1
|
Ding Y, Jia L, Geng Q, Liu Y, Guo S, Zhao S, Kong Y, Jin Q, Xu G, Xu J. Screening and functional characterization of nanobodies targeting the transferrin receptor. Protein Expr Purif 2025; 231:106702. [PMID: 40089126 DOI: 10.1016/j.pep.2025.106702] [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: 01/23/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
The transferrin receptor (TfR1) mediates the cellular uptake of iron and other molecules, playing a vital role in hematology and tumor growth. Nanobodies (NBs) targeting TfR1 offer promising therapeutic potential due to their small size, high specificity and stability. However, rapid identification of effective nanobodies remains challenging.In this study, the truncated extracellular fragment of human TfR1 was expressed in a prokaryotic system and purified. Immunized camelids provided a source for nanobody libraries, which were screened using phage display and high-throughput strategies to identify candidates with specific TfR1 binding.NB 2D7 with nanomolar-level dissociation constants (KD) were successfully identified.The analysis of Cell Counting Kit-8(CCK8) experiments indicates that the combined treatment of NB2D7 with FeCl3 can reduce the survival rate of LoVo cells.This research establishes an efficient platform for anti-TfR1 nanobody screening and highlights the therapeutic potential of these nanobodies in cancer treatment and iron metabolism disorders.
Collapse
Affiliation(s)
- Yuting Ding
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Li Jia
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Qifubo Geng
- School of Information Engineering, Minzu University of China, Beijing, 100081, China
| | - Yan Liu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; College of Pharmacy, Henan University, Kaifeng, Henan, 475000, China
| | - Shaojue Guo
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Shuaiying Zhao
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yingying Kong
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; College of Pharmacy, Henan University, Kaifeng, Henan, 475000, China
| | - Quanfang Jin
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, 201900, China.
| | - Guangxu Xu
- Department of Gynecology, Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Shanghai, 200233, China.
| | - Jianfeng Xu
- Department of Biopharmaceutics, College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
2
|
Baghirov H. Mechanisms of receptor-mediated transcytosis at the blood-brain barrier. J Control Release 2025; 381:113595. [PMID: 40056994 DOI: 10.1016/j.jconrel.2025.113595] [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: 12/21/2024] [Revised: 02/09/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025]
Abstract
In receptor-mediated transcytosis (RMT) of large therapeutics across the blood-brain barrier (BBB), the construct - a macromolecule or a larger carrier with therapeutic payload - binds a protein on brain capillary endothelial cells (BCEC), with internalization and release into the brain parenchyma. The construct's internalization into, trafficking across and release from, but also possible entrapment within BCEC are affected by its engineered properties whose optimization has helped derive insights into transport mechanisms at BCEC. Furthermore, advances in multi-omics, as well as large-scale screening and directed evolution campaigns have helped identify new targets for RMT at BCEC. In this perspective, I raise and reflect on some fundamental questions one can arrive at by comparing the engineered properties of BBB-targeted constructs and the properties of different target proteins. These questions concern the underlying, transcytosis-promoting factors that the optimization of constructs' engineered properties appears to converge on, the precise role of target proteins in RMT, the different mechanisms through which these targets may mediate construct trafficking, and the tentative criteria for target selection on BCEC. Based on these considerations I propose several scenarios and strategies to interfere with the construct's trafficking for more efficient internalization, transport through the endosomal network toward the abluminal membrane, and release from BCEC, both for smaller macromolecules and for larger carriers.
Collapse
Affiliation(s)
- Habib Baghirov
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500 Turku, Finland.
| |
Collapse
|
3
|
Cuypers ML, Jaspers T, Clerckx J, Leekens S, Cawthorne C, Bormans G, Cleeren F, Geukens N, De Strooper B, Dewilde M. Increasing brain half-life of antibodies by additional binding to myelin oligodendrocyte glycoprotein, a CNS specific protein. Fluids Barriers CNS 2025; 22:11. [PMID: 39885527 PMCID: PMC11783731 DOI: 10.1186/s12987-025-00624-1] [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: 09/27/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Therapeutic antibodies for the treatment of neurological disease show great potential, but their applications are rather limited due to limited brain exposure. The most well-studied approach to enhance brain influx of protein therapeutics, is receptor-mediated transcytosis (RMT) by targeting nutrient receptors to shuttle protein therapeutics over the blood-brain barrier (BBB) along with their endogenous cargos. While higher brain exposure is achieved with RMT, the timeframe is short due to rather fast brain clearance. Therefore, we aim to increase the brain half-life of antibodies by binding to myelin oligodendrocyte glycoprotein (MOG), a CNS specific protein. METHODS Alpaca immunization with mouse/human MOG, and subsequent phage selections and screenings for MOG binding single variable domain antibodies (VHHs) were performed to find mouse/human cross-reactive VHHs. Their ability to increase the brain half-life of antibodies was evaluated in healthy wild-type mice by coupling two different MOG VHHs (low/high affinity) in a mono- and bivalent format to a β-secretase 1 (BACE1) inhibiting antibody or a control (anti-SARS-CoV-2) antibody, fused to an anti-transferrin receptor (TfR) VHH for active transport over the BBB. Brain pharmacokinetics and pharmacodynamics, CNS and peripheral biodistribution, and brain toxicity were evaluated after intravenous administration to balb/c mice. RESULTS Additional binding to MOG increases the Cmax and brain half-life of antibodies that are actively shuttled over the BBB. Anti-SARS-CoV-2 antibodies coupled with an anti-TfR VHH and two low affinity anti-MOG VHHs could be detected in brain 49 days after a single intravenous injection, which is a major improvement compared to an anti-SARS-CoV-2 antibody fused to an anti-TfR VHH which cannot be detected in brain anymore one week post treatment. Additional MOG binding of antibodies does not affect peripheral biodistribution but alters brain distribution to white matter localization and less neuronal internalization. CONCLUSIONS We have discovered mouse/human/cynomolgus cross-reactive anti-MOG VHHs which have the ability to drastically increase brain exposure of antibodies. Combining MOG and TfR binding leads to distinct PK, biodistribution, and brain exposure, differentiating it from the highly investigated TfR-shuttling. It is the first time such long brain antibody exposure has been demonstrated after one single dose. This new approach of adding a binding moiety for brain specific targets to RMT shuttling antibodies is a huge advancement for the field and paves the way for further research into brain half-life extension.
Collapse
Affiliation(s)
- Marie-Lynn Cuypers
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium
| | - Tom Jaspers
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium
| | - Jarne Clerckx
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium
| | - Simon Leekens
- Laboratory for Radiopharmaceutical Research, KU Leuven - University of Leuven, O&N II Herestraat 49 box 821, 3000, Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven - University of Leuven, O&N I Herestraat 49 box 505, 3000, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, KU Leuven - University of Leuven, O&N II Herestraat 49 box 821, 3000, Leuven, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, KU Leuven - University of Leuven, O&N II Herestraat 49 box 821, 3000, Leuven, Belgium
| | - Nick Geukens
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium
- PharmAbs - the KU Leuven Antibody Center, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium
| | - Bart De Strooper
- Laboratory for the Research of Neurodegenerative Diseases - VIB Center for Brain and Disease Research, O&N V, Herestraat 49 box 602, 3000, Leuven, Belgium
| | - Maarten Dewilde
- Laboratory for Therapeutic and Diagnostic Antibodies, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium.
- PharmAbs - the KU Leuven Antibody Center, KU Leuven - University of Leuven, O&N II Herestraat 49 box 820, 3000, Leuven, Belgium.
| |
Collapse
|
4
|
Flores-Prieto DE, Stabenfeldt SE. Nanoparticle targeting strategies for traumatic brain injury. J Neural Eng 2024; 21:061007. [PMID: 39622184 DOI: 10.1088/1741-2552/ad995b] [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: 05/06/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems hold immense potential for targeted therapy and diagnosis of neurological disorders, overcoming the limitations of conventional treatment modalities. This review explores the design considerations and functionalization strategies of NPs for precise targeting of the brain and central nervous system. This review discusses the challenges associated with drug delivery to the brain, including the blood-brain barrier and the complex heterogeneity of traumatic brain injury. We also examine the physicochemical properties of NPs, emphasizing the role of size, shape, and surface characteristics in their interactions with biological barriers and cellular uptake mechanisms. The review concludes by exploring the options of targeting ligands designed to augment NP affinity and retention to specific brain regions or cell types. Various targeting ligands are discussed for their ability to mimic receptor-ligand interaction, and brain-specific extracellular matrix components. Strategies to mimic viral mechanisms to increase uptake are discussed. Finally, the emergence of antibody, antibody fragments, and antibody mimicking peptides are discussed as promising targeting strategies. By integrating insights from these scientific fields, this review provides an understanding of NP-based targeting strategies for personalized medicine approaches to neurological disorders. The design considerations discussed here pave the way for the development of NP platforms with enhanced therapeutic efficacy and minimized off-target effects, ultimately advancing the field of neural engineering.
Collapse
Affiliation(s)
- David E Flores-Prieto
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| |
Collapse
|
5
|
Ghezelbash B, Deyhim M, Azarkeivan A, Pourfathollah AA, Eskandari N. The Effects of Glutathione Monoethyl Ester on Different Biochemical, Oxidant, and Antioxidant Levels During Storage in Leukoreduced Red Blood Cells. Adv Biomed Res 2024; 13:111. [PMID: 39717241 PMCID: PMC11665185 DOI: 10.4103/abr.abr_169_23] [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: 05/17/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 12/25/2024] Open
Abstract
Background It is essential to maintain the quality of the stored blood, because various factors affect the stored red blood cells (RBCs) over time, some red blood cell storage lesions (RCSL) develop during storage, and it could reduce the function of the RBCs. The present study aimed to evaluate the effects of glutathione monoethyl ester on different biochemical changes, oxidant, and antioxidant levels in the leukoreduced RBCs (LR-RBCs) during storage. Materials and Methods About 10 units of LR-RBC were collected, processed and stored according to the standard operating procedures (SOPs) of the Iranian Blood Transfusion Organization. Each unit divided into 2 equal parts; LR-RBC treated with glutathione monoethyl ester and a control group. Exposure of phosphatidylserine (PS), reactive oxygen species (ROS) and microvesicle derived from the RBCs (RBC-MVs), were measured by the flow cytometry method. ELISA was used to measure the level of glutathione, and 2, 3-diphosphoglycerate (2,3-DPG). Glucose-6-phosphate dehydrogenase (G6PD) enzyme activity was measured with a chemistry autoanalyzer. Results The levels of glutathione reduced the initial value in the treated group (80%), and the control group (60%), respectively. Exposure of surface PS, ROS and RBC-MVs increased significantly during storage time for consecutive weeks to the amount of GSH. The levels of 2,3-DPG decreased with increasing storage time. Conclusions Overall, The study suggest that glutathione monoethyl ester is effective to reduce the oxidative stress and the quality of RBCs can be improved.
Collapse
Affiliation(s)
- Behrooz Ghezelbash
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Mohammadreza Deyhim
- Department of Biochemistry, Faculty of Medicine, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Azita Azarkeivan
- Department of Biochemistry, Faculty of Medicine, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
6
|
Istomina PV, Gorchakov AA, Paoin C, Yamabhai M. Phage display for discovery of anticancer antibodies. N Biotechnol 2024; 83:205-218. [PMID: 39186973 DOI: 10.1016/j.nbt.2024.08.506] [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: 02/24/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Antibodies and antibody-based immunotherapeutics are the mainstays of cancer immunotherapy. Expanding the repertoire of cancer-specific and cancer-associated epitopes targetable with antibodies represents an important area of research. Phage display is a powerful approach allowing the use of diverse antibody libraries to be screened for binding to a wide range of targets. In this review, we summarize the basics of phage display technology and highlight the advances in anticancer antibody identification and modification via phage display platform. Finally, we describe phage display-derived anticancer monoclonal antibodies that have been approved to date or are in clinical development.
Collapse
Affiliation(s)
- Polina V Istomina
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Andrey A Gorchakov
- Institute of Molecular and Cellular Biology of the Siberian Branch of the Russian Academy of Sciences, Lavrentieva 8/2, Novosibirsk 630090, Russia
| | - Chatchanok Paoin
- Medical Oncology Division, Institute of Medicine, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand
| | - Montarop Yamabhai
- Molecular Biotechnology Laboratory, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Suranaree, Muang, 111 University Avenue, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
7
|
Culkins C, Adomanis R, Phan N, Robinson B, Slaton E, Lothrop E, Chen Y, Kimmel BR. Unlocking the Gates: Therapeutic Agents for Noninvasive Drug Delivery Across the Blood-Brain Barrier. Mol Pharm 2024; 21:5430-5454. [PMID: 39324552 DOI: 10.1021/acs.molpharmaceut.4c00604] [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: 09/27/2024]
Abstract
The blood-brain barrier (BBB) is a highly selective network of various cell types that acts as a filter between the blood and the brain parenchyma. Because of this, the BBB remains a major obstacle for drug delivery to the central nervous system (CNS). In recent years, there has been a focus on developing various modifiable platforms, such as monoclonal antibodies (mAbs), nanobodies (Nbs), peptides, and nanoparticles, as both therapeutic agents and carriers for targeted drug delivery to treat brain cancers and diseases. Methods for bypassing the BBB can be invasive or noninvasive. Invasive techniques, such as transient disruption of the BBB using low pulse electrical fields and intracerebroventricular infusion, lack specificity and have numerous safety concerns. In this review, we will focus on noninvasive transport mechanisms that offer high levels of biocompatibility, personalization, specificity and are regarded as generally safer than their invasive counterparts. Modifiable platforms can be designed to noninvasively traverse the BBB through one or more of the following pathways: passive diffusion through a physio-pathologically disrupted BBB, adsorptive-mediated transcytosis, receptor-mediated transcytosis, shuttle-mediated transcytosis, and somatic gene transfer. Through understanding the noninvasive pathways, new applications, including Chimeric Antigen Receptors T-cell (CAR-T) therapy, and approaches for drug delivery across the BBB are emerging.
Collapse
Affiliation(s)
- Courtney Culkins
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Roman Adomanis
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Nathan Phan
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise Robinson
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ethan Slaton
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Elijah Lothrop
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yinuo Chen
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Blaise R Kimmel
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Engineering, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Pelotonia Institute for Immuno-Oncology, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
8
|
Huang Q, Chan KY, Wu J, Botticello-Romero NR, Zheng Q, Lou S, Keyes C, Svanbergsson A, Johnston J, Mills A, Lin CY, Brauer PP, Clouse G, Pacouret S, Harvey JW, Beddow T, Hurley JK, Tobey IG, Powell M, Chen AT, Barry AJ, Eid FE, Chan YA, Deverman BE. An AAV capsid reprogrammed to bind human transferrin receptor mediates brain-wide gene delivery. Science 2024; 384:1220-1227. [PMID: 38753766 DOI: 10.1126/science.adm8386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Developing vehicles that efficiently deliver genes throughout the human central nervous system (CNS) will broaden the range of treatable genetic diseases. We engineered an adeno-associated virus (AAV) capsid, BI-hTFR1, that binds human transferrin receptor (TfR1), a protein expressed on the blood-brain barrier. BI-hTFR1 was actively transported across human brain endothelial cells and, relative to AAV9, provided 40 to 50 times greater reporter expression in the CNS of human TFRC knockin mice. The enhanced tropism was CNS-specific and absent in wild-type mice. When used to deliver GBA1, mutations of which cause Gaucher disease and are linked to Parkinson's disease, BI-hTFR1 substantially increased brain and cerebrospinal fluid glucocerebrosidase activity compared with AAV9. These findings establish BI-hTFR1 as a potential vector for human CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Ken Y Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jason Wu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Nuria R Botticello-Romero
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Qingxia Zheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Shan Lou
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Casey Keyes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Alexander Svanbergsson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jencilin Johnston
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Allan Mills
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Chin-Yen Lin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Pamela P Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Gabrielle Clouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Simon Pacouret
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - John W Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Thomas Beddow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jenna K Hurley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Isabelle G Tobey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Megan Powell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Albert T Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Andrew J Barry
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Fatma-Elzahraa Eid
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
- Department of Systems and Computer Engineering, Faculty of Engineering, Al-Azhar University, Cairo 11651, Egypt
| | - Yujia A Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| |
Collapse
|
9
|
Huang Q, Chan KY, Lou S, Keyes C, Wu J, Botticello-Romero NR, Zheng Q, Johnston J, Mills A, Brauer PP, Clouse G, Pacouret S, Harvey JW, Beddow T, Hurley JK, Tobey IG, Powell M, Chen AT, Barry AJ, Eid FE, Chan YA, Deverman BE. An AAV capsid reprogrammed to bind human Transferrin Receptor mediates brain-wide gene delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572615. [PMID: 38187643 PMCID: PMC10769326 DOI: 10.1101/2023.12.20.572615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Developing vehicles that efficiently deliver genes throughout the human central nervous system (CNS) will broaden the range of treatable genetic diseases. We engineered an AAV capsid, BI-hTFR1, that binds human Transferrin Receptor (TfR1), a protein expressed on the blood-brain barrier (BBB). BI-hTFR1 was actively transported across a human brain endothelial cell layer and, relative to AAV9, provided 40-50 times greater reporter expression in the CNS of human TFRC knock-in mice. The enhanced tropism was CNS-specific and absent in wild type mice. When used to deliver GBA1, mutations of which cause Gaucher disease and are linked to Parkinson's disease, BI-hTFR1 substantially increased brain and cerebrospinal fluid glucocerebrosidase activity compared to AAV9. These findings establish BI-hTFR1 as a promising vector for human CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Ken Y. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Shan Lou
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Casey Keyes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jason Wu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | | | - Qingxia Zheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jencilin Johnston
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Allan Mills
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Pamela P. Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Gabrielle Clouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Simon Pacouret
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - John W. Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Thomas Beddow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jenna K. Hurley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Isabelle G. Tobey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Megan Powell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Albert T. Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Andrew J. Barry
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Fatma-Elzahraa Eid
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
- Department of Systems and Computer Engineering, Faculty of Engineering, Al-Azhar University; Cairo, Egypt
| | - Yujia A. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Benjamin E. Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| |
Collapse
|
10
|
Zhang Q, Zhang N, Xiao H, Wang C, He L. Small Antibodies with Big Applications: Nanobody-Based Cancer Diagnostics and Therapeutics. Cancers (Basel) 2023; 15:5639. [PMID: 38067344 PMCID: PMC10705070 DOI: 10.3390/cancers15235639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/16/2023] [Accepted: 11/27/2023] [Indexed: 02/13/2025] Open
Abstract
Monoclonal antibodies (mAbs) have exhibited substantial potential as targeted therapeutics in cancer treatment due to their precise antigen-binding specificity. Despite their success in tumor-targeted therapies, their effectiveness is hindered by their large size and limited tissue permeability. Camelid-derived single-domain antibodies, also known as nanobodies, represent the smallest naturally occurring antibody fragments. Nanobodies offer distinct advantages over traditional mAbs, including their smaller size, high stability, lower manufacturing costs, and deeper tissue penetration capabilities. They have demonstrated significant roles as both diagnostic and therapeutic tools in cancer research and are also considered as the next generation of antibody drugs. In this review, our objective is to provide readers with insights into the development and various applications of nanobodies in the field of cancer treatment, along with an exploration of the challenges and strategies for their prospective clinical trials.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| | - Nan Zhang
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China;
| | - Han Xiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Chen Wang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| | - Lian He
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Q.Z.); (C.W.)
| |
Collapse
|
11
|
Baghirov H. Receptor-mediated transcytosis of macromolecules across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1699-1711. [PMID: 37658673 DOI: 10.1080/17425247.2023.2255138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/09/2023] [Accepted: 08/31/2023] [Indexed: 09/03/2023]
Abstract
INTRODUCTION The blood-brain barrier (BBB) restricts brain access of virtually all macromolecules. Receptor-mediated transcytosis (RMT) is one strategy toward their brain delivery. In this strategy, targeting ligands conjugated to therapeutic payload or decorating particles containing the payload interact with targets on brain capillary endothelial cells (BCEC), triggering internalization, trafficking, and release from BCEC. AREAS COVERED RMT at the BBB has leveraged multiple formats of macromolecules and large particles. Interactions between those and BCEC have been studied primarily using antibodies, with findings applicable to the design of larger particles. BBB-penetrant constructs have also been identified in screening campaigns and directed evolution, and subsequently found to interact with RMT targets. In addition, BCEC targeted by constructs incorporating genomic payload can be made to produce therapeutic proteins. EXPERT OPINION While targeting may not be strictly necessary to reach a therapeutic effect for all macromolecules, it can improve a molecule's BBB transport, exposing it to the entire brain parenchyma and enhancing its effect. Constructs with better BCEC transcytosis may be designed rationally, leveraging knowledge about BCEC trafficking, and found in screening campaigns, where this knowledge can reduce the search space and improve iterative refinement. Identification of new targets may also help generate BBB-crossing constructs.
Collapse
Affiliation(s)
- Habib Baghirov
- Roche Informatics, F. Hoffmann-La Roche Ltd, Poznań, Poland
| |
Collapse
|