1
|
Høydahl LS, Berntzen G, Løset GÅ. Engineering T-cell receptor-like antibodies for biologics and cell therapy. Curr Opin Biotechnol 2024; 90:103224. [PMID: 39488859 DOI: 10.1016/j.copbio.2024.103224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/22/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024]
Abstract
A major prevailing challenge limiting our ability to fully harness the potential of the latest-generation therapeutic antibodies is the scarcity of clinically established disease-specific targets. A major next step forward will therefore be to expand this target space. The recent clinical success of immunotherapies such as adoptive T-cell transfer, immune checkpoint inhibition, and chimeric antigen receptor (CAR) T-cell therapy strongly supports focusing on the immunopeptidome of peptides presented by human leukocyte antigen (pHLA) that are normally surveilled by T-cell receptors (TCRs). Directing novel antibody development toward pHLA targets has given rise to TCR-like antibodies, which reached the clinic in 2020, as both bispecific T-cell engaging antibodies and the CARs of CAR-T cell therapies. In this review, we highlight recent advances in TCR-like antibodies, including therapeutic modalities, engineering strategies, and benchmarks for success.
Collapse
Affiliation(s)
| | | | - Geir Å Løset
- Nextera AS, Gaustadalléen 21, N-0349 Oslo, Norway.
| |
Collapse
|
2
|
Weinberg ZY, Soliman SS, Kim MS, Shah DH, Chen IP, Ott M, Lim WA, El-Samad H. De novo-designed minibinders expand the synthetic biology sensing repertoire. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575267. [PMID: 38293112 PMCID: PMC10827046 DOI: 10.1101/2024.01.12.575267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Synthetic and chimeric receptors capable of recognizing and responding to user-defined antigens have enabled "smart" therapeutics based on engineered cells. These cell engineering tools depend on antigen sensors which are most often derived from antibodies. Advances in the de novo design of proteins have enabled the design of protein binders with the potential to target epitopes with unique properties and faster production timelines compared to antibodies. Building upon our previous work combining a de novo-designed minibinder of the Spike protein of SARS-CoV-2 with the synthetic receptor synNotch (SARSNotch), we investigated whether minibinders can be readily adapted to a diversity of cell engineering tools. We show that the Spike minibinder LCB1 easily generalizes to a next-generation proteolytic receptor SNIPR that performs similarly to our previously reported SARSNotch. LCB1-SNIPR successfully enables the detection of live SARS-CoV-2, an improvement over SARSNotch which can only detect cell-expressed Spike. To test the generalizability of minibinders to diverse applications, we tested LCB1 as an antigen sensor for a chimeric antigen receptor (CAR). LCB1-CAR enabled CD8+ T cells to cytotoxically target Spike-expressing cells. We further demonstrate that two other minibinders directed against the clinically relevant epidermal growth factor receptor are able to drive CAR-dependent cytotoxicity with efficacy similar to or better than an existing antibody-based CAR. Our findings suggest that minibinders represent a novel class of antigen sensors that have the potential to dramatically expand the sensing repertoire of cell engineering tools.
Collapse
Affiliation(s)
| | | | - Matthew S. Kim
- Tetrad Gradudate Program, UCSF, San Francisco CA
- Cell Design Institute, San Francisco CA
| | - Devan H. Shah
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, CA
| | - Irene P. Chen
- Gladstone Institutes, San Francisco CA
- Department of Medicine, UCSF, San Francisco CA
| | - Melanie Ott
- Gladstone Institutes, San Francisco CA
- Department of Medicine, UCSF, San Francisco CA
- Chan Zuckerberg Biohub–San Francisco, San Francisco CA
| | - Wendell A. Lim
- Cell Design Institute, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Center for Cellular Construction, University of California, San Francisco, CA, USA
| | - Hana El-Samad
- Department of Biochemistry & Biophysics, UCSF, San Francisco CA
- Cell Design Institute, San Francisco CA
- Chan Zuckerberg Biohub–San Francisco, San Francisco CA
- Altos Labs, San Francisco CA
| |
Collapse
|
3
|
Yarmarkovich M, Marshall QF, Warrington JM, Premaratne R, Farrel A, Groff D, Li W, di Marco M, Runbeck E, Truong H, Toor JS, Tripathi S, Nguyen S, Shen H, Noel T, Church NL, Weiner A, Kendsersky N, Martinez D, Weisberg R, Christie M, Eisenlohr L, Bosse KR, Dimitrov DS, Stevanovic S, Sgourakis NG, Kiefel BR, Maris JM. Targeting of intracellular oncoproteins with peptide-centric CARs. Nature 2023; 623:820-827. [PMID: 37938771 PMCID: PMC10665195 DOI: 10.1038/s41586-023-06706-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023]
Abstract
The majority of oncogenic drivers are intracellular proteins, constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient for generating responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins essential for tumorigenesis. We focused on targeting the unmutated peptide QYNPIRTTF discovered on HLA-A*24:02, which is derived from the neuroblastoma-dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (PC-CARs) through a counter panning strategy using predicted potentially cross-reactive peptides. We further proposed that PC-CARs can recognize peptides on additional HLA allotypes when presenting a similar overall molecular surface. Informed by our computational modelling results, we show that PHOX2B PC-CARs also recognize QYNPIRTTF presented by HLA-A*23:01, the most common non-A2 allele in people with African ancestry. Finally, we demonstrate potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that PC-CARs have the potential to expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and allow targeting through additional HLA allotypes in a clinical setting.
Collapse
Affiliation(s)
- Mark Yarmarkovich
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Quinlen F Marshall
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M Warrington
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Alvin Farrel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David Groff
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Wei Li
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Erin Runbeck
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hau Truong
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jugmohit S Toor
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Sarvind Tripathi
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Son Nguyen
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Helena Shen
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiffany Noel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Amber Weiner
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nathan Kendsersky
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dan Martinez
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca Weisberg
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Molly Christie
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence Eisenlohr
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Nikolaos G Sgourakis
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Robson T, Shah DSH, Welbourn RJL, Phillips SR, Clifton LA, Lakey JH. Fully Aqueous Self-Assembly of a Gold-Nanoparticle-Based Pathogen Sensor. Int J Mol Sci 2023; 24:ijms24087599. [PMID: 37108766 PMCID: PMC10145400 DOI: 10.3390/ijms24087599] [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: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Surface plasmon resonance (SPR) is a very sensitive measure of biomolecular interactions but is generally too expensive for routine analysis of clinical samples. Here we demonstrate the simplified formation of virus-detecting gold nanoparticle (AuNP) assemblies on glass using only aqueous buffers at room temperature. The AuNP assembled on silanized glass and displayed a distinctive absorbance peak due to the localized SPR (LSPR) response of the AuNPs. Next, assembly of a protein engineering scaffold was followed using LSPR and a sensitive neutron reflectometry approach, which measured the formation and structure of the biological layer on the spherical AuNP. Finally, the assembly and function of an artificial flu sensor layer consisting of an in vitro-selected single-chain antibody (scFv)-membrane protein fusion was followed using the LSPR response of AuNPs within glass capillaries. In vitro selection avoids the need for separate animal-derived antibodies and allows for the rapid production of low-cost sensor proteins. This work demonstrates a simple approach to forming oriented arrays of protein sensors on nanostructured surfaces that uses (i) an easily assembled AuNP silane layer, (ii) self-assembly of an oriented protein layer on AuNPs, and (iii) simple highly specific artificial receptor proteins.
Collapse
Affiliation(s)
- Timothy Robson
- Biosciences Institute, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Deepan S H Shah
- Orla Protein Technologies Ltd., Biosciences Centre, International Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, UK
| | - Rebecca J L Welbourn
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 OQX, UK
| | - Sion R Phillips
- Orla Protein Technologies Ltd., Biosciences Centre, International Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, UK
| | - Luke A Clifton
- ISIS Pulsed Neutron and Muon Source, Science and Technology Facilities Council, Rutherford Appleton Laboratory, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 OQX, UK
| | - Jeremy H Lakey
- Biosciences Institute, The Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
5
|
Papadaki GF, Ani O, Florio TJ, Young MC, Danon JN, Sun Y, Dersh D, Sgourakis NG. Decoupling peptide binding from T cell receptor recognition with engineered chimeric MHC-I molecules. Front Immunol 2023; 14:1116906. [PMID: 36761745 PMCID: PMC9905809 DOI: 10.3389/fimmu.2023.1116906] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Major Histocompatibility Complex class I (MHC-I) molecules display self, viral or aberrant epitopic peptides to T cell receptors (TCRs), which employ interactions between complementarity-determining regions with both peptide and MHC-I heavy chain 'framework' residues to recognize specific Human Leucocyte Antigens (HLAs). The highly polymorphic nature of the HLA peptide-binding groove suggests a malleability of interactions within a common structural scaffold. Here, using structural data from peptide:MHC-I and pMHC:TCR structures, we first identify residues important for peptide and/or TCR binding. We then outline a fixed-backbone computational design approach for engineering synthetic molecules that combine peptide binding and TCR recognition surfaces from existing HLA allotypes. X-ray crystallography demonstrates that chimeric molecules bridging divergent HLA alleles can bind selected peptide antigens in a specified backbone conformation. Finally, in vitro tetramer staining and biophysical binding experiments using chimeric pMHC-I molecules presenting established antigens further demonstrate the requirement of TCR recognition on interactions with HLA framework residues, as opposed to interactions with peptide-centric Chimeric Antigen Receptors (CARs). Our results underscore a novel, structure-guided platform for developing synthetic HLA molecules with desired properties as screening probes for peptide-centric interactions with TCRs and other therapeutic modalities.
Collapse
Affiliation(s)
- Georgia F. Papadaki
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Omar Ani
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tyler J. Florio
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael C. Young
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Julia N. Danon
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yi Sun
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Devin Dersh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nikolaos G. Sgourakis
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Irving M, Zoete V, Bassani-Sternberg M, Coukos G. A roadmap for driving CAR T cells toward the oncogenic immunopeptidome. Cancer Cell 2022; 40:20-22. [PMID: 35016027 DOI: 10.1016/j.ccell.2021.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A critical barrier to CAR T cell therapy is the paucity of target antigens that are broadly and stably expressed exclusively in tumors. In their comprehensive multi-omics and pre-clinical study, Yarmarkovich et al. provide proof of principle for the development and efficacy of peptide centric (PC)-CARs targeting the oncogenic immunopeptidome of neuroblastoma.
Collapse
Affiliation(s)
- Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne Teaching Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Vincent Zoete
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne Teaching Hospital and University of Lausanne, Lausanne, Switzerland; SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michal Bassani-Sternberg
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne Teaching Hospital and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne Teaching Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
A Novel Peptide-MHC Targeted Chimeric Antigen Receptor T Cell Forms a T Cell-like Immune Synapse. Biomedicines 2021; 9:biomedicines9121875. [PMID: 34944696 PMCID: PMC8699022 DOI: 10.3390/biomedicines9121875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy is a promising form of adoptive cell therapy that re-engineers patient-derived T cells to express a hybrid receptor specific to a tumour-specific antigen of choice. Many well-characterised tumour antigens are intracellular and therefore not accessible to antibodies at the cell surface. Therefore, the ability to target peptide-MHC tumour targets with antibodies is key for wider applicability of CAR T cell therapy in cancer. One way to evaluate the effectiveness and efficiency of ligating tumour target cells is studying the immune synapse. Here we generated a second-generation CAR to targeting the HLA-A*02:01 restricted H3.3K27M epitope, identified as a possible therapeutic target in ~75% of diffuse midline gliomas, used as a model antigen to study the immune synapse. The pMHCI-specific CAR demonstrated specificity, potent activation, cytokine secretion and cytotoxic function. Furthermore, we characterised killing kinetics using live cell imaging as well as CAR synapse confocal imaging. Here we provide evidence of robust CAR targeting of a model peptide-MHC antigen and that, in contrast to protein-specific CARs, these CARs form a TCR-like immune synapse which facilitates TCR-like killing kinetics.
Collapse
|
8
|
Yarmarkovich M, Marshall QF, Warrington JM, Premaratne R, Farrel A, Groff D, Li W, di Marco M, Runbeck E, Truong H, Toor JS, Tripathi S, Nguyen S, Shen H, Noel T, Church NL, Weiner A, Kendsersky N, Martinez D, Weisberg R, Christie M, Eisenlohr L, Bosse KR, Dimitrov DS, Stevanovic S, Sgourakis NG, Kiefel BR, Maris JM. Cross-HLA targeting of intracellular oncoproteins with peptide-centric CARs. Nature 2021; 599:477-484. [PMID: 34732890 PMCID: PMC8599005 DOI: 10.1038/s41586-021-04061-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 09/23/2021] [Indexed: 12/27/2022]
Abstract
The majority of oncogenic drivers are intracellular proteins, thus constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient to generate responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins that are essential for tumourigenesis and focus on targeting the unmutated peptide QYNPIRTTF, discovered on HLA-A*24:02, which is derived from the neuroblastoma dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (CARs) using a counter-panning strategy with predicted potentially cross-reactive peptides. We further hypothesized that peptide-centric CARs could recognize peptides on additional HLA allotypes when presented in a similar manner. Informed by computational modelling, we showed that PHOX2B peptide-centric CARs also recognize QYNPIRTTF presented by HLA-A*23:01 and the highly divergent HLA-B*14:02. Finally, we demonstrated potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that peptide-centric CARs have the potential to vastly expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and widen the population of patients who would benefit from such therapy by breaking conventional HLA restriction.
Collapse
Affiliation(s)
- Mark Yarmarkovich
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Quinlen F Marshall
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John M Warrington
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Alvin Farrel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David Groff
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Wei Li
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Erin Runbeck
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hau Truong
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jugmohit S Toor
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Sarvind Tripathi
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Son Nguyen
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Helena Shen
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tiffany Noel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Amber Weiner
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nathan Kendsersky
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dan Martinez
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca Weisberg
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Molly Christie
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence Eisenlohr
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kristopher R Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Nikolaos G Sgourakis
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - John M Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Hastie KM, Li H, Bedinger D, Schendel SL, Dennison SM, Li K, Rayaprolu V, Yu X, Mann C, Zandonatti M, Diaz Avalos R, Zyla D, Buck T, Hui S, Shaffer K, Hariharan C, Yin J, Olmedillas E, Enriquez A, Parekh D, Abraha M, Feeney E, Horn GQ, Aldon Y, Ali H, Aracic S, Cobb RR, Federman RS, Fernandez JM, Glanville J, Green R, Grigoryan G, Lujan Hernandez AG, Ho DD, Huang KYA, Ingraham J, Jiang W, Kellam P, Kim C, Kim M, Kim HM, Kong C, Krebs SJ, Lan F, Lang G, Lee S, Leung CL, Liu J, Lu Y, MacCamy A, McGuire AT, Palser AL, Rabbitts TH, Rikhtegaran Tehrani Z, Sajadi MM, Sanders RW, Sato AK, Schweizer L, Seo J, Shen B, Snitselaar JL, Stamatatos L, Tan Y, Tomic MT, van Gils MJ, Youssef S, Yu J, Yuan TZ, Zhang Q, Peters B, Tomaras GD, Germann T, Saphire EO. Defining variant-resistant epitopes targeted by SARS-CoV-2 antibodies: A global consortium study. Science 2021; 374:472-478. [PMID: 34554826 PMCID: PMC9302186 DOI: 10.1126/science.abh2315] [Citation(s) in RCA: 222] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022]
Abstract
Antibody-based therapeutics and vaccines are essential to combat COVID-19 morbidity and mortality after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple mutations in SARS-CoV-2 that could impair antibody defenses propagated in human-to-human transmission and spillover or spillback events between humans and animals. To develop prevention and therapeutic strategies, we formed an international consortium to map the epitope landscape on the SARS-CoV-2 spike protein, defining and structurally illustrating seven receptor binding domain (RBD)–directed antibody communities with distinct footprints and competition profiles. Pseudovirion-based neutralization assays reveal spike mutations, individually and clustered together in variants, that affect antibody function among the communities. Key classes of RBD-targeted antibodies maintain neutralization activity against these emerging SARS-CoV-2 variants. These results provide a framework for selecting antibody treatment cocktails and understanding how viral variants might affect antibody therapeutic efficacy.
Collapse
Affiliation(s)
- Kathryn M. Hastie
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Haoyang Li
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Daniel Bedinger
- Carterra, 825 N. 300 W. Ste C309, Salt Lake City, UT 84103, USA
| | - Sharon L. Schendel
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - S. Moses Dennison
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - Kan Li
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - Vamseedhar Rayaprolu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Xiaoying Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Colin Mann
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Michelle Zandonatti
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Ruben Diaz Avalos
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Dawid Zyla
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Tierra Buck
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Sean Hui
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Kelly Shaffer
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Chitra Hariharan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Jieyun Yin
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Eduardo Olmedillas
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Adrian Enriquez
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Diptiben Parekh
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Milite Abraha
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - Elizabeth Feeney
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - Gillian Q. Horn
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - CoVIC-DB team1
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Carterra, 825 N. 300 W. Ste C309, Salt Lake City, UT 84103, USA
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, Netherlands
- Quadrucept Bio, Ltd., Cambridge CB23 6DW, UK
- Myrio Therapeutics Pty, Ltd., 1 Dalmore Drive, Scoresby, VIC 3179, Australia
- National Resilience, Inc., 13200 NW Nano Ct., Alachua, FL 32615, USA
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
- Activemotif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
- Centivax, Inc., 201 Gateway Blvd., Floor 1, South San Francisco, CA 94080, USA
- Twist Bioscience, 681 Gateway Blvd., South San Francisco, CA 94080, USA
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, 701 West 168th St., HHSC 1102, New York, NY 10032, USA
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital and Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
- Shanghai Henlius Biotech, Inc., 9/F, Innov Tower, Zone A, no. 1801 Hongmei Road, Xuhui District, Shanghai, China
- Kymab, Ltd., The Bennet Building, Babraham Research Campus, Cambridge CB22 3AT, UK
- Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
- Sanyou Biopharmaceuticals Co., Ltd., no. 188 Xinjunhuan Road, Building 6B-C, 3rd Floor, Minhang District, Shanghai 201114, China
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
- Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- AbCipher Biotechnology, 188 Xinjun Ring Road, Building 2, 4th Floor, Minhang District, Shanghai 201114, China
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
- Institute of Cancer Research, Centre for Cancer Drug Discovery, London SM2 5NG, UK
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
- HiFiBiO, Inc., 237 Putnam Avenue, Cambridge, MA 02139, USA
- National Resilience, Inc., 2061 Challenger Dr., Alameda, CA 94501, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Yoann Aldon
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, Netherlands
| | - Hanif Ali
- Quadrucept Bio, Ltd., Cambridge CB23 6DW, UK
| | - Sanja Aracic
- Myrio Therapeutics Pty, Ltd., 1 Dalmore Drive, Scoresby, VIC 3179, Australia
| | - Ronald R. Cobb
- National Resilience, Inc., 13200 NW Nano Ct., Alachua, FL 32615, USA
| | - Ross S. Federman
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
| | - Joseph M. Fernandez
- Activemotif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
| | - Jacob Glanville
- Centivax, Inc., 201 Gateway Blvd., Floor 1, South San Francisco, CA 94080, USA
| | - Robin Green
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
| | - Gevorg Grigoryan
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
| | | | - David D. Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, 701 West 168th St., HHSC 1102, New York, NY 10032, USA
| | - Kuan-Ying A. Huang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital and Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - John Ingraham
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
| | - Weidong Jiang
- Shanghai Henlius Biotech, Inc., 9/F, Innov Tower, Zone A, no. 1801 Hongmei Road, Xuhui District, Shanghai, China
| | - Paul Kellam
- Kymab, Ltd., The Bennet Building, Babraham Research Campus, Cambridge CB22 3AT, UK
- Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
| | - Cheolmin Kim
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
| | - Minsoo Kim
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
| | - Hyeong Mi Kim
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
| | - Chao Kong
- Sanyou Biopharmaceuticals Co., Ltd., no. 188 Xinjunhuan Road, Building 6B-C, 3rd Floor, Minhang District, Shanghai 201114, China
| | - Shelly J. Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Fei Lan
- Activemotif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
- Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Guojun Lang
- Sanyou Biopharmaceuticals Co., Ltd., no. 188 Xinjunhuan Road, Building 6B-C, 3rd Floor, Minhang District, Shanghai 201114, China
| | - Sooyoung Lee
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
| | - Cheuk Lun Leung
- Generate Biomedicines, Inc., 26 Landsdowne Street, Cambridge, MA 02139, USA
| | - Junli Liu
- Shanghai Henlius Biotech, Inc., 9/F, Innov Tower, Zone A, no. 1801 Hongmei Road, Xuhui District, Shanghai, China
| | - Yanan Lu
- Activemotif, Inc., 1914 Palomar Oaks Way, Suite 150, Carlsbad, CA 92008, USA
- AbCipher Biotechnology, 188 Xinjun Ring Road, Building 2, 4th Floor, Minhang District, Shanghai 201114, China
| | - Anna MacCamy
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Andrew T. McGuire
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Anne L. Palser
- Kymab, Ltd., The Bennet Building, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Terence H. Rabbitts
- Quadrucept Bio, Ltd., Cambridge CB23 6DW, UK
- Institute of Cancer Research, Centre for Cancer Drug Discovery, London SM2 5NG, UK
| | - Zahra Rikhtegaran Tehrani
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Mohammad M. Sajadi
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland, Baltimore, MD 21201, USA
| | - Rogier W. Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, Netherlands
| | - Aaron K. Sato
- Twist Bioscience, 681 Gateway Blvd., South San Francisco, CA 94080, USA
| | | | - Jimin Seo
- Celltrion, Inc., Department of Research and Development, 23 Academy-ro Yeonsu-gu Incheon, Republic of Korea
| | - Bingqing Shen
- HiFiBiO, Inc., 237 Putnam Avenue, Cambridge, MA 02139, USA
| | - Jonne L. Snitselaar
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, Netherlands
| | - Leonidas Stamatatos
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Yongcong Tan
- Sanyou Biopharmaceuticals Co., Ltd., no. 188 Xinjunhuan Road, Building 6B-C, 3rd Floor, Minhang District, Shanghai 201114, China
| | - Milan T. Tomic
- National Resilience, Inc., 2061 Challenger Dr., Alameda, CA 94501, USA
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Amsterdam Infection & Immunity Institute, 1105 AZ Amsterdam, Netherlands
| | - Sawsan Youssef
- Centivax, Inc., 201 Gateway Blvd., Floor 1, South San Francisco, CA 94080, USA
| | - Jian Yu
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, 701 West 168th St., HHSC 1102, New York, NY 10032, USA
| | - Tom Z. Yuan
- Twist Bioscience, 681 Gateway Blvd., South San Francisco, CA 94080, USA
| | - Qian Zhang
- HiFiBiO, Inc., 237 Putnam Avenue, Cambridge, MA 02139, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Departments of Surgery, Immunology, and Molecular Genetics and Microbiology and Duke Human Vaccine Institute, Duke University, Durham, NC 27701, USA
| | - Timothy Germann
- Carterra, 825 N. 300 W. Ste C309, Salt Lake City, UT 84103, USA
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
10
|
Abbott RC, Verdon DJ, Gracey FM, Hughes-Parry HE, Iliopoulos M, Watson KA, Mulazzani M, Luong K, D'Arcy C, Sullivan LC, Kiefel BR, Cross RS, Jenkins MR. Novel high-affinity EGFRvIII-specific chimeric antigen receptor T cells effectively eliminate human glioblastoma. Clin Transl Immunology 2021; 10:e1283. [PMID: 33976881 PMCID: PMC8106904 DOI: 10.1002/cti2.1283] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/23/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023] Open
Abstract
Objectives The increasing success of Chimeric Antigen Receptor (CAR) T cell therapy in haematological malignancies is reinvigorating its application in many other cancer types and with renewed focus on its application to solid tumors. We present a novel CAR against glioblastoma, an aggressive, malignant glioma, with a dismal survival rate for which treatment options have remained unchanged for over a decade. Methods We use the human Retained Display (ReD) antibody platform (Myrio Therapeutics) to identify a novel single‐chain variable fragment (scFv) that recognises epidermal growth factor receptor mutant variant III (EGFRvIII), a common and tumor‐specific mutation found in glioblastoma. We use both in vitro functional assays and an in vivo orthotopic xenograft model of glioblastoma to examine the function of our novel CAR, called GCT02, targeted using murine CAR T cells. Results Our EGFRvIII‐specific scFv was found to be of much higher affinity than reported comparators reverse‐engineered from monoclonal antibodies. Despite the higher affinity, GCT02 CAR T cells kill equivalently but secrete lower amounts of cytokine. In addition, GCT02‐CAR T cells also mediate rapid and complete tumor elimination in vivo. Conclusion We present a novel EGFRvIII‐specific CAR, with effective antitumor functions both in in vitro and in a xenograft model of human glioblastoma.
Collapse
Affiliation(s)
- Rebecca C Abbott
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,The Department of Medical Biology The University of Melbourne Parkville VIC Australia
| | - Daniel J Verdon
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | | | - Hannah E Hughes-Parry
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,The Department of Medical Biology The University of Melbourne Parkville VIC Australia
| | - Melinda Iliopoulos
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Katherine A Watson
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Matthias Mulazzani
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Kylie Luong
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Colleen D'Arcy
- Department of Anatomical Pathology Royal Children's Hospital Parkville VIC Australia
| | - Lucy C Sullivan
- Department of Microbiology and Immunology Peter Doherty Institute The University of Melbourne Parkville VIC Australia
| | | | - Ryan S Cross
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Misty R Jenkins
- Immunology Division The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia.,The Department of Medical Biology The University of Melbourne Parkville VIC Australia.,Institute for Molecular Science La Trobe University Bundoora VIC Australia
| |
Collapse
|
11
|
Robson T, Shah DSH, Solovyova AS, Lakey JH. Modular Protein Engineering Approach to the Functionalization of Gold Nanoparticles for Use in Clinical Diagnostics. ACS APPLIED NANO MATERIALS 2018; 1:3590-3599. [PMID: 30101217 PMCID: PMC6083416 DOI: 10.1021/acsanm.8b00737] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/28/2018] [Indexed: 05/21/2023]
Abstract
Functional protein-gold nanoparticle (AuNP) conjugates have a wide variety of applications including biosensing and drug delivery. Correct protein orientation, which is important to maintain functionality on the nanoparticle surface, can be difficult to achieve in practice, and dedicated protein scaffolds have been used on planar gold surfaces to drive the self-assembly of oriented protein arrays. Here we use the transmembrane domain of Escherichia coli outer membrane protein A (OmpATM) to create protein-AuNP conjugates. The addition of a single cysteine residue into a periplasmic loop, to create cysOmpATM, drives oriented assembly and increased equilibrium binding. As the protein surface concentration increases, the sulfur-gold bond in cysOmpATM creates a more densely populated AuNP surface than the poorly organized wtOmpATM layer. The functionalization of AuNP improved both their stability and homogeneity. This was further exploited using multidomain protein chimeras, based on cysOmpATM, which were shown to form ordered protein arrays with their functional domains displayed away from the AuNP surface. A fusion with protein G was shown to specifically bind antibodies via their Fc region. Next, an in vitro selected single chain antibody (scFv)-cysOmpATM fusion protein, bound to AuNP, detected influenza A nucleoprotein, a widely used antigen in diagnostic assays. Finally, using the same scFv-cysOmpATM-AuNP conjugates, a prototype lateral flow assay for influenza demonstrated the utility of fully recombinant self-assembling sensor layers. By simultaneously removing the need for both animal antibodies and a separate immobilization procedure, this technology could greatly simplify the development of a range of in vitro diagnostics.
Collapse
Affiliation(s)
- Timothy Robson
- Institute
for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle
upon Tyne NE2 4HH, U.K.
| | - Deepan S. H. Shah
- Orla
Protein Technologies Ltd., Biosciences Centre, International Centre for Life, Times Square, Newcastle upon Tyne NE1 4EP, U.K.
| | - Alexandra S. Solovyova
- Institute
for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle
upon Tyne NE2 4HH, U.K.
| | - Jeremy H. Lakey
- Institute
for Cell and Molecular Biosciences, The Medical School, Newcastle University, Framlington Place, Newcastle
upon Tyne NE2 4HH, U.K.
- E-mail:
| |
Collapse
|
12
|
Lloyd SB, Niven KP, Kiefel BR, Montefiori DC, Reynaldi A, Davenport MP, Kent SJ, Winnall WR. Exploration of broadly neutralizing antibody fragments produced in bacteria for the control of HIV. Hum Vaccin Immunother 2017; 13:2726-2737. [PMID: 28949787 DOI: 10.1080/21645515.2017.1368935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
While broadly neutralizing antibodies (bnAbs) are a promising preventative and therapeutic tool for HIV infection, production is difficult and expensive. Production of antibody-like fragments in bacterial cytoplasm provides a cheaper alternative. This work explored the transplantation of the complementarity determining regions of the anti-HIV bnAbs PGT121 and 10E8 onto a single-chain variable fragment (scFv) scaffold, previously discovered through a novel screening platform. The scaffolded 10E8 scFv, but not the scaffolded PGT121 scFv, was soluble in bacterial cytoplasm, enabling efficient production in bacteria. Three additional multimeric constructs employing the scaffolded 10E8 scFv were also generated and soluble versions produced in bacteria. However, the constructs were found to have substantially lost anti-HIV binding function and had completely abrogated neutralizing activity. Overall, while this study provides a proof-of-concept for anti-HIV bnAb construct production in bacterial cytoplasm, future refinement of these technologies will be required to realize the goal of producing inexpensive and effective bnAb-like tools for the control of HIV.
Collapse
Affiliation(s)
- Sarah B Lloyd
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia
| | - Keith P Niven
- b Affinity BIO Pty Ltd. , Melbourne , VIC , Australia
| | - Ben R Kiefel
- b Affinity BIO Pty Ltd. , Melbourne , VIC , Australia
| | - David C Montefiori
- c Department of Medicine , Duke University Medical Center , Durham , North Carolina , USA
| | - Arnold Reynaldi
- d Infection Analytics Program, Kirby Institute for Infection and Immunity , University of New South Wales Australia , Sydney , Australia
| | - Miles P Davenport
- d Infection Analytics Program, Kirby Institute for Infection and Immunity , University of New South Wales Australia , Sydney , Australia
| | - Stephen J Kent
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia.,e Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School , Monash University , Melbourne , Australia.,f ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Parkville , Australia
| | - Wendy R Winnall
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia
| |
Collapse
|
13
|
Lee S, Kaku Y, Inoue S, Nagamune T, Kawahara M. Growth signalobody selects functional intrabodies in the mammalian cytoplasm. Biotechnol J 2016; 11:565-73. [PMID: 26647155 DOI: 10.1002/biot.201500364] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/03/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022]
Abstract
A versatile strategy to inhibit protein functions in the cytoplasmic environment is eagerly anticipated for drug discovery. In this study, we demonstrate a novel system to directly select functional intrabodies from a library in the mammalian cytoplasm. In this system, a target homo-oligomeric antigen is expressed together with a single-chain Fv (scFv) library that is linked to the cytoplasmic domain of a receptor tyrosine kinase (RTK) in the cytoplasm of murine interleukin-3 (IL-3)-dependent cells. As the tyrosine kinase is activated by dimerization, only scFv-RTK clones that can bind to the target antigen would be oligomerized and transduce a growth signal under the IL-3-deprived condition, which leads to selection of functional intrabodies. To demonstrate this system, we used rabies virus phosphoprotein (RV-P) that forms dimers in the cytoplasm as a target antigen. As a result, functional intrabodies were selected using our system from a naïve scFv library as well as from a pre-selected anti-RV-P library generated by phage display. This system may be applied for screening intrabodies that can prevent progression of various severe diseases.
Collapse
Affiliation(s)
- Songhee Lee
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kaku
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Satoshi Inoue
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Teruyuki Nagamune
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Masahiro Kawahara
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
14
|
de Marco A. Recombinant antibody production evolves into multiple options aimed at yielding reagents suitable for application-specific needs. Microb Cell Fact 2015; 14:125. [PMID: 26330219 PMCID: PMC4557595 DOI: 10.1186/s12934-015-0320-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 08/20/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Antibodies have been a pillar of basic research, while their relevance in clinical diagnostics and therapy is constantly growing. Consequently, the production of both conventional and fragment antibodies constantly faces more demanding challenges for the improvement of their quantity and quality. The answer to such an increasing need has been the development of a wide array of formats and alternative production platforms. This review offers a critical comparison and evaluation of the different options to help the researchers interested in expressing recombinant antibodies in their choice. RESULTS Rather than the compilation of an exhaustive list of the recent publications in the field, this review intendeds to analyze the development of the most innovative or fast-growing strategies. These have been illustrated with some significant examples and, when possible, compared with the existing alternatives. Space has also been given to those solutions that might represent interesting opportunities or that investigate critical aspects of the production optimization but for which the available data as yet do not allow for a definitive judgment. CONCLUSIONS The take-home message is that there is a clear process of progressive diversification concerning the antibody expression platforms and an effort to yield directly application-adapted immune-reagents rather than generic naked antibodies that need further in vitro modification steps before becoming usable.
Collapse
Affiliation(s)
- Ario de Marco
- Department of Biomedical Sciences and Engineering, University of Nova Gorica, Glavni Trg 9, 5261, Vipava, Slovenia.
| |
Collapse
|