1
|
Ray CMP, Yang H, Spangler JB, Mac Gabhann F. Mechanistic computational modeling of monospecific and bispecific antibodies targeting interleukin-6/8 receptors. PLoS Comput Biol 2024; 20:e1012157. [PMID: 38848446 PMCID: PMC11189202 DOI: 10.1371/journal.pcbi.1012157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/20/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
The spread of cancer from organ to organ (metastasis) is responsible for the vast majority of cancer deaths; however, most current anti-cancer drugs are designed to arrest or reverse tumor growth without directly addressing disease spread. It was recently discovered that tumor cell-secreted interleukin-6 (IL-6) and interleukin-8 (IL-8) synergize to enhance cancer metastasis in a cell-density dependent manner, and blockade of the IL-6 and IL-8 receptors (IL-6R and IL-8R) with a novel bispecific antibody, BS1, significantly reduced metastatic burden in multiple preclinical mouse models of cancer. Bispecific antibodies (BsAbs), which combine two different antigen-binding sites into one molecule, are a promising modality for drug development due to their enhanced avidity and dual targeting effects. However, while BsAbs have tremendous therapeutic potential, elucidating the mechanisms underlying their binding and inhibition will be critical for maximizing the efficacy of new BsAb treatments. Here, we describe a quantitative, computational model of the BS1 BsAb, exhibiting how modeling multivalent binding provides key insights into antibody affinity and avidity effects and can guide therapeutic design. We present detailed simulations of the monovalent and bivalent binding interactions between different antibody constructs and the IL-6 and IL-8 receptors to establish how antibody properties and system conditions impact the formation of binary (antibody-receptor) and ternary (receptor-antibody-receptor) complexes. Model results demonstrate how the balance of these complex types drives receptor inhibition, providing important and generalizable predictions for effective therapeutic design.
Collapse
Affiliation(s)
- Christina M. P. Ray
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Medical-Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Huilin Yang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jamie B. Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for Nano Biotechnology (INBT), Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
2
|
Surowka M, Klein C. A pivotal decade for bispecific antibodies? MAbs 2024; 16:2321635. [PMID: 38465614 PMCID: PMC10936642 DOI: 10.1080/19420862.2024.2321635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.
Collapse
Affiliation(s)
- Marlena Surowka
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharma Research & Early Development, Roche Glycart AG, Schlieren, Switzerland
| |
Collapse
|
3
|
Ray CMP, Yang H, Spangler JB, Mac Gabhann F. Mechanistic computational modeling of monospecific and bispecific antibodies targeting interleukin-6/8 receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.570445. [PMID: 38187701 PMCID: PMC10769311 DOI: 10.1101/2023.12.18.570445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The spread of cancer from organ to organ (metastasis) is responsible for the vast majority of cancer deaths; however, most current anti-cancer drugs are designed to arrest or reverse tumor growth without directly addressing disease spread. It was recently discovered that tumor cell-secreted interleukin-6 (IL-6) and interleukin-8 (IL-8) synergize to enhance cancer metastasis in a cell-density dependent manner, and blockade of the IL-6 and IL-8 receptors (IL-6R and IL-8R) with a novel bispecific antibody, BS1, significantly reduced metastatic burden in multiple preclinical mouse models of cancer. Bispecific antibodies (BsAbs), which combine two different antigen-binding sites into one molecule, are a promising modality for drug development due to their enhanced avidity and dual targeting effects. However, while BsAbs have tremendous therapeutic potential, elucidating the mechanisms underlying their binding and inhibition will be critical for maximizing the efficacy of new BsAb treatments. Here, we describe a quantitative, computational model of the BS1 BsAb, exhibiting how modeling multivalent binding provides key insights into antibody affinity and avidity effects and can guide therapeutic design. We present detailed simulations of the monovalent and bivalent binding interactions between different antibody constructs and the IL-6 and IL-8 receptors to establish how antibody properties and system conditions impact the formation of binary (antibody-receptor) and ternary (receptor-antibody-receptor) complexes. Model results demonstrate how the balance of these complex types drives receptor inhibition, providing important and generalizable predictions for effective therapeutic design.
Collapse
Affiliation(s)
- Christina MP Ray
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Medical-Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Huilin Yang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jamie B Spangler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Institute for Nano Biotechnology (INBT), Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
4
|
Wachter S, Angevin T, Bubna N, Tan A, Cichy A, Brown D, Wolfe LS, Sappington R, Lilla E, Berry L, Grismer D, Orth C, Blanusa M, Mostafa S, Kaufmann H, Felderer K. Application of platform process development approaches to the manufacturing of Mabcalin™ bispecifics. J Biotechnol 2023; 377:13-22. [PMID: 37820750 DOI: 10.1016/j.jbiotec.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Bispecific biotherapeutics offer potent and highly specific treatment options in oncology and immuno-oncology. However, many bispecific formats are prone to high levels of aggregation and instability, leading to prolonged development timelines, inefficient manufacturing, and high costs. The novel class of Mabcalin™ molecules consist of Anticalin® proteins fused to an IgG and are currently being evaluated in pre-clinical and clinical studies. Here, we describe a robust high-yield manufacturing platform for these therapeutic fusion proteins providing data up to commercially relevant scales. A platform upstream process was established for one of the Mabcalin bispecifics and then applied to other clinically relevant drug candidates with different IgG target specificities. Process performance was compared in 3 L bioreactors and production was scaled-up to up to 1000 L for confirmation. The Mabcalin proteins' structural and biophysical similarities enabled a downstream platform approach consisting of initial protein A capture, viral inactivation, mixed-mode anion exchange polishing, second polishing by cation exchange or hydrophobic interaction chromatography, viral filtration, buffer exchange and concentration by ultrafiltration/diafiltration. All three processes met their target specifications and achieved comparable clearance of impurities and product yields across scales. The described platform approach provides a fast and economic path to process confirmation and is well comparable to classical monoclonal antibody approaches in terms of costs and time to clinic.
Collapse
Affiliation(s)
- Stefanie Wachter
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany.
| | - Thibaut Angevin
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - Niket Bubna
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Adelene Tan
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - Adam Cichy
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - David Brown
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Leslie S Wolfe
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Ryan Sappington
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Edward Lilla
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Luke Berry
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Dane Grismer
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Christian Orth
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - Milan Blanusa
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - Sigma Mostafa
- KBI Biopharma, 4117 Emperor Blvd, Suite 200, Durham, NC 27703, USA
| | - Hitto Kaufmann
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| | - Karin Felderer
- Pieris Pharmaceuticals GmbH, Zeppelinstr. 3, Hallbergmoos 85399 Germany
| |
Collapse
|
5
|
Math BA, Waibl F, Lamp LM, Fernández‐Quintero ML, Liedl KR. Cross-linking disulfide bonds govern solution structures of diabodies. Proteins 2023; 91:1316-1328. [PMID: 37376973 PMCID: PMC10952579 DOI: 10.1002/prot.26509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/19/2023] [Indexed: 06/29/2023]
Abstract
In the last years, antibodies have emerged as a promising new class of therapeutics, due to their combination of high specificity with long serum half-life and low risk of side-effects. Diabodies are a popular novel antibody format, consisting of two Fv domains connected with short linkers. Like IgG antibodies, they simultaneously bind two target proteins. However, they offer altered properties, given their smaller size and higher rigidity. In this study, we conducted the-to our knowledge-first molecular dynamics (MD) simulations of diabodies and find a surprisingly high conformational flexibility in the relative orientation of the two Fv domains. We observe rigidifying effects through the introduction of disulfide bonds in the Fv -Fv interface and characterize the effect of different disulfide bond locations on the conformation. Additionally, we compare VH -VL orientations and paratope dynamics between diabodies and an antigen binding fragment (Fab) of the same sequence. We find mostly consistent structures and dynamics, indicating similar antigen binding properties. The most significant differences can be found within the CDR-H2 loop dynamics. Of all CDR loops, the CDR-H2 is located closest to the artificial Fv -Fv interface. All examined diabodies show similar VH -VL orientations, Fv -Fv packing and CDR loop conformations. However, the variant with a P14C-K64C disulfide bond differs most from the Fab in our measures, including the CDR-H3 loop conformational ensemble. This suggests altered antigen binding properties and underlines the need for careful validation of the disulfide bond locations in diabodies.
Collapse
Affiliation(s)
- Barbara A. Math
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Franz Waibl
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Leonida M. Lamp
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Monica L. Fernández‐Quintero
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Klaus R. Liedl
- Institute of General, Inorganic and Theoretical Chemistry, and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| |
Collapse
|
6
|
Iwasaki YW, Tharakaraman K, Subramanian V, Khongmanee A, Hatas A, Fleischer E, Rurak TT, Ngok-ngam P, Tit-oon P, Ruchirawat M, Satayavivad J, Fuangthong M, Sasisekharan R. Generation of bispecific antibodies by structure-guided redesign of IgG constant regions. Front Immunol 2023; 13:1063002. [PMID: 36703993 PMCID: PMC9871890 DOI: 10.3389/fimmu.2022.1063002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Bispecific antibodies (BsAbs) form an exciting class of bio-therapeutics owing to their multispecificity. Although numerous formats have been developed, generation of hetero-tetrameric IgG1-like BsAbs having acceptable safety and pharmacokinetics profiles from a single cell culture system remains challenging due to the heterogeneous pairing between the four chains. Herein, we employed a structure-guided approach to engineer mutations in the constant domain interfaces (CH1-CL and CH3-CH3) of heavy and κ light chains to prevent heavy-light mispairing in the antigen binding fragment (Fab) region and heavy-heavy homodimerization in the Fc region. Transient co-transfection of mammalian cells with heavy and light chains of pre-existing antibodies carrying the engineered constant domains generates BsAbs with percentage purity ranging from 78% to 85%. The engineered BsAbs demonstrate simultaneous binding of both antigens, while retaining the thermal stability, Fc-mediated effector properties and FcRn binding properties of the parental antibodies. Importantly, since the variable domains were not modified, the mutations may enable BsAb formation from antibodies belonging to different germline origins and isotypes. The rationally designed mutations reported in this work could serve as a starting point for generating optimized solutions required for large scale production.
Collapse
Affiliation(s)
- Yordkhwan W. Iwasaki
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand
| | - Kannan Tharakaraman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vidya Subramanian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Amnart Khongmanee
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Andrew Hatas
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Eduardo Fleischer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Troy T. Rurak
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Patchara Ngok-ngam
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Phanthakarn Tit-oon
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mathuros Ruchirawat
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand
| | - Jutamaad Satayavivad
- Program in Environmental Toxicology, Chulabhorn Graduate Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Laboratory of Pharmacology, Chulabhorn Research Institute, Bangkok, Thailand
| | - Mayuree Fuangthong
- Translational Research Unit, Chulabhorn Research Institute, Bangkok, Thailand,Center of Excellence on Environmental Health and Toxicology (EHT), Office of the Permanent Secretary (OPS), Ministry of Higher Education, Science, Research and Innovation (MHESI), Bangkok, Thailand,Program in Applied Biological Sciences, Chulabhorn Graduate Institute, Bangkok, Thailand,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| | - Ram Sasisekharan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States,*Correspondence: Mayuree Fuangthong, ; Ram Sasisekharan,
| |
Collapse
|
7
|
van Diest E, Nicolasen MJT, Kramer L, Zheng J, Hernández-López P, Beringer DX, Kuball J. The making of multivalent gamma delta TCR anti-CD3 bispecific T cell engagers. Front Immunol 2023; 13:1052090. [PMID: 36685546 PMCID: PMC9851377 DOI: 10.3389/fimmu.2022.1052090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction We have recently developed a novel T cell engager concept by utilizing γ9δ2TCR as tumor targeting domain, named gamma delta TCR anti-CD3 bispecific molecule (GAB), targeting the phosphoantigen-dependent orchestration of BTN2A1 and BTN3A1 at the surface of cancer cells. GABs are made by the fusion of the ectodomains of a γδTCR to an anti-CD3 single chain variable fragment (scFv) (γδECTO-αCD3), here we explore alternative designs with the aim to enhance GAB effectivity. Methods The first alternative design was made by linking the variable domains of the γ and δ chain to an anti-CD3 scFv (γδVAR-αCD3). The second alternative design was multimerizing γδVAR-αCD3 proteins to increase the tumor binding valency. Both designs were expressed and purified and the potency to target tumor cells by T cells of the alternative designs was compared to γδECTO-αCD3, in T cell activation and cytotoxicity assays. Results and discussion The γδVAR-αCD3 proteins were poorly expressed, and while the addition of stabilizing mutations based on finding for αβ single chain formats increased expression, generation of meaningful amounts of γδVAR-αCD3 protein was not possible. As an alternative strategy, we explored the natural properties of the original GAB design (γδECTO-αCD3), and observed the spontaneous formation of γδECTO-αCD3-monomers and -dimers during expression. We successfully enhanced the fraction of γδECTO-αCD3-dimers by shortening the linker length between the heavy and light chain in the anti-CD3 scFv, though this also decreased protein yield by 50%. Finally, we formally demonstrated with purified γδECTO-αCD3-dimers and -monomers, that γδECTO-αCD3-dimers are superior in function when compared to similar concentrations of monomers, and do not induce T cell activation without simultaneous tumor engagement. In conclusion, a γδECTO-αCD3-dimer based GAB design has great potential, though protein production needs to be further optimized before preclinical and clinical testing.
Collapse
Affiliation(s)
- Eline van Diest
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Mara J. T. Nicolasen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lovro Kramer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jiali Zheng
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Patricia Hernández-López
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Dennis X. Beringer
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jürgen Kuball
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands,Department of Hematology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands,*Correspondence: Jürgen Kuball,
| |
Collapse
|
8
|
Lu D, Wang X, Su R, Cheng Y, Wang H, Luo L, Xiao Z. Preparation of an Immunoaffinity Column Based on Bispecific Monoclonal Antibody for Aflatoxin B 1 and Ochratoxin A Detection Combined with ic-ELISA. Foods 2022; 11:foods11030335. [PMID: 35159486 PMCID: PMC8833996 DOI: 10.3390/foods11030335] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/21/2022] [Indexed: 01/02/2023] Open
Abstract
A novel and efficient immunoaffinity column (IAC) based on bispecific monoclonal antibody (BsMAb) recognizing aflatoxin B1 (AFB1) and ochratoxin A (OTA) was prepared and applied in simultaneous extraction of AFB1 and OTA from food samples and detection of AFB1/OTA combined with ic-ELISA (indirect competitive ELISA). Two deficient cell lines, hypoxanthine guanine phosphoribosyl-transferase (HGPRT) deficient anti-AFB1 hybridoma cell line and thymidine kinase (TK) deficient anti-OTA hybridoma cell line, were fused to generate a hybrid-hybridoma producing BsMAb against AFB1 and OTA. The subtype of the BsMAb was IgG1 via mouse antibody isotyping kit test. The purity and molecular weight of BsMAb were confirmed by SDS-PAGE method. The cross-reaction rate with AFB2 was 37%, with AFG1 15%, with AFM1 48%, with AFM2 10%, and with OTB 36%. Negligible cross-reaction was observed with other tested compounds. The affinity constant (Ka) was determined by ELISA. The Ka (AFB1) and Ka (OTA) was 2.43 × 108 L/mol and 1.57 × 108 L/mol, respectively. Then the anti-AFB1/OTA BsMAb was coupled with CNBr-Sepharose, and an AFB1/OTA IAC was prepared. The coupling time and elution conditions of IAC were optimized. The coupling time was 1 h with 90% coupling rate, the eluent was methanol–water (60:40, v:v, pH 2.3) containing 1 mol/L NaCl, and the eluent volume was 4 mL. The column capacities of AFB1 and OTA were 165.0 ng and 171.3 ng, respectively. After seven times of repeated use, the preservation rates of column capacity for AFB1 and OTA were 69.3% and 68.0%, respectively. The ic-ELISA for AFB1 and OTA were applied combined with IAC. The IC50 (50% inhibiting concentration) of AFB1 was 0.027 ng/mL, the limit of detection (LOD) was 0.004 ng/mL (0.032 µg/kg), and the linear range was 0.006 ng/mL~0.119 ng/mL. The IC50 of OTA was 0.878 ng/mL, the LOD was 0.126 ng/mL (1.008 µg/kg), and the linear range was 0.259 ng/mL~6.178 ng/mL. Under optimum conditions, corn and wheat samples were pretreated with AFB1-OTA IAC. The recovery rates of AFB1 and OTA were 95.4%~105.0% with ic-ELISA, and the correlations between the detection results and LC-MS were above 0.9. The developed IAC combined with ic-ELISA is reliable and could be applied to the detection of AFB1 and OTA in grains.
Collapse
|
9
|
Boykoff N, Freage L, Lenn J, Mallikaratchy P. Bispecific Aptamer Sensor toward T-Cell Leukemia Detection in the Tumor Microenvironment. ACS OMEGA 2021; 6:32563-32570. [PMID: 34901605 PMCID: PMC8655784 DOI: 10.1021/acsomega.1c04125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/06/2021] [Indexed: 06/14/2023]
Abstract
The current detection methods of malignant cells are mainly based on the high expression levels of certain surface proteins on these cells. However, many of the same surface marker proteins are also expressed in normal cells. Growing evidence suggests that the molecular signatures of the tumor microenvironment (TME) are related to the biological state of a diseased cell. Exploiting the unique molecular signature of the TME, we have designed a molecular sensing agent consisting of a molecular switch that can sense the elevated concentration of a small molecule in the TME and promote precise recognition of a malignant cell. We accomplished this by designing and developing a bispecific aptamer that takes advantage of a high concentration of adenosine 5'-triphosphate in the TME. Thus, we report a prototype of a bispecific aptamer molecule, which serves as a dual detection platform and recognizes tumor cells only when a given metabolite concentration is elevated in the TME. This system overcomes hurdles in detecting tumor cells solely based on the elevated expression of cell surface markers, providing a universal platform for tumor targeting and sensing.
Collapse
Affiliation(s)
- Natalie Boykoff
- Ph.D.
Programs in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, New York 10016, United
States
| | - Lina Freage
- Department
of Chemistry, Lehman College, The City University
of New York, 250 Bedford
Park Blvd., West, Bronx, New York 10468, United
States
| | - Jared Lenn
- The
Bronx High School of Science, 75 W 205th Street, Bronx, New York 10468, United States
| | - Prabodhika Mallikaratchy
- Department
of Chemistry, Lehman College, The City University
of New York, 250 Bedford
Park Blvd., West, Bronx, New York 10468, United
States
- Ph.D.
Programs in Chemistry and Biochemistry, CUNY Graduate Center, 365 Fifth Avenue, New York, New York 10016, United
States
- Ph.D.
Program in Molecular, Cellular and Developmental Biology, CUNY Graduate Center, 365 Fifth Avenue, New York, New York 10016, United
States
| |
Collapse
|
10
|
Lee SC, Ma JSY, Kim MS, Laborda E, Choi SH, Hampton EN, Yun H, Nunez V, Muldong MT, Wu CN, Ma W, Kulidjian AA, Kane CJ, Klyushnichenko V, Woods AK, Joseph SB, Petrassi M, Wisler J, Li J, Jamieson CAM, Schultz PG, Kim CH, Young TS. A PSMA-targeted bispecific antibody for prostate cancer driven by a small-molecule targeting ligand. SCIENCE ADVANCES 2021; 7:eabi8193. [PMID: 34380625 PMCID: PMC8357232 DOI: 10.1126/sciadv.abi8193] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
Despite the development of next-generation antiandrogens, metastatic castration-resistant prostate cancer (mCRPC) remains incurable. Here, we describe a unique semisynthetic bispecific antibody that uses site-specific unnatural amino acid conjugation to combine the potency of a T cell-recruiting anti-CD3 antibody with the specificity of an imaging ligand (DUPA) for prostate-specific membrane antigen. This format enabled optimization of structure and function to produce a candidate (CCW702) with specific, potent in vitro cytotoxicity and improved stability compared with a bispecific single-chain variable fragment format. In vivo, CCW702 eliminated C4-2 xenografts with as few as three weekly subcutaneous doses and prevented growth of PCSD1 patient-derived xenograft tumors in mice. In cynomolgus monkeys, CCW702 was well tolerated up to 34.1 mg/kg per dose, with near-complete subcutaneous bioavailability and a PK profile supporting testing of a weekly dosing regimen in patients. CCW702 is being evaluated in a first in-human clinical trial for men with mCRPC who had progressed on prior therapies (NCT04077021).
Collapse
Affiliation(s)
- Sung Chang Lee
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jennifer S Y Ma
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Min Soo Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eduardo Laborda
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sei-Hyun Choi
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric N Hampton
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Hwayoung Yun
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Vanessa Nunez
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michelle T Muldong
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Christina N Wu
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenxue Ma
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anna A Kulidjian
- Department of Orthopedic Surgery, Scripps MD Anderson Cancer Center, La Jolla, CA 92093, USA
| | - Christopher J Kane
- Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vadim Klyushnichenko
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ashley K Woods
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean B Joseph
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mike Petrassi
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - John Wisler
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jing Li
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christina A M Jamieson
- Department of Urology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Peter G Schultz
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Chan Hyuk Kim
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Travis S Young
- Department of Biology, Calibr, The Scripps Research Institute, La Jolla, CA 92037, USA.
- Department of Chemistry and Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
11
|
Napoleone A, Laurén I, Linkgreim T, Dahllund L, Persson H, Andersson O, Olsson A, Hultqvist G, Frank P, Hall M, Morrison A, Andersson A, Lord M, Mangsbo S. Fed-batch production assessment of a tetravalent bispecific antibody: A case study on piggyBac stably transfected HEK293 cells. N Biotechnol 2021; 65:9-19. [PMID: 34273575 DOI: 10.1016/j.nbt.2021.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/07/2021] [Accepted: 07/11/2021] [Indexed: 12/24/2022]
Abstract
The transition from preclinical biological drug development into clinical trials requires an efficient upscaling process. In this context, bispecific antibody drugs are particularly challenging due to their propensity to form aggregates and generally produce low titers. Here, the upscaling process for a tetravalent bispecific antibody expressed by a piggyBac transposon-mediated stable HEK293 cell pool has been evaluated. The project was performed as a case study at Testa Center, a non-GMP facility for scale-up testing of biologics in Sweden, and encompassed media adaptation strategies, fed-batch optimization and a novel antibody purification technology. The cell pool was adapted to different culture media for evaluation in terms of cell viability and titers compared to its original Expi293 Expression Medium. These parameters were assessed in both sequential stepwise adaption and direct media exchanges. By this, a more affordable medium was identified that did not require stepwise adaptation and with similar titers and viability as in the Expi293 Expression Medium. Fed-batch optimizations resulted in culture densities reaching up to 20 × 106 viable cells/mL with over 90 % viability 12 days post-inoculum, and antibody titers three times higher than corresponding batch cultures. By implementing a novel high-speed protein A fiber technology (Fibro PrismA) with a capture residence time of only 7.5 s, 8 L of supernatant could be purified in 4.5 h without compromising the purity, structural integrity and function of the bispecific antibody. Results from this study related to medium adaptation and design of fed-batch protocols will be highly beneficial during the forthcoming scale-up of this therapeutic antibody.
Collapse
Affiliation(s)
- Antonino Napoleone
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ida Laurén
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Triinu Linkgreim
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Leif Dahllund
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Helena Persson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Oskar Andersson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Anders Olsson
- Science for Life Laboratory, Drug Discovery and Development & School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Greta Hultqvist
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Peter Frank
- Wicket AB, Väderkvarnsgatan 30, 75329 Uppsala, Sweden
| | - Martin Hall
- Cytiva AB, Björkgatan 30, 751 84 Uppsala, Sweden
| | - Annika Morrison
- Cytiva Testa Center AB, Björkgatan 30, 751 84 Uppsala, Sweden
| | | | - Martin Lord
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Barzaman K, Moradi-Kalbolandi S, Hosseinzadeh A, Kazemi MH, Khorramdelazad H, Safari E, Farahmand L. Breast cancer immunotherapy: Current and novel approaches. Int Immunopharmacol 2021; 98:107886. [PMID: 34153663 DOI: 10.1016/j.intimp.2021.107886] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022]
Abstract
The crucial role of the immune system in the progression/regression of breast cancer (BC) should always be taken into account. Various immunotherapy approaches have been investigated for BC, including tumor-targeting antibodies (bispecific antibodies), adoptive T cell therapy, vaccines, and immune checkpoint blockade such as anti-PD-1. In addition, a combination of conventional chemotherapy and immunotherapy approaches contributes to improving patients' overall survival rates. Although encouraging outcomes have been reported in most clinical trials of immunotherapy, some obstacles should still be resolved in this regard. Recently, personalized immunotherapy has been proposed as a potential complementary medicine with immunotherapy and chemotherapy for overcoming BC. Accordingly, this review discusses the brief association of these methods and future directions in BC immunotherapy.
Collapse
Affiliation(s)
- Khadijeh Barzaman
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Moradi-Kalbolandi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Aysooda Hosseinzadeh
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Hossein Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjani University of Medical Sciences, Rafsanjani, Iran; Department of Immunology, School of Medicine, Rafsanjani University of Medical Sciences, Rafsanjani, Iran
| | - Elahe Safari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
13
|
Wang W, Gil-Garcia M, Ventura S. Dual Antibody-Conjugated Amyloid Nanorods to Promote Selective Cell-Cell Interactions. ACS APPLIED MATERIALS & INTERFACES 2021; 13:14875-14884. [PMID: 33759489 PMCID: PMC9262253 DOI: 10.1021/acsami.0c21996] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Grafting biomolecules on nanostructures' surfaces is an increasingly used strategy to target pathogenic cells, with both diagnostic and therapeutic applications. However, nanomaterials monofunctionalized by conjugating a single type of ligand find limited uses in pathologies/therapies that require two or more targets/receptors to be targeted and/or activated with a single molecular entity simultaneously. Therefore, multivalent nanomaterials for dual- or multitargeting are attracting significant interest. This study provides a proof of concept of such nanostructures. We have recently developed a modular methodology that allows obtaining amyloid-based materials decorated with active globular domains. Here, this approach is exploited to generate functional amyloid fibrils displaying antibody capture moieties. A high antibody binding affinity and capacity for the resulting nanofibrils, whose size can be manipulated to obtain homogeneous nanorods with high biocompatibility, are demonstrated. These nanorods are then used for specific antibody-mediated targeting of different cell types. Simultaneous conjugation of these nanorods with different antibodies allows obtaining a mimic of a bispecific antibody that redirects T lymphocytes to tumoral cells, holding high potential for immunotherapy. Overall, the work illustrates a modular and straightforward strategy to obtain preparative quantities of multivalent antibody-functionalized nanomaterials with multitargeting properties without the need for covalent modification.
Collapse
|
14
|
Ke Q, Kroger CJ, Clark M, Tisch RM. Evolving Antibody Therapies for the Treatment of Type 1 Diabetes. Front Immunol 2021; 11:624568. [PMID: 33679717 PMCID: PMC7930374 DOI: 10.3389/fimmu.2020.624568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Type 1 diabetes (T1D) is widely considered to be a T cell driven autoimmune disease resulting in reduced insulin production due to dysfunction/destruction of pancreatic β cells. Currently, there continues to be a need for immunotherapies that selectively reestablish persistent β cell-specific self-tolerance for the prevention and remission of T1D in the clinic. The utilization of monoclonal antibodies (mAb) is one strategy to target specific immune cell populations inducing autoimmune-driven pathology. Several mAb have proven to be clinically safe and exhibit varying degrees of efficacy in modulating autoimmunity, including T1D. Traditionally, mAb therapies have been used to deplete a targeted cell population regardless of antigenic specificity. However, this treatment strategy can prove detrimental resulting in the loss of acquired protective immunity. Nondepleting mAb have also been applied to modulate the function of immune effector cells. Recent studies have begun to define novel mechanisms associated with mAb-based immunotherapy that alter the function of targeted effector cell pools. These results suggest short course mAb therapies may have persistent effects for regaining and maintaining self-tolerance. Furthermore, the flexibility to manipulate mAb properties permits the development of novel strategies to target multiple antigens and/or deliver therapeutic drugs by a single mAb molecule. Here, we discuss current and potential future therapeutic mAb treatment strategies for T1D, and T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Abstract
Bispecific antibodies (bsAbs) target two different epitopes. These are an up-and-coming class of biologics, with two such therapeutics (emicizumab and blinatumomab) FDA approved and on the market, and many more in clinical trials. While the first reported bsAbs were constructed by chemical methods, this approach has fallen out of favour with the advent of modern genetic engineering techniques and, nowadays, the vast majority of bsAbs are produced by protein engineering. However, in recent years, relying on innovations in the fields of bioconjugation and bioorthogonal click chemistry, new chemical methods have appeared that have the potential to be competitive with protein engineering techniques and, indeed, hold some advantages. These approaches offer modularity, reproducibility and batch-to-batch consistency, as well as the integration of handles, whereby additional cargo molecules can be attached easily, e.g. to generate bispecific antibody-drug conjugates. The linker between the antibodies/antibody fragments can also be easily varied, and new formats (types, defined by structural properties or by construction methodology) can be generated rapidly. These attributes offer the potential to revolutionize the field. Here, we review chemical methods for the generation of bsAbs, showing that the newest examples of these techniques are worthy competitors to the industry-standard expression-based strategies.
Collapse
|
16
|
Buschhaus MJ, Becker S, Porter AJ, Barelle CJ. Isolation of highly selective IgNAR variable single-domains against a human therapeutic Fc scaffold and their application as tailor-made bioprocessing reagents. Protein Eng Des Sel 2019; 32:385-399. [PMID: 32119084 DOI: 10.1093/protein/gzaa002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/12/2019] [Accepted: 01/07/2020] [Indexed: 01/03/2023] Open
Abstract
The adaptive immune system of cartilaginous fish (Elasmobranchii), comprising of classical hetero-tetrameric antibodies, is enhanced through the presence of a naturally occurring homodimeric antibody-like immunoglobulin-the new antigen receptor (IgNAR). The binding site of the IgNAR variable single-domain (VNAR) offers advantages of reduced size (<1/10th of classical immunoglobulin) and extended binding topographies, making it an ideal candidate for accessing cryptic epitopes otherwise intractable to conventional antibodies. These attributes, coupled with high physicochemical stability and amenability to phage display, facilitate the selection of VNAR binders to challenging targets. Here, we explored the unique attributes of these single domains for potential application as bioprocessing reagents in the development of the SEED-Fc platform, designed to generate therapeutic bispecific antibodies. A panel of unique VNARs specific to the SEED homodimeric (monospecific) 'by-products' were isolated from a shark semi-synthetic VNAR library via phage display. The lead VNAR candidate exhibited low nanomolar affinity and superior selectivity to SEED homodimer, with functionality being retained upon exposure to extreme physicochemical conditions that mimic their applicability as purification agents. Ultimately, this work exemplifies the robustness of the semi-synthetic VNAR platform, the predisposition of the VNAR paratope to recognise novel epitopes and the potential for routine generation of tailor-made VNAR-based bioprocessing reagents.
Collapse
Affiliation(s)
- Magdalena J Buschhaus
- Elasmogen Ltd, Liberty Building, Foresterhill Health Campus, Foresterhill Road, Aberdeen AB25 2ZP, UK
| | - Stefan Becker
- Merck Biopharma KGaA, Protein Engineering & Antibody Technologies, Global Research and Development, Frankfurter Str. 250 Darmstadt 64293, Germany
| | - Andrew J Porter
- Elasmogen Ltd, Liberty Building, Foresterhill Health Campus, Foresterhill Road, Aberdeen AB25 2ZP, UK.,Department of Molecular and Cell Biology, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Caroline J Barelle
- Elasmogen Ltd, Liberty Building, Foresterhill Health Campus, Foresterhill Road, Aberdeen AB25 2ZP, UK
| |
Collapse
|
17
|
Lu L, Liu N, Fan K, Zhang G, Li C, Yan Y, Liu T, Fu WH. A tetravalent single chain diabody (CD40/HER2) efficiently inhibits tumor proliferation through recruitment of T cells and anti-HER2 functions. Mol Immunol 2019; 109:149-156. [PMID: 30951934 DOI: 10.1016/j.molimm.2019.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 01/05/2023]
Abstract
Our aim was to construct a CD40×HER2 single chain diabody (ScDb) and determine its tumor-specific immune activation and anti-HER2 function. Overlap extension-polymerase chain reaction was applied in the construction of ScDb, and the protein was expressed with the pET28a (+)-Rosetta prokaryotic expression system. Soluble ScDb was purified by a nickel-nitrilotriacetic acid column. Dendritic cells (DC) was stimulated by ScDb and inhibited 4T1 cells proliferation in vitro. In 4T1 tumor mice model, lymphocyte infiltration was prominently detected in ScDb group, Caspase-3 expression was significantly upregulated. ScDb was labeled using quantum dots. Immunofluorescence assay indicated ScDb exhibited high affinity to HER2. T6-17 cells were inhibited by ScDb in vitro. The phosphorylation and expression levels of AKT, ERK were markedly decreased. In T6-17 tumor mice model. Compared to CD40 ScFv, HER2 ScFv and normal saline groups, tumor volume diminished significantly in ScDb group, and tumor cells showed extensive deformation, and pervasive karyopyknosis and karyorrhexis were found. In the present study, we successfully constructed a ScDb fragment and expressed it using a prokaryotic expression system. The in vivo and in vitro experimental results indicated that ScDb could inhibit the proliferation of tumor cells by stimulating the tumor-specific immunoreaction and blocking the HER2-related signaling pathway.
Collapse
Affiliation(s)
- Li Lu
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Ningbo Liu
- Department of Oncology Surgery, The first hospital of Handan, Hebei province China
| | - Kaihu Fan
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Guojing Zhang
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Chuan Li
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Yongjia Yan
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Tong Liu
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China
| | - Wei-Huahua Fu
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, China.
| |
Collapse
|
18
|
Byeon J, Park M, Shin S, Shin YG. Fast and Simple Qualitative/Semi‐Quantitative Analysis of Monoclonal Antibody Mixtures Using Liquid Chromatography–Electrospray Triple Time‐of‐Flight Mass Spectrometry. B KOREAN CHEM SOC 2018. [DOI: 10.1002/bkcs.11610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jin‐Ju Byeon
- College of PharmacyChungnam National University Daejeon 305‐764 South Korea
| | - Min‐Ho Park
- College of PharmacyChungnam National University Daejeon 305‐764 South Korea
| | - Seok‐Ho Shin
- College of PharmacyChungnam National University Daejeon 305‐764 South Korea
| | - Young G. Shin
- College of PharmacyChungnam National University Daejeon 305‐764 South Korea
| |
Collapse
|
19
|
Ellerman D. Bispecific T-cell engagers: Towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2018; 154:102-117. [PMID: 30395966 DOI: 10.1016/j.ymeth.2018.10.026] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023] Open
Abstract
Bispecific molecules redirecting the cytotoxicity of T-cells are a growing class of therapeutics with numerous molecules being tested in clinical trials. However, it has been a long way since the proof of concept studies in the mid 1980's. In the process we have learnt about the impact of different variables related to the bispecific molecule and the target antigen on the potency of this type of drugs. This work reviews the insights gained and how that knowledge has been used to design more potent bispecific T-cell engagers. The more recent advancement of antibodies with this modality into safety studies in non-human primates and as well as in clinical studies has revealed potential toxicity liabilities for the mode of action. Modifications in existing antibody formats and new experimental molecules designed to mitigate these problems are discussed.
Collapse
|
20
|
Andrade C, Arnold L, Motabar D, Aspelund M, Tang A, Hunter A, Chung WK. An Integrated Approach to Aggregate Control for Therapeutic Bispecific Antibodies Using an Improved Three Column Mab Platform-Like Purification Process. Biotechnol Prog 2018; 35:e2720. [PMID: 30298991 PMCID: PMC6667909 DOI: 10.1002/btpr.2720] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/17/2018] [Accepted: 09/17/2018] [Indexed: 12/23/2022]
Abstract
Single chain variable fragment‐IgGs (scFv‐IgG) are a class of bispecific antibodies consisting of two single chain variable fragments (scFv) that are fused to an intact IgG molecule. A common trend observed for expression of scFv‐IgGs in mammalian cell culture is a higher level of aggregates (10%–30%) compared to mAbs, which results in lower purification yields in order to meet product quality targets. Furthermore, the high aggregate levels also pose robustness risks to a conventional mAb three column platform purification process which uses only the polishing steps (e.g., cation exchange chromatography [CEX]) for aggregate removal. Protein A chromatography with pH gradient elution, high performance tangential flow filtration (HP‐TFF) and calcium phosphate precipitation were evaluated at the bench scale as means of introducing orthogonal aggregate removal capabilities into other aspects of the purification process. The two most promising process variants, namely Protein A pH gradient elution followed by calcium phosphate precipitation were evaluated at pilot scale, demonstrating comparable performance. Implementing Protein A chromatography with gradient elution and/or calcium phosphate precipitation removed a sufficient portion of the aggregate burden prior to the CEX polishing step, enabling CEX to be operated robustly under conditions favoring higher monomer yield. From starting aggregate levels ranging from 15% to 23% in the condition media, levels were reduced to between 2% and 3% at the end of the CEX step. The overall yield for the optimal process was 71%. Results of this work suggest an improved three‐column mAb platform‐like purification process for purification of high aggregate scFv‐IgG bispecific antibodies is feasible. © 2018 The Authors. Biotechnology Progress published by Wiley Periodicals, Inc. on behalf of American Institute of Chemical Engineers. Biotechnol. Prog., 35: e2720, 2019
Collapse
Affiliation(s)
- Cassia Andrade
- Purification Process Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| | - Lindsay Arnold
- Process Development Engineering, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| | - Dana Motabar
- Purification Process Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| | - Matthew Aspelund
- Purification Process Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| | - Alison Tang
- Purification Process Sciences, MedImmune LLC, Cambridge, U.K
| | - Alan Hunter
- Purification Process Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| | - Wai Keen Chung
- Purification Process Sciences, MedImmune LLC, One MedImmune Way, Gaithersburg, Maryland, 20878
| |
Collapse
|
21
|
Abstract
Bispecific antibodies have moved from being an academic curiosity with therapeutic promise to reality, with two molecules being currently commercialized (Hemlibra® and Blincyto®) and many more in clinical trials. The success of bispecific antibodies is mainly due to the continuously growing number of mechanisms of actions (MOA) they enable that are not accessible to monoclonal antibodies. One of the earliest MOA of bispecific antibodies and currently the one with the largest number of clinical trials is the redirecting of the cytotoxic activity of T-cells for oncology applications, now extending its use in infective diseases. The use of bispecific antibodies for crossing the blood-brain barrier is another important application because of its potential to advance the therapeutic options for neurological diseases. Another noteworthy application due to its growing trend is enabling a more tissue-specific delivery or activity of antibodies. The different molecular solutions to the initial hurdles that limited the development of bispecific antibodies have led to the current diverse set of bispecific or multispecific antibody formats that can be grouped into three main categories: IgG-like formats, antibody fragment-based formats, or appended IgG formats. The expanded applications of bispecific antibodies come at the price of additional challenges for clinical development. The rising complexity in their structure may increase the risk of immunogenicity and the multiple antigen specificity complicates the selection of relevant species for safety assessment.
Collapse
Affiliation(s)
- Bushra Husain
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Diego Ellerman
- Protein Chemistry Department, Genentech Inc., South San Francisco, CA, 94080, USA.
| |
Collapse
|
22
|
Asano R, Kuroki Y, Honma S, Akabane M, Watanabe S, Mayuzumi S, Hiyamuta S, Kumagai I, Sode K. Comprehensive study of domain rearrangements of single-chain bispecific antibodies to determine the best combination of configurations and microbial host cells. MAbs 2018; 10:854-863. [PMID: 29985753 DOI: 10.1080/19420862.2018.1476815] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Small bispecific antibodies (bsAbs) are important therapeutic molecules and represent the first bsAb format approved by the United States Food and Drug Administration. Diabody (Db), a small bsAb format, has four possible domain orders; we previously reported the differences in the expression levels and cancer growth inhibition effects upon rearranging the domain order of this format. However, there have been no comprehensive reports on domain rearrangements of bispecific single-chain Db (scDb) and tandem single-chain Fv (taFv), which are widely used bsAb formats. In this study, we designed all possible domain orders for scDb and taFv (each with eight variants) with identical Fv pairs and individually expressed all 16 variants using Escherichia coli, Pichia pastoris, and Brevibacillus choshinensis. Comprehensive investigations showed that the intrinsic functions of the variants were similar to each other, regardless of the expression host system, but expression levels varied depending on the format as well as on the host cell. Among the 16 variants, we found a promising candidate that exhibited high activity and productivity. Furthermore, we determined that B. choshinensis is an attractive expression host because of its secretory production of recombinant proteins.
Collapse
Affiliation(s)
- Ryutaro Asano
- a Department of Biotechnology and Life Science, Graduate School of Engineering , Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Yuri Kuroki
- a Department of Biotechnology and Life Science, Graduate School of Engineering , Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Sachiko Honma
- b R&D Department of ProteinExpress Co., Ltd ., Chiba , Japan
| | - Mihoko Akabane
- b R&D Department of ProteinExpress Co., Ltd ., Chiba , Japan
| | | | - Shinzo Mayuzumi
- c Advanced Technology Research Laboratories , Idemitsu Kosan Co., Ltd ., Chiba , Japan
| | - Shuichi Hiyamuta
- c Advanced Technology Research Laboratories , Idemitsu Kosan Co., Ltd ., Chiba , Japan
| | - Izumi Kumagai
- a Department of Biotechnology and Life Science, Graduate School of Engineering , Tokyo University of Agriculture and Technology , Tokyo , Japan
| | - Koji Sode
- a Department of Biotechnology and Life Science, Graduate School of Engineering , Tokyo University of Agriculture and Technology , Tokyo , Japan.,d Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill and North Carolina State University , Chapel Hill , NC , USA
| |
Collapse
|
23
|
Tustian AD, Laurin L, Ihre H, Tran T, Stairs R, Bak H. Development of a novel affinity chromatography resin for platform purification of bispecific antibodies with modified protein a binding avidity. Biotechnol Prog 2018; 34:650-658. [PMID: 29464924 PMCID: PMC6099419 DOI: 10.1002/btpr.2622] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/16/2018] [Indexed: 12/30/2022]
Abstract
There is strong interest in the production of bispecific monoclonal antibodies that can simultaneously bind two distinct targets or epitopes to achieve novel mechanisms of action and efficacy. Regeneron's bispecific technology, based upon a standard IgG, consists of a heterodimer of two different heavy chains, and a common light chain. Coexpression of two heavy chains leads to the formation of two parental IgG impurities, the removal of which is facilitated by a dipeptide substitution in the Fc portion of one of the heavy chains that ablates Fc Protein A binding. Therefore, the affinity capture (Protein A) step of the purification process must perform both bulk capture and high resolution of these mAb impurities, a task current commercially available resins are not designed for. Resolution can be further impaired by the ability of Protein A to bind some antibodies in the variable region of the heavy chain (VH ). This article details development of a novel Protein A resin. This resin combines an alkali stable ligand with a base matrix exhibiting excellent mass transfer properties to allow high capacity single step capture and resolution of bispecific antibodies (bsAbs) with high yields. The developed resin, named MabSelect SuRe™ pcc, is implemented in GMP production processes for several bsAbs. © 2018 The Authors Biotechnology Progress published by Wiley Periodicals, Inc. on behalf of American Institute of Chemical Engineers Biotechnol. Prog., 34:650-658, 2018.
Collapse
Affiliation(s)
| | | | | | - Travis Tran
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| | - Robert Stairs
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| | - Hanne Bak
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591
| |
Collapse
|
24
|
Giese G, Williams A, Rodriguez M, Persson J. Bispecific antibody process development: Assembly and purification of knob and hole bispecific antibodies. Biotechnol Prog 2018; 34:397-404. [PMID: 29193902 DOI: 10.1002/btpr.2590] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/22/2017] [Indexed: 11/10/2022]
Abstract
Production of knob and hole dual light chain bispecific antibodies poses several unique challenges for development of a feasible industrial scale manufacturing process. We developed an efficient process for the assembly and purification of knob and hole dual light chain bispecific antibodies. Two distinct half-antibodies targeting two different antigens were expressed separately in Escherichia coli cells and captured independently using Protein A chromatography. When combined, the knob and hole mutations in the CH3 domains promoted heterodimer formation. The hinge region disulfides were reduced and reoxidized to form the disulfide bridge between the two complementary half antibodies. Unreacted half antibodies, noncovalently linked homodimers, covalently linked homodimers, and noncovalently linked heterodimers are impurities closely related to the product of interest and are challenging to remove by standard processes. Characterization of the molecular properties of the half antibodies and high-throughput screening predicted column chromatography performance and allowed for rapid development of downstream purification steps for removal of unique product-related and process-related impurities. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:397-404, 2018.
Collapse
Affiliation(s)
- Glen Giese
- Department of Purification Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080
| | - Ambrose Williams
- Department of Purification Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080
| | - Maricel Rodriguez
- Department of Purification Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080
| | - Josefine Persson
- Department of Purification Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080
| |
Collapse
|
25
|
Abstract
The common strategy for making bispecific antibodies (BsAbs) involves combining the variable domains of the desired monoclonal antibodies (mAbs) into a single bispecific structure. Bispecific immunotherapeutics has generated many different formats of BsAbs including chemical heteroconjugation of two complete molecules or fragments of monoclonal antibodies, quadroma, F(ab)2, diabodies, tandem diabodies, and single-chain antibodies (scFv). This chapter describes the process of generating activated T cells and arming T cells with heteroconjugated BsAbs to target cancer cells.
Collapse
Affiliation(s)
- Archana Thakur
- Division of Hematology/Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Lawrence G Lum
- Division of Hematology/Oncology, Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
26
|
|
27
|
Loukotová L, Kučka J, Rabyk M, Höcherl A, Venclíková K, Janoušková O, Páral P, Kolářová V, Heizer T, Šefc L, Štěpánek P, Hrubý M. Thermoresponsive β-glucan-based polymers for bimodal immunoradiotherapy – Are they able to promote the immune system? J Control Release 2017; 268:78-91. [DOI: 10.1016/j.jconrel.2017.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 01/05/2023]
|
28
|
Brinkmann U, Kontermann RE. The making of bispecific antibodies. MAbs 2017; 9:182-212. [PMID: 28071970 PMCID: PMC5297537 DOI: 10.1080/19420862.2016.1268307] [Citation(s) in RCA: 672] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022] Open
Abstract
During the past two decades we have seen a phenomenal evolution of bispecific antibodies for therapeutic applications. The 'zoo' of bispecific antibodies is populated by many different species, comprising around 100 different formats, including small molecules composed solely of the antigen-binding sites of two antibodies, molecules with an IgG structure, and large complex molecules composed of different antigen-binding moieties often combined with dimerization modules. The application of sophisticated molecular design and genetic engineering has solved many of the technical problems associated with the formation of bispecific antibodies such as stability, solubility and other parameters that confer drug properties. These parameters may be summarized under the term 'developability'. In addition, different 'target product profiles', i.e., desired features of the bispecific antibody to be generated, mandates the need for access to a diverse panel of formats. These may vary in size, arrangement, valencies, flexibility and geometry of their binding modules, as well as in their distribution and pharmacokinetic properties. There is not 'one best format' for generating bispecific antibodies, and no single format is suitable for all, or even most of, the desired applications. Instead, the bispecific formats collectively serve as a valuable source of diversity that can be applied to the development of therapeutics for various indications. Here, a comprehensive overview of the different bispecific antibody formats is provided.
Collapse
Affiliation(s)
- Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Im Nonnenwald, Penzberg, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, Nobelstraße, Stuttgart, Germany
| |
Collapse
|
29
|
Lloyd SB, Niven KP, Kiefel BR, Montefiori DC, Reynaldi A, Davenport MP, Kent SJ, Winnall WR. Exploration of broadly neutralizing antibody fragments produced in bacteria for the control of HIV. Hum Vaccin Immunother 2017; 13:2726-2737. [PMID: 28949787 DOI: 10.1080/21645515.2017.1368935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
While broadly neutralizing antibodies (bnAbs) are a promising preventative and therapeutic tool for HIV infection, production is difficult and expensive. Production of antibody-like fragments in bacterial cytoplasm provides a cheaper alternative. This work explored the transplantation of the complementarity determining regions of the anti-HIV bnAbs PGT121 and 10E8 onto a single-chain variable fragment (scFv) scaffold, previously discovered through a novel screening platform. The scaffolded 10E8 scFv, but not the scaffolded PGT121 scFv, was soluble in bacterial cytoplasm, enabling efficient production in bacteria. Three additional multimeric constructs employing the scaffolded 10E8 scFv were also generated and soluble versions produced in bacteria. However, the constructs were found to have substantially lost anti-HIV binding function and had completely abrogated neutralizing activity. Overall, while this study provides a proof-of-concept for anti-HIV bnAb construct production in bacterial cytoplasm, future refinement of these technologies will be required to realize the goal of producing inexpensive and effective bnAb-like tools for the control of HIV.
Collapse
Affiliation(s)
- Sarah B Lloyd
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia
| | - Keith P Niven
- b Affinity BIO Pty Ltd. , Melbourne , VIC , Australia
| | - Ben R Kiefel
- b Affinity BIO Pty Ltd. , Melbourne , VIC , Australia
| | - David C Montefiori
- c Department of Medicine , Duke University Medical Center , Durham , North Carolina , USA
| | - Arnold Reynaldi
- d Infection Analytics Program, Kirby Institute for Infection and Immunity , University of New South Wales Australia , Sydney , Australia
| | - Miles P Davenport
- d Infection Analytics Program, Kirby Institute for Infection and Immunity , University of New South Wales Australia , Sydney , Australia
| | - Stephen J Kent
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia.,e Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Health, Central Clinical School , Monash University , Melbourne , Australia.,f ARC Centre of Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Parkville , Australia
| | - Wendy R Winnall
- a Department of Microbiology and Immunology , The University of Melbourne, Peter Doherty Institute for Infection and Immunity , Victoria , Australia
| |
Collapse
|
30
|
Yumura K, Akiba H, Nagatoishi S, Kusano-Arai O, Iwanari H, Hamakubo T, Tsumoto K. Use of SpyTag/SpyCatcher to construct bispecific antibodies that target two epitopes of a single antigen. J Biochem 2017. [DOI: 10.1093/jb/mvx023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Kyohei Yumura
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
| | - Hiroki Akiba
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Satoru Nagatoishi
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Osamu Kusano-Arai
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Komaba, Tokyo 153-8904, Japan
| | - Hiroko Iwanari
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Komaba, Tokyo 153-8904, Japan
| | - Takao Hamakubo
- Department of Quantitative Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Komaba, Tokyo 153-8904, Japan
| | - Kouhei Tsumoto
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
- Medical Proteomics Laboratory, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
31
|
Wang F, Wang H, Shen YD, Li YJ, Dong JX, Xu ZL, Yang JY, Sun YM, Xiao ZL. Bispecific Monoclonal Antibody-Based Multianalyte ELISA for Furaltadone Metabolite, Malachite Green, and Leucomalachite Green in Aquatic Products. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:8054-8061. [PMID: 27706938 DOI: 10.1021/acs.jafc.6b03233] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A new multianalyte immunoassay was designed to screen furaltadone metabolite 5-morpholinomethyl-3-amino-2-oxazolidone (AMOZ), malachite green (MG), and leucomalachite green (LMG) in aquatic products using a bispecific monoclonal antibody (BsMAb). Gradient drug mutagenesis methods were separately used to prepare an anti-3-nitrobenzaldehyde-derivatized AMOZ (3-NPAMOZ) hybridoma cell line that was hypoxanthine-guanine-phosphoribosyltransferase (HGRPT) deficient and an anti-LMG hybridoma cell line that was thymidine kinase (TK) deficient. BsMAb recognizing 3-NPAMOZ and LMG was generated using hybrid-hybridomas of HGRPT and TK deficient cell lines. For AMOZ and LMG, respectively, the BsMAb-based indirect competitive ELSIA (ic-ELISA) values of 1.7 ng/mL and 45.3 ng/mL and detection limits of 0.2 ng/mL and 4.8 ng/mL. To establish the ic-ELISA, 3-NPAMOZ derivatized from AMOZ with 3-nitrobenzaldehyde and LMG reduced from MG by potassium borohydride was recognized by BsMAb. Recoveries of AMOZ, MG, and LMG in aquatic products were satisfactory and correlated with HPLC analysis. Thus, the multianalyte ic-ELISA is suitable for rapid quantification of AMOZ, MG, and LMG in aquatic products.
Collapse
Affiliation(s)
- Feng Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Hong Wang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yu-Dong Shen
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yong-Jun Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
- Zhongshan Quality Supervision & Inspection Institute of Agricultural Products , Zhongshan 528403, China
| | - Jie-Xian Dong
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California , Davis, California 95616, United States
| | - Zhen-Lin Xu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Jin-Yi Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Yuan-Ming Sun
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| | - Zhi-Li Xiao
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University , Guangzhou 510642, China
| |
Collapse
|
32
|
Ströhlein MA, Heiss MM, Jauch KW. The current status of immunotherapy in peritoneal carcinomatosis. Expert Rev Anticancer Ther 2016; 16:1019-27. [PMID: 27530056 DOI: 10.1080/14737140.2016.1224666] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Peritoneal carcinomatosis (PC) is a cancer disease with an urgent need for effective treatment. Conventional chemotherapy failed to show acceptable results. Cytoreductive surgery and hyperthermic chemoperfusion (HIPEC) are only beneficial in few patients with resectable peritoneal metastasis. Immunotherapy could be attractive against PC, as all requirements for immunotherapy are available in the peritoneal cavity. AREAS COVERED This review analyzes the present literature for immunotherapy of PC. Advances from immune stimulators, radionucleotide-conjugated- and bispecific antibodies to future developments like adoptive engineered T-cells with chimeric receptors are discussed. The clinical development of catumaxomab, which was the first intraperitoneal immunotherapy to be approved for clinical treatment, is discussed. The requirements for future developments are illustrated. Expert commentary: Immunotherapy of peritoneal carcinomatosis is manageable, showing striking cancer cell killing. Improved profiles of adverse events by therapy-induced cytokine release, enhanced specific killing and optimal treatment schedules within multimodal treatment will be key factors.
Collapse
Affiliation(s)
- Michael Alfred Ströhlein
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Markus Maria Heiss
- a Department of Abdominal, Vascular and Transplant Surgery, Cologne Merheim Medical Center , Witten/Herdecke University , Cologne , Germany
| | - Karl-Walter Jauch
- b Medical Center of the Ludwig Maximilian University Munich , Munich , Germany
| |
Collapse
|
33
|
Howard CB, Fletcher N, Houston ZH, Fuchs AV, Boase NRB, Simpson JD, Raftery LJ, Ruder T, Jones ML, de Bakker CJ, Mahler SM, Thurecht KJ. Overcoming Instability of Antibody-Nanomaterial Conjugates: Next Generation Targeted Nanomedicines Using Bispecific Antibodies. Adv Healthc Mater 2016; 5:2055-68. [PMID: 27283923 DOI: 10.1002/adhm.201600263] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/20/2016] [Indexed: 12/20/2022]
Abstract
Targeted nanomaterials promise improved therapeutic efficacy, however their application in nanomedicine is limited due to complexities associated with protein conjugations to synthetic nanocarriers. A facile method to generate actively targeted nanomaterials is developed and exemplified using polyethylene glycol (PEG)-functional nanostructures coupled to a bispecific antibody (BsAb) with dual specificity for methoxy PEG (mPEG) epitopes and cancer targets such as epidermal growth factor receptor (EGFR). The EGFR-mPEG BsAb binds with high affinity to recombinant EGFR (KD : 1 × 10(-9) m) and hyperbranched polymer (HBP) consisting of mPEG (KD : 10 × 10(-9) m) and demonstrates higher avidity for HBP compared to linear mPEG. The binding of BsAb-HBP bioconjugate to EGFR on MDA-MB-468 cancer cells is investigated in vitro using a fluorescently labeled polymer, and in in vivo xenograft models by small animal optical imaging. The antibody-targeted nanostructures show improved accumulation in tumor cells compared to non-targeted nanomaterials. This demonstrates a facile approach for tuning targeting ligand density on nanomaterials, by modulating surface functionality. Antibody fragments are tethered to the nanomaterial through simple mixing prior to administration to animals, overcoming the extensive procedures encountered for developing targeted nanomedicines.
Collapse
Affiliation(s)
- Christopher B. Howard
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Nicholas Fletcher
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Zachary H. Houston
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Adrian V. Fuchs
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Nathan R. B. Boase
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Joshua D. Simpson
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Lyndon J. Raftery
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Tim Ruder
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Martina L. Jones
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Christopher J. de Bakker
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Stephen M. Mahler
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| | - Kristofer J. Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN) Centre for Advanced Imaging (CAI) School of Chemical Engineering ARC Centre of Excellence in Convergent BioNano Science and Technology The University of Queensland Brisbane QLD 4072 Australia
| |
Collapse
|
34
|
Abstract
Bispecific antibodies that engage immune cells to kill cancer cells have been actively studied in cancer immunotherapy. In this study, we present a novel bispecific format, S-Fab, fabricated by linking a single-domain anti-carcinoembryonic antigen VHH to a conventional anti-CD3 Fab. In contrast to most bispecific antibodies, the S-Fab bispecific antibody can be efficiently expressed and purified from bacteria. The purified S-Fab is stable in serum and is able to recruit T cells to drive potent cancer cell killing. In xenograft models, the S-Fab antibody suppresses tumor growth in the presence of human immune cells. Our study suggested that the bispecific S-Fab format can be applied to a wide range of immunotherapies.
Collapse
|
35
|
Wang Q, Cheng H, Peng H, Zhou H, Li PY, Langer R. Non-genetic engineering of cells for drug delivery and cell-based therapy. Adv Drug Deliv Rev 2015; 91:125-40. [PMID: 25543006 DOI: 10.1016/j.addr.2014.12.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/04/2014] [Accepted: 12/18/2014] [Indexed: 12/13/2022]
Abstract
Cell-based therapy is a promising modality to address many unmet medical needs. In addition to genetic engineering, material-based, biochemical, and physical science-based approaches have emerged as novel approaches to modify cells. Non-genetic engineering of cells has been applied in delivering therapeutics to tissues, homing of cells to the bone marrow or inflammatory tissues, cancer imaging, immunotherapy, and remotely controlling cellular functions. This new strategy has unique advantages in disease therapy and is complementary to existing gene-based cell engineering approaches. A better understanding of cellular systems and different engineering methods will allow us to better exploit engineered cells in biomedicine. Here, we review non-genetic cell engineering techniques and applications of engineered cells, discuss the pros and cons of different methods, and provide our perspectives on future research directions.
Collapse
|
36
|
Bispecific antibodies. Drug Discov Today 2015; 20:838-47. [PMID: 25728220 DOI: 10.1016/j.drudis.2015.02.008] [Citation(s) in RCA: 440] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 11/23/2022]
Abstract
Bispecific antibodies (bsAbs) combine specificities of two antibodies and simultaneously address different antigens or epitopes. BsAbs with 'two-target' functionality can interfere with multiple surface receptors or ligands associated, for example with cancer, proliferation or inflammatory processes. BsAbs can also place targets into close proximity, either to support protein complex formation on one cell, or to trigger contacts between cells. Examples of 'forced-connection' functionalities are bsAbs that support protein complexation in the clotting cascade, or tumor-targeted immune cell recruiters and/or activators. Following years of research and development (R&D), the first bsAb was approved in 2009. Another bsAb entered the market in December 2014 and several more are in clinical trials. Here, we describe the potentials of bsAbs to become the next wave of antibody-based therapies, focusing on molecules in clinical development.
Collapse
|
37
|
Deyev SM, Lebedenko EN, Petrovskaya LE, Dolgikh DA, Gabibov AG, Kirpichnikov MP. Man-made antibodies and immunoconjugates with desired properties: function optimization using structural engineering. RUSSIAN CHEMICAL REVIEWS 2015. [DOI: 10.1070/rcr4459] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Liu Z, Leng EC, Gunasekaran K, Pentony M, Shen M, Howard M, Stoops J, Manchulenko K, Razinkov V, Liu H, Fanslow W, Hu Z, Sun N, Hasegawa H, Clark R, Foltz IN, Yan W. A novel antibody engineering strategy for making monovalent bispecific heterodimeric IgG antibodies by electrostatic steering mechanism. J Biol Chem 2015; 290:7535-62. [PMID: 25583986 DOI: 10.1074/jbc.m114.620260] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Producing pure and well behaved bispecific antibodies (bsAbs) on a large scale for preclinical and clinical testing is a challenging task. Here, we describe a new strategy for making monovalent bispecific heterodimeric IgG antibodies in mammalian cells. We applied an electrostatic steering mechanism to engineer antibody light chain-heavy chain (LC-HC) interface residues in such a way that each LC strongly favors its cognate HC when two different HCs and two different LCs are co-expressed in the same cell to assemble a functional bispecific antibody. We produced heterodimeric IgGs from transiently and stably transfected mammalian cells. The engineered heterodimeric IgG molecules maintain the overall IgG structure with correct LC-HC pairings, bind to two different antigens with comparable affinity when compared with their parental antibodies, and retain the functionality of parental antibodies in biological assays. In addition, the bispecific heterodimeric IgG derived from anti-HER2 and anti-EGF receptor (EGFR) antibody was shown to induce a higher level of receptor internalization than the combination of two parental antibodies. Mouse xenograft BxPC-3, Panc-1, and Calu-3 human tumor models showed that the heterodimeric IgGs strongly inhibited tumor growth. The described approach can be used to generate tools from two pre-existent antibodies and explore the potential of bispecific antibodies. The asymmetrically engineered Fc variants for antibody-dependent cellular cytotoxicity enhancement could be embedded in monovalent bispecific heterodimeric IgG to make best-in-class therapeutic antibodies.
Collapse
Affiliation(s)
- Zhi Liu
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119,
| | - Esther C Leng
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Kannan Gunasekaran
- From the Departments of Therapeutic Discovery and Amgen Inc., Thousand Oaks, California 91320, and
| | - Martin Pentony
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Min Shen
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Monique Howard
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Janelle Stoops
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Kathy Manchulenko
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Vladimir Razinkov
- Amgen Inc., Seattle, Washington 98119, Process and Product Development
| | - Hua Liu
- Amgen Inc., Seattle, Washington 98119, Therapeutic Innovation Unit
| | - William Fanslow
- Amgen Inc., Seattle, Washington 98119, Therapeutic Innovation Unit
| | - Zhonghua Hu
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Nancy Sun
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Haruki Hasegawa
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Rutilio Clark
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119
| | - Ian N Foltz
- From the Departments of Therapeutic Discovery and Amgen Inc., Burnaby, British Columbia V5A 1V7, Canada
| | - Wei Yan
- From the Departments of Therapeutic Discovery and Amgen Inc., Seattle, Washington 98119,
| |
Collapse
|
39
|
Alkaline cation-exchange chromatography for the reduction of aggregate and a mis-formed disulfide variant in a bispecific antibody purification process. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 975:1-8. [DOI: 10.1016/j.jchromb.2014.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/24/2014] [Accepted: 11/02/2014] [Indexed: 11/19/2022]
|
40
|
McEnaney PJ, Fitzgerald KJ, Zhang AX, Douglass EF, Shan W, Balog A, Kolesnikova MD, Spiegel DA. Chemically synthesized molecules with the targeting and effector functions of antibodies. J Am Chem Soc 2014; 136:18034-43. [PMID: 25514603 PMCID: PMC4291750 DOI: 10.1021/ja509513c] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Indexed: 12/21/2022]
Abstract
This article reports the design, synthesis, and evaluation of a novel class of molecules of intermediate size (approximately 7000 Da), which possess both the targeting and effector functions of antibodies. These compounds—called synthetic antibody mimics targeting prostate cancer (SyAM-Ps)—bind simultaneously to prostate-specific membrane antigen and Fc gamma receptor I, thus eliciting highly selective cancer cell phagocytosis. SyAMs have the potential to combine the advantages of both small-molecule and biologic therapies, and may address many drawbacks associated with available treatments for cancer and other diseases.
Collapse
Affiliation(s)
- Patrick J McEnaney
- Department of Chemistry, Yale University , 225 Prospect Street, New Haven, Connecticut 06511, United States
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Schuster FR, Stanglmaier M, Woessmann W, Winkler B, Siepermann M, Meisel R, Schlegel PG, Hess J, Lindhofer H, Borkhardt A, Buhmann R. Immunotherapy with the trifunctional anti-CD20 x anti-CD3 antibody FBTA05 (Lymphomun) in paediatric high-risk patients with recurrent CD20-positive B cell malignancies. Br J Haematol 2014; 169:90-102. [PMID: 25495919 DOI: 10.1111/bjh.13242] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/04/2014] [Indexed: 12/23/2022]
Abstract
Children with B cell malignancies refractory to standard therapy are known to have a poor prognosis and very limited treatment options. Here, we report on the treatment and follow-up of ten patients diagnosed with relapsed or refractory mature B-cell Non Hodgkin Lymphoma (B-NHL), Burkitt leukaemia (B-AL) or pre B-acute lymphoblastic leukaemia (pre B-ALL). All children were treated with FBTA05 (now designated Lymphomun), an anti-CD3 x anti-CD20 trifunctional bispecific antibody (trAb) in compassionate use. Within individual treatment schedules, Lymphomun was applied (a) after allogeneic stem cell transplantation (allo-SCT, n = 6) to induce sustained long-term remission, or (b) stand alone prior to subsequent chemotherapy to eradicate residual disease before allo-SCT (n = 4). Nine of ten children displayed a clinical response: three stable diseases (SD), one partial remission (PR) and five induced or sustained complete remissions (CR). Five of these nine responders died during follow-up. The other patients still maintain CR with a current overall survival of 874-1424 days (median: 1150 days). In conclusion, despite the dismal clinical prognosis of children refractory to standard therapy, immunotherapy with Lymphomun resulted in a favourable clinical outcome in this cohort of refractory paediatric patients.
Collapse
Affiliation(s)
- Friedhelm R Schuster
- Department of Paediatric Oncology, Haematology and Immunology, University of Duesseldorf, Duesseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Bi- and multispecific antibody derivatives (bsAbs) can be considered as the next generation of targeted biologics for cancer therapy. The general concept of bsAbs is a physical connection of recombinant antibody-derived entities with at least two binding specificities. This generates bsAbs that bind at least two antigens or epitopes, thus altering their binding functionalities and specificities in comparison to "normal" antibodies. Most bsAbs are produced as recombinant proteins, either as large IgG-like proteins that contain Fc regions, or as smaller entities with multiple antigen-binding regions but without Fc. Application of bsAbs in experimental cancer therapy currently includes molecules that bind different cell surface proteins to achieve more complete blockage of proliferative or angiogenesis-associated pathways. This approach of blocking more than one pathway component, or to simultaneously hit complementing pathways, also may limit potential escape mechanisms of cancer cells. BsAbs also are applied in the clinic as vehicles to deliver immune effector cells and/or cytokines to tumors.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharmaceuticals Research and Early Development (pRED), Discovery Oncology (UHW) and Large Molecule Research (UB), Roche Innovation Center Penzberg, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Ulrich Brinkmann
- Roche Pharmaceuticals Research and Early Development (pRED), Discovery Oncology (UHW) and Large Molecule Research (UB), Roche Innovation Center Penzberg, Germany.
| |
Collapse
|
43
|
Luo H, Hong H, Yang SP, Cai W. Design and applications of bispecific heterodimers: molecular imaging and beyond. Mol Pharm 2014; 11:1750-61. [PMID: 24738564 PMCID: PMC4051252 DOI: 10.1021/mp500115x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ligand-based molecular imaging probes have been designed with high affinity and specificity for monitoring biological process and responses. Single-target recognition by traditional probes can limit their applicability for disease detection and therapy because synergistic action between disease mediators and different receptors is often involved in disease progression. Consequently, probes that can recognize multiple targets should demonstrate higher targeting efficacy and specificity than their monospecific peers. This concept has been validated by multiple bispecific heterodimer-based imaging probes that have demonstrated promising results in several animal models. This review summarizes the design strategies for bispecific peptide- and antibody-based heterodimers and their applications in molecular targeting and imaging. The design and application of bispecific heterodimer-conjugated nanomaterials are also discussed.
Collapse
Affiliation(s)
- Haiming Luo
- Department of Radiology, University of Wisconsin-Madison , Madison, Wisconsin 53705-2275, United States
| | | | | | | |
Collapse
|
44
|
Liu Z, Gunasekaran K, Wang W, Razinkov V, Sekirov L, Leng E, Sweet H, Foltz I, Howard M, Rousseau AM, Kozlosky C, Fanslow W, Yan W. Asymmetrical Fc engineering greatly enhances antibody-dependent cellular cytotoxicity (ADCC) effector function and stability of the modified antibodies. J Biol Chem 2013; 289:3571-90. [PMID: 24311787 DOI: 10.1074/jbc.m113.513366] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is mediated through the engagement of the Fc segment of antibodies with Fcγ receptors (FcγRs) on immune cells upon binding of tumor or viral antigen. The co-crystal structure of FcγRIII in complex with Fc revealed that Fc binds to FcγRIII asymmetrically with two Fc chains contacting separate regions of the FcγRIII by utilizing different residues. To fully explore this asymmetrical nature of the Fc-FcγR interaction, we screened more than 9,000 individual clones in Fc heterodimer format in which different mutations were introduced at the same position of two Fc chains using a high throughput competition AlphaLISA® assay. To this end, we have identified a panel of novel Fc variants with significant binding improvement to FcγRIIIA (both Phe-158 and Val-158 allotypes), increased ADCC activity in vitro, and strong tumor growth inhibition in mice xenograft human tumor models. Compared with previously identified Fc variants in conventional IgG format, Fc heterodimers with asymmetrical mutations can achieve similar or superior potency in ADCC-mediated tumor cell killing and demonstrate improved stability in the CH2 domain. Fc heterodimers also allow more selectivity toward activating FcγRIIA than inhibitory FcγRIIB. Afucosylation of Fc variants further increases the affinity of Fc to FcγRIIIA, leading to much higher ADCC activity. The discovery of these Fc variants will potentially open up new opportunities of building the next generation of therapeutic antibodies with enhanced ADCC effector function for the treatment of cancers and infectious diseases.
Collapse
Affiliation(s)
- Zhi Liu
- From the Departments of Therapeutic Discovery and
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Khalili H, Godwin A, Choi JW, Lever R, Khaw PT, Brocchini S. Fab-PEG-Fab as a Potential Antibody Mimetic. Bioconjug Chem 2013; 24:1870-82. [DOI: 10.1021/bc400246z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Hanieh Khalili
- UCL
School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
- NIHR
Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, EC1 V 9EL, United Kingdom
| | - Antony Godwin
- PolyTherics
Ltd, The London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, United Kingdom
| | - Ji-won Choi
- PolyTherics
Ltd, The London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, United Kingdom
| | - Rebecca Lever
- UCL
School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
| | - Peng T. Khaw
- NIHR
Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, EC1 V 9EL, United Kingdom
| | - Steve Brocchini
- UCL
School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, United Kingdom
- NIHR
Biomedical Research Centre, Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, EC1 V 9EL, United Kingdom
| |
Collapse
|
46
|
Gramer MJ, van den Bremer ETJ, van Kampen MD, Kundu A, Kopfmann P, Etter E, Stinehelfer D, Long J, Lannom T, Noordergraaf EH, Gerritsen J, Labrijn AF, Schuurman J, van Berkel PHC, Parren PWHI. Production of stable bispecific IgG1 by controlled Fab-arm exchange: scalability from bench to large-scale manufacturing by application of standard approaches. MAbs 2013; 5:962-73. [PMID: 23995617 PMCID: PMC3896610 DOI: 10.4161/mabs.26233] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The manufacturing of bispecific antibodies can be challenging for a variety of reasons. For example, protein expression problems, stability issues, or the use of non-standard approaches for manufacturing can result in poor yield or poor facility fit. In this paper, we demonstrate the use of standard antibody platforms for large-scale manufacturing of bispecific IgG1 by controlled Fab-arm exchange. Two parental antibodies that each contain a single matched point mutation in the CH3 region were separately expressed in Chinese hamster ovary cells and manufactured at 1000 L scale using a platform fed-batch and purification process that was designed for standard antibody production. The bispecific antibody was generated by mixing the two parental molecules under controlled reducing conditions, resulting in efficient Fab-arm exchange of >95% at kg scale. The reductant was removed via diafiltration, resulting in spontaneous reoxidation of interchain disulfide bonds. Aside from the bispecific nature of the molecule, extensive characterization demonstrated that the IgG1 structural integrity was maintained, including function and stability. These results demonstrate the suitability of this bispecific IgG1 format for commercial-scale manufacturing using standard antibody manufacturing techniques.
Collapse
|
47
|
|
48
|
Fournier P, Schirrmacher V. Bispecific antibodies and trispecific immunocytokines for targeting the immune system against cancer: preparing for the future. BioDrugs 2013; 27:35-53. [PMID: 23329400 DOI: 10.1007/s40259-012-0008-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Monoclonal anti-tumor antibodies (mAbs) that are clinically effective usually recruit, via their constant fragment (Fc) domain, Fc receptor (FcR)-positive accessory cells of the immune system and engage these additionally against the tumor. Since T cells are FcR negative, these important cells are not getting involved. In contrast to mAbs, bispecific antibodies (bsAbs) can be designed in such a way that they involve T cells. bsAbs are artificially designed molecules that bind simultaneously to two different antigens, one on the tumor cell, the other one on an immune effector cell such as CD3 on T cells. Such dual antibody constructs can cross-link tumor cells and T cells. Many such bsAb molecules at the surface of tumor cells can thus build a bridge to T cells and aggregate their CD3 molecules, thereby activating them for cytotoxic activity. BsAbs can also contain a third binding site, for instance a Fc domain or a cytokine that would bind to its respective cytokine receptor. The present review discusses the pros and cons for the use of the Fc fragment during the development of bsAbs using either cell-fusion or recombinant DNA technologies. The recombinant antibody technology allows the generation of very efficient bsAbs containing no Fc domain such as the bi-specific T-cell engager (BiTE). The strong antitumor activity of these molecules makes them very interesting new cancer therapeutics. Over the last decade, we have developed another concept, namely to combine bsAbs and multivalent immunocytokines with a tumor cell vaccine. The latter are patient-derived tumor cells modified by infection with a virus. The virus-Newcastle Disease Virus (NDV)-introduces, at the surface of the tumor cells, viral molecules that can serve as general anchors for the bsAbs. Our strategy aims at redirecting, in an Fc-independent fashion, activities of T cells and accessory cells against autologous tumor antigens. It creates very promising perspectives for a new generation of efficient and safe cancer therapeutics that should confer long-lasting anti-tumor immunity.
Collapse
Affiliation(s)
- Philippe Fournier
- German Cancer Research Center DKFZ, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany,
| | | |
Collapse
|
49
|
Efficient generation of stable bispecific IgG1 by controlled Fab-arm exchange. Proc Natl Acad Sci U S A 2013; 110:5145-50. [PMID: 23479652 DOI: 10.1073/pnas.1220145110] [Citation(s) in RCA: 254] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The promise of bispecific antibodies (bsAbs) to yield more effective therapeutics is well recognized; however, the generation of bsAbs in a practical and cost-effective manner has been a formidable challenge. Here we present a technology for the efficient generation of bsAbs with normal IgG structures that is amenable to both antibody drug discovery and development. The process involves separate expression of two parental antibodies, each containing single matched point mutations in the CH3 domains. The parental antibodies are mixed and subjected to controlled reducing conditions in vitro that separate the antibodies into HL half-molecules and allow reassembly and reoxidation to form highly pure bsAbs. The technology is compatible with standard large-scale antibody manufacturing and ensures bsAbs with Fc-mediated effector functions and in vivo stability typical of IgG1 antibodies. Proof-of-concept studies with HER2×CD3 (T-cell recruitment) and HER2×HER2 (dual epitope targeting) bsAbs demonstrate superior in vivo activity compared with parental antibody pairs.
Collapse
|
50
|
Asano R, Kumagai T, Nagai K, Taki S, Shimomura I, Arai K, Ogata H, Okada M, Hayasaka F, Sanada H, Nakanishi T, Karvonen T, Hayashi H, Katayose Y, Unno M, Kudo T, Umetsu M, Kumagai I. Domain order of a bispecific diabody dramatically enhances its antitumor activity beyond structural format conversion: the case of the hEx3 diabody. Protein Eng Des Sel 2013; 26:359-67. [DOI: 10.1093/protein/gzt009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|