1
|
Vasic V, Dickopf S, Spranger N, Rosenberger RS, Fischer M, Mayer K, Larraillet V, Bates JA, Maier V, Sela T, Nussbaum B, Duerr H, Dengl S, Brinkmann U. Generation of binder-format-payload conjugate-matrices by antibody chain-exchange. Nat Commun 2024; 15:9406. [PMID: 39477939 PMCID: PMC11525586 DOI: 10.1038/s41467-024-53730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
The generation of antibody-drug conjugates with optimal functionality depends on many parameters. These include binder epitope, antibody format, linker composition, conjugation site(s), drug-to-antibody ratio, and conjugation method. The production of matrices that cover all possible parameters is a major challenge in identifying optimal antibody-drug conjugates. To address this bottleneck, we adapted our Format Chain Exchange technology (FORCE), originally established for bispecific antibodies, toward the generation of binder-format-payload matrices (pair-FORCE). Antibody derivatives with exchange-enabled Fc-heterodimers are combined with payload-conjugated Fc donors, and subsequent chain-exchange transfers payloads to antibody derivatives in different formats. The resulting binder-format-conjugate matrices can be generated with cytotoxic payloads, dyes, haptens, and large molecules, resulting in versatile tools for ADC screening campaigns. We show the relevance of pair-FORCE for identifying optimal HER2-targeting antibody-drug conjugates. Analysis of this matrix reveals that the notion of format-defines-function applies not only to bispecific antibodies, but also to antibody-drug conjugates.
Collapse
Affiliation(s)
- Vedran Vasic
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Steffen Dickopf
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Veraxa Biotech, Heidelberg, Germany
| | - Nadine Spranger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Institute of Molecular Immunology, School of Medicine and Health, Technical University Munich (TUM), Munich, Germany
| | - Rose-Sophie Rosenberger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Fischer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Klaus Mayer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Vincent Larraillet
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Jack A Bates
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Verena Maier
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Tatjana Sela
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Bianca Nussbaum
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Harald Duerr
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Stefan Dengl
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany.
| |
Collapse
|
2
|
Tao J, Gu Y, Zhou W, Wang Y. Dual-payload antibody-drug conjugates: Taking a dual shot. Eur J Med Chem 2024; 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] [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
|
3
|
Mathiot L, Baldini C, Letissier O, Hollebecque A, Bahleda R, Gazzah A, Smolenschi C, Sakkal M, Danlos FX, Henon C, Beshiri K, Goldschmidt V, Parisi C, Patrikidou A, Michot JM, Marabelle A, Postel-Vinay S, Bernard-Tessier A, Loriot Y, Ponce S, Champiat S, Ouali K. Exploring the Role of Target Expression in Treatment Efficacy of Antibody-Drug Conjugates (ADCs) in Solid Cancers: A Comprehensive Review. Curr Oncol Rep 2024; 26:1236-1248. [PMID: 39066847 DOI: 10.1007/s11912-024-01576-9] [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] [Accepted: 06/29/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE OF REVIEW Antibody-drug conjugates (ADCs) offer a promising path for cancer therapy, leveraging the specificity of monoclonal antibodies and the cytotoxicity of linked drugs. The success of ADCs hinges on precise targeting of cancer cells based on protein expression levels. This review explores the relationship between target protein expression and ADC efficacy in solid tumours, focusing on results of clinical trials conducted between January 2019 and May 2023. RECENT FINDINGS We hereby highlight approved ADCs, revealing their effectiveness even in low-expressing target populations. Assessing target expression poses challenges, owing to variations in scoring systems and biopsy types. Emerging methods, like digital image analysis, aim to standardize assessment. The complexity of ADC pharmacokinetics, tumour dynamics, and off-target effects emphasises the need for a balanced approach. This review underscores the importance of understanding target protein dynamics and promoting standardized evaluation methods in shaping the future of ADC-based cancer therapies.
Collapse
Affiliation(s)
- Laurent Mathiot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Capucine Baldini
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Octave Letissier
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Antoine Hollebecque
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Rastislav Bahleda
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anas Gazzah
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Cristina Smolenschi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Madona Sakkal
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - François-Xavier Danlos
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Clémence Henon
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Kristi Beshiri
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Vincent Goldschmidt
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Claudia Parisi
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Anna Patrikidou
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Jean-Marie Michot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Aurélien Marabelle
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Sophie Postel-Vinay
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | | | - Yohann Loriot
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Villejuif, France
| | - Santiago Ponce
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
| | - Stéphane Champiat
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Villejuif, France
- Centre d'Investigations Cliniques Biothérapies Pour Une Immunisation in Situ (BIOTHERIS), INSERM, CIC1428, Villejuif, France
| | - Kaïssa Ouali
- Drug Development Department, Gustave Roussy Cancer Campus, Villejuif Cedex, France.
| |
Collapse
|
4
|
Wehrmüller JE, Frei JC, Hechler T, Kulke M, Pahl A, Béhé M, Schibli R, Spycher PR. Site-Specific Modification of Native IgGs with Flexible Drug-Load. Chembiochem 2024:e202400511. [PMID: 39305147 DOI: 10.1002/cbic.202400511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/11/2024] [Indexed: 11/08/2024]
Abstract
Homogeneous, site-specifically conjugated antibodies have shown to result in antibody-drug conjugates (ADCs) with improved efficacy and tolerability compared to stochastically conjugated ADCs. However, precisely controlling the drug load as well as attaching multiple payload moieties to the antibody remains challenging. Here, we demonstrate the simple and direct modification of native IgG-antibodies at the residue glutamine 295 (Q295) without the need for any protein engineering with flexible drug-to-antibody ratios of one or multiple payloads. The conjugation is enabled through short, positively charged lysine containing peptides and native, commercially available microbial transglutaminase. In proof-of-concept studies, HER2-targeting ADCs based on trastuzumab were generated with drug-to-antibody ratios (DARs) of 2 and 4 of the same or different payloads using orthogonal conjugation chemistries. Quantitative biodistribution studies performed with 111In-radiolabeled conjugates showed high tumour uptake and low accumulation of radioactivity in non-targeted tissues. A single dose study of trastuzumab conjugated to the highly potent payload α-Amanitin demonstrated complete and long-lasting tumour remission and was well-tolerated at all dose levels tested.
Collapse
Affiliation(s)
- Jöri E Wehrmüller
- Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Julia C Frei
- Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Torsten Hechler
- Heidelberg Pharma Research GmbH, Gregor-Mendel-Straße 22, 68526, Ladenburg, Germany
| | - Michael Kulke
- Heidelberg Pharma Research GmbH, Gregor-Mendel-Straße 22, 68526, Ladenburg, Germany
| | - Andreas Pahl
- Heidelberg Pharma Research GmbH, Gregor-Mendel-Straße 22, 68526, Ladenburg, Germany
| | - Martin Béhé
- Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Philipp R Spycher
- Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232, Villigen-PSI, Switzerland
- Araris Biotech AG, Riedhofstrasse 11, 8804, Au ZH, Switzerland
| |
Collapse
|
5
|
Szepesi Kovács D, Pásztor B, Ábrányi-Balogh P, Petri L, Imre T, Simon J, Tátrai E, Várady G, Tóvári J, Szijj PA, Keserű GM. Site-Selective Antibody Conjugation with Dibromopyrazines. Bioconjug Chem 2024; 35:1373-1379. [PMID: 39151068 PMCID: PMC11417993 DOI: 10.1021/acs.bioconjchem.4c00296] [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: 06/28/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/18/2024]
Abstract
In recent years, antibody conjugates have evolved as state-of-the-art options for diagnostic and therapeutic applications. During site-selective antibody conjugation, incomplete rebridging of antibody chains limits the homogeneity of conjugates and calls for the development of new rebridging agents. Herein, we report a dibromopyrazine derivative optimized to reach highly homogeneous conjugates rapidly and with high conversion on rebridging of trastuzumab, even providing a feasible route for antibody modification in acidic conditions. Furthermore, coupling a fluorescent dye and a cytotoxic drug resulted in effective antibody conjugates with excellent serum stability and in vitro selectivity, demonstrating the utility of the dibromopyrazine rebridging agent to produce on-demand future antibody conjugates for diagnostic or therapeutic applications.
Collapse
Affiliation(s)
- Dénes Szepesi Kovács
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Bettina Pásztor
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - László Petri
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- Institute
of Chemistry, Faculty of Science, Eötvös
Loránd University, Egyetem t. 1–3, H-1053 Budapest, Hungary
| | - Tímea Imre
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- MS
Metabolomics Research Laboratory, Research
Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - József Simon
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- MS
Metabolomics Research Laboratory, Research
Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Enikő Tátrai
- Department
of Experimental Pharmacology, National Institute
of Oncology, Ráth
György u. 7–9, H-1122 Budapest, Hungary
- National
Tumor Biology Laboratory, Ráth György u. 7–9, H-1122 Budapest, Hungary
| | - György Várady
- Molecular
Cell Biology Research Group, Research Centre
for Natural Sciences, H-1117 Budapest, Hungary
| | - József Tóvári
- Department
of Experimental Pharmacology, National Institute
of Oncology, Ráth
György u. 7–9, H-1122 Budapest, Hungary
- National
Tumor Biology Laboratory, Ráth György u. 7–9, H-1122 Budapest, Hungary
| | - Peter A. Szijj
- Department
of Chemistry, University College London, WC1H 0AJ London, U.K.
| | - György M. Keserű
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
- Department
of Organic Chemistry and Technology, Faculty of Chemical Technology
and Biotechnology, Budapest University of
Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- National
Drug Research and Development Laboratory, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| |
Collapse
|
6
|
Cochran M, Marks I, Albin T, Arias D, Kovach P, Darimont B, Huang H, Etxaniz U, Kwon HW, Shi Y, Diaz M, Tyaglo O, Levin A, Doppalapudi VR. Structure-Activity Relationship of Antibody-Oligonucleotide Conjugates: Evaluating Bioconjugation Strategies for Antibody-Phosphorodiamidate Morpholino Oligomer Conjugates for Drug Development. J Med Chem 2024; 67:14868-14884. [PMID: 39197837 PMCID: PMC11403617 DOI: 10.1021/acs.jmedchem.4c00803] [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: 09/01/2024]
Abstract
Antibody-oligonucleotide conjugates (AOCs) are promising treatments for Duchenne muscular dystrophy (DMD). They work via induction of exon skipping and restoration of dystrophin protein in skeletal and heart muscles. The structure-activity relationships (SARs) of AOCs comprising antibody-phosphorodiamidate morpholino oligomers (PMOs) depend on several aspects of their component parts. We evaluate the SAR of antimouse transferrin receptor 1 antibody (αmTfR1)-PMO conjugates: cleavable and noncleavable linkers, linker location on the PMO, and the impact of drug-to-antibody ratios (DARs) on plasma pharmacokinetics (PK), oligonucleotide delivery to tissues, and exon skipping. AOCs containing a stable linker with a DAR9.7 were the most effective PMO delivery vehicles in preclinical studies. We demonstrate that αmTfR1-PMO conjugates induce dystrophin protein restoration in the skeletal and heart muscles of mdx mice. Our results show that αmTfR1-PMO conjugates are a potentially effective approach for the treatment of DMD.
Collapse
Affiliation(s)
- Michael Cochran
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Isaac Marks
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Tyler Albin
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Danny Arias
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Philip Kovach
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | | | - Hanhua Huang
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Usue Etxaniz
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Hae Won Kwon
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Yunyu Shi
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Matthew Diaz
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Olecya Tyaglo
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Arthur Levin
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| | - Venkata Ramana Doppalapudi
- Avidity Biosciences, Inc., 10578 Science Center Drive Suite 125, San Diego, California 92121, United States
| |
Collapse
|
7
|
Malik S, Sikander M, Bell N, Zubieta D, Bell MC, Yallapu MM, Chauhan SC. Emerging role of mucins in antibody drug conjugates for ovarian cancer therapy. J Ovarian Res 2024; 17:161. [PMID: 39118097 PMCID: PMC11308542 DOI: 10.1186/s13048-024-01485-2] [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: 05/15/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Ovarian cancer stands as the deadliest gynecologic malignancy, responsible for nearly 65% of all gynecologic cancer-related deaths. The challenges in early detection and diagnosis, coupled with the widespread intraperitoneal spread of cancer cells and resistance to chemotherapy, contribute significantly to the high mortality rate of this disease. Due to the absence of specific symptoms and the lack of effective screening methods, most ovarian cancer cases are diagnosed at advanced stages. While chemotherapy is a common treatment, it often leads to tumor recurrence, necessitating further interventions. In recent years, antibody-drug conjugates (ADCs) have emerged as a valuable tool in targeted cancer therapy. These complex biotherapeutics combine an antibody that specifically targets tumor specific/associated antigen(s) with a high potency anti-cancer drug through a linker, offering a promising approach for ovarian cancer treatment. The identification of molecular targets in various human tumors has paved the way for the development of targeted therapies, with ADCs being at the forefront of this innovation. By delivering cytotoxic agents directly to tumors and metastatic lesions, ADCs show potential in managing chemo-resistant ovarian cancers. Mucins such as MUC16, MUC13, and MUC1 have shown significantly higher expression in ovarian tumors as compared to normal and/or benign samples, thus have become promising targets for ADC generation. While traditional markers are limited by their elevated levels in non-cancerous conditions, mucins offer a new possibility for targeted treatment in ovarian cancer. This review comprehensively described the potential of mucins for the generation of ADC therapy, highlighting their importance in the quest to improve the outcome of ovarian cancer patients.
Collapse
Affiliation(s)
- Shabnam Malik
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA
| | - Mohammed Sikander
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA
| | - Natasha Bell
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA
| | - Daniel Zubieta
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA
| | - Maria C Bell
- Sanford Health, Sanford Gynecologic Oncology Clinic, Sioux Falls, SD, 57104, USA
| | - Murali M Yallapu
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA
| | - Subhash C Chauhan
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA.
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX, 78504, USA.
| |
Collapse
|
8
|
Hengel SM, Topletz-Erickson AR, Kadry H, Alley SC. A modelling approach to compare ADC deconjugation and systemic elimination rates of individual drug-load species using native ADC LC-MS data from human plasma. Xenobiotica 2024; 54:492-501. [PMID: 39329288 DOI: 10.1080/00498254.2024.2340741] [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: 02/19/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 09/28/2024]
Abstract
Native liquid chromatography mass spectrometry (LC-MS) is a commonly used approach for intact analysis of inter-chain cysteine conjugated antibody-drug conjugates (ADCs). Coupling native LC-MS with affinity capture provides a platform for intact ADC analysis from in vivo samples and characterisation of individual drug load species, specifically the impact of drug linker deconjugation, hydrolysis, and differential clearance in a biological system.This manuscript describes data generated from native LC-MS analysis of ADCs from human plasma, both in vitro incubations and clinical samples. It also details the pharmacokinetic (PK) model built to specifically characterise the disposition of individual drug load species from MMAE and MMAF interchain cysteine conjugated ADCs.In vitro deconjugation and hydrolysis rates were similar across both ADCs. Differential clearance of higher loaded species in vivo was pronounced for the MMAE conjugated ADC, while systemic elimination after accounting for deconjugation was similar across drug loads for the MMAF conjugated ADC. This is the first report of affinity capture native LC-MS analysis, and subsequent modelling of deconjugation, hydrolysis and clearance rates of individual drug load species using clinical data from cysteine conjugated ADCs.
Collapse
Affiliation(s)
- Shawna M Hengel
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| | | | - Hossam Kadry
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| | - Stephen C Alley
- Clinical Pharmacology and Translational Science, Pfizer Inc, Bothell, Washington, USA
| |
Collapse
|
9
|
Jeon JH, Woo Kim S, Kim YJ, Park JW, Eun Moon J, Beom Lee Y, Yu H, Lee GH, Jin SH, Jeong JH. Synthesis and evaluation of antibody-drug conjugates with high drug-to-antibody ratio using dimaleimide-DM1 as a linker- payload. Bioorg Chem 2024; 149:107504. [PMID: 38850783 DOI: 10.1016/j.bioorg.2024.107504] [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: 10/23/2023] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
The notable characteristics of recently emerged Antibody-Drug Conjugates (ADCs) encompass the targeting of Human Epidermal growth factor Receptor 2 (HER2) through monoclonal antibodies (mAbs) and a high ratio of drug to antibody (DAR). The achievements of Kadcyla® (T-DM1) and Enhertu® (T-Dxd) have demonstrated that HER2-targeting antibodies, such as trastuzumab, have shown to be competitive in terms of efficacy and price for development. Furthermore, with the arrival of T-Dxd and Trodelvy®, high-DAR (7-8) ADCs, which differ from the moderate DAR (3-4) ADCs that were formerly regarded as conventional, are being acknowledged for their worth. Following this trend of drug development, we endeavored to develop a high-DAR ADC using a straightforward approach involving the utilization of DM1, a highly potent substance, in combination with the widely recognized trastuzumab. To achieve a high DAR, DM1 was conjugated to reduced cysteine through the simple design and synthesis of various dimaleimide linkers with differing lengths. Using LC and MS analysis, we have demonstrated that our synthesis methodology is uncomplicated and efficacious, yielding trastuzumab-based ADCs that exhibit a remarkable degree of uniformity. These ADCs have been experimentally substantiated to exert an inhibitory effect on cancer cells in vitro, thus affirming their value as noteworthy additions to the realm of ADCs.
Collapse
Affiliation(s)
- Joo-Hyun Jeon
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Seo Woo Kim
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Yoon-Jung Kim
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jang-Woo Park
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jee Eun Moon
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Yong Beom Lee
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Hana Yu
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Geon-Ho Lee
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Sung-Ha Jin
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea.
| | - Jin-Hyun Jeong
- AbchemBio co., Ltd., D 111, Veritas Hall, Yonsei University, 85 Songdogqahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea; College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea.
| |
Collapse
|
10
|
Zong HF, Li X, Han L, Wang L, Liu JJ, Yue YL, Chen J, Ke Y, Jiang H, Xie YQ, Zhang BH, Zhu JW. A novel bispecific antibody drug conjugate targeting HER2 and HER3 with potent therapeutic efficacy against breast cancer. Acta Pharmacol Sin 2024; 45:1727-1739. [PMID: 38605180 PMCID: PMC11272928 DOI: 10.1038/s41401-024-01279-8] [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: 11/16/2023] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Antibody drug conjugate (ADC) therapy has become one of the most promising approaches in cancer immunotherapy. Bispecific targeting could enhance the efficacy and safety of ADC by improving its specificity, affinity and internalization. In this study we constructed a HER2/HER3-targeting bispecific ADC (BsADC) and characterized its physiochemical properties, target specificity and internalization in vitro, and assessed its anti-tumor activities in breast cancer cell lines and in animal models. The HER2/HER3-targeting BsADC had a drug to antibody ratio (DAR) of 2.89, displayed a high selectivity against the target JIMT-1 breast cancer cells in vitro, as well as a slightly higher level of internalization than HER2- or HER3-monospecific ADCs. More importantly, the bispecific ADC potently inhibited the viability of MCF7, JIMT-1, BT474, BxPC-3 and SKOV-3 cancer cells in vitro. In JIMT-1 breast cancer xenograft mice, a single injection of bispecific ADC (3 mg/kg, i.v.) significantly inhibited the tumor growth with an efficacy comparable to that caused by combined injection of HER2 and HER3-monospecific ADCs (3 mg/kg for each). Our study demonstrates that the bispecific ADC concept can be applied to development of more potent new cancer therapeutics than the monospecific ADCs.
Collapse
Affiliation(s)
- Hui-Fang Zong
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Xi Li
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Han
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Lei Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun-Jun Liu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ya-Li Yue
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jie Chen
- Jecho Institute Co., Ltd., Shanghai, 200240, China
| | - Yong Ke
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Jiang
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA
| | - Yue-Qing Xie
- Jecho Institute Co., Ltd., Shanghai, 200240, China
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA
| | - Bao-Hong Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian-Wei Zhu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education; School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Jecho Institute Co., Ltd., Shanghai, 200240, China.
- Jecho Laboratories, Inc., Frederick, MD, 21704, USA.
| |
Collapse
|
11
|
Medina Pérez VM, Baselga M, Schuhmacher AJ. Single-Domain Antibodies as Antibody-Drug Conjugates: From Promise to Practice-A Systematic Review. Cancers (Basel) 2024; 16:2681. [PMID: 39123409 PMCID: PMC11311928 DOI: 10.3390/cancers16152681] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) represent potent cancer therapies that deliver highly toxic drugs to tumor cells precisely, thus allowing for targeted treatment and significantly reducing off-target effects. Despite their effectiveness, ADCs can face limitations due to acquired resistance and potential side effects. OBJECTIVES This study focuses on advances in various ADC components to improve both the efficacy and safety of these agents, and includes the analysis of several novel ADC formats. This work assesses whether the unique features of VHHs-such as their small size, enhanced tissue penetration, stability, and cost-effectiveness-make them a viable alternative to conventional antibodies for ADCs and reviews their current status in ADC development. METHODS Following PRISMA guidelines, this study focused on VHHs as components of ADCs, examining advancements and prospects from 1 January 2014 to 30 June 2024. Searches were conducted in PubMed, Cochrane Library, ScienceDirect and LILACS using specific terms related to ADCs and single-domain antibodies. Retrieved articles were rigorously evaluated, excluding duplicates and non-qualifying studies. The selected peer-reviewed articles were analyzed for quality and synthesized to highlight advancements, methods, payloads, and future directions in ADC research. RESULTS VHHs offer significant advantages for drug conjugation over conventional antibodies due to their smaller size and structure, which enhance tissue penetration and enable access to previously inaccessible epitopes. Their superior stability, solubility, and manufacturability facilitate cost-effective production and expand the range of targetable antigens. Additionally, some VHHs can naturally cross the blood-brain barrier or be easily modified to favor their penetration, making them promising for targeting brain tumors and metastases. Although no VHH-drug conjugates (nADC or nanoADC) are currently in the clinical arena, preclinical studies have explored various conjugation methods and linkers. CONCLUSIONS While ADCs are transforming cancer treatment, their unique mechanisms and associated toxicities challenge traditional views on bioavailability and vary with different tumor types. Severe toxicities, often linked to compound instability, off-target effects, and nonspecific blood cell interactions, highlight the need for better understanding. Conversely, the rapid distribution, tumor penetration, and clearance of VHHs could be advantageous, potentially reducing toxicity by minimizing prolonged exposure. These attributes make single-domain antibodies strong candidates for the next generation of ADCs, potentially enhancing both efficacy and safety.
Collapse
Affiliation(s)
- Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Marta Baselga
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
12
|
Zhang B, Wang M, Sun L, Liu J, Yin L, Xia M, Zhang L, Liu X, Cheng Y. Recent Advances in Targeted Cancer Therapy: Are PDCs the Next Generation of ADCs? J Med Chem 2024; 67:11469-11487. [PMID: 38980167 DOI: 10.1021/acs.jmedchem.4c00106] [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: 07/10/2024]
Abstract
Antibody-drug conjugates (ADCs) comprise antibodies, cytotoxic payloads, and linkers, which can integrate the advantages of antibodies and small molecule drugs to achieve targeted cancer treatment. However, ADCs also have some shortcomings, such as non-negligible drug resistance, a low therapeutic index, and payload-related toxicity. Many studies have focused on changing the composition of ADCs, and some have even further extended the concept and types of targeted conjugated drugs by replacing the targeted antibodies in ADCs with peptides, revolutionarily introducing peptide-drug conjugates (PDCs). This Perspective summarizes the current research status of ADCs and PDCs and highlights the structural innovations of ADC components. In particular, PDCs are regarded as the next generation of potential targeted drugs after ADCs, and the current challenges of PDCs are analyzed. Our aim is to offer fresh insights for the efficient design and expedited development of innovative targeted conjugated drugs.
Collapse
Affiliation(s)
- Baochen Zhang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Mo Wang
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Li Sun
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, P.R. China
| | - Jiawei Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Libinghan Yin
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Mingjing Xia
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Ling Zhang
- School of Chemical Technology, Shijiazhuang University, Shijiazhuang 050035, P.R. China
| | - Xifu Liu
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| | - Yu Cheng
- Ministry of Education Key Laboratory of Molecular and Cellular Biology, Hebei Anti-Tumor Molecular Target Technology Innovation Center, Hebei Research Center of the Basic Discipline of Cell Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, P.R. China
| |
Collapse
|
13
|
Kwon WA, Lee SY, Jeong TY, Kim HH, Lee MK. Antibody-Drug Conjugates in Urothelial Cancer: From Scientific Rationale to Clinical Development. Cancers (Basel) 2024; 16:2420. [PMID: 39001482 PMCID: PMC11240765 DOI: 10.3390/cancers16132420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have been a significant advancement in cancer therapy, particularly for urothelial cancer (UC). These innovative treatments, originally developed for hematological malignancies, use target-specific monoclonal antibodies linked to potent cytotoxic agents. This rational drug design efficiently delivers cancer cell-killing agents to cells expressing specific surface proteins, which are abundant in UC owing to their high antigen expression. UC is an ideal candidate for ADC therapy, as it enhances on-target efficacy while mitigating systemic toxicity. In recent years, considerable progress has been made in understanding the biology and mechanisms of tumor progression in UC. However, despite the introduction of immune checkpoint inhibitors, advanced UC is characterized by rapid progression and poor survival rates. Targeted therapies that have been developed include the anti-nectin 4 ADC enfortumab vedotin and the fibroblast growth factor receptor inhibitor erdafitinib. Enfortumab vedotin has shown efficacy in prospective studies in patients with advanced UC, alone and in combination with pembrolizumab. The anti-Trop-2 ADC sacituzumab govitecan has also demonstrated effectiveness in single-armed studies. This review highlights the mechanism of action of ADCs, their application in mono- and combination therapies, primary mechanisms of resistance, and future perspectives for their clinical use in UC treatment. ADCs have proven to be an increasingly vital component of the therapeutic landscape for urothelial carcinoma, filling a gap in the treatment of this progressive disease.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Seo-Yeon Lee
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Tae Yoong Jeong
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Hyeon Hoe Kim
- Department of Urology, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Hanyang University College of Medicine, Myongji Hospital, Goyang 10475, Gyeonggi-do, Republic of Korea
| |
Collapse
|
14
|
Chis AA, Dobrea CM, Arseniu AM, Frum A, Rus LL, Cormos G, Georgescu C, Morgovan C, Butuca A, Gligor FG, Vonica-Tincu AL. Antibody-Drug Conjugates-Evolution and Perspectives. Int J Mol Sci 2024; 25:6969. [PMID: 39000079 PMCID: PMC11241239 DOI: 10.3390/ijms25136969] [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: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Antineoplastic therapy is one of the main research themes of this century. Modern approaches have been implemented to target and heighten the effect of cytostatic drugs on tumors and diminish their general/unspecific toxicity. In this context, antibody-drug conjugates (ADCs) represent a promising and successful strategy. The aim of this review was to assess different aspects regarding ADCs. They were presented from a chemical and a pharmacological perspective and aspects like structure, conjugation and development particularities alongside effects, clinical trials, safety issues and perspectives and challenges for future use of these drugs were discussed. Representative examples include but are not limited to the following main structural components of ADCs: monoclonal antibodies (trastuzumab, brentuximab), linkers (pH-sensitive, reduction-sensitive, peptide-based, phosphate-based, and others), and payloads (doxorubicin, emtansine, ravtansine, calicheamicin). Regarding pharmacotherapy success, the high effectiveness expectation associated with ADC treatment is supported by the large number of ongoing clinical trials. Major aspects such as development strategies are first discussed, advantages and disadvantages, safety and efficacy, offering a retrospective insight on the subject. The second part of the review is prospective, focusing on various plans to overcome the previously identified difficulties.
Collapse
Affiliation(s)
| | | | - Anca Maria Arseniu
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Adina Frum
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Luca-Liviu Rus
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Gabriela Cormos
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Cecilia Georgescu
- Faculty of Agriculture Science, Food Industry and Environmental Protection, "Lucian Blaga" University of Sibiu, 550012 Sibiu, Romania
| | - Claudiu Morgovan
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Anca Butuca
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | | | | |
Collapse
|
15
|
Saleh K, Khoury R, Khalife N, Chahine C, Ibrahim R, Tikriti Z, Le Cesne A. Mechanisms of action and resistance to anti-HER2 antibody-drug conjugates in breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:22. [PMID: 39050884 PMCID: PMC11267152 DOI: 10.20517/cdr.2024.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 07/27/2024]
Abstract
Human epidermal growth factor 2 (HER2)-positive breast cancer (BC) represents nearly 20% of all breast tumors. Historically, these patients had a high rate of relapse and dismal prognosis. The advent of HER2-targeting monoclonal antibodies such as trastuzumab followed by pertuzumab had improved the prognosis of HER2-positive metastatic BC. More recently, antibody-drug conjugates (ADCs) are now reshaping the treatment paradigm of solid tumors, especially breast cancer. Tratsuzumab emtansine (T-DM1) was one of the first ADC developed in oncology and was approved for the management of HER2-positive metastatic BC. In a head-to-head comparison, trastuzumab deruxtecan (T-DXd) defeated T-DM1 as a second-line treatment. The efficacy of ADCs is counterbalanced by the appearance of acquired resistance to these agents. In this paper, we summarize the mechanisms of action and resistance of T-DM1 and T-DXd, as well as their clinical efficacy. Additionally, we also discuss potential strategies for addressing resistance to ADC.
Collapse
Affiliation(s)
- Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Nadine Khalife
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Zamzam Tikriti
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, Villejuif 94800, France
| |
Collapse
|
16
|
Martín M, Pandiella A, Vargas-Castrillón E, Díaz-Rodríguez E, Iglesias-Hernangómez T, Martínez Cano C, Fernández-Cuesta I, Winkow E, Perelló MF. Trastuzumab deruxtecan in breast cancer. Crit Rev Oncol Hematol 2024; 198:104355. [PMID: 38621469 DOI: 10.1016/j.critrevonc.2024.104355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 02/06/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) consisting of a humanised, anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody covalently linked to a topoisomerase I inhibitor cytotoxic payload (DXd). The high drug-to-antibody ratio (8:1) ensures a high DXd concentration is delivered to target tumour cells, following internalisation of T-DXd and subsequent cleavage of its tetrapeptide-based linker. DXd's membrane-permeable nature enables it to cross cell membranes and potentially exert antitumour activity on surrounding tumour cells regardless of HER2 expression. T-DXd's unique mechanism of action is reflected in its efficacy in clinical trials in patients with HER2-positive advanced breast cancer (in heavily pretreated populations and in those previously treated with a taxane and trastuzumab), as well as HER2-low metastatic breast cancer. Thus, ADCs such as T-DXd have the potential to change the treatment paradigm of targeting HER2 in metastatic breast cancer, including eventually within the adjuvant/neoadjuvant setting.
Collapse
Affiliation(s)
- Miguel Martín
- Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Universidad Complutense, CIBERONC, Calle Doctor Esquerdo, 46, Madrid 28007, Spain.
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC-IBSAL and CIBERONC, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Emilio Vargas-Castrillón
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, S/N, Madrid 28040, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Plaza de Ramón y Cajal, s/n, Madrid 28040, Spain
| | - Elena Díaz-Rodríguez
- Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC-IBSAL and CIBERONC, Campus Miguel de Unamuno, Salamanca 37007, Spain
| | - Teresa Iglesias-Hernangómez
- Servicio de Farmacología Clínica, Hospital Clínico San Carlos, Calle del Prof Martín Lagos, S/N, Madrid 28040, Spain
| | - Concha Martínez Cano
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| | | | - Elena Winkow
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| | - Maria Francesca Perelló
- Daiichi Sankyo, Paseo Club Deportivo, 1, Edificio 14, Madrid, Pozuelo de Alarcón 28223, Spain
| |
Collapse
|
17
|
Journeaux T, Bernardes GJL. Homogeneous multi-payload antibody-drug conjugates. Nat Chem 2024; 16:854-870. [PMID: 38760431 DOI: 10.1038/s41557-024-01507-y] [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: 09/20/2023] [Accepted: 03/14/2024] [Indexed: 05/19/2024]
Abstract
Many systemic cancer chemotherapies comprise a combination of drugs, yet all clinically used antibody-drug conjugates (ADCs) contain a single-drug payload. These combination regimens improve treatment outcomes by producing synergistic anticancer effects and slowing the development of drug-resistant cell populations. In an attempt to replicate these regimens and improve the efficacy of targeted therapy, the field of ADCs has moved towards developing techniques that allow for multiple unique payloads to be attached to a single antibody molecule with high homogeneity. However, the methods for generating such constructs-homogeneous multi-payload ADCs-are both numerous and complex owing to the plethora of reactive functional groups that make up the surface of an antibody. Here, by summarizing and comparing the methods of both single- and multi-payload ADC generation and their key preclinical and clinical results, we provide a timely overview of this relatively new area of research. The methods discussed range from branched linker installation to the incorporation of unnatural amino acids, with a generalized comparison tool of the most promising modification strategies also provided. Finally, the successes and challenges of this rapidly growing field are critically evaluated, and from this, future areas of research and development are proposed.
Collapse
Affiliation(s)
- Toby Journeaux
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Gonçalo J L Bernardes
- Department of Chemistry, University of Cambridge, Cambridge, UK.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
18
|
Gu Y, Wang Z, Wang Y. Bispecific antibody drug conjugates: Making 1+1>2. Acta Pharm Sin B 2024; 14:1965-1986. [PMID: 38799638 PMCID: PMC11119582 DOI: 10.1016/j.apsb.2024.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 05/29/2024] Open
Abstract
Bispecific antibody‒drug conjugates (BsADCs) represent an innovative therapeutic category amalgamating the merits of antibody‒drug conjugates (ADCs) and bispecific antibodies (BsAbs). Positioned as the next-generation ADC approach, BsADCs hold promise for ameliorating extant clinical challenges associated with ADCs, particularly pertaining to issues such as poor internalization, off-target toxicity, and drug resistance. Presently, ten BsADCs are undergoing clinical trials, and initial findings underscore the imperative for ongoing refinement. This review initially delves into specific design considerations for BsADCs, encompassing target selection, antibody formats, and the linker-payload complex. Subsequent sections delineate the extant progress and challenges encountered by BsADCs, illustrated through pertinent case studies. The amalgamation of BsAbs with ADCs offers a prospective solution to prevailing clinical limitations of ADCs. Nevertheless, the symbiotic interplay among BsAb, linker, and payload necessitates further optimizations and coordination beyond a simplistic "1 + 1" to effectively surmount the extant challenges facing the BsADC domain.
Collapse
Affiliation(s)
- 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, China
| | - Zhijia 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, 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, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
19
|
Taylor KS, McMonagle MM, Guy SC, Human-McKinnon AM, Asamizu S, Fletcher HJ, Davis BW, Suyama TL. Albumin-ruthenium catalyst conjugate for bio-orthogonal uncaging of alloc group. Org Biomol Chem 2024; 22:2992-3000. [PMID: 38526322 DOI: 10.1039/d4ob00234b] [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] [Indexed: 03/26/2024]
Abstract
The employment of antibodies as a targeted drug delivery vehicle has proven successful which is exemplified by the emergence of antibody-drug conjugates (ADCs). However, ADCs are not without their shortcomings. Improvements may be made to the ADC platform by decoupling the cytotoxic drug from the delivery vehicle and conjugating an organometallic catalyst in its place. The resulting protein-metal catalyst conjugate was designed to uncage the masked cytotoxin administered as a separate entity. Macropinocytosis of albumin by cancerous cells suggests the potential of albumin acting as the tumor-targeting delivery vehicle. Herein reported are the first preparation and demonstration of ruthenium catalysts with cyclopentadienyl and quinoline-based ligands conjugated to albumin. The effective uncaging abilities were demonstrated on allyloxy carbamate (alloc)-protected rhodamine 110 and doxorubicin, providing a promising catalytic scaffold for the advancement of selective drug delivery methods in the future.
Collapse
Affiliation(s)
- Kimberly S Taylor
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Madison M McMonagle
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Schaelee C Guy
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Ariana M Human-McKinnon
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Shumpei Asamizu
- Engineering Biology Research Center, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Heidi J Fletcher
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Bradley W Davis
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| | - Takashi L Suyama
- Department of Chemistry and Forensic Science, Waynesburg University, 51 W College St, Waynesburg, PA 15370, USA.
| |
Collapse
|
20
|
Li M, Zhao X, Yu C, Wang L. Antibody-Drug Conjugate Overview: a State-of-the-art Manufacturing Process and Control Strategy. Pharm Res 2024; 41:419-440. [PMID: 38366236 DOI: 10.1007/s11095-023-03649-z] [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: 09/30/2023] [Accepted: 12/16/2023] [Indexed: 02/18/2024]
Abstract
Antibody-drug conjugates (ADCs) comprise an antibody, linker, and drug, which direct their highly potent small molecule drugs to target tumor cells via specific binding between the antibody and surface antigens. The antibody, linker, and drug should be properly designed or selected to achieve the desired efficacy while minimizing off-target toxicity. With a unique and complex structure, there is inherent heterogeneity introduced by product-related variations and the manufacturing process. Here this review primarily covers recent key advances in ADC history, clinical development status, molecule design, manufacturing processes, and quality control. The manufacturing process, especially the conjugation process, should be carefully developed, characterized, validated, and controlled throughout its lifecycle. Quality control is another key element to ensure product quality and patient safety. A patient-centric strategy has been well recognized and adopted by the pharmaceutical industry for therapeutic proteins, and has been successfully implemented for ADCs as well, to ensure that ADC products maintain their quality until the end of their shelf life. Deep product understanding and process knowledge defines attribute testing strategies (ATS). Quality by design (QbD) is a powerful approach for process and product development, and for defining an overall control strategy. Finally, we summarize the current challenges on ADC development and provide some perspectives that may help to give related directions and trigger more cross-functional research to surmount those challenges.
Collapse
Affiliation(s)
- Meng Li
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Xueyu Zhao
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Chuanfei Yu
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China
| | - Lan Wang
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, People's Republic of China.
| |
Collapse
|
21
|
Zhou L, Lu Y, Liu W, Wang S, Wang L, Zheng P, Zi G, Liu H, Liu W, Wei S. Drug conjugates for the treatment of lung cancer: from drug discovery to clinical practice. Exp Hematol Oncol 2024; 13:26. [PMID: 38429828 PMCID: PMC10908151 DOI: 10.1186/s40164-024-00493-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024] Open
Abstract
A drug conjugate consists of a cytotoxic drug bound via a linker to a targeted ligand, allowing the targeted delivery of the drug to one or more tumor sites. This approach simultaneously reduces drug toxicity and increases efficacy, with a powerful combination of efficient killing and precise targeting. Antibody‒drug conjugates (ADCs) are the best-known type of drug conjugate, combining the specificity of antibodies with the cytotoxicity of chemotherapeutic drugs to reduce adverse reactions by preferentially targeting the payload to the tumor. The structure of ADCs has also provided inspiration for the development of additional drug conjugates. In recent years, drug conjugates such as ADCs, peptide‒drug conjugates (PDCs) and radionuclide drug conjugates (RDCs) have been approved by the Food and Drug Administration (FDA). The scope and application of drug conjugates have been expanding, including combination therapy and precise drug delivery, and a variety of new conjugation technology concepts have emerged. Additionally, new conjugation technology-based drugs have been developed in industry. In addition to chemotherapy, targeted therapy and immunotherapy, drug conjugate therapy has undergone continuous development and made significant progress in treating lung cancer in recent years, offering a promising strategy for the treatment of this disease. In this review, we discuss recent advances in the use of drug conjugates for lung cancer treatment, including structure-based drug design, mechanisms of action, clinical trials, and side effects. Furthermore, challenges, potential approaches and future prospects are presented.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Liu
- Department of Geriatrics, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shanglong Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guisha Zi
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, National Health Commission (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Respiratory and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
22
|
Douez E, Allard-Vannier E, Amar IAM, Jolivet L, Boursin F, Maisonial-Besset A, Witkowski T, Chezal JM, Colas C, Letast S, Auvert E, Denevault-Sabourin C, Aubrey N, Joubert N. Branched pegylated linker-auristatin to control hydrophobicity for the production of homogeneous minibody-drug conjugate against HER2-positive breast cancer. J Control Release 2024; 366:567-584. [PMID: 38215985 DOI: 10.1016/j.jconrel.2024.01.012] [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: 07/21/2023] [Revised: 12/28/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Trastuzumab emtansine (Kadcyla®) was the first antibody-drug conjugate (ADC) approved by the Food and Drug Administration in 2013 against a solid tumor, and the first ADC to treat human epidermal growth factor receptor 2 positive (HER2+) breast cancer. However, this second generation ADC is burden by several limitations included heterogeneity, limited activity against heterogeneous tumor (regarding antigen expression) and suboptimal tumor penetration. To address this, different development strategies are oriented towards homogeneous conjugation, new drugs, optimized linkers and/or smaller antibody formats. To reach better developed next generation ADCs, a key parameter to consider is the management of the hydrophobicity associated with the linker-drug, increasing with and limiting the drug-to-antibody ratio (DAR) of the ADC. Here, an innovative branched pegylated linker was developed, to control the hydrophobicity of the monomethyl auristatin E (MMAE) and its cathepsin B-sensitive trigger. This branched pegylated linker-MMAE was then used for the efficient generation of internalizing homogeneous ADC of DAR 8 and minibody-drug conjugate of DAR 4, targeting HER2. Both immunoconjugates were then evaluated in vitro and in vivo on breast cancer models. Interestingly, this study highlighted that the minibody-MMAE conjugate of DAR 4 was the best immunoconjugate regarding in vitro cellular internalization and cytotoxicity, gamma imaging, ex vivo biodistribution profile in mice and efficient reduction of tumor size in vivo. These results are very promising and encourage us to explore further fragment-drug conjugate development.
Collapse
Affiliation(s)
- Emmanuel Douez
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; Pharmacy Department, Tours University Hospital, F-37200 Tours, France
| | - Emilie Allard-Vannier
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France.
| | | | - Louis Jolivet
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Fanny Boursin
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Aurélie Maisonial-Besset
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Tiffany Witkowski
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Jean-Michel Chezal
- Université Clermont Auvergne, Inserm, Imagerie Moléculaire et Stratégies Théranostiques, UMR 1240, F-63000 Clermont-Ferrand, France
| | - Cyril Colas
- UPR 4301 CBM, CNRS, University of Tours, University of Orléans, F-45071 Orléans, France; UMR 7311 ICOA, CNRS, University of Orléans, F-45067 Orléans, France
| | - Stéphanie Letast
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | - Etienne Auvert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France
| | | | - Nicolas Aubrey
- UMR 1282 ISP, INRAE, University of Tours, Team BioMAP, F-37200 Tours, France
| | - Nicolas Joubert
- UMR 1100 CEPR, INSERM, University of Tours, F-37200 Tours, France.
| |
Collapse
|
23
|
Cherifi F, Da Silva A, Martins-Branco D, Awada A, Nader-Marta G. Pharmacokinetics and pharmacodynamics of antibody-drug conjugates for the treatment of patients with breast cancer. Expert Opin Drug Metab Toxicol 2024; 20:45-59. [PMID: 38214896 DOI: 10.1080/17425255.2024.2302460] [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: 09/25/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
INTRODUCTION Currently three antibody-drug-conjugates (ADC) are approved by the European Medicines Agency (EMA) for treatment of breast cancer (BC) patient: trastuzumab-emtansine, trastuzumab-deruxtecan and sacituzumab-govitecan. ADC are composed of a monoclonal antibody (mAb) targeting a specific antigen, a cytotoxic payload and a linker. Pharmacokinetics (PK) and pharmacodynamics (PD) distinguish ADC from conventional chemotherapy and must be understood by clinicians. AREAS COVERED Our review delineates the PK/PD profiles of ADC approved for the treatment of BC with insight for future development. This is an expert opinion literature review based on the EMA's Assessment Reports, enriched by a comprehensive literature search performed on Medline in August 2023. EXPERT OPINION All three ADC distributions are described by a two-compartment structure: tissue and serum. Payload concentration peak is immediate but remains at low concentration. The distribution varied for all ADC only with body weight. mAb will be metabolised firstly by the saturable complex formation of ADC/Tumour-Receptor and secondly by binding of FcgRs in immune cells. They are all excreted in the bile and faeces with minimal urine elimination. Dose adjustments, apart from weight, are not recommended. Novel ADC are composed of cleavable linkers with various targets/payloads with the same PK/PD properties, but novel structures of ADC are in development.
Collapse
Affiliation(s)
- François Cherifi
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Breast Cancer Unit, CLCC François Baclesse, Institut Normand du Sein, Caen, France
| | - Angélique Da Silva
- Departments of Pharmacology and Medical Oncology, Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE, Caen, France
| | - Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Ahmad Awada
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| |
Collapse
|
24
|
Hingorani DV. An overview of site-specific methods for achieving antibody drug conjugates with homogenous drug to antibody ratio. Expert Opin Biol Ther 2024; 24:31-36. [PMID: 38247196 DOI: 10.1080/14712598.2024.2305266] [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: 09/20/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
INTRODUCTION Antibody drug conjugates (ADCs) have emerged as a potent tool in cancer treatment, where cytotoxic drugs are linked to antibodies targeting specific antigens. While conventional ADC synthesis methods have seen success as commercials therapeutics, there is a growing interest in next-generation ADCs, looking at homogeneity of the drug-to-antibody ratio. AREAS COVERED The article provides a high-level overview for achieving said homogeneity by site-directed conjugations via encompassing engineered amino acids, enzyme-mediated strategies, peptide sequences, affinity peptides, and beyond. As the field rapidly evolves with multiple ADCs in clinical trials and the advent of biosimilars, the article explores the benefits and challenges in both conventional and non-platform ADC technologies. EXPERT OPINION The choice of site selection approach must be based on multiple criteria as discussed in this report. Two ADCs made from conjugation to engineered cysteines have been approved by regulatory agencies which have contributed to the excitement in this space. For the others, though successful as proof-of-concept, the true test of merit will be determined as these technologies advance into the clinic. The promise of improving the therapeutics index and decreasing toxicities will continue to drive progress in this area.
Collapse
|
25
|
Li K, Xie G, Deng X, Zhang Y, Jia Z, Huang Z. Antibody-drug conjugates in urinary tumors: clinical application, challenge, and perspectives. Front Oncol 2023; 13:1259784. [PMID: 38173833 PMCID: PMC10761427 DOI: 10.3389/fonc.2023.1259784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Urinary tumors primarily consist of kidney, urothelial, and prostate malignancies, which pose significant treatment challenges, particularly in advanced stages. Antibody-drug conjugates (ADCs) have emerged as a promising therapeutic approach, combining monoclonal antibody specificity with cytotoxic chemotherapeutic payloads. This review highlights recent advancements, opportunities, and challenges in ADC application for urinary tumors. We discuss the FDA-approved ADCs and other novel ADCs under investigation, emphasizing their potential to improve patient outcomes. Furthermore, we explore strategies to address challenges, such as toxicity management, predictive biomarker identification, and resistance mechanisms. Additionally, we examine the integration of ADCs with other treatment modalities, including immune checkpoint inhibitors, targeted therapies, and radiation therapy. By addressing these challenges and exploring innovative approaches, the development of ADCs may significantly enhance therapeutic options and outcomes for patients with advanced urinary tumor.
Collapse
Affiliation(s)
- Keqiang Li
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Guoqing Xie
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiyue Deng
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Zhang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhankui Jia
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenlin Huang
- Department of Urology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
26
|
Sela T, Mansø M, Siegel M, Marban-Doran C, Ducret A, Niewöhner J, Ravn J, Martin RE, Sommer A, Lohmann S, Krippendorff BF, Ladefoged M, Indlekofer A, Quaiser T, Bueddefeld F, Koller E, Mohamed MY, Oelschlaegel T, Gothelf KV, Hofer K, Schumacher FF. Diligent Design Enables Antibody-ASO Conjugates with Optimal Pharmacokinetic Properties. Bioconjug Chem 2023; 34:2096-2111. [PMID: 37916986 DOI: 10.1021/acs.bioconjchem.3c00393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Antisense-oligonucleotides (ASOs) are a promising drug modality for the treatment of neurological disorders, but the currently established route of administration via intrathecal delivery is a major limitation to its broader clinical application. An attractive alternative is the conjugation of the ASO to an antibody that facilitates access to the central nervous system (CNS) after peripheral application and target engagement at the blood-brain barrier, followed by transcytosis. Here, we show that the diligent conjugate design of Brainshuttle-ASO conjugates is the key to generating promising delivery vehicles and thereby establishing design principles to create optimized molecules with drug-like properties. An innovative site-specific transglutaminase-based conjugation technology was chosen and optimized in a stepwise process to identify the best-suited conjugation site, tags, reaction conditions, and linker design. The overall conjugation performance was found to be specifically governed by the choice of buffer conditions and the structure of the linker. The combination of the peptide tags YRYRQ and RYESK was chosen, showing high conjugation fidelity. Elaborate conjugate analysis revealed that one leading differentiating factor was hydrophobicity. The increase of hydrophobicity by the ASO payload could be mitigated by the appropriate choice of conjugation site and the heavy chain position 297 proved to be the most optimal. Evaluating the properties of the linker suggested a short bicyclo[6.1.0]nonyne (BCN) unit as best suited with regards to conjugation performance and potency. Promising in vitro activity and in vivo pharmacokinetic behavior of optimized Brainshuttle-ASO conjugates, based on a microtubule-associated protein tau (MAPT) targeting oligonucleotide, suggest that such designs have the potential to serve as a blueprint for peripherally delivered ASO-based drugs for the CNS in the future.
Collapse
Affiliation(s)
- Tatjana Sela
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
- Department of Biochemistry, Ludwig-Maximilians-Universität, Munich 80539, Germany
| | - Mads Mansø
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Michel Siegel
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Céline Marban-Doran
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Axel Ducret
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Jens Niewöhner
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Jacob Ravn
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Rainer E Martin
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Annika Sommer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Sabine Lohmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Ben-Fillippo Krippendorff
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Mette Ladefoged
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd., Fremtidsvej 3, Hørsholm 2970, Denmark
| | - Annette Indlekofer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Tom Quaiser
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Florian Bueddefeld
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Erich Koller
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| | | | | | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Aarhus 8000, Central Denmark Region, Denmark
| | - Kerstin Hofer
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, Penzberg 82377, Germany
| | - Felix F Schumacher
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
27
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
28
|
Lee G, Iwase T, Matsumoto S, Nabil A, Ebara M. Development of Apoptotic-Cell-Inspired Antibody-Drug Conjugate for Effective Immune Modulation. Int J Mol Sci 2023; 24:16036. [PMID: 38003225 PMCID: PMC10671176 DOI: 10.3390/ijms242216036] [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: 09/12/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Apoptotic cells' phosphoserine (PS) groups have a significant immunosuppressive effect. They inhibit proinflammatory signals by interacting with various immune cells, including macrophages, dendritic cells, and CD4+ cells. Previously, we synthesized PS-group-immobilized polymers and verified their immunomodulatory effects. Despite its confirmed immunomodulatory potential, the PS group has not been considered as a payload for antibody-drug conjugates (ADCs) in a targeted anti-inflammatory approach. AIM We conducted this research to introduce an apoptotic-cell-inspired antibody-drug conjugate for effective immunomodulation. METHOD Poly(2-hydroxyethyl methacrylate-co-2-methacryloyloxyethyl phosphorylserine) (p(HEMA-co-MPS)) was synthesized as a payload using RAFT polymerization, and goat anti-mouse IgG was selected as a model antibody, which was conjugated with the synthesized p(HEMA-co-MPS) via 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide/N-Hydroxysuccinimide (EDC/NHS) reaction. The antibody-binding affinity, anti-inflammatory potential, and cytotoxicity measurements were evaluated. RESULTS We successfully synthesized ADCs with a significant anti-inflammatory effect and optimized the antibody-polymer ratio to achieve the highest antibody-binding affinity. CONCLUSION We successfully introduced p(HEMA-co-MPS) to IgG without decreasing the anti-inflammatory potential of the polymer while maintaining its targeting ability. We suggest that the antibody-polymer ratio be appropriately adjusted for effective therapy. In the future, this technology can be applied to therapeutic antibodies, such as Tocilizumab or Abatacept.
Collapse
Affiliation(s)
- Gyeongwoo Lee
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan (A.N.)
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Taishu Iwase
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan (A.N.)
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Shunsuke Matsumoto
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan (A.N.)
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
| | - Ahmed Nabil
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan (A.N.)
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Mitsuhiro Ebara
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan (A.N.)
- Graduate School of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
- Graduate School of Advanced Engineering, Tokyo University of Science, 6-3-1 Katsushika-ku, Shinjuku, Tokyo 125-8585, Japan
| |
Collapse
|
29
|
Song CH, Jeong M, In H, Kim JH, Lin CW, Han KH. Trends in the Development of Antibody-Drug Conjugates for Cancer Therapy. Antibodies (Basel) 2023; 12:72. [PMID: 37987250 PMCID: PMC10660735 DOI: 10.3390/antib12040072] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023] Open
Abstract
In cancer treatment, the first-generation, cytotoxic drugs, though effective against cancer cells, also harmed healthy ones. The second-generation targeted cancer cells precisely to inhibit their growth. Enter the third-generation, consisting of immuno-oncology drugs, designed to combat drug resistance and bolster the immune system's defenses. These advanced therapies operate by obstructing the uncontrolled growth and spread of cancer cells through the body, ultimately eliminating them effectively. Within the arsenal of cancer treatment, monoclonal antibodies offer several advantages, including inducing cancer cell apoptosis, precise targeting, prolonged presence in the body, and minimal side effects. A recent development in cancer therapy is Antibody-Drug Conjugates (ADCs), initially developed in the mid-20th century. The second generation of ADCs addressed this issue through innovative antibody modification techniques, such as DAR regulation, amino acid substitutions, incorporation of non-natural amino acids, and enzymatic drug attachment. Currently, a third generation of ADCs is in development. This study presents an overview of 12 available ADCs, reviews 71 recent research papers, and analyzes 128 clinical trial reports. The overarching objective is to gain insights into the prevailing trends in ADC research and development, with a particular focus on emerging frontiers like potential targets, linkers, and drug payloads within the realm of cancer treatment.
Collapse
Affiliation(s)
- Chi Hun Song
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Minchan Jeong
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Hyukmin In
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Ji Hoe Kim
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| | - Chih-Wei Lin
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 406, Taiwan;
| | - Kyung Ho Han
- Department of Biological Sciences and Biotechnology, Hannam University, Daejeon 34054, Republic of Korea; (C.H.S.); (M.J.); (H.I.); (J.H.K.)
| |
Collapse
|
30
|
Nguyen TD, Bordeau BM, Balthasar JP. Use of Payload Binding Selectivity Enhancers to Improve Therapeutic Index of Maytansinoid-Antibody-Drug Conjugates. Mol Cancer Ther 2023; 22:1332-1342. [PMID: 37493255 PMCID: PMC10811745 DOI: 10.1158/1535-7163.mct-22-0804] [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: 12/15/2022] [Revised: 05/03/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Systemic exposure to released cytotoxic payload contributes to the dose-limiting off-target toxicities of anticancer antibody-drug conjugates (ADC). In this work, we present an "inverse targeting" strategy to optimize the therapeutic selectivity of maytansinoid-conjugated ADCs. Several anti-maytansinoid sdAbs were generated via phage-display technology with binding IC50 values between 10 and 60 nmol/L. Co-incubation of DM4 with the anti-maytansinoid sdAbs shifted the IC50 value of DM4 up to 250-fold. Tolerability and efficacy of 7E7-DM4 ADC, an anti-CD123 DM4-conjugated ADC, were assessed in healthy and in tumor-bearing mice, with and without co-administration of an anti-DM4 sdAb. Co-administration with anti-DM4 sdAb reduced 7E7-DM4-induced weight loss, where the mean values of percentage weight loss at nadir for mice receiving ADC+saline and ADC+sdAb were 7.9% ± 3% and 3.8% ± 1.3% (P < 0.05). In tumor-bearing mice, co-administration of the anti-maytansinoid sdAb did not negatively affect the efficacy of 7E7-DM4 on tumor growth or survival following dosing of the ADC at 1 mg/kg (P = 0.49) or at 10 mg/kg (P = 0.9). Administration of 7E7-DM4 at 100 mg/kg led to dramatic weight loss, with 80% of treated mice succumbing to toxicity before the appearance of mortality relating to tumor growth in control mice. However, all mice receiving co-dosing of 100 mg/kg 7E7-DM4 with anti-DM4 sdAb were able to tolerate the treatment, which enabled reduction in tumor volume to undetectable levels and to dramatic improvements in survival. In summary, we have demonstrated the utility and feasibility of the application of anti-payload antibody fragments for inverse targeting to improve the selectivity and efficacy of anticancer ADC therapy.
Collapse
Affiliation(s)
- Toan D. Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| | - Brandon M. Bordeau
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214
| |
Collapse
|
31
|
Babbar R, Vanya, Bassi A, Arora R, Aggarwal A, Wal P, Dwivedi SK, Alolayan S, Gulati M, Vargas-De-La-Cruz C, Behl T, Ojha S. Understanding the promising role of antibody drug conjugates in breast and ovarian cancer. Heliyon 2023; 9:e21425. [PMID: 38027672 PMCID: PMC10660083 DOI: 10.1016/j.heliyon.2023.e21425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
A nascent category of anticancer therapeutic drugs called antibody-drug conjugates (ADCs) relate selectivity of aimed therapy using chemotherapeutic medicines with high cytotoxic power. Progressive linker technology led to the advancement of more efficacious and safer treatments. It offers neoteric as well as encouraging therapeutic strategies for treating cancer. ADCs selectively administer a medication by targeting antigens which are abundantly articulated on the membrane surface of tumor cells. Tumor-specific antigens are differently expressed in breast and ovarian cancers and can be utilized to direct ADCs. Compared to conventional chemotherapeutic drugs, this approach enables optimal tumor targeting while minimizing systemic damage. A cleavable linker improves the ADCs because it allows the toxic payload to be distributed to nearby cells that do not express the target protein, operating on assorted tumors with dissimilar cell aggregation. Presently fifteen ADCs are being studied in breast and ovarian carcinoma preclinically, and assortment of few have already undergone promising early-phase clinical trial testing. Furthermore, Phase I and II studies are investigating a wide variety of ADCs, and preliminary findings are encouraging. An expanding sum of ADCs will probably become feasible therapeutic choices as solo agents or in conjunction with chemotherapeutic agents. This review accentuates the most recent preclinical findings, pharmacodynamics, and upcoming applications of ADCs in breast and ovarian carcinoma.
Collapse
Affiliation(s)
- Ritchu Babbar
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Vanya
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Aarti Bassi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Ankur Aggarwal
- Institute of Pharmaceutical Sciences and Research, Gwalior, Madhya Pradesh, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, NH-19 Bhauti, Kanpur, Uttar Pradesh, India
| | | | - Salma Alolayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Buraidah, 51452, Kingdom of Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 1444411, India
- ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW, 20227, Australia
| | - Celia Vargas-De-La-Cruz
- Department of Pharmacology, Faculty of Pharmacy and Biochemistry, Bromatology and Toxicology, Universidad Nacional Mayor de San Marcos, Lima, 150001, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, 15001, Peru
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Punjab, 140306, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
32
|
Fu Z, Gao C, Wu T, Wang L, Li S, Zhang Y, Shi C. Peripheral neuropathy associated with monomethyl auristatin E-based antibody-drug conjugates. iScience 2023; 26:107778. [PMID: 37727735 PMCID: PMC10505985 DOI: 10.1016/j.isci.2023.107778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
Since the successful approval of gemtuzumab ozogamicin, antibody-drug conjugates (ADCs) have emerged as a pivotal category of targeted therapies for cancer. Among these ADCs, the use of monomethyl auristatin E (MMAE) as a payload is prevalent in the development of ADC drugs, which has significantly improved overall therapeutic efficacy against various malignancies. However, increasing clinical observations have raised concerns regarding the potential nervous system toxicity associated with MMAE-based ADCs. Specifically, a higher incidence of peripheral neuropathy has been reported in ADCs incorporating MMAE as payloads. Considering the increasing global use of MMAE-based ADCs, it is imperative to provide an inclusive overview of diagnostic and management strategies for this adverse event. In this review, we examine current information and what future research directions are required to better understand and manage this type of clinical challenge.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan 430000, China
| |
Collapse
|
33
|
Chang HL, Schwettmann B, McArthur HL, Chan IS. Antibody-drug conjugates in breast cancer: overcoming resistance and boosting immune response. J Clin Invest 2023; 133:e172156. [PMID: 37712425 PMCID: PMC10503805 DOI: 10.1172/jci172156] [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] [Indexed: 09/16/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as a revolutionary therapeutic class, combining the precise targeting ability of monoclonal antibodies with the potent cytotoxic effects of chemotherapeutics. Notably, ADCs have rapidly advanced in the field of breast cancer treatment. This innovative approach holds promise for strengthening the immune system through antibody-mediated cellular toxicity, tumor-specific immunity, and adaptive immune responses. However, the development of upfront and acquired resistance poses substantial challenges in maximizing the effectiveness of these therapeutics, necessitating a deeper understanding of the underlying mechanisms. These mechanisms of resistance include antigen loss, derangements in ADC internalization and recycling, drug clearance, and alterations in signaling pathways and the payload target. To overcome resistance, ongoing research and development efforts are focused on urgently identifying biomarkers, integrating immune therapy approaches, and designing novel cytotoxic payloads. This Review provides an overview of the mechanisms and clinical effectiveness of ADCs, and explores their unique immune-boosting function, while also highlighting the complex resistance mechanisms and safety challenges that must be addressed. A continued focus on how ADCs impact the tumor microenvironment will help to identify new payloads that can improve patient outcomes.
Collapse
Affiliation(s)
- Hannah L. Chang
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Blake Schwettmann
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Heather L. McArthur
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
| | - Isaac S. Chan
- Department of Internal Medicine, Division of Hematology and Oncology
- Harold C. Simmons Comprehensive Cancer Center, and
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
34
|
Aggarwal D, Yang J, Salam MA, Sengupta S, Al-Amin MY, Mustafa S, Khan MA, Huang X, Pawar JS. Antibody-drug conjugates: the paradigm shifts in the targeted cancer therapy. Front Immunol 2023; 14:1203073. [PMID: 37671162 PMCID: PMC10475555 DOI: 10.3389/fimmu.2023.1203073] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
Cancer is one of the deadliest diseases, causing million of deaths each year globally. Conventional anti-cancer therapies are non-targeted and have systemic toxicities limiting their versatile applications in many cancers. So, there is an unmet need for more specific therapeutic options that will be effective as well as free from toxicities. Antibody-drug conjugates (ADCs) are suitable alternatives with the right potential and improved therapeutic index for cancer therapy. The ADCs are highly precise new class of biopharmaceutical products that covalently linked a monoclonal antibody (mAb) (binds explicitly to a tumor-associated surface antigen) with a customized cytotoxic drug (kills cancer cells) and tied via a chemical linker (releases the drug). Due to its precise design, it brings about the target cell killing sparing the normal counterpart and free from the toxicities of conventional chemotherapy. It has never been so easy to develop potential ADCs for successful therapeutic usage. With relentless efforts, it took almost a century for scientists to advance the formula and design ADCs for its current clinical applications. Until now, several ADCs have passed successfully through preclinical and clinical trials and because of proven efficacy, a few are approved by the FDA to treat various cancer types. Even though ADCs posed some shortcomings like adverse effects and resistance at various stages of development, with continuous efforts most of these limitations are addressed and overcome to improve their efficacy. In this review, the basics of ADCs, physical and chemical properties, the evolution of design, limitations, and future potentials are discussed.
Collapse
Affiliation(s)
- Devesh Aggarwal
- Department of Chemistry, Purdue University, West Lafayette, IN, United States
| | - Jie Yang
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Md. Abdus Salam
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Malaysia
| | - Sagnik Sengupta
- Department of Chemistry, Purdue University, West Lafayette, IN, United States
| | - Md. Yusuf Al-Amin
- Department of Chemistry, Purdue University, West Lafayette, IN, United States
- Purdue University Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN, United States
| | - Saad Mustafa
- Deen Dayal Upadhyaya (DDU) Kaushal Kendra, Jamia Millia Islamia University, New Delhi, India
| | - Mohammad Aasif Khan
- Division of Hematology and Medical Oncology, Department of Medicine, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, United States
| | - Xun Huang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Medicine, Linyi University, Linyi, Shandong, China
| | - Jogendra Singh Pawar
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- The Ohio State University Comprehensive Cancer Center – Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, United States
| |
Collapse
|
35
|
Kondrashov A, Sapkota S, Sharma A, Riano I, Kurzrock R, Adashek JJ. Antibody-Drug Conjugates in Solid Tumor Oncology: An Effectiveness Payday with a Targeted Payload. Pharmaceutics 2023; 15:2160. [PMID: 37631374 PMCID: PMC10459723 DOI: 10.3390/pharmaceutics15082160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are at the forefront of the drug development revolution occurring in oncology. Formed from three main components-an antibody, a linker molecule, and a cytotoxic agent ("payload"), ADCs have the unique ability to deliver cytotoxic agents to cells expressing a specific antigen, a great leap forward from traditional chemotherapeutic approaches that cause widespread effects without specificity. A variety of payloads can be used, including most frequently microtubular inhibitors (auristatins and maytansinoids), as well as topoisomerase inhibitors and alkylating agents. Finally, linkers play a critical role in the ADCs' effect, as cleavable moieties that serve as linkers impact site-specific activation as well as bystander killing effects, an upshot that is especially important in solid tumors that often express a variety of antigens. While ADCs were initially used in hematologic malignancies, their utility has been demonstrated in multiple solid tumor malignancies, including breast, gastrointestinal, lung, cervical, ovarian, and urothelial cancers. Currently, six ADCs are FDA-approved for the treatment of solid tumors: ado-trastuzumab emtansine and trastuzumab deruxtecan, both anti-HER2; enfortumab-vedotin, targeting nectin-4; sacituzuzmab govitecan, targeting Trop2; tisotumab vedotin, targeting tissue factor; and mirvetuximab soravtansine, targeting folate receptor-alpha. Although they demonstrate utility and tolerable safety profiles, ADCs may become ineffective as tumor cells undergo evolution to avoid expressing the specific antigen being targeted. Furthermore, the current cost of ADCs can be limiting their reach. Here, we review the structure and functions of ADCs, as well as ongoing clinical investigations into novel ADCs and their potential as treatments of solid malignancies.
Collapse
Affiliation(s)
- Aleksei Kondrashov
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD 21229, USA; (A.K.); (S.S.)
| | - Surendra Sapkota
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD 21229, USA; (A.K.); (S.S.)
| | - Aditya Sharma
- Department of Internal Medicine, Dartmouth Health, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; (A.S.); (I.R.)
| | - Ivy Riano
- Department of Internal Medicine, Dartmouth Health, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; (A.S.); (I.R.)
- Division of Hematology and Oncology, Dartmouth Cancer Center, Lebanon, NH 03755, USA
| | - Razelle Kurzrock
- WIN Consortium, 94550 Paris, France;
- MCW Cancer Center, Milwaukee, WI 53226, USA
- Division of Oncology and Hematology, University of Nebraska, Omaha, NE 68198, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Jacob J. Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
36
|
Kanjilal P, Singh K, Das R, Matte J, Thayumanavan S. Antibody Polymer Conjugates (APCs) for Active Targeted Therapeutic Delivery. Biomacromolecules 2023; 24:3638-3646. [PMID: 37478281 PMCID: PMC11145409 DOI: 10.1021/acs.biomac.3c00385] [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] [Indexed: 07/23/2023]
Abstract
Antibody drug conjugates (ADCs) are poised to have an enormous impact on targeted nanomedicine, especially in many cancer pathologies. The reach of the current format of ADCs is limited by their low drug-to-antibody ratio (DAR) because of the associated physiochemical instabilities. Here, we design antibody polymer conjugates (APCs) as a modular strategy to utilize polymers to address ADC's shortcomings. We show here that conjugation of polymer-based therapeutic molecules to antibodies helps increase the DAR, owing to the hydrophilic comonomer in the polymer that helps in masking the increased hydrophobicity caused by high drug loading. We show that the platform exhibits cell targetability and selective cell killing in multiple cell lines expressing disease-relevant antigens, viz., HER2 and EGFR. The ability to use different functionalities in the drug as the handle for polymer attachment further demonstrates the platform nature of APCs. The findings here could serve as an alternative design strategy for the next generation of active targeted nanomedicine.
Collapse
|
37
|
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: 37] [Impact Index Per Article: 37.0] [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
|
38
|
Khoury R, Saleh K, Khalife N, Saleh M, Chahine C, Ibrahim R, Lecesne A. Mechanisms of Resistance to Antibody-Drug Conjugates. Int J Mol Sci 2023; 24:ijms24119674. [PMID: 37298631 DOI: 10.3390/ijms24119674] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
The treatment of cancer patients has dramatically changed over the past decades with the advent of monoclonal antibodies, immune-checkpoint inhibitors, bispecific antibodies, and innovative T-cell therapy. Antibody-drug conjugates (ADCs) have also revolutionized the treatment of cancer. Several ADCs have already been approved in hematology and clinical oncology, such as trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and sacituzumab govitecan (SG) for the treatment of metastatic breast cancer, and enfortumab vedotin (EV) for the treatment of urothelial carcinoma. The efficacy of ADCs is limited by the emergence of resistance due to different mechanisms, such as antigen-related resistance, failure of internalization, impaired lysosomal function, and other mechanisms. In this review, we summarize the clinical data that contributed to the approval of T-DM1, T-DXd, SG, and EV. We also discuss the different mechanisms of resistance to ADCs, as well as the ways to overcome this resistance, such as bispecific ADCs and the combination of ADCs with immune-checkpoint inhibitors or tyrosine-kinase inhibitors.
Collapse
Affiliation(s)
- Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Nadine Khalife
- Department of Head and Neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Mohamad Saleh
- Department of Hematology and Oncology, Lebanese American University Medical Center-Rizk Hopsital, Beirut 1100, Lebanon
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Axel Lecesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
39
|
Passaro A, Jänne PA, Peters S. Antibody-Drug Conjugates in Lung Cancer: Recent Advances and Implementing Strategies. J Clin Oncol 2023:JCO2300013. [PMID: 37224424 DOI: 10.1200/jco.23.00013] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 05/26/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are one of the fastest-growing oncology therapeutics, merging the cytotoxic effect of conjugated payload with the high specific ability and selectivity of monoclonal antibody targeted on a specific cancer cell membrane antigen. The main targets for ADC development are antigens commonly expressed by lung cancer cells, but not in normal tissues. They include human epidermal growth factor receptor 2, human epidermal growth factor receptor 3, trophoblast cell surface antigen 2, c-MET, carcinoembryonic antigen-related cell adhesion molecule 5, and B7-H3, each with one or more specific ADCs that showed encouraging results in the lung cancer field, more in non-small-cell lung cancer than in small-cell lung cancer histology. To date, multiple ADCs are under evaluation, alone or in combination with different molecules (eg, chemotherapy agents or immune checkpoint inhibitors), and the optimal strategy for selecting patients who may benefit from the treatment is evolving, including an improvement of biomarker understanding, involving markers of resistance or response to the payload, besides the antibody target. In this review, we discuss the available evidence and future perspectives on ADCs for lung cancer treatment, including a comprehensive discussion on structure-based drug design, mechanism of action, and resistance concepts. Data were summarized by specific target antigen, biology, efficacy, and safety, differing among ADCs according to the ADC payload and their pharmacokinetics and pharmacodynamics properties.
Collapse
Affiliation(s)
- Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne University, Lausanne, Switzerland
| |
Collapse
|
40
|
Wang S, Wang F, Wang L, Liu Z, Liu M, Li S, Wang Y, Sun X, Jiang J. Detection of antibody-conjugate payload in cynomolgus monkey serum by a high throughput capture LC-MS/MS bioanalysis method. J Pharm Biomed Anal 2023; 227:115069. [PMID: 36854219 DOI: 10.1016/j.jpba.2022.115069] [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: 10/07/2021] [Revised: 08/23/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
Antibody-drug conjugate (ADC) plays a vital role in oncology indications. The efficacy and toxicity of ADC generally depend on the concentration of the drugs in the body system, and physiologically-based pharmacokinetic (P.K.) is a quantitative tool to understand the drug concentration in the body. To characterize the whole drug carefully, sophisticated bioanalysis was required. ADC bioanalysis generally needs multiple analysis strategies, which can accurately quantify total antibody (TAb), antibody-drug conjugate (ADC), antibody-conjugate payload (ac-payload), and free-payload. In this work, we mainly described and validated a high throughput capture Liquid Chromatography tandem-Mass Spectrometry (LC-MS/MS) bioanalysis method to detect the concentrations of ac-payload (such as MMAE) in cynomolgus monkey serum. This method was allowed to determinate the Drug to Antibody Ratio (DAR), obtained by n of ac-payload/ n of TAb. In addition, the technique could significantly improve the throughput of the pre-coated antibody on a 96-well plate. Besides, this method had no interference or carryover in endogenous substances and showed linearity (R2 ≥0.99) in the concentration range within 15.6-2000.0 ng/mL. The inter-run accuracy ranged from 75.8 % to 120.0 %, and precision was within ≤ 20.0 %. Meanwhile, selectivity and the benchtop stability of the method were also validated. This optimization method was successfully applied to the change of average DAR in P.K. study.
Collapse
Affiliation(s)
- Shujuan Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China; Rongchang Industry College, Yantai 264003, Shandong, China
| | - Fengzhu Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China; School of Biological Sciences, University of California, Irvine, CA 92697, United States
| | - Ling Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Zhihao Liu
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Meiling Liu
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Shenjun Li
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Ying Wang
- Pharmaron (Beijing) Inc., Beijing 100176, China
| | | | - Jing Jiang
- Rongchang Industry College, Yantai 264003, Shandong, China; Department of Pharmacology, Binzhou Medical University, Yantai 264003, Shandong, China.
| |
Collapse
|
41
|
Cooper AJ, Heist RS. New Therapies on the Horizon. Hematol Oncol Clin North Am 2023; 37:623-658. [PMID: 37029036 DOI: 10.1016/j.hoc.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Although lung cancer treatment has been transformed by the advent of checkpoint inhibitor immunotherapies, there remains a high unmet need for new effective therapies for patients with progressive disease. Novel treatment strategies include combination therapies with currently available programmed death ligand 1 inhibitors, targeting alternative immune checkpoints, and the use of novel immunomodulatory therapies. In addition, antibody-drug conjugates offer great promise as potent management options. As these agents are further tested in clinical trials, we anticipate that more effective therapies for patients with lung cancer are integrated into regular clinical practice.
Collapse
|
42
|
Cao W, Maza JC, Chernyak N, Flygare JA, Krska SW, Toste FD, Francis MB. Modification of Cysteine-Substituted Antibodies Using Enzymatic Oxidative Coupling Reactions. Bioconjug Chem 2023; 34:510-517. [PMID: 36787347 DOI: 10.1021/acs.bioconjchem.2c00576] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Cysteines are routinely used as site-specific handles to synthesize antibody-drug conjugates for targeted immunotherapy applications. Michael additions between thiols and maleimides are some of the most common methods for modifying cysteines, but these functional groups can be difficult to prepare on scale, and the resulting linkages have been shown to be reversible under some physiological conditions. Here, we show that the enzyme tyrosinase, which oxidizes conveniently accessed phenols to afford reactive ortho-quinone intermediates, can be used to attach phenolic cargo to cysteines engineered on antibody surfaces. The resulting linkages between the thiols and ortho-quinones are shown to be more resistant than maleimides to reversion under physiological conditions. Using this approach, we construct antibody conjugates bearing cytotoxic payloads, which exhibit targeted cell killing, and further demonstrate this method for the attachment of a variety of cargo to antibodies, including fluorophores and oligonucleotides.
Collapse
Affiliation(s)
- Wendy Cao
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Johnathan C Maza
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Natalia Chernyak
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - John A Flygare
- Department of Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Shane W Krska
- Department of Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - F Dean Toste
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| | - Matthew B Francis
- Department of Chemistry, University of California, Berkeley, California 94720, United States
| |
Collapse
|
43
|
Hong Y, Nam SM, Moon A. Antibody-drug conjugates and bispecific antibodies targeting cancers: applications of click chemistry. Arch Pharm Res 2023; 46:131-148. [PMID: 36877356 DOI: 10.1007/s12272-023-01433-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/30/2023] [Indexed: 03/07/2023]
Abstract
Engineering approaches using antibody drug conjugates (ADCs) and bispecific antibodies (bsAbs) are designed to overcome the limitations of conventional chemotherapies and therapeutic antibodies such as drug resistance and non-specific toxicity. Cancer immunotherapies have been shown to be clinically successful with checkpoint blockade and chimeric antigen receptor T cell therapy; however, overactive immune systems still represent a major problem. Given the complexity of a tumor environment, it would be advantageous to have a strategy targeting two or more molecules. We highlight the necessity and importance of a multi-target platform strategy against cancer. Approximately 400 ADCs and over 200 bsAbs are currently being clinically developed for several indications, with promising signs of therapeutic activity. ADCs include antibodies that recognize tumor antigens, linkers that stably connect drugs, and powerful cytotoxic drugs, also known as payloads. ADCs have direct therapeutic effects by targeting cancers with a strong payload. Another type of drug that uses antibodies are bsAbs, targeting two antigens by linking to antigen recognition sites or bridging cytotoxic immune cells to tumor cells, resulting in cancer immunotherapy. Three bsAbs and one ADC have been approved for use by the FDA and the EMA in 2022. Among these, two of the bsAbs and the one ADC are used for cancers. We introduced that bsADC, a combination of ADC and bsAbs, has yet to be approved and several candidates are in the early stages of clinical development in this review. bsADCs technology helps increase the specificity of ADCs or the internalization and killing ability of bsAbs. We also briefly discuss the application of click chemistry in the efficient development of ADCs and bsAbs as a conjugation strategy. The present review summarizes the ADCs, bsAbs, and bsADCs that have been approved for anti-cancer or currently in development. These strategies selectively deliver drugs to malignant tumor cells and can be used as therapeutic approaches for various types of cancer.
Collapse
Affiliation(s)
- Yeji Hong
- College of Pharmacy, Duksung Innovative Drug Center, Duksung Women's University, Seoul, 01369, Korea
| | - Su-Min Nam
- College of Pharmacy, Duksung Innovative Drug Center, Duksung Women's University, Seoul, 01369, Korea
| | - Aree Moon
- College of Pharmacy, Duksung Innovative Drug Center, Duksung Women's University, Seoul, 01369, Korea.
| |
Collapse
|
44
|
Ancheta LR, Shramm PA, Bouajram R, Higgins D, Lappi DA. Streptavidin-Saporin: Converting Biotinylated Materials into Targeted Toxins. Toxins (Basel) 2023; 15:toxins15030181. [PMID: 36977072 PMCID: PMC10059012 DOI: 10.3390/toxins15030181] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/12/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
Streptavidin-Saporin can be considered a type of ‘secondary’ targeted toxin. The scientific community has taken advantage of this conjugate in clever and fruitful ways using many kinds of biotinylated targeting agents to send saporin into a cell selected for elimination. Saporin is a ribosome-inactivating protein that causes inhibition of protein synthesis and cell death when delivered inside a cell. Streptavidin-Saporin, mixed with biotinylated molecules to cell surface markers, results in powerful conjugates that are used both in vitro and in vivo for behavior and disease research. Streptavidin-Saporin harnesses the ‘Molecular Surgery’ capability of saporin, creating a modular arsenal of targeted toxins used in applications ranging from the screening of potential therapeutics to behavioral studies and animal models. The reagent has become a well-published and validated resource in academia and industry. The ease of use and diverse functionality of Streptavidin-Saporin continues to have a significant impact on the life science industry.
Collapse
|
45
|
Abelman RO, Wu B, Spring LM, Ellisen LW, Bardia A. Mechanisms of Resistance to Antibody-Drug Conjugates. Cancers (Basel) 2023; 15:cancers15041278. [PMID: 36831621 PMCID: PMC9954407 DOI: 10.3390/cancers15041278] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Antibody-drug conjugates (ADCs), with antibodies targeted against specific antigens linked to cytotoxic payloads, offer the opportunity for a more specific delivery of chemotherapy and other bioactive payloads to minimize side effects. First approved in the setting of HER2+ breast cancer, more recent ADCs have been developed for triple-negative breast cancer (TNBC) and, most recently, hormone receptor-positive (HR+) breast cancer. While antibody-drug conjugates have compared favorably against traditional chemotherapy in some settings, patients eventually progress on these therapies and require a change in treatment. Mechanisms to explain the resistance to ADCs are highly sought after, in hopes of developing next-line treatment options and expanding the therapeutic windows of existing therapies. These resistance mechanisms are categorized as follows: change in antigen expression, change in ADC processing and resistance, and efflux of the ADC payload. This paper reviews the recently published literature on these mechanisms as well as potential options to overcome these barriers.
Collapse
|
46
|
Holz E, Darwish M, Tesar DB, Shatz-Binder W. A Review of Protein- and Peptide-Based Chemical Conjugates: Past, Present, and Future. Pharmaceutics 2023; 15:600. [PMID: 36839922 PMCID: PMC9959917 DOI: 10.3390/pharmaceutics15020600] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Over the past few decades, the complexity of molecular entities being advanced for therapeutic purposes has continued to evolve. A main propellent fueling innovation is the perpetual mandate within the pharmaceutical industry to meet the needs of novel disease areas and/or delivery challenges. As new mechanisms of action are uncovered, and as our understanding of existing mechanisms grows, the properties that are required and/or leveraged to enable therapeutic development continue to expand. One rapidly evolving area of interest is that of chemically enhanced peptide and protein therapeutics. While a variety of conjugate molecules such as antibody-drug conjugates, peptide/protein-PEG conjugates, and protein conjugate vaccines are already well established, others, such as antibody-oligonucleotide conjugates and peptide/protein conjugates using non-PEG polymers, are newer to clinical development. This review will evaluate the current development landscape of protein-based chemical conjugates with special attention to considerations such as modulation of pharmacokinetics, safety/tolerability, and entry into difficult to access targets, as well as bioavailability. Furthermore, for the purpose of this review, the types of molecules discussed are divided into two categories: (1) therapeutics that are enhanced by protein or peptide bioconjugation, and (2) protein and peptide therapeutics that require chemical modifications. Overall, the breadth of novel peptide- or protein-based therapeutics moving through the pipeline each year supports a path forward for the pursuit of even more complex therapeutic strategies.
Collapse
Affiliation(s)
- Emily Holz
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
47
|
Abelman RO, Keenan JC, Ryan PK, Spring LM, Bardia A. Current and Emerging Role of Antibody-Drug Conjugates in HER2-Negative Breast Cancer. Hematol Oncol Clin North Am 2023; 37:151-167. [PMID: 36435607 DOI: 10.1016/j.hoc.2022.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Antibody-drug conjugates (ADCs) are rapidly evolving therapies that are uniquely able to deliver potent chemotherapy specifically to cancer cells while largely sparing normal cells. ADCs have 3 components: (1) antibody targeted to a tumor-involved antigen, (2) cytotoxic payload, and (3) linker that connects the cytotoxic agent to the antibody. Once the antibody binds the target on the cell surface, the ADC is incorporated into the cell via receptor-mediated endocytosis. Inside the cells, the linker is cleaved in the lysosome and the payload is then released intracellularly. This article will review ADCs in clinical development for HER2-negative metastatic breast cancer.
Collapse
Affiliation(s)
| | - Jennifer C Keenan
- Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Phoebe K Ryan
- Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Laura M Spring
- Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Aditya Bardia
- Breast Cancer Research, Harvard Medical School, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
48
|
Nguyen TD, Bordeau BM, Balthasar JP. Mechanisms of ADC Toxicity and Strategies to Increase ADC Tolerability. Cancers (Basel) 2023; 15:713. [PMID: 36765668 PMCID: PMC9913659 DOI: 10.3390/cancers15030713] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Anti-cancer antibody-drug conjugates (ADCs) aim to expand the therapeutic index of traditional chemotherapy by employing the targeting specificity of monoclonal antibodies (mAbs) to increase the efficiency of the delivery of potent cytotoxic agents to malignant cells. In the past three years, the number of ADCs approved by the Food and Drug Administration (FDA) has tripled. Although several ADCs have demonstrated sufficient efficacy and safety to warrant FDA approval, the clinical use of all ADCs leads to substantial toxicity in treated patients, and many ADCs have failed during clinical development due to their unacceptable toxicity profiles. Analysis of the clinical data has demonstrated that dose-limiting toxicities (DLTs) are often shared by different ADCs that deliver the same cytotoxic payload, independent of the antigen that is targeted and/or the type of cancer that is treated. DLTs are commonly associated with cells and tissues that do not express the targeted antigen (i.e., off-target toxicity), and often limit ADC dosage to levels below those required for optimal anti-cancer effects. In this manuscript, we review the fundamental mechanisms contributing to ADC toxicity, we summarize common ADC treatment-related adverse events, and we discuss several approaches to mitigating ADC toxicity.
Collapse
Affiliation(s)
| | | | - Joseph P. Balthasar
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
49
|
Yang Y, Wang S, Ma P, Jiang Y, Cheng K, Yu Y, Jiang N, Miao H, Tang Q, Liu F, Zha Y, Li N. Drug conjugate-based anticancer therapy - Current status and perspectives. Cancer Lett 2023; 552:215969. [PMID: 36279982 DOI: 10.1016/j.canlet.2022.215969] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
Drug conjugates are conjugates comprising a tumor-homing carrier tethered to a cytotoxic agent via a linker that are designed to deliver an ultra-toxic payload directly to the target cancer cells. This strategy has been successfully used to increase the therapeutic efficacy of cytotoxic agents and reduce their toxic side effects. Drug conjugates are being developed worldwide, with the potential to revolutionize current cancer treatment strategies. Antibody-drug conjugates (ADCs) have developed rapidly, and 14 of them have received market approval since the first approval event by the Food and Drug Administration in 2000. However, there are some limitations in the use of antibodies as carriers. Other classes of drug conjugates are emerging, such as targeted drugs conjugated with peptides (peptide-drug conjugates, PDCs) and polymers (polymer-drug conjugates, PolyDCs) with the remaining constructs similar to those of ADCs. These novel drug conjugates are gaining attention because they overcome the limitations of ADCs. This review summarizes the current state and advancements in knowledge regarding the design, constructs, and clinical efficacy of different drug conjugates.
Collapse
Affiliation(s)
- Yuqi Yang
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Shuhang Wang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiwen Ma
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yale Jiang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Yue Yu
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Jiang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Huilei Miao
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qiyu Tang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Funan Liu
- First Affiliated Hospital of China Medical University, Shenyang, 110002, China
| | - Yan Zha
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| | - Ning Li
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
50
|
Cheng Y, Yuan X, Tian Q, Huang X, Chen Y, Pu Y, Long H, Xu M, Ji Y, Xie J, Tan Y, Zhao X, Song H. Preclinical profiles of SKB264, a novel anti-TROP2 antibody conjugated to topoisomerase inhibitor, demonstrated promising antitumor efficacy compared to IMMU-132. Front Oncol 2022; 12:951589. [PMID: 36620535 PMCID: PMC9817100 DOI: 10.3389/fonc.2022.951589] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/29/2022] [Indexed: 12/25/2022] Open
Abstract
Purpose The aim of this study was to improve the intratumoral accumulation of an antibody-drug conjugate (ADC) and minimize its off-target toxicity, SKB264, a novel anti-trophoblast antigen 2 (TROP2) ADC that was developed using 2-methylsulfonyl pyrimidine as the linker to conjugate its payload (KL610023), a belotecan-derivative topoisomerase I inhibitor. The preclinical pharmacologic profiles of SKB264 were assessed in this study. Methods The in vitro and in vivo pharmacologic profiles of SKB264, including efficacy, pharmacokinetics-pharmacodynamics (PK-PD), safety, and tissue distribution, were investigated using TROP2-positive cell lines, cell-derived xenograft (CDX), patient-derived xenograft (PDX) models, and cynomolgus monkeys. Moreover, some profiles were compared with IMMU-132. Results In vitro, SKB264 and SKB264 monoclonal antibody (mAb) had similar internalization abilities and binding affinities to TROP2. After cellular internalization, KL610023 was released and inhibited tumor cell survival. In vivo, SKB264 significantly inhibited tumor growth in a dose-dependent manner in both CDX and PDX models. After SKB264 administration, the serum or plasma concentration/exposure of SKB264 (conjugated ADC, number of payload units ≥1), total antibody (Tab, unconjugated and conjugated mAb regardless of the number of the payload units), and KL610023 in cynomolgus monkeys increased proportionally with increasing dosage from 1 to 10 mg/kg. The linker stability of SKB264 was significantly enhanced as shown by prolonged payload half-life in vivo (SKB264 vs. IMMU-132, 56.3 h vs. 15.5 h). At the same dose, SKB264's exposure in tumor tissue was 4.6-fold higher than that of IMMU-132. Conclusions Compared with IMMU-132, the longer half-life of SKB264 had a stronger targeting effect and better antitumor activity, suggesting the better therapeutic potential of SKB264 for treating TROP2-positive tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xi Zhao
- Center of Translational Medicine, Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
| | - Hongmei Song
- Center of Translational Medicine, Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
| |
Collapse
|