1
|
Song S, Liu Y, Liu J, Tai W. In Silico-Driven THIOMAB Approach for Stable PROTAC Conjugates by Docking Payloads in Antibody Cavities. Bioconjug Chem 2025; 36:960-970. [PMID: 40196986 DOI: 10.1021/acs.bioconjchem.4c00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The heterobifunctional proteolysis targeting chimeras (PROTACs) are a class of emerging therapeutic modalities that enable selective degradation of target proteins in cells. As antibody payloads, they offer several advantages compared to conventional chemical toxins, such as catalytic nature, potent and long-lasting activity, and precise selectivity to avoid systemic toxicity. However, the relatively large size and high hydrophobicity of these chimeric payloads may result in challenging the stability of antibodies, which complicates the in vivo performance. In this work, we use the highly hydrophobic GNE-987 as model PROTAC to evaluate a THIOMAB approach for mitigating the conjugate's hydrophobicity while maintaining the therapeutic potency. We describe an in silico method to select the less hydrophobic site in an antibody and employ the stable tetrapeptide-aminomethoxy linker to conjugate the PROTAC payloads. The resulting degrader-antibody conjugate (J591 DAC) displays antigen-dependent BRD4 degradation and potent cytotoxic activity in PSMA-positive cancer cells. Finally, this DAC, bearing two highly hydrophobic PROTACs, also exhibits a long blood retention and strong antitumor efficacy in mouse models, likely owing to the homogeneous and stable conjugation from the THIOMAB approach. This work provides an example of the design and construction of antibody conjugates with highly hydrophobic payloads.
Collapse
Affiliation(s)
- Shiwei Song
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Yahui Liu
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiaqi Liu
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
2
|
Le Stum M, Romero E, Molander GA. Photocatalyzed elaboration of antibody-based bioconjugates. Beilstein J Org Chem 2025; 21:616-629. [PMID: 40130177 PMCID: PMC11931643 DOI: 10.3762/bjoc.21.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent a promising class of targeted therapeutics, combining the specificity of antibodies with the potency of cytotoxic drugs to enhance therapeutic efficacy while minimizing off-target effects. The development of new chemical methods for bioconjugation is essential to generate ADCs and to optimize their stability, efficacy, and safety. Traditional conjugation methods often face challenges related to site-selectivity and heterogeneous product mixtures, highlighting the need to develop new, innovative chemical strategies. Photoredox chemistry emerges as a powerful tool in this context, enabling precise, mild, and selective modifications of peptides and proteins. By harnessing light to drive chemical transformations, photoredox techniques can facilitate the synthesis of antibody bioconjugates. This perspective will discuss the drive to develop and empower photoredox methods applied to antibody functionalization.
Collapse
Affiliation(s)
- Marine Le Stum
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Eugénie Romero
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| | - Gary A Molander
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SCBM, 91191 Gif-Sur-Yvette, France
| |
Collapse
|
3
|
Yang Y, Patel JM, Yang RS, Ma F, Niu X, Zhang Y, Niedringhaus T, Al-Sayah M, Yang X. Determination of the decapping efficiency of THIOMAB™ antibodies with the engineered cysteine in the Fc region for making antibody-drug conjugates by specific hinge fragmentation-liquid chromatography. Anal Bioanal Chem 2025; 417:847-859. [PMID: 39688670 DOI: 10.1007/s00216-024-05707-w] [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: 09/24/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The site-specific antibody-drug conjugates (ADCs), particularly those utilizing the engineered cysteine in Fc fragments of mAbs (THIOMAB™ antibodies), have emerged as a novel class of biotherapeutics for cancer treatment. The engineered cysteine residues in these antibodies are capped by cysteine or glutathione through a disulfide bond. Prior to conjugation with linker-payloads, these caps need to be removed through a reduction process. However, monitoring the efficiency of the decapping process has been challenging due to the lack of effective analytical methods. Intact reversed-phase liquid chromatography-mass spectrometry and hydrophobic interaction chromatography methods failed to separate decapped and capped intact THIOMAB™ mAbs in our study. Instead the fragmentation of mAbs provided a novel strategy to analyze the decapping effiency. After cleavage using a hinge specific enzyme, the generated Fc fragments with and without cysteine and/or glutathione caps displayed different hydrophobicity and were well separated by RPLC, allowing quantitative determination of the decapping efficiency. Enzymes that cleave both above and below the hinge disulfide bonds were tested. The use of FabALATICA can determine percentages of molecules with 0, 1, and 2 cysteine and/or glutathione caps, respectively, regardless of whether the antibody contains the hinge LALA mutations. On the other hand, FabRICATOR enzyme can only be utilized for antibodies without LALA mutations for the overall decapping percentage and cannot be used to estimate intact antibody each with 0, 1, and 2 caps. Therefore, FabALACTICA cleavage followed by RPLC provides a wider application of monitoring the decapping efficiency of all antibodies with the engineered cysteine in Fc.
Collapse
Affiliation(s)
- Yun Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Jaymin M Patel
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Rong-Sheng Yang
- Analytical Research and Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Fengfei Ma
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiangfeng Niu
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Yixiao Zhang
- Bioprocess Research & Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Thomas Niedringhaus
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Mohammad Al-Sayah
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiaoyu Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA.
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA.
| |
Collapse
|
4
|
Jiang X, Nik Nabil WN, Ze Y, Dai R, Xi Z, Xu H. Unlocking Natural Potential: Antibody-Drug Conjugates With Naturally Derived Payloads for Cancer Therapy. Phytother Res 2025; 39:789-874. [PMID: 39688127 DOI: 10.1002/ptr.8407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Natural compound-derived chemotherapies remain central to cancer treatment, however, they often cause off-target side effects that negatively impact patients' quality of life. In contrast, antibody-drug conjugates (ADCs) combine cytotoxic payloads with antibodies to specifically target cancer cells. Most approved and clinically investigated ADCs utilize naturally derived payloads, while those with conventional synthetic molecular payloads remain limited. This review focuses on approved ADCs that enhance the efficacy of naturally derived payloads by linking them with antibodies. We provide an overview of the core components of ADCs, their working mechanisms, and FDA-approved ADCs featuring naturally derived payloads, such as calicheamicin, camptothecin, dolastatin 10, maytansine, pyrrolbenzodiazepine (PBD), and the immunotoxin Pseudomonas exotoxin A. This review also explores recent clinical advancements aimed at broadening the therapeutic potential of ADCs, their applicability in treating heterogeneously composed tumors and their potential use beyond oncology. Additionally, this review highlights naturally derived payloads that are currently being clinically investigated but have not yet received approval. By summarizing the current landscape, this review provides insights into promising avenues for exploration and contributes to the refinement of treatment protocols for improved patient outcomes.
Collapse
Affiliation(s)
- Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Pharmaceutical Regulatory Agency, Ministry of Health, Selangor, Malaysia
| | - Yufei Ze
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Bitsch P, Dessin C, Bitsch S, Voss J, Becker J, Sharma P, Biyani N, Kochat H, Sewald N, Kolmar H. Evaluation of Potency and Specificity of Cryptophycin-Loaded Antibody-Drug Conjugates. Chembiochem 2025; 26:e202400738. [PMID: 39462215 DOI: 10.1002/cbic.202400738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
An enhanced variant of the antimitotic toxin cryptophycin was conjugated to the anti-Her2 monoclonal antibody (mAb) Trastuzumab upon Michael addition. Either antibodies with freed hinge-region cysteines or THIOMAB formats with engineered cysteines in the mAbs light chain were added to a maleimide derivative of cryptophycin. These Antibody-Drug Conjugates (ADCs) showed retained binding to Her2 positive tumor cells and highly efficient cell killing in double-digit pM range on high Her2-expressing SK-BR-3 cells. Two ADCs (DAR 6, DAR 3) showed superior cell killing of the cell lines JIMT-1 and RT112 with medium receptor expression level in comparison with a DAR 6 MMAE ADC serving as reference. The observed cell cytotoxicity is target-dependent since no impact on cell viability was observed for low Her2-expressing MDA-MB468 cells. Particularly the DAR 3 ADC in THIOMAB format exhibiting desirable biophysical properties and high potency emerged as a promising candidate for further in vivo investigations.
Collapse
Affiliation(s)
- Peter Bitsch
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Cedric Dessin
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Sebastian Bitsch
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Jona Voss
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Janine Becker
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| | - Panna Sharma
- Lantern Pharma Inc., 1920 McKinney Ave, 7th Floor, Dallas 75201, TX, USA
| | - Neha Biyani
- Lantern Pharma Inc., 1920 McKinney Ave, 7th Floor, Dallas 75201, TX, USA
| | - Harry Kochat
- The University of Tennessee, Health Science Center, 208 S., Dudley Street, Memphis, 38163, TN, USA
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, University of Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Deutschland
| | - Harald Kolmar
- Clemens-Schöpf Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
- Centre of Synthetic Biology, Technical University of Darmstadt, Peter-Grünberg-Str. 4, 64287, Darmstadt, Germany
| |
Collapse
|
6
|
Chen Y, Clay N, Phan N, Lothrop E, Culkins C, Robinson B, Stubblefield A, Ferguson A, Kimmel BR. Molecular Matchmakers: Bioconjugation Techniques Enhance Prodrug Potency for Immunotherapy. Mol Pharm 2025; 22:58-80. [PMID: 39570179 DOI: 10.1021/acs.molpharmaceut.4c00867] [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] [Indexed: 11/22/2024]
Abstract
Cancer patients suffer greatly from the severe off-target side effects of small molecule drugs, chemotherapy, and radiotherapy─therapies that offer little protection following remission. Engineered immunotherapies─including cytokines, immune checkpoint blockade, monoclonal antibodies, and CAR-T cells─provide better targeting and future tumor growth prevention. Still, issues such as ineffective activation, immunogenicity, and off-target effects remain primary concerns. "Prodrug" therapies─classified as therapies administered as inactive and then selectively activated to control the time and area of release─hold significant promise in overcoming these concerns. Bioconjugation techniques (e.g., natural linker conjugation, bioorthogonal reactions, and noncanonical amino acid incorporation) enable the rapid and homogeneous synthesis of prodrugs and offer selective loading of immunotherapeutic agents to carrier molecules and protecting groups to prevent off-target effects after administration. Several prodrug activation mechanisms have been highlighted for cancer therapeutics, including endogenous activation by hypoxic or acidic conditions common in tumors, exogenous activation by targeted bioorthogonal cleavage, or stimuli-responsive light activation, and dual-stimuli activation, which adds specificity by combining these mechanisms. This review will explore modern prodrug conjugation and activation options, focusing on how these strategies can enhance immunotherapy responses and improve patient outcomes. We will also discuss the implications of computational methodology for therapy design and recommend procedures to determine how and where to conjugate carrier systems and "prodrug" groups onto therapeutic agents to enhance the safety and control of these delivery platforms.
Collapse
Affiliation(s)
- Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Natalie Clay
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ariana Stubblefield
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Alani Ferguson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
7
|
Vasco A, Taylor RJ, Méndez Y, Bernardes GJL. On-Demand Thio-Succinimide Hydrolysis for the Assembly of Stable Protein-Protein Conjugates. J Am Chem Soc 2024; 146:20709-20719. [PMID: 39012647 PMCID: PMC11295205 DOI: 10.1021/jacs.4c03721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/17/2024]
Abstract
Chemical post-translational protein-protein conjugation is an important technique with growing applications in biotechnology and pharmaceutical research. Maleimides represent one of the most widely employed bioconjugation reagents. However, challenges associated with the instability of first- and second-generation maleimide technologies are yet to be fully addressed. We report the development of a novel class of maleimide reagents that can undergo on-demand ring-opening hydrolysis of the resulting thio-succinimide. This strategy enables rapid post-translational assembly of protein-protein conjugates. Thio-succinimide hydrolysis, triggered upon application of chemical, photochemical, or enzymatic stimuli, allowed homobifunctional bis-maleimide reagents to be applied in the production of stable protein-protein conjugates, with complete temporal control. Bivalent and bispecific protein-protein dimers constructed from small binders targeting antigens of oncological importance, PD-L1 and HER2, were generated with high purity, stability, and improved functionality compared to monomeric building blocks. The modularity of the approach was demonstrated through elaboration of the linker moiety through a bioorthogonal propargyl handle to produce protein-protein-fluorophore conjugates. Furthermore, extending the functionality of the homobifunctional reagents by temporarily masking reactive thiols included in the linker allowed the assembly of higher order trimeric and tetrameric single-domain antibody conjugates. The potential for the approach to be extended to proteins of greater biochemical complexity was demonstrated in the production of immunoglobulin single-domain antibody conjugates. On-demand control of thio-succinimide hydrolysis combined with the facile assembly of chemically defined homo- and heterodimers constitutes an important expansion of the chemical methods available for generating stable protein-protein conjugates.
Collapse
Affiliation(s)
| | | | - Yanira Méndez
- Yusuf Hamied Department of
Chemistry, University of Cambridge, CB2 1EW Cambridge, U.K.
| | | |
Collapse
|
8
|
Huang Z, Braunstein Z, Chen J, Wei Y, Rao X, Dong L, Zhong J. Precision Medicine in Rheumatic Diseases: Unlocking the Potential of Antibody-Drug Conjugates. Pharmacol Rev 2024; 76:579-598. [PMID: 38622001 DOI: 10.1124/pharmrev.123.001084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/25/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
In the era of precision medicine, antibody-drug conjugates (ADCs) have emerged as a cutting-edge therapeutic strategy. These innovative compounds combine the precision of monoclonal antibodies with the potent cell-killing or immune-modulating abilities of attached drug payloads. This unique strategy not only reduces off-target toxicity but also enhances the therapeutic effectiveness of drugs. Beyond their well established role in oncology, ADCs are now showing promising potential in addressing the unmet needs in the therapeutics of rheumatic diseases. Rheumatic diseases, a diverse group of chronic autoimmune diseases with varying etiologies, clinical presentations, and prognoses, often demand prolonged pharmacological interventions, creating a pressing need for novel, efficient, and low-risk treatment options. ADCs, with their ability to precisely target the immune components, have emerged as a novel therapeutic strategy in this context. This review will provide an overview of the core components and mechanisms behind ADCs, a summary of the latest clinical trials of ADCs for the treatment of rheumatic diseases, and a discussion of the challenges and future prospects faced by the development of next-generation ADCs. SIGNIFICANCE STATEMENT: There is a lack of efficient and low-risk targeted therapeutics for rheumatic diseases. Antibody-drug conjugates, a class of cutting-edge therapeutic drugs, have emerged as a promising targeted therapeutic strategy for rheumatic disease. Although there is limited literature summarizing the progress of antibody-drug conjugates in the field of rheumatic disease, updating the advancements in this area provides novel insights into the development of novel antirheumatic drugs.
Collapse
Affiliation(s)
- Zhiwen Huang
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Zachary Braunstein
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jun Chen
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Yingying Wei
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Xiaoquan Rao
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Lingli Dong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| | - Jixin Zhong
- Departments of Rheumatology and Immunology (Z.H., Y.W., L.D., J.Z.) and Cardiology (X.R.), Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Wexner Medical Center, The Ohio State University, Columbus, Ohio (Z.B.); Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, China (J.C.); Key Laboratory of Vascular Aging (HUST), Ministry of Education, Wuhan, China (J.Z.); and Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (J.Z.)
| |
Collapse
|
9
|
Tanriver M, Müller M, Levasseur MD, Richards D, Majima S, DeMello A, Yamauchi Y, Bode JW. Peptide-Directed Attachment of Hydroxylamines to Specific Lysines of IgG Antibodies for Bioconjugations with Acylboronates. Angew Chem Int Ed Engl 2024; 63:e202401080. [PMID: 38421342 DOI: 10.1002/anie.202401080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
The role of monoclonal antibodies as vehicles to deliver payloads has evolved as a powerful tool in cancer therapy in recent years. The clinical development of therapeutic antibody conjugates with precise payloads holds great promise for targeted therapeutic interventions. The use of affinity-peptide mediated functionalization of native off-the-shelf antibodies offers an effective approach to selectively modify IgG antibodies with a drug-antibody ratio (DAR) of 2. Here, we report the traceless, peptide-directed attachment of two hydroxylamines to native IgGs followed by chemoselective potassium acyltrifluoroborate (KAT) ligation with quinolinium acyltrifluoroborates (QATs), which provide enhanced ligation rates with hydroxylamines under physiological conditions. By applying KAT ligation to the modified antibodies, conjugation of small molecules, proteins, and oligonucleotides to off-the-shelf IgGs proceeds efficiently, in good yields, and with simultaneous cleavage of the affinity peptide-directing moiety.
Collapse
Affiliation(s)
- Matthias Tanriver
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Marco Müller
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Mikail D Levasseur
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Daniel Richards
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Sohei Majima
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Andrew DeMello
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Yohei Yamauchi
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Jeffrey W Bode
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| |
Collapse
|
10
|
Anderson TS, McCormick AL, Smith SL, Lowe DB. Modeling antibody drug conjugate potential using a granzyme B antibody fusion protein. BMC Biol 2024; 22:66. [PMID: 38486229 PMCID: PMC10941411 DOI: 10.1186/s12915-024-01860-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) constitute a promising class of targeted anti-tumor therapeutics that harness the selectivity of monoclonal antibodies with the potency of cytotoxic drugs. ADC development is best suited to initially screening antibody candidates for desired properties that potentiate target cell cytotoxicity. However, validating and producing an optimally designed ADC requires expertise and resources not readily available to certain laboratories. RESULTS In this study, we propose a novel approach to help streamline the identification of potential ADC candidates by utilizing a granzyme B (GrB)-based antibody fusion protein (AFP) for preliminary screening. GrB is a non-immunogenic serine protease expressed by immune effector cells such as CD8 + T cells that induces apoptotic activity and can be leveraged for targeted cell killing. CONCLUSIONS Our innovative model allows critical antibody parameters (including target cell binding, internalization, and cytotoxic potential) to be more reliably evaluated in vitro through the creation of an ADC surrogate. Successful incorporation of this AFP could also significantly expand and enhance ADC development pre-clinically, ultimately leading to the accelerated translation of ADC therapies for patients.
Collapse
Affiliation(s)
- Trevor S Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Amanda L McCormick
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Savanna L Smith
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA
| | - Devin B Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1718 Pine Street, Office 1306, Abilene, TX, 79601, USA.
| |
Collapse
|
11
|
Liao X, Haight A, Welch D, Han L. Selective Reduction of Cysteine Mutant Antibodies for Site-Specific Antibody-Drug Conjugates. Bioconjug Chem 2023; 34:2293-2301. [PMID: 37983167 DOI: 10.1021/acs.bioconjchem.3c00425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Developing site-specific conjugation technologies for antibody-drug conjugates (ADCs) aims to produce more homogeneous and controlled drug-loaded ADCs to reduce variability and thereby improve the therapeutic index. This article presents a technology that uses cysteine mutant antibodies and mild phosphine-based reductants to prepare site-specific ADCs. The two types of cysteine mutant antibodies, designated C6v1 and C6v2, have one of the interchain disulfide-forming cysteines in the Fab region in the light chain (LC214) or in the heavy chain (HC220) substituted by alanine (or other amino acids), respectively. Certain phosphine-based reductants were found to selectively reduce the "unpaired" cysteines, at the heavy chain (HC220) for C6v1 or at the light chain (LC214) for C6v2 while keeping the interchain disulfide bonds in the hinge region intact, resulting in 90% of DAR2 species and more than 95% of the desired specific conjugation at HC or LC following conjugation to maleimide moieties. The reduction method shows consistent selectivity toward various C6v1 or C6v2 antibody backbones. Sensitivity toward buffer pH for some reductants can be used to optimize reductant reactivity and selectivity. The technology can be further expanded to generate site-specific DAR4 or dual-payload ADCs based on C6v1 or C6v2 antibodies. This technology offers a method to control drug-loading and conjugation sites using a mild one-pot process, as compared to the reduction-oxidation methods used in technologies such as THIOMAB, and shows superior DAR profiles and process simplification as compared to other selective reduction methods.
Collapse
Affiliation(s)
- Xiaoli Liao
- Process Chemistry, Small Molecule CMC Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Anthony Haight
- Process Chemistry, Small Molecule CMC Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Dennie Welch
- Process Chemistry, Small Molecule CMC Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Linjie Han
- Analytical Development, Operations S&T, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| |
Collapse
|
12
|
Rakotoarinoro N, Dyck YFK, Krebs SK, Assi MK, Parr MK, Stech M. A disruptive clickable antibody design for the generation of antibody-drug conjugates. Antib Ther 2023; 6:298-310. [PMID: 38107665 PMCID: PMC10720948 DOI: 10.1093/abt/tbad023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/05/2023] [Accepted: 10/14/2023] [Indexed: 12/19/2023] Open
Abstract
Background Antibody-drug conjugates are cancer therapeutics that combine specificity and toxicity. A highly cytotoxic drug is covalently attached to an antibody that directs it to cancer cells. The conjugation of the drug-linker to the antibody is a key point in research and development as well as in industrial production. The consensus is to conjugate the drug to a surface-exposed part of the antibody to ensure maximum conjugation efficiency. However, the hydrophobic nature of the majority of drugs used in antibody-drug conjugates leads to an increased hydrophobicity of the generated antibody-drug conjugates, resulting in higher liver clearance and decreased stability. Methods In contrast, we describe a non-conventional approach in which the drug is conjugated in a buried part of the antibody. To achieve this, a ready-to-click antibody design was created in which an azido-based non-canonical amino acid is introduced within the Fab cavity during antibody synthesis using nonsense suppression technology. The Fab cavity was preferred over the Fc cavity to circumvent issues related to cleavage of the IgG1 lower hinge region in the tumor microenvironment. Results This antibody design significantly increased the hydrophilicity of the generated antibody-drug conjugates compared to the current best-in-class designs based on non-canonical amino acids, while conjugation efficiency and functionality were maintained. The robustness of this native shielding effect and the versatility of this approach were also investigated. Conclusions This pioneer design may become a starting point for the improvement of antibody-drug conjugates and an option to consider for protecting drugs and linkers from unspecific interactions.
Collapse
Affiliation(s)
- Nathanaël Rakotoarinoro
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Yan F K Dyck
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Simon K Krebs
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Miriam-Kousso Assi
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
- Department of Biotechnology, Hamburg University of Applied Sciences, 21033 Hamburg, Germany
| | - Maria K Parr
- Institute of Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marlitt Stech
- Institute for Cell Therapy and Immunology branch Bioanalytics and Bioprocesses, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V., 14476 Potsdam-Golm, Germany
| |
Collapse
|
13
|
Tarantino P, Ricciuti B, Pradhan SM, Tolaney SM. Optimizing the safety of antibody-drug conjugates for patients with solid tumours. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00783-w. [PMID: 37296177 DOI: 10.1038/s41571-023-00783-w] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Over the past 5 years, improvements in the design of antibody-drug conjugates (ADCs) have enabled major advances that have reshaped the treatment of several advanced-stage solid tumours. Considering the intended rationale behind the design of ADCs, which is to achieve targeted delivery of cytotoxic molecules by linking them to antibodies targeting tumour-specific antigens, ADCs would be expected to be less toxic than conventional chemotherapy. However, most ADCs are still burdened by off-target toxicities that resemble those of the cytotoxic payload as well as on-target toxicities and other poorly understood and potentially life-threatening adverse effects. Given the rapid expansion in the clinical indications of ADCs, including use in curative settings and various combinations, extensive efforts are ongoing to improve their safety. Approaches currently being pursued include clinical trials optimizing the dose and treatment schedule, modifications of each ADC component, identification of predictive biomarkers for toxicities, and the development of innovative diagnostic tools. In this Review, we describe the determinants of the toxicities of ADCs in patients with solid tumours, highlighting key strategies that are expected to improve tolerability and enable improvements in the treatment outcomes of patients with advanced-stage and those with early stage cancers in the years to come.
Collapse
Affiliation(s)
- Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Biagio Ricciuti
- Harvard Medical School, Boston, MA, USA
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shan M Pradhan
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Schiemer R, Weggen JT, Schmitt KM, Hubbuch J. An adaptive soft-sensor for advanced real-time monitoring of an antibody-drug conjugation reaction. Biotechnol Bioeng 2023. [PMID: 37190793 DOI: 10.1002/bit.28428] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/31/2023] [Accepted: 05/01/2023] [Indexed: 05/17/2023]
Abstract
In the production of antibody-drug conjugates (ADCs), the conjugation reaction is a central step defining the final product composition and, hence, directly affecting product safety and efficacy. To enable real-time monitoring, spectroscopic sensors in combination with multivariate regression models have gained popularity in recent years. The extended Kalman filter (EKF) can be used as so-called soft-sensor to fuse sensor predictions with long-horizon forecasts by process models. This enables the dynamic update of the current state and provides increased robustness against experimental noise or model errors. Due to the uncertainty associated with sensor and process models in biopharmaceutical applications, the deployment of such soft-sensors is challenging. In this study, we demonstrate the combination of an uncertainty-aware sensor model with a kinetic reaction model using an EKF to monitor a site-directed ADC conjugation reaction. As the sensor model, a Gaussian process regression model is presented to realize a time-variant determination of the sensor uncertainty. The EKF fuses the time-discrete predictions of the amount of conjugated drug from the sensor model with the time-continuous predictions from the kinetic model. While the ADC species are not distinguishable by on-line recorded UV/Vis spectra, the developed soft-sensor is able to dynamically update all relevant reaction species. It could be shown that the use of time-variant process and sensor noise computation approaches improved the performance of the EKF and achieved a reduction of the prediction error of up to 23% compared with the kinetic model. The developed framework proved to enhance robustness against noisy sensor measurements or wrong model initialization and was successfully transferred from batch to fed-batch mode. In future, this framework could be implemented for model-based process control and be adopted for other ADC conjugation reaction types.
Collapse
Affiliation(s)
- Robin Schiemer
- Institute of Process Engineering in Life Sciences-Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Baden-Württemberg, Germany
| | - Jan Tobias Weggen
- Institute of Process Engineering in Life Sciences-Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Baden-Württemberg, Germany
| | - Katrin Marianne Schmitt
- Institute of Process Engineering in Life Sciences-Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Baden-Württemberg, Germany
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences-Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Baden-Württemberg, Germany
| |
Collapse
|
15
|
Weggen JT, Seidel J, Bean R, Wendeler M, Hubbuch J. Kinetic studies and CFD-based reaction modeling for insights into the scalability of ADC conjugation reactions. Front Bioeng Biotechnol 2023; 11:1123842. [PMID: 37082211 PMCID: PMC10111256 DOI: 10.3389/fbioe.2023.1123842] [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: 12/14/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
The manufacturing of antibody-drug conjugates (ADCs) involves the addition of a cytotoxic small-molecule linker-drug (= payload) to a solution of functionalized antibodies. For the development of robust conjugation processes, initially small-scale reaction tubes are used which requires a lot of manual handling. Scale-up to larger reaction vessels is often knowledge-driven and scale-comparability is solely assessed based on final product quality which does not account for the dynamics of the reaction. In addition, information about the influence of process parameters, such as stirrer speed, temperature, or payload addition rates, is limited due to high material costs. Given these limitations, there is a need for a modeling-based approach to investigate conjugation scale-up. In this work, both experimental kinetic studies and computational fluid dynamics (CFD) conjugation simulations were performed to understand the influence of scale and mixing parameters. In the experimental part, conjugation kinetics in small-scale reaction tubes with different mixing types were investigated for two ADC systems and compared to larger bench-scale reactions. It was demonstrated that more robust kinetics can be achieved through internal stirrer mixing instead of external mixing devices, such as orbital shakers. In the simulation part, 3D-reactor models were created by coupling CFD-models for three large-scale reaction vessels with a kinetic model for a site-specific conjugation reaction. This enabled to study the kinetics in different vessels, as well as the effect of process parameter variations in silico. Overall, it was found that for this conjugation type sufficient mixing can be achieved at all scales and the studied parameters cause only deviations during the payload addition period. An additional time-scale analysis demonstrated to aid the assessment of mixing effects during ADC process scale-up when mixing times and kinetic rates are known. In summary, this work highlights the benefit of kinetic models for enhanced conjugation process understanding without the need for large-scale experiments.
Collapse
Affiliation(s)
- Jan Tobias Weggen
- Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Janik Seidel
- Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Ryan Bean
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Michaela Wendeler
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| |
Collapse
|
16
|
André AS, Dias JNR, Aguiar S, Nogueira S, Bule P, Carvalho JI, António JPM, Cavaco M, Neves V, Oliveira S, Vicente G, Carrapiço B, Braz BS, Rütgen B, Gano L, Correia JDG, Castanho M, Goncalves J, Gois PMP, Gil S, Tavares L, Aires-da-Silva F. Rabbit derived VL single-domains as promising scaffolds to generate antibody-drug conjugates. Sci Rep 2023; 13:4837. [PMID: 36964198 PMCID: PMC10038998 DOI: 10.1038/s41598-023-31568-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/14/2023] [Indexed: 03/26/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are among the fastest-growing classes of therapeutics in oncology. Although ADCs are in the spotlight, they still present significant engineering challenges. Therefore, there is an urgent need to develop more stable and effective ADCs. Most rabbit light chains have an extra disulfide bridge, that links the variable and constant domains, between Cys80 and Cys171, which is not found in the human or mouse. Thus, to develop a new generation of ADCs, we explored the potential of rabbit-derived VL-single-domain antibody scaffolds (sdAbs) to selectively conjugate a payload to Cys80. Hence, a rabbit sdAb library directed towards canine non-Hodgkin lymphoma (cNHL) was subjected to in vitro and in vivo phage display. This allowed the identification of several highly specific VL-sdAbs, including C5, which specifically target cNHL cells in vitro and present promising in vivo tumor uptake. C5 was selected for SN-38 site-selective payload conjugation through its exposed free Cys80 to generate a stable and homogenous C5-DAB-SN-38. C5-DAB-SN-38 exhibited potent cytotoxicity activity against cNHL cells while inhibiting DNA-TopoI activity. Overall, our strategy validates a platform to develop a novel class of ADCs that combines the benefits of rabbit VL-sdAb scaffolds and the canine lymphoma model as a powerful framework for clinically translation of novel therapeutics for cancer.
Collapse
Affiliation(s)
- Ana S André
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Joana N R Dias
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Sandra Aguiar
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Sara Nogueira
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Pedro Bule
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Joana Inês Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - João P M António
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Marco Cavaco
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Vera Neves
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Soraia Oliveira
- Technophage SA, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Gonçalo Vicente
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Universidade de Lisboa, Av. da Universidade Técnica, 1300-477, Lisboa, Portugal
| | - Belmira Carrapiço
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Berta São Braz
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Barbara Rütgen
- Department of Pathobiology, Clinical Pathology Unit, University of Veterinary Medicine, Vienna, Austria
| | - Lurdes Gano
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10, 2695-066, Bobadela LRS, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Departamento de Engenharia e Ciências Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10, 2695-066, Bobadela LRS, Portugal
| | - Miguel Castanho
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028, Lisboa, Portugal
| | - Joao Goncalves
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Solange Gil
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Luís Tavares
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Frederico Aires-da-Silva
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal.
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal.
| |
Collapse
|
17
|
van den Berg EBA, Hendriks JCW, Elsinga EW, Eggink M, Dirksen EHC. Switching positions: Assessing the dynamics of conjugational heterogeneity in antibody-drug conjugates using CE-SDS. Electrophoresis 2023; 44:62-71. [PMID: 35907250 PMCID: PMC10086850 DOI: 10.1002/elps.202200140] [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: 05/30/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/01/2023]
Abstract
Antibody-drug conjugates (ADCs) are a prospective class of new oncology therapeutics with the ability to deliver a cytotoxic drug to a targeted location. The concept appears simple, but ADCs are highly complex due to their intrinsic heterogeneity. Randomly conjugated ADCs, for instance, are composed of conjugated species carrying between 0 and 8 linker-drug molecules, with several positional isomers that vary in drug distribution across the antibody. The drug load, expressed as drug-to-antibody ratio (DAR), is a critical quality attribute and should be well controlled, together with the distribution of drug molecules. Here, the impact of the duration of disulfide bond reduction on the DAR was investigated by quantitating the (isomeric) DAR species in ADCs produced with varying reduction times. Although hydrophobic interaction chromatography showed a constant DAR value as a function of reduction time, data obtained by non-reducing CE-SDS revealed an unexpected dynamic in the positional conjugated isomers. The insights obtained have improved our understanding of the correlation between the disulfide bond reduction, an important step in the manufacturing of a cysteine-conjugated ADC, and the conjugational heterogeneity.
Collapse
Affiliation(s)
| | - Jaap C. W. Hendriks
- Analytical Development and Quality Control (ADQC), Byondis B.V.NijmegenThe Netherlands
| | | | - Mark Eggink
- Analytical Development and Quality Control (ADQC), Byondis B.V.NijmegenThe Netherlands
| | - Eef H. C. Dirksen
- Analytical Development and Quality Control (ADQC), Byondis B.V.NijmegenThe Netherlands
| |
Collapse
|
18
|
Yu L, Shang Z, Jin Q, Chan SY, Hong W, Li N, Li P. Antibody-Antimicrobial Conjugates for Combating Antibiotic Resistance. Adv Healthc Mater 2023; 12:e2202207. [PMID: 36300640 DOI: 10.1002/adhm.202202207] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Indexed: 02/03/2023]
Abstract
As the development of new antibiotics lags far behind the emergence of drug-resistant bacteria, alternative strategies to resolve this dilemma are urgently required. Antibody-drug conjugate is a promising therapeutic platform to delivering cytotoxic payloads precisely to target cells for efficient disease treatment. Antibody-antimicrobial conjugates (AACs) have recently attracted considerable interest from researchers as they can target bacteria in the target sites and improve the effectiveness of drugs (i.e., reduced drug dosage and adverse effects), abating the upsurge of antimicrobial resistance. In this review, the selection and progress of three essential blocks that compose the AACs: antibodies, antimicrobial payloads, and linkers are discussed. The commonly used conjugation strategies and the latest applications of AACs in recent years are also summarized. The challenges and opportunities of this booming technology are also discussed at the end of this review.
Collapse
Affiliation(s)
- Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Zifang Shang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong Province, 518026, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology Chinese Academy of Sciences, Beijing, 100101, China
| | - Qizhe Jin
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China.,Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634, Singapore
| | - Weilin Hong
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Nan Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, 710072, China
| |
Collapse
|
19
|
de Faria e Silva AL, Ryder AG. Analyzing protein conjugation reactions for antibody-drug conjugate synthesis using polarized excitation emission matrix spectroscopy. Biotechnol Bioeng 2022; 119:3432-3446. [PMID: 36071600 PMCID: PMC9828061 DOI: 10.1002/bit.28229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 08/23/2022] [Accepted: 09/02/2022] [Indexed: 01/12/2023]
Abstract
Antibody-drug conjugates (ADCs) are promising anticancer therapeutics, which offer important advantages compared to more classical therapies. There are a variety of ADC critical quality attributes (CQAs) such as the protein structure, aggregation, and drug-to-antibody ratio (DAR), which all impact on potency, stability, and toxicity. Production processes can destabilize antibodies via a variety of physical and chemical stresses, and or by increased aggregation after conjugation of hydrophobic drugs. Thus, a proper control strategy for handling, production, and storage is necessary to maintain CQA levels, which requires the use of in-process quality measurements to first identify, then understand, and control the variables which adversely affect ADC CQAs during manufacturing. Here, we show how polarized excitation emission matrix (pEEM) spectroscopy, a sensitive, nondestructive, and potentially fast technique, can be used for rapidly assessing aggregation and DAR in a single measurement. pEEM provides several sources of information for protein analysis: Rayleigh scatter for identifying aggregate/particle formation and fluorescence emission to assess chemical and structural changes induced by attachment of a linker and/or a small molecule drug payload. Here, we used a nontoxic ADC mimic (monoclonal antibody with linker molecule) to demonstrate efficacy of the measurement method. Emission changes caused via light absorption by the attached linker, allowed us to predict DAR with good accuracy using fluorescence signal from the final purified products (6% relative error of prediction [REP]) and also from unpurified alkylation intermediates (11% REP). pEEM changes could also be correlated with size (hydrodynamic radius, Rh ) and aggregate content parameters obtained from dynamic light scattering and size exclusion chromatography (SEC). For the starting material and purified product samples, pEEM correlated better with Rh (R2 = 0.99, 6% REP) than SEC determined aggregate content (18% REP). Combining both fluorescence and light scatter signals also enabled in-process size quantification (6% REP). Overall, combining polarized measurements with EEM and Rayleigh scatter provides a single measurement, multi-attribute test method for ADC manufacturing.
Collapse
Affiliation(s)
- Ana L. de Faria e Silva
- Nanoscale BioPhotonics Laboratory, School of ChemistryNational University of IrelandGalwayIreland
| | - Alan G. Ryder
- Nanoscale BioPhotonics Laboratory, School of ChemistryNational University of IrelandGalwayIreland
| |
Collapse
|
20
|
Debnath U, Verma S, Patra J, Mandal SK. A review on recent synthetic routes and computational approaches for antibody drug conjugation developments used in anti-cancer therapy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
21
|
Murali M, Kumar AR, Nair B, Pavithran K, Devan AR, Pradeep GK, Nath LR. Antibody-drug conjugate as targeted therapeutics against hepatocellular carcinoma: preclinical studies and clinical relevance. Clin Transl Oncol 2022; 24:407-431. [PMID: 34595736 DOI: 10.1007/s12094-021-02707-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/29/2021] [Indexed: 02/05/2023]
Abstract
An antibody-drug conjugate (ADC) is an advanced chemotherapeutic option with immense promises in treating many tumor. They are designed to selectively attack and kill neoplastic cells with minimal toxicity to normal tissues. ADCs are complex engineered immunoconjugates that comprise a monoclonal antibody for site-directed delivery and cytotoxic payload for targeted destruction of malignant cells. Therefore, it enables the reduction of off-target toxicities and enhances the therapeutic index of the drug. Hepatocellular carcinoma (HCC) is a solid tumor that shows high heterogeneity of molecular phenotypes and is considered the second most common cause of cancer-related death. Studies show enormous potential for ADCs targeting GPC3 and CD24 and other tumor-associated antigens in HCC with their high, selective expression and show potential outputs in preclinical evaluations. The review mainly highlights the preclinical evaluation of different antigen-targeted ADCs such as MetFab-DOX, Anti-c-Met IgG-OXA, Anti CD 24, ANC-HN-01, G7mab-DOX, hYP7-DCand hYP7-PC, Anti-CD147 ILs-DOX and AC133-vcMMAF against hepatocellular carcinoma and its future relevance.
Collapse
Affiliation(s)
- M Murali
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - A R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - B Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - K Pavithran
- Department of Medical Oncology and Hematology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - A R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - G K Pradeep
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - L R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| |
Collapse
|
22
|
Kemp JA, Kwon YJ. Cancer nanotechnology: current status and perspectives. NANO CONVERGENCE 2021; 8:34. [PMID: 34727233 PMCID: PMC8560887 DOI: 10.1186/s40580-021-00282-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/05/2021] [Indexed: 05/09/2023]
Abstract
Modern medicine has been waging a war on cancer for nearly a century with no tangible end in sight. Cancer treatments have significantly progressed, but the need to increase specificity and decrease systemic toxicities remains. Early diagnosis holds a key to improving prognostic outlook and patient quality of life, and diagnostic tools are on the cusp of a technological revolution. Nanotechnology has steadily expanded into the reaches of cancer chemotherapy, radiotherapy, diagnostics, and imaging, demonstrating the capacity to augment each and advance patient care. Nanomaterials provide an abundance of versatility, functionality, and applications to engineer specifically targeted cancer medicine, accurate early-detection devices, robust imaging modalities, and enhanced radiotherapy adjuvants. This review provides insights into the current clinical and pre-clinical nanotechnological applications for cancer drug therapy, diagnostics, imaging, and radiation therapy.
Collapse
Affiliation(s)
- Jessica A Kemp
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, 92697, USA.
- Department of Chemical and Biomolecular Engineering, School of Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, School of Engineering, University of California, Irvine, CA, 92697, USA.
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
23
|
Islam M, Kehoe HP, Lissoos JB, Huang M, Ghadban CE, Sánchez GB, Lane HZ, Van Deventer JA. Chemical Diversification of Simple Synthetic Antibodies. ACS Chem Biol 2021; 16:344-359. [PMID: 33482061 PMCID: PMC8096149 DOI: 10.1021/acschembio.0c00865] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibodies possess properties that make them valuable as therapeutics, diagnostics, and basic research tools. However, antibody chemical reactivity and covalent antigen binding are constrained, or even prevented, by the narrow range of chemistries encoded in canonical amino acids. In this work, we investigate strategies for leveraging an expanded range of chemical functionality using yeast displayed antibodies containing noncanonical amino acids (ncAAs) in or near antibody complementarity determining regions (CDRs). To enable systematic characterization of the effects of ncAA incorporation on antibody function, we first investigated whether diversification of a single antibody loop would support the isolation of binding clones against immunoglobulins from three species. We constructed and screened a billion-member library containing canonical amino acid diversity and loop length diversity only within the third complementarity determining region of the heavy chain (CDR-H3). Isolated clones exhibited moderate affinities (double- to triple-digit nanomolar affinities) and, in several cases, single-species specificity, confirming that antibody specificity can be mediated by a single CDR. This constrained diversity enabled the utilization of additional CDRs for the installation of chemically reactive and photo-cross-linkable ncAAs. Binding studies of ncAA-substituted antibodies revealed that ncAA incorporation is reasonably well tolerated, with observed changes in affinity occurring as a function of ncAA side chain identity, substitution site, and the ncAA incorporation machinery used. Multiple azide-containing ncAAs supported copper-catalyzed azide-alkyne cycloaddition (CuAAC) and strain-promoted azide-alkyne cycloaddition (SPAAC) without the abrogation of binding function. Similarly, several alkyne substitutions facilitated CuAAC without the apparent disruption of binding. Finally, antibodies substituted with a photo-cross-linkable ncAA were evaluated for ultraviolet-mediated cross-linking on the yeast surface. Competition-based assays revealed position-dependent covalent linkages, strongly suggesting successful cross-linking. Key findings regarding CuAAC reactions and photo-cross-linking on the yeast surface were confirmed using soluble forms of ncAA-substituted clones. The consistency of findings on the yeast surface and in solution suggest that chemical diversification can be incorporated into yeast display screening approaches. Taken together, our results highlight the power of integrating the use of yeast display and ncAAs in search of proteins with "chemically augmented" binding functions. This includes strategies for systematically introducing small molecule functionality within binding protein structures and evaluating protein-based covalent target binding. The efficient preparation and chemical diversification of antibodies on the yeast surface open up new possibilities for discovering "drug-like" protein leads in high throughput.
Collapse
Affiliation(s)
- Mariha Islam
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Haixing P. Kehoe
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Jacob B. Lissoos
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Manjie Huang
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Christopher E. Ghadban
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Greg B. Sánchez
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Hanan Z. Lane
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - James A. Van Deventer
- Chemical and Biological Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
24
|
Le Gall CM, van der Schoot JMS, Ramos-Tomillero I, Khalily MP, van Dalen FJ, Wijfjes Z, Smeding L, van Dalen D, Cammarata A, Bonger KM, Figdor CG, Scheeren FA, Verdoes M. Dual Site-Specific Chemoenzymatic Antibody Fragment Conjugation Using CRISPR-Based Hybridoma Engineering. Bioconjug Chem 2021; 32:301-310. [PMID: 33476135 PMCID: PMC7898269 DOI: 10.1021/acs.bioconjchem.0c00673] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Functionalized antibodies
and antibody fragments have found applications
in the fields of biomedical imaging, theranostics, and antibody–drug
conjugates (ADC). In addition, therapeutic and theranostic approaches
benefit from the possibility to deliver more than one type of cargo
to target cells, further challenging stochastic labeling strategies.
Thus, bioconjugation methods to reproducibly obtain defined homogeneous
conjugates bearing multiple different cargo molecules, without compromising
target affinity, are in demand. Here, we describe a straightforward
CRISPR/Cas9-based strategy to rapidly engineer hybridoma cells to
secrete Fab′ fragments bearing two distinct site-specific labeling
motifs, which can be separately modified by two different sortase
A mutants. We show that sequential genetic editing of the heavy chain
(HC) and light chain (LC) loci enables the generation of a stable
cell line that secretes a dual tagged Fab′ molecule (DTFab′),
which can be easily isolated. To demonstrate feasibility, we functionalized
the DTFab′ with two distinct cargos in a site-specific manner.
This technology platform will be valuable in the development of multimodal
imaging agents, theranostics, and next-generation ADCs.
Collapse
Affiliation(s)
- Camille M Le Gall
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Johan M S van der Schoot
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Iván Ramos-Tomillero
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Melek Parlak Khalily
- Department of Synthetic Organic Chemistry, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Floris J van Dalen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Zacharias Wijfjes
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Liyan Smeding
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Duco van Dalen
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Anna Cammarata
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands
| | - Kimberly M Bonger
- Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands.,Department of Synthetic Organic Chemistry, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Oncode Institute, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| | - Ferenc A Scheeren
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Martijn Verdoes
- Department of Tumour Immunology, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, The Netherlands.,Institute for Chemical Immunology, Geert Grooteplein Zuid 28, 6525 GA, Nijmegen, Netherlands
| |
Collapse
|
25
|
Park J, Lee S, Kim Y, Yoo TH. Methods to generate site-specific conjugates of antibody and protein. Bioorg Med Chem 2021; 30:115946. [DOI: 10.1016/j.bmc.2020.115946] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
|