151
|
Di Fede G, Bronte G, Rizzo S, Rolfo Cervetto C, Cocorullo G, Gulotta G, Bazan V, Russo A. Monoclonal antibodies and antibody fragments: state of the art and future perspectives in the treatment of non-haematological tumors. Expert Opin Biol Ther 2011; 11:1433-45. [DOI: 10.1517/14712598.2011.594436] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
152
|
Riihimäki TA, Hiltunen S, Rangl M, Nordlund HR, Määttä JAE, Ebner A, Hinterdorfer P, Kulomaa MS, Takkinen K, Hytönen VP. Modification of the loops in the ligand-binding site turns avidin into a steroid-binding protein. BMC Biotechnol 2011; 11:64. [PMID: 21658230 PMCID: PMC3201017 DOI: 10.1186/1472-6750-11-64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 06/09/2011] [Indexed: 01/20/2023] Open
Abstract
Background Engineered proteins, with non-immunoglobulin scaffolds, have become an important alternative to antibodies in many biotechnical and therapeutic applications. When compared to antibodies, tailored proteins may provide advantageous properties such as a smaller size or a more stable structure. Results Avidin is a widely used protein in biomedicine and biotechnology. To tailor the binding properties of avidin, we have designed a sequence-randomized avidin library with mutagenesis focused at the loop area of the binding site. Selection from the generated library led to the isolation of a steroid-binding avidin mutant (sbAvd-1) showing micromolar affinity towards testosterone (Kd ~ 9 μM). Furthermore, a gene library based on the sbAvd-1 gene was created by randomizing the loop area between β-strands 3 and 4. Phage display selection from this library led to the isolation of a steroid-binding protein with significantly decreased biotin binding affinity compared to sbAvd-1. Importantly, differential scanning calorimetry and analytical gel-filtration revealed that the high stability and the tetrameric structure were preserved in these engineered avidins. Conclusions The high stability and structural properties of avidin make it an attractive molecule for the engineering of novel receptors. This methodology may allow the use of avidin as a universal scaffold in the development of novel receptors for small molecules.
Collapse
Affiliation(s)
- Tiina A Riihimäki
- Institute of Biomedical Technology, University of Tampere and Tampere University Hospital, FI-33520 Tampere, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Gaster RS, Xu L, Han SJ, Wilson RJ, Hall DA, Osterfeld SJ, Yu H, Wang SX. Quantification of protein interactions and solution transport using high-density GMR sensor arrays. NATURE NANOTECHNOLOGY 2011; 6:314-20. [PMID: 21478869 PMCID: PMC3089684 DOI: 10.1038/nnano.2011.45] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 03/08/2011] [Indexed: 05/19/2023]
Abstract
Monitoring the kinetics of protein interactions on a high-density sensor array is vital to drug development and proteomic analysis. Label-free kinetic assays based on surface plasmon resonance are the current gold standard, but they have poor detection limits, suffer from non-specific binding, and are not amenable to high-throughput analyses. Here, we show that magnetically responsive nanosensors that have been scaled to over 100,000 sensors per cm² can be used to measure the binding kinetics of various proteins with high spatial and temporal resolution. We present an analytical model that describes the binding of magnetically labelled antibodies to proteins that are immobilized on the sensor surface. This model is able to quantify the kinetics of antibody-antigen binding at sensitivities as low as 20 zeptomoles of solute.
Collapse
Affiliation(s)
- Richard S. Gaster
- Department of Bioengineering, Stanford University, CA 94305, USA
- Medical Scientist Training Program, School of Medicine, Stanford University, CA 94305, USA
| | - Liang Xu
- Department of Materials Science and Engineering, Stanford University, CA 94305, USA
| | - Shu-Jen Han
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598
| | - Robert J. Wilson
- Department of Materials Science and Engineering, Stanford University, CA 94305, USA
| | - Drew A. Hall
- Department of Electrical Engineering, Stanford University, CA 94305, USA
| | | | - Heng Yu
- MagArray Inc., Sunnyvale, CA 94089
| | - Shan X. Wang
- Department of Materials Science and Engineering, Stanford University, CA 94305, USA
- Department of Electrical Engineering, Stanford University, CA 94305, USA
- Correspondence and requests for materials should be addressed to: Shan X. Wang (), Stanford Center for Magnetic Nanotechnology, Phone: 650-723-8671, Mail address: Geballe Laboratory for Advanced Materials, McCullough Building, Room 351, 476 Lomita Mall, Stanford University, Stanford, CA 94305-4045
| |
Collapse
|
154
|
Abstract
Monoclonal antibodies (mAbs) have become one of the largest classes of new therapeutic agents approved for use in oncology, and have revolutionised the treatment of many human malignancies. Clinically useful mAbs can function through several different mechanisms, including inhibition of tumour-related signalling, induction of apoptosis, inhibition of angiogenesis, enhancing host immune response against cancer and targeted delivery of payloads (such as toxins, cytotoxic agents or radioisotopes) to the tumour site. The increasing knowledge of key molecules and cellular pathways involved in tumour induction and progression has led to a rise in the proportion of therapeutic mAbs entering clinical trials. These mAbs consist of various conventional or recombinant, murine, humanised, chimeric or fully human and fusion constructs. In this review, we provide an overview of mAbs approved for use in clinical oncology and those currently in clinical development. We also discuss the mechanisms of action of anti-cancer mAbs, as well as the antigen targets recognised by these antibodies.
Collapse
Affiliation(s)
- Vinochani Pillay
- Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria 3084, Australia
| | | | | |
Collapse
|
155
|
Al-Rubeai M. Production of Antibody Fab Fragments in Escherichia coli. ANTIBODY EXPRESSION AND PRODUCTION 2011. [PMCID: PMC7121915 DOI: 10.1007/978-94-007-1257-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A phage-display library is the most broadly used platform for preparation of recombinant human monoclonal antibody Fab fragments. Panning is effective for the selection of immunoglobulin genes from naïve and immune libraries. However, it is possible to bypass the phage display system if human peripheral lymphocytes are obtained from seropositive patients with infectious diseases as a source of immunoglobulin genes. Direct screening of bacterial colonies producing Fab fragments by colony blotting using filter membranes is practical for the isolation of human Fab fragments to major antigens of pathogens. An oligoclonal culture can also be used, and is a partial application of Epstein-Barr virus transformation of peripheral lymphocytes. Using these procedures, neutralizing antibody Fab fragments to various antigens can be obtained with a sufficient level of cloning efficacy. Chain shuffling and site-directed mutagenesis are also useful ways to improve the quality of the cloned antibody Fab fragments.
Collapse
Affiliation(s)
- Mohamed Al-Rubeai
- Dept. Chemical & Biochemical, Engineering, University College, Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
156
|
Intrabody Expression in Mammalian Cells. ANTIBODY EXPRESSION AND PRODUCTION 2011. [PMCID: PMC7120103 DOI: 10.1007/978-94-007-1257-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
The intracellular expression of antibodies or antibody fragments (intrabodies) in different compartments of mammalian cells allows to block or modulate the function of endogenous molecules. Intrabodies can alter protein folding, protein-protein, protein-DNA, protein-RNA interactions and protein modification. They can induce a phenotypic knockout and work as neutralizing agents by direct binding to the target antigen, by diverting its intracellular traffic or by inhibiting its association with binding partners. They have been largely employed as research tools and are emerging as therapeutic molecules for the treatment of human diseases as viral pathologies, cancer and misfolding diseases. The fast growing bio-market of recombinant antibodies provides intrabodies with enhanced binding specificity, stability and solubility, together with lower immunogenicity, for their use in therapy. This chapter describes the crucial aspects required to express intrabodies in different intracellular compartments of mammalian cells, their various modes of action and gives an update on the applications of intrabodies in human diseases.
Collapse
|
157
|
Ishikawa T, Ito T, Endo R, Nakagawa K, Sawa E, Wakamatsu K. Influence of pH on heat-induced aggregation and degradation of therapeutic monoclonal antibodies. Biol Pharm Bull 2010; 33:1413-7. [PMID: 20686240 DOI: 10.1248/bpb.33.1413] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Monoclonal antibodies are widely used for the treatment of various diseases, and because therapeutic monoclonal antibodies are stored in an aqueous solution or in a lyophilized state, the preparation of a stabilizing formulation that prevents their deterioration (degradation and aggregation) is crucial. Given the structural similarities of the immunoglobulin G (IgG) framework regions and a diversity of only four subclasses, we aimed to find common conditions that stabilize many different antibodies. In this study, we analyzed the effect of pH (the most critical factor in establishing a stable formulation) on human monoclonal antibodies from subclasses IgG1, IgG2, and IgG4, all of which have been utilized in antibody therapeutics. We found that human IgGs are stable with minimal heat-induced degradation and aggregation at pH 5.0-5.5 irrespective of their subclass. We also found that IgG1 is more susceptible to fragmentation, whereas IgG4 is more susceptible to aggregation. This basic information emphasizing the influence of pH on IgG stability should facilitate the optimization of formulation conditions tailored to individual antibodies for specific uses.
Collapse
Affiliation(s)
- Tomoyoshi Ishikawa
- Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd., Takasaki, Gunma 370-0013, Japan.
| | | | | | | | | | | |
Collapse
|
158
|
Cai Z, Zhang H, Liu J, Berezov A, Murali R, Wang Q, Greene MI. Targeting erbB receptors. Semin Cell Dev Biol 2010; 21:961-6. [PMID: 20850557 PMCID: PMC5940346 DOI: 10.1016/j.semcdb.2010.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 09/09/2010] [Indexed: 10/19/2022]
Abstract
Our work is concerned with the origins and therapy of human cancers. Members of the epidermal growth factor receptor (EGFR) family of tyrosine kinases, also known as erbB or HER receptors, are over expressed and/or activated in many types of human tumors and represent important therapeutic targets in cancer therapy. Studies from our laboratory identified targeted therapy as a way to treat cancer. Rational therapeutics targeting and disabling erbB receptors have been developed to reverse the malignant properties of tumors. Reversal of the malignant phenotype, best seen with disabling the HER2 receptors using monoclonal antibodies is a distinct process from that seen with blocking of ligand binding to cognate receptors as has been done for EGFr receptors. Here we review the mechanisms of action deduced from a number of approaches developed in our laboratory and elsewhere, including monoclonal antibodies, peptide mimetics, recombinant proteins and small molecules. The biochemical and biological principles which have been uncovered during these studies of disabling HER2 homomeric or HER2-EGFr heteromeric receptors will help the development of novel and more efficient therapeutics targeting erbB family receptors.
Collapse
Affiliation(s)
- Zheng Cai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | - Hongtao Zhang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| | - Jing Liu
- School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui 230027, China
| | - Alan Berezov
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, S122A Steven Spielberg Building, 8725 Alden Dr., Los Angeles, CA 90048, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Davis Building, # 4092, Los Angeles, CA 90048, USA
| | - Qiang Wang
- Women’s Cancer Research Institute at the Samuel Oschin Comprehensive Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mark I. Greene
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-6082, USA
| |
Collapse
|
159
|
Abstract
Bispecific antibodies, in contrast to conventional monoclonal antibodies, can bind simultaneously two different antigens. Taking advantage of this virtue, they are mostly designed for immune effector cell redirection to tumors and for radionuclide pretargeting to tumors. Bispecific antibodies of the first generation were produced by chemical cross-linking or cell-fusion technologies. More recently, the application of genetic engineering technologies gave rise to numerous formats of bispecific antibody fragments and whole IgG molecules. Because bispecific antibodies enable therapeutic strategies that are not possible with conventional monoclonal antibodies, they attract strong interest. Several bispecific antibody formats have already shown clinical efficacy in cancer patients, catalyzing efforts to translate the imaginative bispecific antibody concepts into effective therapies.
Collapse
Affiliation(s)
- Nurit Hollander
- Department of Human Microbiology, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
160
|
Abstract
A wide variety of full-size monoclonal antibodies (mAbs) and therapeutics derived from alternative antibody formats can be produced through genetic and biological engineering techniques. These molecules are now filling the preclinical and clinical pipelines of every major pharmaceutical company and many biotechnology firms. Metrics for the development of antibody therapeutics, including averages for the number of candidates entering clinical study and development phase lengths for mAbs approved in the United States, were derived from analysis of a dataset of over 600 therapeutic mAbs that entered clinical study sponsored, at least in part, by commercial firms. The results presented provide an overview of the field and context for the evaluation of on-going and prospective mAb development programs. The expansion of therapeutic antibody use through supplemental marketing approvals and the increase in the study of therapeutics derived from alternative antibody formats are discussed.
Collapse
|
161
|
Tropism-modification strategies for targeted gene delivery using adenoviral vectors. Viruses 2010; 2:2290-2355. [PMID: 21994621 PMCID: PMC3185574 DOI: 10.3390/v2102290] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 10/07/2010] [Indexed: 02/08/2023] Open
Abstract
Achieving high efficiency, targeted gene delivery with adenoviral vectors is a long-standing goal in the field of clinical gene therapy. To achieve this, platform vectors must combine efficient retargeting strategies with detargeting modifications to ablate native receptor binding (i.e. CAR/integrins/heparan sulfate proteoglycans) and “bridging” interactions. “Bridging” interactions refer to coagulation factor binding, namely coagulation factor X (FX), which bridges hepatocyte transduction in vivo through engagement with surface expressed heparan sulfate proteoglycans (HSPGs). These interactions can contribute to the off-target sequestration of Ad5 in the liver and its characteristic dose-limiting hepatotoxicity, thereby significantly limiting the in vivo targeting efficiency and clinical potential of Ad5-based therapeutics. To date, various approaches to retargeting adenoviruses (Ad) have been described. These include genetic modification strategies to incorporate peptide ligands (within fiber knob domain, fiber shaft, penton base, pIX or hexon), pseudotyping of capsid proteins to include whole fiber substitutions or fiber knob chimeras, pseudotyping with non-human Ad species or with capsid proteins derived from other viral families, hexon hypervariable region (HVR) substitutions and adapter-based conjugation/crosslinking of scFv, growth factors or monoclonal antibodies directed against surface-expressed target antigens. In order to maximize retargeting, strategies which permit detargeting from undesirable interactions between the Ad capsid and components of the circulatory system (e.g. coagulation factors, erythrocytes, pre-existing neutralizing antibodies), can be employed simultaneously. Detargeting can be achieved by genetic ablation of native receptor-binding determinants, ablation of “bridging interactions” such as those which occur between the hexon of Ad5 and coagulation factor X (FX), or alternatively, through the use of polymer-coated “stealth” vectors which avoid these interactions. Simultaneous retargeting and detargeting can be achieved by combining multiple genetic and/or chemical modifications.
Collapse
|
162
|
Elbakri A, Nelson PN, Abu Odeh RO. The state of antibody therapy. Hum Immunol 2010; 71:1243-50. [PMID: 20849901 DOI: 10.1016/j.humimm.2010.09.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 08/30/2010] [Accepted: 09/09/2010] [Indexed: 12/13/2022]
Abstract
Therapeutic antibodies are widely used in the treatment of various diseases and disease conditions, including cardiovascular diseases, autoimmune disorders, malignancies, and infections. With at least 23 therapeutic agents currently in clinical use and a successful business generating large revenues, major technological advances are now in place to improve the specificity and efficacy of those antibodies already in the market and also generate new, safe and effective macromolecules for the treatment of other ailments. This review provides a summary of the current state of antibody therapy, highlights and discusses recent developments in the field of antibody-based therapeutics production, combination therapy and shows the status of some of the agents that are in clinical trial.
Collapse
Affiliation(s)
- Ali Elbakri
- Department of Medical Laboratory Technology, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| | | | | |
Collapse
|
163
|
Teplyakov A, Obmolova G, Carton JM, Gao W, Zhao Y, Gilliland GL. On the domain pairing in chimeric antibodies. Mol Immunol 2010; 47:2422-6. [PMID: 20554002 DOI: 10.1016/j.molimm.2010.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 04/30/2010] [Accepted: 05/03/2010] [Indexed: 11/28/2022]
Abstract
A chimeric antibody was constructed from two unrelated antibodies by combining their heavy and light chains. The "double chimera" consists of the mouse variable regions of different specificity (IL-13 and EMMPRIN) and the constant regions of different origin (mouse and human). The Fab fragment of this chimeric antibody was expressed in mammalian cells, and the crystal structure was determined at 1.6A resolution. Despite a large number of amino acid substitutions in the double chimera with respect to the parent antibodies, the heavy and light chains associate into a stable molecule. Comparison to the structure of one of the parent antibodies reveals that the variable domain interface, as well as the conformation of antigen-binding loops, is preserved without major rearrangements due to conservation of amino acids in key positions. Comparison to the structures of the all-human and all-mouse constant domains indicates a remarkable plasticity of the inter-chain interface that can tolerate residue relocations of up to 6A.
Collapse
Affiliation(s)
- Alexey Teplyakov
- Centocor R&D, Inc., 145 King of Prussia Road, Radnor, PA 19087, USA.
| | | | | | | | | | | |
Collapse
|
164
|
Balogh Z, Lautner G, Bardóczy V, Komorowska B, Gyurcsányi RE, Mészáros T. Selection and versatile application of virus-specific aptamers. FASEB J 2010; 24:4187-95. [PMID: 20624933 DOI: 10.1096/fj.09-144246] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although the significance of molecular diagnostics in routine plant virus detection is rapidly growing, the preferred methods are still antibody-based enzyme immunoassays. In the past decade, aptamers have been demonstrated to be viable alternatives of antibodies in many applications. We set out to select apple stem pitting virus (ASPV)-specific aptamers and to apply them as antibody substitutes in various immunoassay methods. The applied systematic evolution of ligands by exponential enrichment (SELEX) procedure resulted in highly discriminative aptamers selectively binding to the target virus coat protein even in complex protein matrixes. We developed protocols for exploitation of aptamers in diverse plant virus diagnosis methods, such as dot and Western blot analyses and enzyme-linked oligonucleotide assay (ELONA). Our selected aptamers proved to be superior to the available antibody in all aspects. In contrast to the antibody, the aptamers decorate both native and denaturated proteins, and ELONA produces higher signal intensity than traditional enzyme-linked immunosorbent assay (ELISA) with virus-infected plant extract. Summarily, our results present the selection and practical utilization of first plant virus-specific aptamers. Most important, the first application of ELONA for virus detection is demonstrated, which proposes a novel, more flexible, and cost-effective means of virus diagnostics.
Collapse
Affiliation(s)
- Zsófia Balogh
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
165
|
Abstract
Positron emission tomography (PET) is playing an increasingly important role in the diagnosis, staging, and monitoring response to treatment in a variety of cancers. Recent efforts have focused on immuno-PET, which uses antibody-based radiotracers, to image tumors based on expression of tumor-associated antigens. It is postulated that the specificity afforded by antibody targeting should both improve tumor detection and provide phenotypic information related to primary and metastatic lesions that will guide therapy decisions. Advances in antibody-engineering are providing the tools to develop antibody-based molecules with pharmacokinetic properties optimized for use as immuno-PET radiotracers. Coupled with technical advances in the design of PET scanners, immuno-PET holds promise to improve diagnostic imaging and to guide the use of targeted therapies. An overview of the preclinical immuno-PET studies in cancer models is reviewed here.
Collapse
Affiliation(s)
- Smitha Reddy
- Department of Molecular and Translational Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | |
Collapse
|
166
|
Cuesta AM, Sainz-Pastor N, Bonet J, Oliva B, Alvarez-Vallina L. Multivalent antibodies: when design surpasses evolution. Trends Biotechnol 2010; 28:355-62. [PMID: 20447706 DOI: 10.1016/j.tibtech.2010.03.007] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 03/25/2010] [Accepted: 03/30/2010] [Indexed: 12/21/2022]
Abstract
Evolutionary pressure has selected antibodies as key immune molecules acting against foreign pathogens. The development of monoclonal antibody technology has allowed their widespread use in research, real-time diagnosis and treatment of multiple diseases, including cancer. However, compared with hematologic malignancies, solid tumors have often proven to be relatively resistant to antibody-based therapies. In an attempt to improve the tumor-targeting efficacy of antibodies, new formats with modified, multivalent properties have been generated. Initially, these formats imitated the structure of native IgG, creating mostly monospecific, bivalent antibodies. Recently, novel trivalent antibodies have been developed to maximize tumor targeting capabilities through enhanced biodistribution and functional affinity. We review recent advances in the engineering of multivalent antibodies and further discuss their promise as agents for in vivo diagnostics and therapy.
Collapse
Affiliation(s)
- Angel M Cuesta
- Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Madrid, Spain
| | | | | | | | | |
Collapse
|
167
|
Potential aggregation-prone regions in complementarity-determining regions of antibodies and their contribution towards antigen recognition: a computational analysis. Pharm Res 2010; 27:1512-29. [PMID: 20422267 PMCID: PMC7088613 DOI: 10.1007/s11095-010-0143-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 03/30/2010] [Indexed: 11/03/2022]
Abstract
PURPOSE To analyze contribution of short aggregation-prone regions (APRs), which may self-associate via cross-beta motif and were earlier identified in therapeutic mAbs, towards antigen recognition via structural analyses of antibody-antigen complexes. METHODS A dataset of 29 publically available high-resolution crystal structures of Fab-antigen complexes was collected. Contribution of APRs towards the surface areas of the Fabs buried by the cognate antigens was computed. Propensities of amino acids to occur in APRs and to be involved in antigen binding were compared. Coincidence between APRs and individual CDR loops was examined. RESULTS All Fabs in the dataset contain at least one APR in CDR loops and adjacent framework beta-strands. The average contribution of APRs towards buried surface area of Fabs is 16.0 +/- 10.7%. Aggregation and antigen recognition may be coupled via aromatic residues (Tyr, Trp), which occur with high propensities in both APRs and antigen binding sites. APRs are infrequent in the heavy chain CDR 3 (H3) loops (7%), but are frequent in H2 loops (45%). CONCLUSIONS Co-incidence of APRs with antigen recognition sites can potentially lead to the loss of function upon aggregation. Rational structure-based design or selection strategies are suggested for biotherapeutics with improved druggability while maintaining potency.
Collapse
|
168
|
Huang J, Koide S. Rational conversion of affinity reagents into label-free sensors for Peptide motifs by designed allostery. ACS Chem Biol 2010; 5:273-7. [PMID: 20143785 PMCID: PMC2842456 DOI: 10.1021/cb900284c] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Optical biosensors for short peptide motifs, an important class of biomarkers, have been developed based on "affinity clamps", a new class of recombinant affinity reagents. Affinity clamps are engineered by linking a peptide-binding domain and an antibody mimic domain based on the fibronectin type III scaffold, followed by optimization of the interface between the two. This two-domain architecture allows for the design of allosteric coupling of peptide binding to fluorescence energy transfer between two fluorescent proteins attached to the affinity clamp. Coupled with high affinity and specificity of the underlying affinity clamps and rationally designed mutants with different sensitivity, peptide concentrations in crude cell lysate were determined with a low nanomolar detection limit and over 3 orders of magnitude. Because diverse affinity clamps can be engineered, our strategy provides a general platform to generate a repertoire of genetically encoded, label-free sensors for peptide motifs.
Collapse
Affiliation(s)
- Jin Huang
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, U.S.A
| | - Shohei Koide
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, U.S.A
| |
Collapse
|
169
|
Selvakumar E, Rameshkumar N, Lee SG, Lee SJ, Park HS. In vivo Production of Functional Single-Chain Fv Fragment with an N-Terminal-Specific Bio-orthogonal Reactive Group. Chembiochem 2010; 11:498-501. [DOI: 10.1002/cbic.200900685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
170
|
Shui X, Huang J, Li YH, Xie PL, Li GC. Construction and selection of human Fab antibody phage display library of liver cancer. Hybridoma (Larchmt) 2010; 28:341-7. [PMID: 19857115 DOI: 10.1089/hyb.2009.0028] [Citation(s) in RCA: 196] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study was to construct the fully humanized anti-hepatoma Fab fragment phage libraries and select antibodies against hepatoma specifically. PBMCs of liver cancer patients were immunized in vitro with HpeG(2) cells and were then transformed by Epstein-Barr virus (EBV). After total RNA was extracted, the heavy chain Fd and kappa/lambda light chain were amplified by RT-PCR and cloned into the vector pComb3 to construct the libraries of Fab fragments. The libraries were then panned by HpeG(2) cells. By means of ELISA and immunochemistry, the Fab phage antibodies binding with hepatoma were selected and identified. The Fd and light chain PCR products were subsequently inserted into pComb3, and the volume of Fab libraries reached 1.7 x 10(7). The libraries were enriched about 138-fold by three cycles of panning. 540 phage clones were picked randomly. Using cell ELISA and immunohistochemistry with cultured cells, one clone Fab phage antibody, which had binding activity with hepatoma, was picked out. Fully humanized anti-hepatoma Fab antibody phage display libraries were constructed. One phage clone was selected and confirmed to specifically bind to hepatoma cells. The selected Fab antibody may be further developed and applied to clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Xuan Shui
- Cancer Research Institute, Xiang-Ya School of Medicine, Central South University, Hunan Province, China
| | | | | | | | | |
Collapse
|
171
|
Backovic M, Johansson DX, Klupp BG, Mettenleiter TC, Persson MAA, Rey FA. Efficient method for production of high yields of Fab fragments in Drosophila S2 cells. Protein Eng Des Sel 2010; 23:169-74. [PMID: 20100703 DOI: 10.1093/protein/gzp088] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Fab molecules are used as therapeutic agents, and are invaluable tools in structural biology. We report here a method for production of recombinant Fab in Drosophila S2 cells for use in structural biology. Stably transfected S2 cell lines expressing the Fab were created within weeks. The recombinant Fab was secreted, and after affinity and size exclusion chromatography, 16 mg of pure protein were obtained from a liter of cell culture. The Fab was functional and formed a complex with its cognate antigen as demonstrated by co-precipitation and size exclusion chromatography. Biochemical characterization indicated that the Fab from S2 cells is less extensively glycosylated than the Fab obtained by digestion of antibody produced in hybridoma cells, a feature that may be advantageous for the purposes of crystallogenesis. Taken together, obtaining recombinant Fab from the S2 cells has been a faster and considerably more cost-effective method compared with the enzymatic digestion of the monoclonal antibody.
Collapse
Affiliation(s)
- Marija Backovic
- Institut Pasteur, Unité de Virologie Structurale, Département de Virologie and CNRS Unité de Recherche Associée 3015, Paris, France.
| | | | | | | | | | | |
Collapse
|
172
|
Barakat MR, Kaiser PK. VEGF inhibitors for the treatment of neovascular age-related macular degeneration. Expert Opin Investig Drugs 2010; 18:637-46. [PMID: 19388880 DOI: 10.1517/13543780902855316] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in the Western world for those patients aged 50 years or older. Neovascular AMD, a subtype characterized by the growth of new, pathologic blood vessels, results in most of the cases of severe and rapid vision loss associated with AMD. A critical activator of angiogenesis in neovascular AMD is VEGF. Several therapies have been and are now being developed for neovascular AMD, with the goal of inhibiting VEGF. These VEGF inhibitors include the RNA aptamer pegaptanib, partial and full-length antibodies ranibizumab and bevacizumab, VEGF receptor decoy VEGF Trap, small interfering RNA-based therapies bevasiranib and AGN211745, sirolimus, and tyrosine kinase inhibitors including vatalanib, pazopanib, TG100801, TG101095, AG013958 and AL39324. At present, established therapies have met with great success in reducing the vision loss associated with neovascular AMD, whereas those still investigational in nature offer the potential for further advances.
Collapse
Affiliation(s)
- Mark Rami Barakat
- Cole Eye Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
173
|
Patel R, Andrien BA. Kinetic analysis of a monoclonal therapeutic antibody and its single-chain homolog by surface plasmon resonance. Anal Biochem 2010; 396:59-68. [DOI: 10.1016/j.ab.2009.08.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 10/20/2022]
|
174
|
Ishikawa T, Kobayashi N, Osawa C, Sawa E, Wakamatsu K. Prevention of Stirring-Induced Microparticle Formation in Monoclonal Antibody Solutions. Biol Pharm Bull 2010; 33:1043-6. [DOI: 10.1248/bpb.33.1043] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tomoyoshi Ishikawa
- Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Company Limited
- Department of Chemistry and Chemical Biology, Graduate School of Engineering, Gunma University
| | - Noritaka Kobayashi
- Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Company Limited
| | - Chie Osawa
- Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Company Limited
| | - Eiji Sawa
- Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Company Limited
| | - Kaori Wakamatsu
- Department of Chemistry and Chemical Biology, Graduate School of Engineering, Gunma University
| |
Collapse
|
175
|
Orcutt KD, Ackerman ME, Cieslewicz M, Quiroz E, Slusarczyk AL, Frangioni JV, Wittrup KD. A modular IgG-scFv bispecific antibody topology. Protein Eng Des Sel 2009; 23:221-8. [PMID: 20019028 DOI: 10.1093/protein/gzp077] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Here we present a bispecific antibody (bsAb) format in which a disulfide-stabilized scFv is fused to the C-terminus of the light chain of an IgG to create an IgG-scFv bifunctional antibody. When expressed in mammalian cells and purified by one-step protein A chromatography, the bsAb retains parental affinities of each binding domain, exhibits IgG-like stability and demonstrates in vivo IgG-like tumor targeting and blood clearance. The extension of the C-terminus of the light chain of an IgG with an scFv or even a smaller peptide does appear to disrupt disulfide bond formation between the light and heavy chains; however, this does not appear to affect binding, stability or in vivo properties of the IgG. Thus, we demonstrate here that the light chain of an IgG can be extended with an scFv without affecting IgG function and stability. This format serves as a standardized platform for the construction of functional bsAbs.
Collapse
Affiliation(s)
- Kelly Davis Orcutt
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
176
|
Williams BAR, Diehnelt CW, Belcher P, Greving M, Woodbury NW, Johnston SA, Chaput JC. Creating protein affinity reagents by combining peptide ligands on synthetic DNA scaffolds. J Am Chem Soc 2009; 131:17233-41. [PMID: 19894711 PMCID: PMC2796340 DOI: 10.1021/ja9051735] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A full understanding of the proteome will require ligands to all of the proteins encoded by genomes. While antibodies represent the principle affinity reagents used to bind proteins, their limitations have created a need for new ligands to large numbers of proteins. Here we propose a general concept to obtain protein affinity reagents that avoids animal immunization and iterative selection steps. Central to this process is the idea that small peptide libraries contain sequences that will bind to independent regions on a protein surface and that these ligands can be combined on synthetic scaffolds to create high affinity bivalent reagents. To demonstrate the feasibility of this approach, an array of 4000 unique 12-mer peptides was screened to identify sequences that bind to nonoverlapping sites on the yeast regulatory protein Gal80. Individual peptide ligands were screened at different distances using a novel DNA linking strategy to identify the optimal peptide pair and peptide pair separation distance required to transform two weaker ligands into a single high affinity protein capture reagent. A synthetic antibody or synbody was created with 5 nM affinity to Gal80 that functions in conventional ELISA and pull-down assays. We validated our synthetic antibody approach by creating a second synbody to human transferrin. In both cases, we observed an increase in binding affinity of approximately 1000-fold (DeltaDeltaG = approximately 4.1 kcal/mol) between the individual peptides and final bivalent synbody construct.
Collapse
Affiliation(s)
- Berea A. R. Williams
- Center for BioOptical Nanotechnology, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287-5201
| | - Chris W. Diehnelt
- Center for Innovations in Medicine, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
| | - Paul Belcher
- Center for Innovations in Medicine, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
| | - Matthew Greving
- Center for BioOptical Nanotechnology, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287-5201
| | - Neal W. Woodbury
- Center for BioOptical Nanotechnology, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287-5201
| | - Stephen A. Johnston
- Center for Innovations in Medicine, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287-5201
| | - John C. Chaput
- Center for BioOptical Nanotechnology, The Biodesign Institute, Arizona State University, Tempe, Arizona 85287-5201
- Department of Chemistry and Biochemistry, Arizona State University, Tempe, Arizona 85287-5201
| |
Collapse
|
177
|
Bressler SB. Introduction: Understanding the role of angiogenesis and antiangiogenic agents in age-related macular degeneration. Ophthalmology 2009; 116:S1-7. [PMID: 19800534 DOI: 10.1016/j.ophtha.2009.06.045] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 06/22/2009] [Indexed: 11/18/2022] Open
Abstract
Inhibition of angiogenesis is critical in the prevention and treatment of neovascular age-related macular degeneration (AMD). Pathologic states such as hypoxia, ischemia, or inflammation may tip the balance of proangiogenic and antiangiogenic factors in favor of the formation of new blood vessels. Vascular endothelial growth factor (VEGF) is pivotal in ocular angiogenesis because it is highly selective for endothelial cells, hypoxia drives its synthesis, it diffuses to its target, and it affects multiple components of angiogenesis such as endothelial cell proliferation, survival, and migration. Basic and clinical research implicates VEGF in the pathogenesis of choroidal neovascularization (CNV), although other candidate factors involved with regulation of angiogenesis exist. Intravitreal drugs that block VEGF have revolutionized the care of patients with neovascular AMD, decreasing growth and leakage from choroidal neovascular lesions and preventing moderate and severe vision loss associated with this process.
Collapse
Affiliation(s)
- Susan B Bressler
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| |
Collapse
|
178
|
Leucine-Aspartic Acid-Valine Sequence as Targeting Ligand and Drug Carrier for Doxorubicin Delivery to Melanoma Cells: In Vitro Cellular Uptake and Cytotoxicity Studies. Pharm Res 2009; 26:2578-87. [DOI: 10.1007/s11095-009-9971-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 09/14/2009] [Indexed: 01/14/2023]
|
179
|
Abstract
Immunotherapy for treating illicit drug abuse is a rapidly advancing field. There are currently two major approaches to developing drug-specific immunotherapies: active and passive. Active immunotherapy involves conjugating a drug-like hapten to a carrier protein and using traditional immunization approaches to generate a drug-specific immune response in the patient. In contrast, passive immunotherapy utilizes preformed monoclonal antibodies. Whether generated by active immunization or delivered passively, antibodies act as pharmacokinetic antagonists by binding the drug in the blood-stream and reducing the amount and rate of drug delivery to receptors in the brain. A newly emerging technology in anti-drug immunotherapy is the use of antibody fragments, or scFvs, rather than intact immunoglobulin G (IgG). These scFvs can retain the same binding properties as the original mAbs, and are onethird the molecular weight, providing a scaffold for creating antibody treatments with more customizable properties. Another nascent area of research utilizing the scFv scaffold is in creating drug-specific scFv-nanoparticle conjugates. These conjugates could improve upon current drug-specific antibody paradigms by increasing multivalency and allowing pharmacokinetic customization, while avoiding interactions with endogenous antibody receptor pathways. These parallel approaches to immunotherapy are moving rapidly toward the clinic and may soon provide new therapies for treating drug abuse.
Collapse
Affiliation(s)
- Eric C Peterson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 72205, USA.
| | | |
Collapse
|
180
|
Abstract
The podocytopathies, including minimal-change nephropathy, focal segmental glomerulosclerosis, collapsing glomerulopathy, and diffuse mesangial sclerosis, involve diverse types of injury to podocytes. These injuries can have genetic causes, or can be caused by viral infection, mechanical stress, medication or-probably-immunologic injury. Several lines of evidence-including the immunosuppressive effects of standard therapies-suggest a role for immunologic injury in some cases, but the precise pathologic mechanisms are far from clear. Despite this uncertainty, newly available biologic therapies that target immune cells and cytokines have been used to treat a number of patients with different podocytopathies. Of these therapies, the greatest experience has been gained with rituximab. The data on all such therapies remain too fragmentary to provide firm conclusions, but further clinical research with such agents might help to define pathogenetic pathways and could potentially contribute to new therapies.
Collapse
|
181
|
Ayyadurai N, Neelamegam R, Nagasundarapandian S, Edwardraja S, Park HS, Lee SJ, Yoo TH, Yoon H, Lee SG. Importance of expression system in the production of unnatural recombinant proteins in Escherichia coli. BIOTECHNOL BIOPROC E 2009. [DOI: 10.1007/s12257-009-0009-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
182
|
Löfdahl PA, Nord O, Janzon L, Nygren PA. Selection of TNF-alpha binding affibody molecules using a beta-lactamase protein fragment complementation assay. N Biotechnol 2009; 26:251-9. [PMID: 19576305 DOI: 10.1016/j.nbt.2009.06.980] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 06/11/2009] [Accepted: 06/17/2009] [Indexed: 11/30/2022]
Abstract
Protein fragment complementation assays (PCAs) based on different reporter proteins have been described as powerful tools for monitoring dynamic protein-protein interactions in living cells. The present study describes the construction of a PCA system based on genetic splitting of TEM-1 beta-lactamase for the selection of proteins specifically interacting in the periplasm of Escherichia coli bacterial cells, and its application for the selection of affibody molecules binding human tumour necrosis factor-alpha (TNF-alpha) from a combinatorial library. Vectors encoding individual members of a naïve 10(9) affibody protein library fused to a C-terminal fragment of the beta-lactamase reporter were distributed via phage infection to a culture of cells harbouring a common construct encoding a fusion protein between a non-membrane anchored version of a human TNF-alpha target and the N-terminal segment of the reporter. An initial binding analysis of 29 library variants derived from surviving colonies using selection plates containing ampicillin and in some cases also the beta-lactamase inhibitor tazobactam, indicated a stringent selection for target binding variants. Subsequent analyses showed that the binding affinities (K(D)) for three selected variants studied in more detail were in the range 14-27 nm. The selectivity in binding to TNF-alpha for these variants was further demonstrated in both a cross-target PCA-based challenge and the specific detection of a low nm concentration of TNF-alpha spiked into a complex cell lysate sample. Further, in a biosensor-based competition assay, the binding to TNF-alpha of three investigated affibody variants could be completely blocked by premixing the target with the therapeutic monoclonal antibody adalimumab (Humira), indicating overlapping epitopes between the two classes of reagents. The data indicate that beta-lactamase PCA is a promising methodology for stringent selection of binders from complex naïve libraries to yield high affinity reagents with selective target binding characteristics.
Collapse
Affiliation(s)
- P-A Löfdahl
- Division of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), SE-106 91 Stockholm, Sweden
| | | | | | | |
Collapse
|
183
|
Silva HM, Vieira PMMM, Costa PLN, Pimentel BMS, Moro AM, Kalil J, Maranhão AQ, Coelho V, Brigido MM. Novel humanized anti-CD3 antibodies induce a predominantly immunoregulatory profile in human peripheral blood mononuclear cells. Immunol Lett 2009; 125:129-36. [PMID: 19573559 DOI: 10.1016/j.imlet.2009.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 06/15/2009] [Accepted: 06/22/2009] [Indexed: 01/12/2023]
Abstract
Strategies to minimize the immunogenicity and toxicity of murine anti-CD3 antibodies (e.g. OKT3) are of special interest for organ transplantation and for the treatment of autoimmune diseases. In the present work, we have developed two humanized anti-CD3 antibodies. These molecules were shown to bind to human CD3, though less efficiently, and display less mitogenic activity than OKT3. These results prompted us to investigate whether this reduced mitogenic potential was associated with the development of anti-inflammatory properties. Indeed, in peripheral blood mononuclear cells (PBMCs), the humanized antibody versions induced a predominantly anti-inflammatory cytokine profile, in contrast with the pro-inflammatory profile induced by OKT3. Neither OKT3 nor the humanized versions induced the expression of IL-4, IL-2 or TGF-beta. Both humanized antibodies induced significantly lower production of IFN-gamma and IL-5 and slightly higher production of IL-10 than OKT3. This immunomodulatory profile was most evident by the 80-fold higher ratio of IL-10/IFN-gamma production in PBMCs cultured in the presence of the humanized antibodies, compared to those stimulated with OKT3. Furthermore, these humanized anti-CD3 antibodies induced a late FOXP3 gene expression while OKT3 led to a more transient expression of FOXP3. Taken our results, we suggest that these humanized anti-CD3 antibodies may promote the development of T cells with immunoregulatory activity.
Collapse
Affiliation(s)
- Hernandez M Silva
- Departamento de Biologia Celular, Universidade de Brasília, 70910-900 Brasília, DF, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
|
185
|
Antibody-based sensors: principles, problems and potential for detection of pathogens and associated toxins. SENSORS 2009; 9:4407-45. [PMID: 22408533 PMCID: PMC3291918 DOI: 10.3390/s90604407] [Citation(s) in RCA: 190] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 05/26/2009] [Accepted: 05/26/2009] [Indexed: 01/30/2023]
Abstract
Antibody-based sensors permit the rapid and sensitive analysis of a range of pathogens and associated toxins. A critical assessment of the implementation of such formats is provided, with reference to their principles, problems and potential for 'on-site' analysis. Particular emphasis is placed on the detection of foodborne bacterial pathogens, such as Escherichia coli and Listeria monocytogenes, and additional examples relating to the monitoring of fungal pathogens, viruses, mycotoxins, marine toxins and parasites are also provided.
Collapse
|
186
|
Targeting protein–protein interactions for therapeutic intervention: a challenge for the future. Future Med Chem 2009; 1:65-93. [DOI: 10.4155/fmc.09.12] [Citation(s) in RCA: 187] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Over the last two decades, an increasing research effort in academia and industry has focused on the modulation (both inhibition and stabilization) of protein–protein interactions (PPIs) in order to develop novel therapeutic approaches and target-selective agents in drug discovery. Discussion: The diversity and complexity of highly dynamic systems such as PPIs present many challenges for the identification of drug-like molecules with the ability to modulate the PPI with the necessary selectivity and potency. In this review, a number of these strategies will be presented along with a critical overview of the challenges and potential solutions relating to the exploitation of PPIs as molecular targets. Conclusions: Both traditional drug discovery approaches and some more recently developed innovative strategies have already provided valuable tools for the discovery of PPI modulators, and a number of successful examples have highlighted the potential of targeting PPIs for therapeutic intervention, especially in the oncology area.
Collapse
|
187
|
Gupta A, Heimann AS, Gomes I, Devi LA. Antibodies against G-protein coupled receptors: novel uses in screening and drug development. Comb Chem High Throughput Screen 2009; 11:463-7. [PMID: 18673273 DOI: 10.2174/138620708784911465] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Antibodies are components of the body's humoral immune system that are generated in response to foreign pathogens. Modern biomedical research has employed these very specific and efficient molecules designed by nature in the diagnosis of diseases, localization of gene products as well as in the rapid screening of targets for drug discovery and testing. In addition, the introduction of antibodies with fluorescent or enzymatic tags has significantly contributed to advances in imaging and microarray technology, which are revolutionizing disease research and the search for effective therapeutics. More recently antibodies have been used in the isolation of dimeric G protein-coupled receptor (GPCR) complexes. In this review, we discuss antibodies as powerful research tools for studying GPCRs, and their potential to be developed as drugs themselves.
Collapse
Affiliation(s)
- Achla Gupta
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | | | | | |
Collapse
|
188
|
|
189
|
Paclitaxel palmitate-loaded anti-HER2 cationic immunoemulsion: pharmacokinetic and biodistribution study in healthy mice. J Drug Deliv Sci Technol 2009. [DOI: 10.1016/s1773-2247(09)50051-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
190
|
Rodrigues ME, Costa AR, Henriques M, Azeredo J, Oliveira R. Technological progresses in monoclonal antibody production systems. Biotechnol Prog 2009; 26:332-51. [DOI: 10.1002/btpr.348] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
191
|
Schirrmann T, Krauss J, Arndt MAE, Rybak SM, Dübel S. Targeted therapeutic RNases (ImmunoRNases). Expert Opin Biol Ther 2008; 9:79-95. [DOI: 10.1517/14712590802631862] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
192
|
Anderson GP, Goldman ER. TNT detection using llama antibodies and a two-step competitive fluid array immunoassay. J Immunol Methods 2008; 339:47-54. [DOI: 10.1016/j.jim.2008.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Accepted: 08/05/2008] [Indexed: 11/25/2022]
|
193
|
Tijink BM, Laeremans T, Budde M, Stigter-van Walsum M, Dreier T, de Haard HJ, Leemans CR, van Dongen GAMS. Improved tumor targeting of anti-epidermal growth factor receptor Nanobodies through albumin binding: taking advantage of modular Nanobody technology. Mol Cancer Ther 2008; 7:2288-97. [PMID: 18723476 DOI: 10.1158/1535-7163.mct-07-2384] [Citation(s) in RCA: 215] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The approximately 15-kDa variable domains of camelid heavy-chain-only antibodies (called Nanobodies) can easily be formatted as multivalent or multispecific single-chain proteins. Because of fast excretion, however, they are less suitable for therapy of cancer. In this study, we aimed for improved tumor targeting of a bivalent anti-epidermal growth factor receptor (EGFR) Nanobody (alphaEGFR-alphaEGFR) by fusion to a Nanobody unit binding to albumin (alphaAlb). Biodistributions of alphaEGFR-alphaEGFR, alphaEGFR-alphaEGFR-alphaAlb ( approximately 50 kDa), alphaTNF-alphaTNF-alphaAlb (control, binding tumor necrosis factor-alpha), and the approximately 150-kDa anti-EGFR antibody cetuximab were compared in A431 xenograft-bearing mice. The proteins were radiolabeled with (177)Lu to facilitate quantification. Tumor uptake of (177)Lu-alphaEGFR-alphaEGFR decreased from 5.0 +/- 1.4 to 1.1 +/- 0.1 %ID/g between 6 and 72 h after injection. Due to its rapid blood clearance, tumor-to-blood ratios >80 were obtained within 6 h after injection. Blood clearance became dramatically slower and tumor uptake became significantly higher by introduction of alphaAlb. Blood levels of alphaEGFR-alphaEGFR-alphaAlb were 21.2 +/- 2.5, 11.9 +/- 0.6, and 4.0 +/- 1.4 and tumor levels were 19.4 +/- 5.5, 35.2 +/- 7.5, and 28.0 +/- 6.8 %ID/g at 6, 24, and 72 h after injection, respectively. Tumor uptake was at least as high as for cetuximab (15.5 +/- 3.9, 27.1 +/- 7.9, and 25.6 +/- 6.1 %ID/g) and significantly higher than for alphaTNF-alphaTNF-alphaAlb. alphaEGFR-alphaEGFR-alphaAlb showed faster and deeper tumor penetration than cetuximab. These data show that simple fusion of alphaEGFR and alphaAlb building blocks results in a bifunctional Nanobody format, which seems more favorable for therapy as far as pharmacokinetics and tumor deposition are concerned.
Collapse
Affiliation(s)
- Bernard M Tijink
- Department of Otolaryngology/Head and Neck Surgery, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Debbage P, Jaschke W. Molecular imaging with nanoparticles: giant roles for dwarf actors. Histochem Cell Biol 2008; 130:845-75. [PMID: 18825403 DOI: 10.1007/s00418-008-0511-y] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2008] [Indexed: 10/25/2022]
Abstract
Molecular imaging, first developed to localise antigens in light microscopy, now encompasses all imaging modalities including those used in clinical care: optical imaging, nuclear medical imaging, ultrasound imaging, CT, MRI, and photoacoustic imaging. Molecular imaging always requires accumulation of contrast agent in the target site, often achieved most efficiently by steering nanoparticles containing contrast agent into the target. This entails accessing target molecules hidden behind tissue barriers, necessitating the use of targeting groups. For imaging modalities with low sensitivity, nanoparticles bearing multiple contrast groups provide signal amplification. The same nanoparticles can in principle deliver both contrast medium and drug, allowing monitoring of biodistribution and therapeutic activity simultaneously (theranostics). Nanoparticles with multiple bioadhesive sites for target recognition and binding will be larger than 20 nm diameter. They share functionalities with many subcellular organelles (ribosomes, proteasomes, ion channels, and transport vesicles) and are of similar sizes. The materials used to synthesise nanoparticles include natural proteins and polymers, artificial polymers, dendrimers, fullerenes and other carbon-based structures, lipid-water micelles, viral capsids, metals, metal oxides, and ceramics. Signal generators incorporated into nanoparticles include iron oxide, gadolinium, fluorine, iodine, bismuth, radionuclides, quantum dots, and metal nanoclusters. Diagnostic imaging applications, now appearing, include sentinal node localisation and stem cell tracking.
Collapse
Affiliation(s)
- Paul Debbage
- Department of Anatomy, Division of Histology and Embryology, Medical University Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria.
| | | |
Collapse
|
195
|
Abstract
For the last 30 years, the production of affinity reagents and particularly antibodies for research and therapeutic applications has been dominated by hybridoma and polyclonal technologies, while more modern, reliable and inexpensive approaches have lagged. Here we discuss why this is the case and how a cultural shift in the biomedical research community could bring the new technologies for creating antibodies and other tailor-designed binding proteins into the mainstream, with the potential for myriad new applications in research and medicine.
Collapse
|
196
|
Kelly MP, Lee FT, Tahtis K, Power BE, Smyth FE, Brechbiel MW, Hudson PJ, Scott AM. Tumor targeting by a multivalent single-chain Fv (scFv) anti-Lewis Y antibody construct. Cancer Biother Radiopharm 2008; 23:411-23. [PMID: 18771345 PMCID: PMC2663784 DOI: 10.1089/cbr.2007.0450] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The use of single-chain variable fragment (scFv) constructs has been investigated in cancer radioimmunotherapy (RIT) and radioimmunodetection, as these molecules permit rapid tumor penetration and clearance from the serum relative to whole IgG. Multimerization of scFv constructs has demonstrated improvements in functional affinity (i.e., avidity) and maximal tumor uptake. In this paper, we report the first biodistribution and pharmacokinetics studies of a noncovalent, direct-linked scFv (V(L)-0-V(H)) trimeric/tetrameric "multimer" of the anti-Lewis Y monoclonal antibody, hu3S193. The in vitro binding and in vivo biodistribution of the hu3S193 multimer was characterized alongside the hu3S193 F(ab')(2) following radiolabeling with the Indium-111 ((111)In) radioisotope. Immunoreactivities of the radiolabeled multimer and F(ab')(2) were 73% and 53.2%, and binding affinities (K(a)) were 1.58 x 10(7) M(1) and 4.31 x 10(6) M (1) for the multimer and F(ab')(2), respectively. Maximal tumor uptake in Le(y)-positive MCF-7 breast cancer xenografted BALB/c nude mice was 12.6 +/- 2.5 percent injected dose/per gram (%ID/g) at 6 hours postinjection for the multimer and 15.7 +/- 2.1 %ID/g at 24 hours postinjection for the F(ab')(2). However, limited in vitro stability and high renal localization of radiolabeled constructs were observed, which, despite the observed tumor targeting of the hu3S193 multimer, most likely preclude its use in RIT and imaging modalities.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal, Humanized
- Area Under Curve
- Cell Line, Tumor
- Chromatography, Gel
- Drug Stability
- Female
- Humans
- Immunoconjugates/blood
- Immunoconjugates/pharmacokinetics
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/immunology
- Immunoglobulin Fab Fragments/metabolism
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Immunoglobulin Variable Region/metabolism
- Indium Radioisotopes
- Lewis Blood Group Antigens/immunology
- Mammary Neoplasms, Experimental/diagnostic imaging
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasms/diagnostic imaging
- Neoplasms/immunology
- Neoplasms/metabolism
- Radionuclide Imaging
- Recombinant Proteins/blood
- Recombinant Proteins/immunology
- Recombinant Proteins/pharmacokinetics
- Tissue Distribution
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Marcus P. Kelly
- Tumour Targeting Program, Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria, Australia
| | - F.-T. Lee
- Tumour Targeting Program, Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria, Australia
| | - Kiki Tahtis
- Tumour Targeting Program, Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria, Australia
| | - Barbara E. Power
- CSIRO Molecular and Health Technologies, Parkville, Victoria, Australia
| | - Fiona E. Smyth
- Tumour Targeting Program, Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria, Australia
| | - Martin W. Brechbiel
- Radioimmune & Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Peter J. Hudson
- CSIRO Molecular and Health Technologies, Parkville, Victoria, Australia
| | - Andrew M. Scott
- Tumour Targeting Program, Ludwig Institute for Cancer Research, Austin Hospital, Heidelberg, Victoria, Australia
| |
Collapse
|
197
|
Hofbauer KG, Lecourt AC, Peter JC. Antibodies as pharmacologic tools for studies on the regulation of energy balance. Nutrition 2008; 24:791-7. [PMID: 18662861 DOI: 10.1016/j.nut.2008.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Accepted: 06/03/2008] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Active immunization in rats may serve several purposes: the production of a disease-like phenotype, the generation of pharmacologic tools, and the development of clinically useful therapies. We selected the melanocortin-4 receptor (MC4R) as a target because its blockade could provide a treatment for anorexia and cachexia. METHODS We used a sequence of the N-terminal (NT) domain of the MC4R as an antigen. Rats immunized against the NT peptide produced specific MC4R antibodies (Abs) that were purified and characterized in vitro and in vivo. RESULTS The Abs acted as inverse agonists and reduced under basal conditions the production of cyclic adenosine monophosphate in HEK-293 cells expressing the human MC4R. Rats immunized against the NT peptide developed a phenotype consistent with hypothalamic MC4R blockade, i.e., increased food intake and body weight, liver and fat-pad weights, hepatic steatosis, and increased plasma triacylglycerols. With a high-fat diet, plasma insulin levels were significantly increased. In separate experiments an increase in food intake was observed after injection of purified MC4R Abs into the third ventricle. When lipopolysaccharide was administered in NT-immunized rats the reduction of food intake was partly prevented in this model of cytokine-induced anorexia. CONCLUSION Our results show that active immunization of rats against the MC4R resulted in the generation of specific Abs that stimulated food intake by acting as inverse agonists of the hypothalamic MC4R. Pharmacologically active monoclonal MC4R Abs could be the starting point for the development of novel treatments for patients with anorexia or cachexia.
Collapse
Affiliation(s)
- Karl G Hofbauer
- Applied Pharmacology, Biozentrum, University of Basel, Basel, Switzerland.
| | | | | |
Collapse
|
198
|
Simple method for production of randomized human tenth fibronectin domain III libraries for use in combinatorial screening procedures. Biotechniques 2008; 44:559-62. [PMID: 18476822 DOI: 10.2144/000112726] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Challenges such as the rapid development of detection reagents for emerging or engineered pathogens, the goal of identifying probes for every protein in the human proteome, and the development of therapeutic molecules require systems for development of epitope binding molecules that are faster and cheaper than conventional antibody development. To be practical and effective, antibody mimics must be small, stable molecules that contain exposed loops or surfaces that can be randomized and screened using selective combinatorial assays. The tenth human fibronectin type III domain (10Fn3) fits these requirements and has recently been developed as an antibody mimic for use in detection and therapeutic platforms. Previously described systems for working with 10Fn3 used PCR-based approaches to anneal multiple oligonucleotides to generate randomized 10Fn3 libraries. Here we describe a simplified approach for creating randomized 10Fn3 libraries and report the first use of a T7-based phage display system for screening these libraries.
Collapse
|
199
|
Wang CE, Zhou H, McGuire JR, Cerullo V, Lee B, Li SH, Li XJ. Suppression of neuropil aggregates and neurological symptoms by an intracellular antibody implicates the cytoplasmic toxicity of mutant huntingtin. J Cell Biol 2008; 181:803-16. [PMID: 18504298 PMCID: PMC2396813 DOI: 10.1083/jcb.200710158] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Accepted: 04/30/2008] [Indexed: 12/03/2022] Open
Abstract
Mutant huntingtin accumulates in the neuronal nuclei and processes, which suggests that its subcellular localization is critical for the pathology of Huntington's disease (HD). However, the contribution of cytoplasmic mutant huntingtin and its aggregates in neuronal processes (neuropil aggregates) has not been rigorously explored. We generated an intracellular antibody (intrabody) whose binding to a unique epitope of human huntingtin is enhanced by polyglutamine expansion. This intrabody decreases the cytotoxicity of mutant huntingtin and its distribution in neuronal processes. When expressed in the striatum of HD mice via adenoviral infection, the intrabody reduces neuropil aggregate formation and ameliorates neurological symptoms. Interaction of the intrabody with mutant huntingtin increases the ubiquitination of cytoplasmic huntingtin and its degradation. These findings suggest that the intrabody reduces the specific neurotoxicity of cytoplasmic mutant huntingtin and its associated neurological symptoms by preventing the accumulation of mutant huntingtin in neuronal processes and promoting its clearance in the cytoplasm.
Collapse
Affiliation(s)
- Chuan-En Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
200
|
Rogers J, Schoepp RJ, Schröder O, Clements TL, Holland TF, Li JQ, Li J, Lewis LM, Dirmeier RP, Frey GJ, Tan X, Wong K, Woodnutt G, Keller M, Reed DS, Kimmel BE, Tozer EC. Rapid discovery and optimization of therapeutic antibodies against emerging infectious diseases. Protein Eng Des Sel 2008; 21:495-505. [PMID: 18480090 PMCID: PMC2461042 DOI: 10.1093/protein/gzn027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Using a comprehensive set of discovery and optimization tools, antibodies were produced with the ability to neutralize SARS coronavirus (SARS-CoV) infection in Vero E6 cells and in animal models. These anti-SARS antibodies were discovered using a novel DNA display method, which can identify new antibodies within days. Once neutralizing antibodies were identified, a comprehensive and effective means of converting the mouse sequences to human frameworks was accomplished using HuFR™ (human framework reassembly) technology. The best variant (61G4) from this screen showed a 3.5–4-fold improvement in neutralization of SARS-CoV infection in vitro. Finally, using a complete site-saturation mutagenesis methodology focused on the CDR (complementarity determining regions), a single point mutation (51E7) was identified that improved the 80% plaque reduction neutralization of the virus by greater than 8-fold. These discovery and evolution strategies can be applied to any emerging pathogen or toxin where a causative agent is known.
Collapse
Affiliation(s)
- J Rogers
- Verenium Corporation, 4955 Directors Place, San Diego, CA 92121, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|