1
|
Wehrmüller JE, Frei JC, Hechler T, Kulke M, Pahl A, Béhé M, Schibli R, Spycher PR. Site-Specific Modification of Native IgGs with Flexible Drug-Load. Chembiochem 2025; 26:e202400511. [PMID: 39305147 PMCID: PMC12007069 DOI: 10.1002/cbic.202400511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/11/2024] [Indexed: 11/08/2024]
Abstract
Homogeneous, site-specifically conjugated antibodies have shown to result in antibody-drug conjugates (ADCs) with improved efficacy and tolerability compared to stochastically conjugated ADCs. However, precisely controlling the drug load as well as attaching multiple payload moieties to the antibody remains challenging. Here, we demonstrate the simple and direct modification of native IgG-antibodies at the residue glutamine 295 (Q295) without the need for any protein engineering with flexible drug-to-antibody ratios of one or multiple payloads. The conjugation is enabled through short, positively charged lysine containing peptides and native, commercially available microbial transglutaminase. In proof-of-concept studies, HER2-targeting ADCs based on trastuzumab were generated with drug-to-antibody ratios (DARs) of 2 and 4 of the same or different payloads using orthogonal conjugation chemistries. Quantitative biodistribution studies performed with 111In-radiolabeled conjugates showed high tumour uptake and low accumulation of radioactivity in non-targeted tissues. A single dose study of trastuzumab conjugated to the highly potent payload α-Amanitin demonstrated complete and long-lasting tumour remission and was well-tolerated at all dose levels tested.
Collapse
Affiliation(s)
- Jöri E. Wehrmüller
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Julia C. Frei
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Torsten Hechler
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Michael Kulke
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Andreas Pahl
- Heidelberg Pharma Research GmbHGregor-Mendel-Straße 2268526LadenburgGermany
| | - Martin Béhé
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
| | - Roger Schibli
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
- Department of Chemistry and Applied BiosciencesETH Zürich8093ZürichSwitzerland
| | - Philipp R. Spycher
- Center for Radiopharmaceutical ScienceETH-PSI-USZPaul Scherrer Institute5232Villigen-PSISwitzerland
- Araris Biotech AGRiedhofstrasse 118804Au ZHSwitzerland
| |
Collapse
|
2
|
Wu Z, Mo L, Wang Z, Song L, Kobatake E, Ito Y, Wang Y, Zhang P. Biointerface engineering through amalgamation of gene technology and site-specific growth factor conjugation for efficient osteodifferentiation. Biotechnol Bioeng 2025; 122:80-94. [PMID: 39300684 DOI: 10.1002/bit.28852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/21/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
The development of bone implants through bioinspired immobilization of growth factors remains a key issue in the generation of biological interfaces, especially in enhancing osteodifferentiation ability. In this study, we developed a strategy for surface functionalization of poly(lactide-glycolide) (PLGA) and hydroxyapatite (HA) composite substrates through site-specific conjugation of bone morphogenetic protein 2 containing 3,4-hydroxyphenalyalanine (DOPA-BMP2) mediated by tyrosinase and sortase A (SrtA). Firstly, the growth factor BMP2-LPETG containing LPETG motif was successfully expressed in Escherichia coli through recombinant DNA technology. The excellent binding affinity of binding growth factor (DOPA-BMP2) was achieved by converting the tyrosine residue (Y) of YKYKY-GGG peptide into DOPA (X) by tyrosinase, which bound to the substrates. Then its GGG motif was specifically bound to the end of BMP2-LPETG mediated by SrtA. Therefore, the generated bioactive DOPA-BMP2/PLGA/HA substrates significantly promoted the osteogenic differentiation of MC3T3-E1 cells. Thanks to this microbial-assisted engineering approach, our work presents a facile and highly site-specific strategy to engineer biomimetic materials for orthopedics and dentistry by effectively delivering growth factors, peptides, and other biomacromolecules.
Collapse
Affiliation(s)
- Zhenxu Wu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China
| | - Li Mo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Zongliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China
| | - Liangsong Song
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Eiry Kobatake
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8502, Japan
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN Cluster for Pioneering Research, Saitama, 351-0198, Japan
| | - Yi Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Peibiao Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China
| |
Collapse
|
3
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
4
|
Sela T, Mansø M, Siegel M, Marban-Doran C, Ducret A, Niewöhner J, Ravn J, Martin RE, Sommer A, Lohmann S, Krippendorff BF, Ladefoged M, Indlekofer A, Quaiser T, Bueddefeld F, Koller E, Mohamed MY, Oelschlaegel T, Gothelf KV, Hofer K, Schumacher FF. Diligent Design Enables Antibody-ASO Conjugates with Optimal Pharmacokinetic Properties. Bioconjug Chem 2023; 34:2096-2111. [PMID: 37916986 DOI: 10.1021/acs.bioconjchem.3c00393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Antisense-oligonucleotides (ASOs) are a promising drug modality for the treatment of neurological disorders, but the currently established route of administration via intrathecal delivery is a major limitation to its broader clinical application. An attractive alternative is the conjugation of the ASO to an antibody that facilitates access to the central nervous system (CNS) after peripheral application and target engagement at the blood-brain barrier, followed by transcytosis. Here, we show that the diligent conjugate design of Brainshuttle-ASO conjugates is the key to generating promising delivery vehicles and thereby establishing design principles to create optimized molecules with drug-like properties. An innovative site-specific transglutaminase-based conjugation technology was chosen and optimized in a stepwise process to identify the best-suited conjugation site, tags, reaction conditions, and linker design. The overall conjugation performance was found to be specifically governed by the choice of buffer conditions and the structure of the linker. The combination of the peptide tags YRYRQ and RYESK was chosen, showing high conjugation fidelity. Elaborate conjugate analysis revealed that one leading differentiating factor was hydrophobicity. The increase of hydrophobicity by the ASO payload could be mitigated by the appropriate choice of conjugation site and the heavy chain position 297 proved to be the most optimal. Evaluating the properties of the linker suggested a short bicyclo[6.1.0]nonyne (BCN) unit as best suited with regards to conjugation performance and potency. Promising in vitro activity and in vivo pharmacokinetic behavior of optimized Brainshuttle-ASO conjugates, based on a microtubule-associated protein tau (MAPT) targeting oligonucleotide, suggest that such designs have the potential to serve as a blueprint for peripherally delivered ASO-based drugs for the CNS in the future.
Collapse
Affiliation(s)
- Tatjana Sela
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
- Department of Biochemistry, Ludwig-Maximilians-Universität, Munich 80539, Germany
| | - Mads Mansø
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Michel Siegel
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Céline Marban-Doran
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Axel Ducret
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Jens Niewöhner
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Jacob Ravn
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Rainer E Martin
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Annika Sommer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Sabine Lohmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Ben-Fillippo Krippendorff
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Mette Ladefoged
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Annette Indlekofer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Tom Quaiser
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Florian Bueddefeld
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Erich Koller
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | | | | | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus 8000, Central Denmark Region, Denmark
| | - Kerstin Hofer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Felix F Schumacher
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
5
|
Zhao Y, Kim S, Zheng X, Kim SH, Han A, Chen TH, Wang S, Zhong J, Qiu H, Li N. Investigation of High Molecular Weight Size Variant Formation in Antibody-Drug Conjugates: Microbial Transglutaminase-Mediated Crosslinking. J Pharm Sci 2023; 112:2629-2636. [PMID: 37586591 DOI: 10.1016/j.xphs.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
Microbial transglutaminase (mTG) has become a powerful tool for manufacturing antibody-drug conjugates (ADCs). It enables site-specific conjugation by catalyzing formation of stable isopeptide bond between glutamine (Q) side chain and primary amine. However, the downstream impact of mTG-mediated conjugation on ADC product quality, especially on high molecular weight (HMW) size variant formation has not been studied in a systematic manner. This study investigates the mechanisms underlying the formation of HMW size variants in mTG-mediated ADCs using size exclusion chromatography (SEC) and liquid chromatography-mass spectrometry (LC-MS). Our findings revealed that the mTG-mediated glutamine and lysine (K) crosslinking is the primary source of the increased level of HMW size variants in the ADCs. In the study, two monoclonal antibodies (mAbs) with glutamine engineered for site-specific conjugation were used as model systems. Based on the LC-MS analysis, a single lysine (K56) in the heavy chain (HC) was identified as the major Q-K crosslinking site in one of the two mAbs. The HC C-terminal K was observed to crosslink to the target Q in both mAbs. Quantitative correlation was established between the percentage of HMW size variants determined by SEC and the percentage of crosslinked peptides quantified by MS peptide mapping. Importantly, it was demonstrated that the level of HMW size variants in the second ADC was substantially reduced by the complete removal of HC C-terminal K before conjugation. The current work demonstrates that crosslinking and other side reactions during mTG-mediated conjugation needs to be carefully monitored and controlled to ensure process consistency and high product quality of the final ADC drug product.
Collapse
Affiliation(s)
- Yimeng Zhao
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA.
| | - Sunnie Kim
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Xiang Zheng
- Therapeutic Proteins, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Se Hyun Kim
- Therapeutic Proteins, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Amy Han
- Therapeutic Proteins, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Tse-Hong Chen
- Formulation Development, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Serena Wang
- Formulation Development, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Jieqiang Zhong
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Haibo Qiu
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA.
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| |
Collapse
|
6
|
Debon A, Siirola E, Snajdrova R. Enzymatic Bioconjugation: A Perspective from the Pharmaceutical Industry. JACS AU 2023; 3:1267-1283. [PMID: 37234110 PMCID: PMC10207132 DOI: 10.1021/jacsau.2c00617] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 05/27/2023]
Abstract
Enzymes have firmly established themselves as bespoke catalysts for small molecule transformations in the pharmaceutical industry, from early research and development stages to large-scale production. In principle, their exquisite selectivity and rate acceleration can also be leveraged for modifying macromolecules to form bioconjugates. However, available catalysts face stiff competition from other bioorthogonal chemistries. In this Perspective, we seek to illuminate applications of enzymatic bioconjugation in the face of an expanding palette of new drug modalities. With these applications, we wish to highlight some examples of current successes and pitfalls of using enzymes for bioconjugation along the pipeline and try to illustrate opportunities for further development.
Collapse
Affiliation(s)
- Aaron Debon
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| | - Elina Siirola
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| | - Radka Snajdrova
- Global
Discovery Chemistry, Novartis Institute
for Biomedical Research, Basel 4108, Switzerland
| |
Collapse
|
7
|
Orozco CT, Edgeworth MJ, Devine PWA, Hines AR, Cornwell O, Thompson C, Wang X, Phillips JJ, Ravn P, Jackson SE, Bond NJ. Interconversion of Unexpected Thiol States Affects the Stability, Structure, and Dynamics of Antibody Engineered for Site-Specific Conjugation. Bioconjug Chem 2021; 32:1834-1844. [PMID: 34369158 DOI: 10.1021/acs.bioconjchem.1c00286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody-drug conjugates have become one of the most actively developed classes of drugs in recent years. Their great potential comes from combining the strengths of large and small molecule therapeutics: the exquisite specificity of antibodies and the highly potent nature of cytotoxic compounds. More recently, the approach of engineering antibody-drug conjugate scaffolds to achieve highly controlled drug to antibody ratios has focused on substituting or inserting cysteines to facilitate site-specific conjugation. Herein, we characterize an antibody scaffold engineered with an inserted cysteine that formed an unexpected disulfide bridge during manufacture. A combination of mass spectrometry and biophysical techniques have been used to understand how the additional disulfide bridge forms, interconverts, and changes the stability and structural dynamics of the antibody intermediate. This quantitative and structurally resolved model of the local and global changes in structure and dynamics associated with the engineering and subsequent disulfide-bonded variant can assist future engineering strategies.
Collapse
Affiliation(s)
- Carolina T Orozco
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Matthew J Edgeworth
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| | - Paul W A Devine
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| | - Alistair R Hines
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| | - Owen Cornwell
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| | - Christopher Thompson
- Purification Process Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Xiangyang Wang
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, Gaithersburg, Maryland 20878, United States
| | - Jonathan J Phillips
- Living Systems Institute, Department of Biosciences, University of Exeter, EX4 4QD Exeter, United Kingdom
| | - Peter Ravn
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| | - Sophie E Jackson
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Nicholas J Bond
- Analytical Sciences, Biopharmaceutical Development, R&D, AstraZeneca, CB21 6GH Cambridge, United Kingdom
| |
Collapse
|
8
|
Rudd SE, Van Zuylekom JK, Raicevic A, Pearce LA, Cullinane C, Williams CC, Adams TE, Hicks RJ, Donnelly PS. Enzyme mediated incorporation of zirconium-89 or copper-64 into a fragment antibody for same day imaging of epidermal growth factor receptor. Chem Sci 2021; 12:9004-9016. [PMID: 34276928 PMCID: PMC8261882 DOI: 10.1039/d1sc01422f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022] Open
Abstract
Identification of tumors which over-express Epidermal Growth Factor Receptor (EGFR) is important in selecting patients for anti-EGFR therapies. Enzymatic bioconjugation was used to introduce positron-emitting radionuclides (89Zr, 64Cu) into an anti-EGFR antibody fragment for Positron Emission Tomography (PET) imaging the same day as injection. A monovalent antibody fragment with high affinity for EGFR was engineered to include a sequence that is recognized by the transpeptidase sortase A. Two different metal chelators, one for 89ZrIV and one for 64CuII, were modified with a N-terminal glycine to enable them to act as substrates in sortase A mediated bioconjugation to the antibody fragment. Both fragments provided high-quality PET images of EGFR positive tumors in a mouse model at 3 hours post-injection, a significant advantage when compared to radiolabeled full antibodies that require several days between injection of the tracer and imaging. The use of enzymatic bioconjugation gives reproducible homogeneous products with the metal complexes selectively installed on the C-terminus of the antibody potentially simplifying regulatory approval.
Collapse
Affiliation(s)
- Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science, Biotechnology Institute University of Melbourne Parkville Victoria 3010 Australia
| | | | - Anna Raicevic
- CSIRO Manufacturing Parkville Victoria 3052 Australia
| | | | - Carleen Cullinane
- Research Division, Peter MacCallum Cancer Centre Melbourne Victoria 3000 Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne Parkville Victoria Australia
| | | | | | - Rodney J Hicks
- Sir Peter MacCallum Department of Oncology, University of Melbourne Parkville Victoria Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science, Biotechnology Institute University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
9
|
Greenall SA, McKenzie M, Seminova E, Dolezal O, Pearce L, Bentley J, Kuchibhotla M, Chen SC, McDonald KL, Kornblum HI, Endersby R, Adams TE, Johns TG. Most clinical anti-EGFR antibodies do not neutralize both wtEGFR and EGFRvIII activation in glioma. Neuro Oncol 2021; 21:1016-1027. [PMID: 31002307 DOI: 10.1093/neuonc/noz073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Although epidermal growth factor receptor (EGFR) and its truncated, autoactive mutant EGFR variant (v)III are bona fide drivers of tumorigenesis in some gliomas, therapeutic antibodies developed to neutralize this axis have not improved patient survival in a limited number of trials. Previous studies using cells transduced to exogenously express EGFRvIII may have compromised mechanistic studies of anti-EGFR therapeutics. Therefore, we re-assessed the activity of clinical EGFR antibodies in patient-derived gliomaspheres that endogenously express EGFRvIII. METHODS The antitumor efficacy of antibodies was assessed using in vitro proliferation assays and intracranial orthografts. Receptor activation status, antibody engagement, oncogenic signaling, and mechanism of action after antibody treatment were analyzed by immunoprecipitation and western blotting. Tracking of antibody receptor complexes was conducted using immunofluorescence. RESULTS The EGFR domain III-targeting antibodies cetuximab, necitumumab, nimotuzumab, and matuzumab did not neutralize EGFRvIII activation. Chimeric monoclonal antibody 806 (ch806) neutralized EGFRvIII, but not wild-type (wt)EGFR activation. Panitumumab was the only antibody that neutralized both EGFRvIII and wtEGFR, leading to reduction of p-S6 signaling and superior in vitro and in vivo antitumor activity. Mechanistically, panitumumab induced recycling of receptor but not degradation as previously described. Panitumumab, via its unique avidity, stably cross-linked EGFRvIII to prevent its activation, while ch806 induced a marked reduction in the active EGFRvIII disulphide-bonded dimer. CONCLUSIONS We discovered a previously unknown major resistance mechanism in glioma in that most EGFR domain III-targeting antibodies do not neutralize EGFRvIII. The superior in vitro and in vivo antitumor activity of panitumumab supports further clinical testing of this antibody against EGFRvIII-stratified glioma.
Collapse
Affiliation(s)
- Sameer A Greenall
- Department of Oncology, Monash University and Monash Health, Clayton, Victoria, Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Brain Cancer Discovery Collaborative, New South Wales, Australia
| | - Mathew McKenzie
- School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, Australia.,Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | | | - Olan Dolezal
- CSIRO Manufacturing, Parkville, Victoria, Australia
| | | | - John Bentley
- CSIRO Manufacturing, Parkville, Victoria, Australia
| | - Mani Kuchibhotla
- Cancer Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| | - Shengnan C Chen
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Kerrie L McDonald
- Brain Cancer Discovery Collaborative, New South Wales, Australia.,Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, New South Wales, Australia
| | - Harley I Kornblum
- The Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, California, USA
| | - Raelene Endersby
- Brain Cancer Discovery Collaborative, New South Wales, Australia.,Cancer Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| | | | - Terrance G Johns
- Brain Cancer Discovery Collaborative, New South Wales, Australia.,Cancer Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
10
|
Lieser RM, Yur D, Sullivan MO, Chen W. Site-Specific Bioconjugation Approaches for Enhanced Delivery of Protein Therapeutics and Protein Drug Carriers. Bioconjug Chem 2020; 31:2272-2282. [DOI: 10.1021/acs.bioconjchem.0c00456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel M. Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| |
Collapse
|
11
|
Hofmann T, Krah S, Sellmann C, Zielonka S, Doerner A. Greatest Hits-Innovative Technologies for High Throughput Identification of Bispecific Antibodies. Int J Mol Sci 2020; 21:E6551. [PMID: 32911608 PMCID: PMC7554978 DOI: 10.3390/ijms21186551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Recent years have shown a tremendous increase and diversification in antibody-based therapeutics with advances in production techniques and formats. The plethora of currently investigated bi- to multi-specific antibody architectures can be harnessed to elicit a broad variety of specific modes of actions in oncology and immunology, spanning from enhanced selectivity to effector cell recruitment, all of which cannot be addressed by monospecific antibodies. Despite continuously growing efforts and methodologies, the identification of an optimal bispecific antibody as the best possible combination of two parental monospecific binders, however, remains challenging, due to tedious cloning and production, often resulting in undesired extended development times and increased expenses. Although automated high throughput screening approaches have matured for pharmaceutical small molecule development, it was only recently that protein bioconjugation technologies have been developed for the facile generation of bispecific antibodies in a 'plug and play' manner. In this review, we provide an overview of the most relevant methodologies for bispecific screening purposes-the DuoBody concept, paired light chain single cell production approaches, Sortase A and Transglutaminase, the SpyTag/SpyCatcher system, and inteins-and elaborate on the benefits as well as drawbacks of the different technologies.
Collapse
Affiliation(s)
- Tim Hofmann
- Advanced Cell Culture Technologies, Merck Life Sciences KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany;
| | - Simon Krah
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Carolin Sellmann
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Stefan Zielonka
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| | - Achim Doerner
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Frankfurter Strasse 250, D-64293 Darmstadt, Germany; (S.K.); (C.S.); (S.Z.)
| |
Collapse
|
12
|
Berckman EA, Hartzell EJ, Mitkas AA, Sun Q, Chen W. Biological Assembly of Modular Protein Building Blocks as Sensing, Delivery, and Therapeutic Agents. Annu Rev Chem Biomol Eng 2020; 11:35-62. [PMID: 32155350 DOI: 10.1146/annurev-chembioeng-101519-121526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nature has evolved a wide range of strategies to create self-assembled protein nanostructures with structurally defined architectures that serve a myriad of highly specialized biological functions. With the advent of biological tools for site-specific protein modifications and de novo protein design, a wide range of customized protein nanocarriers have been created using both natural and synthetic biological building blocks to mimic these native designs for targeted biomedical applications. In this review, different design frameworks and synthetic decoration strategies for achieving these functional protein nanostructures are summarized. Key attributes of these designer protein nanostructures, their unique functions, and their impact on biosensing and therapeutic applications are discussed.
Collapse
Affiliation(s)
- Emily A Berckman
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA; .,Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| | - Alexander A Mitkas
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| | - Qing Sun
- Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843, USA
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA;
| |
Collapse
|
13
|
One-step site-specific antibody fragment auto-conjugation using SNAP-tag technology. Nat Protoc 2019; 14:3101-3125. [DOI: 10.1038/s41596-019-0214-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
|
14
|
Lee MD, Tong WY, Nebl T, Pearce LA, Pham TM, Golbaz-Hagh A, Puttick S, Rose S, Adams TE, Williams CC. Dual Site-Specific Labeling of an Antibody Fragment through Sortase A and π-Clamp Conjugation. Bioconjug Chem 2019; 30:2539-2543. [DOI: 10.1021/acs.bioconjchem.9b00639] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Michael D. Lee
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | - Wing Yin Tong
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas Nebl
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | | | - Tam M. Pham
- CSIRO Manufacturing, Parkville, Victoria 3052, Australia
| | - Arghavan Golbaz-Hagh
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, 4072, Australia
| | - Simon Puttick
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, 4072, Australia
- CSIRO Health and Biosecurity, Herston, Queensland 4029, Australia
| | - Stephen Rose
- CSIRO Health and Biosecurity, Herston, Queensland 4029, Australia
| | | | | |
Collapse
|
15
|
Gébleux R, Briendl M, Grawunder U, Beerli RR. Sortase A Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody-Drug Conjugates. Methods Mol Biol 2019; 2012:1-13. [PMID: 31161500 DOI: 10.1007/978-1-4939-9546-2_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Antibody-drug conjugates (ADCs) are highly potent targeted anticancer therapies. They rely on the linking of a selectively targeting antibody moiety with potent cytotoxic payloads to effect antitumoral activity. In recent years, one focus in the ADC field was to create novel methods for site-specifically conjugating payloads to antibodies. The method presented here is based on the S. aureus sortase A-mediated transpeptidation reaction. This method requires antibodies to be engineered in such a way that they possess the sortase recognition pentapeptide motif LPETG on the C-terminus of the immunoglobulin heavy and/or light chains. In addition, the toxin must contain an oligoglycine motif in order to make it a suitable substrate for sortase A. Here we describe a detailed method to conjugate a pentaglycine-modified toxin to the C-termini of LPETG-tagged antibody heavy and light chains using sortase-mediated antibody conjugation (SMAC-Technology™). Highly homogenous, site-specifically conjugated ADCs with controlled drug to antibody ratio and improved overall properties can be obtained with this method.
Collapse
|
16
|
Efficient Production of Homogeneous Lysine-Based Antibody Conjugates Using Microbial Transglutaminase. Methods Mol Biol 2019; 2033:53-65. [PMID: 31332747 DOI: 10.1007/978-1-4939-9654-4_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Random conjugation of chemical linkers to endogenous lysines or cysteines within an antibody yields a heterogeneous mixture of conjugates with various drug-to-antibody ratios. One approach for generating homogeneous antibody conjugates utilizes enzymatic transfer of payloads onto a specific glycan or amino acid residue. Microbial transglutaminase (MTG) is an enzyme that catalyzes the formation of a stable isopeptide bond between a glutamine and a lysine. We have previously identified and reported several sites throughout the antibody structure where an engineered lysine is sufficient for transfer of a glutamine-based substrate onto the antibody. Whereas other enzymatic transfer strategies typically require significant antibody engineering to either modify the N-glycans or introduce a multi-amino acid enzyme recognition site, the lower contextual specificity of MTG for lysines allows just a single lysine point mutation in an antibody to be efficiently transamidated. Here we describe the molecular positioning of these single engineered lysine residues and the conjugation conditions for producing homogeneous antibody conjugates exemplified using azido- and auristatin F-based acyl donor substrates.
Collapse
|
17
|
Pishesha N, Ingram JR, Ploegh HL. Sortase A: A Model for Transpeptidation and Its Biological Applications. Annu Rev Cell Dev Biol 2018; 34:163-188. [PMID: 30110557 DOI: 10.1146/annurev-cellbio-100617-062527] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular biologists and chemists alike have long sought to modify proteins with substituents that cannot be installed by standard or even advanced genetic approaches. We here describe the use of transpeptidases to achieve these goals. Living systems encode a variety of transpeptidases and peptide ligases that allow for the enzyme-catalyzed formation of peptide bonds, and protein engineers have used directed evolution to enhance these enzymes for biological applications. We focus primarily on the transpeptidase sortase A, which has become popular over the past few years for its ability to perform a remarkably wide variety of protein modifications, both in vitro and in living cells.
Collapse
Affiliation(s)
- Novalia Pishesha
- Program in Molecular and Cellular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jessica R Ingram
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Hidde L Ploegh
- Program in Molecular and Cellular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| |
Collapse
|
18
|
Faccio G. From Protein Features to Sensing Surfaces. SENSORS (BASEL, SWITZERLAND) 2018; 18:E1204. [PMID: 29662030 PMCID: PMC5948494 DOI: 10.3390/s18041204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/08/2018] [Accepted: 04/12/2018] [Indexed: 12/25/2022]
Abstract
Proteins play a major role in biosensors in which they provide catalytic activity and specificity in molecular recognition. However, the immobilization process is far from straightforward as it often affects the protein functionality. Extensive interaction of the protein with the surface or significant surface crowding can lead to changes in the mobility and conformation of the protein structure. This review will provide insights as to how an analysis of the physico-chemical features of the protein surface before the immobilization process can help to identify the optimal immobilization approach. Such an analysis can help to preserve the functionality of the protein when on a biosensor surface.
Collapse
Affiliation(s)
- Greta Faccio
- Independent Scientist, St. Gallen 9000, Switzerland.
| |
Collapse
|
19
|
Enzyme-Based Labeling Strategies for Antibody-Drug Conjugates and Antibody Mimetics. Antibodies (Basel) 2018; 7:antib7010004. [PMID: 31544857 PMCID: PMC6698867 DOI: 10.3390/antib7010004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 01/25/2023] Open
Abstract
Strategies for site-specific modification of proteins have increased in number, complexity, and specificity over the last years. Such modifications hold the promise to broaden the use of existing biopharmaceuticals or to tailor novel proteins for therapeutic or diagnostic applications. The recent quest for next-generation antibody–drug conjugates (ADCs) sparked research into techniques with site selectivity. While purely chemical approaches often impede control of dosage or locus of derivatization, naturally occurring enzymes and proteins bear the ability of co- or post-translational protein modifications at particular residues, thus enabling unique coupling reactions or protein fusions. This review provides a general overview and focuses on chemo-enzymatic methods including enzymes such as formylglycine-generating enzyme, sortase, and transglutaminase. Applications for the conjugation of antibodies and antibody mimetics are reported.
Collapse
|
20
|
Spidel JL, Vaessen B, Albone EF, Cheng X, Verdi A, Kline JB. Site-Specific Conjugation to Native and Engineered Lysines in Human Immunoglobulins by Microbial Transglutaminase. Bioconjug Chem 2017; 28:2471-2484. [PMID: 28820579 DOI: 10.1021/acs.bioconjchem.7b00439] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The use of microbial transglutaminase (MTG) to produce site-specific antibody-drug conjugates (ADCs) has thus far focused on the transamidation of engineered acyl donor glutamine residues in an antibody based on the hypothesis that the lower specificity of MTG for acyl acceptor lysines may result in the transamidation of multiple native lysine residues, thereby yielding heterogeneous products. We investigated the utilization of native IgG lysines as acyl acceptor sites for glutamine-based acyl donor substrates. Of the approximately 80 lysines in multiple recombinant IgG monoclonal antibodies (mAbs), none were transamidated. Because recombinant mAbs lack the C-terminal Lys447 due to cleavage by carboxypeptidase B in the production cell host, we explored whether blocking the cleavage of Lys447 by the addition of a C-terminal amino acid could result in transamidation of Lys447 by a variety of acyl donor substrates. MTG efficiently transamidated Lys447 in the presence of any nonacidic, nonproline amino acid residue at position 448. Lysine scanning mutagenesis throughout the antibody further revealed several transamidation sites in both the heavy- and light-chain constant regions. Additionally, scanning mutagenesis of the hinge region in a Fab' fragment revealed sites of transamidation that were not reactive in the context of the full-length mAb. Here, we demonstrate the utility of single lysine substitutions and the C-terminal Lys447 for engineering efficient acyl acceptor sites suitable for site-specific conjugation to a range of glutamine-based acyl donor substrates.
Collapse
Affiliation(s)
- Jared L Spidel
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| | - Benjamin Vaessen
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| | - Earl F Albone
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| | - Xin Cheng
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| | - Arielle Verdi
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| | - J Bradford Kline
- Morphotek Inc. , 210 Welsh Pool Road, Exton, Pennsylvania 19341, United States
| |
Collapse
|
21
|
ESCORTing proteins directly from whole cell-lysate for single-molecule studies. Anal Biochem 2017; 535:35-42. [PMID: 28756135 PMCID: PMC5571901 DOI: 10.1016/j.ab.2017.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 07/19/2017] [Accepted: 07/21/2017] [Indexed: 12/29/2022]
Abstract
We have developed a method for Enzymatic Sortase-assisted Covalent Orientation-specific Restraint Tethering (ESCORT) recombinant proteins onto surfaces directly from cell-lysate. With an improved surface passivation method, we obviate the cumbersome purification steps even for single molecule studies that demand high purity in the sample. We demonstrated high-specificity of the method, high-passivity of the surface and uncompromised functional integrity of anchored proteins using single molecule fluorescence and force-mapping. We anticipate that this method will substantially reduce the investment by way of time, money and energy in the area of single molecule studies.
Collapse
|
22
|
Thérien A, Bédard M, Carignan D, Rioux G, Gauthier-Landry L, Laliberté-Gagné MÈ, Bolduc M, Savard P, Leclerc D. A versatile papaya mosaic virus (PapMV) vaccine platform based on sortase-mediated antigen coupling. J Nanobiotechnology 2017; 15:54. [PMID: 28720097 PMCID: PMC5516373 DOI: 10.1186/s12951-017-0289-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 07/10/2017] [Indexed: 02/04/2023] Open
Abstract
Background Flexuous rod-shaped nanoparticles made of the coat protein (CP) of papaya mosaic virus (PapMV) have been shown to trigger innate immunity through engagement of toll-like receptor 7 (TLR7). PapMV nanoparticles can also serve as a vaccine platform as they can increase the immune response to fused peptide antigens. Although this approach shows great potential, fusion of antigens directly to the CP open reading frame (ORF) is challenging because the fused peptides can alter the structure of the CP and its capacity to self assemble into nanoparticles—a property essential for triggering an efficient immune response to the peptide. This represents a serious limitation to the utility of this approach as fusion of small peptides only is tolerated. Results We have developed a novel approach in which peptides are fused directly to pre-formed PapMV nanoparticles. This approach is based on the use of a bacterial transpeptidase (sortase A; SrtA) that can attach the peptide directly to the nanoparticle. An engineered PapMV CP harbouring the SrtA recognition motif allows efficient coupling. To refine our engineering, and to predict the efficacy of coupling with SrtA, we modeled the PapMV structure based on the known structure of PapMV CP and on recent reports revealing the structure of two closely related potexviruses: pepino mosaic virus (PepMV) and bamboo mosaic virus (BaMV). We show that SrtA can allow the attachment of long peptides [Influenza M2e peptide (26 amino acids) and the HIV-1 T20 peptide (39 amino acids)] to PapMV nanoparticles. Consistent with our PapMV structural model, we show that around 30% of PapMV CP subunits in each nanoparticle can be fused to the peptide antigen. As predicted, engineered nanoparticles were capable of inducing a strong antibody response to the fused antigen. Finally, in a challenge study with influenza virus, we show that mice vaccinated with PapMV-M2e are protected from infection. Conclusions This technology will allow the development of vaccines harbouring long peptides containing several B and/or T cell epitopes that can contribute to a broad and robust protection from infection. The design can be fast, versatile and can be adapted to the development of vaccines for many infectious diseases as well as cancer vaccines. Electronic supplementary material The online version of this article (doi:10.1186/s12951-017-0289-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ariane Thérien
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Mikaël Bédard
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Damien Carignan
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Gervais Rioux
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Louis Gauthier-Landry
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Marie-Ève Laliberté-Gagné
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Marilène Bolduc
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada
| | - Pierre Savard
- Neurosciences, Laval University, 2705 Boul. Laurier, Québec City, PQ, G1V 4G2, Canada
| | - Denis Leclerc
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Center, Laval University, 2705 Boul. Laurier, Quebec City, PQ, G1V 4G2, Canada.
| |
Collapse
|
23
|
Wu Z, Hong H, Zhao X, Wang X. Efficient expression of sortase A from Staphylococcus aureus in Escherichia coli and its enzymatic characterizations. BIORESOUR BIOPROCESS 2017; 4:13. [PMID: 28261538 PMCID: PMC5316389 DOI: 10.1186/s40643-017-0143-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/07/2017] [Indexed: 02/08/2023] Open
Abstract
Background Sortase A (SrtA) is a transpeptidase found in Staphylococcus aureus, which is widely used in site-specific protein modification. However, SrtA was expressed in Escherichia coli (E. coli) in rather low level (ranging from several milligrams to 76.9 mg/L at most). The present study aims to optimize fermentation conditions for improving SrtA expression in E. coli. Results Under the optimized media (0.48 g/L glycerol, 1.37 g/L tryptone, 0.51 g/L yeast extract, MOPS 0.5 g/L, PBS buffer 180 mL/L) and condition (30 °C for 8 h) in a 7-L fermentor, the enzyme activity and the yield of SrtA reached 2458.4 ± 115.9 U/mg DCW and 232.4 ± 21.1 mg/L, respectively, which were higher by 5.8- and 4.5-folds compared with initial conditions, respectively. The yield of SrtA also represented threefold increase than the previously reported maximal level. In addition, the enzymatic characterizations of SrtA (optimal temperature, optimal pH, the influence of metal irons, and tolerance to water-soluble organic solvents) were determined. Conclusions Enhanced expression of SrtA was achieved by optimization of medium and condition. This result will have potential application for production levels of SrtA on an industry scale. Moreover, the detailed enzymatic characterizations of SrtA were examined, which will provide a useful guide for its future application. Electronic supplementary material The online version of this article (doi:10.1186/s40643-017-0143-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhimeng Wu
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, China.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191 China
| | - Haofei Hong
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, China
| | - Xinrui Zhao
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, China
| | - Xun Wang
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 1800 Lihu Road, Wuxi, China
| |
Collapse
|
24
|
Enzyme-Based Strategies to Generate Site-Specifically Conjugated Antibody Drug Conjugates. NEXT GENERATION ANTIBODY DRUG CONJUGATES (ADCS) AND IMMUNOTOXINS 2017. [DOI: 10.1007/978-3-319-46877-8_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
25
|
Adumeau P, Sharma SK, Brent C, Zeglis BM. Site-Specifically Labeled Immunoconjugates for Molecular Imaging--Part 2: Peptide Tags and Unnatural Amino Acids. Mol Imaging Biol 2016; 18:153-65. [PMID: 26754791 DOI: 10.1007/s11307-015-0920-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular imaging using radioisotope- or fluorophore-labeled antibodies is increasingly becoming a critical component of modern precision medicine. Yet despite this promise, the vast majority of these immunoconjugates are synthesized via the random coupling of amine-reactive bifunctional probes to lysines within the antibody, a process that can result in heterogeneous and poorly defined constructs with suboptimal pharmacological properties. In an effort to circumvent these issues, the last 5 years have played witness to a great deal of research focused on the creation of effective strategies for the site-specific attachment of payloads to antibodies. These chemoselective modification methods yield immunoconjugates that are more homogenous and better defined than constructs created using traditional synthetic approaches. Moreover, site-specifically labeled immunoconjugates have also been shown to exhibit superior in vivo behavior compared to their randomly modified cousins. The over-arching goal of this two-part review is to provide a broad yet detailed account of the various site-specific bioconjugation approaches that have been used to create immunoconjugates for positron emission tomography (PET), single photon emission computed tomography (SPECT), and fluorescence imaging. In Part 1, we covered site-specific bioconjugation techniques based on the modification of cysteine residues and the chemoenzymatic manipulation of glycans. In Part 2, we will detail two families of bioconjugation approaches that leverage biochemical tools to achieve site-specificity. First, we will discuss modification methods that employ peptide tags either as sites for enzyme-catalyzed ligations or as radiometal coordination architectures. And second, we will examine bioconjugation strategies predicated on the incorporation of unnatural or non-canonical amino acids into antibodies via genetic engineering. Finally, we will compare the advantages and disadvantages of the modification strategies covered in both parts of the review and offer a brief discussion of the overall direction of the field.
Collapse
Affiliation(s)
- Pierre Adumeau
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Sai Kiran Sharma
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Colleen Brent
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA
| | - Brian M Zeglis
- Department of Chemistry and Biochemistry, Hunter College and the Graduate Center of the City University of New York, 413 East 69th Street, New York, NY, 10021, USA. .,Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Antibody-drug conjugates: Current status and future perspectives. Pharmacol Ther 2016; 167:48-59. [DOI: 10.1016/j.pharmthera.2016.07.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2016] [Indexed: 02/02/2023]
|
27
|
Tsuchikama K, An Z. Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell 2016; 9:33-46. [PMID: 27743348 PMCID: PMC5777969 DOI: 10.1007/s13238-016-0323-0] [Citation(s) in RCA: 508] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/06/2016] [Indexed: 01/11/2023] Open
Abstract
The antibody-drug conjugate (ADC), a humanized or human monoclonal antibody conjugated with highly cytotoxic small molecules (payloads) through chemical linkers, is a novel therapeutic format and has great potential to make a paradigm shift in cancer chemotherapy. This new antibody-based molecular platform enables selective delivery of a potent cytotoxic payload to target cancer cells, resulting in improved efficacy, reduced systemic toxicity, and preferable pharmacokinetics (PK)/pharmacodynamics (PD) and biodistribution compared to traditional chemotherapy. Boosted by the successes of FDA-approved Adcetris® and Kadcyla®, this drug class has been rapidly growing along with about 60 ADCs currently in clinical trials. In this article, we briefly review molecular aspects of each component (the antibody, payload, and linker) of ADCs, and then mainly discuss traditional and new technologies of the conjugation and linker chemistries for successful construction of clinically effective ADCs. Current efforts in the conjugation and linker chemistries will provide greater insights into molecular design and strategies for clinically effective ADCs from medicinal chemistry and pharmacology standpoints. The development of site-specific conjugation methodologies for constructing homogeneous ADCs is an especially promising path to improving ADC design, which will open the way for novel cancer therapeutics.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, 77054, USA
| |
Collapse
|
28
|
Massa S, Vikani N, Betti C, Ballet S, Vanderhaegen S, Steyaert J, Descamps B, Vanhove C, Bunschoten A, van Leeuwen FWB, Hernot S, Caveliers V, Lahoutte T, Muyldermans S, Xavier C, Devoogdt N. Sortase A-mediated site-specific labeling of camelid single-domain antibody-fragments: a versatile strategy for multiple molecular imaging modalities. CONTRAST MEDIA & MOLECULAR IMAGING 2016; 11:328-339. [PMID: 27147480 DOI: 10.1002/cmmi.1696] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/09/2016] [Indexed: 12/20/2022]
Abstract
A generic site-specific conjugation method that generates a homogeneous product is of utmost importance in tracer development for molecular imaging and therapy. We explored the protein-ligation capacity of the enzyme Sortase A to label camelid single-domain antibody-fragments, also known as nanobodies. The versatility of the approach was demonstrated by conjugating independently three different imaging probes: the chelating agents CHX-A"-DTPA and NOTA for single-photon emission computed tomography (SPECT) with indium-111 and positron emission tomography (PET) with gallium-68, respectively, and the fluorescent dye Cy5 for fluorescence reflectance imaging (FRI). After a straightforward purification process, homogeneous single-conjugated tracer populations were obtained in high yield (30-50%). The enzymatic conjugation did not affect the affinity of the tracers, nor the radiolabeling efficiency or spectral characteristics. In vivo, the tracers enabled the visualization of human epidermal growth factor receptor 2 (HER2) expressing BT474M1-tumors with high contrast and specificity as soon as 1 h post injection in all three imaging modalities. These data demonstrate Sortase A-mediated conjugation as a valuable strategy for the development of site-specifically labeled camelid single-domain antibody-fragments for use in multiple molecular imaging modalities. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sam Massa
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
| | - Niravkumar Vikani
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
| | - Cecilia Betti
- Laboratory of Organic Chemistry, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
| | - Steven Ballet
- Laboratory of Organic Chemistry, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
| | - Saskia Vanderhaegen
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium.,Structural Biology Research Center, VIB, 1050, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium.,Structural Biology Research Center, VIB, 1050, Brussels, Belgium
| | - Benedicte Descamps
- Infinity-MEDISIP-iMinds Medical IT, Department of Electronics and Information Systems, Ghent University, 9000, Ghent, Belgium
| | - Christian Vanhove
- Infinity-MEDISIP-iMinds Medical IT, Department of Electronics and Information Systems, Ghent University, 9000, Ghent, Belgium
| | - Anton Bunschoten
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333ZA, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, 2333ZA, Leiden, The Netherlands
| | - Sophie Hernot
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Vicky Caveliers
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.,Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Tony Lahoutte
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.,Nuclear Medicine Department, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium
| | - Catarina Xavier
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium. .,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), 1050, Brussels, Belgium.
| |
Collapse
|
29
|
Yao H, Jiang F, Lu A, Zhang G. Methods to Design and Synthesize Antibody-Drug Conjugates (ADCs). Int J Mol Sci 2016; 17:E194. [PMID: 26848651 PMCID: PMC4783928 DOI: 10.3390/ijms17020194] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 11/16/2022] Open
Abstract
Antibody-drug conjugates (ADCs) have become a promising targeted therapy strategy that combines the specificity, favorable pharmacokinetics and biodistributions of antibodies with the destructive potential of highly potent drugs. One of the biggest challenges in the development of ADCs is the application of suitable linkers for conjugating drugs to antibodies. Recently, the design and synthesis of linkers are making great progress. In this review, we present the methods that are currently used to synthesize antibody-drug conjugates by using thiols, amines, alcohols, aldehydes and azides.
Collapse
Affiliation(s)
- Houzong Yao
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Feng Jiang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Faculty of Materials Science and Chemical Engineering, the State Key Laboratory Base of Novel Functional Materials and Preparation Science, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
30
|
Paterson BM, Donnelly PS. Macrocyclic Bifunctional Chelators and Conjugation Strategies for Copper-64 Radiopharmaceuticals. ADVANCES IN INORGANIC CHEMISTRY 2016. [DOI: 10.1016/bs.adioch.2015.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Siegmund V, Schmelz S, Dickgiesser S, Beck J, Ebenig A, Fittler H, Frauendorf H, Piater B, Betz UAK, Avrutina O, Scrima A, Fuchsbauer H, Kolmar H. Locked by Design: A Conformationally Constrained Transglutaminase Tag Enables Efficient Site‐Specific Conjugation. Angew Chem Int Ed Engl 2015; 54:13420-4. [DOI: 10.1002/anie.201504851] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Vanessa Siegmund
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Stefan Schmelz
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Inhoffenstr. 7, 38124 Braunschweig (Germany)
| | - Stephan Dickgiesser
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Jan Beck
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Aileen Ebenig
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Heiko Fittler
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Holm Frauendorf
- Institut für Organische und Biomolekulare Chemie, Zentrale Analytik/Massenspektrometrie, Georg‐August‐Universität Göttingen, Tammannstr. 2, 37077 Göttingen (Germany)
| | - Birgit Piater
- Merck KGaA, Frankfurterstr. 250, 64293 Darmstadt (Germany)
| | | | - Olga Avrutina
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| | - Andrea Scrima
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Inhoffenstr. 7, 38124 Braunschweig (Germany)
| | - Hans‐Lothar Fuchsbauer
- Fachbereich Chemie‐ und Biotechnologie, Hochschule Darmstadt, Schnittspahnstraße 12, 64287 Darmstadt (Germany)
| | - Harald Kolmar
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Germany)
| |
Collapse
|
32
|
Siegmund V, Schmelz S, Dickgiesser S, Beck J, Ebenig A, Fittler H, Frauendorf H, Piater B, Betz UAK, Avrutina O, Scrima A, Fuchsbauer H, Kolmar H. Durch Design verbrückt: ein konformativ eingeschränkter Transglutaminase‐Marker ermöglicht effiziente ortsspezifische Konjugation. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201504851] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Vanessa Siegmund
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Stefan Schmelz
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Braunschweig (Deutschland)
| | - Stephan Dickgiesser
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Jan Beck
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Aileen Ebenig
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Heiko Fittler
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Holm Frauendorf
- Institut für Organische und Biomolekulare Chemie, Zentrale Analytik/Massenspektrometrie, Universität Göttingen (Deutschland)
| | | | | | - Olga Avrutina
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| | - Andrea Scrima
- Arbeitsgruppe Strukturbiologie der Autophagie, Abteilung Struktur und Funktion der Proteine, Helmholtz‐Zentrum für Infektionsforschung, Braunschweig (Deutschland)
| | | | - Harald Kolmar
- Clemens‐Schöpf‐Institut für Organische Chemie und Biochemie, Technische Universität Darmstadt, Alarich‐Weiss‐Straße 4, 64287 Darmstadt (Deutschland)
| |
Collapse
|
33
|
Niikura K, Horisawa K, Doi N. Endosomal escape efficiency of fusogenic B18 and B55 peptides fused with anti-EGFR single chain Fv as estimated by nuclear translocation. J Biochem 2015; 159:123-32. [PMID: 26338729 DOI: 10.1093/jb/mvv083] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/10/2015] [Indexed: 01/08/2023] Open
Abstract
UNLABELLED Although monoclonal antibodies have been used not only as analytical tools but also as biologic therapeutics, they cannot target intracellular proteins due to their large molecular size and low membrane permeability, which limit their applications. During previous attempts to delivery antibodies intracellularly, the low efficiency of escape from endosomes to the cytosol reduced the bioavailability of antibodies or antibody-conjugated effectors. Recently, we found that the fusogenic peptides (FPs) B18 and B55 from bindin, a sea urchin gamete recognition protein, facilitated the endosomal escape of FP-fused enhanced green fluorescent protein (eGFP) and/or of co-administered cargos such as dextrans [Niikura et al. A fusogenic peptide from a sea urchin fertilization protein promotes intracellular delivery of biomacromolecules by facilitating endosomal escape. J. CONTROL Release 2015;212:85-93]. In this study, we constructed FP-fused anti-epidermal growth factor receptor (EGFR) single-chain Fv (αEGFR[scFv]) proteins and evaluated their endosomal escape efficiency by utilizing a nuclear localization signal). When the FP-fused αEGFR[scFv] proteins were incubated with A431 cells, the estimated endosomal escape efficiency of αEGFR[scFv]-B18 was significantly higher than that of αEGFR[scFv] alone, suggesting that the B18 peptide facilitates endosomal escape of the conjugated scFv in cis. Moreover, αEGFR[scFv]-B55 promoted the intracellular uptake of co-administered eGFP and dextrans in trans. These results imply that B18- and B55-fused antibodies may be useful for the cell-specific intracellular delivery of biomacromolecules.
Collapse
Affiliation(s)
- Keisuke Niikura
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Yokohama 223-8522, Japan
| | - Kenichi Horisawa
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Yokohama 223-8522, Japan
| | - Nobuhide Doi
- Department of Biosciences and Informatics, Keio University, 3-14-1 Hiyoshi, Yokohama 223-8522, Japan
| |
Collapse
|
34
|
Beerli RR, Hell T, Merkel AS, Grawunder U. Sortase Enzyme-Mediated Generation of Site-Specifically Conjugated Antibody Drug Conjugates with High In Vitro and In Vivo Potency. PLoS One 2015; 10:e0131177. [PMID: 26132162 PMCID: PMC4488448 DOI: 10.1371/journal.pone.0131177] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/31/2015] [Indexed: 01/16/2023] Open
Abstract
Antibody drug conjugates (ADCs) have recently been proven to be highly potent anti-tumor drugs, typically exceeding the efficacy of conventional monoclonal antibodies (mAbs). ADCs are currently produced by chemical conjugation of a small-molecule toxin to the mAb through lysine or cysteine side chains. This leads to heterogeneous mixtures of ADCs in which variable numbers of drugs are conjugated to individual antibodies and in which the site of conjugation cannot be defined. Consequently, there is currently significant interest in further development of drug conjugation technologies, with a particular focus on site-specific payload conjugation. Here, we present an enzymatic conjugation platform based on the S. aureus sortase A-mediated transpeptidation reaction, allowing the efficient generation of ADCs with toxins conjugated to pre-defined sites at pre-defined drug-to-antibody ratios. For this, two modifications were introduced: first, immunoglobulin heavy (IgH) and light (IgL) chains were modified at their C-termini by addition of the sortase A recognition motif LPETG, and second, the small molecule tubulin polymerization inhibitors monomethylauristatin E (MMAE) and maytansine were modified by addition of a pentaglycine peptide, thus making them suitable substrates for sortase A-mediated transpeptidation. We demonstrate efficient generation and characterization of the anti-CD30 ADC Ac10-vcPAB-MMAE, an enzymatically conjugated counterpart of brentuximab vedotin (Adcetris), as well as several anti-HER-2 ADCs including trastuzumab-maytansine, the counterpart of trastuzumab emtansine (Kadcyla). ADCs generated in this manner were found to display in vitro cell killing activities indistinguishable from the classic conjugates. Further, when tested in vivo in a HER-2-overexpressing ovarian cancer xenograft mouse model, enzymatically generated trastuzumab-maytansine was found to lead to complete regression of established tumors, similar to Kadcyla.
Collapse
MESH Headings
- Ado-Trastuzumab Emtansine
- Aminoacyltransferases/chemistry
- Aminoacyltransferases/immunology
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Bacterial Proteins/chemistry
- Bacterial Proteins/immunology
- Brentuximab Vedotin
- Cysteine Endopeptidases/chemistry
- Cysteine Endopeptidases/immunology
- Female
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/immunology
- Immunoconjugates/pharmacology
- Ki-1 Antigen/antagonists & inhibitors
- Ki-1 Antigen/genetics
- Ki-1 Antigen/immunology
- Maytansine/analogs & derivatives
- Maytansine/chemistry
- Maytansine/immunology
- Maytansine/pharmacology
- Mice
- Mice, Nude
- Oligopeptides/chemistry
- Oligopeptides/immunology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Protein Engineering
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Staphylococcus aureus/chemistry
- Staphylococcus aureus/enzymology
- Trastuzumab
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Roger R. Beerli
- NBE-Therapeutics AG, Hochbergerstrasse, Basel, Switzerland
- * E-mail:
| | - Tamara Hell
- NBE-Therapeutics AG, Hochbergerstrasse, Basel, Switzerland
| | - Anna S. Merkel
- NBE-Therapeutics AG, Hochbergerstrasse, Basel, Switzerland
| | - Ulf Grawunder
- NBE-Therapeutics AG, Hochbergerstrasse, Basel, Switzerland
| |
Collapse
|
35
|
Alt K, Paterson BM, Westein E, Rudd SE, Poniger SS, Jagdale S, Ardipradja K, Connell TU, Krippner GY, Nair AKN, Wang X, Tochon-Danguy HJ, Donnelly PS, Peter K, Hagemeyer CE. A versatile approach for the site-specific modification of recombinant antibodies using a combination of enzyme-mediated bioconjugation and click chemistry. Angew Chem Int Ed Engl 2015; 54:7515-9. [PMID: 25962581 DOI: 10.1002/anie.201411507] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/24/2015] [Indexed: 12/16/2022]
Abstract
A unique two-step modular system for site-specific antibody modification and conjugation is reported. The first step of this approach uses enzymatic bioconjugation with the transpeptidase Sortase A for incorporation of strained cyclooctyne functional groups. The second step of this modular approach involves the azide-alkyne cycloaddition click reaction. The versatility of the two-step approach has been exemplified by the selective incorporation of fluorescent dyes and a positron-emitting copper-64 radiotracer for fluorescence and positron-emission tomography imaging of activated platelets, platelet aggregates, and thrombi, respectively. This flexible and versatile approach could be readily adapted to incorporate a large array of tailor-made functional groups using reliable click chemistry whilst preserving the activity of the antibody or other sensitive biological macromolecules.
Collapse
Affiliation(s)
- Karen Alt
- Vascular Biotechnology, Baker IDI, Melbourne (Australia).
| | - Brett M Paterson
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Erik Westein
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | - Stacey E Rudd
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Stan S Poniger
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne (Australia)
| | - Shweta Jagdale
- Vascular Biotechnology, Baker IDI, Melbourne (Australia)
| | | | - Timothy U Connell
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia)
| | - Guy Y Krippner
- Vascular Biotechnology, Baker IDI, Melbourne (Australia)
| | - Ashish K N Nair
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | | | - Paul S Donnelly
- School of Chemistry/Bio21 Institute, University of Melbourne (Australia).
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker IDI, Melbourne (Australia)
| | | |
Collapse
|
36
|
Alt K, Paterson BM, Westein E, Rudd SE, Poniger SS, Jagdale S, Ardipradja K, Connell TU, Krippner GY, Nair AKN, Wang X, Tochon-Danguy HJ, Donnelly PS, Peter K, Hagemeyer CE. A Versatile Approach for the Site-Specific Modification of Recombinant Antibodies Using a Combination of Enzyme-Mediated Bioconjugation and Click Chemistry. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201411507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
37
|
Zhai J, Scoble JA, Li N, Lovrecz G, Waddington LJ, Tran N, Muir BW, Coia G, Kirby N, Drummond CJ, Mulet X. Epidermal growth factor receptor-targeted lipid nanoparticles retain self-assembled nanostructures and provide high specificity. NANOSCALE 2015; 7:2905-2913. [PMID: 25516406 DOI: 10.1039/c4nr05200e] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Next generation drug delivery utilising nanoparticles incorporates active targeting to specific sites. In this work, we combined targeting with the inherent advantages of self-assembled lipid nanoparticles containing internal nano-structures. Epidermal growth factor receptor (EGFR)-targeting, PEGylated lipid nanoparticles using phytantriol and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-PEG-maleimide amphiphiles were created. The self-assembled lipid nanoparticles presented here have internal lyotropic liquid crystalline nano-structures, verified by synchrotron small angle X-ray scattering and cryo-transmission electron microscopy, that offer the potential of high drug loading and enhanced cell penetration. Anti-EGFR Fab' fragments were conjugated to the surface of nanoparticles via a maleimide-thiol reaction at a high conjugation efficiency and retained specificity following conjugation to the nanoparticles. The conjugated nanoparticles were demonstrated to have high affinity for an EGFR target in a ligand binding assay.
Collapse
Affiliation(s)
- Jiali Zhai
- CSIRO Manufacturing Flagship, Private Bag 10, Clayton, VIC 3169, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
McCombs JR, Owen SC. Antibody drug conjugates: design and selection of linker, payload and conjugation chemistry. AAPS JOURNAL 2015; 17:339-51. [PMID: 25604608 DOI: 10.1208/s12248-014-9710-8] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 12/19/2014] [Indexed: 11/30/2022]
Abstract
Antibody drug conjugates (ADCs) have emerged as an important pharmaceutical class of drugs designed to harness the specificity of antibodies with the potency of small molecule therapeutics. The three main components of ADCs are the antibody, the linker, and the payload; the majority of early work focused intensely on improving the functionality of these pieces. Recently, considerable attention has been focused on developing methods to control the site and number of linker/drug conjugated to the antibody, with the aim of producing more homogenous ADCs. In this article, we review popular conjugation methods and highlight recent approaches including "click" conjugation and enzymatic ligation. We discuss current linker technology, contrasting the characteristics of cleavable and non-cleavable linkers, and summarize the essential properties of ADC payload, centering on chemotherapeutics. In addition, we report on the progress in characterizing to determine physicochemical properties and on advances in purifying to obtain homogenous products. Establishing a set of selection and analytical criteria will facilitate the translation of novel ADCs and ensure the production of effective biosimilars.
Collapse
Affiliation(s)
- Jessica R McCombs
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S. 2000 E., Salt Lake City, UT, 84112, USA
| | | |
Collapse
|
39
|
Kline T, Steiner AR, Penta K, Sato AK, Hallam TJ, Yin G. Methods to Make Homogenous Antibody Drug Conjugates. Pharm Res 2014; 32:3480-93. [PMID: 25511917 PMCID: PMC4596908 DOI: 10.1007/s11095-014-1596-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
Abstract
Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small molecule chemistries that are currently used and being explored to develop specific, homogenous ADCs.
Collapse
Affiliation(s)
- Toni Kline
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Alexander R Steiner
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Kalyani Penta
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Aaron K Sato
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Trevor J Hallam
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA
| | - Gang Yin
- Sutro Biopharma, Inc, 310 Utah Ave Ste 150, South San Francisco, California, 94080, USA.
| |
Collapse
|
40
|
Baer S, Nigro J, Madej MP, Nisbet RM, Suryadinata R, Coia G, Hong LPT, Adams TE, Williams CC, Nuttall SD. Comparison of alternative nucleophiles for Sortase A-mediated bioconjugation and application in neuronal cell labelling. Org Biomol Chem 2014; 12:2675-85. [PMID: 24643508 DOI: 10.1039/c3ob42325e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Sortase A (SrtA) enzyme from Staphylococcus aureus catalyses covalent attachment of protein substrates to pentaglycine cross-bridges in the Gram positive bacterial cell wall. In vitro SrtA-mediated protein ligation is now an important protein engineering tool for conjugation of substrates containing the LPXTGX peptide recognition sequence to oligo-glycine nucleophiles. In order to explore the use of alternative nucleophiles in this system, five different rhodamine-labelled compounds, with N-terminal nucleophilic amino acids, triglycine, glycine, and lysine, or N-terminal non-amino acid nucleophiles ethylenediamine and cadaverine, were synthesized. These compounds were tested for their relative abilities to function as nucleophiles in SrtA-mediated bioconjugation reactions. N-Terminal triglycine, glycine and ethylenediamine were all efficient in labelling a range of LPETGG containing recombinant antibody and scaffold proteins and peptides, while reduced activity was observed for the other nucleophiles across the range of proteins and peptides studied. Expansion of the range of available nucleophiles which can be utilised in SrtA-mediated bioconjugation expands the range of potential applications for this technology. As a demonstration of the utility of this system, SrtA coupling was used to conjugate the triglycine rhodamine-labelled nucleophile to the C-terminus of an Im7 scaffold protein displaying Aβ, a neurologically important peptide implicated in Alzheimer's disease. Purified, labelled protein showed Aβ-specific targeting to mammalian neuronal cells. Demonstration of targeting neuronal cells with a chimeric protein illustrates the power of this system, and suggests that SrtA-mediated direct cell-surface labelling and visualisation is an achievable goal.
Collapse
Affiliation(s)
- Samuel Baer
- CSIRO Materials Science and Engineering, 343 Royal Parade, Parkville, Victoria 3052, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Bioorthogonal, chemoselective ligation methods are an essential part of the tools utilized to investigate biochemical pathways. Specifically enzymatic approaches are valuable methods in this context due to the inherent specificity of the deployed enzymes and the mild conditions of the modification reactions. One of the most common strategies is based on the transpeptidation catalyzed by sortase A derived from Staphylococcus aureus. The procedure is well established and a wide variety of applications have been published to date. Here, implementations of sortase A, which range from protein labeling using fluorescence dyes and the preparation of cyclic proteins to the modification of entire cells, are summarized. Furthermore, there is a focus on the optimization approaches established to solve the drawbacks of sortase-mediated transpeptidation.
Collapse
Affiliation(s)
- Markus Ritzefeld
- Bielefeld University, Department of Chemistry, Organic and Bioorganic Chemistry (OCIII), Universitätsstrasse 25, 33615 Bielefeld (Germany).
| |
Collapse
|
42
|
Liebscher S, Kornberger P, Fink G, Trost-Gross EM, Höss E, Skerra A, Bordusa F. Derivatization of antibody Fab fragments: a designer enzyme for native protein modification. Chembiochem 2014; 15:1096-100. [PMID: 24782039 DOI: 10.1002/cbic.201400059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Indexed: 01/25/2023]
Abstract
Bioconjugates, such as antibody-drug conjugates, have gained recent attention because of their increasing use in therapeutic and diagnostic applications. Commonly used conjugation reactions based upon chemoselective reagents exhibit a number of drawbacks: most of these reactions lack regio- and stereospecificity, thus resulting in loss of protein functionality due to random modifications. Enzymes provide an obvious solution to this problem, but the intrinsic (natural) substrate specificities of existing enzymes pose severe limitations to the kind of modifications that can be introduced. Here we describe the application of the novel trypsin variant trypsiligase for site-specific modification of the C terminus of a Fab antibody fragment via a stable peptide bond. The suitability of this designed biocatalyst was demonstrated by coupling the Her2-specific Fab to artificial functionalities of either therapeutic (PEG) or diagnostic (fluorescein) relevance. In both cases we obtained homogeneously modified Fab products bearing the artificial functionality exclusively at the desired position.
Collapse
Affiliation(s)
- Sandra Liebscher
- Institute of Biochemistry/Biotechnology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Strasse 3, 06120 Halle/Saale (Germany)
| | | | | | | | | | | | | |
Collapse
|
43
|
Paterson BM, Alt K, Jeffery CM, Price RI, Jagdale S, Rigby S, Williams CC, Peter K, Hagemeyer CE, Donnelly PS. Enzyme-mediated site-specific bioconjugation of metal complexes to proteins: sortase-mediated coupling of copper-64 to a single-chain antibody. Angew Chem Int Ed Engl 2014; 53:6115-9. [PMID: 24777818 DOI: 10.1002/anie.201402613] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Indexed: 01/28/2023]
Abstract
The enzyme-mediated site-specific bioconjugation of a radioactive metal complex to a single-chain antibody using the transpeptidase sortase A is reported. Cage amine sarcophagine ligands that were designed to function as substrates for the sortase A mediated bioconjugation to antibodies were synthesized and enzymatically conjugated to a single-chain variable fragment. The antibody fragment scFv(anti-LIBS) targets ligand-induced binding sites (LIBS) on the glycoprotein receptor GPIIb/IIIa, which is present on activated platelets. The immunoconjugates were radiolabeled with the positron-emitting isotope (64)Cu. The new radiolabeled conjugates were shown to bind selectively to activated platelets. The diagnostic potential of the most promising conjugate was demonstrated in an in vivo model of carotid artery thrombosis using positron emission tomography. This approach gives homogeneous products through site-specific enzyme-mediated conjugation and should be broadly applicable to other metal complexes and proteins.
Collapse
Affiliation(s)
- Brett M Paterson
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Vic (Australia)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Paterson BM, Alt K, Jeffery CM, Price RI, Jagdale S, Rigby S, Williams CC, Peter K, Hagemeyer CE, Donnelly PS. Enzyme-Mediated Site-Specific Bioconjugation of Metal Complexes to Proteins: Sortase-Mediated Coupling of Copper-64 to a Single-Chain Antibody. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201402613] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Kornberger P, Skerra A. Sortase-catalyzed in vitro functionalization of a HER2-specific recombinant Fab for tumor targeting of the plant cytotoxin gelonin. MAbs 2013; 6:354-66. [PMID: 24492291 DOI: 10.4161/mabs.27444] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We report on the preparation of a new type of immunotoxin via in vitro ligation of the αHer2 antigen binding fragment (Fab) of the clinically-validated antibody trastuzumab to the plant toxin gelonin, employing catalysis by the bacterial enzyme sortase A (SrtA). The αHer2 Fab was fused with the extended SrtA recognition motif LPET↓GLEH 6 at the C-terminus of its heavy chain, thereby preventing interference with antigen binding, while the toxin was equipped with a Gly 2 sequence at its N-terminus, distant to the catalytically active site in the C-terminal region. Site-specific in vitro transpeptidation led to a novel antibody-toxin conjugate wherein gelonin had effectively replaced the Fc region of a conventional (monomerized) immunoglobulin. After optimization of reaction conditions and incubation time, the resulting Fab-Gelonin ligation product was purified to homogeneity in a two-step procedure by means of Strep-Tactin affinity chromatography--utilizing the Strep-tag II appended to gelonin--and size exclusion chromatography. Binding activity of the immunotoxin for the Her2 ectodomain was indistinguishable from the unligated Fab as measured by real-time surface plasmon resonance spectroscopy. Specific cytotoxic potency of Fab-Gelonin was demonstrated against two Her2-positive cell lines, resulting in EC 50 values of ~1 nM or lower, indicating a 1000-fold enhanced cell-killing activity compared with gelonin itself. Thus, our strategy provides a convenient route to the modular construction of functional immunotoxins from Fabs of established tumor-specific antibodies with gelonin or related proteotoxins, also avoiding the elevated biosafety levels that would be mandatory for the direct biotechnological preparation of corresponding fusion proteins.
Collapse
Affiliation(s)
- Petra Kornberger
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie; Technische Universität München; Freising-Weihenstephan, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie; Technische Universität München; Freising-Weihenstephan, Germany
| |
Collapse
|
46
|
Perez HL, Cardarelli PM, Deshpande S, Gangwar S, Schroeder GM, Vite GD, Borzilleri RM. Antibody-drug conjugates: current status and future directions. Drug Discov Today 2013; 19:869-81. [PMID: 24239727 DOI: 10.1016/j.drudis.2013.11.004] [Citation(s) in RCA: 336] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/27/2013] [Accepted: 11/04/2013] [Indexed: 01/25/2023]
Abstract
Antibody-drug conjugates (ADCs) aim to take advantage of the specificity of monoclonal antibodies (mAbs) to deliver potent cytotoxic drugs selectively to antigen-expressing tumor cells. Despite the simple concept, various parameters must be considered when designing optimal ADCs, such as selection of the appropriate antigen target and conjugation method. Each component of the ADC (the antibody, linker and drug) must also be optimized to fully realize the goal of a targeted therapy with improved efficacy and tolerability. Advancements over the past several decades have led to a new generation of ADCs comprising non-immunogenic mAbs, linkers with balanced stability and highly potent cytotoxic agents. Although challenges remain, recent clinical success has generated intense interest in this therapeutic class.
Collapse
Affiliation(s)
- Heidi L Perez
- Bristol-Myers Squibb Research & Development, Princeton, NJ 08543, USA
| | - Pina M Cardarelli
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | - Shrikant Deshpande
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | - Sanjeev Gangwar
- Bristol-Myers Squibb Research & Development, Redwood City, CA 94063, USA
| | | | - Gregory D Vite
- Bristol-Myers Squibb Research & Development, Princeton, NJ 08543, USA
| | | |
Collapse
|
47
|
Nisbet RM, Nigro J, Breheney K, Caine J, Hattarki MK, Nuttall SD. Central amyloid-β-specific single chain variable fragment ameliorates Aβ aggregation and neurotoxicity. Protein Eng Des Sel 2013; 26:571-80. [PMID: 23766374 DOI: 10.1093/protein/gzt025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Anti-amyloid-β immunotherapies are a promising therapeutic approach for the treatment and prevention of Alzheimer's disease (AD). Single chain antibody fragments (scFv) are an attractive alternative to whole antibodies due to their small size, single polypeptide format and inability to stimulate potentially undesirable Fc-mediated immune effector functions. We have generated the scFv derivative of anti-Aβ monoclonal antibody, 1E8, known to target residues 17-22 of Aβ. Here we show that the soluble 1E8 scFv binds to the central region of Aβ with an affinity of ~55 nM and significantly reduces fibril formation of Aβ(1-42). Furthermore, 1E8 scFv ameliorates Aβ(1-42)-mediated toxicity in the PC12 cell line and murine primary neuronal cultures. This ability to both target the central region of Aβ and prevent Aβ(1-42) neurotoxicity in vitro makes it a promising therapeutic antibody building block for further functionalization, toward the treatment of AD.
Collapse
Affiliation(s)
- R M Nisbet
- Division of Materials Science and Engineering, CSIRO Preventative Health Flagship, 343 Royal Parade, Parkville, Victoria 3052, Australia
| | | | | | | | | | | |
Collapse
|
48
|
Ta HT, Peter K, Hagemeyer CE. Enzymatic Antibody Tagging: Toward a Universal Biocompatible Targeting Tool. Trends Cardiovasc Med 2012; 22:105-11. [DOI: 10.1016/j.tcm.2012.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|