1
|
Timmins P. Industry update: the latest developments in the field of therapeutic delivery, December 2024. Ther Deliv 2025; 16:305-314. [PMID: 40047525 PMCID: PMC11970750 DOI: 10.1080/20415990.2025.2472722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Affiliation(s)
- Peter Timmins
- Department of Pharmacy, University of Huddersfield, Queensgate, Huddersfield, UK
| |
Collapse
|
2
|
Wharton T, Spring DR. Methods for the Generation of Single-Payload Antibody-Drug Conjugates. ChemMedChem 2025:e202500132. [PMID: 40052373 DOI: 10.1002/cmdc.202500132] [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: 02/16/2025] [Revised: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a powerful form of targeted therapy that can deliver drugs with a high level of selectivity towards a specific cell type, reducing off-target effects and increasing the therapeutic window compared to small molecule therapeutics. However, creating ADCs that are stable, homogeneous, and with controlled drug-to-antibody ratio (DAR) remains a significant challenge. Whilst a myriad of methods have been reported to generate ADCs with a DAR of 2, 4, and 8, strategies to generate DAR 1 constructs are seldom reported despite the advantages of low drug loading to tune ADC properties or to allow access to antibody-antibody and antibody-protein constructs. This concept article highlights the diversity of methods that have been employed to access single-payload ADCs and explores the outlook for the field.
Collapse
Affiliation(s)
- Thomas Wharton
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK, CB2 1EW
| | - David R Spring
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, UK, CB2 1EW
| |
Collapse
|
3
|
Able M, Kasper MA, Vick B, Schwach J, Gao X, Schmitt S, Tizazu B, Fischer A, Künzl S, Leilich M, Mai I, Ochtrop P, Stengl A, de Geus MAR, von Bergwelt-Baildon M, Schumacher D, Helma J, Hackenberger CPR, Götze KS, Jeremias I, Leonhardt H, Feuring M, Spiekermann K. Effective eradication of acute myeloid leukemia stem cells with FLT3-directed antibody-drug conjugates. Leukemia 2025; 39:632-642. [PMID: 39870768 PMCID: PMC11879846 DOI: 10.1038/s41375-024-02510-5] [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: 05/14/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/29/2025]
Abstract
Refractory disease and relapse are major challenges in acute myeloid leukemia (AML) therapy attributed to survival of leukemic stem cells (LSC). To target LSCs, antibody-drug conjugates (ADCs) provide an elegant solution, combining the specificity of antibodies with highly potent payloads. We aimed to investigate if FLT3-20D9h3-ADCs delivering either the DNA-alkylator duocarmycin (DUBA) or the microtubule-toxin monomethyl auristatin F (MMAF) can eradicate quiescent LSCs. We show here that DUBA more potently kills cell-cycle arrested AML cells compared to microtubule-targeting auristatins. Due to limited stability of 20D9h3-DUBA ADC in vivo, we analyzed both ADCs in advanced in vitro stem cell assays. 20D9h3-DUBA successfully eliminated leukemic progenitors in vitro in colony-forming unit and long-term culture initiating cell assays, both in patient cells and in patient-derived xenograft (PDX) cells. Further, it completely prevented engraftment of AML PDX leukemia-initiating cells in NSG mice. 20D9h3-MMAF had a similar effect in engraftment assays, but a less prominent effect in colony assays. Both ADCs did not affect healthy stem and progenitor cells at comparable doses providing the rationale for FLT3 as therapeutic LSC target. Collectively, we show that FLT3-directed ADCs with DUBA or MMAF have potent activity against AML LSCs and represent promising candidates for further clinical development.
Collapse
Affiliation(s)
- Marina Able
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marc-André Kasper
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Binje Vick
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Research Unit Apoptosis in Hematopoietic Stem Cells (AHS), Helmholtz Munich, German Research Center for Environmental Health (HMGU), Munich, Germany
| | - Jonathan Schwach
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Xiang Gao
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Belay Tizazu
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Amrei Fischer
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Künzl
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Marit Leilich
- Technical University of Munich School of Medicine and Health, Department of Medicine III, Technical University of Munich (TUM), Munich, Germany
| | | | - Philipp Ochtrop
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Andreas Stengl
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Mark A R de Geus
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Dominik Schumacher
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
- Tubulis GmbH, Munich, Germany
| | - Jonas Helma
- Tubulis GmbH, Munich, Germany
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Christian P R Hackenberger
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Campus Berlin, Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | - Katharina S Götze
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Technical University of Munich School of Medicine and Health, Department of Medicine III, Technical University of Munich (TUM), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Irmela Jeremias
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany
- Research Unit Apoptosis in Hematopoietic Stem Cells (AHS), Helmholtz Munich, German Research Center for Environmental Health (HMGU), Munich, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU, Munich, Germany
| | - Heinrich Leonhardt
- Faculty of Biology, Human Biology and BioImaging, LMU Munich, Planegg-Martinsried, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Karsten Spiekermann
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU University Hospital, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
4
|
Li X, Patel NL, Kalen J, Schnermann MJ. Benzyl Ammonium Carbamates Undergo Two-Step Linker Cleavage and Improve the Properties of Antibody Conjugates. Angew Chem Int Ed Engl 2025; 64:e202417651. [PMID: 39696914 PMCID: PMC11795738 DOI: 10.1002/anie.202417651] [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: 09/13/2024] [Indexed: 12/20/2024]
Abstract
Targeted payload delivery strategies, such as antibody-drug conjugates (ADCs), have emerged as important therapeutics. Although considerable efforts have been made in the areas of antibody engineering and labeling methodology, improving the overall physicochemical properties of the linker/payload combination remains an important challenge. Here we report an approach to create an intrinsically hydrophilic linker domain. We find that benzyl α-ammonium carbamates (BACs) undergo tandem 1,6-1,2-elimination to release secondary amines. Using a fluorogenic hemicyanine as a model payload component, we show that a zwitterionic BAC linker improves labeling efficiency and reduces antibody aggregation when compared to a commonly used para-amino benzyl (PAB) linker as well as a cationic BAC. Cellular and in vivo fluorescence imaging studies demonstrate that the model payload is specifically released in antigen-expressing cells and tumors. The therapeutic potential of the BAC linker strategy was assessed using an MMAE payload, a potent microtubule-disrupting agent frequently used for ADC applications. The BAC-MMAE combination enhances labeling efficiency and cellular toxicity when compared to the routinely used PAB-Val-Cit ADC analogue. Broadly, this strategy provides a general approach to mask payload hydrophobicity and improve the properties of targeted agents.
Collapse
Affiliation(s)
- Xiaoyi Li
- Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMaryland21702USA
| | - Nimit L. Patel
- Small Animal Imaging ProgramFrederick National Laboratory for Cancer ResearchLeidos Biomedical Research Inc.FrederickMaryland21702USA
| | - Joseph Kalen
- Small Animal Imaging ProgramFrederick National Laboratory for Cancer ResearchLeidos Biomedical Research Inc.FrederickMaryland21702USA
| | - Martin J. Schnermann
- Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMaryland21702USA
| |
Collapse
|
5
|
Boos A, Most J, Cahuzac H, Moreira da Silva L, Daubeuf F, Erb S, Cianférani S, Hernandez-Alba O, Semenchenko C, Dovgan I, Kolodych S, Detappe A, Dantzer F, Wagner A, Zeniou M, Chaubet G. Antibody-Vincristine Conjugates as Potent Anticancer Therapeutic Agents. J Med Chem 2025; 68:695-705. [PMID: 39680648 DOI: 10.1021/acs.jmedchem.4c02425] [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: 12/18/2024]
Abstract
Antibody-drug conjugates (ADCs) are a well-established class of therapeutics primarily used in oncology to selectively deliver highly cytotoxic agents into cancer cells. While ADCs should theoretically spare healthy tissues and diminish side effects in patients, off-target toxicity is still observed, all the more serious, as the drugs are extremely potent. In the quest toward safer payloads, we used the conventional chemotherapeutic drug vincristine to develop antibody-vincristine conjugates. Vincristine was N-alkylated with a cleavable linker and the resulting linker-payload conjugated to free cysteines of antibodies. We show that trastuzumab-vincristine conjugates display subnanomolar potency in vitro on HER2-positive cells, 2 orders of magnitude lower than free vincristine and comparable with marketed ADC. In vivo, trastuzumab-vincristine conjugates led to remarkable efficacy when compared to two standards of care, with complete tumor regression just 9 days after single administration. This highlights the untapped potential of the chemotherapeutic arsenal toward the development of novel ADC.
Collapse
Affiliation(s)
- Agathe Boos
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d'Excellence Medalis, UMR7242, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412 Illkirch, France
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
| | - Julien Most
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- Bio-Functional Chemistry, Laboratoire d'Excellence Medalis, UMR7199, Centre National de la Recherche Scientifique/Université de Strasbourg, Faculté de Pharmacie de Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Héloïse Cahuzac
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- Bio-Functional Chemistry, Laboratoire d'Excellence Medalis, UMR7199, Centre National de la Recherche Scientifique/Université de Strasbourg, Faculté de Pharmacie de Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Louis Moreira da Silva
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- Bio-Functional Chemistry, Laboratoire d'Excellence Medalis, UMR7199, Centre National de la Recherche Scientifique/Université de Strasbourg, Faculté de Pharmacie de Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - François Daubeuf
- PCBIS (CNRS - UAR 3286), ESBS - Pôle API, 300, boulevard Sébastien Brant, CS 10413 67412 Illkirch-Graffenstaden Cedex, France
| | - Stéphane Erb
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- BioOrganic Mass Spectrometry, Laboratoire d'Excellence Medalis, UMR7178, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut pluridisciplinaire Hubert Curien, 23 rue du loess, BP 28 67037, Strasbourg Cedex 2, France
- Infrastructure Nationale de Protéomique ProFI ─ FR2048, 67087 Strasbourg, France
| | - Sarah Cianférani
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- BioOrganic Mass Spectrometry, Laboratoire d'Excellence Medalis, UMR7178, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut pluridisciplinaire Hubert Curien, 23 rue du loess, BP 28 67037, Strasbourg Cedex 2, France
- Infrastructure Nationale de Protéomique ProFI ─ FR2048, 67087 Strasbourg, France
| | - Oscar Hernandez-Alba
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- BioOrganic Mass Spectrometry, Laboratoire d'Excellence Medalis, UMR7178, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut pluridisciplinaire Hubert Curien, 23 rue du loess, BP 28 67037, Strasbourg Cedex 2, France
- Infrastructure Nationale de Protéomique ProFI ─ FR2048, 67087 Strasbourg, France
| | | | - Igor Dovgan
- Carbogen Amcis AG, Hauptstrasse 171, 4416 Bubendorf, Switzerland
| | - Sergii Kolodych
- Syndivia SAS, Syndivia, 8 allée Gaspard Monge, 67083 Strasbourg, France
| | - Alexandre Detappe
- Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
- Équipe labellisée Ligue contre le Cancer, Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
| | - Françoise Dantzer
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d'Excellence Medalis, UMR7242, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412 Illkirch, France
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
| | - Alain Wagner
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- Bio-Functional Chemistry, Laboratoire d'Excellence Medalis, UMR7199, Centre National de la Recherche Scientifique/Université de Strasbourg, Faculté de Pharmacie de Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Maria Zeniou
- Poly(ADP-ribosyl)ation and Genome Integrity, Laboratoire d'Excellence Medalis, UMR7242, Centre National de la Recherche Scientifique/Université de Strasbourg, Institut de Recherche de l'Ecole de Biotechnologie de Strasbourg, 300 bld. S. Brant, CS10413, 67412 Illkirch, France
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
| | - Guilhem Chaubet
- Strasbourg Drug Discovery and Development Institute (IMS), 67000 Strasbourg, France
- Bio-Functional Chemistry, Laboratoire d'Excellence Medalis, UMR7199, Centre National de la Recherche Scientifique/Université de Strasbourg, Faculté de Pharmacie de Strasbourg, 74 Route du Rhin, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
6
|
Tao J, Gu Y, Zhou W, Wang Y. Dual-payload antibody-drug conjugates: Taking a dual shot. Eur J Med Chem 2025; 281:116995. [PMID: 39481229 DOI: 10.1016/j.ejmech.2024.116995] [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: 08/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Antibody-drug conjugates (ADCs) enable the precise delivery of cytotoxic agents by conjugating small-molecule drugs with monoclonal antibodies (mAbs). Over recent decades, ADCs have demonstrated substantial clinical efficacy. However, conventional ADCs often encounter various clinical challenges, including suboptimal efficacy, significant adverse effects, and the development of drug resistance, limiting their broader clinical application. Encouragingly, a next-generation approach-dual-payload ADCs-has emerged as a pioneering strategy to address these challenges. Dual-payload ADCs are characterized by the incorporation of two distinct therapeutic payloads on the same antibody, enhancing treatment efficacy by promoting synergistic effects and reducing the risk of drug resistance. However, the synthesis of dual-payload ADCs is complex due to the presence of multiple functional groups on antibodies. In this review, we comprehensively summarize the construction strategies for dual-payload ADCs, ranging from the design of ADC components to orthogonal chemistry. The subsequent sections explore current challenges and propose prospective strategies, highlighting recent advancements in dual-payload ADC research, thereby laying the foundation for the development of next-generation ADCs.
Collapse
Affiliation(s)
- Junjie Tao
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Zhou
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
7
|
Cui ZH, Zhang H, Zheng FH, Xue JH, Yin QH, Xie XL, Wang YX, Wang T, Zhou L, Fang GM. Generation of antibody-drug conjugates by proximity-driven acyl transfer and sortase-mediated ligation. Org Biomol Chem 2024; 23:188-196. [PMID: 39530194 DOI: 10.1039/d4ob01624f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
We report a sortase-based site-specific antibody-drug conjugation strategy, which involves an affinity peptide-directed acyl transfer reaction and sortase-mediated peptide ligation. Through the affinity peptide-mediated acyl transfer reaction, an LPXTG-containing peptide is conjugated to a specific Lys side chain of an antibody. Under the assistance of sortase, a protein drug bearing a GG motif reacts specifically with the LPXTG moiety to produce an antibody-drug conjugate. Our strategy for antibody conjugation can be applied not only to chemically synthesized drugs, but also to biologically expressed proteins, and will provide a new sortase-based strategy for the preparation of antibody-drug conjugates.
Collapse
Affiliation(s)
- Zhi-Hui Cui
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Hua Zhang
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Feng-Hao Zheng
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Jun-Hao Xue
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Qing-Hong Yin
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Xiao-Lei Xie
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Yu-Xuan Wang
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| | - Tao Wang
- University of Science and Technology of China, Hefei 230026, P. R. China.
| | - Li Zhou
- Anhui Provincial Peptide Drug Engineering Laboratory, Hefei KS-V Peptide Biological Technology Co., Ltd, P. R. China.
| | - Ge-Min Fang
- School of Life Science, Institutes of Physical Science and Information Technology, Institute of Health Sciences, Anhui University, Hefei, 230601, P. R. China.
| |
Collapse
|
8
|
Tsumura R, Anzai T, Koga Y, Takashima H, Matsumura Y, Yasunaga M. Anti-tissue factor antibody conjugated with monomethyl auristatin E or deruxtecan in pancreatic cancer models. Cancer Sci 2024; 115:3986-3996. [PMID: 39322584 PMCID: PMC11611767 DOI: 10.1111/cas.16335] [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: 05/07/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have been recognized as a promising class of cancer therapeutics. Tissue factor (TF), an initiator of the blood coagulation pathway, has been investigated regarding its relationship with cancer, and several preclinical and clinical studies have presented data on anti-TF ADCs, including tisotumab vedotin, which was approved in 2021. However, the feasibility of other payloads in the design of anti-TF ADCs is still unclear because no reports have compared payloads with different cytotoxic mechanisms. For ADCs targeting other antigens, such as Her2, optimizing the payload is also an important issue in order to improve in vivo efficacy. In this study, we prepared humanized anti-TF Ab (clone.1084) conjugated with monomethyl auristatin E (MMAE) or deruxtecan (DXd), and evaluated the efficacy in several cell line- and patient-derived xenograft models of pancreatic cancer. As a result, optimizing the drug / Ab ratio was necessary for each payload in order to prevent pharmacokinetic deterioration and maximize delivery efficiency. In addition, MMAE-conjugated anti-TF ADC showed higher antitumor effects in tumors with strong and homogeneous TF expression, while DXd-conjugated anti-TF ADC was more effective in tumors with weak and heterogeneous TF expression. Analysis of a pancreatic cancer tissue array showed weak and heterogeneous TF expression in most TF-positive specimens, indicating that the response rate to pancreatic cancer might be higher for DXd- than MMAE-conjugated anti-TF ADC. Nevertheless, our findings indicated that optimizing the ADC payloads individually in each patient could maximize the potential of ADC therapeutics.
Collapse
Affiliation(s)
- Ryo Tsumura
- Division of Developmental TherapeuticsEPOC, National Cancer CenterKashiwaJapan
| | - Takahiro Anzai
- Division of Developmental TherapeuticsEPOC, National Cancer CenterKashiwaJapan
- Department of Chemistry and Materials ScienceNational Institute of Technology (KOSEN), Gunma CollegeMaebashiJapan
| | - Yoshikatsu Koga
- Division of Developmental TherapeuticsEPOC, National Cancer CenterKashiwaJapan
| | - Hiroki Takashima
- Division of Developmental TherapeuticsEPOC, National Cancer CenterKashiwaJapan
| | - Yasuhiro Matsumura
- Department of Immune MedicineNational Cancer Center Research InstituteTokyoJapan
- Research DivisionRIN Institute Inc.TokyoJapan
| | - Masahiro Yasunaga
- Division of Developmental TherapeuticsEPOC, National Cancer CenterKashiwaJapan
| |
Collapse
|
9
|
Hua Y, Zou Z, Prescimone A, Ward TR, Mayor M, Köhler V. NSPs: chromogenic linkers for fast, selective, and irreversible cysteine modification. Chem Sci 2024; 15:10997-11004. [PMID: 39027294 PMCID: PMC11253191 DOI: 10.1039/d4sc01710b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
The addition of a sulfhydryl group to water-soluble N-alkyl(o-nitrostyryl)pyridinium ions (NSPs) followed by fast and irreversible cyclization and aromatization results in a stable S-C sp2-bond. The reaction sequence, termed Click & Lock, engages accessible cysteine residues under the formation of N-hydroxy indole pyridinium ions. The accompanying red shift of >70 nm to around 385 nm enables convenient monitoring of the labeling yield by UV-vis spectroscopy at extinction coefficients of ≥2 × 104 M-1 cm-1. The versatility of the linker is demonstrated in the stapling of peptides and the derivatization of proteins, including the modification of reduced trastuzumab with Val-Cit-PAB-MMAE. The high stability of the linker in human plasma, fast reaction rates (k app up to 4.4 M-1 s-1 at 20 °C), high selectivity for cysteine, favorable solubility of the electrophilic moiety and the bathochromic properties of the Click & Lock reaction provide an appealing alternative to existing methods for cysteine conjugation.
Collapse
Affiliation(s)
- Yong Hua
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Zhi Zou
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Alessandro Prescimone
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| | - Thomas R Ward
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
- National Center of Competence in Research (NCCR) "Molecular Systems Engineering" 4058 Basel Switzerland
| | - Marcel Mayor
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
- Institute for Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMFi) Karlsruhe Institute of Technology (KIT) P.O. Box 3640 DE-76021 Karlsruhe Eggenstein-Leopoldshafen Germany
- Lehn Institute of Functional Materials (LIFM), School of Chemistry, Sun Yat-Sen University (SYSU) XinGangXi Road 135 510275 Guangzhou P. R. China
| | - Valentin Köhler
- Department of Chemistry, University of Basel St. Johannsring 19 CH-4056 Basel Switzerland
- Department of Chemistry, University of Basel Mattenstrasse 22 CH-4058 Basel Switzerland
| |
Collapse
|
10
|
Ahangarpour M, Brimble MA, Kavianinia I. Late-Stage Desulfurization Enables Rapid and Efficient Solid-Phase Synthesis of Cathepsin-Cleavable Linkers for Antibody-Drug Conjugates. Bioconjug Chem 2024; 35:1007-1014. [PMID: 38874557 DOI: 10.1021/acs.bioconjchem.4c00199] [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: 06/15/2024]
Abstract
The synthesis of linker-payloads is a critical step in developing antibody-drug conjugates (ADCs), a rapidly advancing therapeutic approach in oncology. The conventional method for synthesizing cathepsin B-labile dipeptide linkers, which are commonly used in ADC development, involves the solution-phase assembly of cathepsin B-sensitive dipeptides, followed by the installation of self-immolative para-aminobenzyl carbonate to facilitate the attachment of potent cytotoxic payloads. However, this approach is often low yield and laborious, especially when extending the peptide chain with components like glutamic acid to improve mouse serum stability or charged amino acids or poly(ethylene glycol) moieties to enhance linker hydrophilicity. Here, we introduce a novel approach utilizing late-stage desulfurization chemistry, enabling safe, facile, and cost-effective access to the cathepsin B-cleavable linker, Val-Ala-PABC-MMAE, on resin for the first time.
Collapse
Affiliation(s)
- Marzieh Ahangarpour
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, 23 Symonds St., Auckland 1010, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - Iman Kavianinia
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
11
|
Tsuchikama K, Anami Y, Ha SYY, Yamazaki CM. Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol 2024; 21:203-223. [PMID: 38191923 DOI: 10.1038/s41571-023-00850-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 149.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Summer Y Y Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chisato M Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
12
|
Schmitt S, Machui P, Mai I, Herterich S, Wunder S, Cyprys P, Gerlach M, Ochtrop P, Hackenberger CP, Schumacher D, Helma J, Vogl AM, Kasper MA. Design and Evaluation of Phosphonamidate-Linked Exatecan Constructs for Highly Loaded, Stable, and Efficacious Antibody-Drug Conjugates. Mol Cancer Ther 2024; 23:199-211. [PMID: 37828728 PMCID: PMC10831470 DOI: 10.1158/1535-7163.mct-23-0359] [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: 06/10/2023] [Revised: 08/30/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody-drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker-payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker-payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB-exatecan moiety. Furthermore, we demonstrate that the identified linker-payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker-payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christian P.R. Hackenberger
- Chemical Biology Department, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
13
|
Chauhan P, V R, Kumar M, Molla R, Mishra SD, Basa S, Rai V. Chemical technology principles for selective bioconjugation of proteins and antibodies. Chem Soc Rev 2024; 53:380-449. [PMID: 38095227 DOI: 10.1039/d3cs00715d] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Proteins are multifunctional large organic compounds that constitute an essential component of a living system. Hence, control over their bioconjugation impacts science at the chemistry-biology-medicine interface. A chemical toolbox for their precision engineering can boost healthcare and open a gateway for directed or precision therapeutics. Such a chemical toolbox remained elusive for a long time due to the complexity presented by the large pool of functional groups. The precise single-site modification of a protein requires a method to address a combination of selectivity attributes. This review focuses on guiding principles that can segregate them to simplify the task for a chemical method. Such a disintegration systematically employs a multi-step chemical transformation to deconvolute the selectivity challenges. It constitutes a disintegrate (DIN) theory that offers additional control parameters for tuning precision in protein bioconjugation. This review outlines the selectivity hurdles faced by chemical methods. It elaborates on the developments in the perspective of DIN theory to demonstrate simultaneous regulation of reactivity, chemoselectivity, site-selectivity, modularity, residue specificity, and protein specificity. It discusses the progress of such methods to construct protein and antibody conjugates for biologics, including antibody-fluorophore and antibody-drug conjugates (AFCs and ADCs). It also briefs how this knowledge can assist in developing small molecule-based covalent inhibitors. In the process, it highlights an opportunity for hypothesis-driven routes to accelerate discoveries of selective methods and establish new targetome in the precision engineering of proteins and antibodies.
Collapse
Affiliation(s)
- Preeti Chauhan
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Ragendu V
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Mohan Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Rajib Molla
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Surya Dev Mishra
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Sneha Basa
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| | - Vishal Rai
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, 462 066, India.
| |
Collapse
|