1
|
Hojjatipour T, Sharifzadeh Z, Maali A, Azad M. Chimeric antigen receptor-natural killer cells: a promising sword against insidious tumor cells. Hum Cell 2023; 36:1843-1864. [PMID: 37477869 DOI: 10.1007/s13577-023-00948-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
Natural killer (NK) cells are a critical component of innate immunity, particularly in initial cancer recognition and inhibition of additional tumor growth or metastasis propagation. NK cells recognize transformed cells without prior sensitization via stimulatory receptors and rapidly eradicate them. However, the protective tumor microenvironment facilitates tumor escaping via induction of an exhaustion state in immune cells, including NK cells. Hence, genetic manipulation of NK cells for specific identification of tumor-associated antigens or a more robust response against tumor cells is a promising strategy for NK cells' tumoricidal augmentation. Regarding the remarkable achievement of engineered CAR-T cells in treating hematologic malignancies, there is evolving interest in CAR-NK cell recruitment in cancer immunotherapy. Innate functionality of NK cells, higher safety, superior in vivo maintenance, and the off-the-shelf potential move CAR-NK-based therapy superior to CAR-T cells treatment. In this review, we have comprehensively discussed the recent genetic manipulations of CAR-NK cell manufacturing regarding different domains of CAR constructs and their following delivery systems into diverse sources of NK cells. Then highlight the preclinical and clinical investigations of CAR-NK cells and examine the current challenges and prospects as an optimistic remedy in cancer immunotherapy.
Collapse
Affiliation(s)
- Tahereh Hojjatipour
- Department of Hematology and Blood Transfusion, Students Research Center, School of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciecnes, Qazvin, Iran
| | - Mehdi Azad
- Department of Medical Laboratory Sciences, School of Paramedicine, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, 3419759811, Iran.
| |
Collapse
|
2
|
França RKA, Studart IC, Bezerra MRL, Pontes LQ, Barbosa AMA, Brigido MM, Furtado GP, Maranhão AQ. Progress on Phage Display Technology: Tailoring Antibodies for Cancer Immunotherapy. Viruses 2023; 15:1903. [PMID: 37766309 PMCID: PMC10536222 DOI: 10.3390/v15091903] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The search for innovative anti-cancer drugs remains a challenge. Over the past three decades, antibodies have emerged as an essential asset in successful cancer therapy. The major obstacle in developing anti-cancer antibodies is the need for non-immunogenic antibodies against human antigens. This unique requirement highlights a disadvantage to using traditional hybridoma technology and thus demands alternative approaches, such as humanizing murine monoclonal antibodies. To overcome these hurdles, human monoclonal antibodies can be obtained directly from Phage Display libraries, a groundbreaking tool for antibody selection. These libraries consist of genetically engineered viruses, or phages, which can exhibit antibody fragments, such as scFv or Fab on their capsid. This innovation allows the in vitro selection of novel molecules directed towards cancer antigens. As foreseen when Phage Display was first described, nowadays, several Phage Display-derived antibodies have entered clinical settings or are undergoing clinical evaluation. This comprehensive review unveils the remarkable progress in this field and the possibilities of using clever strategies for phage selection and tailoring the refinement of antibodies aimed at increasingly specific targets. Moreover, the use of selected antibodies in cutting-edge formats is discussed, such as CAR (chimeric antigen receptor) in CAR T-cell therapy or ADC (antibody drug conjugate), amplifying the spectrum of potential therapeutic avenues.
Collapse
Affiliation(s)
- Renato Kaylan Alves França
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
- Graduate Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Igor Cabral Studart
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Marcus Rafael Lobo Bezerra
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Larissa Queiroz Pontes
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Antonio Marcos Aires Barbosa
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Applied Informatics, University of Fortaleza, Fortaleza 60811-905, Brazil
| | - Marcelo Macedo Brigido
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
| | - Gilvan Pessoa Furtado
- Oswaldo Cruz Foundation, Fiocruz Ceará, Eusébio 61773-270, Brazil; (I.C.S.); (M.R.L.B.); (L.Q.P.); (A.M.A.B.); (G.P.F.)
- Graduate Program in Biotechnology of Natural Resources, Federal University of Ceará, Fortaleza 60440-970, Brazil
| | - Andréa Queiroz Maranhão
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.F.); (M.M.B.)
| |
Collapse
|
3
|
Development of a human phage display-derived anti-PD-1 scFv antibody: an attractive tool for immune checkpoint therapy. BMC Biotechnol 2022; 22:22. [PMID: 35996120 PMCID: PMC9396865 DOI: 10.1186/s12896-022-00752-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/18/2022] [Indexed: 12/13/2022] Open
Abstract
Background The PD-1 checkpoint pathway plays a major role in tumor immune evasion and the development of the tumor microenvironment. Clinical studies show that therapeutic antibodies blocking the PD-1 pathway can restore anti-tumor or anti-virus immune responses by the reinvigoration of exhausted T cells. Because of the promising results of anti-PD-1 monoclonal antibodies in cancer treatment, autoimmune disorders, and infectious diseases, the PD-1 has emerged as an encouraging target for different diseases. Results In the present study, we employed a human semi-synthetic phage library for isolation of some scFvs against the extracellular domain of PD-1 protein by panning process. After the panning, a novel anti-PD-1 scFv (SS107) was found that exhibited specific binding to PD-1 antigen and stimulated Jurkat T cells. The selected anti-PD-1 scFv could restore the production of IL-2 and IFN-γ by Jurkat T cells that were co-cultured with PD-L1 positive tumor cells. Conclusion This anti-PD-1 scFv with high specificity and the ability to reactivate exhausted T cells has the potential to be developed as an anti-cancer agent or to be used in combination with other therapeutic approaches.
Supplementary Information The online version contains supplementary material available at 10.1186/s12896-022-00752-8.
Collapse
|
4
|
França RKADO, Silva JM, Rodrigues LS, Sokolowskei D, Brigido MM, Maranhão AQ. New Anti-Flavivirus Fusion Loop Human Antibodies with Zika Virus-Neutralizing Potential. Int J Mol Sci 2022; 23:ijms23147805. [PMID: 35887153 PMCID: PMC9321016 DOI: 10.3390/ijms23147805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Zika virus infections exhibit recurrent outbreaks and can be responsible for disease complications such as congenital Zika virus syndrome. Effective therapeutic interventions are still a challenge. Antibodies can provide significant protection, although the antibody response may fail due to antibody-dependent enhancement reactions. The choice of the target antigen is a crucial part of the process to generate effective neutralizing antibodies. Human anti-Zika virus antibodies were selected by phage display technology. The antibodies were selected against a mimetic peptide based on the fusion loop region in the protein E of Zika virus, which is highly conserved among different flaviviruses. Four rounds of selection were performed using the synthetic peptide in two strategies: the first was using the acidic elution of bound phages, and the second was by applying a competing procedure. After panning, the selected VH and VL domains were determined by combining NGS and bioinformatic approaches. Three different human monoclonal antibodies were expressed as scFvs and further characterized. All showed a binding capacity to Zika (ZIKV) and showed cross-recognition with yellow fever (YFV) and dengue (DENV) viruses. Two of these antibodies, AZ1p and AZ6m, could neutralize the ZIKV infection in vitro. Due to the conservation of the fusion loop region, these new antibodies can potentially be used in therapeutic intervention against Zika virus and other flavivirus illnesses.
Collapse
Affiliation(s)
- Renato Kaylan Alves de Oliveira França
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Jacyelle Medeiros Silva
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
| | - Lucas Silva Rodrigues
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Dimitri Sokolowskei
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Marcelo Macedo Brigido
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
- III-Immunology Investigation Institute–CNPq-MCT, São Paulo 05403-000, Brazil
- Correspondence:
| | - Andrea Queiroz Maranhão
- Molecular Immunology Laboratory, Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil; (R.K.A.d.O.F.); (J.M.S.); (L.S.R.); (D.S.); (A.Q.M.)
- Graduation Program in Molecular Pathology, University of Brasilia, Brasilia 70910-900, Brazil
- Graduation Program in Molecular Biology, University of Brasilia, Brasilia 70910-900, Brazil
- III-Immunology Investigation Institute–CNPq-MCT, São Paulo 05403-000, Brazil
| |
Collapse
|
5
|
Yue H, Li Y, Yang M, Mao C. T7 Phage as an Emerging Nanobiomaterial with Genetically Tunable Target Specificity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103645. [PMID: 34914854 PMCID: PMC8811829 DOI: 10.1002/advs.202103645] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/27/2021] [Indexed: 05/05/2023]
Abstract
Bacteriophages, also known as phages, are specific antagonists against bacteria. T7 phage has drawn massive attention in precision medicine owing to its distinctive advantages, such as short replication cycle, ease in displaying peptides and proteins, high stability and cloning efficiency, facile manipulation, and convenient storage. By introducing foreign gene into phage DNA, T7 phage can present foreign peptides or proteins site-specifically on its capsid, enabling it to become a nanoparticle that can be genetically engineered to screen and display a peptide or protein capable of recognizing a specific target with high affinity. This review critically introduces the biomedical use of T7 phage, ranging from the detection of serological biomarkers and bacterial pathogens, recognition of cells or tissues with high affinity, design of gene vectors or vaccines, to targeted therapy of different challenging diseases (e.g., bacterial infection, cancer, neurodegenerative disease, inflammatory disease, and foot-mouth disease). It also discusses perspectives and challenges in exploring T7 phage, including the understanding of its interactions with human body, assembly into scaffolds for tissue regeneration, integration with genome editing, and theranostic use in clinics. As a genetically modifiable biological nanoparticle, T7 phage holds promise as biomedical imaging probes, therapeutic agents, drug and gene carriers, and detection tools.
Collapse
Affiliation(s)
- Hui Yue
- School of Materials Science and EngineeringZhejiang UniversityHangzhouZhejiang310027P. R. China
| | - Yan Li
- Institute of Applied Bioresource ResearchCollege of Animal ScienceZhejiang UniversityYuhangtang Road 866HangzhouZhejiang310058P. R. China
| | - Mingying Yang
- Institute of Applied Bioresource ResearchCollege of Animal ScienceZhejiang UniversityYuhangtang Road 866HangzhouZhejiang310058P. R. China
| | - Chuanbin Mao
- School of Materials Science and EngineeringZhejiang UniversityHangzhouZhejiang310027P. R. China
- Department of Chemistry and BiochemistryStephenson Life Science Research CenterInstitute for Biomedical Engineering, Science and TechnologyUniversity of Oklahoma101 Stephenson ParkwayNormanOklahoma73019‐5251USA
| |
Collapse
|
6
|
Schardt JS, Jhajj HS, O’Meara RL, Lwo TS, Smith MD, Tessier PM. Agonist antibody discovery: Experimental, computational, and rational engineering approaches. Drug Discov Today 2022; 27:31-48. [PMID: 34571277 PMCID: PMC8714685 DOI: 10.1016/j.drudis.2021.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/19/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Agonist antibodies that activate cellular signaling have emerged as promising therapeutics for treating myriad pathologies. Unfortunately, the discovery of rare antibodies with the desired agonist functions is a major bottleneck during drug development. Nevertheless, there has been important recent progress in discovering and optimizing agonist antibodies against a variety of therapeutic targets that are activated by diverse signaling mechanisms. Herein, we review emerging high-throughput experimental and computational methods for agonist antibody discovery as well as rational molecular engineering methods for optimizing their agonist activity.
Collapse
Affiliation(s)
- John S. Schardt
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harkamal S. Jhajj
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ryen L. O’Meara
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timon S. Lwo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthew D. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Klangprapan S, Weng CC, Huang WT, Li YK, Choowongkomon K. Selection and Characterization of a Single-Chain Variable Fragment against Porcine Circovirus Type 2 Capsid and Impedimetric Immunosensor Development. ACS OMEGA 2021; 6:24233-24243. [PMID: 34568701 PMCID: PMC8459408 DOI: 10.1021/acsomega.1c03894] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Indexed: 06/13/2023]
Abstract
Porcine circovirus type 2 (PCV2) is the primary causative agent of porcine circovirus-associated disease (PCVAD) that causes huge global economic losses for the swine industry. Effective strategies or rapid detection of PCV2 in pig are essential to control PCVAD. Here, single-chain variable fragments (scFvs) were selected and characterized against the PCV2 capsid using phage display technology. Phage scFv clones were selected from the human scFv phagemid library (Tomlinson I + J) for direct panning against the PCV2 capsid. Eighty-four monoclonal phage scFvs were individually tested for binding to the PCV2 capsid by ELISA. Eight scFv clones showed significant binding to the PCV2 capsid and only three clones (clone nos. 13, 37, and 81) contained both VHCDRs and VLCDRs in the sequence. Clone scFv no. 81 had the highest reactivity to the PCV2 capsid and was constructed in the pET22b (+) expression vector. The recombinant was transformed to Escherichia coli BL21(DE3) for expression and purification. The scFv showed appropriate affinity to the PCV2 capsid by western blot analysis. Kinetics of scFv and the PCV2 capsid were determined using surface plasmon resonance and showed binding affinity in the nanomolar range (K D = 57.2 nM). Our scFv was first applied in the development of an impedimetric immunosensor for PCV2 capsid detection, and results showed that impedance increased with increasing PCV2 capsid expression with limit of detection = 114 nM. Findings demonstrated that our scFv has potential for use as a receptor for biosensor devices.
Collapse
Affiliation(s)
- Supaporn Klangprapan
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University, 50 Ngam Wong Wan Road, Chatuchak, Bangkok 10900, Thailand
| | - Chang-Ching Weng
- Department of Applied Chemistry, National Chiao Tung University, 1001 University Road, Hsinchu 30010, Taiwan
| | - Wan-Ting Huang
- Department of Applied Chemistry, National Chiao Tung University, 1001 University Road, Hsinchu 30010, Taiwan
| | - Yaw-Kuen Li
- Department of Applied Chemistry, National Chiao Tung University, 1001 University Road, Hsinchu 30010, Taiwan
- Center for Emergent Functional Matter Science, National Chiao Tung University, 1001 University Road, Hsinchu 30010, Taiwan
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, 50 Ngam Wong Wan Road, Chatuchak, Bangkok 10900, Thailand
| |
Collapse
|
8
|
Ortega PA, Silva-Miranda M, Torres-Larios A, Campos-Chávez E, Franken KCLCM, Ottenhoff THM, Ivanyi J, Espitia C. Selection of a Single Domain Antibody, Specific for an HLA-Bound Epitope of the Mycobacterial Ag85B Antigen. Front Immunol 2020; 11:577815. [PMID: 33117380 PMCID: PMC7564862 DOI: 10.3389/fimmu.2020.577815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/14/2020] [Indexed: 01/23/2023] Open
Abstract
T cells recognizing epitopes on the surface of mycobacteria-infected macrophages can impart protection, but with associated risk for reactivation to lung pathology. We aimed to identify antibodies specific to such epitopes, which carry potentials for development toward novel therapeutic constructs. Since epitopes presented in the context of major histocompatibility complex alleles are rarely recognized by naturally produced antibodies, we used a phage display library for the identification of monoclonal human single domain antibody producing clones. The selected 2C clone displayed T cell receptor-like recognition of an HLA-A*0201 bound 199KLVANNTRL207 peptide from the Ag85B antigen, which is known to be an immunodominant epitope for human T cells. The specificity of the selected domain antibody was demonstrated by solid phase immunoassay and by immunofluorescent surface staining of peptide loaded cells of the T2 cell line. The antibody affinity binding was determined by biolayer interferometry. Our results validated the used technologies as suitable for the generation of antibodies against epitopes on the surface of Mycobacterium tuberculosis infected cells. The potential approaches forward the development of antibody in immunotherapy of tuberculosis have been outlined in the discussion.
Collapse
Affiliation(s)
- Paola A Ortega
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Mayra Silva-Miranda
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México.,CONACyT-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alfredo Torres-Larios
- Department of Biochemistry and Structural Biology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Eduardo Campos-Chávez
- Department of Biochemistry and Structural Biology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Kees C L C M Franken
- Department of Infectious Diseases, University Medical Centre Leiden, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, University Medical Centre Leiden, Leiden, Netherlands
| | - Juraj Ivanyi
- Center for Host-Microbiome Interactions, King's College London, London, United Kingdom
| | - Clara Espitia
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México.,Center for Host-Microbiome Interactions, King's College London, London, United Kingdom
| |
Collapse
|
9
|
Parray HA, Chiranjivi AK, Asthana S, Yadav N, Shrivastava T, Mani S, Sharma C, Vishwakarma P, Das S, Pindari K, Sinha S, Samal S, Ahmed S, Kumar R. Identification of an anti-SARS-CoV-2 receptor-binding domain-directed human monoclonal antibody from a naïve semisynthetic library. J Biol Chem 2020; 295:12814-12821. [PMID: 32727845 PMCID: PMC7476711 DOI: 10.1074/jbc.ac120.014918] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
There is a desperate need for safe and effective vaccines, therapies, and diagnostics for SARS- coronavirus 2 (CoV-2), the development of which will be aided by the discovery of potent and selective antibodies against relevant viral epitopes. Human phage display technology has revolutionized the process of identifying and optimizing antibodies, providing facile entry points for further applications. Herein, we use this technology to search for antibodies targeting the receptor-binding domain (RBD) of CoV-2. Specifically, we screened a naïve human semisynthetic phage library against RBD, leading to the identification of a high-affinity single-chain fragment variable region (scFv). The scFv was further engineered into two other antibody formats (scFv-Fc and IgG1). All three antibody formats showed high binding specificity to CoV-2 RBD and the spike antigens in different assay systems. Flow cytometry analysis demonstrated specific binding of the IgG1 format to cells expressing membrane-bound CoV-2 spike protein. Docking studies revealed that the scFv recognizes an epitope that partially overlaps with angiotensin-converting enzyme 2 (ACE2)-interacting sites on the CoV-2 RBD. Given its high specificity and affinity, we anticipate that these anti-CoV-2 antibodies will be useful as valuable reagents for accessing the antigenicity of vaccine candidates, as well as developing antibody-based therapeutics and diagnostics for CoV-2.
Collapse
Affiliation(s)
- Hilal Ahmad Parray
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Adarsh Kumar Chiranjivi
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Shailendra Asthana
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Naveen Yadav
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Tripti Shrivastava
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Shailendra Mani
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Chandresh Sharma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Preeti Vishwakarma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Supratik Das
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Kamal Pindari
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Shubbir Ahmed
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Rajesh Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
10
|
Bagheri S, Safaie Qamsari E, Yousefi M, Riazi-Rad F, Sharifzadeh Z. Targeting the 4-1BB costimulatory molecule through single chain antibodies promotes the human T-cell response. Cell Mol Biol Lett 2020; 25:28. [PMID: 32336974 PMCID: PMC7178758 DOI: 10.1186/s11658-020-00219-8] [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: 07/12/2019] [Accepted: 03/27/2020] [Indexed: 11/12/2022] Open
Abstract
Background Adoptive T-cell therapy (ACT) using autologous tumor-reactive T lymphocytes has considerable potential for cancer immunotherapy. In ACT, T cells are isolated from cancer patients and then stimulated and expanded in vitro by cytokines and costimulatory molecules. 4-1BB is an important costimulatory protein belonging to the TNF receptor superfamily. It is involved in T-cell survival, proliferation and activation. Agonistic anti-4-1BB monoclonal antibodies have been introduced as appropriate tools for ACT. Methods Here, various single-chain fragment variable (scFv) antibodies were used to activate T cells isolated from peripheral blood via immune magnetic isolation. The T cells were stimulated with IL-2 and anti-CD-3 mAb and then treated with agonistic anti-4-1BB scFvs. The results showed the remarkable effects of anti-41BB scFvs on the functional properties of T cells, including their activation, proliferation and cytokine production. The flow cytometry analysis revealed a considerable increase in the expression of the T-cell activation marker CD69. Moreover, T-cell proliferation was evidenced in treated cells by CFSE labeling compared to the control groups. Result Anti-4-1BB scFvs significantly increased IFN-γ and IL-2 mRNA and protein expression in T cells, but exhibited no stimulatory effect on IL-4 expression. These findings show that anti-4-1BB scFvs could evoke a Type I immune response. Conclusions Our results demonstrate that targeting the 4-1BB molecule using agonistic scFvs could be an effective strategy for T-cell stimulation as part of an ACT approach to cancer treatment.
Collapse
Affiliation(s)
- Salman Bagheri
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.,2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,3Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Safaie Qamsari
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.,2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,3Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- 2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Riazi-Rad
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
11
|
Xia L, Teng Q, Chen Q, Zhang F. Preparation and Characterization of Anti-GPC3 Nanobody Against Hepatocellular Carcinoma. Int J Nanomedicine 2020; 15:2197-2205. [PMID: 32280214 PMCID: PMC7125335 DOI: 10.2147/ijn.s235058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background Glypican-3 (GPC3) is a newly identified target molecule for the early diagnosis of hepatocellular carcinoma (HCC), while targeted inhibition of GPC3 signaling may help to control the proliferation and metastasis of HCC cells. The purpose of this study was to prepare the anti-GPC3 nanobody and to investigate the affinity of the anti-GPC3 nanobodies in vitro and the anticancer effects on hepatocellular carcinoma in vivo. Methods To screen for unknown anti-GPC3 antibodies, we constructed an antibody phage display library. After three rounds of panning, positive phage clones were identified by enzyme-linked immunosorbent assay (ELISA). Further, the nanobody fusion protein was expressed in E. coli BL21 cells and purified by affinity chromatography. Competitive ELISA and flow cytometry were conducted to confirm the affinity of the anti-GPC3 nanobodies in vitro. The antitumor effects of VHHGPC3 were assessed in vivo. Results The results showed that the nanobody VHHGPC3 had specific high-affinity binding to His-GPC3 antigen. Moreover, VHHGPC3 exhibited specific binding to commercial human GPC3 and recognized the surface GPC3 protein of the hepatoma cell line HepG2. Importantly, in vivo study showed that GPC3 nanobody suppresses the growth of HepG2 and improves the survival rate of tumor mice. Discussion In summary, our new anti-GPC3 nanobody suggests a strong application potential for targeted therapy of liver cancer.
Collapse
Affiliation(s)
- Lijie Xia
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, People's Republic of China
| | - Qiao Teng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, People's Republic of China
| | - Qi Chen
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, People's Republic of China
| | - Fuchun Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, People's Republic of China
| |
Collapse
|
12
|
Jung JH, Han S, Ju M, Jung ST, Yu YG. Isolation of Single Chain Antibodies Specific to Lysophosphatidic Acid Receptor 1 (LPA
1
) from a M13 Phage Display Library Using Purified LPA
1
Stabilized in Nanodiscs. B KOREAN CHEM SOC 2019. [DOI: 10.1002/bkcs.11751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Ji Hae Jung
- Department of Applied ChemistryKookmin University Seoul 02707 South Korea
| | - Seong‐Gu Han
- Department of Applied ChemistryKookmin University Seoul 02707 South Korea
| | - Man‐Seok Ju
- Department of Applied ChemistryKookmin University Seoul 02707 South Korea
| | - Sang Taek Jung
- Graduate School of MedicineKorea University Seoul 02841 South Korea
| | - Yeon Gyu Yu
- Department of Applied ChemistryKookmin University Seoul 02707 South Korea
| |
Collapse
|
13
|
Wu X, Chen S, Lin L, Liu J, Wang Y, Li Y, Li Q, Wang Z. A Single Domain-Based Anti-Her2 Antibody Has Potent Antitumor Activities. Transl Oncol 2018; 11:366-373. [PMID: 29455083 PMCID: PMC5852409 DOI: 10.1016/j.tranon.2018.01.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/27/2018] [Accepted: 01/29/2018] [Indexed: 01/07/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is overexpressed in approximately 20% to 30% of breast cancers and various other types of cancers, which plays a vital role in the cancer progression. Monoclonal antibodies targeting Her2 are now used in the clinic to treat Her2 overexpression cancer patients. However, relapse or resistance is frequent with the current therapies. To generate a new treatment avenue against Her2, we immunized and selected a specific anti-Her2 single domain antibody C3 for further studies. The C3-Fc antibody drove antibody-dependent cell-mediated cytotoxicity against Her2-positive tumor cells in vitro and resulted in potent antitumor growth in vivo. These data suggest that the C3-Fc antibody may provide an alternative avenue for Her2-positive cancer therapy.
Collapse
Affiliation(s)
- Xiaoqiong Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Siqi Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Limin Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Jiayu Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Yanlan Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Yumei Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Qing Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| | - Zhong Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, China 510006; Center for Cellular & Structural Biology, Sun Yat-Sen University, Guangzhou, China 510006.
| |
Collapse
|