1
|
Chen S, Nguyen A, Müller JT, Molbay M, Mehta A, Sheshachala S, Baskaya K, Adams N, Pinto Carneiro S, Merkel OM. Engineered-affibody conjugates contribute to the specific targeting and cellular retention of polyplexes in Erbb3 overexpressed lung cancer cells. Eur J Pharm Sci 2025; 209:107090. [PMID: 40174661 DOI: 10.1016/j.ejps.2025.107090] [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: 10/25/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Ligand-modified nanoparticles have shown the ability to specifically bind to tumor cells, improving retention in tumors after initial accumulation driven by the enhanced permeability and retention effect. These particles are typically engineered to bind to receptors overexpressed in cancer cells compared to healthy cells, such as the HER3 (Erbb3) receptor in lung cancer. In this study, we confirmed the overexpression of Erbb3 in various KRAS mutant lung cancer cell lines. An engineered affibody, well-established in previous research, was selected to target Erbb3 as a proof of concept. The affibody was integrated into the particle system via two distinct strategies. In the pre-functionalization approach, the affibody was conjugated to PEI or C14-PEI using SPDP as a linker. A spectral shift technique was then used to assess the affinity of the affibody and affibody conjugates toward Erbb3, allowing us to estimate the half-maximal effective concentration (EC50). Following synthesis and characterization, various polyplex formulations were prepared, including mRNA complexes with PEI-affibody, C14-PEI/PEI-affibody, and C14-PEI/C14-PEI-affibody. In the post-functionalization approach, polyplex formulations composed of different blends of C14-PEI and functionalized Azido-PEI were initially prepared and subsequently modified with DBCO-functionalized affibody via click chemistry. These formulations were prepared at various nitrogen to phosphate (N/P) ratios and characterized in terms of particle size, polydispersity index (PDI), and zeta potential. We also evaluated cellular uptake and eGFP mRNA expression to understand how the different formulations and conjugates influenced ligand-modified polyplex properties and delivery behavior. Our results demonstrated that affibody conjugates can specifically target Erbb3 and promote polyplex accumulation in KRAS-mutated lung cancer cells. We further analyzed the impact of conjugation methods and affibody density on polyplex design and performance. In conclusion, this study highlights the advantages of using specific targeting ligands. By optimizing formulation components, conjugation methods, and ligand density, various targeting ligands can be attached to polyplexes, enhancing cell-specific targeting, internalization, and retention. These findings provide valuable insights and a foundation for future targeted therapies and polyplex design.
Collapse
Affiliation(s)
- Siyu Chen
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | - Anny Nguyen
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | - Joschka T Müller
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | - Müge Molbay
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | - Aditi Mehta
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | | | - Kemal Baskaya
- NanoTemper Technologies GmbH, Toelzer straße 1, 81379, Munich, Germany
| | - Nathan Adams
- NanoTemper Technologies GmbH, Toelzer straße 1, 81379, Munich, Germany
| | - Simone Pinto Carneiro
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Butenandtstraße 5-13, 81377 Munich, 81377, Germany; Ludwig-Maximilians-Universität Munich, Member of the German Center for Lung Research (DZL), 81377, Munich, Germany.
| |
Collapse
|
2
|
Kang H, Li Y, Sun F, Li F, Zhu M, Zhou Z, Wu J, Wang K, Kutilike B, Miao Z, Xu Y, Liu R, Ding D, Zhang H, Huang X, Zhuang J. Modular Design of T Cell Nanoengagers for Tumor Immunotherapy via Genetically Engineered Lipid-Tagged Antibody Fragments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2503060. [PMID: 40259502 DOI: 10.1002/adma.202503060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/30/2025] [Indexed: 04/23/2025]
Abstract
T cell engagers, which bind tumor-associated antigens and T cell specific molecules, represent a promising class of immunotherapies for enhancing targeted immune responses. Here, a "plug-and-display" platform is introduced for engineering T cell nanoengagers by anchoring antibody fragments into lipid-based nanoparticles. This approach utilizes a genetically engineered lipoprotein fused with single-chain variable fragments (scFv) and nanobodies, which spontaneously integrate into lipid bilayer of the nanoparticles, achieving a high surface density of at least 0.102 scFv nm-2 (≈3200 scFv per particle). Modular bi-specific (Lipo-BiTE) and tri-specific (Lipo-TriTE) immunoliposomes are designed to enhance anti-tumor T cell immune responses. The Lipo-BiTE, integrating anti-CD3 and anti-HER2 scFv at an optimized surface density of 1.28 × 10-3 scFv nm-2, exhibits enhanced CD8+ T cell-mediated cytotoxicity in HER2-positive tumor models by simultaneously engaging tumor cells and T cells. Incorporating anti-PD-L1 nanobodies to create Lipo-TriTE further addresses T cell exhaustion. This modular platform provides a robust foundation for designing immune cell engagers, with broad applications in targeted immunotherapy.
Collapse
Affiliation(s)
- Helong Kang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuanke Li
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Fude Sun
- Key Laboratory of Hebei Province for Molecular Biophysic Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300401, China
| | - Fan Li
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Mingsheng Zhu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zheng Zhou
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jin Wu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Keyu Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Buayishamu Kutilike
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhizhao Miao
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yanqin Xu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ruming Liu
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Dan Ding
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hongkai Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xinglu Huang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jie Zhuang
- School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
3
|
Maksymova L, Pilger YA, Nuhn L, Van Ginderachter JA. Nanobodies targeting the tumor microenvironment and their formulation as nanomedicines. Mol Cancer 2025; 24:65. [PMID: 40033293 PMCID: PMC11877942 DOI: 10.1186/s12943-025-02270-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/12/2025] [Indexed: 03/05/2025] Open
Abstract
Among the emerging strategies for cancer theranostics, nanomedicines offer significant promise in advancing both patients' diagnosis and treatment. In combination with nanobodies, nanomedicines can potentially enhance the precision and efficiency of drug or imaging agent delivery, addressing key limitations of current approaches, such as off-target toxicities. The development of nanomedicines will be further accelerated by the creation of smart nanoparticles, and their integration with immunotherapy. Obviously, the success of nano-immunotherapy will depend on a comprehensive understanding of the tumor microenvironment, including the complex interplay of mechanisms that drive cancer-mediated immunosuppression and immune escape. Hence, effective therapeutic targeting of the tumor microenvironment requires modulation of immune cell function, overcoming resistance mechanisms associated with stromal components or the extracellular matrix, and/or direct elimination of cancer cells. Identifying key molecules involved in cancer progression and drug resistance is, therefore, essential for developing effective therapies and diagnostic tools that can predict patient responses to treatment and monitor therapeutic outcomes. Current nanomedicines are being designed with careful consideration of factors such as the choice of carrier (e.g., biocompatibility, controlled cargo release) and targeting moiety. The unique properties of nanobodies make them an effective engineering tool to target biological molecules with high affinity and specificity. In this review, we focus on the latest applications of nanobodies for targeting various components of the tumor microenvironment for diagnostic and therapeutic purposes. We also explore the main types of nanoparticles used as a carrier for cancer immunotherapies, as well as the strategies for formulating nanoparticle-nanobody conjugates. Finally, we highlight how nanobody-nanoparticle formulations can enhance current nanomedicines.
Collapse
Affiliation(s)
- Liudmyla Maksymova
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yannick A Pilger
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Faculty of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg, Röntgenring 11, Würzburg, 97070, Germany.
| | - Jo A Van Ginderachter
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology (BCIM), Vrije Universiteit Brussel, Pleinlaan 2, Brussels, B-1050, Belgium.
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
4
|
Yuen D, Feeney OM, Noi L, Shengule S, McLeod VM, Reitano P, Tsegay S, Hufton R, Houston ZH, Fletcher NL, Humphries J, Thurecht KJ, Cullinane C, Owen DJ, Porter CJH, Johnston APR. Nanobody-Mediated Cellular Uptake Maximizes the Potency of Polylysine Dendrimers While Preserving Solid Tumor Penetration. ACS NANO 2025; 19:6044-6057. [PMID: 39910852 DOI: 10.1021/acsnano.4c10851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Dendrimers are branched macromolecular structures that are useful nanocarriers for small-molecule drugs, such as cancer therapeutics. Their small size permits penetration into solid tumors, coupled with functionalization with a low-fouling PEG coating that minimizes transient cellular interactions and enhances plasma circulation time. While PEGylated dendrimers show significant promise as anticancer therapeutics, there is potential to increase tumor cell specificity and drive uptake of drugs into cells by conjugating cell-targeting ligands onto the dendrimers. To achieve this, we used an expanded genetic code and bio-orthogonal click chemistry to functionalize monomethyl auristatin E (MMAE)-loaded PEGylated dendrimers with a single tumor cell-targeting nanobody per dendrimer. The uniform addition of a single nanobody ligand facilitated greater intracellular uptake of the drug payload into HER2-positive target cells, while preserving the desirable circulatory characteristics of dendrimers. While the nanobody-dendrimer conjugates show similar levels of tumor infiltration over 24 h compared to unmodified dendrimers, the targeted dendrimers had significantly greater inhibition of tumor growth and long-term retention in the tumors. Our results highlight that biodistribution studies alone are poor predictors of therapeutic performance. The controlled conjugation strategy presented here preserves the size advantage and tissue penetration of dendrimers while maximizing targeted cellular uptake and potency in difficult-to-access solid tumor tissue.
Collapse
Affiliation(s)
- Daniel Yuen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Leo Noi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | | | | | | | - Sammi Tsegay
- Starpharma Ltd., Abbotsford, Victoria 3067, Australia
| | | | - Zachary H Houston
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia, Queensland 4072, Australia
| | - James Humphries
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging, ARC Training Centre for Innovation in Biomedical Imaging Technologies, University of Queensland, St Lucia, Queensland 4072, Australia
| | | | - David J Owen
- Starpharma Ltd., Abbotsford, Victoria 3067, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
5
|
Wu J, Zheng Y, Zhang LN, Gu CL, Chen WL, Chang MQ. Advanced nanomedicines and immunotherapeutics to treat respiratory diseases especially COVID-19 induced thrombosis. World J Clin Cases 2024; 12:2704-2712. [PMID: 38899301 PMCID: PMC11185334 DOI: 10.12998/wjcc.v12.i16.2704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/06/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
Immunotherapy and associated immune regulation strategies gained huge attraction in order to be utilized for treatment and prevention of respiratory diseases. Engineering specifically nanomedicines can be used to regulate host immunity in lungs in the case of respiratory diseases including coronavirus disease 2019 (COVID-19) infection. COVID-19 causes pulmonary embolisms, thus new therapeutic options are required to target thrombosis, as conventional treatment options are either not effective due to the complexity of the immune-thrombosis pathophysiology. In this review, we discuss regulation of immune response in respiratory diseases especially COVID-19. We further discuss thrombosis and provide an overview of some antithrombotic nanoparticles, which can be used to develop nanomedicine against thrombo-inflammation induced by COVID-19 and other respiratory infectious diseases. We also elaborate the importance of immunomodulatory nanomedicines that can block pro-inflammatory signalling pathways, and thus can be recommended to treat respiratory infectious diseases.
Collapse
Affiliation(s)
- Jie Wu
- Department of Respiratory and Oncology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| | - Ying Zheng
- Department of Respiratory and Oncology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| | - Li-Na Zhang
- Department of Respiratory and Oncology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| | - Cai-Li Gu
- Department of Respiratory and Oncology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| | - Wang-Li Chen
- Department of Respiratory and Oncology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| | - Min-Qiang Chang
- Department of Otorhinolaryngology, 72nd Group Army Hospital of PLA, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|
6
|
Morla-Folch J, Ranzenigo A, Fayad ZA, Teunissen AJP. Nanotherapeutic Heterogeneity: Sources, Effects, and Solutions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307502. [PMID: 38050951 PMCID: PMC11045328 DOI: 10.1002/smll.202307502] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Nanomaterials have revolutionized medicine by enabling control over drugs' pharmacokinetics, biodistribution, and biocompatibility. However, most nanotherapeutic batches are highly heterogeneous, meaning they comprise nanoparticles that vary in size, shape, charge, composition, and ligand functionalization. Similarly, individual nanotherapeutics often have heterogeneously distributed components, ligands, and charges. This review discusses nanotherapeutic heterogeneity's sources and effects on experimental readouts and therapeutic efficacy. Among other topics, it demonstrates that heterogeneity exists in nearly all nanotherapeutic types, examines how nanotherapeutic heterogeneity arises, and discusses how heterogeneity impacts nanomaterials' in vitro and in vivo behavior. How nanotherapeutic heterogeneity skews experimental readouts and complicates their optimization and clinical translation is also shown. Lastly, strategies for limiting nanotherapeutic heterogeneity are reviewed and recommendations for developing more reproducible and effective nanotherapeutics provided.
Collapse
Affiliation(s)
- Judit Morla-Folch
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anna Ranzenigo
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zahi Adel Fayad
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Abraham Jozef Petrus Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
7
|
Finnegan JR, FitzGerald LI, Chen MZ, Warne NM, Yuen D, Davis TP, Johnston APR, Kempe K. Length-Dependent Cellular Internalization of Nanobody-Functionalized Poly(2-oxazoline) Nanorods. NANO LETTERS 2024; 24:89-96. [PMID: 37939013 DOI: 10.1021/acs.nanolett.3c03342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
The ability to target specific tissues and to be internalized by cells is critical for successful nanoparticle-based targeted drug delivery. Here, we combined "stealthy" rod-shaped poly(2-oxazoline) (POx) nanoparticles of different lengths with a cancer marker targeting nanobody and a fluorescent cell internalization sensor via a heat-induced living crystallization-driven self-assembly (CDSA) strategy. A significant increase in association and uptake driven by nanobody-receptor interactions was observed alongside nanorod-length-dependent kinetics. Importantly, the incorporation of the internalization sensor allowed for quantitative differentiation between cell surface association and internalization of the targeted nanorods, revealing unprecedented length-dependent cellular interactions of CDSA nanorods. This study highlights the modularity and versatility of the heat-induced CDSA process and further demonstrates the potential of POx nanorods as a modular nanomedicine platform.
Collapse
Affiliation(s)
- John R Finnegan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Laura I FitzGerald
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Moore Zhe Chen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Nicole M Warne
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Daniel Yuen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
8
|
Kim S, Bae Y, Park SH, Chen N, Eom S, Kang S, Park J. Compact and modular bioprobe: Integrating SpyCatcher/SpyTag recombinant proteins with zwitterionic polymer-coated quantum dots. J Colloid Interface Sci 2023; 652:184-194. [PMID: 37595436 DOI: 10.1016/j.jcis.2023.08.016] [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: 01/24/2023] [Revised: 06/05/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023]
Abstract
The development of quantum dot (QD)-based modular bioprobe that has a compact size and enable a facile conjugation of various biofunctional groups is in high demand. To address this, we surface engineered QDs with zwitterion polymer ligands to have an inherent compact size and derivatized them sequentially with the recombinant proteins SpyCatcher/SpyTag (SC/ST) to use their protein ligation system. SC/ST spontaneously form one complex through the isopeptide bond between them. SC-conjugated QDs (QD-SC) were used as base building blocks. Then, ST-biomolecules were added for modular biofunctionalization. We synthesized compact sized (∼15 nm) QD-SC-ST-affibody (antibody-mimicking small protein for tumor detection) conjugates, which showed successful cell-receptor targeting. The target cell-receptor could be easily tuned by changing the type of ST-affibody. We also demonstrated that anti-human-chorionic-gonadotropin mouse IgG1 antibodies can be labeled on the QD surface by mixing QD-SC with the ST-MG1Nb (mouse-IgG1-specific protein). The immunoassay performance of the antibody-labeled QDs was evaluated using a pregnancy test kit, displaying equivalent detection sensitivity to a commercially available kit. This study proposed an innovative strategy for QD biofunctionalization in a modular manner, which can be expanded to a diverse range of colloidal particle derivatization.
Collapse
Affiliation(s)
- Sunghwan Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Yoonji Bae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sung Han Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ning Chen
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Soomin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| | - Jongnam Park
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
9
|
Choi Y, Cho BK, Seok SH, Kim C, Ryu JH, Kwon IC. Controlled spatial characteristics of ligands on nanoparticles: Determinant of cellular functions. J Control Release 2023; 360:672-686. [PMID: 37437847 DOI: 10.1016/j.jconrel.2023.07.020] [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: 03/16/2023] [Revised: 06/27/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Interactions of various ligands and receptors have been extensively investigated because they regulate a series of signal transduction leading to various functional cellular outcomes. The receptors on cell membrane recognize their specific ligands, resulting in specific binding between ligands and receptors. Accumulating evidence reveals that the receptors recognize the difference on the spatial characteristics of ligands as well as the types of ligands. Thus, control on spatial characteristics of multiple ligands presented on therapeutic nanoparticles is believed to impact the cellular functions. Specifically, the localized and multivalent distribution of ligands on nanoparticles can induce receptor oligomerization and receptor clustering, controlling intensity or direction of signal transduction cascades. Here, we will introduce recent studies on the use of material-based nanotechnology to control spatial characteristics of ligands and their effect on cellular functions. These therapeutic nanoparticles with controlled spatial characteristics of ligands may be a promising strategy for maximized therapeutic outcome.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Bo Kyung Cho
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Chansoo Kim
- Computational Science Centre & ASSIST, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; AI-Robot Department, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
10
|
Anzengruber M, Nepustil LM, Kurtaj F, Tahir A, Skoll K, Sami H, Wirth M, Gabor F. A Versatile Brij-Linker for One-Step Preparation of Targeted Nanoparticles. Pharmaceutics 2023; 15:pharmaceutics15051403. [PMID: 37242645 DOI: 10.3390/pharmaceutics15051403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Background: Most frequently the functionalization of nanoparticles is hampered by time-consuming, sometimes harsh conjugation and purification procedures causing premature drug release and/or degradation. A strategy to circumvent multi-step protocols is to synthesize building blocks with different functionalities and to use mixtures thereof for nanoparticle preparation in one step. Methods: BrijS20 was converted into an amine derivative via a carbamate linkage. The Brij-amine readily reacts with pre-activated carboxyl-containing ligands such as folic acid. The structures of the building blocks were confirmed by different spectroscopic methods and their utility was assessed by one-step preparation and characterization of nanoparticles applying PLGA as a matrix polymer. Results: Nanoparticles were about 200 nm in diameter independent of the composition. Experiments with human folate expressing single cells and monolayer revealed that the nanoparticle building block Brij mediates a "stealth" effect and the Brij-amine-folate a "targeting" effect. As compared to plain nanoparticles, the stealth effect decreased the cell interaction by 13%, but the targeting effect increased the cell interaction by 45% in the monolayer. Moreover, the targeting ligand density and thus the cell association of the nanoparticles is easily fine-tuned by selection of the initial ratio of the building blocks. Conclusions: This strategy might be a first step towards the one-step preparation of nanoparticles with tailored functionalities. Relying on a non-ionic surfactant is a versatile approach as it might be extended to other hydrophobic matrix polymers and promising targeting ligands from the biotech pipeline.
Collapse
Affiliation(s)
- Maria Anzengruber
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Lisa Marie Nepustil
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Fatlinda Kurtaj
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Ammar Tahir
- Division of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Katharina Skoll
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Haider Sami
- Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Michael Wirth
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Franz Gabor
- Division of Pharmaceutical Technology and Biopharmaceutics, Faculty of Life Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| |
Collapse
|
11
|
Obeng EM, Steer DL, Fulcher AJ, Wagstaff KM. Sortase A transpeptidation produces seamless, unbranched biotinylated nanobodies for multivalent and multifunctional applications. NANOSCALE ADVANCES 2023; 5:2251-2260. [PMID: 37056610 PMCID: PMC10089078 DOI: 10.1039/d3na00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/15/2023] [Indexed: 06/19/2023]
Abstract
Exploitation of the biotin-streptavidin interaction for advanced protein engineering is used in many bio-nanotechnology applications. As such, researchers have used diverse techniques involving chemical and enzyme reactions to conjugate biotin to biomolecules of interest for subsequent docking onto streptavidin-associated molecules. Unfortunately, the biotin-streptavidin interaction is susceptible to steric hindrance and conformational malformation, leading to random orientations that ultimately impair the function of the displayed biomolecule. To minimize steric conflicts, we employ sortase A transpeptidation to produce quantitative, seamless, and unbranched nanobody-biotin conjugates for efficient display on streptavidin-associated nanoparticles. We further characterize the protein-nanoparticle complex and demonstrate its usefulness in optical microscopy and multivalent severe acute respiratory syndrome coronavirus (SARS-CoV-2) antigen interaction. The approach reported here provides a template for making novel multivalent and multifunctional protein complexes for avidity-inspired technologies.
Collapse
Affiliation(s)
- Eugene M Obeng
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
| | - David L Steer
- Monash Proteomics and Metabolomics Facility, Monash University Clayton VIC 3800 Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University Clayton VIC 3800 Australia
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University Clayton VIC 3800 Australia
| |
Collapse
|
12
|
Lee JH, Chapman DV, Saltzman WM. Nanoparticle Targeting with Antibodies in the Central Nervous System. BME FRONTIERS 2023; 4:0012. [PMID: 37849659 PMCID: PMC10085254 DOI: 10.34133/bmef.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/19/2023] [Indexed: 10/19/2023] Open
Abstract
Treatments for disease in the central nervous system (CNS) are limited because of difficulties in agent penetration through the blood-brain barrier, achieving optimal dosing, and mitigating off-target effects. The prospect of precision medicine in CNS treatment suggests an opportunity for therapeutic nanotechnology, which offers tunability and adaptability to address specific diseases as well as targetability when combined with antibodies (Abs). Here, we review the strategies to attach Abs to nanoparticles (NPs), including conventional approaches of chemisorption and physisorption as well as attempts to combine irreversible Ab immobilization with controlled orientation. We also summarize trends that have been observed through studies of systemically delivered Ab-NP conjugates in animals. Finally, we discuss the future outlook for Ab-NPs to deliver therapeutics into the CNS.
Collapse
Affiliation(s)
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
13
|
Russell P, Esser L, Hagemeyer CE, Voelcker NH. The potential impact of nanomedicine on COVID-19-induced thrombosis. NATURE NANOTECHNOLOGY 2023; 18:11-22. [PMID: 36536042 DOI: 10.1038/s41565-022-01270-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/12/2022] [Indexed: 06/17/2023]
Abstract
Extensive reports of pulmonary embolisms, ischaemic stroke and myocardial infarctions caused by coronavirus disease 2019 (COVID-19), as well as a significantly increased long-term risk of cardiovascular diseases in COVID-19 survivors, have highlighted severe deficiencies in our understanding of thromboinflammation and the need for new therapeutic options. Due to the complexity of the immunothrombosis pathophysiology, the efficacy of treatment with conventional anti-thrombotic medication is questioned. Thrombolytics do appear efficacious, but are hindered by severe bleeding risks, limiting their use. Nanomedicine can have profound impact in this context, protecting delicate (bio)pharmaceuticals from degradation en route and enabling delivery in a targeted and on demand manner. We provide an overview of the most promising nanocarrier systems and design strategies that may be adapted to develop nanomedicine for COVID-19-induced thromboinflammation, including dual-therapeutic approaches with antiviral and immunosuppressants. Resultant targeted and side-effect-free treatment may aid greatly in the fight against the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
- Peije Russell
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
14
|
Functionalization of Nanosystems in Cancer Treatment. Cancer Nanotechnol 2023. [DOI: 10.1007/978-3-031-17831-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
15
|
Mesquita BS, Fens MHAM, Di Maggio A, Bosman EDC, Hennink WE, Heger M, Oliveira S. The Impact of Nanobody Density on the Targeting Efficiency of PEGylated Liposomes. Int J Mol Sci 2022; 23:ijms232314974. [PMID: 36499301 PMCID: PMC9741042 DOI: 10.3390/ijms232314974] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Nanoparticles (NPs) are commonly modified with tumor-targeting moieties that recognize proteins overexpressed on the extracellular membrane to increase their specific interaction with target cells. Nanobodies (Nbs), the variable domain of heavy chain-only antibodies, are a robust targeting ligand due to their small size, superior stability, and strong binding affinity. For the clinical translation of targeted Nb-NPs, it is essential to understand how the number of Nbs per NP impacts the receptor recognition on cells. To study this, Nbs targeting the hepatocyte growth factor receptor (MET-Nbs) were conjugated to PEGylated liposomes at a density from 20 to 800 per liposome and their targeting efficiency was evaluated in vitro. MET-targeted liposomes (MET-TLs) associated more profoundly with MET-expressing cells than non-targeted liposomes (NTLs). MET-TLs with approximately 150-300 Nbs per liposome exhibited the highest association and specificity towards MET-expressing cells and retained their targeting capacity when pre-incubated with proteins from different sources. Furthermore, a MET-Nb density above 300 Nbs per liposome increased the interaction of MET-TLs with phagocytic cells by 2-fold in ex vivo human blood compared to NTLs. Overall, this study demonstrates that adjusting the MET-Nb density can increase the specificity of NPs towards their intended cellular target and reduce NP interaction with phagocytic cells.
Collapse
Affiliation(s)
- Bárbara S. Mesquita
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Marcel H. A. M. Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Alessia Di Maggio
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Esmeralda D. C. Bosman
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Wim E. Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Michal Heger
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing 314041, China
- Membrane Biochemistry and Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, 3584 CG Utrecht, The Netherlands
- Correspondence: (M.H.); (S.O.)
| | - Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Science Faculty, Utrecht University, 3584 CG Utrecht, The Netherlands
- Correspondence: (M.H.); (S.O.)
| |
Collapse
|
16
|
Ma X, Wei B, Wang E. Efficient incorporation of p-azido-l-phenylalanine into the protein using organic solvents. Protein Expr Purif 2022; 200:106158. [PMID: 36007861 DOI: 10.1016/j.pep.2022.106158] [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: 04/17/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022]
Abstract
Azide, the most used photo-crosslinking group, facilitates the analysis of protein structure and function. This group is particularly useful when photochemically label antibodies and examine protein-protein interactions. The use of the expanded genetic code technique allows the special labeling of the functional azide group in proteins by adding the unnatural amino acid (UAA), p-azido-l-phenylalanine (AzF), in response to the amber codon during translation. However, a low UAA uptake rate due to mass transfer resistance in the cell membrane may lead to the early termination of the full-length protein. This study reports a general method for the efficient in vivo incorporation of AzF into the target protein by improving cell permeability using organic solvents. As expected, the yield of the full-length protein was significantly increased, which indicated that the AzF uptake was greatly improved due to the addition of organic solvents. Our method can serve as a good reference for improving the genetic incorporation of other kinds of UAAs into proteins.
Collapse
Affiliation(s)
- Xiaofeng Ma
- Department of Gynecology and Obstetrics, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Bing Wei
- Department of Gynecology and Obstetrics, The Second Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Enlin Wang
- The College of Life Science, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
17
|
Nifontova G, Tsoi T, Karaulov A, Nabiev I, Sukhanova A. Structure-function relationships in polymeric multilayer capsules designed for cancer drug delivery. Biomater Sci 2022; 10:5092-5115. [PMID: 35894444 DOI: 10.1039/d2bm00829g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The targeted delivery of cancer drugs to tumor-specific molecular targets represents a major challenge in modern personalized cancer medicine. Engineering of micron and submicron polymeric multilayer capsules allows the obtaining of multifunctional theranostic systems serving as controllable stimulus-responsive tools with a high clinical potential to be used in cancer therapy and detection. The functionalities of such theranostic systems are determined by the design and structural properties of the capsules. This review (1) describes the current issues in designing cancer cell-targeting polymeric multilayer capsules, (2) analyzes the effects of the interactions of the capsules with the cellular and molecular constituents of biological fluids, and (3) presents the key structural parameters determining the effectiveness of capsule targeting. The influence of the morphological and physicochemical parameters and the origin of the structural components and surface ligands on the functional activity of polymeric multilayer capsules at the molecular, cellular, and whole-body levels are summarized. The basic structural and functional principles determining the future trends of theranostic capsule development are established and discussed.
Collapse
Affiliation(s)
- Galina Nifontova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France.
| | - Tatiana Tsoi
- National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia
| | - Alexander Karaulov
- Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Igor Nabiev
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France. .,National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409 Moscow, Russia.,Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Alyona Sukhanova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France.
| |
Collapse
|
18
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Henry SJW, Dejneka A, Stephanopoulos N, Lunov O. The interactions between DNA nanostructures and cells: A critical overview from a cell biology perspective. Acta Biomater 2022; 146:10-22. [PMID: 35523414 PMCID: PMC9590281 DOI: 10.1016/j.actbio.2022.04.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/18/2022]
Abstract
DNA nanotechnology has yielded remarkable advances in composite materials with diverse applications in biomedicine. The specificity and predictability of building 3D structures at the nanometer scale make DNA nanotechnology a promising tool for uses in biosensing, drug delivery, cell modulation, and bioimaging. However, for successful translation of DNA nanostructures to real-world applications, it is crucial to understand how they interact with living cells, and the consequences of such interactions. In this review, we summarize the current state of knowledge on the interactions of DNA nanostructures with cells. We identify key challenges, from a cell biology perspective, that influence progress towards the clinical translation of DNA nanostructures. We close by providing an outlook on what questions must be addressed to accelerate the clinical translation of DNA nanostructures. STATEMENT OF SIGNIFICANCE: Self-assembled DNA nanostructures (DNs) offers unique opportunities to overcome persistent challenges in the nanobiotechnology field. However, the interactions between engineered DNs and living cells are still not well defined. Critical systematization of current cellular models and biological responses triggered by DNs is a crucial foundation for the successful clinical translation of DNA nanostructures. Moreover, such an analysis will identify the pitfalls and challenges that are present in the field, and provide a basis for overcoming those challenges.
Collapse
Affiliation(s)
- Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 14021, Czech Republic
| | - Skylar J W Henry
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, United States; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ 85281, United States
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic
| | - Nicholas Stephanopoulos
- School of Molecular Sciences, Arizona State University, Tempe, AZ, 85281, United States; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ 85281, United States.
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| |
Collapse
|
19
|
Wu SY, Wu FG, Chen X. Antibody-Incorporated Nanomedicines for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109210. [PMID: 35142395 DOI: 10.1002/adma.202109210] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Antibody-based cancer therapy, one of the most significant therapeutic strategies, has achieved considerable success and progress over the past decades. Nevertheless, obstacles including limited tumor penetration, short circulation half-lives, undesired immunogenicity, and off-target side effects remain to be overcome for the antibody-based cancer treatment. Owing to the rapid development of nanotechnology, antibody-containing nanomedicines that have been extensively explored to overcome these obstacles have already demonstrated enhanced anticancer efficacy and clinical translation potential. This review intends to offer an overview of the advancements of antibody-incorporated nanoparticulate systems in cancer treatment, together with the nontrivial challenges faced by these next-generation nanomedicines. Diverse strategies of antibody immobilization, formats of antibodies, types of cancer-associated antigens, and anticancer mechanisms of antibody-containing nanomedicines are provided and discussed in this review, with an emphasis on the latest applications. The current limitations and future research directions on antibody-containing nanomedicines are also discussed from different perspectives to provide new insights into the construction of anticancer nanomedicines.
Collapse
Affiliation(s)
- Shun-Yu Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
20
|
Gao D, Cheng F, Wang X, Yang H, Liu C, Li C, Yang EM, Cheng G, He W. Developing G value as an indicator for assessing the molecular status of immobilized antibody. Colloids Surf B Biointerfaces 2022; 217:112593. [PMID: 35665639 DOI: 10.1016/j.colsurfb.2022.112593] [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: 02/26/2022] [Revised: 05/08/2022] [Accepted: 05/21/2022] [Indexed: 10/18/2022]
Abstract
Antibody-functionalized nanoparticles (Ab-NPs) are widely used in bioassays due to their excellent affinity, specificity toward antigen, and ease of operation. However, the uncontrollable molecular status of antibodies on NPs severely limits their applications. This work aims at developing a simple method to evaluate the antigen-binding activity of Ab-NPs using two parameters, i.e., antibody adsorption amount and antigen-binding strength. Herein, we proposed a mathematical expression, G, to quantitively describe the amount and strength of Ab-NPs. G value could be used to assess the antigen-binding performance of NPs influenced by surface and solution factors. Seven types of polymers with different surface properties, including four positively and three negatively charged polymer brushes, were grown from silica NPs via surface-initiated atom transfer radical polymerization (SI-ATRP). A pair of antigen and antibody, human chorionic gonadotropin (hCG) and anti-hCG, were selected to screen the antibody immobilization property of polymer brushes. Among them, the G values of 2 polymer-NPs with opposite charges reached maximum, resulting in low detection limits for hCG, where pDMAEA-NP and pMMA-NP represent Poly[N,N-(dimethylamino)ethyl acrylate]-NP and poly(methyl methacrylate)-NP, respectively. The G value of Ab-NPs makes it feasible to estimate the molecular status of the adsorbed antibodies on surfaces, thus showing great potential for in vitro biosensing and bioseparation.
Collapse
Affiliation(s)
- Dongdong Gao
- Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China; Department of Polymer Science & Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China
| | - Fang Cheng
- Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China; Department of Polymer Science & Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China; Ningbo Institute of Dalian University of Technology, Ningbo 315211, China.
| | - Xinglong Wang
- Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China
| | - Heqing Yang
- Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China
| | - Chong Liu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China; Department of Polymer Science & Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China
| | - Chunmei Li
- Tsinglan School, Songshan Lake, Dongguan 523000, China
| | | | - Gang Cheng
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Wei He
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116023, China; Department of Polymer Science & Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116023, China
| |
Collapse
|
21
|
Van Fossen EM, Bednar RM, Jana S, Franklin R, Beckman J, Karplus PA, Mehl RA. Nanobody assemblies with fully flexible topology enabled by genetically encoded tetrazine amino acids. SCIENCE ADVANCES 2022; 8:eabm6909. [PMID: 35522749 PMCID: PMC9075797 DOI: 10.1126/sciadv.abm6909] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/23/2022] [Indexed: 06/14/2023]
Abstract
Assembling nanobodies (Nbs) into polyvalent multimers is a powerful strategy for improving the effectiveness of Nb-based therapeutics and biotechnological tools. However, generally effective approaches to Nb assembly are currently restricted to the amino or carboxyl termini, greatly limiting the diversity of Nb multimer topologies that can be produced. Here, we show that reactive tetrazine groups-site-specifically inserted by genetic code expansion at Nb surface sites-are compatible with Nb folding and function, enabling Nb assembly at any desired point. Using two anti-SARS-CoV-2 Nbs with viral neutralization ability, we created Nb homo- and heterodimers with improved properties compared with conventionally linked Nb homodimers, which, in the case of our tetrazine-conjugated trimer, translated into enhanced viral neutralization. Thus, this tetrazine-based approach is a generally applicable strategy that greatly increases the accessible range of Nb assembly topologies, and thereby adds the optimization of topology as an effective avenue to generate Nb assemblies with improved efficacy.
Collapse
Affiliation(s)
- Elise M. Van Fossen
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Riley M. Bednar
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Subhashis Jana
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Rachel Franklin
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Joseph Beckman
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
- e-MSion, Inc., 2121 NE Jack London Drive, Corvallis, OR 97330, USA
| | - P. Andrew Karplus
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Ryan A. Mehl
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
22
|
Gao S, Guisán JM, Rocha-Martin J. Oriented immobilization of antibodies onto sensing platforms - A critical review. Anal Chim Acta 2022; 1189:338907. [PMID: 34815045 DOI: 10.1016/j.aca.2021.338907] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/08/2021] [Accepted: 07/31/2021] [Indexed: 12/26/2022]
Abstract
The immunosensor has been proven a versatile tool to detect various analytes, such as food contaminants, pathogenic bacteria, antibiotics and biomarkers related to cancer. To fabricate robust and reproducible immunosensors with high sensitivity, the covalent immobilization of immunoglobulins (IgGs) in a site-specific manner contributes to better performance. Instead of the random IgG orientations result from the direct yet non-selective immobilization techniques, this review for the first time introduces the advances of stepwise yet site-selective conjugation strategies to give better biosensing efficiency. Noncovalently adsorbing IgGs is the first but decisive step to interact specifically with the Fc fragment, then following covalent conjugate can fix this uniform and antigens-favorable orientation irreversibly. In this review, we first categorized this stepwise strategy into two parts based on the different noncovalent interactions, namely adhesive layer-mediated interaction onto homofunctional support and layer-free interaction onto heterofunctional support (which displays several different functionalities on its surface that are capable to interact with IgGs). Further, the influence of ligands characteristics (synthesis strategies, spacer requirements and matrices selection) on the heterofunctional support has also been discussed. Finally, conclusions and future perspectives for the real-world application of stepwise covalent conjugation are discussed. This review provides more insights into the fabrication of high-efficiency immunosensor, and special attention has been devoted to the well-orientation of full-length IgGs onto the sensing platform.
Collapse
Affiliation(s)
- Shipeng Gao
- Department of Biocatalysis, Institute of Catalysis and Petrochemistry (ICP) CSIC, Campus UAM, Cantoblanco, 28049, Madrid, Spain
| | - José M Guisán
- Department of Biocatalysis, Institute of Catalysis and Petrochemistry (ICP) CSIC, Campus UAM, Cantoblanco, 28049, Madrid, Spain.
| | - Javier Rocha-Martin
- Department of Biocatalysis, Institute of Catalysis and Petrochemistry (ICP) CSIC, Campus UAM, Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
23
|
Liu T, Huang X, Zhao L, Xiao Z, Li Z, Xin Y, Yang S, Guo D, Zhao W, Mi Y, Li H. Distinguishable Targeting of Non-Small Cell Lung Cancer Using Hyaluronan Functionalized Platinum Nanoclusters and Their Inhibition Behaviors of Proliferation, Invasion, Migration. ChemistryOpen 2021; 10:882-888. [PMID: 34363352 PMCID: PMC8409085 DOI: 10.1002/open.202100070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/04/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and most cancer patients receiving conventional chemotherapy suffer from severe side effects due to the non-selective effects of chemotherapeutic drugs on normal cells. Targeted nanomaterials can obtain excellent accumulation at the tumor site through their active or passive targeting mechanisms, thereby reducing the toxicity of the drugs in various ways. In this study, hyaluronic acid (HA) which could specifically bind to CD44 on the surface of tumor cells, was used to modify amine-caged platinum nanoclusters (Pt NCs-NH2 ) to obtain targeting HA-Pt NCs-NH2 . Based on the differential expression of CD44 on the surface of three lung cells (non-small cell lung cancer cell H1299, small cell lung cancer cell H446, and embryonic lung fibroblast HFL1), HA-Pt NCs-NH2 can differentially enter the three cells and achieve their targeting of non-small cell lung cancer cell (NSCLC) cells. Pt NCs significantly inhibited the proliferation, migration and invasion of NSCLC cells and induced their apoptosis in comparison of classical cisplatin and carboplatin, showing a bright future in early diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Ting Liu
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Xin Huang
- School of TextilesZhongyuan University of TechnologyNo. 41 Zhongyuan Road (M)Zhengzhou450007China
| | - Lingyun Zhao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Zhongqing Xiao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Zengbei Li
- School of TextilesZhongyuan University of TechnologyNo. 41 Zhongyuan Road (M)Zhengzhou450007China
| | - Yi Xin
- Intensive Care UnitZhengzhou Orthopedics HospitalNo. 56 Longhai RoadZhengzhou450052China
| | - Shanshan Yang
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Di Guo
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Wenfei Zhao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI CancerMarshall Medical Research CentreThe Fifth Affiliated Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Hongyun Li
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| |
Collapse
|
24
|
Cremers GAO, Rosier BJHM, Meijs A, Tito NB, van Duijnhoven SMJ, van Eenennaam H, Albertazzi L, de Greef TFA. Determinants of Ligand-Functionalized DNA Nanostructure-Cell Interactions. J Am Chem Soc 2021; 143:10131-10142. [PMID: 34180666 PMCID: PMC8283757 DOI: 10.1021/jacs.1c02298] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Synthesis of ligand-functionalized
nanomaterials with control over
size, shape, and ligand orientation facilitates the design of targeted
nanomedicines for therapeutic purposes. DNA nanotechnology has emerged
as a powerful tool to rationally construct two- and three-dimensional
nanostructures, enabling site-specific incorporation of protein ligands
with control over stoichiometry and orientation. To efficiently target
cell surface receptors, exploration of the parameters that modulate
cellular accessibility of these nanostructures is essential. In this
study, we systematically investigate tunable design parameters of
antibody-functionalized DNA nanostructures binding to therapeutically
relevant receptors, including the programmed cell death protein 1,
the epidermal growth factor receptor, and the human epidermal growth
factor receptor 2. We show that, although the native affinity of antibody-functionalized
DNA nanostructures remains unaltered, the absolute number of bound
surface receptors is lower compared to soluble antibodies due to receptor
accessibility by the nanostructure. We explore structural determinants
of this phenomenon to improve efficiency, revealing that receptor
binding is mainly governed by nanostructure size and DNA handle location.
The obtained results provide key insights in the ability of ligand-functionalized
DNA nanostructures to bind surface receptors and yields design rules
for optimal cellular targeting.
Collapse
Affiliation(s)
- Glenn A O Cremers
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Computational Biology Group, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Bas J H M Rosier
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Computational Biology Group, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ab Meijs
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Computational Biology Group, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Nicholas B Tito
- Electric Ant Lab, Science Park 106, 1098 XG Amsterdam, The Netherlands
| | | | - Hans van Eenennaam
- Aduro Biotech Europe B.V., Kloosterstraat 9, 5349 AB Oss, The Netherlands
| | - Lorenzo Albertazzi
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Molecular Biosensing for Medical Diagnostics, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Tom F A de Greef
- Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Computational Biology Group, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands.,Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ, Nijmegen, The Netherlands.,Center for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
25
|
Huang J, Yuen D, Mintern JD, Johnston APR. Opportunities for innovation: Building on the success of lipid nanoparticle vaccines. Curr Opin Colloid Interface Sci 2021; 55:101468. [PMID: 34093062 PMCID: PMC8164502 DOI: 10.1016/j.cocis.2021.101468] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lipid nanoparticle (LNP) formulations of messenger RNA (mRNA) have demonstrated high efficacy as vaccines against SARS-CoV-2. The success of these nanoformulations underscores the potential of LNPs as a delivery system for next-generation biological therapies. In this article, we highlight the key considerations necessary for engineering LNPs as a vaccine delivery system and explore areas for further optimisation. There remain opportunities to improve the protection of mRNA, optimise cytosolic delivery, target specific cells, minimise adverse side-effects and control the release of RNA from the particle. The modular nature of LNP formulations and the flexibility of mRNA as a payload provide many pathways to implement these strategies. Innovation in LNP vaccines is likely to accelerate with increased enthusiasm following recent successes; however, any advances will have implications for a broad range of therapeutic applications beyond vaccination such as gene therapy.
Collapse
Affiliation(s)
- Jessica Huang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville VIC 3052, Australia
| | - Daniel Yuen
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville VIC 3052, Australia
| | - Justine D Mintern
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, 30 Flemington Rd, Parkville, Victoria 3010, Australia
| | - Angus P R Johnston
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville VIC 3052, Australia
| |
Collapse
|
26
|
Lamoot A, Uvyn A, Kasmi S, De Geest BG. Covalent Cell Surface Conjugation of Nanoparticles by a Combination of Metabolic Labeling and Click Chemistry. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202015625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
| | - Annemiek Uvyn
- Department of Pharmaceutics Ghent University Ghent Belgium
| | - Sabah Kasmi
- Department of Pharmaceutics Ghent University Ghent Belgium
| | | |
Collapse
|
27
|
Lamoot A, Uvyn A, Kasmi S, De Geest BG. Covalent Cell Surface Conjugation of Nanoparticles by a Combination of Metabolic Labeling and Click Chemistry. Angew Chem Int Ed Engl 2021; 60:6320-6325. [PMID: 33368900 DOI: 10.1002/anie.202015625] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 01/23/2023]
Abstract
Conjugation of nanoparticles (NP) to the surface of living cells is of interest in the context of exploiting the tissue homing properties of ex vivo engineered T cells for tumor-targeted delivery of drugs loaded into NP. Cell surface conjugation requires either a covalent or non-covalent reaction. Non-covalent conjugation with ligand-decorated NP (LNP) is challenging and involves a dynamic equilibrium between the bound and unbound state. Covalent NP conjugation results in a permanently bound state of NP, but the current routes for cell surface conjugation face slow reaction kinetics and random conjugation to proteins in the glycocalyx. To address the unmet need for alternative bioorthogonal strategies that allow for efficient covalent cell surface conjugation, we developed a 2-step click conjugation sequence in which cells are first metabolically labeled with azides followed by reaction with sulfo-6-methyl-tetrazine-dibenzyl cyclooctyne (Tz-DBCO) by SPAAC, and subsequent IEDDA with trans-cyclooctene (TCO) functionalized NP. In contrast to using only metabolic azide labeling and subsequent conjugation of DBCO-NP, our 2-step method yields a highly specific cell surface conjugation of LNP, with very low non-specific background binding.
Collapse
Affiliation(s)
| | - Annemiek Uvyn
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Sabah Kasmi
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | | |
Collapse
|
28
|
Tieu T, Wojnilowicz M, Huda P, Thurecht KJ, Thissen H, Voelcker NH, Cifuentes-Rius A. Nanobody-displaying porous silicon nanoparticles for the co-delivery of siRNA and doxorubicin. Biomater Sci 2021; 9:133-147. [DOI: 10.1039/d0bm01335h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Targeted delivery of chemotherapeutics to cancer cells has the potential to yield high drug concentrations in cancer cells while minimizing any unwanted side effects.
Collapse
Affiliation(s)
- Terence Tieu
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Parkville
- Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing
| | - Marcin Wojnilowicz
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing
- Clayton
- Australia
| | - Pie Huda
- Centre for Advanced Imaging
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology
- University of Queensland
- Brisbane
| | - Kristofer J. Thurecht
- Centre for Advanced Imaging
- Australian Institute for Bioengineering and Nanotechnology (AIBN)
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology
- University of Queensland
- Brisbane
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing
- Clayton
- Australia
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Parkville
- Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing
| | - Anna Cifuentes-Rius
- Monash Institute of Pharmaceutical Sciences
- Monash University
- Parkville
- Australia
| |
Collapse
|
29
|
Gettemans J, De Dobbelaer B. Transforming nanobodies into high-precision tools for protein function analysis. Am J Physiol Cell Physiol 2020; 320:C195-C215. [PMID: 33264078 DOI: 10.1152/ajpcell.00435.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Single-domain antibodies, derived from camelid heavy antibodies (nanobodies) or shark variable new antigen receptors, have attracted increasing attention in recent years due to their extremely versatile nature and the opportunities they offer for downstream modification. Discovered more than three decades ago, these 120-amino acid (∼15-kDa) antibody fragments are known to bind their target with high specificity and affinity. Key features of nanobodies that make them very attractive include their single-domain nature, small size, and affordable high-level expression in prokaryotes, and their cDNAs are routinely obtained in the process of their isolation. This facilitates and stimulates new experimental approaches. Hence, it allows researchers to formulate new answers to complex biomedical questions. Through elementary PCR-based technologies and chemical modification strategies, their primary structure can be altered almost at leisure while retaining their specificity and biological activity, transforming them into highly tailored tools that meet the increasing demands of current-day biomedical research. In this review, various aspects of camelid nanobodies are expounded, including intracellular delivery in recombinant format for manipulation of, i.e., cytoplasmic targets, their derivatization to improve nanobody orientation as a capturing device, approaches to reversibly bind their target, their potential as protein-silencing devices in cells, the development of strategies to transfer nanobodies through the blood-brain barrier and their application in CAR-T experimentation. We also discuss some of their disadvantages and conclude with future prospects.
Collapse
Affiliation(s)
- Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Brian De Dobbelaer
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
30
|
Sivaram AJ, Wardiana A, Alcantara S, Sonderegger SE, Fletcher NL, Houston ZH, Howard CB, Mahler SM, Alexander C, Kent SJ, Bell CA, Thurecht KJ. Controlling the Biological Fate of Micellar Nanoparticles: Balancing Stealth and Targeting. ACS NANO 2020; 14:13739-13753. [PMID: 32936613 DOI: 10.1021/acsnano.0c06033] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Integrating nanomaterials with biological entities has led to the development of diagnostic tools and biotechnology-derived therapeutic products. However, to optimize the design of these hybrid bionanomaterials, it is essential to understand how controlling the biological interactions will influence desired outcomes. Ultimately, this knowledge will allow more rapid translation from the bench to the clinic. In this paper, we developed a micellar system that was assembled using modular antibody-polymer amphiphilic materials. The amphiphilic nature was established using either poly(ethylene glycol) (PEG) or a single-chain variable fragment (scFv) from an antibody as the hydrophile and a thermoresponsive polymer (poly(oligoethylene glycol) methyl ether methacrylate) as the hydrophobe. By varying the ratios of these components, a series of nanoparticles with different antibody content was self-assembled, where the surface presentation of targeting ligand was carefully controlled. In vitro and in vivo analysis of these systems identified a mismatch between the optimal targeting ligand density to achieve maximum cell association in vitro compared to tumor accumulation in vivo. For this system, we determined an optimum antibody density for both longer circulation and enhanced targeting to tumors that balanced stealthiness of the particle (to evade immune recognition as determined in both mouse models and in whole human blood) with enhanced accumulation achieved through receptor binding on tumor cells in solid tumors. This approach provides fundamental insights into how different antibody densities affect the interaction of designed nanoparticles with both target cells and immune cells, thereby offering a method to probe the intricate interplay between increased targeting efficiency and the subsequent immune response to nanoparticles.
Collapse
Affiliation(s)
- Amal J Sivaram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Andri Wardiana
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Sheilajen Alcantara
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria 3010, Australia
| | - Stefan E Sonderegger
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Nicholas L Fletcher
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Zachary H Houston
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Christopher B Howard
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Stephen M Mahler
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Training Centre for Biopharmaceutical Innovation, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Cameron Alexander
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Stephen J Kent
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria 3010, Australia
| | - Craig A Bell
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
31
|
Transient Multivalent Nanobody Targeting to CD206-Expressing Cells via PH-Degradable Nanogels. Cells 2020; 9:cells9102222. [PMID: 33019594 PMCID: PMC7600184 DOI: 10.3390/cells9102222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
To target nanomedicines to specific cells, especially of the immune system, nanobodies can be considered as an attractive tool, as they lack the Fc part as compared to traditional antibodies and, thus, prevent unfavorable Fc-receptor mediated mistargeting. For that purpose, we have site-specifically conjugated CD206/MMR-targeting nanobodies to three types of dye-labeled nanogel derivatives: non-degradable nanogels, acid-degradable nanogels (with ketal crosslinks), and single polymer chains (also obtained after nanogel degradation). All of them can be obtained from the same reactive ester precursor block copolymer. After incubation with naïve or MMR-expressing Chinese hamster ovary (CHO) cells, a nanobody mediated targeting and uptake could be confirmed for the nanobody-modified nanocarriers. Thereby, the intact nanogels that display nanobodies on their surface in a multivalent way showed a much stronger binding and uptake compared to the soluble polymers. Based on their acidic pH-responsive degradation potential, ketal crosslinked nanogels are capable of mediating a transient targeting that gets diminished upon unfolding into single polymer chains after endosomal acidification. Such control over particle integrity and targeting performance can be considered as highly attractive for safe and controllable immunodrug delivery purposes.
Collapse
|