1
|
Zhang Z, Guo W, Chen M, Yang Q, Song X, Wang Y. A real-world pharmacovigilance study of blinatumomab based on the FDA adverse event reporting system. Expert Opin Drug Saf 2025:1-8. [PMID: 39912446 DOI: 10.1080/14740338.2025.2464070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Blinatumomab, the first CD3/CD19 bispecific antibody, is FDA-approved for relapsed or refractory precursor B-cell acute lymphoblastic leukemia in adults and children. This study evaluates its safety profile through pharmacovigilance analysis of adverse events (AEs) reported in the FDA Adverse Event Reporting System (FAERS). METHOD We conducted a disproportionality analysis using four algorithms: Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-Item Gamma Poisson Shrinker (MGPS). Data from 2014Q1 to 2023Q4 were analyzed to identify safety signals related to blinatumomab, along with a stratification analysis to examine AE onset timing. RESULT A total of 17,131 AE reports were retrieved from the FAERS database, with 6,266 indicating blinatumomab as the primary suspect. We identified 277 preferred terms (PTs) demonstrating significant disproportionality across all algorithms. Notably, unexpected AEs included Graft Versus Host Disease, myelosuppression, and hypokalaemia. Common AEs were consistent across gender and age groups, predominantly occurring within one month of treatment. CONCLUSION This pharmacovigilance study utilizing the FAERS database identified potential AE signals associated with blinatumomab, providing essential insights for its safe clinical use.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wenhao Guo
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
| | - Minghao Chen
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qianzhi Yang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xia Song
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu, China
- Department of Clinical Pharmacy, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yuping Wang
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Tripathy RK, Pande AH. Nanobody-Oligonucleotide Conjugates (NucleoBodies): The Next Frontier in Oligonucleotide Therapy. Pharm Res 2025; 42:219-236. [PMID: 39953265 DOI: 10.1007/s11095-025-03829-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
As of now, more than 15 oligonucleotide drugs, primarily small interfering RNAs and antisense oligonucleotide classes, have been approved by the US FDA for therapeutic use, and many more are under clinical trials. However, safe and effective delivery of the oligonucleotide-based drugs to the target tissue still remains a major challenge. For enhanced plasma half-life, effective endosomal release, and other multiple functionalities, various carrier molecules have been used over the years. The successful therapeutic application of antibody-drug conjugates has made antibodies a popular choice for the delivery of oligonucleotide payloads into the target tissues. Single-chain variable domains of heavy chain antibodies (nanobodies) have proven a promising alternative to antibodies in recent years due to their small size, high affinity for the target, cell-penetrating potency, simple and easy production. The present review highlights the oligonucleotide drug types and their conjugation with nanobodies called NucleoBodies for effective targeted delivery, detection and diagnostics.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
3
|
Gonzàlez Gutierrez C, Aimard A, Biarnes-Pélicot M, Kerfelec B, Puech PH, Robert P, Piazza F, Chames P, Limozin L. Decoupling Individual Host Response and Immune Cell Engager Cytotoxic Potency. ACS NANO 2025; 19:2089-2098. [PMID: 39791371 DOI: 10.1021/acsnano.4c08541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Immune cell engagers are molecular agents, usually antibody-based constructs, engineered to recruit immune cells against cancer cells and kill them. They are versatile and powerful tools for cancer immunotherapy. Despite the multiplication of engagers tested and accepted in the clinic, how molecular and cellular parameters influence their actions is poorly understood. In particular, disentangling the respective roles of host immune cells and engager biophysical characteristics is needed to improve their design and efficiency. Focusing here on harnessing antibody-dependent Natural Killer cell cytotoxicity, we measure the efficiency of 6 original bispecific antibodies (bsAb), associating an anti-HER2 nanobody and an anti-CD16 nanobody. In vitro cytotoxicity data using primary human NK cells on different target cell lines exposing different antigen densities were collected, exhibiting a wide range of bsAb dose response. In order to rationalize our observations, we introduce a simple multiscale model, postulating that the density of bsAb bridging the two cells is the main parameter triggering the cytotoxic response. We introduce two microscopic parameters: the surface cooperativity describing bsAb affinity at the bridging step and the threshold of bridge density determining the donor-dependent response. Both parameters permit ranking Abs and donors and predicting bsAb potency as a function of antibodies bulk affinities and receptor surface densities on cells. Our approach thus provides a general way to decouple donor response from immune engager characteristics, rationalizing the landscape of molecule design.
Collapse
Affiliation(s)
| | - Adrien Aimard
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | | | - Brigitte Kerfelec
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Pierre-Henri Puech
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| | - Philippe Robert
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
- Assistance Publique Hôpitaux de Marseille, 13005 Marseille, France
| | - Francesco Piazza
- CNRS, Univ. Orleans, CBM, 45000 Orleans, France
- Dipartimento di Fisica e Astronomia, Università di Firenze and INFN sezione di Firenze, 50019 Sesto Fiorentino, Italy
| | - Patrick Chames
- Aix-Marseille Univ., CNRS, INSERM, Institut Paoli Calmettes, CRCM, 13009 Marseille, France
| | - Laurent Limozin
- Aix-Marseille Univ., CNRS, INSERM, LAI, Centuri Living Systems, 13009 Marseille, France
| |
Collapse
|
4
|
Rees M, Abdallah N, Yohannan B, Gonsalves WI. Bispecific antibody targets and therapies in multiple myeloma. Front Immunol 2024; 15:1424925. [PMID: 39450163 PMCID: PMC11499143 DOI: 10.3389/fimmu.2024.1424925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Recently, several bispecific antibodies (BsAbs) have been approved for the treatment of relapsed multiple myeloma (MM) after early phase trials in heavily pre-treated patients demonstrated high response rates and impressive progression-free survival with monotherapy. These BsAbs provide crucial treatment options for relapsed patients and challenging decisions for clinicians. Evidence on the optimal patient population, treatment sequence, and duration of these therapeutics is unknown and subject to active investigation. While rates of cytokine release syndrome and neurotoxicity appear to be lower with BsAbs than with CAR T-cells, morbidity from infection is high and novel pathways of treatment resistance arise from the longitudinal selection pressure of chronic BsAb therapy. Lastly, a wealth of novel T-cell engagers with unique antibody-structures and antigenic targets are under active investigation with promising early outcome data. In this review, we examine the mechanism of action, therapeutic targets, combinational approaches, sequencing and mechanisms of disease relapse for BsAbs in MM.
Collapse
Affiliation(s)
- Matthew Rees
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, St Vincent’s Hospital Melbourne,
Melbourne, VIC, Australia
| | - Nadine Abdallah
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Binoy Yohannan
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
5
|
Sankaran PK, Poskute R, Dewis L, Watanabe Y, Wong V, Fernandez LP, Shannon R, Wong L, Shrubsall R, Carman L, Holt A, Lepore G, Mishra R, Sewell L, Gothard M, Cheeks M, Lindo V. Comprehensive Stress Stability Studies Reveal the Prominent Stability of the Liquid-Formulated Biotherapeutic Asymmetric Monovalent Bispecific IgG1 Monoclonal Antibody Format. J Pharm Sci 2024; 113:2101-2113. [PMID: 38705464 DOI: 10.1016/j.xphs.2024.04.029] [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: 01/17/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/07/2024]
Abstract
The developed asymmetric monovalent bispecific IgG1 or Duet monoclonal antibody (Duet mAb) has two distinct fragment antigen-binding region (Fab) subunits that target two different epitope specificities sequentially or simultaneously. The design features include unique engineered disulfide bridges, knob-into-hole mutations, and kappa and lambda chains to produce Duet mAbs. These make it structurally and functionally complex, so one expects challenging developability linked to instability, degradation of products and pathways, and limited reports available. Here, we have treated the product with different sources of extreme stress over a lengthy period, including varying heat, pH, photo stress, chemical oxidative stress, accelerated stress in physiological conditions, and forced glycation conditions. The effects of different stress conditions on the product were assessed using various analytical characterization tools to measure product-related substances, post-translational modifications (PTMs), structural integrity, higher-order disulfide linkages, and biological activity. The results revealed degradation products and pathways of Duet mAb. A moderate increase in size, charge, and hydrophobic variants, PTMs, including deamidation, oxidation, isomerization, and glycation were observed, with most conditions exhibiting biological activity. In addition, the characterization of fractionated charge variants, including deamidated species, showed satisfactory biological activity. This study demonstrated the prominent stability of the Duet mAb format comparable to most marketed mAbs.
Collapse
Affiliation(s)
| | - Ryte Poskute
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lydia Dewis
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Yasunori Watanabe
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vanessa Wong
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Richard Shannon
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lisa Wong
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Rebecca Shrubsall
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lee Carman
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander Holt
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Giordana Lepore
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Rahul Mishra
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Laura Sewell
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Matt Gothard
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Matthew Cheeks
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Viv Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
6
|
Shatz-Binder W, Azumaya CM, Leonard B, Vuong I, Sudhamsu J, Rohou A, Liu P, Sandoval W, Bol K, Izadi S, Holder PG, Blanchette C, Perozzo R, Kelley RF, Kalia Y. Adapting Ferritin, a Naturally Occurring Protein Cage, to Modulate Intrinsic Agonism of OX40. Bioconjug Chem 2024; 35:593-603. [PMID: 38592684 PMCID: PMC11099885 DOI: 10.1021/acs.bioconjchem.4c00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024]
Abstract
Ferritin is a multivalent, self-assembling protein scaffold found in most human cell types, in addition to being present in invertebrates, higher plants, fungi, and bacteria, that offers an attractive alternative to polymer-based drug delivery systems (DDS). In this study, the utility of the ferritin cage as a DDS was demonstrated within the context of T cell agonism for tumor killing. Members of the tumor necrosis factor receptor superfamily (TNFRSF) are attractive targets for the development of anticancer therapeutics. These receptors are endogenously activated by trimeric ligands that occur in transmembrane or soluble forms, and oligomerization and cell-surface anchoring have been shown to be essential aspects of the targeted agonism of this receptor class. Here, we demonstrated that the ferritin cage could be easily tailored for multivalent display of anti-OX40 antibody fragments on its surface and determined that these arrays are capable of pathway activation through cell-surface clustering. Together, these results confirm the utility, versatility, and developability of ferritin as a DDS.
Collapse
Affiliation(s)
- Whitney Shatz-Binder
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Caleigh M. Azumaya
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Brandon Leonard
- Antibody
Engineering, Genentech Inc., South San Francisco, California 94080, United States
| | - Ivan Vuong
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Pritzker
School of Molecular Engineering, University
of Chicago, 5640 S Ellis Ave, Chicago, Illinois 60637, United States
| | - Jawahar Sudhamsu
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Alexis Rohou
- Structural
Biology, Genentech Inc., South San Francisco, California 94080, United States
| | - Peter Liu
- Microchemistry,
Proteomics and Lipidomics, Genentech Inc., South San Francisco, California 94080, United States
| | - Wendy Sandoval
- Microchemistry,
Proteomics and Lipidomics, Genentech Inc., South San Francisco, California 94080, United States
| | - Karenna Bol
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
- Business
and Program Management, Genentech Inc., South San Francisco, California 94080, United States
| | - Saeed Izadi
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Patrick G. Holder
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Craig Blanchette
- Protein
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Remo Perozzo
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| | - Robert F. Kelley
- Pharmaceutical
Chemistry, Genentech Inc., South San Francisco, California 94080, United States
| | - Yogeshvar Kalia
- Pharmaceutical
Sciences, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
7
|
Poskute R, Sankaran PK, Sewell L, Lepore G, Shrubsall R, Dewis L, Watanabe Y, Wong V, Pascual Fernandez L, Mishra R, Holt A, Sou S, Harris C, Moreno Rodriguez C, Cankorur-Cetinkaya A, Smith J, Lonska N, Powell A, Cui T, Cheeks M, Lindo V. Identification and quantification of chain-pairing variants or mispaired species of asymmetric monovalent bispecific IgG1 monoclonal antibody format using reverse-phase polyphenyl chromatography coupled electrospray ionization mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1237:124085. [PMID: 38513430 DOI: 10.1016/j.jchromb.2024.124085] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/23/2024]
Abstract
Developing a knob-into-hole asymmetric bispecific IgG1 monoclonal antibody (mAb) poses manufacturing challenges due to the expression of chain pairing variants, also called mispaired species, in the desired product. The incorrect pairing of light and heavy chains could result in heterogeneous mispaired species of homodimers, heterodimers, light chain swapping, and low molecular weight species (LMWS). Standard chromatography, capillary electrophoretic, or spectroscopic methods poorly resolve these from the main variants. Here, we report a highly sensitive reverse-phase polyphenyl ultra-high-performance liquid chromatography (RP-UHPLC) method to accurately measure mispaired species of Duet mAb format, an asymmetric IgG1 bispecific mAb, for both process development and quality control analytical tests. Coupled with electrospray ionization mass spectrometry (ESI-MS), it enabled direct online characterization of mispaired species. This single direct assay detected diverse mispaired IgG-like species and LMWS. The method resolved eight disulfide bonds dissociated LMWS and three mispaired LMWS. It also resolved three different types of IgG-like mispaired species, including two homodimers and one heterodimer. The characterization and quantification simultaneously enabled the cell line selection that produces a lesser heterogeneity and lower levels of mispaired species with the desired correctly paired product. The biological activity assessment of samples with increased levels of these species quantified by the method exhibited a linear decline in potency with increasing levels of mispaired species in the desired product. We also demonstrated the utility of the technique for testing in-process intermediate materials to determine and assess downstream purification process capability in removing diverse mispaired IgG-like species and LMWS to a certain level during the downstream purification process. Our investigation demonstrates that adopting this method was vital in developing asymmetric bispecific mAb from the initial stage of cell line development to manufacturing process development. Therefore, this tool could be used in the control strategy to monitor and control mispaired species during manufacturing, thus improving the quality control of the final product.
Collapse
Affiliation(s)
- Ryte Poskute
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Laura Sewell
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Giordana Lepore
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Rebecca Shrubsall
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Lydia Dewis
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Yasunori Watanabe
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Vanessa Wong
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | | | - Rahul Mishra
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Alexander Holt
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Susie Sou
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Claire Harris
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Cristina Moreno Rodriguez
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Ayca Cankorur-Cetinkaya
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Smith
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Nikola Lonska
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Adam Powell
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Tingting Cui
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Matthew Cheeks
- Cell Culture & Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK
| | - Viv Lindo
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
8
|
Shinde SH, Sandeep, Pande AH. Polyvalency: an emerging trend in the development of clinical antibodies. Drug Discov Today 2024; 29:103846. [PMID: 38029835 DOI: 10.1016/j.drudis.2023.103846] [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: 05/02/2023] [Revised: 10/25/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
Medicine has benefited greatly from the development of monoclonal antibody (mAb) technology. First-generation mAbs have seen significant success in the treatment of major diseases, such as autoimmune, inflammation, cancer, infectious, and cardiovascular diseases. Developing next-generation antibodies with improved potency, safety, and non-natural characteristics is a booming field of mAb research. In this review, we discuss the significance of polyvalency and polyvalent antibodies, as well as important findings from preclinical studies and clinical trials involving polyvalent antibodies. We then review the role of tumor necrosis factor-alpha (TNF-α) in inflammatory diseases and the need for polyvalent anti-TNF-α antibodies.
Collapse
Affiliation(s)
- Suraj H Shinde
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Sandeep
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali 160062, Punjab, India.
| |
Collapse
|
9
|
Kuwahara A, Nazuka M, Kuroki Y, Ito K, Watanabe S, Kumagai I, Asano R. Functional integration of protein A binding ability to antibody fragments for convenient and tag-free purification. Bioengineered 2023; 14:2259093. [PMID: 37732741 PMCID: PMC10515673 DOI: 10.1080/21655979.2023.2259093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Although the development of small therapeutic antibodies is important, the affinity tags used for their purification often result in heterogeneous production and immunogenicity. In this study, we integrated Staphylococcus aureus protein A (SpA) binding ability into antibody fragments for convenient and tag-free purification. SpA affinity chromatography is used as a global standard purification method for conventional antibodies owing to its high binding affinity to the Fc region. SpA also has a binding affinity for some variable heavy domains (VH) classified in the VH3 subfamily. Through mutagenesis based on alignment and structural modeling results using the SpA-VH3 cocrystal structure, we integrated the SpA-binding ability into the anti-CD3 single-chain Fv. Furthermore, we applied this mutagenesis approach to more complicated small bispecific antibodies and successfully purified the antibodies using SpA affinity chromatography. The antibodies retained their biological function after purification. Integration of SpA-binding ability into conventional antibody fragments simplifies the purification and monitoring of the production processes and, thus, is an ideal strategy for accelerating the development of small therapeutic antibodies. Furthermore, because of its immunoactivity, the anti-CD3 variable region with SpA-binding ability is an effective building block for developing engineered cancer therapeutic antibodies without the Fc region.
Collapse
Affiliation(s)
- Atsushi Kuwahara
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Misae Nazuka
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Yuri Kuroki
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kohei Ito
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
10
|
Maejima A, Suzuki S, Makabe K, Kumagai I, Asano R. Incorporation of a repeated polypeptide sequence in therapeutic antibodies as a universal masking procedure: A case study of T cell-engaging bispecific antibodies. N Biotechnol 2023; 77:80-89. [PMID: 37467927 DOI: 10.1016/j.nbt.2023.07.004] [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: 03/13/2023] [Revised: 06/06/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023]
Abstract
Prodrug design is a promising approach for reducing the off-target effects of therapeutic antibodies, particularly bispecific antibodies (bsAbs) that recruit T cells for activation; this design uses masking sequences that inhibit antibody binding until they reach the tumor microenvironment, where they are removed. In this study, we propose PAS, a polypeptide sequence composed of repeated Pro, Ala, and Ser residues, as a universal masking sequence. PAS has no specificity, but can inhibit antibody binding through steric hindrance caused by its large fluid dynamic radius and disordered structure; additionally, its length can be adjusted. We fused PAS to the N-terminus of an anti-CD3 single-chain variable fragment (scFv) and a bsAb, that targets both the epidermal growth factor receptor and CD3, via a recognition sequence cleaved by cancer-related proteases. PAS integration inhibited anti-CD3 scFv binding with higher efficacy than the epitope sequence, and the extent of inhibition was proportional to the length of the PAS sequence. For masked bsAbs, T cell-binding ability, cancer growth inhibition effects, and T cell activation effects were also reduced depending on the length of PAS and were fully restored upon removing PAS sequences using protease. The masking procedure using PAS was successfully applied to another scFv. The provision to adjust the masking effects of PAS by tuning its length, makes PAS fusion a valuable tool for the universal design of prodrug antibodies.
Collapse
Affiliation(s)
- Atsushi Maejima
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Saori Suzuki
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Koki Makabe
- Graduate School of Science and Engineering, Faculty of Engineering, Yamagata University, 4-3-16 Jonan, Yonezawa 992-8510, Japan
| | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan; Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 3-8-1, Harumi-cho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
11
|
Porębska N, Ciura K, Chorążewska A, Zakrzewska M, Otlewski J, Opaliński Ł. Multivalent protein-drug conjugates - An emerging strategy for the upgraded precision and efficiency of drug delivery to cancer cells. Biotechnol Adv 2023; 67:108213. [PMID: 37453463 DOI: 10.1016/j.biotechadv.2023.108213] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/20/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
With almost 20 million new cases per year, cancer constitutes one of the most important challenges for public health systems. Unlike traditional chemotherapy, targeted anti-cancer strategies employ sophisticated therapeutics to precisely identify and attack cancer cells, limiting the impact of drugs on healthy cells and thereby minimizing the unwanted side effects of therapy. Protein drug conjugates (PDCs) are a rapidly growing group of targeted therapeutics, composed of a cancer-recognition factor covalently coupled to a cytotoxic drug. Several PDCs, mainly in the form of antibody-drug conjugates (ADCs) that employ monoclonal antibodies as cancer-recognition molecules, are used in the clinic and many PDCs are currently in clinical trials. Highly selective, strong and stable interaction of the PDC with the tumor marker, combined with efficient, rapid endocytosis of the receptor/PDC complex and its subsequent effective delivery to lysosomes, is critical for the efficacy of targeted cancer therapy with PDCs. However, the bivalent architecture of contemporary clinical PDCs is not optimal for tumor receptor recognition or PDCs internalization. In this review, we focus on multivalent PDCs, which represent a rapidly evolving and highly promising therapeutics that overcome most of the limitations of current bivalent PDCs, enhancing the precision and efficiency of drug delivery to cancer cells. We present an expanding set of protein scaffolds used to generate multivalent PDCs that, in addition to folding into well-defined multivalent molecular structures, enable site-specific conjugation of the cytotoxic drug to ensure PDC homogeneity. We provide an overview of the architectures of multivalent PDCs developed to date, emphasizing their efficacy in the targeted treatment of various cancers.
Collapse
Affiliation(s)
- Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Krzysztof Ciura
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Aleksandra Chorążewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| |
Collapse
|
12
|
Sun Z, Gu C, Wang X, Shang A, Quan W, Wu J, Ji P, Yao Y, Liu W, Li D. A novel bivalent anti-c-MET/PD-1 bispecific antibody exhibits potent cytotoxicity against c-MET/PD-L1-positive colorectal cancer. Invest New Drugs 2023; 41:737-750. [PMID: 37646958 DOI: 10.1007/s10637-023-01381-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/04/2023] [Indexed: 09/01/2023]
Abstract
Previously, we generated a novel bispecific antibody (BsAb) simultaneously targeting both c-MET and PD-1 (PDCD1), which can bridge T cells and c-MET positive tumor cells. However, the specific mechanisms and antitumor activities of the BsAb against c-MET/PD-L1 (CD274) positive colorectal cancer (CRC) is not completely understood. In this study, in addition to the tumor intrinsic mechanism investigation with molecular biology assay in vitro, a humanized mouse model was used to evaluate antitumor activity of the BsAb in vivo. The BsAb could inhibit c-MET/PD-L1+ CRC cell migration and show strong antitumor activity against HCT116 tumors in mice, potentially by inducing the degradation of c-MET protein in a dose and time-dependent manner. The BsAb could suppress the phosphorylation of c-MET downstream proteins GRB2-associated-binding protein 1 (Gab1) and focal adhesion kinase (FAK). Considering the tumor extrinsic mechanism, the BsAb may promote phagocytosis of macrophage. Furthermore, the level of plasma exosomal-c-MET/PD-L1 is able to distinguish CRC patients from healthy controls. In summary, the BsAb exhibited potent anti-tumor activities by two distinguished mechanisms: inhibition of c-MET signal transduction and promotion of macrophage-mediated phagocytosis. Our BsAb may provide a novel therapeutic agent for patients with c-MET/PD-L1+ CRC, and the status of exosomal-c-MET/PD-L1 can serve as a biomarker to predict responsiveness to treatment of our BsAb.
Collapse
Affiliation(s)
- Z Sun
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - C Gu
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - X Wang
- Department of Pharmacy, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - A Shang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Department of Laboratory Medicine, The Second People's Hospital of Lianyungang, Lianyungang, 222006, China
| | - W Quan
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - J Wu
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - P Ji
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Y Yao
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - W Liu
- Department of General Surgery, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - D Li
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
13
|
Sun Y, Yu X, Wang X, Yuan K, Wang G, Hu L, Zhang G, Pei W, Wang L, Sun C, Yang P. Bispecific antibodies in cancer therapy: Target selection and regulatory requirements. Acta Pharm Sin B 2023; 13:3583-3597. [PMID: 37719370 PMCID: PMC10501874 DOI: 10.1016/j.apsb.2023.05.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
In recent years, the development of bispecific antibodies (bsAbs) has been rapid, with many new structures and target combinations being created. The boom in bsAbs has led to the successive issuance of industry guidance for their development in the US and China. However, there is a high degree of similarity in target selection, which could affect the development of diversity in bsAbs. This review presents a classification of various bsAbs for cancer therapy based on structure and target selection and examines the advantages of bsAbs over monoclonal antibodies (mAbs). Through database research, we have identified the preferences of available bsAbs combinations, suggesting rational target selection options and warning of potential wastage of medical resources. We have also compared the US and Chinese guidelines for bsAbs in order to provide a reference for their development.
Collapse
Affiliation(s)
- Yanze Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xinmiao Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Gefei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Guoyu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Wenli Pei
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
14
|
Meetze K, Mehta NK, Li B, Michaelson JS, Baeuerle PA. CLN-978, a novel half-life extended CD19/CD3/HSA-specific T cell-engaging antibody construct with potent activity against B-cell malignancies with low CD19 expression. J Immunother Cancer 2023; 11:e007398. [PMID: 37586770 PMCID: PMC10432633 DOI: 10.1136/jitc-2023-007398] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Despite significant progress in the development of T cell-engaging therapies for various B-cell malignancies, a high medical need remains for the refractory disease setting, often characterized by suboptimal target levels. METHODS To address this issue, we have developed a 65-kDa multispecific antibody construct, CLN-978, with affinities tuned to optimize the killing of low-CD19 expressing tumor cells. CLN-978 bound to CD19 on B cells with picomolar affinity, and to CD3ε on T cells with nanomolar affinity. A serum albumin binding domain was incorporated to extend serum half-life. In this setting, we biophysically characterize and report the activities of CLN-978 in cell co-culture assays, multiple mouse models and non-human primates. RESULTS Human T cells redirected by CLN-978 could eliminate target cells expressing less than 300 copies of CD19 on their surface. The half-life extension and high affinity for CD19 led to significant antitumor activity in murine lymphoma models at very low doses of CLN-978. In primates, we observed a long serum half-life, deep and sustained depletion of normal B cells, and remarkable tolerability, in particular, reduced cytokine release when CLN-978 was administered subcutaneously. CONCLUSIONS CLN-978 warrants further exploration. An ongoing clinical phase 1 trial is investigating safety, pharmacokinetics, pharmacodynamics, and the initial therapeutic potential of subcutaneously administered CLN-978 in patients with non-Hodgkin's lymphoma.
Collapse
Affiliation(s)
| | | | - Bochong Li
- Cullinan Oncology Inc, Cambridge, Massachusetts, USA
| | | | - Patrick A Baeuerle
- Cullinan Oncology Inc, Cambridge, Massachusetts, USA
- Institute of Immunology, Ludwig-Maximilians-Universitat Munchen, Planegg, Germany
| |
Collapse
|
15
|
Segaliny AI, Jayaraman J, Chen X, Chong J, Luxon R, Fung A, Fu Q, Jiang X, Rivera R, Ma X, Ren C, Zimak J, Hedde PN, Shang Y, Wu G, Zhao W. A high throughput bispecific antibody discovery pipeline. Commun Biol 2023; 6:380. [PMID: 37029216 PMCID: PMC10082157 DOI: 10.1038/s42003-023-04746-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
Bispecific antibodies (BsAbs) represent an emerging class of immunotherapy, but inefficiency in the current discovery has limited their broad clinical availability. Here we report a high throughput, agnostic, single-cell-based functional screening pipeline, comprising molecular and cell engineering for efficient generation of BsAb library cells, followed by functional interrogation at the single-cell level to identify and sort positive clones and downstream sequence identification and functionality characterization. Using a CD19xCD3 bispecific T cell engager (BiTE) as a model, we demonstrate that our single-cell platform possesses a high throughput screening efficiency of up to one and a half million variant library cells per run and can isolate rare functional clones at a low abundance of 0.008%. Using a complex CD19xCD3 BiTE-expressing cell library with approximately 22,300 unique variants comprising combinatorially varied scFvs, connecting linkers and VL/VH orientations, we have identified 98 unique clones, including extremely rare ones (~ 0.001% abundance). We also discovered BiTEs that exhibit novel properties and insights to design variable preferences for functionality. We expect our single-cell platform to not only increase the discovery efficiency of new immunotherapeutics, but also enable identifying generalizable design principles based on an in-depth understanding of the inter-relationships between sequence, structure, and function.
Collapse
Affiliation(s)
| | - Jayapriya Jayaraman
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Xiaoming Chen
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | | | - Ryan Luxon
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - Audrey Fung
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - Qiwei Fu
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - Xianzhi Jiang
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | | | - Xiaoya Ma
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - Ci Ren
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - Jan Zimak
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Per Niklas Hedde
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA
| | - Yonglei Shang
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA
| | - George Wu
- Amberstone Biosciences, Inc., Irvine, CA, 92618, USA.
| | - Weian Zhao
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, 92697, USA.
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, 92697, USA.
- Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, 92697, USA.
- Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, 92697, USA.
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, 92697, USA.
- Institute for Immunology, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
16
|
Rubio-Pérez L, Lázaro-Gorines R, Harwood SL, Compte M, Navarro R, Tapia-Galisteo A, Bonet J, Blanco B, Lykkemark S, Ramírez-Fernández Á, Ferreras-Gutiérrez M, Domínguez-Alonso C, Díez-Alonso L, Segura-Tudela A, Hangiu O, Erce-Llamazares A, Blanco FJ, Santos C, Rodríguez-Peralto JL, Sanz L, Álvarez-Vallina L. A PD-L1/EGFR bispecific antibody combines immune checkpoint blockade and direct anti-cancer action for an enhanced anti-tumor response. Oncoimmunology 2023; 12:2205336. [PMID: 37114242 PMCID: PMC10128431 DOI: 10.1080/2162402x.2023.2205336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) with antibodies has shown durable clinical responses in a wide range of cancer types, but the overall response rate is still limited. Other effective therapeutic modalities to increase the ICB response rates are urgently needed. New bispecific antibody (bsAb) formats combining the ICB effect and a direct action on cancer cells could improve the efficacy of current immunotherapies. Here, we report the development of a PD-L1/EGFR symmetric bsAb by fusing a dual-targeting tandem trimmer body with the human IgG1 hinge and Fc regions. The bsAb was characterized in vitro and the antitumor efficacy was evaluated in humanized mice bearing xenografts of aggressive triple-negative breast cancer and lung cancer. The IgG-like hexavalent bsAb, designated IgTT-1E, was able to simultaneously bind both EGFR and PD-L1 antigens, inhibit EGF-mediated proliferation, effectively block PD-1/PD-L1 interaction, and induce strong antigen-specific antibody-dependent cellular cytotoxicity activity in vitro. Potent therapeutic efficacies of IgTT-1E in two different humanized mouse models were observed, where tumor growth control was associated with a significantly increased proportion of CD8+ T cells. These results support the development of IgTT-1E for the treatment of EGFR+ cancers.
Collapse
Affiliation(s)
- Laura Rubio-Pérez
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - Rodrigo Lázaro-Gorines
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Seandean L. Harwood
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Rocío Navarro
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Antonio Tapia-Galisteo
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Jaume Bonet
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Simon Lykkemark
- Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus C, Denmark
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alejandro Segura-Tudela
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Oana Hangiu
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Ainhoa Erce-Llamazares
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Department of Antibody Engineering, Leadartis SL, QUBE Technology Park, Madrid, Spain
| | - Francisco J. Blanco
- Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid, Spain
| | - Cruz Santos
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - José L. Rodríguez-Peralto
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Pathology, Universidad Complutense, Madrid, Spain
- Cutaneous Oncology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Laura Sanz
- Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Chair for Immunology UFV/Merck, Universidad Francisco de Vitoria (UFV), Madrid, Spain
- CONTACT Luis Álvarez-Vallina Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Avda. Cordoba s/n, Madrid28041, Spain
| |
Collapse
|
17
|
Huang C, Huang J, Zhu S, Tang T, Chen Y, Qian F. Multivalent nanobodies with rationally optimized linker and valency for intravitreal VEGF neutralization. Chem Eng Sci 2023. [DOI: 10.1016/j.ces.2023.118521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
18
|
Bispecific Antibody Format and the Organization of Immunological Synapses in T Cell-Redirecting Strategies for Cancer Immunotherapy. Pharmaceutics 2022; 15:pharmaceutics15010132. [PMID: 36678761 PMCID: PMC9863865 DOI: 10.3390/pharmaceutics15010132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023] Open
Abstract
T cell-redirecting strategies have emerged as effective cancer immunotherapy approaches. Bispecific antibodies (bsAbs) are designed to specifically recruit T cells to the tumor microenvironment and induce the assembly of the immunological synapse (IS) between T cells and cancer cells or antigen-presenting cells. The way that the quality of the IS might predict the effectiveness of T cell-redirecting strategies, including those mediated by bsAbs or by chimeric antigen receptors (CAR)-T cells, is currently under discussion. Here we review the organization of the canonical IS assembled during natural antigenic stimulation through the T cell receptor (TCR) and to what extent different bsAbs induce T cell activation, canonical IS organization, and effector function. Then, we discuss how the biochemical parameters of different formats of bsAbs affect the effectivity of generating an antigen-induced canonical IS. Finally, the quality of the IS assembled by bsAbs and monoclonal antibodies or CAR-T cells are compared, and strategies to improve bsAb-mediated T cell-redirecting strategies are discussed.
Collapse
|
19
|
Winegar PH, Figg CA, Teplensky MH, Ramani N, Mirkin CA. Modular Nucleic Acid Scaffolds for Synthesizing Monodisperse and Sequence-Encoded Antibody Oligomers. Chem 2022; 8:3018-3030. [PMID: 36405374 PMCID: PMC9674055 DOI: 10.1016/j.chempr.2022.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Synthesizing protein oligomers that contain exact numbers of multiple different proteins in defined architectures is challenging. DNA-DNA interactions can be used to program protein assembly into oligomers; however, existing methods require changes to DNA design to achieve different numbers and oligomeric sequences of proteins. Herein, we develop a modular DNA scaffold that uses only six synthetic oligonucleotides to organize proteins into defined oligomers. As a proof-of-concept, model proteins (antibodies) are oligomerized into dimers and trimers, where antibody function is retained. Illustrating the modularity of this technique, dimer and trimer building blocks are then assembled into pentamers containing three different antibodies in an exact stoichiometry and oligomeric sequence. In sum, this report describes a generalizable method for organizing proteins into monodisperse, sequence-encoded oligomers using DNA. This advance will enable studies into how oligomeric protein sequences affect material properties in areas spanning pharmaceutical development, cascade catalysis, synthetic photosynthesis, and membrane transport.
Collapse
Affiliation(s)
- Peter H. Winegar
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- These authors contributed equally
| | - C. Adrian Figg
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- These authors contributed equally
| | - Michelle H. Teplensky
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Namrata Ramani
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- Department of Materials Science and Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
| | - Chad A. Mirkin
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- Department of Materials Science and Engineering, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA
- Lead contact
| |
Collapse
|
20
|
Development of a self-assembled heptameric nanobody/streptavidin-binding peptide fusion for ultrasensitive detection of serum biomarkers. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Mocquot P, Mossazadeh Y, Lapierre L, Pineau F, Despas F. The pharmacology of blinatumomab: state of the art on pharmacodynamics, pharmacokinetics, adverse drug reactions and evaluation in clinical trials. J Clin Pharm Ther 2022; 47:1337-1351. [PMID: 35906791 PMCID: PMC9796714 DOI: 10.1111/jcpt.13741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Bispecific drugs (BDs) belong to the family of immunotherapies along with checkpoint inhibitors and CAR-T cells. In the field of oncology, BDs are designed to simultaneously bind a tumour antigen on the one side and an antigen present on the surface of effector cells on the other. This review summarizes the information available to date on the first marketed BiTE-format bispecific antibody, blinatumomab BLINCYTO® in acute lymphoblastic leukaemia. METHODS A literature search was conducted in the PubMed database by including studies published in English using the term blinatumomab. Furthermore, bibliographies of selected references were also evaluated for relevant articles. Clinical trial (CT) data were retrieved from clinicaltrials.gov (ongoing trials, adverse events [AEs]) and global pharmacovigilance data were retrieved from VigiBase®. RESULTS AND DISCUSSION Blinatumomab is a fusion protein which consists of two single-chain variable fragments arranged in tandem: the first binds the CD19 surface antigen of all B cells and the second targets the CD3 antigen of T cells. Binding of blinatumomab to B and T cells induces apoptosis of B cells after secretion of granzymes and perforins by T cells. T-cell activation results in secretion of pro-inflammatory cytokines and upregulation of activation markers and adhesion molecules on the surface of T cells. The major CTs that led to an indication show increased overall survival with blinatumomab with better efficacy in patients in haematological remission with minimal residual disease ≥10-3 . The major AEs are cytokine release syndrome, neurotoxicity and hypogammaglobulinemia. The three most frequent system organ classes in CTs are haematological, gastrointestinal and general disorders. These results are also found in VigiBase® but neurological disorders and infections appear more frequently in real life. WHAT IS NEW AND CONCLUSION This review summarizes the current knowledge of blinatumomab in the literature. The subject of many CTs is to improve the route of administration and expand the indications for treatment.
Collapse
Affiliation(s)
- Pauline Mocquot
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Yasmine Mossazadeh
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance
| | - Léopoldine Lapierre
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fanny Pineau
- Département d'Hématologie et de Médecine InterneInstitut Universitaire du Cancer‐Oncopole, CHU de ToulouseToulouseFrance
| | - Fabien Despas
- Département de Pharmacologie Médicale, CHU de ToulouseUniversité Toulouse III ‐ Paul SabatierToulouseFrance,Université Toulouse III ‐ Paul SabatierToulouseFrance,INSERM CIC1436 CIC ToulouseFrance
| |
Collapse
|
22
|
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: 40] [Impact Index Per Article: 13.3] [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
|
23
|
Ordóñez-Reyes C, Garcia-Robledo JE, Chamorro DF, Mosquera A, Sussmann L, Ruiz-Patiño A, Arrieta O, Zatarain-Barrón L, Rojas L, Russo A, de Miguel-Perez D, Rolfo C, Cardona AF. Bispecific Antibodies in Cancer Immunotherapy: A Novel Response to an Old Question. Pharmaceutics 2022; 14:pharmaceutics14061243. [PMID: 35745815 PMCID: PMC9229626 DOI: 10.3390/pharmaceutics14061243] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/14/2023] Open
Abstract
Immunotherapy has redefined the treatment of cancer patients and it is constantly generating new advances and approaches. Among the multiple options of immunotherapy, bispecific antibodies (bsAbs) represent a novel thoughtful approach. These drugs integrate the action of the immune system in a strategy to redirect the activation of innate and adaptive immunity toward specific antigens and specific tumor locations. Here we discussed some basic aspects of the design and function of bsAbs, their main challenges and the state-of-the-art of these molecules in the treatment of hematological and solid malignancies and future perspectives.
Collapse
Affiliation(s)
- Camila Ordóñez-Reyes
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Juan Esteban Garcia-Robledo
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ 85054, USA
| | - Diego F. Chamorro
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Andrés Mosquera
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | - Liliana Sussmann
- Department of Neurology, Fundación Universitaria de Ciencias de la Salud, Bogotá 111221, Colombia;
| | - Alejandro Ruiz-Patiño
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Lucia Zatarain-Barrón
- Thoracic Oncology Unit and Personalized Oncology Laboratory, National Cancer Institute (INCan), Mexico City 14080, Mexico; (O.A.); (L.Z.-B.)
| | - Leonardo Rojas
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
| | | | - Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (D.d.M.-P.); (C.R.)
| | - Andrés F. Cardona
- Foundation for Clinical and Applied Cancer Research—FICMAC, Bogotá 110111, Colombia; (C.O.-R.); (J.E.G.-R.); (D.F.C.); (A.M.); (A.R.-P.); (L.R.)
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad el Bosque, Bogotá 110121, Colombia
- Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC), Bogotá 110131, Colombia
- Correspondence: ; Tel.: +57-(1)-6190052; Fax: +57-(1)-6190053
| |
Collapse
|
24
|
Otsuka Y, Sasaki K, Suyotha W, Furusawa H, Miyazawa K, Konno H, Yano S. Construction of a fusion protein consisting of α-1,3-glucan-binding domains and tetrameric red fluorescent protein, which is involved in the aggregation of α-1,3-glucan and inhibition of fungal biofilm formation. J Biosci Bioeng 2022; 133:524-532. [PMID: 35314116 DOI: 10.1016/j.jbiosc.2022.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 11/24/2022]
Abstract
Agl-KA, an α-1,3-glucan-hydrolyzing enzyme from Bacillus circulans KA-304, has three α-1,3-glucan-binding domains DS1, CB6, and DS2 (DCD). While their individual binding activities toward insoluble α-1,3-glucan and fungal cell-wall are weak, the three domains in combination bind strongly to the α-1,3-glucan and the cell-wall. In this study, we constructed DCD-tetraRFP by fusing DCD with DsRed-Express2, a tetrameric red fluorescent protein. DCD-tetraRFP forms a tetramer in an aqueous solution and contains twelve substrate-binding domains in one complex. We also constructed DCD-monoGFP by fusing DCD with AcGFP1, a monomeric green fluorescent protein. The molecular weight of DCD-tetraRFP and DCD-monoGFP were compared. The results of gel filtration chromatography and dynamic light scattering indicated that DCD-tetraRFP was larger than DCD-monoGFP, suggesting that DCD-tetraRFP had a tetrameric structure. In addition, DCD-tetraRFP bound to insoluble α-1,3-glucan strongly, and the amount of DCD-tetraRFP binding to 0.01% α-1,3-glucan was about twice of DCD-monoGFP. The Kd values of DCD-tetraRFP (measurements per subunit) and DCD-monoGFP were 0.16 and 0.84 μM, respectively. Adding DCD-tetraRFP to a suspension of α-1,3-glucan caused glucan aggregation; however, adding DCD-monoGFP did not. These data suggested that DCD-tetraRFP had four DCDs sterically arranged in different directions so that DCD-tetraRFP cross-linked with the substrate, causing aggregation. Lastly, the aggregates of DCD-tetraRFP and α-1,3-glucan captured Aspergillus oryzae conidia and decreased their biofilm formation by 80% in a 24-well dish.
Collapse
Affiliation(s)
- Yuitsu Otsuka
- Graduate School of Sciences and Engineering, Yamagata University, Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Kai Sasaki
- Graduate School of Sciences and Engineering, Yamagata University, Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Wasana Suyotha
- Department of Industrial Biotechnology, Faculty of Agro-industry, Prince of Songkla University, Hat Yai 90112, Thailand
| | - Hiroyuki Furusawa
- Graduate School of Sciences and Engineering, Yamagata University, Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Ken Miyazawa
- Laboratory of Filamentous Mycoses, Department of Fungal Infection, National Institute of Infectious Diseases, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Hiroyuki Konno
- Graduate School of Sciences and Engineering, Yamagata University, Jonan, Yonezawa, Yamagata 992-8510, Japan
| | - Shigekazu Yano
- Graduate School of Sciences and Engineering, Yamagata University, Jonan, Yonezawa, Yamagata 992-8510, Japan.
| |
Collapse
|
25
|
Wu SY, Wu FG, Chen X. Antibody-Incorporated Nanomedicines for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109210. [PMID: 35142395 DOI: 10.1002/adma.202109210] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Antibody-based cancer therapy, one of the most significant therapeutic strategies, has achieved considerable success and progress over the past decades. Nevertheless, obstacles including limited tumor penetration, short circulation half-lives, undesired immunogenicity, and off-target side effects remain to be overcome for the antibody-based cancer treatment. Owing to the rapid development of nanotechnology, antibody-containing nanomedicines that have been extensively explored to overcome these obstacles have already demonstrated enhanced anticancer efficacy and clinical translation potential. This review intends to offer an overview of the advancements of antibody-incorporated nanoparticulate systems in cancer treatment, together with the nontrivial challenges faced by these next-generation nanomedicines. Diverse strategies of antibody immobilization, formats of antibodies, types of cancer-associated antigens, and anticancer mechanisms of antibody-containing nanomedicines are provided and discussed in this review, with an emphasis on the latest applications. The current limitations and future research directions on antibody-containing nanomedicines are also discussed from different perspectives to provide new insights into the construction of anticancer nanomedicines.
Collapse
Affiliation(s)
- Shun-Yu Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
26
|
Compte M, Sanz L, Álvarez-Vallina L. Applications of trimerbodies in cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:71-87. [PMID: 35777865 DOI: 10.1016/bs.ircmb.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Trimerbodies, with their unique structural and functional properties, are the basis of a new generation of therapeutic antibodies, which due to their small size and plasticity are ideal for the generation of novel biological protein drugs with multiple competitive advantages over conventional full-length monoclonal antibodies. Since their emergence, trimerbodies have been used in preclinical cancer diagnosis and therapy. Trimerbodies are highly adaptable molecules, as they allow target-specific modulation of T cell-mediated anti-tumor immunity to enhance preexisting responses or to generate de novo immune responses. In fact, a tumor-specific humanized 4-1BB-agonistic trimerbody has shown a rather impressive safety and efficacy profile in preclinical studies making it a realistic option for clinical development. Moreover, thanks to the avidity effect they are endowed with considerable therapeutic potential as carriers to deliver cytotoxic payloads to tumors. In addition, molecular imaging studies could benefit from some intermediate-sized trivalent trimerbodies as promising candidates for targeted therapy and tumor imaging.
Collapse
Affiliation(s)
- Marta Compte
- Department of Antibody Engineering, Leadartis S.L., Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario12 de Octubre, Madrid, Spain; Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.
| |
Collapse
|
27
|
Ramírez-Fernández Á, Aguilar-Sopeña Ó, Díez-Alonso L, Segura-Tudela A, Domínguez-Alonso C, Roda-Navarro P, Álvarez-Vallina L, Blanco B. Synapse topology and downmodulation events determine the functional outcome of anti-CD19 T cell-redirecting strategies. Oncoimmunology 2022; 11:2054106. [PMID: 35355682 PMCID: PMC8959521 DOI: 10.1080/2162402x.2022.2054106] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer immunotherapy strategies based on the endogenous secretion of T cell-redirecting bispecific antibodies by engineered T lymphocytes (STAb-T) are emerging as alternative or complementary approaches to those based on chimeric antigen receptors (CAR-T). The antitumor efficacy of bispecific anti-CD19 × anti-CD3 (CD19×CD3) T cell engager (BiTE)-secreting STAb-T cells has been demonstrated in several mouse models of B-cell acute leukemia. Here, we have investigated the spatial topology and downstream signaling of the artificial immunological synapses (IS) that are formed by CAR-T or STAb-T cells. Upon interaction with CD19-positive target cells, STAb-T cells form IS with structure and signal transduction, which more closely resemble those of physiological cognate IS, compared to IS formed by CAR-T cells expressing a second-generation CAR bearing the same CD19-single-chain variable fragment. Importantly, while CD3 is maintained at detectable levels on the surface of STAb-T cells, indicating sustained activation mediated by the secreted BiTE, the anti-CD19 CAR was rapidly downmodulated, which correlated with a more transient downstream signaling. Furthermore, CAR-T cells, but not STAb-T cells, provoke an acute loss of CD19 in target cells. Such differences might represent advantages of the STAb-T strategy over the CAR-T approach and should be carefully considered in order to develop more effective and safer treatments for hematological malignancies.
Collapse
Affiliation(s)
- Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Óscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Laura Díez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Alejandro Segura-Tudela
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain
- Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital, Universitario 12 de Octubre, Madrid, Spain
- Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV), Instituto de Salud Carlos III (RICORS, RD21/0017/0029), Madrid, Spain
| |
Collapse
|
28
|
Obeng EM, Dzuvor CKO, Danquah MK. Anti-SARS-CoV-1 and -2 nanobody engineering towards avidity-inspired therapeutics. NANO TODAY 2022; 42:101350. [PMID: 34840592 PMCID: PMC8608585 DOI: 10.1016/j.nantod.2021.101350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 05/15/2023]
Abstract
In the past two decades, the emergence of coronavirus diseases has been dire distress on both continental and global fronts and has resulted in the search for potent treatment strategies. One crucial challenge in this search is the recurrent mutations in the causative virus spike protein, which lead to viral escape issues. Among the current promising therapeutic discoveries is the use of nanobodies and nanobody-like molecules. While these nanobodies have demonstrated high-affinity interaction with the virus, the unpredictable spike mutations have warranted the need for avidity-inspired therapeutics of potent inhibitors such as nanobodies. This article discusses novel approaches for the design of anti-SARS-CoV-1 and -2 nanobodies to facilitate advanced innovations in treatment technologies. It further discusses molecular interactions and suggests multivalent protein nanotechnology and chemistry approaches to translate mere molecular affinity into avidity.
Collapse
Affiliation(s)
- Eugene M Obeng
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Christian K O Dzuvor
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Michael K Danquah
- Department of Chemical Engineering, University of Tennessee, Chattanooga 615 McCallie Ave, Chattanooga, TN 37403, United States
| |
Collapse
|
29
|
Grunert I, Heinrich K, Ernst J, Hingar M, Briguet A, Leiss M, Wuhrer M, Reusch D, Bulau P. Detailed Analytical Characterization of a Bispecific IgG1 CrossMab Antibody of the Knob-into-Hole Format Applying Various Stress Conditions Revealed Pronounced Stability. ACS OMEGA 2022; 7:3671-3679. [PMID: 35128275 PMCID: PMC8811765 DOI: 10.1021/acsomega.1c06305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 06/14/2023]
Abstract
In recent years, a variety of new antibody formats have been developed. One of these formats allows the binding of one type of antibody to two different epitopes. This can for example be achieved by introduction of the "knob-into-hole" format and a combined CrossMab approach. Due to their complexity, these bispecific antibodies are expected to result in an enhanced variety of different degradation products. Reports on the stability of these molecules are still largely lacking. To address this, a panel of stress conditions, including elevated temperature, pH, oxidizing agents, and forced glycation via glucose incubation, to identify and functionally evaluate critical quality attributes in the complementary-determining and conserved regions of a bispecific antibody was applied in this study. The exertion of various stress conditions combined with an assessment by size exclusion chromatography, ion exchange chromatography, LC-MS/MS peptide mapping, and functional evaluation by cell-based assays was adequate to identify chemical modification sites and assess the stability and integrity, as well as the functionality of a bispecific antibody. Stress conditions induced size variants and post-translational modifications, such as isomerization, deamidation, and oxidation, albeit to a modest extent. Of note, all the observed stress conditions largely maintained functionality. In summary, this study revealed the pronounced stability of IgG1 "knob-into-hole" bispecific CrossMab antibodies compared to already marketed antibody products.
Collapse
Affiliation(s)
- Ingrid Grunert
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Katrin Heinrich
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Juliane Ernst
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Michael Hingar
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Alexandre Briguet
- Pharma
Technical Development, Hoffmann-La Roche, Basel 4070, Switzerland
| | - Michael Leiss
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Manfred Wuhrer
- Center
for Proteomics and Metabolomics, Leiden
University Medical Center, Leiden 2333ZA, The Netherlands
| | - Dietmar Reusch
- Pharma
Technical Development, Roche Diagnostics
GmbH, Penzberg 82377, Germany
| | - Patrick Bulau
- Pharma
Technical Development, Hoffmann-La Roche, Basel 4070, Switzerland
| |
Collapse
|
30
|
Abstract
Antibodies are an integral part of many biological assays and biotherapeutics. However, the sources from which antibodies are derived frequently contain other contaminants which may interfere with assays or cause adverse reactions if administered in vivo. Therefore, a means of isolating these antibodies from their source at high levels of purity is critical. Affinity chromatography is currently one of the most widely applied methods for the purification of antibodies. This method relies on specific and reversible, interactions between antibody structures, or recombinant tags fused to these structures, and ligands immobilized on solid support matrices, generally within a column. Herein, common chromatographic methods applied to antibody purification are described. These include the purification of IgG, and its recombinant forms, through protein A, protein G and immobilized metal affinity chromatography.
Collapse
Affiliation(s)
- Arabelle Cassedy
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Richard O'Kennedy
- School of Biotechnology, Dublin City University, Dublin, Ireland.
- Hamad Bin Khalifa University, Doha, Qatar.
- Qatar Foundation, Doha, Qatar.
| |
Collapse
|
31
|
Qin X, Meng X, Xiong Y, Guo X, Ren Y, Wen L, Zhang Q, Zhu H, Yang Z. Initial evaluation of 99m Tc-labeled anti-CEA scFv for micro-SPECT imaging in mice with colorectal cancer. J Labelled Comp Radiopharm 2021; 65:63-70. [PMID: 34967041 DOI: 10.1002/jlcr.3960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/07/2022]
Abstract
Carcinoembryonic antigen (CEA) has emerged as an important molecular target for several neoplastic diseases, including colorectal cancer with CEA over-expression. In this study, we report the production and radiolabeling of a novel anti-CEA single-chain fragment variable (scFv-96NRT, concentration for 50% of maximal effect 46 ng/mL), and evaluation of [99m Tc]Tc-scFv-96NRT in non-invasive detection of CEA expression. [99m Tc]Tc-scFv-96NRT was synthesized by one step reduction in labeling yield of > 95% with radiochemical purity of > 98% and molar activity of 10-11 GBq/μmol. [99m Tc]Tc-scFv-96NRT showed high stability in 0.01 M phosphate buffered saline (PBS), and 5% human serum albumin (HSA). It exhibited elevated uptake in CEA over-expressing cells. Bio-distribution studies in BALB/c mice revealed that the probe was cleared from blood rapidly, and the highest retention was observed in the kidneys. The micro-single photon emission computed tomography (micro-SPECT) imaging of [99m Tc]Tc-scFv-96NRT showed a specific accumulation pattern, as blocking experiment with excess scFv-96NRT suppressed the tumor uptake. These preliminary results suggest that [99m Tc]Tc-scFv-96NRT is a potential non-invasive molecular probe for imaging tumors with CEA over-expression.
Collapse
Affiliation(s)
- Xue Qin
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yao Xiong
- Chengdu New Radiomedicinetechnology Co.,Ltd
| | - Xiaoyi Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanan Ren
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Li Wen
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qian Zhang
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Zhu
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Yang
- Guizhou University School of Medicine, Guizhou University, Guiyang, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
32
|
Van Holsbeeck K, Martins JC, Ballet S. Downsizing antibodies: Towards complementarity-determining region (CDR)-based peptide mimetics. Bioorg Chem 2021; 119:105563. [PMID: 34942468 DOI: 10.1016/j.bioorg.2021.105563] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/20/2021] [Accepted: 12/12/2021] [Indexed: 12/27/2022]
Abstract
Monoclonal antibodies emerged as an important therapeutic drug class with remarkable specificity and binding affinity. Nonetheless, these heterotetrameric immunoglobulin proteins come with high manufacturing and therapeutic costs which can take extraordinary proportions, besides other limitations such as their limited in cellulo access imposed by their molecular size (ca. 150 kDa). These drawbacks stimulated the development of downsized functional antibody fragments (ca. 15-50 kDa), together with smaller synthetic peptides (ca. 1-3 kDa) derived from the antibodies' crucial complementarity-determining regions (CDR). Despite the general lack of success in the literal translation of CDR loops in peptide mimetics, rational structure-based and computational approaches have shown their potential for obtaining functional CDR-based peptide mimetics. In this review, we describe the efforts made in the development of antibody and nanobody paratope-derived peptide mimetics with particular focus on the used design strategies, in addition to highlighting the challenges associated with their development.
Collapse
Affiliation(s)
- Kevin Van Holsbeeck
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium; NMR and Structure Analysis Unit, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium
| | - José C Martins
- NMR and Structure Analysis Unit, Ghent University, Krijgslaan 281 S4, 9000 Ghent, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
33
|
Pedroso CC, Mann VR, Zuberbühler K, Bohn MF, Yu J, Altoe V, Craik CS, Cohen BE. Immunotargeting of Nanocrystals by SpyCatcher Conjugation of Engineered Antibodies. ACS NANO 2021; 15:18374-18384. [PMID: 34694776 PMCID: PMC9035480 DOI: 10.1021/acsnano.1c07856] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Inorganic nanocrystals such as quantum dots (QDs) and upconverting nanoparticles (UCNPs) are uniquely suited for quantitative live-cell imaging and are typically functionalized with ligands to study specific receptors or cellular targets. Antibodies (Ab) are among the most useful targeting reagents owing to their high affinities and specificities, but common nanocrystal labeling methods may orient Ab incorrectly, be reversible or denaturing, or lead to Ab-NP complexes too large for some applications. Here, we show that SpyCatcher proteins, which bind and spontaneously form covalent isopeptide bonds with cognate SpyTag peptides, can conjugate engineered Ab to nanoparticle surfaces with control over stability, orientation, and stoichiometry. Compact SpyCatcher-functionalized QDs and UCNPs may be labeled with short-chain variable fragment Ab (scFv) engineered to bind urokinase-type plasminogen activator receptors (uPAR) that are overexpressed in many human cancers. Confocal imaging of anti-uPAR scFv-QD conjugates shows the antibody mediates specific binding and internalization by breast cancer cells expressing uPAR. Time-lapse imaging of photostable scFv-UCNP conjugates shows that Ab binding causes uPAR internalization with a ∼20 min half-life on the cell surface, and uPAR is internalized to endolysosomal compartments distinct from general membrane stains and without significant recycling to the cell surface. The controlled and stable conjugation of engineered Ab to NPs enables targeting of diverse receptors for live-cell study of their distribution, trafficking, and physiology.
Collapse
Affiliation(s)
- Cassio C.S. Pedroso
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Victor R. Mann
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Kathrin Zuberbühler
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, United States
| | - Markus-Frederik Bohn
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, United States
| | - Jessica Yu
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Virginia Altoe
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Charles S. Craik
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94143, United States
| | - Bruce E. Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
- Division of Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
- Corresponding Author:
| |
Collapse
|
34
|
Sanz L, Ibáñez-Pérez R, Guerrero-Ochoa P, Lacadena J, Anel A. Antibody-Based Immunotoxins for Colorectal Cancer Therapy. Biomedicines 2021; 9:1729. [PMID: 34829955 PMCID: PMC8615520 DOI: 10.3390/biomedicines9111729] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 01/21/2023] Open
Abstract
Monoclonal antibodies (mAbs) are included among the treatment options for advanced colorectal cancer (CRC). However, while these mAbs effectively target cancer cells, they may have limited clinical activity. A strategy to improve their therapeutic potential is arming them with a toxic payload. Immunotoxins (ITX) combining the cell-killing ability of a toxin with the specificity of a mAb constitute a promising strategy for CRC therapy. However, several important challenges in optimizing ITX remain, including suboptimal pharmacokinetics and especially the immunogenicity of the toxin moiety. Nonetheless, ongoing research is working to solve these limitations and expand CRC patients' therapeutic armory. In this review, we provide a comprehensive overview of targets and toxins employed in the design of ITX for CRC and highlight a wide selection of ITX tested in CRC patients as well as preclinical candidates.
Collapse
Affiliation(s)
- Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute, Hospital Universitario Puerta de Hierro, 28222 Madrid, Spain
| | - Raquel Ibáñez-Pérez
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Patricia Guerrero-Ochoa
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University, 28040 Madrid, Spain
| | - Alberto Anel
- Apoptosis, Immunity and Cancer Group, Aragón Health Research Institute (IIS-Aragón), University of Zaragoza, 50009 Zaragoza, Spain; (R.I.-P.); (P.G.-O.)
| |
Collapse
|
35
|
Blanco B, Domínguez-Alonso C, Alvarez-Vallina L. Bispecific Immunomodulatory Antibodies for Cancer Immunotherapy. Clin Cancer Res 2021; 27:5457-5464. [PMID: 34108185 PMCID: PMC9306338 DOI: 10.1158/1078-0432.ccr-20-3770] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/25/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023]
Abstract
The recent advances in the field of immuno-oncology have dramatically changed the therapeutic strategy against advanced malignancies. Bispecific antibody-based immunotherapies have gained momentum in preclinical and clinical investigations following the regulatory approval of the T cell-redirecting antibody blinatumomab. In this review, we focus on emerging and novel mechanisms of action of bispecific antibodies interacting with immune cells with at least one of their arms to regulate the activity of the immune system by redirecting and/or reactivating effector cells toward tumor cells. These molecules, here referred to as bispecific immunomodulatory antibodies, have the potential to improve clinical efficacy and safety profile and are envisioned as a second wave of cancer immunotherapies. Currently, there are more than 50 bispecific antibodies under clinical development for a range of indications, with promising signs of therapeutic activity. We also discuss two approaches for in vivo secretion, direct gene delivery, and infusion of ex vivo gene-modified cells, which may become instrumental for the clinical application of next-generation bispecific immunomodulatory antibodies.
Collapse
Affiliation(s)
- Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Carmen Domínguez-Alonso
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Luis Alvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Corresponding Author: Luis Alvarez-Vallina, Cancer Immunotherapy Unit, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain. E-mail:
| |
Collapse
|
36
|
Mahmoudi R, Dianat-Moghadam H, Poorebrahim M, Siapoush S, Poortahmasebi V, Salahlou R, Rahmati M. Recombinant immunotoxins development for HER2-based targeted cancer therapies. Cancer Cell Int 2021; 21:470. [PMID: 34488747 PMCID: PMC8422749 DOI: 10.1186/s12935-021-02182-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/26/2021] [Indexed: 01/07/2023] Open
Abstract
Understanding the molecular mechanisms of cancer biology introduces targeted therapy as a complementary method along with other conventional therapies. Recombinant immunotoxins are tumor specific antibodies that their recognizing fragment is utilized for delivering modified toxins into tumor cells. These molecules have been considered as a targeted strategy in the treatment of human cancers. HER2 tumor biomarker is a transmembrane tyrosine kinase receptor that can be used for targeted therapies in the forms of anti-HER2 monoclonal antibodies, antibody-drug conjugates and immunotoxins. There have been many studies on HER2-based immunotoxins in recent years, however, little progress has been made in the clinical field which demanded more improvements. Here, we summarized the HER2 signaling and it's targeting using immunotherapeutic agents in human cancers. Then, we specifically reviewed anti-HER2 immunotoxins, and their strengths and drawbacks to highlight their promising clinical impact.
Collapse
Affiliation(s)
- Reza Mahmoudi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Samaneh Siapoush
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Salahlou
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Woodchuck Hepatitis Virus Post-Transcriptional Regulation Element (WPRE) Promotes Anti-CD19 BiTE Expression in Expi293 Cells. IRANIAN BIOMEDICAL JOURNAL 2021; 25:275-83. [PMID: 34217158 PMCID: PMC8334396 DOI: 10.52547/ibj.25.4.275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background: Bispecific antibodies represent an important class of mAbs, with great therapeutic potentials due to their ability to target simultaneously two distinct epitopes. The generation of functional bispecific antibodies with the highest possible yields is particularly critical for the production of these compounds on industrial scales. Anti- CD3 × CD19 bsAb is a bispecific T-cell engager (BiTE) currently used for treating ALL. Herein, we have tried to optimize the expression level of this antibody in mammalian hosts. Methods: WPRE sequence was incorporated at the 3’ end of the expression cassette. This modification resulted in a notable about two-fold increase in the expression of the bsAb in the Expi293 cell line. Results & Conclusion: Follow-up flow cytometry analysis demonstrated the binding properties of the produced antibody at acceptable levels, and in vitro bioactivity assays showed that this product is potent enough for targeting and destroying CD19-positive cells. Our findings show that WPRE enhances the expression of this type of bispecific mAbs in HEK-293 family cell lines. This approach can be used in biopharma industry for the mass production of anti-CD3 × CD19 bispecific antibody.
Collapse
|
38
|
Miller A, Carr S, Rabbitts T, Ali H. Multimeric antibodies with increased valency surpassing functional affinity and potency thresholds using novel formats. MAbs 2021; 12:1752529. [PMID: 32316838 PMCID: PMC7188389 DOI: 10.1080/19420862.2020.1752529] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The success of therapeutic antibodies is largely attributed for their exquisite specificity, homogeneity, and functionality. There is, however, a need to engineer antibodies to extend and enhance their potency. One parameter is functional affinity augmentation, since antibodies matured in vivo have a natural affinity threshold. Generation of multivalent antibodies is one option capable of surpassing this affinity threshold through increased avidity. In this study, we present a novel platform consisting of an array of multivalent antibody formats, termed Quads, generated using the self-assembling tetramerization domain from p53. We demonstrate the versatility of this tetramerization domain by engineering anti-tumor necrosis factor (TNF) Quads that exhibit major increases in binding potency and in neutralizing TNF-mediated cytotoxicity compared to parental anti-TNF molecules. Further, Quads are amenable to fusion with different binding domains, allowing generation of novel multivalent monospecific and bispecific formats. Quads are thus a novel group of molecules that can be engineered to yield potential therapeutics with novel modalities and potencies.
Collapse
Affiliation(s)
- Ami Miller
- Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Stephen Carr
- Research Complex at Harwell, Rutherford Appleton Laboratory, Oxon, UK
| | - Terry Rabbitts
- Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Hanif Ali
- Quadrucept Bio Limited, Cambridge, UK
| |
Collapse
|
39
|
Divine R, Dang HV, Ueda G, Fallas JA, Vulovic I, Sheffler W, Saini S, Zhao YT, Raj IX, Morawski PA, Jennewein MF, Homad LJ, Wan YH, Tooley MR, Seeger F, Etemadi A, Fahning ML, Lazarovits J, Roederer A, Walls AC, Stewart L, Mazloomi M, King NP, Campbell DJ, McGuire AT, Stamatatos L, Ruohola-Baker H, Mathieu J, Veesler D, Baker D. Designed proteins assemble antibodies into modular nanocages. Science 2021; 372:eabd9994. [PMID: 33795432 PMCID: PMC8592034 DOI: 10.1126/science.abd9994] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/23/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Multivalent display of receptor-engaging antibodies or ligands can enhance their activity. Instead of achieving multivalency by attachment to preexisting scaffolds, here we unite form and function by the computational design of nanocages in which one structural component is an antibody or Fc-ligand fusion and the second is a designed antibody-binding homo-oligomer that drives nanocage assembly. Structures of eight nanocages determined by electron microscopy spanning dihedral, tetrahedral, octahedral, and icosahedral architectures with 2, 6, 12, and 30 antibodies per nanocage, respectively, closely match the corresponding computational models. Antibody nanocages targeting cell surface receptors enhance signaling compared with free antibodies or Fc-fusions in death receptor 5 (DR5)-mediated apoptosis, angiopoietin-1 receptor (Tie2)-mediated angiogenesis, CD40 activation, and T cell proliferation. Nanocage assembly also increases severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus neutralization by α-SARS-CoV-2 monoclonal antibodies and Fc-angiotensin-converting enzyme 2 (ACE2) fusion proteins.
Collapse
MESH Headings
- Angiopoietins/chemistry
- Angiopoietins/immunology
- Angiopoietins/metabolism
- Antibodies/chemistry
- Antibodies/immunology
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- B-Lymphocytes/immunology
- CD40 Antigens/chemistry
- CD40 Antigens/immunology
- CD40 Antigens/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Computer Simulation
- Genes, Synthetic
- Humans
- Immunoglobulin Fc Fragments/chemistry
- Lymphocyte Activation
- Models, Molecular
- Nanostructures
- Protein Binding
- Protein Engineering
- Receptor, TIE-2/metabolism
- Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology
- Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
- SARS-CoV-2/immunology
- Signal Transduction
- T-Lymphocytes/immunology
- T-Lymphocytes/physiology
Collapse
Affiliation(s)
- Robby Divine
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ha V Dang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Jorge A Fallas
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ivan Vulovic
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - William Sheffler
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Shally Saini
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Yan Ting Zhao
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Infencia Xavier Raj
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | | | - Madeleine F Jennewein
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98019, USA
| | - Leah J Homad
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98019, USA
| | - Yu-Hsin Wan
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98019, USA
| | - Marti R Tooley
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Franziska Seeger
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ali Etemadi
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | | | - James Lazarovits
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alex Roederer
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lance Stewart
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Mohammadali Mazloomi
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | | | - Andrew T McGuire
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98019, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Leonidas Stamatatos
- Vaccines and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98019, USA
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
40
|
|
41
|
Divine R, Dang HV, Ueda G, Fallas JA, Vulovic I, Sheffler W, Saini S, Zhao YT, Raj IX, Morawski PA, Jennewein MF, Homad LJ, Wan YH, Tooley MR, Seeger F, Etemadi A, Fahning ML, Lazarovits J, Roederer A, Walls AC, Stewart L, Mazloomi M, King NP, Campbell DJ, McGuire AT, Stamatatos L, Ruohola-Baker H, Mathieu J, Veesler D, Baker D. Designed proteins assemble antibodies into modular nanocages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.12.01.406611. [PMID: 33299994 PMCID: PMC7724662 DOI: 10.1101/2020.12.01.406611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antibodies are widely used in biology and medicine, and there has been considerable interest in multivalent antibody formats to increase binding avidity and enhance signaling pathway agonism. However, there are currently no general approaches for forming precisely oriented antibody assemblies with controlled valency. We describe the computational design of two-component nanocages that overcome this limitation by uniting form and function. One structural component is any antibody or Fc fusion and the second is a designed Fc-binding homo-oligomer that drives nanocage assembly. Structures of 8 antibody nanocages determined by electron microscopy spanning dihedral, tetrahedral, octahedral, and icosahedral architectures with 2, 6, 12, and 30 antibodies per nanocage match the corresponding computational models. Antibody nanocages targeting cell-surface receptors enhance signaling compared to free antibodies or Fc-fusions in DR5-mediated apoptosis, Tie2-mediated angiogenesis, CD40 activation, and T cell proliferation; nanocage assembly also increases SARS-CoV-2 pseudovirus neutralization by α-SARS-CoV-2 monoclonal antibodies and Fc-ACE2 fusion proteins. We anticipate that the ability to assemble arbitrary antibodies without need for covalent modification into highly ordered assemblies with different geometries and valencies will have broad impact in biology and medicine.
Collapse
Affiliation(s)
- Robby Divine
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ha V. Dang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Jorge A. Fallas
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ivan Vulovic
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - William Sheffler
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Shally Saini
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Yan Ting Zhao
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Oral Health Sciences, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Infencia Xavier Raj
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | | | - Madeleine F. Jennewein
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Leah J. Homad
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Yu-Hsin Wan
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
| | - Marti R. Tooley
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Franzika Seeger
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Ali Etemadi
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | | | - James Lazarovits
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alex Roederer
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lance Stewart
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Mohammadali Mazloomi
- Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | | | - Andrew T. McGuire
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
- University of Washington, Department of Global Health, Seattle, WA, USA
| | - Leonidas Stamatatos
- Fred Hutchinson Cancer Research Center, Vaccines and Infectious Diseases Division, Seattle, WA, USA
- University of Washington, Department of Global Health, Seattle, WA, USA
| | - Hannele Ruohola-Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
42
|
Kuwahara A, Nagai K, Nakanishi T, Kumagai I, Asano R. Functional Domain Order of an Anti-EGFR × Anti-CD16 Bispecific Diabody Involving NK Cell Activation. Int J Mol Sci 2020; 21:ijms21238914. [PMID: 33255436 PMCID: PMC7727810 DOI: 10.3390/ijms21238914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (bsAbs) have emerged as promising therapeutics. A bispecific diabody (bsDb) is a small bsAb consisting of two distinct chimeric single-chain components, with two possible arrangements of the domains. We previously reported the effect of domain order on the function of a humanized bsDb targeting the epidermal growth factor receptor (EGFR) on cancer cells, and CD3 on T cells. Notably, the co-localization of a T-cell receptor (TCR) with CD3 is bulky, potentially affecting the cross-linking ability of bsDbs, due to steric hindrance. Here, we constructed and evaluated humanized bsDbs, with different domain orders, targeting EGFR and CD16 on natural killer (NK) cells (hEx16-Dbs). We predicted minimal effects due to steric hindrance, as CD16 lacks accessory molecules. Interestingly, one domain arrangement displayed superior cytotoxicity in growth inhibition assays, despite similar cross-linking abilities for both domain orders tested. In hEx16-Dbs specifically, domain order might affect the agonistic activity of the anti-CD16 portion, which was supported by a cytokine production test, and likely contributed to the superiority of one of the hEx16-Dbs. Our results indicate that both the target antigen and mode of action of an antibody must be considered in the construction of highly functional bsAbs.
Collapse
Affiliation(s)
- Atsushi Kuwahara
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
| | - Keisuke Nagai
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai 980-8579, Japan;
| | - Takeshi Nakanishi
- Department of Applied Chemistry and Bioengineering, Graduate School of Engineering, Osaka City University, Osaka 558-8585, Japan;
| | - Izumi Kumagai
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
| | - Ryutaro Asano
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan; (A.K.); (I.K.)
- Correspondence: ; Tel.: +81-42-388-7512
| |
Collapse
|
43
|
Blanco B, Ramírez-Fernández Á, Alvarez-Vallina L. Engineering Immune Cells for in vivo Secretion of Tumor-Specific T Cell-Redirecting Bispecific Antibodies. Front Immunol 2020; 11:1792. [PMID: 32903593 PMCID: PMC7438551 DOI: 10.3389/fimmu.2020.01792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapeutic approaches based on the redirection of T cell activity toward tumor cells are actively being investigated. The impressive clinical success of the continuously intravenously infused T cell-redirecting bispecific antibody (T-bsAb) blinatumomab (anti-CD19 x anti-CD3), and of engineered T cells expressing anti-CD19 chimeric antigen receptors (CAR-T cells) in hematological malignancies, has led to renewed interest in a novel cancer immunotherapy strategy that combines features of antibody- and cell-based therapies. This emerging approach is based on the endogenous secretion of T-bsAbs by engineered T cells (STAb-T cells). Adoptive transfer of genetically modified STAb-T cells has demonstrated potent anti-tumor activity in both solid tumor and hematologic preclinical xenograft models. We review here the potential benefits of the STAb-T strategy over similar approaches currently being used in clinic, and we discuss the potential combination of this promising strategy with the well-established CAR-T cell approach.
Collapse
Affiliation(s)
- Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Ángel Ramírez-Fernández
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Luis Alvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
44
|
Darwish M, Shatz W, Leonard B, Loyet K, Barrett K, Wong JL, Li H, Abraham R, Lin M, Franke Y, Tam C, Mortara K, Zilberleyb I, Blanchette C. Nanolipoprotein Particles as a Delivery Platform for Fab Based Therapeutics. Bioconjug Chem 2020; 31:1995-2007. [DOI: 10.1021/acs.bioconjchem.0c00349] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Martine Darwish
- Genentech, South San Francisco, California 94088, United States
| | - Whitney Shatz
- Genentech, South San Francisco, California 94088, United States
| | - Brandon Leonard
- Genentech, South San Francisco, California 94088, United States
| | - Kelly Loyet
- Genentech, South San Francisco, California 94088, United States
| | - Kathy Barrett
- Genentech, South San Francisco, California 94088, United States
| | - Janice L. Wong
- Genentech, South San Francisco, California 94088, United States
| | - Hong Li
- Genentech, South San Francisco, California 94088, United States
| | - Ryan Abraham
- Genentech, South San Francisco, California 94088, United States
| | - May Lin
- Genentech, South San Francisco, California 94088, United States
| | - Yvonne Franke
- Genentech, South San Francisco, California 94088, United States
| | - Christine Tam
- Genentech, South San Francisco, California 94088, United States
| | - Kyle Mortara
- Genentech, South San Francisco, California 94088, United States
| | - Inna Zilberleyb
- Genentech, South San Francisco, California 94088, United States
| | | |
Collapse
|
45
|
Li Z, Wang Y, Vasylieva N, Wan D, Yin Z, Dong J, Hammock BD. An Ultrasensitive Bioluminescent Enzyme Immunoassay Based on Nanobody/Nanoluciferase Heptamer Fusion for the Detection of Tetrabromobisphenol A in Sediment. Anal Chem 2020; 92:10083-10090. [PMID: 32559059 DOI: 10.1021/acs.analchem.0c01908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Tetrabromobisphenol A (TBBPA) is a flame retardant and has become a widely concerning environmental pollutant. An ultrasensitive nanobody-based immunoassay was developed to monitor the exposure of TBBPA in sediment. First, the anti-TBBPA nanobody was fused with nanoluciferase, and then a one-step bioluminescent enzyme immunoassay (BLEIA) was developed with high sensitivity for TBBPA, with a maximum half inhibition concentration (IC50) at 187 pg/mL. Although approximately 10-fold higher sensitivity can be achieved by this developed BLEIA than by the classical two-step ELISA (IC50 at 1778 pg/mL), it is still a challenge to detect trace TBBPA in sediment samples reliably due to the relatively high matrix effect. To further improve the performance of this one-step BLEIA, a C4b-binding protein (C4BP) was inserted as a self-assembling linker between the nanobody and nanoluciferase. Therefore, a heptamer fusion containing seven binders and seven tracers was generated. This reagent improved the binding capacity and signal amplification. The one-step heptamer plus BLEIA based on this immune-reagent shows an additional 7-fold improvement of sensitivity, with the IC50 of 28.9 pg/mL and the limit of detection as low as 2.5 pg/mL. The proposed assay was further applied to determine the trace TBBPA in sediment, and the recovery was within 92-103%. Taking advantage of this heptamer fusion, one-step BLEIA can serve as a powerful tool for fast detection of trace TBBPA in the sediment samples.
Collapse
Affiliation(s)
- Zhenfeng Li
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Yi Wang
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States.,Department of Pesticides Science, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Natalia Vasylieva
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Debin Wan
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Zihan Yin
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| | - Jiexian Dong
- Shenzhen Forward Pharma Co., Ltd., Shenzhen 518057, China
| | - Bruce D Hammock
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, California 95616, United States
| |
Collapse
|
46
|
Song W, Das M, Chen X. Nanotherapeutics for Immuno-Oncology: A Crossroad for New Paradigms. Trends Cancer 2020; 6:288-298. [PMID: 32209444 PMCID: PMC7101275 DOI: 10.1016/j.trecan.2020.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 01/17/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
With the rapid increase in the use of nanotechnology and immunotherapy for cancer management in the recent past, there are great implications for using nanotechnology in immuno-oncology. However, to deliver clinical success, the scientific and clinical rationale must be critically evaluated when applying nanotechnology to immuno-oncology challenges. This opinion article distinguishes between designing nanotherapeutics for immunotherapy and the past focus on the placement of chemotherapy agents in nanoparticles. We believe the integration of nanotechnology with cancer immunotherapy for nano-immunotherapeutics provides unique opportunities for both fields, paving the way for entirely new therapeutic paradigms. As a particular focus in our article, we envision the necessities and challenges of nanotechnology in the development of in situ cancer vaccines, immune checkpoint inhibitors, adoptive cell transfer, and bispecific antibody therapy.
Collapse
Affiliation(s)
- Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, Jilin, 130022, China.
| | - Manisit Das
- Division of Pharmacoengineering and Molecular Pharmaceutics, and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, Jilin, 130022, China
| |
Collapse
|
47
|
Asano R, Hosokawa K, Taki S, Konno S, Shimomura I, Ogata H, Okada M, Arai K, Onitsuka M, Omasa T, Nakanishi T, Umetsu M, Kumagai I. Build-up functionalization of anti-EGFR × anti-CD3 bispecific diabodies by integrating high-affinity mutants and functional molecular formats. Sci Rep 2020; 10:4913. [PMID: 32188928 PMCID: PMC7080790 DOI: 10.1038/s41598-020-61840-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/04/2020] [Indexed: 12/26/2022] Open
Abstract
Designing non-natural antibody formats is a practical method for developing highly functional next-generation antibody drugs, particularly for improving the therapeutic efficacy of cancer treatments. One approach is constructing bispecific antibodies (bsAbs). We previously reported a functional humanized bispecific diabody (bsDb) that targeted epidermal growth factor receptor and CD3 (hEx3-Db). We enhanced its cytotoxicity by constructing an Fc fusion protein and rearranging order of the V domain. In this study, we created an additional functional bsAb, by integrating the molecular formats of bsAb and high-affinity mutants previously isolated by phage display in the form of Fv. Introducing the high-affinity mutations into bsDbs successfully increased their affinities and enhanced their cytotoxicity in vitro and in vivo. However, there were some limitations to affinity maturation of bsDb by integrating high-affinity Fv mutants, particularly in Fc-fused bsDb with intrinsic high affinity, because of their bivalency. The tetramers fractionated from the bsDb mutant exhibited the highest in vitro growth inhibition among the small bsAbs and was comparable to the in vivo anti-tumor effects of Fc-fused bsDbs. This molecule shows cost-efficient bacterial production and high therapeutic potential.
Collapse
Affiliation(s)
- Ryutaro Asano
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan. .,Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, Tokyo, 184-8588, Japan.
| | - Katsuhiro Hosokawa
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Shintaro Taki
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Shota Konno
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Ippei Shimomura
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Hiromi Ogata
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Mai Okada
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Kyoko Arai
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Masayoshi Onitsuka
- Institute of Technology and Science, Tokushima University, Tokushima, 770-8506, Japan
| | - Takeshi Omasa
- Institute of Technology and Science, Tokushima University, Tokushima, 770-8506, Japan
| | - Takeshi Nakanishi
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Mitsuo Umetsu
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Izumi Kumagai
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan.
| |
Collapse
|
48
|
Finding the Keys to the CAR: Identifying Novel Target Antigens for T Cell Redirection Immunotherapies. Int J Mol Sci 2020; 21:ijms21020515. [PMID: 31947597 PMCID: PMC7014258 DOI: 10.3390/ijms21020515] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Oncology immunotherapy has been a significant advancement in cancer treatment and involves harnessing and redirecting a patient’s immune response towards their own tumour. Specific recognition and elimination of tumour cells was first proposed over a century ago with Paul Erlich’s ‘magic bullet’ theory of therapy. In the past decades, targeting cancer antigens by redirecting T cells with antibodies using either bispecific T cell engagers (BiTEs) or chimeric antigen receptor (CAR) T cell therapy has achieved impressive clinical responses. Despite recent successes in haematological cancers, linked to a high and uniformly expressed CD19 antigen, the efficacy of T cell therapies in solid cancers has been disappointing, in part due to antigen escape. Targeting heterogeneous solid tumours with T cell therapies will require the identification of novel tumour specific targets. These targets can be found among a range of cell-surface expressed antigens, including proteins, glycolipids or carbohydrates. In this review, we will introduce the current tumour target antigen classification, outline existing approaches to discover novel tumour target antigens and discuss considerations for future design of antibodies with a focus on their use in CAR T cells.
Collapse
|
49
|
Roda-Navarro P, Álvarez-Vallina L. Understanding the Spatial Topology of Artificial Immunological Synapses Assembled in T Cell-Redirecting Strategies: A Major Issue in Cancer Immunotherapy. Front Cell Dev Biol 2020; 7:370. [PMID: 31998721 PMCID: PMC6965029 DOI: 10.3389/fcell.2019.00370] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/16/2019] [Indexed: 11/30/2022] Open
Affiliation(s)
- Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense, Madrid, Spain.,Lymphocyte Immunobiology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| |
Collapse
|
50
|
Mechanisms of noncanonical binding dynamics in multivalent protein-protein interactions. Proc Natl Acad Sci U S A 2019; 116:25659-25667. [PMID: 31776263 DOI: 10.1073/pnas.1902909116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protein multivalency can provide increased affinity and specificity relative to monovalent counterparts, but these emergent biochemical properties and their mechanistic underpinnings are difficult to predict as a function of the biophysical properties of the multivalent binding partners. Here, we present a mathematical model that accurately simulates binding kinetics and equilibria of multivalent protein-protein interactions as a function of the kinetics of monomer-monomer binding, the structure and topology of the multidomain interacting partners, and the valency of each partner. These properties are all experimentally or computationally estimated a priori, including approximating topology with a worm-like chain model applicable to a variety of structurally disparate systems, thus making the model predictive without parameter fitting. We conceptualize multivalent binding as a protein-protein interaction network: ligand and receptor valencies determine the number of interacting species in the network, with monomer kinetics and structural properties dictating the dynamics of each species. As predicted by the model and validated by surface plasmon resonance experiments, multivalent interactions can generate several noncanonical macroscopic binding dynamics, including a transient burst of high-energy configurations during association, biphasic equilibria resulting from interligand competition at high concentrations, and multiexponential dissociation arising from differential lifetimes of distinct network species. The transient burst was only uncovered when extending our analysis to trivalent interactions due to the significantly larger network, and we were able to predictably tune burst magnitude by altering linker rigidity. This study elucidates mechanisms of multivalent binding and establishes a framework for model-guided analysis and engineering of such interactions.
Collapse
|