1
|
Chazeau E, Pipier A, Wegner KD, Ghiringhelli F, Sancey L, Paul C, Goze C. NIR-II aza-BODIPY Platform for the Development of a Fluorescent Antibody Drug Conjugate. J Med Chem 2025; 68:7232-7242. [PMID: 40152348 DOI: 10.1021/acs.jmedchem.4c02777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Real-time imaging of antibody-drug conjugates (ADCs) offers valuable insights for assessing tumor targeting specificity, monitoring therapeutic efficacy, and detecting off-target accumulation that may cause adverse effects. To enable precise tracking, we developed a versatile fluorescent platform based on an NIR-II emitting aza-BODIPY dye, which can be site-specifically grafted onto an IgG1 antibody to generate well-defined fluorescent ADCs. As a proof of concept, we synthesized an HER2-targeting trastuzumab immunoconjugate bearing a NIR-II aza-BODIPY fluorophore. The cytotoxic monomethyl auristatin E (MMAE) payload was introduced in the final step, resulting in a trackable and homogeneous ADC suitable for both in vitro and in vivo investigations. The resulting Trastu-azaNIRII-MMAE selectively accumulated in HER2-positive subcutaneous tumors, significantly reducing the tumor growth. Using NIR-II optical imaging, a single injection of the NIR-II-ADC allowed for the detection of the conjugate over a period of more than one month, highlighting its potential for long-term tracking and therapeutic applications.
Collapse
Affiliation(s)
- Elisa Chazeau
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
| | - Angélique Pipier
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| | - K David Wegner
- Division Biophotonics, Federal Institute for Materials Research and Testing (BAM), 12489 Berlin, Germany
| | - François Ghiringhelli
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
- Plateforme de Transfert en Biologie Cancérologique, CGFL, 21000 Dijon, France
| | - Lucie Sancey
- Université Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000 Grenoble, France
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000 Paris, France
- CTM, UMR1231 INSERM, Université de Bourgogne, 21000 Dijon, France
| | - Christine Goze
- ICMUB, UMR 6302 CNRS, Université Bourgogne Europe, 9 av. A. Savary, BP 47870, 21078 Dijon, France
| |
Collapse
|
2
|
Zhang T, Ouyang Z, Zhang Y, Sun H, Kong L, Xu Q, Qu J, Sun Y. Marine Natural Products in Inflammation-Related Diseases: Opportunities and Challenges. Med Res Rev 2025. [PMID: 40202793 DOI: 10.1002/med.22109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
In recent decades, the potentiality of marine natural products (MNPs) in the medical field has been increasingly recognized. Natural compounds derived from marine microorganisms, algae, and invertebrates have shown significant promise for treating inflammation-related diseases. In this review, we cover the three primary sources of MNPs and their diverse and unique chemical structures and bioactivities. This review aims to summarize the progress of MNPs in combating inflammation-related diseases. Moreover, we cover the functions and mechanisms of MNPs in diseases, highlighting their functions in regulating inflammatory signaling pathways, cellular stress responses, and gut microbiota, among others. Meanwhile, we focus on key technologies and scientific methods to address the current limitations and challenges in MNPs.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Yueran Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Sureda A, Pavlovsky A, Haidar D, Kristo F, Stache V, Zomas A. Real-world outcomes of brentuximab vedotin as consolidation therapy after autologous stem cell transplantation in relapsed/refractory Hodgkin lymphoma: A systematic review and meta-analysis. Bone Marrow Transplant 2025:10.1038/s41409-025-02557-7. [PMID: 40200006 DOI: 10.1038/s41409-025-02557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 04/10/2025]
Abstract
Brentuximab vedotin (BV) as post-autologous stem cell transplantation (ASCT) consolidation was shown to reduce the relapse risk among high-risk patients with relapsed/refractory Hodgkin lymphoma (RRHL) in the clinical trial setting. This systematic review and meta-analysis characterizes real-world evidence (RWE) on the effectiveness and safety of BV as post-ASCT consolidation in 1504 adult and pediatric patients with RRHL from 23 studies across 17 countries. A random-effects model yielded pooled progression-free survival (PFS) and overall survival rates (OS); PFS: 2-year, 74.2%; 5-year, 65.8%; OS: 2-year, 95.8%; 5-year, 91.9%. The most common any-grade adverse events were neuropathy (34.2%) and neutropenia (20.2%). Despite heterogeneity in populations and outcomes, this analysis utilizing real-world data corroborates the efficacy and safety of BV as post-ASCT consolidation in RRHL reported in the experimental arm of the Phase III AETHERA trial. The favorable PFS results in cases exposed to BV prior to ASCT indicate the value of BV in controlling Hodgkin lymphoma (HL) in the salvage setting. Continued research is essential to refine BV treatment strategies amid the evolving treatment landscape.
Collapse
Affiliation(s)
- Anna Sureda
- Institut Català d'Oncologia - Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, Barcelona, Spain.
| | - Astrid Pavlovsky
- FUNDALEU Research Center, Buenos Aires, Argentina
- Centro de Hematologia Pavlovsky, Buenos Aires, Argentina
- Grupo Argentino de Tratamiento de la Leucemia Aguda (GATLA), Buenos Aires, Argentina
| | - Dalah Haidar
- Takeda Pharmaceuticals International AG - Singapore Branch, Singapore, Singapore
| | - Fjoralba Kristo
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Vanessa Stache
- Takeda Pharmaceuticals International AG, Zürich, Switzerland
| | | |
Collapse
|
4
|
Schauenburg D, Weil T. Not So Bioorthogonal Chemistry. J Am Chem Soc 2025; 147:8049-8062. [PMID: 40017419 PMCID: PMC11912343 DOI: 10.1021/jacs.4c15986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
The advent of bioorthogonal chemistry has transformed scientific research, offering a powerful tool for selective and noninvasive labeling of (bio)molecules within complex biological environments. This innovative approach has facilitated the study of intricate cellular processes, protein dynamics, and interactions. Nevertheless, a number of challenges remain to be addressed, including the need for improved reaction kinetics, enhanced biocompatibility, and the development of a more diverse and orthogonal set of reactions. While scientists continue to search for veritable solutions, bioorthogonal chemistry remains a transformative tool with a vast potential for advancing our understanding of biology and medicine. This Perspective offers insights into reactions commonly classified as "bioorthogonal", which, however, may not always demonstrate the desired selectivity regarding the interactions between their components and the additives or catalysts used under the reaction conditions.
Collapse
Affiliation(s)
- Dominik Schauenburg
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Tanja Weil
- Max
Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
5
|
Jiang J, Jian S, Lin F, Zhang Z, Shen Q, Long G, Yang B, Lin J, Fang Y, Ding J, Liu J, Chen Y, Hu Y. Development of ASGR-Mediated Hepatocyte-Targeting Cytotoxic Drug Conjugates with CTSB-Cleavable Linkers Incorporating Succinimide and Succinic Acid Monoamide. J Med Chem 2025; 68:4382-4396. [PMID: 39918134 DOI: 10.1021/acs.jmedchem.4c02232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The ASGR-mediated endocytosis has been successfully applied to the hepatocyte-targeted delivery of therapeutic oligonucleotides via glycoconjugates. However, few studies have explored the conjugated small molecules due to the challenge of cleaving suitable linkers for the release of active small molecules, which is especially different from GalNAc-ONs cleaved by the deoxyribonuclease II in the lysosome. In this study, GalNAc-MMAE conjugates linked by CTSB-cleavable linkers were designed and synthesized. A comprehensive approach revealed that the conjugates were endocytosed by ASGR and subsequently hydrolyzed by CTSB, releasing MMAE. The optimized conjugate with a succinic acid monoamide as the fragment of the linker demonstrated favorable plasma stability, excellent biodistribution, and significant antitumor activities in vivo with weight gain at the effective dose in an orthotopic hepatocellular carcinoma mouse model. This research provides a strategy for developing anti-HCC therapeutic agents using GalNAc drug conjugates with CTSB-cleavable linkers to release active small molecules.
Collapse
Affiliation(s)
- Jingsheng Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Sigang Jian
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feifei Lin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zhuo Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- School of Chinese Materia Medica, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
| | - Qianqian Shen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guozhang Long
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, Xinjiang, China
| | - Biyu Yang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Junzhen Lin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yanfen Fang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | - Jian Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Jia Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, First Xiangshan Branch Alley, Hangzhou 310024, China
| | - Yi Chen
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Youhong Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu-ChongZhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- School of Chinese Materia Medica, College of Pharmacy, Nanjing University of Chinese Medicine, No. 138 Xianlin Road, Nanjing 210023, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, First Xiangshan Branch Alley, Hangzhou 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| |
Collapse
|
6
|
Li S, Zhao X, Fu K, Zhu S, Pan C, Yang C, Wang F, To KK, Fu L. Resistance to antibody-drug conjugates: A review. Acta Pharm Sin B 2025; 15:737-756. [PMID: 40177568 PMCID: PMC11959940 DOI: 10.1016/j.apsb.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 04/05/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are antitumor drugs composed of monoclonal antibodies and cytotoxic payload covalently coupled by a linker. Currently, 15 ADCs have been clinically approved worldwide. More than 100 clinical trials at different phases are underway to investigate the newly developed ADCs. ADCs represent one of the fastest growing classes of targeted antitumor drugs in oncology drug development. It takes advantage of the specific targeting of tumor-specific antigen by antibodies to deliver cytotoxic chemotherapeutic drugs precisely to tumor cells, thereby producing promising antitumor efficacy and favorable adverse effect profiles. However, emergence of drug resistance has severely hindered the clinical efficacy of ADCs. In this review, we introduce the structure and mechanism of ADCs, describe the development of ADCs, summarized the latest research about the mechanisms of ADC resistance, discussed the strategies to overcome ADCs resistance, and predicted biomarkers for treatment response to ADC, aiming to contribute to the development of ADCs in the future.
Collapse
Affiliation(s)
- Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinyu Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kenneth K.W. To
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
7
|
Abdelhamid MS, Wadan AHS, Saad HA, El-Dakroury WA, Hageen AW, Mohammed DH, Mourad S, Mohammed OA, Abdel-Reheim MA, Doghish AS. Nanoparticle innovations in targeted cancer therapy: advancements in antibody-drug conjugates. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03764-7. [PMID: 39825965 DOI: 10.1007/s00210-024-03764-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising strategy in targeted cancer therapy, enabling the precise delivery of cytotoxic agents to tumor sites while minimizing systemic toxicity. However, traditional ADCs face significant limitations, including restricted drug loading capacity, where an optimal drug-to-antibody ratio (DAR) is crucial; low DARs may lead to insufficient potency, while high DARs can cause rapid clearance and increased toxicity. Additionally, ADCs often suffer from instability in circulation due to the potential for premature release of cytotoxic agents, resulting in off-target effects and reduced therapeutic efficacy. Furthermore, their large size can impede adequate penetration into solid tumors, particularly in heterogeneous environments with varying antigen expressions. This review explores the innovative use of nanoparticles as carriers for ADCs, which offers a multifaceted approach to enhance therapeutic efficacy. By leveraging the unique properties of nanoparticles, such as their small size and ability to exploit the enhanced permeability and retention (EPR) effect, researchers can improve drug stability, prolong circulation time, and achieve more effective tumor targeting. Recent studies demonstrate that nanoparticle-encapsulated ADCs can significantly enhance treatment outcomes while reducing off-target effects, as evidenced by improved targeting capabilities and reduced toxicity in preclinical models. Despite the promising advancements, challenges remain, including potential nanoparticle toxicity and manufacturing complexities. This review aims to provide a comprehensive overview of the current research on nanoparticle-encapsulated ADCs. It highlights their potential to transform cancer treatment and offers insights into future directions for optimizing these advanced therapeutic strategies.
Collapse
Affiliation(s)
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, 15888, Suez Governorate, Egypt
| | - Hager Adel Saad
- Faculty of Pharmacy, German University in Cairo (GUC), New Cairo, 11835, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed W Hageen
- Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | | | - Sohaila Mourad
- Faculty of Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
8
|
Akiba H, Ise T, Satoh R, Abe Y, Tsumoto K, Ohno H, Kamada H, Nagata S. Generation of antagonistic biparatopic anti-CD30 antibody from an agonistic antibody by precise epitope determination and utilization of structural characteristics of CD30 molecule. Antib Ther 2025; 8:56-67. [PMID: 39958564 PMCID: PMC11826918 DOI: 10.1093/abt/tbaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/06/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
Background CD30 is a member of the tumor necrosis factor receptor superfamily. Recently, blocking CD30-dependent intracellular signaling has emerged as potential strategy for immunological regulation. Development of antibody-based CD30 antagonists is therefore of significant interest. However, a key challenge is that the bivalent form of natural antibody can crosslink CD30 molecules, leading to signal transduction even in the absence of specific ligand, CD153. Biparatopic antibodies (BpAbs) offer a solution, using two different variable fragments (Fvs) to bind distinct epitopes on a single antigen molecule. BpAbs format is an attractive alternative of natural antibody by potentially avoiding unwanted crosslinking and signaling induction. Methods We systematically characterized 36 BpAbs, each designed with pairs of Fvs binding to nine distinct epitopes across the CD30 extracellular domain. We first identified the precise epitope sites of the nine antibodies by assessing the binding to multiple orthologous CD30 proteins and mutants. We then produced the 36 BpAbs and analyzed their biological activities and binding modes. Results Among 36 BpAbs, we identified both potent ligand-independent agonists and ligand-blocking antagonists, with many displayed reduced signal activation, including 1:1-binding antagonists derived from AC10, a strong agonist developed for lymphoma therapy. Epitope dependency in reduced signaling activity was observed and associated with the flexible nature of CD30 protein. Conclusions We successfully developed antagonistic BpAbs against CD30 by controlling the stoichiometry of antibody-antigen binding mode. This study elucidated the mechanism of signaling induction, informing the design strategies of the development of biparatopic antibodies.
Collapse
Affiliation(s)
- Hiroki Akiba
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Advanced Biopharmaceuticals, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Tomoko Ise
- Laboratory of Antibody Design, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Reiko Satoh
- Laboratory of Advanced Biopharmaceuticals, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Yasuhiro Abe
- Division of Drugs, National Institute of Health Sciences, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | - Kouhei Tsumoto
- School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Advanced Biopharmaceuticals, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Haruhiko Kamada
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Advanced Biopharmaceuticals, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Satoshi Nagata
- Laboratory of Antibody Design, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
9
|
Zhao Y, Qiu W, Weng X, Gu C, Li S. ALK + anaplastic large cell lymphoma involving the bladder: case report and review of the literature. Diagn Pathol 2024; 19:157. [PMID: 39695732 DOI: 10.1186/s13000-024-01585-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
We reported a case of anaplastic lymphoma kinase (ALK)-positive anaplastic large cell lymphoma (ALK + ALCL) involving the bladder. The patient was a 27-year-old female, whose main clinical symptoms included fever, painless lymphadenopathy, and hematuria. Imaging studies suggested a bladder mass. The bladder mass was maximally resected through transurethral bladder tumor resection. The pathology report indicated a malignant tumor of the bladder. Based on immunohistochemical and gene rearrangement results, the diagnosis was confirmed as ALK + ALCL. After undergoing five cycles of treatment with the BV + CHP chemotherapy regimen, the patient's condition is currently stable, and no tumor recurrence was observed upon re-examination. ALK + ALCL involving the bladder is very rare, and early diagnosis is challenging. By reviewing the diagnostic and treatment process of this patient, and in conjunction with a review of modern literature on the disease's incidence characteristics, treatment protocols, and prognosis, this aims to provide a reference for clinicians in diagnosing and treating this condition, thereby reducing delays.
Collapse
Affiliation(s)
- Ying Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenqiang Qiu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiangtao Weng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Traditional Chinese Medicine), Guangzhou, China
| | - Chiming Gu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Traditional Chinese Medicine), Guangzhou, China.
| | - Siyi Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Traditional Chinese Medicine), Guangzhou, China.
| |
Collapse
|
10
|
Saberi SA, Cheng D, Nambudiri VE. Antibody-drug conjugates: A review of cutaneous adverse effects. J Am Acad Dermatol 2024; 91:922-931. [PMID: 39047980 DOI: 10.1016/j.jaad.2024.07.1463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/30/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
Antibody-drug conjugates (ADCs) are an emerging class of anticancer agents that combine targeting antibodies with potent cytotoxic agents. Their molecular configuration allows for increased therapeutic efficacy and reduced adverse-effect profiles compared to monoclonal antibodies or cytotoxic chemotherapy alone. ADCs cause off-target toxicities through several mechanisms, including premature deconjugation of the cytotoxic agent in the serum and the presence of the targeted antigen on normal tissues. Given cutaneous adverse events comprise 31.3% of all-grade adverse events in clinical trials involving ADCs, dermatologists are increasingly called upon to manage the cutaneous toxicities caused by these drugs. In this review, we summarize known cutaneous toxicities of the ADCs that have been approved for use by the US Food and Drug Administration to date. Dermatologists can play a key role in recognizing cutaneous reactions associated with ADCs, contributing to guidelines for their management, and aiding during clinical trials to generate detailed morphologic and histopathologic descriptions of cutaneous toxicities caused by ADCs.
Collapse
Affiliation(s)
- Shahin A Saberi
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Debby Cheng
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Vinod E Nambudiri
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
11
|
Cao F, Tang C, Chen X, Tu Z, Jin Y, Turk OM, Nishimura RN, Ebens A, Dubljevic V, Campbell JA, Zhou J, Hansen JE. Cathepsin B Nuclear Flux in a DNA-Guided "Antinuclear Missile" Cancer Therapy. ACS CENTRAL SCIENCE 2024; 10:1562-1572. [PMID: 39220699 PMCID: PMC11363321 DOI: 10.1021/acscentsci.4c00559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 09/04/2024]
Abstract
Some antinuclear antibodies (ANAs) bind extracellular nucleic acids released into tumor environments and are pulled into the nuclei of live cancer cells through nucleoside salvage pathways, independent of tumor-specific surface antigens. Here we show that ANA nuclear penetration induces nuclear flux by the lysosomal protease cathepsin B and leverage this mechanism to design an antinuclear antibody-drug conjugate (ANADC) with cathepsin B-labile drug linker. The ANADC targets nucleic acid exhaust from necrotic tumors and crosses membrane barriers through nucleoside salvage as a DNA-seeking and tumor agnostic "antinuclear missile" cancer therapy.
Collapse
Affiliation(s)
- Fei Cao
- Department
of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut 06510, United States
| | - Caroline Tang
- Department
of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut 06510, United States
| | - Xiaoyong Chen
- Department
of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut 06510, United States
| | - Zewei Tu
- Department
of Neurosurgery, Yale School of Medicine, New Haven, Connecticut 06510, United States
| | - Ying Jin
- Division
of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale School of Medicine, New Haven, Connecticut 06510, United States
| | - Olivia M. Turk
- Department
of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut 06510, United States
| | - Robert N. Nishimura
- Department
of Research & Development, Greater Los
Angeles Veterans Affairs Healthcare System, Los Angeles, California 90073, United States
- Department
of Neurology, David Geffen School of Medicine
at UCLA, Los Angeles, California 90095, United States
| | - Allen Ebens
- Adanate, Palo Alto, California 94305, United States
| | | | | | - Jiangbing Zhou
- Department
of Neurosurgery, Yale School of Medicine, New Haven, Connecticut 06510, United States
- Yale Cancer
Center, New Haven, Connecticut 06510, United States
| | - James E. Hansen
- Department
of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut 06510, United States
- Yale Cancer
Center, New Haven, Connecticut 06510, United States
| |
Collapse
|
12
|
Gray ME, Zielinski KM, Xu F, Elder KK, McKay SJ, Ojo VT, Benjamin SR, Yaseen AA, Brooks TA, Tumey LN. A comparison of the activity, lysosomal stability, and efficacy of legumain-cleavable and cathepsin-cleavable ADC linkers. Xenobiotica 2024; 54:458-468. [PMID: 38738708 PMCID: PMC11436314 DOI: 10.1080/00498254.2024.2352051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
Over the past two decades, antibody-drug conjugates (ADCs) have emerged as a highly effective drug delivery technology. ADCs utilise a monoclonal antibody, a chemical linker, and a therapeutic payload to selectively deliver highly potent pharmaceutical agents to specific cell types.Challenges such as premature linker cleavage and clearance due to linker hydrophobicity have adversely impacted the stability and safety of ADCs. While there are various solutions to these challenges, our team has focused on replacement of hydrophobic ValCit linkers (cleaved by CatB) with Asn-containing linkers that are cleaved by lysosomal legumain.Legumain is abundantly present in lysosomes and is known to play a role in tumour microenvironment dynamics. Herein, we directly compare the lysosomal cleavage, cytotoxicity, plasma stability, and efficacy of a traditional cathepsin-cleavable ADC to a matched Asn-containing legumain-cleavable ADC.We demonstrate that Asn-containing linker sequences are specifically cleaved by lysosomal legumain and that Asn-linked MMAE ADCs are broadly active against a variety of tumours, even those with low legumain expression. Finally, we show that AsnAsn-linked ADCs exhibit comparable or improved efficacy to traditional ValCit-linked ADCs. Our study paves the way for replacement of the traditional ValCit linker technology with more hydrophilic Asn-containing peptide linker sequences.
Collapse
Affiliation(s)
- Meghan E Gray
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Karina M Zielinski
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Fanny Xu
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Kayla K Elder
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Steven J McKay
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Victor T Ojo
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Samantha R Benjamin
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Aiman A Yaseen
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - Tracy A Brooks
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| | - L Nathan Tumey
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, NY, USA
| |
Collapse
|
13
|
Pang S, Duong A, Siu C, Indorf A. Antibody drug conjugates: Design implications for clinicians. J Oncol Pharm Pract 2024; 30:907-918. [PMID: 38651308 DOI: 10.1177/10781552241228827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
OBJECTIVE There are currently 11 antibody-drug conjugates (ADC) that are FDA approved for use in oncologic disease states, with many more in the pipeline. The authors aim to review the pharmacokinetic profiles of the components of ADCs to engage pharmacist practitioners in practical considerations in the care of patients. This article provides an overview on the use of ADCs in the setting of organ dysfunction, drug-drug interactions, and management of on- and off-target adverse effects. DATA SOURCES A systematic search of the literature on ADCs through September 2023 was conducted. Clinical trials as well as articles on ADC design and functional components, adverse effects, and pharmacokinetics were reviewed. Reviewed literature included prescribing information as well as tertiary sources and primary literature. DATA SUMMARY A total of 11 ADCs were reviewed for the purpose of this article. A description of the mechanism of action and structure of ADCs is outlined, and a table containing description of each currently FDA-approved ADC is included. Various mechanisms of ADC toxicity are reviewed, including how ADC structure may be implicated. CONCLUSION It is imperative that pharmacist clinicians understand the design and function of each component of an ADC to continue to assess new approvals for use in oncology patients. Understanding the design of the ADC can help a pharmacy practitioner compare and contrast adverse effect profiles to support their multidisciplinary teams and to engage patients in education and management of their care.
Collapse
Affiliation(s)
- Stephanie Pang
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Arianne Duong
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Chloe Siu
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| | - Amy Indorf
- Department of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Chis AA, Dobrea CM, Arseniu AM, Frum A, Rus LL, Cormos G, Georgescu C, Morgovan C, Butuca A, Gligor FG, Vonica-Tincu AL. Antibody-Drug Conjugates-Evolution and Perspectives. Int J Mol Sci 2024; 25:6969. [PMID: 39000079 PMCID: PMC11241239 DOI: 10.3390/ijms25136969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Antineoplastic therapy is one of the main research themes of this century. Modern approaches have been implemented to target and heighten the effect of cytostatic drugs on tumors and diminish their general/unspecific toxicity. In this context, antibody-drug conjugates (ADCs) represent a promising and successful strategy. The aim of this review was to assess different aspects regarding ADCs. They were presented from a chemical and a pharmacological perspective and aspects like structure, conjugation and development particularities alongside effects, clinical trials, safety issues and perspectives and challenges for future use of these drugs were discussed. Representative examples include but are not limited to the following main structural components of ADCs: monoclonal antibodies (trastuzumab, brentuximab), linkers (pH-sensitive, reduction-sensitive, peptide-based, phosphate-based, and others), and payloads (doxorubicin, emtansine, ravtansine, calicheamicin). Regarding pharmacotherapy success, the high effectiveness expectation associated with ADC treatment is supported by the large number of ongoing clinical trials. Major aspects such as development strategies are first discussed, advantages and disadvantages, safety and efficacy, offering a retrospective insight on the subject. The second part of the review is prospective, focusing on various plans to overcome the previously identified difficulties.
Collapse
Affiliation(s)
| | | | - Anca Maria Arseniu
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Adina Frum
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Luca-Liviu Rus
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Gabriela Cormos
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Cecilia Georgescu
- Faculty of Agriculture Science, Food Industry and Environmental Protection, "Lucian Blaga" University of Sibiu, 550012 Sibiu, Romania
| | - Claudiu Morgovan
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | - Anca Butuca
- Faculty of Medicine, "Lucian Blaga" University of Sibiu, 550169 Sibiu, Romania
| | | | | |
Collapse
|
15
|
Wang Y, Kilic O, Rozumalski L, Distefano MD, Wagner CR. Targeted Drug Delivery by MMAE Farnesyl-Bioconjugated Multivalent Chemically Self-Assembled Nanorings Induces Potent Receptor-Dependent Immunogenic Cell Death. Bioconjug Chem 2024; 35:582-592. [PMID: 38701361 PMCID: PMC11633779 DOI: 10.1021/acs.bioconjchem.4c00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Antibody-drug conjugates, nanoparticles, and liposomes have been used for anticancer drug delivery. The success of targeted killing of cancer cells relies heavily on the selectivity of the drug delivery systems. In most systems, antibodies or their fragments were used as targeting ligands. In this study, we have investigated the potential for protein-based octomeric chemically self-assembled nanorings (CSANs) to be used for anticancer drug delivery. The CSANs are composed of a DHFR-DHFR fusion protein incorporating an EGFR-targeting fibronectin and the anticancer drug MMAE conjugated through a C-terminal farnesyl azide. The anti-EGFR-MMAE CSANs were shown to undergo rapid internalization and have potent cytotoxicity to cancer cells across a 9000-fold difference in EGFR expression. In addition, anti-EGFR-MMAE CSANs were shown to induce immunological cell death. Thus, multivalent and modular CSANs are a potential alternative anticancer drug delivery platform with the capability of targeting tumor cells with heterogeneous antigen expression while activating the anticancer immune response.
Collapse
Affiliation(s)
- Yiao Wang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Ozgun Kilic
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Lakmal Rozumalski
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Mark D. Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Carston R. Wagner
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
16
|
Uchida S, Serada S, Suzuki Y, Funajima E, Kitakami K, Dobashi K, Tamatani S, Sato Y, Beppu T, Ogasawara K, Naka T. Glypican-1-targeted antibody-drug conjugate inhibits the growth of glypican-1-positive glioblastoma. Neoplasia 2024; 50:100982. [PMID: 38417223 PMCID: PMC10915784 DOI: 10.1016/j.neo.2024.100982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/21/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
Glioblastoma is the deadliest form of brain tumor. The presence of the blood-brain barrier (BBB) significantly hinders chemotherapy, necessitating the development of innovative treatment options for this tumor. This report presents the in vitro and in vivo efficacy of an antibody-drug conjugate (ADC) that targets glypican-1 (GPC1) in glioblastoma. The GPC1-ADC was created by conjugating a humanized anti-GPC1 antibody (clone T2) with monomethyl auristatin E (MMAE) via maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl linkers. Immunohistochemical staining analysis of a glioblastoma tissue microarray revealed that GPC1 expression was elevated in more than half of the cases. GPC1-ADC, when bound to GPC1, was efficiently and rapidly internalized in glioblastoma cell lines. It inhibited the growth of GPC1-positive glioma cell lines by inducing cell cycle arrest in the G2/M phase and triggering apoptosis in vitro. We established a heterotopic xenograft model by subcutaneously implanting KALS-1 and administered GPC1-ADC intravenously. GPC1-ADC significantly inhibited tumor growth and increased the number of mitotic cells. We also established an orthotopic xenograft model by intracranially implanting luciferase-transfected KS-1-Luc#19. After injecting Evans blue and resecting brain tissues, dye leakage was observed in the implantation area, confirming BBB disruption. We administered GPC1-ADC intravenously and measured the luciferase activity using an in vivo imaging system. GPC1-ADC significantly inhibited tumor growth and extended survival. In conclusion, GPC1-ADC demonstrated potent intracranial activity against GPC1-positive glioblastoma in an orthotopic xenograft model. These results indicate that GPC1-ADC could represent a groundbreaking new therapy for treating glioblastoma beyond the BBB.
Collapse
Affiliation(s)
- Shun Uchida
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Satoshi Serada
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan.
| | - Yuji Suzuki
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Eiji Funajima
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kei Kitakami
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | - Kazumasa Dobashi
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan
| | | | - Yuichi Sato
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Takaaki Beppu
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Kuniaki Ogasawara
- Department of Neurosurgery, School of Medicine Iwate Medical University, Yahaba, Japan
| | - Testuji Naka
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, School of Medicine Iwate Medical University, Yahaba, Japan.
| |
Collapse
|
17
|
Liu K, Li M, Li Y, Li Y, Chen Z, Tang Y, Yang M, Deng G, Liu H. A review of the clinical efficacy of FDA-approved antibody‒drug conjugates in human cancers. Mol Cancer 2024; 23:62. [PMID: 38519953 PMCID: PMC10960395 DOI: 10.1186/s12943-024-01963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/21/2024] [Indexed: 03/25/2024] Open
Abstract
While strategies such as chemotherapy and immunotherapy have become the first-line standard therapies for patients with advanced or metastatic cancer, acquired resistance is still inevitable in most cases. The introduction of antibody‒drug conjugates (ADCs) provides a novel alternative. ADCs are a new class of anticancer drugs comprising the coupling of antitumor mAbs with cytotoxic drugs. Compared with chemotherapeutic drugs, ADCs have the advantages of good tolerance, accurate target recognition, and small effects on noncancerous cells. ADCs occupy an increasingly important position in the therapeutic field. Currently, there are 13 Food and Drug Administration (FDA)‒approved ADCs and more than 100 ADC drugs at different stages of clinical trials. This review briefly describes the efficacy and safety of FDA-approved ADCs, and discusses the related problems and challenges to provide a reference for clinical work.
Collapse
Affiliation(s)
- Kaifeng Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meijia Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yudong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yutong Li
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Zixin Chen
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yiqi Tang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Meitian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Guoquan Deng
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China
| | - Hongwei Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, China.
| |
Collapse
|
18
|
Zhang M, Zuo Y, Chen S, Li Y, Xing Y, Yang L, Wang H, Guo R. Antibody-drug conjugates in urothelial carcinoma: scientometric analysis and clinical trials analysis. Front Oncol 2024; 14:1323366. [PMID: 38665947 PMCID: PMC11044263 DOI: 10.3389/fonc.2024.1323366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/12/2024] [Indexed: 04/28/2024] Open
Abstract
In 2020, bladder cancer, which commonly presents as urothelial carcinoma, became the 10th most common malignancy. For patients with metastatic urothelial carcinoma, the standard first-line treatment remains platinum-based chemotherapy, with immunotherapy serving as an alternative in cases of programmed death ligand 1 expression. However, treatment options become limited upon resistance to platinum and programmed death 1 or programmed death ligand 1 agents. Since the FDA's approval of Enfortumab Vedotin and Sacituzumab Govitecan, the therapeutic landscape has expanded, heralding a shift towards antibody-drug conjugates as potential first-line therapies. Our review employed a robust scientometric approach to assess 475 publications on antibody-drug conjugates in urothelial carcinoma, revealing a surge in related studies since 2018, predominantly led by U.S. institutions. Moreover, 89 clinical trials were examined, with 36 in Phase II and 13 in Phase III, exploring antibody-drug conjugates as both monotherapies and in combination with other agents. Promisingly, novel targets like HER-2 and EpCAM exhibit substantial therapeutic potential. These findings affirm the increasing significance of antibody-drug conjugates in urothelial carcinoma treatment, transitioning them from posterior-line to frontline therapies. Future research is poised to focus on new therapeutic targets, combination therapy optimization, treatment personalization, exploration of double antibody-coupled drugs, and strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| | - Yuanye Zuo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Siyi Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yaonan Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Xing
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Department of Clinical Laboratory, First Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Tsuchikama K, Anami Y, Ha SYY, Yamazaki CM. Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol 2024; 21:203-223. [PMID: 38191923 DOI: 10.1038/s41571-023-00850-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Summer Y Y Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chisato M Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Makiyama J, Ishitsuka K, Munakata W, Maruyama D, Nagai H. An update on the developments in the treatment of adult T-cell leukemia-lymphoma: current knowledge and future perspective. Jpn J Clin Oncol 2023; 53:1104-1111. [PMID: 37592900 DOI: 10.1093/jjco/hyad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/05/2023] [Indexed: 08/19/2023] Open
Abstract
Adult T-cell leukemia-lymphoma is defined as peripheral T-cell lymphoma caused by the human T-cell leukemia virus type I. Adult T-cell leukemia-lymphoma is classified into indolent (favorable chronic or smoldering) or aggressive (acute, lymphoma or unfavorable chronic) types. This review discusses the therapeutic developments for patients with adult T-cell leukemia-lymphoma and unmet issues in treating adult T-cell leukemia-lymphoma. For indolent adult T-cell leukemia-lymphoma, a watchful waiting strategy is recommended until the disease progresses to aggressive adult T-cell leukemia-lymphoma. For aggressive adult T-cell leukemia-lymphoma, multi-agent chemotherapy with or without allogeneic hematopoietic stem cell transplantation has been recommended. However, many patients with adult T-cell leukemia-lymphoma relapse, and their prognosis is poor. Recently, novel agents, including mogamulizumab, lenalidomide, brentuximab vedotin, tucidinostat and valemetostat, have been approved for patients with relapsed or refractory aggressive adult T-cell leukemia-lymphoma, and the combination of mogamulizumab with multi-agent chemotherapy or brentuximab vedotin with cyclophosphamide, doxorubicin and prednisone has been approved for patients with untreated aggressive adult T-cell leukemia-lymphoma in Japan. Importantly, the aging of patients with adult T-cell leukemia-lymphoma has recently been reported, and no standard of care for elderly patients with adult T-cell leukemia-lymphoma has been established. New evidence must be obtained from prospective clinical trials to improve the prognosis of patients with adult T-cell leukemia-lymphoma.
Collapse
Affiliation(s)
- Junya Makiyama
- Department of Hematology, Sasebo City General Hospital, Sasebo, Japan
| | - Kenji Ishitsuka
- Department of Hematology and Rheumatology, Kagoshima University, Kagoshima, Japan
| | - Wataru Munakata
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Maruyama
- Department of Hematology Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hirokazu Nagai
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
21
|
Gulyak EL, Alferova VA, Korshun VA, Sapozhnikova KA. Introduction of Carbonyl Groups into Antibodies. Molecules 2023; 28:7890. [PMID: 38067618 PMCID: PMC10707781 DOI: 10.3390/molecules28237890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Antibodies and their derivatives (scFv, Fabs, etc.) represent a unique class of biomolecules that combine selectivity with the ability to target drug delivery. Currently, one of the most promising endeavors in this field is the development of molecular diagnostic tools and antibody-based therapeutic agents, including antibody-drug conjugates (ADCs). To meet this challenge, it is imperative to advance methods for modifying antibodies. A particularly promising strategy involves the introduction of carbonyl groups into the antibody that are amenable to further modification by biorthogonal reactions, namely aliphatic, aromatic, and α-oxo aldehydes, as well as aliphatic and aryl-alkyl ketones. In this review, we summarize the preparation methods and applications of site-specific antibody conjugates that are synthesized using this approach.
Collapse
Affiliation(s)
| | | | | | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (E.L.G.); (V.A.A.); (V.A.K.)
| |
Collapse
|
22
|
Rodrigues WF, Miguel CB, de Abreu MCM, Neto JM, Oliveira CJF. Potential Associations between Vascular Biology and Hodgkin's Lymphoma: An Overview. Cancers (Basel) 2023; 15:5299. [PMID: 37958472 PMCID: PMC10649902 DOI: 10.3390/cancers15215299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Hodgkin's lymphoma (HL) is a lymphatic neoplasm typically found in the cervical lymph nodes. The disease is multifactorial, and in recent years, the relationships between various vascular molecules have been explored in the field of vascular biology. The connection between vascular biology and HL is intricate and the roles of several pathways remain unclear. This review summarizes the cellular and molecular relationships between vascular biology and HL. Proteins associated with various functions in vascular biology, including cytokines (TNF-α, IL-1, IL-13, and IL-21), chemokines (CXCL10, CXCL12, and CCL21), adhesion molecules (ELAM-1/VCAM-1), and growth factors (BDNF/NT-3, platelet-derived growth factor receptor-α), have been linked to tumor activity. Notable tumor activities include the induction of paracrine activation of NF-kB-dependent pathways, upregulation of adhesion molecule regulation, genome amplification, and effective loss of antigen presentation mediated by MHC-II. Preclinical study models, primarily those using cell culture, have been optimized for HL. Animal models, particularly mice, are also used as alternatives to complex biological systems, with studies primarily focusing on the physiopathogenic evaluation of the disease. These biomolecules warrant further study because they may shed light on obscure pathways and serve as targets for prevention and/or treatment interventions.
Collapse
Affiliation(s)
- Wellington Francisco Rodrigues
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | - Camila Botelho Miguel
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | | | - Jamil Miguel Neto
- University Center of Mineiros, Unifimes, Mineiros 75833-130, GO, Brazil; (M.C.M.d.A.); (J.M.N.)
| | - Carlo José Freire Oliveira
- Postgraduate Course in Tropical Medicine and Infectious Diseases, Federal University of Triangulo Mineiro, UFTM, Uberaba 38025-440, MG, Brazil; (C.B.M.); (C.J.F.O.)
| |
Collapse
|
23
|
Riccardi F, Dal Bo M, Macor P, Toffoli G. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Front Pharmacol 2023; 14:1274088. [PMID: 37790810 PMCID: PMC10544916 DOI: 10.3389/fphar.2023.1274088] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-cancer compounds that are widely used in the treatment of hematologic malignancies and solid tumors. Unlike conventional chemotherapeutic drug-based therapies, that are mainly associated with modest specificity and therapeutic benefit, the three key components that form an ADC (a monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety) achieve remarkable improvement in terms of targeted killing of cancer cells and, while sparing healthy tissues, a reduction in systemic side effects caused by off-tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been approved to date by the market approval by the Food and Drug Administration (FDA), the European Medicines Agency (EMA) and/or other international governmental agencies for use in clinical oncology, and hundreds are undergoing evaluation in the preclinical and clinical phases. Here, our aim is to provide a comprehensive overview of the key features revolving around ADC therapeutic strategy including their structural and targeting properties, mechanism of action, the role of the tumor microenvironment and review the approved ADCs in clinical oncology, providing discussion regarding their toxicity profile, clinical manifestations and use in novel combination therapies. Finally, we briefly review ADCs in other pathological contexts and provide key information regarding ADC manufacturing and analytical characterization.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
24
|
Rivera-Pérez C, Ponce González XP, Hernández-Savedra NY. Antimicrobial and anticarcinogenic activity of bioactive peptides derived from abalone viscera (Haliotis fulgens and Haliotis corrugata). Sci Rep 2023; 13:15185. [PMID: 37704667 PMCID: PMC10499822 DOI: 10.1038/s41598-023-41491-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023] Open
Abstract
Bioactive peptides have been studied in several sources due to their valuable potential in the pharmaceutical and food industries. Abalone viscera, which are normally discarded as byproducts, are a rich source of protein. Thus, the aim of this study was to explore the potential bioactivity of peptides derived from abalone viscera (Haliotis fulgens and Haliotis corrugata) after hydrolysis with a commercial mixture of enzymes. The hydrolysates obtained were fractionated using gel filtration chromatography. The resulting hydrolysate fractions were investigated for their antimicrobial and cytotoxic activities, including the expression of gelatinases mmp-2 and mmp-9 in human prostate cancer cell lines (PC3). Results showed antimicrobial activity for protein fractions of H. corrugata against Proteus mirabilis and Pseudomona aeuroginosa (66.2-116.25 kDa), Bacillus subtilis (6.5-21.5 kDa), and Aspergillus niger (97.4-116.25 kDa), while H. fulgens peptide fractions (200-31 kDa) displayed activity against six bacterial strains, and fractions from 116.25 to 21.5 kDa had effects on the fungus A. niger, Alternaria alternata, and Aspergillus flavus. Additionally, protein fractions displayed cytotoxic activity, inhibiting 30.4-53.8% of PC3 cellular growth. Selected fractions decreased the PMA-induced and not-induced expressions of mmp-2 and mmp-9 in PC3 cells. Abalone viscera, as byproducts, can be used as a potential source of antimicrobial and anticancer peptides.
Collapse
Affiliation(s)
- Crisalejandra Rivera-Pérez
- Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, C.P. 23096, La Paz, BCS, México
| | - Xolotl Paloma Ponce González
- Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, C.P. 23096, La Paz, BCS, México
| | - Norma Yolanda Hernández-Savedra
- Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, C.P. 23096, La Paz, BCS, México.
| |
Collapse
|
25
|
Hansen RA, Märcher A, Pedersen KN, Gothelf KV. Insertion of Chemical Handles into the Backbone of DNA during Solid-Phase Synthesis by Oxidative Coupling of Amines to Phosphites. Angew Chem Int Ed Engl 2023; 62:e202305373. [PMID: 37119479 DOI: 10.1002/anie.202305373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/01/2023]
Abstract
Conjugation of molecules or proteins to oligonucleotides can improve their functional and therapeutic capacity. However, such modifications are often limited to the 5' and 3' end of oligonucleotides. Herein, we report the development of an inexpensive and simple method that allows for the insertion of chemical handles into the backbone of oligonucleotides. This method is compatible with standardized automated solid-phase oligonucleotide synthesis, and relies on formation of phosphoramidates. A unique phosphoramidite is incorporated into a growing oligonucleotide, and oxidized to the desired phosphoramidate using iodine and an amine of choice. Azides, alkynes, amines, and alkanes have been linked to oligonucleotides via internally positioned phosphoramidates with oxidative coupling yields above 80 %. We show the design of phosphoramidates from secondary amines that specifically hydrolyze to the phosphate only at decreased pH. Finally, we show the synthesis of an antibody-DNA conjugate, where the oligonucleotide can be selectively released in a pH 5.5 buffer.
Collapse
Affiliation(s)
- Rikke A Hansen
- sDepartment of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Anders Märcher
- sDepartment of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Kristian Nørgaard Pedersen
- sDepartment of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Kurt V Gothelf
- sDepartment of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| |
Collapse
|
26
|
Lordick F, Merz M, Büch E, Aigner A. Antibody-Drug Conjugates as a Targeted Therapeutic Approach Across Entities in Oncology. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:329-336. [PMID: 37073718 PMCID: PMC10408283 DOI: 10.3238/arztebl.m2023.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/06/2022] [Accepted: 04/03/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND Cancer is no longer treated on the basis of its histological lineage alone; more and more drugs are being developed that are directed toward specific molecular and immunological features. Monoclonal antibodies are one type of selectively acting therapeutic agent. As part of this development, antibody-drug conjugates ("ADCs") have been approved in recent years for the treatment of hematologic and solid malignancies. METHODS This review is based on pertinent articles retrieved by a selective search in PubMed, as well as on papers presented at international congresses of specialist societies such as the European Society for Medical Oncology, the American Society of Clinical Oncology, and the American Association for Cancer Research, and information published on the websites of the European Medicines Agency, the Food and Drug Administration, and the German Joint Federal Committee. RESULTS The efficacy of the nine ADCs currently approved in the European Union (as of 12/2022) is derived from technical improvements in the conjugation process, the introduction of novel linkers for the covalent binding of cytotoxic agents to the Fc portion of the antibody, and the development of new, potent cytotoxic agents. Compared to conventional cancer therapies, the approved ADCs improve treatment outcomes with respect to tumor remission, time to tumor progression and, in some cases, overall survival by specifically channeling cytotoxic agents into the malignant target cells and thereby limiting, at least to some extent, the exposure of healthy tissue to adverse effects. Various potential side effects still require attention, including venous occlusive disease, pneumonitis, ocular keratopathy, and skin rash. The development of effective ADCs requires the identification of tumor-selective targets to which ADCs can bind. CONCLUSION ADCs are a novel category of drugs for the treatment of cancer. Their approval is mainly, but not exclusively, based on the favorable findings of randomized, controlled phase III trials. ADCs are already helping to improve the outcomes of treatment for cancer.
Collapse
Affiliation(s)
- Florian Lordick
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, University of Leipzig Medical Center, Comprehensive Cancer Center Central Germany, Leipzig, Germany
| | - Maximilian Merz
- Department of Hematology, Cell therapy und Hemostaseology, University of Leipzig Medical Center, Comprehensive Cancer Center Central Germany, Leipzig, Germany
| | - Eva Büch
- Department of Oncology, Gastroenterology, Hepatology, Pulmonology, University of Leipzig Medical Center, Comprehensive Cancer Center Central Germany, Leipzig, Germany
| | - Achim Aigner
- Rudolf Boehm Institute of Pharmacology and Toxicology, Independent Department of Clinical Pharmacology, Medical Faculty, University of Leipzig, Comprehensive Cancer Center Central Germany, Leipzig, Germany
| |
Collapse
|
27
|
Köksalan D, Sözen M, Selek A, Gezer E, Cantürk Z, Çetinarslan B. Brentuximab vedotin-associated diabetic ketoacidosis: a case report. Int J Diabetes Dev Ctries 2023; 43:120-124. [PMID: 35875342 PMCID: PMC9287688 DOI: 10.1007/s13410-022-01116-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/29/2022] [Indexed: 11/08/2022] Open
Abstract
Background Diabetic ketoacidosis (DKA) is a life-threatening complication of diabetes mellitus (DM). It is characterized by hyperglycemia, metabolic acidosis, and ketonemia. Fortunately, drug-induced hyperglycemias are usually mild and not life-threatening. However, rarely some cases may present with ketoacidosis. In this case report, we aimed to present a brentuximab vedotin (BV) associated with DKA. Case presentation A 23-year-old Caucasian man presented with abdominal pain, nausea, and vomiting for 1-2 weeks. The patient had a previous diagnosis of Hodgkin's lymphoma and primer hypothyroidism. He is using levothyroxine 150 μg per day and received BV treatment for Hodgkin lymphoma (HL) 10 days ago. No steroid treatment was administered for premedication before BV. Except for obesity, all system examinations are normal. There were no signs of any infection. Laboratory data revealed hyperglycemia, metabolic acidosis, and ketonemia. The patient was admitted to the service with a diagnosis of DKA. After the patient was admitted to our clinic, insulin treatment and hydration started immediately. Despite the insulin infusion reaching 1700 units per day, the patient's diabetic ketoacidosis extended to 1 week. Anti-insulin, anti-glutamic acid decarboxylase, and islet cell autoantibodies were negative, which were checked to exclude type 1 DM. Fasting C-peptide was 28 ng/mL (normal range, 0.9-7.1 ng/mL). With all these, the diabetic ketoacidosis status of the patient was evaluated as a BV side effect. Conclusion This patient is a rare case of BV-associated DKA. It is very important to know this relationship since BV treatment has turned into a standard treatment for relapsed Hodgkin lymphoma. Our case highlights that this diagnosis should be kept in mind as a complication of each dose of BV administration.
Collapse
Affiliation(s)
- Damla Köksalan
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| | - Mehmet Sözen
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| | - Alev Selek
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| | - Emre Gezer
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| | - Zeynep Cantürk
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| | - Berrin Çetinarslan
- Department of Endocrinology and Metabolism, Kocaeli University Faculty of Medicine, 41000 Kocaeli, Turkey
| |
Collapse
|
28
|
Cieslak C, Mitteldorf C, Krömer-Olbrisch T, Kempf W, Stadler R. QuPath Analysis for CD30+ Cutaneous T-Cell Lymphoma. Am J Dermatopathol 2023; 45:93-98. [PMID: 36669072 DOI: 10.1097/dad.0000000000002330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/10/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mycosis fungoides is the most common subtype of cutaneous T-cell lymphoma, in which the expression of cluster of differentiation 30 (CD30)+ subtype can now be treated with the CD30 antibody conjugate brentuximab vedotin. Diagnostic methods are based on immunohistochemical (IHC) staining followed by manual assessment by pathologists, which is always a subjective calculation. QuPath, an open-source software for digital pathology image analysis, satisfies the requirements of objective approaches. METHODS Ten samples from mycosis fungoides patients with CD30 expression at different stages were stained for CD3 and CD30 by IHC staining, scanned, and quantitative analysis was performed using QuPath (version 2.1). Each slide was independently assessed by 3 board-certified dermatopathologists. RESULTS Individual estimates for CD30+/CD3+ cells varied among the individual histopathologists (mean coefficient of variation, 0.46; range, 0-0.78). QuPath analysis showed excellent separation between the positively stained cells for CD3 and CD30 IHC and other cells and tissue structures, although the results correlated strongly with the respective mean estimates of the 3 histopathologists (Pearson-R 0.93). CONCLUSIONS The results show a high interobserver variability evaluation of IHC markers, although quantitative image analysis offer a significant advantage for comparison. This is not only relevant for clinical routine but also especially critical in therapeutic studies addressing targeted molecules.
Collapse
Affiliation(s)
- Cassandra Cieslak
- University Clinic for Dermatology, Johannes Wesling Medical Centre, Minden, Germany
- University Hospital of Ruhr-University, Bochum, Germany
| | - Christina Mitteldorf
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany; and
| | - Tanja Krömer-Olbrisch
- University Clinic for Dermatology, Johannes Wesling Medical Centre, Minden, Germany
- University Hospital of Ruhr-University, Bochum, Germany
| | - Werner Kempf
- Kempf und Pfaltz Histologische Diagnostik, Zurich, Switzerland
| | - Rudolf Stadler
- University Clinic for Dermatology, Johannes Wesling Medical Centre, Minden, Germany
- University Hospital of Ruhr-University, Bochum, Germany
| |
Collapse
|
29
|
Sun Y, Sha Y, Cui G, Meng F, Zhong Z. Lysosomal-mediated drug release and activation for cancer therapy and immunotherapy. Adv Drug Deliv Rev 2023; 192:114624. [PMID: 36435229 DOI: 10.1016/j.addr.2022.114624] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/10/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
The development of carrier systems that are able to transport and release therapeutics to target cells is an emergent strategy to treat cancer; however, they following endocytosis are usually trapped in the endo/lysosomal compartments. The efficacy of drug conjugates and nanotherapeutics relies critically on their intracellular drug release ability, for which advanced systems responding to the unique lysosomal environment such as acidic pH and abundant enzymes (e.g. cathepsin B, sulfatase and β-glucuronidase) or equipped with photochemical internalization property have been energetically pursued. In this review, we highlight the recent designs of smart systems that promote efficient lysosomal release and/or escape of anticancer agents including chemotherapeutics (e.g. doxorubicin, platinum, chloroquine and hydrochloroquine) and biotherapeutics (e.g. proteins, siRNA, miRNA, mRNA and pDNA) to cancer cells or immunotherapeutic agents (e.g. antigens, mRNA and immunoadjuvants) to antigen-presenting cells (APCs), thereby boosting cancer therapy and immunotherapy. Lysosomal-mediated drug release presents an appealing approach to develop innovative cancer therapeutics and immunotherapeutics.
Collapse
Affiliation(s)
- Yinping Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, PR China
| | - Yongjie Sha
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, PR China
| | - Guanhong Cui
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, PR China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
30
|
Busato D, Mossenta M, Dal Bo M, Macor P, Toffoli G. The Proteoglycan Glypican-1 as a Possible Candidate for Innovative Targeted Therapeutic Strategies for Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2022; 23:ijms231810279. [PMID: 36142190 PMCID: PMC9499405 DOI: 10.3390/ijms231810279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/04/2022] [Indexed: 11/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of all pancreatic cancers, with a 5-year survival rate of 7% and 80% of patients diagnosed with advanced or metastatic malignancies. Despite recent advances in diagnostic testing, surgical techniques, and systemic therapies, there remain limited options for the effective treatment of PDAC. There is an urgent need to develop targeted therapies that are able to differentiate between cancerous and non-cancerous cells to reduce side effects and better inhibit tumor growth. Antibody-targeted strategies are a potentially effective option for introducing innovative therapies. Antibody-based immunotherapies and antibody-conjugated nanoparticle-based targeted therapies with antibodies targeting specific tumor-associated antigens (TAA) can be proposed. In this context, glypican-1 (GPC1), which is highly expressed in PDAC and not expressed or expressed at very low levels in non-malignant lesions and healthy pancreatic tissues, is a useful TAA that can be achieved by a specific antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy. In this review, we describe the main clinical features of PDAC. We propose the proteoglycan GPC1 as a useful TAA for PDAC-targeted therapies. We also provide a digression on the main developed approaches of antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy, which can be used to target GPC1.
Collapse
Affiliation(s)
- Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Correspondence: ; Tel.: +39-0434-659816
| | - Monica Mossenta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| |
Collapse
|
31
|
Ha SYY, Anami Y, Yamazaki CM, Xiong W, Haase CM, Olson SD, Lee J, Ueno NT, Zhang N, An Z, Tsuchikama K. An Enzymatically Cleavable Tripeptide Linker for Maximizing the Therapeutic Index of Antibody-Drug Conjugates. Mol Cancer Ther 2022; 21:1449-1461. [PMID: 35793453 PMCID: PMC9452487 DOI: 10.1158/1535-7163.mct-22-0362] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022]
Abstract
Valine-citrulline is a protease-cleavable linker commonly used in many drug delivery systems, including antibody-drug conjugates (ADC) for cancer therapy. However, its suboptimal in vivo stability can cause various adverse effects such as neutropenia and hepatotoxicity, leading to dose delays or treatment discontinuation. Here, we report that glutamic acid-glycine-citrulline (EGCit) linkers have the potential to solve this clinical issue without compromising the ability of traceless drug release and ADC therapeutic efficacy. We demonstrate that our EGCit ADC resists neutrophil protease-mediated degradation and spares differentiating human neutrophils. Notably, our anti-HER2 ADC shows almost no sign of blood and liver toxicity in healthy mice at 80 mg kg-1. In contrast, at the same dose level, the FDA-approved anti-HER2 ADCs Kadcyla and Enhertu show increased levels of serum alanine aminotransferase and aspartate aminotransferase and morphologic changes in liver tissues. Our EGCit conjugates also exert greater antitumor efficacy in multiple xenograft tumor models compared with Kadcyla and Enhertu. This linker technology could substantially broaden the therapeutic windows of ADCs and other drug delivery agents, providing clinical options with improved efficacy and safety.
Collapse
Affiliation(s)
- Summer Y. Y. Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Chisato M. Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Wei Xiong
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Candice M. Haase
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Scott D. Olson
- Department of Pediatric Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| | - Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, 1881 East Rd., Houston, TX 77054, USA
| |
Collapse
|
32
|
Khatib SE, Salla M. The mosaic puzzle of the therapeutic monoclonal antibodies and antibody fragments - A modular transition from full-length immunoglobulins to antibody mimetics. Leuk Res Rep 2022; 18:100335. [PMID: 35832747 PMCID: PMC9272380 DOI: 10.1016/j.lrr.2022.100335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 01/07/2023] Open
Abstract
The use of monoclonal antibodies represents an important and efficient diagnostic and therapeutic tool in disease management and modern science but remains limited by several factors including the uneven distribution in diseased tissues as well as undesired activation of side immune reactions. Major scientific advancements including Recombinant DNA Technology, Hybridoma Technology, and Polymerase Chain Reaction have considerably impacted the use of monoclonal antibodies providing technical and effective solutions to overcome the shortcomings encountered with conventional antibodies. Initially, the introduction of antibody fragments allowed a more uniform and deeper penetration of the targeted tissue and reduced unwanted activation of Fc-mediated immune reactions. On another level, the immunogenicity of murine-derived antibodies was overcome by humanizing their encoding genes with specific sequences of human origin andtransgenic mice able to synthesize fully human antibodies were successfully created. Moreover, the advancement of genetic engineering techniques supported by the modular structure of antibody coding genes paved the way for the development of a new generation of antibody fragments with a wide spectrum of monospecific and bispecific agents. These later could be monovalent, bivalent, or multivalent, and either expressed as a single chain, assembled in multimeric forms or stringed in tandem. This has conferred improved affinity, stability, and solubility to antibody targetting. Lately, a new array of monoclonal antibody fragments was introduced with the engineering of nanobody and antibody mimetics as non-immunoglobulin-derived fragments with promising diagnostic and therapeutic applications. In this review, we decipher the molecular basis of monoclonal antibody engineering with a detailed screening of the antibody derivatives that provides new perspectives to expand the use of monoclonal fragments into previously unexplored fields.
Collapse
Affiliation(s)
- Sami El Khatib
- Lebanese International University, Department of Biomedical Sciences, Bekaa Campus, Khiyara, West Bekaa, Lebanon
| | - Mohamed Salla
- University of Alberta. Biochemistry Department, Faculty of Medicine and Dentistry,116St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
33
|
Debnath U, Verma S, Patra J, Mandal SK. A review on recent synthetic routes and computational approaches for antibody drug conjugation developments used in anti-cancer therapy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Chia CSB. A Patent Review on FDA-Approved Antibody-Drug Conjugates, Their Linkers and Drug Payloads. ChemMedChem 2022; 17:e202200032. [PMID: 35384350 DOI: 10.1002/cmdc.202200032] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/22/2022] [Indexed: 12/30/2022]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising class of biologics since the first approval of Gemtuzumab ozogamicin in 2000. Compared to small molecule drugs, ADCs are structurally much more complex as they comprise of an antibody conjugated to cytotoxic payloads by specially-designed linkers. Correspondingly, the ADC patent landscape is also much more complex. This review collates and discusses the patents protecting ADCs approved by the FDA up to 31 December 2021, with particular emphasis on their linker and cytotoxin payload technologies.
Collapse
Affiliation(s)
- C S Brian Chia
- Experimental Drug Development Centre, 10 Biopolis Road, Chromos #08-01, 138670, Singapore, Singapore
| |
Collapse
|
35
|
Fu Z, Li S, Han S, Shi C, Zhang Y. Antibody drug conjugate: the "biological missile" for targeted cancer therapy. Signal Transduct Target Ther 2022; 7:93. [PMID: 35318309 PMCID: PMC8941077 DOI: 10.1038/s41392-022-00947-7] [Citation(s) in RCA: 676] [Impact Index Per Article: 225.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
Antibody-drug conjugate (ADC) is typically composed of a monoclonal antibody (mAbs) covalently attached to a cytotoxic drug via a chemical linker. It combines both the advantages of highly specific targeting ability and highly potent killing effect to achieve accurate and efficient elimination of cancer cells, which has become one of the hotspots for the research and development of anticancer drugs. Since the first ADC, Mylotarg® (gemtuzumab ozogamicin), was approved in 2000 by the US Food and Drug Administration (FDA), there have been 14 ADCs received market approval so far worldwide. Moreover, over 100 ADC candidates have been investigated in clinical stages at present. This kind of new anti-cancer drugs, known as "biological missiles", is leading a new era of targeted cancer therapy. Herein, we conducted a review of the history and general mechanism of action of ADCs, and then briefly discussed the molecular aspects of key components of ADCs and the mechanisms by which these key factors influence the activities of ADCs. Moreover, we also reviewed the approved ADCs and other promising candidates in phase-3 clinical trials and discuss the current challenges and future perspectives for the development of next generations, which provide insights for the research and development of novel cancer therapeutics using ADCs.
Collapse
Affiliation(s)
- Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Shijun Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, (Parkville Campus) 381 Royal Parade,, Parkville, VIC, 3052, Australia
- Faculty of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, People's Republic of China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
36
|
Xu J, Li X, Du Y. Antibody-Pattern Recognition Receptor Agonist Conjugates: A Promising Therapeutic Strategy for Cancer. Adv Biol (Weinh) 2022; 6:e2101065. [PMID: 35122418 DOI: 10.1002/adbi.202101065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/27/2021] [Indexed: 01/15/2023]
Abstract
Antibody-drug conjugates (ADCs) are composed of monoclonal antibodies linked to cytotoxic payload drugs, each of which can be diversely designed in accordance with pharmacological and clinical requirements. The use of ADCs is effective for the treatment of different diseases, including cancers, and is gaining widespread attention. To date, 12 ADCs have been approved by the U.S. Food and Drug Administration for treating cancer and improving the quality of life of patients. To expand the application of ADCs and improve their treatment efficiency, various formats have recently been manufactured, including pattern recognition receptor (PRR) agonist-based ADCs. The antibody has a unique structure that enables the specific delivery of PRR agonists to the tumor area, and this improves the therapeutic efficacy while minimizing systemic toxicity. This review briefly discusses the current landscape and future perspectives of antibody-PRR agonist conjugates for cancer therapy.
Collapse
Affiliation(s)
- Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiang Li
- Beijing Kawin Technology Share-Holding Co., Ltd, BDA, Beijing, 100176, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| |
Collapse
|
37
|
Ottevanger R, Melchers RC, Quint KD. Remittance of primary cutaneous CD30+ lymphoproliferative disorder in a patient on adalimumab. JAAD Case Rep 2022; 22:34-37. [PMID: 35274033 PMCID: PMC8904182 DOI: 10.1016/j.jdcr.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
|
38
|
CD30-targeted therapy induces apoptosis of inflammatory cytokine-stimulated synovial fibroblasts and ameliorates collagen antibody-induced arthritis in mice. Inflamm Res 2022; 71:215-226. [DOI: 10.1007/s00011-021-01537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 12/20/2021] [Indexed: 11/05/2022] Open
|
39
|
Lerchen HG, Stelte-Ludwig B, Kopitz C, Heroult M, Zubov D, Willuda J, Schlange T, Kahnert A, Wong H, Izumi R, Hamdy A. A Small Molecule–Drug Conjugate (SMDC) Consisting of a Modified Camptothecin Payload Linked to an αVß3 Binder for the Treatment of Multiple Cancer Types. Cancers (Basel) 2022; 14:cancers14020391. [PMID: 35053556 PMCID: PMC8773721 DOI: 10.3390/cancers14020391] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/07/2021] [Accepted: 01/04/2022] [Indexed: 12/27/2022] Open
Abstract
To improve tumor selectivity of cytotoxic agents, we designed VIP236, a small molecule–drug conjugate consisting of an αVβ3 integrin binder linked to a modified camptothecin payload (VIP126), which is released by the enzyme neutrophil elastase (NE) in the tumor microenvironment (TME). The tumor targeting and pharmacokinetics of VIP236 were studied in tumor-bearing mice by in vivo near-infrared imaging and by analyzing tumor and plasma samples. The efficacy of VIP236 was investigated in a panel of cancer cell lines in vitro, and in MX-1, NCI-H69, and SW480 murine xenograft models. Imaging studies with the αVβ3 binder demonstrated efficient tumor targeting. Administration of VIP126 via VIP236 resulted in a 10-fold improvement in the tumor/plasma ratio of VIP126 compared with VIP126 administered alone. Unlike SN38, VIP126 is not a substrate of P-gp and BCRP drug transporters. VIP236 presented strong cytotoxic activity in the presence of NE. VIP236 treatment resulted in tumor regressions and very good tolerability in all in vivo models tested. VIP236 represents a novel approach for delivering a potent cytotoxic agent by utilizing αVβ3 as a targeting moiety and NE in the TME to release the VIP126 payload—designed for high permeability and low efflux—directly into the tumor stroma.
Collapse
Affiliation(s)
- Hans-Georg Lerchen
- Vincerx Pharma GmbH, 40789 Monheim am Rhein, Germany;
- Correspondence: ; Tel.: +49-157-31993091
| | | | | | - Melanie Heroult
- Crop Science Division, Bayer AG, 65926 Frankfurt am Main, Germany;
| | - Dmitry Zubov
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Joerg Willuda
- Pharmaceuticals R&D, Bayer AG, 13353 Berlin, Germany;
| | - Thomas Schlange
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Antje Kahnert
- Pharmaceuticals R&D, Bayer AG, 42096 Wuppertal, Germany; (D.Z.); (T.S.); (A.K.)
| | - Harvey Wong
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| | - Raquel Izumi
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| | - Ahmed Hamdy
- Vincerx Pharma Inc., Palo Alto, CA 94306, USA; (H.W.); (R.I.); (A.H.)
| |
Collapse
|
40
|
Tsujii S, Serada S, Fujimoto M, Uemura S, Namikawa T, Nomura T, Murakami I, Hanazaki K, Naka T. Glypican-1 Is a Novel Target for Stroma and Tumor Cell Dual-Targeting Antibody-Drug Conjugates in Pancreatic Cancer. Mol Cancer Ther 2021; 20:2495-2505. [PMID: 34583978 DOI: 10.1158/1535-7163.mct-21-0335] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/21/2021] [Accepted: 09/20/2021] [Indexed: 02/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a stroma-rich cancer. Extracellular matrix proteins produced by cancer-associated fibroblasts (CAFs) found in tumor stroma that impedes effective delivery of chemotherapeutic agents results in poor response in patients with PDAC. Previously, our group reported that glypican-1 (GPC1) was overexpressed in human PDAC and negatively correlated with patient survival. Immunohistochemical analysis of 25 patients with PDAC tumor specimens revealed elevated expression of GPC1 in stromal cells and pancreatic cancer cells in 80% of patients. Interestingly, GPC1 was expressed on CAFs in PDAC. We generated a GPC1 antibody-drug conjugate conjugated with monomethyl auristatin E [GPC1-ADC(MMAE)] and evaluated its preclinical antitumor activity by targeting GPC1-positive CAF and cancer cells in PDAC. GPC1-ADC(MMAE) inhibited the growth of GPC1-positive PDAC cell lines in vitro Furthermore, GPC1-ADC(MMAE) showed a potent antitumor effect in the PDAC patient-derived tumor xenograft (PDX) model against GPC1-positive CAF and heterogeneous GPC1-expressing cancer cells. Notably, GPC1-ADC(MMAE) showed robust preclinical efficacy against GPC1 in a stroma-positive/cancer-negative PDAC PDX model. GPC1-ADC(MMAE) was delivered and internalized to CAFs. Although apoptosis was not observed in CAFs, the released MMAE from CAFs via MDR-1 induced apoptosis of cancer cells neighboring CAFs and efficiently inhibited PDAC tumor growth. GPC1-ADC(MMAE) exhibited potent and unique antitumor activity in GPC1-positive PDAC PDX models, which suggests that GPC1 is a novel therapeutic target in PDAC and other stromal GPC1-positive solid tumors. These findings show that targeting GPC1 on CAF using GPC1-ADC(MMAE) is a useful approach in case of stroma-rich tumors such as PDAC.
Collapse
Affiliation(s)
- Shigehiro Tsujii
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Satoshi Serada
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan.
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan
| | - Minoru Fujimoto
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| | - Sunao Uemura
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Tsutomu Namikawa
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Taisei Nomura
- Animal Models of Human Diseases, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Ichiro Murakami
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Kazuhiro Hanazaki
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Tetsuji Naka
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan.
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan
- Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| |
Collapse
|
41
|
Pei M, Liu T, Ouyang L, Sun J, Deng X, Sun X, Wu W, Huang P, Chen YL, Tan X, Liu X, Zhu P, Liu Y, Wang D, Wu J, Wang Q, Wang G, Gong L, Qin Q, Wang C. Enzyme-linked immunosorbent assays for quantification of MMAE-Conjugated ADCs and total antibodies in cynomolgus monkey sera. J Pharm Anal 2021; 12:645-652. [PMID: 36105165 PMCID: PMC9463470 DOI: 10.1016/j.jpha.2021.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are commonly heterogeneous and require extensive assessment of exposure-efficacy and exposure-safety relationships in preclinical and clinical studies. In this study, we report the generation of a monoclonal antibody against monomethyl auristatin E (MMAE) and the development, validation, and application of sensitive and high-throughput enzyme-linked immunosorbent assays (ELISA) to measure the concentrations of MMAE-conjugated ADCs and total antibodies (tAb, antibodies in ADC plus unconjugated antibodies) in cynomolgus monkey sera. These assays were successfully applied to in vitro plasma stability and pharmacokinetic (PK) studies of SMADC001, an MMAE-conjugated ADC against trophoblast cell surface antigen 2 (TROP-2). The plasma stability of SMADC001 was better than that of similar ADCs coupled with PEG4-Val-Cit, Lys (m-dPEG24)-Cit, and Val-Cit linkers. The developed ELISA methods for the calibration standards of ADC and tAb revealed a correlation between serum concentrations and the OD450 values, with R2 at 1.000, and the dynamic range was 0.3–35.0 ng/mL and 0.2–22.0 ng/mL, respectively; the intra- and inter-assay accuracy bias% ranged from −12.2% to −5.2%, precision ranged from −12.4% to −1.4%, and the relative standard deviation (RSD) was less than 6.6% and 8.7%, respectively. The total error was less than 20.4%. The development and validation steps of these two assays met the acceptance criteria for all addressed validation parameters, which suggested that these can be applied to quantify MMAE-conjugated ADCs, as well as in PK studies. Furthermore, these assays can be easily adopted for development of other similar immunoassays. A specific monoclonal antibody against MMAE was generated via hybridoma technology. ELISA was used to quantify MMAE-ADCs with a calibration range of 0.5–35 ng/mL. ELISA was used to quantify total antibodies with a calibration range of 0.6–22 ng/mL. Pharmacokinetic profiles of SMADC001 in cynomolgus monkeys were investigated.
Collapse
Affiliation(s)
- Min Pei
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tingting Liu
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu Ouyang
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianhua Sun
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaojie Deng
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaomin Sun
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei Wu
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Peng Huang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi-Li Chen
- Research and Development Center, Dartsbio Pharmaceuticals Ltd., Zhongshan, Guangdong, 528400, China
| | - Xiaorong Tan
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaoyue Liu
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peng Zhu
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yongzhen Liu
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Deheng Wang
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Junliang Wu
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qi Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guifeng Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Likun Gong
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, 528400, China
- Corresponding author. Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Qiuping Qin
- Center for Drug Safety Evaluation and Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Corresponding author.
| | - Chunhe Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
- Research and Development Center, Dartsbio Pharmaceuticals Ltd., Zhongshan, Guangdong, 528400, China
- School of Pharmacy, Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai, 201203, China
- Corresponding author. Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
42
|
Kapoor S, Champion G, Basu A, Mariampillai A, Olnes MJ. Immune Therapies for Myelodysplastic Syndromes and Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5026. [PMID: 34638510 PMCID: PMC8507987 DOI: 10.3390/cancers13195026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are hematologic malignancies arising from the bone marrow. Despite recent advances in treating these diseases, patients with higher-risk MDS and AML continue to have a poor prognosis with limited survival. It has long been recognized that there is an immune component to the pathogenesis of MDS and AML, but until recently, immune therapies have played a limited role in treating these diseases. Immune suppressive therapy exhibits durable clinical responses in selected patients with MDS, but the question of which patients are most suitable for this treatment remains unclear. Over the past decade, there has been remarkable progress in identifying genomic features of MDS and AML, which has led to an improved discernment of the molecular pathogenesis of these diseases. An improved understanding of immune and inflammatory molecular mechanisms of MDS and AML have also recently revealed novel therapeutic targets. Emerging treatments for MDS and AML include monoclonal antibodies such as immune checkpoint inhibitors, bispecific T-cell-engaging antibodies, antibody drug conjugates, vaccine therapies, and cellular therapeutics including chimeric antigen receptor T-cells and NK cells. In this review, we provide an overview of the current understanding of immune dysregulation in MDS and AML and an update on novel immune therapies for these bone marrow malignancies.
Collapse
Affiliation(s)
- Sargam Kapoor
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Grace Champion
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Aparna Basu
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
| | - Anu Mariampillai
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Matthew J. Olnes
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
- WWAMI School of Medical Education, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| |
Collapse
|
43
|
Märcher A, Nijenhuis MAD, Gothelf KV. A Wireframe DNA Cube: Antibody Conjugate for Targeted Delivery of Multiple Copies of Monomethyl Auristatin E. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107221] [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)
- Anders Märcher
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Minke A. D. Nijenhuis
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| | - Kurt V. Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO) Aarhus University Gustav Wieds Vej 14 8000 Aarhus Denmark
| |
Collapse
|
44
|
Yokota K, Serada S, Tsujii S, Toya K, Takahashi T, Matsunaga T, Fujimoto M, Uemura S, Namikawa T, Murakami I, Kobayashi S, Eguchi H, Doki Y, Hanazaki K, Naka T. Anti-Glypican-1 Antibody-drug Conjugate as Potential Therapy Against Tumor Cells and Tumor Vasculature for Glypican-1-Positive Cholangiocarcinoma. Mol Cancer Ther 2021; 20:1713-1722. [PMID: 34224365 DOI: 10.1158/1535-7163.mct-21-0015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/27/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
Cholangiocarcinoma is a highly malignant cancer. Many patients need systemic chemotherapy to prevent tumor development and recurrence; however, their prognosis is poor due to the lack of effective therapy. Therefore, a new treatment option is urgently required. We recently identified glypican-1 (GPC1) as a novel cancer antigen of esophageal squamous cell carcinoma. We also demonstrated the efficacy and safety of GPC1-targeted ADC (GPC1-ADC) conjugating anti-GPC1 mAb possessing high internalization activity with monomethyl auristatin F (MMAF), which is a potent tubulin polymerizing inhibitor. In this study, we confirmed that GPC1 was highly expressed in cholangiocarcinoma cells and tissues. IHC analysis of 49 extrahepatic cholangiocarcinoma patient tumor specimens revealed high expression of GPC1 in 47% of patients. These patients demonstrated significantly poorer prognosis compared with the low-expression group in terms of disease-free survival and overall survival (P < 0.05). GPC1 was also expressed in tumor vessels of cholangiocarcinoma, but not on the vessels of nontumor tissues. MMAF-conjugated GPC1-ADC showed potent tumor growth inhibition against GPC1-positive cholangiocarcinoma cells in vitro and in vivo In a GPC1 knockout xenograft model, GPC1-ADC partially inhibited tumor growth. Vascular endothelial cells in tumor tissues of GPC1-negative xenograft mice expressed GPC1 and were arrested in the G2-M phase of cell cycle by GPC1-ADC. GPC1-ADC exhibits direct as well as indirect antitumor effects via inhibition of tumor angiogenesis. Our preclinical data highlight GPC1-ADC as a promising therapy for GPC1-positive cholangiocarcinoma.
Collapse
Affiliation(s)
- Keiichiro Yokota
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Satoshi Serada
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Japan.
| | - Shigehiro Tsujii
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Keisuke Toya
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tsuyoshi Takahashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takashi Matsunaga
- Department of Medical course, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Minoru Fujimoto
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Sunao Uemura
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Tsutomu Namikawa
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Ichiro Murakami
- Department of Pathology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kazuhiro Hanazaki
- Department of Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Tetsuji Naka
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Japan.
| |
Collapse
|
45
|
Munekage E, Serada S, Tsujii S, Yokota K, Kiuchi K, Tominaga K, Fujimoto M, Kanda M, Uemura S, Namikawa T, Nomura T, Murakami I, Hanazaki K, Naka T. A glypican-1-targeted antibody-drug conjugate exhibits potent tumor growth inhibition in glypican-1-positive pancreatic cancer and esophageal squamous cell carcinoma. Neoplasia 2021; 23:939-950. [PMID: 34332450 PMCID: PMC8340053 DOI: 10.1016/j.neo.2021.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/08/2023]
Abstract
An antibody-drug conjugate (ADC) is a promising therapeutic modality because selective and effective delivery of an anti-cancer drug is achieved by drug-conjugated antibody-targeting cancer antigen. Glypican 1 (GPC1) is highly expressed in malignant tumors, including pancreatic ductal adenocarcinoma (PDAC) and esophageal squamous cell carcinoma (ESCC). Herein, we describe the usefulness of GPC1-targeting ADC. Humanized anti-GPC1 antibody (clone T2) was developed and conjugated with monomethyl auristatin E (MMAE) via maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl (mc-vc-PABC) linkers (humanized GPC1-ADC[MMAE]). Humanized GPC1-ADC(MMAE) inhibited the growth of GPC1-positive PDAC and ESCC cell lines via inducing cycle arrest in the G2/M phase and apoptosis in vitro. The binding activity of humanized GPC1-ADC(MMAE) with GPC1 was comparable with that of the unconjugated anti-GPC1 antibody. The humanized GPC1-ADC(MMAE) was effective in GPC1-positive BxPC-3 subcutaneously xenografted mice but not in GPC1-negative BxPC-3-GPC1-KO xenografted mice. To assess the bystander killing activity of the humanized GPC1-ADC(MMAE), a mixture of GPC1-positive BxPC-3 and GPC1-negative BxPC-3-GPC1-KO-Luc cells were subcutaneously inoculated, and a heterogenous GPC1-expressing tumor model was developed. The humanized GPC1-ADC(MMAE) inhibited the tumor growth and decreased the luciferase signal, measured with an in vivo imaging system (IVIS), which suggests that the suppression of the BxPC-3-GPC1-KO-Luc population. The humanized GPC1-ADC(MMAE) also inhibited the established liver metastases of BxPC-3 cells and significantly improved the overall survival of the mice. It exhibited a potent antitumor effect on the GPC1-positive PDAC and ESCC patient-derived xenograft (PDX) models. Our preclinical data demonstrate that GPC1 is a promising therapeutic target for ADC.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/metabolism
- Antigens, Neoplasm/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/physiology
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/immunology
- Esophageal Neoplasms/metabolism
- Esophageal Squamous Cell Carcinoma/drug therapy
- Esophageal Squamous Cell Carcinoma/immunology
- Esophageal Squamous Cell Carcinoma/metabolism
- Glypicans/antagonists & inhibitors
- Glypicans/metabolism
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/metabolism
- Humans
- Immunoconjugates/administration & dosage
- Immunoconjugates/metabolism
- Mice
- Mice, Knockout
- Mice, SCID
- Mice, Transgenic
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Eri Munekage
- Department of Surgery, Kochi University, Nankoku, Kochi, Japan; Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Satoshi Serada
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan.
| | - Shigehiro Tsujii
- Department of Surgery, Kochi University, Nankoku, Kochi, Japan; Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Keiichiro Yokota
- Department of Surgery, Kochi University, Nankoku, Kochi, Japan; Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Keita Kiuchi
- Department of Medical course, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Kenji Tominaga
- Department of Medical course, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Minoru Fujimoto
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan
| | - Mizuki Kanda
- Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan
| | - Sunao Uemura
- Department of Surgery, Kochi University, Nankoku, Kochi, Japan
| | | | - Taisei Nomura
- Animal Models of Human Diseases, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| | - Ichiro Murakami
- Department of Pathology, School of Medicine, Kochi University, Nankoku, Kochi, Japan
| | | | - Tetsuji Naka
- Department of Clinical Immunology, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan; Institute for Biomedical Sciences Molecular Pathophysiology, Iwate Medical University, Yahaba, Iwate, Japan; Division of Allergy and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba, Iwate, Japan.
| |
Collapse
|
46
|
The ROR1 antibody-drug conjugate huXBR1-402-G5-PNU effectively targets ROR1+ leukemia. Blood Adv 2021; 5:3152-3162. [PMID: 34424320 DOI: 10.1182/bloodadvances.2020003276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 04/18/2021] [Indexed: 11/20/2022] Open
Abstract
Antibody-drug conjugates directed against tumor-specific targets have allowed targeted delivery of highly potent chemotherapy to malignant cells while sparing normal cells. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein with limited expression on normal adult tissues and is overexpressed on the surface of malignant cells in mantle cell lymphoma, acute lymphocytic leukemia with t(1;19)(q23;p13) translocation, and chronic lymphocytic leukemia. This differential expression makes ROR1 an attractive target for antibody-drug conjugate therapy, especially in malignancies such as mantle cell lymphoma and acute lymphocytic leukemia, in which systemic chemotherapy remains the gold standard. Several preclinical and phase 1 clinical studies have established the safety and effectiveness of anti-ROR1 monoclonal antibody-based therapies. Herein we describe a humanized, first-in-class anti-ROR1 antibody-drug conjugate, huXBR1-402-G5-PNU, which links a novel anti-ROR1 antibody (huXBR1-402) to a highly potent anthracycline derivative (PNU). We found that huXBR1-402-G5-PNU is cytotoxic to proliferating ROR1+ malignant cells in vitro and suppressed leukemia proliferation and extended survival in multiple models of mice engrafted with human ROR1+ leukemia. Lastly, we show that the B-cell lymphoma 2 (BCL2)-dependent cytotoxicity of huXBR1-402-G5-PNU can be leveraged by combined treatment strategies with the BCL2 inhibitor venetoclax. Together, our data present compelling preclinical evidence for the efficacy of huXBR1-402-G5-PNU in treating ROR1+ hematologic malignancies.
Collapse
|
47
|
Matsuda Y, Leung M, Tawfiq Z, Fujii T, Mendelsohn BA. In-situ Reverse Phased HPLC Analysis of Intact Antibody-Drug Conjugates. ANAL SCI 2021; 37:1171-1176. [PMID: 33518587 DOI: 10.2116/analsci.20p424] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/22/2021] [Indexed: 08/09/2023]
Abstract
The field of oncology has recently seen an exponential growth in antibody-drug conjugates (ADCs) as a biopharmaceutical class with seven ADCs being launched onto the market in the last ten years. Despite the increase in the industrial research and development of these compounds, their structural complexity and heterogeneity continue to present various challenges regarding their analysis including reaction monitoring. Robust and simple reaction monitoring analysis are in demand in the view of at-line in-process monitoring, and can instill control, confidence and reliability in the ADC manufacturing process. Aiming at providing chromatographic methods for conjugation monitoring, we evaluated herein the potential of utilizing reverse phase HPLC analysis, without sample pretreatment, for characterization of traditional cysteine-based ADCs. This analysis can be used for estimation of drug antibody ratio (DAR), which has shown the same trends and results as other well-established HPLC techniques. This methodology was also applied to three ADCs derived from three different antibodies. Additionally, we analyzed unpurified ADC samples existing in a complex reaction matrix and separated ADC species and payload compounds. This investigation was conducted using three different ADCs based on different payloads. The results described herein indicate the potential application of this RP-HPLC methodology in reaction monitoring studies.
Collapse
Affiliation(s)
- Yutaka Matsuda
- Ajinomoto Co., Inc., 1-1 Suzuki-cho, Kawasaki, Kawasaki, Kanagawa, 210-8681, Japan.
| | - Monica Leung
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA, 92121, United States
| | - Zhala Tawfiq
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA, 92121, United States
| | - Tomohiro Fujii
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA, 92121, United States
| | - Brian A Mendelsohn
- Ajinomoto Bio-Pharma Services, 11040 Roselle Street, San Diego, CA, 92121, United States.
- Exelixis Inc, 1851 Harbor Bay Pkwy, Alameda, CA, 94502, United States.
| |
Collapse
|
48
|
Märcher A, Nijenhuis MAD, Gothelf KV. A Wireframe DNA Cube: Antibody Conjugate for Targeted Delivery of Multiple Copies of Monomethyl Auristatin E. Angew Chem Int Ed Engl 2021; 60:21691-21696. [PMID: 34309988 DOI: 10.1002/anie.202107221] [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] [Received: 05/30/2021] [Revised: 07/18/2021] [Indexed: 11/08/2022]
Abstract
In recent years, several antibody drug conjugates (ADC) have been accepted by the FDA as therapeutics against cancer. It is well-known that control of drug-to-antibody ratio (DAR) is vital for the success of an ADC, which inspires the advancement of better and simpler methods for tight control of DAR. We present the development of an antibody DNA wireframe cube conjugate for precise control of DAR. The DNA wireframe cube consists of four single strands, which when folded present eight single stranded domains. One domain is bound to a monofunctionalized antibody DNA conjugate, and the seven others are attached to DNA functionalized with the potent tubulin inhibitor MMAE, thereby preparing an ADC with a DAR of precisely seven. The formation of the ADC is investigated by gel electrophoresis and atomic force microscopy. Lastly, the developed MMAE loaded ADC was used for targeted drug delivery in vitro.
Collapse
Affiliation(s)
- Anders Märcher
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Minke A D Nijenhuis
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Kurt V Gothelf
- Department of Chemistry and Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| |
Collapse
|
49
|
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
|
50
|
Matsumiya W, Karaca I, Ghoraba H, Akhavanrezayat A, Mobasserian A, Hassan M, Regenold J, Yasar C, Liedtke M, Kitazawa K, Nguyen QD. Structural changes of corneal epithelium in belantamab-associated superficial keratopathy using anterior segment optical coherence tomography. Am J Ophthalmol Case Rep 2021; 23:101133. [PMID: 34169181 PMCID: PMC8208963 DOI: 10.1016/j.ajoc.2021.101133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/26/2021] [Accepted: 06/05/2021] [Indexed: 10/26/2022] Open
Abstract
Purpose To describe structural changes in corneal epithelium using anterior segment optical coherence tomography (AS-OCT) in two relapsed and refractory multiple myeloma (RRMM) patients with bilateral belantamab-associated superficial keratopathy (BASK). Observations case 1 A 56-year-old male who was diagnosed with RRMM and initiated on belantamab mafodotin, presented on day 42 (three weeks after the second infusion) with decreased pinhole visual acuity from 20/20 and 20/25 to 20/70 and 20/50 in the right eye and left eye, respectively. Slit-lamp examination revealed moderate superficial keratopathy with microcystic-like epithelial changes (MECs) in the paracentral cornea in both eyes. AS-OCT demonstrated increased bilateral heterogeneous signal intensity and hyperreflective lesions as well as increased thickness in the paracentral corneal epithelium with uninvolved central cornea. Given bilateral MECs, the third infusion was withheld, and then given on day 62 after five weeks of drug-free interval. Although MECs had improved on day 82, pinhole visual acuity remained at 20/50 and 20/40 in the right eye and the left eye. AS-OCT showed that hyperreflective lesions mostly resolved and corneal epithelial thickness returned to baseline, despite a slightly increased persisting heterogeneous signal intensity in the peripheral corneal epithelium in both eyes. Case 2 A 77-year-old male with RRMM was started on belantamab mafodotin infusions. His pinhole visual acuity decreased from 20/40 and 20/30 at baseline to 20/60 and 20/40 on day 41 (three weeks after the second infusion) in the right eye and left eye, respectively. Slit-lamp examination showed diffuse, moderate MECs in both eyes, which was more severe in the peripheral cornea. AS-OCT demonstrated increased bilateral heterogeneous signal intensity and hyperreflective lesions in the corneal epithelium, which are more severe in the right eye along with increased corneal epithelial thickness. Therefore, belantamab mafodotin was withheld. Conclusions and Impotance AS-OCT objectively demonstrated structural changes such as signal intensity and thickness alterations with hyperreflective lesions in the corneal epithelium related to BASK. AS-OCT might be useful for clinicians to monitor ocular surface adverse events in RRMM patients receiving belantamab mafodotin and to adjust therapeutic plans for the patients.
Collapse
Affiliation(s)
- Wataru Matsumiya
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA.,Department of Surgery, Division of Ophthalmology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Irmak Karaca
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Hashem Ghoraba
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Amir Akhavanrezayat
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Azadeh Mobasserian
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Muhammad Hassan
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Jonathan Regenold
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Cigdem Yasar
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Koji Kitazawa
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Buck Institute for Research on Aging, Novato, CA, USA
| | - Quan Dong Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, USA
| |
Collapse
|