1
|
McClelland S, Maxwell PJ, Branco C, Barry ST, Eberlein C, LaBonte MJ. Targeting IL-8 and Its Receptors in Prostate Cancer: Inflammation, Stress Response, and Treatment Resistance. Cancers (Basel) 2024; 16:2797. [PMID: 39199570 PMCID: PMC11352248 DOI: 10.3390/cancers16162797] [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: 07/01/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
This review delves into the intricate roles of interleukin-8 (IL-8) and its receptors, CXCR1 and CXCR2, in prostate cancer (PCa), particularly in castration-resistant (CRPC) and metastatic CRPC (mCRPC). This review emphasizes the crucial role of the tumour microenvironment (TME) and inflammatory cytokines in promoting tumour progression and response to tumour cell targeting agents. IL-8, acting through C-X-C chemokine receptor type 1 (CXCR1) and type 2 (CXCR2), modulates multiple signalling pathways, enhancing the angiogenesis, proliferation, and migration of cancer cells. This review highlights the shift in PCa research focus from solely tumour cells to the non-cancer-cell components, including vascular endothelial cells, the extracellular matrix, immune cells, and the dynamic interactions within the TME. The immunosuppressive nature of the PCa TME significantly influences tumour progression and resistance to emerging therapies. Current treatment modalities, including androgen deprivation therapy and chemotherapeutics, encounter persistent resistance and are complicated by prostate cancer's notably "immune-cold" nature, which limits immune system response to the tumour. These challenges underscore the critical need for novel approaches that both overcome resistance and enhance immune engagement within the TME. The therapeutic potential of inhibiting IL-8 signalling is explored, with studies showing enhanced sensitivity of PCa cells to treatments, including radiation and androgen receptor inhibitors. Clinical trials, such as the ACE trial, demonstrate the efficacy of combining CXCR2 inhibitors with existing treatments, offering significant benefits, especially for patients with resistant PCa. This review also addresses the challenges in targeting cytokines and chemokines, noting the complexity of the TME and the need for precision in therapeutic targeting to avoid side effects and optimize outcomes.
Collapse
Affiliation(s)
- Shauna McClelland
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Pamela J. Maxwell
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Cristina Branco
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| | - Simon T. Barry
- Bioscience Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK; (S.T.B.); (C.E.)
| | - Cath Eberlein
- Bioscience Early Oncology, AstraZeneca, Cambridge CB2 0AA, UK; (S.T.B.); (C.E.)
| | - Melissa J. LaBonte
- Patrick G Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK; (S.M.); (P.J.M.); (C.B.)
| |
Collapse
|
2
|
Zamani MR, Hadzima M, Blažková K, Šubr V, Ormsby T, Celis-Gutierrez J, Malissen B, Kostka L, Etrych T, Šácha P, Konvalinka J. Polymer-based antibody mimetics (iBodies) target human PD-L1 and function as a potent immune checkpoint blocker. J Biol Chem 2024; 300:107325. [PMID: 38685532 PMCID: PMC11154707 DOI: 10.1016/j.jbc.2024.107325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
Immune checkpoint blockade (ICB) using monoclonal antibodies against programmed cell death protein 1 (PD-1) or programmed death-ligand 1 (PD-L1) is the treatment of choice for cancer immunotherapy. However, low tissue permeability, immunogenicity, immune-related adverse effects, and high cost could be possibly improved using alternative approaches. On the other hand, synthetic low-molecular-weight (LMW) PD-1/PD-L1 blockers have failed to progress beyond in vitro studies, mostly due to low binding affinity or poor pharmacological characteristics resulting from their limited solubility and/or stability. Here, we report the development of polymer-based anti-human PD-L1 antibody mimetics (α-hPD-L1 iBodies) by attaching the macrocyclic peptide WL12 to a N-(2-hydroxypropyl)methacrylamide copolymer. We characterized the binding properties of iBodies using surface plasmon resonance, enzyme-linked immunosorbent assay, flow cytometry, confocal microscopy, and a cellular ICB model. We found that the α-hPD-L1 iBodies specifically target human PD-L1 (hPD-L1) and block the PD-1/PD-L1 interaction in vitro, comparable to the atezolizumab, durvalumab, and avelumab licensed monoclonal antibodies targeting PD-L1. Our findings suggest that iBodies can be used as experimental tools to target hPD-L1 and could serve as a platform to potentiate the therapeutic effect of hPD-L1-targeting small molecules by improving their affinity and pharmacokinetic properties.
Collapse
Affiliation(s)
- Mohammad Reza Zamani
- Faculty of Science, Department of Cell Biology, Charles University, Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Hadzima
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Department of Organic Chemistry, Charles University, Prague, Czech Republic
| | - Kristýna Blažková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Vladimír Šubr
- Department of Biomedical polymers, Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Tereza Ormsby
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Javier Celis-Gutierrez
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, INSERM, CNRS, Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, INSERM, CNRS, Marseille, France
| | - Libor Kostka
- Department of Biomedical polymers, Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Etrych
- Department of Biomedical polymers, Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Department of Biochemistry, Charles University, Prague, Czech Republic.
| |
Collapse
|
3
|
Heremans J, Maximilian Awad R, Bridoux J, Ertveldt T, Caveliers V, Madder A, Hoogenboom R, Devoogdt N, Ballet S, Hernot S, Breckpot K, Martin C. Sustained release of a human PD-L1 single-domain antibody using peptide-based hydrogels. Eur J Pharm Biopharm 2024; 196:114183. [PMID: 38246566 DOI: 10.1016/j.ejpb.2024.114183] [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: 01/07/2024] [Accepted: 01/14/2024] [Indexed: 01/23/2024]
Abstract
Monoclonal antibodies (mAbs) targeting the immune checkpoint axis, which contains the programmed cell death protein-1 (PD-1) and its ligand PD-L1, revolutionized the field of oncology. Unfortunately, the large size of mAbs and the presence of an Fc fraction limit their tumor penetrative capacities and support off-target effects, potentially resulting in unresponsive patients and immune-related adverse events (irAEs) respectively. Single-domain antibodies (sdAbs) are ten times smaller than conventional mAbs and represent an emerging antibody subclass that has been proposed as next generation immune checkpoint inhibitor (ICI) therapeutics. They demonstrate favorable characteristics, such as an excellent stability, high antigen-binding affinity and an enhanced tumor penetration. Because sdAbs have a short half-life, methods to prolong their presence in the circulation and at the target site might be necessary in some cases to unfold their full therapeutic potential. In this study, we investigated a peptide-based hydrogel as an injectable biomaterial depot formulation for the sustained release of the human PD-L1 sdAb K2. We showed that a hydrogel composed of the amphipathic hexapeptide hydrogelator H-FQFQFK-NH2 prolonged the in vivo release of K2 after subcutaneous (s.c.) injection, up to at least 72 h, as monitored by SPECT/CT and fluorescence imaging. Additionally, after encapsulation in the hydrogel and s.c. administration, a significantly extended systemic presence and tumor uptake of K2 was observed in mice bearing a melanoma tumor expressing human PD-L1. Altogether, this study describes how peptide hydrogels can be exploited to provide the sustained release of sdAbs, thereby potentially enhancing its clinical and therapeutic effects.
Collapse
Affiliation(s)
- Julie Heremans
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Jessica Bridoux
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Vicky Caveliers
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Annemieke Madder
- Organic and Biomimetic Chemistry Research Group, Ghent University, 9000 Ghent, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Charlotte Martin
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| |
Collapse
|
4
|
Zhang X, Wang J, Tan Y, Chen C, Tang S, Zhao S, Qin Q, Huang H, Duan S. Nanobodies in cytokine‑mediated immunotherapy and immunoimaging (Review). Int J Mol Med 2024; 53:12. [PMID: 38063273 DOI: 10.3892/ijmm.2023.5336] [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: 08/31/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Cytokines are the main regulators of innate and adaptive immunity, mediating communications between the cells of the immune system and regulating biological functions, including cell motility, differentiation, growth and apoptosis. Cytokines and cytokine receptors have been used in the treatment of tumors and autoimmune diseases, and to intervene in cytokine storms. Indeed, the use of monoclonal antibodies to block cytokine‑receptor interactions, as well as antibody‑cytokine fusion proteins has exhibited immense potential for the treatment of tumors and autoimmune diseases. Compared with these traditional types of antibodies, nanobodies not only maintain a high affinity and specificity, but also have the advantages of high thermal stability, a high capacity for chemical manipulation, low immunogenicity, good tissue permeability, rapid clearance and economic production. Thus, nanobodies have extensive potential for use in the diagnosis and treatment of cytokine‑related diseases. The present review summarizes the application of nanobodies in cytokine‑mediated immunotherapy and immunoimaging.
Collapse
Affiliation(s)
- Xiaochen Zhang
- Department of Medicine, Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Jin Wang
- Department of Medicine, Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Ying Tan
- Department of Medicine, Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Chaoting Chen
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Shuang Tang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Shimei Zhao
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Qiuhong Qin
- Department of Medicine, Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Hansheng Huang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Siliang Duan
- Department of Medicine, Guangxi University of Science and Technology, Guangxi Zhuang Autonomous Region 545005, P.R. China
| |
Collapse
|
5
|
Nguyen H, Nguyen HL, Lan PD, Thai NQ, Sikora M, Li MS. Interaction of SARS-CoV-2 with host cells and antibodies: experiment and simulation. Chem Soc Rev 2023; 52:6497-6553. [PMID: 37650302 DOI: 10.1039/d1cs01170g] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the devastating global COVID-19 pandemic announced by WHO in March 2020. Through unprecedented scientific effort, several vaccines, drugs and antibodies have been developed, saving millions of lives, but the fight against COVID-19 continues as immune escape variants of concern such as Delta and Omicron emerge. To develop more effective treatments and to elucidate the side effects caused by vaccines and therapeutic agents, a deeper understanding of the molecular interactions of SARS-CoV-2 with them and human cells is required. With special interest in computational approaches, we will focus on the structure of SARS-CoV-2 and the interaction of its spike protein with human angiotensin-converting enzyme-2 (ACE2) as a prime entry point of the virus into host cells. In addition, other possible viral receptors will be considered. The fusion of viral and human membranes and the interaction of the spike protein with antibodies and nanobodies will be discussed, as well as the effect of SARS-CoV-2 on protein synthesis in host cells.
Collapse
Affiliation(s)
- Hung Nguyen
- Institute of Physics, Polish Academy of Sciences, al. Lotnikow 32/46, 02-668 Warsaw, Poland.
| | - Hoang Linh Nguyen
- Institute of Fundamental and Applied Sciences, Duy Tan University, Ho Chi Minh City 700000, Vietnam
- Faculty of Environmental and Natural Sciences, Duy Tan University, Da Nang 550000, Vietnam
| | - Pham Dang Lan
- Life Science Lab, Institute for Computational Science and Technology, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, 729110 Ho Chi Minh City, Vietnam
- Faculty of Physics and Engineering Physics, VNUHCM-University of Science, 227, Nguyen Van Cu Street, District 5, 749000 Ho Chi Minh City, Vietnam
| | - Nguyen Quoc Thai
- Dong Thap University, 783 Pham Huu Lau Street, Ward 6, Cao Lanh City, Dong Thap, Vietnam
| | - Mateusz Sikora
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, al. Lotnikow 32/46, 02-668 Warsaw, Poland.
| |
Collapse
|
6
|
Avrov KO, Shatik SV, Zaitsev VV, Al-Shehadat RI, Shashkova OA, Terekhina LA, Malakhov IS, Samoylovich MP. Application of Magnetic Particles for Fast Determination of Immunoreactive Fraction of 68Ga-Labelled VHH Antibodies to PD-L1. Sovrem Tekhnologii Med 2023; 15:26-33. [PMID: 38435480 PMCID: PMC10904357 DOI: 10.17691/stm2023.15.3.03] [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: 12/19/2022] [Indexed: 03/05/2024] Open
Abstract
Quantification of the immunoreactive fraction (IRF) of radioactive isotope-labeled antibodies or their fragments is necessary to assess the specific activity of radiopharmaceuticals. Traditionally, cells expressing the target molecules on their surface are used to determine IRF, but such analysis is time-consuming and has difficulties with standardization. The aim of the study was to develop a fast and reliable method for quantitative determination of IRF by 68Ga-labeled VHH antibodies to PD-L1 based on the use of magnetic particles coated with antigen molecules. Materials and Methods Commercially available magnetic particles coated with protein A have been used in our study. The antigen conjugated with the Fc fragment (PD-L1-Fc) was immobilized on the particles. The IRF value of 68Ga radionuclide-labeled nanobodies (VHH) against PD-L1 (68Ga-VHH-PD-L1) was determined using magnetic particles coated with antigen molecules and cells expressing the antigen on their surface. When VHH antibodies were conjugated to 68Ga radionuclide, protein molecules were modified using bifunctional chelating agents: tetraazacyclododecanetetraacetic acid (DOTA) or deferoxamine (DFO). The magnitude of IRF was defined as the ratio of radioactivity specifically bound to particles or cells to the total radioactivity added to the sample. Results The specificity of the 68Ga-VHH-PD-L1 radioimmunoconjugate binding to the antigen-coated magnetic particles has been proved. Some special aspects, which should be taken into consideration when using this method, have been established. The comparison of the IRF estimates using the antigen-expressing cells and magnetic particles has not revealed any significant differences in the results obtained in our study. Nevertheless, the presented method based on magnetic particles with immobilized antigen molecules requires only 15 min to determine the radioimmunoconjugate IRF, which is of fundamental importance for the routine assessment of the specificity of radiopharmaceuticals containing short-lived isotopes.
Collapse
Affiliation(s)
- K O Avrov
- Senior Researcher, Laboratory of Hybridome Technology; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - S V Shatik
- Head of the Department of Cyclotron-Produced Radiopharmaceuticals; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - V V Zaitsev
- Head of the Group for Radiopharmaceuticals Synthesis; Leading Technologist, Department of Cyclotron-Produced Radiopharmaceuticals; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - R I Al-Shehadat
- General Director; Innova plus LLC, 13 Kalinina St., Lit. A, Office 18-N, Saint Petersburg, 197198, Russia
| | - O A Shashkova
- Senior Researcher, Laboratory of Hybridome Technology; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - L A Terekhina
- Researcher, Laboratory of Hybridome Technology; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - I S Malakhov
- Senior Researcher, Laboratory of Hybridome technology; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| | - M P Samoylovich
- Chief Researcher; Head of the Laboratory of Hybridome technology; A.M. Granov Russian Research Center of Radiology and Surgical Technologies of the Ministry of Health of the Russian Federation, 70 Leningradskaya St., Saint Petersburg, Pesochniy Settlement, 197758, Russia
| |
Collapse
|
7
|
Yong Joon Kim J, Sang Z, Xiang Y, Shen Z, Shi Y. Nanobodies: Robust miniprotein binders in biomedicine. Adv Drug Deliv Rev 2023; 195:114726. [PMID: 36754285 DOI: 10.1016/j.addr.2023.114726] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 12/30/2022] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Variable domains of heavy chain-only antibodies (VHH), also known as nanobodies (Nbs), are monomeric antigen-binding domains derived from the camelid heavy chain-only antibodies. Nbs are characterized by small size, high target selectivity, and marked solubility and stability, which collectively facilitate high-quality drug development. In addition, Nbs are readily expressed from various expression systems, including E. coli and yeast cells. For these reasons, Nbs have emerged as preferred antibody fragments for protein engineering, disease diagnosis, and treatment. To date, two Nb-based therapies have been approved by the U.S. Food and Drug Administration (FDA). Numerous candidates spanning a wide spectrum of diseases such as cancer, immune disorders, infectious diseases, and neurodegenerative disorders are under preclinical and clinical investigation. Here, we discuss the structural features of Nbs that allow for specific, versatile, and strong target binding. We also summarize emerging technologies for identification, structural analysis, and humanization of Nbs. Our main focus is to review recent advances in using Nbs as a modular scaffold to facilitate the engineering of multivalent polymers for cutting-edge applications. Finally, we discuss remaining challenges for Nb development and envision new opportunities in Nb-based research.
Collapse
Affiliation(s)
- Jeffrey Yong Joon Kim
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA; Medical Scientist Training Program, University of Pittsburgh School of Medicine and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Zhe Sang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Yufei Xiang
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA
| | - Zhuolun Shen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Shi
- Center of Protein Engineering and Therapeutics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1, Gustave L. Levy Pl, New York, NY 10029, USA.
| |
Collapse
|
8
|
Maali A, Gholizadeh M, Feghhi-Najafabadi S, Noei A, Seyed-Motahari SS, Mansoori S, Sharifzadeh Z. Nanobodies in cell-mediated immunotherapy: On the road to fight cancer. Front Immunol 2023; 14:1012841. [PMID: 36761751 PMCID: PMC9905824 DOI: 10.3389/fimmu.2023.1012841] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
The immune system is essential in recognizing and eliminating tumor cells. The unique characteristics of the tumor microenvironment (TME), such as heterogeneity, reduced blood flow, hypoxia, and acidity, can reduce the efficacy of cell-mediated immunity. The primary goal of cancer immunotherapy is to modify the immune cells or the TME to enable the immune system to eliminate malignancies successfully. Nanobodies, known as single-domain antibodies, are light chain-free antibody fragments produced from Camelidae antibodies. The unique properties of nanobodies, including high stability, reduced immunogenicity, enhanced infiltration into the TME of solid tumors and facile genetic engineering have led to their promising application in cell-mediated immunotherapy. They can promote the cancer therapy either directly by bridging between tumor cells and immune cells and by targeting cancer cells using immune cell-bound nanobodies or indirectly by blocking the inhibitory ligands/receptors. The T-cell activation can be engaged through anti-CD3 and anti-4-1BB nanobodies in the bispecific (bispecific T-cell engagers (BiTEs)) and trispecific (trispecific T-cell engager (TriTEs)) manners. Also, nanobodies can be used as natural killer (NK) cell engagers (BiKEs, TriKEs, and TetraKEs) to create an immune synapse between the tumor and NK cells. Nanobodies can redirect immune cells to attack tumor cells through a chimeric antigen receptor (CAR) incorporating a nanobody against the target antigen. Various cancer antigens have been targeted by nanobody-based CAR-T and CAR-NK cells for treating both hematological and solid malignancies. They can also cause the continuation of immune surveillance against tumor cells by stopping inappropriate inhibition of immune checkpoints. Other roles of nanobodies in cell-mediated cancer immunotherapy include reprogramming macrophages to reduce metastasis and angiogenesis, as well as preventing the severe side effects occurring in cell-mediated immunotherapy. Here, we highlight the critical functions of various immune cells, including T cells, NK cells, and macrophages in the TME, and discuss newly developed immunotherapy methods based on the targeted manipulation of immune cells and TME with nanobodies.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Monireh Gholizadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Sheila Seyed-Motahari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Zahra Sharifzadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,*Correspondence: Zahra Sharifzadeh,
| |
Collapse
|
9
|
Zhou M, Liang S, Liu D, Ma K, Peng Y, Wang Z. Engineered Nanoprobes for Immune Activation Monitoring. ACS NANO 2022; 16:19940-19958. [PMID: 36454191 DOI: 10.1021/acsnano.2c09743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The activation of the immune system is critical for cancer immunotherapy and treatments of inflammatory diseases. Non-invasive visualization of immunoactivation is designed to monitor the dynamic nature of the immune response and facilitate the assessment of therapeutic outcomes, which, however, remains challenging. Conventional imaging modalities, such as positron emission tomography, computed tomography, etc., were utilized for imaging immune-related biomarkers. To explore the dynamic immune monitoring, probes with signals correlated to biomarkers of immune activation or prognosis are urgently needed. These emerging molecular probes, which turn on the signal only in the presence of the intended biomarker, can improve the detection specificity. These probes with "turn on" signals enable non-invasive, dynamic, and real-time imaging with high sensitivity and efficiency, showing significance for multifunctionality/multimodality imaging. As a result, more and more innovative engineered nanoprobes combined with diverse imaging modalities were developed to assess the activation of the immune system. In this work, we comprehensively review the recent and emerging advances in engineered nanoprobes for monitoring immune activation in cancer or other immune-mediated inflammatory diseases and discuss the potential in predicting the efficacy following treatments. Research on real-time in vivo immunoimaging is still under exploration, and this review can provide guidance and facilitate the development and application of next-generation imaging technologies.
Collapse
Affiliation(s)
- Mengli Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuang Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Kongshuo Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yuxuan Peng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Zhaohui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
10
|
Wang P, Gu Y, Yang J, Qiu J, Xu Y, Xu Z, Gao J, Wan C. The prognostic value of NLRP1/NLRP3 and its relationship with immune infiltration in human gastric cancer. Aging (Albany NY) 2022; 14:9980-10008. [PMID: 36541912 PMCID: PMC9831740 DOI: 10.18632/aging.204438] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Inflammasomes are related to tumorigenesis and immune-regulation. Here, we investigated the prognostic value of the NLR family pyrin domain containing (NLRP) 1/NLRP3 inflammasome and its potential mechanisms in immune-regulation in gastric cancer (GC). METHODS We analyzed the differential expression of NLRP1/NLRP3 between tumor and normal tissues using the Oncomine and Tumor Immune Estimate Resource (TIMER) databases. Immunohistochemistry and western blotting were used to detect NLRP1/NLRP3 protein expression in GC tissues. Correlations between NLRP1/NLRP3 expression levels and patient survival were analyzed using Kaplan-Meier survival curves. The relationships of NLRP1/NLRP3 expression and tumor-infiltrating immune cells/marker genes were assessed using the TIMER database. NLRP1/NLRP3 and immune checkpoint gene correlations were verified by single-gene co-expression analyses, and tumor immune-related pathways involving NLRP1/NLRP3 were analyzed using gene set enrichment analysis (GSEA). RESULTS Elevated NLRP1/NLRP3 expression was significantly correlated with lymph node metastasis, poor survival, immune-infiltrating cell abundances, and immune cell markers. NLRP3 showed stronger correlations with immune infiltration and the prognosis of gastric cancer. NLRP1 and NLRP3 might be involved in the same tumor immune-related pathways. Thus, high NLRP1/NLRP3 expression promotes immune cell infiltration and poor prognosis in GC. NLRP1/NLRP3, particularly NLRP3, may have important roles in immune infiltration and may serve as a prognostic biomarker for GC. CONCLUSIONS NLRP1/NLRP3, particularly NLRP3, may have important roles in immune infiltration and may serve as a prognostic biomarker for GC.
Collapse
Affiliation(s)
- Ping Wang
- School of Preclinical Medicine, Wannan Medical College, Wuhu 241001, China
| | - Yulan Gu
- Department of Oncology, Changshu Second People’s Hospital, Changshu 215500, China
| | - Jianke Yang
- School of Preclinical Medicine, Wannan Medical College, Wuhu 241001, China
| | - Jiamin Qiu
- Department of Pathology, Changshu Second People’s Hospital, Changshu 215500, China
| | - Yeqiong Xu
- Central laboratory of Changshu Medical examination Institute, Changshu 215500, China
| | - Zengxiang Xu
- School of Preclinical Medicine, Wannan Medical College, Wuhu 241001, China
| | - Jiguang Gao
- School of Preclinical Medicine, Wannan Medical College, Wuhu 241001, China
| | - Chuandan Wan
- Central laboratory of Changshu Medical examination Institute, Changshu 215500, China
| |
Collapse
|
11
|
Lu Q, Chen R, Du S, Chen C, Pan Y, Luan X, Yang J, Zeng F, He B, Han X, Song Y. Activation of the cGAS-STING pathway combined with CRISPR-Cas9 gene editing triggering long-term immunotherapy. Biomaterials 2022; 291:121871. [DOI: 10.1016/j.biomaterials.2022.121871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/04/2022] [Accepted: 10/20/2022] [Indexed: 11/28/2022]
|
12
|
Lyu M, Shi X, Liu X, Liu Y, Zhu X, Liao L, Zhao H, Sun N, Wang S, Chen L, Fan L, Xu Q, Zhu Q, Gao K, Chen H, Zhu Y, Li Z, Guo W, Zheng Y, Gu Y, Liu L, Wang M, Liu Y. Generation and Screening of Antigen-Specific Nanobodies from Mammalian Cells Expressing the BCR Repertoire Library Using Droplet-Based Microfluidics. Anal Chem 2022; 94:7970-7980. [PMID: 35604850 DOI: 10.1021/acs.analchem.2c00865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nanobodies, also known as VHHs, originate from the serum of Camelidae. Nanobodies have considerable advantages over conventional antibodies, including smaller size, more modifiable, and deeper tissue penetration, making them promising tools for immunotherapy and antibody-drug development. A high-throughput nanobody screening platform is critical to the rapid development of nanobodies. To date, droplet-based microfluidic systems have exhibited improved performance compared to the traditional phage display technology in terms of time and throughput. In realistic situations, however, it is difficult to directly apply the technology to the screening of nanobodies. Requirements of plasma cell enrichment and high cell viability, as well as a lack of related commercial reagents, are leading causes for impeding the development of novel methods. We overcame these obstacles by constructing a eukaryotic display system that secretes nanobodies utilizing homologous recombination and eukaryotic transformation technologies, and the significant advantages are that it is independent of primary cell viability and it does not require plasma cell enrichment in advance. Next, a signal capture system of "SA-beads + Biotin-antigen + nanobody-6 × His + fluorescence-labeled anti-6 × His (secondary antibody)" was designed for precise localization of the eukaryotic-expressed nanobodies in a droplet. Based on this innovation, we screened 293T cells expressing anti-PD-L1 nanobodies with a high positive rate of targeted cells (up to 99.8%). Then, single-cell transcriptomic profiling uncovered the intercellular heterogeneity and BCR sequence of target cells at a single-cell level. The complete complementarity determining region (CDR3) structure was obtained, which was totally consistent with the BCR reference. This study expanded the linkage between microfluidic technology and nanobody applications and also showed potential to accelerate the rapid transformation of nanobodies in the large-scale market.
Collapse
Affiliation(s)
- Menghua Lyu
- BGI-Shenzhen, Shenzhen 518083, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuyang Shi
- BGI-Shenzhen, Shenzhen 518083, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Yang Liu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xijun Zhu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Na Sun
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Linzhe Chen
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Linyuan Fan
- BGI-Shenzhen, Shenzhen 518083, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qumiao Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | - Kai Gao
- BGI-Shenzhen, Shenzhen 518083, China
| | - Huaying Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Yonggang Zhu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China
| | - Zida Li
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Weijin Guo
- Department of Biomedical Engineering, Shantou University, Shantou 515063, China
| | - Yue Zheng
- BGI-Shenzhen, Shenzhen 518083, China
| | - Ying Gu
- BGI-Shenzhen, Shenzhen 518083, China
| | | | | | - Ya Liu
- BGI-Shenzhen, Shenzhen 518083, China.,Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518100, China
| |
Collapse
|
13
|
Vaněk O, Kalousková B, Abreu C, Nejadebrahim S, Skořepa O. Natural killer cell-based strategies for immunotherapy of cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 129:91-133. [PMID: 35305726 DOI: 10.1016/bs.apcsb.2022.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Natural killer (NK) cells are a family of lymphocytes with a natural ability to kill infected, harmed, or malignantly transformed cells. As these cells are part of the innate immunity, the cytotoxic mechanisms are activated upon recognizing specific patterns without prior antigen sensitization. This recognition is crucial for NK cell function in the maintenance of homeostasis and immunosurveillance. NK cells not only act directly toward malignant cells but also participate in the complex immune response by producing cytokines or cross-talk with other immune cells. Cancer may be seen as a break of all immune defenses when malignant cells escape the immunity and invade surrounding tissues creating a microenvironment supporting tumor progression. This process may be reverted by intervening immune response with immunotherapy, which may restore immune recognition. NK cells are important effector cells for immunotherapy. They may be used for adoptive cell transfer, genetically modified with chimeric antigen receptors, or triggered with appropriate antibodies and other antibody-fragment-based recombinant therapeutic proteins tailored specifically for NK cell engagement. NK cell receptors, responsible for target recognition and activation of cytotoxic response, could also be targeted in immunotherapy, for example, by various bi-, tri-, or multi-specific fusion proteins designed to bridge the gap between tumor markers present on target cells and activation receptors expressed on NK cells. However, this kind of immunoactive therapeutics may be developed only with a deep functional and structural knowledge of NK cell receptor: ligand interactions. This review describes the recent developments in the fascinating protein-engineering field of NK cell immunotherapeutics.
Collapse
Affiliation(s)
- Ondřej Vaněk
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Barbora Kalousková
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Celeste Abreu
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Shiva Nejadebrahim
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ondřej Skořepa
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
14
|
Zebardast A, Hosseini P, Hasanzadeh A, latifi T. The role of single-domain antibodies (or nanobodies) in SARS-CoV-2 neutralization. Mol Biol Rep 2022; 49:647-656. [PMID: 34648139 PMCID: PMC8514607 DOI: 10.1007/s11033-021-06819-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/08/2021] [Indexed: 01/06/2023]
Abstract
The severe acute respiratory syndrome (SARS-CoV-2), a newly emerging of coronavirus, continues to infect humans in the absence of a viable treatment. Neutralizing antibodies that disrupt the interaction of RBD and ACE2 has been under the spotlight as a way of developing the COVID-19 treatment. Some animals, such as llamas, manufacture heavy-chain antibodies that have a single variable domain (VHH) instead of two variable domains (VH/VL) as opposed to typical antibodies. Nanobodies are antigen-specific, single-domain, changeable segments of camelid heavy chain-only antibodies that are recombinantly produced. These types of antibodies exhibit a wide range of strong physical and chemical properties, like high solubility, and stability. The VHH's high-affinity attachment to the receptor-binding domain (RBD) allowed the neutralization of SARS-CoV-2. To tackle COVID-19, some nanobodies are being developed against SARS-CoV-2, some of which have been recently included in clinical trials. Nanobody therapy may be useful in managing the COVID-19 pandemic as a potent and low-cost treatment. This paper describes the application of nanobodies as a new class of recombinant antibodies in COVID-19 treatment.
Collapse
Affiliation(s)
- Arghavan Zebardast
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Parastoo Hosseini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hasanzadeh
- Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Golestan, Iran
| | - Tayebeh latifi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Wang P, Tang L, Zhou B, Cheng L, Zhao RC, Zhang J. Analytical methods for the detection of PD-1/PD-L1 and other molecules related to immune checkpoints. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2021.116505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Sun S, Ding Z, Yang X, Zhao X, Zhao M, Gao L, Chen Q, Xie S, Liu A, Yin S, Xu Z, Lu X. Nanobody: A Small Antibody with Big Implications for Tumor Therapeutic Strategy. Int J Nanomedicine 2021; 16:2337-2356. [PMID: 33790553 PMCID: PMC7997558 DOI: 10.2147/ijn.s297631] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. However, the efficacy of antibody-based therapy is still confined and desperately needs further improvement. Nanobodies are the recombinant variable domains of heavy-chain-only antibodies, with many unique properties such as small size (~15kDa), excellent solubility, superior stability, ease of manufacture, quick clearance from blood, and deep tissue penetration, which gain increasing acceptance as therapeutical tools and are considered also as building blocks for chimeric antigen receptors as well as for targeted drug delivery. Thus, one of the promising novel developments that may address the deficiency of monoclonal antibody-based therapies is the utilization of nanobodies. This article provides readers the significant factors that the structural and biochemical properties of nanobodies and the research progress on nanobodies in the fields of tumor treatment, as well as their application prospect.
Collapse
Affiliation(s)
- Shuyang Sun
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Ziqiang Ding
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiaomei Yang
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xinyue Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Minlong Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Li Gao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qu Chen
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shenxia Xie
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Aiqun Liu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shihua Yin
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xiaoling Lu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|