1
|
Vasic V, Dickopf S, Spranger N, Rosenberger RS, Fischer M, Mayer K, Larraillet V, Bates JA, Maier V, Sela T, Nussbaum B, Duerr H, Dengl S, Brinkmann U. Generation of binder-format-payload conjugate-matrices by antibody chain-exchange. Nat Commun 2024; 15:9406. [PMID: 39477939 PMCID: PMC11525586 DOI: 10.1038/s41467-024-53730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
The generation of antibody-drug conjugates with optimal functionality depends on many parameters. These include binder epitope, antibody format, linker composition, conjugation site(s), drug-to-antibody ratio, and conjugation method. The production of matrices that cover all possible parameters is a major challenge in identifying optimal antibody-drug conjugates. To address this bottleneck, we adapted our Format Chain Exchange technology (FORCE), originally established for bispecific antibodies, toward the generation of binder-format-payload matrices (pair-FORCE). Antibody derivatives with exchange-enabled Fc-heterodimers are combined with payload-conjugated Fc donors, and subsequent chain-exchange transfers payloads to antibody derivatives in different formats. The resulting binder-format-conjugate matrices can be generated with cytotoxic payloads, dyes, haptens, and large molecules, resulting in versatile tools for ADC screening campaigns. We show the relevance of pair-FORCE for identifying optimal HER2-targeting antibody-drug conjugates. Analysis of this matrix reveals that the notion of format-defines-function applies not only to bispecific antibodies, but also to antibody-drug conjugates.
Collapse
Affiliation(s)
- Vedran Vasic
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Steffen Dickopf
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Veraxa Biotech, Heidelberg, Germany
| | - Nadine Spranger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Institute of Molecular Immunology, School of Medicine and Health, Technical University Munich (TUM), Munich, Germany
| | - Rose-Sophie Rosenberger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Fischer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Klaus Mayer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Vincent Larraillet
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Jack A Bates
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Verena Maier
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Tatjana Sela
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Bianca Nussbaum
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Harald Duerr
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Stefan Dengl
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany.
| |
Collapse
|
2
|
Fan Z, Iqbal H, Ni J, Khan NU, Irshad S, Razzaq A, Alfaifi MY, Elbehairi SEI, Shati AA, Zhou J, Cheng H. Rationalized landscape on protein-based cancer nanomedicine: Recent progress and challenges. Int J Pharm X 2024; 7:100238. [PMID: 38511068 PMCID: PMC10951516 DOI: 10.1016/j.ijpx.2024.100238] [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: 01/11/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
The clinical advancement of protein-based nanomedicine has revolutionized medical professionals' perspectives on cancer therapy. Protein-based nanoparticles have been exploited as attractive vehicles for cancer nanomedicine due to their unique properties derived from naturally biomacromolecules with superior biocompatibility and pharmaceutical features. Furthermore, the successful translation of Abraxane™ (paclitaxel-based albumin nanoparticles) into clinical application opened a new avenue for protein-based cancer nanomedicine. In this mini-review article, we demonstrate the rational design and recent progress of protein-based nanoparticles along with their applications in cancer diagnosis and therapy from recent literature. The current challenges and hurdles that hinder clinical application of protein-based nanoparticles are highlighted. Finally, future perspectives for translating protein-based nanoparticles into clinic are identified.
Collapse
Affiliation(s)
- Zhechen Fan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Haroon Iqbal
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| | - Naveed Ullah Khan
- Department of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, China
| | - Shahla Irshad
- Department of Allied Health Sciences, Faculty of Health and Medical Sciences, Mirpur University of Science and Technology (MUST), Mirpur, Azad Jammu and Kashmir 10250, Pakistan
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mohammad Y. Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Ali A. Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
3
|
Katrini J, Boldrini L, Santoro C, Valenza C, Trapani D, Curigliano G. Biomarkers for Antibody-Drug Conjugates in Solid Tumors. Mol Cancer Ther 2024; 23:436-446. [PMID: 38363729 DOI: 10.1158/1535-7163.mct-23-0482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/18/2024]
Abstract
The clinical development and then the progressive entry in clinical practice of antibody-drug conjugates (ADC) have marked a transformative advancement in the overall cancer treatment. ADCs have been extensively tested for a large number of tumors, reporting heterogeneous clinical efficacy and safety results. In some diseases, the advent of ADCs has yielded significant changes in the prognostic trajectory, portending an improvement of the survival and/or quality of life. ADCs are targeted agents, capable of delivering highly cytotoxic payloads selectively to antigen-expressing cancer cells. As such, they have been intended as perfect "bullets" to enable the promise of precision medicine, toward high-efficacy and limited-toxicity treatment options. However, only some approved ADCs are intended for the use in biomarker-selected patient populations, restricting potentially the opportunity to be more precise. Yet, key characteristics of modern ADCs might allow the activity of ADCs in tumors with heterogeneous or low expression of cancer antigens, resulting in a clinical activity that could sublimate the classic paradigm of a drug-to-target perfect match. In our review, we portrayed the current landscape of approved ADCs, reporting data of activity as related to the expression of the cancer antigens, and elucidating possible determinants of the safety and efficacy, including when used in a therapeutic sequence.
Collapse
Affiliation(s)
- Jalissa Katrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Laura Boldrini
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Cherifi F, Da Silva A, Martins-Branco D, Awada A, Nader-Marta G. Pharmacokinetics and pharmacodynamics of antibody-drug conjugates for the treatment of patients with breast cancer. Expert Opin Drug Metab Toxicol 2024; 20:45-59. [PMID: 38214896 DOI: 10.1080/17425255.2024.2302460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
INTRODUCTION Currently three antibody-drug-conjugates (ADC) are approved by the European Medicines Agency (EMA) for treatment of breast cancer (BC) patient: trastuzumab-emtansine, trastuzumab-deruxtecan and sacituzumab-govitecan. ADC are composed of a monoclonal antibody (mAb) targeting a specific antigen, a cytotoxic payload and a linker. Pharmacokinetics (PK) and pharmacodynamics (PD) distinguish ADC from conventional chemotherapy and must be understood by clinicians. AREAS COVERED Our review delineates the PK/PD profiles of ADC approved for the treatment of BC with insight for future development. This is an expert opinion literature review based on the EMA's Assessment Reports, enriched by a comprehensive literature search performed on Medline in August 2023. EXPERT OPINION All three ADC distributions are described by a two-compartment structure: tissue and serum. Payload concentration peak is immediate but remains at low concentration. The distribution varied for all ADC only with body weight. mAb will be metabolised firstly by the saturable complex formation of ADC/Tumour-Receptor and secondly by binding of FcgRs in immune cells. They are all excreted in the bile and faeces with minimal urine elimination. Dose adjustments, apart from weight, are not recommended. Novel ADC are composed of cleavable linkers with various targets/payloads with the same PK/PD properties, but novel structures of ADC are in development.
Collapse
Affiliation(s)
- François Cherifi
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Breast Cancer Unit, CLCC François Baclesse, Institut Normand du Sein, Caen, France
| | - Angélique Da Silva
- Departments of Pharmacology and Medical Oncology, Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE, Caen, France
| | - Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| | - Ahmad Awada
- Oncology Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guilherme Nader-Marta
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (U.L.B), Brussels, Belgium
| |
Collapse
|
5
|
Hejmady S, Pradhan R, Kumari S, Pandey M, Dubey SK, Taliyan R. Pharmacokinetics and toxicity considerations for antibody-drug conjugates: an overview. Bioanalysis 2023; 15:1193-1202. [PMID: 37724472 DOI: 10.4155/bio-2023-0104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023] Open
Abstract
Antibody-drug conjugates (ADCs) is one of the fastest-growing drug-delivery systems. It involves a monoclonal antibody conjugated with payload via a ligand that directly targets the expressive protein of diseased cell. Hence, it reduces systemic exposure and provides site-specific delivery along with reduced toxicity. Because of this advantage, researchers have gained interest in this novel system. ADCs have displayed great promise in drug delivery and biomedical applications. However, a lack of understanding exists on their mechanisms of biodistribution, metabolism and side effects. To gain a better understanding of the therapeutics, careful consideration of the pharmacokinetics and toxicity needs to be undertaken. In this review, different pharmacokinetics parameters including distribution, bioanalysis and heterogeneity are discussed for developing novel therapeutics.
Collapse
Affiliation(s)
- Siddhanth Hejmady
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Rajesh Pradhan
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Shobha Kumari
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Meghna Pandey
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Sunil K Dubey
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India
- Medical Research, R&D Healthcare Division, Emami Ltd, Kolkata 700056, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Pilani Campus, Rajasthan, 333031, India
| |
Collapse
|
6
|
Lim RK, Buschman M, Khasanov A, Ledesma A, Chen J, Nguyen T, Guo J, Li L, Huang J, Niu J, Kerwin L, Wang R, Guo Y, Zhu T, Kaufmann G, Zhang Y, Zhou H, Ji H, Fu Y. Discovery of novel cMET-targeting antibody Fab drug conjugates as potential treatment for solid tumors with highly expressed cMET. Expert Opin Biol Ther 2023; 23:1137-1149. [PMID: 38078403 DOI: 10.1080/14712598.2023.2292633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Solid tumors are becoming prevalent affecting both old and young populations. Numerous solid tumors are associated with high cMET expression. The complexity of solid tumors combined with the highly interconnected nature of the cMET/HGF pathway with other cellular pathways make the pursuit of finding an effective treatment extremely challenging. The current standard of care for these malignancies is mostly small molecule-based chemotherapy. Antibody-based therapeutics as well as antibody drug conjugates are promising emerging classes against cMET-overexpressing solid tumors. RESEARCH DESIGN AND METHODS In this study, we described the design, synthesis, in vitro and in vivo characterization of cMET-targeting Fab drug conjugates (FDCs) as an alternative therapeutic strategy. The format is comprised of a Fab conjugated to a potent cytotoxic drug via a cleavable linker employing lysine-based and cysteine-based conjugation chemistries. RESULTS We found that the FDCs have potent anti-tumor efficacies in cancer cells with elevated overexpression of cMET. Moreover, they demonstrated a remarkable anti-tumor effect in a human gastric xenograft mouse model. CONCLUSIONS The FDC format has the potential to overcome some of the challenges presented by the other classes of therapeutics. This study highlights the promise of antibody fragment-based drug conjugate formats for the treatment of solid tumors.
Collapse
Affiliation(s)
- Reyna Kv Lim
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Matthew Buschman
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Alisher Khasanov
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Arthur Ledesma
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - John Chen
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Thanhtruc Nguyen
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Joanna Guo
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Lingna Li
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Jonathan Huang
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Jin Niu
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Lisa Kerwin
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Rengang Wang
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Yurong Guo
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Tong Zhu
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Gunnar Kaufmann
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Yanliang Zhang
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Heyou Zhou
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Henry Ji
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| | - Yanwen Fu
- Antibody Discovery & Technology, Sorrento Therapeutics, Inc, San Diego, CA, USA
| |
Collapse
|
7
|
Maiti R, Patel B, Patel N, Patel M, Patel A, Dhanesha N. Antibody drug conjugates as targeted cancer therapy: past development, present challenges and future opportunities. Arch Pharm Res 2023; 46:361-388. [PMID: 37071273 PMCID: PMC11345756 DOI: 10.1007/s12272-023-01447-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/26/2023] [Indexed: 04/19/2023]
Abstract
Antibody drug conjugates (ADCs) are promising cancer therapeutics with minimal toxicity as compared to small cytotoxic molecules alone and have shown the evidence to overcome resistance against tumor and prevent relapse of cancer. The ADC has a potential to change the paradigm of cancer chemotherapeutic treatment. At present, 13 ADCs have been approved by USFDA for the treatment of various types of solid tumor and haematological malignancies. This review covers the three structural components of an ADC-antibody, linker, and cytotoxic payload-along with their respective structure, chemistry, mechanism of action, and influence on the activity of ADCs. It covers comprehensive insight on structural role of linker towards efficacy, stability & toxicity of ADCs, different types of linkers & various conjugation techniques. A brief overview of various analytical techniques used for the qualitative and quantitative analysis of ADC is summarized. The current challenges of ADCs, such as heterogeneity, bystander effect, protein aggregation, inefficient internalization or poor penetration into tumor cells, narrow therapeutic index, emergence of resistance, etc., are outlined along with recent advances and future opportunities for the development of more promising next-generation ADCs.
Collapse
Affiliation(s)
- Ritwik Maiti
- Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Bhumika Patel
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| | - Nrupesh Patel
- Department of Pharmaceutical Analysis, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Mehul Patel
- Department of Pharmaceutical Chemistry and Analysis, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India
| | - Alkesh Patel
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, 388421, Gujarat, India
| | - Nirav Dhanesha
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
8
|
Wang Y, Zhang J, Han B, Tan L, Cai W, Li Y, Su Y, Yu Y, Wang X, Duan X, Wang H, Shi X, Wang J, Yang X, Liu T. Noncanonical amino acids as doubly bio-orthogonal handles for one-pot preparation of protein multiconjugates. Nat Commun 2023; 14:974. [PMID: 36810592 PMCID: PMC9944564 DOI: 10.1038/s41467-023-36658-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Genetic encoding of noncanonical amino acid (ncAA) for site-specific protein modification has been widely applied for many biological and therapeutic applications. To efficiently prepare homogeneous protein multiconjugates, we design two encodable noncanonical amino acids (ncAAs), 4-(6-(3-azidopropyl)-s-tetrazin-3-yl) phenylalanine (pTAF) and 3-(6-(3-azidopropyl)-s-tetrazin-3-yl) phenylalanine (mTAF), containing mutually orthogonal and bioorthogonal azide and tetrazine reaction handles. Recombinant proteins and antibody fragments containing the TAFs can easily be functionalized in one-pot reactions with combinations of commercially available fluorophores, radioisotopes, PEGs, and drugs in a plug-and-play manner to afford protein dual conjugates to assess combinations of tumor diagnosis, image-guided surgery, and targeted therapy in mouse models. Furthermore, we demonstrate that simultaneously incorporating mTAF and a ketone-containing ncAA into one protein via two non-sense codons allows preparation of a site-specific protein triconjugate. Our results demonstrate that TAFs are doubly bio-orthogonal handles for efficient and scalable preparation of homogeneous protein multiconjugates.
Collapse
Affiliation(s)
- Yong Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Jingming Zhang
- Department of Nuclear Medicine, Peking University First Hospital, 100034, Beijing, China
| | - Boyang Han
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Linzhi Tan
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Wenkang Cai
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Yuxuan Li
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Yeyu Su
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Yutong Yu
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Xin Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, 100034, Beijing, China
| | - Haoyu Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Xiaomeng Shi
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, 100034, Beijing, China. .,Institute of Medical Technology, Peking University Health Science Center, 100191, Beijing, China.
| | - Tao Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, 100191, Beijing, China.
| |
Collapse
|
9
|
Tarantino P, Tolaney SM. The Dawn of the Antibody-Drug Conjugates Era: How T-DM1 Reinvented the Future of Chemotherapy for Solid Tumors. Cancer Res 2022; 82:3659-3661. [PMID: 36245247 DOI: 10.1158/0008-5472.can-22-2324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022]
Abstract
Delivering targeted chemotherapy through antibody-drug conjugates (ADC) has emerged as an extremely effective therapeutic strategy for multiple types of cancer. The first agent of this class to be established for treating a solid tumor was trastuzumab emtansine (T-DM1), approved in 2013 for the treatment of HER2-positive metastatic breast cancer. Much of the knowledge that led to this approval came from the landmark Cancer Research publication by Lewis Phillips and colleagues in 2008, where they described the in vitro and in vivo efficacy, pharmacokinetics, and toxicity of T-DM1, demonstrating its relevant preclinical activity against HER2-positive breast cancer models. In this article, the authors also explored the use of different linkers to conjugate the cytotoxic payload to the trastuzumab vehicle, demonstrating improved stability, efficacy, and tolerability of the compound when adopting a specific thioether linker. The findings from this work not only set the stage for the clinical development of T-DM1, but also highlighted the modularity of ADCs, with small changes in their components able to dramatically impact their activity and toxicity. This finding would prove key for the development of novel ADCs, several of which are now reshaping the way we treat breast cancer and other cancer types. In this commentary, we discuss the key implications of the work by Phillips and colleagues, putting it in context of the current and anticipated expansion in the use of ADCs to treat cancer. See related article by Phillips et al., Cancer Res 2008;68:9280-90.
Collapse
Affiliation(s)
- Paolo Tarantino
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sara M Tolaney
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
10
|
Marei HE, Cenciarelli C, Hasan A. Potential of antibody-drug conjugates (ADCs) for cancer therapy. Cancer Cell Int 2022; 22:255. [PMID: 35964048 PMCID: PMC9375290 DOI: 10.1186/s12935-022-02679-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/05/2022] [Indexed: 11/10/2022] Open
Abstract
The primary purpose of ADCs is to increase the efficacy of anticancer medications by minimizing systemic drug distribution and targeting specific cells. Antibody conjugates (ADCs) have changed the way cancer is treated. However, because only a tiny fraction of patients experienced long-term advantages, current cancer preclinical and clinical research has been focused on combination trials. The complex interaction of ADCs with the tumor and its microenvironment appear to be reliant on the efficacy of a certain ADC, all of which have significant therapeutic consequences. Several clinical trials in various tumor types are now underway to examine the potential ADC therapy, based on encouraging preclinical results. This review tackles the potential use of ADCs in cancer therapy, emphasizing the essential processes underlying their positive therapeutic impacts on solid and hematological malignancies. Additionally, opportunities are explored to understand the mechanisms of ADCs action, the mechanism of resistance against ADCs, and how to overcome potential resistance following ADCs administration. Recent clinical findings have aroused interest, leading to a large increase in the number of ADCs in clinical trials. The rationale behind ADCs, as well as their primary features and recent research breakthroughs, will be discussed. We then offer an approach for maximizing the potential value that ADCs can bring to cancer patients by highlighting key ideas and distinct strategies.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
| |
Collapse
|
11
|
Intra-Domain Cysteines (IDC), a New Strategy for the Development of Original Antibody Fragment–Drug Conjugates (FDCs). Pharmaceutics 2022; 14:pharmaceutics14081524. [PMID: 35893780 PMCID: PMC9331466 DOI: 10.3390/pharmaceutics14081524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
Antibody–drug conjugates (ADCs) derived from a full immunoglobulin-G (IgG) are associated with suboptimal solid-tumor penetration and Fc-mediated toxicities. Antibody fragment–drug conjugates (FDCs) could be an alternative. Nevertheless, innovative solutions are needed to implant cysteines as conjugation sites in the single-chain fragment variable (scFv) format, which is the backbone from which many other antibody formats are built. In addition, the bioconjugation site has the utmost importance to optimize the safety and efficacy of bioconjugates. Our previous intra-tag cysteine (ITC) strategy consisted of introducing a bioconjugation motif at the C-terminal position of the 4D5.2 scFv, but this motif was subjected to proteolysis when the scFv was produced in CHO cells. Considering these data, using three intra-domain cysteine (IDC) strategies, several parameters were studied to assess the impact of different locations of a site-specific bioconjugation motif in the variable domains of an anti-HER2 scFv. In comparison to the ITC strategy, our new IDC strategy allowed us to identify new fragment–drug conjugates (FDCs) devoid of proteolysis and exhibiting enhanced stability profiles, better affinity, and better ability to kill selectively HER2-positive SK-BR-3 cells in vitro at picomolar concentrations. Thus, this work represents an important optimization step in the design of more complex and effective conjugates.
Collapse
|
12
|
Synthesis and Evaluation of Small Molecule Drug Conjugates Harnessing Thioester-Linked Maytansinoids. Pharmaceutics 2022; 14:pharmaceutics14071316. [PMID: 35890212 PMCID: PMC9323955 DOI: 10.3390/pharmaceutics14071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
Ligand-targeting drug conjugates are a class of clinically validated biopharmaceutical drugs constructed by conjugating cytotoxic drugs with specific disease antigen targeting ligands through appropriate linkers. The integrated linker-drug motif embedded within such a system can prevent the premature release during systemic circulation, thereby allowing the targeting ligand to engage with the disease antigen and selective accumulation. We have designed and synthesized new thioester-linked maytansinoid conjugates. By performing in vitro cytotoxicity, targeting ligand binding assay, and in vivo pharmacokinetic studies, we investigated the utility of this new linker-drug moiety in the small molecule drug conjugate (SMDC) system. In particular, we conjugated the thioester-linked maytansinoids to the phosphatidylserine-targeting small molecule zinc dipicolylamine and showed that Zn8_DM1 induced tumor regression in the HCC1806 triple-negative breast cancer xenograft model. Moreover, in a spontaneous sorafenib-resistant liver cancer model, Zn8_DM1 exhibited potent antitumor growth efficacy. From quantitative mRNA analysis of Zn8_DM1 treated-tumor tissues, we observed the elevation of gene expressions associated with a “hot inflamed tumor” state. With the identification and validation of a plethora of cancer-associated antigens in the “omics” era, this work provided the insight that antibody- or small molecule-based targeting ligands can be conjugated similarly to generate new ligand-targeting drug conjugates.
Collapse
|
13
|
Shah P, Chandra S. Review on emergence of nanomaterial coatings in bio-engineered cardiovascular stents. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Fertal SA, Poterala JE, Ponik SM, Wisinski KB. Stromal Characteristics and Impact on New Therapies for Metastatic Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1238. [PMID: 35267548 PMCID: PMC8909697 DOI: 10.3390/cancers14051238] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/25/2022] Open
Abstract
The heterogenous nature of triple-negative breast cancer (TNBC) is an underlying factor in therapy resistance, metastasis, and overall poor patient outcome. The lack of hormone and growth factor receptors lends to the use of chemotherapy as the first-line treatment for TNBC. However, the failure of chemotherapy demonstrates the need to develop novel immunotherapies, antibody-drug conjugates (ADCs), and other tumor- and stromal-targeted therapeutics for TNBC patients. The potential for stromal-targeted therapy is driven by studies indicating that the interactions between tumor cells and the stromal extracellular matrix (ECM) activate mechanisms of therapy resistance. Here, we will review recent outcomes from clinical trials targeting metastatic TNBC with immunotherapies aimed at programed death ligand-receptor interactions, and ADCs specifically linked to trophoblast cell surface antigen 2 (Trop-2). We will discuss how biophysical and biochemical cues from the ECM regulate the pathophysiology of tumor and stromal cells toward a pro-tumor immune environment, therapy resistance, and poor TNBC patient outcome. Moreover, we will highlight how ECM-mediated resistance is motivating the development of new stromal-targeted therapeutics with potential to improve therapy for this disease.
Collapse
Affiliation(s)
- Shelby A. Fertal
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Johanna E. Poterala
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| | - Suzanne M. Ponik
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Cell and Regenerative Biology, UW School of Medicine and Public Health, Madison, WI 53705, USA
| | - Kari B. Wisinski
- University of Wisconsin (UW) Carbone Cancer Center, Madison, WI 53705, USA; (S.A.F.); (J.E.P.); (S.M.P.)
- Department of Medicine, UW School of Medicine and Public Health, Madison, WI 53726, USA
| |
Collapse
|
15
|
Abstract
Polyclonal immunoglobulin (Ig) preparations have been used for several decades for treatment of primary and secondary immunodeficiencies and for treatment of some infections and intoxications. This has demonstrated the importance of Igs, also called antibodies (Abs) for prevention and elimination of infections. Moreover, elucidation of the structure and functions of Abs has suggested that they might be useful for targeted treatment of several diseases, including cancers and autoimmune diseases. The development of technologies for production of specific monoclonal Abs (MAbs) in large amounts has led to the production of highly effective therapeutic antibodies (TAbs), a collective term for MAbs (MAbs) with demonstrated clinical efficacy in one or more diseases. The number of approved TAbs is currently around hundred, and an even larger number is under development, including several engineered and modified Ab formats. The use of TAbs has provided new treatment options for many severe diseases, but prediction of clinical effect is difficult, and many patients eventually lose effect, possibly due to development of Abs to the TAbs or to other reasons. The therapeutic efficacy of TAbs can be ascribed to one or more effects, including binding and neutralization of targets, direct cytotoxicity, Ab-dependent complement-dependent cytotoxicity, Ab-dependent cellular cytotoxicity or others. The therapeutic options for TAbs have been expanded by development of several new formats of TAbs, including bispecific Abs, single domain Abs, TAb-drug conjugates, and the use of TAbs for targeted activation of immune cells. Most promisingly, current research and development can be expected to increase the number of clinical conditions, which may benefit from TAbs.
Collapse
Affiliation(s)
- Gunnar Houen
- Department of Neurology, Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
16
|
Fatima SW, Khare SK. Benefits and challenges of antibody drug conjugates as novel form of chemotherapy. J Control Release 2021; 341:555-565. [PMID: 34906604 DOI: 10.1016/j.jconrel.2021.12.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022]
Abstract
Antibody drug conjugates (ADCs) are an emerging therapeutic modality for targeted cancer treatment. They represent the unique amalgamation of chemotherapy and immunotherapy. ADCs comprise of monoclonal antibodies linked with drugs (payloads) through a chemical linker designed to deliver the cytotoxic moiety to the cancer cells. The present paper is a review of recent clinical advances of each component of ADCs (antibody/linker/payload) and how the individual component influences the activity of ADCs. The review discusses opportunities for improving ADCs efficiency and ways to have a better antibody-based molecular platform, which could substantially increase chemotherapy outcomes. This review casts an outlook on how ADCs enhancement in terms of their pharmacokinetics, therapeutic indexes and safety profiles can overcome the prevailing challenges like drug resistance in cancer treatment. A novel strategy of augmenting antibodies with nanoparticles anticipates a huge success in terms of targeted delivery of drugs in several diseases. Antibody conjugated nanoparticles (ACNPs) are a very promising strategy for the cutting-edge development of chemo/immunotherapies for efficient delivery of payloads at the targeted cancer cells. The avenues of a high drug to antibody ratio (DAR) owing to the selection of broad chemotherapy payloads, regulating drug release eliciting higher avidity of ACNPs over ADCs will be the modern immunotherapeutics. ACNPs carry immense potential to mark a paradigm shift in cancer chemotherapy that may be a substitute for ADCs.
Collapse
Affiliation(s)
- Syeda Warisul Fatima
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sunil K Khare
- Enzyme and Microbial Biochemistry Laboratory, Department of Chemistry, Indian Institute of Technology Delhi, New Delhi 110016, India.
| |
Collapse
|
17
|
Drago JZ, Modi S, Chandarlapaty S. Unlocking the potential of antibody-drug conjugates for cancer therapy. Nat Rev Clin Oncol 2021; 18:327-344. [PMID: 33558752 PMCID: PMC8287784 DOI: 10.1038/s41571-021-00470-8] [Citation(s) in RCA: 629] [Impact Index Per Article: 157.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Nine different antibody-drug conjugates (ADCs) are currently approved as cancer treatments, with dozens more in preclinical and clinical development. The primary goal of ADCs is to improve the therapeutic index of antineoplastic agents by restricting their systemic delivery to cells that express the target antigen of interest. Advances in synthetic biochemistry have ushered in a new generation of ADCs, which promise to improve upon the tissue specificity and cytotoxicity of their predecessors. Many of these drugs have impressive activity against treatment-refractory cancers, although hurdles impeding their broader use remain, including systemic toxicity, inadequate biomarkers for patient selection, acquired resistance and unknown benefit in combination with other cancer therapies. Emerging evidence indicates that the efficacy of a given ADC depends on the intricacies of how the antibody, linker and payload components interact with the tumour and its microenvironment, all of which have important clinical implications. In this Review, we discuss the current state of knowledge regarding the design, mechanism of action and clinical efficacy of ADCs as well as the apparent limitations of this treatment class. We then propose a path forward by highlighting several hypotheses and novel strategies to maximize the potential benefit that ADCs can provide to patients with cancer.
Collapse
Affiliation(s)
- Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weil Cornell Medicine, New York, NY, USA.
| | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weil Cornell Medicine, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Knudsen C, Jürgensen JA, Føns S, Haack AM, Friis RUW, Dam SH, Bush SP, White J, Laustsen AH. Snakebite Envenoming Diagnosis and Diagnostics. Front Immunol 2021; 12:661457. [PMID: 33995385 PMCID: PMC8113877 DOI: 10.3389/fimmu.2021.661457] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Snakebite envenoming is predominantly an occupational disease of the rural tropics, causing death or permanent disability to hundreds of thousands of victims annually. The diagnosis of snakebite envenoming is commonly based on a combination of patient history and a syndromic approach. However, the availability of auxiliary diagnostic tests at the disposal of the clinicians vary from country to country, and the level of experience within snakebite diagnosis and intervention may be quite different for clinicians from different hospitals. As such, achieving timely diagnosis, and thus treatment, is a challenge faced by treating personnel around the globe. For years, much effort has gone into developing novel diagnostics to support diagnosis of snakebite victims, especially in rural areas of the tropics. Gaining access to affordable and rapid diagnostics could potentially facilitate more favorable patient outcomes due to early and appropriate treatment. This review aims to highlight regional differences in epidemiology and clinical snakebite management on a global scale, including an overview of the past and ongoing research efforts within snakebite diagnostics. Finally, the review is rounded off with a discussion on design considerations and potential benefits of novel snakebite diagnostics.
Collapse
Affiliation(s)
- Cecilie Knudsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
- BioPorto Diagnostics A/S, Hellerup, Denmark
| | - Jonas A. Jürgensen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sofie Føns
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Aleksander M. Haack
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Rasmus U. W. Friis
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Søren H. Dam
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Sean P. Bush
- Department of Emergency Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Julian White
- Toxinology Department, Women’s and Children’s Hospital, North Adelaide, SA, Australia
| | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
19
|
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: 227] [Impact Index Per Article: 45.4] [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
|
20
|
Rahmati M, Silva EA, Reseland JE, A Heyward C, Haugen HJ. Biological responses to physicochemical properties of biomaterial surface. Chem Soc Rev 2020; 49:5178-5224. [PMID: 32642749 DOI: 10.1039/d0cs00103a] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biomedical scientists use chemistry-driven processes found in nature as an inspiration to design biomaterials as promising diagnostic tools, therapeutic solutions, or tissue substitutes. While substantial consideration is devoted to the design and validation of biomaterials, the nature of their interactions with the surrounding biological microenvironment is commonly neglected. This gap of knowledge could be owing to our poor understanding of biochemical signaling pathways, lack of reliable techniques for designing biomaterials with optimal physicochemical properties, and/or poor stability of biomaterial properties after implantation. The success of host responses to biomaterials, known as biocompatibility, depends on chemical principles as the root of both cell signaling pathways in the body and how the biomaterial surface is designed. Most of the current review papers have discussed chemical engineering and biological principles of designing biomaterials as separate topics, which has resulted in neglecting the main role of chemistry in this field. In this review, we discuss biocompatibility in the context of chemistry, what it is and how to assess it, while describing contributions from both biochemical cues and biomaterials as well as the means of harmonizing them. We address both biochemical signal-transduction pathways and engineering principles of designing a biomaterial with an emphasis on its surface physicochemistry. As we aim to show the role of chemistry in the crosstalk between the surface physicochemical properties and body responses, we concisely highlight the main biochemical signal-transduction pathways involved in the biocompatibility complex. Finally, we discuss the progress and challenges associated with the current strategies used for improving the chemical and physical interactions between cells and biomaterial surface.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway. h.j.haugen.odont.uio.no
| | | | | | | | | |
Collapse
|
21
|
Khowsathit J, Bazzoli A, Cheng H, Karanicolas J. Computational Design of an Allosteric Antibody Switch by Deletion and Rescue of a Complex Structural Constellation. ACS CENTRAL SCIENCE 2020; 6:390-403. [PMID: 32232139 PMCID: PMC7099597 DOI: 10.1021/acscentsci.9b01065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Indexed: 05/08/2023]
Abstract
Therapeutic monoclonal antibodies have transformed medicine, especially with regards to treating cancers and disorders of the immune system. More than 50 antibody-derived drugs have already reached the clinic, the majority of which target cytokines or cell-surface receptors. Unfortunately, many of these targets have pleiotropic functions: they serve multiple different roles, and often not all of these roles are disease-related. This can be problematic because antibodies act throughout the body, and systemic neutralization of such targets can lead to safety concerns. To address this, we have developed a strategy whereby an antibody's ability to recognize its antigen is modulated by a second layer of control, relying on addition of an exogenous small molecule. In previous studies, we began to explore this idea by introducing a deactivating tryptophan-to-glycine mutation in the domain-domain interface of a single-chain variable fragment (scFv), and then restoring activity by adding back indole to fit the designed cavity. Here, we now describe a novel computational strategy for enumerating larger cavities that can be formed by simultaneously introducing multiple adjacent large-to-small mutations; we then carry out a complementary virtual screen to identify druglike compounds to match each candidate cavity. We first demonstrate the utility of this strategy in a fluorescein-binding single-chain variable fragment (scFv) and experimentally characterize a triple mutant with reduced antigen-binding (Rip-3) that can be rescued using a complementary ligand (Stitch-3). Because our design is built upon conserved residues in the antibody framework, we then show that the same mutation/ligand pair can also be used to modulate antigen-binding in an scFv build from a completely unrelated framework. This set of residues is present in many therapeutic antibodies as well, suggesting that this mutation/ligand pair may serve as a general starting point for introducing ligand-dependence into many clinically relevant antibodies.
Collapse
Affiliation(s)
- Jittasak Khowsathit
- Program
in Molecular Therapeutics, Fox Chase Cancer
Center, Philadelphia, Pennsylvania 19111, United States
- Department of Molecular
Biosciences and Center for Computational Biology, University
of Kansas, Lawrence, Kansas 66045, United
States
| | - Andrea Bazzoli
- Department of Molecular
Biosciences and Center for Computational Biology, University
of Kansas, Lawrence, Kansas 66045, United
States
| | - Hong Cheng
- Program
in Molecular Therapeutics, Fox Chase Cancer
Center, Philadelphia, Pennsylvania 19111, United States
| | - John Karanicolas
- Program
in Molecular Therapeutics, Fox Chase Cancer
Center, Philadelphia, Pennsylvania 19111, United States
- Department of Molecular
Biosciences and Center for Computational Biology, University
of Kansas, Lawrence, Kansas 66045, United
States
| |
Collapse
|
22
|
Tsao KK, Lee AC, Racine KÉ, Keillor JW. Site-Specific Fluorogenic Protein Labelling Agent for Bioconjugation. Biomolecules 2020; 10:E369. [PMID: 32121143 PMCID: PMC7175205 DOI: 10.3390/biom10030369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 01/29/2023] Open
Abstract
Many clinically relevant therapeutic agents are formed from the conjugation of small molecules to biomolecules through conjugating linkers. In this study, two novel conjugating linkers were prepared, comprising a central coumarin core, functionalized with a dimaleimide moiety at one end and a terminal alkyne at the other. In our first design, we developed a protein labelling method that site-specifically introduces an alkyne functional group to a dicysteine target peptide tag that was genetically fused to a protein of interest. This method allows for the subsequent attachment of azide-functionalized cargo in the facile synthesis of novel protein-cargo conjugates. However, the fluorogenic aspect of the reaction between the linker and the target peptide was less than we desired. To address this shortcoming, a second linker reagent was prepared. This new design also allowed for the site-specific introduction of an alkyne functional group onto the target peptide, but in a highly fluorogenic and rapid manner. The site-specific addition of an alkyne group to a protein of interest was thus monitored in situ by fluorescence increase, prior to the attachment of azide-functionalized cargo. Finally, we also demonstrated that the cargo can also be attached first, in an azide/alkyne cycloaddition reaction, prior to fluorogenic conjugation with the target peptide-fused protein.
Collapse
Affiliation(s)
| | | | | | - Jeffrey W. Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (K.K.T.); (A.C.L.); (K.É.R.)
| |
Collapse
|
23
|
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: 0.6] [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
|
24
|
Abstract
The prototypical ADC mechanism involving antigen-mediated uptake and lysosomal release is both elegantly simple and scientifically compelling. However, recent clinical-stage failures have prompted a reevaluation of this delivery paradigm and have resulted in an array of new technologies that have the potential to improve the safety and efficacy of up and coming programs. These innovations can generally be categorized into seven areas that will be elaborated on in this chapter: (1) Exploiting new payload mechanisms; (2) Increasing the drug-antibody ratio (DAR); (3) Increasing the antibody penetration; (4) Overcoming ADC resistance mechanisms; (5) Increasing the efficiency of ADC uptake and processing; (6) Mitigating off-target payload exposure; and (7) Employment of noncytotoxic payloads. It is our belief that these seven areas capture the current "landscape" of innovations that are taking place in the design of next-generation ADCs. Together, these advancements are reshaping the ADC field and providing a path forward in the face of the recent clinical setbacks.
Collapse
Affiliation(s)
- L Nathan Tumey
- Department of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA.
- Pfizer Inc., Groton, CT, USA.
| |
Collapse
|
25
|
Khongorzul P, Ling CJ, Khan FU, Ihsan AU, Zhang J. Antibody–Drug Conjugates: A Comprehensive Review. Mol Cancer Res 2019; 18:3-19. [DOI: 10.1158/1541-7786.mcr-19-0582] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/22/2019] [Accepted: 10/22/2019] [Indexed: 11/16/2022]
|
26
|
Yang K, Chen B, Gianolio DA, Stefano JE, Busch M, Manning C, Alving K, Gregory RC, Brondyk WH, Miller RJ, Dhal PK. Convergent synthesis of hydrophilic monomethyl dolastatin 10 based drug linkers for antibody-drug conjugation. Org Biomol Chem 2019; 17:8115-8124. [PMID: 31460552 DOI: 10.1039/c9ob01639b] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We report a modular approach to synthesize maleimido group containing hydrophilic dolastatin 10 (Dol10) derivatives as drug-linkers for the syntheses of antibody-drug conjugates (ADCs). Discrete polyethylene glycol (PEG) moieties of different chain lengths were introduced as part of the linker to impart hydrophilicity to these drug linkers. The synthesis process involved construction of PEG maleimido derivatives of the tetrapeptide intermediate (N-methylvaline-valine-dolaisoleucine-dolaproine), which were subsequently coupled with dolaphenine to generate the desired drug linkers. The synthetic method reported in this manuscript circumvents the use of highly cytotoxic Dol10 in its native form. By using trastuzumab (Herceptin®) as the antibody we have synthesized Dol10 containing ADCs. The presence of a discrete PEG chain in the drug linkers resulted in ADCs free from aggregation. The effect of PEG chain length on the biological activities of these Dol10 containing ADCs was investigated by in vitro cytotoxicity assays. ADCs containing PEG6 and PEG8 spacers exhibited the highest level of in vitro anti-proliferative activity against HER2-positive (SK-BR-3) human tumor cells. ADCs derived from Herceptin® and PEG8-Dol10, at a dose of 10 mg kg-1, effectively delayed the tumor growth and prolonged the survival time in mice bearing human ovarian SKOV-3 xenografts.
Collapse
Affiliation(s)
- Kanwen Yang
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Bo Chen
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | - James E Stefano
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Michelle Busch
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | - Kim Alving
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | | | | | - Robert J Miller
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| | - Pradeep K Dhal
- Sanofi Global R&D, 153 Second Avenue, Waltham, MA 02139, USA.
| |
Collapse
|
27
|
Liu YW, Chen YY, Hsu CY, Chiu TY, Liu KL, Lo CF, Fang MY, Huang YC, Yeh TK, Pak KY, Gray BD, Hsu TA, Huang KH, Shih C, Shia KS, Chen CT, Tsou LK. Linker Optimization and Therapeutic Evaluation of Phosphatidylserine-Targeting Zinc Dipicolylamine-based Drug Conjugates. J Med Chem 2019; 62:6047-6062. [PMID: 31181158 DOI: 10.1021/acs.jmedchem.9b00173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We report that compound 13, a novel phosphatidylserine-targeting zinc(II) dipicolylamine drug conjugate, readily triggers a positive feedback therapeutic loop through the in situ generation of phosphatidylserine in the tumor microenvironment. Linker modifications, pharmacokinetics profiling, in vivo antitumor studies, and micro-Western array of treated-tumor tissues were employed to show that this class of conjugates induced regeneration of apoptotic signals, which facilitated subsequent recruitment of the circulating conjugates through the zinc(II) dipicolylamine-phosphatidylserine association and resulted in compounding antitumor efficacy. Compared to the marketed compound 17, compound 13 not only induced regressions in colorectal and pancreatic tumor models, it also exhibited at least 5-fold enhancement in antitumor efficacy with only 40% of the drug employed during treatment, culminating in a >12.5-fold increase in therapeutic potential. Our study discloses a chemically distinct apoptosis-targeting theranostic, with built-in complementary functional moieties between the targeting module and the drug mechanism to expand the arsenal of antitumor therapy.
Collapse
Affiliation(s)
- Yu-Wei Liu
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Yun-Yu Chen
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Chia-Yu Hsu
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Tai-Yu Chiu
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Yu-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc. , West Chester , Pennsylvania 19380 , United States
| | - Brian D Gray
- Molecular Targeting Technologies, Inc. , West Chester , Pennsylvania 19380 , United States
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Kuan-Hsun Huang
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| | - Lun K Tsou
- Institute of Biotechnology and Pharmaceutical Research , National Health Research Institutes , Miaoli 35053 , Taiwan , ROC
| |
Collapse
|
28
|
Liu W, Zhao W, Bai X, Jin S, Li Y, Qiu C, Pan L, Ding D, Xu Y, Zhou Z, Chen S. High antitumor activity of Sortase A-generated anti-CD20 antibody fragment drug conjugates. Eur J Pharm Sci 2019; 134:81-92. [DOI: 10.1016/j.ejps.2019.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/27/2019] [Accepted: 04/11/2019] [Indexed: 12/29/2022]
|
29
|
Aubrey N, Allard-Vannier E, Martin C, Bryden F, Letast S, Colas C, Lakhrif Z, Collinet N, Dimier-Poisson I, Chourpa I, Viaud-Massuard MC, Joubert N. Site-Specific Conjugation of Auristatins onto Engineered scFv Using Second Generation Maleimide to Target HER2-positive Breast Cancer in Vitro. Bioconjug Chem 2018; 29:3516-3521. [DOI: 10.1021/acs.bioconjchem.8b00668] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Nicolas Aubrey
- ISP UMR 1282, INRA, Université de Tours, Team BioMAP, 31 avenue Monge, 37200 Tours, France
| | | | - Camille Martin
- GICC EA7501, Université de Tours, Team IMT, 31 avenue Monge, 37200 Tours, France
| | - Francesca Bryden
- GICC EA7501, Université de Tours, Team IMT, 31 avenue Monge, 37200 Tours, France
| | - Stéphanie Letast
- GICC EA7501, Université de Tours, Team IMT, 31 avenue Monge, 37200 Tours, France
| | - Cyril Colas
- ICOA UMR 7311, Université d’Orléans, CNRS, rue de Chartres, 45067 Orléans, France
- CBM UPR 4311, CNRS, Université d’Orléans, rue Charles Sadron, 45071 Orléans, France
| | - Zineb Lakhrif
- ISP UMR 1282, INRA, Université de Tours, Team BioMAP, 31 avenue Monge, 37200 Tours, France
| | - Nils Collinet
- NMNS EA 6295, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | | | - Igor Chourpa
- NMNS EA 6295, Université de Tours, 31 avenue Monge, 37200 Tours, France
| | | | - Nicolas Joubert
- GICC EA7501, Université de Tours, Team IMT, 31 avenue Monge, 37200 Tours, France
| |
Collapse
|
30
|
Rossin R, Versteegen RM, Wu J, Khasanov A, Wessels HJ, Steenbergen EJ, Ten Hoeve W, Janssen HM, van Onzen AHAM, Hudson PJ, Robillard MS. Chemically triggered drug release from an antibody-drug conjugate leads to potent antitumour activity in mice. Nat Commun 2018; 9:1484. [PMID: 29728559 PMCID: PMC5935733 DOI: 10.1038/s41467-018-03880-y] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/20/2018] [Indexed: 11/25/2022] Open
Abstract
Current antibody-drug conjugates (ADCs) target internalising receptors on cancer cells leading to intracellular drug release. Typically, only a subset of patients with solid tumours has sufficient expression of such a receptor, while there are suitable non-internalising receptors and stroma targets. Here, we demonstrate potent therapy in murine tumour models using a non-internalising ADC that releases its drugs upon a click reaction with a chemical activator, which is administered in a second step. This was enabled by the development of a diabody-based ADC with a high tumour uptake and very low retention in healthy tissues, allowing systemic administration of the activator 2 days later, leading to efficient and selective activation throughout the tumour. In contrast, the analogous ADC comprising the protease-cleavable linker used in the FDA approved ADC Adcetris is not effective in these tumour models. This first-in-class ADC holds promise for a broader applicability of ADCs across patient populations. Current antibody-drug conjugates (ADCs) target internalising receptors on cancer cells. Here, the authors report the development and in vivo validation of a non-internalising ADC with the capacity to target cancer cells and release its therapeutic cargo extracellularly via a chemical trigger.
Collapse
Affiliation(s)
- Raffaella Rossin
- Tagworks Pharmaceuticals, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Ron M Versteegen
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | - Jeremy Wu
- Avipep Pty Ltd, 343 Royal Parade, Parkville, VIC, 3052, Australia
| | - Alisher Khasanov
- Levena Biopharma, 4955 Directors Place, Suite 300, San Diego, CA, 92121, USA
| | - Hans J Wessels
- Radboud Proteomics Centre, Department of Laboratory Medicine, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Erik J Steenbergen
- Department of Pathology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - Henk M Janssen
- SyMO-Chem B.V., Den Dolech 2, 5612 AZ, Eindhoven, The Netherlands
| | | | - Peter J Hudson
- Avipep Pty Ltd, 343 Royal Parade, Parkville, VIC, 3052, Australia
| | - Marc S Robillard
- Tagworks Pharmaceuticals, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
31
|
Pye H, Butt MA, Funnell L, Reinert HW, Puccio I, Rehman Khan SU, Saouros S, Marklew JS, Stamati I, Qurashi M, Haidry R, Sehgal V, Oukrif D, Gandy M, Whitaker HC, Rodriguez-Justo M, Novelli M, Hamoudi R, Yahioglu G, Deonarain MP, Lovat LB. Using antibody directed phototherapy to target oesophageal adenocarcinoma with heterogeneous HER2 expression. Oncotarget 2018; 9:22945-22959. [PMID: 29796164 PMCID: PMC5955430 DOI: 10.18632/oncotarget.25159] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/28/2018] [Indexed: 12/22/2022] Open
Abstract
Early oesophageal adenocarcinoma (OA) and pre-neoplastic dysplasia may be treated with endoscopic resection and ablative techniques such as photodynamic therapy (PDT). Though effective, discrete areas of disease may be missed leading to recurrence. PDT further suffers from the side effects of off-target photosensitivity. A tumour specific and light targeted therapeutic agent with optimised pharmacokinetics could be used to destroy residual cancerous cells left behind after resection. A small molecule antibody-photosensitizer conjugate was developed targeting human epidermal growth factor receptor 2 (HER2). This was tested in an in vivo mouse model of human OA using a xenograft flank model with clinically relevant low level HER2 expression and heterogeneity. In vitro we demonstrate selective binding of the conjugate to tumour versus normal tissue. Light dependent cytotoxicity of the phototherapy agent in vitro was observed. In an in vivo OA mouse xenograft model the phototherapy agent had desirable pharmacokinetic properties for tumour uptake and blood clearance time. PDT treatment caused tumour growth arrest in all the tumours despite the tumours having a clinically defined low/negative HER2 expression level. This new phototherapy agent shows therapeutic potential for treatment of both HER2 positive and borderline/negative OA.
Collapse
Affiliation(s)
- Hayley Pye
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Mohammed Adil Butt
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| | - Laura Funnell
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Halla W Reinert
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Ignazio Puccio
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Saif U Rehman Khan
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Savvas Saouros
- Antikor BioPharma, Stevenage, UK.,Imperial College London, London, UK
| | | | | | - Maryam Qurashi
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Imperial College London, London, UK
| | - Rehan Haidry
- Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| | - Vinay Sehgal
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| | - Dahmane Oukrif
- Department of Pathology, University College London, London, UK
| | - Michael Gandy
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | - Hayley C Whitaker
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK
| | | | - Marco Novelli
- Department of Pathology, University College London, London, UK
| | - Rifat Hamoudi
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, UAE
| | - Gokhan Yahioglu
- Antikor BioPharma, Stevenage, UK.,Imperial College London, London, UK
| | - Mahendra P Deonarain
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Antikor BioPharma, Stevenage, UK.,Imperial College London, London, UK
| | - Laurence B Lovat
- Department for Tissue and Energy, Division of Surgery and Interventional Science, University College London, London, UK.,Upper Gastrointestinal Service, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
32
|
Kaiser CE, Rincon Pabon JP, Khowsathit J, Castaldi MP, Kazmirski SL, Weis DD, Zhang AX, Karanicolas J. Modulating Antibody Structure and Function through Directed Mutations and Chemical Rescue. ACS Synth Biol 2018; 7:1152-1162. [PMID: 29609459 DOI: 10.1021/acssynbio.8b00124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Monoclonal antibody therapeutics have revolutionized the treatment of diseases such as cancer and autoimmune disorders, and also serve as research reagents for diverse and unparalleled applications. To extend their utility in both contexts, we have begun development of tunable antibodies, whose activity can be controlled by addition of a small molecule. Conceptually, we envision that incorporating cavity-forming mutations into an antibody can disrupt its structure, thereby reducing its affinity for antigen; addition of a small molecule may then restore the active structure, and thus rescue antigen binding. As a first proof of concept toward implementing this strategy, we have incorporated individual tryptophan to glycine mutations into FITC-E2, an anti-fluorescein single-chain variable fragment (scFv). We find that these can disrupt the protein structure and diminish antigen binding, and further that both structure and function can be rescued by addition of indole to complement the deleted side chain. While the magnitude of the affinity difference triggered by indole is modest in this first model system, it nonetheless provides a framework for future mutation/ligand pairs that may induce more dramatic responses. Disrupting and subsequently rescuing antibody activity, as exemplified by this first example, may represent a new approach to "design in" fine-tuned control of antibody activity for a variety of future applications.
Collapse
Affiliation(s)
- Christine E. Kaiser
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Juan Pablo Rincon Pabon
- Department of Chemistry and Ralph Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Jittasak Khowsathit
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, United States
| | - M. Paola Castaldi
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Steven L. Kazmirski
- Structure and Biophysics, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - David D. Weis
- Department of Chemistry and Ralph Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, Kansas 66045, United States
| | - Andrew X. Zhang
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts 02451, United States
| | - John Karanicolas
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, United States
| |
Collapse
|
33
|
Warnders FJ, Lub-de Hooge MN, de Vries EGE, Kosterink JGW. Influence of protein properties and protein modification on biodistribution and tumor uptake of anticancer antibodies, antibody derivatives, and non-Ig scaffolds. Med Res Rev 2018; 38:1837-1873. [PMID: 29635825 DOI: 10.1002/med.21498] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/30/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022]
Abstract
Newly developed protein drugs that target tumor-associated antigens are often modified in order to increase their therapeutic effect, tumor exposure, and safety profile. During the development of protein drugs, molecular imaging is increasingly used to provide additional information on their in vivo behavior. As a result, there are increasing numbers of studies that demonstrate the effect of protein modification on whole body distribution and tumor uptake of protein drugs. However, much still remains unclear about how to interpret obtained biodistribution data correctly. Consequently, there is a need for more insight in the correct way of interpreting preclinical and clinical imaging data. Summarizing the knowledge gained to date may facilitate this interpretation. This review therefore provides an overview of specific protein properties and modifications that can affect biodistribution and tumor uptake of anticancer antibodies, antibody fragments, and nonimmunoglobulin scaffolds. Protein properties that are discussed in this review are molecular size, target interaction, FcRn binding, and charge. Protein modifications that are discussed are radiolabeling, fluorescent labeling drug conjugation, glycosylation, humanization, albumin binding, and polyethylene glycolation.
Collapse
Affiliation(s)
- Frank-Jan Warnders
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjolijn N Lub-de Hooge
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jos G W Kosterink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,PharmacoTherapy, Epidemiology & Economy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Deonarain MP, Yahioglu G, Stamati I, Pomowski A, Clarke J, Edwards BM, Diez-Posada S, Stewart AC. Small-Format Drug Conjugates: A Viable Alternative to ADCs for Solid Tumours? Antibodies (Basel) 2018; 7:E16. [PMID: 31544868 PMCID: PMC6698822 DOI: 10.3390/antib7020016] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/16/2022] Open
Abstract
Antibody-Drug Conjugates (ADCs) have been through multiple cycles of technological innovation since the concept was first practically demonstrated ~40 years ago. Current technology is focusing on large, whole immunoglobulin formats (of which there are approaching 100 in clinical development), many with site-specifically conjugated payloads numbering 2 or 4. Despite the success of trastuzumab-emtansine in breast cancer, ADCs have generally failed to have an impact in solid tumours, leading many to explore alternative, smaller formats which have better penetrating properties as well as more rapid pharmacokinetics (PK). This review describes research and development progress over the last ~10 years obtained from the primary literature or conferences covering over a dozen different smaller format-drug conjugates from 80 kDa to around 1 kDa in total size. In general, these agents are potent in vitro, particularly more recent ones incorporating ultra-potent payloads such as auristatins or maytansinoids, but this potency profile changes when testing in vivo due to the more rapid clearance. Strategies to manipulate the PK properties, whilst retaining the more effective tumour penetrating properties could at last make small-format drug conjugates viable alternative therapeutics to the more established ADCs.
Collapse
Affiliation(s)
- Mahendra P Deonarain
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
- Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, UK.
| | - Gokhan Yahioglu
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
- Department of Chemistry, Imperial College London, Exhibition Road, London SW72AZ, UK.
| | - Ioanna Stamati
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Anja Pomowski
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - James Clarke
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Bryan M Edwards
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Soraya Diez-Posada
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| | - Ashleigh C Stewart
- Antikor Biopharma Ltd., Stevenage Bioscience Catalyst, Gunnels Wood Road, Stevenage Herts SG12FX, UK.
| |
Collapse
|
35
|
Beaudoin S, Paquette M, Fafard-Couture L, Tremblay MA, Lecomte R, Guérin B, Leyton JV. Initial Evaluation of Antibody-conjugates Modified with Viral-derived Peptides for Increasing Cellular Accumulation and Improving Tumor Targeting. J Vis Exp 2018. [PMID: 29578523 DOI: 10.3791/55440] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Antibody-conjugates (ACs) modified with virus-derived peptides are a potentially powerful class of tumor cell delivery agents for molecular payloads used in cancer treatment and imaging due to increased cellular accumulation over current ACs. During early AC in vitro development, fluorescence techniques and radioimmunoassays are sufficient for determining intracellular localization, accumulation efficiency, and target cell specificity. Currently, there is no consensus on standardized methods for preparing cells for evaluating AC intracellular accumulation and localization. The initial testing of ACs modified with virus-derived peptides is critical especially if several candidates have been constructed. Determining intracellular accumulation by fluorescence can be affected by background signal from ACs at the cell surface and complicate the interpretation of accumulation. For radioimmunoassays, typically treated cells are fractionated and the radioactivity in different cell compartments measured. However, cell lysis varies from cell to cell and often nuclear and cytoplasmic compartments are not adequately isolated. This can produce misleading data on payload delivery properties. The intravenous injection of radiolabeled virus-derived peptide-modified ACs in tumor bearing mice followed by radionuclide imaging is a powerful method for determining tumor targeting and payload delivery properties at the in vivo phase of development. However, this is a relatively recent advancement and few groups have evaluated virus-derived peptide-modified ACs in this manner. We describe the processing of treated cells to more accurately evaluate virus-derived peptide-modified AC accumulation when using confocal microscopy and radioimmunoassays. Specifically, a method for trypsinizing cells to remove cell surface bound ACs. We also provide a method for improving cellular fractionation. Lastly, this protocol provides an in vivo method using positron emission tomography (PET) for evaluating initial tumor targeting properties in tumor-bearing mice. We use the radioisotope 64Cu (t1/2 = 12.7 h) as an example payload in this protocol.
Collapse
Affiliation(s)
- Simon Beaudoin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | - Michel Paquette
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | | | - Mylene A Tremblay
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke
| | - Roger Lecomte
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology
| | - Jeffrey V Leyton
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke; Sherbrooke Molecular Imaging Center (CIMS), Université de Sherbrooke; Sherbrooke Institute of Pharmacology;
| |
Collapse
|
36
|
Martin C, Kizlik-Masson C, Pèlegrin A, Watier H, Viaud-Massuard MC, Joubert N. Antibody-drug conjugates: Design and development for therapy and imaging in and beyond cancer, LabEx MAbImprove industrial workshop, July 27-28, 2017, Tours, France. MAbs 2018; 10:210-221. [PMID: 29239690 PMCID: PMC5825198 DOI: 10.1080/19420862.2017.1412130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The annual "Antibody Industrial Symposium", co organized by LabEx MAbImprove, MabDesign and Polepharma, was held in Tours, France on June 27-28, 2017. The focus was on antibody-drug-conjugates (ADCs), new entities which realize the hope of Paul Ehrlich's magic bullet. ADCs result from the bioconjugation of a highly cytotoxic drug to a selective monoclonal antibody, which acts as a vector. Building on knowledge gained during the development of three approved ADCs, brentuximab vedotin (Adcetris®), ado trastuzumab emtansine (Kadcyla®) and inotuzumab ozogamicin (Besponsa®), and the many ADCs in development, this meeting addressed strategies and the latest innovations in the field from fundamental research to manufacturing.
Collapse
Affiliation(s)
- Camille Martin
- a Equipe 4 IMT GICC, Université François Rabelais , Tours , France
| | | | - André Pèlegrin
- c IRCM, Institut de Recherche en Cancérologie de Montpellier , Université de Montpellier, Institut régional du Cancer de Montpellier , Montpellier , France
| | - Hervé Watier
- b Equipe 1 FRAME GICC, Université François Rabelais , Tours , France.,d Service d'Immunologie, CHRU de Tours , Tours , France
| | | | - Nicolas Joubert
- a Equipe 4 IMT GICC, Université François Rabelais , Tours , France
| |
Collapse
|
37
|
Montoliu-Gaya L, Murciano-Calles J, Martinez JC, Villegas S. Towards the improvement in stability of an anti-Aβ single-chain variable fragment, scFv-h3D6, as a way to enhance its therapeutic potential. Amyloid 2017; 24:167-175. [PMID: 28699800 DOI: 10.1080/13506129.2017.1348347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ScFv-h3D6 is a single-chain variable fragment derived from the monoclonal antibody bapineuzumab that prevents Aβ-induced cytotoxicity by capturing Aβ oligomers. The benefits of scFv-h3D6 treatment in Alzheimer's disease are known at the behavioural, cellular and molecular levels in the 3xTg-AD mouse model. Antibody-based therapeutics are only stable in a limited temperature range, so their benefit in vivo depends on their capability for maintaining the proper fold. Here, we have stabilized the scFv-h3D6 folding by introducing the mutation VH-K64R and combining it with the previously described elongation of the VL domain (C3). The stabilities of the different scFv-h3D6 constructs were calculated from urea and thermal denaturation followed by Trp-fluorescence, CD and DSC and resulted in the order C3 > K64R/C3 > VH-K64R ≥ scFv-h3D6; showing that the combination of both mutations was not additive, instead they partially cancelled each other. The three mutants assayed showed a decreased aggregation tendency but maintained their capability to aggregate in the form of worm-like fibrils, basis of the protective effect of scFv-h3D6. Cytotoxicity assays showed that all the mutants recovered cell viability of Aβ-treated neuroblastoma cell cultures in a dose-dependent manner and with efficiencies that correlated with stability, therefore improving the therapeutic ability of this antibody.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- a Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular , Unitat de Biociències, Universitat Autònoma de Barcelona , Spain
| | - Javier Murciano-Calles
- b Department of Physical Chemistry and Institute of Biotechnology, Faculty of Sciences , University of Granada, Campus Fuentenueva , Granada , Spain
| | - Jose C Martinez
- b Department of Physical Chemistry and Institute of Biotechnology, Faculty of Sciences , University of Granada, Campus Fuentenueva , Granada , Spain
| | - Sandra Villegas
- a Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular , Unitat de Biociències, Universitat Autònoma de Barcelona , Spain
| |
Collapse
|
38
|
Doubly Caged Linker for AND-Type Fluorogenic Construction of Protein/Antibody Bioconjugates and In Situ Quantification. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201702748] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Liu G, Shi G, Sheng H, Jiang Y, Liang H, Liu S. Doubly Caged Linker for AND-Type Fluorogenic Construction of Protein/Antibody Bioconjugates and In Situ Quantification. Angew Chem Int Ed Engl 2017; 56:8686-8691. [PMID: 28524357 DOI: 10.1002/anie.201702748] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Indexed: 12/15/2022]
Abstract
In situ quantification of the conjugation efficiency of azide-terminated synthetic polymers/imaging probes and thiol-functionalized antibodies/proteins/peptides was enabled by a doubly caged profluorescent and heterodifunctional core molecule C1 as a self-sorting bridging unit. Orthogonal dual "click" coupling of C1 with azide- and thiol-functionalized precursors led to highly fluorescent bioconjugates, whereas single-click products remained essentially nonfluorescent. Integration with FRET processes was also possible. For the construction of antibody-probe conjugates from an anti-carcinoembryonic antigen and a quinone-caged profluorescent naphthalimide derivative, the dual "click" coupling process with C1 was monitored on the basis of the emission turn-on of C1, whereas prominent changes in FRET ratios occurred for antibody-imaging-probe conjugates when specifically triggered by quinone oxidoreductase (NQO1), which is overexpressed in various types of cancer cells.
Collapse
Affiliation(s)
- Guhuan Liu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'An Road, Shanghai, 200032, China
| | - Haoyue Sheng
- Department of Urology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'An Road, Shanghai, 200032, China
| | - Yanyan Jiang
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, China
| | - Haojun Liang
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, iChem (Collaborative Innovation Center of Chemistry for Energy Materials), Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui Province, 230026, China
| |
Collapse
|
40
|
Liu YW, Shia KS, Wu CH, Liu KL, Yeh YC, Lo CF, Chen CT, Chen YY, Yeh TK, Chen WH, Jan JJ, Huang YC, Huang CL, Fang MY, Gray BD, Pak KY, Hsu TA, Huang KH, Tsou LK. Targeting Tumor Associated Phosphatidylserine with New Zinc Dipicolylamine-Based Drug Conjugates. Bioconjug Chem 2017; 28:1878-1892. [PMID: 28581724 DOI: 10.1021/acs.bioconjchem.7b00225] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A series of zinc(II) dipicolylamine (ZnDPA)-based drug conjugates have been synthesized to probe the potential of phosphatidylserine (PS) as a new antigen for small molecule drug conjugate (SMDC) development. Using in vitro cytotoxicity and plasma stability studies, PS-binding assay, in vivo pharmacokinetic studies, and maximum tolerated dose profiles, we provided a roadmap and the key parameters required for the development of the ZnDPA based drug conjugate. In particular, conjugate 24 induced tumor regression in the COLO 205 xenograft model and exhibited a more potent antitumor effect with a 70% reduction of cytotoxic payload compared to that of the marketed irinotecan when dosed at the same regimen. In addition to the validation of PS as an effective pharmacodelivery target for SMDC, our work also provided the foundation that, if applicable, a variety of therapeutic agents could be conjugated in the same manner to treat other PS-associated diseases.
Collapse
Affiliation(s)
- Yu-Wei Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chien-Huang Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kuan-Liang Liu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yu-Cheng Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chen-Fu Lo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yun-Yu Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Wei-Han Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Jiing-Jyh Jan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Ming-Yu Fang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Brian D Gray
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Koon Y Pak
- Molecular Targeting Technologies, Inc. , West Chester, Pennsylvania 19380, United States
| | - Tsu-An Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Kuan-Hsun Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| | - Lun K Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes , Miaoli 35053, Taiwan, ROC
| |
Collapse
|
41
|
Spidel JL, Albone EF, Cheng X, Vaessen B, Jacob S, Milinichik AZ, Verdi A, Kline JB, Grasso L. Engineering humanized antibody framework sequences for optimal site-specific conjugation of cytotoxins. MAbs 2017; 9:907-915. [PMID: 28541812 DOI: 10.1080/19420862.2017.1330734] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The prevailing techniques used to generate antibody-drug conjugates (ADCs) involve random conjugation of the linker-drug to multiple lysines or cysteines in the antibody. Engineering natural and non-natural amino acids into an antibody has been demonstrated to be an effective strategy to produce homogeneous ADC products with defined drug-to-antibody ratios. We recently reported an efficient residue-specific conjugation technology (RESPECT) where thiol-reactive payloads can be efficiently conjugated to a native unpaired cysteine in position 80 (C80) of rabbit light chains. Deimmunizing the rabbit variable domains through humanization is necessary to reduce the risk of anti-drug antibody responses in patients. However, we found that first-generation humanized RESPECT ADCs showed high levels of aggregation and low conjugation efficiency. We correlated these negative properties to the phenylalanine at position 83 present in most human variable kappa frameworks. When position 83 was substituted with selected amino acids, conjugation was restored and aggregation was reduced to levels similar to the chimeric ADC. This engineering strategy allows for development of second-generation humanized RESPECT ADCs with desirable biopharmaceutical properties.
Collapse
|
42
|
Albone EF, Spidel JL, Cheng X, Park YC, Jacob S, Milinichik AZ, Vaessen B, Butler J, Kline JB, Grasso L. Generation of therapeutic immunoconjugates via Residue-Specific Conjugation Technology (RESPECT) utilizing a native cysteine in the light chain framework of Oryctolagus cuniculus. Cancer Biol Ther 2017; 18:347-357. [PMID: 28394698 DOI: 10.1080/15384047.2017.1312232] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The conjugation of toxins, dyes, peptides, or proteins to monoclonal antibodies is often performed via free thiol groups generated by either partial reduction methods or engineering free cysteine residues into the antibody sequence. Antibodies from the rabbit Oryctolagus cuniculus have an additional intrachain disulfide bond, whereby the light chain variable kappa domain is bridged to the constant kappa region between cysteine residues at positions 80 and 171, respectively. Chimerization of rabbit antibodies with human constant domains allows for the generation of a free thiol group at the light chain position 80 (C80) that can be used for site-specific conjugation. An efficient process for the purification and simultaneous removal of cysteinylation at the C80 site was developed. The unpaired C80 was shown to be efficiently conjugated using several different maleimido-based ligands. REsidue SPEcific Conjugation Technology (RESPECT) antibody-drug conjugates prepared using rabbit-human chimeric anti-human mesothelin rabbit antibodies and maleimido-PEG2-auristatin conjugated to C80 were shown to be highly potent and specific in vitro and effective in vivo in reduction of tumor growth in a highly aggressive mesothelin-expressing xenograft tumor model.
Collapse
|
43
|
Beck A, Goetsch L, Dumontet C, Corvaïa N. Strategies and challenges for the next generation of antibody-drug conjugates. Nat Rev Drug Discov 2017; 16:315-337. [PMID: 28303026 DOI: 10.1038/nrd.2016.268] [Citation(s) in RCA: 1525] [Impact Index Per Article: 190.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) are one of the fastest growing classes of oncology therapeutics. After half a century of research, the approvals of brentuximab vedotin (in 2011) and trastuzumab emtansine (in 2013) have paved the way for ongoing clinical trials that are evaluating more than 60 further ADC candidates. The limited success of first-generation ADCs (developed in the early 2000s) informed strategies to bring second-generation ADCs to the market, which have higher levels of cytotoxic drug conjugation, lower levels of naked antibodies and more-stable linkers between the drug and the antibody. Furthermore, lessons learned during the past decade are now being used in the development of third-generation ADCs. In this Review, we discuss strategies to select the best target antigens as well as suitable cytotoxic drugs; the design of optimized linkers; the discovery of bioorthogonal conjugation chemistries; and toxicity issues. The selection and engineering of antibodies for site-specific drug conjugation, which will result in higher homogeneity and increased stability, as well as the quest for new conjugation chemistries and mechanisms of action, are priorities in ADC research.
Collapse
Affiliation(s)
- Alain Beck
- Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, 5 Avenue Napoleon III, 74160 Saint Julien en Genevois, France
| | - Liliane Goetsch
- Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, 5 Avenue Napoleon III, 74160 Saint Julien en Genevois, France
| | - Charles Dumontet
- Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS, 69000 Lyon, France.,University of Lyon, 69000 Lyon, France.,Hospices Civils de Lyon, 69000 Lyon, France
| | - Nathalie Corvaïa
- Institut de Recherche Pierre Fabre, Centre d'Immunologie Pierre Fabre, 5 Avenue Napoleon III, 74160 Saint Julien en Genevois, France
| |
Collapse
|
44
|
Dormeshkin D, Gilep A, Sergeev G, Usanov S. Development of CYB5-fusion monitoring system for efficient periplasmic expression of multimeric proteins in Escherichia coli. Protein Expr Purif 2016; 128:60-6. [DOI: 10.1016/j.pep.2016.08.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/05/2016] [Accepted: 08/10/2016] [Indexed: 11/15/2022]
|
45
|
Jang S, Kim G, Oh J, Lee S, Kim D, Kim KH, Kim YH, Rhee DK, Lee S. Molecular characterization of a single-chain antibody variable fragment (scFv) specific for PspA from Streptococcus pneumoniae. Biochem Biophys Res Commun 2016; 482:141-146. [PMID: 27845043 DOI: 10.1016/j.bbrc.2016.10.150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 10/31/2016] [Indexed: 10/20/2022]
Abstract
Streptococcus pneumoniae is a major infectious agent responsible for pneumonia, otitis media, sepsis and meningitis. Pneumococcal surface protein A (PspA) is a well-characterized virulence factor localized on the surface and a target for vaccine development. In this study, we screened a single-chain antibody variable fragment (scFv) using phage display from a human synthetic library to select a clone 2B11. Affinity (Kd) of 2B11 was measured to be 5 nM using biolayer interferometry. 2B11 exhibited a dose-dependent recognition of recombinant PspA with no cross-reactivity towards pneumococcal antigens. The epitope on PspA was defined to residues 231-242 by mutational analysis. Molecular docking analysis supported the experimentally determined epitope, suggesting that the helix spanning residues 231-242 can bind to 2B11 with residues in the CDR-H3 (complementarity determining region 3 in the heavy chain) actively participating in the molecular contacts. Comparison of 2B11 with a commercial PspA antibody revealed that 2B11 exhibited a better specificity towards recombinant PspA antigen. 2B11 was capable of detecting endogenous PspA from pneumococcal lysates with affinity similar to that of the commercial antibody. Our study provides a molecular tool for biosensors detecting pneumococcal diseases.
Collapse
Affiliation(s)
- ShinA Jang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Gyuhee Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Jihye Oh
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Seungyeop Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Dongho Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Kook-Han Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, South Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, 16419, South Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, South Korea.
| |
Collapse
|
46
|
Pye H, Butt MA, Reinert HW, Maruani A, Nunes JPM, Marklew JS, Qurashi M, Funnell L, May A, Stamati I, Hamoudi R, Baker JR, Smith MEB, Caddick S, Deonarain MP, Yahioglu G, Chudasama V, Lovat LB. A HER2 selective theranostic agent for surgical resection guidance and photodynamic therapy. Photochem Photobiol Sci 2016; 15:1227-1238. [PMID: 27501936 DOI: 10.1039/c6pp00139d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In many cancers early intervention involves surgical resection of small localised tumour masses. Inadequate resection leads to recurrence whereas overzealous treatment can lead to organ damage. This work describes production of a HER2 targeting antibody Fab fragment dual conjugated to achieve both real time near-infrared fluorescent imaging and photodynamic therapy. The use of fluorescence emission from a NIR-dye could be used to guide resection of tumour bulk, for example during endoscopic diagnosis for oesophago-gastric adenocarcinoma, this would then be followed by activation of the photodynamic therapeutic agent to destroy untreated localised areas of cancer infiltration and tumour infiltrated lymph nodes. This theranostic agent was prepared from the Fab fragment of trastuzumab initially by functional disulfide re-bridging and site-specific click reaction of a NIR-dye. This was followed by further reaction with a novel pre-activated form of the photosensitiser chlorin e6 with the exposed fragments' lysine residues. Specific binding of the theranostic agent was observed in vitro with a HER2 positive cell line and cellular near-infrared fluorescence was observed with flow cytometry. Specific photo-activity of the conjugates when exposed to laser light was observed with HER2 positive but not HER2 negative cell lines in vitro, this selectivity was not seen with the unconjugated drug. This theranostic agent demonstrates that two different photo-active functions can be coupled to the same antibody fragment with little interference to their independent activities.
Collapse
Affiliation(s)
- H Pye
- Department for Tissue & Energy, Division of Surgery & Interventional Science, University College London, Cruciform Building, Gower Street, London, WC1E 6AE, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
48
|
Ginn C, Choi JW, Brocchini S. Disulfide-bridging PEGylation during refolding for the more efficient production of modified proteins. Biotechnol J 2016; 11:1088-99. [DOI: 10.1002/biot.201600035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/07/2016] [Accepted: 05/17/2016] [Indexed: 01/18/2023]
Affiliation(s)
| | - Ji-won Choi
- PolyTherics Ltd; Babraham Research Campus, Babraham; Cambridge UK
| | | |
Collapse
|
49
|
Abstract
Antibody drug conjugates (ADCs) have emerged as a viable option in targeted delivery of highly potent cytotoxic drugs in treatment of solid tumors. At the time of writing, only two ADCs have received regulatory approval with >40 others in clinical development. The first generation ADCs suffered from a lack of specificity in amino acid site-conjugations, yielding statistically heterogeneous stoichiometric ratios of drug molecules per antibody molecule. For the second generation ADCs, however, site-specific amino acid conjugation using enzymatic ligation, introduction of unnatural amino acids, and site-specific protein engineering hold promise to alleviate some of the current technical limitations. The rapid progress in technology platforms and antibody engineering has introduced novel linkers, site-specific conjugation chemistry, and new payload candidates that could possibly be exploited in the context of ADCs. A search using the Clinical Trial Database registry ( www.clinicaltrials.gov ), using the keyword 'antibody drug conjugate', yielded ~270 hits. The main focus of this article is to present a brief overview of the recent developments and current challenges related to ADC development.
Collapse
|
50
|
Massa S, Xavier C, Muyldermans S, Devoogdt N. Emerging site-specific bioconjugation strategies for radioimmunotracer development. Expert Opin Drug Deliv 2016; 13:1149-63. [DOI: 10.1080/17425247.2016.1178235] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sam Massa
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catarina Xavier
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging laboratory, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|