1
|
Tagawa H, Saeki R, Yamamoto C, Tanito K, Tanaka C, Munekawa S, Nii T, Kishimura A, Murakami H, Mori T, Katayama Y. The effect of Fc region affinity of protein-based antibody-recruiting molecules on antibody-dependent cellular cytotoxicity. RSC Adv 2024; 14:22860-22866. [PMID: 39040702 PMCID: PMC11262565 DOI: 10.1039/d4ra03391d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024] Open
Abstract
Previously, we reported anticancer molecules, Fc-binding antibody-recruiting molecules (Fc-ARMs), which crosslink proteins on cancer cells with endogenous immunoglobulin Gs (IgGs) via their Fc region. The mobilized IgGs on cancer cells can accommodate natural killer cells to induce antibody-dependent cellular cytotoxicity (ADCC). Because previous Fc-ARMs utilized Fc-binding peptides, their affinity to IgGs is weak, which resulted in the limited induction capability of ADCC. Previous Fc-ARMs also unitized small molecular ligands to cancer cells, which limited their universal applicability to any cancer cells. A recent study reported that protein-based Fc-ARMs might overcome the issues associated with non-proteinous Fc-ARMs. Here, we examined the universality of a protein-based Fc-ARM by replacing its tumor-binding domain with a human epidermal growth factor receptor 2 (HER2)-specific affibody (ZHER2:342). We also examined the requirement of its Fc-binding domain affinity. We found that the Fc-ARMs accepted an affibody as a tumor-binding domain to induce ADCC. Furthermore, the required residence time of the complex between Fc-ARM and IgG was ∼102 min, which was comparable to that when monoclonal antibodies bind to their specific antigens. However, we found that the extent of ADCC induced by Fc-ARM was lower than that of conventional IgG-mediated ADCC, indicating that further enhancement of the affinity of the antibody-binding terminus and tumor-binding terminus of Fc-ARM may be needed to achieve ADCC equivalent to that of conventional IgG-mediated ADCC.
Collapse
Affiliation(s)
- Hiroshi Tagawa
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Riku Saeki
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Chihaya Yamamoto
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Kenta Tanito
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Chihiro Tanaka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
| | - Shoki Munekawa
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Teruki Nii
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- International Research Center for Molecular Systems, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Hiroshi Murakami
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University Furo-cho, Chikusa-ku Nagoya 464-8603 Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
- Center for Future Chemistry, Kyushu University 744 Motooka, Nishi-ku Fukuoka 819-0395 Japan
| |
Collapse
|
2
|
Bruce HA, Singer AU, Blazer LL, Luu K, Ploder L, Pavlenco A, Kurinov I, Adams JJ, Sidhu SS. Antigen-binding fragments with improved crystal lattice packing and enhanced conformational flexibility at the elbow region as crystallization chaperones. Protein Sci 2024; 33:e5081. [PMID: 38924648 PMCID: PMC11201802 DOI: 10.1002/pro.5081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
It has been shown previously that a set of three modifications-termed S1, Crystal Kappa, and elbow-act synergistically to improve the crystallizability of an antigen-binding fragment (Fab) framework. Here, we prepared a phage-displayed library and performed crystallization screenings to identify additional substitutions-located near the heavy-chain elbow region-which cooperate with the S1, Crystal Kappa, and elbow modifications to increase expression and improve crystallizability of the Fab framework even further. One substitution (K141Q) supports the signature Crystal Kappa-mediated Fab:Fab crystal lattice packing interaction. Another substitution (E172G) improves the compatibility of the elbow modification with the Fab framework by alleviating some of the strain incurred by the shortened and bulkier elbow linker region. A third substitution (F170W) generates a split-Fab conformation, resulting in a powerful crystal lattice packing interaction comprising the biological interaction interface between the variable heavy and light chain domains. In sum, we have used K141Q, E172G, and F170W substitutions-which complement the S1, Crystal Kappa, and elbow modifications-to generate a set of highly crystallizable Fab frameworks that can be used as chaperones to enable facile elucidation of Fab:antigen complex structures by x-ray crystallography.
Collapse
Affiliation(s)
| | | | - Levi L. Blazer
- School of PharmacyUniversity of WaterlooWaterlooOntarioCanada
| | - Khanh Luu
- School of PharmacyUniversity of WaterlooWaterlooOntarioCanada
| | - Lynda Ploder
- School of PharmacyUniversity of WaterlooWaterlooOntarioCanada
| | | | | | | | | |
Collapse
|
3
|
Bruce HA, Singer AU, Filippova EV, Blazer LL, Adams JJ, Enderle L, Ben‐David M, Radley EH, Mao DYL, Pau V, Orlicky S, Sicheri F, Kurinov I, Atwell S, Kossiakoff AA, Sidhu SS. Engineered antigen-binding fragments for enhanced crystallization of antibody:antigen complexes. Protein Sci 2024; 33:e4824. [PMID: 37945533 PMCID: PMC10731619 DOI: 10.1002/pro.4824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/09/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
The atomic-resolution structural information that X-ray crystallography can provide on the binding interface between a Fab and its cognate antigen is highly valuable for understanding the mechanism of interaction. However, many Fab:antigen complexes are recalcitrant to crystallization, making the endeavor a considerable effort with no guarantee of success. Consequently, there have been significant steps taken to increase the likelihood of Fab:antigen complex crystallization by altering the Fab framework. In this investigation, we applied the surface entropy reduction strategy coupled with phage-display technology to identify a set of surface substitutions that improve the propensity of a human Fab framework to crystallize. In addition, we showed that combining these surface substitutions with previously reported Crystal Kappa and elbow substitutions results in an extraordinary improvement in Fab and Fab:antigen complex crystallizability, revealing a strong synergistic relationship between these sets of substitutions. Through comprehensive Fab and Fab:antigen complex crystallization screenings followed by structure determination and analysis, we defined the roles that each of these substitutions play in facilitating crystallization and how they complement each other in the process.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Daniel Y. L. Mao
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoCanada
| | - Victor Pau
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoCanada
| | - Stephen Orlicky
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoCanada
| | - Frank Sicheri
- Lunenfeld‐Tanenbaum Research InstituteSinai Health SystemTorontoCanada
- Departments of Biochemistry and Molecular GeneticsUniversity of TorontoOntarioCanada
| | | | | | - Anthony A. Kossiakoff
- Department of Biochemistry and Molecular BiologyUniversity of ChicagoChicagoIllinoisUSA
| | | |
Collapse
|
4
|
Ullah SF, Moreira G, Datta SPA, McLamore E, Vanegas D. An Experimental Framework for Developing Point-of-Need Biosensors: Connecting Bio-Layer Interferometry and Electrochemical Impedance Spectroscopy. BIOSENSORS 2022; 12:938. [PMID: 36354449 PMCID: PMC9688365 DOI: 10.3390/bios12110938] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Biolayer interferometry (BLI) is a well-established laboratory technique for studying biomolecular interactions important for applications such as drug development. Currently, there are interesting opportunities for expanding the use of BLI in other fields, including the development of rapid diagnostic tools. To date, there are no detailed frameworks for implementing BLI in target-recognition studies that are pivotal for developing point-of-need biosensors. Here, we attempt to bridge these domains by providing a framework that connects output(s) of molecular interaction studies with key performance indicators used in the development of point-of-need biosensors. First, we briefly review the governing theory for protein-ligand interactions, and we then summarize the approach for real-time kinetic quantification using various techniques. The 2020 PRISMA guideline was used for all governing theory reviews and meta-analyses. Using the information from the meta-analysis, we introduce an experimental framework for connecting outcomes from BLI experiments (KD, kon, koff) with electrochemical (capacitive) biosensor design. As a first step in the development of a larger framework, we specifically focus on mapping BLI outcomes to five biosensor key performance indicators (sensitivity, selectivity, response time, hysteresis, operating range). The applicability of our framework was demonstrated in a study of case based on published literature related to SARS-CoV-2 spike protein to show the development of a capacitive biosensor based on truncated angiotensin-converting enzyme 2 (ACE2) as the receptor. The case study focuses on non-specific binding and selectivity as research goals. The proposed framework proved to be an important first step toward modeling/simulation efforts that map molecular interactions to sensor design.
Collapse
Affiliation(s)
- Sadia Fida Ullah
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC 29634, USA
| | - Geisianny Moreira
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC 29634, USA
- Global Alliance for Rapid Diagnostics, Michigan State University, East Lancing, MI 48824, USA
| | - Shoumen Palit Austin Datta
- MIT Auto-ID Labs, Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
- Medical Device (MDPnP) Interoperability and Cybersecurity Labs, Biomedical Engineering Program, Deparment of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Eric McLamore
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC 29634, USA
- Global Alliance for Rapid Diagnostics, Michigan State University, East Lancing, MI 48824, USA
- Agricultural Sciences, Clemson University, 821 McMillan Rd, Clemson, SC 29631, USA
| | - Diana Vanegas
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC 29634, USA
- Global Alliance for Rapid Diagnostics, Michigan State University, East Lancing, MI 48824, USA
- Interdisciplinary Group for Biotechnology Innovation and Ecosocial Change-BioNovo, Universidad del Valle, Cali 76001, Colombia
| |
Collapse
|
5
|
Wei W, Corbeil CR, Gaudreault F, Deprez C, Purisima EO, Sulea T. Antibody mutations favoring
pH
‐dependent binding in solid tumor microenvironments: Insights from large‐scale structure‐based calculations. Proteins 2022; 90:1538-1546. [DOI: 10.1002/prot.26340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/26/2022] [Accepted: 03/23/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Wanlei Wei
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Christopher R. Corbeil
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Francis Gaudreault
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Christophe Deprez
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Enrico O. Purisima
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| | - Traian Sulea
- Human Health Therapeutics Research Center National Research Council Canada Montreal Quebec Canada
| |
Collapse
|
6
|
Understanding and Modulating Antibody Fine Specificity: Lessons from Combinatorial Biology. Antibodies (Basel) 2022; 11:antib11030048. [PMID: 35892708 PMCID: PMC9326607 DOI: 10.3390/antib11030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Combinatorial biology methods such as phage and yeast display, suitable for the generation and screening of huge numbers of protein fragments and mutated variants, have been useful when dissecting the molecular details of the interactions between antibodies and their target antigens (mainly those of protein nature). The relevance of these studies goes far beyond the mere description of binding interfaces, as the information obtained has implications for the understanding of the chemistry of antibody–antigen binding reactions and the biological effects of antibodies. Further modification of the interactions through combinatorial methods to manipulate the key properties of antibodies (affinity and fine specificity) can result in the emergence of novel research tools and optimized therapeutics.
Collapse
|
7
|
Harwardt J, Bogen JP, Carrara SC, Ulitzka M, Grzeschik J, Hock B, Kolmar H. A Generic Strategy to Generate Bifunctional Two-in-One Antibodies by Chicken Immunization. Front Immunol 2022; 13:888838. [PMID: 35479092 PMCID: PMC9036444 DOI: 10.3389/fimmu.2022.888838] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 12/21/2022] Open
Abstract
Various formats of bispecific antibodies exist, among them Two-in-One antibodies in which each Fab arm can bind to two different antigens. Their IgG-like architecture accounts for low immunogenicity and also circumvents laborious engineering and purification steps to facilitate correct chain pairing. Here we report for the first time the identification of a Two‐in‐One antibody by yeast surface display (YSD) screening of chicken-derived immune libraries. The resulting antibody simultaneously targets the epidermal growth factor receptor (EGFR) and programmed death‐ligand 1 (PD-L1) at the same Fv fragment with two non-overlapping paratopes. The dual action Fab is capable of inhibiting EGFR signaling by binding to dimerization domain II as well as blocking the PD-1/PD-L1 interaction. Furthermore, the Two-in-One antibody demonstrates specific cellular binding properties on EGFR/PD-L1 double positive tumor cells. The presented strategy relies solely on screening of combinational immune-libraries and obviates the need for any additional CDR engineering as described in previous reports. Therefore, this study paves the way for further development of therapeutic antibodies derived from avian immunization with novel and tailor-made binding properties.
Collapse
Affiliation(s)
- Julia Harwardt
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Jan P. Bogen
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Michael Ulitzka
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Ferring Darmstadt Laboratory, Biologics Technology and Development, Darmstadt, Germany
| | - Julius Grzeschik
- Ferring Biologics Innovation Centre, Biologics Technology and Development, Epalinges, Switzerland
| | - Björn Hock
- Ferring Biologics Innovation Centre, Biologics Technology and Development, Epalinges, Switzerland
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
- Centre for Synthtic Biology, Technical University of Darmstadt, Darmstadt, Germany
- *Correspondence: Harald Kolmar,
| |
Collapse
|
8
|
Bordeau BM, Abuqayyas L, Nguyen TD, Chen P, Balthasar JP. Development and Evaluation of Competitive Inhibitors of Trastuzumab-HER2 Binding to Bypass the Binding-Site Barrier. Front Pharmacol 2022; 13:837744. [PMID: 35250584 PMCID: PMC8895951 DOI: 10.3389/fphar.2022.837744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/27/2022] [Indexed: 12/31/2022] Open
Abstract
Our group has developed and experimentally validated a strategy to increase antibody penetration in solid tumors through transient inhibition of antibody-antigen binding. In prior work, we demonstrated that 1HE, an anti-trastuzumab single domain antibody that transiently inhibits trastuzumab binding to HER2, increased the penetration of trastuzumab and increased the efficacy of ado-trastuzumab emtansine (T-DM1) in HER2+ xenograft bearing mice. In the present work, 1HE variants were developed using random mutagenesis and phage display to enable optimization of tumor penetration and efficacy of trastuzumab-based therapeutics. To guide the rational selection of a particular 1HE mutant for a specific trastuzumab-therapy, we developed a mechanistic pharmacokinetic (PK) model to predict within-tumor exposure of trastuzumab/T-DM1. A pharmacodynamic (PD) component was added to the model to predict the relationship between intratumor exposure to T-DM1 and the corresponding therapeutic effect in HER2+ xenografts. To demonstrate the utility of the competitive inhibition approach for immunotoxins, PK parameters specific for a recombinant immunotoxin were incorporated into the model structure. Dissociation half-lives for variants ranged from 1.1 h (for variant LG11) to 107.9 h (for variant HE10). Simulations predicted that 1HE co-administration can increase the tumor penetration of T-DM1, with inhibitors with longer trastuzumab binding half-lives relative to 1HE (15.5 h) further increasing T-DM1 penetration at the expense of total tumor uptake of T-DM1. The PK/PD model accurately predicted the response of NCI-N87 xenografts to treatment with T-DM1 or T-DM1 co-administered with 1HE. Model predictions indicate that the 1HE mutant HF9, with a trastuzumab binding half-life of 51.1 h, would be the optimal inhibitor for increasing T-DM1 efficacy with a modest extension in the median survival time relative to T-DM1 with 1HE. Model simulations predict that LG11 co-administration will dramatically increase immunotoxin penetration within all tumor regions. We expect that the mechanistic model structure and the wide range of inhibitors developed in this work will enable optimization of trastuzumab-cytotoxin penetration and efficacy in solid tumors.
Collapse
Affiliation(s)
| | | | | | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
9
|
Shi RL, Xiao G, Dillon TM, McAuley A, Ricci MS, Bondarenko PV. Identification of critical chemical modifications by size exclusion chromatography of stressed antibody-target complexes with competitive binding. MAbs 2021; 13:1887612. [PMID: 33616001 PMCID: PMC7899689 DOI: 10.1080/19420862.2021.1887612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chemical modifications (attributes) in the binding regions of stressed therapeutic proteins may affect binding to target and efficacy of therapeutic proteins. The method presented here describes the criticality assessment of therapeutic antibody modifications by size-exclusion chromatography (SEC) of competitive binding between a stressed antibody and its target, human epidermal growth factor receptor-2 (HER2), followed by SEC fractionation and peptide mapping characterization of bound and unbound antibodies. When stressed antibody and its target were mixed at a stoichiometric molar ratio of 1:2, only antibody-receptor complex eluted from SEC, indicating that binding was not decreased to break the complex. When a smaller amount of the receptor was provided (1:1), the antibody species with modifications reducing binding eluted as unbound from SEC, while the antibody-receptor complex eluted as the bound fraction. Peptide mapping revealed ratios of modifications between unbound and bound fractions. Statistical analysis after triplicate measurements (n = 3) indicated that heavy chain (HC) D102 isomerization and light chain (LC) N30 deamidation were four-fold higher in unbound fraction with high statistical significance. Although HC N55 deamidation and M107 oxidation were also abundant, they were not statistically different between unbound and bound. Our findings agree with previously published potency measurements of collected CEX fractions and the crystal structure of antibody and HER2. Overall, competitive SEC of stressed antibody-receptor mixture followed by peptide mapping is a useful tool in revealing critical residues and modifications involved in the antibody-target binding, even if they elute as a complex from SEC when mixed at 1:2 stoichiometric ratio.
Collapse
Affiliation(s)
- Rachel Liuqing Shi
- Attribute Sciences, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| | - Gang Xiao
- Attribute Sciences, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| | - Thomas M Dillon
- Attribute Sciences, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| | - Arnold McAuley
- Drug Product Technologies, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| | - Margaret S Ricci
- Attribute Sciences, Process Development, Amgen Inc , Thousand Oaks, CA, USA.,Drug Product Technologies, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| | - Pavel V Bondarenko
- Attribute Sciences, Process Development, Amgen Inc , Thousand Oaks, CA, USA
| |
Collapse
|
10
|
|
11
|
Betts A, Clark T, Jasper P, Tolsma J, van der Graaf PH, Graziani EI, Rosfjord E, Sung M, Ma D, Barletta F. Use of translational modeling and simulation for quantitative comparison of PF-06804103, a new generation HER2 ADC, with Trastuzumab-DM1. J Pharmacokinet Pharmacodyn 2020; 47:513-526. [PMID: 32710210 PMCID: PMC7520420 DOI: 10.1007/s10928-020-09702-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/07/2020] [Indexed: 12/26/2022]
Abstract
A modeling and simulation approach was used for quantitative comparison of a new generation HER2 antibody drug conjugate (ADC, PF-06804103) with trastuzumab-DM1 (T-DM1). To compare preclinical efficacy, the pharmacokinetic (PK)/pharmacodynamic (PD) relationship of PF-06804103 and T-DM1 was determined across a range of mouse tumor xenograft models, using a tumor growth inhibition model. The tumor static concentration was assigned as the minimal efficacious concentration. PF-06804103 was concluded to be more potent than T-DM1 across cell lines studied. TSCs ranged from 1.0 to 9.8 µg/mL (n = 7) for PF-06804103 and from 4.7 to 29 µg/mL (n = 5) for T-DM1. Two experimental models which were resistant to T-DM1, responded to PF-06804103 treatment. A mechanism-based target mediated drug disposition (TMDD) model was used to predict the human PK of PF-06804103. This model was constructed and validated based on T-DM1 which has non-linear PK at doses administered in the clinic, driven by binding to shed HER2. Non-linear PK is predicted for PF-06804103 in the clinic and is dependent upon circulating HER2 extracellular domain (ECD) concentrations. The models were translated to human and suggested greater efficacy for PF-06804103 compared to T-DM1. In conclusion, a fit-for-purpose translational PK/PD strategy for ADCs is presented and used to compare a new generation HER2 ADC with T-DM1.
Collapse
Affiliation(s)
- Alison Betts
- Department of Biomedicine Design, Pfizer Inc, 610 Main Street, Cambridge, MA, 02139, USA.
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands.
- Applied Biomath, 561 Virginia Rd, Suite 220, Concord, MA, 01742, USA.
| | - Tracey Clark
- Worldwide Research Procurement, Pfizer Inc, Eastern Point Rd, Groton, CT, 06340, USA
| | - Paul Jasper
- RES Group, Inc, 75 Second Avenue, Needham, MA, 02494, USA
| | - John Tolsma
- RES Group, Inc, 75 Second Avenue, Needham, MA, 02494, USA
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, 2300 RA, Leiden, The Netherlands
| | | | - Edward Rosfjord
- Oncology Research & Development, Pfizer Inc, 401 N Middletown Rd, Pearl River, NY, 10965, USA
| | - Matthew Sung
- Oncology Research & Development, Pfizer Inc, 401 N Middletown Rd, Pearl River, NY, 10965, USA
| | - Dangshe Ma
- Department of Therapeutic Proteins, Regeneron, Tarrytown, NY, 10591, USA
| | - Frank Barletta
- Oncology Research & Development, Pfizer Inc, 401 N Middletown Rd, Pearl River, NY, 10965, USA.
- Department of Biomedicine, Design Pfizer Inc, Design Pfizer Inc, Pearl River, NY, 10965, USA.
| |
Collapse
|
12
|
Arslan M, Karadag D, Kalyoncu S. Conformational changes in a Vernier zone region: Implications for antibody dual specificity. Proteins 2020; 88:1447-1457. [PMID: 32526069 DOI: 10.1002/prot.25964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/23/2020] [Accepted: 06/06/2020] [Indexed: 11/07/2022]
Abstract
Understanding the determinants of antibody specificity is one of the challenging tasks in antibody development. Monospecific antibodies are still dominant in approved antibody therapeutics but there is a significant body of work to show that multispecific antibodies can increase the overall therapeutic effect. Dual-specific or "Two-in-One" antibodies can bind to two different antigens separately with the same antigen-binding site as opposed to bispecifics, which simultaneously bind to two different antigens through separate antigen-binding units. These nonstandard dual-specific antibodies were recently shown to be promising for new antibody-based therapeutics. Here, we physicochemically and structurally analyzed six different antibodies of which two are monospecific and four are dual-specific antibodies derived from monospecific templates to gain insight about dual-specificity determinants. These dual-specific antibodies can target both human epidermal growth factor receptor 2 and vascular endothelial growth factor at different binding affinities. We showed that a particular region of clustered Vernier zone residues might play key roles in gaining dual specificity. While there are minimal intramolecular interactions between a certain Vernier zone region, namely LV4 and LCDR1 of monospecific template, there is a significant structural change and consequently close contact formation between LV4-LCDR1 loops of derived dual-specific antibodies. Although Vernier zone residues were previously shown to be important for humanization applications, they are mostly underestimated in the literature. Here, we also aim to resurrect Vernier zone residues for antibody engineering efforts.
Collapse
Affiliation(s)
- Merve Arslan
- Izmir Biomedicine and Genome Center, Izmir, Turkey.,Izmir Biomedicine and Genome Institute, Dokuz Eylül University, Izmir, Turkey
| | | | | |
Collapse
|
13
|
Kuroda D, Tsumoto K. Engineering Stability, Viscosity, and Immunogenicity of Antibodies by Computational Design. J Pharm Sci 2020; 109:1631-1651. [DOI: 10.1016/j.xphs.2020.01.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/25/2019] [Accepted: 01/10/2020] [Indexed: 12/18/2022]
|
14
|
van Faassen H, Ryan S, Henry KA, Raphael S, Yang Q, Rossotti MA, Brunette E, Jiang S, Haqqani AS, Sulea T, MacKenzie CR, Tanha J, Hussack G. Serum albumin‐binding V
H
Hs with variable pH sensitivities enable tailored half‐life extension of biologics. FASEB J 2020; 34:8155-8171. [DOI: 10.1096/fj.201903231r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/14/2020] [Accepted: 04/02/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Henk van Faassen
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Shannon Ryan
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Kevin A. Henry
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
- Department of Biochemistry, Microbiology & Immunology University of Ottawa Ottawa ON Canada
| | - Shalini Raphael
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Qingling Yang
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Martin A. Rossotti
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Eric Brunette
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Susan Jiang
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Arsalan S. Haqqani
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre National Research Council Canada Montréal QC Canada
| | - C. Roger MacKenzie
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| | - Jamshid Tanha
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
- Department of Biochemistry, Microbiology & Immunology University of Ottawa Ottawa ON Canada
| | - Greg Hussack
- Human Health Therapeutics Research Centre National Research Council Canada Ottawa ON Canada
| |
Collapse
|
15
|
Surface plasmon resonance sensing of Ebola virus: a biological threat. Anal Bioanal Chem 2020; 412:4101-4112. [PMID: 32306070 DOI: 10.1007/s00216-020-02641-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/19/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022]
Abstract
Here, different monoclonal antibodies (mAb1, mAb2 and mAb3) of Ebola virus were screened in a real-time and label-free manner using surface plasmon resonance (SPR) to select an appropriate antibody for biosensor applications against a biological warfare agent. For this purpose, a gold SPR chip was modified with 4-mercaptobenzoic acid (4-MBA), and modification was confirmed by FTIR-ATR and EIS. The 4-MBA-modified gold SPR chip was used for immobilization of the recombinant nucleoprotein of Ebola (EBOV-rNP), and the interactions of mAb1, mAb2 and mAb3 were then investigated to determine the best mAb based on the affinity constant (KD), expressed as equilibrium dissociation constant. KD values of 809 nM, 350 pM and 52 pM were found for the interaction of mAb1, mAb2 and mAb3 of Ebola with the immobilized EBOV-rNP, respectively, thus reflecting the high affinity of mAb3. This was confirmed by ELISA results. The thermodynamic parameters (ΔG, ΔH and ΔS) for the interaction between mAb3 and EBOV-rNP were also determined, which revealed that the interaction was spontaneous, endothermic and driven by entropy. The SPR limit of detection of EBOV-rNP with mAb3 was 0.5 pg ml-1, showing mAb3 to be the best high-affinity antibody in our study. This study has opened up new possibilities for SPR screening of different monoclonal antibodies of BWA through the convergence of materials science and optical techniques.
Collapse
|
16
|
Domínguez JM, Pérez-Chacón G, Guillén MJ, Muñoz-Alonso MJ, Somovilla-Crespo B, Cibrián D, Acosta-Iborra B, Adrados M, Muñoz-Calleja C, Cuevas C, Sánchez-Madrid F, Avilés P, Zapata JM. CD13 as a new tumor target for antibody-drug conjugates: validation with the conjugate MI130110. J Hematol Oncol 2020; 13:32. [PMID: 32264921 PMCID: PMC7140356 DOI: 10.1186/s13045-020-00865-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/27/2020] [Indexed: 11/19/2022] Open
Abstract
Background In the search for novel antibody-drug conjugates (ADCs) with therapeutic potential, it is imperative to identify novel targets to direct the antibody moiety. CD13 seems an attractive ADC target as it shows a differential pattern of expression in a variety of tumors and cell lines and it is internalized upon engagement with a suitable monoclonal antibody. PM050489 is a marine cytotoxic compound tightly binding tubulin and impairing microtubule dynamics which is currently undergoing clinical trials for solid tumors. Methods Anti-CD13 monoclonal antibody (mAb) TEA1/8 has been used to prepare a novel ADC, MI130110, by conjugation to the marine compound PM050489. In vitro and in vivo experiments have been carried out to demonstrate the activity and specificity of MI130110. Results CD13 is readily internalized upon TEA1/8 mAb binding, and the conjugation with PM050489 did not have any effect on the binding or the internalization of the antibody. MI130110 showed remarkable activity and selectivity in vitro on CD13-expressing tumor cells causing the same effects than those described for PM050489, including cell cycle arrest at G2, mitosis with disarrayed and often multipolar spindles consistent with an arrest at metaphase, and induction of cell death. In contrast, none of these toxic effects were observed in CD13-null cell lines incubated with MI130110. Furthermore, in vivo studies showed that MI130110 exhibited excellent antitumor activity in a CD13-positive fibrosarcoma xenograft murine model, with total remissions in a significant number of the treated animals. Mitotic catastrophes, typical of the payload mechanism of action, were also observed in the tumor cells isolated from mice treated with MI130110. In contrast, MI130110 failed to show any activity in a xenograft mouse model of myeloma cells not expressing CD13, thereby corroborating the selectivity of the ADC to its target and its stability in circulation. Conclusion Our results show that MI130110 ADC combines the antitumor potential of the PM050489 payload with the selectivity of the TEA1/8 monoclonal anti-CD13 antibody and confirm the correct intracellular processing of the ADC. These results demonstrate the suitability of CD13 as a novel ADC target and the effectiveness of MI130110 as a promising antitumor therapeutic agent.
Collapse
Affiliation(s)
| | - Gema Pérez-Chacón
- Instituto de Investigaciones Biomedicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Instituto de Investigacion Sanitaria La Paz, IdiPAZ, Madrid, Spain
| | | | | | - Beatriz Somovilla-Crespo
- Department of Immunology, Instituto de Investigacion Sanitaria Hospital de la Princesa, IIS-IP, Madrid, Spain
| | - Danay Cibrián
- Department of Immunology, Instituto de Investigacion Sanitaria Hospital de la Princesa, IIS-IP, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | - Magdalena Adrados
- Department of Pathology, Instituto de Investigacion Sanitaria Hospital de la Princesa, IIS-IP, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigacion Sanitaria Hospital de la Princesa, IIS-IP, Madrid, Spain
| | - Carmen Cuevas
- Research Department, PharmaMar S.A., Colmenar Viejo, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Department of Immunology, Instituto de Investigacion Sanitaria Hospital de la Princesa, IIS-IP, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Pablo Avilés
- Research Department, PharmaMar S.A., Colmenar Viejo, Madrid, Spain.
| | - Juan M Zapata
- Instituto de Investigaciones Biomedicas "Alberto Sols", CSIC-UAM, Madrid, Spain. .,Instituto de Investigacion Sanitaria La Paz, IdiPAZ, Madrid, Spain.
| |
Collapse
|
17
|
Menezes B, Cilliers C, Wessler T, Thurber GM, Linderman JJ. An Agent-Based Systems Pharmacology Model of the Antibody-Drug Conjugate Kadcyla to Predict Efficacy of Different Dosing Regimens. AAPS JOURNAL 2020; 22:29. [PMID: 31942650 DOI: 10.1208/s12248-019-0391-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
The pharmaceutical industry has invested significantly in antibody-drug conjugates (ADCs) with five FDA-approved therapies and several more showing promise in late-stage clinical trials. The FDA-approved therapeutic Kadcyla (ado-trastuzumab emtansine or T-DM1) can extend the survival of patients with tumors overexpressing HER2. However, tumor histology shows that most T-DM1 localizes perivascularly, but coadministration with its unconjugated form (trastuzumab) improves penetration of the ADC into the tumor and subsequent treatment efficacy. ADC dosing schedule, e.g., dose fractionation, has also been shown to improve tolerability. However, it is still not clear how coadministration with carrier doses impacts efficacy in terms of receptor expression, dosing regimens, and payload potency. Here, we develop a hybrid agent-based model (ABM) to capture ADC and/or antibody delivery and to predict tumor killing and growth kinetics. The results indicate that a carrier dose improves efficacy when the increased number of cells targeted by the ADC outweighs the reduced fractional killing of the targeted cells. The threshold number of payloads per cell required for killing plays a pivotal role in defining this cutoff. Likewise, fractionated dosing lowers ADC efficacy due to lower tissue penetration from a reduced maximum plasma concentration. It is only beneficial when an increase in tolerability from fractionation allows a higher ADC/payload dose that more than compensates for the loss in efficacy from fractionation. Overall, the multiscale model enables detailed depictions of heterogeneous ADC delivery, cancer cell death, and tumor growth to show how carrier dosing impacts efficacy to design the most efficacious regimen.
Collapse
Affiliation(s)
- Bruna Menezes
- Department of Chemical Engineering, University of Michigan, NCRC B28, 2800 Plymouth Road, Ann Arbor, Michigan, 48109-2800, USA
| | - Cornelius Cilliers
- Department of Chemical Engineering, University of Michigan, NCRC B28, 2800 Plymouth Road, Ann Arbor, Michigan, 48109-2800, USA
| | - Timothy Wessler
- Department of Chemical Engineering, University of Michigan, NCRC B28, 2800 Plymouth Road, Ann Arbor, Michigan, 48109-2800, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, NCRC B28, 2800 Plymouth Road, Ann Arbor, Michigan, 48109-2800, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, NCRC B28, 2800 Plymouth Road, Ann Arbor, Michigan, 48109-2800, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
18
|
Zwaagstra JC, Sulea T, Baardsnes J, Radinovic S, Cepero-Donates Y, Robert A, O’Connor-McCourt MD, Tikhomirov IA, Jaramillo ML. Binding and functional profiling of antibody mutants guides selection of optimal candidates as antibody drug conjugates. PLoS One 2019; 14:e0226593. [PMID: 31891584 PMCID: PMC6938348 DOI: 10.1371/journal.pone.0226593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/30/2019] [Indexed: 12/15/2022] Open
Abstract
An increasingly appreciated conundrum in the discovery of antibody drug conjugates (ADCs) is that an antibody that was selected primarily for strong binding to its cancer target may not serve as an optimal ADC. In this study, we performed mechanistic cell-based experiments to determine the correlation between antibody affinity, avidity, internalization and ADC efficacy. We used structure-guided design to assemble a panel of antibody mutants with predicted Her2 affinities ranging from higher to lower relative to the parent antibody, Herceptin. These antibodies were ranked for binding via SPR and via flow-cytometry on high-Her2 SKOV3 cells and low-Her2 MCF7 cells, the latter acting as a surrogate for low-Her2 normal cells. A subpanel of variants, representative of different Her2-binding affinities (2 strong, 2 moderate and 3 weak), were further screened via high-content imaging for internalization efficacies in high versus low-Her2 cells. Finally, these antibodies were evaluated in ADC cytotoxicity screening assays (using DM1 and MMAE secondary antibodies) and as antibody-drug conjugates (DM1 and PNU159682). Our results identified specific but weak Her2-binding variants as optimal candidates for developing DM1 and PNU ADCs since they exhibited high potencies (low to sub-nM) in high-Her2 SKOV3 cells and low toxicities in low-Her2 cells. The 2 strong-affinity variants were highly potent in SKOV3 cells but also showed significant toxicities in low-Her2 cells and therefore are predicted to be toxic in normal tissues. Our findings show that pharmacological profiling of an antibody library in multiple binding and functional assays allows for selection of optimal ADCs.
Collapse
Affiliation(s)
- John C. Zwaagstra
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
- * E-mail: (JZ); (MJ)
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Stevo Radinovic
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Yuneivy Cepero-Donates
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | - Alma Robert
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
| | | | | | - Maria Luz. Jaramillo
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, Quebec, Canada
- * E-mail: (JZ); (MJ)
| |
Collapse
|
19
|
The biological activity of bispecific trastuzumab/pertuzumab plant biosimilars may be drastically boosted by disulfiram increasing formaldehyde accumulation in cancer cells. Sci Rep 2019; 9:16168. [PMID: 31700025 PMCID: PMC6838051 DOI: 10.1038/s41598-019-52507-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023] Open
Abstract
Studies of breast cancer therapy have examined the improvement of bispecific trastuzumab/pertuzumab antibodies interacting simultaneously with two different epitopes of the human epidermal growth factor receptor 2 (HER2). Here, we describe the creation and production of plant-made bispecific antibodies based on trastuzumab and pertuzumab plant biosimilars (bi-TPB-PPB). Using surface plasmon resonance analysis of bi-TPB-PPB antibodies binding with the HER2 extracellular domain, we showed that the obtained Kd values were within the limits accepted for modified trastuzumab and pertuzumab. Despite the ability of bi-TPB-PPB antibodies to bind to Fcγ receptor IIIa and HER2 oncoprotein on the cell surface, a proliferation inhibition assay did not reveal any effect until α1,3-fucose and β1,2-xylose in the Asn297-linked glycan were removed. Another approach to activating bi-TPB-PPB may be associated with the use of disulfiram (DSF) a known aldehyde dehydrogenase 2 (ALDH2) inhibitor. We found that disulfiram is capable of killing breast cancer cells with simultaneous formaldehyde accumulation. Furthermore, we investigated the capacity of DSF to act as an adjuvant for bi-TPB-PPB antibodies. Although the content of ALDH2 mRNA was decreased after BT-474 cell treatment with antibodies, we only observed cell proliferation inhibiting activity of bi-TPB-PPB in the presence of disulfiram. We concluded that disulfiram can serve as a booster and adjuvant for anticancer immunotherapy.
Collapse
|
20
|
Gale P. Towards a thermodynamic mechanistic model for the effect of temperature on arthropod vector competence for transmission of arboviruses. MICROBIAL RISK ANALYSIS 2019; 12:27-43. [PMID: 32289057 PMCID: PMC7104215 DOI: 10.1016/j.mran.2019.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 03/03/2019] [Accepted: 03/03/2019] [Indexed: 05/21/2023]
Abstract
Arboviruses such as West Nile virus (WNV), bluetongue virus (BTV), dengue virus (DENV) and chikungunya virus (CHIKV) infect their arthropod vectors over a range of average temperatures depending on the ambient temperature. How the transmission efficiency of an arbovirus (i.e. vector competence) varies with temperature influences not only the short term risk of arbovirus outbreaks in humans and livestock but also the long term impact of climate change on the geographical range of the virus. The strength of the interaction between viral surface (glyco)protein (GP) and the host cell receptor (Cr) on binding of virus to host cell is defined by the thermodynamic dissociation constant Kd_receptor which is assumed to equal 10-3 M (at 37 °C) for binding of a sialic acid (SA) on the arthropod midgut epithelial cell surface to a SA-binding site on the surface of BTV, for example. Here virus binding affinity is modelled with increasing number of GP/Cr contacts at temperatures from 10 °C to 35 °C taking into account the change in entropy on immobilization of the whole virus on binding (ΔSa_immob). Based on published data, three thermodynamic GP/Cr binding scenarios, namely enthalpy-driven, entropy-assisted and entropy-driven, are shown to affect the temperature sensitivity of virus binding in different ways. Thus for enthalpy-driven GP/Cr binding, viruses bind host cells much more strongly at 10 °C than 35 °C. A mechanistic model is developed for the number of arthropod midgut cells with bound virus and by building in a kinetic component for the rate of arbovirus replication and subsequent spread to the arthropod salivary glands, a model for the effect of temperature on vector competence is developed. The model separates the opposing effects of temperature on midgut cell binding affinity from the kinetic component of virogenesis. It successfully accommodates both increases in vector competence with temperature as for DENV and WNV in mosquitoes and decreases as for the CHIKV 2010-1909 strain in various populations of Aedes albopictus mosquitoes. Enhanced cell binding at lower temperatures through enthalpy-driven GP/Cr binding compensates for the lower replication rate to some degree such that some transmission can still occur at lower temperatures. In contrast, the strength of entropy-driven GP/Cr binding diminishes at low temperatures although there is no minimum temperature threshold for transmission efficiency. The magnitude of ΔSa_immob is an important data gap. It is concluded that thermodynamic and kinetic data obtained at the molecular level will prove important in modelling vector competence with temperature.
Collapse
Key Words
- AIV, avian influenza virus
- Arbovirus
- BBF, brush border fragments from midgut
- C.VT, number of arthropod midgut cells with bound arbovirus at temperature T
- CHIKV, chikungunya virus
- Cfree, number of midgut epithelial cells which can bind virus with no virus bound
- Cr, host cell receptor
- Ctotal_midgut, number of midgut epithelial cells which can bind virus
- DENV, dengue fever virus
- EA, activation energy
- EBOV, Zaire ebolavirus
- EIP, extrinsic incubation period
- Enthalpy
- Entropy
- Fc, fraction of arthropod midgut cells with bound virus at temperature T
- GP, viral (glyco)protein on virus surface that binds to Cr
- HA, haemagglutinin
- HRV3, human rhinovirus serotype 3
- ICAM-1, intercellular adhesion molecule-1
- IDR, intrinsically disordered region of a protein
- Ka, binding affinity for virus to host cells at temperature T
- Kd_receptor, dissociation constant for GP from Cr
- Kd_virus, dissociation constant for virus from host cell
- M, molar (moles dm−3)
- NA, neuraminidase
- R, ideal gas constant
- RdRp, RNA dependent RNA polymerase
- SA, sialic acid
- Temperature
- VEEV, Venezuelan equine encephalitis virus
- VSV, vesicular stomatitis virus
- Vector competence
- Vfree, virus not bound to cells
- Vtotal, virus challenge dose to midgut
- WEEV, Western equine encephalitis virus
- WNV, West Nile virus
- k, rate of reaction
- n, number of GP/Cr contacts made on virus binding to cell
- pcompleteT, probability, given a virion has bound to the surface of a midgut cell, that that midgut cell becomes infected and that its progeny viruses go on to infect the salivary gland so completing the arthropod infection process within the life time of the arthropod at temperature T
- pfu, plaque-forming unit
- ptransmissionT, probability of successful infection of the arthropod salivary glands given oral exposure at temperature T
- ΔGa_receptor, change in Gibbs free energy on association of GP and Cr receptor
- ΔHa_receptor, change in enthalpy for binding of virus GP to host Cr receptor
- ΔHa_virus, change in enthalpy for binding of virus to host cell
- ΔSa_immob, change in entropy on immobilization of virus to cell surface
- ΔSa_receptor, change in entropy for binding of virus GP to host Cr receptor
- ΔSa_virus, change in entropy for binding of virus to host cell
- ΔSconf, change in conformation entropy within GP or Cr
Collapse
Affiliation(s)
- Paul Gale
- 15 Weare Close, Portland, Dorset DT5 1JP, United Kingdom
| |
Collapse
|
21
|
Schrag JD, Picard MÈ, Gaudreault F, Gagnon LP, Baardsnes J, Manenda MS, Sheff J, Deprez C, Baptista C, Hogues H, Kelly JF, Purisima EO, Shi R, Sulea T. Binding symmetry and surface flexibility mediate antibody self-association. MAbs 2019; 11:1300-1318. [PMID: 31318308 PMCID: PMC6748613 DOI: 10.1080/19420862.2019.1632114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Solution stability is an important factor in the optimization of engineered biotherapeutic candidates such as monoclonal antibodies because of its possible effects on manufacturability, pharmacology, efficacy and safety. A detailed atomic understanding of the mechanisms governing self-association of natively folded protein monomers is required to devise predictive tools to guide screening and re-engineering along the drug development pipeline. We investigated pairs of affinity-matured full-size antibodies and observed drastically different propensities to aggregate from variants differing by a single amino-acid. Biophysical testing showed that antigen-binding fragments (Fabs) from the aggregating antibodies also reversibly associated with equilibrium dissociation constants in the low-micromolar range. Crystal structures (PDB accession codes 6MXR, 6MXS, 6MY4, 6MY5) and bottom-up hydrogen-exchange mass spectrometry revealed that Fab self-association occurs in a symmetric mode that involves the antigen complementarity-determining regions. Subtle local conformational changes incurred upon point mutation of monomeric variants foster formation of complementary polar interactions and hydrophobic contacts to generate a dimeric Fab interface. Testing of popular in silico tools generally indicated low reliabilities for predicting the aggregation propensities observed. A structure-aggregation data set is provided here in order to stimulate further improvements of in silico tools for prediction of native aggregation. Incorporation of intermolecular docking, conformational flexibility, and short-range packing interactions may all be necessary features of the ideal algorithm.
Collapse
Affiliation(s)
- Joseph D Schrag
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Marie-Ève Picard
- Département de Biochimie, de Microbiologie et de Bio-informatique, PROTEO, and Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Pavillon Charles-Eugène-Marchand , Québec City, QC G1V 0A6 , Canada
| | - Francis Gaudreault
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Louis-Patrick Gagnon
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Mahder S Manenda
- Département de Biochimie, de Microbiologie et de Bio-informatique, PROTEO, and Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Pavillon Charles-Eugène-Marchand , Québec City, QC G1V 0A6 , Canada
| | - Joey Sheff
- Human Health Therapeutics Research Centre, National Research Council Canada , Ottawa , ON K1A 0R6 , Canada
| | - Christophe Deprez
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Cassio Baptista
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Hervé Hogues
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - John F Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada , Ottawa , ON K1A 0R6 , Canada
| | - Enrico O Purisima
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| | - Rong Shi
- Département de Biochimie, de Microbiologie et de Bio-informatique, PROTEO, and Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Pavillon Charles-Eugène-Marchand , Québec City, QC G1V 0A6 , Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council Canada , Montreal , QC H4P 2R2 , Canada
| |
Collapse
|
22
|
Hwang D, Nilchan N, Nanna AR, Li X, Cameron MD, Roush WR, Park H, Rader C. Site-Selective Antibody Functionalization via Orthogonally Reactive Arginine and Lysine Residues. Cell Chem Biol 2019; 26:1229-1239.e9. [PMID: 31231031 DOI: 10.1016/j.chembiol.2019.05.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 04/23/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023]
Abstract
Homogeneous antibody-drug conjugates (ADCs) that use a highly reactive buried lysine (Lys) residue embedded in a dual variable domain (DVD)-IgG1 format can be assembled with high precision and efficiency under mild conditions. Here we show that replacing the Lys with an arginine (Arg) residue affords an orthogonal ADC assembly that is site-selective and stable. X-ray crystallography confirmed the location of the reactive Arg residue at the bottom of a deep pocket. As the Lys-to-Arg mutation is confined to a single residue in the heavy chain of the DVD-IgG1, heterodimeric assemblies that combine a buried Lys in one arm, a buried Arg in the other arm, and identical light chains, are readily assembled. Furthermore, the orthogonal conjugation chemistry enables the loading of heterodimeric DVD-IgG1s with two different cargos in a one-pot reaction and thus affords a convenient platform for dual-warhead ADCs and other multifaceted antibody conjugates.
Collapse
Affiliation(s)
- Dobeen Hwang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Napon Nilchan
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Alex R Nanna
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Xiaohai Li
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Michael D Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - William R Roush
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - HaJeung Park
- X-Ray Crystallography Core, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
23
|
Modeling to capture bystander-killing effect by released payload in target positive tumor cells. BMC Cancer 2019; 19:194. [PMID: 30832603 PMCID: PMC6399851 DOI: 10.1186/s12885-019-5336-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Background Antibody-drug conjugates (ADCs) are intended to bind to specific positive target antigens and eradicate only tumor cells from an intracellular released payload through the lysosomal protease. Payloads, such as MMAE, have the capacity to kill adjacent antigen-negative (Ag–) tumor cells, which is called the bystander-killing effect, as well as directly kill antigen-positive (Ag+) tumor cells. We propose that a dose-response curve should be independently considered to account for target antigen-positive/negative tumor cells. Methods A model was developed to account for the payload in Ag+/Ag– cells and the associated parameters were applied. A tumor growth inhibition (TGI) effect was explored based on an ordinary differential equation (ODE) after substituting the payload concentration in Ag+/Ag– cells into an Emax model, which accounts for the dose-response curve. To observe the bystander-killing effects based on the amount of Ag+/Ag– cells, the Emax model is used independently. TGI models based on ODE are unsuitable for describing the initial delay through a tumor–drug interaction. This was solved using an age-structured model based on the stochastic process. Results β∈(0,1] is a fraction parameter that determines the proportion of cells that consist of Ag+/Ag– cells. The payload concentration decreases when the ratio of efflux to influx increases. The bystander-killing effect differs with varying amounts of Ag+ cells. The larger β is, the less bystander-killing effect. The decrease of the bystander-killing effect becomes stronger as Ag+ cells become larger than the Ag– cells. Overall, the ratio of efflux to influx, the amount of released payload, and the proportion of Ag+ cells determine the efficacy of the ADC. The tumor inhibition delay through a payload-tumor interaction, which goes through several stages, may be solved using an age-structured model. Conclusions The bystander-killing effect, one of the most important topics of ADCs, has been explored in several studies without the use of modeling. We propose that the bystander-killing effect can be captured through a mathematical model when considering the Ag+ and Ag– cells. In addition, the TGI model based on the age-structure can capture the initial delay through a drug interaction as well as the bystander-killing effect. Electronic supplementary material The online version of this article (10.1186/s12885-019-5336-7) contains supplementary material, which is available to authorized users.
Collapse
|
24
|
Site-specific conjugation of recognition tags to trastuzumab for peptide nucleic acid-mediated radionuclide HER2 pretargeting. Biomaterials 2019; 203:73-85. [PMID: 30877838 DOI: 10.1016/j.biomaterials.2019.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 01/11/2023]
Abstract
Pretargeting is a promising strategy to reach high imaging contrast in a shorter time than by targeting with directly radiolabeled monoclonal antibodies (mAbs). One of problems in pretargeting is a site-specific, reproducible and uniform conjugation of recognition tags to mAbs. To solve this issue we propose a photoconjugation to covalently couple a recognition tag to a mAb via a photoactivatable Z domain. The Z-domain, a 58-amino acid protein derived from the IgG-binding B-domain of Staphylococcus aureus protein A, has a well-characterized binding site in the Fc portion of IgG. We tested the feasibility of this approach using pretargeting based on hybridization between peptide nucleic acids (PNAs). We have used photoconjugation to couple trastuzumab with the PNA-based hybridization probe, HP1. A complementary [57Co]Co-labeled PNA hybridization probe ([57Co]Co-HP2) was used as the secondary targeting probe. In vitro studies demonstrated that trastuzumab-ZHP1 bound specifically to human epidermal growth factor receptor 2 (HER2)-expressing cells with nanomolar affinity. The binding of the secondary [57Co]Co-HP2 probe to trastuzumab-PNA-pretreated cells was in the picomolar affinity range. A two-fold increase in SKOV-3 tumor targeting was achieved when [57Co]Co-HP2 (0.7 nmol) was injected 48 h after injection of trastuzumab-ZHP1 (0.5 nmol) compared with trastuzumab-ZHP1 alone (0.8 ± 0.2 vs. 0.33 ± 0.06 %ID/g). Tumor accumulation of [57Co]Co-HP2 was significantly reduced by pre-saturation with trastuzumab or when no trastuzumab-ZHP1 was preinjected. A tumor-to-blood uptake ratio of 1.5 ± 0.3 was achieved resulting in a clear visualization of HER2-expressing xenografts as confirmed by SPECT imaging. In conclusion, the feasibility of stable site-specific coupling of a PNA-based recognition tag to trastuzumab and successful pretargeting has been demonstrated. This approach can hopefully be used for a broad range of mAbs and recognition tags.
Collapse
|
25
|
Kroetsch A, Qiao C, Heavey M, Guo L, Shah DK, Park S. Engineered pH-dependent recycling antibodies enhance elimination of Staphylococcal enterotoxin B superantigen in mice. MAbs 2018; 11:411-421. [PMID: 30526311 DOI: 10.1080/19420862.2018.1545510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
A new modality in antibody engineering has emerged in which the antigen affinity is designed to be pH dependent (PHD). In particular, combining high affinity binding at neutral pH with low affinity binding at acidic pH leads to a novel antibody that can more effectively neutralize the target antigen while avoiding antibody-mediated antigen accumulation. Here, we studied how the in vivo pharmacokinetics of the superantigen, Staphylococcal enterotoxin B (SEB), is affected by an engineered antibody with pH-dependent binding. PHD anti-SEB antibodies were engineered by introducing mutations into a high affinity anti-SEB antibody, 3E2, by rational design and directed evolution. Three antibody mutants engineered in the study have an affinity at pH 6.0 that is up to 68-fold weaker than the control antibody. The pH dependency of each mutant, measured as the pH-dependent affinity ratio (PAR - ratio of affinity at pH 7.4 and pH 6.0), ranged from 6.7-11.5 compared to 1.5 for the control antibody. The antibodies were characterized in mice by measuring their effects on the pharmacodynamics and pharmacokinetics (PK) of SEB after co-administration. All antibodies were effective in neutralizing the toxin and reducing the toxin-induced cytokine production. However, engineered PHD antibodies led to significantly faster elimination of the toxin from the circulation than wild type 3E2. The area under the curve computed from the SEB PK profile correlated well with the PAR value of antibody, indicating the importance of fine tuning the pH dependency of binding. These results suggest that a PHD recycling antibody may be useful to treat intoxication from a bacterial toxin by accelerating its clearance.
Collapse
Affiliation(s)
- Andrew Kroetsch
- a Department of Chemical and Biological Engineering , University at Buffalo , Buffalo , New York , USA
| | - Chunxia Qiao
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Mairead Heavey
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Leiming Guo
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Dhaval K Shah
- b Department of Pharmaceutical Sciences , University at Buffalo , Buffalo , New York , USA
| | - Sheldon Park
- a Department of Chemical and Biological Engineering , University at Buffalo , Buffalo , New York , USA
| |
Collapse
|
26
|
Lin TJ, Beal KM, Brown PW, DeGruttola HS, Ly M, Wang W, Chu CH, Dufield RL, Casperson GF, Carroll JA, Friese OV, Figueroa B, Marzilli LA, Anderson K, Rouse JC. Evolution of a comprehensive, orthogonal approach to sequence variant analysis for biotherapeutics. MAbs 2018; 11:1-12. [PMID: 30303443 PMCID: PMC6343769 DOI: 10.1080/19420862.2018.1531965] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Amino acid sequence variation in protein therapeutics requires close monitoring during cell line and cell culture process development. A cross-functional team of Pfizer colleagues from the Analytical and Bioprocess Development departments worked closely together for over 6 years to formulate and communicate a practical, reliable sequence variant (SV) testing strategy with state-of-the-art techniques that did not necessitate more resources or lengthen project timelines. The final Pfizer SV screening strategy relies on next-generation sequencing (NGS) and amino acid analysis (AAA) as frontline techniques to identify mammalian cell clones with genetic mutations and recognize cell culture process media/feed conditions that induce misincorporations, respectively. Mass spectrometry (MS)-based techniques had previously been used to monitor secreted therapeutic products for SVs, but we found NGS and AAA to be equally informative, faster, less cumbersome screening approaches. MS resources could then be used for other purposes, such as the in-depth characterization of product quality in the final stages of commercial-ready cell line and culture process development. Once an industry-wide challenge, sequence variation is now routinely monitored and controlled at Pfizer (and other biopharmaceutical companies) through increased awareness, dedicated cross-line efforts, smart comprehensive strategies, and advances in instrumentation/software, resulting in even higher product quality standards for biopharmaceutical products.
Collapse
Affiliation(s)
- T Jennifer Lin
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Kathryn M Beal
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Paul W Brown
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | | | - Mellisa Ly
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Wenge Wang
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Chia H Chu
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Robert L Dufield
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Gerald F Casperson
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - James A Carroll
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Olga V Friese
- b Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Chesterfield , MO , USA
| | - Bruno Figueroa
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Lisa A Marzilli
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Karin Anderson
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| | - Jason C Rouse
- a Biotherapeutics Pharmaceutical Sciences , Pfizer, Inc , Andover , MA , USA
| |
Collapse
|
27
|
Mizukami M, Onishi H, Hanagata H, Miyauchi A, Ito Y, Tokunaga H, Ishibashi M, Arakawa T, Tokunaga M. Efficient production of Trastuzumab Fab antibody fragments in Brevibacillus choshinensis expression system. Protein Expr Purif 2018; 150:109-118. [DOI: 10.1016/j.pep.2018.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 10/16/2022]
|
28
|
Gale P. Using thermodynamic parameters to calibrate a mechanistic dose-response for infection of a host by a virus. MICROBIAL RISK ANALYSIS 2018; 8:1-13. [PMID: 32289059 PMCID: PMC7103988 DOI: 10.1016/j.mran.2018.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/29/2017] [Accepted: 01/03/2018] [Indexed: 05/21/2023]
Abstract
Assessing the risk of infection from emerging viruses or of existing viruses jumping the species barrier into novel hosts is limited by the lack of dose response data. The initial stages of the infection of a host by a virus involve a series of specific contact interactions between molecules in the host and on the virus surface. The strength of the interaction is quantified in the literature by the dissociation constant (Kd) which is determined experimentally and is specific for a given virus molecule/host molecule combination. Here, two stages of the initial infection process of host intestinal cells are modelled, namely escape of the virus in the oral challenge dose from the innate host defenses (e.g. mucin proteins in mucus) and the subsequent binding of any surviving virus to receptor molecules on the surface of the host epithelial cells. The strength of virus binding to host cells and to mucins may be quantified by the association constants, Ka and Kmucin, respectively. Here, a mechanistic dose-response model for the probability of infection of a host by a given virus dose is constructed using Ka and Kmucin which may be derived from published Kd values taking into account the number of specific molecular interactions. It is shown that the effectiveness of the mucus barrier is determined not only by the amount of mucin but also by the magnitude of Kmucin. At very high Kmucin values, slight excesses of mucin over virus are sufficient to remove all the virus according to the model. At lower Kmucin values, high numbers of virus may escape even with large excesses of mucin. The output from the mechanistic model is the probability (p1) of infection by a single virion which is the parameter used in conventional dose-response models to predict the risk of infection of the host from the ingested dose. It is shown here how differences in Ka (due to molecular differences in an emerging virus strain or new host) affect p1, and how these differences in Ka may be quantified in terms of two thermodynamic parameters, namely enthalpy and entropy. This provides the theoretical link between sequencing data and risk of infection. Lack of data on entropy is a limitation at present and may also affect our interpretation of Kd in terms of infectivity. It is concluded that thermodynamic approaches have a major contribution to make in developing dose-response models for emerging viruses.
Collapse
Key Words
- Asp, aspartate
- CRD, carbohydrate-recognition domain
- Cr, host cell receptor
- Dose-response
- EBOV, Zaire ebolavirus
- Enthalpy
- Entropy
- G, Gibbs free energy
- GI, gastrointestinal
- GP, glycoprotein
- H, enthalpy
- HA, haemagglutinin
- HBGA, histoblood group antigen
- HeV, Hendra virus
- Ka, Kmucin, association constants
- Kd, dissociation constant for two molecules bound to each other
- L, Avogadro number
- M, molar (moles dm−3)
- MBP, mannose binding protein
- MERS-CoV, MERS coronavirus
- MRA, microbiological risk assessment
- Mucin
- NPC1, Niemann-Pick C1 protein
- NiV, Nipah virus
- NoV, norovirus
- PL, phospholipid
- PRR, pathogen recognition receptor
- Phe, phenylalanine
- R, ideal gas constant
- S, entropy
- SPR, surface plasmon resonance
- T, temperature
- TIM-1, T-cell immunoglobulin and mucin domain protein 1
- VSV, vesicular stomatitis virus
- Virus
- k, on/off rate constant
- n, number of GP/Cr molecular contacts per virus/host cell binding
- pfu, plaque-forming unit
- ΔGa, change in Gibbs free energy on association of virus and cell
- ΔHa, change in enthalpy on association of virus and cell
- ΔSa, change in entropy on association of virus and cell
- ΔΔHa, change in ΔHa
Collapse
|
29
|
Khalili H, Brocchini S, Khaw PT, Filippov SK. Comparative thermodynamic analysis in solution of a next generation antibody mimetic to VEGF. RSC Adv 2018; 8:35787-35793. [PMID: 35547916 PMCID: PMC9088213 DOI: 10.1039/c8ra07059h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022] Open
Abstract
An antibody mimetic known as Fab–PEG–Fab (FpF) is a stable bivalent molecule that may have some potential therapeutic advantages over IgG antibodies due to differences in their binding kinetics as determined by surface plasmon resonance. Here we describe the thermodynamic binding properties to vascular endothelial growth factor (VEGF) of the FpF antibody mimetics derived from bevacizumab and ranibizumab. Bevacizumab is an IgG antibody and ranibizumab is an antibody fragment (Fab). Both are used clinically to target VEGF to inhibit angiogenesis. FpFbeva displayed comparable binding affinity (KD) and binding thermodynamics (ΔH = −25.7 kcal mole−1 and ΔS = 14 kcal mole−1) to bevacizumab (ΔH = −25 kcal mole−1, ΔS = 13.3 kcal mole−1). FpFrani interactions with VEGF were characterised by large favourable enthalpy (ΔH = −42 kcal mole−1) and unfavourable entropy (ΔS = 31 kcal mole−1) changes compared to ranibizumab (ΔH = −18.5 kcal mole−1 and ΔS = 6.7 kcal mole−1), which being a Fab, is mono-valent. A large negative entropy change resulting in binding of bivalent FpF to homodimer VEGF might be due to the conformational change of the flexible regions of the FpF upon ligand binding. Mono-valent Fab (i.e. ranibizumab or the Fab derived from bevacizumab) displayed a larger degree of freedom (smaller unfavourable entropy) upon binding to homodimer VEGF. Our report describes the first comprehensive enthalpy and entropy compensation analysis for FpF antibody mimetics. While the FpFs displayed similar thermodynamics and binding affinity to the full IgG (i.e. bevacizumab), their enhanced protein stability, slower dissociation rate and lack of Fc effector functions could make FpF a potential next-generation therapy for local tissue-targeted indications. ITC illustrated similar binding thermodynamics for anti-VEGF IgG and FpFs. Bivalent FpFrani displayed larger enthalpy and entropy than monovalent ranibizumab.![]()
Collapse
Affiliation(s)
- Hanieh Khalili
- UEL School of Health
- Sport and Bioscience
- London
- UK
- UCL School of Pharmacy
| | - Steve Brocchini
- UCL School of Pharmacy
- London
- UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust
- UCL Institute of Ophthalmology
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust
- UCL Institute of Ophthalmology
- London
- UK
| | | |
Collapse
|
30
|
Hoffman F, Gavaghan D, Osborne J, Barrett I, You T, Ghadially H, Sainson R, Wilkinson R, Byrne H. A mathematical model of antibody-dependent cellular cytotoxicity (ADCC). J Theor Biol 2018; 436:39-50. [DOI: 10.1016/j.jtbi.2017.09.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
|
31
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 605] [Impact Index Per Article: 86.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
32
|
Mahmud MN, Oda M, Usui D, Inoshima Y, Ishiguro N, Kamatari YO. A multispecific monoclonal antibody G2 recognizes at least three completely different epitope sequences with high affinity. Protein Sci 2017; 26:2162-2169. [PMID: 28791742 DOI: 10.1002/pro.3263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/18/2017] [Accepted: 07/29/2017] [Indexed: 11/09/2022]
Abstract
A monoclonal antibody (mAb) G2 possesses an unusual characteristic of reacting with at least three proteins (ATP6V1C1, SEPT3, and C6H10orf76) other than its original antigen, chicken prion protein (ChPrP). The epitopes on ChPrP and ATP6V1C1 have been identified previously. In this study, we identified the epitope in the third protein, SEPT3. Interestingly, there was no amino acid sequence similarity among the epitopes on the three proteins. These epitopes had high binding affinities to G2 (KD = ∼10-7 M for monovalent binding and KD = ∼10-9 M for divalent binding), as determined using a SPR biosensor. This is the first report on a three-in-one mAb recognizing completely different epitope sequences with high affinity. Additionally, competitive ELISA indicated that the binding sites on G2, specific for the three different epitopes, overlapped, suggesting that the antigen-binding site may be flexible in the free form and capable of adapting to at least three different conformations to enable interactions with three different antigens.
Collapse
Affiliation(s)
- Md Nuruddin Mahmud
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Masayuki Oda
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Daiki Usui
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Yasuo Inoshima
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan.,Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Naotaka Ishiguro
- The United Graduate School of Veterinary Sciences, Gifu University, Gifu, 501-1193, Japan.,Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Gifu, 501-1193, Japan
| | - Yuji O Kamatari
- Life Science Research Center, Gifu University, Gifu, 501-1193, Japan
| |
Collapse
|
33
|
Vivcharuk V, Baardsnes J, Deprez C, Sulea T, Jaramillo M, Corbeil CR, Mullick A, Magoon J, Marcil A, Durocher Y, O’Connor-McCourt MD, Purisima EO. Assisted Design of Antibody and Protein Therapeutics (ADAPT). PLoS One 2017; 12:e0181490. [PMID: 28750054 PMCID: PMC5531539 DOI: 10.1371/journal.pone.0181490] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/30/2017] [Indexed: 11/19/2022] Open
Abstract
Effective biologic therapeutics require binding affinities that are fine-tuned to their disease-related molecular target. The ADAPT (Assisted Design of Antibody and Protein Therapeutics) platform aids in the selection of mutants that improve/modulate the affinity of antibodies and other biologics. It uses a consensus z-score from three scoring functions and interleaves computational predictions with experimental validation, significantly enhancing the robustness of the design and selection of mutants. The platform was tested on three antibody Fab-antigen systems that spanned a wide range of initial binding affinities: bH1-VEGF-A (44 nM), bH1-HER2 (3.6 nM) and Herceptin-HER2 (0.058 nM). Novel triple mutants were obtained that exhibited 104-, 46- and 32-fold improvements in binding affinity for each system, respectively. Moreover, for all three antibody-antigen systems over 90% of all the intermediate single and double mutants that were designed and tested showed higher affinities than the parent sequence. The contributions of the individual mutants to the change in binding affinity appear to be roughly additive when combined to form double and triple mutants. The new interactions introduced by the affinity-enhancing mutants included long-range electrostatics as well as short-range nonpolar interactions. This diversity in the types of new interactions formed by the mutants was reflected in SPR kinetics that showed that the enhancements in affinities arose from increasing on-rates, decreasing off-rates or a combination of the two effects, depending on the mutation. ADAPT is a very focused search of sequence space and required only 20-30 mutants for each system to be made and tested to achieve the affinity enhancements mentioned above.
Collapse
Affiliation(s)
- Victor Vivcharuk
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Jason Baardsnes
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Christophe Deprez
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Traian Sulea
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Maria Jaramillo
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | | | - Alaka Mullick
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Joanne Magoon
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Anne Marcil
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Yves Durocher
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | | | - Enrico O. Purisima
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| |
Collapse
|
34
|
Peiris D, Spector AF, Lomax-Browne H, Azimi T, Ramesh B, Loizidou M, Welch H, Dwek MV. Cellular glycosylation affects Herceptin binding and sensitivity of breast cancer cells to doxorubicin and growth factors. Sci Rep 2017; 7:43006. [PMID: 28223691 PMCID: PMC5320443 DOI: 10.1038/srep43006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/12/2017] [Indexed: 01/07/2023] Open
Abstract
Alterations in protein glycosylation are a key feature of oncogenesis and have been shown to affect cancer cell behaviour perturbing cell adhesion, favouring cell migration and metastasis. This study investigated the effect of N-linked glycosylation on the binding of Herceptin to HER2 protein in breast cancer and on the sensitivity of cancer cells to the chemotherapeutic agent doxorubicin (DXR) and growth factors (EGF and IGF-1). The interaction between Herceptin and recombinant HER2 protein and cancer cell surfaces (on-rate/off-rate) was assessed using a quartz crystal microbalance biosensor revealing an increase in the accessibility of HER2 to Herceptin following deglycosylation of cell membrane proteins (deglycosylated cells Bmax: 6.83 Hz; glycosylated cells Bmax: 7.35 Hz). The sensitivity of cells to DXR and to growth factors was evaluated using an MTT assay. Maintenance of SKBR-3 cells in tunicamycin (an inhibitor of N-linked glycosylation) resulted in an increase in sensitivity to DXR (0.1 μM DXR P < 0.001) and a decrease in sensitivity to IGF-1 alone and to IGF-1 supplemented with EGF (P < 0.001). This report illustrates the importance of N-linked glycosylation in modulating the response of cancer cells to chemotherapeutic and biological treatments and highlights the potential of glycosylation inhibitors as future combination treatments for breast cancer.
Collapse
Affiliation(s)
- Diluka Peiris
- Attana AB, Bjornnasvagen 21, SE-11419, Stockholm, Sweden
| | - Alexander F Spector
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Hannah Lomax-Browne
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| | - Tayebeh Azimi
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| | - Bala Ramesh
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Hazel Welch
- Division of Surgery and Interventional Science, UCL Medical School Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Miriam V Dwek
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, 115 New Cavendish St, W1W 6UW, UK
| |
Collapse
|
35
|
Jacquot G, Lécorché P, Malcor JD, Laurencin M, Smirnova M, Varini K, Malicet C, Gassiot F, Abouzid K, Faucon A, David M, Gaudin N, Masse M, Ferracci G, Dive V, Cisternino S, Khrestchatisky M. Optimization and in Vivo Validation of Peptide Vectors Targeting the LDL Receptor. Mol Pharm 2016; 13:4094-4105. [PMID: 27656777 DOI: 10.1021/acs.molpharmaceut.6b00687] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Active targeting and delivery to pathophysiological organs of interest is of paramount importance to increase specific accumulation of therapeutic drugs or imaging agents while avoiding systemic side effects. We recently developed a family of new peptide ligands of the human and rodent LDL receptor (LDLR), an attractive cell-surface receptor with high uptake activity and local enrichment in several normal or pathological tissues (Malcor et al., J. Med. Chem. 2012, 55 (5), 2227). Initial chemical optimization of the 15-mer, all natural amino acid compound 1/VH411 (DSGL[CMPRLRGC]cDPR) and structure-activity relationship (SAR) investigation led to the cyclic 8 amino acid analogue compound 22/VH445 ([cMPRLRGC]c) which specifically binds hLDLR with a KD of 76 nM and has an in vitro blood half-life of ∼3 h. Further introduction of non-natural amino acids led to the identification of compound 60/VH4106 ([(d)-"Pen"M"Thz"RLRGC]c), which showed the highest KD value of 9 nM. However, this latter analogue displayed the lowest in vitro blood half-life (∼1.9 h). In the present study, we designed a new set of peptide analogues, namely, VH4127 to VH4131, with further improved biological properties. Detailed analysis of the hLDLR-binding kinetics of previous and new analogues showed that the latter all displayed very high on-rates, in the 106 s-1.M-1 range, and off-rates varying from the low 10-2 s-1 to the 10-1 s-1 range. Furthermore, all these new analogues showed increased blood half-lives in vitro, reaching ∼7 and 10 h for VH4129 and VH4131, respectively. Interestingly, we demonstrate in cell-based assays using both VH445 and the most balanced optimized analogue VH4127 ([cM"Thz"RLRG"Pen"]c), showing a KD of 18 nM and a blood half-life of ∼4.3 h, that its higher on-rate correlated with a significant increase in both the extent of cell-surface binding to hLDLR and the endocytosis potential. Finally, intravenous injection of tritium-radiolabeled 3H-VH4127 in wild-type or ldlr -/- mice confirmed their active LDLR targeting in vivo. Overall, this study extends our previous work toward a diversified portfolio of LDLR-targeted peptide vectors with validated LDLR-targeting potential in vivo.
Collapse
Affiliation(s)
- Guillaume Jacquot
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Pascaline Lécorché
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Jean-Daniel Malcor
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Mathieu Laurencin
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France.,CEA-Saclay, Service d'Ingénierie Moléculaire de Protéines (SIMOPRO), Labex LERMIT, CEA-DSV-iBiTecS , 91191 Gif/Yvette Cedex, France
| | - Maria Smirnova
- Variabilité de réponse aux psychotropes, INSERM U1144 , Paris 75006, France.,Faculté de Pharmacie, Université Paris Descartes, INSERM UMR S-1144 , Paris 75006, France
| | - Karine Varini
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France.,Aix Marseille Univ, CNRS, NICN , Marseille, France
| | - Cédric Malicet
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Fanny Gassiot
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Karima Abouzid
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Aude Faucon
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Marion David
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | - Nicolas Gaudin
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France.,Aix Marseille Univ, CNRS, NICN , Marseille, France
| | - Maxime Masse
- VECT-HORUS SAS, Faculté de Médecine secteur Nord , 51 Boulevard Pierre Dramard, CS80011, 13344 Marseille Cedex 15, France
| | | | - Vincent Dive
- CEA-Saclay, Service d'Ingénierie Moléculaire de Protéines (SIMOPRO), Labex LERMIT, CEA-DSV-iBiTecS , 91191 Gif/Yvette Cedex, France
| | - Salvatore Cisternino
- Variabilité de réponse aux psychotropes, INSERM U1144 , Paris 75006, France.,Faculté de Pharmacie, Université Paris Descartes, INSERM UMR S-1144 , Paris 75006, France
| | | |
Collapse
|
36
|
Gale P. Could Bat Cell Temperature and Filovirus Filament Length Explain the Emergence of Ebola Virus in Mammals? Predictions of a Thermodynamic Model. Transbound Emerg Dis 2016; 64:1676-1693. [PMID: 27670273 DOI: 10.1111/tbed.12580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Indexed: 02/07/2023]
Abstract
The host reservoir of Zaire ebolavirus (EBOV) remains elusive. One suggestion is that EBOV emerges in mammals when the precursor virus jumps from mayflies (or other riverine insects) to insectivorous bats. However, this does not fit with the current view that filoviruses cannot infect arthropods. Here, it is first argued that the evidence that arthropods are refractory is not definitive. Second, it is proposed that a combination of filovirus filament length and the high temperature (~42°C) experienced by an insect virus ingested by a flying bat, together with the large number of insects eaten by bats (e.g. during an ephemeral mass emergence of mayflies), facilitate jumping the species barrier. The length of a filovirus filament is related to the number of genome copies (GC). Predictions from a preliminary thermodynamic model developed here suggest that filament length could greatly affect EBOV infectivity to mammalian cells with infectivity peaking for filaments of a certain length. Importantly, the infectivity to mammals of even short filaments may be more than one million-fold higher than that for the single GC virion. Third, it is proposed that at the high temperature within the bat, the phospholipid phosphatidylserine in the virus envelope promotes filament formation through fusion of single GC particles within the ingested insect, thus hugely increasing their infectivity to bats. Forth, according to the thermodynamic model, increasing the temperature from 27°C (insect cell temperature at average air temperature in Guinea, West Africa) to 42°C (bat) could increase the affinity of the filaments for bat cells by 1-2 orders of magnitude, while having no effect on the binding affinity of the single GC virions. The thermodynamic model developed here is supported by the counterintuitive observation that high glycoprotein densities on the EBOV surface reduce its infectivity in contrast to other viruses such as HIV.
Collapse
Affiliation(s)
- P Gale
- Independent Scientist, Tilehurst, Reading, Berkshire, UK
| |
Collapse
|
37
|
Cilliers C, Guo H, Liao J, Christodolu N, Thurber GM. Multiscale Modeling of Antibody-Drug Conjugates: Connecting Tissue and Cellular Distribution to Whole Animal Pharmacokinetics and Potential Implications for Efficacy. AAPS JOURNAL 2016; 18:1117-1130. [PMID: 27287046 DOI: 10.1208/s12248-016-9940-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/27/2016] [Indexed: 11/30/2022]
Abstract
Antibody-drug conjugates exhibit complex pharmacokinetics due to their combination of macromolecular and small molecule properties. These issues range from systemic concerns, such as deconjugation of the small molecule drug during the long antibody circulation time or rapid clearance from nonspecific interactions, to local tumor tissue heterogeneity, cell bystander effects, and endosomal escape. Mathematical models can be used to study the impact of these processes on overall distribution in an efficient manner, and several types of models have been used to analyze varying aspects of antibody distribution including physiologically based pharmacokinetic (PBPK) models and tissue-level simulations. However, these processes are quantitative in nature and cannot be handled qualitatively in isolation. For example, free antibody from deconjugation of the small molecule will impact the distribution of conjugated antibodies within the tumor. To incorporate these effects into a unified framework, we have coupled the systemic and organ-level distribution of a PBPK model with the tissue-level detail of a distributed parameter tumor model. We used this mathematical model to analyze new experimental results on the distribution of the clinical antibody-drug conjugate Kadcyla in HER2-positive mouse xenografts. This model is able to capture the impact of the drug-antibody ratio (DAR) on tumor penetration, the net result of drug deconjugation, and the effect of using unconjugated antibody to drive ADC penetration deeper into the tumor tissue. This modeling approach will provide quantitative and mechanistic support to experimental studies trying to parse the impact of multiple mechanisms of action for these complex drugs.
Collapse
Affiliation(s)
- Cornelius Cilliers
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Hans Guo
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Jianshan Liao
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Nikolas Christodolu
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA
| | - Greg M Thurber
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Rd., Ann Arbor, Michigan, 48109, USA. .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, 48109, USA.
| |
Collapse
|
38
|
N.Tinianow J, Pandya DN, Pailloux SL, Ogasawara A, Vanderbilt AN, Gill HS, Williams SP, Wadas TJ, Magda D, Marik J. Evaluation of a 3-hydroxypyridin-2-one (2,3-HOPO) Based Macrocyclic Chelator for (89)Zr(4+) and Its Use for ImmunoPET Imaging of HER2 Positive Model of Ovarian Carcinoma in Mice. Am J Cancer Res 2016; 6:511-21. [PMID: 26941844 PMCID: PMC4775861 DOI: 10.7150/thno.14261] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/05/2016] [Indexed: 11/15/2022] Open
Abstract
A novel octadentate 3-hydroxypyridin-2-one (2,3-HOPO) based di-macrocyclic ligand was evaluated for chelation of 89Zr; subsequently, it was used as a bi-functional chelator for preparation of 89Zr-labeled antibodies. Quantitative chelation of 89Zr4+ with the octadentate ligand forming 89ZrL complex was achieved under mild conditions within 15 minutes. The 89Zr-complex was stable in vitro in presence of DTPA, but a slow degradation was observed in serum. In vivo, the hydrophilic 89Zr-complex showed prevalently renal excretion; and an elevated bone uptake of radioactivity suggested a partial release of 89Zr4+ from the complex. The 2,3-HOPO based ligand was conjugated to the monoclonal antibodies, HER2-specific trastuzumab and an isotypic anti-gD antibody, using a p-phenylene bis-isothiocyanate linker to yield products with an average loading of less than 2 chelates per antibody. Conjugated antibodies were labeled with 89Zr under mild conditions providing the PET tracers in 60-69% yield. Despite the limited stability in mouse serum; the PET tracers performed very well in vivo. The PET imaging in mouse model of HER2 positive ovarian carcinoma showed tumor uptake of 89Zr-trastuzumab (29.2 ± 12.9 %ID/g) indistinguishable (p = 0.488) from the uptake of positive control 89Zr-DFO-trastuzumab (26.1 ± 3.3 %ID/g). In conclusion, the newly developed 3-hydroxypyridin-2-one based di-macrocyclic chelator provides a viable alternative to DFO-based heterobifunctional ligands for preparation of 89Zr-labeled monoclonal antibodies for immunoPET studies.
Collapse
|
39
|
Bispecific CD3/HER2 Targeting FynomAb Induces Redirected T Cell-Mediated Cytolysis with High Potency and Enhanced Tumor Selectivity. Antibodies (Basel) 2015. [DOI: 10.3390/antib4040426] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
40
|
Frei JC, Kielian M, Lai JR. Comprehensive mapping of functional epitopes on dengue virus glycoprotein E DIII for binding to broadly neutralizing antibodies 4E11 and 4E5A by phage display. Virology 2015; 485:371-82. [PMID: 26339794 DOI: 10.1016/j.virol.2015.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/04/2015] [Accepted: 08/12/2015] [Indexed: 11/16/2022]
Abstract
Here we investigated the binding of Dengue virus envelope glycoprotein domain III (DIII) by two broadly neutralizing antibodies (bNAbs), 4E11 and 4E5A. There are four serotypes of Dengue virus (DENV-1 to -4), whose DIII sequences vary by up to 49%. We used combinatorial alanine scanning mutagenesis, a phage display approach, to map functional epitopes (those residues that contribute most significantly to the energetics of antibody-antigen interaction) on these four serotypes. Our results showed that 4E11, which binds strongly to DENV-1, -2, and -3, and moderately to DENV-4, recognized a common conserved core functional epitope involving DIII residues K310, L/I387, L389, and W391. There were also unique recognition features for each serotype, suggesting that 4E11 has flexible recognition requirements. Similar scanning studies for the related bNAb 4E5A, which binds more tightly to DENV-4, identified broader functional epitopes on DENV-1. These results provide useful information for immunogen and therapeutic antibody design.
Collapse
Affiliation(s)
- Julia C Frei
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Margaret Kielian
- Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States.
| |
Collapse
|
41
|
Houlihan G, Gatti-Lafranconi P, Lowe D, Hollfelder F. Directed evolution of anti-HER2 DARPins by SNAP display reveals stability/function trade-offs in the selection process. Protein Eng Des Sel 2015; 28:269-79. [PMID: 26134501 PMCID: PMC4550541 DOI: 10.1093/protein/gzv029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 01/08/2023] Open
Abstract
In vitro display technologies have proved to be powerful tools for obtaining high-affinity protein binders. We recently described SNAP display, an entirely in vitro DNA display system that uses the SNAP-tag to link protein with its encoding DNA in water-in-oil emulsions. Here, we apply SNAP display for the affinity maturation of a designed ankyrin repeat proteins (DARPin) that binds to the extracellular domain of HER2 previously isolated by ribosome display. After four SNAP display selection cycles, proteins that bound specifically to HER2 in vitro, with dissociation constants in the low- to sub-nanomolar range, were isolated. In vitro affinities of the panel of evolved DARPins directly correlated with the fluorescence intensities of evolved DARPins bound to HER2 on a breast cancer cell line. A stability trade-off is observed as the most improved DARPins have decreased thermostability, when compared with the parent DARPin used as a starting point for affinity maturation. Dissection of the framework mutations of the highest affinity variant, DARPin F1, shows that functionally destabilising and compensatory mutations accumulated throughout the four rounds of evolution.
Collapse
Affiliation(s)
- Gillian Houlihan
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK MedImmune Ltd, Milstein Building, Granta Park, Cambridge CB1 6GH, UK
| | - Pietro Gatti-Lafranconi
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| | - David Lowe
- MedImmune Ltd, Milstein Building, Granta Park, Cambridge CB1 6GH, UK
| | - Florian Hollfelder
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK
| |
Collapse
|
42
|
Varela MA. Identification of sequences common to more than one therapeutic target to treat complex diseases: simulating the high variance in sequence interactivity evolved to modulate robust phenotypes. BMC Genomics 2015; 16:530. [PMID: 26187740 PMCID: PMC4506634 DOI: 10.1186/s12864-015-1727-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 06/26/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Genome-wide association studies show that most human traits and diseases are caused by a combination of environmental and genetic causes, with each one of these having a relatively small effect. In contrast, most therapies based on macromolecules like antibodies, antisense oligonucleotides or peptides focus on a single gene product. On the other hand, complex organisms seem to have a plethora of functional molecules able to bind specifically to multiple genes or genes products based on their sequences but the mechanisms that lead organisms to recruit these multispecific regulators remain unclear. RESULTS The mutational biases inferred from the genomic sequences of six organisms show an increase in the variance of sequence interactivity in complex organisms. The high variance in the interactivity of sequences presents an ideal evolutionary substrate to recruit sequence-specific regulators able to target multiple gene products. For example, here it is shown how the 3'UTR can fluctuate between sequences likely to be complementary to other sites in the genome in the search for advantageous interactions. A library of nucleotide- and peptide-based tools was built using a script to search for candidates (e.g. peptides, antigens to raise antibodies or antisense oligonucleotides) to target sequences shared by key pathways in human disorders, such as cancer and immune diseases. This resource will be accessible to the community at www.wikisequences.org . CONCLUSIONS This study describes and encourages the adoption of the same multitarget strategy (e.g., miRNAs, Hsp90) that has evolved in organisms to modify complex traits to treat diseases with robust pathological phenotypes. The increase in the variance of sequence interactivity detected in the human and mouse genomes when compared with less complex organisms could have expedited the evolution of regulators able to interact to multiple gene products and modulate robust phenotypes. The identification of sequences common to more than one therapeutic target carried out in this study could facilitate the design of new multispecific methods able to modify simultaneously key pathways to treat complex diseases.
Collapse
Affiliation(s)
- Miguel Angel Varela
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| |
Collapse
|
43
|
Koenig P, Lee CV, Sanowar S, Wu P, Stinson J, Harris SF, Fuh G. Deep Sequencing-guided Design of a High Affinity Dual Specificity Antibody to Target Two Angiogenic Factors in Neovascular Age-related Macular Degeneration. J Biol Chem 2015; 290:21773-86. [PMID: 26088137 DOI: 10.1074/jbc.m115.662783] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Indexed: 11/06/2022] Open
Abstract
The development of dual targeting antibodies promises therapies with improved efficacy over mono-specific antibodies. Here, we engineered a Two-in-One VEGF/angiopoietin 2 antibody with dual action Fab (DAF) as a potential therapeutic for neovascular age-related macular degeneration. Crystal structures of the VEGF/angiopoietin 2 DAF in complex with its two antigens showed highly overlapping binding sites. To achieve sufficient affinity of the DAF to block both angiogenic factors, we turned to deep mutational scanning in the complementarity determining regions (CDRs). By mutating all three CDRs of each antibody chain simultaneously, we were able not only to identify affinity improving single mutations but also mutation pairs from different CDRs that synergistically improve both binding functions. Furthermore, insights into the cooperativity between mutations allowed us to identify fold-stabilizing mutations in the CDRs. The data obtained from deep mutational scanning reveal that the majority of the 52 CDR residues are utilized differently for the two antigen binding function and permit, for the first time, the engineering of several DAF variants with sub-nanomolar affinity against two structurally unrelated antigens. The improved variants show similar blocking activity of receptor binding as the high affinity mono-specific antibodies against these two proteins, demonstrating the feasibility of generating a dual specificity binding surface with comparable properties to individual high affinity mono-specific antibodies.
Collapse
Affiliation(s)
| | | | | | - Ping Wu
- Structural Biology, Genentech Research and Early Development, South San Francisco, California 94080
| | | | - Seth F Harris
- Structural Biology, Genentech Research and Early Development, South San Francisco, California 94080
| | | |
Collapse
|
44
|
Study of the interaction of trastuzumab and SKOV3 epithelial cancer cells using a quartz crystal microbalance sensor. SENSORS 2015; 15:5884-94. [PMID: 25763651 PMCID: PMC4435181 DOI: 10.3390/s150305884] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/31/2014] [Accepted: 03/02/2015] [Indexed: 12/20/2022]
Abstract
Analytical methods founded upon whole cell-based assays are of importance in early stage drug development and in fundamental studies of biomolecular recognition. Here we have studied the binding of the monoclonal antibody trastuzumab to human epidermal growth factor receptor 2 (HER2) on human ovary adenocarcinoma epithelial cancer cells (SKOV3) using quartz crystal microbalance (QCM) technology. An optimized procedure for immobilizing the cells on the chip surface was established with respect to fixation procedure and seeding density. Trastuzumab binding to the cell decorated sensor surface was studied, revealing a mean dissociation constant, KD, value of 7 ± 1 nM (standard error of the mean). This study provides a new perspective on the affinity of the antibody-receptor complex presented a more natural context compared to purified receptors. These results demonstrate the potential for using whole cell-based QCM assay in drug development, the screening of HER2 selective antibody-based drug candidates, and for the study of biomolecular recognition. This real time, label free approach for studying interactions with target receptors present in their natural environment afforded sensitive and detailed kinetic information about the binding of the analyte to the target.
Collapse
|
45
|
Kieber-Emmons T, Saha S, Pashov A, Monzavi-Karbassi B, Murali R. Carbohydrate-mimetic peptides for pan anti-tumor responses. Front Immunol 2014; 5:308. [PMID: 25071769 PMCID: PMC4075079 DOI: 10.3389/fimmu.2014.00308] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/17/2014] [Indexed: 11/26/2022] Open
Abstract
Molecular mimicry is fundamental to biology and transcends to many disciplines ranging from immune pathology to drug design. Structural characterization of molecular partners has provided insight into the origins and relative importance of complementarity in mimicry. Chemical complementarity is easy to understand; amino acid sequence similarity between peptides, for example, can lead to cross-reactivity triggering similar reactivity from their cognate receptors. However, conformational complementarity is difficult to decipher. Molecular mimicry of carbohydrates by peptides is often considered one of those. Extensive studies of innate and adaptive immune responses suggests the existence of carbohydrate mimicry, but the structural basis for this mimicry yields confounding details; peptides mimicking carbohydrates in some cases fail to exhibit both chemical and conformational mimicry. Deconvolution of these two types of complementarity in mimicry and its relationship to biological function can nevertheless lead to new therapeutics. Here, we discuss our experience examining the immunological aspects and implications of carbohydrate-peptide mimicry. Emphasis is placed on the rationale, the lessons learned from the methodologies to identify mimics, a perspective on the limitations of structural analysis, the biological consequences of mimicking tumor-associated carbohydrate antigens, and the notion of reverse engineering to develop carbohydrate-mimetic peptides in vaccine design strategies to induce responses to glycan antigens expressed on cancer cells.
Collapse
Affiliation(s)
- Thomas Kieber-Emmons
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Somdutta Saha
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anastas Pashov
- Stephan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Behjatolah Monzavi-Karbassi
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ramachandran Murali
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
46
|
Schanzer JM, Wartha K, Croasdale R, Moser S, Künkele KP, Ries C, Scheuer W, Duerr H, Pompiati S, Pollman J, Stracke J, Lau W, Ries S, Brinkmann U, Klein C, Umana P. A novel glycoengineered bispecific antibody format for targeted inhibition of epidermal growth factor receptor (EGFR) and insulin-like growth factor receptor type I (IGF-1R) demonstrating unique molecular properties. J Biol Chem 2014; 289:18693-706. [PMID: 24841203 DOI: 10.1074/jbc.m113.528109] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In the present study, we have developed a novel one-arm single chain Fab heterodimeric bispecific IgG (OAscFab-IgG) antibody format targeting the insulin-like growth factor receptor type I (IGF-1R) and the epidermal growth factor receptor (EGFR) with one binding site for each target antigen. The bispecific antibody XGFR is based on the "knob-into-hole" technology for heavy chain heterodimerization with one heavy chain consisting of a single chain Fab to prevent wrong pairing of light chains. XGFR was produced with high expression yields and showed simultaneous binding to IGF-1R and EGFR with high affinity. Due to monovalent binding of XGFR to IGF-1R, IGF-1R internalization was strongly reduced compared with the bivalent parental antibody, leading to enhanced Fc-mediated cellular cytotoxicity. To further increase immune effector functions triggered by XGFR, the Fc portion of the bispecific antibody was glycoengineered, which resulted in strong antibody-dependent cell-mediated cytotoxicity activity. XGFR-mediated inhibition of IGF-1R and EGFR phosphorylation as well as A549 tumor cell proliferation was highly effective and was comparable with a combined treatment with EGFR (GA201) and IGF-1R (R1507) antibodies. XGFR also demonstrated potent anti-tumor efficacy in multiple mouse xenograft tumor models with a complete growth inhibition of AsPC1 human pancreatic tumors and improved survival of SCID beige mice carrying A549 human lung tumors compared with treatment with antibodies targeting either IGF-1R or EGFR. In summary, we have applied rational antibody engineering technology to develop a heterodimeric OAscFab-IgG bispecific antibody, which combines potent signaling inhibition with antibody-dependent cell-mediated cytotoxicity induction and results in superior molecular properties over two established tetravalent bispecific formats.
Collapse
Affiliation(s)
| | | | | | - Samuel Moser
- Roche Glycart AG, CH-8952 Schlieren, Switzerland, and
| | | | | | | | - Harald Duerr
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Sandra Pompiati
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Jan Pollman
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Jan Stracke
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Wilma Lau
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Stefan Ries
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | - Ulrich Brinkmann
- Large Molecule Research, Pharmaceutical Research and Early Development, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany
| | | | - Pablo Umana
- Roche Glycart AG, CH-8952 Schlieren, Switzerland, and
| |
Collapse
|
47
|
Lee CV, Koenig P, Fuh G. A two-in-one antibody engineered from a humanized interleukin 4 antibody through mutation in heavy chain complementarity-determining regions. MAbs 2014; 6:622-7. [PMID: 24618680 DOI: 10.4161/mabs.28483] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A mono-specific antibody may recruit a second antigen binding specificity, thus converting to a dual-specific Two-in-One antibody through mutation at the light chain complementarity-determining regions (CDRs). It is, however, unknown whether mutation at the heavy chain CDRs may evolve such dual specificity. Herein, we examined the CDRs of a humanized interleukin 4 (IL4) antibody using alanine scanning and structural modeling, designed libraries of mutants in regions that tolerate mutation, and isolated dual specific antibodies harboring mutation at the heavy chain CDRs only. We then affinity improved an IL4/IL5 dual specific antibody to variants with dissociation constants in the low nanomolar range for both antigens. The results demonstrate the full capacity of antibodies to evolve dual binding specificity.
Collapse
Affiliation(s)
- Chingwei V Lee
- Department of Antibody Engineering; Genentech Inc.; South San Francisco, CA USA
| | - Patrick Koenig
- Department of Antibody Engineering; Genentech Inc.; South San Francisco, CA USA
| | - Germaine Fuh
- Department of Antibody Engineering; Genentech Inc.; South San Francisco, CA USA
| |
Collapse
|
48
|
Liu P, Cai Z, Kang JW, Boyle AJ, Adams J, Lu Y, Ngo Ndjock Mbong G, Sidhu S, Reilly RM, Winnik MA. Intracellular routing in breast cancer cells of streptavidin-conjugated trastuzumab Fab fragments linked to biotinylated doxorubicin-functionalized metal chelating polymers. Biomacromolecules 2014; 15:715-25. [PMID: 24506198 DOI: 10.1021/bm401483a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We describe the synthesis of a heterotelechelic metal-chelating polymer (Bi-MCP-Dox), a polyacrylamide with a number average degree of polymerization DPn = 50 (PDI = 1.2), with biotin (Bi) and doxorubicin (Dox) as functional chain ends and diethylenetriaminepentaacetic acid (DTPA) pendant groups as the binding sites for metal ions. We compared its behavior in cell-uptake experiments with a similar polymer (Bi-MCP) without Dox. These MCPs were complexed with trastuzumab Fab (tmFab) fragments covalently linked to streptavidin (SAv) to form tmFab-SAv-Bi-MCP-Dox and tmFab-SAv-Bi-MCP via the strong affinity between Bi and SAv. tmFab targets human epidermal growth factor receptor-2 (HER2), which is overexpressed on certain human breast cancer cells. Surface plasmon resonance (SPR) experiments with the extracellular domain (ECD) of HER2 showed that incorporation of the MCPs in these complexes had no significant effect on the association or dissociation rate with the HER2 ECD and the dissociation constants. The tmFab-complexed MCPs were subsequently labeled with (111)In (an Auger electron emitting radionuclide). Auger electrons can cause lethal DNA double strand breaks (DSBs) but only if they are emitted intracellularly and especially, in close proximity to the nucleus. To evaluate the cellular and nuclear uptake of tmFab-SAv-Bi-MCP-Dox, we incubated HER2+ SK-BR-3 human breast cancer cells with the complexes saturated with stable In(3+) and visualized their distribution by confocal fluorescence microscopy, monitoring the fluorescence of Dox. In parallel, we carried out cell fractionation studies on tmFab-SAv-Bi-MCP-Dox and on tmFab-SAv-Bi-MCP labeled with (111)In. Both radiolabeled complexes showed cell internalization and nuclear localization. We conclude that metal-chelating polymers with this composition appear to encourage internalization, nuclear uptake, and chromatin (DNA) binding of trastuzumab fragments modified with streptavidin in human breast cancer cells expressing HER2. Further study is needed to understand the impact of polymer charge on cellular uptake and distribution to intracellular compartments.
Collapse
Affiliation(s)
- Peng Liu
- Department of Chemistry, University of Toronto , 80 St. George Street, Toronto, Ontario, Canada , M5S 3H6
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Li L, Gardner I, Rose R, Jamei M. Incorporating Target Shedding Into a Minimal PBPK-TMDD Model for Monoclonal Antibodies. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e96. [PMID: 24477089 PMCID: PMC3910015 DOI: 10.1038/psp.2013.73] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 11/12/2013] [Indexed: 11/09/2022]
Abstract
Shedding of a pharmacological target from cells, giving rise to a soluble target that can also bind therapeutic proteins, is a common phenomenon. In this study, a minimal physiologically based pharmacokinetic model was used to simulate the pharmacokinetics of trastuzumab and the simultaneous binding of the compound to soluble (in blood and tissue interstitial space) and membrane-bound (in the tissue interstitial space) forms of human epidermal growth factor receptor 2 (HER2). The parameter values describing binding of trastuzumab to HER2 were largely derived from in vitro data, and the effects of varying HER2 levels, the affinity difference between membrane-bound HER2 and shed antigen, and slow binding kinetics were investigated. The model simulates a sharp decrease in trough drug concentrations at concentrations of soluble target between 500 and 1,000 ng/ml in plasma. This corresponds with the clinical concentration range of soluble target wherein changes in half-life of trastuzumab have been observed.
Collapse
Affiliation(s)
- L Li
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, UK
| | - I Gardner
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, UK
| | - R Rose
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, UK
| | - M Jamei
- Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, UK
| |
Collapse
|
50
|
Understanding the role of cross-arm binding efficiency in the activity of monoclonal and multispecific therapeutic antibodies. Methods 2014; 65:95-104. [DOI: 10.1016/j.ymeth.2013.07.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 01/09/2023] Open
|