1
|
Hayashi H, Mak TW, Tanaka Y, Kubo Y, Izumida M, Urae R, Matsuyama T. Development of a highly sensitive platform for protein-protein interaction detection and regulation of T cell function. Proc Natl Acad Sci U S A 2024; 121:e2318190121. [PMID: 39106307 PMCID: PMC11331103 DOI: 10.1073/pnas.2318190121] [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: 10/19/2023] [Accepted: 06/28/2024] [Indexed: 08/09/2024] Open
Abstract
We developed a highly sensitive assay for detecting protein-protein interaction using chimeric receptors comprising two molecules of interest in the extracellular domain and interferon alpha and beta receptor subunit 1 or 2 (IFNAR1/2) in the intracellular domain. This intracellular IFNAR1/2 reconstitution system (IFNARRS) proved markedly more sensitive than the NanoBiT system, currently considered one of the best detection systems for protein interaction. Employing chimeric receptors with extracellular domains from the IFNγ or IL-2 receptor and the intracellular domains of IFNAR1/2, the IFNARRS system effectively identifies low IFNγ or IL-2 levels. Cells stably expressing these chimeric receptors responded to IFNγ secreted by activated T cells following various stimuli, including a specific peptide-antigen. The activation signals were further enhanced by the expression of relevant genes, such as costimulators, via IFN-stimulated response elements in the promoters. Besides IFNγ or IL-2, the IFNARRS system demonstrated the capability to detect other cytokines by using the corresponding extracellular domains from these target cytokine receptors.
Collapse
Affiliation(s)
- Hideki Hayashi
- Medical University Research Administrator, Nagasaki University School of Medicine, Nagasaki852-8523, Japan
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong Special Administrative Region 999077, China
| | - Tak Wah Mak
- Center for Medical Innovation, Nagasaki University, Nagasaki852-8588, Japan
- Princess Margaret Cancer Center, University Health Network, Toronto, ONM5G 2M9, Canada
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region 999077, China
| | - Yoshimasa Tanaka
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong Special Administrative Region 999077, China
| | - Yoshinao Kubo
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki852-8523, Japan
| | - Mai Izumida
- Department of Clinical Medicine, Institute of Tropical Medicine, Nagasaki University, Nagasaki852-8523, Japan
| | - Ryuji Urae
- Souseikai Clinical Research Center, Fukuoka812-0025, Japan
| | - Toshifumi Matsuyama
- Department of Forensic Pathology and Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki852-8523, Japan
| |
Collapse
|
2
|
Tennenhouse A, Khmelnitsky L, Khalaila R, Yeshaya N, Noronha A, Lindzen M, Makowski EK, Zaretsky I, Sirkis YF, Galon-Wolfenson Y, Tessier PM, Abramson J, Yarden Y, Fass D, Fleishman SJ. Computational optimization of antibody humanness and stability by systematic energy-based ranking. Nat Biomed Eng 2024; 8:30-44. [PMID: 37550425 PMCID: PMC10842793 DOI: 10.1038/s41551-023-01079-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 07/13/2023] [Indexed: 08/09/2023]
Abstract
Conventional methods for humanizing animal-derived antibodies involve grafting their complementarity-determining regions onto homologous human framework regions. However, this process can substantially lower antibody stability and antigen-binding affinity, and requires iterative mutational fine-tuning to recover the original antibody properties. Here we report a computational method for the systematic grafting of animal complementarity-determining regions onto thousands of human frameworks. The method, which we named CUMAb (for computational human antibody design; available at http://CUMAb.weizmann.ac.il ), starts from an experimental or model antibody structure and uses Rosetta atomistic simulations to select designs by energy and structural integrity. CUMAb-designed humanized versions of five antibodies exhibited similar affinities to those of the parental animal antibodies, with some designs showing marked improvement in stability. We also show that (1) non-homologous frameworks are often preferred to highest-homology frameworks, and (2) several CUMAb designs that differ by dozens of mutations and that use different human frameworks are functionally equivalent.
Collapse
Affiliation(s)
- Ariel Tennenhouse
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Lev Khmelnitsky
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Razi Khalaila
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Yeshaya
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ashish Noronha
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
| | - Moshit Lindzen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Emily K Makowski
- Biointerfaces Institute and Departments of Chemical Engineering, Pharmaceutical Sciences and Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ira Zaretsky
- Antibody Engineering Unit, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Peter M Tessier
- Biointerfaces Institute and Departments of Chemical Engineering, Pharmaceutical Sciences and Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deborah Fass
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sarel J Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Wu Z, Yoshikawa T, Inoue S, Ito Y, Kasuya H, Nakashima T, Zhang H, Kotaka S, Hosoda W, Suzuki S, Kagoya Y. CD83 expression characterizes precursor exhausted T cell population. Commun Biol 2023; 6:258. [PMID: 36906640 PMCID: PMC10008643 DOI: 10.1038/s42003-023-04631-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023] Open
Abstract
T cell exhaustion is a main obstacle against effective cancer immunotherapy. Exhausted T cells include a subpopulation that maintains proliferative capacity, referred to as precursor exhausted T cells (TPEX). While functionally distinct and important for antitumor immunity, TPEX possess some overlapping phenotypic features with the other T-cell subsets within the heterogeneous tumor-infiltrating T-lymphocytes (TIL). Here we explore surface marker profiles unique to TPEX using the tumor models treated by chimeric antigen receptor (CAR)-engineered T cells. We find that CD83 is predominantly expressed in the CCR7+PD1+ intratumoral CAR-T cells compared with the CCR7-PD1+ (terminally differentiated) and CAR-negative (bystander) T cells. The CD83+CCR7+ CAR-T cells exhibit superior antigen-induced proliferation and IL-2 production compared with the CD83- T cells. Moreover, we confirm selective expression of CD83 in the CCR7+PD1+ T-cell population in primary TIL samples. Our findings identify CD83 as a marker to discriminate TPEX from terminally exhausted and bystander TIL.
Collapse
Affiliation(s)
- Zhiwen Wu
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Toshiaki Yoshikawa
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Inoue
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Ito
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hitomi Kasuya
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takahiro Nakashima
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Haosong Zhang
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Saki Kotaka
- Department of Gynecologic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Waki Hosoda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Shiro Suzuki
- Department of Gynecologic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Yuki Kagoya
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan.
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
4
|
Rosato F, Pasupuleti R, Tomisch J, Meléndez AV, Kolanovic D, Makshakova ON, Wiltschi B, Römer W. A bispecific, crosslinking lectibody activates cytotoxic T cells and induces cancer cell death. J Transl Med 2022; 20:578. [PMID: 36494671 PMCID: PMC9733292 DOI: 10.1186/s12967-022-03794-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Aberrant glycosylation patterns play a crucial role in the development of cancer cells as they promote tumor growth and aggressiveness. Lectins recognize carbohydrate antigens attached to proteins and lipids on cell surfaces and represent potential tools for application in cancer diagnostics and therapy. Among the emerging cancer therapies, immunotherapy has become a promising treatment modality for various hematological and solid malignancies. Here we present an approach to redirect the immune system into fighting cancer by targeting altered glycans at the surface of malignant cells. We developed a so-called "lectibody", a bispecific construct composed of a lectin linked to an antibody fragment. This lectibody is inspired by bispecific T cell engager (BiTEs) antibodies that recruit cytotoxic T lymphocytes (CTLs) while simultaneously binding to tumor-associated antigens (TAAs) on cancer cells. The tumor-related glycosphingolipid globotriaosylceramide (Gb3) represents the target of this proof-of-concept study. It is recognized with high selectivity by the B-subunit of the pathogen-derived Shiga toxin, presenting opportunities for clinical development. METHODS The lectibody was realized by conjugating an anti-CD3 single-chain antibody fragment to the B-subunit of Shiga toxin to target Gb3+ cancer cells. The reactive non-canonical amino acid azidolysine (AzK) was inserted at predefined single positions in both proteins. The azido groups were functionalized by bioorthogonal conjugation with individual linkers that facilitated selective coupling via an alternative bioorthogonal click chemistry reaction. In vitro cell-based assays were conducted to evaluate the antitumoral activity of the lectibody. CTLs, Burkitt´s lymphoma-derived cells and colorectal adenocarcinoma cell lines were screened in flow cytometry and cytotoxicity assays for activation and lysis, respectively. RESULTS This proof-of-concept study demonstrates that the lectibody activates T cells for their cytotoxic signaling, redirecting CTLs´ cytotoxicity in a highly selective manner and resulting in nearly complete tumor cell lysis-up to 93%-of Gb3+ tumor cells in vitro. CONCLUSIONS This research highlights the potential of lectins in targeting certain tumors, with an opportunity for new cancer treatments. When considering a combinatorial strategy, lectin-based platforms of this type offer the possibility to target glycan epitopes on tumor cells and boost the efficacy of current therapies, providing an additional strategy for tumor eradication and improving patient outcomes.
Collapse
Affiliation(s)
- Francesca Rosato
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Rajeev Pasupuleti
- ACIB - The Austrian Centre of Industrial Biotechnology, Graz, Austria
- Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria
| | - Jana Tomisch
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Ana Valeria Meléndez
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Dajana Kolanovic
- ACIB - The Austrian Centre of Industrial Biotechnology, Graz, Austria
- Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria
| | - Olga N Makshakova
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Kazan Institute for Biochemistry and Biophysics, FRC Kazan Scientific Center of RAS, Kazan, Russian Federation
| | - Birgit Wiltschi
- ACIB - The Austrian Centre of Industrial Biotechnology, Graz, Austria.
- Institute of Molecular Biotechnology, Graz University of Technology, Graz, Austria.
- Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria.
| | - Winfried Römer
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
5
|
Gao Y, Yang T, Liu H, Song N, Dai C, Ding Y. Development and characterization of a novel human CD137 agonistic antibody with anti-tumor activity and a good safety profile in non-human primates. FEBS Open Bio 2022; 12:2166-2178. [PMID: 36176235 PMCID: PMC9714380 DOI: 10.1002/2211-5463.13494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/01/2022] [Accepted: 09/28/2022] [Indexed: 01/25/2023] Open
Abstract
CD137 (4-1BB, TNFRSF9), an inducible T-cell costimulatory receptor, is expressed on activated T cells, activated NK cells, Treg cells, and several innate immune cells, including DCs, monocytes, neutrophils, mast cells, and eosinophils. In animal models and clinical trials, anti-CD137 agonistic monoclonal antibodies have shown anti-tumor potential, but balancing the efficacy and toxicity of anti-CD137 agonistic monoclonal antibodies is a considerable hindrance for clinical applications. Here, we describe a novel fully human CD137 agonistic antibody (PE0116) generated from immunized harbor H2L2 human transgenic mice. PE0116 is a ligand blocker, which is also the case for Utomilumab (one of the leading CD137 agonistic drugs); PE0116 partially overlaps with Urelumab's recognized epitope. In vitro, PE0116 activates NF-κB signaling, significantly promotes T-cell proliferation, and increases cytokine secretion in the presence of cross-linking. Importantly, PE0116 possesses robust anti-tumor activity in the MC38 tumor model. In vivo, PE0116 exhibits a good safety profile and has typical pharmacokinetic characteristics of an IgG antibody in preclinical studies of non-human primates. In summary, PE0116 is a promising anti-CD137 antibody with a good safety profile in preclinical studies.
Collapse
Affiliation(s)
- Yingying Gao
- State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiChina
- Biologics DiscoveryShanghai ChemPartner Co., Ltd.China
| | - Teddy Yang
- Biologics DiscoveryShanghai ChemPartner Co., Ltd.China
| | - Hu Liu
- Biologics DiscoveryShanghai ChemPartner Co., Ltd.China
| | - Ningning Song
- Biologics DiscoveryShanghai ChemPartner Co., Ltd.China
| | - Chaohui Dai
- Biologics DiscoveryShanghai Hyamab Biotechnology Co., Ltd.China
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life SciencesFudan UniversityShanghaiChina
| |
Collapse
|
6
|
Chen X, Xue L, Ding X, Zhang J, Jiang L, Liu S, Hou H, Jiang B, Cheng L, Zhu Q, Zhang L, Zhou X, Ma J, Liu Q, Li Y, Ren Z, Jiang B, Song X, Song J, Jin W, Wei M, Shen Z, Liu X, Wang L, Li K, Zhang T. An Fc-Competent Anti-Human TIGIT Blocking Antibody Ociperlimab (BGB-A1217) Elicits Strong Immune Responses and Potent Anti-Tumor Efficacy in Pre-Clinical Models. Front Immunol 2022; 13:828319. [PMID: 35273608 PMCID: PMC8902820 DOI: 10.3389/fimmu.2022.828319] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
TIGIT (T-cell immunoglobulin and ITIM domain) has emerged as a promising target in cancer immunotherapy. It is an immune “checkpoint” inhibitor primarily expressed on activated T cells, NK cells and Tregs. Engagement of TIGIT to its ligands PVR and PVR-L2 leads to inhibitory signaling in T cells, promoting functional exhaustion of tumor-infiltrating T lymphocytes. Here, we described the pre-clinical characterization of Ociperlimab (BGB-A1217), a novel humanized IgG1 anti-TIGIT antibody (mAb), and systemically evaluated the contribution of Fc functions in the TIGIT mAb-mediated anti-tumor activities. BGB-A1217 binds to the extracellular domain of human TIGIT with high affinity (KD = 0.135 nM) and specificity, and efficiently blocks the interaction between TIGIT and its ligands PVR or PVR-L2. Cell-based assays show that BGB-A1217 significantly enhances T-cell functions. In addition, BGB-A1217 induces antibody dependent cellular cytotoxicity (ADCC) against Treg cells, activates NK cells and monocytes, and removes TIGIT from T cell surfaces in an Fc-dependent manner, In vivo, BGB-A1217, either alone or in combination with an anti-PD-1 mAb elicits strong immune responses and potent anti-tumor efficacy in pre-clinical models. Moreover, the Fc effector function is critical for the anti-tumor activity of BGB-A1217 in a syngeneic human TIGIT-knock-in mouse model. The observed anti-tumor efficacy is associated with a pharmacodynamic change of TIGIT down-regulation and Treg reduction. These data support the selection of BGB-A1217 with an effector function competent Fc region for clinical development for the treatment of human cancers.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Liu Xue
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiao Ding
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jing Zhang
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lei Jiang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Sha Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Hongjia Hou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Bin Jiang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Liang Cheng
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Qing Zhu
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lijie Zhang
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiaosui Zhou
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jie Ma
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Qi Liu
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Yucheng Li
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Zhiying Ren
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Beibei Jiang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xiaomin Song
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Jing Song
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Wei Jin
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Min Wei
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Zhirong Shen
- Department of Discovery Biomarkers, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Xuesong Liu
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Lai Wang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Kang Li
- Department of Biologics, BeiGene (Beijing) Co., Ltd., Beijing, China
| | - Tong Zhang
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
7
|
Selection and characterization of scFv antibody against nucleocapsid protein of Porcine reproductive and respiratory syndrome virus. ACTA VET BRNO 2020. [DOI: 10.2754/avb202089010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a widespread infectious agent in pigs. Nucleocapsid (N) protein of PRRSV has been identified as the most immunodominant viral protein. The main goal of the work was the selection and characterization of a single-chain antibody fragments (scFv) antibody specific to the N protein. Specific scFv antibody clone D5 was selected from the Tomlinson phagemid library and purified by immobilized metal affinity chromatography from the periplasmatic space of E. coli cells. The antibody was then characterized by sequencing and the ability to recognize the native virus N protein by Western blot and competitive ELISA. Pepscan analysis identified the position of the binding epitope between amino acids 62–84 of the N protein. Our study could help to improve the diagnostics and prevention of PRRSV in Central Europe.
Collapse
|
8
|
Ahamadi-Fesharaki R, Fateh A, Vaziri F, Solgi G, Siadat SD, Mahboudi F, Rahimi-Jamnani F. Single-Chain Variable Fragment-Based Bispecific Antibodies: Hitting Two Targets with One Sophisticated Arrow. Mol Ther Oncolytics 2019; 14:38-56. [PMID: 31011631 PMCID: PMC6463744 DOI: 10.1016/j.omto.2019.02.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite the success of monoclonal antibodies (mAbs) to treat some disorders, the monospecific molecular entity of mAbs as well as the presence of multiple factors and pathways involved in the pathogenesis of disorders, such as various malignancies, infectious diseases, and autoimmune disorders, and resistance to therapy have restricted the therapeutic efficacy of mAbs in clinical use. Bispecific antibodies (bsAbs), by concurrently recognizing two targets, can partly circumvent these problems. Serial killing of tumor cells by bsAb-redirected T cells, simultaneous blocking of two antigens involved in the HIV-1 infection, and concurrent targeting of the activating and inhibitory receptors on B cells to modulate autoimmunity are part of the capabilities of bsAbs. After designing and developing a large number of bsAbs for years, catumaxomab, a full-length bsAb targeting EpCAM and CD3, was approved in 2009 to treat EpCAM-positive carcinomas besides blinatumomab, a bispecific T cell engager antibody targeting CD19 and CD3, which was approved in 2014 to treat relapsed or refractory acute lymphoblastic leukemia. Furthermore, approximately 60 bsAbs are under investigation in clinical trials. The current review aims at portraying different formats of the single-chain variable fragment (scFv)-based bsAbs and shedding light on the scFv-based bsAbs in preclinical development, different phases of clinical trials, and the market.
Collapse
Affiliation(s)
- Raoufeh Ahamadi-Fesharaki
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Davar Siadat
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Rahimi-Jamnani
- Human Antibody Lab, Innovation Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
9
|
Ma H, Ó'Fágáin C, O'Kennedy R. Unravelling enhancement of antibody fragment stability – Role of format structure and cysteine modification. J Immunol Methods 2019; 464:57-63. [DOI: 10.1016/j.jim.2018.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 01/08/2023]
|
10
|
Helsen CW, Hammill JA, Lau VWC, Mwawasi KA, Afsahi A, Bezverbnaya K, Newhook L, Hayes DL, Aarts C, Bojovic B, Denisova GF, Kwiecien JM, Brain I, Derocher H, Milne K, Nelson BH, Bramson JL. The chimeric TAC receptor co-opts the T cell receptor yielding robust anti-tumor activity without toxicity. Nat Commun 2018; 9:3049. [PMID: 30076299 PMCID: PMC6076291 DOI: 10.1038/s41467-018-05395-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 06/25/2018] [Indexed: 01/06/2023] Open
Abstract
Engineering T cells with chimeric antigen receptors (CARs) is an effective method for directing T cells to attack tumors, but may cause adverse side effects such as the potentially lethal cytokine release syndrome. Here the authors show that the T cell antigen coupler (TAC), a chimeric receptor that co-opts the endogenous TCR, induces more efficient anti-tumor responses and reduced toxicity when compared with past-generation CARs. TAC-engineered T cells induce robust and antigen-specific cytokine production and cytotoxicity in vitro, and strong anti-tumor activity in a variety of xenograft models including solid and liquid tumors. In a solid tumor model, TAC-T cells outperform CD28-based CAR-T cells with increased anti-tumor efficacy, reduced toxicity, and faster tumor infiltration. Intratumoral TAC-T cells are enriched for Ki-67+ CD8+ T cells, demonstrating local expansion. These results indicate that TAC-T cells may have a superior therapeutic index relative to CAR-T cells.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD28 Antigens/immunology
- Cell Line, Tumor
- Cytokines/blood
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Female
- Genetic Engineering
- HEK293 Cells
- Humans
- Immunotherapy, Adoptive/methods
- Lentivirus/genetics
- Lymphocyte Activation
- Male
- Mice
- Mice, Inbred NOD
- Protein Engineering
- Receptor, ErbB-2/immunology
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Recombinant Proteins/immunology
- Single-Domain Antibodies
- T-Cell Antigen Receptor Specificity/genetics
- T-Cell Antigen Receptor Specificity/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Vision, Ocular
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Christopher W Helsen
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Joanne A Hammill
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Vivian W C Lau
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Kenneth A Mwawasi
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Arya Afsahi
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Lisa Newhook
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Danielle L Hayes
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Craig Aarts
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Bojana Bojovic
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Galina F Denisova
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Jacek M Kwiecien
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
- Department of Clinical Pathomorphology, Medical University of Lublin, Racławickie 1 Street, 20-059, Lublin, Poland
| | - Ian Brain
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Heather Derocher
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, 2410 Lee Ave, Victoria, BC, V8R 6V5, Canada
| | - Katy Milne
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, 2410 Lee Ave, Victoria, BC, V8R 6V5, Canada
| | - Brad H Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, 2410 Lee Ave, Victoria, BC, V8R 6V5, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
11
|
Schwaigerlehner L, Pechlaner M, Mayrhofer P, Oostenbrink C, Kunert R. Lessons learned from merging wet lab experiments with molecular simulation to improve mAb humanization. Protein Eng Des Sel 2018; 31:257-265. [PMID: 29757445 PMCID: PMC6277173 DOI: 10.1093/protein/gzy009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/04/2018] [Accepted: 04/12/2018] [Indexed: 12/12/2022] Open
Abstract
Humanized monoclonal antibodies (mAbs) are among the most promising modern therapeutics, but defined engineering strategies are still not available. Antibody humanization often leads to a loss of affinity, as it is the case for our model antibody Ab2/3H6 (PDB entry 3BQU). Identifying appropriate back-to-mouse mutations is needed to restore binding affinity, but highly challenging. In order to get more insight, we have applied molecular dynamics simulations and correlated them to antibody binding and expression in wet lab experiments. In this study, we discuss six mAb variants and investigate a tyrosine conglomeration, an isopolar substitution and the improvement of antibody binding towards wildtype affinity. In the 3D structure of the mouse wildtype, residue R94h is surrounded by three tyrosines which form a so-called 'tyrosine cage'. We demonstrate that the tyrosine cage has a supporting function for the CDRh3 loop conformation. The isopolar substitution is not able to mimic the function appropriately. Finally, we show that additional light chain mutations can restore binding to wildtype-comparable level, and also improve the expression of the mAb significantly. We conclude that the variable light chain of Ab2/3H6 is of underestimated importance for the interaction with its antigen mAb 2F5.
Collapse
Affiliation(s)
- L Schwaigerlehner
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria
| | - M Pechlaner
- Department of Material Sciences and Process Engineering, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria
| | - P Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria
| | - C Oostenbrink
- Department of Material Sciences and Process Engineering, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria
| | - R Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences, Muthgasse 18, Vienna, Austria
| |
Collapse
|
12
|
Ellwanger K, Reusch U, Fucek I, Knackmuss S, Weichel M, Gantke T, Molkenthin V, Zhukovsky EA, Tesar M, Treder M. Highly Specific and Effective Targeting of EGFRvIII-Positive Tumors with TandAb Antibodies. Front Oncol 2017; 7:100. [PMID: 28596941 PMCID: PMC5442391 DOI: 10.3389/fonc.2017.00100] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/01/2017] [Indexed: 12/31/2022] Open
Abstract
To harness the cytotoxic capacity of immune cells for the treatment of solid tumors, we developed tetravalent, bispecific tandem diabody (TandAb) antibodies that recognize EGFRvIII, the deletion variant III of the epidermal growth factor receptor (EGFR), and CD3 on T-cells, thereby directing immune cells to eliminate EGFRvIII-positive tumor cells. Using phage display, we identified scFv antibodies selectively binding to EGFRvIII. These highly EGFRvIII-specific, fully human scFv were substantially improved by affinity maturation, achieving KDs in the picomolar range, and were used to construct a set of bispecific EGFRvIII-targeting TandAbs with a broad range of binding and cytotoxic properties. These antibodies exhibited an exquisite specificity for a distinguished epitope in the N-terminal portion of EGFRvIII, as shown on recombinant antigen in Western Blot, SPR, and ELISA, as well as on antigen-expressing cells in FACS assays, and did not bind to the wild-type EGFR. High-affinity EGFRvIII/CD3 TandAbs were most potent in killing assays, displaying cytotoxicity toward EGFRvIII-expressing CHO, F98 glioma, or human DK-MG cells with EC50 values in the range of 1-10 pM in vitro. They also demonstrated dose-dependent growth control in vivo in an EGFRvIII-positive subcutaneous xenograft tumor model. Together with the tumor-exclusive expression of EGFRvIII, the EGFRvIII/CD3 TandAbs' high specificity and strictly target-dependent activation with no off-target activity provide an opportunity to target tumor cells and spare normal tissues, thereby reducing the side effects associated with other anti-EGFR therapies. In summary, EGFRvIII/CD3 TandAbs are highly attractive therapeutic antibody candidates for selective immunotherapy of EGFRvIII-positive tumors.
Collapse
|
13
|
Ayyar BV, Arora S, Ravi SS. Optimizing antibody expression: The nuts and bolts. Methods 2017; 116:51-62. [PMID: 28163103 DOI: 10.1016/j.ymeth.2017.01.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/28/2017] [Accepted: 01/28/2017] [Indexed: 01/07/2023] Open
Abstract
Antibodies are extensively utilized entities in biomedical research, and in the development of diagnostics and therapeutics. Many of these applications require high amounts of antibodies. However, meeting this ever-increasing demand of antibodies in the global market is one of the outstanding challenges. The need to maintain a balance between demand and supply of antibodies has led the researchers to discover better means and methods for optimizing their expression. These strategies aim to increase the volumetric productivity of the antibodies along with the reduction of associated manufacturing costs. Recent years have witnessed major advances in recombinant protein technology, owing to the introduction of novel cloning strategies, gene manipulation techniques, and an array of cell and vector engineering techniques, together with the progress in fermentation technologies. These innovations were also highly beneficial for antibody expression. Antibody expression depends upon the complex interplay of multiple factors that may require fine tuning at diverse levels to achieve maximum yields. However, each antibody is unique and requires individual consideration and customization for optimizing the associated expression parameters. This review provides a comprehensive overview of several state-of-the-art approaches, such as host selection, strain engineering, codon optimization, gene optimization, vector modification and process optimization that are deemed suitable for enhancing antibody expression.
Collapse
Affiliation(s)
- B Vijayalakshmi Ayyar
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sushrut Arora
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Shiva Shankar Ravi
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
14
|
Wu H, Yao L, Chou L, Yang JH, Zhang YX, Li XL, Shan BE. Construction and functional analysis of an anti-human cervical carcinoma/anti-human CD3 single-chain bispecific antibody. Mol Med Rep 2016; 14:804-10. [PMID: 27220396 DOI: 10.3892/mmr.2016.5292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 03/09/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to construct a single-chain bispecific antibody (scBsAb) against cervical carcinoma and to investigate its biological activities. The scBsAb was constructed using a genetic cloning technique and antigen binding activities were detected by ELISA. The iodogen method was used to analyze the pharmacokinetics. The Rosette formation test was used to detect the binding ability between peripheral blood lymphocytes (PBLs) and Cs1213 cervical cancer cells. In addition, the MTT method was performed to detect the killing effect of PBLs. The molecular weight of the scBsAb was ~60 kDa. The antigen binding activities of scBsAbs were compared with the anti‑human cervical carcinoma antibody single‑chain Fv fragment (CSAs‑1 scFv) and anti‑cluster of differentiation (CD)3 scFv (P>0.05). In addition, a pharmacokinetics assay demonstrated that compared with the two corresponding scFvs, scBsAbs exhibited a significantly prolonged retention time in the body (P<0.01). In addition, the number of rosettes formed by PBLs and Cs1213 cells in the scBsAb group was markedly greater than that in the scFv groups or the RPMI‑1640 group (P<0.05 and P<0.01, respectively). The killing activity of PBLs against scBsAb‑mediated Cs1213 cells was significantly greater than that mediated by the other antibodies (P<0.05). When the concentration of scBsAb was 40 µg/ml, the killing rate was 64.5%. Thus, anti‑human cervical carcinoma/anti‑CD3 scBsAbs may possess two types of antigen binding activity, prolong the duration in vivo and improve the killing activity of PBLs against cancer cells.
Collapse
Affiliation(s)
- Hong Wu
- Department of Gynecology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Li Yao
- Department of Gynecology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Lin Chou
- Department of Gynecology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Jin-Hua Yang
- Department of Pathology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Yun-Xiu Zhang
- Department of Gynecology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Xiao-Li Li
- Department of Gynecology, Zhengzhou People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Bo-Er Shan
- Department of Gynecologic Oncology, Cancer Hospital, Fudan University, Shanghai 200438, P.R. China
| |
Collapse
|
15
|
Kapelski S, Boes A, Spiegel H, de Almeida M, Klockenbring T, Reimann A, Fischer R, Barth S, Fendel R. Fast track antibody V-gene rescue, recombinant expression in plants and characterization of a PfMSP4-specific antibody. Malar J 2015; 14:50. [PMID: 25651860 PMCID: PMC4323031 DOI: 10.1186/s12936-015-0577-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/25/2015] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Monoclonal antibodies (mAbs) are essential tools in biological research, diagnosis and therapy, and are conventionally produced in murine hybridoma cell lines. Professional applications of mAbs depend on the steady supply of material. Because hybridoma cultures can stop producing the antibody or even die, preservation of the unique epitope specificity of mAbs by rescue of the sequences encoding the antibody variable domains (V regions) is important. The availability of these sequences enables not only the recombinant expression of the original antibody for further applications, but opens the road for antibody engineering towards innovative diagnostic or therapeutic applications. A time- and cost-efficient production system enabling the detailed analysis of the antibodies is an essential requirement in this context. METHODS Sequences were rescued from three hybridoma cell lines, subjected to sequence analysis, subcloned into binary expression vectors and recombinantly expressed as chimeric mAb (constant regions of human IgG1:k1) in Nicotiana benthamiana plants. The properties of the recombinant and the murine mAbs were compared using competition enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR) spectroscopy. The recognition of native PfMSP4 by the recombinant mAb was analysed by immunofluorescence staining of Pf 3D7A schizonts and by western blot analysis of merozoite extract. RESULTS The rescued sequences of all three hybridoma cell lines were identical. The recombinant mAb was successfully expressed as IgG in plants at moderate levels (45 mg/kg fresh leaf weight). Preservation of the original epitope was demonstrated in a competition ELISA, using recombinant mAb and the three murine mAbs. EGF_PfMSP4-specific affinities were determined by SPR spectroscopy to 8 nM and 10 nM for the murine or recombinant mAb, respectively. Binding to parasite PfMSP4 was confirmed in an immunofluorescence assay showing a characteristic staining pattern and by western blot analysis using merozoite extract. CONCLUSIONS As demonstrated by the example of an EGF_PfMSP4-specific antibody, the described combination of a simple and efficient hybridoma antibody cloning approach with the flexible, robust and cost-efficient transient expression system suitable to rapidly produce mg-amounts of functional recombinant antibodies provides an attractive method for the generation of mAbs and their derivatives as research tool, novel therapeutics or diagnostics.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Protozoan/genetics
- Antibodies, Protozoan/immunology
- Antibodies, Protozoan/isolation & purification
- Antigens, Protozoan/immunology
- Blotting, Western
- Enzyme-Linked Immunosorbent Assay
- Gene Expression
- Humans
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Immunoglobulin Variable Region/isolation & purification
- Mice, Inbred BALB C
- Microscopy, Fluorescence
- Organisms, Genetically Modified/genetics
- Organisms, Genetically Modified/metabolism
- Protozoan Proteins/immunology
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/isolation & purification
- Surface Plasmon Resonance
- Nicotiana/genetics
- Nicotiana/metabolism
Collapse
Affiliation(s)
- Stephanie Kapelski
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
- RWTH Aachen University, Institute for Molecular Biotechnology, Worringer Weg 1, 52074, Aachen, Germany.
- Institute for Applied Medical Engineering at RWTH Aachen University and Hospital, Department of Experimental Medicine and Immunotherapy, Pauwelsstraße 20, 52074, Aachen, Germany.
| | - Alexander Boes
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - Melanie de Almeida
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
- RWTH Aachen University, Institute for Molecular Biotechnology, Worringer Weg 1, 52074, Aachen, Germany.
| | - Torsten Klockenbring
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - Andreas Reimann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
- RWTH Aachen University, Institute for Molecular Biotechnology, Worringer Weg 1, 52074, Aachen, Germany.
| | - Stefan Barth
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
- Institute for Applied Medical Engineering at RWTH Aachen University and Hospital, Department of Experimental Medicine and Immunotherapy, Pauwelsstraße 20, 52074, Aachen, Germany.
| | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstraße 6, 52074, Aachen, Germany.
- RWTH Aachen University, Institute for Molecular Biotechnology, Worringer Weg 1, 52074, Aachen, Germany.
- Institute for Applied Medical Engineering at RWTH Aachen University and Hospital, Department of Experimental Medicine and Immunotherapy, Pauwelsstraße 20, 52074, Aachen, Germany.
| |
Collapse
|
16
|
Baum V, Bühler P, Gierschner D, Herchenbach D, Fiala GJ, Schamel WW, Wolf P, Elsässer-Beile U. Antitumor activities of PSMA×CD3 diabodies by redirected T-cell lysis of prostate cancer cells. Immunotherapy 2013; 5:27-38. [PMID: 23256796 DOI: 10.2217/imt.12.136] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Although prostate cancer is one of the most commonly diagnosed malignancies in men, there is no effective curative therapy for the advanced disease. Therefore, the aim of the present study was to generate prostate-specific membrane antigen (PSMA)×CD3 diabodies as a novel treatment option for this tumor. METHODS A PSMA×CD3 diabody and a covalently linked single-chain diabody were constructed from the anti-PSMA single-chain Fv fragment D7 and an anti-CD3 single-chain Fv fragment. The fusion proteins were periplasmatically expressed in Escherichia coli. The binding properties were tested on PSMA-expressing C4-2 prostate cancer cells and CD3(+) Jurkat cells by flow cytometry. For in vitro functional analysis, a cell viability assay was used. T-cell activation was determined by flow cytometry. In vivo activity of the diabody was tested in SCID mice reconstituted with human peripheral blood lymphocytes bearing C4-2 tumor xenografts. RESULTS Bacterial expression levels were significantly higher for the diabody (1-1.5 mg/l culture) compared with the single-chain diabody (0.2-0.4 mg/l culture). Specific binding on CD3-expressing Jurkat cells and PSMA-expressing C4-2 cells was shown with both diabody formats. In vitro, both diabodies proved to be potent agents for retargeting human CD4(+) and CD8(+) lymphocytes to lyse C4-2 prostate cancer cells. The formation of conjugates between T cells and target cells with clustering of the diabody at sites of interaction could be shown. SCID mice reconstituted with human peripheral blood lymphocytes bearing C4-2 tumor xenografts with the diabody showed an efficient inhibition of tumor growth. CONCLUSION Both diabody formats showed a highly efficient and specific T cell-mediated killing of prostate cancer cells and are encouraging for further development in preclinical and clinical studies.
Collapse
Affiliation(s)
- Volker Baum
- Department of Urology, Experimental Urology, University Hospital, University of Freiburg, Breisacher Street 117, 79106 Freiburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Corrada D, Morra G, Colombo G. Investigating allostery in molecular recognition: insights from a computational study of multiple antibody-antigen complexes. J Phys Chem B 2013; 117:535-52. [PMID: 23240736 DOI: 10.1021/jp310753z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibody-antigen recognition plays a key role in the immune response against pathogens. Here, we have investigated various aspects of this problem by analyzing a large and diverse set of antibodies and their respective complexes with protein antigens through atomistic simulations. Common features of antibody response to the presence of antigens are elucidated by the analysis of the proteins' internal dynamics and coordination in different ligand states, combined with the analysis of the interaction networks implicated in the stabilization of functional structures. The use of a common structural reference reveals preferential changes in the dynamic coordination and intramolecular interaction networks induced by antigen binding and shared by all antibodies. Such changes propagate from the binding region through the whole immunoglobulin domains. Overall, complexed antibodies show more diffuse networks of nonbonded interactions and a general higher internal dynamic coordination, which preferentially involve the immunoglobulin (Ig) domains of the heavy chain. The combined results provide atomistic insights into the correlations between the modulation of conformational dynamics, structural stability, and allosteric signal transduction. In particular, the results suggest that specific networks of residues, shared among all the analyzed proteins, define the molecular pathways by which antibody structures respond to antigen binding. Our studies may have implications in practical use, such as the rational design of antibodies with specifically modulated antigen-binding affinities.
Collapse
Affiliation(s)
- Dario Corrada
- Istituto di Chimica del Riconoscimento Molecolare - Consiglio Nazionale delle Ricerche (CNR-ICRM), via Mario Bianco 9, 20131 Milano, Italy
| | | | | |
Collapse
|
18
|
Friedrich M, Raum T, Lutterbuese R, Voelkel M, Deegen P, Rau D, Kischel R, Hoffmann P, Brandl C, Schuhmacher J, Mueller P, Finnern R, Fuergut M, Zopf D, Slootstra JW, Baeuerle PA, Rattel B, Kufer P. Regression of human prostate cancer xenografts in mice by AMG 212/BAY2010112, a novel PSMA/CD3-Bispecific BiTE antibody cross-reactive with non-human primate antigens. Mol Cancer Ther 2012; 11:2664-73. [PMID: 23041545 DOI: 10.1158/1535-7163.mct-12-0042] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For treatment of patients with prostate cancer (PCa), we developed a novel T cell-engaging (BiTE) antibody designated AMG 212 or BAY2010112 that is bispecific for prostate-specific membrane antigen (PSMA) and the CD3 epsilon subunit of the T cell receptor complex. AMG 212/BAY2010112 induced target cell-dependent activation and cytokine release of T cells, and efficiently redirected T cells for lysis of target cells. In addition to Chinese hamster ovary cells stably expressing human or cynomolgus monkey PSMA, T cells redirected by AMG 212/BAY2010112 also lysed human PCa cell lines VCaP, 22Rv1, MDA PCa 2b, C4-2, PC-3-huPSMA, and LnCaP at half maximal BiTE concentrations between 0.1 and 4 ng/mL (1.8-72 pmol/L). No lysis of PSMA-negative human PCa cell lines PC-3 and DU145 was observed. The subcutaneous (s.c.) formation of tumors from PC-3-huPSMA cells in NOD/SCID mice was significantly prevented by once daily intravenous (i.v.) injection of AMG 212/BAY2010112 at a dose level as low as 0.005 mg/kg/d. Rapid tumor shrinkage with complete remissions were observed in NOD/SCID mice bearing established s.c. 22Rv1 xenografts after repeated daily treatment with AMG 212/BAY2010112 by either the i.v. or s.c. route. Of note, 22Rv1 tumors were grown in the absence of human T cells followed by intraperitoneal injection of T cells 3 days before BiTE treatment. No effects on tumor growth were observed in the absence of human T cells or AMG 212/BAY2010112. On the basis of these preclinical results, AMG 212/BAY2010112 appears as a promising new BiTE antibody for the treatment of patients with PSMA-expressing PCa.
Collapse
|
19
|
Le Gall F, Reusch U, Bakulina AY, Kiprijanov SM. WITHDRAWN: Tetravalent Bispecific Single-Chain Fv Antibodies for Lysis of Leukemia Cells by Autologous T Cells. J Mol Biol 2012:S0022-2836(12)00741-3. [PMID: 22985968 DOI: 10.1016/j.jmb.2012.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 08/26/2012] [Accepted: 08/30/2012] [Indexed: 11/17/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Fabrice Le Gall
- Affimed Therapeutics AG, Im Neuenheimer Feld 582, Technologiepark, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
20
|
Abstract
Despite the rising impact of the generation of antibodies by phage display and other technologies, hybridoma technology still provides a valuable tool for the generation of high-affinity binders against different targets. But there exist several limitations of using hybridoma-derived antibodies. The source of the hybridoma clones are mostly rat or mouse B-lymphocytes. Therefore a human-anti-mouse or human-anti-rat antibody response may result in immunogenicity of these antibodies. This leads to the necessity of humanization of these antibodies where the knowledge of the amino acid sequence of the proteins is inalienable. Furthermore, additional in vitro modifications, e.g., affinity maturation or fusion to other proteins, are dependent on cloning of the antigen-binding domains.Here we describe the isolation of RNA from hybridoma cells and the primers that can be used for the amplification of VL and VH as well as the cloning of the antibody in scFv format and its expression in Escherichia coli.
Collapse
Affiliation(s)
- Lars Toleikis
- Protein Engineering and Antibody Technologies NBE Technologies, Merck Serono, Merck KGaA, Darmstadt, Germany
| | | |
Collapse
|
21
|
Wang T, Duan Y. Probing the stability-limiting regions of an antibody single-chain variable fragment: a molecular dynamics simulation study. Protein Eng Des Sel 2011; 24:649-57. [PMID: 21729946 DOI: 10.1093/protein/gzr029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Antibody single-chain variable fragments (scFvs) offer particular advantages over the full-size antibodies, including easy expression, efficient local concentration and fast body clearance. However, scFvs typically show low thermal stability that limits their biomedical and biotechnological applications. In this study, we examined the thermal stability of the human and murine vascular endothelial growth factor antibody scFv fragment by molecular dynamics simulations. A consistent observation was the dissociation of the light-chain (VL) and heavy-chain (VH) domains and loss of the native structures of both domains in the simulations at the elevated temperatures. The stability-limiting structural elements in the protein were revealed from the detailed analyses on the native contacts. We found that dissociation of the VL-VH domains was the first event leading to the unfolding of the native structure of the protein and the disruption of the VL-VH interface was largely due to the break of the interfacial hydrophobic and aromatic interactions while the hydrogen-bonding interaction between Gln38 in VL and Gln39 in VH remained. Within the β-barrel structure of the VL and VH domains, β-strands β6, β2 and β11 appeared to be the least stable. In addition, we found that the VH domain was more thermally resistant than the VL domain. Based on these findings, we discussed potential strategies to improve the stability of this therapeutically important scFv fragment.
Collapse
Affiliation(s)
- Ting Wang
- Department of Applied Science, University of California, Davis, CA 95616-8816, USA
| | | |
Collapse
|
22
|
Fortmüller K, Alt K, Gierschner D, Wolf P, Baum V, Freudenberg N, Wetterauer U, Elsässer-Beile U, Bühler P. Effective targeting of prostate cancer by lymphocytes redirected by a PSMA × CD3 bispecific single-chain diabody. Prostate 2011; 71:588-96. [PMID: 20945402 DOI: 10.1002/pros.21274] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Accepted: 08/24/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND For redirecting T-lymphocytes to induce prostate cancer cell lysis, we constructed a novel bispecific single-chain (bsc) diabody directed to the prostate specific membrane antigen (PSMA) and the T-cell receptor (TCR)-associated CD3 molecule on T-cells. METHODS The PSMA × CD3 bsc diabody was generated from an anti-CD3 single chain Fv fragment (scFv) and the anti-PSMA scFv D7. It was expressed in E. coli and purified from the periplasmic extract and culture supernatant by immobilized metal affinity chromatography (IMAC). The binding properties were tested on PSMA-expressing prostate cancer cells and PSMA-negative cell lines as well as on Jurkat cells by flow cytometry. For in vitro functional analysis, a cell viability test (WST-1) was used and activation of T-cells was determined by measuring the surface marker expression of CD25 and CD69. For in vivo evaluation, the diabody was administered in combination with human peripheral blood lymphocytes (Ly) in a C4-2 xenograft-SCID mouse model. RESULTS Specific binding of the PSMA × CD3 bsc diabody both to CD3-positive Jurkat cells and PSMA-expressing C4-2 cells was shown by flow cytometry. In vitro, the PSMA × CD3 bsc diabody proved to be a potent agent for retargeting CD4+ and CD8+ human lymphocytes to lyse C4-2 prostate cancer cells. Treatment of SCID mice bearing C4-2 tumor xenografts with the diabody and human lymphocytes efficiently inhibited tumor growth. CONCLUSIONS The PSMA × CD3 bsc diabody bears a high potential for the immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Kerstin Fortmüller
- Department of Urology, Experimental Urology, University Hospital Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Pai JC, Culver JA, Drury JE, Motani RS, Lieberman RL, Maynard JA. Conversion of scFv peptide-binding specificity for crystal chaperone development. Protein Eng Des Sel 2011; 24:419-28. [PMID: 21217145 PMCID: PMC3077810 DOI: 10.1093/protein/gzq120] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 11/18/2010] [Accepted: 11/26/2010] [Indexed: 12/26/2022] Open
Abstract
In spite of advances in protein expression and purification over the last decade, many proteins remain recalcitrant to structure determination by X-ray crystallography. One emerging tactic to obtain high-quality protein crystals for structure determination, particularly in the case of membrane proteins, involves co-crystallization with a protein-specific antibody fragment. Here, we report the development of new recombinant single-chain antibody fragments (scFv) capable of binding a specific epitope that can be introduced into internal loops of client proteins. The previously crystallized hexa-histidine-specific 3D5 scFv antibody was modified in the complementary determining region and by random mutagenesis, in conjunction with phage display, to yield scFvs with new biochemical characteristics and binding specificity. Selected variants include those specific for the hexa-histidine peptide with increased expression, solubility (up to 16.6 mg/ml) and sub-micromolar affinity, and those with new specificity for the EE hexa-peptide (EYMPME) and nanomolar affinity. Complexes of one such chaperone with model proteins harboring either an internal or a terminal EE tag were isolated by gel filtration. The 3.1 Å resolution structure of this chaperone reveals a binding surface complementary to the EE peptide and a ∼52 Å channel in the crystal lattice. Notably, in spite of 85% sequence identity, and nearly identical crystallization conditions, the engineered scFv crystallizes in a different space group than the parent 3D5 scFv, and utilizes two new crystal contacts. These engineered scFvs represent a new class of chaperones that may eliminate the need for de novo identification of candidate chaperones from large antibody libraries.
Collapse
Affiliation(s)
- Jennifer C. Pai
- Department of Chemical Engineering, University of Texas at Austin, MC0400, 1 University Station, Austin TX 78712, USA
| | - Jeffrey A. Culver
- School of Chemistry and Biochemistry and Institute for Bioscience and Bioengineering, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
| | - Jason E. Drury
- School of Chemistry and Biochemistry and Institute for Bioscience and Bioengineering, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
| | - Rakesh S. Motani
- Department of Microbial Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Raquel L. Lieberman
- School of Chemistry and Biochemistry and Institute for Bioscience and Bioengineering, Georgia Institute of Technology, 901 Atlantic Drive NW, Atlanta, GA 30332, USA
| | - Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, MC0400, 1 University Station, Austin TX 78712, USA
| |
Collapse
|
24
|
Antibody engineering using phage display with a coiled-coil heterodimeric Fv antibody fragment. PLoS One 2011; 6:e19023. [PMID: 21552519 PMCID: PMC3084267 DOI: 10.1371/journal.pone.0019023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/14/2011] [Indexed: 11/19/2022] Open
Abstract
A Fab-like antibody binding unit, ccFv, in which a pair of heterodimeric coiled-coil domains was fused to VH and VL for Fv stabilization, was constructed for an anti-VEGF antibody. The anti-VEGF ccFv showed the same binding affinity as scFv but significantly improved stability and phage display level. Furthermore, phage display libraries in the ccFv format were constructed for humanization and affinity maturation of the anti-VEGF antibody. A panel of VH frameworks and VH-CDR3 variants, with a significant improvement in affinity and expressibility in both E. coli and yeast systems, was isolated from the ccFv phage libraries. These results demonstrate the potential application of the ccFv antibody format in antibody engineering.
Collapse
|
25
|
Erlandsson A, Holm P, Jafari R, Stigbrand T, Sundström BE. Functional mapping of the anti-idiotypic antibody anti-TS1 scFv using site-directed mutagenesis and kinetic analysis. MAbs 2010; 2:662-9. [PMID: 21124071 DOI: 10.4161/mabs.2.6.13275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recombinant antibodies may be engineered to obtain improved functional properties. Functional mapping of the residues in the binding surfaces is of importance for predicting alterations needed to yield the desired properties. In this investigation, 17 single mutation mutant single-chain variable fragments (scFvs) of the anti-idiotypic antibody anti-TS1 were generated in order to functionally map amino acid residues important for the interaction with its idiotype TS1. Residues in anti-TS1 determined to be very important for the interaction were identified, Y32L, K50L, K33H, and Y52H, and they were distributed adjacent to a centrally located hydrophobic area, and contributed extensively to the interaction energy (≥2.5 kcal/mol) in the interaction. Quantitative ELISA assays, BIAcore technologies and three-dimensional surface analysis by modeling were employed to visualize the consequences of the mutations. The expression levels varied between 2 - 1,800 nM as determined by ELISA. All the 17 scFvs displayed higher dissociation rates (60 - 1,300 times) and all but two of them also faster association rates (1.3 - 56 times). The decrease in affinity was determined to be 1.6 - 12,200 times. Two of the mutants displayed almost identical affinity with the wild type anti-TS1, but with a change in both association and dissociation rates. The present investigation demonstrates that it is possible to generate a large panorama of anti-idiotypic antibodies, and single out a few that might be of potential use for future clearing and pre-targeting purposes of idiotypic-anti-idiotypic interactions.
Collapse
Affiliation(s)
- Ann Erlandsson
- Department of Chemistry and Biomedical Sciences, Karlstad University, Karlstad, Sweden
| | | | | | | | | |
Collapse
|
26
|
Cheng WW, Allen TM. The use of single chain Fv as targeting agents for immunoliposomes: an update on immunoliposomal drugs for cancer treatment. Expert Opin Drug Deliv 2010; 7:461-78. [PMID: 20331354 PMCID: PMC4006819 DOI: 10.1517/17425240903579963] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPORTANCE OF THE FIELD Targeted liposomal drugs represent the next evolution of liposomal drug delivery in cancer treatment. In various preclinical cancer models, antibody-targeted PEGylated liposomal drugs have demonstrated superior therapeutic effects over their non-targeted counterparts. Single chain Fv (scFv) has gained popularity in recent years as the targeting agent of choice over traditional targeting agents such as monoclonal antibodies (mAb) and antibody fragments (e.g., Fab'). AREAS COVERED IN THIS REVIEW This review is focused mainly on advances in scFv-targeted liposomal drug delivery for the treatment of cancers, based on a survey of the recent literature, and on experiments done in a murine model of human B-lymphoma, using anti-CD19 targeted liposomes targeted with whole mAb, Fab' fragments and scFv fragments. WHAT THE READER WILL GAIN This review examines the recent advances in PEGylated immunoliposomal drug delivery, focusing on scFv fragments as targeting agents, in comparison with Fab' and mAb. TAKE HOME MESSAGE For clinical development, scFv are potentially preferred targeting agents for PEGylated liposomes over mAb and Fab', owing to factors such as decreased immunogenicity, and pharmacokinetics/biodistribution profiles that are similar to non-targeted PEGylated (Stealth) liposomes.
Collapse
Affiliation(s)
- W W Cheng
- Centre for Drug Research & Development, 364-2259 Lower Mall, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
27
|
Min WK, Cho YJ, Park JB, Bae YH, Kim EJ, Park K, Park YC, Seo JH. Production and characterization of monoclonal antibody and its recombinant single chain variable fragment specific for a food-born mycotoxin, fumonisin B1. Bioprocess Biosyst Eng 2009; 33:109-15. [DOI: 10.1007/s00449-009-0350-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2009] [Accepted: 06/23/2009] [Indexed: 11/28/2022]
|
28
|
Honegger A, Malebranche AD, Röthlisberger D, Plückthun A. The influence of the framework core residues on the biophysical properties of immunoglobulin heavy chain variable domains. Protein Eng Des Sel 2009; 22:121-34. [DOI: 10.1093/protein/gzn077] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
29
|
Abstract
Conventionally, antibody phage display has been used to isolate recombinant antibodies that are monovalent in their interaction with target antigens. These antibodies can be reengineered for expression in mammalian cell culture as full-length, monospecific immunoglobulins. An emerging branch of research has sought to generate bivalent recombinant antibodies by manipulating the length of the linker separating heavy- and light-chain variable domains in single-chain Fv proteins, thereby promoting inter-scFv interaction and the formation of "diabodies." With careful control, this can generate scFv-based proteins able to bind two very distinct targets, "bispecific diabodies." Further manipulation enables the assembly of higher-order complexes.
Collapse
|
30
|
Scott N, Reynolds CB, Wright MJ, Qazi O, Fairweather N, Deonarain MP. Single-chain Fv phage display propensity exhibits strong positive correlation with overall expression levels. BMC Biotechnol 2008; 8:97. [PMID: 19113995 PMCID: PMC2630973 DOI: 10.1186/1472-6750-8-97] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 12/29/2008] [Indexed: 01/09/2023] Open
Abstract
Background Single chain Fvs (scFvs) are widely applied in research, diagnostics and therapeutic settings. Display and selection from combinatorial libraries is the main route to their discovery and many factors influence the success of this process. They exhibit low thermodynamic stability, resulting in low levels of premature cytosolic folding or aggregation which facilitates sec YEG-mediated translocation and phage in E. coli. However, there is little data analysing how this is related to and influenced by scFv protein expression. Results We characterised the relationship between overall scFv expression and display propensity for a panel of 15 anti-tetanus toxin scFvs and found a strong positive correlation (Rho = 0.88, p < 0.005) between the two parameters. Display propensity, overall expression and soluble localisation to the periplasm and extracellular fractions were clone specific characteristics which varied despite high levels of sequence homology. There was no correlation between display of scFv or its expression in non-fused (free) form with soluble scFv localisation to the periplasm or culture supernatant. This suggests that divergence in the fate of scFv-pIII and non-fused scFv after translocation to the periplasm accounts for the observed disparity. Differential degrees of periplasmic aggregation of non-fused scFv between clones may affect the partitioning of scFv in the periplasm and culture supernatant abrogating any correlation. We suggest that these factors do not apply to the scFv-pIII fusion since it remains anchored to the bacterial inner membrane as part of the innate phage packaging and budding process. Conclusion We conclude that in the absence of premature cytosolic aggregation or folding, the propensity of a scFv to be displayed on phage is directly related to its overall expression level and is thus indirectly influenced by factors such as codon bias, mRNA abundance or putative DNA motifs affecting expression. This suggests that scFvs capable of high overall expression and display levels may not produce high yields of non phage-fused soluble protein in either the periplasmic or extracellular fractions of E. coli. This should be considered when screening clones selected from combinatorial libraries for further study. The nucleotide and amino acid sequences of the anti-tetanus toxin scFvs have been deposited in the EMBL data base: accession numbers-C1: AM749134, C2: AM749135, C3: AM749136, C4: AM749137, C5: AM749138, N1: AM749139, N2: AM749140, N3: AM749141, N4: AM749142, N5: AM749143 J1; AM749144, J2: AM749145, J3: AM749146, J4: AM749147, J5: AM749148.
Collapse
Affiliation(s)
- Nathan Scott
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, Exhibition Road, London, SW7 2AZ, UK.
| | | | | | | | | | | |
Collapse
|
31
|
Develter J, Booth NA, Declerck PJ, Gils A. Bispecific targeting of thrombin activatable fibrinolysis inhibitor and plasminogen activator inhibitor-1 by a heterodimer diabody. J Thromb Haemost 2008; 6:1884-91. [PMID: 18752580 DOI: 10.1111/j.1538-7836.2008.03137.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVES Thrombin activatable fibrinolysis inhibitor (TAFI) and plasminogen activator inhibitor-1 (PAI-1) play important roles in fibrinolysis. Both reduce plasmin generation, but they exert their antifibrinolytic effects via different mechanisms. This study reports the cloning and characterization of a heterodimer diabody that inhibits TAFI and PAI-1 simultaneously. METHODS AND RESULTS The diabody was derived from two inhibiting monoclonal antibodies, i.e. MA-33H1F7, an anti-PAI-1 antibody that induces non-inhibitory substrate behavior of PAI-1, and MA-T12D11, an anti-TAFI antibody that inhibits activation of TAFI by the thrombin-thrombomodulin complex. A single-chain variable fragment (scFv) was derived from MA-T12D11 that displayed slightly reduced binding and inhibitory properties as compared to MA-T12D11. Characterization of the diabody revealed a similar affinity for TAFI and PAI-1 as that of the parental antibodies. Furthermore, the inhibitory properties of MA-33H1F7 and MA-T12D11 were fully preserved in the diabody format. In platelet-free plasma (PFP) clots, addition of the diabody had a stronger effect in shortening lysis times than either MA-T12D11 or MA-33H1F7. A similar reduction in clot lysis time was observed in platelet-rich plasma (PRP) clots. The same effect on clot lysis times in PFP and PRP was also achieved by the combined addition of MA-T12D11 and MA-33H1F7. The lysis rate of human model thrombi, made from whole blood, was approximately doubled after addition of the diabody. Moreover, this effect was significantly better than after the combined addition of the individual antibodies. CONCLUSIONS These observations demonstrate that simultaneous inhibition of TAFI and PAI-1 results in faster lysis of the formed thrombus.
Collapse
Affiliation(s)
- J Develter
- Laboratory for Pharmaceutical Biology, Faculty of Pharmaceutical Sciences, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
32
|
Eder M, Wängler B, Knackmuss S, LeGall F, Little M, Haberkorn U, Mier W, Eisenhut M. Tetrafluorophenolate of HBED-CC: a versatile conjugation agent for 68Ga-labeled small recombinant antibodies. Eur J Nucl Med Mol Imaging 2008; 35:1878-86. [DOI: 10.1007/s00259-008-0816-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 04/18/2008] [Indexed: 11/25/2022]
|
33
|
Thorpe SJ, Ball C, Fox B, Thompson KM, Thorpe R, Bristow A. Anti-D and anti-i activities are inseparable in V4-34-encoded monoclonal anti-D: the same framework 1 residues are required for both reactivities. Transfusion 2008. [DOI: 10.1111/j.1537-2995.2007.01624.x-i2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
34
|
Thorpe SJ, Ball C, Fox B, Thompson KM, Thorpe R, Bristow A. Anti-D and anti-i activities are inseparable in V4-34-encoded monoclonal anti-D: the same framework 1 residues are required for both reactivities. Transfusion 2008; 48:930-40. [PMID: 18346025 DOI: 10.1111/j.1537-2995.2007.01624.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The heavy-chain V4-34 germline gene segment is mandatory for pathologic cold-reacting autoantibodies with anti-I/i specificity (cold agglutinins) and is also preferentially used by monoclonal immunoglobulin M alloantibodies against D and other Rh antigens. The use of the V4-34 segment by monoclonal anti-D has previously been shown to also confer anti-I/i reactivity (cold agglutinin activity), which has implications for the use of such antibodies for Rh blood typing. V4-34 framework 1 (FR1) sequence is believed to be critical for cold agglutinin activity of cold agglutinins. STUDY DESIGN AND METHODS The aim of this investigation was to use site-directed mutagenesis of a recombinant V4-34-encoded anti-D to determine the contribution of V4-34 FR1 sequence to anti-D activity and whether mutational modifications in the FR1 region could separately alter anti-D and anti-i activities. RESULTS The results show that amino acid changes in V4-34 FR1 at W7, A23, and Y25 have a profound effect on anti-D activity as well as on anti-i activity. It was not possible to substantially reduce or remove anti-i activity without reducing anti-D activity to a comparable extent. CONCLUSIONS The same nonpolar hydrophobic amino acids in FR1 are critical for maintaining both anti-D and anti-i activity. It is proposed that these residues influence the conformation of the antigen-binding site.
Collapse
Affiliation(s)
- Susan J Thorpe
- Biotherapeutics Group and Technology Development and Infrastructure Group, National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, UK.
| | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Antibody Phage Display. SPRINGER PROTOCOLS HANDBOOKS 2008. [PMCID: PMC7123299 DOI: 10.1007/978-1-60327-375-6_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
37
|
Asano R, Sone Y, Makabe K, Tsumoto K, Hayashi H, Katayose Y, Unno M, Kudo T, Kumagai I. Humanization of the bispecific epidermal growth factor receptor x CD3 diabody and its efficacy as a potential clinical reagent. Clin Cancer Res 2007; 12:4036-42. [PMID: 16818703 DOI: 10.1158/1078-0432.ccr-06-0059] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Bispecific antibodies (BsAb) have been exploited as both cancer immunodiagnostics and cancer therapeutics and show promise in clinical trials of cancer imaging and therapy. For development of BsAbs as clinical reagents, we have focused on construction of small recombinant BsAbs, called bispecific diabodies. Here, we constructed and characterized a humanized bispecific diabody. EXPERIMENTAL DESIGN We have reported significant antitumor activity of an anti-epidermal growth factor receptor (EGFR) x anti-CD3 bispecific diabody (Ex3) in in vitro cytotoxicity assays and in vivo. We humanized the Ex3 diabody (hEx3) by grafting the complementarity-determining region and compared its biological properties with those of Ex3. We also tested its physiologic stability and ability to alter survival in xenografted mice. RESULTS The final yield of hEx3 was 10 times that of Ex3, and refolded hEx3 and Ex3 showed identical binding profiles in EGFR-positive cell lines and EGFR-transfected Chinese hamster ovary cells. hEx3 showed dose-dependent cytotoxicity to EGFR-positive cell lines, which could be specifically inhibited by parental monoclonal antibody IgGs against EGFR or CD3 antigens. The heterodimeric structure was retained in PBS for 6 months, and growth inhibition was maintained after incubation under physiologic conditions. Coadministration of hEx3 with T-LAK cells and interleukin-2 prolonged the survival of nude mice with human colon carcinoma. CONCLUSIONS The humanized diabody hEx3 is an attractive molecule for cancer therapy and may provide important insights into the development of EGFR-based cancer-targeting reagents.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/therapeutic use
- Antigen-Antibody Reactions
- Antineoplastic Agents/immunology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- CD3 Complex/immunology
- CHO Cells
- Carcinoma/drug therapy
- Carcinoma/immunology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cloning, Molecular
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/immunology
- Cricetinae
- Cricetulus
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- ErbB Receptors/immunology
- Flow Cytometry
- Humans
- Mice
- Mice, Nude
- Protein Binding
- Transplantation, Heterologous
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ryutaro Asano
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bühler P, Wolf P, Gierschner D, Schaber I, Katzenwadel A, Schultze-Seemann W, Wetterauer U, Tacke M, Swamy M, Schamel WWA, Elsässer-Beile U. A bispecific diabody directed against prostate-specific membrane antigen and CD3 induces T-cell mediated lysis of prostate cancer cells. Cancer Immunol Immunother 2007; 57:43-52. [PMID: 17579857 PMCID: PMC2755730 DOI: 10.1007/s00262-007-0348-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 05/23/2007] [Indexed: 12/03/2022]
Abstract
Background Although cancer of the prostate is one of the most commonly diagnosed cancers in men, no curative treatment currently exists after its progression beyond resectable boundaries. Therefore, new agents for targeted treatment strategies are needed. Cross-linking of tumor antigens with T-cell associated antigens by bispecific monoclonal antibodies have been shown to increase antigen-specific cytotoxicity in T-cells. Since the prostate-specific membrane antigen (PSMA) represents an excellent tumor target, immunotherapy with bispecific diabodies could be a promising novel treatment option for prostate cancer. Methods A heterodimeric diabody specific for human PSMA and the T-cell antigen CD3 was constructed from the DNA of anti-CD3 and anti-PSMA single chain Fv fragments (scFv). It was expressed in E. coli using a vector containing a bicistronic operon for co-secretion of the hybrid scFv VHCD3-VLPSMA and VHPSMA-VLCD3. The resulting PSMAxCD3 diabody was purified from the periplasmic extract by immobilized metal affinity chromatography (IMAC). The binding properties were tested on PSMA-expressing prostate cancer cells and PSMA-negative cell lines as well as on Jurkat cells by flow cytometry. For in vitro functional analysis, a cell viability test (WST) was used. For in vivo evaluation the diabody was applied together with human peripheral blood lymphocytes (PBL) in a C4-2 xenograft-SCID mouse model. Results By Blue Native gel electrophoresis, it could be shown that the PSMAxCD3 diabody is mainly a tetramer. Specific binding both to CD3-expressing Jurkat cells and PSMA-expressing C4-2 cells was shown by flow cytometry. In vitro, the diabody proved to be a potent agent for retargeting PBL to lyze C4-2 prostate cancer cells. Treatment of SCID mice inoculated with C4-2 tumor xenografts with the diabody and PBL efficiently inhibited tumor growth. Conclusions The PSMAxCD3 diabody bears the potential for facilitating immunotherapy of prostate cancer and for the elimination of minimal residual disease.
Collapse
Affiliation(s)
- P. Bühler
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzle Str. 1, 79104 Freiburg, Germany
| | - P. Wolf
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - D. Gierschner
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - I. Schaber
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - A. Katzenwadel
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - W. Schultze-Seemann
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - U. Wetterauer
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - M. Tacke
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Stefan-Meier-Strasse 17, 79106 Freiburg, Germany
| | - M. Swamy
- Department of Molecular Immunology, Max-Planck-Institute for Immunobiology and University of Freiburg, Biologie III, Stübeweg 51, 79108 Freiburg, Germany
| | - W. W. A. Schamel
- Department of Molecular Immunology, Max-Planck-Institute for Immunobiology and University of Freiburg, Biologie III, Stübeweg 51, 79108 Freiburg, Germany
| | - U. Elsässer-Beile
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| |
Collapse
|
39
|
Gasser B, Mattanovich D. Antibody production with yeasts and filamentous fungi: on the road to large scale? Biotechnol Lett 2006; 29:201-12. [PMID: 17120087 DOI: 10.1007/s10529-006-9237-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 10/11/2006] [Accepted: 10/11/2006] [Indexed: 10/23/2022]
Abstract
Yeasts and filamentous fungi have gained significant interest for the production of recombinant antibodies and antibody fragments. The opportunities and constraints of antibody (fragment) production in these hosts are highlighted as well as cell engineering strategies to overcome the constraints. Following aspects are addressed: folding, assembly and secretion of antibody related proteins, process optimization to improve productivity and quality, proteolysis, and, as a major point of interest, glycosylation.
Collapse
Affiliation(s)
- Brigitte Gasser
- Institute of Applied Microbiology, Department of Biotechnology, University of Natural Resources and Applied Life Sciences, Vienna, Austria
| | | |
Collapse
|
40
|
Johansson DX, Drakenberg K, Hopmann KH, Schmidt A, Yari F, Hinkula J, Persson MAA. Efficient expression of recombinant human monoclonal antibodies in Drosophila S2 cells. J Immunol Methods 2006; 318:37-46. [PMID: 17137589 DOI: 10.1016/j.jim.2006.08.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/25/2006] [Accepted: 08/28/2006] [Indexed: 11/24/2022]
Abstract
We have explored the Drosophila S2 cell line for expression of Ig molecules isolated as Fab or scFv cDNA from phage-displayed libraries. We present a series of vectors for inducible expression and secretion of human Ig heavy (HC) and light chains (LC), both on separate plasmids and in combination constructs. Both HC (tested as human gamma(1)) and LC (human kappa) could be expressed separately and were secreted into the medium, confirming previous reports. When the combination vector carrying both the HC and LC cDNA, as well as when the HC and LC vectors were co-transfected, complete IgG1 was found in the medium. Transient transfection resulted in production levels of 0.5-1 mg/l. Stable cell lines could be established within 2-3 weeks. After 10-12 days of expression from such cell lines, Ig molecules accumulated and the medium contained typically 5-35 mg/l of IgG1. The IgG in these preparations was purified to more than 90% purity on protein G columns. Binding characteristics for IgG of the same clone expressed in S2 cells or mammalian cells were indistinguishable. The main advantages with this system compared to mammalian expression were its robustness and the much faster establishment of stable, high level producing cell lines.
Collapse
Affiliation(s)
- Daniel X Johansson
- Karolinska Institutet, Department of Medicine at Center for Molecular Medicine, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
41
|
Wark KL, Hudson PJ. Latest technologies for the enhancement of antibody affinity. Adv Drug Deliv Rev 2006; 58:657-70. [PMID: 16828920 DOI: 10.1016/j.addr.2006.01.025] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2006] [Accepted: 05/06/2006] [Indexed: 11/24/2022]
Abstract
High affinity antibodies are crucial both for the discovery and validation of biomarkers for human health and disease and as clinical diagnostic and therapeutic reagents. This review describes some of the latest technologies for the design, mutation and selection of high affinity antibodies that provide a paradigm for molecular evolution of a far wider range of proteins including enzymes. Strategies include both in vivo and in vitro methods and embrace the latest concepts for antibody display and selection. Specifically, affinity enhancement can be tailored to the target-binding surface, typically the complementary determining region (CDR) loops in antibodies, whereas enhanced stability, expression or catalytic properties can be affected by selected changes to the core protein scaffold. Together, these technologies provide a rapid and powerful strategy to drive the next generation of protein-based reagents for numerous clinical, environmental and agribusiness applications.
Collapse
Affiliation(s)
- Kim L Wark
- CRC for Diagnostics at CSIRO Molecular and Health Technologies, 343 Royal Parade, Parkville 3052, Australia.
| | | |
Collapse
|
42
|
Abstract
Maximizing the expression yields of recombinant whole antibodies and antibody fragments such as Fabs, single-chain Fvs and single-domain antibodies is highly desirable since it leads to lower production costs. Various eukaryotic and prokaryotic expression systems have been exploited to accommodate antibody expression but Escherichia coli systems have enjoyed popularity, in particular with respect to antibody fragments, because of their low cost and convenience. In many instances, product yields have been less than adequate and intrinsic and extrinsic variables have been investigated in an effort to improve yields. This review deals with various aspects of antibody expression in E. coli with a particular focus on single-domain antibodies.
Collapse
Affiliation(s)
- Mehdi Arbabi-Ghahroudi
- Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario.
| | | | | |
Collapse
|
43
|
Albrecht H, Denardo GL, Denardo SJ. Monospecific bivalent scFv-SH: effects of linker length and location of an engineered cysteine on production, antigen binding activity and free SH accessibility. J Immunol Methods 2006; 310:100-16. [PMID: 16499921 DOI: 10.1016/j.jim.2005.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2005] [Revised: 09/16/2005] [Accepted: 12/20/2005] [Indexed: 02/04/2023]
Abstract
Development of tumor targeting pharmaceuticals on a modular platform is an attractive paradigm. Design choices for bispecific (anti-tumor and anti-chelate) pretargeting molecules are increased by the use of scFvs. Because a scFv is monovalent and small in size, its functional affinity and in vivo residence time can be improved through multimerization. ScFv multimers can be covalent or non-covalent. In vivo studies indicate that covalent scFv multimers are preferable. Attachment of scFv modules to scaffolds offers a wide range of possibilities for size and valency. A free thiol introduced at the C terminal end of a scFv (scFv-SH) allows for site-specific covalent attachment to a PEG scaffold without interfering with its antigen (Ag) binding. Although in theory, multimerization of 3 or 4 scFvs can be achieved by direct conjugation, as scFv-SH, to a tri or tetrafunctionalized PEG, it is not a practical option since homogeneous tri and tetrafunctionalized PEG are not readily available. However, the generation of (scFv)(3-4)-PEG molecules through attachment of combinations of di-scFv-SH (tandemly expressed scFvs) and scFv-SH or 2 di-scFv-SH to a bifunctional PEG is a sound approach that also allows for better control of the scFv-PEG conjugate molecular composition. Optimization of the molecular format of the di-scFv-SH module for production as soluble proteins in E. coli, Ag binding and conjugation is reported in this study. ScFvs in the VH-VL format were used for the di-scFv constructs since Fv domain inversion to VL-VH, while not yielding more protein, also abolished Ag binding. The effects on production yield, Ag binding and conjugation potential of the scFv joining linker length and the presence and location of an engineered cysteine were assessed in vitro. Our data indicate that for di-scFv-SH, an increase of the scFv joining linker length results in higher production and better Ag binding; a 20 aa long linker (G(4)S)(4) was the longest linker tested. For the engineered cysteine, three locations were tested; within the scFv joining linker, at the C terminus upstream of the E Tag and as the carboxy terminal aa. The accessibility of the free SH assessed by conjugation of di-scFv-SH to HRP-Mal demonstrated that di-scFv-HRP conjugates are formed with comparable efficiencies when the cysteine is located at the scFv carboxy end. This empirical work provides a framework for the development of bispecific scFv multimers via site-specific attachment of scFv-SH and di-scFv-SH modules to a scaffold.
Collapse
Affiliation(s)
- Huguette Albrecht
- University of California Davis Medical Center, Sacramento, 95816, USA.
| | | | | |
Collapse
|
44
|
Reusch U, Le Gall F, Hensel M, Moldenhauer G, Ho AD, Little M, Kipriyanov SM. Effect of tetravalent bispecific CD19×CD3 recombinant antibody construct and CD28 costimulation on lysis of malignant B cells from patients with chronic lymphocytic leukemia by autologous T cells. Int J Cancer 2004; 112:509-18. [PMID: 15382079 DOI: 10.1002/ijc.20417] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
To develop an effective antitumor immunotherapy for B-lineage non-Hodgkin's lymphoma, we constructed a tetravalent tandem diabody (tanDb) specific for both human CD19 (B-cell marker) and CD3 (T-cell antigen). Here, we report the effective killing of malignant primary B cells from patients with B-cell chronic lymphocytic leukemia (B-CLL) by autologous T cells induced by tanDb at very low E:T ratios. Mononuclear cells from patients with B-CLL were cultured with bispecific antibody fragments in either the presence or absence of monospecific anti-CD28 antibody. Use of tetravalent tanDbs caused almost quantitative elimination of malignant B cells from the blood samples of 19 patients and some cytotoxic activity in 3 of 23 analyzed cases. In contrast, the structurally similar but bivalent diabody and single-chain diabody demonstrated nearly no antitumor activity in an autologous system. tanDb-induced activation and proliferation of T cells occurred only in the presence of CD19+ target cells. Expression of the B7-1 (CD80) and B7-2 (CD86) molecules on the surface of leukemia cells made unnecessary the additional CD28-costimulation of T cells. When only a few tanDb molecules were present, the effect of CD28 costimulation on T-cell activation was more pronounced. Depending on the patient sample, we observed a 10- to 1,000-fold decrease of the half-maximal concentrations of tanDb for cell lysis. Upon CD28 crosslinking by agonistic MAb, specific tumor cell lysis was found at tanDb concentrations as low as 0.5 pM. These data demonstrate that the tetravalent CD19xCD3 tanDb might be a promising tool for the immunotherapy of human B-cell leukemias and lymphomas.
Collapse
MESH Headings
- Aged
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- Antibodies, Monoclonal/immunology
- Antigens, CD/metabolism
- Antigens, CD19/immunology
- B7-1 Antigen/metabolism
- B7-2 Antigen
- CD28 Antigens/immunology
- CD3 Complex/immunology
- Cytotoxicity, Immunologic
- Female
- Humans
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphocyte Activation
- Male
- Membrane Glycoproteins/metabolism
- Middle Aged
- Recombinant Proteins/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Uwe Reusch
- Affimed Therapeutics, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Arndt MAE, Krauss J, Schwarzenbacher R, Vu BK, Greene S, Rybak SM. Generation of a highly stable, internalizing anti-CD22 single-chain Fv fragment for targeting non-Hodgkin's lymphoma. Int J Cancer 2004; 107:822-9. [PMID: 14566834 DOI: 10.1002/ijc.11451] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The generation of a single chain Fv (scFv) fragment derived from the anti-CD22 monoclonal antibody LL2 resulted in a molecule with good antigen binding but very poor stability properties, thus hampering its clinical applicability. Here we report on the construction of an engineered LL2 scFv fragment by rational mutagenesis. The contribution of uncommon wild-type sequence residues for providing stability to the conserved common core structure of immunoglobulins was examined. Aided by computer homology modeling, 3 destabilizing residues within the core of the wild-type VH domain were identified. Owing to the conserved nature of the buried core structure, mutagenesis of these sites to respective consensus residues markedly stabilized the molecule but did not influence its antigen binding properties: the engineered scFv MJ-7 exhibited exceptional biophysical stability with a half-life not reached after 6 days of incubation in human serum at 37 degrees C, while fully retaining the epitope specificity of the monoclonal antibody, and antigen binding affinity of the wild-type scFv. Furthermore, both the monoclonal antibody LL2 and the engineered scFv fragment became fully internalized after only 30 min of incubation at 37 degrees C with CD22+ tumor cells. These properties predict scFv MJ-7 could become a novel powerful tool to selectively deliver cytotoxic agents to malignant CD22+ cells.
Collapse
|
46
|
Antibody Fragments. Antibodies (Basel) 2004. [DOI: 10.1007/978-1-4419-8875-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
47
|
Albrecht H, Burke PA, Natarajan A, Xiong CY, Kalicinsky M, DeNardo GL, DeNardo SJ. Production of Soluble ScFvs with C-Terminal-Free Thiol for Site-Specific Conjugation or Stable Dimeric ScFvs on Demand. Bioconjug Chem 2004; 15:16-26. [PMID: 14733579 DOI: 10.1021/bc030018+] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
ScFv recombinant antibody fragments can provide specific tumor binding modules for targeting drugs. In the process of building multimeric tumor targeting pharmaceuticals, a prerequisite is the conservation of functional scFv antigen binding domains, thereby excluding scFv random conjugation to a carrier molecule or to another scFv. The pCANTAB 5E phage display/expression vector was genetically engineered to express any scFv gene as scFv with an additional C-terminal cysteine (scFv-Cys) such that the specific conjugation site is removed from the binding domain. Selected scFvs derived from an anti-MUC-1 scFv phage library were expressed in pCANTAB 5E and its modified version pCANTAB 5E Cys vectors, and compared for key characteristics. Production yields of scFv and scFv-Cys in shaker flask and biofermentor were compared. In the absence of a reducing agent, stable dimers (covalent scFv homodimers (scFv-Cys)2) were the major form of scFv-Cys. These diabodies provided substantial signal enhancement for immunohistochemical staining of tissues. In the presence of a reducing agent, scFv-Cys molecules remained monomeric, with the free SH available for conjugation to a PEG(maleimide)2 scaffold to form immunoreactive PEG(scFv)2 bioconjugates. ScFv expression from pCANTAB 5E Cys allowed for the production of soluble scFv-Cys protein from E.coli, either as stable scFv-Cys or (scFv-Cys)2. ScFv-Cys can be used for conjugation to PEG to form bivalent PEG (scFv-Cys)2 molecules or used as (scFv-Cys)2 for increased sensitivity in IHC.
Collapse
Affiliation(s)
- Huguette Albrecht
- Radiodiagnosis and Therapy, Molecular Cancer Institute, University of California Davis Medical Center, Sacramento, California 95816, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Kipriyanov SM, Moldenhauer G, Braunagel M, Reusch U, Cochlovius B, Le Gall F, Kouprianova OA, Von der Lieth CW, Little M. Effect of domain order on the activity of bacterially produced bispecific single-chain Fv antibodies. J Mol Biol 2003; 330:99-111. [PMID: 12818205 DOI: 10.1016/s0022-2836(03)00526-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bispecific single-chain Fv antibodies comprise four covalently linked immunoglobulin variable (VH and VL) domains of two different specificities. Depending on the order of the VH and VL domains and on the length of peptides separating them, the single-chain molecule either forms two single-chain Fv (scFv) modules from the adjacent domains of the same specificity, a so-called scFv-scFv tandem [(scFv)(2)], or folds head-to-tail with the formation of a diabody-like structure, a so-called bispecific single-chain diabody (scBsDb). We generated a number of four-domain constructs composed of the same VH and VL domains specific either for human CD19 or CD3, but arranged in different orders. When expressed in bacteria, all (scFv)(2) variants appeared to be only half-functional, binding to CD19 and demonstrating no CD3-binding activity. Only the diabody-like scBsDb could bind both antigens. Comparison of the scBsDb with a structurally similar non-covalent dimer (diabody) demonstrated a stabilizing effect of the linker in the middle of the scBsDb molecule. We demonstrated that the mechanism of inactivation of CD19xCD3 diabody under physiological conditions is initiated by a dissociation of the weaker (anti-CD3) VH/VL interface followed by domain swapping with the formation of non-active homodimers. The instability of one homodimer makes the process of diabody dissociation/reassociation irreversible, thus gradually decreasing the fraction of active molecules. The structural parameters influencing the formation of functional bispecific single-chain antibodies are indicated and ways of making relatively stable bispecific molecules are proposed.
Collapse
Affiliation(s)
- Sergey M Kipriyanov
- Recombinant Antibody Research Group, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bradbury A, Velappan N, Verzillo V, Ovecka M, Chasteen L, Sblattero D, Marzari R, Lou J, Siegel R, Pavlik P. Antibodies in proteomics I: generating antibodies. Trends Biotechnol 2003; 21:275-81. [PMID: 12788548 DOI: 10.1016/s0167-7799(03)00112-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The explosion in genome sequencing, and in subsequent DNA array experiments, has provided extensive information on gene sequence, organization and expression. This has resulted in a desire to perform similarly broad experiments on all the proteins encoded by a genome. Panels of specific antibodies, or other binding ligands, will be essential tools in this endeavour. Because traditional immunization will be unlikely to generate antibodies in sufficient quantity, and of the required quality and reproducibility, in vitro selection methods will probably be used. This review--the first of two--examines the strategies available for in vitro antibody selection. The second review discusses the adaptation of these methods to high throughput and the uses to which antibodies, once derived, can be put.
Collapse
Affiliation(s)
- Andrew Bradbury
- B Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
There are great demands on the stability, expression yield and resistance to aggregation of antibody fragments. To untangle intrinsic domain effects from domain interactions, we present first a systematic evaluation of the isolated human immunoglobulin variable heavy (V(H)) and light (V(L)) germline family consensus domains and then a systematic series of V(H)-V(L) combinations in the scFv format. The constructs were evaluated in terms of their expression behavior, oligomeric state in solution and denaturant-induced unfolding equilibria under non-reducing conditions. The seven V(H) and seven V(L) domains represent the consensus sequences of the major human germline subclasses, derived from the Human Combinatorial Antibody Library (HuCAL). The isolated V(H) and V(L) domains with the highest thermodynamic stability and yield of soluble protein were V(H)3 and V(kappa)3, respectively. Similar measurements on all domain combinations in scFv fragments allowed the scFv fragments to be classified according to thermodynamic stability and in vivo folding yield. The scFv fragments containing the variable domain combinations H3kappa3, H1bkappa3, H5kappa3 and H3kappa1 show superior properties concerning yield and stability. Domain interactions diminish the intrinsic differences of the domains. ScFv fragments containing V(lambda) domains show high levels of stability, even though V(lambda) domains are surprisingly unstable by themselves. This is due to a strong interaction with the V(H) domain and depends on the amino acid sequence of the CDR-L3. On the basis of these analyses and model structures, we suggest possibilities for further improvement of the biophysical properties of individual frameworks and give recommendations for library design.
Collapse
Affiliation(s)
- Stefan Ewert
- Biochemisches Institut, Universität Zürich, Winterthurerstr 190, CH-8057 Zürich, Switzerland
| | | | | | | |
Collapse
|