1
|
Yu Y, Hu W, Xu Y, Xu HB, Gao J. Advancements in delivery Systems for Proteolysis-Targeting Chimeras (PROTACs): Overcoming challenges and expanding biomedical applications. J Control Release 2025; 382:113719. [PMID: 40268200 DOI: 10.1016/j.jconrel.2025.113719] [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: 01/24/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
PROTAC (Proteolysis-Targeting Chimera), an emerging drug development strategy based on small molecule technology, has garnered widespread attention due to its high efficiency, broad applicability, low resistance, and dosage advantages. However, PROTAC molecules still exhibit certain limitations that require urgent resolution. Although significant progress has been made in designing PROTACs that target various disease-related proteins, research on drug delivery systems (DDS) for PROTACs remains relatively limited. This review aims to explore the critical role of delivery system design in addressing the inherent challenges associated with PROTAC molecules from a novel perspective. Beginning with five major challenges-insufficient targeting, poor pharmacokinetic properties, low cell permeability, limited accessibility, and the Hook effect-this article introduces formulation strategies to mitigate these deficiencies. It discusses potential solutions through targeted modifications, nano-delivery systems, intelligent response systems, and membrane biomimetic technologies, among others. Furthermore, it elucidates the mechanisms and principles underlying these approaches and analyzes the advantages of various delivery strategies. The insights provided in this review offer insights for designing delivery systems tailored to PROTACs with diverse characteristics for different disease applications.
Collapse
Affiliation(s)
- Yawei Yu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weitong Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihua Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong-Bin Xu
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Zhe Jiang 315010, China.
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
2
|
Cristina V, Giulia R, Yin W, Mathilde H, Dipambita R, Torbjörn G, Riccardo C, Stefano R. Affibodies as valuable tool to prevent β 2m aggregation under lysosomal-like conditions. Biol Direct 2025; 20:67. [PMID: 40481566 PMCID: PMC12142832 DOI: 10.1186/s13062-025-00659-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Beta-2 microglobulin (β2m) is a small protein that forms the invariant subunit of the Major Histocompatibility Complex I. Monomeric β2m is stable under physiological conditions, however high local concentrations can induce misfolding, leading to amyloid deposition. This accumulation has been recently observed in the lysosomes of tumour-associated macrophages from patients affected by multiple myeloma. Such aggregation has been linked to inflammation and tumour progression. Stabilizing the native state of β2m could be the first step towards preventing this cancer-promoting process. To achieve this goal, the effect of affibody molecules, small and stress-resistant affinity proteins, was tested. Three affibodies molecules were selected against β2m. Affibody-β2m complex formation was initially assessed by size exclusion chromatography and subsequently confirmed by microscale thermophoresis and isothermal titration calorimetry. In parallel, in presence of one of the affibody (Zβ2m_01) a significant reduction in β2m aggregation was observed. The inhibition of amyloid formation was also confirmed by transmission electron microscopy. Taken together, these results indicate that Zβ2m_01 has the potential to act as β2m aggregation inhibitor under lysosomal-like pH values.
Collapse
Affiliation(s)
- Visentin Cristina
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.
| | - Rizzi Giulia
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Wen Yin
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hotot Mathilde
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Roy Dipambita
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Gräslund Torbjörn
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Capelli Riccardo
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy
| | - Ricagno Stefano
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
3
|
Rybarczyk A, Sultan T, Hussain N, Azam HMH, Rafique S, Zdarta J, Jesionowski T. Fusion of enzymatic proteins: Enhancing biological activities and facilitating biological modifications. Adv Colloid Interface Sci 2025; 340:103473. [PMID: 40086016 DOI: 10.1016/j.cis.2025.103473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
The fusion of enzymatic proteins represents a dynamic frontier in biotechnology and enzymatic engineering. This in-depth review looks at the many different ways that fusion proteins can be used, showing their importance in biosensing, gene therapy, targeted drug delivery, and biocatalysis. Fusion proteins have shown an astounding ability to improve and fine-tune biological functions by combining the most beneficial parts of different enzymes. Our first step is to explain how protein fusion increases biological functions. This will provide a broad picture of how this phenomenon has changed many fields. We dissect the intricate mechanisms through which fusion proteins orchestrate cellular processes, underscoring their potential to revolutionize the landscape of molecular biology. We also explore the complicated world of structural analysis and design strategies, stressing the importance of molecular insights for making the fusion protein approach work better. These insights broaden understanding of the underlying principles and illuminate the path toward unlocking untapped potential. The review also introduces cutting-edge techniques for constructing fusion protein libraries, such as DNA shuffling and phage display. These new methods allow scientists to build a molecular orchestra with an unprecedented level of accuracy, and thus use fusion proteins to their full potential in various situations. In conclusion, we provide a glimpse into the current challenges and prospects in fusion protein research, shedding light on recent advancements that promise to reshape the future of biotechnology. As we make this interesting journey through the field of enzymatic protein combination, it becomes clear that the fusion paradigm is about to start a new era of innovation that will push the limits of what is possible in biology and molecular engineering.
Collapse
Affiliation(s)
- Agnieszka Rybarczyk
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland
| | - Talha Sultan
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Hafiz Muhammad Husnain Azam
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany
| | - Safa Rafique
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Jakub Zdarta
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| | - Teofil Jesionowski
- Institute of Chemical Technology and Engineering, Faculty of Chemical Technology, Poznan University of Technology, Berdychowo 4, PL-60965 Poznan, Poland.
| |
Collapse
|
4
|
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
|
5
|
Karbasi F, Emamzadeh R, Nazari M. Development and evaluation of a new affiprobe that targets HER 3 positive cells using protein engineering. Biochimie 2025:S0300-9084(25)00098-7. [PMID: 40414339 DOI: 10.1016/j.biochi.2025.05.008] [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: 07/20/2024] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 05/27/2025]
Abstract
Engineered affiprobes that detect a specific target by generating a target-dependent signal, play an important role in biomedical sciences such as breast cancer monitoring. In this study, a new bioluminescence affiprobe consisting of an HER3-binding affibody and a bioluminescent protein, C-SRLc8, a thermostable variant of Renilla luciferase has been developed. The bioluminescent affibody, called C-SRLuc8‒Z08699 was expressed in the Escherichia coli and purified by affinity chromatography. The suitability of the new affiprobe to detected HER3 positive cells was evaluated. It was found that the new affiprobe allowed the detection of HER3 expressing cells with a detection limit of 14578 cells per 200 μl assay volume and a working range of 14000 to 50000 cells. The new affiprobe can be used for detecting, studying and monitoring the HER3-expressing cells using a luminometer in fast and inexpensive assays.
Collapse
Affiliation(s)
- Farnaz Karbasi
- Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Rahman Emamzadeh
- Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran.
| | - Mahboobeh Nazari
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| |
Collapse
|
6
|
Zhu H, Ding Y. Nanobodies: From Discovery to AI-Driven Design. BIOLOGY 2025; 14:547. [PMID: 40427736 PMCID: PMC12109276 DOI: 10.3390/biology14050547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
Nanobodies, derived from naturally occurring heavy-chain antibodies in camelids (VHHs) and sharks (VNARs), are unique single-domain antibodies that have garnered significant attention in therapeutic, diagnostic, and biotechnological applications due to their small size, stability, and high specificity. This review first traces the historical discovery of nanobodies, highlighting key milestones in their isolation, characterization, and therapeutic development. We then explore their structure-function relationship, emphasizing features like their single-domain architecture and long CDR3 loop that contribute to their binding versatility. Additionally, we examine the growing interest in multiepitope nanobodies, in which binding to different epitopes on the same antigen not only enhances neutralization and specificity but also allows these nanobodies to be used as controllable modules for precise antigen manipulation. This review also discusses the integration of AI in nanobody design and optimization, showcasing how machine learning and deep learning approaches are revolutionizing rational design, humanization, and affinity maturation processes. With continued advancements in structural biology and computational design, nanobodies are poised to play an increasingly vital role in addressing both existing and emerging biomedical challenges.
Collapse
Affiliation(s)
- Haoran Zhu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Shanghai 200433, China;
- Quzhou Fudan Institute, Quzhou 324002, China
| | - Yu Ding
- State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Fudan University, Shanghai 200433, China;
- Quzhou Fudan Institute, Quzhou 324002, China
| |
Collapse
|
7
|
Murugan D, Thirumalaiswamy HV, Murugesan V, Venkatesan J, Balachandran U, Lakshminarayanan K, Satpati D, Nikolić S, Rangasamy L. Unlocking the power of affibody conjugated radioactive metallopharmaceuticals for targeted cancer diagnosis and therapy. Pharmacol Ther 2025:108863. [PMID: 40294752 DOI: 10.1016/j.pharmthera.2025.108863] [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: 10/28/2024] [Revised: 03/02/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025]
Abstract
Cancer is the second-largest death-causing disease after cardiovascular diseases. Effective research on cancer diagnosis and subsequent elimination plays a vital role in reducing the cancer-related death toll. Radiotherapy is one of the best strategies that could kill masses of solid tumour tissues; however, the efficacy is limited due to the bystander effect. This issue could be solved by the emergence of targeted delivery of radiometallic complexes, enabling clinicians to monitor the tumour regions and effectively destroy the tumour. Affibody® molecules are a class of synthetic peptides known as antibody mimics having the binding sites of an antibody. The specificity of affibodies is found to be greater than that of antibodies due to their small size. This review intends to highlight the recent developments in the field of affibody-targeted radiometallopharmaceuticals. These approaches could be essential for early cancer detection, tumour staging, and monitoring the response to therapy and could produce better therapeutic outcomes. In an attempt to provide ideas and inspiration for the researchers to design affibody-conjugated radiopharmaceuticals that are clinically applicable, we have provided an in-depth exploration of the various types of affibody-conjugated radiopharmaceuticals that are currently in clinical trials and various other pre-clinically tested conjugates in this article. Only a few review reports on affibody-conjugated radiometallopharmaceuticals, typically focusing on a specific molecular target or radionuclides reported. In this review, we provide a comprehensive overview of most radiometals, such as 111In, 68Ga, 64Cu, 55Co, 57Co, 44Sc, 99mTc, 89Zr, 90Y, 211At, 188Re, and 177Lu, choice of chelators, and potential cancer-associated molecular targets such HER2, EGFR or HER1, HER3, IGF-1R, PDGFRβ, VEGFR2, PD-L1, CAIX, PD-L1, neonatal Fc receptor (FcRn) and B7-H3. This approach highlights the advancements made over the past twenty years in affibody conjugates for radio imaging and therapy in oncology.
Collapse
Affiliation(s)
- Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Harashkumar Vasanthakumari Thirumalaiswamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Vasanth Murugesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Janarthanan Venkatesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Unnikrishnan Balachandran
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Kalaiarasu Lakshminarayanan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India; School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Drishty Satpati
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, Maharashtra 400085, India; Homi Bhabha National Institute, Mumbai 400094, India
| | - Stefan Nikolić
- Innovative Centre of the Faculty of Chemistry Belgrade, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
8
|
Westerberg C, Mestre Borras A, Ståhl S, Löfblom J. Affibody-based HER2 prodrug shows conditional cytotoxic effect on HER2-positive cancer cells. Biochem Biophys Res Commun 2025; 758:151660. [PMID: 40117970 DOI: 10.1016/j.bbrc.2025.151660] [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: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Therapeutic affinity proteins offer a targeted mode of action due to their typically high affinity and specificity for disease-associated molecules. In cancer therapy, such target molecules are often overexpressed receptors on tumor cells. However, their presence in healthy tissues can lead to on-target, off-tumor toxicity, necessitating strategies to enhance tumor selectivity. Here, we present an affibody-based prodrug concept that exploits tumor-associated proteases for selective activation. As proof of concept, we designed, produced, and characterized HER2-specific prodrug candidates, each incorporating a distinct protease substrate for selective activation by tumor-associated proteases. Their activation by corresponding proteases and subsequent HER2 binding were assessed. The most promising prodrug candidate was conjugated to the cytotoxic agent DM1 and evaluated for cytotoxicity in HER2-positive cancer cells. The results demonstrated potent, HER2-dependent cell killing, with markedly reduced cytotoxicity in the absence of prodrug activation. These findings support the feasibility of affibody-based prodrugs as a strategy to enhance tumor selectivity and minimize off-tumor toxicity in targeted cancer therapy.
Collapse
Affiliation(s)
- Cornelia Westerberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anna Mestre Borras
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
9
|
Sadler C, Creamer A, Giang KA, Darmawan KK, Shamsabadi A, Richards DA, Nilvebrant J, Wojciechowski JP, Charchar P, Burdis R, Smith F, Yarovsky I, Nygren PÅ, Stevens MM. Adding a Twist to Lateral Flow Immunoassays: A Direct Replacement of Antibodies with Helical Affibodies, from Selection to Application. J Am Chem Soc 2025; 147:11925-11940. [PMID: 40135773 PMCID: PMC11987028 DOI: 10.1021/jacs.4c17452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Immunoreagents, most commonly antibodies, are integral components of lateral flow immunoassays. However, the use of antibodies comes with limitations, particularly relating to their reproducible production, and poor thermal and chemical stability. Here, we employ phage display to develop affibodies, a class of nonimmunoglobulin affinity proteins based on a small three-helix bundle scaffold, against SARS-CoV-2 Spike protein. Subsequently, we demonstrate the utility and viability of affibodies to directly replace antibodies in lateral flow immunoassays. In addition, we highlight several physiochemical advantages of affibodies, including their ability to withstand exposure to high temperature and humidity while maintaining superior performance compared to their antibody counterparts. Furthermore, we investigate the adsorption mechanism of affibodies to the surface of gold nanoparticle probes via a His6-tag, introduced to also facilitate recombinant purification. Through molecular dynamics simulations, we elucidate the structural and physical characteristics of affibody dimers which result in high-performing detection probes when immobilized on nanoparticle surfaces. This work demonstrates that affibodies can be used as direct replacements to antibodies in immunoassays and should be further considered as alternatives owing to their improved physiochemical properties and modular design.
Collapse
Affiliation(s)
- Christy
J. Sadler
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Department
of Physiology, Anatomy and Genetics, Department of Engineering Science,
Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K.
| | - Adam Creamer
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Department
of Physiology, Anatomy and Genetics, Department of Engineering Science,
Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K.
| | - Kim Anh Giang
- Department
of Protein Science, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm SE-114 21, Sweden
| | | | - André Shamsabadi
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Department
of Physiology, Anatomy and Genetics, Department of Engineering Science,
Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K.
| | - Daniel A. Richards
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Institute
for Chemical and Bioengineering, ETH Zurich, 8093 Zürich, Switzerland
| | - Johan Nilvebrant
- Department
of Protein Science, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm SE-114 21, Sweden
| | - Jonathan P. Wojciechowski
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Department
of Physiology, Anatomy and Genetics, Department of Engineering Science,
Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K.
| | - Patrick Charchar
- School
of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | - Ross Burdis
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
| | - Francesca Smith
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
| | - Irene Yarovsky
- School
of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | - Per-Åke Nygren
- Department
of Protein Science, AlbaNova University Center, KTH Royal Institute of Technology, Stockholm SE-114 21, Sweden
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, Institute of Biomedical Engineering Imperial College London, London SW7 2AZ, U.K.
- Department
of Physiology, Anatomy and Genetics, Department of Engineering Science,
Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, U.K.
| |
Collapse
|
10
|
Liu J, Liu Z, Hu J, Fan B, Zhang S, Chang K, Mao X, Huang G, Liu Z, Ma L. Anti-breast cancer activity of a novel genetically engineered fusion protein composed of HER2 affibody and proapoptotic peptide R8-KLA. Med Oncol 2025; 42:155. [PMID: 40205290 DOI: 10.1007/s12032-025-02707-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
HER2-positive breast cancer is an aggressive subtype with unfavorable prognoses. Although HER2-targeted agents represented by monoclonal antibodies have achieved remarkable success in the clinic, there are still a substantial number of patients with disease relapse. Recently, multifunctional fusion proteins obtained via genetic engineering technology have received much attention in targeted tumor therapy, especially in breast cancer. In this study, we genetically engineered a novel recombinant fusion protein, named HMK, which was designed as a bifunctional construct including the HER2-specific affibody ZHER2:342 for targeted receptor recognition, and a proapoptotic module featuring a cell-penetrating octa-arginine (R8) motif conjugated to an antimicrobial peptide KLA. High-purity HMK proteins were successfully obtained using E. coli expression system and Ni-Nitrilotriacetic acid affinity purification method. HMK exhibited higher cytotoxicity in HER2-positive breast cancer cells SK-BR-3 (IC50 of 8.36 ± 0.62 μM) compared to normal breast epithelial cells MCF-10A (IC50 of 32.40 ± 2.93 μM), demonstrating favorable selectivity. HMK induced apoptosis in SK-BR-3 cells via activating both endogenous and exogenous apoptotic pathways, as evidenced by the cleavage of Caspase 8, Caspase 9, Caspase 3, and PARP. Caspase inhibitor Z-VAD significantly reversed the function of HMK in SK-BR-3 cells, suggesting that caspase-dependent apoptosis was crucial for the anti-breast cancer activity of HMK. Our results suggested that HMK protein may have the potential to become a candidate molecule for HER2-positive breast cancer treatment.
Collapse
Affiliation(s)
- Jian Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zi Liu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| | - Junfeng Hu
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Binru Fan
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Shizhun Zhang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Kaili Chang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Xiuping Mao
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Guozheng Huang
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China
| | - Zhi Liu
- Department of Pathology, Ma'anshan Municipal People's Hospital, Ma'anshan, Anhui, 243000, China
| | - Liang Ma
- Department of Chemical Biology and Pharmaceutical Engineering, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, Anhui, 243002, China.
| |
Collapse
|
11
|
Gao B, Sabnis R, Kotnis S, Feliciano S, Poling K, Mei T, Feng M, Das JK, Song J, Sun Q. Modular Platform for Efficient Assembly of Multifunctional Antibodies Using Orthogonal Protein-Protein Interactions. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20685-20692. [PMID: 40159649 PMCID: PMC11986891 DOI: 10.1021/acsami.4c21958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Multifunctional antibodies, capable of simultaneously engaging multiple targets, are a unique class of antibodies that have sparked growing interest. Current approaches for making multifunctional antibodies, including chemical conjugation or genetic modifications, suffer from low product yield, complex structure design, and complicated manufacturing processes. In this study, we report a modular post-translational platform with highly specific protein-protein interactions for multifunctional antibody assembly and an elastin-like polypeptide (ELP) for easy purification. We generated and purified multifunctional antibodies with over 90% assembled scaffold and overall product purity. Additionally, we assembled antibodies with diverse applications, including detecting cancer, inhibiting cancer cell growth, and directing T cells to cancer cells for enhanced therapeutic efficacy. This platform offers high assembly efficiency, easy purification, and modularity for the redesign of antibody functions.
Collapse
Affiliation(s)
- Baizhen Gao
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Rushant Sabnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Siddhi Kotnis
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Sofia Feliciano
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Kyge Poling
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Tracy Mei
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Min Feng
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
| | - Jugal Kishore Das
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Jianxun Song
- Department
of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas 77807, United States
| | - Qing Sun
- Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77840, United States
- Interdisciplinary
Graduate Program in Genetics and Genomics, Texas A&M University, College
Station, Texas 77843, United States
| |
Collapse
|
12
|
López-García P, Tejero-Ojeda MM, Vaquero ME, Carrión-Vázquez M. Current amyloid inhibitors: Therapeutic applications and nanomaterial-based innovations. Prog Neurobiol 2025; 247:102734. [PMID: 40024279 DOI: 10.1016/j.pneurobio.2025.102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Amyloid proteins have long been in the spotlight for being involved in many degenerative diseases including Alzheimer´s, Parkinson´s or type 2 diabetes, which currently cannot be prevented and for which there is no effective treatment or cure. Here we provide a comprehensive review of inhibitors that act directly on the amyloidogenic pathway (at the monomer, oligomer or fibril level) of key pathological amyloids, focusing on the most representative amyloid-related diseases. We discuss the latest advances in preclinical and clinical trials, focusing on cutting-edge developments, particularly on nanomaterials-based inhibitors, which offer unprecedented opportunities to address the complexity of protein misfolding disorders and are revolutionizing the landscape of anti-amyloid therapeutics. Notably, nanomaterials are impacting critical areas such as bioavailability, penetrability and functionality of compounds currently used in biomedicine, paving the way for more specific therapeutic solutions tailored to various amyloid-related diseases. Finally, we highlight the window of opportunity opened by comparative analysis with so-called functional amyloids for the development of innovative therapeutic approaches for these devastating diseases.
Collapse
|
13
|
Huang Y, Stankevych M, Gujrati V, Klemm U, Mohammed A, Wiesner D, Saccomano M, Tost M, Feuchtinger A, Mishra K, Bruns O, Geerlof A, Ntziachristos V, Stiel AC. Photoswitching protein-XTEN fusions as injectable optoacoustic probes. Acta Biomater 2025; 195:536-546. [PMID: 39914636 DOI: 10.1016/j.actbio.2025.02.002] [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: 05/29/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Optoacoustic imaging (OAI) is a unique in vivo imaging technique combining deep tissue penetration with high resolution and molecular sensitivity. OAI relying on strong intrinsic contrast, such as blood hemoglobin, already shows its value in medical diagnostics. However, OAI sensitivity to current extrinsic contrast agents is insufficient and limits its role in detecting disease-related biomarkers. The recently introduced concept of photoswitching and temporal unmixing techniques for OAI allows detecting extrinsic contrast with high sensitivity, allowing the visualization of small populations of cells labeled with photoswitching proteins deep within the tissue. However, transgene modification might not be permitted in some cases, such as for diagnostic use. Therefore, it is desirable to leverage the concept of photoswitching OAI towards injectable formulations. Since photoswitchable synthetic dyes are mainly excited by blue wavelengths unsuited for imaging in tissue, we propose exploiting the addition of XTENs to photoswitching proteins towards yielding injectable agents. The addition of XTEN to a protein enhances its plasma half-life and bioavailability, thus allowing its use, for example, in targeted labeling approaches. In this pilot study, we show that intravenously injected near-infrared absorbing photoswitchable proteins, ReBphP-PCM, coupled to XTEN, allow highly sensitive optoacoustic visualization of a tumor xenograft in vivo. The sensitivity to XTENs-ReBphP-PCM determined by ex vivo analysis of labeled cells is one to two orders of magnitude beyond conventional synthetic dyes used currently in OAI. The enhanced sensitivity afforded by photoswitching OAI, in combination with the increased bioavailability and biocompatibility of XTENs-ReBphP-PCM, makes this fusion protein a promising tool for facilitating sensitive detection of biomarkers in OAI with a potential for future use in diagnostics. STATEMENT OF SIGNIFICANCE: Optoacoustic imaging (OAI) is a unique in vivo imaging technique that combines deep tissue penetration with high resolution. OAI, which relies on intrinsic contrast, such as blood hemoglobin, could already be valuable in medical diagnostics. However, the use of extrinsic contrast agents to augment disease-related biomarkers in research and diagnostics suffers from very limited sensitivity of the generated contrast agent. We present an intravenously injected photoswitchable protein, ReBphP-PCM, coupled to XTEN, allowing highly sensitive OAI. The sensitivity is one to two orders of magnitude greater than that of conventional synthetic dyes used currently in OA imaging. The high sensitivity afforded by photoswitching together with the enhanced bioavailability and biocompatibility of the XTENs-ReBphP-PCM make this a standard agent for high-quality detection of OAI with potential for clinical use.
Collapse
Affiliation(s)
- Yishu Huang
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Mariia Stankevych
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Vipul Gujrati
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Uwe Klemm
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Azeem Mohammed
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - David Wiesner
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Mara Saccomano
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Monica Tost
- Core Facility Pathology & Tissue Analytics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology & Tissue Analytics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kanuj Mishra
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
| | - Oliver Bruns
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Arie Geerlof
- Protein Expression and Purification Facility, Institute of Structural Biology, Helmholtz Center Munich for Environmental Health, Neuherberg, Germany
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Andre C Stiel
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Biological Imaging, Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health & School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; Protein Engineering for Superresolution Microscopy Lab, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
14
|
Tolmachev V, Papalanis E, Bezverkhniaia EA, Rosly AH, Vorobyeva A, Orlova A, Carlqvist M, Frejd FY, Oroujeni M. Impact of Radiometal Chelates on In Vivo Visualization of Immune Checkpoint Protein Using Radiolabeled Affibody Molecules. ACS Pharmacol Transl Sci 2025; 8:706-717. [PMID: 40109742 PMCID: PMC11915182 DOI: 10.1021/acsptsci.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
The immune checkpoint protein B7-H3 (CD276) is overexpressed in various cancers and is an attractive target for the treatment of malignant tumors. Radionuclide molecular imaging of B7-H3 expression using engineered scaffold proteins such as Affibody molecules is a promising strategy for the selection of potential responders to B7-H3-targeted therapy. Feasibility of B7-H3 imaging was demonstrated using two 99mTc-labeled probes, AC12 and an affinity-matured SYNT179 using a [99mTc]Tc-GGGC label. This study aimed to evaluate whether the use of a residualizing 111In-based label provides better imaging contrast compared with a nonresidualizing label. To do that, SYNT179 and AC12-GGGC Affibody molecules were labeled with 111In using (4,10-bis-carboxymethyl-7-{[2-(2,5-dioxo-3-thioxo-pyrrolidin-1-yl)-ethylcarbamoyl]-methyl}-1,4,7,10-tetraaza-cyclododec-1-yl)-acetic acid (maleimide-DOTA) chelator, site-specifically coupled to the C-terminus of Affibody molecules. The binding affinities of the 111In-labeled conjugates to B7-H3-expressing living cells were higher compared with the affinities of the 99mTc-labeled variants. In mice with B7-H3-expressing xenografts, the tumor uptake of 111In-labeled proteins (3.6 ± 0.3 and 1.8 ± 0.5%ID/g for [111In]In-SYNT179-DOTA and [111In]In-AC12-DOTA, respectively) was significantly (p < 0.05, ANOVA) higher than those for 99mTc-labeled counterparts (1.6 ± 0.2%ID/g and 0.8 ± 0.2%ID/g for [99mTc]Tc-SYNT179 and [99mTc]Tc-AC12-GGGC, respectively). The best variant, [111In]In-SYNT179-DOTA, provided a tumor-to-blood ratio of 31.1 ± 2.9, which was twice higher than that for [99mTc]Tc-SYNT179 and 7-fold higher than that for [99mTc]Tc-AC12-GGGC. Both 111In-labeled Affibody molecules had higher renal retention compared with 99mTc-labeled ones, but the hepatobiliary excretion of 111In-labeled proteins was appreciably lower, potentially improving the imaging of abdominal metastases. Overall, [111In]In-SYNT179-DOTA is the most promising tracer for visualization of B7-H3 expression.
Collapse
Affiliation(s)
- Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Eleftherios Papalanis
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | | | - Alia Hani Rosly
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 83 Uppsala, Sweden
| | | | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
- Affibody AB, 171 65 Solna, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
15
|
Zhang H, Zheng M, Cai Y, Kamara S, Chen J, Zhu S, Zhang L. Novel affibody molecules targeting the AXL extracellular structural domain for molecular imaging and targeted therapy of gastric cancer. Gastric Cancer 2025; 28:174-186. [PMID: 39644434 PMCID: PMC11842530 DOI: 10.1007/s10120-024-01568-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/10/2024] [Indexed: 12/09/2024]
Abstract
Gastric cancer (GC) has a poor prognosis and high mortality because it is often diagnosed at an advanced stage. Targeted therapeutics are considered an important class for advanced GC treatment. However, the fewer effective therapeutic targets and the poor coverage of the GC population limit the use of GC targeted therapies. Recent research suggests that the AXL receptor tyrosine kinase (AXL) plays an vital role in the survival and proliferation of GC cells, and blocking AXL pathway may be an effective strategy for targeted therapies. On the other hand, the affibody molecule, with its small size and faster penetration of tissue, has great potential in tumor imaging and targeted therapy. In this study, we report the novel AXL-binding affibody molecules (ZAXL:239) screened by a phage-displayed peptide library. The ZAXL:239 could specifically bind and interact with AXL proteins in vitro and in vivo, as demonstrated by surface plasmon resonance, co-immunoprecipitation, immuno-fluorescence co-localization, and near infrared fluorescent imaging. In addition, ZAXL:239 affibody molecules could significantly inhibit the proliferative activity and induce apoptosis of AXL-positive GC cells by decreasing the phosphorylation levels of the PI3K/AKT1 and MEK/ERK pathway, leading to the suppression of the downstream nuclear protein c-myc. Moreover, ZAXL:239 was found to have significant anti-tumor effects in AXL-positive GC transplantation tumor nude mouse models. In brief, we provide strong evidence that the novel ZAXL:239 affibody molecules have great potential as a potent tumor-specific molecular imaging and targeted therapeutic agents for GC.
Collapse
Affiliation(s)
- HuiHui Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Maolin Zheng
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - YiQi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Saidu Kamara
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Jun Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Shanli Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| |
Collapse
|
16
|
Ou L, Setegne MT, Elliot J, Shen F, Dassama LMK. Protein-Based Degraders: From Chemical Biology Tools to Neo-Therapeutics. Chem Rev 2025; 125:2120-2183. [PMID: 39818743 PMCID: PMC11870016 DOI: 10.1021/acs.chemrev.4c00595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
The nascent field of targeted protein degradation (TPD) could revolutionize biomedicine due to the ability of degrader molecules to selectively modulate disease-relevant proteins. A key limitation to the broad application of TPD is its dependence on small-molecule ligands to target proteins of interest. This leaves unstructured proteins or those lacking defined cavities for small-molecule binding out of the scope of many TPD technologies. The use of proteins, peptides, and nucleic acids (otherwise known as "biologics") as the protein-targeting moieties in degraders addresses this limitation. In the following sections, we provide a comprehensive and critical review of studies that have used proteins and peptides to mediate the degradation and hence the functional control of otherwise challenging disease-relevant protein targets. We describe existing platforms for protein/peptide-based ligand identification and the drug delivery systems that might be exploited for the delivery of biologic-based degraders. Throughout the Review, we underscore the successes, challenges, and opportunities of using protein-based degraders as chemical biology tools to spur discoveries, elucidate mechanisms, and act as a new therapeutic modality.
Collapse
Affiliation(s)
- Lisha Ou
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Mekedlawit T. Setegne
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Jeandele Elliot
- Department
of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Fangfang Shen
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry, Stanford University, Stanford, California 94305, United States
- Sarafan
ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology & Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
17
|
Brennecke B, Civili B, Sabale PM, Barluenga S, Meyer B, Winssinger N. Self-assembled proteomimetic (SAP) with antibody-like binding from short PNA-peptide conjugates. Proc Natl Acad Sci U S A 2025; 122:e2412850122. [PMID: 39951509 PMCID: PMC11848287 DOI: 10.1073/pnas.2412850122] [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: 06/28/2024] [Accepted: 01/08/2025] [Indexed: 02/16/2025] Open
Abstract
Affinity proteins based on a three-helix bundle (affibodies, alphabodies, and computationally de novo designed ones) have been shown to be a general platform to discover binders with properties reminiscent of antibodies, combining high target specificity with affinities reaching well below the nanomolar. Herein, we report a strategy, coined self-assembled proteomimetic (SAP), to mimic such three-helix bundle architecture with a hybridization-enforced two-helix coiled coil that is obtained by templated native chemical ligation (T-NCL) of PNA-peptide conjugates. This SAP strategy stands out by its synthetic accessibility, reducing the length on the longest synthetic peptide to less than 30 amino acids which is readily attainable by standard SPPS methodologies. We show that the T-NCL dramatically accelerates the ligation, enabling this chemistry to proceed in a combinatorial fashion at low micromolar concentrations. We demonstrate that small combinatorial libraries of SAPs can be prepared in one operation and used directly in affinity selections against a target of interest with an LC-MS analysis of the fittest binders. Moreover, we show that the underlying design paradigm is functional for SAPs based on structurally distinct three-helix peptides aimed at different therapeutic targets, namely HER2 and spike's RBD, reaching picomolar affinities. We further illustrate that the affinity of the SAP can be allosterically regulated using a toehold displacement of the hybridizing PNAs to disrupt the coiled coil stabilization. Finally, we show that an RBD-targeting SAP effectively inhibits viral entry of SARS-CoV-2 with an IC50 of 2.8 nM.
Collapse
Affiliation(s)
- Benjamin Brennecke
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva1211, Switzerland
| | - Beatrice Civili
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva1211, Switzerland
| | - Pramod M. Sabale
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva1211, Switzerland
| | - Sofia Barluenga
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva1211, Switzerland
| | - Benjamin Meyer
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva1211, Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry, Faculty of Science, University of Geneva, Geneva1211, Switzerland
| |
Collapse
|
18
|
Liu Y, Valji K, Monsky W, Zheng C, Yang X. Optical imaging guidance in oncologic surgery and interventional oncology. Pharmacol Res 2025; 212:107612. [PMID: 39826822 PMCID: PMC12057765 DOI: 10.1016/j.phrs.2025.107612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Over recent decades, optical imaging (OI) has become an integral part of medical imaging, offering significant advantages over other modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI). OI is distinguished by its real-time imaging capability, cost-effectiveness, portability, absence of ionizing radiation, and high patient acceptability. The introduction of advanced optical dyes (including FDA-approved agents like indocyanine green, Cytalux, and Gleolan) has greatly enhanced its clinical utility. OI has shown clear benefits in the management of patients with cancer, originally by open surgery and now extending to minimally invasive, image-guided interventional procedures. This review highlights recent developments in OI for oncology, emphasizing its benefits for clinicians in guiding surgical and interventional procedures.
Collapse
Affiliation(s)
- Yiming Liu
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA; Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Karim Valji
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Wayne Monsky
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoming Yang
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA.
| |
Collapse
|
19
|
Bobolowski H, Fiedler E, Haupts U, Lilie H, Weininger U. A functional helix shuffled variant of the B domain of Staphylococcus aureus. Protein Sci 2025; 34:e70012. [PMID: 39840789 PMCID: PMC11751873 DOI: 10.1002/pro.70012] [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: 07/26/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025]
Abstract
The B domain of protein A is a biotechnologically important three-helix bundle protein. It binds the Fc fragment of antibodies with helix 1/2 and the Fab region with helix 2/3. Here we designed a helix shuffled variant by changing the connectivity of the helices, in order to redesign the helix bundle, yielding altered helix-loop-helix properties. The new loops that generate the new connectivity were created in several protein libraries, and Fc binding variants were selected for a detailed biochemical characterization. We were able to create variants with Fc binding affinity at the same level as the wild type B but with significantly reduced thermal stability. The NMR structure proved that the overall three-dimensional structure was maintained not only in the helix shuffled variant but also points to some potential local differences to wild-type B, which could be the reason for the reduced thermal stability. Therefore, protein A is an example of an optimized structure being more important for stability than for function. Using the helix shuffled variant as a ligand on an affinity column facilitates a robust and straightforward purification of antibodies, but allows for a milder elution at less extreme pH. Therefore, the helix shuffled variant is a suitable ligand to purify more pH-sensitive antibodies.
Collapse
Affiliation(s)
| | | | | | - Hauke Lilie
- Department of Biotechnology and BiochemistryMartin‐Luther‐University Halle‐WittenbergHalleGermany
| | - Ulrich Weininger
- Institute of Physics, BiophysicsMartin‐Luther‐University Halle‐WittenbergHalle (Saale)Germany
| |
Collapse
|
20
|
Cheung P, Persson J, Zhang B, Vasylovska S, Lau J, Invast S, Korsgren O, Ståhl S, Löfblom J, Eriksson O. DGCR2 targeting affibody molecules for delivery of drugs and imaging reagents to human beta cells. Sci Rep 2025; 15:417. [PMID: 39747317 PMCID: PMC11695922 DOI: 10.1038/s41598-024-84574-y] [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: 10/09/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
A distinctive feature of both type 1 and type 2 diabetes is the waning of insulin-secreting beta cells in the pancreas. New methods for direct and specific targeting of the beta cells could provide platforms for delivery of pharmaceutical reagents. Imaging techniques such as Positron Emission Tomography (PET) rely on the efficient and specific delivery of imaging reagents, and could greatly improve our understanding of diabetes etiology as well as providing biomarkers for viable beta-cell mass in tissue, in both pancreas and in islet grafts.The DiGeorge Syndrome Critical Region Gene 2 (DGCR2) protein has been suggested as a beta-cell specific protein in the pancreas, but so far there has been a lack of available high-affinity binders suitable for targeted drug delivery or molecular imaging. Affibody molecules belong to a class of small affinity proteins with excellent properties for molecular imaging. Here, we further validate the presence of DGCR2 in pancreatic and stem cell (SC)-derived beta cells, and then describe the generation and selection of several Affibody molecules candidates that target human DGCR2. Using an in-house developed directed evolution method, new DGCR2-binding Affibody molecules were generated and evaluated for thermal stability and affinity. The Affibody molecules variants were further developed as targeting agents for delivering imaging reagents to beta cell. The Affibody molecule ZDGCR2:AM106 displayed nanomolar affinity, suitable stability and biodistribution, with negligible toxicity to islets, qualifying it as a suitable lead candidate for further development as a tool for specific delivery of drugs and imaging reagents to beta cells.
Collapse
Affiliation(s)
- Pierre Cheung
- Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jonas Persson
- Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bo Zhang
- Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Joey Lau
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sofie Invast
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Ståhl
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, Division of Protein Engineering, KTH Royal Institute of Technology, Stockholm, Sweden.
- KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Olof Eriksson
- Science For Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.
- Uppsala University, Uppsala, Sweden.
| |
Collapse
|
21
|
Liu Y, Bai X, Wang H, Wang J, Li S, Zhang H, Wang F, Hong Z. PEG-modification enhances the targeted photothermal therapy of affibody-conjugated indocyanine green for precision cancer treatment. Biochem Biophys Res Commun 2025; 742:151155. [PMID: 39662453 DOI: 10.1016/j.bbrc.2024.151155] [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: 09/18/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Photothermal therapy (PTT) is an innovative cancer treatment that leverages heat generated from near-infrared light exposure to induce tumor cell death. A major challenge in PTT is achieving precise delivery of the photothermal agent to tumor tissues to maximize efficacy and minimize off-target effects. In this study, we introduce a novel ligand-coupled photothermal reagent that addresses this challenge by leveraging the high-affinity HER2 affibody ZHER2:2891 (referred to as ZHER2), conjugated with indocyanine green (ICG) for targeted delivery. Polyethylene glycol (PEG) was incorporated as a hydrophilic linker to further optimize photothermal conversion efficiency and enhance tumor-specific targeting. Among the conjugates tested, ZHER2-PEG1000-ICG, modified with a PEG chain of 1000 Da molecular weight, demonstrated exceptional performance. In vitro studies revealed that ZHER2-PEG1000-ICG specifically bound to HER2-expressing cells and effectively induced cell death. In vivo experiments using HER2-positive N87 tumor-bearing mice showed that ZHER2-PEG1000-ICG accumulated highly and specifically in tumor tissues over an extended period. Upon light irradiation, this conjugate caused a significant rise in temperature at the tumor site, resulting in complete tumor elimination with a single photothermal treatment. This PEG-modified affibody-ICG conjugate represents a precise and effective approach to PTT, offering a promising new therapeutic strategy for cancer treatment with the potential to significantly impact future cancer therapies.
Collapse
Affiliation(s)
- Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, 453100, PR China; State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Xuerui Bai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Henan Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Jian Wang
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, PR China.
| | - Shuang Li
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, PR China.
| | - Hongru Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| | - Fengwei Wang
- People's Hospital of Tianjin, School of Medicine, Nankai University, Tianjin, 300071, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
22
|
Martins SA, Correia JDG. 99mTc(I)-Labeled His-Tagged Proteins: Impact in the Development of Novel Imaging Probes and in Drug Discovery. Chembiochem 2024; 25:e202400645. [PMID: 39158861 DOI: 10.1002/cbic.202400645] [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: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Abstract
Technetium-99 m (99mTc) remains the cornerstone of nuclear medicine for single photon emission computed tomography (SPECT) due to its widespread availability and chemical and physical features. Its multiple oxidation states allow for the design and production of radiopharmaceuticals with versatile properties, namely in terms of pharmacokinetic profile. 99mTc(V) is the most common oxidation state, but 99mTc(I) gained traction after the pioneering work of Alberto and colleagues, which resulted in the introduction of the organometallic core fac-[99mTc(CO)3(H2O)3]+. This core is readily available from [99mTcO4]- and displays three labile water molecules that can be easily swapped for ligands with different denticity and/or donor atoms in aqueous environment. This makes it possible to radiolabel small molecules as well as high molecular weight molecules, such as antibodies or other proteins, while assuring biological activity. Direct radiolabelling of those proteins with fac-[99mTc(CO)3]+ under mild conditions is accomplished through incorporation of a polyhistidine tag (His-tag), a commonly used tag for purification of recombinant proteins. This review aims to address the direct radiolabelling of His-tagged macromolecules with fac-[99mTc(CO)3]+ for development of molecular imaging agents and the impact of this technology in the discovery and development of imaging and/or therapeutic agents towards clinical application.
Collapse
Affiliation(s)
- Sofia A Martins
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | - João D G Correia
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
- Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, CTN, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| |
Collapse
|
23
|
Li M, Yang J, Zhou L, Zhang J, Li Y, Chen J, Dong H, Zhang L, Zhu S. Efficacy of a Novel Affitoxin Targeting Major Outer Membrane Protein Against Chlamydia trachomatis In Vitro and In Vivo. J Infect Dis 2024; 230:1476-1487. [PMID: 38723186 DOI: 10.1093/infdis/jiae257] [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: 10/19/2023] [Accepted: 05/04/2024] [Indexed: 12/17/2024] Open
Abstract
Targeted therapy is an attractive approach for treating infectious diseases. Affibody molecules have similar capability to antibodies that facilitate molecular recognition in both diagnostic and therapeutic applications. Targeting major outer membrane protein (MOMP) for treating infection of Chlamydia trachomatis, one of the most common sexually transmitted pathogens, is a promising therapeutic approach. Previously, we have reported a MOMP-specific affibody (ZMOMP:461) from phage display library. Here, we first fused it with modified Pseudomonas exotoxin (PE38KDEL) and a cell-penetrating peptide (CPP) to develop an affitoxin, Z461X-CPP. We then verified the addition of both toxin and CPPs that did not affect the affinitive capability of ZMOMP:461 to MOMP. Upon uptake by C trachomatis-infected cells, Z461X-CPP induced cell apoptosis in vitro. In an animal model, Z461X significantly shortened the duration of C trachomatis infection and prevented pathological damage in the mouse reproductive system. These findings provide compelling evidence that the MOMP-specific affitoxin has great potential for targeting therapy of C trachomatis infection.
Collapse
Affiliation(s)
- Mingyang Li
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Jia Yang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Luqi Zhou
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Jing Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Yang Li
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Jun Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Haiyan Dong
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| | - Shanli Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
24
|
Li CL, Ma XY, Yi P. Bispecific Antibodies, Immune Checkpoint Inhibitors, and Antibody-Drug Conjugates Directing Antitumor Immune Responses: Challenges and Prospects. Cell Biochem Funct 2024; 42:e70011. [PMID: 39463028 DOI: 10.1002/cbf.70011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/27/2024] [Accepted: 10/13/2024] [Indexed: 10/29/2024]
Abstract
Tumor immunotherapy includes bispecific antibodies (BsAbs), immune checkpoint inhibitors (ICIs), vaccines, and adoptive cell immunotherapy. BsAbs belong to the family of antibodies that can specifically target two or more different antigens and are a promising option for tumor immunotherapy. Immune checkpoints are antibodies targeting PD-1, PD-L1, and CTLA4 and have demonstrated remarkable therapeutic efficacy in the treatment of hematological and solid tumors, whose combination therapies have been shown to synergistically enhance the antitumor effects of BsAbs. In addition, the clinical efficacy of existing monoclonal antibodies targeting PD-1 (e.g., ipilimumab, nivolumab, pembrolizumab, and cemiplimab) and PD-L1 (e.g., atezolizumab, avelumab, and durvalumab) could also be enhanced by conjugation to small drugs as antibody-drug conjugates (ADCs). The development of truly effective therapies for patients with treatment-resistant cancers can be achieved by optimizing the various components of ADCs.
Collapse
Affiliation(s)
- Chen Lu Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yuan Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yi
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Swenson CS, Mandava G, Thomas DM, Moellering RE. Tackling Undruggable Targets with Designer Peptidomimetics and Synthetic Biologics. Chem Rev 2024; 124:13020-13093. [PMID: 39540650 PMCID: PMC12036645 DOI: 10.1021/acs.chemrev.4c00423] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The development of potent, specific, and pharmacologically viable chemical probes and therapeutics is a central focus of chemical biology and therapeutic development. However, a significant portion of predicted disease-causal proteins have proven resistant to targeting by traditional small molecule and biologic modalities. Many of these so-called "undruggable" targets feature extended, dynamic protein-protein and protein-nucleic acid interfaces that are central to their roles in normal and diseased signaling pathways. Here, we discuss the development of synthetically stabilized peptide and protein mimetics as an ever-expanding and powerful region of chemical space to tackle undruggable targets. These molecules aim to combine the synthetic tunability and pharmacologic properties typically associated with small molecules with the binding footprints, affinities and specificities of biologics. In this review, we discuss the historical and emerging platforms and approaches to design, screen, select and optimize synthetic "designer" peptidomimetics and synthetic biologics. We examine the inspiration and design of different classes of designer peptidomimetics: (i) macrocyclic peptides, (ii) side chain stabilized peptides, (iii) non-natural peptidomimetics, and (iv) synthetic proteomimetics, and notable examples of their application to challenging biomolecules. Finally, we summarize key learnings and remaining challenges for these molecules to become useful chemical probes and therapeutics for historically undruggable targets.
Collapse
Affiliation(s)
- Colin S Swenson
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Gunasheil Mandava
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Deborah M Thomas
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Raymond E Moellering
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
26
|
Yang B, Gomes DEB, Liu Z, Santos MS, Li J, Bernardi RC, Nash MA. Engineering the Mechanical Stability of a Therapeutic Complex between Affibody and Programmed Death-Ligand 1 by Anchor Point Selection. ACS NANO 2024; 18:31912-31922. [PMID: 39514863 DOI: 10.1021/acsnano.4c09220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Protein-protein complexes can vary in mechanical stability depending on the direction from which force is applied. Here, we investigated the mechanical stability of a complex between a binding scaffold called Affibody and an immune checkpoint protein Programmed Death-Ligand 1 (PD-L1). We used AFM single-molecule force spectroscopy with bioorthogonal clickable peptide handles, shear stress bead adhesion assays, molecular modeling, and steered molecular dynamics (SMD) to understand the pulling point dependency of the mechanostability of the Affibody:(PD-L1) complex. We observed a wide range of rupture forces depending on the anchor point. Pulling from residue #22 on Affibody generated an intermediate state attributed to partially unfolded PD-L1, while pulling from Affibody's N-terminus generated a force-activated catch bond. Pulling from residue #22 or #47 on Affibody generated high rupture forces, with the complex breaking at up to ∼190 pN under loading rates of ∼104-105 pN/s, representing a ∼4-fold increase as compared with low-force N-terminal pulling. SMD simulations showed relative tendencies in rupture forces that were consistent with experiments and, through visualization of force propagation networks, provided mechanistic insights. These results demonstrate how the mechanical properties of protein-protein interfaces can be controlled by informed choice of site-specific bioconjugation points within molecules, with implications for optimal bioconjugation strategies in drug delivery vehicles.
Collapse
Affiliation(s)
- Byeongseon Yang
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Diego E B Gomes
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| | - Zhaowei Liu
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Mariana Sá Santos
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Jiajun Li
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Rafael C Bernardi
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| | - Michael A Nash
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| |
Collapse
|
27
|
Colas K, Bindl D, Suga H. Selection of Nucleotide-Encoded Mass Libraries of Macrocyclic Peptides for Inaccessible Drug Targets. Chem Rev 2024; 124:12213-12241. [PMID: 39451037 PMCID: PMC11565579 DOI: 10.1021/acs.chemrev.4c00422] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024]
Abstract
Technological advances and breakthrough developments in the pharmaceutical field are knocking at the door of the "undruggable" fortress with increasing insistence. Notably, the 21st century has seen the emergence of macrocyclic compounds, among which cyclic peptides are of particular interest. This new class of potential drug candidates occupies the vast chemical space between classic small-molecule drugs and larger protein-based therapeutics, such as antibodies. As research advances toward clinical targets that have long been considered inaccessible, macrocyclic peptides are well-suited to tackle these challenges in a post-rule of 5 pharmaceutical landscape. Facilitating their discovery is an arsenal of high-throughput screening methods that exploit massive randomized libraries of genetically encoded compounds. These techniques benefit from the incorporation of non-natural moieties, such as non- proteinogenic amino acids or stabilizing hydrocarbon staples. Exploiting these features for the strategic architectural design of macrocyclic peptides has the potential to tackle challenging targets such as protein-protein interactions, which have long resisted research efforts. This Review summarizes the basic principles and recent developments of the main high-throughput techniques for the discovery of macrocyclic peptides and focuses on their specific deployment for targeting undruggable space. A particular focus is placed on the development of new design guidelines and principles for the cyclization and structural stabilization of cyclic peptides and the resulting success stories achieved against well-known inaccessible drug targets.
Collapse
Affiliation(s)
- Kilian Colas
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Daniel Bindl
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Suga
- University of Tokyo, Department of Chemistry, Graduate School of Science 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
28
|
Ståhl S, Lindberg H, Hjelm LC, Löfblom J, Dahlsson Leitao C. Engineering of Affibody Molecules. Cold Spring Harb Protoc 2024; 2024:pdb.top107760. [PMID: 37491082 DOI: 10.1101/pdb.top107760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Affibody molecules are small, robust, and versatile affinity proteins currently being explored for therapeutic, diagnostic, and biotechnological applications. Surface-exposed residues on the affibody scaffold are randomized to create large affibody libraries from which novel binding specificities to virtually any protein target can be generated using combinatorial protein engineering. Affibody molecules have the potential to complement-or even surpass-current antibody-based technologies, exhibiting multiple desirable properties, such as high stability, affinity, and specificity, efficient tissue penetration, and straightforward modular extension of functional domains. It has been shown in both preclinical and clinical studies that affibody molecules are safe, efficacious, and valuable alternatives to antibodies for specific targeting in the context of in vivo diagnostics and therapy. Here, we provide a general background of affibody molecules, give examples of reported applications, and briefly summarize the methodology for affibody generation.
Collapse
Affiliation(s)
- Stefan Ståhl
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Hanna Lindberg
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Linnea Charlotta Hjelm
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | | |
Collapse
|
29
|
Zhu J, Yu H, Xie L, Shuai D, Huang Z, Chen Y, Ni C, Jia C, Rong X, Zhang L, Chu M. A novel format of TNF-α binding affibody molecule ameliorate coronary artery endothelial injury in a mouse model of Kawasaki disease. Int J Biol Macromol 2024; 281:136255. [PMID: 39366611 DOI: 10.1016/j.ijbiomac.2024.136255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/19/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Kawasaki disease (KD) is a disease characterized by systemic immune vasculitis that often involves coronary arteries and can result in long-term cardiovascular sequelae. Different strategies for treatment of KD-and KD-induced coronary artery lesions are currently under investigation, including passive immunization with anti-TNFα monoclonal antibodies (mAbs). Herein, we examine the potential therapeutic capabilities of a novel type of TNFα-targeting agent based on an affibody molecule possessing fundamentally different properties than mAbs. Using phage display technology, we successfully screened and obtained three TNF-α binding affibody molecules and confirmed their high binding affinity and specificity for recombinant and native TNF-α by surface plasmon resonance (SPR), confocal double immunofluorescence and coimmunoprecipitation assays. Moreover, by binding to TNF-α, the affibody molecules could effectively neutralize TNFα-induced L929 cytotoxicity. To increase the targeting properties and serum half-life, one preferred affibody molecule ZTNF-α263 was redesigned to assemble drugs with bivalent TNFα binding with added specificity for serum albumin (ZTNF-α263-ABD035-ZTNF-α263, hereinafter denoted ZTAT). We further determined its binding ability, TNF-α signal blocking and neutralizing capacity, serum half-life and immunogenicity. Most importantly, our study provides strong evidence that the engineered ZTAT protein was therapeutically effective against KD induced-endothelial injury, as judged by both in vitro and in vivo assessments. These data suggested that because of the flexibility inherent, low-molecular weight anti-TNFα affibody construct ZTAT, can be developed into a potent therapeutic agent that can be produced and purified cost-effectively.
Collapse
Affiliation(s)
- Jinshun Zhu
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huan Yu
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Longzhi Xie
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dujuan Shuai
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhixian Huang
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yufei Chen
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chao Ni
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chang Jia
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xing Rong
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
| | - Maoping Chu
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, 325027 Wenzhou, Zhejiang, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
30
|
Liu Y, Oroujeni M, Liao Y, Vorobyeva A, Bodenko V, Orlova A, Konijnenberg M, Carlqvist M, Wahlberg E, Loftenius A, Frejd FY, Tolmachev V. Evaluation of a novel 177Lu-labelled therapeutic Affibody molecule with a deimmunized ABD domain and improved biodistribution profile. Eur J Nucl Med Mol Imaging 2024; 51:4038-4048. [PMID: 39008065 PMCID: PMC11527907 DOI: 10.1007/s00259-024-06840-5] [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: 01/25/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE Fusion of Affibody molecules with an albumin-binding domain (ABD) provides targeting agents, which are suitable for radionuclide therapy. To facilitate clinical translation, the low immunogenic potential of such constructs with targeting properties conserved is required. METHODS The HER2-targeting Affibody molecule ZHER2:2891 was fused with a deimmunized ABD variant and DOTA was conjugated to a unique C-terminal cysteine. The novel construct, PEP49989, was labelled with 177Lu. Affinity, specificity, and in vivo targeting properties of [177Lu]Lu-PEP49989 were characterised. Experimental therapy in mice with human HER2-expressing xenografts was evaluated. RESULTS The maximum molar activity of 52 GBq/µmol [177Lu]Lu-PEP49989 was obtained. [177Lu]Lu-PEP49989 bound specifically to HER2-expressing cells in vitro and in vivo. The HER2 binding affinity of [177Lu]Lu-PEP49989 was similar to the affinity of [177Lu]Lu-ABY-027 containing the parental ABD035 variant. The renal uptake of [177Lu]Lu-PEP49989 was 1.4-fold higher, but hepatic and splenic uptake was 1.7-2-fold lower than the uptake of [177Lu]Lu-ABY-027. The median survival of xenograft-bearing mice treated with 21 MBq [177Lu]Lu-PEP49989 (> 90 days) was significantly longer than the survival of mice treated with vehicle (38 days) or trastuzumab (45 days). Treatment using a combination of [177Lu]Lu-PEP49989 and trastuzumab increased the number of complete tumour remissions. The renal and hepatic toxicity was minimal to mild. CONCLUSION In preclinical studies, [177Lu]Lu-PEP49989 demonstrated favourable biodistribution and a strong antitumour effect, which was further enhanced by co-treatment with trastuzumab.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Affibody AB, Solna, 171 65, Sweden
| | - Yunqi Liao
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Vitalina Bodenko
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, 751 23, Sweden
| | - Mark Konijnenberg
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden
- Affibody AB, Solna, 171 65, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 751 85, Sweden.
| |
Collapse
|
31
|
Lechi F, Eriksson J, Odell LR, Wegrzyniak O, Löfblom J, Frejd FY, Zhang B, Eriksson O. Optimized method for fluorine-18 radiolabeling of Affibody molecules using RESCA. EJNMMI Radiopharm Chem 2024; 9:73. [PMID: 39460878 PMCID: PMC11512968 DOI: 10.1186/s41181-024-00304-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND In recent years, the interest in Al[18F]F as a labeling agent for Positron Emission Tomography (PET) radiotracers has risen, as it allows for fast and efficient fluorine-18 labeling by harnessing chelation chemistry. The introduction of Restrained Complexing Agent (RESCA) as a chelator has also shown that chelator-based radiolabeling reactions can be performed in mild conditions, making the radiolabeling process attractively more facile than most conventional radiofluorination methods. The aim of the study was to establish optimized conditions for Al[18F]F labeling of Affibody molecules using RESCA as a complexing agent, using Z09591 and Z0185, two Affibody proteins targeting PDGFRβ and TNFα, respectively, as model compounds. RESULTS The Al[18F]F labeling of RESCA-conjugated Z09591 was tested at different temperatures (rt to 60 °C) and with varying reaction times (12 to 60 min), and optimal conditions were then implemented on RESCA-Z0185. The optimized synthesis method was: 1.5-2.5 GBq of cyclotron produced fluorine-18 were trapped on a QMA cartridge and eluted with saline solution to react with 12 nmol of AlCl3 and form Al[18F]F. The respective RESCA-conjugated Affibody molecule (14 nmol) in NaOAc solution was added to the Al[18F]F solution and left to react at 60 °C for 12 min. The mixture was purified on a NAP5 size exclusion column and then analyzed by HPLC. The entire process took approximately 35 min, was highly reproducible, indicating the efficiency and reliability of the method. The labeled compounds demonstrated retained biological function for their respective targets after purification. CONCLUSIONS We present a general and optimized method for Al[18F]F labeling of RESCA-conjugated Affibody molecules, which can be widely applied to this class of peptide-based imaging agents. Moreover, radiochemical yields were improved when the labeling was conducted at 37 °C or above. In vitro and in vivo assessment of the respective tracers was promising, showing retained binding capacity as well as moderate defluorination, which is usually regarded as a potential downside for RESCA-conjugated tracers.
Collapse
Affiliation(s)
- Francesco Lechi
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Jonas Eriksson
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- PET center, Uppsala University Hospital, Uppsala, Sweden
| | - Luke R Odell
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Olivia Wegrzyniak
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - John Löfblom
- Division of Protein Engineering, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Bo Zhang
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Department of Medicinal Chemistry, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
32
|
Wang S, Zhang C, Liu H, Fan X, Fu S, Li W, Zhang H. Targeted PHA Microsphere-Loaded Triple-Drug System with Sustained Drug Release for Synergistic Chemotherapy and Gene Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1657. [PMID: 39452993 PMCID: PMC11510473 DOI: 10.3390/nano14201657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
The combination of paclitaxel (PTX) with other chemotherapy drugs (e.g., gemcitabine, GEM) or genetic drugs (e.g., siRNA) has been shown to enhance therapeutic efficacy against tumors, reduce individual drug dosages, and prevent drug resistance associated with single-drug treatments. However, the varying solubility of chemotherapy drugs and genetic drugs presents a challenge in co-delivering these agents. In this study, nanoparticles loaded with PTX were prepared using the biodegradable polymer material poly(3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx). These nanoparticles were surface-modified with target proteins (Affibody molecules) and RALA cationic peptides to create core-shell structured microspheres with targeted and cationic functionalization. A three-drug co-delivery system (PTX@PHBHHx-ARP/siRNAGEM) were developed by electrostatically adsorbing siRNA chains containing GEM onto the microsphere surface. The encapsulation efficiency of PTX in the nanodrug was found to be 81.02%, with a drug loading of 5.09%. The chemogene adsorption capacity of siRNAGEM was determined to be 97.3%. Morphological and size characterization of the nanodrug revealed that PTX@PHBHHx-ARP/siRNAGEM is a rough-surfaced microsphere with a particle size of approximately 150 nm. This nanodrug exhibited targeting capabilities toward BT474 cells with HER2 overexpression while showing limited targeting ability toward MCF-7 cells with low HER2 expression. Results from the MTT assay demonstrated that PTX@PHBHHx-ARP/siRNAGEM exhibits high cytotoxicity and excellent combination therapy efficacy compared to physically mixed PTX/GEM/siRNA. Additionally, Western blot analysis confirmed that siRNA-mediated reduction of Bcl-2 expression significantly enhanced cell apoptosis mediated by PTX or GEM in tumor cells, thereby increasing cell sensitivity to PTX and GEM. This study presents a novel targeted nanosystem for the co-delivery of chemotherapy drugs and genetic drugs.
Collapse
Affiliation(s)
- Shuo Wang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| | - Chao Zhang
- Department of Life Science, Hengshui University, Hengshui 053000, China;
| | - Huandi Liu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| | - Xueyu Fan
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| | - Shuangqing Fu
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| | - Wei Li
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| | - Honglei Zhang
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Materials Science, Hebei University, Baoding 071002, China; (S.W.); (H.L.); (X.F.); (S.F.)
| |
Collapse
|
33
|
Li Q, Yang X, Xia X, Xia XX, Yan D. Affibody-Functionalized Elastin-like Peptide-Drug Conjugate Nanomicelle for Targeted Ovarian Cancer Therapy. Biomacromolecules 2024; 25:6474-6484. [PMID: 39235966 DOI: 10.1021/acs.biomac.4c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Recombinant elastin-like polypeptides (ELPs) have emerged as an attractive nanoplatform for drug delivery due to their tunable genetically encoded sequence, biocompatibility, and stimuli-responsive self-assembly behaviors. Here, we designed and biosynthesized an HER2 (human epidermal growth factor receptor 2)-targeted affibody-ELP fusion protein (Z-ELP), which was subsequently conjugated with monomethyl auristatin E (MMAE) to build a protein-drug conjugate (Z-ELP-M). Due to its thermal response, Z-ELP-M can immediately self-assemble into a nanomicelle at physiological temperature. Benefiting from its active targeting and nanomorphology, Z-ELP-M exhibits enhanced cellular internalization and deep tumor penetration in vitro. Moreover, Z-ELP-M shows excellent tumor targeting and superior antitumor efficacy in HER2-positive ovarian cancer, demonstrating a relative tumor growth inhibition of 104.6%. These findings suggest that an affibody-functionalized elastin-like peptide-drug conjugate nanomicelle is an efficient strategy to improve antitumor efficacy and biosafety in cancer therapy.
Collapse
Affiliation(s)
- Qingrong Li
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xuelin Xia
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
34
|
Shao H, Lv K, Wang P, Jin J, Cai Y, Chen J, Kamara S, Zhu S, Zhu G, Zhang L. Novel anti-CEA affibody for rapid tumor-targeting and molecular imaging diagnosis in mice bearing gastrointestinal cancer cell lines. Front Microbiol 2024; 15:1464088. [PMID: 39444679 PMCID: PMC11496145 DOI: 10.3389/fmicb.2024.1464088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
Gastrointestinal cancer is a common malignant tumor with a high incidence worldwide. Despite continuous improvements in diagnosis and treatment strategies, the overall prognosis of gastrointestinal tumors remains poor. Carcinoembryonic antigen (CEA) is highly expressed in various types of cancers, especially in gastrointestinal cancers, making it a potential target for therapeutic intervention. Therefore, the expression of CEA can be used as an indication of the existence of tumors, chosen as a target for molecular imaging diagnosis, and effectively utilized in the targeted therapy of gastrointestinal cancers. In this study, we report the selection and characterization of affibody molecules (ZCEA539, ZCEA546, and ZCEA919) specific to the CEA protein. Their ability to bind to recombinant and native CEA protein has been confirmed by surface plasmon resonance (SPR), immunofluorescence, and immunohistochemistry assays. Furthermore, Dylight755-labeled ZCEA affibody showed accumulation within the tumor site 1 h post injection and was continuously enhanced for 4 h. The Dylight755-labeled ZCEA affibody exhibited high tumor-targeting specificity in CEA+ xenograft-bearing mice and possesses promising characteristics for tumor-targeting imaging. Overall, our results suggest the potential use of ZCEA affibodies as fluorescent molecular imaging probes for detecting CEA expression in gastrointestinal cancer.
Collapse
Affiliation(s)
- Huanyi Shao
- Department of Pediatric Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Kaiji Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Pengfei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinji Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiqi Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jun Chen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Saidu Kamara
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Shanli Zhu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Guanbao Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lifang Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Institute of Molecular Virology and Immunology, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Wohlrab J, Eichner A. [Variants of biotechnological drugs in dermatology : Status quo and future]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:775-780. [PMID: 39172184 DOI: 10.1007/s00105-024-05403-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 08/23/2024]
Abstract
Biotechnological drugs, so-called biopharmaceuticals, have complex structures, have special physicochemical characteristics and are subject to special regulatory laws. In addition to recombinant monoclonal antibodies, proteins and fusion proteins, a large number of biotechnological variations have been developed, of which antibody fragments, nanobodies, peptides, and antibody-drug conjugates in particular have found their way into clinical application. In addition to strategies for the treatment of oncological diseases, chronic inflammatory diseases in particular are being addressed, which are also becoming of great importance in dermatology. The advantages of biopharmaceuticals are increasingly being recognised and developed as part of special strategies for topical application. Due to the rapid development of molecular medicine, new targets for biopharmaceuticals are constantly being identified, and pharmacokinetically favourable biotechnological molecules are being created using refined methods. It can be assumed that this development will lead to even more highly innovative therapeutic and diagnostic options.
Collapse
Affiliation(s)
- Johannes Wohlrab
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie, Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Str. 40, 06097, Halle (Saale), Deutschland.
- An-Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Deutschland.
| | - Adina Eichner
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie, Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Str. 40, 06097, Halle (Saale), Deutschland
- An-Institut für angewandte Dermatopharmazie, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Deutschland
| |
Collapse
|
36
|
Dahlsson Leitao C, Ståhl S, Löfblom J. Surface-engineered bacteria in drug development. Microb Biotechnol 2024; 17:e70033. [PMID: 39403960 PMCID: PMC11474283 DOI: 10.1111/1751-7915.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Bacterial surface display in combination with fluorescence-activated cell sorting is a versatile and robust system and an interesting alternative approach to phage display for the generation of therapeutic affinity proteins. The system enables real-time monitoring and sorting of cell populations, which presents unique possibilities for drug development. It has been used to develop several affibody molecules currently being evaluated preclinically for the treatment and diagnosis of, for example, cancer and neurodegenerative diseases. Additionally, it can be implemented in other areas of drug design, such as for mapping epitopes and evolving enzyme specificities.
Collapse
Affiliation(s)
| | - Stefan Ståhl
- Department of Protein ScienceKTH Royal Institute of TechnologyStockholmSweden
| | - John Löfblom
- Department of Protein ScienceKTH Royal Institute of TechnologyStockholmSweden
| |
Collapse
|
37
|
Saemundsson SA, Curry SD, Bower BM, DeBoo EJ, Goodwin AP, Cha JN. Controlling cellular packing and hypoxia in 3D tumor spheroids via DNA interactions. Biomater Sci 2024; 12:4759-4769. [PMID: 39136101 PMCID: PMC11320176 DOI: 10.1039/d4bm00688g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Tumor spheroids represent valuable in vitro models for studying cancer biology and evaluating therapeutic strategies. In this study, we investigated the impact of varying lengths of DNA-modified cell surfaces on spheroid formation, cellular adhesion molecule expression, and hypoxia levels within 4T1 mouse breast cancer spheroids. Through a series of experiments, we demonstrated that modifying cell surfaces with biotinylated DNA strands of different lengths facilitated spheroid formation without significantly altering the expression of fibronectin and e-cadherin, key cellular adhesion molecules. However, our findings revealed a notable influence of DNA length on hypoxia levels within the spheroids. As DNA length increased, hypoxia levels decreased, indicating enhanced intercellular spacing and porosity within the spheroid structure. These results contribute to a better understanding of how DNA modification of cell surfaces can modulate spheroid architecture and microenvironmental conditions. Such insights may have implications for developing therapeutic interventions targeting the tumor microenvironment to improve cancer treatment efficacy.
Collapse
Affiliation(s)
- Sven A Saemundsson
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
| | - Shane D Curry
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
| | - Bryce M Bower
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
| | - Ethan J DeBoo
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
| | - Andrew P Goodwin
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA
| | - Jennifer N Cha
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA.
- Materials Science and Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA
- Biomedical Engineering Program, University of Colorado, Boulder, 596 UCB, Boulder, CO, 80303, USA
| |
Collapse
|
38
|
Hu X, Hu H, Li D, Wang P, Cai J. Affibody-based molecular probe 99mTc-(HE) 3Z HER2:V2 for non-invasive HER2 detection in ovarian and breast cancer xenografts. Open Med (Wars) 2024; 19:20241027. [PMID: 39247440 PMCID: PMC11377979 DOI: 10.1515/med-2024-1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose This study aimed to assess the biodistribution and bioactivity of the affibody molecular probe 99mTc-(HE)3ZHER2:V2, prepared by genetic recombination, and to investigate its potential for targeted human epidermal growth factor receptor 2 (HER2) imaging in SKOV3 ovarian cancer and MDA-MB-361 breast cancer xenografts. Methods Affibody molecules were generated through genetic recombination. The radiochemical purity of the 99mTc-labeled HER2 affibody was determined using reverse phase high performance liquid chromatography (RP-HPLC). Evaluation of HER2 affinity in SKOV3 ovarian cancer cells and MDA-MB-361 breast cancer cells (HER2-positive) was conducted by calculating equilibrium dissociation constants. Biodistribution of the 99mTc-labeled affibody molecular probe was assessed in Balb/c mice bearing SKOV3 tumors. Tumor targeting specificity was evaluated in Balb/c mice using SKOV3, MDA-MB-361, and AT-3 (HER2-negative) xenografts. Results Affibody (HE)3ZHER2:V2, generated through recombinant gene expression, was successfully labeled with 99mTc, achieving a radiochemical purity of (96.0 ± 1.7)% (n = 3) as determined by RP-HPLC. This molecular probe exhibited specific binding to HER2-positive SKOV3 cells, demonstrating intense radioactive uptake. Biodistribution analysis showed rapid accumulation of 99mTc-(HE)3ZHER2:V2 in HER2-positive tumors post-administration, primarily clearing through the urinary system. Single-photon emission computed tomography imaging conducted 1-3 h after intravenous injection of 99mTc-(HE)3ZHER2:V2 into HER2-positive SKOV3 and MDA-MB-361 nude mouse models confirmed targeted uptake of the molecular probe by the tumors. Conclusions The molecular probe 99mTc-(HE)3ZHER2:V2 developed in this study effectively targets HER2 for imaging HER2-positive SKOV3 and MDA-MB-361 xenografts in vivo. It exhibits rapid blood clearance without evident toxic effects, suggesting its potential as a valuable marker for detecting HER2 expression in tumor cells.
Collapse
Affiliation(s)
- Xianwen Hu
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Huichuan, Zunyi, 563003, China
| | - Hongyu Hu
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Huichuan, Zunyi, 563003, China
| | - Dandan Li
- Department of Obstetrics, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou, Zunyi, 563003, China
| | - Pan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, China
| | - Jiong Cai
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan, Zunyi, 563003, China
| |
Collapse
|
39
|
Gomes DEB, Yang B, Vanella R, Nash MA, Bernardi RC. Integrating Dynamic Network Analysis with AI for Enhanced Epitope Prediction in PD-L1:Affibody Interactions. J Am Chem Soc 2024; 146:23842-23853. [PMID: 39146039 DOI: 10.1021/jacs.4c05869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Understanding binding epitopes involved in protein-protein interactions and accurately determining their structure are long-standing goals with broad applicability in industry and biomedicine. Although various experimental methods for binding epitope determination exist, these approaches are typically low throughput and cost-intensive. Computational methods have potential to accelerate epitope predictions; however, recently developed artificial intelligence (AI)-based methods frequently fail to predict epitopes of synthetic binding domains with few natural homologues. Here we have developed an integrated method employing generalized-correlation-based dynamic network analysis on multiple molecular dynamics (MD) trajectories, initiated from AlphaFold2Multimer structures, to unravel the structure and binding epitope of the therapeutic PD-L1:Affibody complex. Both AlphaFold2 and conventional molecular dynamics trajectory analysis were ineffective in distinguishing between two proposed binding models, parallel and perpendicular. However, our integrated approach, utilizing dynamic network analysis, demonstrated that the perpendicular mode was significantly more stable. These predictions were validated using a suite of experimental epitope mapping protocols, including cross-linking mass spectrometry and next-generation sequencing-based deep mutational scanning. Conversely, AlphaFold3 failed to predict a structure bound in the perpendicular pose, highlighting the necessity for exploratory research in the search for binding epitopes and challenging the notion that AI-generated protein structures can be accepted without scrutiny. Our research underscores the potential of employing dynamic network analysis to enhance AI-based structure predictions for more accurate identification of protein-protein interaction interfaces.
Collapse
Affiliation(s)
- Diego E B Gomes
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| | - Byeongseon Yang
- Institute of Physical Chemistry, Department of Chemistry, University of Basel, Basel 4058, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel 4058, Switzerland
| | - Rosario Vanella
- Institute of Physical Chemistry, Department of Chemistry, University of Basel, Basel 4058, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel 4058, Switzerland
| | - Michael A Nash
- Institute of Physical Chemistry, Department of Chemistry, University of Basel, Basel 4058, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel 4058, Switzerland
| | - Rafael C Bernardi
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
40
|
Xia X, Yang X, Gao W, Huang W, Xia X, Yan D. A novel HER2 targeting nanoagent self-assembled from affibody-epothilone B conjugate for cancer therapy. J Nanobiotechnology 2024; 22:502. [PMID: 39169343 PMCID: PMC11337599 DOI: 10.1186/s12951-024-02754-4] [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: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Epothilone B (Epo B), a promising antitumor compound effective against various types of cancer cells in vitro. However, its poor selectivity for tumor cells and inadequate therapeutic windows significantly limit its potential clinical application. Affibody is a class of non-immunoglobulin affinity proteins with precise targeting capability to overexpressed molecular receptors on cancer cells, has been intensively investigated due to its exceptional affinity properties. In this study, we present a targeted nanoagent self-assembled from the precursor of an affibody conjugated with Epo B via a linker containing the thioketal (tk) group that is sensitive to reactive oxygen species (ROS). The core-shell structure of the ZHER2:342-Epo B Affibody-Drug Conjugate Nanoagent (Z-E ADCN), with the cytotoxin Epo B encapsulated within the ZHER2:342 affibody corona, leads to significantly reduced side effects on normal organs. Moreover, the abundant presence of ZHER2:342 on the surface effectively enhances the targeting capacity and tumor accumulation of the drug. Z-E ADCN can be internalized by cancer cells via HER2 receptor-mediated endocytosis followed by Epo B release in response to high levels of ROS, resulting in excellent anticancer efficacy in HER2-positive tumor models.
Collapse
Affiliation(s)
- Xuelin Xia
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Wenhui Gao
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Xiaoxia Xia
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
41
|
Pham CV, Chowdhury R, Patel S, Melke H, Hou Y, Xu H, Jia L, Duan A, Duan W, Xiang D. The role of the size of affinity ligands in the detection and characterization of extracellular vesicles. Biosens Bioelectron 2024; 258:116381. [PMID: 38744116 DOI: 10.1016/j.bios.2024.116381] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Surface proteins on the membrane of nano-sized extracellular vesicles (EVs) not only play crucial roles in cell-to-cell communication, but also are specific binding targets for EV detection, isolation and tracking. The low abundance of protein biomarkers on EV surface, the formation of clusters and the complex EV surface network impose significant challenges to the study of EVs. Employing bulky sized affinity ligands, such as antibodies, in the detection and characterization of these vesicles often result in reduced sensitivity of detection or poor quantification of proteins on the EV surface. By virtue of their small size and high specificity, Affibody molecules emerge as a potential alternative to their monoclonal antibody counterparts as robust affinity ligands in EV research. In this study, we present a theoretical framework on the superiority of anti-HER2 Affibodies over anti-HER2 antibodies in labeling and detecting HER2-positive EVs, followed by the demonstration of the advantages of HER2 Affibodies in accessing EV surface and the detection of EVs through multiple types of approaches including fluorescence intensity, colorimetry, and fluorescence polarization. HER2 Affibodies outperformed by 10-fold over three HER2 antibody clones in accessing HER2-positive EVs derived from different human cancer cell lines. Furthermore, HRP-Affibody molecules could detect EVs from cancer cells spiked into human serum with at least a 2-fold higher sensitivity compared with that of their antibody counterparts. In addition, in fluorescence polarization assays in which no separation of free from bound ligand is required, FITC-labeled HER2 Affibodies could sensitively detect HER2-positive EVs with a clinically relevant limit of detection, whilst HER2 antibodies failed to detect EVs in the same conditions. With the demonstrated superiority in accessing and detecting surface targets over bulky-sized antibodies in EVs, Affibodies may become the next-generation of affinity ligands in the precise characterization and quantification of molecular architecture on the surface of EVs.
Collapse
Affiliation(s)
- Cuong Viet Pham
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Rocky Chowdhury
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Shweta Patel
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Haben Melke
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Yingchu Hou
- Laboratory of Tumor Molecular and Cellular Biology College of Life Sciences, Shaanxi Normal University 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Lee Jia
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Andrew Duan
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic, Australia
| | - Wei Duan
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia.
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, The Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
42
|
Ek M, Nilvebrant J, Nygren PÅ, Ståhl S, Lindberg H, Löfblom J. An anti-sortilin affibody-peptide fusion inhibits sortilin-mediated progranulin degradation. Front Immunol 2024; 15:1437886. [PMID: 39185427 PMCID: PMC11342335 DOI: 10.3389/fimmu.2024.1437886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Heterozygous loss-of-function mutations in the GRN gene are a common cause of frontotemporal dementia. Such mutations lead to decreased plasma and cerebrospinal fluid levels of progranulin (PGRN), a neurotrophic factor with lysosomal functions. Sortilin is a negative regulator of extracellular PGRN levels and has shown promise as a therapeutic target for frontotemporal dementia, enabling increased extracellular PGRN levels through inhibition of sortilin-mediated PGRN degradation. Here we report the development of a high-affinity sortilin-binding affibody-peptide fusion construct capable of increasing extracellular PGRN levels in vitro. By genetic fusion of a sortilin-binding affibody generated through phage display and a peptide derived from the progranulin C-terminus, an affinity protein (A3-PGRNC15*) with 185-pM affinity for sortilin was obtained. Treating PGRN-secreting and sortilin-expressing human glioblastoma U-251 cells with the fusion protein increased extracellular PGRN levels up to 2.5-fold, with an EC50 value of 1.3 nM. Our results introduce A3-PGRNC15* as a promising new agent with therapeutic potential for the treatment of frontotemporal dementia. Furthermore, the work highlights means to increase binding affinity through synergistic contribution from two orthogonal polypeptide units.
Collapse
Affiliation(s)
| | | | | | | | | | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
43
|
Pierzynowska K, Morcinek-Orłowska J, Gaffke L, Jaroszewicz W, Skowron PM, Węgrzyn G. Applications of the phage display technology in molecular biology, biotechnology and medicine. Crit Rev Microbiol 2024; 50:450-490. [PMID: 37270791 DOI: 10.1080/1040841x.2023.2219741] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 10/17/2022] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
The phage display technology is based on the presentation of peptide sequences on the surface of virions of bacteriophages. Its development led to creation of sophisticated systems based on the possibility of the presentation of a huge variability of peptides, attached to one of proteins of bacteriophage capsids. The use of such systems allowed for achieving enormous advantages in the processes of selection of bioactive molecules. In fact, the phage display technology has been employed in numerous fields of biotechnology, as diverse as immunological and biomedical applications (in both diagnostics and therapy), the formation of novel materials, and many others. In this paper, contrary to many other review articles which were focussed on either specific display systems or the use of phage display in selected fields, we present a comprehensive overview of various possibilities of applications of this technology. We discuss an usefulness of the phage display technology in various fields of science, medicine and the broad sense of biotechnology. This overview indicates the spread and importance of applications of microbial systems (exemplified by the phage display technology), pointing to the possibility of developing such sophisticated tools when advanced molecular methods are used in microbiological studies, accompanied with understanding of details of structures and functions of microbial entities (bacteriophages in this case).
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | | | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Weronika Jaroszewicz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
44
|
Imai M, Colas K, Suga H. Protein Grafting Techniques: From Peptide Epitopes to Lasso-Grafted Neobiologics. Chempluschem 2024; 89:e202400152. [PMID: 38693599 DOI: 10.1002/cplu.202400152] [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: 02/26/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/03/2024]
Abstract
Protein engineering techniques have vastly expanded their domain of impact, notably following the success of antibodies. Likewise, smaller peptide therapeutics have carved an increasingly significant niche for themselves in the pharmaceutical landscape. The concept of grafting such peptides onto larger protein scaffolds, thus harvesting the advantages of both, has given rise to a variety of protein engineering strategies that are reviewed herein. We also describe our own "Lasso-Grafting" approach, which combines traditional grafting concepts with mRNA display to streamline the production of multiple grafted drug candidates for virtually any target.
Collapse
Affiliation(s)
- Mikio Imai
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kilian Colas
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
45
|
Gralewska P, Gajek A, Marczak A, Rogalska A. Targeted Nanocarrier-Based Drug Delivery Strategies for Improving the Therapeutic Efficacy of PARP Inhibitors against Ovarian Cancer. Int J Mol Sci 2024; 25:8304. [PMID: 39125873 PMCID: PMC11312858 DOI: 10.3390/ijms25158304] [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: 06/19/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The current focus of ovarian cancer (OC) research is the improvement of treatment options through maximising drug effectiveness. OC remains the fifth leading cause of cancer-induced mortality in women worldwide. In recent years, nanotechnology has revolutionised drug delivery systems. Nanoparticles may be utilised as carriers in gene therapy or to overcome the problem of drug resistance in tumours by limiting the number of free drugs in circulation and thereby minimising undesired adverse effects. Cell surface receptors, such as human epidermal growth factor 2 (HER2), folic acid (FA) receptors, CD44 (also referred to as homing cell adhesion molecule, HCAM), and vascular endothelial growth factor (VEGF) are highly expressed in ovarian cancer cells. Generation of active targeting nanoparticles involves modification with ligands that recognise cell surface receptors and thereby promote internalisation by cancer cells. Several poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are currently used for the treatment of high-grade serous ovarian carcinomas (HGSOC) or platinum-sensitive relapsed OC. However, PARP resistance and poor drug bioavailability are common challenges, highlighting the urgent need to develop novel, effective strategies for ovarian cancer treatment. This review evaluates the utility of nanoparticles in ovarian cancer therapy, with a specific focus on targeted approaches and the use of PARPi nanocarriers to optimise treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Aneta Rogalska
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90–236 Lodz, Poland; (P.G.); (A.G.); (A.M.)
| |
Collapse
|
46
|
Kolesnikova OA, Komedchikova EN, Zvereva SD, Obozina AS, Dorozh OV, Afanasev I, Nikitin PI, Mochalova EN, Nikitin MP, Shipunova VO. Albumin incorporation into recognising layer of HER2-specific magnetic nanoparticles as a tool for optimal targeting of the acidic tumor microenvironment. Heliyon 2024; 10:e34211. [PMID: 39100472 PMCID: PMC11296017 DOI: 10.1016/j.heliyon.2024.e34211] [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: 06/25/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Cancer is unquestionably a global healthcare challenge, spurring the exporation of novel treatment approaches. In recent years, nanomaterials have garnered significant interest with the greatest hopes for targeted nanoformulations due to their cell-specific delivery, improved therapeutic efficacy, and reduced systemic toxicity for the organism. The problem of successful clinical translation of nanoparticles may be related to the fact that most in vitro tests are performed at pH values of normal cells and tissues, ranging from 7.2 to 7.4. The extracellular pH values of tumors are characterized by a shift to a more acidic region in the range of 5.6-7.0 and represent a crucial target for enhancing nanoparticle delivery to cancer cells. Here we show the method of non-active protein incorporation into the surface of HER2-targeted nanoparticles to achieve optimal cellular uptake within the pH range of the tumor microenvironment. The method efficacy was confirmed in vitro and in vivo showing the maximum binding of nanoparticles to cells at a pH value 6.4. Namely, fluorescent magnetic nanoparticles, modified with HER2-recognising affibody ZHER2:342, with proven specificity in terms of HER2 recognition (with 62-fold higher cellular uptake compared to control nanoparticles) were designed for targeting cancer cells at slightly acidic pH values. The stabilizing protein, namely, bovine serum albumin, one of the major blood components with widespread availability and biocompatibility, was used for the decoration of the nanoparticle surface to alter the pH response of the targeting magnetic conjugates. The optimally designed nanoparticles showed a bell-shaped dependency of interaction with cancer cells in the pH range of 5.6-8.0 with maximum cellular uptake at pH value 6.4 close to that of the tumor microenvironment. In vivo experiments revealed that after i.v. administration, BSA-decorated nanoparticles exhibited 2 times higher accumulation in tumors compared to magnetic nanoparticles modified with affibody only. Thus, we demonstrated a valid method for enhancing the specificity of targeted nanoparticle delivery to cancer cells without changing the functional components of nanoparticles.
Collapse
Affiliation(s)
- Olga A. Kolesnikova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
| | - Elena N. Komedchikova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Svetlana D. Zvereva
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | | | - Olha V. Dorozh
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
| | - Iurii Afanasev
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Petr I. Nikitin
- Prokhorov General Physics Institute, Russian Academy of Sciences, 38 Vavilov Street, 119991, Moscow, Russia
| | - Elizaveta N. Mochalova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Moscow Institute of Physics and Technology, 9 Institutskiy Per., 141701, Dolgoprudny, Russia
| | - Maxim P. Nikitin
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| | - Victoria O. Shipunova
- Moscow Center for Advanced Studies, Kulakova str. 20, 123592, Moscow, Russia
- Department of Nanobiomedicine, Sirius University of Science and Technology, 1 Olympic Ave., 354340, Sochi, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., 117997, Moscow, Russia
| |
Collapse
|
47
|
Mao M, Ahrens L, Luka J, Contreras F, Kurkina T, Bienstein M, Sárria Pereira de Passos M, Schirinzi G, Mehn D, Valsesia A, Desmet C, Serra MÁ, Gilliland D, Schwaneberg U. Material-specific binding peptides empower sustainable innovations in plant health, biocatalysis, medicine and microplastic quantification. Chem Soc Rev 2024; 53:6445-6510. [PMID: 38747901 DOI: 10.1039/d2cs00991a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Material-binding peptides (MBPs) have emerged as a diverse and innovation-enabling class of peptides in applications such as plant-/human health, immobilization of catalysts, bioactive coatings, accelerated polymer degradation and analytics for micro-/nanoplastics quantification. Progress has been fuelled by recent advancements in protein engineering methodologies and advances in computational and analytical methodologies, which allow the design of, for instance, material-specific MBPs with fine-tuned binding strength for numerous demands in material science applications. A genetic or chemical conjugation of second (biological, chemical or physical property-changing) functionality to MBPs empowers the design of advanced (hybrid) materials, bioactive coatings and analytical tools. In this review, we provide a comprehensive overview comprising naturally occurring MBPs and their function in nature, binding properties of short man-made MBPs (<20 amino acids) mainly obtained from phage-display libraries, and medium-sized binding peptides (20-100 amino acids) that have been reported to bind to metals, polymers or other industrially produced materials. The goal of this review is to provide an in-depth understanding of molecular interactions between materials and material-specific binding peptides, and thereby empower the use of MBPs in material science applications. Protein engineering methodologies and selected examples to tailor MBPs toward applications in agriculture with a focus on plant health, biocatalysis, medicine and environmental monitoring serve as examples of the transformative power of MBPs for various industrial applications. An emphasis will be given to MBPs' role in detecting and quantifying microplastics in high throughput, distinguishing microplastics from other environmental particles, and thereby assisting to close an analytical gap in food safety and monitoring of environmental plastic pollution. In essence, this review aims to provide an overview among researchers from diverse disciplines in respect to material-(specific) binding of MBPs, protein engineering methodologies to tailor their properties to application demands, re-engineering for material science applications using MBPs, and thereby inspire researchers to employ MBPs in their research.
Collapse
Affiliation(s)
- Maochao Mao
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Leon Ahrens
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Julian Luka
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Francisca Contreras
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Tetiana Kurkina
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | - Marian Bienstein
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| | | | | | - Dora Mehn
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Andrea Valsesia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Cloé Desmet
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Ulrich Schwaneberg
- Lehrstuhl für Biotechnologie, RWTH Aachen University, Worringerweg 3, 52074 Aachen, Germany.
| |
Collapse
|
48
|
Zhang J, Bodenko V, Larkina M, Bezverkhniaia E, Xu T, Liao Y, Abouzayed A, Plotnikov E, Tretyakova M, Yuldasheva F, Belousov MV, Orlova A, Tolmachev V, Gräslund T, Vorobyeva A. Half-life extension via ABD-fusion leads to higher tumor uptake of an affibody-drug conjugate compared to PAS- and XTENylation. J Control Release 2024; 370:468-478. [PMID: 38697314 DOI: 10.1016/j.jconrel.2024.04.051] [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: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
A critical parameter during the development of protein therapeutics is to endow them with suitable pharmacokinetic and pharmacodynamic properties. Small protein drugs are quickly eliminated by kidney filtration, and in vivo half-life extension is therefore often desired. Here, different half-life extension technologies were studied where PAS polypeptides (PAS300, PAS600), XTEN polypeptides (XTEN288, XTEN576), and an albumin binding domain (ABD) were compared for half-life extension of an anti-human epidermal growth factor receptor 2 (HER2) affibody-drug conjugate. The results showed that extension with the PAS or XTEN polypeptides or the addition of the ABD lowered the affinity for HER2 to some extent but did not negatively affect the cytotoxic potential. The half-lives in mice ranged from 7.3 h for the construct including PAS300 to 11.6 h for the construct including PAS600. The highest absolute tumor uptake was found for the construct including the ABD, which was 60 to 160% higher than the PASylated or XTENylated constructs, even though it did not have the longest half-life (9.0 h). A comparison of the tumor-to-normal-organ ratios showed the best overall performance of the ABD-fused construct. In conclusion, PASylation, XTENylation, and the addition of an ABD are viable strategies for half-life extension of affibody-drug conjugates, with the best performance observed for the construct including the ABD.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Scientific and Educational Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Maria Larkina
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Ekaterina Bezverkhniaia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Tianqi Xu
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Yunqi Liao
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Evgenii Plotnikov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Maria Tretyakova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Feruza Yuldasheva
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia
| | - Mikhail V Belousov
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk 634050, Russia; Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk 634050, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, Roslagstullsbacken 21, Stockholm 114 17, Sweden.
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
49
|
Yang B, Gomes DEB, Liu Z, Santos MS, Li J, Bernardi RC, Nash MA. Engineering the Mechanical Stability of a Therapeutic Affibody/PD-L1 Complex by Anchor Point Selection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595133. [PMID: 38826272 PMCID: PMC11142103 DOI: 10.1101/2024.05.21.595133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Protein-protein complexes can vary in mechanical stability depending on the direction from which force is applied. Here we investigated the anisotropic mechanical stability of a molecular complex between a therapeutic non-immunoglobulin scaffold called Affibody and the extracellular domain of the immune checkpoint protein PD-L1. We used a combination of single-molecule AFM force spectroscopy (AFM-SMFS) with bioorthogonal clickable peptide handles, shear stress bead adhesion assays, molecular modeling, and steered molecular dynamics (SMD) simulations to understand the pulling point dependency of mechanostability of the Affibody:(PD-L1) complex. We observed diverse mechanical responses depending on the anchor point. For example, pulling from residue #22 on Affibody generated an intermediate unfolding event attributed to partial unfolding of PD-L1, while pulling from Affibody's N-terminus generated force-activated catch bond behavior. We found that pulling from residue #22 or #47 on Affibody generated the highest rupture forces, with the complex breaking at up to ~ 190 pN under loading rates of ~104-105 pN/sec, representing a ~4-fold increase in mechanostability as compared with low force N-terminal pulling. SMD simulations provided consistent tendencies in rupture forces, and through visualization of force propagation networks provided mechanistic insights. These results demonstrate how mechanostability of therapeutic protein-protein interfaces can be controlled by informed selection of anchor points within molecules, with implications for optimal bioconjugation strategies in drug delivery vehicles.
Collapse
Affiliation(s)
- Byeongseon Yang
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Diego E. B. Gomes
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| | - Zhaowei Liu
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
- Present address: Department of Bionanoscience, Delft University of Technology, 2629HZ Delft, the Netherlands
| | - Mariana Sá Santos
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Jiajun Li
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| | - Rafael C. Bernardi
- Department of Physics, Auburn University, Auburn, Alabama 36849, United States
| | - Michael A. Nash
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4056 Basel, Switzerland
| |
Collapse
|
50
|
Russo F, Civili B, Winssinger N. Bright Red Bioluminescence from Semisynthetic NanoLuc (sNLuc). ACS Chem Biol 2024; 19:1035-1039. [PMID: 38717306 PMCID: PMC11106743 DOI: 10.1021/acschembio.4c00033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
Red-shifted bioluminescence is highly desirable for diagnostic and imaging applications. Herein, we report a semisynthetic NanoLuc (sNLuc) based on complementation of a split NLuc (LgBiT) with a synthetic peptide (SmBiT) functionalized with a fluorophore for BRET emission. We observed exceptional BRET ratios with diverse fluorophores, notably in the red (I674/I450 > 14), with a brightness that is sufficient for naked eye detection in blood or through tissues. To exemplify its utility, LgBiT was fused to a miniprotein that binds HER2 (affibody, ZHER2), and the selective detection of HER2+ SK-BR-3 cells over HER2- HeLa cells was demonstrated.
Collapse
Affiliation(s)
- Francesco Russo
- Department of Organic Chemistry,
Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Beatrice Civili
- Department of Organic Chemistry,
Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Nicolas Winssinger
- Department of Organic Chemistry,
Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|