1
|
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
|
2
|
Site-Specific Antibody Conjugation with Payloads beyond Cytotoxins. Molecules 2023; 28:molecules28030917. [PMID: 36770585 PMCID: PMC9921355 DOI: 10.3390/molecules28030917] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/12/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023] Open
Abstract
As antibody-drug conjugates have become a very important modality for cancer therapy, many site-specific conjugation approaches have been developed for generating homogenous molecules. The selective antibody coupling is achieved through antibody engineering by introducing specific amino acid or unnatural amino acid residues, peptides, and glycans. In addition to the use of synthetic cytotoxins, these novel methods have been applied for the conjugation of other payloads, including non-cytotoxic compounds, proteins/peptides, glycans, lipids, and nucleic acids. The non-cytotoxic compounds include polyethylene glycol, antibiotics, protein degraders (PROTAC and LYTAC), immunomodulating agents, enzyme inhibitors and protein ligands. Different small proteins or peptides have been selectively conjugated through unnatural amino acid using click chemistry, engineered C-terminal formylglycine for oxime or click chemistry, or specific ligation or transpeptidation with or without enzymes. Although the antibody protamine peptide fusions have been extensively used for siRNA coupling during early studies, direct conjugations through engineered cysteine or lysine residues have been demonstrated later. These site-specific antibody conjugates containing these payloads other than cytotoxic compounds can be used in proof-of-concept studies and in developing new therapeutics for unmet medical needs.
Collapse
|
3
|
Xu T, Zhang F, Chen D, Sun L, Tomazela D, Fayadat-Dilman L. Interrogating heterogeneity of cysteine-engineered antibody-drug conjugates and antibody-oligonucleotide conjugates by capillary zone electrophoresis-mass spectrometry. MAbs 2023; 15:2229102. [PMID: 37381585 DOI: 10.1080/19420862.2023.2229102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 06/11/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
Production of site-specific cysteine-engineered antibody-drug conjugates (ADCs) in mammalian cells may produce developability challenges, fragments, and heterogenous molecules, leading to potential product critical quality attributes in later development stages. Liquid phase chromatography with mass spectrometry (LC-MS) is widely used to evaluate antibody impurities and drug-to-antibody ratio, but faces challenges in analysis of fragment product variants of cysteine-engineered ADCs and oligonucleotide-to-antibody ratio (OAR) species of antibody-oligonucleotide conjugates (AOCs). Here, for the first time, we report novel capillary zone electrophoresis (CZE)-MS approaches to address the challenges above. CZE analysis of six ADCs made with different parent monoclonal antibodies (mAbs) and small molecule drug-linker payloads revealed that various fragment impurities, such as half mAbs with one/two drugs, light chains with one/two drugs, light chains with C-terminal cysteine truncation, heavy chain clippings, were well resolved from the main species. However, most of these fragments were coeluted or had signal suppression during LC-MS analysis. Furthermore, the method was optimized on both ionization and separation aspects to enable the characterization of two AOCs. The method successfully achieved baseline separation and accurate quantification of their OAR species, which were also highly challenging using conventional LC-MS methods. Finally, we compared the migration time and CZE separation profiles among ADCs and their parent mAbs, and found that properties of mAbs and linker payloads significantly influenced the separation of product variants by altering their size or charge. Our study showcases the good performance and broad applicability of CZE-MS techniques for monitoring the heterogeneity of cysteine-engineered ADCs and AOCs.
Collapse
Affiliation(s)
- Tian Xu
- Department of Chemistry Michigan State University, East Lansing MI 48824 USA
| | - Fan Zhang
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | - Daoyang Chen
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | - Liangliang Sun
- Department of Chemistry Michigan State University, East Lansing MI 48824 USA
| | - Daniela Tomazela
- Discovery Biologics, Protein Sciences, Merck & Co., Inc, South San Francisco, CA 94080 USA
| | | |
Collapse
|
4
|
18F Site-Specific Labelling of a Single-Chain Antibody against Activated Platelets for the Detection of Acute Thrombosis in Positron Emission Tomography. Int J Mol Sci 2022; 23:ijms23136886. [PMID: 35805892 PMCID: PMC9267009 DOI: 10.3390/ijms23136886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
Positron emission tomography is the imaging modality of choice when it comes to the high sensitivity detection of key markers of thrombosis and inflammation, such as activated platelets. We, previously, generated a fluorine-18 labelled single-chain antibody (scFv) against ligand-induced binding sites (LIBS) on activated platelets, binding it to the highly abundant platelet glycoprotein integrin receptor IIb/IIIa. We used a non-site-specific bio conjugation approach with N-succinimidyl-4-[18F]fluorobenzoate (S[18F]FB), leading to a mixture of products with reduced antigen binding. In the present study, we have developed and characterised a novel fluorine-18 PET radiotracer, based on this antibody, using site-specific bio conjugation to engineer cysteine residues with N-[2-(4-[18F]fluorobenzamido)ethyl]maleimide ([18F]FBEM). ScFvanti-LIBS and control antibody mut-scFv, with engineered C-terminal cysteine, were reduced, and then, they reacted with N-[2-(4-[18F]fluorobenzamido)ethyl]maleimide ([18F]FBEM). Radiolabelled scFv was injected into mice with FeCl3-induced thrombus in the left carotid artery. Clots were imaged in a PET MR imaging system, and the amount of radioactivity in major organs was measured using an ionisation chamber and image analysis. Assessment of vessel injury, as well as the biodistribution of the radiolabelled scFv, was studied. In the in vivo experiments, we found uptake of the targeted tracer in the injured vessel, compared with the non-injured vessel, as well as a high uptake of both tracers in the kidney, lung, and muscle. As expected, both tracers cleared rapidly via the kidney. Surprisingly, a large quantity of both tracers was taken up by organs with a high glutathione content, such as the muscle and lung, due to the instability of the maleimide cysteine bond in vivo, which warrants further investigations. This limits the ability of the novel antibody radiotracer 18F-scFvanti-LIBS to bind to the target in vivo and, therefore, as a useful agent for the sensitive detection of activated platelets. We describe the first fluorine-18 variant of the scFvanti-LIBS against activated platelets using site-specific bio conjugation.
Collapse
|
5
|
Espinosa LA, Ramos Y, Andújar I, Torres EO, Cabrera G, Martín A, Roche D, Chinea G, Becquet M, González I, Canaán-Haden C, Nelson E, Rojas G, Pérez-Massón B, Pérez-Martínez D, Boggiano T, Palacio J, Lozada Chang SL, Hernández L, de la Luz Hernández KR, Markku S, Vitikainen M, Valdés-Balbín Y, Santana-Medero D, Rivera DG, Vérez-Bencomo V, Emalfarb M, Tchelet R, Guillén G, Limonta M, Pimentel E, Ayala M, Besada V, González LJ. In-solution buffer-free digestion allows full-sequence coverage and complete characterization of post-translational modifications of the receptor-binding domain of SARS-CoV-2 in a single ESI-MS spectrum. Anal Bioanal Chem 2021; 413:7559-7585. [PMID: 34739558 PMCID: PMC8569510 DOI: 10.1007/s00216-021-03721-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/16/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022]
Abstract
Subunit vaccines based on the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 provide one of the most promising strategies to fight the COVID-19 pandemic. The detailed characterization of the protein primary structure by mass spectrometry (MS) is mandatory, as described in ICHQ6B guidelines. In this work, several recombinant RBD proteins produced in five expression systems were characterized using a non-conventional protocol known as in-solution buffer-free digestion (BFD). In a single ESI-MS spectrum, BFD allowed very high sequence coverage (≥ 99%) and the detection of highly hydrophilic regions, including very short and hydrophilic peptides (2-8 amino acids), and the His6-tagged C-terminal peptide carrying several post-translational modifications at Cys538 such as cysteinylation, homocysteinylation, glutathionylation, truncated glutathionylation, and cyanylation, among others. The analysis using the conventional digestion protocol allowed lower sequence coverage (80-90%) and did not detect peptides carrying most of the above-mentioned PTMs. The two C-terminal peptides of a dimer [RBD(319-541)-(His)6]2 linked by an intermolecular disulfide bond (Cys538-Cys538) with twelve histidine residues were only detected by BFD. This protocol allows the detection of the four disulfide bonds present in the native RBD, low-abundance scrambling variants, free cysteine residues, O-glycoforms, and incomplete processing of the N-terminal end, if present. Artifacts generated by the in-solution BFD protocol were also characterized. BFD can be easily implemented; it has been applied to the characterization of the active pharmaceutical ingredient of two RBD-based vaccines, and we foresee that it can be also helpful to the characterization of mutated RBDs.
Collapse
Affiliation(s)
- Luis Ariel Espinosa
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Yassel Ramos
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Ivan Andújar
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Enso Onill Torres
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Gleysin Cabrera
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Alejandro Martín
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Diamilé Roche
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Glay Chinea
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Mónica Becquet
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Isabel González
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Camila Canaán-Haden
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Elías Nelson
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Gertrudis Rojas
- Center of Molecular Immunology, 216 St., P.O. Box 16040, Havana, Cuba
| | | | | | - Tamy Boggiano
- Center of Molecular Immunology, 216 St., P.O. Box 16040, Havana, Cuba
| | - Julio Palacio
- Center of Molecular Immunology, 216 St., P.O. Box 16040, Havana, Cuba
| | | | - Lourdes Hernández
- Center of Molecular Immunology, 216 St., P.O. Box 16040, Havana, Cuba
| | | | - Saloheimo Markku
- VTT Technical Research Centre of Finland Ltd, P.O. Box 1000, 02044 VTT, Espoo, Finland
| | - Marika Vitikainen
- VTT Technical Research Centre of Finland Ltd, P.O. Box 1000, 02044 VTT, Espoo, Finland
| | | | | | - Daniel G Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Zapata & G, 10400, Havana, Cuba
| | | | - Mark Emalfarb
- Dyadic International, Inc, 140 Intercoastal Pointe Drive, Suite #404, Jupiter, FL, 33477, USA
| | - Ronen Tchelet
- Dyadic International, Inc, 140 Intercoastal Pointe Drive, Suite #404, Jupiter, FL, 33477, USA
| | - Gerardo Guillén
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Miladys Limonta
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Eulogio Pimentel
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Marta Ayala
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Vladimir Besada
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba
| | - Luis Javier González
- Center for Genetic Engineering and Biotechnology, Ave 31, e/ 158 y 190, Cubanacán, Playa, Havana, Cuba.
| |
Collapse
|
6
|
Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J Control Release 2021; 340:1-34. [PMID: 34673122 DOI: 10.1016/j.jconrel.2021.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
The antibody-drug conjugates (ADCs) are one the fastest growing biotherapeutics in oncology and are still in their infancy in gastrointestinal (GI) cancer for clinical applications to improve patient survival. The ADC based approach is developed with tumor specific antigen, antibody carrying cytotoxic agents to precisely target and deliver chemotherapeutics at the tumor site. To date, 11 ADCs have been approved by US-FDA, and more than 80 are in the clinical development phase for different oncological indications. However, The ADCs based therapies in GI cancers are still far from having high-efficient clinical outcomes. The limited success of these ADCs and lessons learned from the past are now being used to develop a newer generation of ADC against GI cancers. In this review, we did a comprehensive assessment of the key components of ADCs, including tumor marker, antibody, cytotoxic payload, and linkage strategy, with a focus on technical improvement and some future trends in the pipeline for clinical translation. The various preclinical and clinical ADCs used in gastrointestinal malignancies, their target, composition and bioconjugation, along with preclinical and clinical outcomes, are discussed. The emphasis is also given to new generation ADCs employing novel mAb, payload, linker, and bioconjugation methods are also included.
Collapse
Affiliation(s)
- Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Dheer
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilash Samykutty
- Stephenson Comprehensive Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
7
|
Site-Specific Antibody Conjugation to Engineered Double Cysteine Residues. Pharmaceuticals (Basel) 2021; 14:ph14070672. [PMID: 34358098 PMCID: PMC8308878 DOI: 10.3390/ph14070672] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 01/02/2023] Open
Abstract
Site-specific antibody conjugations generate homogeneous antibody-drug conjugates with high therapeutic index. However, there are limited examples for producing the site-specific conjugates with a drug-to-antibody ratio (DAR) greater than two, especially using engineered cysteines. Based on available Fc structures, we designed and introduced free cysteine residues into various antibody CH2 and CH3 regions to explore and expand this technology. The mutants were generated using site-directed mutagenesis with good yield and properties. Conjugation efficiency and selectivity were screened using PEGylation. The top single cysteine mutants were then selected and combined as double cysteine mutants for expression and further investigation. Thirty-six out of thirty-eight double cysteine mutants display comparable expression with low aggregation similar to the wild-type antibody. PEGylation screening identified seventeen double cysteine mutants with good conjugatability and high selectivity. PEGylation was demonstrated to be a valuable and efficient approach for quickly screening mutants for high selectivity as well as conjugation efficiency. Our work demonstrated the feasibility of generating antibody conjugates with a DAR greater than 3.4 and high site-selectivity using THIOMABTM method. The top single or double cysteine mutants identified can potentially be applied to site-specific antibody conjugation of cytotoxin or other therapeutic agents as a next generation conjugation strategy.
Collapse
|
8
|
Mills BJ, Kruger T, Bruncko M, Zhang X, Jameel F. Effect of Linker-Drug Properties and Conjugation Site on the Physical Stability of ADCs. J Pharm Sci 2020; 109:1662-1672. [PMID: 32027921 DOI: 10.1016/j.xphs.2020.01.029] [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] [Received: 07/01/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 11/30/2022]
Abstract
The physical stability of antibody drug conjugates is dictated by the properties of the antibody, linker-drug, and conjugation site. Two linker-drugs were chosen that are different in terms of hydrophobicity and polar surface area to evaluate the effect of linker-drug properties on antibody-drug conjugate (ADC) behavior. Site-specific and non-site-specific conjugation was used to investigate the role of conjugation site in conformational and colloidal stability. Finally, 2 antibodies were selected to determine if the observed results were antibody-specific. The conformational stability is affected, with the highest degree of destabilization observed when conjugation results in the removal of interchain disulfide bonds. Although conformational destabilization occurred in the domain in which conjugation occurred and domains distinct from the conjugation site, no correlation could be drawn between linker-drug properties and conformational stability. Evaluation of aggregation by size exclusion HPLC confirmed a relationship between linker-drug hydrophobicity and aggregation propensity under thermal stress in all ADCs tested. The extent of aggregation was far greater in the conjugates generated with a more hydrophobic antibody, illustrating that the properties of both the antibody and linker-drug contribute to aggregation. These studies emphasize that the distinct properties of the molecule as a whole warrant a case-by-case evaluation of each ADC.
Collapse
Affiliation(s)
- Brittney J Mills
- Drug Product Development, AbbVie Inc., North Chicago, Illinois 60064.
| | - Terra Kruger
- Drug Product Development, AbbVie Inc., North Chicago, Illinois 60064; Division of Pharmaceutics, University of Iowa, Iowa City, Iowa 52242
| | - Milan Bruncko
- Global Biologics, AbbVie Inc., North Chicago, Illinois 60064
| | - Xinxin Zhang
- Global Biologics, AbbVie Inc., North Chicago, Illinois 60064
| | - Feroz Jameel
- Drug Product Development, AbbVie Inc., North Chicago, Illinois 60064
| |
Collapse
|
9
|
Bai C, Reid EE, Wilhelm A, Shizuka M, Maloney EK, Laleau R, Harvey L, Archer KE, Vitharana D, Adams S, Kovtun Y, Miller ML, Chari R, Keating TA, Yoder NC. Site-Specific Conjugation of the Indolinobenzodiazepine DGN549 to Antibodies Affords Antibody-Drug Conjugates with an Improved Therapeutic Index as Compared with Lysine Conjugation. Bioconjug Chem 2019; 31:93-103. [PMID: 31747250 DOI: 10.1021/acs.bioconjchem.9b00777] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibody-drug conjugates have elicited great interest recently as targeted chemotherapies for cancer. Recent preclinical and clinical data have continued to raise questions about optimizing the design of these complex therapeutics. Biochemical methods for site-specific antibody conjugation have been a design feature of recent clinical ADCs, and preclinical reports suggest that site-specifically conjugated ADCs generically offer improved therapeutic indices (i.e., the fold difference between efficacious and maximum tolerated doses). Here we present the results of a systematic preclinical comparison of ADCs embodying the DNA-alkylating linker-payload DGN549 generated with both heterogeneous lysine-directed and site-specific cysteine-directed conjugation chemistries. Importantly, the catabolites generated by each ADC are the same regardless of the conjugation format. In two different model systems evaluated, the site-specific ADC showed a therapeutic index benefit. However, the therapeutic index benefit is different in each case: both show evidence of improved tolerability, though with different magnitudes, and in one case significant efficacy improvement is also observed. These results support our contention that conjugation chemistry of ADCs is best evaluated in the context of a particular antibody, target, and linker-payload, and ideally across multiple disease models.
Collapse
Affiliation(s)
- Chen Bai
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Emily E Reid
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Alan Wilhelm
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Manami Shizuka
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Erin K Maloney
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Rassol Laleau
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Lauren Harvey
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Katie E Archer
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Dilrukshi Vitharana
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Sharlene Adams
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Yelena Kovtun
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Michael L Miller
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Ravi Chari
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Thomas A Keating
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| | - Nicholas C Yoder
- Science, Technology, and Translation , ImmunoGen, Inc. , 830 Winter Street , Waltham , Massachusetts 02451 , United States
| |
Collapse
|
10
|
Yoder NC, Bai C, Tavares D, Widdison WC, Whiteman KR, Wilhelm A, Wilhelm SD, McShea MA, Maloney EK, Ab O, Wang L, Jin S, Erickson HK, Keating TA, Lambert JM. A Case Study Comparing Heterogeneous Lysine- and Site-Specific Cysteine-Conjugated Maytansinoid Antibody-Drug Conjugates (ADCs) Illustrates the Benefits of Lysine Conjugation. Mol Pharm 2019; 16:3926-3937. [PMID: 31287952 DOI: 10.1021/acs.molpharmaceut.9b00529] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates are an emerging class of cancer therapeutics constructed from monoclonal antibodies conjugated with small molecule effectors. First-generation molecules of this class often employed heterogeneous conjugation chemistry, but many site-specifically conjugated ADCs have been described recently. Here, we undertake a systematic comparison of ADCs made with the same antibody and the same macrocyclic maytansinoid effector but conjugated either heterogeneously at lysine residues or site-specifically at cysteine residues. Characterization of these ADCs in vitro reveals generally similar properties, including a similar catabolite profile, a key element in making a meaningful comparison of conjugation chemistries. In a mouse model of cervical cancer, the lysine-conjugated ADC affords greater efficacy on a molar payload basis. Rather than making general conclusions about ADCs conjugated by a particular chemistry, we interpret these results as highlighting the complexity of ADCs and the interplay between payload class, linker chemistry, target antigen, and other variables that determine efficacy in a given setting.
Collapse
|
11
|
Davydova M, Dewaele Le Roi G, Adumeau P, Zeglis BM. Synthesis and Bioconjugation of Thiol-Reactive Reagents for the Creation of Site-Selectively Modified Immunoconjugates. J Vis Exp 2019. [PMID: 30907883 DOI: 10.3791/59063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Maleimide-bearing bifunctional probes have been employed for decades for the site-selective modification of thiols in biomolecules, especially antibodies. Yet maleimide-based conjugates display limited stability in vivo because the succinimidyl thioether linkage can undergo a retro-Michael reaction. This, of course, can lead to the release of the radioactive payload or its exchange with thiol-bearing biomolecules in circulation. Both of these processes can produce elevated activity concentrations in healthy organs as well as decreased activity concentrations in target tissues, resulting in reduced imaging contrast and lower therapeutic ratios. In 2018, we reported the creation of a modular, stable, and easily accessible phenyloxadiazolyl methyl sulfone reagent - dubbed 'PODS' - as a platform for thiol-based bioconjugations. We have clearly demonstrated that PODS-based site-selective bioconjugations reproducibly and robustly create homogenous, well-defined, highly immunoreactive, and highly stable radioimmunoconjugates. Furthermore, preclinical experiments in murine models of colorectal cancer have shown that these site-selectively labeled radioimmunoconjugates exhibit far superior in vivo performance compared to radiolabeled antibodies synthesized via maleimide-based conjugations. In this protocol, we will describe the four-step synthesis of PODS, the creation of a bifunctional PODS-bearing variant of the ubiquitous chelator DOTA (PODS-DOTA), and the conjugation of PODS-DOTA to the HER2-targeting antibody trastuzumab.
Collapse
Affiliation(s)
- Maria Davydova
- Department of Chemistry, Hunter College of the City University of New York
| | - Guillaume Dewaele Le Roi
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York
| | - Pierre Adumeau
- Department of Chemistry, Hunter College of the City University of New York
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York; Ph.D. Program in Chemistry, Graduate Center of the City University of New York; Department of Radiology, Memorial Sloan Kettering Cancer Center; Department of Radiology, Weill Cornell Medical College;
| |
Collapse
|
12
|
Alves NJ. Antibody conjugation and formulation. Antib Ther 2019; 2:33-39. [PMID: 33928219 PMCID: PMC7990145 DOI: 10.1093/abt/tbz002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/31/2019] [Accepted: 02/08/2019] [Indexed: 11/14/2022] Open
Abstract
In an era where ultra-high antibody concentrations, high viscosities, low volumes, auto-injectors and long storage requirements are already complex problems with the current unconjugated monoclonal antibodies on the market, the formulation demands for antibody-drug conjugates (ADCs) are significant. Antibodies have historically been administered at relatively low concentrations through intravenous (IV) infusion due to their large size and the inability to formulate for oral delivery. Due to the high demands associated with IV infusion and the development of novel antibody targets and unique antibody conjugates, more accessible routes of administration such as intramuscular and subcutaneous are being explored. This review will summarize various site-specific and non-site-specific antibody conjugation techniques in the context of ADCs and the demands of formulation for high concentration clinical implementation.
Collapse
Affiliation(s)
- Nathan J Alves
- Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
13
|
Morais M, Ma MT. Site-specific chelator-antibody conjugation for PET and SPECT imaging with radiometals. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 30:91-104. [PMID: 30553525 PMCID: PMC6291455 DOI: 10.1016/j.ddtec.2018.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/17/2022]
Abstract
Antibodies and their derivatives radiolabelled with positron- and gamma-emitting radiometals enable sensitive and quantitative molecular Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT) imaging of antibody distribution in vivo. Chelators that are covalently attached to antibodies allow radiolabelling with metallic PET and SPECT radioisotopes. Conventional strategies for chelator-protein conjugation generate heterogeneous mixtures of bioconjugates that can exhibit reduced affinity for their receptor targets, and undesirable biodistribution and pharmacokinetics. Recent advances in bioconjugation technology enable site-specific modification to generate well-defined constructs with superior properties. Herein we survey existing site-specific chelator-protein conjugation methods. These include chelator attachment to cysteines/disulfide bonds or the glycan region of the antibody, enzyme-mediated chelator conjugation, and incorporation of sequences of amino acids that chelate the radiometal. Such technology will allow better use of PET and SPECT imaging in the development of antibody-based therapies.
Collapse
Affiliation(s)
- Mauricio Morais
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom.
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| |
Collapse
|
14
|
Tuwahatu CA, Yeung CC, Lam YW, Roy VAL. The molecularly imprinted polymer essentials: curation of anticancer, ophthalmic, and projected gene therapy drug delivery systems. J Control Release 2018; 287:24-34. [PMID: 30110614 DOI: 10.1016/j.jconrel.2018.08.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/10/2018] [Accepted: 08/11/2018] [Indexed: 02/06/2023]
Abstract
The development of polymeric materials as drug delivery systems has advanced from systems that rely on classical passive targeting to carriers that can sustain the precisely controlled release of payloads upon physicochemical triggers in desired microenvironment. Molecularly imprinted polymers (MIP), materials designed to capture specific molecules based on their molecular shape and charge distribution, are attractive candidates for fulfilling these purposes. In particular, drug-imprinted polymers coupled with active targeting mechanisms have been explored as potential drug delivery systems. In this review, we have curated important recent efforts in the development of drug-imprinted polymers in a variety of clinical applications, especially oncology and ophthalmology. MIP possesses properties that may complement the traditional delivery systems of these two disciplines, such as passive enhanced permeability and retention effect (EPR) in cancer tumors, and passive drug diffusion in delivering ophthalmic therapeutics. Furthermore, the prospects of MIP integration with the emerging gene therapies will be discussed.
Collapse
Affiliation(s)
- Christian Antonio Tuwahatu
- Department of Materials Science and Engineering and State Key Laboratory of Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Chi Chung Yeung
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Vellaisamy Arul Lenus Roy
- Department of Materials Science and Engineering and State Key Laboratory of Millimeter Waves, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
15
|
Adumeau P, Davydova M, Zeglis BM. Thiol-Reactive Bifunctional Chelators for the Creation of Site-Selectively Modified Radioimmunoconjugates with Improved Stability. Bioconjug Chem 2018; 29:1364-1372. [PMID: 29509393 DOI: 10.1021/acs.bioconjchem.8b00081] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Maleimide-bearing bifunctional chelators have been used extensively for the site-selective bioconjugation and radiolabeling of peptides and proteins. However, bioconjugates obtained using these constructs inevitably suffer from limited stability in vivo, a trait that translates into suboptimal activity concentrations in target tissues and higher uptake levels in healthy, nontarget tissues. To circumvent this issue, phenyloxadiazolyl methylsulfones have previously been reported as alternatives to maleimides for thiol-based ligations, but these constructs have scarcely been used in the field of radiochemistry. In this report, we describe the synthesis of two thiol-reactive bifunctional chelators for 89Zr and 177Lu based on a new, easy-to-make phenyloxadiazolyl methylsulfone reagent, PODS. Radioimmunoconjugates created using these novel bifunctional chelators displayed in vitro stability that was higher than that of their maleimide-derived cousins. More importantly, positron emission tomography imaging in murine models of cancer revealed that a 89Zr-labeled radioimmunoconjugate created using a PODS-bearing bifunctional chelator produced a rate of uptake in nontarget tissues that is significantly lower than that of its analogous maleimide-based counterpart.
Collapse
Affiliation(s)
- Pierre Adumeau
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10028 , United States.,Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Maria Davydova
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10028 , United States
| | - Brian M Zeglis
- Department of Chemistry , Hunter College of the City University of New York , New York , New York 10028 , United States.,Department of Radiology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Ph.D. Program in Chemistry , Graduate Center of the City University of New York , New York , New York 10016 , United States.,Department of Radiology , Weill Cornell Medical College , New York , New York 10065 , United States
| |
Collapse
|
16
|
Sussman D, Westendorf L, Meyer DW, Leiske CI, Anderson M, Okeley NM, Alley SC, Lyon R, Sanderson RJ, Carter PJ, Benjamin DR. Engineered cysteine antibodies: an improved antibody-drug conjugate platform with a novel mechanism of drug-linker stability. Protein Eng Des Sel 2018; 31:47-54. [DOI: 10.1093/protein/gzx067] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/08/2018] [Indexed: 12/16/2022] Open
|
17
|
Gilroy JJ, Eakin CM. Characterization of drug load variants in a thiol linked antibody-drug conjugate using multidimensional chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2017. [DOI: 10.1016/j.jchromb.2017.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
18
|
Tumey LN, Li F, Rago B, Han X, Loganzo F, Musto S, Graziani EI, Puthenveetil S, Casavant J, Marquette K, Clark T, Bikker J, Bennett EM, Barletta F, Piche-Nicholas N, Tam A, O'Donnell CJ, Gerber HP, Tchistiakova L. Site Selection: a Case Study in the Identification of Optimal Cysteine Engineered Antibody Drug Conjugates. AAPS JOURNAL 2017; 19:1123-1135. [PMID: 28439809 DOI: 10.1208/s12248-017-0083-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/03/2017] [Indexed: 11/30/2022]
Abstract
As the antibody drug conjugate (ADC) community continues to shift towards site-specific conjugation technology, there is a growing need to understand how the site of conjugation impacts the biophysical and biological properties of an ADC. In order to address this need, we prepared a carefully selected series of engineered cysteine ADCs and proceeded to systematically evaluate their potency, stability, and PK exposure. The site of conjugation did not have a significant influence on the thermal stability and in vitro cytotoxicity of the ADCs. However, we demonstrate that the rate of cathepsin-mediated linker cleavage is heavily dependent upon site and is closely correlated with ADC hydrophobicity, thus confirming other recent reports of this phenomenon. Interestingly, conjugates with high rates of cathepsin-mediated linker cleavage did not exhibit decreased plasma stability. In fact, the major source of plasma instability was shown to be retro-Michael mediated deconjugation. This process is known to be impeded by succinimide hydrolysis, and thus, we undertook a series of mutational experiments demonstrating that basic residues located nearby the site of conjugation can be a significant driver of succinimide ring opening. Finally, we show that total antibody PK exposure in rat was loosely correlated with ADC hydrophobicity. It is our hope that these observations will help the ADC community to build "design rules" that will enable more efficient prosecution of next-generation ADC discovery programs.
Collapse
Affiliation(s)
- L Nathan Tumey
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, P.O. Box 6000, Binghamton, New York, 13902-6000, USA.
| | - Fengping Li
- Biomedicine Design, Pfizer, Inc., Cambridge, Massachusetts, 06379, USA
| | - Brian Rago
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | - Xiaogang Han
- PKDM, Amgen, Inc., 360 Binney Street, AMA 1, Cambridge, Massachusetts, 02142, USA
| | - Frank Loganzo
- Oncology Research and Development, Pfizer, Inc., 401 N. Middletown Rd., Pearl River, New York, 10965, USA
| | - Sylvia Musto
- Oncology Research and Development, Pfizer, Inc., 401 N. Middletown Rd., Pearl River, New York, 10965, USA
| | - Edmund I Graziani
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | | | - Jeffrey Casavant
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | | | - Tracey Clark
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | - Jack Bikker
- International Flavors and Fragrances, 521 West 57th Street, New York, New York, 10019, USA
| | - Eric M Bennett
- Biomedicine Design, Pfizer, Inc., Cambridge, Massachusetts, 06379, USA
| | - Frank Barletta
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | | | - Amy Tam
- Biomedicine Design, Pfizer, Inc., Cambridge, Massachusetts, 06379, USA
| | - Christopher J O'Donnell
- Worldwide Research and Development, Pfizer, Inc., 445 Eastern Point Road, Groton, Connecticut, 06379, USA
| | - Hans Peter Gerber
- Maverick Therapeutics, Inc, 3260 Bayshore Blvd, Brisbane, California, 94005, USA
| | | |
Collapse
|
19
|
Dimasi N, Fleming R, Zhong H, Bezabeh B, Kinneer K, Christie RJ, Fazenbaker C, Wu H, Gao C. Efficient Preparation of Site-Specific Antibody-Drug Conjugates Using Cysteine Insertion. Mol Pharm 2017; 14:1501-1516. [PMID: 28245132 DOI: 10.1021/acs.molpharmaceut.6b00995] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Antibody-drug conjugates (ADCs) are a class of biopharmaceuticals that combine the specificity of antibodies with the high-potency of cytotoxic drugs. Engineering cysteine residues in the antibodies using mutagenesis is a common method to prepare site-specific ADCs. With this approach, solvent accessible amino acids in the antibody have been selected for substitution with cysteine for conjugating maleimide-bearing cytotoxic drugs, resulting in homogeneous and stable site-specific ADCs. Here we describe a cysteine engineering approach based on the insertion of cysteines before and after selected sites in the antibody, which can be used for site-specific preparation of ADCs. Cysteine-inserted antibodies have expression level and monomeric content similar to the native antibodies. Conjugation to a pyrrolobenzodiazepine dimer (SG3249) resulted in comparable efficiency of site-specific conjugation between cysteine-inserted and cysteine-substituted antibodies. Cysteine-inserted ADCs were shown to have biophysical properties, FcRn, and antigen binding affinity similar to the cysteine-substituted ADCs. These ADCs were comparable for serum stability to the ADCs prepared using cysteine-mutagenesis and had selective and potent cytotoxicity against human prostate cancer cells. Two of the cysteine-inserted variants abolish binding of the resulting ADCs to FcγRs in vitro, thereby potentially preventing non-target mediated uptake of the ADCs by cells of the innate immune system that express FcγRs, which may result in mitigating off-target toxicities. A selected cysteine-inserted ADC demonstrated potent dose-dependent anti-tumor activity in a xenograph tumor mouse model of human breast adenocarcinoma expressing the oncofetal antigen 5T4.
Collapse
Affiliation(s)
- Nazzareno Dimasi
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Ryan Fleming
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Haihong Zhong
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Binyam Bezabeh
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Krista Kinneer
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Ronald J Christie
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Christine Fazenbaker
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Herren Wu
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| | - Changshou Gao
- Antibody Discovery and Protein Engineering and ‡Oncology Research, MedImmune , Gaithersburg, Maryland 20878, United States
| |
Collapse
|
20
|
Zhong X, He T, Prashad AS, Wang W, Cohen J, Ferguson D, Tam AS, Sousa E, Lin L, Tchistiakova L, Gatto S, D'Antona A, Luan YT, Ma W, Zollner R, Zhou J, Arve B, Somers W, Kriz R. Mechanistic understanding of the cysteine capping modifications of antibodies enables selective chemical engineering in live mammalian cells. J Biotechnol 2017; 248:48-58. [PMID: 28300660 DOI: 10.1016/j.jbiotec.2017.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/02/2017] [Accepted: 03/06/2017] [Indexed: 12/21/2022]
Abstract
Protein modifications by intricate cellular machineries often redesign the structure and function of existing proteins to impact biological networks. Disulfide bond formation between cysteine (Cys) pairs is one of the most common modifications found in extracellularly-destined proteins, key to maintaining protein structure. Unpaired surface cysteines on secreted mammalian proteins are also frequently found disulfide-bonded with free Cys or glutathione (GSH) in circulation or culture, the mechanism for which remains unknown. Here we report that these so-called Cys-capping modifications take place outside mammalian cells, not in the endoplasmic reticulum (ER) where oxidoreductase-mediated protein disulfide formation occurs. Unpaired surface cysteines of extracellularly-arrived proteins such as antibodies are uncapped upon secretion before undergoing disulfide exchange with cystine or oxidized GSH in culture medium. This observation has led to a feasible way to selectively modify the nucleophilic thiol side-chain of cell-surface or extracellular proteins in live mammalian cells, by applying electrophiles with a chemical handle directly into culture medium. These findings provide potentially an effective approach for improving therapeutic conjugates and probing biological systems.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States.
| | - Tao He
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Amar S Prashad
- Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, Pearl River, NY 10965,United States
| | | | - Justin Cohen
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Darren Ferguson
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Amy S Tam
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Eric Sousa
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Laura Lin
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Lioudmila Tchistiakova
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Scott Gatto
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Aaron D'Antona
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | | | - Weijun Ma
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Richard Zollner
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Jing Zhou
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Bo Arve
- Pharmaceutical Sciences, Medicinal Sciences, Pfizer Worldwide R&D, Pearl River, NY 10965,United States
| | - Will Somers
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| | - Ronald Kriz
- BioMedicine Design, Medicinal Sciences, Pfizer Worldwide R&D, Cambridge, MA 02139, United States
| |
Collapse
|
21
|
Coming-of-Age of Antibodies in Cancer Therapeutics. Trends Pharmacol Sci 2016; 37:1009-1028. [DOI: 10.1016/j.tips.2016.09.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023]
|
22
|
Soni KS, Desale SS, Bronich TK. Nanogels: An overview of properties, biomedical applications and obstacles to clinical translation. J Control Release 2016; 240:109-126. [PMID: 26571000 PMCID: PMC4862943 DOI: 10.1016/j.jconrel.2015.11.009] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/01/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
Nanogels have emerged as a versatile hydrophilic platform for encapsulation of guest molecules with a capability to respond to external stimuli that can be used for a multitude of applications. These are soft materials capable of holding small molecular therapeutics, biomacromolecules, and inorganic nanoparticles within their crosslinked networks, which allows them to find applications for therapy as well as imaging of a variety of disease conditions. Their stimuli-responsive behavior can be easily controlled by selection of constituent polymer and crosslinker components to achieve a desired response at the site of action, which imparts nanogels the ability to participate actively in the intended function of the carrier system rather than being passive carriers of their cargo. These properties not only enhance the functionality of the carrier system but also help in overcoming many of the challenges associated with the delivery of cargo molecules, and this review aims to highlight the distinct and unique capabilities of nanogels as carrier systems for the delivery of an array of cargo molecules over other nanomaterials. Despite their obvious usefulness, nanogels are still not a commonplace occurrence in clinical practice. We have also made an attempt to highlight some of the major challenges that need to be overcome to advance nanogels further in the field of biomedical applications.
Collapse
Affiliation(s)
- Kruti S Soni
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Swapnil S Desale
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| |
Collapse
|
23
|
Adumeau P, Sharma SK, Brent C, Zeglis BM. Site-Specifically Labeled Immunoconjugates for Molecular Imaging--Part 1: Cysteine Residues and Glycans. Mol Imaging Biol 2016; 18:1-17. [PMID: 26754790 PMCID: PMC4722084 DOI: 10.1007/s11307-015-0919-4] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Due to their remarkable selectivity and specificity for cancer biomarkers, immunoconjugates have emerged as extremely promising vectors for the delivery of diagnostic radioisotopes and fluorophores to malignant tissues. Paradoxically, however, these tools for precision medicine are synthesized in a remarkably imprecise way. Indeed, the vast majority of immunoconjugates are created via the random conjugation of bifunctional probes (e.g., DOTA-NCS) to amino acids within the antibody (e.g., lysines). Yet antibodies have multiple copies of these residues throughout their macromolecular structure, making control over the location of the conjugation reaction impossible. This lack of site specificity can lead to the formation of poorly defined, heterogeneous immunoconjugates with suboptimal in vivo behavior. Over the past decade, interest in the synthesis and development of site-specifically labeled immunoconjugates—both antibody-drug conjugates as well as constructs for in vivo imaging—has increased dramatically, and a number of reports have suggested that these better defined, more homogeneous constructs exhibit improved performance in vivo compared to their randomly modified cousins. In this two-part review, we seek to provide an overview of the various methods that have been developed to create site-specifically modified immunoconjugates for positron emission tomography, single photon emission computed tomography, and fluorescence imaging. We will begin with an introduction to the structure of antibodies and antibody fragments. This is followed by the core of the work: sections detailing the four different approaches to site-specific modification strategies based on cysteine residues, glycans, peptide tags, and unnatural amino acids. These discussions will be divided into two installments: cysteine residues and glycans will be detailed in Part 1 of the review, while peptide tags and unnatural amino acids will be addressed in Part 2. Ultimately, we sincerely hope that this review fosters interest and enthusiasm for site-specific immunoconjugates within the nuclear medicine and molecular imaging communities.
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 Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY10065, NY, 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, NY10065, NY, USA.
| |
Collapse
|
24
|
Kim Y, Park EJ, Na DH. Antibody–drug conjugates for targeted anticancer drug delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0254-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
25
|
Shinmi D, Taguchi E, Iwano J, Yamaguchi T, Masuda K, Enokizono J, Shiraishi Y. One-Step Conjugation Method for Site-Specific Antibody-Drug Conjugates through Reactive Cysteine-Engineered Antibodies. Bioconjug Chem 2016; 27:1324-31. [PMID: 27074832 DOI: 10.1021/acs.bioconjchem.6b00133] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineered cysteine residues are particularly convenient for site-specific conjugation of antibody-drug conjugates (ADC), because no cell engineering and additives are required. Usually, unpaired cysteine residues form mixed disulfides during fermentation in Chinese hamster ovarian (CHO) cells; therefore, additional reduction and oxidization steps are required prior to conjugation. In this study, we prepared light chain (Lc)-Q124C variants in IgG and examined the conjugation efficiency. Intriguingly, Lc-Q124C exhibited high thiol reactivity and directly generated site-specific ADC without any pretreatment (named active thiol antibody: Actibody). Most of the cysteine-maleimide conjugates including Lc-Q124C showed retro-Michael reaction with cysteine 34 in albumin and were decomposed over time. In order to acquire resistance to a maleimide exchange reaction, the facile procedure for succinimide hydrolysis on anion exchange resin was employed. Hydrolyzed Lc-Q124C conjugate prepared with anion exchange procedure retained high stability in plasma. Recently, various stable linkage schemes for cysteine conjugation have been reported. The combination with direct conjugation by the use of Actibody and stable linker technology could enable the generation of stable site-specific ADC through a simple method. Actibody technology with Lc-Q124C at a less exposed position opens a new path for cysteine-based conjugation, and contributes to reducing entry barriers to the preparation and evaluation of ADC.
Collapse
Affiliation(s)
- Daisuke Shinmi
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Eri Taguchi
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Junko Iwano
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Tsuyoshi Yamaguchi
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Kazuhiro Masuda
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Junichi Enokizono
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| | - Yasuhisa Shiraishi
- Research Core Function Laboratories and §Innovative Technology Laboratories, Research Functions Unit, #R&D Planning Department, R&D Division, and ‡Bio Process Research and Development Laboratories, Production Division, Kyowa Hakko Kirin Co., Ltd. , Tokyo, 100-8185, Japan
| |
Collapse
|
26
|
Schumacher D, Hackenberger CPR, Leonhardt H, Helma J. Current Status: Site-Specific Antibody Drug Conjugates. J Clin Immunol 2016; 36 Suppl 1:100-7. [PMID: 27003914 PMCID: PMC4891387 DOI: 10.1007/s10875-016-0265-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 12/04/2022]
Abstract
Antibody drug conjugates (ADCs), a promising class of cancer biopharmaceuticals, combine the specificity of therapeutic antibodies with the pharmacological potency of chemical, cytotoxic drugs. Ever since the first ADCs on the market, a plethora of novel ADC technologies has emerged, covering as diverse aspects as antibody engineering, chemical linker optimization and novel conjugation strategies, together aiming at constantly widening the therapeutic window for ADCs. This review primarily focuses on novel chemical and biotechnological strategies for the site-directed attachment of drugs that are currently validated for 2nd generation ADCs to promote conjugate homogeneity and overall stability.
Collapse
Affiliation(s)
- Dominik Schumacher
- Chemical Biology and Department of Chemistry, Leibniz-Institut für Molekulare Pharmakologie and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christian P R Hackenberger
- Chemical Biology and Department of Chemistry, Leibniz-Institut für Molekulare Pharmakologie and Humboldt Universität zu Berlin, Berlin, Germany
| | - Heinrich Leonhardt
- Department of Biology II, Ludwig-Maximilians-Universität München and Center for Integrated Protein Science Munich, Planegg-Martinsried, Germany
| | - Jonas Helma
- Department of Biology II, Ludwig-Maximilians-Universität München and Center for Integrated Protein Science Munich, Planegg-Martinsried, Germany.
| |
Collapse
|
27
|
Yoon A, Shin JW, Kim S, Kim H, Chung J. Chicken scFvs with an Artificial Cysteine for Site-Directed Conjugation. PLoS One 2016; 11:e0146907. [PMID: 26764487 PMCID: PMC4713166 DOI: 10.1371/journal.pone.0146907] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/23/2015] [Indexed: 01/08/2023] Open
Abstract
For the site-directed conjugation of chemicals and radioisotopes to the chicken-derived single-chain variable fragment (scFv), we investigated amino acid residues replaceable with cysteine. By replacing each amino acid of the 157 chicken variable region framework residues (FR, 82 residues on VH and 75 on VL) with cysteine, 157 artificial cysteine mutants were generated and characterized. At least 27 residues on VL and 37 on VH could be replaced with cysteine while retaining the binding activity of the original scFv. We prepared three VL (L5, L6 and L7) and two VH (H13 and H16) mutants as scFv-Ckappa fusion proteins and showed that PEG-conjugation to the sulfhydryl group of the artificial cysteine was achievable in all five mutants. Because the charge around the cysteine residue affects the in vivo stability of thiol-maleimide conjugation, we prepared 16 charge-variant artificial cysteine mutants by replacing the flanking residues of H13 with charged amino acids and determined that the binding activity was not affected in any of the mutants except one. We prepared four charge-variant H13 artificial cysteine mutants (RCK, DCE, ECD and ECE) as scFv-Ckappa fusion proteins and confirmed that the reactivity of the sulfhydryl group on cysteine is active and their binding activity is retained after the conjugation process.
Collapse
Affiliation(s)
- Aerin Yoon
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
| | - Jung Won Shin
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
| | - Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyori Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, South Korea
- * E-mail:
| |
Collapse
|
28
|
|
29
|
Shiraishi Y, Muramoto T, Nagatomo K, Shinmi D, Honma E, Masuda K, Yamasaki M. Identification of Highly Reactive Cysteine Residues at Less Exposed Positions in the Fab Constant Region for Site-Specific Conjugation. Bioconjug Chem 2015; 26:1032-40. [DOI: 10.1021/acs.bioconjchem.5b00080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Yasuhisa Shiraishi
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Takashige Muramoto
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Kazutaka Nagatomo
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Daisuke Shinmi
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Emiko Honma
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Kazuhiro Masuda
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| | - Motoo Yamasaki
- Innovative Technology Laboratories, Research Functions Unit, R&D Division, Kyowa Hakko Kirin Co., Ltd., Tokyo 194-8533, Japan
| |
Collapse
|
30
|
Agarwal P, Bertozzi CR. Site-specific antibody-drug conjugates: the nexus of bioorthogonal chemistry, protein engineering, and drug development. Bioconjug Chem 2015; 26:176-92. [PMID: 25494884 PMCID: PMC4335810 DOI: 10.1021/bc5004982] [Citation(s) in RCA: 450] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Antibody–drug
conjugates (ADCs) combine the specificity
of antibodies with the potency of small molecules to create targeted
drugs. Despite the simplicity of this concept, generation of clinically
successful ADCs has been very difficult. Over the past several decades,
scientists have learned a great deal about the constraints on antibodies,
linkers, and drugs as they relate to successful construction of ADCs.
Once these components are in hand, most ADCs are prepared by nonspecific
modification of antibody lysine or cysteine residues with drug-linker
reagents, which results in heterogeneous product mixtures that cannot
be further purified. With advances in the fields of bioorthogonal
chemistry and protein engineering, there is growing interest in producing
ADCs by site-specific conjugation to the antibody, yielding more homogeneous
products that have demonstrated benefits over their heterogeneous
counterparts in vivo. Here, we chronicle the development
of a multitude of site-specific conjugation strategies for assembly
of ADCs and provide a comprehensive account of key advances and their
roots in the fields of bioorthogonal chemistry and protein engineering.
Collapse
Affiliation(s)
- Paresh Agarwal
- Departments of Chemistry and ‡Molecular and Cell Biology and §Howard Hughes Medical Institute, University of California , Berkeley, California 94720, United States
| | | |
Collapse
|
31
|
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: 239] [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
|
32
|
Shefet-Carasso L, Benhar I. Antibody-targeted drugs and drug resistance--challenges and solutions. Drug Resist Updat 2014; 18:36-46. [PMID: 25476546 DOI: 10.1016/j.drup.2014.11.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/16/2014] [Accepted: 11/17/2014] [Indexed: 11/24/2022]
Abstract
Antibody-based therapy of various human malignancies has shown efficacy in the past 30 years and is now one of the most successful and leading strategies for targeted treatment of patients harboring hematological malignancies and solid tumors. Antibody-drug conjugates (ADCs) aim to take advantage of the affinity and specificity of monoclonal antibodies (mAbs) to selectively deliver potent cytotoxic drugs to antigen-expressing tumor cells. Key parameters for ADC include choosing the optimal components of the ADC (the antibody, the linker and the cytotoxic drug) and selecting the suitable cell-surface target antigen. Building on the success of recent FDA approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla), ADCs are currently a class of drugs with a robust pipeline with clinical applications that are rapidly expanding. The more ADCs are being evaluated in preclinical models and clinical trials, the clearer are becoming the parameters and the challenges required for their therapeutic success. This rapidly growing knowledge and clinical experience are revealing novel modalities and mechanisms of resistance to ADCs, hence offering plausible solutions to such challenges. Here, we review the key parameters for designing a powerful ADC, focusing on how ADCs are addressing the challenge of multiple drug resistance (MDR) and its rational overcoming.
Collapse
Affiliation(s)
- LeeRon Shefet-Carasso
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel
| | - Itai Benhar
- Department of Molecular Microbiology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
33
|
Lacek K, Urbanowicz RA, Troise F, De Lorenzo C, Severino V, Di Maro A, Tarr AW, Ferrara F, Ploss A, Temperton N, Ball JK, Nicosia A, Cortese R, Pessi A. Dramatic potentiation of the antiviral activity of HIV antibodies by cholesterol conjugation. J Biol Chem 2014; 289:35015-28. [PMID: 25342747 PMCID: PMC4263897 DOI: 10.1074/jbc.m114.591826] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The broadly neutralizing antibodies HIV 2F5 and 4E10, which bind to overlapping epitopes in the membrane-proximal external region of the fusion protein gp41, have been proposed to use a two-step mechanism for neutralization; first, they bind and preconcentrate at the viral membrane through their long, hydrophobic CDRH3 loops, and second, they form a high affinity complex with the protein epitope. Accordingly, mutagenesis of the CDRH3 can abolish their neutralizing activity, with no change in the affinity for the peptide epitope. We show here that we can mimic this mechanism by conjugating a cholesterol group outside of the paratope of an antibody. Cholesterol-conjugated antibodies bind to lipid raft domains on the membrane, and because of this enrichment, they show increased antiviral potency. In particular, we find that cholesterol conjugation (i) rescues the antiviral activity of CDRH3-mutated 2F5, (ii) increases the antiviral activity of WT 2F5, (iii) potentiates the non-membrane-binding HIV antibody D5 10–100-fold (depending on the virus strain), and (iv) increases synergy between 2F5 and D5. Conjugation can be made at several positions, including variable and constant domains. Cholesterol conjugation therefore appears to be a general strategy to boost the potency of antiviral antibodies, and, because membrane affinity is engineered outside of the antibody paratope, it can complement affinity maturation strategies.
Collapse
Affiliation(s)
- Krzysztof Lacek
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Laboratory of Virus Molecular Biology, University of Gdansk, 80-822 Gdansk, Poland
| | - Richard A Urbanowicz
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Fulvia Troise
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| | - Claudia De Lorenzo
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Napoli (NA), Italy
| | - Valeria Severino
- the Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Via Vivaldi 43, 81100 Caserta (CE), Italy
| | - Antimo Di Maro
- the Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples, Via Vivaldi 43, 81100 Caserta (CE), Italy
| | - Alexander W Tarr
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Francesca Ferrara
- the Viral Pseudotype Unit, Infectious Diseases and Allergy group, School of Pharmacy, University of Kent, Kent ME4 4TB, United Kingdom
| | - Alexander Ploss
- the Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, and
| | - Nigel Temperton
- the Viral Pseudotype Unit, Infectious Diseases and Allergy group, School of Pharmacy, University of Kent, Kent ME4 4TB, United Kingdom
| | - Jonathan K Ball
- the School of Life Sciences and Nottingham Digestive Diseases Centre Biomedical Research Unit, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, United Kingdom
| | - Alfredo Nicosia
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, the Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini 5, 80131 Napoli (NA), Italy
| | - Riccardo Cortese
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| | - Antonello Pessi
- From Ceinge Biotecnologie Avanzate S.C.R.L., Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy, JV Bio, Via Gaetano Salvatore 486, 80145 Napoli (NA), Italy
| |
Collapse
|
34
|
Hui JZ, Al Zaki A, Cheng Z, Popik V, Zhang H, Luning Prak ET, Tsourkas A. Facile method for the site-specific, covalent attachment of full-length IgG onto nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:3354-63. [PMID: 24729432 PMCID: PMC4142076 DOI: 10.1002/smll.201303629] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/20/2014] [Indexed: 05/18/2023]
Abstract
Antibodies, most commonly IgGs, have been widely used as targeting ligands in research and therapeutic applications due to their wide array of targets, high specificity and proven efficacy. Many of these applications require antibodies to be conjugated onto surfaces (e.g. nanoparticles and microplates); however, most conventional bioconjugation techniques exhibit low crosslinking efficiencies, reduced functionality due to non-site-specific labeling and random surface orientation, and/or require protein engineering (e.g. cysteine handles), which can be technically challenging. To overcome these limitations, we have recombinantly expressed Protein Z, which binds the Fc region of IgG, with an UV active non-natural amino acid benzoylphenyalanine (BPA) within its binding domain. Upon exposure to long wavelength UV light, the BPA is activated and forms a covalent link between the Protein Z and the bound Fc region of IgG. This technology was combined with expressed protein ligation (EPL), which allowed for the introduction of a fluorophore and click chemistry-compatible azide group onto the C-terminus of Protein Z during the recombinant protein purification step. This enabled the crosslinked-Protein Z-IgG complexes to be efficiently and site-specifically attached to aza-dibenzocyclooctyne-modified nanoparticles, via copper-free click chemistry.
Collapse
Affiliation(s)
- James Zhe Hui
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Ajlan Al Zaki
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Zhiliang Cheng
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| | - Vladimir Popik
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Hongtao Zhang
- Department of Pathology and Lab Medicine, University of Pennsylvania, PA 19104, USA
| | - Eline T. Luning Prak
- Department of Pathology and Lab Medicine, University of Pennsylvania, PA 19104, USA
| | - Andrew Tsourkas
- Department of Bioengineering, University of Pennsylvania, 210 S. 33 Street, 240 Skirkanich Hall, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Massa S, Xavier C, De Vos J, Caveliers V, Lahoutte T, Muyldermans S, Devoogdt N. Site-specific labeling of cysteine-tagged camelid single-domain antibody-fragments for use in molecular imaging. Bioconjug Chem 2014; 25:979-88. [PMID: 24815083 DOI: 10.1021/bc500111t] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Site-specific labeling of molecular imaging probes allows the development of a homogeneous tracer population. The resulting batch-to-batch reproducible pharmacokinetic and pharmacodynamic properties are of great importance for clinical translation. Camelid single-domain antibody-fragments (sdAbs)-the recombinantly produced antigen-binding domains of heavy-chain antibodies, also called Nanobodies-are proficient probes for molecular imaging. To safeguard their intrinsically high binding specificity and affinity and to ensure the tracer's homogeneity, we developed a generic strategy for the site-specific labeling of sdAbs via a thio-ether bond. The unpaired cysteine was introduced at the carboxyl-terminal end of the sdAb to eliminate the risk of antigen binding interference. The spontaneous dimerization and capping of the unpaired cysteine required a reduction step prior to conjugation. This was optimized with the mild reducing agent 2-mercaptoethylamine in order to preserve the domain's stability. As a proof-of-concept the reduced probe was subsequently conjugated to maleimide-DTPA, for labeling with indium-111. A single conjugated tracer was obtained and confirmed via mass spectrometry. The specificity and affinity of the new sdAb-based imaging probe was validated in a mouse xenograft tumor model using a modified clinical lead compound targeting the human epidermal growth factor receptor 2 (HER2) cancer biomarker. These data provide a versatile and standardized strategy for the site-specific labeling of sdAbs. The conjugation to the unpaired cysteine results in the production of a homogeneous group of tracers and is a multimodal alternative to the technetium-99m labeling of sdAbs.
Collapse
Affiliation(s)
- Sam Massa
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel (VUB) , 1090 Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
36
|
Chari RVJ, Miller ML, Widdison WC. Antibody-drug conjugates: an emerging concept in cancer therapy. Angew Chem Int Ed Engl 2014; 53:3796-827. [PMID: 24677743 DOI: 10.1002/anie.201307628] [Citation(s) in RCA: 711] [Impact Index Per Article: 71.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Indexed: 01/17/2023]
Abstract
Traditional cancer chemotherapy is often accompanied by systemic toxicity to the patient. Monoclonal antibodies against antigens on cancer cells offer an alternative tumor-selective treatment approach. However, most monoclonal antibodies are not sufficiently potent to be therapeutically active on their own. Antibody-drug conjugates (ADCs) use antibodies to deliver a potent cytotoxic compound selectively to tumor cells, thus improving the therapeutic index of chemotherapeutic agents. The recent approval of two ADCs, brentuximab vedotin and ado-trastuzumab emtansine, for cancer treatment has spurred tremendous research interest in this field. This Review touches upon the early efforts in the field, and describes how the lessons learned from the first-generation ADCs have led to improvements in every aspect of this technology, i.e., the antibody, the cytotoxic compound, and the linker connecting them, leading to the current successes. The design of ADCs currently in clinical development, and results from mechanistic studies and preclinical and clinical evaluation are discussed. Emerging technologies that seek to further advance this exciting area of research are also discussed.
Collapse
Affiliation(s)
- Ravi V J Chari
- ImmunoGen, Inc. 830 Winter St, Waltham, MA 02451 (USA) http://www.immunogen.com.
| | | | | |
Collapse
|
37
|
Chari RVJ, Miller ML, Widdison WC. Antikörper-Wirkstoff-Konjugate: ein neues Konzept in der Krebstherapie. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201307628] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
38
|
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: 319] [Impact Index Per Article: 29.0] [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
|
39
|
Buchanan A, Clementel V, Woods R, Harn N, Bowen MA, Mo W, Popovic B, Bishop SM, Dall'Acqua W, Minter R, Jermutus L, Bedian V. Engineering a therapeutic IgG molecule to address cysteinylation, aggregation and enhance thermal stability and expression. MAbs 2013; 5:255-62. [PMID: 23412563 PMCID: PMC3893235 DOI: 10.4161/mabs.23392] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Antibodies can undergo a variety of covalent and non-covalent degradation reactions that have adverse effects on efficacy, safety, manufacture and storage. We had identified an antibody to Angiopoietin 2 (Ang2 mAb) that neutralizes Ang2 binding to its receptor in vitro and inhibits tumor growth in vivo. Despite favorable pharmacological activity, the Ang2 mAb preparations were heterogeneous, aggregated rapidly and were poorly expressed. Here, we report the engineering of the antibody variable and constant domains to generate an antibody with reduced propensity to aggregate, enhanced homogeneity, 11°C elevated T(m), 26-fold improved level of expression and retained activity. The engineered molecule, MEDI-3617, is now compatible with the large scale material supply required for clinical trials and is currently being evaluated in Phase 1 in cancer patients. This is the first report to describe the stability engineering of a therapeutic antibody addressing non canonical cysteine residues and the design strategy reported here is generally applicable to other therapeutic antibodies and proteins.
Collapse
|
40
|
Adair JR, Howard PW, Hartley JA, Williams DG, Chester KA. Antibody–drug conjugates – a perfect synergy. Expert Opin Biol Ther 2012; 12:1191-206. [DOI: 10.1517/14712598.2012.693473] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Rose RJ, Labrijn AF, van den Bremer ETJ, Loverix S, Lasters I, van Berkel PHC, van de Winkel JGJ, Schuurman J, Parren PWHI, Heck AJR. Quantitative analysis of the interaction strength and dynamics of human IgG4 half molecules by native mass spectrometry. Structure 2011; 19:1274-82. [PMID: 21893287 DOI: 10.1016/j.str.2011.06.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 05/18/2011] [Accepted: 06/13/2011] [Indexed: 01/20/2023]
Abstract
Native mass spectrometry (MS) is a powerful technique for studying noncovalent protein-protein interactions. Here, native MS was employed to examine the noncovalent interactions involved in homodimerization of antibody half molecules (HL) in hinge-deleted human IgG4 (IgG4Δhinge). By analyzing the concentration dependence of the relative distribution of monomer HL and dimer (HL)(2) species, the apparent dissociation constant (K(D)) for this interaction was determined. In combination with site-directed mutagenesis, the relative contributions of residues at the CH3-CH3 interface to this interaction could be characterized and corresponding K(D) values quantified over a range of 10(-10)-10(-4) M. The critical importance of this noncovalent interaction in maintaining the intact dimeric structure was also proven for the full-length IgG4 backbone. Using time-resolved MS, the kinetics of the interaction could be measured, reflecting the dynamics of IgG4 HL exchange. Hence, native MS has provided a quantitative view of local structural features that define biological properties of IgG4.
Collapse
Affiliation(s)
- Rebecca J Rose
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Challenges in developing bioanalytical assays for characterization of antibody–drug conjugates. Bioanalysis 2011; 3:677-700. [DOI: 10.4155/bio.11.30] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With more than 34 targets being investigated and nearly 20 clinical trials at various phases of development, antibody–drug conjugates (ADCs) hold a lot of promise for improving oncological malignancy therapy. This therapeutic strategy designed to specifically or preferentially deliver a cytotoxic agent to tumor cells through conjugation to a monoclonal antibody is not new. Although this approach is relatively simple conceptually, the history of ADCs clearly attests to the high degree of complexity in their development. Each component of an ADC is important to achieve efficacy with minimal toxicity, and the ability to monitor this multicomponent therapeutic entity is deemed to be critical for their successful optimization. In this article we review the different bioanalytical strategies that have been implemented to characterize various ADCs and discuss the challenges and issues associated with these approaches.
Collapse
|
43
|
Bioconjugation using mutant glycosyltransferases for the site-specific labeling of biomolecules with sugars carrying chemical handles. Methods Mol Biol 2011; 751:281-96. [PMID: 21674337 DOI: 10.1007/978-1-61779-151-2_17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This chapter presents a technique that employs mutant glycosyltransferase enzymes for the site-specific bioconjugation of biomolecules via a glycan moiety to facilitate the development of a targeted drug delivery system. The target specificity of this methodology is based on unique sugar residues that are present on glycoproteins or engineered glycopeptides. The glycosyltransferases used in this approach have been manipulated in a way that confers the ability to transfer a modified sugar residue with a chemical handle to a sugar moiety of the glycoprotein or to a polypeptide tag of an engineered nonglycoprotein. The availability of the modified sugar moiety thus makes it possible to link cargo molecules at specific sites. The cargo may be comprised of, for example, biotin or fluorescent tags for detection, imaging agents for magnetic resonance imaging (MRI), or cytotoxic drugs for cancer therapy.
Collapse
|
44
|
Voynov V, Chennamsetty N, Kayser V, Wallny HJ, Helk B, Trout BL. Design and application of antibody cysteine variants. Bioconjug Chem 2010; 21:385-92. [PMID: 20092294 DOI: 10.1021/bc900509s] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Antibodies are multidomain proteins that are extensively used as a research tool in molecular biology and as therapeutics in medicine. In many cases, antibodies are engineered to contain surface cysteines for the site-specific conjugation of payloads. These antibodies can serve as payload vehicles in targeting a diseased cell to which the conjugated molecules exercise their activity. Here, we design and analyze a set of fourteen new IgG1 cysteine variants, with at least one variant per immunoglobulin fold domain. The cross-linking propensity of these mutants correlates very well with a tool we have developed for measuring aggregation propensity in silico, called spatial aggregation propensity (SAP). Our results indicate the utility of the SAP technology in selecting antibody cysteine variants with desired properties. Moreover, the different oligomerization propensity of the variants suggests a variety of applications in molecular biology and medicine, such as payload delivery, structural analysis, electrophoresis, and chromatography.
Collapse
Affiliation(s)
- Vladimir Voynov
- Massachusetts Institute of Technology, Chemical Engineering, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
45
|
Hofer T, Skeffington LR, Chapman CM, Rader C. Molecularly defined antibody conjugation through a selenocysteine interface. Biochemistry 2010; 48:12047-57. [PMID: 19894757 DOI: 10.1021/bi901744t] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Antibody conjugates have broad utility in basic, preclinical, and clinical applications. Conventional antibody conjugation through the amine group of lysine or the thiol group of cysteine residues yields heterogeneous products of undefined stoichiometry and considerable batch-to-batch variability. To preserve the two hallmarks of the antibody molecule, precision and predictability, methods that enable site-specific antibody conjugation are in high demand. On the basis of a mammalian cell expression system, we describe the utilization of the 21st natural amino acid selenocysteine for the generation of IgG and Fab molecules with unique nucleophilic reactivity that affords site-specific conjugation to electrophilic derivatives of biotin, fluorescein, and poly(ethylene glycol). The resulting antibody conjugates were found to fully retain their antigen binding capability and, in the case of IgG, the ability to mediate effector functions. Gain of function was demonstrated in vitro and in vivo. While these antibody conjugates are relevant for a variety of proteomic, diagnostic, and therapeutic applications, they also constitute a proof of principle for the generation of molecularly defined antibody-drug conjugates and radioimmunoconjugates. Compared to other site-specific antibody conjugation methods, selenocysteine interface technology (i) only involves a minor modification at the C-terminus that does not interfere with disulfide bridges, (ii) does not require activation, and (iii) generates unique 1:1 stoichiometries of biological and chemical components. Collectively, our method affords the generation of highly defined antibody conjugates with broad utility from proteomic applications to therapeutic intervention.
Collapse
Affiliation(s)
- Thomas Hofer
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1203, USA
| | | | | | | |
Collapse
|
46
|
Chen XN, Nguyen M, Jacobson F, Ouyang J. Charge-based analysis of antibodies with engineered cysteines: from multiple peaks to a single main peak. MAbs 2009; 1:563-71. [PMID: 20068389 DOI: 10.4161/mabs.1.6.10058] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
THIOMABs are antibodies with an engineered unpaired cysteine residue on each heavy chain that can be used as intermediates to generate antibody-drug conjugates. Multiple charge variant peaks were observed during cation-exchange chromatography (CEX) and imaged capillary isoelectric focusing (cIEF) analysis of several different THIOMABs. This charge heterogeneity was due to cysteinylation and/or glutathionylation at the engineered and unpaired cysteines through disulfide bonds formed during the cell culture process. Cysteine treatment followed by analysis using CEX, LC/MS and electrophoresis demonstrates that cysteine is a mild reductant that can remove glutathione and cysteine bound to the engineered cysteines without disrupting the inter- or intra-chain disulfide bonds of antibodies. We further demonstrated that using a cysteine/cystine redox pair (rather than cysteine alone) can not only effectively remove glutathione at the engineered cysteines, but also generate homogeneously cysteinylated species, which resulted in one main peak in both CEX-HPLC and imaged cIEF assays for antibodies with engineered and unpaired cysteines.
Collapse
Affiliation(s)
- Xiaoying Nancy Chen
- Protein Analytical Chemistry, Genentech Inc., 1 DNA Way, South San Francisco, CA, USA.
| | | | | | | |
Collapse
|
47
|
Ramakrishnan B, Boeggeman E, Manzoni M, Zhu Z, Loomis K, Puri A, Dimitrov DS, Qasba PK. Multiple site-specific in vitro labeling of single-chain antibody. Bioconjug Chem 2009; 20:1383-9. [PMID: 19507852 PMCID: PMC3402211 DOI: 10.1021/bc900149r] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
For multiple site-specific conjugations of bioactive molecules to a single-chain antibody (scFv) molecule, we have constructed a human anti HER2 receptor, scFv, with a C-terminal fusion polypeptide containing 1, 3, or 17 threonine (Thr) residues. The C-terminal extended fusion polypeptides of these recombinant scFv fusion proteins are used as the acceptor substrate for human polypeptide-alpha-Nu-acetylgalactosaminyltransferase II (h-ppGalNAc-T2) that transfers either GalNAc or 2-keto-Gal, a modified galactose with a chemical handle, from their respective UDP-sugars to the side-chain hydroxyl group of the Thr residue(s). The recombinant scFv fusion proteins are expressed in E. coli as inclusion bodies and in vitro refolded and glycosylated with h-ppGalNAc-T2. Upon protease cleavage, the MALDI-TOF spectra of the glycosylated C-terminal fusion polypeptides showed that the glycosylated scFv fusion protein with a single Thr residue is fully glycosylated with a single 2-keto-Gal, whereas the glycosylated scFv fusion protein with 3 and 17 Thr residues is found as an equal mixture of 2-3 and 5-8 2-keto-Gal glycosylated fusion proteins, respectively. These fusion scFv proteins with the modified galactose are then conjugated with a fluorescence probe, Alexa488, that carries an orthogonal reactive group. The fluorescence labeled scFv proteins bind specifically to a human breast cancer cell line (SK-BR-3) that overexpresses the HER2 receptor, indicating that the in vitro folded scFv fusion proteins are biologically active and the presence of conjugated multiple Alexa488 probes in their C-terminal end does not interfere with their binding to the antigen.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pradman K. Qasba
- Corresponding author. Structural Glycobiology Section, CCRNP, CCR, NCI-Frederick, Building 469, Room 221, Frederick, Maryland 21702; . Phone: (301) 846-1934; Fax: (301) 846-7149
| |
Collapse
|
48
|
Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic index. Nat Biotechnol 2008; 26:925-32. [DOI: 10.1038/nbt.1480] [Citation(s) in RCA: 946] [Impact Index Per Article: 59.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 06/19/2008] [Indexed: 11/09/2022]
|
49
|
|
50
|
Miao Z, McCoy MR, Singh DD, Barrios B, Hsu OL, Cheal SM, Meares CF. Cysteinylated protein as reactive disulfide: an alternative route to affinity labeling. Bioconjug Chem 2007; 19:15-9. [PMID: 18062660 DOI: 10.1021/bc700330j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Engineering the permanent formation of a receptor-ligand complex has a number of promising applications in chemistry, biology, and medicine. Antibodies and other proteins can be excellent receptors for synthetic ligands such as probes or drugs. Because proteins possess an array of nucleophilic sites, the placement of an electrophile on the synthetic ligand to react with a nucleophile on the macromolecule is a standard practice. Previously, we have used the site-directed incorporation of cysteine nucleophiles at the periphery of an antibody's binding site, paired with the chemical design of weakly electrophilic ligands, to produce receptor-ligand pairs that conjugate specifically and permanently (Corneillie et al. (2004) Bioconjugate Chem. 15, 1392-1402 and references therein). After protein expression in Drosophila S2 cells, we found, as is frequently observed, that the engineered cysteine was reversibly blocked by disulfide linkage to a cysteine monomer (cysteinylated). Removal of the cysteine monomer requires some care because of the need to preserve other disulfide linkages in the protein. Here, we report that cysteinylation can be used to advantage by treating the cysteine monomer as a leaving group and the protein disulfide as an electrophile with special affinity for thiols. Two ligands bearing thiol side chains were synthesized and incubated with the cysteinylated antibody Fab fragment 2D12.5 G54C, with the finding that both ligands become covalently attached within a few minutes under physiological conditions. The attachment is robust even in the presence of excess thiol reagents. This rapid, specific conjugation is particularly interesting for biomedical applications.
Collapse
Affiliation(s)
- Zheng Miao
- Department of Chemistry, University of California, Davis, California 95616, USA
| | | | | | | | | | | | | |
Collapse
|