1
|
Schnider ST, Vigano MA, Affolter M, Aguilar G. Functionalized Protein Binders in Developmental Biology. Annu Rev Cell Dev Biol 2024; 40:119-142. [PMID: 39038471 DOI: 10.1146/annurev-cellbio-112122-025214] [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] [Indexed: 07/24/2024]
Abstract
Developmental biology has greatly profited from genetic and reverse genetic approaches to indirectly studying protein function. More recently, nanobodies and other protein binders derived from different synthetic scaffolds have been used to directly dissect protein function. Protein binders have been fused to functional domains, such as to lead to protein degradation, relocalization, visualization, or posttranslational modification of the target protein upon binding. The use of such functionalized protein binders has allowed the study of the proteome during development in an unprecedented manner. In the coming years, the advent of the computational design of protein binders, together with further advances in scaffold engineering and synthetic biology, will fuel the development of novel protein binder-based technologies. Studying the proteome with increased precision will contribute to a better understanding of the immense molecular complexities hidden in each step along the way to generate form and function during development.
Collapse
Affiliation(s)
| | | | | | - Gustavo Aguilar
- Current affiliation: Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
- Biozentrum, Universität Basel, Basel, Switzerland;
| |
Collapse
|
2
|
Freitag PC, Kolibius J, Wieboldt R, Weber R, Hartmann KP, van Gogh M, Brücher D, Läubli H, Plückthun A. DARPin-fused T cell engager for adenovirus-mediated cancer therapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200821. [PMID: 39021370 PMCID: PMC11253662 DOI: 10.1016/j.omton.2024.200821] [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: 11/13/2023] [Revised: 04/03/2024] [Accepted: 05/28/2024] [Indexed: 07/20/2024]
Abstract
Bispecific T cell engagers are a promising class of therapeutic proteins for cancer therapy. Their potency and small size often come with systemic toxicity and short half-life, making intravenous administration cumbersome. These limitations can be overcome by tumor-specific in situ expression, allowing high local accumulation while reducing systemic concentrations. However, encoding T cell engagers in viral or non-viral vectors and expressing them in situ ablates all forms of quality control performed during recombinant protein production. It is therefore vital to design constructs that feature minimal domain mispairing, and increased homogeneity of the therapeutic product. Here, we report a T cell engager architecture specifically designed for vector-mediated immunotherapy. It is based on a fusion of a designed ankyrin repeat protein (DARPin) to a CD3-targeting single-chain antibody fragment, termed DATE (DARPin-fused T cell Engager). The DATE induces potent T cell-mediated killing of HER2+ cancer cells, both as recombinantly produced therapeutic protein and as in situ expressed payload from a HER2+-retargeted high-capacity adenoviral vector (HC-AdV). We report remarkable tumor remission, DATE accumulation, and T cell infiltration through in situ expression mediated by a HER2+-retargeted HC-AdV in vivo. Our results support further investigations and developments of DATEs as payloads for vector-mediated immunotherapy.
Collapse
Affiliation(s)
- Patrick C. Freitag
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jonas Kolibius
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ronja Wieboldt
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Remi Weber
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - K. Patricia Hartmann
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Merel van Gogh
- Department of Physiology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Dominik Brücher
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University Hospital and University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
3
|
Xie S, Shi W, Duan S, Huang X, Liu A, Hou X, Lin X, Zhong D, Sun S, Ding Z, Yang X, Chen X, Lu X. A nanobody-guided multifunctional T cell engager promotes strong anti-tumor responses via synergistic immuno-photothermal effects. J Nanobiotechnology 2024; 22:561. [PMID: 39272205 PMCID: PMC11401270 DOI: 10.1186/s12951-024-02781-1] [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: 04/30/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND T cell-based immunotherapies are facing great challenges in the recruitment and activation of tumor-specific T cells against solid tumors. Among which, utilizing nanobody (Nb) or nanobodies (Nbs) to construct T cell engager has emerged as a more practical potential for enhancing the anti-tumor effectiveness of T cells. Here, we designed a new Nb-guided multifunctional T cell engager (Nb-MuTE) that not only recruited effector T cells into the tumor tissues, but also efficiently activated T cells anti-tumor immunity when synergies with photothermal effect. RESULTS The Nb-MuTE, which was constructed based on an indocyanine green (ICG)-containing liposome with surface conjugation of CD105 and CD3 Nbs, and showed excellent targetability to both tumor and T cells, following enhancement of activation, proliferation and cytokine secretion of tumor-specific T cells. Notably, the immunological anti-tumor functions of Nb-MuTE-mediated T cells were further enhanced by the ICG-induced photothermal effect in vitro and in vivo. CONCLUSIONS Such a new platform Nb-MuTE provides a practical and "all-in-one" strategy to potentiate T cell responses for the treatment of solid tumor in clinic.
Collapse
Affiliation(s)
- Shenxia Xie
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Wei Shi
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Siliang Duan
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545005, P. R. China
| | - Xianing Huang
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Aiqun Liu
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Xiaoqiong Hou
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
- School of Basic Medical Science, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Xuandong Lin
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Dani Zhong
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Shuyang Sun
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Ziqiang Ding
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China
| | - Xiaomei Yang
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China.
- School of Basic Medical Science, Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China.
| | - Xiaoyuan Chen
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| | - Xiaoling Lu
- Guangxi Key Laboratory of Nanobody Research, Guangxi Nanobody Engineering Research Center, College of Stomatology, Hospital of Stomatology Guangxi Medical University, Nanning, Guangxi, 530021, P. R. China.
| |
Collapse
|
4
|
Wang X, Ingavat N, Liew JM, Dzulkiflie N, Loh HP, Kok YJ, Bi X, Yang Y, Zhang W. Effects of molecule hydrophobicity and structural flexibility of appended bispecific antibody on Protein A chromatography. J Chromatogr A 2024; 1731:465206. [PMID: 39053253 DOI: 10.1016/j.chroma.2024.465206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Appended bispecific antibody (aBsAb) with two single chain variable fragments (scFv) linked at the c-terminus of its heavy chains is one of the promising formats in bispecific therapeutics. The presence of hydrophobic and flexible scFv fragments render aBsAb molecules higher molecule hydrophobicity and structural flexibility compared to monoclonal antibody (mAb), thus making its purification more challenging. We set out to investigate how the unique molecular properties of aBsAb affect its performance on Protein A chromatography. We showed that aBsAb has a high propensity for chromatography-induced aggregation due to its high molecule hydrophobicity, and this couldn't be improved by the addition of common chaotropic salts. Moreover, the presence of chaotropic salts, such as arginine hydrochloride (Arg-HCl), retarded aBsAb elution during Protein A chromatography rather than facilitating which was widely observed in mAb Protein A elution. Nevertheless, we were able to overcome the aggregation issue by optimizing elution condition and improved aBsAb purity from 29 % to 93 % in Protein A eluate with a high molecular weight (HMW) species of less than 5 %. We also showed that the high molecular flexibility of aBsAb leads to different hydrodynamic sizes of the aBsAb molecule post Protein A elution, neutralization, and re-acidification, which are pH dependent. This is different from mAbs where their sizes do not change post neutralization even with re-exposure to acid. The above unique observations of aBsAb in Protein A chromatography were clearly explained from the perspectives of its high molecular hydrophobicity and structural flexibility.
Collapse
Affiliation(s)
- Xinhui Wang
- Downstream Processing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Nattha Ingavat
- Downstream Processing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Jia Min Liew
- Downstream Processing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Nuruljannah Dzulkiflie
- Downstream Processing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Han Ping Loh
- Cell Line Development, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Yee Jiun Kok
- Analytical Science & Technology (Protein Analytics), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Xuezhi Bi
- Analytical Science & Technology (Protein Analytics), Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Yuansheng Yang
- Cell Line Development, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Wei Zhang
- Downstream Processing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore.
| |
Collapse
|
5
|
Choi J, Chen W, Liao H, Li X, Shendure J. A molecular proximity sensor based on an engineered, dual-component guide RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.14.553235. [PMID: 37645782 PMCID: PMC10461971 DOI: 10.1101/2023.08.14.553235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
One of the goals of synthetic biology is to enable the design of arbitrary molecular circuits with programmable inputs and outputs. Such circuits bridge the properties of electronic and natural circuits, processing information in a predictable manner within living cells. Genome editing is a potentially powerful component of synthetic molecular circuits, whether for modulating the expression of a target gene or for stably recording information to genomic DNA. However, programming molecular events such as protein-protein interactions or induced proximity as triggers for genome editing remains challenging. Here we demonstrate a strategy termed "P3 editing", which links protein-protein proximity to the formation of a functional CRISPR-Cas9 dual-component guide RNA. By engineering the crRNA:tracrRNA interaction, we demonstrate that various known protein-protein interactions, as well as the chemically-induced dimerization of protein domains, can be used to activate prime editing or base editing in human cells. Additionally, we explore how P3 editing can incorporate outputs from ADAR-based RNA sensors, potentially allowing specific RNAs to induce specific genome edits within a larger circuit. Our strategy enhances the controllability of CRISPR-based genome editing, facilitating its use in synthetic molecular circuits deployed in living cells.
Collapse
Affiliation(s)
- Junhong Choi
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wei Chen
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Hanna Liao
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
- Seattle Hub for Synthetic Biology, Seattle, WA 98195, USA
| | - Xiaoyi Li
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Seattle Hub for Synthetic Biology, Seattle, WA 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA 98195, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA 98195, USA
- Seattle Hub for Synthetic Biology, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Tadokoro T, Tsuboi H, Nakamura K, Hayakawa T, Ohmura R, Kato I, Inoue M, Tsunoda S, Niizuma S, Okada Y, Otsuguro S, Maenaka K. Thermostability and binding properties of single-chained Fv fragments derived from therapeutic antibodies. Protein Sci 2024; 33:e5084. [PMID: 38923711 PMCID: PMC11201803 DOI: 10.1002/pro.5084] [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: 02/10/2024] [Revised: 05/15/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
Small antibody fragments have recently been used as alternatives to full-length monoclonal antibodies in therapeutic applications. One of the most popular fragment antibodies is single-chain fragment variables (scFvs), consisting of variable heavy (VH) and variable light (VL) domains linked by a flexible peptide linker. scFvs have small molecular sizes, which enables good tissue penetration and low immunogenicity. Despite these advantages, the use of scFvs, especially for therapeutic purpose, is still limited because of the difficulty to regulate the binding activity and conformational stability. In this study, we constructed and analyzed 10 scFv fragments derived from 10 representatives of FDA-approved mAbs to evaluate their physicochemical properties. Differential scanning calorimetry analysis showed that scFvs exhibited relatively high but varied thermostability, from 50 to 70°C of melting temperatures, and different unfolding cooperativity. Surface plasmon resonance analysis revealed that scFvs fragments that exhibit high stability and cooperative unfolding likely tend to maintain antigen binding. This study demonstrated the comprehensive physicochemical properties of scFvs derived from FDA-approved antibodies, providing insights into antibody design and development.
Collapse
Affiliation(s)
- Takashi Tadokoro
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Harumi Tsuboi
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Kota Nakamura
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Tetsushi Hayakawa
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Reo Ohmura
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Izumi Kato
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Masaki Inoue
- Laboratory of Cellular and Molecular Physiology, The Faculty of Pharmaceutical SciencesKobe Gakuin UniversityKobeJapan
| | - Shin‐ichi Tsunoda
- Laboratory of Cellular and Molecular Physiology, The Faculty of Pharmaceutical SciencesKobe Gakuin UniversityKobeJapan
| | - Sayaka Niizuma
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Yukari Okada
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Satoko Otsuguro
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
| | - Katsumi Maenaka
- Laboratory of Biomolecular Science, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
- Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical SciencesHokkaido UniversitySapporoJapan
- Institute for Vaccine Research and Development (HU‐IVReD)Hokkaido UniversitySapporoJapan
- Global Station for Biosurfaces and Drug DiscoveryHokkaido UniversitySapporoJapan
| |
Collapse
|
7
|
Verhaar ER, van Keizerswaard WJC, Knoflook A, Balligand T, Ploegh HL. Nanobody-based CAR NK cells for possible immunotherapy of MICA + tumors. PNAS NEXUS 2024; 3:pgae184. [PMID: 38756234 PMCID: PMC11096969 DOI: 10.1093/pnasnexus/pgae184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
The glycoproteins MICA and MICB are upregulated on the surface of cells undergoing stress, for instance due to (viral) infection or malignant transformation. MICA/B are the ligands for the activating receptor NKG2D, found on cytotoxic immune cells like NK cells, CD8+ T cells, and γδ T cells. Upon engagement of NKG2D, these cells are activated to eradicate the MICA/B-positive targets, assisted by the secretion of cytokines. Nanobodies, or VHHs, are derived from the variable regions of camelid heavy-chain only immunoglobulins. Nanobodies are characterized by their small size, ease of production, stability, and specificity of recognition. We generated nanobodies that recognize membrane-bound MICA with high affinity. Here, we use these nanobodies as building blocks for a chimeric antigen receptor (CAR) to establish VHH-based CAR NK cells. These anti-MICA nanobody-based CAR NK cells recognize and selectively kill MICA-positive tumor cells in vitro and in vivo. We track localization of the VHH-based CAR NK cells to MICA-positive lung metastases by immuno-positron emission tomography imaging.
Collapse
Affiliation(s)
- Elisha R Verhaar
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | | | - Anouk Knoflook
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Balligand
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
8
|
Zhu B, Yin H, Zhang D, Zhang M, Chao X, Scimeca L, Wu MR. Synthetic biology approaches for improving the specificity and efficacy of cancer immunotherapy. Cell Mol Immunol 2024; 21:436-447. [PMID: 38605087 PMCID: PMC11061174 DOI: 10.1038/s41423-024-01153-x] [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: 09/10/2023] [Accepted: 03/03/2024] [Indexed: 04/13/2024] Open
Abstract
Immunotherapy has shown robust efficacy in treating a broad spectrum of hematological and solid cancers. Despite the transformative impact of immunotherapy on cancer treatment, several outstanding challenges remain. These challenges include on-target off-tumor toxicity, systemic toxicity, and the complexity of achieving potent and sustainable therapeutic efficacy. Synthetic biology has emerged as a promising approach to overcome these obstacles, offering innovative tools for engineering living cells with customized functions. This review provides an overview of the current landscape and future prospects of cancer immunotherapy, particularly emphasizing the role of synthetic biology in augmenting its specificity, controllability, and efficacy. We delineate and discuss two principal synthetic biology strategies: those targeting tumor surface antigens with engineered immune cells and those detecting intratumoral disease signatures with engineered gene circuits. This review concludes with a forward-looking perspective on the enduring challenges in cancer immunotherapy and the potential breakthroughs that synthetic biology may contribute to the field.
Collapse
Affiliation(s)
- Bo Zhu
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Hang Yin
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Di Zhang
- Drug Safety Research & Evaluation, Takeda Pharmaceuticals International Company, Cambridge, MA, 02139, USA
| | - Meiling Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Xiaojuan Chao
- Department of Liver Surgery, Center of Hepato-Pancreato-Biliary Surgery, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Luca Scimeca
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ming-Ru Wu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
9
|
Mii Y. Understanding and manipulating extracellular behaviors of Wnt ligands. In Vitro Cell Dev Biol Anim 2024; 60:441-448. [PMID: 38379096 DOI: 10.1007/s11626-024-00856-9] [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: 11/13/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024]
Abstract
Wnt, a family of secreted signaling proteins, serves diverse functions in embryogenesis, organogenesis, cancer, and stem cell functions. In the context of development, Wnt has been considered a representative morphogen, forming concentration gradients to give positional information to cells or tissues. However, although gradients are often illustrated in schemata, the reality of concentration gradients, or in other words, actual spatial distribution of Wnt ligands, and their behaviors in the extracellular space still remain poorly known. To understand extracellular behavior of Wnt ligands, quantitative analyses such as fluorescence correlation spectroscopy (FCS) and fluorescence recovery after photobleaching (FRAP) are highly informative because Wnt dispersal involves physical and biochemical processes, such as diffusion and binding to or dissociation from cell surface molecules, including heparan sulfate proteoglycans (HSPGs). Here, I briefly discuss representative methods to quantify morphogen dynamics. In addition, I discuss molecular manipulations of morphogens, mainly focusing on use of protein binders, and synthetic biology of morphogens as indicators of current and future directions in this field.
Collapse
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology (NIBB) and Exploratory Research Center On Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi, 444-8787, Japan.
- The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Aichi, 444-8787, Japan.
| |
Collapse
|
10
|
Zhu L, Man CW, Harrison RE, Wu Z, Limsakul P, Peng Q, Hashimoto M, Mamaril AP, Xu H, Liu L, Wang Y. Engineering a Programmed Death-Ligand 1-Targeting Monobody Via Directed Evolution for SynNotch-Gated Cell Therapy. ACS NANO 2024; 18:8531-8545. [PMID: 38456901 PMCID: PMC10958600 DOI: 10.1021/acsnano.4c01597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Programmed death-ligand 1 (PD-L1) is a promising target for cancer immunotherapy due to its ability to inhibit T cell activation; however, its expression on various noncancer cells may cause on-target off-tumor toxicity when designing PD-L1-targeting Chimeric Antigen Receptor (CAR) T cell therapies. Combining rational design and directed evolution of the human fibronectin-derived monobody scaffold, "PDbody" was engineered to bind to PD-L1 with a preference for a slightly lower pH, which is typical in the tumor microenvironment. PDbody was further utilized as a CAR to target the PD-L1-expressing triple negative MDA-MB-231 breast cancer cell line. To mitigate on-target off-tumor toxicity associated with targeting PD-L1, a Cluster of Differentiation 19 (CD19)-recognizing SynNotch IF THEN gate was integrated into the system. This CD19-SynNotch PDbody-CAR system was then expressed in primary human T cells to target CD19-expressing MDA-MB-231 cancer cells. These CD19-SynNotch PDbody-CAR T cells demonstrated both specificity and efficacy in vitro, accurately eradicating cancer targets in cytotoxicity assays. Moreover, in an in vivo bilateral murine tumor model, they exhibited the capability to effectively restrain tumor growth. Overall, CD19-SynNotch PDbody-CAR T cells represent a distinct development over previously published designs due to their increased efficacy, proliferative capability, and mitigation of off-tumor toxicity for solid tumor treatment.
Collapse
Affiliation(s)
- Linshan Zhu
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Alfred
E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Chi-Wei Man
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California, 92093 United States
| | - Reed E.S. Harrison
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Zhuohang Wu
- Alfred
E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Praopim Limsakul
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Division
of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand
- Center of
Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand
| | - Qin Peng
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Institute
of Systems and Physical Biology, Shenzhen
Bay Laboratory, Shenzhen 518132, P.R. China
| | - Matthew Hashimoto
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Anthony P. Mamaril
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Hongquan Xu
- Department
of Statistics, University of California, Los Angeles, California 90095, United States
| | - Longwei Liu
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Alfred
E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Yingxiao Wang
- Department
of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Alfred
E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
11
|
Madsen AV, Pedersen LE, Kristensen P, Goletz S. Design and engineering of bispecific antibodies: insights and practical considerations. Front Bioeng Biotechnol 2024; 12:1352014. [PMID: 38333084 PMCID: PMC10850309 DOI: 10.3389/fbioe.2024.1352014] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Bispecific antibodies (bsAbs) have attracted significant attention due to their dual binding activity, which permits simultaneous targeting of antigens and synergistic binding effects beyond what can be obtained even with combinations of conventional monospecific antibodies. Despite the tremendous therapeutic potential, the design and construction of bsAbs are often hampered by practical issues arising from the increased structural complexity as compared to conventional monospecific antibodies. The issues are diverse in nature, spanning from decreased biophysical stability from fusion of exogenous antigen-binding domains to antibody chain mispairing leading to formation of antibody-related impurities that are very difficult to remove. The added complexity requires judicious design considerations as well as extensive molecular engineering to ensure formation of high quality bsAbs with the intended mode of action and favorable drug-like qualities. In this review, we highlight and summarize some of the key considerations in design of bsAbs as well as state-of-the-art engineering principles that can be applied in efficient construction of bsAbs with diverse molecular formats.
Collapse
Affiliation(s)
- Andreas V. Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lasse E. Pedersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
12
|
Zhou J, Shi F, Luo X, Lei B, Shi Z, Huang C, Zhang Y, Li X, Wang H, Li XY, He X. The persistence and antitumor efficacy of CAR-T cells are modulated by tonic signaling within the CDR. Int Immunopharmacol 2024; 126:111239. [PMID: 37979453 DOI: 10.1016/j.intimp.2023.111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated remarkable clinical efficacy, but challenges related to relapse and CAR-T cell exhaustion persist. One contributing factor to this exhaustion is CAR tonic signaling, where CAR-T cells self-activate without antigen stimulation, leading to reduced persistence and impaired antitumor activity. To address this issue, we conducted a preclinical study evaluating tonic signaling using nanobody-derived CAR-T cells. Our investigation revealed that specific characteristics of the complementary determining regions (CDRs), including low solubility, polarity, positive charge, energy, and area of ionic and positive CDR patches of amino acids, were associated with low antigen-independent tonic signaling. Significantly, we observed that stronger tonic signaling directly impacted CAR-T cell proliferation in vitro, consequently leading to CAR-T cell exhaustion and diminished persistence and effectiveness in vivo. Our findings provide compelling preclinical evidence and lay the foundation for the clinical assessment of CAR-T cells with distinct tonic signaling patterns. Understanding the role of CDRs in modulating tonic signaling holds promise for advancing the development of more efficient and durable CAR-T cell therapies, thereby enhancing the treatment of cancer and addressing the challenges of relapse in CAR-T cell therapy.
Collapse
Affiliation(s)
- Jincai Zhou
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| | - Feifei Shi
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xinran Luo
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Bixia Lei
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Zhongjun Shi
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Chenyu Huang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Yuting Zhang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xiaopei Li
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Huajing Wang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xian-Yang Li
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| | - Xiaowen He
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
13
|
Starkie DO, Arber C, Baker T, Lightwood DJ, Wray S. Antibody-mediated degradation of 4R-tau restores mitochondrial membrane polarization in human induced pluripotent stem cell-derived neurons with the MAPT 10+16 mutation. MAbs 2024; 16:2436102. [PMID: 39665388 PMCID: PMC11790244 DOI: 10.1080/19420862.2024.2436102] [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: 09/27/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Microtubule-associated protein tau is inextricably linked to a group of clinically diverse neurodegenerative diseases termed tauopathies. The ratio balance of the major tau splicing isoform groups (3 R- and 4 R-tau) is critical in maintaining healthy neurons. An imbalance causing excess 4 R tau is associated with diseases such as progressive supranuclear palsy and frontotemporal dementia. The mechanisms by which increased 4 R results in neuronal dysfunction and neurodegeneration are not fully understood, and progress has been limited partly by a lack of suitable tools to investigate tau isoform imbalance. This work generated novel 3 R- and 4 R-specific antibody tools and 4 R-tau degrading intracellular antibody fragment "degrabodies". These were used to probe the molecular mechanisms of excess 4 R-tau in disease-mutant induced pluripotent stem cell-derived neurons. For the first time, we demonstrate a causative link between excess 4 R-tau and mitochondrial membrane hyperpolarization with wide-ranging potential for elucidating novel therapeutic approaches to treat neurodegenerative disease.
Collapse
Affiliation(s)
- Dale O. Starkie
- Antibody Discovery and Optimization, UCB Pharma, Slough, Berkshire, UK
| | - Charles Arber
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Terry Baker
- Antibody Discovery and Optimization, UCB Pharma, Slough, Berkshire, UK
| | | | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
14
|
Fawcett C, Tickle JR, Coles CH. Facilitating high throughput bispecific antibody production and potential applications within biopharmaceutical discovery workflows. MAbs 2024; 16:2311992. [PMID: 39674918 DOI: 10.1080/19420862.2024.2311992] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 01/25/2024] [Indexed: 12/17/2024] Open
Abstract
A major driver for the recent investment surge in bispecific antibody (bsAb) platforms and products is the multitude of distinct mechanisms of action that bsAbs offer compared to a combination of two monoclonal antibodies. Four bsAb products were granted first regulatory approvals in the US or EU during 2023 and the biopharmaceutical industry pipeline is brimming with bsAb candidates across a broad range of therapeutic applications. In previously reported bsAb discovery campaigns, following a hypothesis-based choice of two specific target proteins, selections and screening activities have often been performed in mono-specific formats. The conversion to bispecific modalities has usually been positioned toward the end of the discovery process and has involved small numbers of lead molecules, largely due to challenges in expressing, purifying, and analyzing large numbers of bsAbs. In this review, we discuss emerging strategies to facilitate the production of expanded bsAb panels, focusing particularly upon combinatorial methods to generate bsAb matrices. Such technologies will enable screening in. bispecific formats at earlier stages of discovery campaigns, not only widening the accessible protein space to maximize chances of success, but also advancing empirical bi-target validation activities to assess initial target selection hypotheses.
Collapse
Affiliation(s)
- Caitlin Fawcett
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, UK
| | - Joseph R Tickle
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| | - Charlotte H Coles
- Large Molecule Discovery, GSK, GSK Medicines Research Centre, Stevenage, UK
| |
Collapse
|
15
|
Qin Y, Jin J, Zhang J, Wang H, Liu L, Zhang Y, Ling S, Hu J, Li N, Wang J, Lv C, Yang X. A fully human monoclonal antibody targeting Semaphorin 5A alleviates the progression of rheumatoid arthritis. Biomed Pharmacother 2023; 168:115666. [PMID: 37832409 DOI: 10.1016/j.biopha.2023.115666] [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: 05/19/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune disease worldwide. Although progress has been made in RA treatment in recent decades, remission cannot be effectively achieved for a considerable proportion of RA patients. Thus, novel potential targets for therapeutic strategies are needed. Semaphorin 5A (SEMA5A) plays a pivotal role in RA progression by facilitating pannus formation, and it is a promising therapeutic target. In this study, we sought to develop an antibody treatment strategy targeting SEMA5A and evaluate its therapeutic effect using a collagen-induced arthritis (CIA) model. We generated SYD12-12, a fully human SEMA5A blocking antibody, through phage display technology. SYD12-12 intervention effectively inhibited angiogenesis and aggressive phenotypes of RA synoviocytes in vitro and dose-dependently inhibited synovial hyperplasia, pannus formation, bone destruction in CIA mice. Notably, SYD12-12 also improved the Treg/Th17 imbalance in CIA mice. We confirmed through immunofluorescence and molecular docking that SYD12-12 integrated with the unique TSP-1 domain of SEMA5A. In conclusion, we developed and characterized a fully human SEMA5A-blocking antibody for the first time. SYD12-12 effectively alleviated disease progression in CIA mice by inhibiting pannus formation and improving the Treg/Th17 imbalance, demonstrating its potential for the RA treatment.
Collapse
Affiliation(s)
- Yang Qin
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Jin
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiani Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanwen Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunwang Ling
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinzhu Hu
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Nuan Li
- Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianguang Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Chen Lv
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
16
|
Ding Z, Sun S, Wang X, Yang X, Shi W, Huang X, Xie S, Mo F, Hou X, Liu A, Jiang X, Tang Z, Lu X. Nanobody-based trispecific T cell engager (Nb-TriTE) enhances therapeutic efficacy by overcoming tumor-mediated immunosuppression. J Hematol Oncol 2023; 16:115. [PMID: 38031188 PMCID: PMC10688028 DOI: 10.1186/s13045-023-01507-4] [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: 09/06/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND T cell engagers (TCEs) have been established as an emerging modality for hematologic malignancies, but solid tumors remain refractory. However, the upregulation of programmed cell death 1 (PD-1) is correlated with T cell dysfunction that confer tumor-mediated immunosuppression. Developing a novel nanobody-based trispecific T cell engager (Nb-TriTE) would be a potential strategy to improve therapeutic efficacy. METHODS Given the therapeutic potential of nanobodies (Nbs), we first screened Nb targeting fibroblast activation protein (FAP) and successfully generated a Nb-based bispecific T cell engager (Nb-BiTE) targeting FAP. Then, we developed a Nb-TriTE by fusing an anti-PD-1 Nb to the Nb-BiTE. The biological activity and antitumor efficacy of the Nb-TriTE were evaluated in vitro and in both cell line-derived and patient-derived xenograft mouse models. RESULTS We had for the first time successfully selected a FAP Nb for the generation of novel Nb-BiTE and Nb-TriTE, which showed good binding ability to their targets. Nb-TriTE not only induced robust tumor antigen-specific killing, potent T cell activation and enhanced T cell function in vitro, but also suppressed tumor growth, improved survival and mediated more T cell infiltration than Nb-BiTE in mouse models of different solid tumors without toxicity. CONCLUSIONS This novel Nb-TriTE provides a promising and universal platform to overcome tumor-mediated immunosuppression and improve patient outcomes in the future.
Collapse
Affiliation(s)
- Ziqiang Ding
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Shuyang Sun
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaomei Yang
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Wei Shi
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xianing Huang
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Shenxia Xie
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Fengzhen Mo
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoqiong Hou
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Aiqun Liu
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhuoran Tang
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China.
| | - Xiaoling Lu
- School of Basic Medical Sciences/College of Stomatology/Hospital of Stomatology/Guangxi Key Laboratory of Nanobody Research/Guangxi Nanobody Engineering Research Center/Laboratory Animal Center/Pharmaceutical College/Affiliated Tumor Hospital, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
17
|
Bakavayev S, Stavsky A, Argueti-Ostrovsky S, Yehezkel G, Fridmann-Sirkis Y, Barak Z, Gitler D, Israelson A, Engel S. Blocking an epitope of misfolded SOD1 ameliorates disease phenotype in a model of amyotrophic lateral sclerosis. Brain 2023; 146:4594-4607. [PMID: 37394908 DOI: 10.1093/brain/awad222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/01/2023] [Accepted: 06/11/2023] [Indexed: 07/04/2023] Open
Abstract
The current strategies to mitigate the toxicity of misfolded superoxide dismutase 1 (SOD1) in familial amyotrophic lateral sclerosis via blocking SOD1 expression in the CNS are indiscriminative for misfolded and intact proteins, and as such, entail a risk of depriving CNS cells of their essential antioxidant potential. As an alternative approach to neutralize misfolded and spare unaffected SOD1 species, we developed scFv-SE21 antibody that blocks the β6/β7 loop epitope exposed exclusively in misfolded SOD1. The β6/β7 loop epitope has previously been proposed to initiate amyloid-like aggregation of misfolded SOD1 and mediate its prion-like activity. The adeno-associated virus-mediated expression of scFv-SE21 in the CNS of hSOD1G37R mice rescued spinal motor neurons, reduced the accumulation of misfolded SOD1, decreased gliosis and thus delayed disease onset and extended survival by 90 days. The results provide evidence for the role of the exposed β6/β7 loop epitope in the mechanism of neurotoxic gain-of-function of misfolded SOD1 and open avenues for the development of mechanism-based anti-SOD1 therapeutics, whose selective targeting of misfolded SOD1 species may entail a reduced risk of collateral oxidative damage to the CNS.
Collapse
Affiliation(s)
- Shamchal Bakavayev
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Alexandra Stavsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Shirel Argueti-Ostrovsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Galit Yehezkel
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yael Fridmann-Sirkis
- Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zeev Barak
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Daniel Gitler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Stanislav Engel
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
- The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
18
|
Smith R. Bringing cell therapy to tumors: considerations for optimal CAR binder design. Antib Ther 2023; 6:225-239. [PMID: 37846297 PMCID: PMC10576856 DOI: 10.1093/abt/tbad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells have revolutionized the immunotherapy of B-cell malignancies and are poised to expand the range of their impact across a broad range of oncology and non-oncology indications. Critical to the success of a given CAR is the choice of binding domain, as this is the key driver for specificity and plays an important role (along with the rest of the CAR structure) in determining efficacy, potency and durability of the cell therapy. While antibodies have proven to be effective sources of CAR binding domains, it has become apparent that the desired attributes for a CAR binding domain do differ from those of a recombinant antibody. This review will address key factors that need to be considered in choosing the optimal binding domain for a given CAR and how binder properties influence and are influenced by the rest of the CAR.
Collapse
Affiliation(s)
- Richard Smith
- Department of Research, Kite, a Gilead Company, 5858 Horton Street, Suite 240, Emeryville, CA 94070, USA
| |
Collapse
|
19
|
Qian L, Lin X, Gao X, Khan RU, Liao JY, Du S, Ge J, Zeng S, Yao SQ. The Dawn of a New Era: Targeting the "Undruggables" with Antibody-Based Therapeutics. Chem Rev 2023. [PMID: 37186942 DOI: 10.1021/acs.chemrev.2c00915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The high selectivity and affinity of antibodies toward their antigens have made them a highly valuable tool in disease therapy, diagnosis, and basic research. A plethora of chemical and genetic approaches have been devised to make antibodies accessible to more "undruggable" targets and equipped with new functions of illustrating or regulating biological processes more precisely. In this Review, in addition to introducing how naked antibodies and various antibody conjugates (such as antibody-drug conjugates, antibody-oligonucleotide conjugates, antibody-enzyme conjugates, etc.) work in therapeutic applications, special attention has been paid to how chemistry tools have helped to optimize the therapeutic outcome (i.e., with enhanced efficacy and reduced side effects) or facilitate the multifunctionalization of antibodies, with a focus on emerging fields such as targeted protein degradation, real-time live-cell imaging, catalytic labeling or decaging with spatiotemporal control as well as the engagement of antibodies inside cells. With advances in modern chemistry and biotechnology, well-designed antibodies and their derivatives via size miniaturization or multifunctionalization together with efficient delivery systems have emerged, which have gradually improved our understanding of important biological processes and paved the way to pursue novel targets for potential treatments of various diseases.
Collapse
Affiliation(s)
- Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xue Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544
| |
Collapse
|
20
|
Gonzalez-Garcia P, Muñoz-Miranda JP, Fernandez-Cisnal R, Olvera L, Moares N, Gabucio A, Fernandez-Ponce C, Garcia-Cozar F. Specific Activation of T Cells by an ACE2-Based CAR-Like Receptor upon Recognition of SARS-CoV-2 Spike Protein. Int J Mol Sci 2023; 24:ijms24087641. [PMID: 37108807 PMCID: PMC10145580 DOI: 10.3390/ijms24087641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causative agent of the Coronavirus Disease 2019 (COVID-19) pandemic, which is still a health issue worldwide mostly due to a high rate of contagiousness conferred by the high-affinity binding between cell viral receptors, Angiotensin-Converting Enzyme 2 (ACE2) and SARS-CoV-2 Spike protein. Therapies have been developed that rely on the use of antibodies or the induction of their production (vaccination), but despite vaccination being still largely protective, the efficacy of antibody-based therapies wanes with the advent of new viral variants. Chimeric Antigen Receptor (CAR) therapy has shown promise for tumors and has also been proposed for COVID-19 treatment, but as recognition of CARs still relies on antibody-derived sequences, they will still be hampered by the high evasion capacity of the virus. In this manuscript, we show the results from CAR-like constructs with a recognition domain based on the ACE2 viral receptor, whose ability to bind the virus will not wane, as Spike/ACE2 interaction is pivotal for viral entry. Moreover, we have developed a CAR construct based on an affinity-optimized ACE2 and showed that both wild-type and affinity-optimized ACE2 CARs drive activation of a T cell line in response to SARS-CoV-2 Spike protein expressed on a pulmonary cell line. Our work sets the stage for the development of CAR-like constructs against infectious agents that would not be affected by viral escape mutations and could be developed as soon as the receptor is identified.
Collapse
Affiliation(s)
| | - Juan P Muñoz-Miranda
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | | | - Lucia Olvera
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Noelia Moares
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Antonio Gabucio
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Cecilia Fernandez-Ponce
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Francisco Garcia-Cozar
- Institute of Biomedical Research Cadiz (INIBICA), 11009 Cadiz, Spain
- Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| |
Collapse
|
21
|
Zekri L, Lutz M, Prakash N, Manz T, Klimovich B, Mueller S, Hoerner S, Hagelstein I, Engel M, Chashchina A, Pfluegler M, Heitmann JS, Jung G, Salih HR. An optimized IgG-based B7-H3xCD3 bispecific antibody for treatment of gastrointestinal cancers. Mol Ther 2023; 31:1033-1045. [PMID: 36793213 PMCID: PMC10124076 DOI: 10.1016/j.ymthe.2023.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
T cell-based immunotherapy has revolutionized oncological treatment. However, many patients do not respond to treatment, and long-term remissions remain rare, particularly in gastrointestinal cancers like colorectal cancer (CRC). B7-H3 is overexpressed in multiple cancer entities including CRC on both tumor cells and tumor vasculature, the latter facilitating influx of effector cells into the tumor site upon therapeutic targeting. We generated a panel of T cell-recruiting B7-H3xCD3 bispecific antibodies (bsAbs) and show that targeting a membrane-proximal B7-H3 epitope allows for a 100-fold reduction of CD3 affinity. In vitro, our lead compound CC-3 showed superior tumor cell killing, T cell activation, proliferation, and memory formation, whereas undesired cytokine release was reduced. In vivo, CC-3 mediated potent antitumor activity in three independent models using immunocompromised mice adoptively transferred with human effector cells with regard to prevention of lung metastasis and flank tumor growth as well as elimination of large established tumors. Thus, fine-tuning of both target and CD3 affinities as well as binding epitopes allowed for the generation of a B7-H3xCD3 bsAbs with promising therapeutic activity. CC-3 is presently undergoing good manufacturing practice (GMP) production to enable evaluation in a clinical "first-in-human" study in CRC.
Collapse
Affiliation(s)
- Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Martina Lutz
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Nisha Prakash
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Timo Manz
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Stefanie Mueller
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Sebastian Hoerner
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Monika Engel
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Anna Chashchina
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Martin Pfluegler
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Jonas S Heitmann
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Gundram Jung
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; Department of Immunology, Institute for Cell Biology, Eberhard Karls University, Tuebingen, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tuebingen, Tuebingen 72076, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen 72076, Germany; DKFZ Partner Site Tuebingen, German Cancer Consortium (DKTK), Tuebingen 72076, Germany.
| |
Collapse
|
22
|
Qin Y, Wu G, Jin J, Wang H, Zhang J, Liu L, Zhao H, Wang J, Yang X. A fully human connective tissue growth factor blocking monoclonal antibody ameliorates experimental rheumatoid arthritis through inhibiting angiogenesis. BMC Biotechnol 2023; 23:6. [PMID: 36869335 PMCID: PMC9985226 DOI: 10.1186/s12896-023-00776-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Connective tissue growth factor (CTGF) plays a pivotal role in the pathogenesis of rheumatoid arthritis (RA) by facilitating angiogenesis and is a promising therapeutic target for RA treatment. Herein, we generated a fully human CTGF blocking monoclonal antibody (mAb) through phage display technology. RESULTS A single-chain fragment variable (scFv) with a high affinity to human CTGF was isolated through screening a fully human phage display library. We carried out affinity maturation to elevate its affinity for CTGF and reconstructed it into a full-length IgG1 format for further optimization. Surface plasmon resonance (SPR) data showed that full-length antibody IgG mut-B2 bound to CTGF with a dissociation constant (KD) as low as 0.782 nM. In the collagen-induced arthritis (CIA) mice, IgG mut-B2 alleviated arthritis and decreased the level of pro-inflammatory cytokines in a dose-dependent manner. Furthermore, we confirmed that the TSP-1 domain of CTGF is essential for the interaction. Additionally, the results of Transwell assays, tube formation experiments, and chorioallantoic membrane (CAM) assays showed that IgG mut-B2 could effectively inhibit angiogenesis. CONCLUSION The fully human mAb that antagonizes CTGF could effectively alleviate arthritis in CIA mice, and its mechanism is tightly associated with the TSP-1 domain of CTGF.
Collapse
Affiliation(s)
- Yang Qin
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Gan Wu
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China.,Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying, Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Jin
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Hao Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Jiani Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Li Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Heping Zhao
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Jianguang Wang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, China. .,Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying, Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
23
|
Farokhi-Fard A, Bayat E, Beig Parikhani A, Komijani S, Aghamirza Moghim Aliabadi H, Sardari S, Gharib B, Barkhordari F, Azadmanesh K, Karimipoor M, Bakhshandeh H, Davami F. Bacterial production and biophysical characterization of a hard-to-fold scFv against myeloid leukemia cell surface marker, IL-1RAP. Mol Biol Rep 2023; 50:1191-1202. [PMID: 36435922 DOI: 10.1007/s11033-022-07972-3] [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: 06/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Interleukin-1 receptor accessory protein (IL-1RAP) is one of the most promising therapeutic targets proposed for myeloid leukemia. Antibodies (Abs) specific to IL-1RAP could be valuable tools for targeted therapy of this lethal malignancy. This study is about the preparation of a difficult-to-produce single-chain variable fragment (scFv) construct against the membrane-bound isoform of human IL-1RAP using Escherichia coli (E. coli). METHODS Different approaches were examined for refolding and characterization of the scFv. Binding activities of antibody fragments were comparatively evaluated using cell-based enzyme-linked immunosorbent assay (ELISA). Homogeneity and secondary structure of selected scFv preparation were analyzed using analytical size exclusion chromatography (SEC) and circular dichroism (CD) spectroscopy, respectively. The activity of the selected preparation was evaluated after long-term storage, repeated freeze-thaw cycles, or following incubation with normal and leukemic serum. RESULTS Strategies for soluble expression of the scFv failed. Even with the help of Trx, ≥ 98% of proteins were expressed as inclusion bodies (IBs). Among three different refolding methods, the highest recovery rate was obtained from the dilution method (11.2%). Trx-tag substantially enhanced the expression level (18%, considering the molecular weight (MW) differences), recovery rate (˃1.6-fold), and binding activity (˃2.6-fold increase in absorbance450nm). The produced scFv exhibited expected secondary structure as well as acceptable bio-functionality, homogeneity, and stability. CONCLUSION We were able to produce 21 mg/L culture functional and stable anti-IL-1RAP scFv via recovering IBs by pulse dilution procedure. The produced scFv as a useful targeting agent could be used in scheming new therapeutics or diagnostics for myeloid malignancies.
Collapse
Affiliation(s)
- Aref Farokhi-Fard
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Elham Bayat
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran
| | - Arezoo Beig Parikhani
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran
| | - Samira Komijani
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran
| | - Hooman Aghamirza Moghim Aliabadi
- Protein Chemistry Laboratory, Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran, Tehran, Iran
- Advance Chemical Studies Laboratory, Faculty of Chemistry, K.N. Toosi University, Tehran, Iran
| | - Soroush Sardari
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farzaneh Barkhordari
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran
| | | | - Morteza Karimipoor
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Haleh Bakhshandeh
- Department of Nanobiotechnology, New Technology Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Davami
- Medical Biotechnology Department, Biotechnology research center, Pasteur Institute of Iran (IPI), No. 69, Pasteur Ave, Tehran, Iran.
| |
Collapse
|
24
|
Shin J, Raissi S, Phelan P, Bullock PA. Rational design of a Nivolumab-based ANTI-PD-1 single chain variable fragment that blocks the interaction between PD-1 expressed on T-CELLS and PD-L1 ON CHO cells. Protein Expr Purif 2023; 202:106196. [PMID: 36280166 DOI: 10.1016/j.pep.2022.106196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/16/2022] [Indexed: 11/05/2022]
Abstract
Antibodies that block the interaction between PD-1 expressing T-cells and cancer cells expressing PD-L1 play a central role in contemporary immunotherapy regimes [1-3]. We previously reported the isolation of a single chain variable fragment (scFv) of the monoclonal anti-PD-1 antibody Nivolumab, that binds to purified PD-1 and blocked its interaction with PD-L1 [4]. This anti-PD-1 scFv did not, however, function in a cell-based assay designed to detect the disruption of the PD-1/PD-L1 interaction, a result likely due to its poor solubility in tissue culture media. Herein we report that following a series of structure-based rational design analyses, including Aggreescan3D, we have isolated a variant of the anti-PD-1 scFv having significantly improved solubility in tissue culture medium. Moreover, this soluble anti-PD-1 scFv variant disrupted the interaction between PD-1 expressed on Jurkat Cells and PD-L1 expressed on CHO cells. These findings are discussed in terms of the related observation that the residues mutated to form the anti-PD-1 variant are conserved in many other scFvs; thus, the properties of a range of scFvs will likely be enhanced by similar mutations of the conserved residues.
Collapse
Affiliation(s)
- Jong Shin
- Department of Pathology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Siavash Raissi
- Department of Developmental, Molecular and Chemical Biology Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA
| | - Paul Phelan
- Joinn Biologics, 2600 Hilltop Drive, Building L, Richmond, CA, 94806, USA
| | - Peter A Bullock
- Department of Developmental, Molecular and Chemical Biology Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA.
| |
Collapse
|
25
|
Amatsu S, Matsumura T, Zuka M, Fujinaga Y. Molecular engineering of a minimal E-cadherin inhibitor protein derived from Clostridium botulinum hemagglutinin. J Biol Chem 2023; 299:102944. [PMID: 36707052 PMCID: PMC9958082 DOI: 10.1016/j.jbc.2023.102944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Hemagglutinin (HA), a nontoxic component of the botulinum neurotoxin (BoNT) complex, binds to E-cadherin and inhibits E-cadherin-mediated cell-cell adhesion. HA is a 470 kDa protein complex comprising six HA1, three HA2, and three HA3 subcomponents. Thus, to prepare recombinant full-length HA in vitro, it is necessary to reconstitute the macromolecular complex from purified HA subcomponents, which involves multiple purification steps. In this study, we developed NanoHA, a minimal E-cadherin inhibitor protein derived from Clostridium botulinum HA with a simple purification strategy needed for production. NanoHA, containing HA2 and a truncated mutant of HA3 (amino acids 380-626; termed as HA3mini), is a 47 kDa single polypeptide (one-tenth the molecular weight of full-length HA, 470 kDa) engineered with three types of modifications: (i) a short linker sequence between the C terminus of HA2 and N terminus of HA3; (ii) a chimeric complex composed of HA2 derived from the serotype C BoNT complex and HA3mini from the serotype B BoNT complex; and (iii) three amino acid substitutions from hydrophobic to hydrophilic residues on the protein surface. We demonstrated that NanoHA inhibits E-cadherin-mediated cell-cell adhesion of epithelial cells (e.g., Caco-2 and Madin-Darby canine kidney cells) and disrupts their epithelial barrier. Finally, unlike full-length HA, NanoHA can be transported from the basolateral side to adherens junctions via passive diffusion. Overall, these results indicate that the rational design of NanoHA provides a minimal E-cadherin inhibitor with a wide variety of applications as a lead molecule and for further molecular engineering.
Collapse
Affiliation(s)
- Sho Amatsu
- Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan; Department of Forensic Medicine and Pathology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| | - Takuhiro Matsumura
- Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Masahiko Zuka
- Department of Forensic Medicine and Pathology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Yukako Fujinaga
- Department of Bacteriology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
26
|
Madsen AV, Kristensen P, Buell AK, Goletz S. Generation of robust bispecific antibodies through fusion of single-domain antibodies on IgG scaffolds: a comprehensive comparison of formats. MAbs 2023; 15:2189432. [PMID: 36939220 PMCID: PMC10038023 DOI: 10.1080/19420862.2023.2189432] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Bispecific antibodies (bsAbs) enable dual binding of different antigens with potential synergistic targeting effects and innovative therapeutic possibilities. The formation of bsAbs is, however, often dependent on complex engineering strategies with a high risk of antibody chain mispairing leading to contamination of the final product with incorrectly assembled antibody species. This study demonstrates formation of bsAbs in a generic and conceptually easy manner through fusion of single-domain antibodies (sdAbs) onto IgG scaffolds through flexible 10 amino acid linkers to form high-quality bsAbs with both binding functionalities intact and minimal product-related impurities. SdAbs are attractive fusion partners due to their small and monomeric nature combined with antigen-binding capabilities comparable to conventional human antibodies. By systematically comparing a comprehensive panel of symmetric αPD-L1×αHER2 antibodies, including reversely mirrored antigen specificities, we investigate how the molecular geometry affects production, stability, antigen binding and CD16a binding. SdAb fusion of the heavy chain was generally preferred over light chain fusion for promoting good expression and high biophysical stability as well as maintaining efficient binding to both antigens. We find that N-terminal sdAb fusion might sterically hinder antigen-binding to the Fv region of the IgG scaffold, whereas C-terminal fusion might disturb antigen-binding to the fused sdAb. Our work demonstrates a toolbox of complementary methods for in-depth analysis of key features, such as in-solution dual antigen binding, thermal stability, and aggregation propensity, to ensure high bsAb quality. These techniques can be executed at high-throughput and/or with very low material consumption and thus represent valuable tools for bsAb screening and development.
Collapse
Affiliation(s)
- Andreas V Madsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Peter Kristensen
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Alexander K Buell
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
27
|
Strubel A, Münick P, Chaikuad A, Dreier B, Schaefer J, Gebel J, Osterburg C, Tuppi M, Schäfer B, Knapp S, Plückthun A, Dötsch V. Designed Ankyrin Repeat Proteins as a tool box for analyzing p63. Cell Death Differ 2022; 29:2445-2458. [PMID: 35717504 PMCID: PMC9751120 DOI: 10.1038/s41418-022-01030-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 01/31/2023] Open
Abstract
The function of the p53 transcription factor family is dependent on several folded domains. In addition to a DNA-binding domain, members of this family contain an oligomerization domain. p63 and p73 also contain a C-terminal Sterile α-motif domain. Inhibition of most transcription factors is difficult as most of them lack deep pockets that can be targeted by small organic molecules. Genetic knock-out procedures are powerful in identifying the overall function of a protein, but they do not easily allow one to investigate roles of individual domains. Here we describe the characterization of Designed Ankyrin Repeat Proteins (DARPins) that were selected as tight binders against all folded domains of p63. We determine binding affinities as well as specificities within the p53 protein family and show that DARPins can be used as intracellular inhibitors for the modulation of transcriptional activity. By selectively inhibiting DNA binding of the ΔNp63α isoform that competes with p53 for the same promoter sites, we show that p53 can be reactivated. We further show that inhibiting the DNA binding activity stabilizes p63, thus providing evidence for a transcriptionally regulated negative feedback loop. Furthermore, the ability of DARPins to bind to the DNA-binding domain and the Sterile α-motif domain within the dimeric-only and DNA-binding incompetent conformation of TAp63α suggests a high structural plasticity within this special conformation. In addition, the developed DARPins can also be used to specifically detect p63 in cell culture and in primary tissue and thus constitute a very versatile research tool for studying the function of p63.
Collapse
Affiliation(s)
- Alexander Strubel
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Philipp Münick
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Apirat Chaikuad
- grid.7839.50000 0004 1936 9721Institute of Pharmaceutical Chemistry, Goethe University, 60438 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721Structural Genomics Consortium, Goethe University, 60438 Frankfurt, Germany
| | - Birgit Dreier
- grid.7400.30000 0004 1937 0650Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Jonas Schaefer
- grid.7400.30000 0004 1937 0650Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Jakob Gebel
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Christian Osterburg
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Marcel Tuppi
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Birgit Schäfer
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| | - Stefan Knapp
- grid.7839.50000 0004 1936 9721Institute of Pharmaceutical Chemistry, Goethe University, 60438 Frankfurt, Germany ,grid.7839.50000 0004 1936 9721Structural Genomics Consortium, Goethe University, 60438 Frankfurt, Germany
| | - Andreas Plückthun
- grid.7400.30000 0004 1937 0650Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Volker Dötsch
- grid.7839.50000 0004 1936 9721Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438 Frankfurt, Germany
| |
Collapse
|
28
|
Arvedson T, Bailis JM, Urbig T, Stevens JL. Considerations for design, manufacture, and delivery for effective and safe T-cell engager therapies. Curr Opin Biotechnol 2022; 78:102799. [PMID: 36179408 DOI: 10.1016/j.copbio.2022.102799] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 08/12/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022]
Abstract
T-cell engager (TCE) molecules provide a targeted immunotherapy approach to treat hematologic malignancies and solid tumors. Since the approval of the CD19-targeted BiTE® (bispecific T-cell engager) molecule blinatumomab, multiple TCE molecules against different targets have been developed in several tumor types, with the approval of three additional TCE molecules in 2022. Some of the initial challenges, such as the need for continuous intravenous administration and low productivity, have been addressed in subsequent iterations of the platform by advancing half-life extended, Fc-based molecules. As clinical data from these molecules emerge, additional optimization of formats and manufacturability will be necessary. Ongoing efforts are focused on further improving TCE efficacy, safety, and convenience of administration.
Collapse
Affiliation(s)
- Tara Arvedson
- Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Julie M Bailis
- Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
29
|
Hambach J, Mann AM, Bannas P, Koch-Nolte F. Targeting multiple myeloma with nanobody-based heavy chain antibodies, bispecific killer cell engagers, chimeric antigen receptors, and nanobody-displaying AAV vectors. Front Immunol 2022; 13:1005800. [PMID: 36405759 PMCID: PMC9668101 DOI: 10.3389/fimmu.2022.1005800] [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: 07/28/2022] [Accepted: 10/21/2022] [Indexed: 11/29/2022] Open
Abstract
Nanobodies are well suited for constructing biologics due to their high solubility. We generated nanobodies directed against CD38, a tumor marker that is overexpressed by multiple myeloma and other hematological malignancies. We then used these CD38-specific nanobodies to construct heavy chain antibodies, bispecific killer cell engagers (BiKEs), chimeric antigen receptor (CAR)-NK cells, and nanobody-displaying AAV vectors. Here we review the utility of these nanobody-based constructs to specifically and effectively target CD38-expressing myeloma cells. The promising results of our preclinical studies warrant further clinical studies to evaluate the potential of these CD38-specific nanobody-based constructs for treatment of multiple myeloma.
Collapse
Affiliation(s)
- Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Marei Mann
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Friedrich Koch-Nolte,
| |
Collapse
|
30
|
Muñoz-López P, Ribas-Aparicio RM, Becerra-Báez EI, Fraga-Pérez K, Flores-Martínez LF, Mateos-Chávez AA, Luria-Pérez R. Single-Chain Fragment Variable: Recent Progress in Cancer Diagnosis and Therapy. Cancers (Basel) 2022; 14:cancers14174206. [PMID: 36077739 PMCID: PMC9455005 DOI: 10.3390/cancers14174206] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Recombinant antibody fragments have shown remarkable potential as diagnostic and therapeutic tools in the fight against cancer. The single-chain fragment variable (scFv) that contains the complete antigen-binding domains of a whole antibody, has several advantages such as a high specificity and affinity for antigens, a low immunogenicity, and the proven ability to penetrate tumor tissues and diffuse. This review provides an overview of the current studies on the principle, generation, and applications of scFvs, particularly in the diagnosis and therapy of cancer, and underscores their potential use in clinical trials. Abstract Cancer remains a public health problem worldwide. Although conventional therapies have led to some excellent outcomes, some patients fail to respond to treatment, they have few therapeutic alternatives and a poor survival prognosis. Several strategies have been proposed to overcome this issue. The most recent approach is immunotherapy, particularly the use of recombinant antibodies and their derivatives, such as the single-chain fragment variable (scFv) containing the complete antigen-binding domains of a whole antibody that successfully targets tumor cells. This review describes the recent progress made with scFvs as a cancer diagnostic and therapeutic tool, with an emphasis on preclinical approaches and their potential use in clinical trials.
Collapse
Affiliation(s)
- Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Rosa María Ribas-Aparicio
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Prolongación de Carpio y Plan de Ayala S/N, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Hospital Infantil de México Federico Gómez, Doctor Márquez 162, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-(55)-5228-9917 (ext. 4401)
| |
Collapse
|
31
|
Frosi Y, Lin YC, Shimin J, Ramlan SR, Hew K, Engman AH, Pillai A, Yeung K, Cheng YX, Cornvik T, Nordlund P, Goh M, Lama D, Gates ZP, Verma CS, Thean D, Lane DP, Asial I, Brown CJ. Engineering an autonomous VH domain to modulate intracellular pathways and to interrogate the eIF4F complex. Nat Commun 2022; 13:4854. [PMID: 35982046 PMCID: PMC9388512 DOI: 10.1038/s41467-022-32463-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/02/2022] [Indexed: 11/12/2022] Open
Abstract
An attractive approach to target intracellular macromolecular interfaces and to model putative drug interactions is to design small high-affinity proteins. Variable domains of the immunoglobulin heavy chain (VH domains) are ideal miniproteins, but their development has been restricted by poor intracellular stability and expression. Here we show that an autonomous and disufhide-free VH domain is suitable for intracellular studies and use it to construct a high-diversity phage display library. Using this library and affinity maturation techniques we identify VH domains with picomolar affinity against eIF4E, a protein commonly hyper-activated in cancer. We demonstrate that these molecules interact with eIF4E at the eIF4G binding site via a distinct structural pose. Intracellular overexpression of these miniproteins reduce cellular proliferation and expression of malignancy-related proteins in cancer cell lines. The linkage of high-diversity in vitro libraries with an intracellularly expressible miniprotein scaffold will facilitate the discovery of VH domains suitable for intracellular applications.
Collapse
Affiliation(s)
- Yuri Frosi
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Yen-Chu Lin
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Insilico Medicine Taiwan Ltd., Taipei City, 110208, Taiwan
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei City, 112304, Taiwan
| | - Jiang Shimin
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Siti Radhiah Ramlan
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
| | - Kelly Hew
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Alf Henrik Engman
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Anil Pillai
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Kit Yeung
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Yue Xiang Cheng
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Tobias Cornvik
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
| | - Par Nordlund
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Megan Goh
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - Dilraj Lama
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Biomedicum Quarter 7B-C Solnavägen 9, 17165, Solna, Sweden
| | - Zachary P Gates
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), A*STAR, 8 A Biomedical Grove, #07-01 Neuros Building, 138665, Singapore, Singapore
| | - Chandra S Verma
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, 138671, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore, Singapore
| | - Dawn Thean
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - David P Lane
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore
| | - Ignacio Asial
- DotBio Pte. Ltd., 1 Research Link, Singapore, 117604, Singapore.
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore.
| | - Christopher J Brown
- p53 Laboratory (A*STAR), 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138648, Singapore.
- Disease Intervention Technology Laboratory (DITL), Institute of Molecular and Cell Biology, A*STAR, Singapore, 138673, Singapore.
| |
Collapse
|
32
|
IgG-Based Bispecific Anti-CD95 Antibodies for the Treatment of B Cell-Derived Malignancies and Autoimmune Diseases. Cancers (Basel) 2022; 14:cancers14163941. [PMID: 36010934 PMCID: PMC9405798 DOI: 10.3390/cancers14163941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Therapeutic antibodies have become a crucial cornerstone of the standard therapy for lymphoma and autoimmune diseases. However, the respective target antigens are also expressed on healthy B cells resulting in unspecific effects. In this article, we present a novel approach to selectively induce apoptosis in lymphoma cells and autoreactive B cells that express the CD95 death receptor. Therefore, we developed an improved IgG-based bispecific antibody format with favorable production properties and pharmacokinetics for CD20- and CD19-directed induction of apoptosis via CD95. We could show that our bispecific anti-CD95 antibodies are very efficient in the depletion of malignant and autoreactive B cells in vitro and in vivo. Therefore, our antibodies could help to provide a more selective therapy for patients with B cell-derived malignancies and autoimmune diseases. Abstract Antibodies against the B cell-specific antigens CD20 and CD19 have markedly improved the treatment of B cell-derived lymphoma and autoimmune diseases by depleting malignant and autoreactive B cells. However, since CD20 and CD19 are also expressed on healthy B cells, such antibodies lack disease specificity. Here, we optimize a previously developed concept that uses bispecific antibodies to induce apoptosis selectively in malignant and autoreactive B cells that express the death receptor CD95. We describe the development and characterization of bispecific antibodies with CD95xCD20 and CD95xCD19 specificity in a new IgG-based format. We could show that especially the CD95xCD20 antibody mediated a strong induction of apoptosis in malignant B cells in vitro. In vivo, the antibody was clearly superior to the previously used Fabsc format with identical specificities. In addition, both IgGsc antibodies depleted activated B cells in vitro, leading to a significant reduction in antibody production and cytokine secretion. The killing of resting B cells and hepatocytes that lack CD95 and CD20/CD19, respectively, was marginal. Thus, our results imply that bispecific anti-CD95 antibodies in the IgGsc format are an attractive tool for a more selective and efficient depletion of malignant as well as autoreactive B cells.
Collapse
|
33
|
Wang Y, Yuan W, Guo S, Li Q, Chen X, Li C, Liu Q, Sun L, Chen Z, Yuan Z, Luo C, Chen S, Tong S, Nassal M, Wen YM, Wang YX. A 33-residue peptide tag increases solubility and stability of Escherichia coli produced single-chain antibody fragments. Nat Commun 2022; 13:4614. [PMID: 35941164 PMCID: PMC9359998 DOI: 10.1038/s41467-022-32423-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Single-chain variable fragments (scFvs), composed of variable domains of heavy and light chains of an antibody joined by a linker, share antigen binding capacity with their parental antibody. Due to intrinsically low solubility and stability, only two Escherichia coli-produced scFvs have been approved for therapy. Here we report that a 33-residue peptide, termed P17 tag, increases the solubility of multiple scFvs produced in Escherichia coli SHuffle strain by up to 11.6 fold. Hydrophilic sequence, especially charged residues, but not the predicted α-helical secondary structure of P17 tag, contribute to the solubility enhancement. Notably, the P17 tag elevates the thermostability of scFv as efficiently as intra-domain disulfide bonds. Moreover, a P17-tagged scFv targeting hepatitis B virus surface proteins shows over two-fold higher antigen-binding affinity and virus-neutralizing activity than the untagged version. These data strongly suggest a type I intramolecular chaperone-like activity of the P17 tag. Hence, the P17 tag could benefit the research, production, and application of scFv. Low solubility and stability of Escherichia coli produced single chain variable fragments (scFvs) restrict their applications. Here the authors report a 33-residue peptide tag which simultaneously increases the solubility and thermostability of multiple scFvs produced in Escherichia coli SHuffle strain.
Collapse
Affiliation(s)
- Yang Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjie Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Siqi Guo
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmacy, Nanchang University, Nanchang, China
| | - Qiqi Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaomei Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qianying Liu
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Lei Sun
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Zhenguo Chen
- Institutes of Biomedical Science, Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Luo
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, China
| | - Shijie Chen
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Shuping Tong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Michael Nassal
- Department of Internal Medicine II/Molecular Biology, University Hospital Freiburg, Freiburg, Germany
| | - Yu-Mei Wen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong-Xiang Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
34
|
Birtel M, Voss RH, Reinhard K, Rengstl B, Ouchan Y, Michel K, Hayduk N, Tillmann B, Becker R, Suchan M, Theobald M, Oehm P, Türeci Ö, Sahin U. A TCR-like CAR Promotes Sensitive Antigen Recognition and Controlled T-cell Expansion Upon mRNA Vaccination. CANCER RESEARCH COMMUNICATIONS 2022; 2:827-841. [PMID: 36923303 PMCID: PMC10010320 DOI: 10.1158/2767-9764.crc-21-0154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/10/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Chimeric antigen receptor (CAR) T cells are efficacious in patients with B-cell malignancies, while their activity is limited in patients with solid tumors. We developed a novel heterodimeric TCR-like CAR (TCAR) designed to achieve optimal chain pairing and integration into the T-cell CD3 signaling complex. The TCAR mediated high antigen sensitivity and potent antigen-specific T-cell effector functions in short-term in vitro assays. Both persistence and functionality of TCAR T cells were augmented by provision of costimulatory signals, which improved proliferation in vitro and in vivo. Combination with a nanoparticulate RNA vaccine, developed for in vivo expansion of CAR T cells, promoted tightly controlled expansion, survival, and antitumor efficacy of TCAR T cells in vivo. Significance A novel TCAR is tightly controlled by RNA vaccine-mediated costimulation and may provide an alternative to second-generation CARs for the treatment of solid tumors.
Collapse
Affiliation(s)
- Matthias Birtel
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Ralf-Holger Voss
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Department of Research Center for Immunotherapy (FZI), University Medical Center (UMC) of the Johannes Gutenberg University, Mainz, Germany
| | - Katharina Reinhard
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Benjamin Rengstl
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Yasmina Ouchan
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Kristina Michel
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Nina Hayduk
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Bodo Tillmann
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - René Becker
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - Martin Suchan
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
| | - Matthias Theobald
- Department of Hematology, Oncology, and Pneumology, University Cancer Center (UCT), University Medical Center (UMC) of Johannes Gutenberg University, Mainz, Germany
| | - Petra Oehm
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Özlem Türeci
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
| | - Ugur Sahin
- TRON – Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH (non-profit), Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, BioNTech Cell & Gene Therapies GmbH, Mainz, Germany
- Department of Research Center for Immunotherapy (FZI), University Medical Center (UMC) of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
35
|
Khatib SE, Salla M. The mosaic puzzle of the therapeutic monoclonal antibodies and antibody fragments - A modular transition from full-length immunoglobulins to antibody mimetics. Leuk Res Rep 2022; 18:100335. [PMID: 35832747 PMCID: PMC9272380 DOI: 10.1016/j.lrr.2022.100335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 06/09/2022] [Accepted: 06/25/2022] [Indexed: 01/07/2023] Open
Abstract
The use of monoclonal antibodies represents an important and efficient diagnostic and therapeutic tool in disease management and modern science but remains limited by several factors including the uneven distribution in diseased tissues as well as undesired activation of side immune reactions. Major scientific advancements including Recombinant DNA Technology, Hybridoma Technology, and Polymerase Chain Reaction have considerably impacted the use of monoclonal antibodies providing technical and effective solutions to overcome the shortcomings encountered with conventional antibodies. Initially, the introduction of antibody fragments allowed a more uniform and deeper penetration of the targeted tissue and reduced unwanted activation of Fc-mediated immune reactions. On another level, the immunogenicity of murine-derived antibodies was overcome by humanizing their encoding genes with specific sequences of human origin andtransgenic mice able to synthesize fully human antibodies were successfully created. Moreover, the advancement of genetic engineering techniques supported by the modular structure of antibody coding genes paved the way for the development of a new generation of antibody fragments with a wide spectrum of monospecific and bispecific agents. These later could be monovalent, bivalent, or multivalent, and either expressed as a single chain, assembled in multimeric forms or stringed in tandem. This has conferred improved affinity, stability, and solubility to antibody targetting. Lately, a new array of monoclonal antibody fragments was introduced with the engineering of nanobody and antibody mimetics as non-immunoglobulin-derived fragments with promising diagnostic and therapeutic applications. In this review, we decipher the molecular basis of monoclonal antibody engineering with a detailed screening of the antibody derivatives that provides new perspectives to expand the use of monoclonal fragments into previously unexplored fields.
Collapse
Affiliation(s)
- Sami El Khatib
- Lebanese International University, Department of Biomedical Sciences, Bekaa Campus, Khiyara, West Bekaa, Lebanon
| | - Mohamed Salla
- University of Alberta. Biochemistry Department, Faculty of Medicine and Dentistry,116St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
36
|
Hambach J, Fumey W, Stähler T, Gebhardt AJ, Adam G, Weisel K, Koch-Nolte F, Bannas P. Half-Life Extended Nanobody-Based CD38-Specific Bispecific Killercell Engagers Induce Killing of Multiple Myeloma Cells. Front Immunol 2022; 13:838406. [PMID: 35651607 PMCID: PMC9150782 DOI: 10.3389/fimmu.2022.838406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/19/2022] [Indexed: 01/14/2023] Open
Abstract
CD38 is a target for immunotherapy of multiple myeloma. Llama-derived CD38-specific nanobodies allow easy reformatting into mono-, bi- and multispecific proteins. To evaluate the utility of nanobodies for constructing CD38-specific nanobody-based killer cell engagers (nano-BiKEs), we generated half-life extended nano-BiKEs (HLE-nano-BiKEs) by fusing a CD38-specific nanobody to a CD16-specific nanobody for binding to the Fc-receptor on NK cells and further to an albumin-specific nanobody to extend the half-life in vivo. HLE-nano-BiKEs targeting three different epitopes (E1, E2, E3) of CD38 were expressed in transiently transfected HEK-6E cells. We verified specific and simultaneous binding to CD38 on myeloma cells, CD16 on NK cells, and to albumin. We tested the capacity of these HLE-nano-BiKEs to mediate cytotoxicity against CD38-expressing multiple myeloma cell lines and primary myeloma cells from human bone marrow biopsies in bioluminescence and flowcytometry assays with NK92 cells as effector cells. The results revealed specific time- and dose-dependent cytolysis of CD38+ myeloma cell lines and effective depletion of CD38-expressing multiple myeloma cells from primary human bone marrow samples. Our results demonstrate the efficacy of CD38-specific HLE-nano-BiKEs in vitro and ex vivo, warranting further preclinical evaluation in vivo of their therapeutic potential for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - William Fumey
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Josephine Gebhardt
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Weisel
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Peter Bannas,
| |
Collapse
|
37
|
Effect of non-repetitive linker on in vitro and in vivo properties of an anti-VEGF scFv. Sci Rep 2022; 12:5449. [PMID: 35361822 PMCID: PMC8971466 DOI: 10.1038/s41598-022-09324-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/03/2022] [Indexed: 11/08/2022] Open
Abstract
Single chain antibody fragments (scFvs) are favored in diagnostic and therapeutic fields thanks to their small size and the availability of various engineering approaches. Linker between variable heavy (VH) and light (VL) chains of scFv covalently links these domains and it can affect scFv’s bio-physical/chemical properties and in vivo activity. Thus, scFv linker design is important for a successful scFv construction, and flexible linkers are preferred for a proper pairing of VH–VL. The flexibility of the linker is determined by length and sequence content and glycine-serine (GS) linkers are commonly preferred for scFvs based on their highly flexible profiles. Despite the advantage of this provided flexibility, GS linkers carry repeated sequences which can cause problems for PCR-based engineering approaches and immunogenicity. Here, two different linkers, a repetitive GS linker and an alternative non-repetitive linker with similar flexibility but lower immunogenicity are employed to generate anti-Vascular Endothelial Growth Factor scFvs derived from bevacizumab. Our findings highlight a better in vitro profile of the non-repetitive linker such as a higher monomer ratio, higher thermal stability while there was no significant difference in in vivo efficacy in a zebrafish embryonic angiogenesis model. This is the first study to compare in vivo efficacy of scFvs with different linkers in a zebrafish model.
Collapse
|
38
|
Mirzaeinia S, Zeinali S, Budisa N, Karbalaei-Heidari HR. Targeted Codelivery of Prodigiosin and Simvastatin Using Smart BioMOF: Functionalization by Recombinant Anti-VEGFR1 scFv. Front Bioeng Biotechnol 2022; 10:866275. [PMID: 35402395 PMCID: PMC8987009 DOI: 10.3389/fbioe.2022.866275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 11/18/2022] Open
Abstract
Biological metal-organic frameworks (BioMOFs) are hybrid compounds in which metal nodes are linked to biocompatible organic ligands and have potential for medical application. Herein, we developed a novel BioMOF modified with an anti-VEGFR1 scFv antibody (D16F7 scFv). Our BioMOF is co-loaded with a combination of an anticancer compound and a lipid-lowering drug to simultaneously suppress the proliferation, growth rate and metastases of cancer cells in cell culture model system. In particular, Prodigiosin (PG) and Simvastatin (SIM) were co-loaded into the newly synthesized Ca-Gly BioMOF nanoparticles coated with maltose and functionalized with a recombinant maltose binding protein-scFv fragment of anti-VEGFR1 (Ca-Gly-Maltose-D16F7). The nanoformulation, termed PG + SIM-NP-D16F7, has been shown to have strong active targeting behavior towards VEGFR1-overexpresing cancer cells. Moreover, the co-delivery of PG and SIM not only effectively inhibits the proliferation of cancer cells, but also prevents their invasion and metastasis. The PG + SIM-NP-D16F7 nanocarrier exhibited stronger cytotoxic and anti-metastatic effects compared to mono-treatment of free drugs and drug-loaded nanoparticles. Smart co-delivery of PG and SIM on BioMOF nanoparticles had synergistic effects on growth inhibition and prevented cancer cell metastasis. The present nanoplatform can be introduced as a promising tool for chemotherapy compared with mono-treatment and/or non-targeted formulations.
Collapse
Affiliation(s)
- Somayyeh Mirzaeinia
- Molecular Biotechnology Lab, Department of Biology, Faculty of Science, Shiraz University, Shiraz, Iran
| | - Sedighe Zeinali
- Department of Nanochemical Engineering, School of Advanced Technologies, Nanotechnology Research Institute, Shiraz University, Shiraz, Iran
| | - Nediljko Budisa
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
- Institut für Chemie, Technische Universität Berlin, Berlin, Germany
| | - Hamid Reza Karbalaei-Heidari
- Molecular Biotechnology Lab, Department of Biology, Faculty of Science, Shiraz University, Shiraz, Iran
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
39
|
Uchino S, Ito Y, Sato Y, Handa T, Ohkawa Y, Tokunaga M, Kimura H. Live imaging of transcription sites using an elongating RNA polymerase II-specific probe. J Cell Biol 2022; 221:212888. [PMID: 34854870 PMCID: PMC8647360 DOI: 10.1083/jcb.202104134] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/12/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
In eukaryotic nuclei, most genes are transcribed by RNA polymerase II (RNAP2), whose regulation is a key to understanding the genome and cell function. RNAP2 has a long heptapeptide repeat (Tyr1-Ser2-Pro3-Thr4-Ser5-Pro6-Ser7), and Ser2 is phosphorylated on an elongation form. To detect RNAP2 Ser2 phosphorylation (RNAP2 Ser2ph) in living cells, we developed a genetically encoded modification-specific intracellular antibody (mintbody) probe. The RNAP2 Ser2ph-mintbody exhibited numerous foci, possibly representing transcription “factories,” and foci were diminished during mitosis and in a Ser2 kinase inhibitor. An in vitro binding assay using phosphopeptides confirmed the mintbody’s specificity. RNAP2 Ser2ph-mintbody foci were colocalized with proteins associated with elongating RNAP2 compared with factors involved in the initiation. These results support the view that mintbody localization represents the sites of RNAP2 Ser2ph in living cells. RNAP2 Ser2ph-mintbody foci showed constrained diffusional motion like chromatin, but they were more mobile than DNA replication domains and p300-enriched foci, suggesting that the elongating RNAP2 complexes are separated from more confined chromatin domains.
Collapse
Affiliation(s)
- Satoshi Uchino
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuma Ito
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Yuko Sato
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Tetsuya Handa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Makio Tokunaga
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Hiroshi Kimura
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan.,Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Japan
| |
Collapse
|
40
|
Dong S, He K, Guan L, Shi Q, Wu J, Feng J, Yang W, Shi X. Construction and Characterization of a Single-Chain Variable Fragment (scFv) for the Detection of Cry1Ab and Cry1Ac Toxins from the Anti-Cry1Ab Monoclonal Antibody. FOOD ANAL METHOD 2022. [DOI: 10.1007/s12161-021-02223-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Liu Y, Tsang K, Mays M, Hansen G, Chiecko J, Crames M, Wei Y, Zhou W, Fredrick C, Hu J, Liu D, Gebhard D, Huang ZF, Datar A, Kronkaitis A, Gueneva-Boucheva K, Seeliger D, Han F, Sen S, Kasturirangan S, Scheer JM, Nixon AE, Panavas T, Marlow MS, Kumar S. An adapted consensus protein design strategy for identifying globally optimal biotherapeutics. MAbs 2022; 14:2073632. [PMID: 35613320 PMCID: PMC9135432 DOI: 10.1080/19420862.2022.2073632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Biotherapeutic optimization, whether to improve general properties or to engineer specific attributes, is a time-consuming process with uncertain outcomes. Conversely, Consensus Protein Design has been shown to be a viable approach to enhance protein stability while retaining function. In adapting this method for a more limited number of protein sequences, we studied 21 consensus single-point variants from eight publicly available CD3 binding sequences with high similarity but diverse biophysical and pharmacological properties. All single-point consensus variants retained CD3 binding and performed similarly in cell-based functional assays. Using Ridge regression analysis, we identified the variants and sequence positions with overall beneficial effects on developability attributes of the CD3 binders. A second round of sequence generation that combined these substitutions into a single molecule yielded a unique CD3 binder with globally optimized developability attributes. In this first application to therapeutic antibodies, adapted Consensus Protein Design was found to be highly beneficial within lead optimization, conserving resources and minimizing iterations. Future implementations of this general strategy may help accelerate drug discovery and improve success rates in bringing novel biotherapeutics to market.
Collapse
Affiliation(s)
- Yanyun Liu
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Kenny Tsang
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Michelle Mays
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Gale Hansen
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Jeffrey Chiecko
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Maureen Crames
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Yangjie Wei
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Weijie Zhou
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Chase Fredrick
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - James Hu
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Dongmei Liu
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Douglas Gebhard
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Zhong-Fu Huang
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Akshita Datar
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Anthony Kronkaitis
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | | | - Daniel Seeliger
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Fei Han
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Saurabh Sen
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Srinath Kasturirangan
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Justin M Scheer
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Andrew E Nixon
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Tadas Panavas
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Michael S Marlow
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| | - Sandeep Kumar
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceutical Inc., Ridgefield, CT, USA
| |
Collapse
|
42
|
Soleimanizadeh A, Dinter H, Schindowski K. Central Nervous System Delivery of Antibodies and Their Single-Domain Antibodies and Variable Fragment Derivatives with Focus on Intranasal Nose to Brain Administration. Antibodies (Basel) 2021; 10:antib10040047. [PMID: 34939999 PMCID: PMC8699001 DOI: 10.3390/antib10040047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
IgG antibodies are some of the most important biopharmaceutical molecules with a high market volume. In spite of the fact that clinical therapies with antibodies are broadly utilized in oncology, immunology and hematology, their delivery strategies and biodistribution need improvement, their limitations being due to their size and poor ability to penetrate into tissues. In view of their small size, there is a rising interest in derivatives, such as single-domain antibodies and single-chain variable fragments, for clinical diagnostic but also therapeutic applications. Smaller antibody formats combine several benefits for clinical applications and can be manufactured at reduced production costs compared with full-length IgGs. Moreover, such formats have a relevant potential for targeted drug delivery that directs drug cargo to a specific tissue or across the blood–brain barrier. In this review, we give an overview of the challenges for antibody drug delivery in general and focus on intranasal delivery to the central nervous system with antibody formats of different sizes.
Collapse
Affiliation(s)
- Arghavan Soleimanizadeh
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Faculty of Medicine, University of Ulm, 89081 Ulm, Germany
| | - Heiko Dinter
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Department of Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Correspondence:
| |
Collapse
|
43
|
Farokhinejad F, Lane RE, Lobb RJ, Edwardraja S, Wuethrich A, Howard CB, Trau M. Generation of Nanoyeast Single-Chain Variable Fragments as High-Avidity Biomaterials for Dengue Virus Detection. ACS Biomater Sci Eng 2021; 7:5850-5860. [PMID: 34738789 DOI: 10.1021/acsbiomaterials.1c01001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bioengineered yeast bio-nanomaterials termed nanoyeasts displaying antibody single-chain variable fragments (scFvs) against diagnostic targets are a promising alternative to monoclonal antibodies (mAbs). A potential limitation for translating nanoyeasts into diagnostic tools is batch-to-batch variability. Herein, we demonstrate a systematic approach for cost-efficient production of highly specific nanoyeasts that enabled accurate dengue virus (DENV) detection by immunoassay (2.5% CV). Yeasts bioengineered to surface express DENV-specific scFvs (up to 66% of the total cell population) were fragmented into nanoyeast fractions trialing sonication, bead beating, and high-pressure disruption methods. Nanoyeast fractions from sonication had optimal target binding, uniform particle size (±89 nm), were stable, and retained diagnostic activity for 7 days at 37 °C compared to traditional mAbs that lost activity after 1 day at 37 °C. We engineered a panel of nanoyeast scFvs targeting DENV nonstructural protein 1 (NS1): (i) specific for serotyping DENV 1-4 and (ii) cross-reactive anti-DENV scFvs that are suitable for "yes/no" diagnostic applications. We demonstrate highly specific nanoyeast scFvs for serotyping DENV. We show that nanoyeast scFvs specifically detect NS1 in simulated patient plasma with a limit of detection of 250 ng/mL, the concentration found in infected patients.
Collapse
Affiliation(s)
- Fahimeh Farokhinejad
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rebecca E Lane
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Richard J Lobb
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Selvakumar Edwardraja
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Alain Wuethrich
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Christopher B Howard
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Matt Trau
- Centre of Personalized Nanomedicine, Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
44
|
Tomar DS, Licari G, Bauer J, Singh SK, Li L, Kumar S. Stress-dependent flexibility of a full-length human monoclonal antibody: Insights from molecular dynamics to support biopharmaceutical development. J Pharm Sci 2021; 111:628-637. [PMID: 34742728 DOI: 10.1016/j.xphs.2021.10.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/30/2021] [Accepted: 10/30/2021] [Indexed: 01/15/2023]
Abstract
After several decades of advancements in drug discovery, product development of biopharmaceuticals remains a time- and resource-consuming endeavor. One of the main reasons is associated to the lack of fundamental understanding of conformational dynamics of such biologic entities, and how they respond to various stresses encountered during manufacturing. In this work, we have studied the conformational dynamics of human IgG1κ b12 monoclonal antibody (mAb) using molecular dynamics simulations. The hundreds of nanoseconds long trajectories reveal that b12 mAb is highly flexible. Its variable domains show greater conformational fluctuations than the constant domains. Additionally, it collapses towards a more globular shape in response to thermal stress, leading to decrease in the total solvent exposed surface area and radius of gyration. This behavior is more pronounced for the deglycosylated b12 mAb, and it appears to correlate with increase in inter-domain contacts between specific regions of the antibody. Conformational fluctuations also cause temporary formation and disruption of hydrophobic and charged patches on the antibody surface, which is particularly important for the prediction of CMC properties during development phases of antibody-based biotherapeutics. The insights gained through these simulations may help the development of biologic drugs, especially with regards to manufacturing processes where antibodies may undergo significant thermal stress.
Collapse
Affiliation(s)
- Dheeraj S Tomar
- Biotherapeutics Pharmaceutical Sciences Research and Development, Pfizer Inc., 700 Chesterfield Parkway West, Chesterfield, MO, 63017, USA
| | - Giuseppe Licari
- Pharmaceuticals Development Biologicals, Boehringer Ingelheim Pharmaceuticals, Inc., D-88397 Biberach an der Riss, Germany
| | - Joschka Bauer
- Pharmaceuticals Development Biologicals, Boehringer Ingelheim Pharmaceuticals, Inc., D-88397 Biberach an der Riss, Germany
| | - Satish K Singh
- Biotherapeutics Pharmaceutical Sciences Research and Development, Pfizer Inc., 700 Chesterfield Parkway West, Chesterfield, MO, 63017, USA
| | - Li Li
- Biotherapeutics Pharmaceutical Sciences Research and Development, Pfizer Inc., 1 Burtt Road, Andover, Massachusetts, 01810, USA
| | - Sandeep Kumar
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877.
| |
Collapse
|
45
|
Meric G, Naik S, Hunter AK, Robinson AS, Roberts CJ. Challenges for design of aggregation-resistant variants of granulocyte colony-stimulating factor. Biophys Chem 2021; 277:106630. [PMID: 34119805 DOI: 10.1016/j.bpc.2021.106630] [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] [Received: 03/07/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 01/15/2023]
Abstract
Non-native protein aggregation is a long-standing issue in pharmaceutical biotechnology. A rational design approach was used in order to identify variants of recombinant human granulocyte colony-stimulating factor (rhG-CSF) with lower aggregation propensity at solution conditions that are typical of commercial formulation. The approach used aggregation-prone-region (APR) predictors to select single amino acid substitutions that were predicted to decrease intrinsic aggregation propensity (IAP). The results of static light scattering temperature-ramps and chemical unfolding experiments demonstrated that none of the selected variants exhibited improved aggregation resistance, and the apparent conformational stability of each variant was lower than that of WT. Aggregation studies under partly denaturing conditions suggested that the IAP of at least one variant remained unaltered. Overall, this study highlights a general challenge in designing aggregation resistance for proteins, due to the need to accurately predict both APRs and conformational stability.
Collapse
Affiliation(s)
- Gulsum Meric
- Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States.
| | - Subhashchandra Naik
- Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States.
| | - Alan K Hunter
- Biopharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, MD 20878, United States.
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States.
| | - Christopher J Roberts
- Chemical & Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
46
|
Dyson MR, Masters E, Pazeraitis D, Perera RL, Syrjanen JL, Surade S, Thorsteinson N, Parthiban K, Jones PC, Sattar M, Wozniak-Knopp G, Rueker F, Leah R, McCafferty J. Beyond affinity: selection of antibody variants with optimal biophysical properties and reduced immunogenicity from mammalian display libraries. MAbs 2021; 12:1829335. [PMID: 33103593 PMCID: PMC7592150 DOI: 10.1080/19420862.2020.1829335] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The early phase of protein drug development has traditionally focused on target binding properties leading to a desired mode of therapeutic action. As more protein therapeutics pass through the development pipeline; however, it is clear that non-optimal biophysical properties can emerge, particularly as proteins are formulated at high concentrations, causing aggregation or polyreactivity. Such late-stage "developability" problems can lead to delay or failure in traversing the development process. Aggregation propensity is also correlated with increased immunogenicity, resulting in expensive, late-stage clinical failures. Using nucleases-directed integration, we have constructed large mammalian display libraries where each cell contains a single antibody gene/cell inserted at a single locus, thereby achieving transcriptional normalization. We show a strong correlation between poor biophysical properties and display level achieved in mammalian cells, which is not replicated by yeast display. Using two well-documented examples of antibodies with poor biophysical characteristics (MEDI-1912 and bococizumab), a library of variants was created based on surface hydrophobic and positive charge patches. Mammalian display was used to select for antibodies that retained target binding and permitted increased display level. The resultant variants exhibited reduced polyreactivity and reduced aggregation propensity. Furthermore, we show in the case of bococizumab that biophysically improved variants are less immunogenic than the parental molecule. Thus, mammalian display helps to address multiple developability issues during the earliest stages of lead discovery, thereby significantly de-risking the future development of protein drugs.
Collapse
Affiliation(s)
| | | | | | | | - Johanna L Syrjanen
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory , NY, USA
| | | | | | | | | | | | - Gordana Wozniak-Knopp
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences , Vienna, Austria
| | - Florian Rueker
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences , Vienna, Austria
| | | | | |
Collapse
|
47
|
Fernández-Taboada G, Riaño-Umbarila L, Olvera-Rodríguez A, Gómez-Ramírez IV, Losoya-Uribe LF, Becerril B. The venom of the scorpion Centruroides limpidus, which causes the highest number of stings in Mexico, is neutralized by two recombinant antibody fragments. Mol Immunol 2021; 137:247-255. [PMID: 34298407 DOI: 10.1016/j.molimm.2021.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/05/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022]
Abstract
Phage display and directed evolution have made it possible to generate recombinant antibodies in the format of single chain variable fragments (scFvs) capable of neutralizing different toxins and venoms of Mexican scorpions. Despite having managed to neutralize a significant number of venoms, some others have not yet been completely neutralized, due to the diversity of the toxic components present in them. An example is the venom of the scorpion Centruroides limpidus, which contains three toxins of medical importance, called Cll1, Cll2 and Cl13. The first two are neutralized by scFv 10FG2, while Cl13, due to its sequence divergence, was not even recognized. For this reason, the aim of the present work was the generation of a new scFv capable of neutralizing Cl13 toxin and thereby helping to neutralize the whole venom of this scorpion. By hybridoma technology, a monoclonal antibody (mAb B7) was generated, which was able to recognize and partially neutralize Cl13 toxin. From mAb B7, its scFv format was obtained, named scFv B7 and subjected to three cycles of directed evolution. At the end of these processes, scFv 11F which neutralized Cl13 toxin was obtained. This scFv, administered in conjunction with scFv 10FG2, allowed to fully neutralize the whole venom of Centruroides limpidus scorpion.
Collapse
Affiliation(s)
- Guillermo Fernández-Taboada
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Lidia Riaño-Umbarila
- Cátedra CONACYT, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Alejandro Olvera-Rodríguez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Ilse Viridiana Gómez-Ramírez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Luis Fernando Losoya-Uribe
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| | - Baltazar Becerril
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, UNAM, Apartado Postal 510-3, Cuernavaca, Morelos, 62250, Mexico.
| |
Collapse
|
48
|
Enderle L, Shalaby KH, Gorelik M, Weiss A, Blazer LL, Paduch M, Cardarelli L, Kossiakoff A, Adams JJ, Sidhu SS. A T cell redirection platform for co-targeting dual antigens on solid tumors. MAbs 2021; 13:1933690. [PMID: 34190031 PMCID: PMC8253144 DOI: 10.1080/19420862.2021.1933690] [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] [Indexed: 11/05/2022] Open
Abstract
In order to direct T cells to specific features of solid cancer cells, we engineered a bispecific antibody format, named Dual Antigen T cell Engager (DATE), by fusing a single-chain variable fragment targeting CD3 to a tumor-targeting antigen-binding fragment. In this format, multiple novel paratopes against different tumor antigens were able to recruit T-cell cytotoxicity to tumor cells in vitro and in an in vivo pancreatic ductal adenocarcinoma xenograft model. Since unique surface antigens in solid tumors are limited, in order to enhance selectivity, we further engineered “double-DATEs” targeting two tumor antigens simultaneously. The double-DATE contains an additional autonomous variable heavy-chain domain, which binds a second tumor antigen without itself eliciting a cytotoxic response. This novel modality provides a strategy to enhance the selectivity of immune redirection through binary targeting of native tumor antigens. The modularity and use of a common, stable human framework for all components enables a pipeline approach to rapidly develop a broad repertoire of tailored DATEs and double-DATEs with favorable biophysical properties and high potencies and selectivities.
Collapse
Affiliation(s)
- Leonie Enderle
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Karim H Shalaby
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Maryna Gorelik
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Alexander Weiss
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Levi L Blazer
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Marcin Paduch
- Institute for Biophysical Dynamics, Gordon Center for Integrative Science, Chicago, USA
| | - Lia Cardarelli
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Anthony Kossiakoff
- Institute for Biophysical Dynamics, Gordon Center for Integrative Science, Chicago, USA.,Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, USA
| | - Jarrett J Adams
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Sachdev S Sidhu
- Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
49
|
Sokullu E, Gauthier MS, Coulombe B. Discovery of Antivirals Using Phage Display. Viruses 2021; 13:v13061120. [PMID: 34200959 PMCID: PMC8230593 DOI: 10.3390/v13061120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
The latest coronavirus disease outbreak, COVID-19, has brought attention to viral infections which have posed serious health threats to humankind throughout history. The rapid global spread of COVID-19 is attributed to the increased human mobility of today's world, yet the threat of viral infections to global public health is expected to increase continuously in part due to increasing human-animal interface. Development of antiviral agents is crucial to combat both existing and novel viral infections. Recently, there is a growing interest in peptide/protein-based drug molecules. Antibodies are becoming especially predominant in the drug market. Indeed, in a remarkably short period, four antibody therapeutics were authorized for emergency use in COVID-19 treatment in the US, Russia, and India as of November 2020. Phage display has been one of the most widely used screening methods for peptide/antibody drug discovery. Several phage display-derived biologics are already in the market, and the expiration of intellectual property rights of phage-display antibody discovery platforms suggests an increment in antibody drugs in the near future. This review summarizes the most common phage display libraries used in antiviral discovery, highlights the approaches employed to enhance the antiviral potency of selected peptides/antibody fragments, and finally provides a discussion about the present status of the developed antivirals in clinic.
Collapse
Affiliation(s)
- Esen Sokullu
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| | - Marie-Soleil Gauthier
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
| | - Benoit Coulombe
- Department of Translational Proteomics, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada;
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
- Correspondence: (E.S.); (B.C.)
| |
Collapse
|
50
|
Xi Z, Liu X, Lin R, Persons JD, Ilina TV, Li W, Dimitrov DS, Ishima R. The reduced form of the antibody CH2 domain. Protein Sci 2021; 30:1895-1903. [PMID: 34107549 DOI: 10.1002/pro.4142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022]
Abstract
Among the immunoglobulin domains, the CH2 domain has the lowest thermal stability, which also depends on amino acid sequence and buffer conditions. To further identify factors that influence CH2 folding and stability, we characterized the domain in the reduced form using differential scanning fluorimetry and nuclear magnetic resonance. We show that the CH2 domain can fold, similarly to the disulfide-bridged form, without forming a disulfide-bridge, even though the protein contains two Cys residues. Although the reduced form exhibits thermal stability more than 15°C lower than the disulfide-bridged form, it does not undergo immediate full oxidization. To explain this phenomenon, we compared CH2 oxidization at different conditions and demonstrate a need for significant fluctuation of the folded conformation to enhance CH2 disulfide-bridge formation. We conclude that, since CH2 can be purified as a folded, semi-stable, reduced protein that can coexist with the oxidized form, verification of the level of oxidization at each step is critical in CH2 engineering studies.
Collapse
Affiliation(s)
- Zhaoyong Xi
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Xianglei Liu
- Center for Antibody Therapeutics, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rui Lin
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Gastroenterology and Hepatology, Tianjin Medical University, General Hospital, Tianjin, China
| | - John D Persons
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Tatiana V Ilina
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Wei Li
- Center for Antibody Therapeutics, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|