3,962
Views
0
CrossRef citations to date
0
Altmetric
Report

Robust production of monovalent bispecific IgG antibodies through novel electrostatic steering mutations at the CH1-Cλ interface

, , , , , , , , , , , , , , , , , , , , , & show all
Article: 2273449 | Received 17 Aug 2023, Accepted 17 Oct 2023, Published online: 06 Nov 2023

ABSTRACT

Bispecific antibodies represent an increasingly large fraction of biologics in therapeutic development due to their expanded scope in functional capabilities. Asymmetric monovalent bispecific IgGs (bsIgGs) have the additional advantage of maintaining a native antibody-like structure, which can provide favorable pharmacology and pharmacokinetic profiles. The production of correctly assembled asymmetric monovalent bsIgGs, however, is a complex engineering endeavor due to the propensity for non-cognate heavy and light chains to mis-pair. Previously, we introduced the DuetMab platform as a general solution for the production of bsIgGs, which utilizes an engineered interchain disulfide bond in one of the CH1-CL domains to promote orthogonal chain pairing between heavy and light chains. While highly effective in promoting cognate heavy and light chain pairing, residual chain mispairing could be detected for specific combinations of Fv pairs. Here, we present enhancements to the DuetMab design that improve chain pairing and production through the introduction of novel electrostatic steering mutations at the CH1-CL interface with lambda light chains (CH1-Cλ). These mutations work together with previously established charge-pair mutations at the CH1-CL interface with kappa light chains (CH1-Cκ) and Fab disulfide engineering to promote cognate heavy and light chain pairing and enable the reliable production of bsIgGs. Importantly, these enhanced DuetMabs do not require engineering of the variable domains and are robust when applied to a panel of bsIgGs with diverse Fv sequences. We present a comprehensive biochemical, biophysical, and functional characterization of the resulting DuetMabs to demonstrate compatibility with industrial production benchmarks. Overall, this enhanced DuetMab platform substantially streamlines process development of these disruptive biotherapeutics.

Introduction

Over the past decade, bispecific antibodies have emerged as a leading class of biological therapeutics with extensive therapeutic potential. Unlike conventional monoclonal antibodies which bind a single target, bispecific antibodies are capable of engaging two distinct epitopes. This ostensibly simple change in binding capabilities has transformed the therapeutic landscape of antibodies due to the expanded functional capabilities that are accessible with bispecific antibodies. Simultaneous targeting of two independent disease markers offers improved therapeutic efficacy and greater resilience to escape mechanisms than mono-targeted therapies.Citation1,Citation2 Additionally, dual-antigen avidity binding can enhance target selectivity on double-positive target cells over single-positive normal tissue, leading to reduced toxicity and improved therapeutic index.Citation3 In addition to enhanced targeting capabilities, bispecific antibodies also provide access to an expanded set of mechanisms of action, including T-cell engagers, half-life extension, trans-membrane delivery, and targeted protein degradation.Citation4,Citation5 These disruptive modalities are rapidly progressing and show exciting promise in many therapeutic areas, including cancer, autoimmunity, and metabolic diseases.Citation6

Asymmetric monovalent bispecific IgG (bsIgG) therapeutics have the additional advantage of maintaining a native antibody-like structure, which provides favorable bioavailability and pharmacokinetic (PK) profiles and leverages the production expertise associated with conventional antibodies. The synthesis of bsIgGs, however, often requires the production of asymmetric molecules, which adds substantial complexity to the manufacturing process. A monovalent bispecific IgG, for example, is composed of two heavy chains (HCs) and two light chains (LCs), which can fold into a total of 10 different molecule combinations, only one of which contains the desired assembly of HCs and LCs.Citation7 Several techniques have been developed to drive heterodimerization of the heavy chains during production, including the knobs-into-holes approach,Citation8 electrostatic steering,Citation9 and disulfide engineering.Citation10 Alternative approaches include the use of Fab-arm exchange to produce the bispecific after production,Citation11 or bypassing the problem through the use of a common HC.Citation12 An additional challenge in asymmetric bsIgG production is the correct pairing of HCs and LCs. Although initially addressed through the use of a common LC,Citation13 several protein engineering solutions have been developed to promote orthogonal chain pairing, including domain cross-over between CH1 and CL,Citation14 engineering of the antigen-binding fragment (Fab) interface,Citation15,Citation16 disulfide bond engineering,Citation17,Citation18 and the introduction of charged residues in the HC-LC interface.Citation19,Citation20

We have previously introduced the DuetMab platform for the production of monovalent bispecific IgG antibodies.Citation17,Citation21,Citation22 This approach combined knobs-into-holes Fc mutations with the introduction of an engineered disulfide at the CH1-CL interface to promote correct chain associations. Compatibility with both kappa (κ) and lambda (λ) LCs was demonstrated, and several of the presented DuetMabs contained Fabs with a combination of both LCs.

A persistent challenge in the development and application of bsIgGs is the significant influence that VH and VL sequences have on HC-LC pairing, which can overwhelm engineering methods that promote specific chain associations.Citation15,Citation16,Citation23 There thus remains a need for improvements in strategies for bsIgG production that build toward robust platforms that accommodate diverse Fv sequences. Additionally, the development of bispecifics that maintain the structural integrity and manufacturability of the native IgG remains a rate-limiting step for advancement of these biotherapeutics. Here, we present enhancements to the DuetMab platform that improve bsIgG pairing and production through the combination of disulfide engineering and electrostatic steering mutations to drive correct HC-LC pairing. Although electrostatic steering was explored and reported in the literature previously, no solution has been provided for antibodies with a lambda LC. Here, we expand the bispecific antibody toolbox by disclosing for the first time electrostatic steering mutations that promote cognate chain pairing for lambda LCs. Additionally, we also present a comprehensive biochemical, biophysical, and functional characterization of the resulting DuetMabs to demonstrate compatibility with industrial production benchmarks.

Results

Design of CH1-Cλ interface variants for orthogonal Fab pairing

Previous engineering efforts by Dillon et al. to drive orthogonal HC-LC pairing identified the CH1 S183K and CL V133E mutations, which provide significant improvements to bispecific antibody production.Citation19 However, electrostatic steering mutations have not yet been identified that promote correct chain pairing in the context of a CH1-CL interface with a lambda constant domain (Cλ). At the CH1-Cλ interface, a tyrosine (Y178) is predicted to clash with the S183K-V133E interaction, whereas the corresponding threonine residue in kappa LCs is able to accommodate these mutations due to its smaller size (). We sought to identify charge pair mutations that would maintain compatibility with lambda LCs. In an effort to identify engineering solutions that could serve as a robust platform that functions independently of Fv sequence, we restricted our search to pairs of amino acids at the CH1-Cλ interface. To increase the likelihood that selected pairs have a direct interaction, only amino acids in which the Cβ atom were oriented toward the interface were selected (in the case of glycine residues, we required the carbonyl to be oriented away from interface). Additionally, we required that selected residues have sufficient space beyond the Cβ atom to introduce amino acids with longer side chains (i.e., arginine and glutamic acid). The last criteria for selection involved substituting amino acids at candidate positions and searching for possible steric clashes. Substituted residues included Arg, Lys, Asp, Glu, Ser, and Thr to introduce electrostatic interactions that may promote orthogonal chain pairing. Substitutions of these amino acids were explored in a pair-wise fashion. Candidate positions where substitutions could be made without steric clash for at least one combination of permitted Chi angles were selected for experimental evaluation. In total, these criteria identified the amino acid pairs and substitutions shown in .

Figure 1. Structural analysis of the CH1-CL interface with kappa and lambda LCs. Structural alignment of the engineered CH1-Cκ interface (S183K CH1, V133E Cκ; PDB ID 5TDN; gray) with the wild-type CH1-Cλ complex (PDB ID 4LLD; CH1, green; Cλ, red). In this alignment, the Cλ residue Y178 exhibits significant steric clash with the engineered CH1 residue S183K.

Figure 1. Structural analysis of the CH1-CL interface with kappa and lambda LCs. Structural alignment of the engineered CH1-Cκ interface (S183K CH1, V133E Cκ; PDB ID 5TDN; gray) with the wild-type CH1-Cλ complex (PDB ID 4LLD; CH1, green; Cλ, red). In this alignment, the Cλ residue Y178 exhibits significant steric clash with the engineered CH1 residue S183K.

Table 1. Proposed electrostatic steering mutations at the CH1-Cλ interface.

Charge pairs in the CH1-Cλ interface promote correct heavy chain and light chain pairing

Paired mutations were evaluated in a model DuetMab bsIgG carrying the variable heavy (VH) and variable light (VL) domains of in-house anti-epidermal growth factor receptor (αEGFR) and anti-receptor tyrosine-protein kinase 2 (αHER2) mAbs. Specifically, the VH and VL genes of αEGFR were inserted into a human constant gamma-1 heavy chain (CH1–CH3) carrying the ‘hole’ mutations and a constant Kappa LC (Cκ), whereas the VH and VL genes of αHER2 were inserted into a human constant gamma-1 heavy chain carrying the ‘knob’ mutation and a constant Lambda LC (Cλ). The previously reported engineered V12 disulfide was incorporated into the ‘knob’ arm by mutation of F126C and S121C in the HC and LC, respectively, and mutation of the native disulfide cysteine residues to valine.Citation17 For initial screening efforts, production of bsIgG was carried out in a high-throughput fashion at a scale of 3 mL. In this production, the “wild-type” version of this bsIgG (control 1) exhibited a correct HC-LC pairing of 70% (, ). Incorporation of CH1 S183K and Cκ V133E mutations into the hole arm (control 2) improved chain pairing to 92%. We then assessed the chain pairing of bsIgGs containing both S183K/V133E in ‘hole’ arm and one of the 58 candidate mutation pairs in the ‘knob’ arm (, ). In this cohort, five charge-pair combinations exhibited improved chain pairing to values between 93% and 96%: variants 33, 34, 35, 36, and 41.

Figure 2. Screening CH1-Cλ interface mutants for improved chain pairing properties. Bispecific αEGFR/αHER2 antibodies containing electrostatic steering mutations () were generated via a small-scale, high-throughput production and purification platform. Relative abundances of kappa and lambda LCs in each antibody were measured by capillary electrophoresis, and the resulting stoichiometric ratios were used to calculate chain pairing percentages.

Figure 2. Screening CH1-Cλ interface mutants for improved chain pairing properties. Bispecific αEGFR/αHER2 antibodies containing electrostatic steering mutations (Table 2) were generated via a small-scale, high-throughput production and purification platform. Relative abundances of kappa and lambda LCs in each antibody were measured by capillary electrophoresis, and the resulting stoichiometric ratios were used to calculate chain pairing percentages.

Table 2. Screening of candidate variants for improved chain pairing properties.

Liquid chromatography-mass spectrometry (LCMS) analysis of the intact, non-reduced antibodies identified the prominent mis-paired species in variant 1 as a heterodimer with two EGFR LCs (Supplementary Figure S1). Notably, this results in antibodies in which the EGFR LC containing the native C220 residue mis-pairs with the HER2 HC containing the engineered F126C residue. This pairing does not result in disulfide bond formation between the HC and LC, as evidenced by dissociation of the EGFR LC during mass analysis.

To evaluate potential effects on production, the selected charge-pair variants with improved pairing and control molecules were expressed at 100 mL scale. All DuetMabs exhibited similar titers with the exception of variant #41, which was reduced 2.5-fold (). Chain pairing at the 100 mL scale was consistent with the 3 mL scale productions and showed an average difference of 3 ± 2%. Overall, CH1-Cλ charge-pair combinations improved HC-LC pairing compared to S183K/V133E alone, with variants 33, 34, 36, and 41 exhibiting >96% correct pairing after protein A purification. Secondary affinity purification and preparative size-exclusion chromatography (SEC) were used to remove mis-paired bsIgGs and aggregates in preparation for subsequent biochemical characterization. Bispecific IgG complexes were confirmed by subunit mass spectrometry analysis of the Fab-digested fragments, which identified masses consistent with the correctly paired bispecific antibodies (Supplementary Figure S2, Supplementary Table S1).

Table 3. Production and chain pairing profiles of engineered DuetMab variants.

DuetMabs with charge pair variants have similar physicochemical properties

A crucial feature for the general application of electrostatic steering mutations in bsIgG production is that they have minimal impacts on the native physicochemical and biological properties of the antibody. To determine whether the CH1-Cλ charge pair mutations altered antibody function or structure, we performed a series of biochemical and biophysical assessments on the selected charge pair variants.

We assessed the effects of CH1-Cλ mutations on the binding affinity of the αHER2 arm by biolayer interferometry. DuetMabs without charge mutations in the αHER2 Fab (controls 1 and 2) had KD values of 0.35 and 0.51 nM, respectively, whereas variants 33, 34, 35, 36, and 41 had similar kinetic values from 0.24 to 0.49 nM (Supplementary Figure S3 and Supplementary Table S2). Differential scanning calorimetry (DSC) was used to measure the thermal stability of the DuetMab antibodies. Thermograms of control 2 and the selected variants exhibited highly similar profiles (, ). Furthermore, the Tonset value of control 2 was 48.4°C, whereas selected variants had values of 48.3–52.5°C. Differential scanning fluorimetry (DSF) further identified no significant difference in Tonset or Tm values between electrostatic steering mutants and control molecules (Supplementary Table S3). Aggregation and fragmentation propensity of the variants was further assessed by an accelerated stability heat stress study wherein SEC was used to monitor the levels of aggregates and fragments before and after incubation at 45°C for 14 days. No significant increase in aggregation was observed for any of the DuetMab variants, and only variant 35 exhibited a low-level increase in fragmentation (Supplementary Table S3). Additionally, all variants exhibited SEC retention times consistent with that of an IgG1 control. Collectively, these results provide evidence that the incorporation of the selected electrostatic steering mutations into the CH1-Cλ interface had minimal effects on the physicochemical properties of the antibody.

Figure 3. Differential scanning calorimetry on engineered DuetMabs. Thermograms show no difference between control 2 and the engineered variants. Differences between control 1 and control 2 are likely due to the S183K and V133E mutations in the CH1 and Cκ domains, respectively.

Figure 3. Differential scanning calorimetry on engineered DuetMabs. Thermograms show no difference between control 2 and the engineered variants. Differences between control 1 and control 2 are likely due to the S183K and V133E mutations in the CH1 and Cκ domains, respectively.

Table 4. Differential scanning calorimetry on engineered DuetMab variants.

DuetMabs with charge pair variants have similar biological and pharmacokinetic properties

Anti-EGFR antibodies have been shown to inhibit EGFR ligand binding, EGFR/HER2 heterodimerization, downstream signaling, and cell proliferation.Citation24 The cytotoxic activity of the engineered DuetMab variants was assessed using a viability assay with a non-small cell lung cancer cell line, NCI H358, which expresses both EGFR and HER2 on the cell surface. Half-maximal effective concentration (EC50) values of control 1 and 2 for inducing cell cytotoxicity were 0.57 and 1.08 nM, respectively, and EC50 values of the selected variants all fell within a similar range from 0.40 to 0.81 nM (). Maximum killing values of the controls and variants were also comparable and ranged between 45% and 48%.

Figure 4. Biological activity of engineered DuetMabs. Controls and select engineered variants were assayed for cytotoxic activity on NCI H358 cells. Each point represents the mean values of triplicate wells and error bars represent ± the standard error of the mean (SEM). R347 is an isotype control antibody.

Figure 4. Biological activity of engineered DuetMabs. Controls and select engineered variants were assayed for cytotoxic activity on NCI H358 cells. Each point represents the mean values of triplicate wells and error bars represent ± the standard error of the mean (SEM). R347 is an isotype control antibody.

The PK profile of control 1 and variants 33, 34, and 41 was evaluated in human FcRn transgenic Tg32 mice. Mice were dosed intravenously at 5 mg/kg and the PK properties were assessed over 35 days. Calculated antibody half-lives were ~180 hours and estimates in human as determined by allometric scaling from mouse PK data were ~17 days (, Supplementary Figure S4). Variant 34 exhibited slightly faster clearance and a reduced half-life by ~1.3-fold. Altogether, these results demonstrate that electrostatic charge pair variants preserve the biological and PK properties of the parent antibodies.

Table 5. Pharmacokinetic properties of engineered DuetMab variants.

Structural investigation of charge pair mutations in the CH1-Cλ interface

To determine the molecular implications of electrostatic steering mutations at the CH1-Cλ interface, X-ray crystallographic structures were determined for two Fab molecules containing A141D/T117R and A141E/T117R (variants 33 and 34), which showed excellent pairing and developability profiles. Crystals of these Fabs diffracted to resolutions of 2.1 Å and 2.0 Å, respectively (Supplementary Table S4). The resulting refined structures show close alignment with the wild-type CH1-Cλ structure (RMSD < 0.7 Å) and determined that the mutated amino acids engage in a salt bridge interaction (). These residues are in a buried position in the CH1-Cλ interface and have calculated solvent-accessible surface areas of 15–29%. Any energetic penalty due to the desolvation of these charged residues appears to be compensated by the favorable interactions of the protein–protein interface.Citation25 The observed improvements in HC-LC pairing likely arise due to a combination of the favorable interactions of the engineered salt bridge, as well as unfavorable interactions that may form during chain mispairing where unsatisfied hydrogen bond donors and acceptors may be present.

Figure 5. Crystallographic analysis of engineered DuetMabs. (a) Crystal structure of the Fab fragment in engineered variant #33 (CH1 A141D, Cλ T117R, PDB ID 8TJF) and (b) engineered variant #34 (CH1 A141E, Cλ T117R, PDB ID 8TI4) in structural alignment with the wild-type IgG1 CH1:Cλ complex (PDB ID 4LLD). RMSD values in these alignments were 0.669 Å and 0.577 Å, respectively. Dotted lines highlight hydrogen bonds present in engineered salt bridge interactions (A141D/T117R, 2.6 Å; A141E/T117R, 3.1 Å).

Figure 5. Crystallographic analysis of engineered DuetMabs. (a) Crystal structure of the Fab fragment in engineered variant #33 (CH1 A141D, Cλ T117R, PDB ID 8TJF) and (b) engineered variant #34 (CH1 A141E, Cλ T117R, PDB ID 8TI4) in structural alignment with the wild-type IgG1 CH1:Cλ complex (PDB ID 4LLD). RMSD values in these alignments were 0.669 Å and 0.577 Å, respectively. Dotted lines highlight hydrogen bonds present in engineered salt bridge interactions (A141D/T117R, 2.6 Å; A141E/T117R, 3.1 Å).

Engineered variants exhibit robust improvements in chain pairing across multiple antibody pair combinations

Chain-pairing in bsIgGs has been demonstrated to exhibit significant Fv dependence.Citation15,Citation16,Citation23 To determine whether the observed improvements in chain pairing from the selected variants extend to other antibody sequences, we designed four additional model DuetMab bsIgGs: αEGFR(clone A, Cκ)-αCD3(clone A, Cλ), αPD1(Cκ)-ISO(isotype control, Cλ), αSTEAP2(Cκ)-αCD3(clone B, Cλ), and αCMET(Cκ)-αEGFR(clone B, Cλ). Notably, although the charge pair S183K/V133E in CH1/Cκ alone exhibited >95% correct chain pairing for αSTEAP2-αCD3, this value was reduced to 66–83% in the remaining three DuetMabs. In contrast, the charge variants significantly improved pairing ratios independent of Fv sequence (, Supplementary Table S5). In particular, variants #33, 34, and 41 exhibited excellent chain pair ratios ranging from 90% to 100% for all five DuetMab designs. These data suggest that the engineered charge-pair variants show consistent improvement in chain pairing across several Fv sequences.

Figure 6. Chain pairing of engineered DuetMabs with diverse Fv sequences. Controls and selected charge pair variants (33, 34, 35, 36, and 41) were incorporated into a total of five DuetMabs with different Fv sequence combinations. Chain pairing is displayed after protein A as determined by kappa and lambda light chain stoichiometry.

Figure 6. Chain pairing of engineered DuetMabs with diverse Fv sequences. Controls and selected charge pair variants (33, 34, 35, 36, and 41) were incorporated into a total of five DuetMabs with different Fv sequence combinations. Chain pairing is displayed after protein A as determined by kappa and lambda light chain stoichiometry.

Discussion

The efficient and reliable production of bispecific IgG antibodies presents an engineering challenge that requires delicate balance. On one hand, natural antibody sequences exhibit promiscuous binding when co-expressed with other antibody chains, and protein engineering strategies are needed to drive the correct assembly of complex bispecific antibodies. On the other hand, each departure from the native antibody sequence can be a liability due to potentially unfavorable effects on production, stability, or immunogenicity. Indeed, the therapeutic field is trending toward more native IgG-like structures that can leverage the efficient production techniques of conventional mAbs while limiting developability risks. A further complication is that HC-LC pairing properties exhibit significant Fv dependence, and solutions for one bsIgG may have mixed success when applied to other Fv sequences (, Supplementary Table S5).Citation15,Citation16,Citation23 The ideal engineering solutions for bsIgG production, therefore, are those that can efficiently achieve cognate chain pairing across a variety of bsIgGs with minimal alteration to native sequences.

Previous studies have shown that the incorporation of pairs of charged residues into the CH1-CL interface of bsIgGs promotes orthogonal chain pairing.Citation19,Citation20 This work introduced substantial contributions to the antibody engineering toolkit for bsIgG production, but the efficacy of this strategy has only been established for the Kappa CL domain (Cκ) and has not been demonstrated for Lambda CL domains (Cλ). A possible explanation may be due to the presence of a tyrosine in the CH1-Cλ interface that is predicted to clash with the CH1 S183K mutation ().

To address this unmet need, we sought to identify electrostatic steering mutations at the CH1-Cλ interface that promote cognate chain pairing and exhibit compatibility with the DuetMab platform for bsIgG production. DuetMab antibodies consist of four independent chains and drive cognate HC-LC pairing by the substitution of the native CH1-CL disulfide bond on one Fab arm with an engineered V12 disulfide.Citation17,Citation21,Citation22 We used a structure-guided approach to identify 58 candidate pairs of electrostatic steering mutations at the CH1-Cλ interface (). In order to ensure compatibility with the V12 disulfide as well as previously established CH1-Cκ charge pair mutations, we incorporated candidate mutation pairs into a model DuetMab targeting EGFR and HER2. The EGFR Fab arm contained the native HC-LC disulfide and the CH1 S183K and Cκ V133E mutations, while the HER2 Fab arm contained the engineered V12 disulfide and the candidate CH1-Cλ electrostatic steering mutations. Generation and subsequent screening of these candidates identified a select set of variants with highly specific chain pairing properties: CH1/Cλ mutations A141D/T117R, A141E/T117R, A141S/T117R, A141T/T117R, and A141D/T117K ( and ). Subsequent analysis of these DuetMabs demonstrated that these mutations largely preserve the production, physicochemical, and biological properties of the parent antibodies.

Structural characterization of Fabs containing the CH1/Cλ mutations A141D/T117R and A141E/T117R confirmed the presence of salt bridge interactions between the mutated interface residues (). The energetic favorability of these interactions may directly promote orthogonal HC-LC pairing. Additionally, potential unfavorable interactions that may occur at the CH1-Cλ interface in the context of mis-paired chains could also contribute to correct pairing. The extent to which the observed improvements in chain pairing are due to one or a combination of these phenomena is unknown.

In summary, we introduced novel electrostatic steering mutations at the CH1-Cλ interface that, together with the engineered V12 disulfide, provide substantial improvements to chain pairing and preserve favorable antibody developability profiles. Importantly, this enhanced DuetMab design does not require any engineering to Fv sequences and is robust when applied to a variety of bsIgG combinations (). We anticipate that this improved DuetMab design will serve as a platform for the reliable production of bsIgGs, as well as the expansion into more complex multispecific antibody designs with enhanced functional capabilities.

Materials and methods

Expression and purification of bispecific antibodies

Bispecific antibodies were produced using two bicistronic mammalian expression vectors: one plasmid containing two heavy chains (pDuet-Heavy) and a second containing two LCs (pDuet-Light) similar to as described previously.Citation17 Briefly, the first operon of pDuet-Heavy contained a heavy chain with ‘hole’ (T366S, L368A, and Y407V), heterodimer disulfide (Y349C), and RF (H435R/Y436F) mutations in the CH3 domain, while the second operon contained the ‘knob’ (T366W) and heterodimer disulfide (S354C) mutations in CH3.Citation13,Citation26 Additional V12 disulfide (F126C, C220V) and/or electrostatic steering mutations were incorporated into CH1 domains for molecules as listed in .Citation17 The first operon of the pDuet-Light vector contained LCs with a constant kappa (Cκ) domain, while the second operon contained LCs with a constant lambda (Cλ) domain and the V12 disulfide mutations (S121C, C212V). Additional electrostatic steering mutations were incorporated into Cκ and Cλ domains for molecules as listed in . Antibody amino acid sequences are available in Supplementary Table S6.

Antibodies were produced by transient co-transfection of Chinese hamster ovary (CHO) cells with pDuet-Heavy and pDuet-Light expression vectors using PEI-MAX (Polysciences) as a transfection reagent. CHO cells were maintained using a proprietary culture medium. Cell supernatants were collected 7 and 13 days after transfection and filtered through a 0.22 μm sterile filter in preparation for titer measurements. Antibody concentrations were determined using protein A sensors (Sartorius) on an Octet384 instrument using the manufacturer’s recommended protocols. Antibodies were purified by either protein A magnetic bead affinity purification (GenScript) or standard protein A affinity chromatography (Cytiva), followed by LC affinity chromatography if necessary, in accordance with the manufacturer’s protocol, and were subsequently buffer exchanged into phosphate-buffered saline (PBS; pH 7.2).

The purity and oligomeric state of purified molecules were assessed by microfluidics-based electrophoresis and analytical SEC. Microfluidics-based electrophoresis was performed using a 2100 Bioanalyzer system (Agilent) in accordance with the manufacturer’s protocols. In this analysis, we observed that kappa and lambda LCs, despite being similar molecular weight, are efficiently resolved, possibly due to differences in amino acid composition between the Cκ and Cλ domains which may contribute to different CE-SDS mobilities. Relative abundances of antibody kappa and lambda LCs were used to calculate the percentage of correctly paired antibody.

Analytical SEC-HPLC was performed on an Agilent 1260 Infinity HPLC system using a TSK-gel G3000SWxL column (Tosoh Biosciences). Preparative SEC-HPLC was carried out using a Superdex 200 column (Cytiva) to remove protein aggregates. Concentrations of purified antibodies were determined by absorbance measurements at 280 nm.

Biolayer interferometry

Binding measurements were made using an Octet RED384 instrument (Forte Bio). Briefly, streptavidin (SA) biosensors were loaded with biotinylated HER2 protein (ACRO Biosystems) in PBS (pH 7.2) with 1 mg/ml bovine serum albumin (BSA) and 0.05% (v/v) Tween. Loaded biosensors were washed in the same buffer before carrying out association and dissociation measurements with various antibodies at the indicated times. Kinetic parameters were determined using a non-linear fit of the data with the Octet software (v.12.2.1.24).

Mass spectrometry

Proteins were deglycosylated using PNGase F (Promega) before intact mass analysis or digested using IgdE (Genovis) before sub-unit mass analysis. The protein mass was acquired by a UPLC coupled to a Xevo G2-XS QTof mass spectrometer (Waters). A BEH C4 column (300 Å, 1.7 µm, 2.1 × 50 mm; Waters) was used for the reversed-phase separation. Mobile phases A and B were 0.02% TFA in water and in acetonitrile, respectively. Mass spectra were collected at a mass-to-charge ratio range of 800–4,500. Molecular mass was determined by deconvolution using the MaxEnt I algorithm within Waters MassLynx.

High performance-SEC

Antibody samples were analyzed using HP-SEC to determine levels of aggregate, monomer, and fragment. Samples (100 μg in PBS buffer) were injected on an Agilent 1200 series high-performance liquid chromatography (HPLC) instrument and separated using a TSKgel G3000SWxl size-exclusion column (Tosoh Bioscience #08541). The mobile phase was 100 mM sodium phosphate (pH 6.8), and sample flow rate was 1 mL/min. Ultraviolet (UV) detection was performed at 280 nm.

Heat stress accelerated stability

Samples were diluted to 1 mg/mL in PBS (pH 7.2) and incubated for 2 weeks at either 4°C or 45°C. Samples were analyzed by HP-SEC to determine monomer, aggregate, and fragment percentages based on curve integration using the HPLC ChemStation software (Agilent). The change in monomer, aggregate, and fragment content was calculated from the difference between each sample incubated at 45°C versus 4°C.

Differential scanning fluorimetry

DSF measurements were performed using a previously established method with minor modifications.Citation27 Samples were prepared by combining 20 µL of protein sample at 1 mg/mL in PBS (pH 7.2) with 5 µL of SYPRO Orange dye (Invitrogen S-6651) diluted to 40X in PBS (pH 7.2) in a 96-well PCR plate in duplicate. The plate was sealed and measurements performed in a QuantStudio 7 Flex Real-Time PCR System (Applied Biosystems). Samples were subjected to an initial equilibration step at 25°C for 2 min, followed by a temperature ramp to 99°C at 0.05°C/sec increments. The fluorescence emission was monitored using the FAM filter set. The Tm value for each sample was calculated in the Protein Thermal Shift software (Applied Biosystems) using the Boltzmann method, and Tonset values were approximated from these curves.

Cell viability assays

Cell viability was determined using the CellTiter-Glo™ Luminescent Cell Viability Assay (Promega), which measures ATP abundance as an indicator for metabolically active cells. In brief, NCI H358 cells were seeded in 96-well plates at a density of ~1 × 10Citation4 cells/well in RPMI 1640 media supplemented with 0.1% BSA, and 0.2 ng/mL human recombinant EGF. Antibodies at various concentrations were added to samples in triplicate, and cells were incubated for 72 hr at 37°C and 5% CO2 in a humidified incubator. After treatment, the cells were exposed to CellTiter-Glo® reagent (Promega) for ~15 min and OD409 was measured using an EnVision 2104 Multilabel plate reader (PerkinElmer). Cell viability was determined by comparing ATP signal relative to that of control cells with no antibody treatment.

Expression and purification of antibody Fabs

Fabs were transiently expressed in a suspension of human embryonic kidney (HEK) 293 cells, using 293fectin Transfection Reagent (Life Technologies) following the manufacturer’s protocols. Cells were grown in FreeStyle 293-F Expression Medium (Life Technologies) for 10 days and fed with a proprietary cell feed solution. Cells were harvested, and the supernatant was filtered through a 0.2 µM filter prior to purification. Fabs were purified using a 5 ml CaptureSelect CH1-XL column (Thermo Fisher Scientific), dialyzed against 25 mM Hepes (pH 7) and further purified using a 5 ml HiTrap SP HP cation exchange column (Cytiva) in a NaCl gradient.

Crystallization, harvesting, and X-ray diffraction

Fab fragments were purified using a Superdex 200 Increase 10/300 GL column (Cytiva) pre-equilibrated with 25 mM HEPES, pH 7.5, and 100 mM NaCl to ensure homogeneity of the samples. Initial crystallization trials were carried out by the sitting-drop vapor-diffusion method at 20°C. Crystallization drops were dispensed in 96-well crystallization plates (Intelli-Plate 102-0001-20, Art Robbins Instruments) using a Phoenix crystallization robot (Art Robbins Instruments) and commercially available crystallization screens. The drops were composed of equal volumes of protein and reservoir buffer.

Crystals were harvested from sitting drop plates from the following crystallization solutions: for A141E/T117R, 0.1 M Bis-Tris at pH 6.5, 25% w/v PEG 3350, and a protein concentration of 18.4 mg/ml; for A141D/T117R, 200 mM sodium chloride, 0.1 M Bis-Tris at pH 5.5, 25% w/v PEG 3350, and a protein concentration of 9 mg/ml. Harvested crystals were flash-cooled in liquid nitrogen and diffraction experiments were performed on a beamline B14–1 at Stanford Synchrotron Radiation Lightsource at 100 K. Diffraction data were collected from a single crystal for each Fab and were processed, integrated, and scaled with the XDS software package.Citation28

Structures of both Fab molecules were determined using molecular replacement method with the MolRep programCitation29 from the CCP4Citation30 suite of crystallographic software. Model building was performed using CootCitation31, and Refmac5Citation32 was used for refinement. Experimental structure factors and corresponding refined models for CH1 A141D:Cλ T117R and A141E:Cλ T117R Fab structures were deposited with the RCSB PDB with accession codes 8TJF and 8TI4, respectively.Citation33

Pharmacokinetic analysis

Female Tg32 mice with human neonatal Fc receptor (FcRn) between 8 and 12 weeks-old (Jackson Laboratory), were injected with antibodies intravenously via tail vein at a dose of 5 mg/kg into 6 animals per group. Serial submandibular bleeds were obtained from mice at 1 hr, 6 hr, 24 hr, 2 d, 4 d, 7 d, 11 d, 14 d, 21 d and 28 d. Terminal bleeds were taken with cardiac puncture on day 36. After 30 min at room temperature, blood samples were centrifuged at 2500 rpm for 15 min and sera were collected for analyses. All PK studies were approved by the Institutional Animal Care and Use Committee at AstraZeneca.

The concentration of antibodies in mouse serum samples was determined using a universal ELISA assay. The method procedure was a stepwise format in which wash steps followed each incubation step. Microtiter plates were coated with a sheep anti-human IgG antibody. A standard curve of each antibody was prepared in 10% pooled naïve mouse serum. Three levels (high, medium, and low) of quality control (QC) samples were prepared in 100% pooled naïve mouse serum. Following a blocking step with Casein buffer, standard curve calibrators, QCs, and samples diluted to the method minimum required dilution (MRD) of 1:10 were added to the microtiter plates and incubated for 1 h with shaking at room temperature. After a wash, HRP-labeled goat anti-human IgG antibody was added to the plate and incubated for 1 hr with shaking. Reactions were stopped with acid, and the plate was read on a spectrophotometer at 450 nm. Data were analyzed with SoftMax® Pro (SMP), version 7.1. The standard curve was established using a 4-parameter logistical curve fit model without weighting. The quantitative range of this assay was 15.625 ng/mL (lower limit of quantitation [LLOQ]) to 2000 ng/mL (upper limit of quantitation [ULOQ]). PK parameters were estimated via non-compartmental analysis using the software Phoenix WinNonlin version 8.2 (Certera).

Abbreviations

Standard 3-letter and 1-letter amino acid abbreviations are used throughout this manuscript. Antibody amino acids are numbered according to the Eu convention.
bsIgG=

Bispecific immunoglobulin G

CH1=

Immunoglobulin constant domain 1

CH3=

Immunoglobulin constant domain 3

CL=

Constant light chain domain

Cλ=

Lambda constant light chain domain

Cκ=

Kappa constant light chain domain

CD3=

Cluster of differentiation 3

CMET=

Tyrosine-protein kinase Met

DSC=

Differential scanning calorimetry

DSF=

Differential scanning fluorimetry

EC50=

Half-maximal effective concentration

EGFR=

Epidermal growth factor receptor

Fab=

Antigen-binding fragment

FcRn=

Neonatal crystallizable fragment receptor

Fv=

Variable fragment

HC=

Heavy chain

HEK=

Human embryonic kidney

HER2=

Receptor tyrosine protein kinase 2

HPLC=

High-performance liquid chromatography

ISO=

Isotype control

LC=

Light chain

LCMS=

Liquid chromatography-mass spectrometry

LLOQ=

Lower limit of quantitation

MRD=

Minimum required dilution

PD1=

Programmed cell death protein 1 receptor

PK=

Pharmacokinetic

SA=

Streptavidin

SEC=

Size exclusion chromatography

SEM=

Standard error of the mean

TFA=

Trifluoroacetic acid

ULOQ=

Upper limit of quantification

QC=

Quality control

Supplemental material

Supplemental Material

Download Zip (560.4 KB)

Disclosure statement

The authors were employed by AstraZeneca at the time this work was performed.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19420862.2023.2273449

Additional information

Funding

Use of the Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, is supported by the U.S. Department of Energy, Office of Science, Office of Basic Energy Sciences under Contract No. DE-AC02-76SF00515. The SSRL Structural Molecular Biology Program is supported by the DOE Office of Biological and Environmental Research, and by the National Institutes of Health, National Institute of General Medical Sciences (including P41GM103393).

References

  • Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol. 2015;67:95–10. doi:10.1016/j.molimm.2015.01.003.
  • Kontermann RE, Brinkmann U. Bispecific antibodies. Drug Discov Today. 2015;20(7):838–47. doi:10.1016/j.drudis.2015.02.008.
  • Mazor Y, Sachsenmeier KF, Yang C, Hansen A, Filderman J, Mulgrew K, Wu H, Dall’acqua WF. Enhanced tumor-targeting selectivity by modulating bispecific antibody binding affinity and format valence. Sci Rep. 2017;7(1):40098. doi:10.1038/srep40098.
  • Gera N. The evolution of bispecific antibodies. Expert Opin Biol Ther. 2022;22(8):945–49. doi:10.1080/14712598.2022.2040987.
  • Cotton AD, Nguyen DP, Gramespacher JA, Seiple IB, Wells JA. Development of antibody-based PROTACs for the degradation of the cell-surface immune checkpoint protein PD-L1. J Am Chem Soc. 2021;143(2):593–98. doi:10.1021/jacs.0c10008.
  • Brinkmann U, Kontermann RE. Bispecific antibodies. Sci. 2021;372(6545):916–17. doi:10.1126/science.abg1209.
  • Schaefer W, Völger HR, Lorenz S, Imhof-Jung S, Regula JT, Klein C, Mølhøj M. Heavy and light chain pairing of bivalent quadroma and knobs-into-holes antibodies analyzed by UHR-ESI-QTOF mass spectrometry. MAbs. 2016;8(1):49–55. doi:10.1080/19420862.2015.1111498.
  • Ridgway JBB, Presta LG, Carter P. ‘Knobs-into-holes’ engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Eng Des Sel. 1996;9(7):617–21. doi:10.1093/protein/9.7.617.
  • Gunasekaran K, Pentony M, Shen M, Garrett L, Forte C, Woodward A, Ng SB, Born T, Retter M, Manchulenko K. Enhancing antibody Fc heterodimer formation through electrostatic steering effects: applications to bispecific molecules and monovalent IgG. J Biol Chem. 2010;285(25):19637–46. doi:10.1074/jbc.M110.117382.
  • Atwell S, Ridgway JB, Wells JA, Carter P. Stable heterodimers from remodeling the domain interface of a homodimer using a phage display library. Journal Of Molecular Biology. 1997;270(1):26–35. PMID: 9231898. doi:10.1006/jmbi.1997.1116.
  • Labrijn AF, Meesters JI, de Goeij BECG, van den Bremer ETJ, Neijssen J, van Kampen MD, Strumane K, Verploegen S, Kundu A, Gramer MJ, et al. Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc Natl Acad Sci USA. 2013;110(13):5145–50. doi:10.1073/pnas.1220145110.
  • Fischer N, Elson G, Magistrelli G, Dheilly E, Fouque N, Laurendon A, Gueneau F, Ravn U, Depoisier J-F, Moine V, et al. Exploiting light chains for the scalable generation and platform purification of native human bispecific IgG. Nat Commun. 2015;6:6113. doi:10.1038/ncomms7113.
  • Merchant AM, Zhu Z, Yuan JQ, Goddard A, Adams CW, Presta LG, Carter P. An efficient route to human bispecific IgG. Nat Biotechnol. 1998;16:677–81. doi:10.1038/nbt0798-677.
  • Schaefer W, Regula JT, Bahner M, Schanzer J, Croasdale R, Durr H, Gassner C, Georges G, Kettenberger H, Imhof-Jung S, et al. Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci U S A. 2011;108:11187–92. doi:10.1073/pnas.1019002108. PMID: 21690412.
  • Lewis SM, Wu X, Pustilnik A, Sereno A, Huang F, Rick HL, Guntas G, Leaver-Fay A, Smith EM, Ho C, et al. Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface. Nat Biotechnol. 2014;32(2):191–98. PMID: 24463572. doi:10.1038/nbt.2797.
  • Froning KJ, Leaver-Fay A, Wu X, Phan S, Gao L, Huang F, Pustilnik A, Bacica M, Houlihan K, Chai Q, et al. Computational design of a specific heavy chain/κ light chain interface for expressing fully IgG bispecific antibodies. Protein Sci. 2017;26(10):2021–38. PMID: 28726352. doi:10.1002/pro.3240.
  • Mazor Y, Oganesyan V, Yang C, Hansen A, Wang J, Liu H, Sachsenmeier K, Carlson M, Gadre DV, Borrok MJ, et al. Improving target cell specificity using a novel monovalent bispecific IgG design. MAbs. 2015;7(2):377–89. doi:10.1080/19420862.2015.1007816.
  • Vaks L, Litvak-Greenfeld D, Dror S, Shefet-Carasso L, Matatov G, Nahary L, Shapira S, Hakim R, Alroy I, Benhar I. Design principles for bispecific IgGs, opportunities and pitfalls of artificial disulfide bonds. Antibodies. 2018;7(3):27. https://www.mdpi.com/2073-4468/7/3/27 https://mdpi-res.com/d_attachment/antibodies/antibodies-07-00027/article_deploy/antibodies-07-00027-v3.pdf?version=1533721209
  • Dillon M, Yin Y, Zhou J, McCarty L, Ellerman D, Slaga D, Junttila TT, Han G, Sandoval W, Ovacik MA, et al. Efficient production of bispecific IgG of different isotypes and species of origin in single mammalian cells. MAbs. 2017;9(2):213–30. doi:10.1080/19420862.2016.1267089.
  • Liu Z, Leng EC, Gunasekaran K, Pentony M, Shen M, Howard M, Stoops J, Manchulenko K, Razinkov V, Liu H, et al. A novel antibody engineering strategy for making monovalent bispecific heterodimeric IgG antibodies by electrostatic steering mechanism*. J Biol Chem. 2015;290(12):7535–62. doi:10.1074/jbc.M114.620260.
  • Dovedi SJ, Elder MJ, Yang C, Sitnikova SI, Irving L, Hansen A, Hair J, Jones DC, Hasani S, Wang B, et al. Design and efficacy of a monovalent bispecific PD-1/CTLA4 antibody that enhances CTLA4 blockade on PD-1(+) activated T cells. Cancer Discov. 2021;11(5):1100–17. PMID: 33419761. doi:10.1158/2159-8290.CD-20-1445.
  • Mazor Y, Hansen A, Yang C, Chowdhury PS, Wang J, Stephens G, Wu H, Dall’acqua WF. Insights into the molecular basis of a bispecific antibody’s target selectivity. MAbs. 2015;7:461–69. doi:10.1080/19420862.2015.1022695. PMID: 25730144.
  • Joshi KK, Phung W, Han G, Yin Y, Kim I, Sandoval W, Carter PJ. Elucidating heavy/light chain pairing preferences to facilitate the assembly of bispecific IgG in single cells. MAbs. 2019;11(7):1254–65. PMID: 31286843. doi:10.1080/19420862.2019.1640549.
  • Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, Moessner E, Freytag O, Friess T, Ries CH, et al. GA201 (RG7160): a novel, humanized, glycoengineered anti-EGFR antibody with enhanced ADCC and superior in vivo efficacy compared with Cetuximab. Clin Cancer Res. 2013;19(5):1126–38. doi:10.1158/1078-0432.Ccr-12-0989.
  • Xu D, Tsai CJ, Nussinov R. Hydrogen bonds and salt bridges across protein-protein interfaces. Protein Eng Des Sel. 1997;10(9):999–1012. doi:10.1093/protein/10.9.999.
  • Jendeberg L, Nilsson P, Larsson A, Denker P, Uhlén M, Nilsson B, Nygren P-Å. Engineering of Fc(1) and Fc(3) from human immunoglobulin G to analyse subclass specificity for staphylococcal protein a. J Immunol Methods. 1997;201(1):25–34. doi:10.1016/S0022-1759(96)00215-3.
  • Shan L, Mody N, Sormani P, Rosenthal KL, Damschroder MM, Esfandiary R. Developability assessment of engineered monoclonal antibody variants with a complex self-association behavior using complementary Analytical and in silico tools. Mol Pharm. 2018;15(12):5697–710. doi:10.1021/acs.molpharmaceut.8b00867.
  • Kabsch W. Integration, scaling, space-group assignment and post-refinement. Acta Crystallogr D Biol Crystallogr. 2010;66(2):133–44. doi:10.1107/S0907444909047374.
  • Vagin A, Teplyakov A. MOLREP: an automated program for molecular replacement. J Appl Crystallogr. 1997;30(6):1022–25. doi:10.1107/S0021889897006766.
  • Winn MD, Ballard CC, Cowtan KD, Dodson EJ, Emsley P, Evans PR, Keegan RM, Krissinel EB, Leslie AG, McCoy A. Overview of the CCP4 suite and current developments. Acta Crystallogr D Biol Crystallogr. 2011;67(4):235–42. doi:10.1107/S0907444910045749.
  • Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. 2004;60(12):2126–32. doi:10.1107/S0907444904019158.
  • Kovalevskiy O, Nicholls RA, Long F, Carlon A, Murshudov GN. Overview of refinement procedures within REFMAC5: utilizing data from different sources. Acta Crystallogr D Struct Biol. 2018;74(3):215–27. doi:10.1107/S2059798318000979.
  • Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, Shindyalov IN, Bourne PE. The protein data bank. Nucleic Acids Res. 2000;28(1):235–42. doi:10.1093/nar/28.1.235.