51
|
Asano R, Shimomura I, Konno S, Ito A, Masakari Y, Orimo R, Taki S, Arai K, Ogata H, Okada M, Furumoto S, Onitsuka M, Omasa T, Hayashi H, Katayose Y, Unno M, Kudo T, Umetsu M, Kumagai I. Rearranging the domain order of a diabody-based IgG-like bispecific antibody enhances its antitumor activity and improves its degradation resistance and pharmacokinetics. MAbs 2014; 6:1243-54. [PMID: 25517309 PMCID: PMC4623410 DOI: 10.4161/mabs.29445] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
One approach to creating more beneficial therapeutic antibodies is to develop bispecific antibodies (bsAbs), particularly IgG-like formats with tetravalency, which may provide several advantages such as multivalent binding to each target antigen. Although the effects of configuration and antibody-fragment type on the function of IgG-like bsAbs have been studied, there have been only a few detailed studies of the influence of the variable fragment domain order. Here, we prepared four types of hEx3-scDb-Fc, IgG-like bsAbs, built from a single-chain hEx3-Db (humanized bispecific diabody [bsDb] that targets epidermal growth factor receptor and CD3), to investigate the influence of domain order and fusion manner on the function of a bsDb with an Fc fusion format. Higher cytotoxicities were observed with hEx3-scDb-Fcs with a variable light domain (VL)-variable heavy domain (VH) order (hEx3-scDb-Fc-LHs) compared with a VH-VL order, indicating that differences in the Fc fusion manner do not affect bsDb activity. In addition, flow cytometry suggested that the higher cytotoxicities of hEx3-scDb-Fc-LH may be attributable to structural superiority in cross-linking. Interestingly, enhanced degradation resistance and prolonged in vivo half-life were also observed with hEx3-scDb-Fc-LH. hEx3-scDb-Fc-LH and its IgG2 variant exhibited intense in vivo antitumor effects, suggesting that Fc-mediated effector functions are dispensable for effective anti-tumor activities, which may cause fewer side effects. Our results show that merely rearranging the domain order of IgG-like bsAbs can enhance not only their antitumor activity, but also their degradation resistance and in vivo half-life, and that hEx3-scDb-Fc-LHs are potent candidates for next-generation therapeutic antibodies.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- AUC, area-under-the-curve
- CD3
- EGFR, epidermal growth factor receptor
- FITC-CD3ϵγ, fluorescein isothiocyanate-labeled CD3ϵγ; DVD-IgTM, dual variable domain immunoglobulin
- FITC-sEGFR, FITC-labeled sEGFR
- Fv, variable fragment
- ICR, imprinting control region
- IgG-like bispecific antibody
- MTS, 3-(4, 5-dimethylthiazole-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt
- PBMCs, peripheral blood mononuclear cells
- PBS, phosphate-buffered saline
- SDS-PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis
- SPR, surface plasmon resonance
- SUV, standardized uptake value
- T-LAK cells, lymphokine-activated killer cells with the T-cell phenotype
- VH, variable heavy domain
- VL, variable light domain
- antibody engineering
- bispecific diabody
- bsAb, bispecific antibody
- bsDb, bispecific diabody
- cancer immunotherapy
- effective domain order
- epidermal growth factor receptor
- sEGFR, soluble EGFR
- scDb, single-chain diabody
- scFv, single-chain Fv
- taFv, tandem scFv
Collapse
Affiliation(s)
- Ryutaro Asano
- a Department of Biomolecular Engineering ; Graduate School of Engineering; Tohoku University ; Sendai , Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Cheng M, Ahmed M, Xu H, Cheung NKV. Structural design of disialoganglioside GD2 and CD3-bispecific antibodies to redirect T cells for tumor therapy. Int J Cancer 2014; 136:476-86. [PMID: 24895182 DOI: 10.1002/ijc.29007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 04/21/2014] [Accepted: 05/08/2014] [Indexed: 02/02/2023]
Abstract
Antibody-based immunotherapy has proven efficacy for patients with high-risk neuroblastoma. However, despite being the most efficient tumoricidal effectors, T cells are underutilized because they lack Fc receptors. Using a monovalent single-chain fragment (ScFv) platform, we engineered tandem scFv bispecific antibodies (BsAbs) that specifically target disialoganglioside (GD2) on tumor cells and CD3 on T cells. Structural variants of BsAbs were constructed and ranked based on binding to GD2, and on competency in inducing T-cell-mediated tumor cytotoxicity. In vitro thermal stability and binding measurements were used to characterize each of the constructs, and in silico molecular modeling was used to show how the orientation of the variable region heavy (VH) and light (VL) chains of the anti-GD2 ScFv could alter the conformations of key residues responsible for high affinity binding. We showed that the VH-VL orientation, the (GGGGS)3 linker, disulfide bond stabilization of scFv, when combined with an affinity matured mutation provided the most efficient BsAb to direct T cells to lyse GD2-positive tumor cells. In vivo, the optimized BsAb could efficiently inhibit melanoma and neuroblastoma xenograft growth. These findings provide preclinical validation of a structure-based method to assist in designing BsAb for T-cell-mediated therapy.
Collapse
Affiliation(s)
- Ming Cheng
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | |
Collapse
|
53
|
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: 3.9] [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
|
54
|
Construction and production of an IgG-Like tetravalent bispecific antibody, IgG-single-chain Fv fusion. Methods Mol Biol 2014; 1060:185-213. [PMID: 24037843 DOI: 10.1007/978-1-62703-586-6_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
In recent years, both laboratory and clinical studies have demonstrated that bispecific antibodies (BsAbs) may have significant potential application in cancer therapy either by targeting tumor cells with cytotoxic agents including effector cells, radionuclides, drugs, and toxins, or by simultaneously blocking two tumor-associated targets, e.g., tumor growth factors and/or their cell surface receptors. A major obstacle in the development of BsAb has been the difficulty of producing the materials in sufficient quality and quantity by traditional technologies such as the hybrid hybridoma and chemical conjugation methods. The development of recombinant BsAbs as therapeutic agents will depend heavily on the advances made in the design of the constructs (or formats) and production efficiency. Here we describe a recombinant method for the construction and production of a tetravalent IgG-like BsAb molecule, IgG-scFv fusion, in which, a single-chain Fv (scFv) antibody fragment of one antigen specificity is genetically fused to the c-terminal of a conventional IgG of a different antigen specificity.
Collapse
|
55
|
Generation of bispecific IgG antibodies by structure-based design of an orthogonal Fab interface. Nat Biotechnol 2014; 32:191-8. [PMID: 24463572 DOI: 10.1038/nbt.2797] [Citation(s) in RCA: 198] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 12/10/2013] [Indexed: 01/22/2023]
Abstract
Robust generation of IgG bispecific antibodies has been a long-standing challenge. Existing methods require extensive engineering of each individual antibody, discovery of common light chains, or complex and laborious biochemical processing. Here we combine computational and rational design approaches with experimental structural validation to generate antibody heavy and light chains with orthogonal Fab interfaces. Parental monoclonal antibodies incorporating these interfaces, when simultaneously co-expressed, assemble into bispecific IgG with improved heavy chain-light chain pairing. Bispecific IgGs generated with this approach exhibit pharmacokinetic and other desirable properties of native IgG, but bind target antigens monovalently. As such, these bispecific reagents may be useful in many biotechnological applications.
Collapse
|
56
|
Müller MR, Saunders K, Grace C, Jin M, Piche-Nicholas N, Steven J, O'Dwyer R, Wu L, Khetemenee L, Vugmeyster Y, Hickling TP, Tchistiakova L, Olland S, Gill D, Jensen A, Barelle CJ. Improving the pharmacokinetic properties of biologics by fusion to an anti-HSA shark VNAR domain. MAbs 2014; 4:673-85. [PMID: 23676205 DOI: 10.4161/mabs.22242] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Advances in recombinant antibody technology and protein engineering have provided the opportunity to reduce antibodies to their smallest binding domain components and have concomitantly driven the requirement for devising strategies to increase serum half-life to optimise drug exposure, thereby increasing therapeutic efficacy. In this study, we adopted an immunization route to raise picomolar affinity shark immunoglobulin new antigen receptors (IgNARs) to target human serum albumin (HSA). From our model shark species, Squalus acanthias, a phage display library encompassing the variable binding domain of IgNAR (VNAR) was constructed, screened against target, and positive clones were characterized for affinity and specificity. N-terminal and C-terminal molecular fusions of our lead hit in complex with a naïve VNAR domain were expressed, purified and exhibited the retention of high affinity binding to HSA, but also cross-selectivity to mouse, rat and monkey serum albumin both in vitro and in vivo. Furthermore, the naïve VNAR had enhanced pharmacokinetic (PK) characteristics in both N- and C-terminal orientations and when tested as a three domain construct with naïve VNAR flanking the HSA binding domain at both the N and C termini. Molecules derived from this platform technology also demonstrated the potential for clinical utility by being available via the subcutaneous route of delivery. This study thus demonstrates the first in vivo functional efficacy of a VNAR binding domain with the ability to enhance PK properties and support delivery of multifunctional therapies.
Collapse
|
57
|
Gu J, Ghayur T. Rationale and development of multispecific antibody drugs. Expert Rev Clin Pharmacol 2014; 3:491-508. [DOI: 10.1586/ecp.10.28] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
58
|
Spencer D, Novarra S, Zhu L, Mugabe S, Thisted T, Baca M, Depaz R, Barton C. O-xylosylation in a Recombinant Protein is Directed at a Common Motif on Glycine–Serine Linkers. J Pharm Sci 2013; 102:3920-4. [DOI: 10.1002/jps.23733] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/30/2013] [Accepted: 09/03/2013] [Indexed: 11/10/2022]
|
59
|
Lee CC, Perchiacca JM, Tessier PM. Toward aggregation-resistant antibodies by design. Trends Biotechnol 2013; 31:612-20. [DOI: 10.1016/j.tibtech.2013.07.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/30/2013] [Accepted: 07/05/2013] [Indexed: 12/19/2022]
|
60
|
A tale of two specificities: bispecific antibodies for therapeutic and diagnostic applications. Trends Biotechnol 2013; 31:621-32. [PMID: 24094861 PMCID: PMC7114091 DOI: 10.1016/j.tibtech.2013.08.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/01/2013] [Accepted: 08/27/2013] [Indexed: 12/20/2022]
Abstract
Recombinant DNA technologies are leading the rapid expansion of bispecific antibody formats. The therapeutic potential of bispecific antibodies is being realized through creative design. Bispecific antibodies are potentially underutilized reagents for diagnostics.
Artificial manipulation of antibody genes has facilitated the production of several unique recombinant antibody formats, which have highly important therapeutic and biotechnological applications. Although bispecific antibodies (bsAbs) are not new, they are coming to the forefront as our knowledge of the potential efficacy of antibody-based therapeutics expands. The next generation of bsAbs is developing due to significant improvements in recombinant antibody technologies. This review focuses on recent advances with a particular focus on improvements in format and design that are contributing to the resurgence of bsAbs, and in particular, on innovative structures applicable to next generation point-of-care (POC) devices with applicability to low resource environments.
Collapse
|
61
|
Fennell BJ, McDonnell B, Tam ASP, Chang L, Steven J, Broadbent ID, Gao H, Kieras E, Alley J, Luxenberg D, Edmonds J, Fitz LJ, Miao W, Whitters MJ, Medley QG, Guo YJ, Darmanin-Sheehan A, Autin B, Shúilleabháin DN, Cummins E, King A, Krebs MRH, Grace C, Hickling TP, Boisvert A, Zhong X, McKenna M, Francis C, Olland S, Bloom L, Paulsen J, Somers W, Jensen A, Lin L, Finlay WJJ, Cunningham O. CDR-restricted engineering of native human scFvs creates highly stable and soluble bifunctional antibodies for subcutaneous delivery. MAbs 2013; 5:882-95. [PMID: 23995618 PMCID: PMC3896602 DOI: 10.4161/mabs.26201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
While myriad molecular formats for bispecific antibodies have been examined to date, the simplest structures are often based on the scFv. Issues with stability and manufacturability in scFv-based bispecific molecules, however, have been a significant hindrance to their development, particularly for high-concentration, stable formulations that allow subcutaneous delivery. Our aim was to generate a tetravalent bispecific molecule targeting two inflammatory mediators for synergistic immune modulation. We focused on an scFv-Fc-scFv format, with a flexible (A4T)3 linker coupling an additional scFv to the C-terminus of an scFv-Fc. While one of the lead scFvs isolated directly from a naïve library was well-behaved and sufficiently potent, the parental anti-CXCL13 scFv 3B4 required optimization for affinity, stability, and cynomolgus ortholog cross-reactivity. To achieve this, we eschewed framework-based stabilizing mutations in favor of complementarity-determining region (CDR) mutagenesis and re-selection for simultaneous improvements in both affinity and thermal stability. Phage-displayed 3B4 CDR-mutant libraries were used in an aggressive "hammer-hug" selection strategy that incorporated thermal challenge, functional, and biophysical screening. This approach identified leads with improved stability and>18-fold, and 4,100-fold higher affinity for both human and cynomolgus CXCL13, respectively. Improvements were exclusively mediated through only 4 mutations in VL-CDR3. Lead scFvs were reformatted into scFv-Fc-scFvs and their biophysical properties ranked. Our final candidate could be formulated in a standard biopharmaceutical platform buffer at 100 mg/ml with<2% high molecular weight species present after 7 weeks at 4 °C and viscosity<15 cP. This workflow has facilitated the identification of a truly manufacturable scFv-based bispecific therapeutic suitable for subcutaneous administration.
Collapse
Affiliation(s)
- Brian J Fennell
- Pfizer; Global Biotherapeutics Technologies; Dublin, Ireland
| | - Barry McDonnell
- Pfizer; Global Biotherapeutics Technologies; Dublin, Ireland
| | - Amy Sze Pui Tam
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Lijun Chang
- Pfizer; Global Biotherapeutics Technologies, Foresterhill; Aberdeen, UK
| | - John Steven
- Pfizer; Global Biotherapeutics Technologies, Foresterhill; Aberdeen, UK
| | - Ian D Broadbent
- Pfizer; Global Biotherapeutics Technologies, Foresterhill; Aberdeen, UK
| | - Huilan Gao
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | | | | | | | | | | | | | | | | | - Yongjing J Guo
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | | | - Bénédicte Autin
- Pfizer; Global Biotherapeutics Technologies; Dublin, Ireland
| | | | - Emma Cummins
- Pfizer; Global Biotherapeutics Technologies; Dublin, Ireland
| | - Amy King
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Mark R H Krebs
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | | | | | - Angela Boisvert
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Xiaotian Zhong
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Matthew McKenna
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | | | - Stephane Olland
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Laird Bloom
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Janet Paulsen
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Will Somers
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Allan Jensen
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | - Laura Lin
- Pfizer; Global Biotherapeutics Technologies; Cambridge, MA USA
| | | | - Orla Cunningham
- Pfizer; Global Biotherapeutics Technologies; Dublin, Ireland
| |
Collapse
|
62
|
Fournier P, Schirrmacher V. Bispecific antibodies and trispecific immunocytokines for targeting the immune system against cancer: preparing for the future. BioDrugs 2013; 27:35-53. [PMID: 23329400 DOI: 10.1007/s40259-012-0008-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Monoclonal anti-tumor antibodies (mAbs) that are clinically effective usually recruit, via their constant fragment (Fc) domain, Fc receptor (FcR)-positive accessory cells of the immune system and engage these additionally against the tumor. Since T cells are FcR negative, these important cells are not getting involved. In contrast to mAbs, bispecific antibodies (bsAbs) can be designed in such a way that they involve T cells. bsAbs are artificially designed molecules that bind simultaneously to two different antigens, one on the tumor cell, the other one on an immune effector cell such as CD3 on T cells. Such dual antibody constructs can cross-link tumor cells and T cells. Many such bsAb molecules at the surface of tumor cells can thus build a bridge to T cells and aggregate their CD3 molecules, thereby activating them for cytotoxic activity. BsAbs can also contain a third binding site, for instance a Fc domain or a cytokine that would bind to its respective cytokine receptor. The present review discusses the pros and cons for the use of the Fc fragment during the development of bsAbs using either cell-fusion or recombinant DNA technologies. The recombinant antibody technology allows the generation of very efficient bsAbs containing no Fc domain such as the bi-specific T-cell engager (BiTE). The strong antitumor activity of these molecules makes them very interesting new cancer therapeutics. Over the last decade, we have developed another concept, namely to combine bsAbs and multivalent immunocytokines with a tumor cell vaccine. The latter are patient-derived tumor cells modified by infection with a virus. The virus-Newcastle Disease Virus (NDV)-introduces, at the surface of the tumor cells, viral molecules that can serve as general anchors for the bsAbs. Our strategy aims at redirecting, in an Fc-independent fashion, activities of T cells and accessory cells against autologous tumor antigens. It creates very promising perspectives for a new generation of efficient and safe cancer therapeutics that should confer long-lasting anti-tumor immunity.
Collapse
Affiliation(s)
- Philippe Fournier
- German Cancer Research Center DKFZ, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany,
| | | |
Collapse
|
63
|
Biological Insights into Therapeutic Protein Modifications throughout Trafficking and Their Biopharmaceutical Applications. Int J Cell Biol 2013; 2013:273086. [PMID: 23690780 PMCID: PMC3652174 DOI: 10.1155/2013/273086] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/20/2013] [Indexed: 12/16/2022] Open
Abstract
Over the lifespan of therapeutic proteins, from the point of biosynthesis to the complete clearance from tested subjects, they undergo various biological modifications. Therapeutic influences and molecular mechanisms of these modifications have been well appreciated for some while remained less understood for many. This paper has classified these modifications into multiple categories, according to their processing locations and enzymatic involvement during the trafficking events. It also focuses on the underlying mechanisms and structural-functional relationship between modifications and therapeutic properties. In addition, recent advances in protein engineering, cell line engineering, and process engineering, by exploring these complex cellular processes, are discussed and summarized, for improving functional characteristics and attributes of protein-based biopharmaceutical products.
Collapse
|
64
|
Proctor EA, Kota P, Demarest SJ, Caravella JA, Dokholyan NV. Highly covarying residues have a functional role in antibody constant domains. Proteins 2013; 81:884-95. [PMID: 23280585 DOI: 10.1002/prot.24247] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 12/05/2012] [Accepted: 12/14/2012] [Indexed: 01/25/2023]
Abstract
The ability to generate and design antibodies recognizing specific targets has revolutionized the pharmaceutical industry and medical imaging. Engineering antibody therapeutics in some cases requires modifying their constant domains to enable new and altered interactions. Engineering novel specificities into antibody constant domains has proved challenging due to the complexity of inter-domain interactions. Covarying networks of residues that tend to cluster on the protein surface and near binding sites have been identified in some proteins. However, the underlying role these networks play in the protein resulting in their conservation remains unclear in most cases. Resolving their role is crucial, because residues in these networks are not viable design targets if their role is to maintain the fold of the protein. Conversely, these networks of residues are ideal candidates for manipulating specificity if they are primarily involved in binding, such as the myriad interdomain interactions maintained within antibodies. Here, we identify networks of evolutionarily-related residues in C-class antibody domains by evaluating covariation, a measure of propensity with which residue pairs vary dependently during evolution. We computationally test whether mutation of residues in these networks affects stability of the folded antibody domain, determining their viability as design candidates. We find that members of covarying networks cluster at domain-domain interfaces, and that mutations to these residues are diverse and frequent during evolution, precluding their importance to domain stability. These results indicate that networks of covarying residues exist in antibody domains for functional reasons unrelated to thermodynamic stability, making them ideal targets for antibody design.
Collapse
Affiliation(s)
- Elizabeth A Proctor
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, North Carolina 27599-7260, USA
| | | | | | | | | |
Collapse
|
65
|
Xu L, Kohli N, Rennard R, Jiao Y, Razlog M, Zhang K, Baum J, Johnson B, Tang J, Schoeberl B, Fitzgerald J, Nielsen U, Lugovskoy AA. Rapid optimization and prototyping for therapeutic antibody-like molecules. MAbs 2013; 5:237-54. [PMID: 23392215 PMCID: PMC3893234 DOI: 10.4161/mabs.23363] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Multispecific antibody-like molecules have the potential to advance the standard-of-care in many human diseases. The design of therapeutic molecules in this class, however, has proven to be difficult and, despite significant successes in preclinical research, only one trivalent antibody, catumaxomab, has demonstrated clinical utility. The challenge originates from the complexity of the design space where multiple parameters such as affinity, avidity, effector functions, and pharmaceutical properties need to be engineered in concurrent fashion to achieve the desired therapeutic efficacy. Here, we present a rapid prototyping approach that allows us to successfully optimize these parameters within one campaign cycle that includes modular design, yeast display of structure focused antibody libraries and high throughput biophysical profiling. We delineate this approach by presenting a design case study of MM-141, a tetravalent bispecific antibody targeting two compensatory signaling growth factor receptors: insulin-like growth factor 1 receptor (IGF-1R) and v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (ErbB3). A MM-141 proof-of-concept (POC) parent molecule did not meet initial design criteria due to modest bioactivity and poor stability properties. Using a combination of yeast display, structured-guided antibody design and library-scale thermal challenge assay, we discovered a diverse set of stable and active anti-IGF-1R and anti-ErbB3 single-chain variable fragments (scFvs). These optimized modules were reformatted to create a diverse set of full-length tetravalent bispecific antibodies. These re-engineered molecules achieved complete blockade of growth factor induced pro-survival signaling, were stable in serum, and had adequate activity and pharmaceutical properties for clinical development. We believe this approach can be readily applied to the optimization of other classes of bispecific or even multispecific antibody-like molecules.
Collapse
Affiliation(s)
- Lihui Xu
- Merrimack Pharmaceuticals, Inc. Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Weatherill EE, Cain KL, Heywood SP, Compson JE, Heads JT, Adams R, Humphreys DP. Towards a universal disulphide stabilised single chain Fv format: importance of interchain disulphide bond location and vL-vH orientation. Protein Eng Des Sel 2012; 25:321-9. [PMID: 22586154 DOI: 10.1093/protein/gzs021] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Engineered introduction of interface interchain disulphide bonds is perceived to be a simple method to increase the stability of single chain Fv (scFv). Six disulphide bond locations have been cited within the literature but the potential for the broad use of each has not been examined. Five of these disulphide bond locations were introduced into one scFv in order to compare their relative effects on expression, thermal stability, percent monomer formation and retention of antigen binding. The disulphide bond position vH44-vL100 was observed to enable the most favourable balance of biophysical properties. The vH44-vL100 disulphide bond was introduced into five additional scFv in both vL-vH and vH-vL orientations in order to investigate its general applicability. Data are presented to show the relative influence of scFv sequence, v-region organisation and interchain disulphide bond on expression yield, thermal stability and percent monomer. Introduction of the vH44-vL100 disulphide bond typically resulted in no or little increase in thermal stability and no change in percent monomer but did confer the benefit of permanently fixing monomer:dimer ratios during purification and analysis.
Collapse
Affiliation(s)
- Eve E Weatherill
- Protein Expression and Purification Group, UCB, Slough, Berkshire SL1 3WE, England
| | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
The advent of modern antibody engineering has led to numerous successes in the application of these proteins for cancer therapy in the 13 years since the first Food and Drug Administration approval, which has stimulated active interest in developing more and better drugs based on these molecules. A wide range of tools for discovering and engineering antibodies has been brought to bear on this challenge in the past two decades. Here, we summarize mechanisms of monoclonal antibody therapeutic activity, challenges to effective antibody-based treatment, existing technologies for antibody engineering, and current concepts for engineering new antibody formats and antibody alternatives as next generation biopharmaceuticals for cancer treatment.
Collapse
Affiliation(s)
- Eric T Boder
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996-2200, USA.
| | | |
Collapse
|
68
|
A Cassette Vector System for the Rapid Cloning and Production of Bispecific Tetravalent Antibodies. Antibodies (Basel) 2012. [DOI: 10.3390/antib1010019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
69
|
Abstract
The ability of antibodies to bind to target molecules with high affinity and specificity has led to their widespread use in diagnostic and therapeutic applications. Nevertheless, a limitation of antibodies is their propensity to self-associate and aggregate at high concentrations and elevated temperatures. The large size and multidomain architecture of full-length monoclonal antibodies have frustrated systematic analysis of how antibody sequence and structure regulate antibody solubility. In contrast, analysis of single and multidomain antibody fragments that retain the binding activity of mono-clonal antibodies has provided valuable insights into the determinants of antibody aggregation. Here we review advances in engineering antibody frameworks, domain interfaces, and antigen-binding loops to prevent aggregation of natively and nonnatively folded antibody fragments. We also highlight advances and unmet challenges in developing robust strategies for engineering large, multidomain antibodies to resist aggregation.
Collapse
Affiliation(s)
- Joseph M Perchiacca
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | | |
Collapse
|
70
|
Abstract
Monoclonal antibodies are widely used for the treatment of cancer, inflammatory and infectious diseases and other disorders. Most of the marketed antibodies are monospecific and therefore capable of interacting and interfering with a single target. However, complex diseases are often multifactorial in nature, and involve redundant or synergistic action of disease mediators or upregulation of different receptors, including crosstalk between their signaling networks. Consequently, blockade of multiple, different pathological factors and pathways may result in improved therapeutic efficacy. This result can be achieved by combining different drugs, or use of the dual targeting strategies applying bispecific antibodies that have emerged as an alternative to combination therapy. This review discusses the various dual targeting strategies for which bispecific antibodies have been developed and provides an overview of the established bispecific antibody formats.
Collapse
Affiliation(s)
- Roland E Kontermann
- Institut für Zellbiologie und Immunologie; Universität Stuttgart; Stuttgart, Germany
| |
Collapse
|
71
|
Kamperidis P, Kamalati T, Ferrari M, Jones M, Garrood T, Smith MD, Diez-Posada S, Hughes C, Finucane C, Mather S, Nissim A, George AJT, Pitzalis C. Development of a novel recombinant biotherapeutic with applications in targeted therapy of human arthritis. ACTA ACUST UNITED AC 2012; 63:3758-67. [PMID: 21953304 DOI: 10.1002/art.30650] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To isolate recombinant antibodies with specificity for human arthritic synovium and to develop targeting reagents with joint-specific delivery capacity for therapeutic and/or diagnostic applications. METHODS In vivo single-chain Fv (scFv) antibody phage display screening using a human synovial xenograft model was used to isolate antibodies specific to the microvasculature of human arthritic synovium. Single-chain Fv antibody tissue-specific reactivity was assessed by immunostaining of synovial tissues from normal controls and from patients with rheumatoid arthritis and osteoarthritis, normal human tissue arrays, and tissues from other patients with inflammatory diseases displaying neovasculogenesis. In vivo scFv antibody tissue-specific targeting capacity was examined in the human synovial xenograft model using both (125)I-labeled and biotinylated antibody. RESULTS We isolated a novel recombinant human antibody, scFv A7, with specificity for the microvasculature of human arthritic synovium. We showed that in vivo, this antibody could efficiently target human synovial microvasculature in SCID mice transplanted with human arthritic synovial xenografts. Our results demonstrated that scFv A7 antibody had no reactivity with the microvasculature or with other cellular components found in a comprehensive range of normal human tissues including normal human synovium. Further, we showed that the reactivity of the scFv A7 antibody was not a common feature of neovasculogenesis associated with chronic inflammatory conditions. CONCLUSION Here we report for the first time the identification of an scFv antibody, A7, that specifically recognizes an epitope expressed in the microvasculature of human arthritic synovium and that has the potential to be developed as a joint-specific pharmaceutical.
Collapse
Affiliation(s)
- Panagiotis Kamperidis
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Vendel MC, Favis M, Snyder WB, Huang F, Capili AD, Dong J, Glaser SM, Miller BR, Demarest SJ. Secretion from bacterial versus mammalian cells yields a recombinant scFv with variable folding properties. Arch Biochem Biophys 2012; 526:188-93. [PMID: 22230329 DOI: 10.1016/j.abb.2011.12.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 12/19/2011] [Accepted: 12/21/2011] [Indexed: 11/24/2022]
Abstract
Escherichia coli (E. coli) is the most commonly used organism for expressing antibody fragments such as single chain antibody Fvs (scFvs). Previously, we have utilized E. coli to express well-folded scFvs for characterization and engineering purposes with the goal of using these engineered proteins as building blocks for generating IgG-like bispecific antibodies (BsAbs). In the study, described here, we observed a significant difference in the secondary structure of an scFv produced in E. coli and the same scFv expressed and secreted from chinese hamster ovary (CHO) cells as part of a BsAb. We devised a proteolytic procedure to separate the CHO-derived scFv from its antibody-fusion partner and compared its properties with those of the E. coli-derived scFv. In comparison to the CHO-derived scFv, the E. coli-derived scFv was found trapped in a misfolded, but monomeric state that was stable for months at 4 °C. The misfolded state bound antigen in a heterogeneous fashion that included non-specific binding, which made functional characterization challenging. This odd incidence of obtaining a misfolded scFv from bacteria suggests careful characterization of the folded properties of bacterially expressed scFvs is warranted if anomalous issues with antigen-binding or non-specificity occur during an engineering campaign. Additionally, our proteolytic methodology for obtaining significant levels of intact scFvs from highly expressed IgG-like antibody proteins serves as a robust method for producing scFvs in CHO without the use of designed cleavage motifs.
Collapse
|
73
|
Moore GL, Bautista C, Pong E, Nguyen DHT, Jacinto J, Eivazi A, Muchhal US, Karki S, Chu SY, Lazar GA. A novel bispecific antibody format enables simultaneous bivalent and monovalent co-engagement of distinct target antigens. MAbs 2011; 3:546-57. [PMID: 22123055 DOI: 10.4161/mabs.3.6.18123] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Bispecific antibodies based on full-length antibody structures are more optimal than fragment-based formats because they benefit from the favorable properties of the Fc region. However, the homodimeric nature of Fc effectively imposes bivalent binding on all current full-length bispecific antibodies, an attribute that can result in nonspecific activation of cross-linked receptors. We engineered a novel bispecific format, referred to as mAb-Fv, that utilizes a heterodimeric Fc region to enable monovalent co-engagement of a second target antigen in a full-length context. mAb-Fv constructs co-targeting CD16 and CD3 were expressed and purified as heterodimeric species, bound selectively to their co-target antigens, and mediated potent cytotoxic activity by NK cells and T cells, respectively. The capacity to co-engage distinct target antigens simultaneously with different valencies is an improved feature for bispecific antibodies with promising therapeutic implications.
Collapse
|
74
|
Demarest SJ, Hariharan M, Elia M, Salbato J, Jin P, Bird C, Short JM, Kimmel BE, Dudley M, Woodnutt G, Hansen G. Neutralization of Clostridium difficile toxin A using antibody combinations. MAbs 2011; 2:190-8. [PMID: 20150758 DOI: 10.4161/mabs.2.2.11220] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The pathogenicity of Clostridium difficile (C. difficile) is mediated by the release of two toxins, A and B. Both toxins contain large clusters of repeats known as cell wall binding (CWB) domains responsible for binding epithelial cell surfaces. Several murine monoclonal antibodies were generated against the CWB domain of toxin A and screened for their ability to neutralize the toxin individually and in combination. Three antibodies capable of neutralizing toxin A all recognized multiple sites on toxin A, suggesting that the extent of surface coverage may contribute to neutralization. Combination of two noncompeting antibodies, denoted 3358 and 3359, enhanced toxin A neutralization over saturating levels of single antibodies. Antibody 3358 increased the level of detectable CWB domain on the surface of cells, while 3359 inhibited CWB domain cell surface association. These results suggest that antibody combinations that cover a broader epitope space on the CWB repeat domains of toxin A (and potentially toxin B) and utilize multiple mechanisms to reduce toxin internalization may provide enhanced protection against C. difficile-associated diarrhea.
Collapse
|
75
|
Abstract
The use of monoclonal antibodies (mAbs) has become a general approach for specifically targeting and treating human disease. In oncology, the therapeutic utility of mAbs is usually evaluated in the context of treatment with standard of care, as well as other small molecule targeted therapies. Many anti-cancer antibody modalities have achieved validation, including the targeting of growth factor and angiogenesis pathways, the induction of tumor cell killing or apoptosis, and the blocking of immune inhibitory mechanisms to stimulate anti-tumor responses. But, as with other targeted therapies, few antibodies are curative because of biological complexities that underlie tumor formation and redundancies in molecular pathways that enable tumors to adapt and show resistance to treatment. This review discusses the combinations of antibody therapeutics that are emerging to improve efficacy and durability within a specific biological mechanism (e.g., immunomodulation or the inhibition of angiogenesis) and across multiple biological pathways (e.g., inhibition of tumor growth and induction of tumor cell apoptosis).
Collapse
|
76
|
Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci U S A 2011; 108:11187-92. [PMID: 21690412 DOI: 10.1073/pnas.1019002108] [Citation(s) in RCA: 347] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We describe a generic approach to assemble correctly two heavy and two light chains, derived from two existing antibodies, to form human bivalent bispecific IgG antibodies without use of artificial linkers. Based on the knobs-into-holes technology that enables heterodimerization of the heavy chains, correct association of the light chains and their cognate heavy chains is achieved by exchange of heavy-chain and light-chain domains within the antigen binding fragment (Fab) of one half of the bispecific antibody. This "crossover" retains the antigen-binding affinity but makes the two arms so different that light-chain mispairing can no longer occur. Applying the three possible "CrossMab" formats, we generated bispecific antibodies against angiopoietin-2 (Ang-2) and vascular endothelial growth factor A (VEGF-A) and show that they can be produced by standard techniques, exhibit stabilities comparable to natural antibodies, and bind both targets simultaneously with unaltered affinity. Because of its superior side-product profile, the CrossMab(CH1-CL) was selected for in vivo profiling and showed potent antiangiogenic and antitumoral activity.
Collapse
|
77
|
Igawa T, Tsunoda H, Kuramochi T, Sampei Z, Ishii S, Hattori K. Engineering the variable region of therapeutic IgG antibodies. MAbs 2011; 3:243-52. [PMID: 21406966 DOI: 10.4161/mabs.3.3.15234] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Since the first generation of humanized IgG1 antibodies reached the market in the late 1990s, IgG antibody molecules have been extensively engineered. The success of antibody therapeutics has introduced severe competition in developing novel therapeutic monoclonal antibodies, especially for promising or clinically validated targets. Such competition has led researchers to generate so-called second or third generation antibodies with clinical differentiation utilizing various engineering and optimization technologies. Parent IgG antibodies can be engineered to have improved antigen binding properties, effector functions, pharmacokinetics, pharmaceutical properties and safety issues. Although the primary role of the antibody variable region is to bind to the antigen, it is also the main source of antibody diversity and its sequence affects various properties important for developing antibody therapeutics. Here we review recent research activity in variable region engineering to generate superior antibody therapeutics.
Collapse
Affiliation(s)
- Tomoyuki Igawa
- Chugai Pharmaceutical Co. Ltd., Fuji-Gotemba Research Laboratories, Shizuoka, Japan.
| | | | | | | | | | | |
Collapse
|
78
|
Dong J, Sereno A, Aivazian D, Langley E, Miller BR, Snyder WB, Chan E, Cantele M, Morena R, Joseph IBJK, Boccia A, Virata C, Gamez J, Yco G, Favis M, Wu X, Graff CP, Wang Q, Rohde E, Rennard R, Berquist L, Huang F, Zhang Y, Gao SX, Ho SN, Demarest SJ, Reff ME, Hariharan K, Glaser SM. A stable IgG-like bispecific antibody targeting the epidermal growth factor receptor and the type I insulin-like growth factor receptor demonstrates superior anti-tumor activity. MAbs 2011; 3:273-88. [PMID: 21393993 PMCID: PMC3149708 DOI: 10.4161/mabs.3.3.15188] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 02/16/2011] [Indexed: 01/01/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) and the type I insulin-like growth factor receptor (IGF-1R) are two cell surface receptor tyrosine kinases known to cooperate to promote tumor progression and drug resistance. Combined blockade of EGFR and IGF-1R has shown improved anti-tumor activity in preclinical models. Here, we report the characterization of a stable IgG-like bispecific antibody (BsAb) dual-targeting EGFR and IGF-1R that was developed for cancer therapy. The BsAb molecule (EI-04), constructed with a stability-engineered single chain variable fragment (scFv) against IGF-1R attached to the carboxyl-terminus of an IgG against EGFR, displays favorable biophysical properties for biopharmaceutical development. Biochemically, EI-04 bound to human EGFR and IGF-1R with sub nanomolar affinity, co-engaged the two receptors simultaneously, and blocked the binding of their respective ligands with similar potency compared to the parental monoclonal antibodies (mAbs). In tumor cells, EI-04 effectively inhibited EGFR and IGF-1R phosphorylation, and concurrently blocked downstream AKT and ERK activation, resulting in greater inhibition of tumor cell growth and cell cycle progression than the single mAbs. EI-04, likely due to its tetravalent bispecific format, exhibited high avidity binding to BxPC3 tumor cells co-expressing EGFR and IGF-1R, and consequently improved potency at inhibiting IGF-driven cell growth over the mAb combination. Importantly, EI-04 demonstrated enhanced in vivo anti-tumor efficacy over the parental mAbs in two xenograft models, and even over the mAb combination in the BxPC3 model. Our data support the clinical investigation of EI-04 as a superior cancer therapeutic in treating EGFR and IGF-1R pathway responsive tumors.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibody Affinity/immunology
- Antibody Specificity/immunology
- Blotting, Western
- CHO Cells
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/immunology
- Cricetinae
- Cricetulus
- Dose-Response Relationship, Drug
- ErbB Receptors/immunology
- ErbB Receptors/metabolism
- Humans
- Immunoglobulin G/immunology
- Mice
- Mice, Nude
- Mice, SCID
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/pathology
- Phosphorylation/drug effects
- Receptor, IGF Type 1/immunology
- Receptor, IGF Type 1/metabolism
- Signal Transduction/drug effects
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/pharmacology
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
Collapse
|
79
|
Fitzgerald J, Lugovskoy A. Rational engineering of antibody therapeutics targeting multiple oncogene pathways. MAbs 2011; 3:299-309. [PMID: 21393992 DOI: 10.4161/mabs.3.3.15299] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Monoclonal antibodies have significantly advanced our ability to treat cancer, yet clinical studies have shown that many patients do not adequately respond to monospecific therapy. This is in part due to the multifactorial nature of the disease, where tumors rely on multiple and often redundant pathways for proliferation. Bi- or multi- specific antibodies capable of blocking multiple growth and survival pathways at once have a potential to better meet the challenge of blocking cancer growth, and indeed many of them are advancing in clinical development. ( 1) However, bispecific antibodies present significant design challenges mostly due to the increased number of variables to consider. In this perspective we describe an innovative integrated approach to the discovery of bispecific antibodies with optimal molecular properties, such as affinity, avidity, molecular format and stability. This approach combines simulations of potential inhibitors using mechanistic models of the disease-relevant biological system to reveal optimal inhibitor characteristics with antibody engineering techniques that yield manufacturable therapeutics with robust pharmaceutical properties. We illustrate how challenges of meeting the optimal design criteria and chemistry, manufacturing and control concerns can be addressed simultaneously in the context of an accelerated therapeutic design cycle. Finally, to demonstrate how this rational approach can be applied, we present a case study where the insights from mechanistic modeling were used to guide the engineering of an IgG-like bispecific antibody.
Collapse
|
80
|
Arnett SO, Teillaud JL, Wurch T, Reichert JM, Dunlop C, Huber M. IBC's 21st Annual Antibody Engineering and 8th Annual Antibody Therapeutics International Conferences and 2010 Annual Meeting of the Antibody Society. December 5-9, 2010, San Diego, CA USA. MAbs 2011; 3:133-52. [PMID: 21304271 PMCID: PMC3092615 DOI: 10.4161/mabs.3.2.14939] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The 21st Annual Antibody Engineering and 8th Annual Antibody Therapeutics international conferences, and the 2010 Annual Meeting of The Antibody Society, organized by IBC Life Sciences with contributions from The Antibody Society and two Scientific Advisory Boards, was held December 5–9, 2010 in San Diego, CA. The conferences featured over 100 presentations and 100 posters, and included a pre-conference workshop on deep-sequencing of antibody genes. The total number of delegates exceeded 800, which set a new attendance record for the conference. The conferences were organized with a focus on antibody engineering only on the first day and a joint engineering/therapeutics session on the last day. Delegates could select from presentations that occurred in two simultaneous sessions on days 2 and 3. Day 1 included presentations on neutralizing antibodies and the identification of vaccine targets, as well as a historical overview of 20 years of phage display utilization. Topics presented in the Antibody Engineering sessions on day 2 and 3 included antibody biosynthesis, structure and stability; antibodies in a complex environment; antibody half-life; and targeted nanoparticle therapeutics. In the Antibody Therapeutics sessions on days 2 and 3, preclinical and early stage development and clinical updates of antibody therapeutics, including TRX518, SYM004, MM111, PRO140, CVX-241, ASG-5ME, U3-1287 (AMG888), R1507 and trastuzumab emtansine, were discussed and perspectives were provided on the development of biosimilar and biobetter antibodies, including coverage of regulatory and intellectual property issues. The joint engineering/therapeutics session on the last day focused on bispecific and next-generation antibodies. Summaries of most of the presentations are included here, but, due to the large number of speakers, it was not possible to include summaries for every presentation. Delegates enjoyed the splendid views of the San Diego Bay and proximity to the Gaslamp Quarter provided by the venue. The 22nd Annual Antibody Engineering and 9th Annual Antibody Therapeutics conferences, and the 2011 Annual Meeting of The Antibody Society, are planned for December 5–8, 2011 at the same location in San Diego, and will include two two-day short courses on Introduction to Antibody Engineering and Protein Characterization for Biotechnology Product Development.
Collapse
Affiliation(s)
- Samantha O Arnett
- Department of Immunology & Microbial Science, The Scripps Research Institute, La Jolla, USA
| | | | | | | | | | | |
Collapse
|
81
|
Beck A, Wurch T, Reichert JM. 6th Annual European Antibody Congress 2010: November 29-December 1, 2010, Geneva, Switzerland. MAbs 2011; 3:111-32. [PMID: 21441785 PMCID: PMC3092614 DOI: 10.4161/mabs.3.2.14788] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 01/11/2011] [Indexed: 12/31/2022] Open
Abstract
The 6th European Antibody Congress (EAC), organized by Terrapinn Ltd., was held in Geneva, Switzerland, which was also the location of the 4th and 5th EAC. As was the case in 2008 and 2009, the EAC was again the largest antibody congress held in Europe, drawing nearly 250 delegates in 2010. Numerous pharmaceutical and biopharmaceutical companies active in the field of therapeutic antibody development were represented, as were start-up and academic organizations and representatives from the US Food and Drug Administration FDA. The global trends in antibody research and development were discussed, including success stories of recent marketing authorizations of golimumab (Simponi®) and canakinumab (Ilaris®) by Johnson & Johnson and Novartis, respectively, updates on antibodies in late clinical development (obinutuzumab/GA101, farletuzumab/MORAb-003 and itolizumab/T1 h, by Glycart/Roche, Morphotek and Biocon, respectively) and success rates for this fast-expanding class of therapeutics (Tufts Center for the Study of Drug Development). Case studies covering clinical progress of girentuximab (Wilex), evaluation of panobacumab (Kenta Biotech), characterization of therapeutic antibody candidates by protein microarrays (Protagen), antibody-drug conjugates (sanofi-aventis, ImmunoGen, Seattle Genetics, Wyeth/Pfizer), radio-immunoconjugates (Bayer Schering Pharma, Université de Nantes) and new scaffolds (Ablynx, AdAlta, Domantis/GlaxoSmithKline, Fresenius, Molecular Partners, Pieris, Scil Proteins, Pfizer, University of Zurich) were presented. Major antibody structural improvements were showcased, including the latest selection engineering of the best isotypes (Abbott, Pfizer, Pierre Fabre), hinge domain (Pierre Fabre), dual antibodies (Abbott), IgG-like bispecific antibodies (Biogen Idec), antibody epitope mapping case studies (Eli Lilly), insights in FcγRII receptor (University of Cambridge), as well as novel tools for antibody fragmentation (Genovis). Improvements of antibody druggability (Abbott, Bayer, Pierre Fabre, Merrimack, Pfizer), enhancing IgG pharmacokinetics (Abbott, Chugai), progress in manufacturing (Genmab, Icosagen Cell Factory, Lonza, Pierre Fabre) and the development of biosimilar antibodies (Biocon, Sandoz, Triskel) were also discussed. Last but not least, identification of monoclonal antibodies (mAbs) against new therapeutic targets (Genentech, Genmab, Imclone/Lilly, Vaccinex) including Notch, cMet, TGFbRII, SEMA4D, novel development in immunotherapy and prophylaxis against influenza (Crucell), anti-tumor activity of immunostimulatory antibodies (MedImmune/Astra Zeneca) and translations to clinical studies including immunogenicity issues (Amgen, Novartis, University of Debrecen) were presented.
Collapse
Affiliation(s)
- Alain Beck
- Physio-Chemistry Department, Centre d'Immunologie Pierre-Fabre, Saint julien en Genevois, France.
| | | | | |
Collapse
|
82
|
Dong J, Sereno A, Snyder WB, Miller BR, Tamraz S, Doern A, Favis M, Wu X, Tran H, Langley E, Joseph I, Boccia A, Kelly R, Wortham K, Wang Q, Berquist L, Huang F, Gao SX, Zhang Y, Lugovskoy A, Martin S, Gouvis H, Berkowitz S, Chiang G, Reff M, Glaser SM, Hariharan K, Demarest SJ. Stable IgG-like bispecific antibodies directed toward the type I insulin-like growth factor receptor demonstrate enhanced ligand blockade and anti-tumor activity. J Biol Chem 2011; 286:4703-17. [PMID: 21123183 PMCID: PMC3039382 DOI: 10.1074/jbc.m110.184317] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Revised: 11/02/2010] [Indexed: 12/11/2022] Open
Abstract
Bispecific antibodies (BsAbs) target multiple epitopes on the same molecular target or different targets. Although interest in BsAbs has persisted for decades, production of stable and active BsAbs has hindered their clinical evaluation. Here, we describe the production and characterization of tetravalent IgG-like BsAbs that combine the activities of allosteric and competitive inhibitors of the type-I insulin-like growth factor receptor (IGF-1R). The BsAbs, which were engineered for thermal stability, express well, demonstrate favorable biophysical properties, and recognize both epitopes on IGF-1R. Only one BsAb with a unique geometry, denoted BIIB4-5scFv, was capable of engaging all four of its binding arms simultaneously. All the BsAbs (especially BIIB4-5scFv) demonstrated enhanced ligand blocking over the single monoclonal antibodies (mAbs), particularly at high ligand concentrations. The pharmacokinetic profiles of two IgG-like BsAbs were tested in nude mice and shown to be comparable with that of the parental mAbs. The BsAbs, especially BIIB4-5scFv, demonstrated an improved ability to reduce the growth of multiple tumor cell lines and to inhibit ligand-induced IGF-1R signaling in tumor cells over the parental mAbs. BIIB4-5scFv also led to superior tumor growth inhibition over its parental mAbs in vivo. In summary, BsAbs that bridge multiple inhibitory mechanisms against a single target may generally represent a more effective strategy for intervention in oncology or other indications compared with traditional mAb therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Monoclonal, Murine-Derived/pharmacokinetics
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Drug Stability
- Humans
- Immunoglobulin G
- Ligands
- Mice
- Mice, Nude
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Protein Stability
- Receptor, IGF Type 1/antagonists & inhibitors
- Receptor, IGF Type 1/immunology
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
| | | | | | | | | | - Adam Doern
- From Biogen Idec, San Diego, California 92122
| | | | - Xiufeng Wu
- From Biogen Idec, San Diego, California 92122
| | - Hon Tran
- From Biogen Idec, San Diego, California 92122
| | | | | | | | | | | | - Qin Wang
- From Biogen Idec, San Diego, California 92122
| | | | - Flora Huang
- From Biogen Idec, San Diego, California 92122
| | | | - Ying Zhang
- From Biogen Idec, San Diego, California 92122
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Development of tetravalent, bispecific CCR5 antibodies with antiviral activity against CCR5 monoclonal antibody-resistant HIV-1 strains. Antimicrob Agents Chemother 2011; 55:2369-78. [PMID: 21300827 DOI: 10.1128/aac.00215-10] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we describe novel tetravalent, bispecific antibody derivatives that bind two different epitopes on the HIV coreceptor CCR5. The basic protein formats that we applied were derived from Morrison-type bispecific antibodies: whole IgGs to which we connected single-chain antibodies (scFvs) via (Gly4Ser)n sequences at either the C or N terminus of the light chain or heavy chain. By design optimization, including disulfide stabilization of scFvs or introduction of 30-amino-acid linkers, stable molecules could be obtained in amounts that were within the same range as or no less than 4-fold lower than those observed with monoclonal antibodies in transient expression assays. In contrast to monospecific CCR5 antibodies, bispecific antibody derivatives block two alternative docking sites of CCR5-tropic HIV strains on the CCR5 coreceptor. Consequently, these molecules showed 18- to 57-fold increased antiviral activities compared to the parent antibodies. Most importantly, one prototypic tetravalent CCR5 antibody had antiviral activity against virus strains resistant to the single parental antibodies. In summary, physical linkage of two CCR5 antibodies targeting different epitopes on the HIV coreceptor CCR5 resulted in tetravalent, bispecific antibodies with enhanced antiviral potency against wild-type and CCR5 antibody-resistant HIV-1 strains.
Collapse
|
84
|
Miller BR, Demarest SJ, Lugovskoy A, Huang F, Wu X, Snyder WB, Croner LJ, Wang N, Amatucci A, Michaelson JS, Glaser SM. Stability engineering of scFvs for the development of bispecific and multivalent antibodies. Protein Eng Des Sel 2010; 23:549-57. [PMID: 20457695 DOI: 10.1093/protein/gzq028] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Single-chain Fvs (scFvs) are commonly used building blocks for creating engineered diagnostic and therapeutic antibody molecules. Bispecific antibodies (BsAbs) hold particular interest due to their ability to simultaneously bind and engage two distinct targets. We describe a technology for producing stable, scalable IgG-like bispecific and multivalent antibodies based on methods for rapidly engineering thermally stable scFvs. Focused libraries of mutant scFvs were designed using a combination of sequence-based statistical analyses and structure-, and knowledge-based methods. Libraries encoding these designs were expressed in E. coli and culture supernatants-containing soluble scFvs screened in a high-throughput assay incorporating a thermal challenge prior to an antigen-binding assay. Thermally stable scFvs were identified that retain full antigen-binding affinity. Single mutations were found that increased the measured T(m) of either the V(H) or V(L) domain by as much as 14 degrees C relative to the wild-type scFv. Combinations of mutations further increased the T(m) by as much as an additional 12 degrees C. Introduction of a stability-engineered scFv as part of an IgG-like BsAb enabled scalable production and purification of BsAb with favorable biophysical properties.
Collapse
Affiliation(s)
- Brian R Miller
- Biogen Idec, Inc., 5200 Research Place, San Diego, CA 92122, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Mabry R, Gilbertson DG, Frank A, Vu T, Ardourel D, Ostrander C, Stevens B, Julien S, Franke S, Meengs B, Brody J, Presnell S, Hamacher NB, Lantry M, Wolf A, Bukowski T, Rosler R, Yen C, Anderson-Haley M, Brasel K, Pan Q, Franklin H, Thompson P, Dodds M, Underwood S, Peterson S, Sivakumar PV, Snavely M. A dual-targeting PDGFRbeta/VEGF-A molecule assembled from stable antibody fragments demonstrates anti-angiogenic activity in vitro and in vivo. MAbs 2010; 2:20-34. [PMID: 20065654 DOI: 10.4161/mabs.2.1.10498] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Targeting angiogenesis is a promising approach to the treatment of solid tumors and age-related macular degeneration (AMD). Inhibition of vascularization has been validated by the successful marketing of monoclonal antibodies (mAbs) that target specific growth factors or their receptors, but there is considerable room for improvement in existing therapies. Combination of mAbs targeting both the VEGF and PDGF pathways has the potential to increase the efficacy of anti-angiogenic therapy without the accompanying toxicities of tyrosine kinase inhibitors and the inability to combine efficiently with traditional chemotherapeutics. However, development costs and regulatory issues have limited the use of combinatorial approaches for the generation of more efficacious treatments. The concept of mediating disease pathology by targeting two antigens with one therapeutic was proposed over two decades ago. While mAbs are particularly suitable candidates for a dual-targeting approach, engineering bispecificity into one molecule can be difficult due to issues with expression and stability, which play a significant role in manufacturability. Here, we address these issues upstream in the process of developing a bispecific antibody (bsAb). Single-chain antibody fragments (scFvs) targeting PDGFRbeta and VEGF-A were selected for superior stability. The scFvs were fused to both termini of human Fc to generate a bispecific, tetravalent molecule. The resulting molecule displays potent activity, binds both targets simultaneously, and is stable in serum. The assembly of a bsAb using stable monomeric units allowed development of an anti-PDGFRB/VEGF-A antibody capable of attenuating angiogenesis through two distinct pathways and represents an efficient method for rapid engineering of dual-targeting molecules.
Collapse
Affiliation(s)
- Robert Mabry
- Antibody Discovery and Assay Technology, ZymoGenetics, Inc., Seattle, WA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|