1
|
De Groof TWM, Lauwers Y, De Pauw T, Saxena M, Vincke C, Van Craenenbroeck J, Chapon C, Le Grand R, Raes G, Naninck T, Van Ginderachter JA, Devoogdt N. Specific imaging of CD8 + T-Cell dynamics with a nanobody radiotracer against human CD8β. Eur J Nucl Med Mol Imaging 2024; 52:193-207. [PMID: 39218831 PMCID: PMC11599301 DOI: 10.1007/s00259-024-06896-3] [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: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE While immunotherapy has revolutionized the oncology field, variations in therapy responsiveness limit the broad applicability of these therapies. Diagnostic imaging of immune cell, and specifically CD8+ T cell, dynamics could allow early patient stratification and result in improved therapy efficacy and safety. In this study, we report the development of a nanobody-based immunotracer for non-invasive SPECT and PET imaging of human CD8+ T-cell dynamics. METHODS Nanobodies targeting human CD8β were generated by llama immunizations and subsequent biopanning. The lead anti-human CD8β nanobody was characterized on binding, specificity, stability and toxicity. The lead nanobody was labeled with technetium-99m, gallium-68 and copper-64 for non-invasive imaging of human T-cell lymphomas and CD8+ T cells in human CD8 transgenic mice and non-human primates by SPECT/CT or PET/CT. Repeated imaging of CD8+ T cells in MC38 tumor-bearing mice allowed visualization of CD8+ T-cell dynamics. RESULTS The nanobody-based immunotracer showed high affinity and specific binding to human CD8 without unwanted immune activation. CD8+ T cells were non-invasively visualized by SPECT and PET imaging in naïve and tumor-bearing mice and in naïve non-human primates with high sensitivity. The nanobody-based immunotracer showed enhanced specificity for CD8+ T cells and/or faster in vivo pharmacokinetics compared to previous human CD8-targeting immunotracers, allowing us to follow human CD8+ T-cell dynamics already at early timepoints. CONCLUSION This study describes the development of a more specific human CD8+ T-cell-targeting immunotracer, allowing follow-up of immunotherapy responses by non-invasive imaging of human CD8+ T-cell dynamics.
Collapse
Affiliation(s)
- Timo W M De Groof
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Yoline Lauwers
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tessa De Pauw
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mohit Saxena
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Cécile Vincke
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jolien Van Craenenbroeck
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Catherine Chapon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Geert Raes
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thibaut Naninck
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Fontenay-aux-Roses & Le Kremlin-Bicêtre, Inserm, Paris, CEA, France
| | - Jo A Van Ginderachter
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Molecular Imaging and Therapy Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
2
|
Basuli F, Shi J, Lindberg E, Fayn S, Lee W, Ho M, Hammoud DA, Cheloha RW, Swenson RE, Escorcia FE. Sortase-Mediated Site-Specific Conjugation to Prepare Fluorine-18-Labeled Nanobodies. Bioconjug Chem 2024; 35:1335-1342. [PMID: 39172920 DOI: 10.1021/acs.bioconjchem.4c00264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Single-domain antibodies, or nanobodies (Nbs), are promising biomolecules for use in molecular imaging due to their excellent affinity, specificity, and fast clearance from the blood. Given their short blood half-life, pairing Nbs with short-lived imaging radioisotopes is desirable. Because fluorine-18 (18F) is routinely used for clinical imaging, it is an attractive radioisotope for Nbs. We report a novel sortase-based, site-specific 18F-labeling method applied to three nanobodies. Labeled nanobodies were synthesized either by a two-step indirect radiolabeling method in one pot or by a one-step direct labeling method using a sortase-mediated conjugation of either the radiolabeled chelator (H-GGGK((±)-Al[18F]FH3RESCA)-NH2) or the unlabeled chelator (H-GGGK((±)-H3RESCA)-NH2) followed by labeling with Al[18F]F, respectively. The overall radiochemical yields were 15-43% (n = 22, decay-corrected) in 70 min (indirect labeling) and 23-58% (n = 12, decay-corrected) in 50 min (direct labeling). The radiochemical purities of the labeled nanobodies prepared by both methods were >98% with a specific activity of 400-600 Ci/mmol (n = 22) for each of the three Nbs tested and exhibited excellent stability profiles under physiological conditions. This simple, site-specific, reproducible, and generalizable 18F-labeling method to prepare nanobodies (Nb-Al[18F]F-RESCA) or other low molecular weight biomolecules can easily be adopted in various settings for preclinical and clinical studies.
Collapse
Affiliation(s)
- Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Jianfeng Shi
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Eric Lindberg
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Stanley Fayn
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, U.K
| | - Woonghee Lee
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Ross W Cheloha
- Chemical Biology in Signaling Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, Maryland 20892-0001, United States
| | - Freddy E Escorcia
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
3
|
Nemecz D, Nowak WA, Nemecz Á. VHH Nanobody Versatility against Pentameric Ligand-Gated Ion Channels. J Med Chem 2024; 67:8502-8518. [PMID: 38829690 PMCID: PMC11181324 DOI: 10.1021/acs.jmedchem.4c00231] [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: 01/26/2024] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
Pentameric ligand-gated ion channels provide rapid chemical-electrical signal transmission between cells in the central and peripheral nervous system. Their dysfunction is associated with many nervous system disorders. They are composed of five identical (homomeric receptors) or homologous (heteromeric receptors) subunits. VHH nanobodies, or single-chain antibodies, are the variable domain, VHH, of antibodies that are composed of the heavy chain only from camelids. Their unique structure results in many specific biochemical and biophysical properties that make them an excellent alternative to conventional antibodies. This Perspective explores the published VHH nanobodies which have been isolated against pentameric ligand-gated ion channel subfamilies. It outlines the genetic and chemical modifications available to alter nanobody function. An assessment of the available functional and structural data indicate that it is feasible to create therapeutic agents and impart, through their modification, a given desired modulatory effect of its target receptor for current stoichiometric-specific VHH nanobodies.
Collapse
Affiliation(s)
- Dorota Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Weronika A. Nowak
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| | - Ákos Nemecz
- Biochemistry
Department, Nicolaus Copernicus University
in Torun, 87-100 Torun, Poland
| |
Collapse
|
4
|
Tripathy RK, Pande AH. Molecular and functional insight into anti-EGFR nanobody: Theranostic implications for malignancies. Life Sci 2024; 345:122593. [PMID: 38554946 DOI: 10.1016/j.lfs.2024.122593] [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: 01/09/2024] [Revised: 02/27/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Targeted therapy and imaging are the most popular techniques for the intervention and diagnosis of cancer. A potential therapeutic target for the treatment of cancer is the epidermal growth factor receptor (EGFR), primarily for glioblastoma, lung, and breast cancer. Over-production of ligand, transcriptional up-regulation due to autocrine/paracrine signalling, or point mutations at the genomic locus may contribute to the malfunction of EGFR in malignancies. This exploit makes use of EGFR, an established biomarker for cancer diagnostics and treatment. Despite considerable development in the last several decades in making EGFR inhibitors, they are still not free from limitations like toxicity and a short serum half-life. Nanobodies and antibodies share similar binding properties, but nanobodies have the additional advantage that they can bind to antigenic epitopes deep inside the target that conventional antibodies are unable to access. For targeted therapy, anti-EGFR nanobodies can be conjugated to various molecules such as drugs, peptides, toxins and photosensitizers. These nanobodies can be designed as novel immunoconjugates using the universal modular antibody-based platform technology (UniCAR). Furthermore, Anti-EGFR nanobodies can be expressed in neural stem cells and visualised by effective fluorescent and radioisotope labelling.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, (Mohali) 160062, Punjab, India.
| |
Collapse
|
5
|
Vandewalle N, Satilmis H, Verheye E, Fan R, Wang Y, De Groof TW, Bridoux J, Kerre T, De Beule N, De Becker A, De Bruyne E, Menu E, Vanderkerken K, Breckpot K, Devoogdt N, De Veirman K. AXL-specific single domain antibodies show diagnostic potential and anti-tumor activity in Acute Myeloid Leukemia. Theranostics 2024; 14:2656-2674. [PMID: 38773967 PMCID: PMC11103505 DOI: 10.7150/thno.91456] [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: 10/24/2023] [Accepted: 03/16/2024] [Indexed: 05/24/2024] Open
Abstract
Rationale: AXL expression has been identified as a prognostic factor in acute myeloid leukemia (AML) and is detectable in approximately 50% of AML patients. In this study, we developed AXL-specific single domain antibodies (sdAbs), cross-reactive for both mouse and human AXL protein, to non-invasively image and treat AXL-expressing cancer cells. Methods: AXL-specific sdAbs were induced by immunizing an alpaca with mouse and human AXL proteins. SdAbs were characterized using ELISA, flow cytometry, surface plasmon resonance and the AlphaFold2 software. A lead compound was selected and labeled with 99mTc for evaluation as a diagnostic tool in mouse models of human (THP-1 cells) or mouse (C1498 cells) AML using SPECT/CT imaging. For therapeutic purposes, the lead compound was fused to a mouse IgG2a-Fc tail and in vitro functionality tests were performed including viability, apoptosis and proliferation assays in human AML cell lines and primary patient samples. Using these in vitro models, its anti-tumor effect was evaluated as a single agent, and in combination with standard of care agents venetoclax or cytarabine. Results: Based on its cell binding potential, cross-reactivity, nanomolar affinity and GAS6/AXL blocking capacity, we selected sdAb20 for further evaluation. Using SPECT/CT imaging, we observed tumor uptake of 99mTc-sdAb20 in mice with AXL-positive THP-1 or C1498 tumors. In THP-1 xenografts, an optimized protocol using pre-injection of cold sdAb20-Fc was required to maximize the tumor-to-background signal. Besides its diagnostic value, we observed a significant reduction in tumor cell proliferation and viability using sdAb20-Fc in vitro. Moreover, combining sdAb20-Fc and cytarabine synergistically induced apoptosis in human AML cell lines, while these effects were less clear when combined with venetoclax. Conclusions: Because of their diagnostic potential, sdAbs could be used to screen patients eligible for AXL-targeted therapy and to follow-up AXL expression during treatment and disease progression. When fused to an Fc-domain, sdAbs acquire additional therapeutic properties that can lead to a multidrug approach for the treatment of AXL-positive cancer patients.
Collapse
Affiliation(s)
- Niels Vandewalle
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Hatice Satilmis
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Emma Verheye
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Brussels Center of Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rong Fan
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Yanmeng Wang
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Timo W.M. De Groof
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Jessica Bridoux
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Tessa Kerre
- Department of Hematology, Ghent University Hospital, Ghent, Belgium
| | - Nathan De Beule
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ann De Becker
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Translational Oncology Research Center (TORC), Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Laboratory of Molecular Imaging and Therapy (MITH), Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Vrije Universiteit Brussel, Brussels, Belgium
- Translational Oncology Research Center (TORC), team Hematology and Immunology (HEIM), Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
6
|
Kunz S, Durandy M, Seguin L, Feral CC. NANOBODY ® Molecule, a Giga Medical Tool in Nanodimensions. Int J Mol Sci 2023; 24:13229. [PMID: 37686035 PMCID: PMC10487883 DOI: 10.3390/ijms241713229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Although antibodies remain the most widely used tool for biomedical research, antibody technology is not flawless. Innovative alternatives, such as Nanobody® molecules, were developed to address the shortcomings of conventional antibodies. Nanobody® molecules are antigen-binding variable-domain fragments derived from the heavy-chain-only antibodies of camelids (VHH) and combine the advantageous properties of small molecules and monoclonal antibodies. Nanobody® molecules present a small size (~15 kDa, 4 nm long and 2.5 nm wide), high solubility, stability, specificity, and affinity, ease of cloning, and thermal and chemical resistance. Recombinant production in microorganisms is cost-effective, and VHH are also building blocks for multidomain constructs. These unique features led to numerous applications in fundamental research, diagnostics, and therapy. Nanobody® molecules are employed as biomarker probes and, when fused to radioisotopes or fluorophores, represent ideal non-invasive in vivo imaging agents. They can be used as neutralizing agents, receptor-ligand antagonists, or in targeted vehicle-based drug therapy. As early as 2018, the first Nanobody®, Cablivi (caplacizumab), a single-domain antibody (sdAb) drug developed by French pharmaceutical giant Sanofi for the treatment of adult patients with acquired thrombocytopenic purpura (aTTP), was launched. Nanobody® compounds are ideal tools for further development in clinics for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Sarah Kunz
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
- Department of Oncology, Sanofi Research Center, 94400 Vitry-sur-Seine, France
| | - Manon Durandy
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| | - Laetitia Seguin
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| | - Chloe C. Feral
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| |
Collapse
|
7
|
Ionescu RE. Ultrasensitive Electrochemical Immunosensors Using Nanobodies as Biocompatible Sniffer Tools of Agricultural Contaminants and Human Disease Biomarkers. MICROMACHINES 2023; 14:1486. [PMID: 37630022 PMCID: PMC10456424 DOI: 10.3390/mi14081486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023]
Abstract
Nanobodies (Nbs) are known as camelid single-domain fragments or variable heavy chain antibodies (VHH) that in vitro recognize the antigens (Ag) similar to full-size antibodies (Abs) and in vivo allow immunoreactions with biomolecule cavities inaccessible to conventional Abs. Currently, Nbs are widely used for clinical treatments due to their remarkably improved performance, ease of production, thermal robustness, superior physical and chemical properties. Interestingly, Nbs are also very promising bioreceptors for future rapid and portable immunoassays, compared to those using unstable full-size antibodies. For all these reasons, Nbs are excellent candidates in ecological risk assessments and advanced medicine, enabling the development of ultrasensitive biosensing platforms. In this review, immobilization strategies of Nbs on conductive supports for enhanced electrochemical immune detection of food contaminants (Fcont) and human biomarkers (Hbio) are discussed. In the case of Fcont, the direct competitive immunoassay detection using coating antigen solid surface is the most commonly used approach for efficient Nbs capture which was characterized with cyclic voltammetry (CV) and differential pulse voltammetry (DPV) when the signal decays for increasing concentrations of free antigen prepared in aqueous solutions. In contrast, for the Hbio investigations on thiolated gold electrodes, increases in amperometric and electrochemical impedance spectroscopy (EIS) signals were recorded, with increases in the antigen concentrations prepared in PBS or spiked real human samples.
Collapse
Affiliation(s)
- Rodica Elena Ionescu
- Light, Nanomaterials and Nanotechnology (L2n) Laboratory, CNRS EMR 7004, University of Technology of Troyes, 12 Rue Marie Curie CS 42060, 10004 Troyes, France
| |
Collapse
|
8
|
Hurley K, Cao M, Huang H, Wang Y. Targeted Alpha Therapy (TAT) with Single-Domain Antibodies (Nanobodies). Cancers (Basel) 2023; 15:3493. [PMID: 37444603 DOI: 10.3390/cancers15133493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Meiyun Cao
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Haiming Huang
- Research Center, Forlong Biotechnology Inc., Suzhou 215004, China
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
9
|
De Pauw T, De Mey L, Debacker JM, Raes G, Van Ginderachter JA, De Groof TWM, Devoogdt N. Current status and future expectations of nanobodies in oncology trials. Expert Opin Investig Drugs 2023; 32:705-721. [PMID: 37638538 DOI: 10.1080/13543784.2023.2249814] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Monoclonal antibodies have revolutionized personalized medicine for cancer in recent decades. Despite their broad application in oncology, their large size and complexity may interfere with successful tumor targeting for certain applications of cancer diagnosis and therapy. Nanobodies have unique structural and pharmacological features compared to monoclonal antibodies and have successfully been used as complementary anti-cancer diagnostic and/or therapeutic tools. AREAS COVERED Here, an overview is given of the nanobody-based diagnostics and therapeutics that have been or are currently being tested in oncological clinical trials. Furthermore, preclinical developments, which are likely to be translated into the clinic in the near future, are highlighted. EXPERT OPINION Overall, the presented studies show the application potential of nanobodies in the field of oncology, making it likely that more nanobodies will be clinically approved in the upcoming future.
Collapse
Affiliation(s)
- Tessa De Pauw
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn De Mey
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Timo W M De Groof
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
10
|
Maier A, Toner YC, Munitz J, Sullivan NA, Sakurai K, Meerwaldt AE, Brechbühl EE, Prévot G, van Elsas Y, Maas RJ, Ranzenigo A, Soultanidis G, Rashidian M, Pérez-Medina C, Heo GS, Gropler RJ, Liu Y, Reiner T, Nahrendorf M, Swirski FK, Strijkers GJ, Teunissen AJ, Calcagno C, Fayad ZA, Mulder WJ, van Leent MM. Multiparametric Immunoimaging Maps Inflammatory Signatures in Murine Myocardial Infarction Models. JACC Basic Transl Sci 2023; 8:801-816. [PMID: 37547068 PMCID: PMC10401290 DOI: 10.1016/j.jacbts.2022.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 08/08/2023]
Abstract
In the past 2 decades, research on atherosclerotic cardiovascular disease has uncovered inflammation to be a key driver of the pathophysiological process. A pressing need therefore exists to quantitatively and longitudinally probe inflammation, in preclinical models and in cardiovascular disease patients, ideally using non-invasive methods and at multiple levels. Here, we developed and employed in vivo multiparametric imaging approaches to investigate the immune response following myocardial infarction. The myocardial infarction models encompassed either transient or permanent left anterior descending coronary artery occlusion in C57BL/6 and Apoe-/-mice. We performed nanotracer-based fluorine magnetic resonance imaging and positron emission tomography (PET) imaging using a CD11b-specific nanobody and a C-C motif chemokine receptor 2-binding probe. We found that immune cell influx in the infarct was more pronounced in the permanent occlusion model. Further, using 18F-fluorothymidine and 18F-fluorodeoxyglucose PET, we detected increased hematopoietic activity after myocardial infarction, with no difference between the models. Finally, we observed persistent systemic inflammation and exacerbated atherosclerosis in Apoe-/- mice, regardless of which infarction model was used. Taken together, we showed the strengths and capabilities of multiparametric imaging in detecting inflammatory activity in cardiovascular disease, which augments the development of clinical readouts.
Collapse
Affiliation(s)
- Alexander Maier
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Cardiology and Angiology I, Heart Center of Freiburg University, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yohana C. Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Nathaniel A.T. Sullivan
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ken Sakurai
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Anu E. Meerwaldt
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Biomedical Magnetic Resonance Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht/Utrecht University, Utrecht, the Netherlands
| | - Eliane E.S. Brechbühl
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Geoffrey Prévot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yuri van Elsas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne J.F. Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Georgios Soultanidis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Gyu Seong Heo
- Department of Radiology, Washington University, St Louis, Missouri, USA
| | - Robert J. Gropler
- Department of Radiology, Washington University, St Louis, Missouri, USA
| | - Yongjian Liu
- Department of Radiology, Washington University, St Louis, Missouri, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Department of Radiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Filip K. Swirski
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gustav J. Strijkers
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Abraham J.P. Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zahi A. Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Willem J.M. Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Chemical Biology, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mandy M.T. van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
11
|
Benloucif A, Meyer D, Balasse L, Goubard A, Danner L, Bouhlel A, Castellano R, Guillet B, Chames P, Kerfelec B. Rapid nanobody-based imaging of mesothelin expressing malignancies compatible with blocking therapeutic antibodies. Front Immunol 2023; 14:1200652. [PMID: 37388728 PMCID: PMC10303918 DOI: 10.3389/fimmu.2023.1200652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/25/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction Mesothelin (MSLN) is overexpressed in a wide variety of cancers with few therapeutic options and has recently emerged as an attractive target for cancer therapy, with a large number of approaches currently under preclinical and clinical investigation. In this respect, developing mesothelin specific tracers as molecular companion tools for predicting patient eligibility, monitoring then response to mesothelin-targeting therapies, and tracking the evolution of the disease or for real-time visualisation of tumours during surgery is of growing importance. Methods We generated by phage display a nanobody (Nb S1) and used enzymatic approaches were used to site-directed conjugate Nb S1 with either ATTO 647N fluorochrome or NODAGA chelator for fluorescence and positron emission tomography imaging (PET) respectively. Results We demonstrated that Nb S1 displays a high apparent affinity and specificity for human mesothelin and demonstrated that the binding, although located in the membrane distal domain of mesothelin, is not impeded by the presence of MUC16, the only known ligand of mesothelin, nor by the therapeutic antibody amatuximab. In vivo experiments showed that both ATTO 647N and [68Ga]Ga-NODAGA-S1 rapidly and specifically accumulated in mesothelin positive tumours compared to mesothelin negative tumours or irrelevant Nb with a high tumour/background ratio. The ex vivo biodistribution profile analysis also confirmed a significantly higher uptake of Nb S1 in MSLN-positive tumours than in MSLNlow tumours. Conclusion We demonstrated for the first time the use of an anti-MSLN nanobody as PET radiotracer for same day imaging of MSLN+ tumours, targeting an epitope compatible with the monitoring of amatuximab-based therapies and current SS1-derived-drug conjugates.
Collapse
Affiliation(s)
- Abdennour Benloucif
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Damien Meyer
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Laure Balasse
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Armelle Goubard
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Lucile Danner
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ahlem Bouhlel
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Rémy Castellano
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Benjamin Guillet
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Brigitte Kerfelec
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
12
|
Jiang Y, Lin Y, Krishnaswamy S, Pan R, Wu Q, Sandusky-Beltran LA, Liu M, Kuo MH, Kong XP, Congdon EE, Sigurdsson EM. Single-domain antibody-based noninvasive in vivo imaging of α-synuclein or tau pathology. SCIENCE ADVANCES 2023; 9:eadf3775. [PMID: 37163602 PMCID: PMC10171817 DOI: 10.1126/sciadv.adf3775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/11/2023] [Indexed: 05/12/2023]
Abstract
Intracellular deposition of α-synuclein and tau are hallmarks of synucleinopathies and tauopathies, respectively. Recently, several dye-based imaging probes with selectivity for tau aggregates have been developed, but suitable imaging biomarkers for synucleinopathies are still unavailable. Detection of both of these aggregates early in the disease process may allow for prophylactic therapies before functional impairments have manifested, highlighting the importance of developing specific imaging probes for these lesions. In contrast to the β sheet dyes, single-domain antibodies, found in camelids and a few other species, are highly specific, and their small size allows better brain entry and distribution than whole antibodies. Here, we have developed such imaging ligands via phage display libraries derived from llamas immunized with α-synuclein and tau preparations, respectively. These probes allow noninvasive and specific in vivo imaging of α-synuclein versus tau pathology in mice, with the brain signal correlating strongly with lesion burden. These small antibody derivatives have great potential for in vivo diagnosis of these diseases.
Collapse
Affiliation(s)
- Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Yan Lin
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Senthilkumar Krishnaswamy
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Qian Wu
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Leslie A. Sandusky-Beltran
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Erin E. Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Einar M. Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| |
Collapse
|
13
|
Ma L, Brecher M, Soufal A, Gaiotto T, Tian S, Chandramouli S, Dewar V, Ferrant L, Zhang M, Zhou X, Roy V. Structural interrogation of a trimeric prefusion RSV fusion protein vaccine candidate by a camelid nanobody. Vaccine 2023; 41:3308-3316. [PMID: 37085457 DOI: 10.1016/j.vaccine.2023.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
In the past decade, camelid nanobodies have been developed for multiple applications, including immuno-imaging, cancer immunotherapy, and antiviral therapeutics. Despite the prevalence of these approaches, nanobodies have rarely been used to assess the potency of vaccine antigen candidates, which are primarily based on mAb binding approaches. In this work, we demonstrate that a nanobody-based ELISA method is suitable for characterization of a leading respiratory syncytial virus (RSV) vaccine candidate, RSVPreF3. This nanobody, F-VHH-L66, compares similarly with AM14, an antibody well-known to be specific for the prefusion form of the RSV surface fusion glycoprotein, RSV F. ELISA assays based on F-VHH-L66 were specific for the trimeric, prefusion form of RSV F, the antigen conformation that best generates neutralizing antibodies. Additionally, the F-VHH-L66-based ELISA proved accurate, linear, and stability-indicating. Statistical analysis of 65 independent F-VHH-L66-based ELISA experiments indicated assay performance similar to that of ELISA assays based on AM14. Moreover, the binding kinetics of F-VHH-L66 to RSVPreF3 are comparable to those of AM14 when measured by surface plasmon resonance (SPR). Finally, F-VHH-L66 neutralized RSV(A) with similar efficacy as AM14; this bioactivity data further supports its use as an alternative to AM14 for pre-fusion specific structural characterization of RSVPreF3.
Collapse
Affiliation(s)
- Li Ma
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Matthew Brecher
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States.
| | - Allison Soufal
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | | | - Sai Tian
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Sumana Chandramouli
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | | | | | - Meng Zhang
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Xianzhi Zhou
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Varnika Roy
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States.
| |
Collapse
|
14
|
Tang H, Gao Y, Han J. Application Progress of the Single Domain Antibody in Medicine. Int J Mol Sci 2023; 24:ijms24044176. [PMID: 36835588 PMCID: PMC9967291 DOI: 10.3390/ijms24044176] [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: 01/17/2023] [Revised: 02/07/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The camelid-derived single chain antibody (sdAb), also termed VHH or nanobody, is a unique, functional heavy (H)-chain antibody (HCAb). In contrast to conventional antibodies, sdAb is a unique antibody fragment consisting of a heavy-chain variable domain. It lacks light chains and a first constant domain (CH1). With a small molecular weight of only 12~15 kDa, sdAb has a similar antigen-binding affinity to conventional Abs but a higher solubility, which exerts unique advantages for the recognition and binding of functional, versatile, target-specific antigen fragments. In recent decades, with their unique structural and functional features, nanobodies have been considered promising agents and alternatives to traditional monoclonal antibodies. As a new generation of nano-biological tools, natural and synthetic nanobodies have been used in many fields of biomedicine, including biomolecular materials, biological research, medical diagnosis and immune therapies. This article briefly overviews the biomolecular structure, biochemical properties, immune acquisition and phage library construction of nanobodies and comprehensively reviews their applications in medical research. It is expected that this review will provide a reference for the further exploration and unveiling of nanobody properties and function, as well as a bright future for the development of drugs and therapeutic methods based on nanobodies.
Collapse
Affiliation(s)
- Huaping Tang
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuan Gao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
- Correspondence:
| | - Jiangyuan Han
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
15
|
Mulero F, Oteo M, Garaulet G, Magro N, Rebollo L, Medrano G, Santiveri C, Romero E, Sellek RE, Margolles Y, Campos-Olivas R, Arroyo AG, Fernández LA, Morcillo MA, Martínez-Torrecuadrada JL. Development of anti-membrane type 1-matrix metalloproteinase nanobodies as immunoPET probes for triple negative breast cancer imaging. Front Med (Lausanne) 2022; 9:1058455. [PMID: 36507540 PMCID: PMC9729729 DOI: 10.3389/fmed.2022.1058455] [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: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by aggressiveness and high rates of metastasis. The identification of relevant biomarkers is crucial to improve outcomes for TNBC patients. Membrane type 1-matrix metalloproteinase (MT1-MMP) could be a good candidate because its expression has been reported to correlate with tumor malignancy, progression and metastasis. Moreover, single-domain variable regions (VHHs or Nanobodies) derived from camelid heavy-chain-only antibodies have demonstrated improvements in tissue penetration and blood clearance, important characteristics for cancer imaging. Here, we have developed a nanobody-based PET imaging strategy for TNBC detection that targets MT1-MMP. A llama-derived library was screened against the catalytic domain of MT1-MMP and a panel of specific nanobodies were identified. After a deep characterization, two nanobodies were selected to be labeled with gallium-68 (68Ga). ImmunoPET imaging with both ([68Ga]Ga-NOTA-3TPA14 and [68Ga]Ga-NOTA-3CMP75) in a TNBC mouse model showed precise tumor-targeting capacity in vivo with high signal-to-background ratios. (68Ga)Ga-NOTA-3CMP75 exhibited higher tumor uptake compared to (68Ga)Ga-NOTA-3TPA14. Furthermore, imaging data correlated perfectly with the immunohistochemistry staining results. In conclusion, we found a promising candidate for nanobody-based PET imaging to be further investigated as a diagnostic tool in TNBC.
Collapse
Affiliation(s)
- Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Oteo
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Natalia Magro
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Lluvia Rebollo
- Protein Production Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Clara Santiveri
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Eduardo Romero
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Ricela E. Sellek
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, (CNB-CSIC), Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alicia G. Arroyo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Luis Angel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, (CNB-CSIC), Madrid, Spain
| | - Miguel Angel Morcillo
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain,*Correspondence: Miguel Angel Morcillo,
| | | |
Collapse
|
16
|
Tran LH, Graulus GJ, Vincke C, Smiejkowska N, Kindt A, Devoogdt N, Muyldermans S, Adriaensens P, Guedens W. Nanobodies for the Early Detection of Ovarian Cancer. Int J Mol Sci 2022; 23:ijms232213687. [PMID: 36430166 PMCID: PMC9691119 DOI: 10.3390/ijms232213687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Ovarian cancer ranks fifth in cancer-related deaths among women. Since ovarian cancer patients are often asymptomatic, most patients are diagnosed only at an advanced stage of disease. This results in a 5-year survival rate below 50%, which is in strong contrast to a survival rate as high as 94% if detected and treated at an early stage. Monitoring serum biomarkers offers new possibilities to diagnose ovarian cancer at an early stage. In this study, nanobodies targeting the ovarian cancer biomarkers human epididymis protein 4 (HE4), secretory leukocyte protease inhibitor (SLPI), and progranulin (PGRN) were evaluated regarding their expression levels in bacterial systems, epitope binning, and antigen-binding affinity by enzyme-linked immunosorbent assay and surface plasmon resonance. The selected nanobodies possess strong binding affinities for their cognate antigens (KD~0.1-10 nM) and therefore have a pronounced potential to detect ovarian cancer at an early stage. Moreover, it is of utmost importance that the limits of detection (LOD) for these biomarkers are in the pM range, implying high specificity and sensitivity, as demonstrated by values in human serum of 37 pM for HE4, 163 pM for SLPI, and 195 pM for PGRN. These nanobody candidates could thus pave the way towards multiplexed biosensors.
Collapse
Affiliation(s)
- Lan-Huong Tran
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, BE-3590 Diepenbeek, Belgium
| | - Geert-Jan Graulus
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, BE-3590 Diepenbeek, Belgium
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, BE-1050 Brussels, Belgium
| | - Natalia Smiejkowska
- Laboratory of Medical Biochemistry, University of Antwerp, Prinsstraat 13, BE-2000 Antwerpen, Belgium
| | - Anne Kindt
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, BE-1050 Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Pleinlaan 2, BE-1050 Brussels, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, BE-1050 Brussels, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, BE-3590 Diepenbeek, Belgium
- Analytical and Circular Chemistry, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, BE-3590 Diepenbeek, Belgium
- Correspondence:
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, BE-3590 Diepenbeek, Belgium
| |
Collapse
|
17
|
Lugat A, Bailly C, Chérel M, Rousseau C, Kraeber-Bodéré F, Bodet-Milin C, Bourgeois M. Immuno-PET: Design options and clinical proof-of-concept. Front Med (Lausanne) 2022; 9:1026083. [PMID: 36314010 PMCID: PMC9613928 DOI: 10.3389/fmed.2022.1026083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Radioimmunoconjugates have been used for over 30 years in nuclear medicine applications. In the last few years, advances in cancer biology knowledge have led to the identification of new molecular targets specific to certain patient subgroups. The use of these targets in targeted therapies approaches has allowed the developments of specifically tailored therapeutics for patients. As consequence of the PET-imaging progresses, nuclear medicine has developed powerful imaging tools, based on monoclonal antibodies, to in vivo characterization of these tumor biomarkers. This imaging modality known as immuno-positron emission tomography (immuno-PET) is currently in fastest-growing and its medical value lies in its ability to give a non-invasive method to assess the in vivo target expression and distribution and provide key-information on the tumor targeting. Currently, immuno-PET presents promising probes for different nuclear medicine topics as staging/stratification tool, theranostic approaches or predictive/prognostic biomarkers. To develop a radiopharmaceutical drug that can be used in immuno-PET approach, it is necessary to find the best compromise between the isotope choice and the immunologic structure (full monoclonal antibody or derivatives). Through some clinical applications, this paper review aims to discuss the most important aspects of the isotope choice and the usable proteic structure that can be used to meet the clinical needs.
Collapse
Affiliation(s)
- Alexandre Lugat
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France
| | - Clément Bailly
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Michel Chérel
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Caroline Rousseau
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest (ICO) – Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France
| | - Mickaël Bourgeois
- Nantes-Angers Cancer Research Center CRCI2NA, University of Nantes, INSERM UMR1307, CNRS-ERL6075, Nantes, France,Nuclear Medicine Department, University Hospital, Nantes, France,ARRONAX Cyclotron, Saint-Herblain, France,*Correspondence: Mickaël Bourgeois
| |
Collapse
|
18
|
Mulero F. ImmunoPET in oncology. Rev Esp Med Nucl Imagen Mol 2022; 41:332-339. [PMID: 35961857 DOI: 10.1016/j.remnie.2022.08.001] [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: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 01/14/2023]
Abstract
Due to increase of immunotherapy in oncology, it is essential to have a biological characterization of tumors. Knowing which antigens are expressed both on the surface of the tumor cell and at tumor microenvironment in order to predict the tretment response different therapeutic antibodies, has become a need. ImmunoPET is a non-invasive diagnostic imaging tool that combines the high specificity of antibodies against antigens with the high sensitivity, resolution and quantification capacity of PET imaging. With ImmunoPET we obtain a virtual biopsy of tumors, it has a big present and future in preclinical-clinical research, being already a reality in predicting and monitoring the response to treatments with monoclonal antibodies, allowing a selection of patients and therapies reaching a personalized medicine contributing to improve clinical decisions.
Collapse
Affiliation(s)
- Francisca Mulero
- Unidad de Imagen Molecular, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro, 3, Madrid, Spain.
| |
Collapse
|
19
|
InmunoPET en oncología. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
20
|
Wang Y, Wang C, Huang M, Qin S, Zhao J, Sang S, Zheng M, Bian Y, Huang C, Zhang H, Guo L, Jiang J, Xu C, Dai N, Zheng Y, Han J, Yang M, Xu T, Miao L. Pilot study of a novel nanobody 68 Ga-NODAGA-SNA006 for instant PET imaging of CD8 + T cells. Eur J Nucl Med Mol Imaging 2022; 49:4394-4405. [PMID: 35829748 DOI: 10.1007/s00259-022-05903-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/30/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Positron emission tomography (PET) with specific diagnostic probes for quantifying CD8+ T cells has emerged as a powerful technique for monitoring the immune response. However, most CD8+ T cell radiotracers are based on antibodies or antibody fragments, which are slowly cleared from circulation. Herein, we aimed to develop and assess 68 Ga-NODAGA-SNA006 for instant PET (iPET) imaging of CD8+ T cells. METHODS A novel nanobody without a hexahistidine (His6) tag, SNA006-GSC, was designed, site-specifically conjugated with NODAGA-maleimide and radiolabelled with 68 Ga. The PET imaging profiles of 68 Ga-NODAGA-SNA006 were evaluated in BALB/c MC38-CD8+/CD8- tumour models and cynomolgus monkeys. Three volunteers with lung cancer underwent whole-body PET/CT imaging after 68 Ga-NODAGA-SNA006 administration. The biodistribution, pharmacokinetics and dosimetry of patients were also investigated. In addition, combined with immunohistochemistry (IHC), the quantitative performance of the tracer for monitoring CD8 expression was evaluated in BALB/c MC38-CD8+/CD8- and human subjects. RESULTS 68 Ga-NODAGA-SNA006 was prepared with RCP > 98% and SA > 100 GBq/μmol. 68 Ga-NODAGA-SNA006 exhibited specific uptake in MC38-CD8+ xenografts tumours, CD8-rich tissues (such as the spleen) in monkeys and CD8+ tumour lesions in patients within 1 h. Fast washout from circulation was observed in three volunteers (t1/2 < 20 min). A preliminary quantitative linear relationship (R2 = 0.9668, p < 0.0001 for xenografts and R2 = 0.7924, p = 0.0013 for lung patients) appeared between 68 Ga-NODAGA-SNA006 uptake and CD8 expression. 68 Ga-NODAGA-SNA006 was well tolerated by all patients. CONCLUSION 68 Ga-NODAGA-SNA006 PET imaging can instantly quantify CD8 expression with an ideal safety profile and is expected to be important for dynamically tracking CD8+ T cells and monitoring immune responses for individualised cancer immunotherapy. TRIAL REGISTRATION NCT05126927 (19 November 2021, retrospectively registered).
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chao Wang
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd., Suzhou, 215125, Jiangsu, China
| | - Minzhou Huang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Songbing Qin
- Department of Radiotherapy, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shibiao Sang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Meng Zheng
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yicong Bian
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chenrong Huang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Hua Zhang
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China.,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Lingchuan Guo
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiwei Jiang
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chun Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Na Dai
- Department of Nuclear Medicine, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yushuang Zheng
- Department of Pathology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiajun Han
- Department of Thoracic Surgery, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, No. 20 Qian-Rong Rd., Wuxi, 214063, Jiangsu, China.
| | - Tao Xu
- Smart-Nuclide Biotech, No. 218 Xing-Hu Rd., Suzhou, 215125, Jiangsu, China.
| | - Liyan Miao
- Department of Clinical Pharmacology, the First Affiliated Hospital of Soochow University, No. 899 Ping-Hai Rd., Jiangsu, 215006, Suzhou, China. .,Institute for Interdisciplinary Drug Research and Translational Sciences, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
21
|
Shan X, Gong X, Li J, Wen J, Li Y, Zhang Z. Current approaches of nanomedicines in the market and various stage of clinical translation. Acta Pharm Sin B 2022; 12:3028-3048. [PMID: 35865096 PMCID: PMC9293719 DOI: 10.1016/j.apsb.2022.02.025] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/16/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Compared with traditional drug therapy, nanomedicines exhibit intriguing biological features to increase therapeutic efficiency, reduce toxicity and achieve targeting delivery. This review provides a snapshot of nanomedicines that have been currently launched or in the clinical trials, which manifests a diversified trend in carrier types, applied indications and mechanisms of action. From the perspective of indications, this article presents an overview of the applications of nanomedicines involving the prevention, diagnosis and treatment of various diseases, which include cancer, infections, blood disorders, cardiovascular diseases, immuno-associated diseases and nervous system diseases, etc. Moreover, the review provides some considerations and perspectives in the research and development of nanomedicines to facilitate their translations in clinic.
Collapse
Affiliation(s)
- Xiaoting Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Gong
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Jingyuan Wen
- School of Pharmacy, University of Auckland, Auckland 1142, New Zealand
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiwen Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Moliner-Morro A, McInerney GM, Hanke L. Nanobodies in the limelight: Multifunctional tools in the fight against viruses. J Gen Virol 2022; 103. [PMID: 35579613 DOI: 10.1099/jgv.0.001731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antibodies are natural antivirals generated by the vertebrate immune system in response to viral infection or vaccination. Unsurprisingly, they are also key molecules in the virologist's molecular toolbox. With new developments in methods for protein engineering, protein functionalization and application, smaller antibody-derived fragments are moving in focus. Among these, camelid-derived nanobodies play a prominent role. Nanobodies can replace full-sized antibodies in most applications and enable new possible applications for which conventional antibodies are challenging to use. Here we review the versatile nature of nanobodies, discuss their promise as antiviral therapeutics, for diagnostics, and their suitability as research tools to uncover novel aspects of viral infection and disease.
Collapse
Affiliation(s)
- Ainhoa Moliner-Morro
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
23
|
Gong J, Zhao L, Yang J, Zhu M, Zhao J. [99mTc]Tc-Labeled Plectin-Targeting Peptide as a Novel SPECT Probe for Tumor Imaging. Pharmaceutics 2022; 14:pharmaceutics14050996. [PMID: 35631582 PMCID: PMC9146797 DOI: 10.3390/pharmaceutics14050996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
Certain receptors are often overexpressed during tumor occurrence and development and closely correlate with carcinogenesis. Owing to its overexpression on the cell membrane and cytoplasm of various tumors, plectin, which is involved in tumor proliferation, migration, and invasion, has been viewed as a promising target for cancer imaging. Hence, plectin-targeting agents have great potential as imaging probes for tumor diagnosis. In this study, we developed a [99mTc]Tc-labeled plectin-targeted peptide (PTP) as a novel single-photon emission computed tomography (SPECT) probe for tumor imaging and investigated its pharmacokinetics, biodistribution, and targeting ability in several types of tumor-bearing mouse models. The PTP had good biocompatibility and targeting ability to tumor cells in vitro and could be readily labeled with [99mTc]Tc after modification with the bifunctional chelator 6-hydrazino nicotinamide (HYNIC). Furthermore, the prepared [99mTc]Tc-labeled PTP ([99mTc]Tc-HYNIC-PTP) showed high radiochemical purity and excellent stability in vitro. In addition, favorable biodistribution, fast blood clearance, and clear accumulation of [99mTc]Tc-HYNIC-PTP in several types of tumors were observed, with a good correlation between tumor uptake and plectin expression levels. These results indicate the potential of [99mTc]Tc-HYNIC-PTP as a novel SPECT probe for tumor imaging.
Collapse
Affiliation(s)
- Jiali Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
| | - Jiqin Yang
- Department of Nuclear Medicine, General Hospital of Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| | - Meilin Zhu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; (J.G.); (L.Z.)
- Correspondence: (J.Y.); (M.Z.); (J.Z.); Tel.: +86-21-3779-8352 (J.Z.)
| |
Collapse
|
24
|
DeLuca KF, Mick JE, Ide AH, Lima WC, Sherman L, Schaller KL, Anderson SM, Zhao N, Stasevich TJ, Varma D, Nilsson J, DeLuca JG. Generation and diversification of recombinant monoclonal antibodies. eLife 2021; 10:72093. [PMID: 34970967 PMCID: PMC8763395 DOI: 10.7554/elife.72093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Antibodies are indispensable tools used for a large number of applications in both foundational and translational bioscience research; however, there are drawbacks to using traditional antibodies generated in animals. These include a lack of standardization leading to problems with reproducibility, high costs of antibodies purchased from commercial sources, and ethical concerns regarding the large number of animals used to generate antibodies. To address these issues, we have developed practical methodologies and tools for generating low-cost, high-yield preparations of recombinant monoclonal antibodies and antibody fragments directed to protein epitopes from primary sequences. We describe these methods here, as well as approaches to diversify monoclonal antibodies, including customization of antibody species specificity, generation of genetically encoded small antibody fragments, and conversion of single chain antibody fragments (e.g. scFv) into full-length, bivalent antibodies. This study focuses on antibodies directed to epitopes important for mitosis and kinetochore function; however, the methods and reagents described here are applicable to antibodies and antibody fragments for use in any field.
Collapse
Affiliation(s)
- Keith F DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Jeanne E Mick
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Amy Hodges Ide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Wanessa C Lima
- Geneva Antibody Facility, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lori Sherman
- CU Cancer Center Cell Technologies Shared Resource, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Kristin L Schaller
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, United States
| | - Ning Zhao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Timothy J Stasevich
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| | - Dileep Varma
- Department of Cell and Developmental Biology, Northwestern University, Chicago, United States
| | - Jakob Nilsson
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Germany
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, United States
| |
Collapse
|
25
|
Lecocq Q, Debie P, Puttemans J, Awad RM, De Beck L, Ertveldt T, De Vlaeminck Y, Goyvaerts C, Raes G, Keyaerts M, Breckpot K, Devoogdt N. Evaluation of single domain antibodies as nuclear tracers for imaging of the immune checkpoint receptor human lymphocyte activation gene-3 in cancer. EJNMMI Res 2021; 11:115. [PMID: 34727262 PMCID: PMC8563901 DOI: 10.1186/s13550-021-00857-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022] Open
Abstract
Recent advancements in the field of immune-oncology have led to a significant increase in life expectancy of patients with diverse forms of cancer, such as hematologic malignancies, melanoma and lung cancer. Unfortunately, these encouraging results are not observed in the majority of patients, who remain unresponsive and/or encounter adverse events. Currently, researchers are collecting more insight into the cellular and molecular mechanisms that underlie these variable responses. As an example, the human lymphocyte activation gene-3 (huLAG-3), an inhibitory immune checkpoint receptor, is increasingly studied as a therapeutic target in immune-oncology. Noninvasive molecular imaging of the immune checkpoint programmed death protein-1 (PD-1) or its ligand PD-L1 has shown its value as a strategy to guide and monitor PD-1/PD-L1-targeted immune checkpoint therapy. Yet, radiotracers that allow dynamic, whole body imaging of huLAG-3 expression are not yet described. We here developed single-domain antibodies (sdAbs) that bind huLAG-3 and showed that these sdAbs can image huLAG-3 in tumors, therefore representing promising tools for further development into clinically applicable radiotracers.
Collapse
Affiliation(s)
- Q Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - P Debie
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - J Puttemans
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium
| | - R M Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - L De Beck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - T Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - Y De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - C Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium
| | - G Raes
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium.,Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - M Keyaerts
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - K Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E, 1090, Brussels, Belgium.
| | - N Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K, 1090, Brussels, Belgium.
| |
Collapse
|