51
|
Alameddine R, Mallea P, Shahab F, Zakharia Y. Antibody Drug Conjugates in Bladder Cancer: Current Milestones and Future Perspectives. Curr Treat Options Oncol 2023; 24:1167-1182. [PMID: 37403009 DOI: 10.1007/s11864-023-01114-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 07/06/2023]
Abstract
OPINION STATEMENT Over the last several years, the treatment landscape of urothelial carcinoma has witnessed an unprecedented expansion of therapeutic options including checkpoint inhibitors, tyrosine kinase inhibitors, and antibody drug conjugates (ADC). Early trial data has shown that ADCs are safer and potentially effective treatment options in advanced bladder cancer as well as in the early disease. In particular, enfortumab-vedotin (EV) has shown promising results with a recent cohort of a clinical trial demonstrating that EV is effective as neoadjuvant monotherapy as well as in combination with pembrolizumab in metastatic setting. Similar promising results have been shown by other classes of ADC in other trials including sacituzumab-govitecan (SG) and oportuzumab monatox (OM). ADCs are likely to become a mainstay treatment option in the urothelial carcinoma playbook as either a monotherapy or combination therapy. The cost of the drug presents a real challenge, but further trial data may justify the use of the drug as mainstay treatment.
Collapse
Affiliation(s)
- Raafat Alameddine
- Division of Hematology Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Patrick Mallea
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Farhan Shahab
- Department of Emergency Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Yousef Zakharia
- Division of Hematology Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| |
Collapse
|
52
|
Zafar S, Armaghan M, Khan K, Hassan N, Sharifi-Rad J, Habtemariam S, Kieliszek M, Butnariu M, Bagiu IC, Bagiu RV, Cho WC. New insights into the anticancer therapeutic potential of maytansine and its derivatives. Biomed Pharmacother 2023; 165:115039. [PMID: 37364476 DOI: 10.1016/j.biopha.2023.115039] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Maytansine is a pharmacologically active 19-membered ansamacrolide derived from various medicinal plants and microorganisms. Among the most studied pharmacological activities of maytansine over the past few decades are anticancer and anti-bacterial effects. The anticancer mechanism of action is primarily mediated through interaction with the tubulin thereby inhibiting the assembly of microtubules. This ultimately leads to decreased stability of microtubule dynamics and cause cell cycle arrest, resulting in apoptosis. Despite its potent pharmacological effects, the therapeutic applications of maytansine in clinical medicine are quite limited due to its non-selective cytotoxicity. To overcome these limitations, several derivatives have been designed and developed mostly by modifying the parent structural skeleton of maytansine. These structural derivatives exhibit improved pharmacological activities as compared to maytansine. The present review provides a valuable insight into maytansine and its synthetic derivatives as anticancer agents.
Collapse
Affiliation(s)
- Sameen Zafar
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan.
| | - Nazia Hassan
- Department of Biochemistry, University of Agriculture Faisalabad, Pakistan
| | | | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK.
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland.
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, 300645, Calea Aradului 119, Timis, Romania.
| | - Iulia-Cristina Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Multidisciplinary Research Center on Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Preventive Medicine Study Center, Timisoara, Romania
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
53
|
Mckertish CM, Kayser V. A Novel Dual-Payload ADC for the Treatment of HER2+ Breast and Colon Cancer. Pharmaceutics 2023; 15:2020. [PMID: 37631234 PMCID: PMC10459570 DOI: 10.3390/pharmaceutics15082020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Antibody-drug conjugates (ADCs) have demonstrated a great therapeutic potential against cancer due to their target specificity and cytotoxicity. To exert a maximum therapeutic effect on cancerous cells, we have conjugated two different payloads to different amino acids, cysteines (cys) and lysines (lys), on trastuzumab, which is a humanised anti-HER2 monoclonal antibody. First, trastuzumab was conjugated with monomethyl auristatin E (MMAE), an antimitotic agent, through a cleavable linker (Val-Cit) to prepare ADC (Tmab-VcMMAE). Then, the ADC (Tmab-VcMMAE) was conjugated with a second antimitotic agent, Mertansine (DM1), via a non-cleavable linker Succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC) to form a dual conjugate (Tmab-VcMMAE-SMCC-DM1). Our results indicated that the dual-payload conjugate, Tmab-VcMMAE-SMCC-DM1, had a synergistic and superior cytotoxic effect compared to trastuzumab alone. Ultimately employing a dual conjugation approach has the potential to overcome treatment-resistance and tumour recurrences and could pave the way to employ other payloads to construct dual (or multiple) payload complexes.
Collapse
Affiliation(s)
| | - Veysel Kayser
- Sydney School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
54
|
Egorova VS, Kolesova EP, Lopus M, Yan N, Parodi A, Zamyatnin AA. Smart Delivery Systems Responsive to Cathepsin B Activity for Cancer Treatment. Pharmaceutics 2023; 15:1848. [PMID: 37514035 PMCID: PMC10386206 DOI: 10.3390/pharmaceutics15071848] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Cathepsin B is a lysosomal cysteine protease, contributing to vital cellular homeostatic processes including protein turnover, macroautophagy of damaged organelles, antigen presentation, and in the extracellular space, it takes part in tissue remodeling, prohormone processing, and activation. However, aberrant overexpression of cathepsin B and its enzymatic activity is associated with different pathological conditions, including cancer. Cathepsin B overexpression in tumor tissues makes this enzyme an important target for smart delivery systems, responsive to the activity of this enzyme. The generation of technologies which therapeutic effect is activated as a result of cathepsin B cleavage provides an opportunity for tumor-targeted therapy and controlled drug release. In this review, we summarized different technologies designed to improve current cancer treatments responsive to the activity of this enzyme that were shown to play a key role in disease progression and response to the treatment.
Collapse
Affiliation(s)
- Vera S Egorova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Ekaterina P Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Kalina Campus, Vidyanagari, Mumbai 400098, India
| | - Neng Yan
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Andrey A Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, Sochi 354340, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| |
Collapse
|
55
|
Sun T, Niu X, He Q, Liu M, Qiao S, Qi RQ. Development, efficacy and side effects of antibody‑drug conjugates for cancer therapy (Review). Mol Clin Oncol 2023; 18:47. [PMID: 37206431 PMCID: PMC10189422 DOI: 10.3892/mco.2023.2643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/22/2023] [Indexed: 05/21/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are anticancer drugs that combine cytotoxic small-molecule drugs (payloads) with monoclonal antibodies through a chemical linker and that transfer toxic payloads to tumor cells expressing target antigens. All ADCs are based on human IgG. In 2009, the Food and Drug Administration (FDA) approved gemtuzumab ozogamicin as the initial first-generation ADC. Since then, at least 100 ADC-related projects have been initiated, and 14 ADCs are currently being tested in clinical trials. The limited success of gemtuzumab ozogamicin has led to the development of optimization strategies for the next generation of drugs. Subsequently, experts have improved the first-generation ADCs and have developed second-generation ADCs such as ado-trastuzumab emtansine. Second-generation ADCs have higher specific antigen levels, more stable linkers and longer half-lives and show great potential to transform cancer treatment models. Since the first two generations of ADCs have served as a good foundation, the development of ADCs is accelerating, and third-generation ADCs, represented by trastuzumab deruxtecan, are ready for wide application. Third-generation ADCs are characterized by strong pharmacokinetics and high pharmaceutical activity, and their drug-to-antibody ratio mainly ranges from 2 to 4. In the past decade, the research prospects of ADCs have broadened, and an increasing number of specific antigen targets and mechanisms of cytotoxic drug release have been discovered and studied. To date, seven ADCs have been approved by the FDA for lymphoma, and three have been approved to treat breast cancer. The present review explores the function and development of ADCs and their clinical use in cancer treatment.
Collapse
Affiliation(s)
- Te Sun
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Xueli Niu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Qing He
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
| | - Min Liu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Shuai Qiao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
- Correspondence to: Professor Rui-Qun Qi or Mrs. Shuai Qiao, Department of Dermatology, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, Liaoning 110001, P.R. China
| | - Rui-Qun Qi
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
- Key Laboratory of Immunodermatology, National Joint Engineering Research Center for Theranostics of Immunological Skin Diseases, Ministry of Education and NHC, Shenyang, Liaoning 110001, P.R. China
- Correspondence to: Professor Rui-Qun Qi or Mrs. Shuai Qiao, Department of Dermatology, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
56
|
Gabriele F, Palerma M, Ippoliti R, Angelucci F, Pitari G, Ardini M. Recent Advances on Affibody- and DARPin-Conjugated Nanomaterials in Cancer Therapy. Int J Mol Sci 2023; 24:ijms24108680. [PMID: 37240041 DOI: 10.3390/ijms24108680] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Affibodies and designed ankyrin repeat proteins (DARPins) are synthetic proteins originally derived from the Staphylococcus aureus virulence factor protein A and the human ankyrin repeat proteins, respectively. The use of these molecules in healthcare has been recently proposed as they are endowed with biochemical and biophysical features heavily demanded to target and fight diseases, as they have a strong binding affinity, solubility, small size, multiple functionalization sites, biocompatibility, and are easy to produce; furthermore, impressive chemical and thermal stability can be achieved. especially when using affibodies. In this sense, several examples reporting on affibodies and DARPins conjugated to nanomaterials have been published, demonstrating their suitability and feasibility in nanomedicine for cancer therapy. This minireview provides a survey of the most recent studies describing affibody- and DARPin-conjugated zero-dimensional nanomaterials, including inorganic, organic, and biological nanoparticles, nanorods, quantum dots, liposomes, and protein- and DNA-based assemblies for targeted cancer therapy in vitro and in vivo.
Collapse
Affiliation(s)
- Federica Gabriele
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Marta Palerma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
57
|
Conjugation site characterization of antibody-drug conjugates using electron-transfer/higher-energy collision dissociation (EThcD). Anal Chim Acta 2023; 1251:340978. [PMID: 36925279 DOI: 10.1016/j.aca.2023.340978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Antibody-drug conjugates (ADCs) are formed by binding of cytotoxic drugs to monoclonal antibodies (mAbs) through chemical linkers. A comprehensive evaluation of the critical quality attributes (CQAs) of ADCs is vital for drug development but remains challenging owing to ADC structural heterogeneity than mAbs. Drug conjugation sites can considerably affect ADC properties, such as stability and pharmacokinetics, however, few studies have focused on method development in this area owing to technical challenges. Hybrid electron-transfer/higher-energy collision dissociation (EThcD) produces more fragment ions than conventional higher-energy collision dissociation (HCD) fragmentation, which aids in identifying and localizing post-translational modifications. Herein, we systematically employ EThcD to assess the fragmentation mode impact on conjugation site characterization for randomly conjugated and site-specific ADCs. EThcD generates more fragment ions in tandem mass spectrometry (MS/MS) spectra compared with HCD. Additional ions aid in pinpointing the correct conjugation sites that bear complex linker payload structures. Our study may contribute to the quality control of various preclinical and clinical ADCs.
Collapse
|
58
|
Rasic P, Jeremic M, Jeremic R, Dusanovic Pjevic M, Rasic M, Djuricic SM, Milickovic M, Vukadin M, Mijovic T, Savic D. Targeting B7-H3-A Novel Strategy for the Design of Anticancer Agents for Extracranial Pediatric Solid Tumors Treatment. Molecules 2023; 28:molecules28083356. [PMID: 37110590 PMCID: PMC10145344 DOI: 10.3390/molecules28083356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Recent scientific data recognize the B7-H3 checkpoint molecule as a potential target for immunotherapy of pediatric solid tumors (PSTs). B7-H3 is highly expressed in extracranial PSTs such as neuroblastoma, rhabdomyosarcoma, nephroblastoma, osteosarcoma, and Ewing sarcoma, whereas its expression is absent or very low in normal tissues and organs. The influence of B7-H3 on the biological behavior of malignant solid neoplasms of childhood is expressed through different molecular mechanisms, including stimulation of immune evasion and tumor invasion, and cell-cycle disruption. It has been shown that B7-H3 knockdown decreased tumor cell proliferation and migration, suppressed tumor growth, and enhanced anti-tumor immune response in some pediatric solid cancers. Antibody-drug conjugates targeting B7-H3 exhibited profound anti-tumor effects against preclinical models of pediatric solid malignancies. Moreover, B7-H3-targeting chimeric antigen receptor (CAR)-T cells demonstrated significant in vivo activity against different xenograft models of neuroblastoma, Ewing sarcoma, and osteosarcoma. Finally, clinical studies demonstrated the potent anti-tumor activity of B7-H3-targeting antibody-radioimmunoconjugates in metastatic neuroblastoma. This review summarizes the established data from various PST-related studies, including in vitro, in vivo, and clinical research, and explains all the benefits and potential obstacles of targeting B7-H3 by novel immunotherapeutic agents designed to treat malignant extracranial solid tumors of childhood.
Collapse
Affiliation(s)
- Petar Rasic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Marija Jeremic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Rada Jeremic
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marija Dusanovic Pjevic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Rasic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Slavisa M Djuricic
- Department of Clinical Pathology, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Maja Milickovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Miroslav Vukadin
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Tanja Mijovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Djordje Savic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
59
|
Huysamen A, Fadeyi OE, Mayuni G, Dogbey DM, Mungra N, Biteghe FAN, Hardcastle N, Ramamurthy D, Akinrinmade OA, Naran K, Cooper S, Lang D, Richter W, Hunter R, Barth S. Click Chemistry-Generated Auristatin F-Linker-Benzylguanine for a SNAP-Tag-Based Recombinant Antibody-Drug Conjugate Demonstrating Selective Cytotoxicity toward EGFR-Overexpressing Tumor Cells. ACS OMEGA 2023; 8:4026-4037. [PMID: 36743041 PMCID: PMC9893251 DOI: 10.1021/acsomega.2c06844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/03/2023] [Indexed: 06/18/2023]
Abstract
Antibody-drug conjugates (ADCs) are bifunctional molecules combining the targeting potential of monoclonal antibodies with the cancer-killing ability of cytotoxic drugs. This simple yet intelligently designed system directly addresses the lack of specificity encountered with conventional anti-cancer treatment regimes. However, despite their initial success, the generation of clinically sustainable and effective ADCs has been plagued by poor tumor penetration, undefined chemical linkages, unpredictable pharmacokinetic profiles, and heterogeneous mixtures of products. To this end, we generated a SNAP-tag-based fusion protein targeting the epidermal growth factor receptor (EGFR)-a biomarker of aggressive and drug-resistant cancers. Here, we demonstrate the use of a novel click coupling strategy to engineer a benzylguanine (BG)-linker-auristatin F (AuriF) piece that can be covalently tethered to the EGFR-targeting SNAP-tag-based fusion protein in an irreversible 1:1 stoichiometric reaction to form a homogeneous product. Furthermore, using these recombinant ADCs to target EGFR-overexpressing tumor cells, we provide a proof-of-principle for generating biologically active antimitotic therapeutic proteins capable of inducing cell death in a dose-dependent manner, thus alleviating some of the challenges of early ADC development.
Collapse
Affiliation(s)
- Allan
M. Huysamen
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Olaolu E. Fadeyi
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Grace Mayuni
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dennis M. Dogbey
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Neelakshi Mungra
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Centre
for Immunity and Immunotherapies, Seattle
Children’s Research Institute, Seattle, Washington 98101, United States
| | - Fleury A. N. Biteghe
- Department
of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, California 90048, United States
| | - Natasha Hardcastle
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Dharanidharan Ramamurthy
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Olusiji A. Akinrinmade
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- Department
of Molecular Pharmacology, Albert Einstein
College of Medicine, Bronx, New York 10461, United States
| | - Krupa Naran
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
| | - Susan Cooper
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | - Dirk Lang
- Division
of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | | | - Roger Hunter
- Department
of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Stefan Barth
- Medical
Biotechnology and Immunotherapy Research Unit, Institute of Infectious
Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7700, South Africa
- South
African Research Chair in Cancer Biotechnology, Department of Integrative
Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape
Town 7700, South Africa
| |
Collapse
|
60
|
Antibody-drug conjugates in lung cancer: dawn of a new era? NPJ Precis Oncol 2023; 7:5. [PMID: 36631624 PMCID: PMC9834242 DOI: 10.1038/s41698-022-00338-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are one of fastest growing classes of oncology drugs in modern drug development. By harnessing the powers of both cytotoxic chemotherapy and targeted therapy, ADCs are unique in offering the potential to deliver highly potent cytotoxic agents to cancer cells which express a pre-defined cell surface target. In lung cancer, the treatment paradigm has shifted dramatically in recent years, and now ADCs are now joining the list as potential options for lung cancer patients. Since 2020, the first ADC for NSCLC patients has been FDA-approved (trastuzumab deruxtecan) and two ADCs have been granted FDA Breakthrough Therapy Designation, currently under evaluation (patritumab deruxtecan, telisotuzumab vedotin). Furthermore, several early-phase trials are assessing various novel ADCs, either as monotherapy or in combinations with advanced lung cancer, and more selective and potent ADCs are expected to become therapeutic options in clinic soon. In this review, we discuss the structure and mechanism of action of ADCs, including insights from pre-clinical work; we summarize the ADCs' recent progress in lung cancer, describe toxicity profiles of ADCs, and explore strategies designed to enhance ADC potency and overcome resistance. In addition, we discuss novel ADC strategies of interest in lung cancer, including non-cytotoxic payloads, such as immunomodulatory and anti-apoptotic agents.
Collapse
|
61
|
Yao P, Zhang Y, Zhang S, Wei X, Liu Y, Du C, Hu M, Feng C, Li J, Zhao F, Li C, Li Z, Du L. Knowledge atlas of antibody-drug conjugates on CiteSpace and clinical trial visualization analysis. Front Oncol 2023; 12:1039882. [PMID: 36686767 PMCID: PMC9850101 DOI: 10.3389/fonc.2022.1039882] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Objective Antibody-drugs conjugates (ADCs) are novel drugs with highly targeted and tumor-killing abilities and developing rapidly. This study aimed to evaluate drug discovery and clinical trials of and explore the hotspots and frontiers from 2012 to 2022 using bibliometric methods. Methods Publications on ADCs were retrieved between 2012 and 2022 from Web of Science (WoS) and analyzed with CiteSpace 6.1.R2 software for the time, region, journals, institutions, etc. Clinical trials were downloaded from clinical trial.org and visualized with Excel software. Results A total of 696 publications were obtained and 187 drug trials were retrieved. Since 2012, research on ADCs has increased year by year. Since 2020, ADC-related research has increased dramatically, with the number of relevant annual publications exceeding 100 for the first time. The United States is the most authoritative and superior country and region in the field of ADCs. The University of Texas MD Anderson Cancer Center is the most authoritative institution in this field. Research on ADCs includes two clinical trials and one review, which are the most influential references. Clinical trials of ADCs are currently focused on phase I and phase II. Comprehensive statistics and analysis of the published literature and clinical trials in the field of ADCs, have shown that the most studied drug is brentuximab vedotin (BV), the most popular target is human epidermal growth factor receptor 2 (HER2), and breast cancer may become the main trend and hotspot for ADCs indications in recent years. Conclusion Antibody-drug conjugates have become the focus of targeted therapies in the field of oncology. The innovation of technology and combination application strategy will become the main trend and hotspots in the future.
Collapse
Affiliation(s)
- Peizhuo Yao
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yinbin Zhang
- School of Chemistry, Xi’an Jiaotong University, Xi’an, China,*Correspondence: Yinbin Zhang, ; Shuqun Zhang,
| | - Shuqun Zhang
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,*Correspondence: Yinbin Zhang, ; Shuqun Zhang,
| | - Xinyu Wei
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanbin Liu
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chong Du
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Mingyou Hu
- School of Chemistry, Xi’an Jiaotong University, Xi’an, China
| | - Cong Feng
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jia Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fang Zhao
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chaofan Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Zhen Li
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lisha Du
- Department of Oncology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
62
|
Xue J, Lou X, Ning D, Shao R, Chen G. Mechanism and treatment of α-amanitin poisoning. Arch Toxicol 2023; 97:121-131. [PMID: 36271256 DOI: 10.1007/s00204-022-03396-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 12/31/2024]
Abstract
Amanita poisoning has a high mortality rate. The α-amanitin toxin in Amanita is the main lethal toxin. There is no specific detoxification drug for α-amanitin, and the clinical treatment mainly focuses on symptomatic and supportive therapy. The pathogenesis of α-amanitin mainly includes: α-amanitin can inhibit the activity of RNA polymeraseII in the nucleus, including the inhibition of the largest subunit of RNA polymeraseII, RNApb1, bridge helix, and trigger loop. In addition, α-amanitin acts in vivo through the enterohepatic circulation and transport system. α-Amanitin can cause the cell death. The existing mechanisms of cell damage mainly focus on apoptosis, oxidative stress, and autophagy. In addition to the pathogenic mechanism, α-amanitin also has a role in cancer treatment, which is the focus of current research. The mechanism of action of α-amanitin on the body is still being explored.
Collapse
Affiliation(s)
- Jinfang Xue
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Xiran Lou
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Deyuan Ning
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Ruifei Shao
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Guobing Chen
- Department of Emergency Medicine, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Xishan District, Kunming, 650032, People's Republic of China.
| |
Collapse
|
63
|
Yamazaki S, Matsuda Y. Tag‐Free Enzymatic Modification for Antibody−Drug Conjugate Production. ChemistrySelect 2022. [DOI: 10.1002/slct.202203753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | - Yutaka Matsuda
- Ajinomoto Bio-Pharma Services 11040 Roselle Street San Diego CA 92121 United States
| |
Collapse
|
64
|
Barreca M, Lang N, Tarantelli C, Spriano F, Barraja P, Bertoni F. Antibody-drug conjugates for lymphoma patients: preclinical and clinical evidences. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:763-794. [PMID: 36654819 PMCID: PMC9834635 DOI: 10.37349/etat.2022.00112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/08/2022] [Indexed: 12/28/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are a recent, revolutionary approach for malignancies treatment, designed to provide superior efficacy and specific targeting of tumor cells, compared to systemic cytotoxic chemotherapy. Their structure combines highly potent anti-cancer drugs (payloads or warheads) and monoclonal antibodies (Abs), specific for a tumor-associated antigen, via a chemical linker. Because the sensitive targeting capabilities of monoclonal Abs allow the direct delivery of cytotoxic payloads to tumor cells, these agents leave healthy cells unharmed, reducing toxicity. Different ADCs have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of a wide range of malignant conditions, both as monotherapy and in combination with chemotherapy, including for lymphoma patients. Over 100 ADCs are under preclinical and clinical investigation worldwide. This paper it provides an overview of approved and promising ADCs in clinical development for the treatment of lymphoma. Each component of the ADC design, their mechanism of action, and the highlights of their clinical development progress are discussed.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Noémie Lang
- Division of Oncology, Department of Oncology, Faculty of Medicine, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Chiara Tarantelli
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| |
Collapse
|
65
|
Fontes MS, Vargas Pivato de Almeida D, Cavalin C, Tagawa ST. Targeted Therapy for Locally Advanced or Metastatic Urothelial Cancer (mUC): Therapeutic Potential of Sacituzumab Govitecan. Onco Targets Ther 2022; 15:1531-1542. [PMID: 36575731 PMCID: PMC9790156 DOI: 10.2147/ott.s339348] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Urothelial carcinoma is the second most frequent genitourinary malignancy. Despite the poor prognosis, new treatment options have emerged and have expanded the therapeutic landscape for the disease. Although major improvements have been achieved, many patients experience rapid disease progression and low responses in subsequent lines of therapy. Sacituzumab govitecan is an ADC that targets Trop-2, which is highly expressed in urothelial cancers. Promising results in early clinical trials have led to further drug development which confirmed encouraging efficacy. Sacituzumab govitecan has been given accelerated approval in 2021 for patients with locally advanced and metastatic urothelial cancer who previously received a platinum containing chemotherapy and either a programmed death receptor-1 or programmed death ligand inhibitor. The results are promising, with encouraging efficacy and safety, however responses are not universal. There is a growing comprehension of mechanisms of resistance and predictive biomarkers that are crucial to improving outcomes. In this review, we summarize the current knowledge on antibody-drug conjugates and the clinical findings that led to the approval of Sacituzumab govitecan and discuss the therapeutic potential of new combinations, mechanisms of resistance and predictive biomarkers.
Collapse
Affiliation(s)
- Mariane S Fontes
- Oncology Department, Oncoclinicas Group, Rio de Janeiro, Brazil
- LACOG, Latin American Cooperative Oncology Group, Brazil
| | | | | | | |
Collapse
|
66
|
Unveiling the antibody-drug conjugates portfolio in battling Triple-negative breast cancer: Therapeutic trends and Future horizon. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:25. [PMID: 36456774 DOI: 10.1007/s12032-022-01884-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/02/2022] [Indexed: 12/03/2022]
Abstract
Triple-negative breast cancer (TNBC) showcases a labyrinthine network exhibiting deficient expression of Estrogen receptor (ER), Progesterone receptor (PR), and Human-epidermal growth factor receptor-2 (HER2). This restricts the conventional chemotherapeutic, hormonal, and few targeted regimens in showing efficient anti-tumor response. Antibody-drug conjugates (ADCs) are target-specific conjugates comprising a monoclonal antibody attached to the desired cytotoxic payload with the support of a stable linker. They are designated as one of the encouraging sets of targeted therapies that have unveiled affirmative outcomes owing to increased specificity in targeting the undetectable or deficiently expressed targets. Another virtue of ADCs lending superiority to this approach is the presence of inherent bystander effect which has a detrimental influence on the tumor microenvironment (TME) devoid of antigen expression. In the current scenario, FDA-approved Sacituzumab govitecan is widely being utilized to mitigate TNBC while many other ADCs are being studied in clinical trials. Additionally, a focus has been set on revelation of application of Trastuzumab deruxtecan in HER2-low metastatic breast cancer which widens the current therapeutic horizon dealing with such carcinomas. After making an effort towards sketching ADCs profile, we conclude that this novel approach deserves to be investigated through future campaigns owing to its remarkable bystander effect, ability to precisely recognize the antigen and spare the naïve cells from detrimental toxicity. Exploration of the remarkable potential of Sacituzumab govitecan in multiple indications including TNBC portrays the prominence of ADCs and prompts the bright future of this therapeutic approach. In this review, we present the basic foundation of ADCs alongside summarizing the building blocks of several ADCs used in TNBC. Furthermore, by shedding light on the therapeutic regimens and concomitant effects of various ADCs derived from the supportive backbone of clinical trials, we have attempted to convene several segments of ADCs and portray their potentialities time ahead.
Collapse
|
67
|
Plant-Derived Type I Ribosome Inactivating Protein-Based Targeted Toxins: A Review of the Clinical Experience. Toxins (Basel) 2022; 14:toxins14080563. [PMID: 36006226 PMCID: PMC9412999 DOI: 10.3390/toxins14080563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Targeted toxins (TT) for cancer treatment are a class of hybrid biologic comprised of a targeting domain coupled chemically or genetically to a proteinaceous toxin payload. The targeting domain of the TT recognises and binds to a defined target molecule on the cancer cell surface, thereby delivering the toxin that is then required to internalise to an appropriate intracellular compartment in order to kill the target cancer cell. Toxins from several different sources have been investigated over the years, and the two TTs that have so far been licensed for clinical use in humans; both utilise bacterial toxins. Relatively few clinical studies have, however, been undertaken with TTs that utilise single-chain type I ribosome inactivating proteins (RIPs). This paper reviews the clinical experience that has so far been obtained for a range of TTs based on five different type I RIPs and concludes that the majority studied in early phase trials show significant clinical activity that justifies further clinical investigation. A range of practical issues relating to the further clinical development of TT’s are also covered briefly together with some suggested solutions to outstanding problems.
Collapse
|
68
|
Singh S, Serwer L, DuPage A, Elkins K, Chauhan N, Ravn M, Buchanan F, Wang L, Krimm M, Wong K, Sagert J, Tipton K, Moore SJ, Huang Y, Jang A, Ureno E, Miller A, Patrick S, Duvur S, Liu S, Vasiljeva O, Li Y, Henriques T, Badagnani I, Jeffries S, Schleyer S, Leanna R, Krebber C, Viswanathan S, Desnoyers L, Terrett J, Belvin M, Morgan-Lappe S, Kavanaugh WM, Richardson J. Nonclinical Efficacy and Safety of CX-2029, an Anti-CD71 Probody-Drug Conjugate. Mol Cancer Ther 2022; 21:1326-1336. [PMID: 35666803 PMCID: PMC9662867 DOI: 10.1158/1535-7163.mct-21-0193] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/19/2021] [Accepted: 05/20/2022] [Indexed: 01/07/2023]
Abstract
Probody therapeutics (Pb-Txs) are conditionally activated antibody-drug conjugates (ADCs) designed to remain inactive until proteolytically activated in the tumor microenvironment, enabling safer targeting of antigens expressed in both tumor and normal tissue. Previous attempts to target CD71, a highly expressed tumor antigen, have failed to establish an acceptable therapeutic window due to widespread normal tissue expression. This study evaluated whether a probody-drug conjugate targeting CD71 can demonstrate a favorable efficacy and tolerability profile in preclinical studies for the treatment of cancer. CX-2029, a Pb-Tx conjugated to maleimido-caproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E, was developed as a novel cancer therapeutic targeting CD71. Preclinical studies were performed to evaluate the efficacy and safety of this anti-CD71 PDC in patient-derived xenograft (PDX) mouse models and cynomolgus monkeys, respectively. CD71 expression was detected at high levels by IHC across a broad range of tumor and normal tissues. In vitro, the masked Pb-Tx form of the anti-CD71 PDC displayed a >50-fold reduced affinity for binding to CD71 on cells compared with protease-activated, unmasked anti-CD71 PDC. Potent in vivo tumor growth inhibition (stasis or regression) was observed in >80% of PDX models (28/34) at 3 or 6 mg/kg. Anti-CD71 PDC remained mostly masked (>80%) in circulation throughout dosing in cynomolgus monkeys at 2, 6, and 12 mg/kg and displayed a 10-fold improvement in tolerability compared with an anti-CD71 ADC, which was lethal. Preclinically, anti-CD71 PDC exhibits a highly efficacious and acceptable safety profile that demonstrates the utility of the Pb-Tx platform to target CD71, an otherwise undruggable target. These data support further clinical development of the anti-CD71 PDC CX-2029 as a novel cancer therapeutic.
Collapse
Affiliation(s)
- Shweta Singh
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Laura Serwer
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Amy DuPage
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Kristi Elkins
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | | | | | - Leyu Wang
- AbbVie Inc., North Chicago, Illinois
| | - Michael Krimm
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Ken Wong
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Jason Sagert
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | | | - Yuanhui Huang
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Andrew Jang
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Eric Ureno
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Adam Miller
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Sarah Patrick
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Shanti Duvur
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Shouchun Liu
- CytomX Therapeutics, Inc, South San Francisco, California
| | - Olga Vasiljeva
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | | | | | | | - Siew Schleyer
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | - Claus Krebber
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | - Luc Desnoyers
- CytomX Therapeutics, Inc, South San Francisco, California
| | | | - Marcia Belvin
- CytomX Therapeutics, Inc, South San Francisco, California
- Corresponding Author: Marcia Belvin, CytomX Therapeutics, Inc., South San Francisco, CA 94080. Phone: (650)-892-9803; E-mail:
| | | | | | | |
Collapse
|
69
|
Procopiou G, Jackson PJM, di Mascio D, Auer JL, Pepper C, Rahman KM, Fox KR, Thurston DE. DNA sequence-selective G-A cross-linking ADC payloads for use in solid tumour therapies. Commun Biol 2022; 5:741. [PMID: 35906376 PMCID: PMC9338023 DOI: 10.1038/s42003-022-03633-0] [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: 07/14/2021] [Accepted: 06/24/2022] [Indexed: 11/11/2022] Open
Abstract
Antibody-Drug Conjugates (ADCs) are growing in importance for the treatment of both solid and haematological malignancies. There is a demand for new payloads with novel mechanisms of action that may offer enhanced therapeutic efficacy, especially in patients who develop resistance. We report here a class of Cyclopropabenzindole-Pyridinobenzodiazepine (CBI-PDD) DNA cross-linking payloads that simultaneously alkylate guanine (G) and adenine (A) bases in the DNA minor groove with a defined sequence selectivity. The lead payload, FGX8-46 (6), produces sequence-selective G-A cross-links and affords cytotoxicity in the low picomolar region across a panel of 11 human tumour cell lines. When conjugated to the antibody cetuximab at an average Drug-Antibody Ratio (DAR) of 2, an ADC is produced with significant antitumour activity at 1 mg/kg in a target-relevant human tumour xenograft mouse model with an unexpectedly high tolerability (i.e., no weight loss observed at doses as high as 45 mg/kg i.v., single dose). A class of Cyclopropabenzindole-Pyridinobenzodiazepine (CBI-PDD) DNA cross-linking payloads, used in Antibody-Drug Conjugates, alkylate guanine and adenine bases in the DNA minor groove with a defined sequence selectivity.
Collapse
Affiliation(s)
- George Procopiou
- Femtogenix, Lawes Open Innovation Hub, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK
| | - Paul J M Jackson
- Femtogenix, Lawes Open Innovation Hub, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK
| | - Daniella di Mascio
- School of Biological Sciences, Life Sciences Building B85, University of Southampton, Southampton, Hampshire, SO17 1BJ, UK
| | - Jennifer L Auer
- Femtogenix, Lawes Open Innovation Hub, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK
| | - Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, UK
| | - Khondaker Miraz Rahman
- Femtogenix, Lawes Open Innovation Hub, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK.,School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK
| | - Keith R Fox
- School of Biological Sciences, Life Sciences Building B85, University of Southampton, Southampton, Hampshire, SO17 1BJ, UK
| | - David E Thurston
- Femtogenix, Lawes Open Innovation Hub, Rothamsted Research, West Common, Harpenden, Hertfordshire, AL5 2JQ, UK. .,School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London, SE1 9NH, UK.
| |
Collapse
|
70
|
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
|
71
|
Qi X, Li Y, Liu W, Wang Y, Chen Z, Lin L. Research Trend of Publications Concerning Antibody-Drug Conjugate in Solid Cancer: A Bibliometric Study. Front Pharmacol 2022; 13:921385. [PMID: 35795565 PMCID: PMC9252465 DOI: 10.3389/fphar.2022.921385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Antibody-drug conjugate (ADC) is a promising therapy for solid cancer that has raised global concern. Although several papers have reviewed the current state of ADCs in different solid cancers, a quantitative analysis of the publications in this field is scarce. Methods: Publications related to ADC in the field of solid cancer were obtained from the Web of Science Core Collection. Data analyses were performed with VOSviewer 1.6.9, HistCite 2.1, CiteSpace V and R package Bibliometrix. Results: A total of 3,482 records were obtained in the holistic field and 1,197 in the clinical field. Steady growth in the number of publications was observed. The United States was the leading contributor in this field. Krop IE was the most influential author. The most productive institution was Genentech Inc., while Mem Sloan Kettering Canc Ctr was the most cited one. The most impactful journal was the Journal of Clinical Oncology. A total of 37 burst references and five burst references were identified between 2017–2022 in the holistic and clinical fields, respectively. Keywords analysis indicated that ADCs research mainly involved breast cancer, triple-negative breast cancer, ovarian cancer, small cell lung cancer, prostate cancer, gastric cancer, and urothelial carcinoma. ADC agents including trastuzumab emtansine, trastuzumab deruxtecan, sacituzumab govitecan, enfortumab vedotin, and rovalpituzumab tesirine were highly studied. Targets including HER2, trophoblast cell-surface antigen, mesothelin, delta-like ligand 3, and nectin-4 were the major concerns. Conclusion: This study analyzed publications concerning ADCs in the field of solid cancer with bibliometric analysis. Further clinical trials of ADCs and designs of the next generation of ADCs are the current focuses of the field. Acquired resistance of ADCs and biomarkers for ADC therapy efficacy monitoring are future concerns.
Collapse
Affiliation(s)
- Xiangjun Qi
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanlong Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifan Wang
- School of Chinese Classics Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuangzhong Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Lizhu Lin,
| |
Collapse
|
72
|
Zaib S, Areeba BS, Nehal Rana BS, Wattoo JI, Alsaab HO, Alzhrani RM, Awwad NS, Ibrahium HA, Khan I. Nanomedicines Targeting Heat Shock Protein 90 Gene Expression in the Therapy of Breast Cancer. ChemistrySelect 2022. [DOI: 10.1002/slct.202104553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sumera Zaib
- Department of Biochemistry Faculty of Life Sciences University of Central Punjab Lahore 54590 Pakistan
| | - B. S. Areeba
- Department of Biochemistry Faculty of Life Sciences University of Central Punjab Lahore 54590 Pakistan
| | - B. S. Nehal Rana
- Department of Biochemistry Faculty of Life Sciences University of Central Punjab Lahore 54590 Pakistan
| | - Javed Iqbal Wattoo
- Department of Biotechnology Faculty of Life Sciences University of Central Punjab Lahore 54590 Pakistan
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology Taif University, P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Rami M. Alzhrani
- Department of Pharmaceutics and Industrial Pharmacy College of Pharmacy Taif University, P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Nasser S. Awwad
- Chemistry Department Faculty of Science King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Hala A. Ibrahium
- Biology Department Faculty of Science King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
- Department of Semi Pilot Plant Nuclear Materials Authority P.O. Box 530 El Maadi Egypt
| | - Imtiaz Khan
- Manchester Institute of Biotechnology The University of Manchester 131 Princess Street Manchester M1 7DN United Kingdom
| |
Collapse
|
73
|
Cavaco M, Castanho MARB, Neves V. The Use of Antibody-Antibiotic Conjugates to Fight Bacterial Infections. Front Microbiol 2022; 13:835677. [PMID: 35330773 PMCID: PMC8940529 DOI: 10.3389/fmicb.2022.835677] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/14/2022] [Indexed: 12/26/2022] Open
Abstract
The emergence of antimicrobial resistance (AMR) is rapidly increasing and it is one of the significant twenty-first century's healthcare challenges. Unfortunately, the development of effective antimicrobial agents is a much slower and complex process compared to the spread of AMR. Consequently, the current options in the treatment of AMR are limited. One of the main alternatives to conventional antibiotics is the use of antibody-antibiotic conjugates (AACs). These innovative bioengineered agents take advantage of the selectivity, favorable pharmacokinetic (PK), and safety of antibodies, allowing the administration of more potent antibiotics with less off-target effects. Although AACs' development is challenging due to the complexity of the three components, namely, the antibody, the antibiotic, and the linker, some successful examples are currently under clinical studies.
Collapse
Affiliation(s)
| | | | - Vera Neves
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
74
|
Murali M, Kumar AR, Nair B, Pavithran K, Devan AR, Pradeep GK, Nath LR. Antibody-drug conjugate as targeted therapeutics against hepatocellular carcinoma: preclinical studies and clinical relevance. Clin Transl Oncol 2022; 24:407-431. [PMID: 34595736 DOI: 10.1007/s12094-021-02707-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/29/2021] [Indexed: 02/05/2023]
Abstract
An antibody-drug conjugate (ADC) is an advanced chemotherapeutic option with immense promises in treating many tumor. They are designed to selectively attack and kill neoplastic cells with minimal toxicity to normal tissues. ADCs are complex engineered immunoconjugates that comprise a monoclonal antibody for site-directed delivery and cytotoxic payload for targeted destruction of malignant cells. Therefore, it enables the reduction of off-target toxicities and enhances the therapeutic index of the drug. Hepatocellular carcinoma (HCC) is a solid tumor that shows high heterogeneity of molecular phenotypes and is considered the second most common cause of cancer-related death. Studies show enormous potential for ADCs targeting GPC3 and CD24 and other tumor-associated antigens in HCC with their high, selective expression and show potential outputs in preclinical evaluations. The review mainly highlights the preclinical evaluation of different antigen-targeted ADCs such as MetFab-DOX, Anti-c-Met IgG-OXA, Anti CD 24, ANC-HN-01, G7mab-DOX, hYP7-DCand hYP7-PC, Anti-CD147 ILs-DOX and AC133-vcMMAF against hepatocellular carcinoma and its future relevance.
Collapse
Affiliation(s)
- M Murali
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - A R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - B Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - K Pavithran
- Department of Medical Oncology and Hematology, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - A R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - G K Pradeep
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - L R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| |
Collapse
|
75
|
Tsai CH, Chiu TY, Chen CT, Hsu CY, Tsai YR, Yeh TK, Huang KH, Tsou LK. Click Chemistry and Multicomponent Reaction for Linker Diversification of Zinc Dipicolylamine-Based Drug Conjugates. Front Chem 2022; 9:822587. [PMID: 35242746 PMCID: PMC8886374 DOI: 10.3389/fchem.2021.822587] [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: 11/26/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
An efficient Ugi multicomponent reaction with strain promoted azide-alkyne cycloaddition protocol has been utilized in concert or independently to prepare a small family of bioactive zinc(II) dipicolylamine (ZnDPA)-based SN-38 conjugates. With sequential click chemistry coupling between the cytotoxic payload and phosphatidylserine-targeting ZnDPA ligand derived from structurally diverse carboxylic acids, aldehyde or ketones, and isocyanides, we demonstrated that this convergent synthetic strategy could furnish conjugates harnessing diversified linkers that exhibited different pharmacokinetic profiles in systemic circulation in vivo. Among the eight new conjugates, comparative studies on in vitro cytotoxicities, plasma stabilities, in vivo pharmacokinetic properties, and maximum tolerated doses were then carried out to identify a potent ZnDPA-based SN-38 conjugate that resulted in pancreatic cancer growth regression with an 80% reduction of cytotoxic payload used when compared to that of the marketed irinotecan. Our work provided the roadmap to construct a variety of theranostic agents in a similar manner for cancer treatment.
Collapse
|
76
|
Davis TK, Jennings ME. Site-Specific Conjugation Quantitation of a Cysteine-Conjugated Antibody-Drug Conjugate Using Stable Isotope Labeling Peptide Mapping LC-MS/MS Analysis. Anal Chem 2022; 94:2772-2778. [PMID: 35100801 DOI: 10.1021/acs.analchem.1c04025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Drug-load (DL) characterization of antibody-drug conjugates (ADCs) is an important analytical task due to its designation as a critical quality attribute (CQA) affecting potency and stability. Intact and subunit liquid chromatography-mass spectrometry (LC-MS) analyses can determine global drug-to-antibody ratios (DARs) that correlate well with other orthogonal analytical methods; however, peptide mapping liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis has struggled to provide complementary site-specific quantitation of drug conjugation sites. The peptide mapping method described herein utilizes stable isotope labeling to accurately quantitate the site-specific conjugation levels of a cysteine-conjugated ADC to provide "bottom-up" DAR characterization in parallel with protein sequence and post-translational modification (PTM) characterization in one multi-attribute analytical method (MAM).
Collapse
Affiliation(s)
- Tyler K Davis
- AbbVie, Analytical Research and Development, 1401 Sheridan Road, North Chicago, Illinois 60064, United States
| | - Mark E Jennings
- GlaxoSmithKline, CMC Analytical, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
77
|
Zhang H, Jin C, Zhang L, Peng B, Zhang Y, Liu Y, Li L, Ye M, Xiong W, Tan W. CD71-Specific Aptamer Conjugated with Monomethyl Auristatin E for the Treatment of Uveal Melanoma. ACS APPLIED MATERIALS & INTERFACES 2022; 14:32-40. [PMID: 34928139 DOI: 10.1021/acsami.1c13980] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy among adults. Despite significant advances in diagnosis and treatment, the general mortality of UM remains alarmingly high. This calls for the development of new approaches for the treatment of UM, such as targeted cancer therapy. CD71, also known as transferrin receptor 1, is overexpressed in UM cell lines and tissues. Herein, we report the development of a CD71-specific aptamer targeting the XQ-2d-MMAE conjugate that can distinguish UM cells from normal human uveal melanocytes. The cytotoxic drug monomethyl auristatin E (MMAE) could be easily coupled onto XQ-2d, a DNA aptamer that specifically targets CD71, to achieve efficiently targeted cancer growth inhibition in a mouse xenograft model, thus implying that XQ-2d-MMAE might be developed into a promising novel anti-tumor agent for the treatment of UM. Collectively, our results demonstrated that CD71 is a reliable target for drug delivery in UM and could be utilized as a model to explore aptamer-mediated targeted UM treatment strategies.
Collapse
Affiliation(s)
- Hui Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Cheng Jin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Lin Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Bo Peng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yibin Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yan Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Ling Li
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wei Xiong
- Department of Ophthalmology, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
78
|
Lu J, Ding J, Liu Z, Chen T. Retrospective analysis of the preparation and application of immunotherapy in cancer treatment (Review). Int J Oncol 2022; 60:12. [PMID: 34981814 PMCID: PMC8759346 DOI: 10.3892/ijo.2022.5302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
Monoclonal antibody technology plays a vital role in biomedical and immunotherapy, which greatly promotes the study of the structure and function of genes and proteins. To date, monoclonal antibodies have gone through four stages: murine monoclonal antibody, chimeric monoclonal antibody, humanised monoclonal antibody and fully human monoclonal antibody; thousands of monoclonal antibodies have been used in the fields of biology and medicine, playing a special role in the pathogenesis, diagnosis and treatment of disease. In this review, we compare the advantages and disadvantages of hybridoma technology, phage display technology, ribosome display technology, transgenic mouse technology, single B cell monoclonal antibody generation technologies, and forecast the promising applications of these technologies in clinical medicine, disease diagnosis and tumour treatment.
Collapse
Affiliation(s)
- Jiachen Lu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianing Ding
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
79
|
Jin Y, Schladetsch MA, Huang X, Balunas MJ, Wiemer AJ. Stepping forward in antibody-drug conjugate development. Pharmacol Ther 2022; 229:107917. [PMID: 34171334 PMCID: PMC8702582 DOI: 10.1016/j.pharmthera.2021.107917] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/03/2023]
Abstract
Antibody-drug conjugates (ADCs) are cancer therapeutic agents comprised of an antibody, a linker and a small-molecule payload. ADCs use the specificity of the antibody to target the toxic payload to tumor cells. After intravenous administration, ADCs enter circulation, distribute to tumor tissues and bind to the tumor surface antigen. The antigen then undergoes endocytosis to internalize the ADC into tumor cells, where it is transported to lysosomes to release the payload. The released toxic payloads can induce apoptosis through DNA damage or microtubule inhibition and can kill surrounding cancer cells through the bystander effect. The first ADC drug was approved by the United States Food and Drug Administration (FDA) in 2000, but the following decade saw no new approved ADC drugs. From 2011 to 2018, four ADC drugs were approved, while in 2019 and 2020 five more ADCs entered the market. This demonstrates an increasing trend for the clinical development of ADCs. This review summarizes the recent clinical research, with a specific focus on how the in vivo processing of ADCs influences their design. We aim to provide comprehensive information about current ADCs to facilitate future development.
Collapse
Affiliation(s)
- Yiming Jin
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Megan A Schladetsch
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Xueting Huang
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Marcy J Balunas
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Andrew J Wiemer
- Division of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA; Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
80
|
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
|
81
|
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
|
82
|
Peng H. Perspectives on the development of antibody-drug conjugates targeting ROR1 for hematological and solid cancers. Antib Ther 2021; 4:222-227. [PMID: 34805745 PMCID: PMC8597957 DOI: 10.1093/abt/tbab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/13/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are targeted therapeutics generated by conjugation of cytotoxic small molecules to monoclonal antibodies (mAbs) via chemical linkers. Due to their selective delivery of toxic payloads to antigen-positive cancer cells, ADCs demonstrate wider therapeutic indexes compared with conventional chemotherapy. After decades of intensive research and development, significant advances have been made in the field, leading to a total of 10 U.S. food and drug administration (FDA)-approved ADCs to treat cancer patients. Currently, ~80 ADCs targeting different antigens are under clinical evaluation for treatment of either hematological or solid malignancies. Notably, three ADCs targeting the same oncofetal protein, receptor tyrosine kinase like orphan receptor 1 (ROR1), have attracted considerable attention when they were acquired or licensed successively in the fourth quarter of 2020 by three major pharmaceutical companies. Apparently, ROR1 has emerged as an attractive target for cancer therapy. Since all the components of ADCs, including the antibody, linker and payload, as well as the conjugation method, play critical roles in ADC’s efficacy and performance, their choice and combination will determine how far they can be advanced. This review summarizes the design and development of current anti-ROR1 ADCs and highlights an emerging trend to target ROR1 for cancer therapy.
Collapse
Affiliation(s)
- Haiyong Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, 130 Scripps Way, C278, Jupiter, FL 33458, USA
| |
Collapse
|
83
|
Tracey SR, Smyth P, Barelle CJ, Scott CJ. Development of next generation nanomedicine-based approaches for the treatment of cancer: we've barely scratched the surface. Biochem Soc Trans 2021; 49:2253-2269. [PMID: 34709394 PMCID: PMC8589425 DOI: 10.1042/bst20210343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/23/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022]
Abstract
Interest in nanomedicines has grown rapidly over the past two decades, owing to the promising therapeutic applications they may provide, particularly for the treatment of cancer. Personalised medicine and 'smart' actively targeted nanoparticles represent an opportunity to deliver therapies directly to cancer cells and provide sustained drug release, in turn providing overall lower off-target toxicity and increased therapeutic efficacy. However, the successful translation of nanomedicines from encouraging pre-clinical findings to the clinic has, to date, proven arduous. In this review, we will discuss the use of nanomedicines for the treatment of cancer, with a specific focus on the use of polymeric and lipid nanoparticle delivery systems. In particular, we examine approaches exploring the surface functionalisation of nanomedicines to elicit active targeting and therapeutic effects as well as challenges and future directions for nanoparticles in cancer treatment.
Collapse
Affiliation(s)
- Shannon R. Tracey
- The Patrick G Johnston Centre for Cancer Research, Queen's University, 97 Lisburn Road, Belfast BT9 7AE, U.K
| | - Peter Smyth
- The Patrick G Johnston Centre for Cancer Research, Queen's University, 97 Lisburn Road, Belfast BT9 7AE, U.K
| | - Caroline J. Barelle
- Elasmogen Ltd, Liberty Building, Foresterhill Health Campus, Aberdeen AB25 2ZP, U.K
| | - Christopher J. Scott
- The Patrick G Johnston Centre for Cancer Research, Queen's University, 97 Lisburn Road, Belfast BT9 7AE, U.K
| |
Collapse
|
84
|
Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J Control Release 2021; 340:1-34. [PMID: 34673122 DOI: 10.1016/j.jconrel.2021.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
The antibody-drug conjugates (ADCs) are one the fastest growing biotherapeutics in oncology and are still in their infancy in gastrointestinal (GI) cancer for clinical applications to improve patient survival. The ADC based approach is developed with tumor specific antigen, antibody carrying cytotoxic agents to precisely target and deliver chemotherapeutics at the tumor site. To date, 11 ADCs have been approved by US-FDA, and more than 80 are in the clinical development phase for different oncological indications. However, The ADCs based therapies in GI cancers are still far from having high-efficient clinical outcomes. The limited success of these ADCs and lessons learned from the past are now being used to develop a newer generation of ADC against GI cancers. In this review, we did a comprehensive assessment of the key components of ADCs, including tumor marker, antibody, cytotoxic payload, and linkage strategy, with a focus on technical improvement and some future trends in the pipeline for clinical translation. The various preclinical and clinical ADCs used in gastrointestinal malignancies, their target, composition and bioconjugation, along with preclinical and clinical outcomes, are discussed. The emphasis is also given to new generation ADCs employing novel mAb, payload, linker, and bioconjugation methods are also included.
Collapse
Affiliation(s)
- Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Dheer
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilash Samykutty
- Stephenson Comprehensive Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
85
|
Humeau J, Le Naour J, Galluzzi L, Kroemer G, Pol JG. Trial watch: intratumoral immunotherapy. Oncoimmunology 2021; 10:1984677. [PMID: 34676147 PMCID: PMC8526014 DOI: 10.1080/2162402x.2021.1984677] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
While chemotherapy and radiotherapy remain the first-line approaches for the management of most unresectable tumors, immunotherapy has emerged in the past two decades as a game-changing treatment, notably with the clinical success of immune checkpoint inhibitors. Immunotherapies aim at (re)activating anticancer immune responses which occur in two main steps: (1) the activation and expansion of tumor-specific T cells following cross-presentation of tumor antigens by specialized myeloid cells (priming phase); and (2) the immunological clearance of malignant cells by these antitumor T lymphocytes (effector phase). Therapeutic vaccines, adjuvants, monoclonal antibodies, cytokines, immunogenic cell death-inducing agents including oncolytic viruses, anthracycline-based chemotherapy and radiotherapy, as well as adoptive cell transfer, all act at different levels of this cascade to (re)instate cancer immunosurveillance. Intratumoral delivery of these immunotherapeutics is being tested in clinical trials to promote superior antitumor immune activity in the context of limited systemic toxicity.
Collapse
Affiliation(s)
- Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Institut Universitaire de France, Paris, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Jonathan G. Pol
- Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin Bicêtre, France
| |
Collapse
|
86
|
Franzese O, Torino F, Giannetti E, Cioccoloni G, Aquino A, Faraoni I, Fuggetta MP, De Vecchis L, Giuliani A, Kaina B, Bonmassar E. Abscopal Effect and Drug-Induced Xenogenization: A Strategic Alliance in Cancer Treatment? Int J Mol Sci 2021; 22:ijms221910672. [PMID: 34639014 PMCID: PMC8509363 DOI: 10.3390/ijms221910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The current state of cancer treatment is still far from being satisfactory considering the strong impairment of patients' quality of life and the high lethality of malignant diseases. Therefore, it is critical for innovative approaches to be tested in the near future. In view of the crucial role that is played by tumor immunity, the present review provides essential information on the immune-mediated effects potentially generated by the interplay between ionizing radiation and cytotoxic antitumor agents when interacting with target malignant cells. Therefore, the radiation-dependent abscopal effect (i.e., a biological effect of ionizing radiation that occurs outside the irradiated field), the influence of cancer chemotherapy on the antigenic pattern of target neoplastic cells, and the immunogenic cell death (ICD) caused by anticancer agents are the main topics of this presentation. It is widely accepted that tumor immunity plays a fundamental role in generating an abscopal effect and that anticancer drugs can profoundly influence not only the host immune responses, but also the immunogenic pattern of malignant cells. Remarkably, several anticancer drugs impact both the abscopal effect and ICD. In addition, certain classes of anticancer agents are able to amplify already expressed tumor-associated antigens (TAA). More importantly, other drugs, especially triazenes, induce the appearance of new tumor neoantigens (TNA), a phenomenon that we termed drug-induced xenogenization (DIX). The adoption of the abscopal effect is proposed as a potential therapeutic modality when properly applied concomitantly with drug-induced increase in tumor cell immunogenicity and ICD. Although little to no preclinical or clinical studies are presently available on this subject, we discuss this issue in terms of potential mechanisms and therapeutic benefits. Upcoming investigations are aimed at evaluating how chemical anticancer drugs, radiation, and immunotherapies are interacting and cooperate in evoking the abscopal effect, tumor xenogenization and ICD, paving the way for new and possibly successful approaches in cancer therapy.
Collapse
Affiliation(s)
- Ornella Franzese
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Elisa Giannetti
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Giorgia Cioccoloni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK
| | - Angelo Aquino
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Isabella Faraoni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Maria Pia Fuggetta
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Liana De Vecchis
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Anna Giuliani
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, D-55131 Mainz, Germany
- Correspondence: (B.K.); (E.B.)
| | - Enzo Bonmassar
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
- Correspondence: (B.K.); (E.B.)
| |
Collapse
|
87
|
Hasan MM, Laws M, Jin P, Rahman KM. Factors influencing the choice of monoclonal antibodies for antibody-drug conjugates. Drug Discov Today 2021; 27:354-361. [PMID: 34597756 DOI: 10.1016/j.drudis.2021.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/20/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
In antibody-drug conjugates (ADCs), monoclonal antibodies (mAbs) act as carriers for a cytotoxic payload providing the therapy with targeted action against cells expressing a target cell surface antigen. An appropriate choice of mAb is crucial to developing a successful ADC for clinical development. However, problems such as immunogenicity, poor pharmacokinetic (PK) and pharmacodynamic (PD) profiles and variable drug-antibody ratios (DARs) plague ADCs. In this review, we detail recent mAb-based innovations and factors that should be considered to overcome these problems to achieve a new generation of more effective ADC therapeutics.
Collapse
Affiliation(s)
- Md Mahbub Hasan
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Mark Laws
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Peiqin Jin
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Khondaker Miraz Rahman
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
88
|
Shin C, Kim SS, Jo YH. Extending traditional antibody therapies: Novel discoveries in immunotherapy and clinical applications. Mol Ther Oncolytics 2021; 22:166-179. [PMID: 34514097 PMCID: PMC8416972 DOI: 10.1016/j.omto.2021.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Immunotherapy has been well regarded as one of the safer and antigen-specific anti-cancer treatments compared to first-generation chemotherapy. Since Coley's discovery, researchers focused on engineering novel antibody-based therapies. Including artificial and modified antibodies, such as antibody fragments, antibody-drug conjugates, and synthetic mimetics, the variety of immunotherapy has been rapidly expanding in the last few decades. Genetic and chemical modifications to monoclonal antibody have been brought into academia, in vivo trials, and clinical applications. Here, we have looked around antibodies overall. First, we elucidate the antibody structure and its cytotoxicity mechanisms. Second, types of therapeutic antibodies are presented. Additionally, there is a summarized list of US Food and Drug Administration (FDA)-approved therapeutic antibodies and recent clinical trials. This review provides a comprehensive overview of both the general function of therapeutic antibodies and a few main variations in development, including recent advent with the proposed mechanism of actions, and we introduce types of therapeutic antibodies, clinical trials, and approved commercial immunotherapeutic drugs.
Collapse
Affiliation(s)
- Charles Shin
- Chadwick International, Incheon 22002, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Hwa Jo
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
89
|
Lucas AT, Moody A, Schorzman AN, Zamboni WC. Importance and Considerations of Antibody Engineering in Antibody-Drug Conjugates Development from a Clinical Pharmacologist's Perspective. Antibodies (Basel) 2021; 10:30. [PMID: 34449544 PMCID: PMC8395454 DOI: 10.3390/antib10030030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/04/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Antibody-drug conjugates (ADCs) appear to be in a developmental boom, with five FDA approvals in the last two years and a projected market value of over $4 billion by 2024. Major advancements in the engineering of these novel cytotoxic drug carriers have provided a few early success stories. Although the use of these immunoconjugate agents are still in their infancy, valuable lessons in the engineering of these agents have been learned from both preclinical and clinical failures. It is essential to appreciate how the various mechanisms used to engineer changes in ADCs can alter the complex pharmacology of these agents and allow the ADCs to navigate the modern-day therapeutic challenges within oncology. This review provides a global overview of ADC characteristics which can be engineered to alter the interaction with the immune system, pharmacokinetic and pharmacodynamic profiles, and therapeutic index of ADCs. In addition, this review will highlight some of the engineering approaches being explored in the creation of the next generation of ADCs.
Collapse
Affiliation(s)
- Andrew T. Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Amber Moody
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Allison N. Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
| | - William C. Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (A.T.L.); (A.N.S.)
- Carolina Center of Cancer Nanotechnology Excellence, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Glolytics, LLC, Chapel Hill, NC 27517, USA
| |
Collapse
|
90
|
Mckertish CM, Kayser V. Advances and Limitations of Antibody Drug Conjugates for Cancer. Biomedicines 2021; 9:872. [PMID: 34440076 PMCID: PMC8389690 DOI: 10.3390/biomedicines9080872] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
The popularity of antibody drug conjugates (ADCs) has increased in recent years, mainly due to their unrivalled efficacy and specificity over chemotherapy agents. The success of the ADC is partly based on the stability and successful cleavage of selective linkers for the delivery of the payload. The current research focuses on overcoming intrinsic shortcomings that impact the successful development of ADCs. This review summarizes marketed and recently approved ADCs, compares the features of various linker designs and payloads commonly used for ADC conjugation, and outlines cancer specific ADCs that are currently in late-stage clinical trials for the treatment of cancer. In addition, it addresses the issues surrounding drug resistance and strategies to overcome resistance, the impact of a narrow therapeutic index on treatment outcomes, the impact of drug-antibody ratio (DAR) and hydrophobicity on ADC clearance and protein aggregation.
Collapse
Affiliation(s)
| | - Veysel Kayser
- Sydney School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
91
|
Site-specific glycan-conjugated NISTmAb antibody drug conjugate mimetics: synthesis, characterization, and utility. Anal Bioanal Chem 2021; 413:4989-5001. [PMID: 34231000 DOI: 10.1007/s00216-021-03460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 10/20/2022]
Abstract
Antibody drug conjugates (ADCs) represent a rapidly growing modality for the treatment of numerous oncology indications. The complexity of analytical characterization method development is increased due to the potential for synthetic intermediates and process-related impurities. In addition, the cytotoxicity of such materials provides an additional challenge with regard to handling products and/or sharing materials with analytical collaborators and/or vendors for technology development. Herein, we have utilized a site-specific chemoenzymatic glycoconjugation strategy for preparing ADC mimetics composed of the NIST monoclonal antibody (NISTmAb) conjugated to non-cytotoxic payloads representing both small molecules and peptides. The materials were exhaustively characterized with high-resolution mass spectrometry-based approaches to demonstrate the utility of each analytical method for confirming the conjugation fidelity as well as deep characterization of low-abundance synthetic intermediates and impurities arising from payload raw material heterogeneity. These materials therefore represent a widely available test metric to develop novel ADC analytical methods as well as a platform to discuss best practices for extensive characterization.
Collapse
|
92
|
Sadraeian M, da Cruz EF, Boyle RW, Bahou C, Chudasama V, Janini LMR, Diaz RS, Guimarães FEG. Photoinduced Photosensitizer-Antibody Conjugates Kill HIV Env-Expressing Cells, Also Inactivating HIV. ACS OMEGA 2021; 6:16524-16534. [PMID: 34235324 PMCID: PMC8246456 DOI: 10.1021/acsomega.1c01721] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/27/2021] [Indexed: 06/01/2023]
Abstract
HIV-infected cells persist for decades in patients administered with antiretroviral therapy (ART). Meanwhile, an alarming surge in drug-resistant HIV viruses has been occurring. Addressing these issues, we propose the application of photoimmunotherapy (PIT) against not only HIV Env-expressing cells but also HIV. Previously, we showed that a human anti-gp41 antibody (7B2) conjugated to cationic or anionic photosensitizers (PSs) could specifically target and kill the HIV Env-expressing cells. Here, our photolysis studies revealed that the binding of photoimmunoconjugates (PICs) on the membrane of HIV Env-expressing cells is sufficient to induce necrotic cell death due to physical damage to the membrane by singlet oxygen, which is independent of the type of PSs. This finding persuaded us to study the virus photoinactivation of PICs using two HIV-1 strains, X4 HIV-1 NL4-3 and JR-CSF virus. We observed that the PICs could destroy the viral strains, probably via physical damage on the HIV envelope. In conclusion, we report the application of PIT as a possible dual-tool for HIV immunotherapy and ART by killing HIV-expressing cells and cell-free HIV, respectively.
Collapse
Affiliation(s)
- Mohammad Sadraeian
- São Carlos Institute of Physics, University of São Paulo, Caixa Postal 369, São Carlos, SP CEP 13560-970, Brazil
| | | | - Ross W Boyle
- Department of Chemistry, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Calise Bahou
- Department of Chemistry, University College London, London WC1H 0AJ, U.K
| | - Vijay Chudasama
- Department of Chemistry, University College London, London WC1H 0AJ, U.K
| | | | - Ricardo Sobhie Diaz
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Francisco E G Guimarães
- São Carlos Institute of Physics, University of São Paulo, Caixa Postal 369, São Carlos, SP CEP 13560-970, Brazil
| |
Collapse
|
93
|
Su D, Zhang D. Linker Design Impacts Antibody-Drug Conjugate Pharmacokinetics and Efficacy via Modulating the Stability and Payload Release Efficiency. Front Pharmacol 2021; 12:687926. [PMID: 34248637 PMCID: PMC8262647 DOI: 10.3389/fphar.2021.687926] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 01/03/2023] Open
Abstract
The development of antibody-drug conjugates (ADCs) has significantly been advanced in the past decade given the improvement of payloads, linkers and conjugation methods. In particular, linker design plays a critical role in modulating ADC stability in the systemic circulation and payload release efficiency in the tumors, which thus affects ADC pharmacokinetic (PK), efficacy and toxicity profiles. Previously, we have investigated key linker parameters such as conjugation chemistry (e.g., maleimide vs. disulfide), linker length and linker steric hindrance and their impacts on PK and efficacy profiles. Herein, we discuss our perspectives on development of integrated strategies for linker design to achieve a balance between ADC stability and payload release efficiency for desired efficacy in antigen-expressing xenograft models. The strategies have been successfully applied to the design of site-specific THIOMABTM antibody-drug conjugates (TDCs) with different payloads. We also propose to conduct dose fractionation studies to gain guidance for optimal dosing regimens of ADCs in pre-clinical models.
Collapse
Affiliation(s)
- Dian Su
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| | - Donglu Zhang
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
94
|
Matsuda Y, Chakrabarti A, Takahashi K, Yamada K, Nakata K, Okuzumi T, Mendelsohn BA. Chromatographic analysis of site-specific antibody-drug conjugates produced by AJICAP first-generation technology using a recombinant FcγIIIa receptor-ligand affinity column. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1177:122753. [PMID: 34098178 DOI: 10.1016/j.jchromb.2021.122753] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/24/2021] [Accepted: 04/29/2021] [Indexed: 01/08/2023]
Abstract
Commercially approved conventional antibody-drug conjugates (ADCs) are produced as heterogeneous mixtures containing a stochastic distribution of payloads decorating the antibody molecules resulting in decreased efficacy and thus lowering their therapeutic index. Control of the DAR and conjugation site in the development of next-generation ADCs is believed to assist in increasing the therapeutic index of these targeted biologics leading to overall enhanced clinical efficacy and reduced toxicity. A chemical site-specific conjugation technology termed AJICAP® allows ADC developers to control both the location and quantity of the payload conjugation to an antibody. Furthermore, this simplified ADC composition enables a streamlined chemical analysis. Here we report the chromatographic separation of site-specific ADCs produced by AJICAP® technology using an analytical affinity chromatography HPLC column containing a recombinant FcγIIIa receptor-ligand immobilized on a non-porous polymer resin (NPR). These HPLC analyses provided visually clear chromatogram results reflecting the heterogeneity of each ADC. The affinity strength was also measured by biolayer interferometry (BLI) and predicted by molecular structure analysis. The results indicate that AJICAP® technology is a promising solution to link hydrophobic payloads to antibodies without compromising antibody receptor function. This study also shows that FcγIIIa-NPR column can be used to characterize site-specific conjugated ADCs compared to ADCs synthesized using conventional methods.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan.
| | - Atis Chakrabarti
- Tosoh Bioscience, 3604 Horizon Drive, Suite 100, King of Prussia, PA 19406, USA
| | | | - Kei Yamada
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Kunio Nakata
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Tatsuya Okuzumi
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kanagawa 210-8681, Japan
| | - Brian A Mendelsohn
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA 92121, USA
| |
Collapse
|
95
|
Bocharova EA, Kopytina NI, Slynko ЕЕ. Anti-tumour drugs of marine origin currently at various stages of clinical trials (review). REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oncological diseases for a long time have remained one of the most significant health problems of modern society, which causes great losses in its labour and vital potential. Contemporary oncology still faces unsolved issues as insufficient efficacy of treatment of progressing and metastatic cancer, chemoresistance, and side-effects of the traditional therapy which lead to disabilities among or death of a high number of patients. Development of new anti-tumour preparations with a broad range of pharmaceutical properties and low toxicity is becoming increasingly relevant every year. The objective of the study was to provide a review of the recent data about anti-tumour preparations of marine origin currently being at various phases of clinical trials in order to present the biological value of marine organisms – producers of cytotoxic compounds, and the perspectives of their use in modern biomedical technologies. Unlike the synthetic oncological preparations, natural compounds are safer, have broader range of cytotoxic activity, can inhibit the processes of tumour development and metastasis, and at the same time have effects on several etiopathogenic links of carcinogenesis. Currently, practical oncology uses 12 anti-tumour preparations of marine origin (Fludarabine, Cytarabine, Midostaurin, Nelarabine, Eribulin mesylate, Brentuximab vedotin, Trabectedin, Plitidepsin, Enfortumab vedotin, Polatuzumab vedotin, Belantamab mafodotin, Lurbinectedin), 27 substances are at different stages of clinical trials. Contemporary approaches to the treatment of oncological diseases are based on targeted methods such as immune and genetic therapies, antibody-drug conjugates, nanoparticles of biopolymers, and metals. All those methods employ bioactive compounds of marine origin. Numerous literature data from recent years indicate heightened attention to the marine pharmacology and the high potential of marine organisms for the biomedicinal and pharmaceutic industries.
Collapse
|
96
|
Edupuganti VVSR, Tyndall JDA, Gamble AB. Self-immolative Linkers in Prodrugs and Antibody Drug Conjugates in Cancer Treatment. Recent Pat Anticancer Drug Discov 2021; 16:479-497. [PMID: 33966624 DOI: 10.2174/1574892816666210509001139] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/09/2021] [Accepted: 03/02/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND The design of anti-cancer therapies with high anti-tumour efficacy and reduced toxicity continues to be challenging. Anti-cancer prodrug and antibody-drug-conjugate (ADC) strategies that can specifically and efficiently deliver cytotoxic compounds to cancer cells have been used to overcome some of the challenges. Key to the success of many of these strategies is a self-immolative linker, which after activation can release the drug payload. Various types of triggerable self-immolative linkers are used in prodrugs and ADCs to improve their efficacy and safety. OBJECTIVE Numerous patents have reported the significance of self-immolative linkers in prodrugs and ADCs in cancer treatment. Based on the recent patent literature, we summarise methods for designing the site-specific activation of non-toxic prodrugs and ADCs in order to improve selectivity for killing cancer cells. METHODS In this review, an integrated view of the potential use of prodrugs and ADCs in cancer treatment are provided. This review presents recent patents and related publications over the past ten years to 2020. RESULTS The recent patent literature has been summarised for a wide variety of self-immolative PABC linkers, which are cleaved by factors including responding to the difference between the extracellular and intracellular environments (pH, ROS, glutathione), by over-expressed enzymes (cathepsin, plasmin, β-glucuronidase) or bioorthogonal activation. The mechanism for self-immolation involves the linker undergoing a 1,4- or 1,6-elimination (via electron cascade) or intramolecular cyclisation to release cytotoxic drug at the targeted site. CONCLUSION This review provides the commonly used strategies from recent patent literature in the development of prodrugs based on targeted cancer therapy and antibody-drug conjugates, which show promising results in therapeutic applications.
Collapse
Affiliation(s)
| | - Joel D A Tyndall
- School of Pharmacy, University of Otago, Dunedin, 9054. New Zealand
| | - Allan B Gamble
- School of Pharmacy, University of Otago, Dunedin, 9054. New Zealand
| |
Collapse
|
97
|
Capone E, Lattanzio R, Gasparri F, Orsini P, Rossi C, Iacobelli V, De Laurenzi V, Natali PG, Valsasina B, Iacobelli S, Sala G. EV20/NMS-P945, a Novel Thienoindole Based Antibody-Drug Conjugate Targeting HER-3 for Solid Tumors. Pharmaceutics 2021; 13:pharmaceutics13040483. [PMID: 33918158 PMCID: PMC8066800 DOI: 10.3390/pharmaceutics13040483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
HER-3 is becoming an attractive target for antibody-drug conjugate (ADC)-based therapy. Indeed, this receptor and its ligands are found to be overexpressed in several malignancies, and re-activation of its downstream signaling axis is known to play a critical role in modulating the sensitivity of targeted therapeutics in different tumors. In this study, we generated a novel ADC named EV20/NMS-P945 by coupling the anti-HER-3 antibody EV20 with a duocarmycin-like derivative, the thienoindole (TEI) NMS-P528, a DNA minor groove alkylating agent through a peptidic cleavable linker. This ADC showed target-dependent cytotoxic activity in vitro on several tumor cell lines and therapeutic activity in mouse xenograft tumor models, including those originating from pancreatic, prostatic, head and neck, gastric and ovarian cancer cells and melanoma. Pharmacokinetics and toxicological studies in monkeys demonstrated that this ADC possesses a favorable terminal half-life and stability and it is well tolerated. These data support further EV20/NMS-P945 clinical development as a therapeutic agent against HER-3-expressing malignancies.
Collapse
Affiliation(s)
- Emily Capone
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Fabio Gasparri
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Paolo Orsini
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Cosmo Rossi
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | - Valentina Iacobelli
- Department of Gynecology and Obstetrics, Catholic University, 00168 Rome, Italy;
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
| | | | - Barbara Valsasina
- Nerviano Medical Sciences Srl, 20014 Milan, Italy; (F.G.); (P.O.); (B.V.)
| | - Stefano Iacobelli
- MediaPharma s.r.l., Via della Colonnetta 50/A, 66100 Chieti, Italy;
- Correspondence: or (S.I.); (G.S.); Tel.: +39-08-7154-1504 (G.S.)
| | - Gianluca Sala
- Department of Innovative Technologies in Medicine & Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (E.C.); (R.L.); (V.D.L.)
- Center for Advanced Studies and Technology (CAST), Via Polacchi 11, 66100 Chieti, Italy;
- Correspondence: or (S.I.); (G.S.); Tel.: +39-08-7154-1504 (G.S.)
| |
Collapse
|
98
|
Merkul E, Muns JA, Sijbrandi NJ, Houthoff H, Nijmeijer B, Rheenen G, Reedijk J, Dongen GAMS. An Efficient Conjugation Approach for Coupling Drugs to Native Antibodies via the Pt
II
Linker
Lx
for Improved Manufacturability of Antibody–Drug Conjugates. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202011593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Eugen Merkul
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Joey A. Muns
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Niels J. Sijbrandi
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Hendrik‐Jan Houthoff
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Bart Nijmeijer
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Gerro Rheenen
- Chemistry Department LinXis BV De Boelelaan 1085c Amsterdam 1081 HV The Netherlands
| | - Jan Reedijk
- Leiden Institute of Chemistry Leiden University PO Box 9502 2300 RA Leiden The Netherlands
| | - Guus A. M. S. Dongen
- Department of Radiology and Nuclear Medicine Amsterdam UMC, location VU medical center Amsterdam The Netherlands
| |
Collapse
|
99
|
Merkul E, Muns JA, Sijbrandi NJ, Houthoff H, Nijmeijer B, van Rheenen G, Reedijk J, van Dongen GAMS. An Efficient Conjugation Approach for Coupling Drugs to Native Antibodies via the Pt II Linker Lx for Improved Manufacturability of Antibody-Drug Conjugates. Angew Chem Int Ed Engl 2021; 60:3008-3015. [PMID: 33185916 PMCID: PMC7986738 DOI: 10.1002/anie.202011593] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/29/2020] [Indexed: 12/20/2022]
Abstract
The PtII linker [ethylenediamineplatinum(II)]2+ , coined Lx, has emerged as a novel non-conventional approach to antibody-drug conjugates (ADCs) and has shown its potential in preclinical in vitro and in vivo benchmark studies. A crucial improvement of the Lx conjugation reaction from initially <15 % to ca. 75-90 % conjugation efficiency is described, resulting from a systematic screening of all relevant reaction parameters. NaI, a strikingly simple inorganic salt additive, greatly improves the conjugation efficiency as well as the conjugation selectivity simply by exchanging the leaving chloride ligand on Cl-Lx-drug complexes (which are direct precursors for Lx-ADCs) for iodide, thus generating I-Lx-drug complexes as more reactive species. Using this iodide effect, we developed a general and highly practical conjugation procedure that is scalable: our lead Lx-ADC was produced on a 5 g scale with an outstanding conjugation efficiency of 89 %.
Collapse
Affiliation(s)
- Eugen Merkul
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Joey A. Muns
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Niels J. Sijbrandi
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | | | - Bart Nijmeijer
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Gerro van Rheenen
- Chemistry DepartmentLinXis BVDe Boelelaan 1085cAmsterdam1081HVThe Netherlands
| | - Jan Reedijk
- Leiden Institute of ChemistryLeiden UniversityPO Box 95022300RALeidenThe Netherlands
| | - Guus A. M. S. van Dongen
- Department of Radiology and Nuclear MedicineAmsterdam UMC, location VU medical centerAmsterdamThe Netherlands
| |
Collapse
|
100
|
Zavoiura O, Brunner B, Casteels P, Zimmermann L, Ozog M, Boutton C, Helms MW, Wagenaar T, Adam V, Peterka J, Metz-Weidmann C, Deschaght P, Scheidler S, Jahn-Hofmann K. Nanobody-siRNA Conjugates for Targeted Delivery of siRNA to Cancer Cells. Mol Pharm 2021; 18:1048-1060. [PMID: 33444501 DOI: 10.1021/acs.molpharmaceut.0c01001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Targeted extrahepatic delivery of siRNA remains a challenging task in the field of nucleic acid therapeutics. An ideal delivery tool must internalize siRNA exclusively into the cells of interest without affecting the silencing activity of siRNA. Here, we report the use of anti-EGFR Nanobodies (trademark of Ablynx N.V.) as tools for targeted siRNA delivery. A straightforward procedure for site-specific conjugation of siRNA to an engineered C-terminal cysteine residue on the Nanobody (trademark of Ablynx N.V.) is described. We show that siRNA-conjugated Nanobodies (Nb-siRNA) retain their binding to EGFR and enter EGFR-positive cells via receptor-mediated endocytosis. The activity of Nb-siRNAs was assessed by measuring the knockdown of a housekeeping gene (AHSA1) in EGFR-positive and EGFR-negative cells. We demonstrate that Nb-siRNAs are active in vitro and induce mRNA cleavage in the targeted cell line. In addition, we discuss the silencing activity of siRNA conjugated to fused Nbs with various combinations of EGFR-binding building blocks. Finally, we compare the performance of Nb-siRNA joined by four different linkers and discuss the advantages and limitations of using cleavable and noncleavable linkers in the context of Nanobody-mediated siRNA delivery.
Collapse
Affiliation(s)
- Oleksandr Zavoiura
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Bodo Brunner
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Peter Casteels
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Luciana Zimmermann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Matthias Ozog
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Carlo Boutton
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Mike W Helms
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Timothy Wagenaar
- Sanofi, Research, 640 Memorial Drive, Cambridge, 02139 Massachusetts, United States
| | - Volker Adam
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Josefine Peterka
- Sanofi, TIDES platform, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | | | - Pieter Deschaght
- Ablynx, a Sanofi Company, Technologiepark 21, 9052 Zwijnaarde, Belgium
| | - Sabine Scheidler
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Kerstin Jahn-Hofmann
- Sanofi, Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| |
Collapse
|