1
|
Zheng Y, Xu R, Cheng H, Tai W. Mono-amino acid linkers enable highly potent small molecule-drug conjugates by conditional release. Mol Ther 2024; 32:1048-1060. [PMID: 38369752 PMCID: PMC11163218 DOI: 10.1016/j.ymthe.2024.02.020] [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: 09/15/2023] [Revised: 01/18/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
The endosome cleavable linkers have been widely employed by antibody-drug conjugates and small molecule-drug conjugates (SMDCs) to control the accurate release of payloads. An effective linker should provide stability in systemic circulation but efficient payload release at its targeted tumor sites. This conflicting requirement always leads to linker design with increasing structural complexity. Balance of the effectiveness and structural complexity presents a linker design challenge. Here, we explored the possibility of mono-amino acid as so far the simplest cleavable linker (X-linker) for SMDC-based auristatin delivery. Within a diverse set of X-linkers, the SMDCs differed widely in bioactivity, with one (Asn-linker) having significantly improved potency (IC50 = 0.1 nM) and fast response to endosomal cathepsin B cleavage. Notably, this SMDC, once grafted with effector protein fragment crystallizable (Fc), demonstrated a profound in vivo therapeutic effect in aspects of targetability, circulation half-life (t1/2 = 73 h), stability, and anti-tumor efficacy. On the basis of these results, we believe that this mono-amino acid linker, together with the new SMDC-Fc scaffold, has significant potential in targeted delivery application.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Ruolin Xu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Hong Cheng
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
2
|
Subhan MA, Torchilin VP. Advances in Targeted Therapy of Breast Cancer with Antibody-Drug Conjugate. Pharmaceutics 2023; 15:1242. [PMID: 37111727 PMCID: PMC10144345 DOI: 10.3390/pharmaceutics15041242] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a potential and promising therapy for a wide variety of cancers, including breast cancer. ADC-based drugs represent a rapidly growing field of breast cancer therapy. Various ADC drug therapies have progressed over the past decade and have generated diverse opportunities for designing of state-of-the-art ADCs. Clinical progress with ADCs for the targeted therapy of breast cancer have shown promise. Off-target toxicities and drug resistance to ADC-based therapy have hampered effective therapy development due to the intracellular mechanism of action and limited antigen expression on breast tumors. However, innovative non-internalizing ADCs targeting the tumor microenvironment (TME) component and extracellular payload delivery mechanisms have led to reduced drug resistance and enhanced ADC effectiveness. Novel ADC drugs may deliver potent cytotoxic agents to breast tumor cells with reduced off-target effects, which may overcome difficulties related to delivery efficiency and enhance the therapeutic efficacy of cytotoxic cancer drugs for breast cancer therapy. This review discusses the development of ADC-based targeted breast cancer therapy and the clinical translation of ADC drugs for breast cancer treatment.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, North Eastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, North Eastern University, Boston, MA 02115, USA
| |
Collapse
|
3
|
Marzullo P, Boiarska Z, Pérez-Peña H, Abel AC, Álvarez-Bernad B, Lucena-Agell D, Vasile F, Sironi M, Altmann KH, Prota AE, Díaz JF, Pieraccini S, Passarella D. Maytansinol Derivatives: Side Reactions as a Chance for New Tubulin Binders. Chemistry 2021; 28:e202103520. [PMID: 34788896 PMCID: PMC9299702 DOI: 10.1002/chem.202103520] [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: 09/28/2021] [Indexed: 11/07/2022]
Abstract
Maytansinol is a valuable precursor for the preparation of maytansine derivatives (known as maytansinoids). Inspired by the intriguing structure of the macrocycle and the success in targeted cancer therapy of the derivatives, we explored the maytansinol acylation reaction. As a result, we were able to obtain a series of derivatives with novel modifications of the maytansine scaffold. We characterized these molecules by docking studies, by a comprehensive biochemical evaluation, and by determination of their crystal structures in complex with tubulin. The results shed further light on the intriguing chemical behavior of maytansinoids and confirm the relevance of this peculiar scaffold in the scenario of tubulin binders.
Collapse
Affiliation(s)
- Paola Marzullo
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Zlata Boiarska
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Helena Pérez-Peña
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Anne-Catherine Abel
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Forschungsstrasse 111, 5232, Villigen PSI, Switzerland
| | - Beatriz Álvarez-Bernad
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Daniel Lucena-Agell
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Francesca Vasile
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Maurizio Sironi
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH, Zürich, Vladimir-Prelog Weg 4, HCI H405, 8093, Zürich, Switzerland
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Paul Scherrer Institute, Forschungsstrasse 111, 5232, Villigen PSI, Switzerland
| | - J Fernando Díaz
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Stefano Pieraccini
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| | - Daniele Passarella
- Department of Chemistry, Università degli Studi di Milano, Via Golgi 19, 20133, Milan, Italy
| |
Collapse
|
4
|
Tedeschini T, Campara B, Grigoletto A, Bellini M, Salvalaio M, Matsuno Y, Suzuki A, Yoshioka H, Pasut G. Polyethylene glycol-based linkers as hydrophilicity reservoir for antibody-drug conjugates. J Control Release 2021; 337:431-447. [PMID: 34329685 DOI: 10.1016/j.jconrel.2021.07.041] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 01/12/2023]
Abstract
Antibody-drug conjugates (ADCs) are an established therapeutic entity in which potent cytotoxic drugs are conjugated to a monoclonal antibody. In parallel with the great emphasis put on novel site-specific bioconjugation technologies, future advancements in this field also rely on exploring novel linker-drug architectures that improve the efficacy and stability of ADCs. In this context, the use of hydrophilic linkers represents a valid strategy to mask or reduce the inherent hydrophobicity of the most used cytotoxic drugs and positively impact the physical stability and in vivo performance of ADCs. Here, we describe the use of linkers containing monodisperse poly(ethylene glycol) (PEG) moieties for the construction of highly-loaded lysine-conjugated ADCs. The studied ADCs differ in the positioning of PEG (linear or pendant), the bonding type with the antibody (amide or carbamate), and the drug-to-antibody ratio (DAR). These ADCs were first evaluated for their stability in solution under thermal stress, showing that both the drug-linker-polymer design and the nature of the antibody-linker bonding are of great importance for their physical and chemical stability. Amide-coupled ADCs bearing two pendant 12-unit poly(ethylene glycol) chains within the drug-linker structure were the best performing conjugates, distancing themselves from the ADCs obtained with a conventional linear 24-unit PEG oligomer or the linker of Kadcyla®. The pharmacokinetic profiles of amide-linked ADCs, with a linear or pendant configuration of the PEG, were tested in mice in comparison to Kadcyla®. Total antibody pharmacokinetics paralleled the trends in aggregation tendency, with slower clearance rates for the ADCs based on the pendant drug-linker format. The above-mentioned findings have provided important clues on the drug-linker design and revealed that the positioning and configuration of a PEG unit have to be carefully tuned to achieve ADCs with improved stability and pharmacokinetics.
Collapse
Affiliation(s)
- T Tedeschini
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - B Campara
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - A Grigoletto
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - M Bellini
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - M Salvalaio
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - Y Matsuno
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - A Suzuki
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - H Yoshioka
- NOF CORPORATION, DDS Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - G Pasut
- University of Padova, Dept. Pharmaceutical Sciences, Via Marzolo 5, 35131 Padova, Italy.
| |
Collapse
|
5
|
Li XF, Liu CF, Rao GW. Monoclonal Antibodies, Small Molecule Inhibitors and Antibody-drug Conjugates as HER2 Inhibitors. Curr Med Chem 2021; 28:3339-3360. [PMID: 32900344 DOI: 10.2174/0929867327666200908112847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 11/22/2022]
Abstract
Overexpression of human epidermal growth factor receptor (HER)-2 is found in a variety of cancers, often portending poor clinical outcomes. Therefore, HER2 is an attractive target for treatment. This review describes the research progress of HER2 targeted inhibitors in recent years. Excellent reviews are available, so we focus on the development, mechanisms of action, and structure-activity relationships of different types of inhibitors, including monoclonal antibodies, small molecule inhibitors, and antibody-drug conjugates (ADCs). In addition, the differences among them are compared.
Collapse
Affiliation(s)
- Xiu-Fang Li
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chen-Fu Liu
- School of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Guo-Wu Rao
- College of Pharmaceutical Science, Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
6
|
Miller JT, Vitro CN, Fang S, Benjamin SR, Tumey LN. Enzyme-Agnostic Lysosomal Screen Identifies New Legumain-Cleavable ADC Linkers. Bioconjug Chem 2021; 32:842-858. [PMID: 33788548 DOI: 10.1021/acs.bioconjchem.1c00124] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Over the past two decades, antibody drug conjugates (ADCs) and small molecule drug conjugates (SMDCs) have widely employed valine-citruline and related cathepsin-cleavable linkers due to their stability in plasma and their rapid cleavage by lysosomal cathepsins. However, a number of recent studies have illustrated that these linkers are subject to cleavage by exogenous enzymes such as Ces1C and neutrophil elastase, thus resulting in off-target release of drug. As such, there is a need to diversify the portfolio of ADC linkers in order to overcome nonspecific drug release. Rather than targeting cathepsins, we began with an "enzyme agnostic" screen in which a panel of 75 peptide FRET pairs were screened for cleavage in lysosomal extracts and in plasma. Unexpectedly, a series of Asn-containing peptides emerged from this screen as being cleaved far more quickly than traditional ValCit-type linkers while retaining excellent stability in plasma. Catabolism studies demonstrated that these linkers were cleaved by legumain, an asparaginyl endopeptidase that is overexpressed in a variety of cancers and is known to be present in the lysosome. MMAE-containing ADCs that incorporated these new linkers were shown to exhibit highly potent and selective cytotoxicity, comparable to analogous ValCit ADCs. Importantly, the Asn-containing linkers were shown to be completely stable to human neutrophil elastase, an enzyme thought to be responsible for the neutropenia and thrombocytopenia associated with ValCitPABC-MMAE ADCs. The legumain-cleavable ADCs were shown to have excellent stability in both mouse and human serum, retaining >85% of the drug after 1 week of incubation. Moreover, the corresponding small molecule FRET pairs exhibited <10% cleavage after 18 h in mouse and human serum. On the basis of these results, we believe that these new linkers (AsnAsn in particular) have significant potential in both ADC and SMDC drug delivery applications.
Collapse
Affiliation(s)
- Jared T Miller
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, P.O. Box 6000, Binghamton, New York 13902, United States
| | - Caitlin N Vitro
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, P.O. Box 6000, Binghamton, New York 13902, United States
| | - Siteng Fang
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, P.O. Box 6000, Binghamton, New York 13902, United States
| | - Samantha R Benjamin
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, P.O. Box 6000, Binghamton, New York 13902, United States
| | - L Nathan Tumey
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, P.O. Box 6000, Binghamton, New York 13902, United States
| |
Collapse
|
7
|
Anderson TS, Wooster AL, La-Beck NM, Saha D, Lowe DB. Antibody-drug conjugates: an evolving approach for melanoma treatment. Melanoma Res 2021; 31:1-17. [PMID: 33165241 DOI: 10.1097/cmr.0000000000000702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melanoma continues to be an aggressive and deadly form of skin cancer while therapeutic options are continuously developing in an effort to provide long-term solutions for patients. Immunotherapeutic strategies incorporating antibody-drug conjugates (ADCs) have seen varied levels of success across tumor types and represent a promising approach for melanoma. This review will explore the successes of FDA-approved ADCs to date compared to the ongoing efforts of melanoma-targeting ADCs. The challenges and opportunities for future therapeutic development are also examined to distinguish how ADCs may better impact individuals with malignancies such as melanoma.
Collapse
Affiliation(s)
| | | | - Ninh M La-Beck
- Departments of Immunotherapeutics and Biotechnology
- Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas, USA
| | | | - Devin B Lowe
- Departments of Immunotherapeutics and Biotechnology
| |
Collapse
|
8
|
Joubert N, Beck A, Dumontet C, Denevault-Sabourin C. Antibody-Drug Conjugates: The Last Decade. Pharmaceuticals (Basel) 2020; 13:ph13090245. [PMID: 32937862 PMCID: PMC7558467 DOI: 10.3390/ph13090245] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 01/01/2023] Open
Abstract
An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of.
Collapse
Affiliation(s)
- Nicolas Joubert
- GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France;
- Correspondence:
| | - Alain Beck
- Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France;
| | - Charles Dumontet
- Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France;
- Hospices Civils de Lyon, 69000 Lyon, France
| | - Caroline Denevault-Sabourin
- GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France;
| |
Collapse
|
9
|
Staben LR, Chen J, Cruz-Chuh JD, Del Rosario G, Go MA, Guo J, Khojasteh SC, Kozak KR, Li G, Ng C, Lewis Phillips GD, Pillow TH, Rowntree RK, Wai J, Wei B, Xu K, Xu Z, Yu SF, Zhang D, Dragovich PS. Systematic Variation of Pyrrolobenzodiazepine (PBD)-Dimer Payload Physicochemical Properties Impacts Efficacy and Tolerability of the Corresponding Antibody-Drug Conjugates. J Med Chem 2020; 63:9603-9622. [PMID: 32787101 DOI: 10.1021/acs.jmedchem.0c00691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytotoxic pyrrolobenzodiazepine (PBD)-dimer molecules are frequently utilized as payloads for antibody-drug conjugates (ADCs), and many examples are currently in clinical development. In order to further explore this ADC payload class, the physicochemical properties of various PBD-dimer molecules were modified by the systematic introduction of acidic and basic moieties into their chemical structures. The impact of these changes on DNA binding, cell membrane permeability, and in vitro antiproliferation potency was, respectively, determined using a DNA alkylation assay, PAMPA assessments, and cell-based cytotoxicity measurements conducted with a variety of cancer lines. The modified PBD-dimer compounds were subsequently incorporated into CD22-targeting ADCs, and these entities were profiled in a variety of in vitro and in vivo experiments. The introduction of a strongly basic moiety into the PBD-dimer scaffold afforded a conjugate with dramatically worsened mouse tolerability properties relative to ADCs derived from related payloads, which lacked the basic group.
Collapse
Affiliation(s)
- Leanna R Staben
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jinhua Chen
- WuXi AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | | | - Geoff Del Rosario
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Mary Ann Go
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jun Guo
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - S Cyrus Khojasteh
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Katherine R Kozak
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Guangmin Li
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Carl Ng
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Thomas H Pillow
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rebecca K Rowntree
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - BinQing Wei
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Keyang Xu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Zijin Xu
- WuXi AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Shang-Fan Yu
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Donglu Zhang
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Peter S Dragovich
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
10
|
Zhao P, Zhang Y, Li W, Jeanty C, Xiang G, Dong Y. Recent advances of antibody drug conjugates for clinical applications. Acta Pharm Sin B 2020; 10:1589-1600. [PMID: 33088681 PMCID: PMC7564033 DOI: 10.1016/j.apsb.2020.04.012] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/25/2020] [Accepted: 03/08/2020] [Indexed: 02/06/2023] Open
Abstract
Antibody drug conjugates (ADCs) normally compose of a humanized antibody and small molecular drug via a chemical linker. After decades of preclinical and clinical studies, a series of ADCs have been widely used for treating specific tumor types in the clinic such as brentuximab vedotin (Adcetris®) for relapsed Hodgkin's lymphoma and systemic anaplastic large cell lymphoma, gemtuzumab ozogamicin (Mylotarg®) for acute myeloid leukemia, ado-trastuzumab emtansine (Kadcyla®) for HER2-positive metastatic breast cancer, inotuzumab ozogamicin (Besponsa®) and most recently polatuzumab vedotin-piiq (Polivy®) for B cell malignancies. More than eighty ADCs have been investigated in different clinical stages from approximately six hundred clinical trials to date. This review summarizes the key elements of ADCs and highlights recent advances of ADCs, as well as important lessons learned from clinical data, and future directions.
Collapse
|
11
|
Garofalo M, Grazioso G, Cavalli A, Sgrignani J. How Computational Chemistry and Drug Delivery Techniques Can Support the Development of New Anticancer Drugs. Molecules 2020; 25:E1756. [PMID: 32290224 PMCID: PMC7180704 DOI: 10.3390/molecules25071756] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 01/17/2023] Open
Abstract
The early and late development of new anticancer drugs, small molecules or peptides can be slowed down by some issues such as poor selectivity for the target or poor ADME properties. Computer-aided drug design (CADD) and target drug delivery (TDD) techniques, although apparently far from each other, are two research fields that can give a significant contribution to overcome these problems. Their combination may provide mechanistic understanding resulting in a synergy that makes possible the rational design of novel anticancer based therapies. Herein, we aim to discuss selected applications, some also from our research experience, in the fields of anticancer small organic drugs and peptides.
Collapse
Affiliation(s)
- Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Giovanni Grazioso
- Department of Pharmaceutical Sciences, University of Milano, 20133 Milan, Italy
| | - Andrea Cavalli
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| | - Jacopo Sgrignani
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), 6500 Bellinzona, Switzerland
| |
Collapse
|
12
|
Beck A, Dumontet C, Joubert N. [Antibody-drug conjugates in oncology. New strategies in development]. Med Sci (Paris) 2020; 35:1043-1053. [PMID: 31903916 DOI: 10.1051/medsci/2019228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An Antibody-Drug Conjugate (armed antibody) is a vectorized chemotherapy that results from the grafting of a cytotoxic agent on a monoclonal antibody via a judiciously designed spacer arm. ADCs have made considerable progress in 10 years. In 2009, only gemtuzumab ozogamicin (Mylotarg®) was used clinically. In 2019, 4 other ADCs have been approved and more than 80 others are in active clinical trials. The second part of this review will focus on new emerging strategies to address ADCs drawbacks and attempt to broaden their therapeutic window. Finally, combinations with conventional chemotherapy or checkpoint inhibitors will be discussed, in the pursuit to make Antibody-Drug Conjugates the embodiment of Paul Ehrlich's dream of the magic bullet.
Collapse
Affiliation(s)
- Alain Beck
- Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien-en-Genevois, France
| | - Charles Dumontet
- Cancer Research Center of Lyon (CRCL), Inserm 1052/CNRS, 69000 Lyon, France - Université de Lyon, 69000 Lyon, France - Hospices Civils de Lyon, 69000 Lyon, France
| | - Nicolas Joubert
- GICC EA7501, Université de Tours, équipe IMT, 31 avenue Monge, 37200 Tours, France
| |
Collapse
|
13
|
Scales SJ, Tsai SP, Zacharias N, Cruz-Chuh JD, Bullen G, Velasquez E, Chang J, Bruguera E, Kozak KR, Sadowsky J. Development of a Cysteine-Conjugatable Disulfide FRET Probe: Influence of Charge on Linker Cleavage and Payload Trafficking for an Anti-HER2 Antibody Conjugate. Bioconjug Chem 2019; 30:3046-3056. [PMID: 31726009 DOI: 10.1021/acs.bioconjchem.9b00678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Disulfide-linked bioconjugates allow the delivery of pharmacologically active or other cargo to specific tissues in a redox-sensitive fashion. However, an understanding of the kinetics, subcellular distribution, and mechanism of disulfide cleavage in such bioconjugates is generally lacking. Here, we report a modular disulfide-linked TAMRA-BODIPY based FRET probe that can be readily synthesized, modified, and conjugated to a cysteine-containing biomolecule to enable real-time monitoring of disulfide cleavage during receptor-mediated endocytosis in cells. We demonstrate the utility of this probe to study disulfide reduction during HER2 receptor-mediated uptake of a Cys-engineered anti-HER2 THIOMAB antibody. We found that introduction of positive, but not negative, charges in the probe improved retention of the BODIPY catabolite. This permitted the observation of significant disulfide cleavage in endosomes or lysosomes on par with proteolytic cleavage of a similarly charged valine-citrulline peptide-based probe. In general, the FRET probe we describe should enable real-time cellular monitoring of disulfide cleavage in other targeted delivery systems for mechanistic or diagnostic applications. Furthermore, modifications to the released BODIPY moiety permit evaluation of physicochemical properties that govern lysosomal egress or retention, which may have implications for the development of next-generation antibody-drug conjugates.
Collapse
Affiliation(s)
- Suzie J Scales
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Siao Ping Tsai
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Neelie Zacharias
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Josefa Dela Cruz-Chuh
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Gordy Bullen
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Erick Velasquez
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Julie Chang
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Elise Bruguera
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Katherine R Kozak
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Jack Sadowsky
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| |
Collapse
|
14
|
Burton JK, Bottino D, Secomb TW. A Systems Pharmacology Model for Drug Delivery to Solid Tumors by Antibody-Drug Conjugates: Implications for Bystander Effects. AAPS JOURNAL 2019; 22:12. [PMID: 31828446 DOI: 10.1208/s12248-019-0390-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 10/30/2019] [Indexed: 01/08/2023]
Abstract
Antibody-drug conjugates (ADCs) are cancer drugs composed of a humanized antibody linked to a cytotoxic payload, allowing preferential release of payload in cancer cells expressing the antibody-targeted antigen. Here, a systems pharmacology model is used to simulate ADC transport from blood to tumor tissue and ADC uptake by tumor cells. The model includes effects of spatial gradients in drug concentration in a three-dimensional network of tumor blood vessels with realistic geometry and accounts for diffusion of ADC in the tumor extracellular space, binding to antigen, internalization, intracellular processing, and payload efflux from cells. Cells that process an internalized ADC-antigen complex may release payload that can be taken up by other "bystander" cells. Such bystander effects are included in the model. The model is used to simulate conditions in previous experiments, showing good agreement with experimental results. Simulations are used to analyze the relationship of bystander effects to payload properties and single-dose administrations. The model indicates that exposure of payload to cells distant from vessels is sensitive to the free payload diffusivity in the extracellular space. When antigen expression is heterogeneous, the model indicates that the amount of payload accumulating in non-antigen-expressing cells increases linearly with dose but depends only weakly on the percentage of antigen-expressing cells. The model provides an integrated mechanistic framework for understanding the effects of spatial gradients on drug distribution using ADCs and for designing ADCs to achieve more effective payload distribution in solid tumors, thereby increasing the therapeutic index of the ADC.
Collapse
Affiliation(s)
| | - Dean Bottino
- Takeda Pharmaceuticals, Cambridge, Massachusetts, USA
| | - Timothy W Secomb
- Program in Applied Mathematics, University of Arizona, Tucson, Arizona, USA. .,Department of Physiology, University of Arizona, Tucson, Arizona, 85724-5051, USA.
| |
Collapse
|
15
|
Salomon PL, Reid EE, Archer KE, Harris L, Maloney EK, Wilhelm AJ, Miller ML, Chari RVJ, Keating TA, Singh R. Optimizing Lysosomal Activation of Antibody–Drug Conjugates (ADCs) by Incorporation of Novel Cleavable Dipeptide Linkers. Mol Pharm 2019; 16:4817-4825. [DOI: 10.1021/acs.molpharmaceut.9b00696] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Paulin L. Salomon
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Emily E. Reid
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Katie E. Archer
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Luke Harris
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Alan J. Wilhelm
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Michael L. Miller
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Thomas A. Keating
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rajeeva Singh
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
16
|
Costoplus JA, Veale KH, Qiu Q, Ponte JF, Lanieri L, Setiady Y, Dong L, Skaletskaya A, Bartle LM, Salomon P, Wu R, Maloney EK, Kovtun YV, Ab O, Lai K, Chari RVJ, Widdison WC. Peptide-Cleavable Self-immolative Maytansinoid Antibody-Drug Conjugates Designed To Provide Improved Bystander Killing. ACS Med Chem Lett 2019; 10:1393-1399. [PMID: 31620224 PMCID: PMC6792174 DOI: 10.1021/acsmedchemlett.9b00310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/27/2019] [Indexed: 11/29/2022] Open
Abstract
![]()
A new type of antibody–drug
conjugate (ADC) has been prepared that contains a sulfur-bearing maytansinoid
attached to an antibody via a highly stable tripeptide linker. Once
internalized by cells, proteases in catabolic vesicles cleave the
peptide of the ADC’s linker causing self-immolation that releases
a thiol-bearing metabolite, which is then S-methylated.
Conjugates were prepared with peptide linkers containing only alanyl
residues, which were all l isomers or had a single d residue in one of the three positions. A d-alanyl residue
in the linker did not significantly impair a conjugate’s cytotoxicity
or bystander killing unless it was directly attached to the immolative
moiety. Increasing the number of methylene units in the maytansinoid
side chain of a conjugate did not typically affect an ADC’s
cytotoxicity to targeted cells but did increase bystander killing
activity. ADCs with the highest in vitro bystander
killing were then evaluated in vivo in mice, where
they displayed improved efficacy compared to previously described
types of maytansinoid conjugates.
Collapse
Affiliation(s)
- Juliet A. Costoplus
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Karen H. Veale
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Qifeng Qiu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jose F. Ponte
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Leanne Lanieri
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yulius Setiady
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ling Dong
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Anna Skaletskaya
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Laura M. Bartle
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Paulin Salomon
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rui Wu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yelena V. Kovtun
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Olga Ab
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Kate Lai
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Wayne C. Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
17
|
Li W, Veale KH, Qiu Q, Sinkevicius KW, Maloney EK, Costoplus JA, Lau J, Evans HL, Setiady Y, Ab O, Abbott SM, Lee J, Wisitpitthaya S, Skaletskaya A, Wang L, Keating TA, Chari RVJ, Widdison WC. Synthesis and Evaluation of Camptothecin Antibody-Drug Conjugates. ACS Med Chem Lett 2019; 10:1386-1392. [PMID: 31620223 PMCID: PMC6792168 DOI: 10.1021/acsmedchemlett.9b00301] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/06/2019] [Indexed: 01/23/2023] Open
Abstract
Antibody-drug conjugates (ADCs) that incorporate the exatecan derivative DXd in their payload are showing promising clinical results in solid tumor indications. The payload has an F-ring that also contains a second chiral center, both of which complicate its synthesis and derivatization. Here we report on new camptothecin-ADCs that do not have an F-ring in their payloads yet behave similarly to DXd-bearing conjugates in vitro and in vivo. This simplification allows easier derivatization of camptothecin A and B rings for structure-activity relationship studies and payload optimization. ADCs having different degrees of bystander killing and the ability to release hydroxyl or thiol-bearing metabolites following peptide linker cleavage were investigated.
Collapse
Affiliation(s)
- Wei Li
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Karen H. Veale
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Qifeng Qiu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | | | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Juliet A. Costoplus
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Janet Lau
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Helen L. Evans
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Yulius Setiady
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Olga Ab
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Stephen M. Abbott
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Jenny Lee
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | | | - Anna Skaletskaya
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Lintao Wang
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Thomas A. Keating
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Wayne C. Widdison
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
18
|
Abstract
The concept of exploiting the specific binding properties of monoclonal antibodies as a mechanism for selective delivery of cytotoxic agents to tumor cells is an attractive solution to the challenge of increasing the therapeutic index of cell-killing agents for treating cancer. All three parts of an antibody-drug conjugate (ADC)-the antibody, the cytotoxic payload, and the linker chemistry that joins them together-as well as the biologic properties of the cell-surface target antigen are important in designing an effective anticancer agent. The approval of brentuximab vedotin in 2011 for treating relapsed Hodgkin's lymphoma and systemic anaplastic large cell lymphoma, and the approval of ado-trastuzumab emtansine in 2013 for treating HER2-positive metastatic breast cancer, have sparked vigorous research in the field, with >65 ADCs currently in clinical evaluation. This review highlights the ADCs that are approved for marketing, in pivotal clinical trials, or in at least phase II clinical development for treating both hematologic malignancies and solid tumors.
Collapse
|
19
|
Reid EE, Archer KE, Shizuka M, Wilhelm A, Yoder NC, Bai C, Fishkin NE, Harris L, Maloney EK, Salomon P, Hong E, Wu R, Ab O, Jin S, Lai KC, Sikka S, Chari RVJ, Miller ML. Effect of Linker Stereochemistry on the Activity of Indolinobenzodiazepine Containing Antibody-Drug Conjugates (ADCs). ACS Med Chem Lett 2019; 10:1193-1197. [PMID: 31413805 DOI: 10.1021/acsmedchemlett.9b00240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/10/2019] [Indexed: 01/09/2023] Open
Abstract
Antibody-drug conjugates (ADCs) that incorporate potent indolinobenzodiazepine DNA alkylators as the payload component are currently undergoing clinical evaluation. In one ADC design, the payload molecules are linked to the antibody through a peptidase-labile l-Ala-l-Ala linker. In order to determine the role of amino acid stereochemistry on antitumor activity and tolerability, we incorporated l- and d-alanyl groups in the dipeptide, synthesized all four diastereomers, and prepared and tested the corresponding ADCs. Results of our preclinical evaluation showed that the l-Ala-l-Ala configuration provided the ADC with the highest therapeutic index (antitumor activity vs toxicity).
Collapse
Affiliation(s)
- Emily E. Reid
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Katie E. Archer
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Manami Shizuka
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Alan Wilhelm
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Nicholas C. Yoder
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Chen Bai
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Nathan E. Fishkin
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Luke Harris
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Erin K. Maloney
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Paulin Salomon
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Erica Hong
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Rui Wu
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Olga Ab
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Shan Jin
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Katharine C. Lai
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Surina Sikka
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Ravi V. J. Chari
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| | - Michael L. Miller
- ImmunoGen, Inc., 830 Winter Street, Waltham, Massachusetts 02451, United States
| |
Collapse
|
20
|
Yaghoubi S, Karimi MH, Lotfinia M, Gharibi T, Mahi-Birjand M, Kavi E, Hosseini F, Sineh Sepehr K, Khatami M, Bagheri N, Abdollahpour-Alitappeh M. Potential drugs used in the antibody-drug conjugate (ADC) architecture for cancer therapy. J Cell Physiol 2019; 235:31-64. [PMID: 31215038 DOI: 10.1002/jcp.28967] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 05/20/2019] [Indexed: 01/04/2023]
Abstract
Cytotoxic small-molecule drugs have a major influence on the fate of antibody-drug conjugates (ADCs). An ideal cytotoxic agent should be highly potent, remain stable while linked to ADCs, kill the targeted tumor cell upon internalization and release from the ADCs, and maintain its activity in multidrug-resistant tumor cells. Lessons learned from successful and failed experiences in ADC development resulted in remarkable progress in the discovery and development of novel highly potent small molecules. A better understanding of such small-molecule drugs is important for development of effective ADCs. The present review discusses requirements making a payload appropriate for antitumor ADCs and focuses on the main characteristics of commonly-used cytotoxic payloads that showed acceptable results in clinical trials. In addition, the present study represents emerging trends and recent advances of payloads used in ADCs currently under clinical trials.
Collapse
Affiliation(s)
- Sajad Yaghoubi
- Department of Clinical Microbiology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Majid Lotfinia
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran.,Core Research Lab, Kashan University of Medical Sciences, Kashan, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Motahare Mahi-Birjand
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Esmaeil Kavi
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Fahimeh Hosseini
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koushan Sineh Sepehr
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Khatami
- NanoBioelectrochemistry Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | |
Collapse
|
21
|
Zheng Y, Shen Y, Meng X, Wu Y, Zhao Y, Wu C. Stabilizing p-Dithiobenzyl Urethane Linkers without Rate-Limiting Self-Immolation for Traceless Drug Release. ChemMedChem 2019; 14:1196-1203. [PMID: 31020782 DOI: 10.1002/cmdc.201900248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Indexed: 12/31/2022]
Abstract
Exploiting the redox sensitivity of disulfide bonds is a prevalent strategy in targeted prodrug designs. In contrast to aliphatic disulfides, p-thiobenzyl-based disulfides have rarely been used for prodrug designs, given their intrinsic instability caused by the low pKa of aromatic thiols. Here, we examined the interplay between steric hindrance and the low-pKa effect on thiol-disulfide exchange reactions and uncovered a new thiol-disulfide exchange process for the self-immolation of p-thiobenzyl-based disulfides. We observed a central leaving group shifting effect in the α,α-dimethyl-substituted p-dithiobenzyl urethane linkers (DMTB linkers), which leads to increased disulfide stability by more than two orders of magnitude, an extent that is significantly greater than that observed with typical aliphatic disulfides. In particular, the DMTB linkers display not only high stability, but also rapid self-immolation kinetics due to the low pKa of the aromatic thiol, which can be used as a general and robust linkage between targeting reagents and cytotoxic drugs for targeted prodrug designs. The unique and promising stability characteristics of the present DMTB linker will likely inspire the development of novel targeted prodrugs to achieve traceless release of drugs into cells.
Collapse
Affiliation(s)
- Yiwu Zheng
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yang Shen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Xiaoting Meng
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yaqi Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Yibing Zhao
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| | - Chuanliu Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, 361005, China
| |
Collapse
|
22
|
Nittoli T, Kelly MP, Delfino F, Rudge J, Kunz A, Markotan T, Spink J, Chen Z, Shan J, Navarro E, Tait M, Provoncha K, Giurleo J, Zhao F, Jiang X, Hylton D, Makonnen S, Hickey C, Kirshner JR, Thurston G, Papadopoulos N. Antibody drug conjugates of cleavable amino-alkyl and aryl maytansinoids. Bioorg Med Chem 2018; 26:2271-2279. [PMID: 29605304 DOI: 10.1016/j.bmc.2018.02.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 02/09/2018] [Accepted: 02/15/2018] [Indexed: 10/18/2022]
Abstract
Natural products have been used for many medicinal purposes for centuries. Antibody drug conjugates (ADCs) have utilized this rich source of small molecule therapeutics to produce several clinically useful treatments. ADCs based on the natural product maytansine have been successful clinically. The authors further the utility of the anti-cancer natural product maytansine by developing efficacious payloads and linker-payloads for conjugating to antibodies. The success of our approach was realized in the EGFRvIII targeting ADC EGFRvIII-16. The ADC was able to regress tumors in 2 tumor models (U251/EGFRvIII and MMT/EGFRvIII). When compared to a positive control ADC, the efficacy observed was similar or improved while the isotype control ADCs had no effect.
Collapse
Affiliation(s)
- Thomas Nittoli
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States.
| | - Marcus P Kelly
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Frank Delfino
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - John Rudge
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Arthur Kunz
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Thomas Markotan
- Abzena, 360 George Patterson Blvd, Bristol, PA 19007, United States
| | - Jan Spink
- Abzena, 360 George Patterson Blvd, Bristol, PA 19007, United States
| | - Zhaoyuan Chen
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jing Shan
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Elizabeth Navarro
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Michele Tait
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Kathleen Provoncha
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jason Giurleo
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Feng Zhao
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Xiaobo Jiang
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Donna Hylton
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Sosina Makonnen
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Carlos Hickey
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Jessica R Kirshner
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Gavin Thurston
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| | - Nicholas Papadopoulos
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, NY 10591, United States
| |
Collapse
|
23
|
Miller ML, Shizuka M, Wilhelm A, Salomon P, Reid EE, Lanieri L, Sikka S, Maloney EK, Harvey L, Qiu Q, Archer KE, Bai C, Vitharana D, Harris L, Singh R, Ponte JF, Yoder NC, Kovtun Y, Lai KC, Ab O, Pinkas J, Keating TA, Chari RV. A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody–Drug Conjugates (ADCs). Mol Cancer Ther 2018; 17:650-660. [DOI: 10.1158/1535-7163.mct-17-0940] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/15/2017] [Accepted: 12/28/2017] [Indexed: 11/16/2022]
|
24
|
Zheng Y, Ren J, Wu Y, Meng X, Zhao Y, Wu C. Proteolytic Unlocking of Ultrastable Twin-Acylhydrazone Linkers for Lysosomal Acid-Triggered Release of Anticancer Drugs. Bioconjug Chem 2017; 28:2620-2626. [DOI: 10.1021/acs.bioconjchem.7b00471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yiwu Zheng
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Jing Ren
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Yaqi Wu
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Xiaoting Meng
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Yibing Zhao
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| | - Chuanliu Wu
- Department of Chemistry,
College of Chemistry and Chemical Engineering, State Key Laboratory
of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of
Spectrochemical Analysis and Instrumentation, Xiamen University, Xiamen, 361005, P.R. China
| |
Collapse
|
25
|
Singh R, Setiady YY, Ponte J, Kovtun YV, Lai KC, Hong EE, Fishkin N, Dong L, Jones GE, Coccia JA, Lanieri L, Veale K, Costoplus JA, Skaletskaya A, Gabriel R, Salomon P, Wu R, Qiu Q, Erickson HK, Lambert JM, Chari RVJ, Widdison WC. A New Triglycyl Peptide Linker for Antibody-Drug Conjugates (ADCs) with Improved Targeted Killing of Cancer Cells. Mol Cancer Ther 2016; 15:1311-20. [PMID: 27197308 DOI: 10.1158/1535-7163.mct-16-0021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/19/2016] [Indexed: 11/16/2022]
Abstract
A triglycyl peptide linker (CX) was designed for use in antibody -: drug conjugates (ADC), aiming to provide efficient release and lysosomal efflux of cytotoxic catabolites within targeted cancer cells. ADCs comprising anti-epithelial cell adhesion molecule (anti-EpCAM) and anti-EGFR antibodies with maytansinoid payloads were prepared using CX or a noncleavable SMCC linker (CX and SMCC ADCs). The in vitro cytotoxic activities of CX and SMCC ADCs were similar for several cancer cell lines; however, the CX ADC was more active (5-100-fold lower IC50) than the SMCC ADC in other cell lines, including a multidrug-resistant line. Both CX and SMCC ADCs showed comparable MTDs and pharmacokinetics in CD-1 mice. In Calu-3 tumor xenografts, antitumor efficacy was observed with the anti-EpCAM CX ADC at a 5-fold lower dose than the corresponding SMCC ADC in vivo Similarly, the anti-EGFR CX ADC showed improved antitumor activity over the respective SMCC conjugate in HSC-2 and H1975 tumor models; however, both exhibited similar activity against FaDu xenografts. Mechanistically, in contrast with the charged lysine-linked catabolite of SMCC ADC, a significant fraction of the carboxylic acid catabolite of CX ADC could be uncharged in the acidic lysosomes, and thus diffuse out readily into the cytosol. Upon release from tumor cells, CX catabolites are charged at extracellular pH and do not penetrate and kill neighboring cells, similar to the SMCC catabolite. Overall, these data suggest that CX represents a promising linker option for the development of ADCs with improved therapeutic properties. Mol Cancer Ther; 15(6); 1311-20. ©2016 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ling Dong
- ImmunoGen, Inc., Waltham, Massachusetts
| | | | | | | | | | | | | | | | | | - Rui Wu
- ImmunoGen, Inc., Waltham, Massachusetts
| | | | | | | | | | | |
Collapse
|