1
|
Grairi M, Le Borgne M. Antibody-drug conjugates: prospects for the next generation. Drug Discov Today 2024; 29:104241. [PMID: 39542204 DOI: 10.1016/j.drudis.2024.104241] [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: 07/02/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
The concept of a 'magic bullet' was first introduced by Paul Ehrlich in the early 1900s, he foresaw the advent of targeted therapies and the specific killing of harmful cells and/or microorganisms. However, these therapies were only used in the clinic after the second half of the 20th century with the development of specific monoclonal antibodies. To date, 13 antibody-drug conjugates (ADCs) are commercially available. Many advances have been made by modifying one or several of the three main components of an ADC, namely the antibody, the cleavable or non-cleavable linker or the payload, and by integrating conjugation chemistry. Despite these efforts, some problems have emerged and thus limit their effectiveness. New strategies could overcome these problems and identify the next generation of ADC.
Collapse
Affiliation(s)
- Meriem Grairi
- Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Faculté de Pharmacie, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France
| | - Marc Le Borgne
- Institut des Sciences Pharmaceutiques et Biologiques (ISPB), Faculté de Pharmacie, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France; Small Molecules for Biological Targets Team, Centre de recherche en cancérologie de Lyon, Centre Léon Bérard, CNRS 5286, INSERM 1052, Université Claude Bernard Lyon 1, Univ Lyon, 69373 Lyon, France.
| |
Collapse
|
2
|
Tedeschini T, Campara B, Grigoletto A, Zanotto I, Cannella L, Gabbia D, Matsuno Y, Suzuki A, Yoshioka H, Armirotti A, De Martin S, Pasut G. Optimization of a pendant-shaped PEGylated linker for antibody-drug conjugates. J Control Release 2024; 375:74-89. [PMID: 39216599 DOI: 10.1016/j.jconrel.2024.08.049] [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: 02/12/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
In this work, we conceived and developed antibody-drug conjugates (ADCs) that could efficiently release the drug after enzymatic cleavage of the linker moiety by tumoral proteases. The antibody-drug linkers we used are the result of a rational optimization of a previously reported PEGylated linker, PUREBRIGHT® MA-P12-PS, which showed excellent drug loading capacities but lacked an inbuilt drug discharge mechanism, thus limiting the potency of the resulting ADCs. To address this limitation, we chose to incorporate a protease-sensitive trigger into the linker to favor the release of a "PEGless" drug inside the tumor cells and, therefore, obtain potent ADCs. Currently, most marketed ADCs are based on the Val-Cit dipeptide followed by a self-immolative spacer for releasing the drug in its unmodified form. Here, we selected two untraditional peptide sequences, a Phe-Gly dipeptide and a Val-Ala-Gly tripeptide and placed one or the other in between the drug on one side (N-terminus) and the rest of the linker, including the PEG moiety, on the other side (C-terminus), without a self-immolative group. We found that both linkers responded to cathepsin B, a reference lysosomal enzyme, and liberated a PEG-free drug catabolite, as desired. We then used the two linkers to generate ADCs based on trastuzumab (a HER2-targeting antibody) and DM1 (a microtubule-targeted cytotoxic agent) with an average drug-to-antibody ratio (DAR) of 4 or 8. The ADCs showed restored cytotoxicity in vitro, which was proportional to the DM1 loading and generally higher for the ADCs bearing Val-Ala-Gly in their structure. In an ovarian cancer mouse model, the DAR 8 ADC based on Val-Ala-Gly behaved better than Kadcyla® (an approved ADC of DAR 3.5 used as control throughout this study), leading to a higher tumor volume reduction and more prolonged median survival. Taken together, our results depict a successful linker optimization process and encourage the application of the Val-Ala-Gly tripeptide as an alternative to other existing protease-sensitive triggers for ADCs.
Collapse
Affiliation(s)
- T Tedeschini
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy.
| | - B Campara
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - A Grigoletto
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - I Zanotto
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - L Cannella
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - D Gabbia
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - Y Matsuno
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - A Suzuki
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - H Yoshioka
- NOF CORPORATION, Life Science Research Laboratory, 3-3 Chidori-Cho, Kawasaki-Ku, Kawasaki, Kanagawa 210-0865, Japan
| | - A Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - S De Martin
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy
| | - G Pasut
- University of Padova, Dept. Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 35131 Padova, Italy.
| |
Collapse
|
3
|
Yang Q, Liu Y. Technical, preclinical, and clinical developments of Fc-glycan-specific antibody-drug conjugates. RSC Med Chem 2024:d4md00637b. [PMID: 39568595 PMCID: PMC11575643 DOI: 10.1039/d4md00637b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have emerged as a powerful avenue in the therapeutic treatment of cancer. Site-specific antibody-drug conjugations represent the latest trend in the development of ADCs, addressing the limitations of traditional random conjugation technologies. This article summarizes the innovative development of Fc-glycan-specific ADCs (gsADCs), which utilize the conserved Fc N-glycan as the anchor point for site-specific conjugation. This approach offers significant strengths, including improved ADC homogeneity and overall hydrophilicity, enhanced pharmacokinetics and therapeutic index, and potentially reduced Fc receptor-mediated side effects. Currently dozens of gsADCs are in different preclinical and clinical development stages. Notably, JSKN003 and IBI343 have demonstrated promising results in phase 1 trials and are advancing into phase 3 studies. This review discusses the advantages of Fc-glycan-conjugation, various glycan-specific conjugation techniques, and the preclinical and clinical development of gsADCs. While challenges such as increased manufacturing cost for large-scale production need continuous innovation to overcome and there are different opinions regarding the pros and cons of reduced/diminished affinities to Fc gamma receptors, ongoing research and clinical progress underscore the potential of gsADCs to renovate ADC cancer therapy.
Collapse
Affiliation(s)
- Qiang Yang
- Brilliant BioConsultation Ellicott City MD 21043 USA
| | | |
Collapse
|
4
|
Hao JL, Li XY, Liu YT, Lang JX, Liu DJ, Zhang CD. Antibody-drug conjugates in gastric cancer: from molecular landscape to clinical strategies. Gastric Cancer 2024; 27:887-906. [PMID: 38963593 DOI: 10.1007/s10120-024-01529-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a crucial component of targeted therapies in gastric cancer, potentially altering traditional treatment paradigms. Many ADCs have entered rigorous clinical trials based on biological theories and preclinical experiments. Modality trials have also been conducted in combination with monoclonal antibody therapies, chemotherapies, immunotherapies, and other treatments to enhance the efficacy of drug coordination effects. However, ADCs exhibit limitations in treating gastric cancer, including resistance triggered by their structure or other factors. Ongoing intensive researches and preclinical experiments are yielding improvements, while enhancements in drug development processes and concomitant diagnostics during the therapeutic period actively boost ADC efficacy. The optimal treatment strategy for gastric cancer patients is continually evolving. This review summarizes the clinical progress of ADCs in treating gastric cancer, analyzes the mechanisms of ADC combination therapies, discusses resistance patterns, and offers a promising outlook for future applications in ADC drug development and companion diagnostics.
Collapse
Affiliation(s)
- Jia-Lin Hao
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yu-Tong Liu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Ji-Xuan Lang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Di-Jie Liu
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Chun-Dong Zhang
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| |
Collapse
|
5
|
Guo Y, Shen Z, Zhao W, Lu J, Song Y, Shen L, Lu Y, Wu M, Shi Q, Zhuang W, Qiu Y, Sheng J, Zhou Z, Fang L, Che J, Dong X. Rational Identification of Novel Antibody-Drug Conjugate with High Bystander Killing Effect against Heterogeneous Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306309. [PMID: 38269648 PMCID: PMC10987111 DOI: 10.1002/advs.202306309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 12/15/2023] [Indexed: 01/26/2024]
Abstract
Bystander-killing payloads can significantly overcome the tumor heterogeneity issue and enhance the clinical potential of antibody-drug conjugates (ADC), but the rational design and identification of effective bystander warheads constrain the broader implementation of this strategy. Here, graph attention networks (GAT) are constructed for a rational bystander killing scoring model and ADC construction workflow for the first time. To generate efficient bystander-killing payloads, this model is utilized for score-directed exatecan derivatives design. Among them, Ed9, the most potent payload with satisfactory permeability and bioactivity, is further used to construct ADC. Through linker optimization and conjugation, novel ADCs are constructed that perform excellent anti-tumor efficacy and bystander-killing effect in vivo and in vitro. The optimal conjugate T-VEd9 exhibited therapeutic efficacy superior to DS-8201 against heterogeneous tumors. These results demonstrate that the effective scoring approach can pave the way for the discovery of novel ADC with promising bystander payloads to combat tumor heterogeneity.
Collapse
Affiliation(s)
- Yu Guo
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Zheyuan Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Wenbin Zhao
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityHangzhou310018P. R. China
| | - Jialiang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yi Song
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Liteng Shen
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yang Lu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Mingfei Wu
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Qiuqiu Shi
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Weihao Zhuang
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Yueping Qiu
- The Department of PharmacyZhejiang Cancer HospitalHangzhou310022P. R. China
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated Hospital, Zhejiang University School of MedicineHangzhou310002P. R. China
| | - Zhan Zhou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang UniversityHangzhou310018P. R. China
| | - Luo Fang
- The Department of PharmacyZhejiang Cancer HospitalHangzhou310022P. R. China
| | - Jinxin Che
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical SciencesZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhou310058P. R. China
- Department of PharmacySecond Affiliated HospitalZhejiang University School of MedicineHangzhou310009P. R. China
| |
Collapse
|
6
|
Tsuchikama K, Anami Y, Ha SYY, Yamazaki CM. Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol 2024; 21:203-223. [PMID: 38191923 DOI: 10.1038/s41571-023-00850-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Summer Y Y Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chisato M Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
7
|
Zimmerman BS, Esteva FJ. Next-Generation HER2-Targeted Antibody-Drug Conjugates in Breast Cancer. Cancers (Basel) 2024; 16:800. [PMID: 38398191 PMCID: PMC10887217 DOI: 10.3390/cancers16040800] [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: 01/07/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) tyrosine kinase is overexpressed in 20% of breast cancers and associated with a less favorable prognosis compared to HER2-negative disease. Patients have traditionally been treated with a combination of chemotherapy and HER2-targeted monoclonal antibodies such as trastuzumab and pertuzumab. The HER2-targeted antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd) represent a novel class of therapeutics in breast cancer. These drugs augment monoclonal antibodies with a cytotoxic payload, which is attached by a linker, forming the basic structure of an ADC. Novel combinations and sequential approaches are under investigation to overcome resistance to T-DM1 and T-DXd. Furthermore, the landscape of HER2-targeted therapy is rapidly advancing with the development of ADCs designed to attack cancer cells with greater precision and reduced toxicity. This review provides an updated summary of the current state of HER2-targeted ADCs as well as a detailed review of investigational agents on the horizon. Clinical trials are crucial in determining the optimal dosing regimens, understanding resistance mechanisms, and identifying patient populations that would derive the most benefit from these treatments. These novel ADCs are at the forefront of a new era in targeted cancer therapy, holding the potential to improve outcomes for patients with HER2-positive and HER2-Low breast cancer.
Collapse
Affiliation(s)
- Brittney S. Zimmerman
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| | - Francisco J. Esteva
- Northwell, New Hyde Park, NY 11042, USA;
- Northwell Health Cancer Institute, Lake Success, NY 11042, USA
| |
Collapse
|
8
|
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
|
9
|
Schmitt S, Machui P, Mai I, Herterich S, Wunder S, Cyprys P, Gerlach M, Ochtrop P, Hackenberger CP, Schumacher D, Helma J, Vogl AM, Kasper MA. Design and Evaluation of Phosphonamidate-Linked Exatecan Constructs for Highly Loaded, Stable, and Efficacious Antibody-Drug Conjugates. Mol Cancer Ther 2024; 23:199-211. [PMID: 37828728 PMCID: PMC10831470 DOI: 10.1158/1535-7163.mct-23-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/30/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody-drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker-payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker-payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB-exatecan moiety. Furthermore, we demonstrate that the identified linker-payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker-payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christian P.R. Hackenberger
- Chemical Biology Department, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
10
|
Timmins P. Industry update: the latest developments in the field of therapeutic delivery, October 2023. Ther Deliv 2024; 15:77-94. [PMID: 38197132 DOI: 10.4155/tde-2023-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Affiliation(s)
- Peter Timmins
- Department of Pharmacy, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK
| |
Collapse
|
11
|
Postupalenko V, Marx L, Pantin M, Viertl D, Gsponer N, Giudice G, Gasilova N, Schottelius M, Lévy F, Garrouste P, Segura JM, Nyanguile O. Site-selective template-directed synthesis of antibody Fc conjugates with concomitant ligand release. Chem Sci 2024; 15:1324-1337. [PMID: 38274063 PMCID: PMC10806771 DOI: 10.1039/d3sc04324j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/13/2023] [Indexed: 01/27/2024] Open
Abstract
Template-directed methods are emerging as some of the most effective means to conjugate payloads at selective sites of monoclonal antibodies (mAbs). We have previously reported a method based on an engineered Fc-III reactive peptide to conjugate a radionuclide chelator to K317 of antibodies with the concomitant release of the Fc-III peptide ligand. Here, our method was redesigned to target two lysines proximal to the Fc-III binding site, K248 and K439. Using energy minimization predictions and a semi-combinatorial synthesis approach, we sampled multiple Fc-III amino acid substituents of A3, H5, L6 and E8, which were then converted into Fc-III reactive conjugates. Middle-down MS/MS subunit analysis of the resulting trastuzumab conjugates revealed that K248 and K439 can be selectively targeted using the Fc-III reactive variants L6Dap, L6Orn, L6Y and A3K or A3hK, respectively. Across all variants tested, L6Orn-carbonate appeared to be the best candidate, yielding a degree and yield of conjugation of almost 2 and 100% for a broad array of payloads including radionuclide chelators, fluorescent dyes, click-chemistry reagents, pre-targeted imaging reagents, and some cytotoxic small molecules. Furthermore, L6Orn carbonate appeared to yield similar conjugation results across multiple IgG subtypes. In vivo proof of concept was achieved by conjugation of NODAGA to the PD1/PD-L1 immune checkpoint inhibitor antibody atezolizumab, followed by PET imaging of PD-L1 expression in mice bearing PD-L1 expressing tumor xenograft using radiolabeled [64Cu]Cu-atezolizumab.
Collapse
Affiliation(s)
- Viktoriia Postupalenko
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Léo Marx
- Debiopharm Research & Manufacturing SA Campus "après-demain", Rue du Levant 146 1920 Martigny Switzerland
| | - Mathilde Pantin
- Debiopharm Research & Manufacturing SA Campus "après-demain", Rue du Levant 146 1920 Martigny Switzerland
| | - David Viertl
- Translational Radiopharmaceutical Sciences, Departments of Nuclear Medicine and of Oncology, CHUV/UNIL 1011 Lausanne Switzerland
- In Vivo Imaging Facility, Department of Research and Training, University of Lausanne CH-1011 Lausanne
| | - Nadège Gsponer
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Gaëlle Giudice
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Natalia Gasilova
- EPFL Valais Wallis, MSEAP, ISIC-GE-VS rue de l'Industrie 17 1951 Sion Switzerland
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Departments of Nuclear Medicine and of Oncology, CHUV/UNIL 1011 Lausanne Switzerland
- Agora, pôle de recherche sur le cancer 1011 Lausanne Switzerland
| | - Frédéric Lévy
- Debiopharm International SA Forum "après-demain", Chemin Messidor 5-7, Case postale 5911 1002 Lausanne Switzerland
| | - Patrick Garrouste
- Debiopharm Research & Manufacturing SA Campus "après-demain", Rue du Levant 146 1920 Martigny Switzerland
| | - Jean-Manuel Segura
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| | - Origène Nyanguile
- Institute of Life Technologies, HES-SO Valais-Wallis Rue de l'Industrie 23 CH-1950 Sion Switzerland
| |
Collapse
|
12
|
Schöffski P, Wang CC, Schöffski MP, Wozniak A. Current Role of Topoisomerase I Inhibitors for the Treatment of Mesenchymal Malignancies and Their Potential Future Use as Payload of Sarcoma-Specific Antibody-Drug Conjugates. Oncol Res Treat 2023; 47:18-41. [PMID: 38016427 PMCID: PMC10860894 DOI: 10.1159/000535491] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Topoisomerase I is an enzyme that plays a crucial part in DNA replication and transcription by the relaxation of supercoiled double-stranded DNA. Topoisomerase I inhibitors bind to the topoisomerase I cleavage complex, thereby stabilizing it and preventing the religation of the DNA strands, leading to DNA damage, cell cycle arrest, and apoptosis. Various topoisomerase I inhibitors have been evaluated in solid tumors, and irinotecan and topotecan have been approved for the treatment of epithelial malignancies. None of them have been approved for sarcoma, a diverse group of rare solid tumors with an unmet need for effective treatments. SUMMARY Topoisomerase I inhibitors have been evaluated in preclinical studies as single agents or in combination in solid tumors, some of which have included sarcomas where activity was observed. Clinical trials evaluating topoisomerase I inhibitors for the treatment of sarcoma have shown limited efficacy as monotherapy. In combination with other cytotoxic agents, topoisomerase I inhibitors have become part of clinical routine in selected sarcoma subtypes. Regimens such as irinotecan/vincristine/temozolomide are used in relapsed rhabdomyosarcoma, irinotecan/temozolomide and vincristine/topotecan/cyclophosphamide are commonly given in refractory Ewing sarcoma, and topotecan/carboplatin showed some activity in advanced soft tissue sarcoma. This review provides an overview of key studies with topoisomerase I inhibitors for the treatment of sarcoma. Topoisomerase I inhibitors are currently also being assessed as "payloads" for antibody-drug conjugates (ADCs), allowing for the targeting of specific antigen-expressing tumor cells and the delivery of the inhibitor directly to the tumor cells with the potential of enhancing therapeutic efficacy while minimizing systemic toxicity. Here, we also provide a brief overview on topoisomerase I-ADCs. KEY MESSAGE Topoisomerase I inhibitors are an important component of some systemic therapies for selected sarcomas and have potent cytotoxic properties and pharmacological characteristics that make them relevant candidates as payloads for the development of sarcoma-specific ADCs. ADCs are antibody-based targeted agents allowing for efficient and specific delivery of a given drug to the tumor cell. Topoisomerase I-ADCs are a novel targeted delivery approach which may have the potential to improve the therapeutic index of topoisomerase I inhibitors in the treatment of sarcoma and warrants investigation in a broad variety of mesenchymal malignancies.
Collapse
Affiliation(s)
- Patrick Schöffski
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | - Chao-Chi Wang
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| | | | - Agnieszka Wozniak
- Department of Oncology, Laboratory of Experimental Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
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: 13] [Impact Index Per Article: 6.5] [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
|
14
|
Dumontet C, Reichert JM, Senter PD, Lambert JM, Beck A. Antibody-drug conjugates come of age in oncology. Nat Rev Drug Discov 2023; 22:641-661. [PMID: 37308581 DOI: 10.1038/s41573-023-00709-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 06/14/2023]
Abstract
Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with the potency of highly cytotoxic agents, potentially reducing the severity of side effects by preferentially targeting their payload to the tumour site. ADCs are being increasingly used in combination with other agents, including as first-line cancer therapies. As the technology to produce these complex therapeutics has matured, many more ADCs have been approved or are in late-phase clinical trials. The diversification of antigenic targets as well as bioactive payloads is rapidly broadening the scope of tumour indications for ADCs. Moreover, novel vector protein formats as well as warheads targeting the tumour microenvironment are expected to improve the intratumour distribution or activation of ADCs, and consequently their anticancer activity for difficult-to-treat tumour types. However, toxicity remains a key issue in the development of these agents, and better understanding and management of ADC-related toxicities will be essential for further optimization. This Review provides a broad overview of the recent advances and challenges in ADC development for cancer treatment.
Collapse
Affiliation(s)
- Charles Dumontet
- CRCL INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, Lyon, France.
| | | | | | | | - Alain Beck
- Institut de Recherche Pierre Fabre, CIPF, Saint-Julien-en-Genevois, France
| |
Collapse
|
15
|
Shastry M, Gupta A, Chandarlapaty S, Young M, Powles T, Hamilton E. Rise of Antibody-Drug Conjugates: The Present and Future. Am Soc Clin Oncol Educ Book 2023; 43:e390094. [PMID: 37229614 DOI: 10.1200/edbk_390094] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Antibody-drug conjugates (ADCs) embody a simple, but elegant, vision for cancer therapy-the delivery of a potent cytotoxic agent to tumor cells with minimal damage to normal cells-so-called smart chemo. Although there were significant challenges in achieving this milestone culminating in the first Food and Drug Administration approval in 2000, subsequent advancements in technology have led to rapid drug development with regulatory approvals for ADCs targeting a variety of tumor types. The most successful application for solid tumors has been in breast cancer, with ADCs becoming the standard of care across traditional human epidermal growth factor receptor 2 (HER2)+, hormone receptor+ (HR+) and triple-negative disease subtypes. Moreover, the improved features and gains in potency with the development of ADCs have expanded the treatment-eligible population to those with low/heterogeneous expression of the target antigen on the tumor with trastuzumab deruxtecan or in the case of sacituzumab govitecan, agnostic to target expression. Despite their antibody-directed homing, these novel agents come with their share of toxicities obligating appropriate patient selection and vigilant monitoring while on treatment. As more ADCs are included in the treatment armamentarium, mechanisms of resistance need to be studied and understood for optimal sequencing. Modifying the payload to use immune-stimulating agents or combination therapies with immunotherapy and other effective targeted therapies may further extend the utility of these agents in the treatment of solid tumors.
Collapse
Affiliation(s)
| | - Avantika Gupta
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Sarat Chandarlapaty
- Department of Medicine, Human Oncology and Pathogenesis Program, MSKCC, New York, NY
| | - Matthew Young
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Erika Hamilton
- Sarah Cannon Research Institute, Nashville, TN
- Tennessee Oncology, Nashville, TN
| |
Collapse
|
16
|
Yang J, Jia L, He Z, Wang Y. Recent advances in SN-38 drug delivery system. Int J Pharm 2023; 637:122886. [PMID: 36966982 DOI: 10.1016/j.ijpharm.2023.122886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
DNA topoisomerase I plays a key role in lubricatingthe wheels of DNA replication or RNA transcription through breaking and reconnecting DNA single-strand. It is widely known that camptothecin and its derivatives (CPTs) have inhibitory effects on topoisomerases I, and have obtained some clinical benefits in cancer treatment. The potent cytotoxicity makes 7-ethyl-10-hydroxycamptothecin (SN-38) become a brilliant star among these derivatives. However, some undesirable physical and chemical properties of this compound, including poor solubility and stability, seriously hinder its effective delivery to tumor sites. In recent years, strategies to alleviate these defects have aroused extensive research interest. By focusing on the loading mechanism, basic nanodrug delivery systems with SN-38 loaded, like nanoparticles, liposomes and micelles, are demonstrated here. Additionally, functionalized nanodrug delivery systems of SN-38 including prodrug and active targeted nanodrug delivery systems and delivery systems designed to overcome drug resistance are also reviewed. At last, challenges for future research in formulation development and clinical translation of SN-38 drug delivery system are discussed.
Collapse
|
17
|
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: 8.0] [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
|
18
|
Payload diversification: a key step in the development of antibody-drug conjugates. J Hematol Oncol 2023; 16:3. [PMID: 36650546 PMCID: PMC9847035 DOI: 10.1186/s13045-022-01397-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Antibody-drug conjugates (ADCs) is a fast moving class of targeted biotherapeutics that currently combines the selectivity of monoclonal antibodies with the potency of a payload consisting of cytotoxic agents. For many years microtubule targeting and DNA-intercalating agents were at the forefront of ADC development. The recent approval and clinical success of trastuzumab deruxtecan (Enhertu®) and sacituzumab govitecan (Trodelvy®), two topoisomerase 1 inhibitor-based ADCs, has shown the potential of conjugating unconventional payloads with differentiated mechanisms of action. Among future developments in the ADC field, payload diversification is expected to play a key role as illustrated by a growing number of preclinical and clinical stage unconventional payload-conjugated ADCs. This review presents a comprehensive overview of validated, forgotten and newly developed payloads with different mechanisms of action.
Collapse
|
19
|
Tashima T. Delivery of Drugs into Cancer Cells Using Antibody-Drug Conjugates Based on Receptor-Mediated Endocytosis and the Enhanced Permeability and Retention Effect. Antibodies (Basel) 2022; 11:antib11040078. [PMID: 36546903 PMCID: PMC9774242 DOI: 10.3390/antib11040078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Innumerable people worldwide die of cancer every year, although pharmaceutical therapy has actualized many benefits in human health. For background, anti-cancer drug development is difficult due to the multifactorial pathogenesis and complicated pathology of cancers. Cancer cells excrete hydrophobic low-molecular anti-cancer drugs by overexpressed efflux transporters such as multiple drug resistance 1 (MDR1) at the apical membrane. Mutation-driven drug resistance is also developed in cancer. Moreover, the poor distribution of drug to cancer cells is a serious problem, because patients suffer from off-target side effects. Thus, highly selective and effective drug delivery into solid cancer cells across the membrane should be established. It is known that substances (10-100 nm in diameter) such as monoclonal antibodies (mAbs) (approximately 14.2 nm in diameter) or nanoparticles spontaneously gather in solid tumor stroma or parenchyma through the capillary endothelial fenestration, ranging from 200-2000 nm, in neovasculatures due to the enhanced permeability and retention (EPR) effect. Furthermore, cancer antigens, such as HER2, Nectin-4, or TROP2, highly selectively expressed on the surface of cancer cells act as a receptor for receptor-mediated endocytosis (RME) using mAbs against such antigens. Thus, antibody-drug conjugates (ADCs) are promising anti-cancer pharmaceutical agents that fulfill accurate distribution due to the EPR effect and due to antibody-antigen binding and membrane permeability owing to RME. In this review, I introduce the implementation and possibility of highly selective anti-cancer drug delivery into solid cancer cells based on the EPR effect and RME using anti-cancer antigens ADCs with payloads through suitable linkers.
Collapse
Affiliation(s)
- Toshihiko Tashima
- Tashima Laboratories of Arts and Sciences, 1239-5 Toriyama-cho, Kohoku-ku, Yokohama 222-0035, Japan
| |
Collapse
|
20
|
Nguyen KA, Conilh L, Falson P, Dumontet C, Boumendjel A. The first ADC bearing the ferroptosis inducer RSL3 as a payload with conservation of the fragile electrophilic warhead. Eur J Med Chem 2022; 244:114863. [DOI: 10.1016/j.ejmech.2022.114863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022]
|
21
|
Ma Z, Zhang Y, Zhu M, Feng L, Zhang Y, An Z. Interstitial lung disease associated with anti-HER2 anti-body drug conjugates: results from clinical trials and the WHO's pharmacovigilance database. Expert Rev Clin Pharmacol 2022; 15:1351-1361. [DOI: 10.1080/17512433.2022.2121705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Zhuo Ma
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yi Zhang
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Lin Feng
- Department of Clinical Epidemiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Zhuoling An
- Department of Pharmacy, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Han S, Lim KS, Blackburn BJ, Yun J, Putnam CW, Bull DA, Won YW. The Potential of Topoisomerase Inhibitor-Based Antibody–Drug Conjugates. Pharmaceutics 2022; 14:pharmaceutics14081707. [PMID: 36015333 PMCID: PMC9413092 DOI: 10.3390/pharmaceutics14081707] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 12/17/2022] Open
Abstract
DNA topoisomerases are essential enzymes that stabilize DNA supercoiling and resolve entanglements. Topoisomerase inhibitors have been widely used as anti-cancer drugs for the past 20 years. Due to their selectivity as topoisomerase I (TOP1) inhibitors that trap TOP1 cleavage complexes, camptothecin and its derivatives are promising anti-cancer drugs. To increase accumulation of TOP1 inhibitors in cancer cells through the targeting of tumors, TOP1 inhibitor antibody–drug conjugates (TOP1-ADC) have been developed and marketed. Some TOP1-ADCs have shown enhanced therapeutic efficacy compared to prototypical anti-cancer ADCs, such as T-DM1. Here, we review various types of camptothecin-based TOP1 inhibitors and recent developments in TOP1-ADCs. We then propose key points for the design and construction of TOP1-ADCs. Finally, we discuss promising combinatorial strategies, including newly developed approaches to maximizing the therapeutic potential of TOP1-ADCs.
Collapse
Affiliation(s)
- Seungmin Han
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Kwang Suk Lim
- Department of Biotechnology and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Korea
- Department of Smart Health Science and Technology, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Korea
| | - Brody J. Blackburn
- Department of Medical Pharmacology, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Jina Yun
- Division of Hematology-Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Korea
| | - Charles W. Putnam
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - David A. Bull
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
| | - Young-Wook Won
- Division of Cardiothoracic Surgery, Department of Surgery, University of Arizona College of Medicine—Tucson, Tucson, AZ 85724, USA
- Correspondence:
| |
Collapse
|
23
|
Jo U, Murai Y, Agama KK, Sun Y, Saha LK, Yang X, Arakawa Y, Gayle S, Jones K, Paralkar V, Sundaram RK, Doorn JV, Vasquez JC, Bindra RS, Choi WS, Pommier Y. TOP1-DNA Trapping by Exatecan and Combination Therapy with ATR Inhibitor. Mol Cancer Ther 2022; 21:1090-1102. [PMID: 35439320 PMCID: PMC9256811 DOI: 10.1158/1535-7163.mct-21-1000] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/18/2022] [Accepted: 04/12/2022] [Indexed: 01/07/2023]
Abstract
Exatecan and deruxtecan are antineoplastic camptothecin derivatives in development as tumor-targeted-delivery warheads in various formulations including peptides, liposomes, polyethylene glycol nanoparticles, and antibody-drug conjugates. Here, we report the molecular pharmacology of exatecan compared with the clinically approved topoisomerase I (TOP1) inhibitors and preclinical models for validating biomarkers and the combination of exatecan with ataxia telangiectasia and Rad3-related kinase (ATR) inhibitors. Modeling exatecan binding at the interface of a TOP1 cleavage complex suggests two novel molecular interactions with the flanking DNA base and the TOP1 residue N352, in addition to the three known interactions of camptothecins with the TOP1 residues R364, D533, and N722. Accordingly, exatecan showed much stronger TOP1 trapping, higher DNA damage, and apoptotic cell death than the classical TOP1 inhibitors used clinically. We demonstrate the value of SLFN11 expression and homologous recombination (HR) deficiency (HRD) as predictive biomarkers of response to exatecan. We also show that exatecan kills cancer cells synergistically with the clinical ATR inhibitor ceralasertib (AZD6738). To establish the translational potential of this combination, we tested CBX-12, a clinically developed pH-sensitive peptide-exatecan conjugate that selectively targets cancer cells and is currently in clinical trials. The combination of CBX-12 with ceralasertib significantly suppressed tumor growth in mouse xenografts. Collectively, our results demonstrate the potency of exatecan as a TOP1 inhibitor and its clinical potential in combination with ATR inhibitors, using SLFN11 and HRD as predictive biomarkers.
Collapse
Affiliation(s)
- Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhisa Murai
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Keli K. Agama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yilun Sun
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liton Kumar Saha
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Xi Yang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Yasuhiro Arakawa
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Kelli Jones
- Cybrexa Therapeutics, New Haven, CT 06511, USA
| | | | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Jinny Van Doorn
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Juan C. Vasquez
- Department of Pediatrics, Yale School of Medicine, New Haven, CT 06511, USA
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Woo Suk Choi
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Angelastro A, Barkhanskiy A, Mattey AP, Pallister EG, Spiess R, Goundry W, Barran P, Flitsch SL. Galactose Oxidase Enables Modular Assembly of Conjugates from Native Antibodies with High Drug-to-Antibody Ratios. CHEMSUSCHEM 2022; 15:e202102592. [PMID: 34931761 PMCID: PMC9303943 DOI: 10.1002/cssc.202102592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/20/2021] [Indexed: 05/31/2023]
Abstract
The potential of antibody conjugates with high drug loading in anticancer therapy has recently been highlighted by the approval of Trastuzumab deruxtecan and Sacituzumab govitecan. These biopharmaceutical approaches have spurred interest in bioconjugation strategies with high and defined degrees of drug-to-antibody ratio (DAR), in particular on native antibodies. Here, a glycoengineering methodology was developed to generate antibody drug conjugates with DAR of up to eight, by combining highly selective enzymatic galactosylation and oxidation with biorthogonal tandem Knoevenagel-Michael addition chemistry. This four-step approach offers a selective route to conjugates from native antibodies with high drug loading, and thus illustrates how biocatalysis can be used for the generation of biopharmaceuticals using mild reaction conditions.
Collapse
Affiliation(s)
- Antonio Angelastro
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Alexey Barkhanskiy
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Ashley P. Mattey
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Edward G. Pallister
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Reynard Spiess
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - William Goundry
- The Department of Pharmaceutical SciencesAstraZenecaSilk Road Business ParkMacclesfieldSK10 2NAUK
| | - Perdita Barran
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| | - Sabine L. Flitsch
- School of Chemistry and Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DN
| |
Collapse
|
25
|
Díaz-Rodríguez E, Gandullo-Sánchez L, Ocaña A, Pandiella A. Novel ADCs and Strategies to Overcome Resistance to Anti-HER2 ADCs. Cancers (Basel) 2021; 14:154. [PMID: 35008318 PMCID: PMC8750930 DOI: 10.3390/cancers14010154] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 12/23/2022] Open
Abstract
During recent years, a number of new compounds against HER2 have reached clinics, improving the prognosis and quality of life of HER2-positive breast cancer patients. Nonetheless, resistance to standard-of-care drugs has motivated the development of novel agents, such as new antibody-drug conjugates (ADCs). The latter are a group of drugs that benefit from the potency of cytotoxic agents whose action is specifically guided to the tumor by the target-specific antibody. Two anti-HER2 ADCs have reached the clinic: trastuzumab-emtansine and, more recently, trastuzumab-deruxtecan. In addition, several other HER2-targeted ADCs are in preclinical or clinical development, some of them with promising signs of activity. In the present review, the structure, mechanism of action, and potential resistance to all these ADCs will be described. Specific attention will be given to discussing novel strategies to circumvent resistance to ADCs.
Collapse
Affiliation(s)
- Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
- Departamento de Bioquímica y Biología Molecular, University of Salamanca, 37007 Salamanca, Spain
| | - Lucía Gandullo-Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| | - Alberto Ocaña
- Hospital Clínico San Carlos, Centro de Investigación Biomédica en Red de Oncología (CIBERONC), 28040 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-IBSAL and CIBERONC, 37007 Salamanca, Spain; (E.D.-R.); (L.G.-S.)
| |
Collapse
|
26
|
Theocharopoulos C, Lialios PP, Samarkos M, Gogas H, Ziogas DC. Antibody-Drug Conjugates: Functional Principles and Applications in Oncology and Beyond. Vaccines (Basel) 2021; 9:1111. [PMID: 34696218 PMCID: PMC8538104 DOI: 10.3390/vaccines9101111] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/28/2022] Open
Abstract
In the era of precision medicine, antibody-based therapeutics are rapidly enriched with emerging advances and new proof-of-concept formats. In this context, antibody-drug conjugates (ADCs) have evolved to merge the high selectivity and specificity of monoclonal antibodies (mAbs) with the cytotoxic potency of attached payloads. So far, ten ADCs have been approved by FDA for oncological indications and many others are currently being tested in clinical and preclinical level. This paper summarizes the essential components of ADCs, from their functional principles and structure up to their limitations and resistance mechanisms, focusing on all latest bioengineering breakthroughs such as bispecific mAbs, dual-drug platforms as well as novel linkers and conjugation chemistries. In continuation of our recent review on anticancer implication of ADC's technology, further insights regarding their potential usage outside of the oncological spectrum are also presented. Better understanding of immunoconjugates could maximize their efficacy and optimize their safety, extending their use in everyday clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Dimitrios C. Ziogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Laiko General Hospital, 115 27 Athens, Greece; (C.T.); (P.-P.L.); (M.S.); (H.G.)
| |
Collapse
|
27
|
Khaiwa N, Maarouf NR, Darwish MH, Alhamad DWM, Sebastian A, Hamad M, Omar HA, Orive G, Al-Tel TH. Camptothecin's journey from discovery to WHO Essential Medicine: Fifty years of promise. Eur J Med Chem 2021; 223:113639. [PMID: 34175539 DOI: 10.1016/j.ejmech.2021.113639] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022]
Abstract
Nature represents a rich source of compounds used for the treatment of many diseases. Camptothecin (CPT), isolated from the bark of Camptotheca acuminata, is a cytotoxic alkaloid that attenuates cancer cell replication by inhibiting DNA topoisomerase 1. Despite its promising and wide spectrum antiproliferative activity, its use is limited due to low solubility, instability, acquired tumour cell resistance, and remarkable toxicity. This has led to the development of numerous CPT analogues with improved pharmacodynamic and pharmacokinetic profiles. Three natural product-inspired drugs, namely, topotecan, irinotecan, and belotecan, are clinically approved and prescribed drugs for the treatment of several types of cancer, whereas other derivatives are in clinical trials. In this review, which covers literature from 2015 to 2020, we aim to provide a comprehensive overview and describe efforts that led to the development of a variety of CPT analogues. These efforts have led to the discovery of potent, first-in-class chemotherapeutic agents inspired by CPT. In addition, the mechanism of action, SAR studies, and recent advances of novel CPT drug delivery systems and antibody drug conjugates are discussed.
Collapse
Affiliation(s)
- Noura Khaiwa
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Noor R Maarouf
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Mhd H Darwish
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Dima W M Alhamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Anusha Sebastian
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Mohamad Hamad
- Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates; College of Health Sciences, 27272, Sharjah, United Arab Emirates
| | - Hany A Omar
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
| | - Taleb H Al-Tel
- College of Pharmacy, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, 27272, Sharjah, United Arab Emirates.
| |
Collapse
|