug conjugate mimics. Moreover, the method demonstrated suitability for the analysis of protein amounts of <1 µg, which suggests applicability during early-stage development activities.


Subject(s)
Immunoconjugates , Staphylococcal Protein A , Antibodies, Monoclonal , Chromatography, Ion Exchange , Immunoconjugates/analysis , Mass Spectrometry
4.
Anal Chem ; 93(15): 6135-6144, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33835773

ABSTRACT

Antibody-drug conjugates (ADCs) pose challenges to bioanalysis because of their inherently intricate structures and potential for very complex catabolism. Common bioanalysis strategy is to measure the concentration of ADCs and Total Antibody (Ab) as well as deconjugated warhead in circulation. The ADCs and the Total Ab can be quantified with ligand binding assays (LBA) or with hybrid immunocapture-liquid chromatography coupled with multiple reaction monitoring mass spectrometry (LBA-LC-MRM). With the LBA-LC-MRM approach, a surrogate analyte, often the signature peptide, and released warhead can be used for the quantification of the Total Ab and ADCs, respectively. Recent advances in analytical instrumentation, especially the development of high resolution mass spectrometers (HRMS), have enabled characterization and quantification of intact macromolecules such as ADCs. The LBA-LC-HRMS approach employs immunocapture, followed by chromatographic separation at the macromolecule level and detection of the intact analyte. We developed an intact quantification method with 1-10 µg/mL linear dynamic range using 25 µL of plasma sample volume. This method was qualified for the measurement of naked monoclonal antibody (mAb), a site-specific cysteine-conjugated ADC with drug to antibody ratio ∼2 (DAR2) and a site-nonspecific cysteine-conjugated ADC (DAR8) in rat plasma. Samples from a rat pharmacokinetic (PK) study were analyzed with both methods. For the naked mAb, the results from both assays matched well. For ADCs, new species were observed from the LBA-HRMS method. The results demonstrated that potential biotransformation of the ADC was unveiled using the intact quantification approach while not being observed with traditional LBA-LC-MRM approach. Our work demonstrated an application of novel intact quantification by supporting animal PK studies. Moreover, our results suggest that the intact quantification method can provide novel perspectives on ADC in vivo characterization and quantification, which can benefit future drug candidate optimization as well as the immunogenicity impact evaluation and safety assessment.


Subject(s)
Immunoconjugates , Animals , Antibodies, Monoclonal , Biotransformation , Chromatography, Liquid , Immunoconjugates/analysis , Mass Spectrometry , Rats
5.
Anal Chem ; 93(38): 12930-12937, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34519496

ABSTRACT

The therapeutic efficacy and pharmacokinetics of antibody-drug conjugates (ADCs) in general, and antibody-oligonucleotide conjugates (AOCs) in particular, depend on the drug-to-antibody ratio (DAR) distribution and average value. The DAR is considered a critical quality attribute, and information pertaining to it needs to be gathered during ADC/AOC development, production, and storage. However, because of the high structural complexity of ADC/AOC samples, particularly in the initial drug-development stages, the application of the current state-of-the-art mass spectrometric approaches can be limited for DAR analysis. Here, we demonstrate a novel approach for the analysis of complex ADC/AOC samples, following native size-exclusion chromatography Orbitrap Fourier transform mass spectrometry (FTMS). The approach is based on the integration of the proteoform-level mass spectral peaks in order to provide an estimate of the DAR distribution and its average value with less than 10% error. The peak integration is performed via a truncation of the Orbitrap's unreduced time-domain ion signals (transients) before mass spectra generation via FT processing. Transient recording and processing are undertaken using an external data acquisition system, FTMS Booster X2, coupled to a Q Exactive HF Orbitrap FTMS instrument. This approach has been applied to the analysis of whole and subunit-level trastuzumab conjugates with oligonucleotides. The obtained results indicate that ADC/AOC sample purification or simplification procedures, for example, deglycosylation, could be omitted or minimized prior to the DAR analysis, streamlining the drug-development process.


Subject(s)
Immunoconjugates , Pharmaceutical Preparations , Fourier Analysis , Immunoconjugates/analysis , Mass Spectrometry , Oligonucleotides
6.
Analyst ; 146(3): 747-769, 2021 Feb 07.
Article in English | MEDLINE | ID: mdl-33410843

ABSTRACT

Accelerated development of new therapeutics in an increasingly competitive landscape requires the use of high throughput analytical platforms. In addition, the complexity of novel biotherapeutic formats (e.g. fusion proteins, protein-polymer conjugates, co-formulations, etc.) reinforces the need to improve the selectivity and resolution of conventional one-dimensional (1D) liquid chromatography (LC). Liquid chromatography-mass spectrometry (LC-MS)-based technologies such as native LC-MS for intact mass analysis or peptide mapping (also called bottom-up approach)-based multi-attribute methods (MAM) have already demonstrated their potential to complement the conventional analytical toolbox for monoclonal antibody (mAb) characterization. Two-dimensional liquid-chromatography (2D-LC-MS) methods have emerged in the last ten years as promising approaches to address the increasing analytical challenges faced with novel antibody formats. However, off-line sample preparation procedures are still required for conventional 1D and 2D-LC-MS methods for the in-depth variant characterization at the peptide level. Multi-dimensional LC-MS (mD-LC-MS) combine sample preparation and multi-level (i.e. intact, reduced, middle-up and peptide) analysis within the same chromatographic set-up. This review presents an overview of the benefits and limitations of mD-LC-MS approaches in comparison to conventional chromatographic methods (i.e. 1D-LC-UV methods at intact protein level and 1D-LC-MS methods at peptide level). The current analytical trends in antibody characterization by mD-LC-MS approaches, beyond the 2D-LC-MS workhorse, are also reviewed, and our vision on a more integrated multi-level mD-LC-MS characterization platform is shared.


Subject(s)
Immunoconjugates , Tandem Mass Spectrometry , Antibodies, Monoclonal , Chromatography, Liquid , Immunoconjugates/analysis , Peptides
7.
Chem Pharm Bull (Tokyo) ; 69(10): 976-983, 2021.
Article in English | MEDLINE | ID: mdl-34602579

ABSTRACT

Antibody-drug conjugates (ADCs) are biopharmaceuticals produced by chemically linking small molecules (payloads) to antibodies that possess specific affinity for the target cell. The ADCs currently on the commercially market are the result of a stochastic conjugation of highly-potent payloads to multiple sites on the monoclonal antibody, resulting in a heterogeneous drug-antibody ratio (DAR) and drug distribution. The heterogeneity inherent to ADCs not produced site-specifically may not only be detrimental to the quality of the drug but also is less-desirable from the perspective of regulatory science. An ideal method or unified approach used to measure the DAR for ADCs, a critical aspect of their analysis and characterization, has not yet been established in the ADC field and remains an often-challenging issue for bioanalytical chemists. In this review we describe, compare, and evaluate the characteristics of various DAR determination methods for ADCs featuring recently reported technologies. The future landscape of bioconjugate DAR analysis is also discussed.


Subject(s)
Immunoconjugates/analysis , Humans , Molecular Structure
8.
Anal Chem ; 92(10): 6839-6843, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32309925

ABSTRACT

There are many pharmacokinetic challenges associated with administering protein therapeutics, including biotransformation via clipping, deamidation, isomerization, oxidation, etc. In the case of engineered multivalent tethered antibody formats, proteolysis or deconjugation at the fusion or conjugation site present further issues. Unlike degradations associated with antibody drug conjugates, such biotransformations of tethered antibody formats usually result in degraded products with large mass differences. These large differences can result in processing or mass spectrometry response bias among the resulting product species that can lead to inaccurate stability quantitation. Herein, we describe an assay strategy for characterizing and quantitating degradations accurately for multivalent antibodies by incorporating response bias corrections. For the multivalent tethered antibody molecules selected, an ∼30-80% difference in response, compared to the cleaved product, was observed. To correct for the response bias, selected tethered multivalent antibodies and an IgG antibody (representing the stable intact and the degraded product species, respectively) were spiked in serum at known ratios for analysis. Following affinity capture, we generated calibration curves (five-parameter logistic fit p < 0.05) by plotting the measured ratios of the MS ion responses against the known spiked-in ratios (CVs < 8% for calibration standards). The qualified calibration curve (accuracy within 8% and 2% for measuring degradations of 5% and 15% product, respectively) was then used, through interpolation, to determine stability profiles for the same multivalent tethered antibody formats from both in vitro serum and pharmacokinetic study samples.


Subject(s)
Antibodies/analysis , Immunoconjugates/analysis , Chromatography, Liquid , Mass Spectrometry
9.
Anal Chem ; 92(12): 8170-8177, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32407621

ABSTRACT

Conventional antibody-drug conjugate (ADC) manufacturing methods are based on the nonselective bioconjugation of cytotoxic drugs to lysine and cysteine residues. This results in highly heterogeneous mixtures of different drug-antibody ratios (DAR) that can significantly affect the safety and efficacy of the ADC product. Recently, an innovative procedure named GlyCLICK was suggested, consisting of a two-step enzymatic procedure to transform Fc-glycans present on IgG mAbs into two site-specific anchor points for the conjugation of any alkyne-containing payload of choice. Here, we evaluated the conjugation process by comparing trastuzumab and trastuzumab conjugated with DM1, following the GlyCLICK procedure. Complementary reversed phase liquid chromatography (RPLC) and hydrophilic interaction chromatography (HILIC) coupled to high-resolution mass spectrometry (HRMS) were used to analyze the protein subunits (ca. 25-100 kDa) obtained after different levels of enzymatic digestion and chemical reduction. Our results demonstrated that the hydrophobic character of the drug molecule allows to rapidly confirm the Fc-drug conjugation at the chromatographic level. Furthermore, the hyphenation to MS detection provided accurate mass information on the ADC subunits and facilitated the DAR determination of 2.0. Therefore, this work illustrates how middle-up analysis using LC/HRMS can provide accurate and complementary information on the critical quality attributes of these novel site-specific ADC products.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoconjugates/analysis , Polysaccharides/chemistry , Chromatography, Liquid , Mass Spectrometry , Molecular Conformation
10.
Anal Chem ; 92(12): 8268-8277, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32392410

ABSTRACT

Complex biotherapeutics present challenges from drug discovery, screening, and development perspectives. While monoclonal antibody drugs are not monitored for metabolites in the same manner as small molecules, biotherapeutics such as fusion proteins, antibody-drug conjugates, or bispecific antibodies may undergo biotransformation (such as clipping, deamidation, or oxidation) in vivo, resulting in catabolites that can have a direct impact on drug safety or efficacy. Here antibody subunit LC-MS is utilized for evaluation of two classes of complex biotherapeutics: an antibody-drug conjugate and a mAb-fusion biotherapeutic. Pharmacokinetic concentration, biotransformation, and DAR data are collectively presented using the subunit LC-MS approach for the two molecules, and the methods shared in detail can be applied to any humanized IgG1 mAb biotherapeutic for preclinical study support. Overall, the data generated from antibody LC-MS analyses can provide key information in early phase development and deliver multiple study end points with a single data set.


Subject(s)
Antibodies, Monoclonal/analysis , Immunoconjugates/analysis , Animals , Antibodies, Monoclonal/pharmacokinetics , Biotransformation , Chromatography, Liquid , Immunoconjugates/pharmacokinetics , Macaca mulatta , Mass Spectrometry , Rats
11.
Anal Chem ; 92(22): 15096-15103, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33108180

ABSTRACT

Antibody drug conjugates (ADCs), which harness the high targeting specificity of monoclonal antibodies (mAb) with the potency of small molecule therapeutics, are one of the fastest growing pharmaceutical classes. Nevertheless, ADC conjugation techniques and processes may introduce intrinsic heterogeneity including primary sequence variants, varied drug-to-antibody ratio (DAR) species, and drug positional isomers, which must be monitored to ensure the safety and efficacy of ADCs. Liquid chromatography coupled to mass spectrometry (LC-MS) is a powerful tool for characterization of ADCs. However, the conventional bottom-up MS analysis workflows require an enzymatic digestion step which can be time consuming and may introduce artifactual modifications. Herein, we develop an online LC-MS/MS method for rapid analysis of reduced ADCs without digestion, enabling determination of DAR, characterization of the primary sequence, and localization of the drug conjugation site of the ADC using high-resolution Fourier transform ion cyclotron resonance (FTICR) MS. Specifically, a model cysteine-linked ADC was reduced to generate six unique subunits: light chain (Lc) without drug (Lc0), Lc with 1 drug (Lc1), heavy chain (Hc) without drug (Hc0), and Hc with 1-3 drugs (Hc1-3, respectively). A concurrent reduction strategy is applied to assess ADC subunits in both the partially reduced (intrachain disulfide bonds remain intact) and fully reduced (all disulfide bonds are cleaved) forms. The entire procedure including the sample preparation and LC-MS/MS takes less than 55 min, enabling rapid multiattribute analysis of ADCs.


Subject(s)
Chromatography, Liquid/methods , Cyclotrons , Fourier Analysis , Immunoconjugates/analysis , Tandem Mass Spectrometry/instrumentation , Immunoconjugates/chemistry , Isomerism , Time Factors
12.
Anal Chem ; 92(18): 12168-12175, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786429

ABSTRACT

Immunoaffinity (IA) LC-MS/MS pharmacokinetic (PK) assays are widely used in the field for antibody drug conjugates (ADCs) containing peptide linkers that are enzymatically cleavable, such as MC-ValCit-PAB. Conjugate PK assay strategies for these ADCs involve cleavage with cathepsin B or papain to release and measure the antibody-conjugated drug (acDrug) concentration. However, robust acDrug PK methods for disulfide-linked self-immolating ADCs are lacking as they are a different conjugation modality. We developed acDrug PK assays for next-generation disulfide-linked ADCs involving immunoaffinity capture, chemical cleavage, and LC-MS/MS. Disulfide-linked ADCs captured from plasma were chemically reduced at basic pH to release the linker-drug, followed by self-immolation to liberate the active drug, and quantified by MRM LC-MS/MS. Herein, we detail the development and optimization of this chemical cleavage acDrug PK assay, resulting in robust accuracy and precision (±20%). The conjugation site of the linker-drug on the antibody was found to affect the kinetics of drug release. Multiple biophysical and chemical characteristics, such as tertiary structure, fractional solvent accessibility, pKa of the conjugation site, surrounding residue's pI, and electrostatic charge, may directly impact the drug release kinetics. Similar site-specific stability has been previously reported for ADCs in vivo. The assay development and qualification data for this original assay format are presented along with its application to multiple in vitro and in vivo studies across species.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Disulfides/pharmacokinetics , Immunoconjugates/pharmacokinetics , Antibodies, Monoclonal/analysis , Chromatography, Liquid , Disulfides/analysis , Humans , Immunoassay , Immunoconjugates/analysis , Molecular Structure , Tandem Mass Spectrometry
13.
Anal Chem ; 92(13): 9001-9007, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32441508

ABSTRACT

Characterization of antibody-drug conjugates (ADCs) using mass spectrometry (MS) is important in drug discovery and formulation development and as part of the quality control processes. To facilitate electrospray ionization (ESI) and produce high-quality mass spectra, common components of storage solutions for monoclonal antibodies (mAbs) and ADCs, such as nonvolatile phosphate-buffered saline (PBS), should be replaced before analysis. Centrifugal spin-type kits are extensively used for the desalting or buffer-exchange of mAbs and ADCs samples. The commercially available kits commonly require at least 100 µL of a sample at 1 mg/mL for optimal recovery. However, most ESI-MS based analyses require no more than 25 µg of protein for triplicate injection. In this study, we present a novel method for desalting of ADCs and mAbs building on the SP3 approach with nonfunctionalized carboxylate coated magnetic beads without affinity ligands. The analytes bind to the hydrophilic beads upon the addition of organic solvent, and various solutions of volatile salts or acids can be used in the elution step. The optimized protocol allowed for 88% recovery of ADC at a 25 µL sample volume and 90% recovery at 100 µL. More than 90% of the salts were removed using a process of 20 min. The intra- and interday precision showed little variation with an RSD of 1% and 2%, respectively. The compatibility of this new workflow with low sample volumes is highly valuable since a smaller fraction of the sample is wasted for analysis of the expensive analytes, without compromising recovery.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoconjugates/analysis , Magnetics , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Hydrogen-Ion Concentration , Immunoconjugates/chemistry , Mass Spectrometry , Oligopeptides/chemistry , Reproducibility of Results , Solvents/chemistry , Trastuzumab/chemistry
14.
Anal Chem ; 92(2): 2186-2193, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31880920

ABSTRACT

With the rapid rise of therapeutic antibodies and antibody-drug conjugates, significant investments have been made in developing workflows that utilize mass spectrometry to detect these intact molecules, the large fragments generated by their selective digestion, and the peptides generated by traditional proteomics workflows. The resultant data is used to gain insight into a wide range of parameters, including primary sequence, disulfide bonding, glycosylation patterns, biotransformation, and more. However, many of the technologies utilized to couple these workflows to mass spectrometers have significant limitations that force nonoptimal modifications to upstream sample preparation steps, limit the throughput of high-volume workflows, and prevent the harmonization of diverse experiments onto a single hardware platform. Here, we describe a new analytical platform that enables direct and high-throughput coupling to electrospray ionization mass spectrometry. The SampleStream platform is compatible with both native and denaturing electrospray, operates with a throughput of up to 15 s/sample, provides extensive concentration of dilute samples, and affords similar sensitivity to comparable liquid chromatographic methods.


Subject(s)
Antibodies, Monoclonal/analysis , High-Throughput Screening Assays , Immunoconjugates/analysis , High-Throughput Screening Assays/instrumentation , Software , Spectrometry, Mass, Electrospray Ionization/instrumentation
15.
Drug Metab Dispos ; 48(5): 368-377, 2020 05.
Article in English | MEDLINE | ID: mdl-32086295

ABSTRACT

Antibody-drug conjugates (ADCs) employ overexpressed cell surface antigens to deliver cytotoxic payloads inside cancer cells. However, the relationship between target expression and ADC efficacy remains ambiguous. In this manuscript, we have addressed a part of this ambiguity by quantitatively investigating the effect of antigen expression levels on ADC exposure within cancer cells. Trastuzumab-valine-citrulline-monomethyl auristatin E was used as a model ADC, and four different cell lines with diverse levels of human epidermal growth factor receptor 2 (HER2) expression were used as model cells. The pharmacokinetics (PK) of total trastuzumab, released monomethyl auristatin E (MMAE), and total MMAE were measured inside the cells and in the cell culture media following incubation with two different concentrations of ADC. In addition, target expression levels, target internalization rate, and cathepsin B and MDR1 protein concentrations were determined for each cell line. All the PK data were mathematically characterized using a cell-level systems PK model for ADC. It was found that SKBR-3, MDA-MB-453, MCF-7, and MDA-MB-468 cells had ∼800,000, ∼250,000, ∼50,000, and ∼10,000 HER2 receptors per cell, respectively. A strong linear relationship (R 2 > 0.9) was observed between HER2 receptor count and released MMAE exposure inside the cancer cells. There was an inverse relationship found between HER2 expression level and internalization rate, and cathepsin B and multidrug resistance protein 1 (MDR1) expression level varied slightly among the cell lines. The PK model was able to simultaneously capture all the PK profiles reasonably well while estimating only two parameters. Our results demonstrate a strong quantitative relationship between antigen expression level and intracellular exposure of ADCs in cancer cells. SIGNIFICANCE STATEMENT: In this manuscript, we have demonstrated a strong linear relationship between target expression level and antibody-drug conjugate (ADC) exposure inside cancer cells. We have also shown that this relationship can be accurately captured using the cell-level systems pharmacokinetics model developed for ADCs. Our results indirectly suggest that the lack of relationship between target expression and efficacy of ADC may stem from differences in the pharmacodynamic properties of cancer cells.


Subject(s)
Antineoplastic Agents, Immunological/pharmacokinetics , Immunoconjugates/pharmacokinetics , Neoplasms/drug therapy , Oligopeptides/pharmacokinetics , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/analysis , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents, Immunological/analysis , Antineoplastic Agents, Immunological/therapeutic use , Cathepsin B/analysis , Cathepsin B/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Immunoconjugates/analysis , Immunoconjugates/therapeutic use , Models, Biological , Neoplasms/immunology , Neoplasms/pathology , Oligopeptides/analysis , Oligopeptides/therapeutic use , Receptor, ErbB-2/analysis , Receptor, ErbB-2/antagonists & inhibitors , Trastuzumab/analysis , Trastuzumab/therapeutic use
16.
Bioorg Chem ; 94: 103391, 2020 01.
Article in English | MEDLINE | ID: mdl-31761409

ABSTRACT

Thermostability of monoclonal antibodies (mAbs) and antibody-drug conjugates (ADCs), as a critical property of biotherapeutics, is important for their physicochemical processes, pharmacodynamics, and pharmacokinetics. Fc glycosylation of mAbs plays a crucial role in antibody functions including thermostability, however, due to the lack of homogeneous glycosylation for comparison, the precise impact of glycoforms on thermostability of mAbs and ADCs remains challenging to elucidate. In this paper, we employed the technique of differential scanning fluorimetry (DSF) to investigate the thermostability of Fc domains, glycoengineered mAbs, and ADCs, carrying well-defined N-glycan structures for comparison. The results revealed that high-mannose-type N-glycans dramatically reduce the Tm value of Fc, compared to complex-type N-glycans. We also found that core-fucose contributes to the thermostability of mAbs, and the unnatural modification on non-reducing end of biantennary N-glycan can compensate the reduced stability of afucosylated mAbs and maintain the advantage of enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). DSF analysis of lysine-linked and glycosite-specific ADCs indicated that thermostability of glycan-linked ADCs is reduced, but it could be improved by using an optimized linkage. This work provides an in-depth analysis on thermostability of mAbs and ADCs with homogeneous glycoforms, and also proposes new strategies for optimizing glycoengineered mAbs and glycosite-specific ADCs using unnatural glycan and stabilized linkage.


Subject(s)
Antibodies, Monoclonal/analysis , Fluorometry , Immunoconjugates/analysis , Temperature , Antibodies, Monoclonal/immunology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glycosylation , Humans , Immunoconjugates/immunology , Molecular Structure , Structure-Activity Relationship
17.
Anal Chem ; 91(4): 2805-2812, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30661356

ABSTRACT

The synthesis of antibody-drug conjugates (ADCs) using the interchain cysteines of the antibody inherently gives a mixture of proteins with varying drug-to-antibody ratio. The drug distribution profiles of ADCs are routinely characterized by hydrophobic interaction chromatography (HIC). Because HIC is not in-line compatible with mass spectrometry (MS) due to the high salt levels, it is laborious to identify the constituents of HIC peaks. An MS-compatible alternative to HIC is reported here: native reversed phase liquid chromatography (nRPLC). This novel technique employs a mobile phase 50 mM ammonium acetate for high sensitivity in MS and elution with a gradient of water/isopropanol. The key to the enhancement is a bonded phase giving weaker drug-surface interactions compared to the noncovalent interactions holding the antibody-drug conjugates together. The hydrophobicity of the bonded phase is varied, and the least hydrophobic bonded phase in the series, poly(methyl methacrylate), is found to resolve the intact constituents of a model ADC (Ab095-PZ) and a commercial ADC (brentuximab vedotin) under the MS-compatible conditions. The nRPLC-MS data show that all species, ranging from drug-to-antibody ratios of 1 to 8, remained intact in the column. Another desired advantage of the nRPLC is the ability of resolving multiple positional isomers of ADC that are not well-resolved in other chromatographic modes. This supports the premise that lower hydrophobicity of the bonded phase is the key to enabling online nRPLC-MS analysis of antibody-drug conjugates.


Subject(s)
Antineoplastic Agents, Immunological/analysis , Brentuximab Vedotin/analysis , Chromatography, Reverse-Phase/methods , Immunoconjugates/analysis , Acetates/chemistry , Chromatography, High Pressure Liquid/methods , Hydrophobic and Hydrophilic Interactions
18.
Anal Chem ; 91(20): 12724-12732, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31478640

ABSTRACT

Antibody-drug conjugates (ADCs) have become a major class of oncology biopharmaceuticals. Traditional ADCs have a stochastic distribution of cytotoxic drugs attached at several different sites on the antibody. The heterogeneous nature of stochastic ADCs results in a complex compositional analysis. To improve on traditional ADC technology, we have developed a chemical conjugation platform termed "AJICAP" for the site-specific modification of native antibodies using a class of IgG Fc affinity reagents. Here we report further investigation focusing on several analyses of a first-generation AJICAP-ADC (Angew. Chem., Int. Ed. 2019, 58, 5592-5597). For drug-antibody ratio (DAR) determination, we examined and compared six different analytical methods. To the best of our knowledge, this is the first report of a comparison of analytical techniques to measure the DAR for ADCs produced by a site-specific technology such as AJICAP. Furthermore, a rapid analytical process for confirmation of the site selectivity of AJICAP conjugation was established by SEC-Q-TOF-MS. The analytical strategy reported here can be applied to the DAR determination of site-specific ADCs.


Subject(s)
Immunoconjugates/analysis , Mass Spectrometry/methods , Antineoplastic Agents/chemistry , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Immunoglobulin Fc Fragments/chemistry , Oligopeptides/chemistry , Trastuzumab/chemistry
19.
Anal Chem ; 91(18): 11661-11669, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31442030

ABSTRACT

Antibody-drug conjugates (ADCs) are designed to combine the target specificity of monoclonal antibodies and potent cytotoxin drugs to achieve better therapeutic outcomes. Comprehensive evaluation of the quality attributes of ADCs is critical for drug development but remains challenging due to heterogeneity of the construct. Currently, peptide mapping with reversed-phase liquid chromatography (RPLC) coupled to mass spectrometry (MS) is the predominant approach to characterize ADCs. However, it is suboptimal for sequence characterization and quantification of ADCs because it lacks a comprehensive view of coexisting variants and suffers from varying ionization effects of drug-conjugated peptides compared to unconjugated counterparts. Here, we present the first middle-down RPLC-MS analysis of both cysteine (Adcetris; BV) and lysine (Kadcyla; T-DM1) conjugated ADCs at the subunit level (∼25 kDa) with electron transfer dissociation (ETD). We successfully achieved high-resolution separation of subunit isomers arising from different drug conjugation and subsequently localized the conjugation sites. Moreover, we obtained a comprehensive overview of the microvariants associated with each subunits and characterized them such as oxidized variants with different sites. Furthermore, we observed relatively high levels of conjugation near complementarity-determining regions (CDRs) from the heavy chain but no drug conjugation near CDRs of light chain (Lc) from lysine conjugated T-DM1. Based on the extracted ion chromatograms, we accurately measured average drug to antibody ratio (DAR) values and relative occupancy of drug-conjugated subunits. Overall, the middle-down MS approach enables the evaluation of multiple quality attributes including DAR, positional isomers, conjugation sites, occupancy, and microvariants, which potentially opens up a new avenue to characterize ADCs.


Subject(s)
Ado-Trastuzumab Emtansine/chemistry , Brentuximab Vedotin/chemistry , Immunoconjugates/analysis , Immunoconjugates/chemistry , Ado-Trastuzumab Emtansine/analysis , Brentuximab Vedotin/analysis , Chromatography, Reverse-Phase , Cysteine/chemistry , Electron Transport , Lysine/chemistry , Tandem Mass Spectrometry/methods
20.
Anal Chem ; 91(23): 14896-14903, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31621311

ABSTRACT

This study describes the use of a multidimensional HPLC (2D and 4D) system for a faster and more effective characterization of an antibody-drug conjugate (ADC) product, compared to the standard off-line approach of fraction collection and off-line variant characterization. The size variants of an interchain cysteine-linked ADC were characterized to understand the effect of the different drug-to-antibody ratio (DAR) species on aggregate formation. For this purpose, the ADC product and a full panel of stressed samples were analyzed. The dimeric ADC species were baseline resolved from the main peak (Rs = 2.7) by UHP-SEC (ultra-high-performance size exclusion chromatography) under nondenaturing conditions using a buffered mobile phase containing 5% 2-propanol. A 2D-LC (SEC-HIC) method was then developed to compare the average DAR values of the main peak species vs the aggregates. A 4D-LC/MS method (SEC-reduction-digestion-RPHPLC) was also developed to determine levels of potential critical quality attributes (pCQAs) including aggregation, average DAR, oxidation, and deamidation, in a 2 h run. An average DAR value of 3.5-3.6 was found for the main peak using both 2D-LC and 4D-LC methods, and these values were consistent with DAR determined by the in-house reference hydrophobic interaction chromatography (HIC) method. The multidimensional LC approaches also showed an increase in the content of high-DAR species in the SEC fractions containing the aggregates. Overall the entire workflow of data acquisition is completed within a day using the multidimensional on-line approach, in comparison to multiple days required with the traditional off-line approaches.


Subject(s)
Antibodies/analysis , Chromatography, High Pressure Liquid/standards , Immunoconjugates/analysis , Peptide Fragments/analysis , Antibodies/chemistry , Cysteine/chemistry , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/chemistry , Light , Oxidation-Reduction , Peptide Mapping , Proteolysis , Solutions , Trypsin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL