1
|
Eden T, Schaffrath AZ, Wesolowski J, Stähler T, Tode N, Richter N, Schäfer W, Hambach J, Hermans-Borgmeyer I, Woens J, Le Gall CM, Wendler S, Linke-Winnebeck C, Stobbe M, Budnicki I, Wanney A, Heitz Y, Schimmelpfennig L, Schweitzer L, Zimmer D, Stahl E, Seyfried F, Gebhardt AJ, Dieckow L, Riecken K, Fehse B, Bannas P, Magnus T, Verdoes M, Figdor CG, Hartlepp KF, Schleer H, Füner J, Tomas NM, Haag F, Rissiek B, Mann AM, Menzel S, Koch-Nolte F. Generation of nanobodies from transgenic 'LamaMice' lacking an endogenous immunoglobulin repertoire. Nat Commun 2024; 15:4728. [PMID: 38830864 PMCID: PMC11148044 DOI: 10.1038/s41467-024-48735-x] [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: 10/23/2023] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
Due to their exceptional solubility and stability, nanobodies have emerged as powerful building blocks for research tools and therapeutics. However, their generation in llamas is cumbersome and costly. Here, by inserting an engineered llama immunoglobulin heavy chain (IgH) locus into IgH-deficient mice, we generate a transgenic mouse line, which we refer to as 'LamaMouse'. We demonstrate that LamaMice solely express llama IgH molecules without association to Igκ or λ light chains. Immunization of LamaMice with AAV8, the receptor-binding domain of the SARS-CoV-2 spike protein, IgE, IgG2c, and CLEC9A enabled us to readily select respective target-specific nanobodies using classical hybridoma and phage display technologies, single B cell screening, and direct cloning of the nanobody-repertoire into a mammalian expression vector. Our work shows that the LamaMouse represents a flexible and broadly applicable platform for a facilitated selection of target-specific nanobodies.
Collapse
Affiliation(s)
- Thomas Eden
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alessa Z Schaffrath
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janusz Wesolowski
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Natalie Tode
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Richter
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Schäfer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jannis Woens
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Camille M Le Gall
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sabrina Wendler
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | - Martina Stobbe
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | | | | | | | | | | | | | - Fabienne Seyfried
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna J Gebhardt
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lynn Dieckow
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martijn Verdoes
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Klaus F Hartlepp
- ChromoTek GmbH, Martinsried, Germany - A part of Proteintech Group, Martinsried, Germany
| | | | | | - Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Mann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Bonn, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
2
|
Vitayathikornnasak S, Rattanapisit K, Malla A, Suwanchaikasem P, Strasser R, Khorattanakulchai N, Pothisamutyothin K, Arunmanee W, Phoolcharoen W. Characterization of plant produced V HH antibodies against cobra venom toxins for antivenom therapy. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2024; 42:e00841. [PMID: 38707206 PMCID: PMC11066514 DOI: 10.1016/j.btre.2024.e00841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 05/07/2024]
Abstract
Cobra (Naja kaouthia) venom contains many toxins including α-neurotoxin (αNTX) and phospholipase A2 (PLA2), which can cause neurodegeneration, respiratory failure, and even death. The traditional antivenom derived from animal serum faces many challenges and limitations. Heavy-chain-only antibodies (HCAb), fusing VHH with human IgG Fc region, offer advantages in tissue penetration, antigen binding, and extended half-life. This research involved the construction and transient expression of two types of VHH-FC which are specific to α-Neurotoxin (VHH-αNTX-FC) and Phospholipase A2 (VHH-PLA2-FC) in Nicotiana benthamiana leaves. The recombinant HCAbs were incubated for up to six days to optimize expression levels followed by purification by affinity chromatography and characterization using LC/Q-TOF mass spectrometry (MS). Purified proteins demonstrated over 92 % sequence coverage and an average mass of around 82 kDa with a high-mannose N-glycan profile. An antigen binding assay showed that the VHH-αNTX-Fc has a greater ability to bind to crude venom than VHH-PLA2-Fc.
Collapse
Affiliation(s)
| | | | | | | | - Richard Strasser
- Department of Applied Genetics and Cell Biology, Institute of Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Vienna, Austria
| | | | - Kanokporn Pothisamutyothin
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Wanatchaporn Arunmanee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Waranyoo Phoolcharoen
- Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
3
|
Leighton PA, Ching K, Reynolds K, Vuong CN, Zeng B, Zhang Y, Gupta A, Morales J, Rivera GS, Srivastava DB, Cotter R, Pedersen D, Collarini E, Izquierdo S, van de Lavoir MC, Harriman W. Chickens with a Truncated Light Chain Transgene Express Single-Domain H Chain-Only Antibodies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1744-1753. [PMID: 38629917 PMCID: PMC11102025 DOI: 10.4049/jimmunol.2300617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/22/2024] [Indexed: 05/20/2024]
Abstract
H chain-only Igs are naturally produced in camelids and sharks. Because these Abs lack the L chain, the Ag-binding domain is half the size of a traditional Ab, allowing this type of Ig to bind to targets in novel ways. Consequently, the H chain-only single-domain Ab (sdAb) structure has the potential to increase the repertoire and functional range of an active humoral immune system. The majority of vertebrates use the standard heterodimeric (both H and L chains) structure and do not produce sdAb format Igs. To investigate if other animals are able to support sdAb development and function, transgenic chickens (Gallus gallus) were designed to produce H chain-only Abs by omitting the L chain V region and maintaining only the LC region to serve as a chaperone for Ab secretion from the cell. These birds produced 30-50% normal B cell populations within PBMCs and readily expressed chicken sequence sdAbs. Interestingly, the H chains contained a spontaneous CH1 deletion. Although no isotype switching to IgY or IgA occurred, the IgM repertoire was diverse, and immunization with a variety of protein immunogens rapidly produced high and specific serum titers. mAbs of high affinity were efficiently recovered by single B cell screening. In in vitro functional assays, the sdAbs produced by birds immunized against SARS-CoV-2 were also able to strongly neutralize and prevent viral replication. These data suggest that the truncated L chain design successfully supported sdAb development and expression in chickens.
Collapse
|
4
|
Hanssens H, Meeus F, Gesquiere EL, Puttemans J, De Vlaeminck Y, De Veirman K, Breckpot K, Devoogdt N. Anti-Idiotypic VHHs and VHH-CAR-T Cells to Tackle Multiple Myeloma: Different Applications Call for Different Antigen-Binding Moieties. Int J Mol Sci 2024; 25:5634. [PMID: 38891821 PMCID: PMC11171536 DOI: 10.3390/ijms25115634] [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: 03/25/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
CAR-T cell therapy is at the forefront of next-generation multiple myeloma (MM) management, with two B-cell maturation antigen (BCMA)-targeted products recently approved. However, these products are incapable of breaking the infamous pattern of patient relapse. Two contributing factors are the use of BCMA as a target molecule and the artificial scFv format that is responsible for antigen recognition. Tackling both points of improvement in the present study, we used previously characterized VHHs that specifically target the idiotype of murine 5T33 MM cells. This idiotype represents one of the most promising yet challenging MM target antigens, as it is highly cancer- but also patient-specific. These VHHs were incorporated into VHH-based CAR modules, the format of which has advantages compared to scFv-based CARs. This allowed a side-by-side comparison of the influence of the targeting domain on T cell activation. Surprisingly, VHHs previously selected as lead compounds for targeted MM radiotherapy are not the best (CAR-) T cell activators. Moreover, the majority of the evaluated VHHs are incapable of inducing any T cell activation. As such, we highlight the importance of specific VHH selection, depending on its intended use, and thereby raise an important shortcoming of current common CAR development approaches.
Collapse
Affiliation(s)
- Heleen Hanssens
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Emma L. Gesquiere
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Janik Puttemans
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Kim De Veirman
- Laboratory for Hematology and Immunology (HEIM), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/D0, 1090 Brussels, Belgium;
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Translational Oncology Research Center, Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/E2, 1090 Brussels, Belgium; (F.M.); (Y.D.V.); (K.B.)
| | - Nick Devoogdt
- Molecular Imaging and Therapy Research Group (MITH), Department of Biomedical Sciences, Vrije Universiteit Brussel, Laarbeeklaan 103/K0, 1090 Brussels, Belgium; (H.H.); (E.L.G.); (J.P.)
| |
Collapse
|
5
|
Chidkoksung K, Parakasikron N, Nuanualsuwan S, Khantasup K. Development of a latex agglutination test based on VH antibody fragment for detection of Streptococcus suis serotype 2. PLoS One 2024; 19:e0299691. [PMID: 38568909 PMCID: PMC10990187 DOI: 10.1371/journal.pone.0299691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/15/2024] [Indexed: 04/05/2024] Open
Abstract
Streptococcus suis serotype 2 (SS2) is an important porcine pathogen that causes diseases in both swine and human. For rapid SS2 identification, a novel latex agglutination test (LAT) based on heavy-chain variable domain antibody (VH) was developed. Firstly, the soluble 47B3 VH antibody fragment from a phage display library, in which cysteine residues were engineered at the C-terminus, was expressed in Escherichia coli. The purified protein was then gently reduced to form monomeric soluble 47B3 VH subsequently used to coat with latex beads by means of site-specific conjugation. The resulting VH-coated beads gave a good agglutination reaction with SS2. The LAT was able to distinguish S. suis serotype 2 from serotype 1/2, which shares some common sugar residues, and showed no cross-reaction with other serotypes of S. suis or other related bacteria. The detection sensitivity was found to be as high as 1.85x106 cells. The LAT was stable at 4°C for at least six months without loss of activity. To the best of our knowledge, this is the first LAT based on a VH antibody fragment that can be considered as an alternative for conventional antibody-based LAT where VHs are the most favored recombinant antibody.
Collapse
Affiliation(s)
- Kiratika Chidkoksung
- The Medical Microbiology Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Nattihda Parakasikron
- The Medical Microbiology Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Suphachai Nuanualsuwan
- Department of Veterinary Public Health, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok, Thailand
- Food Risk Hub, Research Unit of Chulalongkorn University, Bangkok, Thailand
| | - Kannika Khantasup
- Food Risk Hub, Research Unit of Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Mikkilineni L, Natrakul DA, Lam N, Manasanch EE, Mann J, Weissler KA, Wong N, Brudno JN, Goff SL, Yang JC, Ganaden M, Patel R, Zheng Z, Gartner JJ, Martin KR, Wang HW, Yuan CM, Lowe T, Maric I, Shao L, Jin P, Stroncek DF, Highfill SL, Rosenberg SA, Kochenderfer JN. Rapid anti-myeloma activity by T cells expressing an anti-BCMA CAR with a human heavy-chain-only antigen-binding domain. Mol Ther 2024; 32:503-526. [PMID: 38155568 PMCID: PMC10861980 DOI: 10.1016/j.ymthe.2023.12.018] [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: 10/09/2023] [Revised: 12/08/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Multiple myeloma (MM) is a rarely curable malignancy of plasma cells. MM expresses B cell maturation antigen (BCMA). We developed a fully human anti-BCMA chimeric antigen receptor (CAR) with a heavy-chain-only antigen-recognition domain, a 4-1BB domain, and a CD3ζ domain. The CAR was designated FHVH33-CD8BBZ. We conducted the first-in-humans clinical trial of T cells expressing FHVH33-CD8BBZ (FHVH-T). Twenty-five patients with relapsed MM were treated. The stringent complete response rate (sCR) was 52%. Median progression-free survival (PFS) was 78 weeks. Of 24 evaluable patients, 6 (25%) had a maximum cytokine-release syndrome (CRS) grade of 3; no patients had CRS of greater than grade 3. Most anti-MM activity occurred within 2-4 weeks of FHVH-T infusion as shown by decreases in the rapidly changing MM markers serum free light chains, urine light chains, and bone marrow plasma cells. Blood CAR+ cell levels peaked during the time that MM elimination was occurring, between 7 and 15 days after FHVH-T infusion. C-C chemokine receptor type 7 (CCR7) expression on infusion CD4+ FHVH-T correlated with peak blood FHVH-T levels. Single-cell RNA sequencing revealed a shift toward more differentiated FHVH-T after infusion. Anti-CAR antibody responses were detected in 4 of 12 patients assessed. FHVH-T has powerful, rapid, and durable anti-MM activity.
Collapse
Affiliation(s)
- Lekha Mikkilineni
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Danielle A Natrakul
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Norris Lam
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Jennifer Mann
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Katherine A Weissler
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nathan Wong
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research in the CCR Collaborative Bioinformatics Resource, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer N Brudno
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie L Goff
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James C Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Micaela Ganaden
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rashmika Patel
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhili Zheng
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jared J Gartner
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kathryn R Martin
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Hao-Wei Wang
- Flow Cytometry Unit, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Constance M Yuan
- Flow Cytometry Unit, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tyler Lowe
- Flow Cytometry Unit, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Irina Maric
- Hematology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Lipei Shao
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Ping Jin
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD, USA
| | - David F Stroncek
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Steven L Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Keri D, Walker M, Singh I, Nishikawa K, Garces F. Next generation of multispecific antibody engineering. Antib Ther 2024; 7:37-52. [PMID: 38235376 PMCID: PMC10791046 DOI: 10.1093/abt/tbad027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 01/19/2024] Open
Abstract
Multispecific antibodies recognize two or more epitopes located on the same or distinct targets. This added capability through protein design allows these man-made molecules to address unmet medical needs that are no longer possible with single targeting such as with monoclonal antibodies or cytokines alone. However, the approach to the development of these multispecific molecules has been met with numerous road bumps, which suggests that a new workflow for multispecific molecules is required. The investigation of the molecular basis that mediates the successful assembly of the building blocks into non-native quaternary structures will lead to the writing of a playbook for multispecifics. This is a must do if we are to design workflows that we can control and in turn predict success. Here, we reflect on the current state-of-the-art of therapeutic biologics and look at the building blocks, in terms of proteins, and tools that can be used to build the foundations of such a next-generation workflow.
Collapse
Affiliation(s)
- Daniel Keri
- Department of Protein Therapeutics, Research, Gilead Research, 324 Lakeside Dr, Foster City, CA 94404, USA
| | - Matt Walker
- Department of Protein Therapeutics, Research, Gilead Research, 324 Lakeside Dr, Foster City, CA 94404, USA
| | - Isha Singh
- Department of Protein Therapeutics, Research, Gilead Research, 324 Lakeside Dr, Foster City, CA 94404, USA
| | - Kyle Nishikawa
- Department of Protein Therapeutics, Research, Gilead Research, 324 Lakeside Dr, Foster City, CA 94404, USA
| | - Fernando Garces
- Department of Protein Therapeutics, Research, Gilead Research, 324 Lakeside Dr, Foster City, CA 94404, USA
| |
Collapse
|
8
|
Vuong CN, Reynolds KM, Rivera GS, Zeng B, Karimpourkalou Z, Norng M, Zhang Y, Chowdhury R, Pedersen D, Pantoja M, Collarini E, Garimalla S, Izquierdo S, Vajda EG, Antonio B, Srivastava DB, van de Lavoir MC, Abdiche Y, Harriman W, Leighton PA. Heavy chain-only antibodies with a stabilized human VH in transgenic chickens for therapeutic antibody discovery. MAbs 2024; 16:2435476. [PMID: 39607037 DOI: 10.1080/19420862.2024.2435476] [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: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
Heavy chain-only antibodies have found many applications where conventional heavy-light heterodimeric antibodies are not favorable. Heavy chain-only antibodies with their single antigen-binding domain offer the advantage of a smaller size and higher stability relative to conventional antibodies, and thus, the potential for novel targeting modalities. Domain antibodies have commonly been sourced from camelids with ex-vivo humanization or transgenic rodents expressing heavy chains without light chains, but these host species are all mammalian, limiting their capacity to elicit robust immune responses to conserved mammalian targets. We have developed transgenic chickens expressing heavy chain-only antibodies with a human variable region to combine the superior target recognition advantages of a divergent, non-mammalian host with the ability to discover single-domain binders. These birds produce robust immune responses, consisting of antigen-specific antibodies targeting diverse epitopes with a range of affinities. Biophysical attributes are favorable, with good developability profiles and low predicted immunogenicity.
Collapse
|
9
|
Sun Y, Yang XN, Yang SS, Lyu YZ, Zhang B, Liu KW, Li N, Cui JC, Huang GX, Liu CL, Xu J, Mi JQ, Chen Z, Fan XH, Chen SJ, Chen S. Antigen-induced chimeric antigen receptor multimerization amplifies on-tumor cytotoxicity. Signal Transduct Target Ther 2023; 8:445. [PMID: 38062078 PMCID: PMC10703879 DOI: 10.1038/s41392-023-01686-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 12/18/2023] Open
Abstract
Ligand-induced receptor dimerization or oligomerization is a widespread mechanism for ensuring communication specificity, safeguarding receptor activation, and facilitating amplification of signal transduction across the cellular membrane. However, cell-surface antigen-induced multimerization (dubbed AIM herein) has not yet been consciously leveraged in chimeric antigen receptor (CAR) engineering for enriching T cell-based therapies. We co-developed ciltacabtagene autoleucel (cilta-cel), whose CAR incorporates two B-cell maturation antigen (BCMA)-targeted nanobodies in tandem, for treating multiple myeloma. Here we elucidated a structural and functional model in which BCMA-induced cilta-cel CAR multimerization amplifies myeloma-targeted T cell-mediated cytotoxicity. Crystallographic analysis of BCMA-nanobody complexes revealed atomic details of antigen-antibody hetero-multimerization whilst analytical ultracentrifugation and small-angle X-ray scattering characterized interdependent BCMA apposition and CAR juxtaposition in solution. BCMA-induced nanobody CAR multimerization enhanced cytotoxicity, alongside elevated immune synapse formation and cytotoxicity-mediating cytokine release, towards myeloma-derived cells. Our results provide a framework for contemplating the AIM approach in designing next-generation CARs.
Collapse
Affiliation(s)
- Yan Sun
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiu-Na Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Shuang-Shuang Yang
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi-Zhu Lyu
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Hematology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bing Zhang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Kai-Wen Liu
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Na Li
- National Facility for Protein Science Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Jia-Chen Cui
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang-Xiang Huang
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cheng-Lin Liu
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Xu
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Hu Fan
- Legend Biotech China, Nanjing, 211112, China.
- Wondercel Biotechnology, Shenzhen, 518052, China.
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Shuo Chen
- Shanghai Institute of Hematology, National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Immune Therapy Institute, Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
10
|
Ravi G, Costa LJ. Bispecific T-cell engagers for treatment of multiple myeloma. Am J Hematol 2023; 98 Suppl 2:S13-S21. [PMID: 35702871 DOI: 10.1002/ajh.26628] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 11/08/2022]
Abstract
Bispecific T cell engagers (TCE) derive from monoclonal antibodies and concomitantly engage a target on the surface of cancer cell and CD3 on the surface of T-cells. TCEs promote T cell activation and lysis of tumor cells. Most TCEs in development for multiple myeloma (MM) target the B cell maturation antigen (BCMA) and differ among themselves in structure, pharmacokinetics, route and schedule of administration. CD3/BCMA TCEs produce response in ~60% of patients treated in phase 1 trials. TCEs are also in development targeting the G protein-coupled receptor, class C group 5 member D (GPRC5D) and the Fc receptor homologue 5 (FcRH5). Main toxicities are cytokine release syndrome and cytopenias. Here we review the current development and future directions of TCEs in MM.
Collapse
Affiliation(s)
- Gayathri Ravi
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Luciano J Costa
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
11
|
Rubio-Pérez L, Lázaro-Gorines R, Harwood SL, Compte M, Navarro R, Tapia-Galisteo A, Bonet J, Blanco B, Lykkemark S, Ramírez-Fernández Á, Ferreras-Gutiérrez M, Domínguez-Alonso C, Díez-Alonso L, Segura-Tudela A, Hangiu O, Erce-Llamazares A, Blanco FJ, Santos C, Rodríguez-Peralto JL, Sanz L, Álvarez-Vallina L. A PD-L1/EGFR bispecific antibody combines immune checkpoint blockade and direct anti-cancer action for an enhanced anti-tumor response. Oncoimmunology 2023; 12:2205336. [PMID: 37114242 PMCID: PMC10128431 DOI: 10.1080/2162402x.2023.2205336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) with antibodies has shown durable clinical responses in a wide range of cancer types, but the overall response rate is still limited. Other effective therapeutic modalities to increase the ICB response rates are urgently needed. New bispecific antibody (bsAb) formats combining the ICB effect and a direct action on cancer cells could improve the efficacy of current immunotherapies. Here, we report the development of a PD-L1/EGFR symmetric bsAb by fusing a dual-targeting tandem trimmer body with the human IgG1 hinge and Fc regions. The bsAb was characterized in vitro and the antitumor efficacy was evaluated in humanized mice bearing xenografts of aggressive triple-negative breast cancer and lung cancer. The IgG-like hexavalent bsAb, designated IgTT-1E, was able to simultaneously bind both EGFR and PD-L1 antigens, inhibit EGF-mediated proliferation, effectively block PD-1/PD-L1 interaction, and induce strong antigen-specific antibody-dependent cellular cytotoxicity activity in vitro. Potent therapeutic efficacies of IgTT-1E in two different humanized mouse models were observed, where tumor growth control was associated with a significantly increased proportion of CD8+ T cells. These results support the development of IgTT-1E for the treatment of EGFR+ cancers.
Collapse
Affiliation(s)
- Laura Rubio-Pérez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - Rodrigo Lázaro-Gorines
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Seandean L. Harwood
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Rocío Navarro
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Antonio Tapia-Galisteo
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Jaume Bonet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Simon Lykkemark
- Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus C, Denmark
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alejandro Segura-Tudela
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Oana Hangiu
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Ainhoa Erce-Llamazares
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Francisco J. Blanco
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| | - Cruz Santos
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - José L. Rodríguez-Peralto
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Pathology, Universidad Complutense, Madrid, Spain
- Cutaneous Oncology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Laura Sanz
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Madrid, Spain
- CONTACT Luis Álvarez-Vallina Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Avda. Cordoba s/n, Madrid28041, Spain
| |
Collapse
|
12
|
Avanzino BC, Prabhakar K, Dalvi P, Hartstein S, Kehm H, Balasubramani A, Boudreau AA, Buelow B, Chang K, Davison LM, Iyer S, Kalwit V, Lewis Wilson K, Malik-Chaudhry HK, Pierson W, Pineda G, Rangaswamy US, Saiganesh S, Schellenberger U, Ugamraj HS, Yabut RD, Buelow R, Chapman J, Trinklein ND, Harris KE. A T-cell engaging bispecific antibody with a tumor-selective bivalent folate receptor alpha binding arm for the treatment of ovarian cancer. Oncoimmunology 2022; 11:2113697. [PMID: 36016696 PMCID: PMC9397469 DOI: 10.1080/2162402x.2022.2113697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
The use of T-cell engagers (TCEs) to treat solid tumors is challenging, and several have been limited by narrow therapeutic windows due to substantial on-target, off-tumor toxicities due to the expression of low levels of target antigens on healthy tissues. Here, we describe TNB-928B, a fully human TCE that has a bivalent binding arm for folate receptor alpha (FRα) to selectively target FRα overexpressing tumor cells while avoiding the lysis of cells with low levels of FRα expression. The bivalent design of the FRα binding arm confers tumor selectivity due to low-affinity but high-avidity binding to high FRα antigen density cells. TNB-928B induces preferential effector T-cell activation, proliferation, and selective cytotoxic activity on high FRα expressing cells while sparing low FRα expressing cells. In addition, TNB-928B induces minimal cytokine release compared to a positive control TCE containing OKT3. Moreover, TNB-928B exhibits substantial ex vivo tumor cell lysis using endogenous T-cells and robust tumor clearance in vivo, promoting T-cell infiltration and antitumor activity in mouse models of ovarian cancer. TNB-928B exhibits pharmacokinetics similar to conventional antibodies, which are projected to enable favorable administration in humans. TNB-928B is a novel TCE with enhanced safety and specificity for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Brian C. Avanzino
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Kirthana Prabhakar
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Pranjali Dalvi
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Sharon Hartstein
- Teneobio, Inc, Newark, CA, United States
- Therapeutic Discovery, Amgen Inc., Newark, CA, USA
| | | | - Aarti Balasubramani
- Teneobio, Inc, Newark, CA, United States
- Therapeutic Discovery, Amgen Inc., Newark, CA, USA
| | | | - Ben Buelow
- Teneobio, Inc, Newark, CA, United States
| | | | | | | | - Vidyut Kalwit
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Kristin Lewis Wilson
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Will Pierson
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Geovanni Pineda
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Udaya S. Rangaswamy
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | - Sowmya Saiganesh
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| | | | - Harshad S. Ugamraj
- Teneobio, Inc, Newark, CA, United States
- Process Development, Amgen Inc., Newark, CA, USA
| | - Rodolfovan D. Yabut
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Jocelyn Chapman
- Division of Gynecologic Oncology, University of California, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | | | - Katherine E. Harris
- Teneobio, Inc, Newark, CA, United States
- Oncology Research, Amgen Inc., Newark, CA, USA
| |
Collapse
|
13
|
Wang SY, Zhao LN, Cheng H, Shi M, Chen W, Qi KM, Sun C, Wang X, Cao J, Xu KL. [Long-term safety and activity of humanized CD19 chimeric antigen receptor T cells for children and young adults with relapsed/refractory acute lymphoblastic leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:557-561. [PMID: 36709132 PMCID: PMC9395560 DOI: 10.3760/cma.j.issn.0253-2727.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Indexed: 11/17/2022]
Abstract
Objective: To investigate the efficacy and safety of humanized CD19-specific chimeric antigen receptor T cells (hCART19s) in treating children and young adults with relapsed/refractory acute lymphoblastic leukemia (R/R ALL) and to analyze relevant factors affecting its curative effect and prognosis. Methods: We conducted a single-center clinical trial involving 31 children and young adult patients with R/R B-ALL who were treated with humanized CD19-specific CAR-T cells (hCART19s) from May 2016 to September 2021. Results: Results showed that 27 (87.1%) patients achieved complete remission (CR) or CR with incomplete count recovery (CRi) one month after CAR-T cell infusion. During treatment, 20 (64.5%) patients developed grade 1-2 cytokine release syndrome (CRS) , and 4 (12.9%) developed grade 3 CRS. Additionally, two patients had grade 1 neurological events. During the follow-up with a median time of 19.3 months, the median event-free survival (EFS) was 15.7 months (95% CI 8.7-22.5) , and the median overall survival (OS) was 32.2 months (95% CI 10.6-53.9) . EFS and OS rates were higher in patients who have undergone hemopoietic stem cell transplantation (HSCT) than in those without [EFS: (75.0 ± 12.5) % vs (21.1 ± 9.4) %, P=0.012; OS: (75.0 ± 12.5) % vs (24.6 ± 10.2) %, P=0.035]. The EFS and OS rates were significantly lower in patients with >3 treatment lines than in those with <3 treatment lines [EFS: 0 vs (49.5±10.4) %, P<0.001; OS: 0 vs (52.0±10.8) %, P<0.001]. To the cutoff date, 12 patients presented with CD19(+) relapse, and 1 had CD19(-) relapse. Conclusion: hCART19s are effective in treating pediatric and young adult R/R ALL patients, with a low incidence of severe adverse events and reversible symptoms. Following HSCT, the number of treatment lines can affect the long-term efficacy and prognosis of pediatric and young adult R/R ALL patients.
Collapse
Affiliation(s)
- S Y Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - L N Zhao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - H Cheng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - M Shi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - W Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - K M Qi
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - C Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - X Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - J Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - K L Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
14
|
Zhang Q, Zu C, Hu Y, Huang H. CAR-T cells for cancer immunotherapy-the barriers ahead and the paths through. Int Rev Immunol 2022; 41:567-581. [PMID: 35635212 DOI: 10.1080/08830185.2022.2080820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This review discusses the major concerns and changes emerged during the rapidly extended clinical application of chimeric antigen receptor (CAR) T therapy based on our experience and understanding. In the past decades, the CAR-T cells have been questioned, sequentially, about their capability of inducing initial remission, their safety profile, their ability to sustain long-term persistence and response, and their potential to be industrialized. Significant advances, novel targeting strategies, innovative molecular structure, fine tuning of both CAR-T and host immune system, combination with other therapies, streamlined manufacturing, and etc., have been made to overcome these challenges. Although not perfectly resolved, rational pathways have been proposed to pass through the barriers. Here, we present the recent achievements on these pathways, and look into the possible future directions.
Collapse
Affiliation(s)
- Qiqi Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Cheng Zu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yongxian Hu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.,Institute of Hematology, Zhejiang University, Hangzhou, China.,Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, China
| |
Collapse
|
15
|
Rizzo D, Cerofolini L, Giuntini S, Iozzino L, Pergola C, Sacco F, Palmese A, Ravera E, Luchinat C, Baroni F, Fragai M. Epitope Mapping and Binding Assessment by Solid-State NMR Provide a Way for the Development of Biologics under the Quality by Design Paradigm. J Am Chem Soc 2022; 144:10006-10016. [PMID: 35617699 PMCID: PMC9185746 DOI: 10.1021/jacs.2c03232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
Multispecific biologics
are an emerging class of drugs, in which
antibodies and/or proteins designed to bind pharmacological targets
are covalently linked or expressed as fusion proteins to increase
both therapeutic efficacy and safety. Epitope mapping on the target
proteins provides key information to improve the affinity and also
to monitor the manufacturing process and drug stability. Solid-state
NMR has been here used to identify the pattern of the residues of
the programmed cell death ligand 1 (PD-L1) ectodomain that are involved
in the interaction with a new multispecific biological drug. This
is possible because the large size and the intrinsic flexibility of
the complexes are not limiting factors for solid-state NMR.
Collapse
Affiliation(s)
- Domenico Rizzo
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Stefano Giuntini
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Luisa Iozzino
- Analytical Development Biotech Department, Merck Serono S.p.a, Via Luigi Einaudi, 11, 00012 Guidonia, RM, Italy
| | - Carlo Pergola
- Analytical Development Biotech Department, Merck Serono S.p.a, Via Luigi Einaudi, 11, 00012 Guidonia, RM, Italy
| | - Francesca Sacco
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Analytical Development Biotech Department, Merck Serono S.p.a, Via Luigi Einaudi, 11, 00012 Guidonia, RM, Italy
| | - Angelo Palmese
- Analytical Development Biotech Department, Merck Serono S.p.a, Via Luigi Einaudi, 11, 00012 Guidonia, RM, Italy
| | - Enrico Ravera
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Fabio Baroni
- Analytical Development Biotech Department, Merck Serono S.p.a, Via Luigi Einaudi, 11, 00012 Guidonia, RM, Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy.,Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3, 50019 Sesto Fiorentino, Italy.,Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
16
|
Liu H, Liu Y, Zhao Z, Li Y, Mustafa B, Chen Z, Barve A, Jain A, Yao X, Li G, Cheng K. Discovery of Anti-PD-L1 Human Domain Antibodies for Cancer Immunotherapy. Front Immunol 2022; 13:838966. [PMID: 35444660 PMCID: PMC9013927 DOI: 10.3389/fimmu.2022.838966] [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] [Received: 12/18/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy using monoclonal antibodies targeting the PD-1/PD-L1 interaction has shown enormous success for various cancers. Despite their encouraging results in clinics, antibody-based checkpoint inhibitors have several limitations, such as poor tumor penetration. To address these limitations of monoclonal antibodies, there is a growing interest in developing low-molecular-weight checkpoint inhibitors, such as antibody fragments. Several antibody fragments targeting PD-1/PD-L1 were recently discovered using phage libraries from camel or alpaca. However, animal-derived antibody fragments may elicit unwanted immune responses, which limit their therapeutic applications. For the first time, we used a human domain antibody phage library and discovered anti-human PD-L1 human single-domain antibodies (dAbs) that block the PD-1/PD-L1 interaction. Among them, the CLV3 dAb shows the highest affinity to PD-L1. The CLV3 dAb also exhibits the highest blocking efficacy of the PD-1/PD-L1 interaction. Moreover, the CLV3 dAb significantly inhibits tumor growth in mice implanted with CT26 colon carcinoma cells. These results suggest that CLV3 dAb can be potentially used as an anti-PD-L1 inhibitor for cancer immunotherapy.
Collapse
Affiliation(s)
- Hao Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Bahaa Mustafa
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Zhijin Chen
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Ashutosh Barve
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Akshay Jain
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Xiaolan Yao
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Guangfu Li
- Department of Surgery, and Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
17
|
Ugamraj HS, Dang K, Ouisse LH, Buelow B, Chini EN, Castello G, Allison J, Clarke SC, Davison LM, Buelow R, Deng R, Iyer S, Schellenberger U, Manika SN, Bijpuria S, Musnier A, Poupon A, Cuturi MC, van Schooten W, Dalvi P. TNB-738, a biparatopic antibody, boosts intracellular NAD+ by inhibiting CD38 ecto-enzyme activity. MAbs 2022; 14:2095949. [PMID: 35867844 PMCID: PMC9311320 DOI: 10.1080/19420862.2022.2095949] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cluster of differentiation 38 (CD38) is an ecto-enzyme expressed primarily on immune cells that metabolize nicotinamide adenine dinucleotide (NAD+) to adenosine diphosphate ribose or cyclic ADP-ribose and nicotinamide. Other substrates of CD38 include nicotinamide adenine dinucleotide phosphate and nicotinamide mononucleotide, a critical NAD+ precursor in the salvage pathway. NAD+ is an important coenzyme involved in several metabolic pathways and is a required cofactor for the function of sirtuins (SIRTs) and poly (adenosine diphosphate-ribose) polymerases. Declines in NAD+ levels are associated with metabolic and inflammatory diseases, aging, and neurodegenerative disorders. To inhibit CD38 enzyme activity and boost NAD+ levels, we developed TNB-738, an anti-CD38 biparatopic antibody that pairs two non-competing heavy chain-only antibodies in a bispecific format. By simultaneously binding two distinct epitopes on CD38, TNB-738 potently inhibited its enzymatic activity, which in turn boosted intracellular NAD+ levels and SIRT activities. Due to its silenced IgG4 Fc, TNB-738 did not deplete CD38-expressing cells, in contrast to the clinically available anti-CD38 antibodies, daratumumab, and isatuximab. TNB-738 offers numerous advantages compared to other NAD-boosting therapeutics, including small molecules, and supplements, due to its long half-life, specificity, safety profile, and activity. Overall, TNB-738 represents a novel treatment with broad therapeutic potential for metabolic and inflammatory diseases associated with NAD+ deficiencies.Abbreviations: 7-AAD: 7-aminoactinomycin D; ADCC: antibody dependent cell-mediated cytotoxicity; ADCP: antibody dependent cell-mediated phagocytosis; ADPR: adenosine diphosphate ribose; APC: allophycocyanin; cADPR: cyclic ADP-ribose; cDNA: complementary DNA; BSA: bovine serum albumin; CD38: cluster of differentiation 38; CDC: complement dependent cytotoxicity; CFA: Freund's complete adjuvant; CHO: Chinese hamster ovary; CCP4: collaborative computational project, number 4; COOT: crystallographic object-oriented toolkit; DAPI: 4',6-diamidino-2-phenylindole; DNA: deoxyribonucleic acid; DSC: differential scanning calorimetry; 3D: three dimensional; εNAD+: nicotinamide 1,N6-ethenoadenine dinucleotide; ECD: extracellular domain; EGF: epidermal growth factor; FACS: fluorescence activated cell sorting; FcγR: Fc gamma receptors; FITC: fluorescein isothiocyanate; HEK: human embryonic kidney; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IgG: immunoglobulin; IFA: incomplete Freund's adjuvant; IFNγ: Interferon gamma; KB: kinetic buffer; kDa: kilodalton; KEGG: kyoto encyclopedia of genes and genomes; LDH: lactate dehydrogenase; M: molar; mM: millimolar; MFI: mean fluorescent intensity; NA: nicotinic acid; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; NAM: nicotinamide; NGS: next-generation sequencing; NHS/EDC: N-Hydroxysuccinimide/ ethyl (dimethylamino propyl) carbodiimide; Ni-NTA: nickel-nitrilotriacetic acid; nL: nanoliter; NK: natural killer; NMN: nicotinamide mononucleotide; OD: optical density; PARP: poly (adenosine diphosphate-ribose) polymerase; PBS: phosphate-buffered saline; PBMC: peripheral blood mononuclear cell; PDB: protein data bank; PE: phycoerythrin; PISA: protein interfaces, surfaces, and assemblies: PK: pharmacokinetics; mol: picomolar; RNA: ribonucleic acid; RLU: relative luminescence units; rpm: rotations per minute; RU: resonance unit; SEC: size exclusion chromatography; SEM: standard error of the mean; SIRT: sirtuins; SPR: surface plasmon resonance; µg: microgram; µM: micromolar; µL: microliter.
Collapse
Affiliation(s)
| | | | - Laure-Hélène Ouisse
- INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Université, Nantes, France
| | | | - Eduardo N Chini
- Department of Anesthesiology and Perioperative Medicine, Kogod Center on Aging, Mitochondrial Care Center, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | | | | | | | - Rong Deng
- R&D Q-Pharm consulting LLC, Pleasanton, California, USA
| | | | | | | | | | | | | | - Maria Cristina Cuturi
- INSERM, Centre de Recherche en Transplantation et Immunologie UMR1064, Université, Nantes, France
| | | | | |
Collapse
|
18
|
Silva-Pilipich N, Smerdou C, Vanrell L. A Small Virus to Deliver Small Antibodies: New Targeted Therapies Based on AAV Delivery of Nanobodies. Microorganisms 2021; 9:microorganisms9091956. [PMID: 34576851 PMCID: PMC8465657 DOI: 10.3390/microorganisms9091956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
Nanobodies are camelid-derived single-domain antibodies that present some advantages versus conventional antibodies, such as a smaller size, and higher tissue penetrability, stability, and hydrophilicity. Although nanobodies can be delivered as proteins, in vivo expression from adeno-associated viral (AAV) vectors represents an attractive strategy. This is due to the fact that AAV vectors, that can provide long-term expression of recombinant genes, have shown an excellent safety profile, and can accommodate genes for one or several nanobodies. In fact, several studies showed that AAV vectors can provide sustained nanobody expression both locally or systemically in preclinical models of human diseases. Some of the pathologies addressed with this technology include cancer, neurological, cardiovascular, infectious, and genetic diseases. Depending on the indication, AAV-delivered nanobodies can be expressed extracellularly or inside cells. Intracellular nanobodies or “intrabodies” carry out their function by interacting with cell proteins involved in disease and have also been designed to help elucidate cellular mechanisms by interfering with normal cell processes. Finally, nanobodies can also be used to retarget AAV vectors, when tethered to viral capsid proteins. This review covers applications in which AAV vectors have been used to deliver nanobodies, with a focus on their therapeutic use.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain;
- Correspondence: (C.S.); (L.V.); Tel.: +34-948194700 (C.S.); +508-29021505 (L.V.); Fax: +34-948194717 (C.S.)
| | - Lucía Vanrell
- Biotechnology Laboratory, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, Montevideo 11100, Uruguay
- Nanogrow Biotech, CIE BIO Incubator, Mercedes 1237, Montevideo 11100, Uruguay
- Correspondence: (C.S.); (L.V.); Tel.: +34-948194700 (C.S.); +508-29021505 (L.V.); Fax: +34-948194717 (C.S.)
| |
Collapse
|
19
|
Lower Somatic Mutation Levels in the λ Light-Chain Repertoires with Chronic HBV Infection. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5525369. [PMID: 34239579 PMCID: PMC8203402 DOI: 10.1155/2021/5525369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/27/2021] [Indexed: 11/21/2022]
Abstract
To investigate the characteristics of the immunoglobulin light-chain repertoires with chronic HBV infection, the high-throughput sequencing and IMGT/HighV-QUEST were adapted to analyze the κ (IgK) and λ (IgL) light-chain repertoires from the inactive HBV carriers (IHB) and the healthy adults (HH). The comparative analysis revealed high similarity between the κ light-chain repertoires of the HBV carriers and the healthy adults. Nevertheless, the proportion of IGLV genes with ≥90% identity as the germline genes was higher in the IgL light-chain repertoire of the IHB library compared with that of HH library (74.6% vs. 69.1%). Besides, the frequency of amino acid mutations in the CDR1 regions was significantly lower in the IgL light-chain repertoire of the IHB library than that of the HH library (65.52% vs. 56.0%). These results suggested the lower somatic mutation level in the IgL repertoire of IHB library, which might indicate the biased selection of IGLV genes in the IgL repertoire with chronic HBV infection. These findings might lead to a better understanding of the characteristics of the light-chain repertoires of HBV chronically infected individuals.
Collapse
|
20
|
Dang K, Castello G, Clarke SC, Li Y, Balasubramani A, Boudreau A, Davison L, Harris KE, Pham D, Sankaran P, Ugamraj HS, Deng R, Kwek S, Starzinski A, Iyer S, van Schooten W, Schellenberger U, Sun W, Trinklein ND, Buelow R, Buelow B, Fong L, Dalvi P. Attenuating CD3 affinity in a PSMAxCD3 bispecific antibody enables killing of prostate tumor cells with reduced cytokine release. J Immunother Cancer 2021; 9:e002488. [PMID: 34088740 PMCID: PMC8183203 DOI: 10.1136/jitc-2021-002488] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Therapeutic options currently available for metastatic castration-resistant prostate cancer (mCRPC) do not extend median overall survival >6 months. Therefore, the development of novel and effective therapies for mCRPC represents an urgent medical need. T cell engagers (TCEs) have emerged as a promising approach for the treatment of mCRPC due to their targeted mechanism of action. However, challenges remain in the clinic due to the limited efficacy of TCEs observed thus far in solid tumors as well as the toxicities associated with cytokine release syndrome (CRS) due to the usage of high-affinity anti-CD3 moieties such as OKT3. METHODS Using genetically engineered transgenic rats (UniRat and OmniFlic) that express fully human IgG antibodies together with an NGS-based antibody discovery pipeline, we developed TNB-585, an anti-CD3xPSMA TCE for the treatment of mCRPC. TNB-585 pairs a tumor-targeting anti-PSMA arm together with a unique, low-affinity anti-CD3 arm in bispecific format. We tested TNB-585 in T cell-redirected cytotoxicity assays against PSMA+ tumor cells in both two-dimensional (2D) cultures and three-dimensional (3D) spheroids as well as against patient-derived prostate tumor cells. Cytokines were measured in culture supernatants to assess the ability of TNB-585 to induce tumor killing with low cytokine release. TNB-585-mediated T cell activation, proliferation, and cytotoxic granule formation were measured to investigate the mechanism of action. Additionally, TNB-585 efficacy was evaluated in vivo against C4-2 tumor-bearing NCG mice. RESULTS In vitro, TNB-585 induced activation and proliferation of human T cells resulting in the killing of PSMA+ prostate tumor cells in both 2D cultures and 3D spheroids with minimal cytokine release and reduced regulatory T cell activation compared with a positive control antibody that contains the same anti-PSMA arm but a higher affinity anti-CD3 arm (comparable with OKT3). In addition, TNB-585 demonstrated potent efficacy against patient-derived prostate tumors ex vivo and induced immune cell infiltration and dose-dependent tumor regression in vivo. CONCLUSIONS Our data suggest that TNB-585, with its low-affinity anti-CD3, may be efficacious while inducing a lower incidence and severity of CRS in patients with prostate cancer compared with TCEs that incorporate high-affinity anti-CD3 domains.
Collapse
Affiliation(s)
| | | | | | - Yuping Li
- Teneobio, Inc, Newark, California, USA
| | | | | | | | | | - Duy Pham
- Teneobio, Inc, Newark, California, USA
| | | | | | - Rong Deng
- Teneobio, Inc, Newark, California, USA
| | - Serena Kwek
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | - Alec Starzinski
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | | - Lawrence Fong
- Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
21
|
Harris KE, Lorentsen KJ, Malik-Chaudhry HK, Loughlin K, Basappa HM, Hartstein S, Ahmil G, Allen NS, Avanzino BC, Balasubramani A, Boudreau AA, Chang K, Cuturi MC, Davison LM, Ho DM, Iyer S, Rangaswamy US, Sankaran P, Schellenberger U, Buelow R, Trinklein ND. A bispecific antibody agonist of the IL-2 heterodimeric receptor preferentially promotes in vivo expansion of CD8 and NK cells. Sci Rep 2021; 11:10592. [PMID: 34011961 PMCID: PMC8134639 DOI: 10.1038/s41598-021-90096-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/06/2021] [Indexed: 11/09/2022] Open
Abstract
The use of recombinant interleukin-2 (IL-2) as a therapeutic protein has been limited by significant toxicities despite its demonstrated ability to induce durable tumor-regression in cancer patients. The adverse events and limited efficacy of IL-2 treatment are due to the preferential binding of IL-2 to cells that express the high-affinity, trimeric receptor, IL-2Rαβγ such as endothelial cells and T-regulatory cells, respectively. Here, we describe a novel bispecific heavy-chain only antibody which binds to and activates signaling through the heterodimeric IL-2Rβγ receptor complex that is expressed on resting T-cells and NK cells. By avoiding binding to IL-2Rα, this molecule circumvents the preferential T-reg activation of native IL-2, while maintaining the robust stimulatory effects on T-cells and NK-cells in vitro. In vivo studies in both mice and cynomolgus monkeys confirm the molecule's in vivo biological activity, extended pharmacodynamics due to the Fc portion of the molecule, and enhanced safety profile. Together, these results demonstrate that the bispecific antibody is a safe and effective IL-2R agonist that harnesses the benefits of the IL-2 signaling pathway as a potential anti-cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ghenima Ahmil
- Inserm, Centre de Recherche en Transplantation Et Immunologie, UMR 1064, Nantes Université, 44000, Nantes, France
| | | | | | | | | | | | - Maria-Cristina Cuturi
- Inserm, Centre de Recherche en Transplantation Et Immunologie, UMR 1064, Nantes Université, 44000, Nantes, France
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bélanger K, Tanha J. High-efficacy, high-manufacturability human VH domain antibody therapeutics from transgenic sources. Protein Eng Des Sel 2021; 34:6276122. [PMID: 33991089 DOI: 10.1093/protein/gzab012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 11/14/2022] Open
Abstract
Interest in single-domain antibodies (sdAbs) stems from their unique structural/pronounced, hence therapeutically desirable, features. From the outset-as therapeutic modalities-human antibody heavy chain variable domains (VHs) attracted a particular attention compared with 'naturally-occurring' camelid and shark heavy-chain-only antibody variable domains (VHHs and VNARs, respectively) due to their perceived lack of immunogenicity. However, they have not quite lived up to their initial promise as the VH hits, primarily mined from synthetic VH phage display libraries, have too often been plagued with aggregation tendencies, low solubility and low affinity. Largely unexplored, synthetic camelized human VH display libraries appeared to have remediated the aggregation problem, but the low affinity of the VH hits still persisted, requiring undertaking additional, laborious affinity maturation steps to render VHs therapeutically feasible. A wholesome resolution has recently emerged with the development of non-canonical transgenic rodent antibody discovery platforms that appear to facilely and profusely generate high affinity, high solubility and aggregation-resistant human VHs.
Collapse
Affiliation(s)
- Kasandra Bélanger
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Jamshid Tanha
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada.,Department of Biochemistry, Microbiology & Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| |
Collapse
|
23
|
Chenouard V, Remy S, Tesson L, Ménoret S, Ouisse LH, Cherifi Y, Anegon I. Advances in Genome Editing and Application to the Generation of Genetically Modified Rat Models. Front Genet 2021; 12:615491. [PMID: 33959146 PMCID: PMC8093876 DOI: 10.3389/fgene.2021.615491] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
The rat has been extensively used as a small animal model. Many genetically engineered rat models have emerged in the last two decades, and the advent of gene-specific nucleases has accelerated their generation in recent years. This review covers the techniques and advances used to generate genetically engineered rat lines and their application to the development of rat models more broadly, such as conditional knockouts and reporter gene strains. In addition, genome-editing techniques that remain to be explored in the rat are discussed. The review also focuses more particularly on two areas in which extensive work has been done: human genetic diseases and immune system analysis. Models are thoroughly described in these two areas and highlight the competitive advantages of rat models over available corresponding mouse versions. The objective of this review is to provide a comprehensive description of the advantages and potential of rat models for addressing specific scientific questions and to characterize the best genome-engineering tools for developing new projects.
Collapse
Affiliation(s)
- Vanessa Chenouard
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- genOway, Lyon, France
| | - Séverine Remy
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Laurent Tesson
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | - Séverine Ménoret
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
- CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes Université, Nantes, France
| | - Laure-Hélène Ouisse
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| | | | - Ignacio Anegon
- CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Université de Nantes, Nantes, France
| |
Collapse
|
24
|
Rossotti MA, Bélanger K, Henry KA, Tanha J. Immunogenicity and humanization of single‐domain antibodies. FEBS J 2021; 289:4304-4327. [DOI: 10.1111/febs.15809] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/26/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Martin A. Rossotti
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
| | - Kasandra Bélanger
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
| | - Kevin A. Henry
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine University of Ottawa Canada
| | - Jamshid Tanha
- Life Sciences Division Human Health Therapeutics Research Centre National Research Council Canada Ottawa Canada
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine University of Ottawa Canada
| |
Collapse
|
25
|
Wagner DL, Fritsche E, Pulsipher MA, Ahmed N, Hamieh M, Hegde M, Ruella M, Savoldo B, Shah NN, Turtle CJ, Wayne AS, Abou-El-Enein M. Immunogenicity of CAR T cells in cancer therapy. Nat Rev Clin Oncol 2021; 18:379-393. [PMID: 33633361 PMCID: PMC8923136 DOI: 10.1038/s41571-021-00476-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2021] [Indexed: 12/14/2022]
Abstract
Patient-derived T cells genetically reprogrammed to express CD19-specific chimeric antigen receptors (CARs) have shown remarkable clinical responses and are commercially available for the treatment of patients with certain advanced-stage B cell malignancies. Nonetheless, several trials have revealed pre-existing and/or treatment-induced immune responses to the mouse-derived single-chain variable fragments included in these constructs. These responses might have contributed to both treatment failure and the limited success of redosing strategies observed in some patients. Data from early phase clinical trials suggest that CAR T cells are also associated with immunogenicity-related events in patients with solid tumours. Generally, the clinical implications of anti-CAR immune responses are poorly understood and highly variable between different CAR constructs and malignancies. These observations highlight an urgent need to uncover the mechanisms of immunogenicity in patients receiving CAR T cells and develop validated assays to enable clinical detection. In this Review, we describe the current clinical evidence of anti-CAR immune responses and discuss how new CAR T cell technologies might impact the risk of immunogenicity. We then suggest ways to reduce the risks of anti-CAR immune responses to CAR T cell products that are advancing towards the clinic. Finally, we summarize measures that investigators could consider in order to systematically monitor and better comprehend the possible effects of immunogenicity during trials involving CAR T cells as well as in routine clinical practice.
Collapse
Affiliation(s)
- Dimitrios L Wagner
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Transfusion Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Enrico Fritsche
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael A Pulsipher
- Section of Transplantation and Cellular Therapy, Children's Hospital Los Angeles Cancer and Blood Disease Institute, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Nabil Ahmed
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Mohamad Hamieh
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, NY, USA
| | - Meenakshi Hegde
- Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania Philadelphia, Philadelphia, PA, USA.,Division of Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cameron J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA.,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Alan S Wayne
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mohamed Abou-El-Enein
- Berlin Center for Advanced Therapies (BeCAT) and Berlin Institute of Health (BIH) Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Division of Medical Oncology, Department of Medicine, and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. .,Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Flego M, Colotti G, Ascione A, Dupuis ML, Petrucci E, Riccioni R, Andreotti M, Raggi C, Boe A, Barca S, Gellini M, Vella S, Mallano A. Isolation and preliminary characterization of a human 'phage display'-derived antibody against neural adhesion molecule-1 antigen interfering with fibroblast growth factor receptor-1 binding. Hum Antibodies 2021; 29:63-84. [PMID: 33164927 DOI: 10.3233/hab-200431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
BACKGROUND The NCAM or CD56 antigen is a cell surface glycoprotein belonging to the immunoglobulin super-family involved in cell-cell and cell-matrix adhesion. NCAM is also over-expressed in many tumour types and is considered a tumour associated antigen, even if its role and biological mechanisms implicated in tumour progression and metastasis have not yet to be elucidated. In particular, it is quite well documented the role of the interaction between the NCAM protein and the fibroblast growth factor receptor-1 in metastasis and invasion, especially in the ovarian cancer progression. OBJECTIVE Here we describe the isolation and preliminary characterization of a novel human anti-NCAM single chain Fragment variable antibody able to specifically bind NCAM-expressing cells, including epithelial ovarian cancer cells. METHODS The antibody was isolate by phage display selection and was characterized by ELISA, FACS analysis and SPR experiments. Interference in EOC migration was analyzed by scratch test. RESULTS It binds a partially linear epitope lying in the membrane proximal region of two fibronectin-like domains with a dissociation constant of 3.43 × 10-8 M. Interestingly, it was shown to interfere with the NCAM-FGFR1 binding and to partially decrease migration of EOC cells. CONCLUSIONS According to our knowledge, this is the first completely human antibody able to interfere with this newly individuated cancer mechanism.
Collapse
Affiliation(s)
- Michela Flego
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council, c/o Department Biochemical Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandro Ascione
- National Center for Control and Evaluation of Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Luisa Dupuis
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Eleonora Petrucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta Riccioni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Andreotti
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Carla Raggi
- National Center for Control and Evaluation of Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Barca
- National Center for Drug Research and Evaluation Unit of Preclinical and Clinical Evaluation of Medicinal Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Mara Gellini
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Vella
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Mallano
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
27
|
Malik-Chaudhry HK, Prabhakar K, Ugamraj HS, Boudreau AA, Buelow B, Dang K, Davison LM, Harris KE, Jorgensen B, Ogana H, Pham D, Schellenberger U, Van Schooten W, Buelow R, Iyer S, Trinklein ND, Rangaswamy US. TNB-486 induces potent tumor cell cytotoxicity coupled with low cytokine release in preclinical models of B-NHL. MAbs 2021; 13:1890411. [PMID: 33818299 PMCID: PMC8023237 DOI: 10.1080/19420862.2021.1890411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
The therapeutic potential of targeting CD19 in B cell malignancies has garnered attention in the past decade, resulting in the introduction of novel immunotherapy agents. Encouraging clinical data have been reported for T cell-based targeting agents, such as anti-CD19/CD3 bispecific T-cell engager blinatumomab and chimeric antigen receptor (CAR)-T therapies, for acute lymphoblastic leukemia and B cell non-Hodgkin lymphoma (B-NHL). However, clinical use of both blinatumomab and CAR-T therapies has been limited due to unfavorable pharmacokinetics (PK), significant toxicity associated with cytokine release syndrome and neurotoxicity, and manufacturing challenges. We present here a fully human CD19xCD3 bispecific antibody (TNB-486) for the treatment of B-NHL that could address the limitations of the current approved treatments. In the presence of CD19+ target cells and T cells, TNB-486 induces tumor cell lysis with minimal cytokine release, when compared to a positive control. In vivo, TNB-486 clears CD19+ tumor cells in immunocompromised mice in the presence of human peripheral blood mononuclear cells in multiple models. Additionally, the PK of TNB-486 in mice or cynomolgus monkeys is similar to conventional antibodies. This new T cell engaging bispecific antibody targeting CD19 represents a novel therapeutic that induces potent T cell-mediated tumor-cell cytotoxicity uncoupled from high levels of cytokine release, making it an attractive candidate for B-NHL therapy.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antigens, CD19/immunology
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacology
- CD3 Complex/antagonists & inhibitors
- CD3 Complex/immunology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Coculture Techniques
- Cytokines/metabolism
- Cytotoxicity, Immunologic/drug effects
- Humans
- K562 Cells
- Lymphocyte Activation/drug effects
- Lymphoma, Non-Hodgkin/drug therapy
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/metabolism
- Macaca fascicularis
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, SCID
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
| | | | | | | | | | - Kevin Dang
- Teneobio, Inc., Newark, CA, United States
| | | | | | | | - Heather Ogana
- Graduate Program in Cancer Biology and Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Duy Pham
- Teneobio, Inc., Newark, CA, United States
| | | | | | | | | | | | | |
Collapse
|
28
|
Transgenic Animals for the Generation of Human Antibodies. LEARNING MATERIALS IN BIOSCIENCES 2021. [DOI: 10.1007/978-3-030-54630-4_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Sawant MS, Streu CN, Wu L, Tessier PM. Toward Drug-Like Multispecific Antibodies by Design. Int J Mol Sci 2020; 21:E7496. [PMID: 33053650 PMCID: PMC7589779 DOI: 10.3390/ijms21207496] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/02/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
The success of antibody therapeutics is strongly influenced by their multifunctional nature that couples antigen recognition mediated by their variable regions with effector functions and half-life extension mediated by a subset of their constant regions. Nevertheless, the monospecific IgG format is not optimal for many therapeutic applications, and this has led to the design of a vast number of unique multispecific antibody formats that enable targeting of multiple antigens or multiple epitopes on the same antigen. Despite the diversity of these formats, a common challenge in generating multispecific antibodies is that they display suboptimal physical and chemical properties relative to conventional IgGs and are more difficult to develop into therapeutics. Here we review advances in the design and engineering of multispecific antibodies with drug-like properties, including favorable stability, solubility, viscosity, specificity and pharmacokinetic properties. We also highlight emerging experimental and computational methods for improving the next generation of multispecific antibodies, as well as their constituent antibody fragments, with natural IgG-like properties. Finally, we identify several outstanding challenges that need to be addressed to increase the success of multispecific antibodies in the clinic.
Collapse
Affiliation(s)
- Manali S. Sawant
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Craig N. Streu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemistry, Albion College, Albion, MI 49224, USA
| | - Lina Wu
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M. Tessier
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (M.S.S.); (C.N.S.)
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
30
|
Xu S, Yang H, Zhuo Y, Yu Y, Liao H, Li S, Yue Y, Su K, Zhang Z. Production of Autoreactive Heavy Chain-Only Antibodies in Systemic Lupus Erythematosus. Front Immunol 2020; 11:632. [PMID: 32431693 PMCID: PMC7214812 DOI: 10.3389/fimmu.2020.00632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/19/2020] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by the overproduction of high-affinity autoreactive antibodies. Here, we show that more than 65.8% of 222 recombinant antibodies derived from 8 SLE patients can be secreted as heavy chain-only antibodies (HCAbs) when expressed in HEK-293T cells. The secretion of HCAbs follows the conventional endoplasmic reticulum-Golgi apparatus pathway, despite triggering a weaker unfolded protein response (UPR). Many of the purified SLE HCAbs remain autoreactive and have an even higher affinity for dsDNA, Sm, nucleosome, and cardiolipin than HCAbs from healthy individuals. Extended analyses of the CDR3 region and the heavy chain variable (VH) region of HCAb F3 show that the VH region is responsible for IgH secretion, while the CDR3 region determines its reactivity. Such a high frequency of HCAb secretion cannot fully concur with our current understanding of antibody assembly and secretion. The presence of a large proportion of autoreactive HCAbs in SLE reveals a novel mechanism for the generation of autoreactive antibodies in lupus.
Collapse
Affiliation(s)
- Shu Xu
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Hong Yang
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yue Zhuo
- Health Management Center, Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yangsheng Yu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Hongyan Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Song Li
- Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, China
| | - Yinshi Yue
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kaihong Su
- Education-Microbiology/Immunology, Department of Medical Education, California University of Science and Medicine, San Bernardino, CA, United States
| | - Zhixin Zhang
- Health Management Center, Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
31
|
Li Z, Zhang M, Zheng S, Song Y, Cheng X, Yu D, Du L, Ren L, Han H, Zhao Y. Genetic removal of the CH1 exon leads to the production of hypofunctional heavy chain-only IgG2a in rats. Transgenic Res 2020; 29:199-213. [PMID: 32078126 DOI: 10.1007/s11248-020-00189-9] [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: 11/26/2019] [Accepted: 01/04/2020] [Indexed: 12/01/2022]
Abstract
Despite great values in many applications, heavy chain-only antibodies (HcAbs) are naturally only produced in camelids and sharks, which are not easy to access and handle. Production of the type of antibodies in small laboratory animals would remarkably facilitate their applications. We previously reported a mouse line in which the CH1 exon of mouse γ1 was deleted that could express heavy chain-only IgG1 antibodies. However, these mice showed an extremely weak IgG1 response to specific antigens when immunized, and we could only achieve single VH domains with low affinity to antigens using these mice. One possibility is that the mouse germline VH repertoire was not sufficient to support the expression of functional heavy chain-only antibodies. In this study, we report the generation of a rat line in which the CH1 exon of the γ2a gene was removed and the γ1 and γ2b genes were silenced. Although the genetically modified rats expressed heavy chain-only IgG2a, they also exhibited a very weak IgG2a response to antigen immunization. Panning of a phage library constructed using IgG2a VH segments amplified from immunized rats identified antigen-specific single VH antibodies, which also exhibited much lower affinity than that of commercial mAbs. Together with our previous report, this study suggests that the simple genetic removal of the CH1 exon does not guarantee the successful expression of functional heavy chain-only antibodies.
Collapse
Affiliation(s)
- Zhenrong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Ming Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Shunan Zheng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Yu Song
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Xueqian Cheng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Di Yu
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Lijuan Du
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Liming Ren
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Haitang Han
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China.
| | - Yaofeng Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Science, National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing, 100193, People's Republic of China.
| |
Collapse
|
32
|
Anti-BCMA chimeric antigen receptors with fully human heavy-chain-only antigen recognition domains. Nat Commun 2020; 11:283. [PMID: 31941907 PMCID: PMC6962219 DOI: 10.1038/s41467-019-14119-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 12/13/2019] [Indexed: 12/25/2022] Open
Abstract
Chimeric antigen receptor (CAR)-expressing T cells targeting B-cell maturation antigen (BCMA) have activity against multiple myeloma, but improvements in anti-BCMA CARs are needed. We demonstrated recipient anti-CAR T-cell responses against a murine single-chain variable fragment (scFv) used clinically in anti-BCMA CARs. To bypass potential anti-CAR immunogenicity and to reduce CAR binding domain size, here we designed CARs with antigen-recognition domains consisting of only a fully human heavy-chain variable domain without a light-chain domain. A CAR designated FHVH33-CD8BBZ contains a fully human heavy-chain variable domain (FHVH) plus 4-1BB and CD3ζ domains. T cells expressing FHVH33-CD8BBZ exhibit similar cytokine release, degranulation, and mouse tumor eradication as a CAR that is identical except for substitution of a scFv for FHVH33. Inclusion of 4-1BB is critical for reducing activation-induced cell death and promoting survival of T cells expressing FHVH33-containing CARs. Our results indicate that heavy-chain-only anti-BCMA CARs are suitable for evaluation in a clinical trial.
Collapse
|
33
|
Teng Y, Young JL, Edwards B, Hayes P, Thompson L, Johnston C, Edwards C, Sanders Y, Writer M, Pinto D, Zhang Y, Roode M, Chovanec P, Matheson L, Corcoran AE, Fernandez A, Montoliu L, Rossi B, Tosato V, Gjuracic K, Nikitin D, Bruschi C, McGuinness B, Sandal T, Romanos M. Diverse human V H antibody fragments with bio-therapeutic properties from the Crescendo Mouse. N Biotechnol 2019; 55:65-76. [PMID: 31600579 DOI: 10.1016/j.nbt.2019.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/04/2019] [Accepted: 10/04/2019] [Indexed: 01/26/2023]
Abstract
We describe the 'Crescendo Mouse', a human VH transgenic platform combining an engineered heavy chain locus with diverse human heavy chain V, D and J genes, a modified mouse Cγ1 gene and complete 3' regulatory region, in a triple knock-out (TKO) mouse background devoid of endogenous immunoglobulin expression. The addition of the engineered heavy chain locus to the TKO mouse restored B cell development, giving rise to functional B cells that responded to immunization with a diverse response that comprised entirely 'heavy chain only' antibodies. Heavy chain variable (VH) domain libraries were rapidly mined using phage display technology, yielding diverse high-affinity human VH that had undergone somatic hypermutation, lacked aggregation and showed enhanced expression in E. coli. The Crescendo Mouse produces human VH fragments, or Humabody® VH, with excellent bio-therapeutic potential, as exemplified here by the generation of antagonistic Humabody® VH specific for human IL17A and IL17RA.
Collapse
Affiliation(s)
- Yumin Teng
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Joyce L Young
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Bryan Edwards
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Philip Hayes
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Lorraine Thompson
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Colette Johnston
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Carolyn Edwards
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Yun Sanders
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Michele Writer
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Debora Pinto
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Yanjing Zhang
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Mila Roode
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Peter Chovanec
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Louise Matheson
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Anne E Corcoran
- Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Almudena Fernandez
- Centro Nacional de Biotecnologia (CNB-CSIC) & CIBER de Enfermedades Raras (CIBERER-ISCIII), Darwin 3, 28049, Madrid, Spain
| | - Lluis Montoliu
- Centro Nacional de Biotecnologia (CNB-CSIC) & CIBER de Enfermedades Raras (CIBERER-ISCIII), Darwin 3, 28049, Madrid, Spain
| | - Beatrice Rossi
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Valentina Tosato
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Kresimir Gjuracic
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Dmitri Nikitin
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Carlo Bruschi
- International Centre for Genetic Engineering and Biotechnology, Yeast Molecular Genetics Laboratory, Padriciano 99, 34149, Trieste, Italy
| | - Brian McGuinness
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Thomas Sandal
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Mike Romanos
- Crescendo Biologics Ltd, Babraham Research Campus, Cambridge, CB22 3AT, UK
| |
Collapse
|
34
|
Peptide Conjugates with Small Molecules Designed to Enhance Efficacy and Safety. Molecules 2019; 24:molecules24101855. [PMID: 31091786 PMCID: PMC6572008 DOI: 10.3390/molecules24101855] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/17/2022] Open
Abstract
Peptides constitute molecular diversity with unique molecular mechanisms of action that are proven indispensable in the management of many human diseases, but of only a mere fraction relative to more traditional small molecule-based medicines. The integration of these two therapeutic modalities offers the potential to enhance and broaden pharmacology while minimizing dose-dependent toxicology. This review summarizes numerous advances in drug design, synthesis and development that provide direction for next-generation research endeavors in this field. Medicinal studies in this area have largely focused upon the application of peptides to selectively enhance small molecule cytotoxicity to more effectively treat multiple oncologic diseases. To a lesser and steadily emerging extent peptides are being therapeutically employed to complement and diversify the pharmacology of small molecule drugs in diseases other than just cancer. No matter the disease, the purpose of the molecular integration remains constant and it is to achieve superior therapeutic outcomes with diminished adverse effects. We review linker technology and conjugation chemistries that have enabled integrated and targeted pharmacology with controlled release. Finally, we offer our perspective on opportunities and obstacles in the field.
Collapse
|
35
|
Trinklein ND, Pham D, Schellenberger U, Buelow B, Boudreau A, Choudhry P, Clarke SC, Dang K, Harris KE, Iyer S, Jorgensen B, Pratap PP, Rangaswamy US, Ugamraj HS, Vafa O, Wiita AP, van Schooten W, Buelow R, Force Aldred S. Efficient tumor killing and minimal cytokine release with novel T-cell agonist bispecific antibodies. MAbs 2019; 11:639-652. [PMID: 30698484 PMCID: PMC6601548 DOI: 10.1080/19420862.2019.1574521] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.
Collapse
Affiliation(s)
| | - Duy Pham
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Ben Buelow
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | - Priya Choudhry
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | - Kevin Dang
- a Teneobio, Inc ., Menlo Park , CA , USA
| | | | | | | | | | | | | | - Omid Vafa
- a Teneobio, Inc ., Menlo Park , CA , USA
| | - Arun P Wiita
- b Department of Laboratory Medicine , University of California , San Francisco , CA , USA
| | | | | | | |
Collapse
|