1
|
Lisowska M, Worrall EG, Zavadil-Kokas F, Charlton K, Murray E, Mohtar MA, Krejcir R, Hrabal V, Brydon J, Urionabarrenetxea AG, Saliba DG, Grima M, Kalathiya U, Muller P, Krejci A, Vojtesek B, Ball KL, Fahraeus R, Argyle DJ, Parys M, Hupp TR. The development of a canine single-chain phage antibody library to isolate recombinant antibodies for use in translational cancer research. CELL REPORTS METHODS 2025; 5:101008. [PMID: 40132540 PMCID: PMC12049728 DOI: 10.1016/j.crmeth.2025.101008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/10/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
The development of canine immunotolerant monoclonal antibodies can accelerate the invention of new medicines for both canine and human diseases. We develop a methodology to clone the naive, somatically mutated variable domain repertoire from canine B cell mRNA using 5'RACE PCR. A set of degenerate primers were then designed and used to clone variable domain genes into archival "holding" plasmid libraries. These archived variable domain genes were then combinatorially ligated to produce a scFv M13 phage library. Next-generation long-read and short-read DNA sequencing methodologies were developed to annotate features of the cloned library including CDR diversity and IGHV/IGKV/IGLV subfamily distribution. A synthetic immunoglobulin G was developed from this scFv library to the canine immune checkpoint receptor PD-1. This synthetic platform can be used to clone and annotate archived antibody variable domain genes for use in perpetuity in order to develop improved preclinical models for the treatment of complex human diseases.
Collapse
Affiliation(s)
- Małgorzata Lisowska
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland.
| | - Erin G Worrall
- University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK
| | - Filip Zavadil-Kokas
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Keith Charlton
- University of Aberdeen, Polwarth Building, Foresterhill, Aberdeen, UK
| | - Euan Murray
- University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK; Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St Louis, Paris, France
| | - M Aiman Mohtar
- University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK
| | - Radovan Krejcir
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Vaclav Hrabal
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Jack Brydon
- University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK
| | | | - David G Saliba
- Faculty of Health Sciences/Department Applied Biomedical Science, Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Mariana Grima
- Faculty of Health Sciences/Department Applied Biomedical Science, Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Umesh Kalathiya
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland
| | - Petr Muller
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Adam Krejci
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic; Laboratory of Growth Regulators, Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Kathryn L Ball
- University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK
| | - Robin Fahraeus
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St Louis, Paris, France
| | - David J Argyle
- The University of Edinburgh, Royal (Dick) School of Veterinary Studies and Roslin Institute, Edinburgh, UK
| | - Maciej Parys
- The University of Edinburgh, Royal (Dick) School of Veterinary Studies and Roslin Institute, Edinburgh, UK
| | - Ted R Hupp
- International Centre for Cancer Vaccine Science, University of Gdansk, Gdansk, Poland; University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, Scotland, UK
| |
Collapse
|
2
|
Lim CC, Lim TS. Profiling the broad antibody diversity of lymphatic filariasis immune antibody repertoire by deep sequencing. Int J Biol Macromol 2025; 290:140037. [PMID: 39828167 DOI: 10.1016/j.ijbiomac.2025.140037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Lymphatic filariasis is caused by infections of thread-like filarial worms, namely Wuchereria bancrofti, Brugia Malayi and Brugia timori. However, in-depth analysis of the antibody repertoire against Lymphatic filariasis is lacking. Using high-throughput sequencing of antibody repertoires, immunome analysis of IgG (LG) and IgM (LM) repertoires were studied. Despite significant differences between LG and LM in V(D)J gene usage, IGHV4-34, IGHV6-1, IGHD3-10 and IGHJ4 were preferred in both repertoires. The CDR3 in the LG repertoire showed a longer length than LM. Higher SHM level were observed in LG sequences and presence of oligoclonal sequences indicates the extent of clonal expansion. The prevalence of rare clonotypes in LM repertoire depicts the high clonal diversity when compared to LG repertoire. Monoclonal antibodies against closely related parasitic infections were present within the LG repertoire suggesting that immune repertoires may not be as exclusive and biased against the target infection as initially thought. The characterization of the immune repertoire can provide critical insight into the antibody response patterns in disease state, antibody generation process during infections and future antibody designs.
Collapse
Affiliation(s)
- Chia Chiu Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
3
|
Erasmus MF, Spector L, Ferrara F, DiNiro R, Pohl TJ, Perea-Schmittle K, Wang W, Tessier PM, Richardson C, Turner L, Kumar S, Bedinger D, Sormanni P, Fernández-Quintero ML, Ward AB, Loeffler JR, Swanson OM, Deane CM, Raybould MIJ, Evers A, Sellmann C, Bachas S, Ruffolo J, Nastri HG, Ramesh K, Sørensen J, Croasdale-Wood R, Hijano O, Leal-Lopes C, Shahsavarian M, Qiu Y, Marcatili P, Vernet E, Akbar R, Friedensohn S, Wagner R, Kurella VB, Malhotra S, Kumar S, Kidger P, Almagro JC, Furfine E, Stanton M, Graff CP, Villalba SD, Tomszak F, Teixeira AAR, Hopkins E, Dovner M, D'Angelo S, Bradbury ARM. AIntibody: an experimentally validated in silico antibody discovery design challenge. Nat Biotechnol 2024; 42:1637-1642. [PMID: 39496931 DOI: 10.1038/s41587-024-02469-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Affiliation(s)
| | | | | | | | | | | | - Wei Wang
- University of California, San Diego, USA
| | | | | | - Laure Turner
- GENEWIZ from Azenta Life Sciences, South Plainfield, USA
| | - Sumit Kumar
- GENEWIZ from Azenta Life Sciences, South Plainfield, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yu Qiu
- Sanofi, Inc, Cambridge, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Kim M, Bai X, Im H, Yang J, Kim Y, Kim MMJ, Oh Y, Jeon Y, Kwon H, Lee S, Lee CH. Construction and validation of a synthetic phage-displayed nanobody library. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:457-467. [PMID: 39198226 PMCID: PMC11361996 DOI: 10.4196/kjpp.2024.28.5.457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 09/01/2024]
Abstract
Nanobodies derived from camelids and sharks offer unique advantages in therapeutic applications due to their ability to bind to epitopes that were previously inaccessible. Traditional methods of nanobody development face challenges such as ethical concerns and antigen toxicity. Our study presents a synthetic, phagedisplayed nanobody library using trinucleotide-directed mutagenesis technology, which allows precise amino acid composition in complementarity-determining regions (CDRs), with a focus on CDR3 diversity. This approach avoids common problems such as frameshift mutations and stop codon insertions associated with other synthetic antibody library construction methods. By analyzing FDA-approved nanobodies and Protein Data Bank sequences, we designed sub-libraries with different CDR3 lengths and introduced amino acid substitutions to improve solubility. The validation of our library through the successful isolation of nanobodies against targets such as PD-1, ATXN1 and STAT3 demonstrates a versatile and ethical platform for the development of high specificity and affinity nanobodies and represents a significant advance in biotechnology.
Collapse
Affiliation(s)
- Minju Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Xuelian Bai
- Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea
| | - Hyewon Im
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jisoo Yang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Youngju Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Minjoo MJ Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yeonji Oh
- Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea
| | - Yuna Jeon
- Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea
| | - Hayoung Kwon
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seunghyun Lee
- Research Center, EPD Biotherapeutics Inc., Seoul 08378, Korea
| | - Chang-Han Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 03080, Korea
- Wide River Institute of Immunology, Seoul National University, Hongcheon 25159, Korea
- SNU Dementia Research Center, College of Medicine, Seoul National University, Seoul 03080, Korea
- Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
5
|
Hutchings CJ, Sato AK. Phage display technology and its impact in the discovery of novel protein-based drugs. Expert Opin Drug Discov 2024; 19:887-915. [PMID: 39074492 DOI: 10.1080/17460441.2024.2367023] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/07/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Phage display technology is a well-established versatile in vitro display technology that has been used for over 35 years to identify peptides and antibodies for use as reagents and therapeutics, as well as exploring the diversity of alternative scaffolds as another option to conventional therapeutic antibody discovery. Such successes have been responsible for spawning a range of biotechnology companies, as well as many complementary technologies devised to expedite the drug discovery process and resolve bottlenecks in the discovery workflow. AREAS COVERED In this perspective, the authors summarize the application of phage display for drug discovery and provide examples of protein-based drugs that have either been approved or are being developed in the clinic. The amenability of phage display to generate functional protein molecules to challenging targets and recent developments of strategies and techniques designed to harness the power of sampling diverse repertoires are highlighted. EXPERT OPINION Phage display is now routinely combined with cutting-edge technologies to deep-mine antibody-based repertoires, peptide, or alternative scaffold libraries generating a wealth of data that can be leveraged, e.g. via artificial intelligence, to enable the potential for clinical success in the discovery and development of protein-based therapeutics.
Collapse
|
6
|
Trepl J, Pasin C, Schneidawind D, Mueller NJ, Manz MG, Bankova AK, Abela IA. Evaluating tixagevimab/cilgavimab prophylaxis in allogeneic haematopoietic cell transplantation recipients for COVID-19 prevention. Br J Haematol 2024; 204:1908-1912. [PMID: 38327109 DOI: 10.1111/bjh.19321] [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: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
Allogeneic haematopoietic cell transplantation (allo-HCT) recipients exhibit an increased risk of COVID-19, particularly in the early post-transplant phase, due to insufficient vaccine responses. This retrospective study investigated the incidence of SARS-CoV-2 infection in allo-HCT recipients who received tixagevimab/cilgavimab pre-exposure prophylaxis (T/C PrEP) compared to those who did not. Logistic regression, adjusted for sex, age, SARS-CoV-2 vaccination status and immunosuppressive treatment, revealed a significant reduction in the likelihood of SARS-CoV-2 infection risk with T/C PrEP (adjusted odds ratio aOR = 0.26 [0.07, 0.91]). These findings suggest the potential efficacy of monoclonal antibody PrEP in protecting this vulnerable patient population from COVID-19.
Collapse
Affiliation(s)
- Julia Trepl
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Chloé Pasin
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Dominik Schneidawind
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolas J Mueller
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Andriyana K Bankova
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- National Specialized Hospital for Hematological Diseases, Sofia, Bulgaria
| | - Irene A Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Veggiani G, Sidhu SS. Beyond Natural Immune Repertoires: Synthetic Antibodies. Cold Spring Harb Protoc 2024; 2024:107768. [PMID: 37295822 DOI: 10.1101/pdb.top107768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Synthetic antibody libraries, in which the antigen-binding sites are precisely designed, offer unparalleled precision in antibody engineering, exceeding the potential of natural immune repertoires and constituting a novel generation of research tools and therapeutics. Recent advances in artificial intelligence-driven technologies and their integration into synthetic antibody discovery campaigns hold the promise to further streamline and effectively develop antibodies. Here, we provide an overview of synthetic antibodies. Our associated protocol describes how to develop highly diverse and functional synthetic antibody phage display libraries.
Collapse
Affiliation(s)
- Gianluca Veggiani
- The Anvil Institute, Kitchener, Ontario N2G 1H6, Canada
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | |
Collapse
|
8
|
Ferrara F, Fanni A, Teixeira AAR, Molina E, Leal-Lopes C, DeAguero A, D'Angelo S, Erasmus MF, Spector L, Rodriguez Carnero LA, Li J, Pohl TJ, Suslov N, Desrumeaux K, McMahon C, Kathuria S, Bradbury ARM. A next-generation Fab library platform directly yielding drug-like antibodies with high affinity, diversity, and developability. MAbs 2024; 16:2394230. [PMID: 39192463 DOI: 10.1080/19420862.2024.2394230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
We previously described an in vitro single-chain fragment (scFv) library platform originally designed to generate antibodies with excellent developability properties. The platform design was based on the use of clinical antibodies as scaffolds into which replicated natural complementarity-determining regions purged of sequence liabilities were inserted, and the use of phage and yeast display to carry out antibody selection. In addition to being developable, antibodies generated using our platform were extremely diverse, with most campaigns yielding sub-nanomolar binders. Here, we describe a platform advancement that incorporates Fab phage display followed by single-chain antibody-binding fragment Fab (scFab) yeast display. The scFab single-gene format provides balanced expression of light and heavy chains, with enhanced conversion to IgG, thereby combining the advantages of scFvs and Fabs. A meticulously engineered, quality-controlled Fab phage library was created using design principles similar to those used to create the scFv library. A diverse panel of binding scFabs, with high conversion efficiency to IgG, was isolated against two targets. This study highlights the compatibility of phage and yeast display with a Fab semi-synthetic library design, offering an efficient approach to generate drug-like antibodies directly, facilitating their conversion to potential therapeutic candidates.
Collapse
Affiliation(s)
| | - Adeline Fanni
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | - Esteban Molina
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | | | - Sara D'Angelo
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | - Laura Spector
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | - Jianquan Li
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | - Thomas J Pohl
- Specifica LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | | | | | | | | |
Collapse
|
9
|
Leal-Lopes C, D'Angelo S, Erasmus MF, Teixeira AAR, Temples G, Zhou J, Bradbury ARM, Ferrara F. High throughput purification of monoclonal recombinant antibodies using a Protein-A coated membrane plate system. N Biotechnol 2023; 77:111-119. [PMID: 37648151 DOI: 10.1016/j.nbt.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
The therapeutic use of monoclonal antibodies (mAbs) ranges from cancer treatment to immune-mediated conditions, covering infectious and cardiovascular disorders, among others. The development of improved methods for therapeutic antibody discovery has accelerated the identification of numerous mAbs: a discovery campaign can be deeply mined, resulting in hundreds, even thousands, of potential antibody leads for a given target of interest. High throughput mAb expression and purification methods are required for the rapid validation of those leads. In this work, we describe the implementation of a Protein-A coated membrane plate system, the Purexa™ AHT membrane plate, for robust preparative purification of hundreds of recombinant mAbs, without the need for automation. The high efficiency (>80%) recovery generated sufficient mAb for downstream screening analyses such as ELISA and surface plasmon resonance (SPR). This new system allows the functional validation of hundreds of lead antibodies from discovery campaigns in a timely manner regardless of operational size.
Collapse
Affiliation(s)
- Camila Leal-Lopes
- New Mexico Consortium, 4200 W. Jemez Rd, Suite 301, Los Alamos, NM 87544, USA
| | - Sara D'Angelo
- Specifica Inc, a Q2 Solution Company, 1607 Alcaldesa Street, Santa Fe, NM 87501, USA
| | - M Frank Erasmus
- Specifica Inc, a Q2 Solution Company, 1607 Alcaldesa Street, Santa Fe, NM 87501, USA
| | - Andre A R Teixeira
- New Mexico Consortium, 4200 W. Jemez Rd, Suite 301, Los Alamos, NM 87544, USA
| | - Graham Temples
- Purilogics, a Donaldson Brand, 900B W Faris Rd., Greenville, SC 29605, USA
| | - Jinxiang Zhou
- Purilogics, a Donaldson Brand, 900B W Faris Rd., Greenville, SC 29605, USA
| | - Andrew R M Bradbury
- Specifica Inc, a Q2 Solution Company, 1607 Alcaldesa Street, Santa Fe, NM 87501, USA.
| | - Fortunato Ferrara
- Specifica Inc, a Q2 Solution Company, 1607 Alcaldesa Street, Santa Fe, NM 87501, USA.
| |
Collapse
|
10
|
Erasmus MF, Ferrara F, D'Angelo S, Spector L, Leal-Lopes C, Teixeira AA, Sørensen J, Nagpal S, Perea-Schmittle K, Choudhary A, Honnen W, Calianese D, Antonio Rodriguez Carnero L, Cocklin S, Greiff V, Pinter A, Bradbury ARM. Insights into next generation sequencing guided antibody selection strategies. Sci Rep 2023; 13:18370. [PMID: 37884618 PMCID: PMC10603065 DOI: 10.1038/s41598-023-45538-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/20/2023] [Indexed: 10/28/2023] Open
Abstract
Therapeutic antibody discovery often relies on in-vitro display methods to identify lead candidates. Assessing selected output diversity traditionally involves random colony picking and Sanger sequencing, which has limitations. Next-generation sequencing (NGS) offers a cost-effective solution with increased read depth, allowing a comprehensive understanding of diversity. Our study establishes NGS guidelines for antibody drug discovery, demonstrating its advantages in expanding the number of unique HCDR3 clusters, broadening the number of high affinity antibodies, expanding the total number of antibodies recognizing different epitopes, and improving lead prioritization. Surprisingly, our investigation into the correlation between NGS-derived frequencies of CDRs and affinity revealed a lack of association, although this limitation could be moderately mitigated by leveraging NGS clustering, enrichment and/or relative abundance across different regions to enhance lead prioritization. This study highlights NGS benefits, offering insights, recommendations, and the most effective approach to leverage NGS in therapeutic antibody discovery.
Collapse
Affiliation(s)
| | | | - Sara D'Angelo
- Specifica LLC, a Q2 Solutions Company, Santa Fe, USA
| | - Laura Spector
- Specifica LLC, a Q2 Solutions Company, Santa Fe, USA
| | | | | | | | | | | | - Alok Choudhary
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - William Honnen
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | - David Calianese
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | | | - Simon Cocklin
- Specifica LLC, a Q2 Solutions Company, Santa Fe, USA
| | | | - Abraham Pinter
- Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, 07103, USA
| | | |
Collapse
|
11
|
Li Y, Yang KD, Kong DC, Ye JF. Advances in phage display based nano immunosensors for cholera toxin. Front Immunol 2023; 14:1224397. [PMID: 37781379 PMCID: PMC10534012 DOI: 10.3389/fimmu.2023.1224397] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Cholera, a persistent global public health concern, continues to cause outbreaks in approximately 30 countries and territories this year. The imperative to safeguard water sources and food from Vibrio cholerae, the causative pathogen, remains urgent. The bacterium is mainly disseminated via ingestion of contaminated water or food. Despite the plate method's gold standard status for detection, its time-consuming nature, taking several days to provide results, remains a challenge. The emergence of novel virulence serotypes raises public health concerns, potentially compromising existing detection methods. Hence, exploiting Vibrio cholerae toxin testing holds promise due to its inherent stability. Immunobiosensors, leveraging antibody specificity and sensitivity, present formidable tools for detecting diverse small molecules, encompassing drugs, hormones, toxins, and environmental pollutants. This review explores cholera toxin detection, highlighting phage display-based nano immunosensors' potential. Engineered bacteriophages exhibit exceptional cholera toxin affinity, through specific antibody fragments or mimotopes, enabling precise quantification. This innovative approach promises to reshape cholera toxin detection, offering an alternative to animal-derived methods. Harnessing engineered bacteriophages aligns with ethical detection and emphasizes sensitivity and accuracy, a pivotal stride in the evolution of detection strategies. This review primarily introduces recent advancements in phage display-based nano immunosensors for cholera toxin, encompassing technical aspects, current challenges, and future prospects.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- School of Nursing, Jilin University, Changchun, China
| | - De-cai Kong
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
12
|
Choi HL, Yang HR, Shin HG, Hwang K, Kim JW, Lee JH, Ryu T, Jung Y, Lee S. Generation and Next-Generation Sequencing-Based Characterization of a Large Human Combinatorial Antibody Library. Int J Mol Sci 2023; 24:ijms24066011. [PMID: 36983085 PMCID: PMC10057307 DOI: 10.3390/ijms24066011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Antibody phage display is a key technology for the discovery and development of target-specific monoclonal antibodies (mAbs) for use in research, diagnostics, and therapy. The construction of a high-quality antibody library, with larger and more diverse antibody repertoires, is essential for the successful development of phage display-derived mAbs. In this study, a large human combinatorial single-chain variable fragment library (1.5 × 1011 colonies) was constructed from Epstein-Barr virus-infected human peripheral blood mononuclear cells stimulated with a combination of two of the activators of human B cells, the Toll-like receptor 7/8 agonist R848 and interleukin-2. Next-generation sequencing analysis with approximately 1.9 × 106 and 2.7 × 106 full-length sequences of heavy chain variable (VH) and κ light chain variable (Vκ) domains, respectively, revealed that the library consists of unique VH (approximately 94%) and Vκ (approximately 91%) sequences with greater diversity than germline sequences. Lastly, multiple unique mAbs with high affinity and broad cross-species reactivity could be isolated from the library against two therapeutically relevant target antigens, validating the library quality. These findings suggest that the novel antibody library we have developed may be useful for the rapid development of target-specific phage display-derived recombinant human mAbs for use in therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Hye Lim Choi
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Kyusang Hwang
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Woong Kim
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Ji Hyun Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Taehoon Ryu
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Yushin Jung
- ATG Lifetech Inc., Seoul 08507, Republic of Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Department of Applied Chemistry, Kookmin University, Seoul 02707, Republic of Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
13
|
Discovery and Optimization of Neutralizing SARS-CoV-2 Antibodies Using ALTHEA Gold Plus Libraries™. Int J Mol Sci 2023; 24:ijms24054609. [PMID: 36902040 PMCID: PMC10003557 DOI: 10.3390/ijms24054609] [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: 01/24/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
We recently reported the isolation and characterization of anti-SARS-CoV-2 antibodies from a phage display library built with the VH repertoire of a convalescent COVID-19 patient, paired with four naïve synthetic VL libraries. One of the antibodies, called IgG-A7, neutralized the Wuhan, Delta (B.1.617.2) and Omicron (B.1.1.529) strains in authentic neutralization tests (PRNT). It also protected 100% transgenic mice expressing the human angiotensin-converting enzyme 2 (hACE-2) from SARS-CoV-2 infection. In this study, the four synthetic VL libraries were combined with the semi-synthetic VH repertoire of ALTHEA Gold Libraries™ to generate a set of fully naïve, general-purpose, libraries called ALTHEA Gold Plus Libraries™. Three out of 24 specific clones for the RBD isolated from the libraries, with affinity in the low nanomolar range and sub-optimal in vitro neutralization in PRNT, were affinity optimized via a method called "Rapid Affinity Maturation" (RAM). The final molecules reached sub-nanomolar neutralization potency, slightly superior to IgG-A7, while the developability profile over the parental molecules was improved. These results demonstrate that general-purpose libraries are a valuable source of potent neutralizing antibodies. Importantly, since general-purpose libraries are "ready-to-use", it could expedite isolation of antibodies for rapidly evolving viruses such as SARS-CoV-2.
Collapse
|
14
|
Velappan N, Ferrara F, D’Angelo S, Close D, Naranjo L, Bolding MR, Mozden SC, Troup CB, McCullough DK, Gomez A, Kedge M, Bradbury ARM. Direct selection of functional fluorescent-protein antibody fusions by yeast display. PLoS One 2023; 18:e0280930. [PMID: 36827414 PMCID: PMC9956592 DOI: 10.1371/journal.pone.0280930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/11/2023] [Indexed: 02/26/2023] Open
Abstract
Antibodies are important reagents for research, diagnostics, and therapeutics. Many examples of chimeric proteins combining the specific target recognition of antibodies with complementing functionalities such as fluorescence, toxicity or enzymatic activity have been described. However, antibodies selected solely on the basis of their binding specificities are not necessarily ideal candidates for the construction of chimeras. Here, we describe a high throughput method based on yeast display to directly select antibodies most suitable for conversion to fluorescent chimera. A library of scFv binders was converted to a fluorescent chimeric form, by cloning thermal green protein into the linker between VH and VL, and directly selecting for both binding and fluorescent functionality. This allowed us to directly identify antibodies functional in the single chain TGP format, that manifest higher protein expression, easier protein purification, and one-step binding assays.
Collapse
Affiliation(s)
- Nileena Velappan
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | | | - Sara D’Angelo
- Specifica Inc., Santa Fe, NM, United States of America
| | - Devin Close
- Arup Laboratories, Salt Lake City, UT, United States of America
| | | | - Madeline R. Bolding
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Sarah C. Mozden
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | | | - Donna K. McCullough
- Microbiology Department, University of Tennessee, Knoxville, TN, United States of America
| | - Analyssa Gomez
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | - Marijo Kedge
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, United States of America
| | | |
Collapse
|
15
|
Erasmus MF, Dovner M, Ferrara F, D'Angelo S, Teixeira AA, Leal-Lopes C, Spector L, Hopkins E, Bradbury ARM. Determining the affinities of high-affinity antibodies using KinExA and surface plasmon resonance. MAbs 2023; 15:2291209. [PMID: 38088807 DOI: 10.1080/19420862.2023.2291209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Accurate and efficient affinity measurement techniques are essential for the biophysical characterization of therapeutic monoclonal antibodies, one of the fastest growing drug classes. Surface plasmon resonance (SPR) is widely used for determining antibody affinity, but does not perform well with extremely high affinity (low picomolar to femtomolar range) molecules. In this study, we compare the SPR-based Carterra LSA and the kinetic exclusion assay (KinExA) for measuring the affinities of 48 antibodies generated against the SARS-CoV-2 receptor-binding domain. These data reveal that high-affinity antibodies can be generated straight from selections using high-quality in vitro library platforms with 54% correspondence between affinities measured using LSA and KinExA. Generally, where there was a 2-fold or greater difference between LSA and KinExA, KinExA reported that affinities were tighter. We highlight the differences between LSA and KinExA, identifying the benefits and pitfalls of each in terms of dynamic range and throughput. Furthermore, we demonstrate for the first time that single-point screening with KinExA can significantly improve throughput while maintaining a strong correlation with full binding curve equilibrium measurements, enabling the accurate rank-ordering of clones with exceptionally tight binding properties.
Collapse
Affiliation(s)
| | | | | | - Sara D'Angelo
- Specifica, LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | | | - Laura Spector
- Specifica, LLC, a Q2 Solutions Company, Santa Fe, NM, USA
| | | | | |
Collapse
|
16
|
Dormeshkin D, Shapira M, Dubovik S, Kavaleuski A, Katsin M, Migas A, Meleshko A, Semyonov S. Isolation of an escape-resistant SARS-CoV-2 neutralizing nanobody from a novel synthetic nanobody library. Front Immunol 2022; 13:965446. [PMID: 36189235 PMCID: PMC9524272 DOI: 10.3389/fimmu.2022.965446] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
The COVID−19 pandemic not only resulted in a global crisis, but also accelerated vaccine development and antibody discovery. Herein we report a synthetic humanized VHH library development pipeline for nanomolar-range affinity VHH binders to SARS-CoV-2 variants of concern (VoC) receptor binding domains (RBD) isolation. Trinucleotide-based randomization of CDRs by Kunkel mutagenesis with the subsequent rolling-cycle amplification resulted in more than 1011 diverse phage display library in a manageable for a single person number of electroporation reactions. We identified a number of nanomolar-range affinity VHH binders to SARS-CoV-2 variants of concern (VoC) receptor binding domains (RBD) by screening a novel synthetic humanized antibody library. In order to explore the most robust and fast method for affinity improvement, we performed affinity maturation by CDR1 and CDR2 shuffling and avidity engineering by multivalent trimeric VHH fusion protein construction. As a result, H7-Fc and G12x3-Fc binders were developed with the affinities in nM and pM range respectively. Importantly, these affinities are weakly influenced by most of SARS-CoV-2 VoC mutations and they retain moderate binding to BA.4\5. The plaque reduction neutralization test (PRNT) resulted in IC50 = 100 ng\ml and 9.6 ng\ml for H7-Fc and G12x3-Fc antibodies, respectively, for the emerging Omicron BA.1 variant. Therefore, these VHH could expand the present landscape of SARS-CoV-2 neutralization binders with the therapeutic potential for present and future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Dmitri Dormeshkin
- Laboratory of Molecular Diagnostics and Biotechnology, Institute of Bioorganic Chemistry of the National academy of Sciences of Belarus, Minsk, Belarus
- *Correspondence: Dmitri Dormeshkin,
| | - Michail Shapira
- Laboratory of Molecular Diagnostics and Biotechnology, Institute of Bioorganic Chemistry of the National academy of Sciences of Belarus, Minsk, Belarus
| | - Simon Dubovik
- Department of Biology, Belarusian State University, Minsk, Belarus
| | - Anton Kavaleuski
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Mikalai Katsin
- Imunovakcina, UAB, Vilnius, Lithuania
- Immunofusion, LLC, Minsk, Belarus
| | - Alexandr Migas
- Imunovakcina, UAB, Vilnius, Lithuania
- Immunofusion, LLC, Minsk, Belarus
| | | | - Sergei Semyonov
- Laboratory of Biosafety With Pathogens Collection, Republican Research and Practical Center for Epidemiology & Microbiology, Minsk, Belarus
| |
Collapse
|
17
|
Pantaleo G, Correia B, Fenwick C, Joo VS, Perez L. Antibodies to combat viral infections: development strategies and progress. Nat Rev Drug Discov 2022; 21:676-696. [PMID: 35725925 PMCID: PMC9207876 DOI: 10.1038/s41573-022-00495-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 12/11/2022]
Abstract
Monoclonal antibodies (mAbs) are appealing as potential therapeutics and prophylactics for viral infections owing to characteristics such as their high specificity and their ability to enhance immune responses. Furthermore, antibody engineering can be used to strengthen effector function and prolong mAb half-life, and advances in structural biology have enabled the selection and optimization of potent neutralizing mAbs through identification of vulnerable regions in viral proteins, which can also be relevant for vaccine design. The COVID-19 pandemic has stimulated extensive efforts to develop neutralizing mAbs against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with several mAbs now having received authorization for emergency use, providing not just an important component of strategies to combat COVID-19 but also a boost to efforts to harness mAbs in therapeutic and preventive settings for other infectious diseases. Here, we describe advances in antibody discovery and engineering that have led to the development of mAbs for use against infections caused by viruses including SARS-CoV-2, respiratory syncytial virus (RSV), Ebola virus (EBOV), human cytomegalovirus (HCMV) and influenza. We also discuss the rationale for moving from empirical to structure-guided strategies in vaccine development, based on identifying optimal candidate antigens and vulnerable regions within them that can be targeted by antibodies to result in a strong protective immune response.
Collapse
Affiliation(s)
- Giuseppe Pantaleo
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Bruno Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Craig Fenwick
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Victor S Joo
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Laurent Perez
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland.
| |
Collapse
|
18
|
Wilman W, Wróbel S, Bielska W, Deszynski P, Dudzic P, Jaszczyszyn I, Kaniewski J, Młokosiewicz J, Rouyan A, Satława T, Kumar S, Greiff V, Krawczyk K. Machine-designed biotherapeutics: opportunities, feasibility and advantages of deep learning in computational antibody discovery. Brief Bioinform 2022; 23:bbac267. [PMID: 35830864 PMCID: PMC9294429 DOI: 10.1093/bib/bbac267] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/09/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Antibodies are versatile molecular binders with an established and growing role as therapeutics. Computational approaches to developing and designing these molecules are being increasingly used to complement traditional lab-based processes. Nowadays, in silico methods fill multiple elements of the discovery stage, such as characterizing antibody-antigen interactions and identifying developability liabilities. Recently, computational methods tackling such problems have begun to follow machine learning paradigms, in many cases deep learning specifically. This paradigm shift offers improvements in established areas such as structure or binding prediction and opens up new possibilities such as language-based modeling of antibody repertoires or machine-learning-based generation of novel sequences. In this review, we critically examine the recent developments in (deep) machine learning approaches to therapeutic antibody design with implications for fully computational antibody design.
Collapse
|
19
|
Bemani P, Moazen S, Nadimi E, Nejatollahi F. Development of Human Recombinant Antibodies Against ROR1 Tumor Antigen. Rep Biochem Mol Biol 2022; 11:282-288. [PMID: 36164620 PMCID: PMC9455193 DOI: 10.52547/rbmb.11.2.282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal antigen expressed on many types of cancer cells, but not normal adult cells. ROR1 antigen contributes to cancer development and progression by several signaling pathways. ROR1 expression has been associated with tumor growth, survival, and metastasis. In this study specific human recombinant antibodies were selected against ROR1 antigen for their use in cancer immunotherapy. METHODS Phage display technology was used to produce phage antibody from a human scFv library. Phage concentration was determined to confirm the phage rescue process. Panning procedure was performed to isolate specific scFv clones against ROR1 epitope. Phage ELISA was done to evaluate the reactivity of the selected scFvs. RESULTS Two specific human scFvs with frequencies of 20% and 25% were selected against ROR1 peptide. The antibodies showed specific reaction to the corresponding epitopes in phage ELISA. DISCUSSION Cancer targeted therapy using human specific antibodies is a new strategy, which is used in cancer therapy. The selected specific scFvs that target ROR1 epitope are human antibodies that originated from a human library and have the potential to be used in clinic in cancer immunotherapy of ROR1 positive tumors without induction of human anti mouse antibody (HAMA) response.
Collapse
Affiliation(s)
- Peyman Bemani
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz Iran.
| | - Setareh Moazen
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver BC, Canada.
| | - Elham Nadimi
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz Iran.
| | - Foroogh Nejatollahi
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz Iran.
| |
Collapse
|
20
|
Strohl WR, Ku Z, An Z, Carroll SF, Keyt BA, Strohl LM. Passive Immunotherapy Against SARS-CoV-2: From Plasma-Based Therapy to Single Potent Antibodies in the Race to Stay Ahead of the Variants. BioDrugs 2022; 36:231-323. [PMID: 35476216 PMCID: PMC9043892 DOI: 10.1007/s40259-022-00529-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 12/15/2022]
Abstract
The COVID-19 pandemic is now approaching 2 years old, with more than 440 million people infected and nearly six million dead worldwide, making it the most significant pandemic since the 1918 influenza pandemic. The severity and significance of SARS-CoV-2 was recognized immediately upon discovery, leading to innumerable companies and institutes designing and generating vaccines and therapeutic antibodies literally as soon as recombinant SARS-CoV-2 spike protein sequence was available. Within months of the pandemic start, several antibodies had been generated, tested, and moved into clinical trials, including Eli Lilly's bamlanivimab and etesevimab, Regeneron's mixture of imdevimab and casirivimab, Vir's sotrovimab, Celltrion's regdanvimab, and Lilly's bebtelovimab. These antibodies all have now received at least Emergency Use Authorizations (EUAs) and some have received full approval in select countries. To date, more than three dozen antibodies or antibody combinations have been forwarded into clinical trials. These antibodies to SARS-CoV-2 all target the receptor-binding domain (RBD), with some blocking the ability of the RBD to bind human ACE2, while others bind core regions of the RBD to modulate spike stability or ability to fuse to host cell membranes. While these antibodies were being discovered and developed, new variants of SARS-CoV-2 have cropped up in real time, altering the antibody landscape on a moving basis. Over the past year, the search has widened to find antibodies capable of neutralizing the wide array of variants that have arisen, including Alpha, Beta, Gamma, Delta, and Omicron. The recent rise and dominance of the Omicron family of variants, including the rather disparate BA.1 and BA.2 variants, demonstrate the need to continue to find new approaches to neutralize the rapidly evolving SARS-CoV-2 virus. This review highlights both convalescent plasma- and polyclonal antibody-based approaches as well as the top approximately 50 antibodies to SARS-CoV-2, their epitopes, their ability to bind to SARS-CoV-2 variants, and how they are delivered. New approaches to antibody constructs, including single domain antibodies, bispecific antibodies, IgA- and IgM-based antibodies, and modified ACE2-Fc fusion proteins, are also described. Finally, antibodies being developed for palliative care of COVID-19 disease, including the ramifications of cytokine release syndrome (CRS) and acute respiratory distress syndrome (ARDS), are described.
Collapse
Affiliation(s)
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Sciences Center, Houston, TX USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Sciences Center, Houston, TX USA
| | | | | | | |
Collapse
|
21
|
Ferrara F, D’Angelo S, Erasmus MF, Teixeira AA, Leal-Lopes C, Spector LP, Pohl T, Fanni A, Cocklin S, Bradbury ARM. Pandemic's silver lining. MAbs 2022; 14:2133666. [PMID: 36253351 PMCID: PMC9578449 DOI: 10.1080/19420862.2022.2133666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The intense international focus on the COVID-19 pandemic has provided a unique opportunity to use a wide array of novel tools to carry out scientific studies on the SARS-CoV-2 virus. The value of these comparative studies extends far beyond their consequences for SARS-CoV-2, providing broad implications for health-related science. Here we specifically discuss the impacts of these comparisons on advances in vaccines, the analysis of host humoral immunity, and antibody discovery. As an extension, we also discuss potential synergies between these areas.Abbreviations: CoVIC: The Coronavirus Immunotherapeutic Consortium; EUA: Emergency Use Authorization.
Collapse
Affiliation(s)
| | - Sara D’Angelo
- Specifica Inc., A Q2 Solutions Company, Santa Fe, NM, USA
| | | | | | | | | | - Tom Pohl
- Specifica Inc., A Q2 Solutions Company, Santa Fe, NM, USA
| | - Adeline Fanni
- Specifica Inc., A Q2 Solutions Company, Santa Fe, NM, USA
| | - Simon Cocklin
- Specifica Inc., A Q2 Solutions Company, Santa Fe, NM, USA
| | - Andrew R. M. Bradbury
- Specifica Inc., A Q2 Solutions Company, Santa Fe, NM, USA,CONTACT Andrew R. M. Bradbury Specifica Inc, Los Alamos, NM, USA
| |
Collapse
|