1
|
Callahan V, Sutton MS, Gardner CL, Kenchegowda D, Dunagan MM, Gosavi M, Green C, Chen TY, Prado-Smith J, Long D, Vogel JL, Kristie TM, Clancy CS, Burke CW, Roederer M, Fox JM. Requirement for Fc effector function is overcome by binding potency for broadly reactive anti-alphavirus antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.03.619087. [PMID: 39605662 PMCID: PMC11601667 DOI: 10.1101/2024.11.03.619087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Alphaviruses are emerging public health threats. Broadly reactive anti-alphavirus monoclonal antibodies (mAbs) have been shown to be protective in mouse models of infection. However, the mechanism of Fc-dependent or Fc-independent heterologous protection remains ill-defined in vivo. Here, we used two vaccine-elicited, broadly reactive, anti-alphavirus mAbs, SKT05 and SKT20, to establish correlates of mAb-mediated protection during Venezuelan equine encephalitis virus (VEEV) challenge. SKT20 required Fc effector functions to prevent lethality. In contrast, SKT05-mediated survival was independent of Fc effector functions, which is likely linked to early viral control through potent egress inhibition. However, control of virus replication and spread with SKT05 was Fc-dependent; these findings extended to additional in vivo models with alternative VEEV subtypes and chikungunya virus. During therapeutic delivery of SKT05, Fc effector functions were only required at 3 days post-infection. The necessity of Fc effector functions for SKT20 was related to mAb binding avidity rather than epitope and could be overcome by increasing the dose of SKT20 relative to the functional avidity of SKT05. Collectively, this study identified antibody avidity as a correlate for in vivo efficacy and associated Fc-dependent mechanisms that can be leveraged for therapeutic development of monoclonal antibodies against alphaviruses.
Collapse
Affiliation(s)
- Victoria Callahan
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew S. Sutton
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christina L. Gardner
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, USA
| | - Doreswamy Kenchegowda
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan M. Dunagan
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mrunal Gosavi
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Courtney Green
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tammy Y. Chen
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Prado-Smith
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Daniel Long
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jodi L. Vogel
- Molecular Genetics Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Thomas M. Kristie
- Molecular Genetics Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Crystal W. Burke
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie M. Fox
- Emerging Virus Immunity Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Gardner CL, Erwin-Cohen RA, Lewis BS, Bakken RR, Honnold SP, Glass PJ, Burke CW. Syrian Hamsters Model Does Not Reflect Human-like Disease after Aerosol Exposure to Encephalitic Alphaviruses. Methods Protoc 2024; 7:42. [PMID: 38804336 PMCID: PMC11130913 DOI: 10.3390/mps7030042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Venezuelan (VEE), eastern (EEE), and western (WEE) equine encephalitis viruses are encephalitic New World alphaviruses that cause periodic epizootic and epidemic outbreaks in horses and humans that may cause severe morbidity and mortality. Currently there are no FDA-licensed vaccines or effective antiviral therapies. Each year, there are a limited number of human cases of encephalitic alphaviruses; thus, licensure of a vaccine or therapeutic would require approval under the FDA animal rule. Approval under the FDA animal rule requires the disease observed in the animal model to recapitulate what is observed in humans. Currently, initial testing of vaccines and therapeutics is performed in the mouse model. Unfortunately, alphavirus disease manifestations in a mouse do not faithfully recapitulate human disease; the VEEV mouse model is lethal whereas in humans VEEV is rarely lethal. In an effort to identify a more appropriate small animal model, we evaluated hamsters in an aerosol exposure model of encephalitic alphavirus infection. The pathology, lethality, and viremia observed in the infected hamsters was inconsistent with what is observed in NHP models and humans. These data suggest that hamsters are not an appropriate model for encephalitic alphaviruses to test vaccines or potential antiviral therapies.
Collapse
Affiliation(s)
- Christina L. Gardner
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.L.G.); (R.A.E.-C.); (R.R.B.); (P.J.G.)
| | - Rebecca A. Erwin-Cohen
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.L.G.); (R.A.E.-C.); (R.R.B.); (P.J.G.)
| | - Bridget S. Lewis
- Pathology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (B.S.L.); (S.P.H.)
| | - Russell R. Bakken
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.L.G.); (R.A.E.-C.); (R.R.B.); (P.J.G.)
| | - Shelley P. Honnold
- Pathology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (B.S.L.); (S.P.H.)
| | - Pamela J. Glass
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.L.G.); (R.A.E.-C.); (R.R.B.); (P.J.G.)
- Risk Management Office, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Crystal W. Burke
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.L.G.); (R.A.E.-C.); (R.R.B.); (P.J.G.)
| |
Collapse
|
3
|
Schwedler JL, Stefan MA, Thatcher CE, McIlroy PR, Sinha A, Phillips AM, Sumner CA, Courtney CM, Kim CY, Weilhammer DR, Harmon B. Therapeutic efficacy of a potent anti-Venezuelan equine encephalitis virus antibody is contingent on Fc effector function. MAbs 2024; 16:2297451. [PMID: 38170638 PMCID: PMC10766394 DOI: 10.1080/19420862.2023.2297451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
The development of specific, safe, and potent monoclonal antibodies (Abs) has led to novel therapeutic options for infectious disease. In addition to preventing viral infection through neutralization, Abs can clear infected cells and induce immunomodulatory functions through engagement of their crystallizable fragment (Fc) with complement proteins and Fc receptors on immune cells. Little is known about the role of Fc effector functions of neutralizing Abs in the context of encephalitic alphavirus infection. To determine the role of Fc effector function in therapeutic efficacy against Venezuelan equine encephalitis virus (VEEV), we compared the potently neutralizing anti-VEEV human IgG F5 (hF5) Ab with intact Fc function (hF5-WT) or containing the loss of function Fc mutations L234A and L235A (hF5-LALA) in the context of VEEV infection. We observed significantly reduced binding to complement and Fc receptors, as well as differential in vitro kinetics of Fc-mediated cytotoxicity for hF5-LALA compared to hF5-WT. The in vivo efficacy of hF5-LALA was comparable to hF5-WT at -24 and + 24 h post infection, with both Abs providing high levels of protection. However, when hF5-WT and hF5-LALA were administered + 48 h post infection, there was a significant decrease in the therapeutic efficacy of hF5-LALA. Together these results demonstrate that optimal therapeutic Ab treatment of VEEV, and possibly other encephalitic alphaviruses, requires neutralization paired with engagement of immune effectors via the Fc region.
Collapse
Affiliation(s)
- Jennifer L. Schwedler
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Maxwell A. Stefan
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Christine E. Thatcher
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Peter R. McIlroy
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Anupama Sinha
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Ashlee M. Phillips
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Christopher A. Sumner
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Colleen M. Courtney
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Christina Y. Kim
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| | - Dina R. Weilhammer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Brooke Harmon
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, CA, USA
| |
Collapse
|
4
|
Burke CW, Gardner CL, Goodson AI, Piper AE, Erwin-Cohen RA, White CE, Glass PJ. Defining the Cynomolgus Macaque ( Macaca fascicularis) Animal Model for Aerosolized Venezuelan Equine Encephalitis: Importance of Challenge Dose and Viral Subtype. Viruses 2023; 15:2351. [PMID: 38140592 PMCID: PMC10748030 DOI: 10.3390/v15122351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Venezuelan equine encephalitis virus (VEEV) outbreaks occur sporadically. Additionally, VEEV has a history of development as a biothreat agent. Yet, no FDA-approved vaccine or therapeutic exists for VEEV disease. The sporadic outbreaks present a challenge for testing medical countermeasures (MCMs) in humans; therefore, well-defined animal models are needed for FDA Animal Rule licensure. The cynomolgus macaque (CM) model has been studied extensively at high challenge doses of the VEEV Trinidad donkey strain (>1.0 × 108 plaque-forming units [PFU]), doses that are too high to be a representative human dose. Based on viremia of two subtypes of VEEV, IC, and IAB, we found the CM infectious dose fifty (ID50) to be low, 12 PFU, and 6.7 PFU, respectively. Additionally, we characterized the pattern of three clinical parameters (viremia, temperature, and lymphopenia) across a range of doses to identify a challenge dose producing consistent signs of infection. Based on these studies, we propose a shift to using a lower challenge dose of 1.0 × 103 PFU in the aerosol CM model of VEEV disease. At this dose, NHPs had the highest viremia, demonstrated a fever response, and had a measurable reduction in complete lymphocyte counts-biomarkers that can demonstrate MCM efficacy.
Collapse
Affiliation(s)
- Crystal W. Burke
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
| | - Christina L. Gardner
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
| | - Aimee I. Goodson
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
| | - Ashley E. Piper
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
| | - Rebecca A. Erwin-Cohen
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
| | - Charles E. White
- Statistics Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| | - Pamela J. Glass
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA (A.I.G.)
- Risk Management Office, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA
| |
Collapse
|
5
|
Guliy OI, Evstigneeva SS, Khanadeev VA, Dykman LA. Antibody Phage Display Technology for Sensor-Based Virus Detection: Current Status and Future Prospects. BIOSENSORS 2023; 13:640. [PMID: 37367005 DOI: 10.3390/bios13060640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023]
Abstract
Viruses are widespread in the environment, and many of them are major pathogens of serious plant, animal, and human diseases. The risk of pathogenicity, together with the capacity for constant mutation, emphasizes the need for measures to rapidly detect viruses. The need for highly sensitive bioanalytical methods to diagnose and monitor socially significant viral diseases has increased in the past few years. This is due, on the one hand, to the increased incidence of viral diseases in general (including the unprecedented spread of a new coronavirus infection, SARS-CoV-2), and, on the other hand, to the need to overcome the limitations of modern biomedical diagnostic methods. Phage display technology antibodies as nano-bio-engineered macromolecules can be used for sensor-based virus detection. This review analyzes the commonly used virus detection methods and approaches and shows the prospects for the use of antibodies prepared by phage display technology as sensing elements for sensor-based virus detection.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospect Entuziastov, Saratov 410049, Russia
| | - Stella S Evstigneeva
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospect Entuziastov, Saratov 410049, Russia
| | - Vitaly A Khanadeev
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospect Entuziastov, Saratov 410049, Russia
| | - Lev A Dykman
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 13 Prospect Entuziastov, Saratov 410049, Russia
| |
Collapse
|
6
|
Samsel J, Boswell KL, Watkins T, Ambrozak DR, Mason R, Yamamoto T, Ko S, Yang Y, Zhou T, Doria-Rose NA, Foulds KE, Roederer M, Mascola JR, Kwong PD, Gama L, Koup RA. Rhesus macaque Bcl-6/Bcl-xL B cell immortalization: Discovery of HIV-1 neutralizing antibodies from lymph node. J Immunol Methods 2023; 516:113445. [PMID: 36848985 DOI: 10.1016/j.jim.2023.113445] [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: 01/03/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Abstract
Many HIV-1 vaccines are designed to elicit neutralizing antibodies, and pre-clinical testing is often carried out in rhesus macaques (RMs). We have therefore adapted a method of B cell immortalization for use with RM B cells. In this system, RM B cells are activated with CD40 ligand and RM IL-21 before transduction with a retroviral vector encoding Bcl-6, Bcl-xL, and green fluorescent protein. Importantly, RM B cells from lymph nodes are more effectively immortalized by this method than B cells from PBMC, a difference not seen in humans. We suggest the discrepancy between these two tissues is due to increased expression of CD40 on RM lymph node B cells. Immortalized RM B cells expand long-term, undergo minimal somatic hypermutation, express surface B cell receptor, and secrete antibodies into culture. This allows for the identification of cells based on antigen specificity and/or functional assays. Here, we show the characterization of this system and its application for the isolation of HIV-1 neutralizing antibodies from a SHIV.CH505-infected animal, both with and without antigen probe. Taken together, we show that Bcl-6/xL immortalization is a valuable and flexible tool for antibody discovery in RMs, but with important distinctions from application of the system in human cells.
Collapse
Affiliation(s)
- Jakob Samsel
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America; Institute for Biomedical Sciences, George Washington University, Washington, D.C., United States of America.
| | - Kristin L Boswell
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| | - Timothy Watkins
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| | - David R Ambrozak
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| | - Rosemarie Mason
- ImmunoTechnology Section, VRC; Humoral Immunology Section, VRC
| | - Takuya Yamamoto
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| | | | | | | | | | | | | | | | | | - Lucio Gama
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center (VRC), NIAID, NIH, Bethesda, MD, United States of America
| |
Collapse
|
7
|
Mahdavi SZB, Oroojalian F, Eyvazi S, Hejazi M, Baradaran B, Pouladi N, Tohidkia MR, Mokhtarzadeh A, Muyldermans S. An overview on display systems (phage, bacterial, and yeast display) for production of anticancer antibodies; advantages and disadvantages. Int J Biol Macromol 2022; 208:421-442. [PMID: 35339499 DOI: 10.1016/j.ijbiomac.2022.03.113] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 03/17/2022] [Indexed: 11/05/2022]
Abstract
Antibodies as ideal therapeutic and diagnostic molecules are among the top-selling drugs providing considerable efficacy in disease treatment, especially in cancer therapy. Limitations of the hybridoma technology as routine antibody generation method in conjunction with numerous developments in molecular biology led to the development of alternative approaches for the streamlined identification of most effective antibodies. In this regard, display selection technologies such as phage display, bacterial display, and yeast display have been widely promoted over the past three decades as ideal alternatives to traditional methods. The display of antibodies on phages is probably the most widespread of these methods, although surface display on bacteria or yeast have been employed successfully, as well. These methods using various sizes of combinatorial antibody libraries and different selection strategies possessing benefits in screening potency, generating, and isolation of high affinity antibodies with low risk of immunogenicity. Knowing the basics of each method assists in the design and retrieval process of antibodies suitable for different diseases, including cancer. In this review, we aim to outline the basics of each library construction and its display method, screening and selection steps. The advantages and disadvantages in comparison to alternative methods, and their applications in antibody engineering will be explained. Finally, we will review approved or non-approved therapeutic antibodies developed by employing these methods, which may serve as therapeutic antibodies in cancer therapy.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Shirin Eyvazi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Maryam Hejazi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Serge Muyldermans
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, China..
| |
Collapse
|
8
|
Avril A, Tournier JN, Paucod JC, Fournes B, Thullier P, Pelat T. Antibodies against Anthrax Toxins: A Long Way from Benchlab to the Bedside. Toxins (Basel) 2022; 14:172. [PMID: 35324669 PMCID: PMC8955606 DOI: 10.3390/toxins14030172] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
Anthrax is an acute disease caused by the bacterium Bacillus anthracis, and is a potential biowarfare/bioterrorist agent. Its pulmonary form, caused by inhalation of the spores, is highly lethal and is mainly related to injury caused by the toxins secretion. Antibodies neutralizing the toxins of B. anthracis are regarded as promising therapeutic drugs, and two are already approved by the Federal Drug Administration. We developed a recombinant human-like humanized antibody, 35PA83 6.20, that binds the protective antigen and that neutralized anthrax toxins in-vivo in White New Zealand rabbits infected with the lethal 9602 strain by intranasal route. Considering these promising results, the preclinical and clinical phase one development was funded and a program was started. Unfortunately, after 5 years, the preclinical development was cancelled due to industrial and scientific issues. This shutdown underlined the difficulty particularly, but not only, for an academic laboratory to proceed to clinical development, despite the drug candidate being promising. Here, we review our strategy and some preliminary results, and we discuss the issues that led to the no-go decision of the pre-clinical development of 35PA83 6.20 mAb. Our review provides general information to the laboratories planning a (pre-)clinical development.
Collapse
Affiliation(s)
- Arnaud Avril
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Jean-Nicolas Tournier
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
- Ecole du Val-de-Grâce, 75005 Paris, France
| | - Jean-Charles Paucod
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Bénédicte Fournes
- Laboratoire Français du Fractionnement et des Biotechnologies, 91940 Les Ulis, France;
| | - Philippe Thullier
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| | - Thibaut Pelat
- Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France; (J.-N.T.); (J.-C.P.); (P.T.); (T.P.)
| |
Collapse
|
9
|
Calvert AE, Bennett SL, Hunt AR, Fong RH, Doranz BJ, Roehrig JT, Blair CD. Exposing cryptic epitopes on the Venezuelan equine encephalitis virus E1 glycoprotein prior to treatment with alphavirus cross-reactive monoclonal antibody allows blockage of replication early in infection. Virology 2021; 565:13-21. [PMID: 34626907 PMCID: PMC8765347 DOI: 10.1016/j.virol.2021.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 01/17/2023]
Abstract
Eastern equine encephalitis virus (EEEV), western equine encephalitis virus (WEEV) and Venezuelan equine encephalitis virus (VEEV) can cause fatal encephalitis in humans and equids. Some MAbs to the E1 glycoprotein are known to be cross-reactive, weakly neutralizing in vitro but can protect from disease in animal models. We investigated the mechanism of neutralization of VEEV infection by the broadly cross-reactive E1-specific MAb 1A4B-6. 1A4B-6 protected 3-week-old Swiss Webster mice prophylactically from lethal VEEV challenge. Likewise, 1A4B-6 inhibited virus growth in vitro at a pre-attachment step after virions were incubated at 37 °C and inhibited virus-mediated cell fusion. Amino acid residue N100 in the fusion loop of E1 protein was identified as critical for binding. The potential to elicit broadly cross-reactive MAbs with limited virus neutralizing activity in vitro but that can inhibit virus entry and protect animals from infection merits further exploration for vaccine and therapeutic developmental research.
Collapse
Affiliation(s)
- Amanda E Calvert
- Arboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA.
| | - Susan L Bennett
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Ann R Hunt
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | - John T Roehrig
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Carol D Blair
- Center for Vector-borne Infectious Diseases, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
10
|
Guerrero-Arguero I, Tellez-Freitas CM, Weber KS, Berges BK, Robison RA, Pickett BE. Alphaviruses: Host pathogenesis, immune response, and vaccine & treatment updates. J Gen Virol 2021; 102. [PMID: 34435944 DOI: 10.1099/jgv.0.001644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human pathogens belonging to the Alphavirus genus, in the Togaviridae family, are transmitted primarily by mosquitoes. The signs and symptoms associated with these viruses include fever and polyarthralgia, defined as joint pain and inflammation, as well as encephalitis. In the last decade, our understanding of the interactions between members of the alphavirus genus and the human host has increased due to the re-appearance of the chikungunya virus (CHIKV) in Asia and Europe, as well as its emergence in the Americas. Alphaviruses affect host immunity through cytokines and the interferon response. Understanding alphavirus interactions with both the innate immune system as well as the various cells in the adaptive immune systems is critical to developing effective therapeutics. In this review, we summarize the latest research on alphavirus-host cell interactions, underlying infection mechanisms, and possible treatments.
Collapse
Affiliation(s)
- Israel Guerrero-Arguero
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Bradford K Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Brett E Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
11
|
Roth KDR, Wenzel EV, Ruschig M, Steinke S, Langreder N, Heine PA, Schneider KT, Ballmann R, Fühner V, Kuhn P, Schirrmann T, Frenzel A, Dübel S, Schubert M, Moreira GMSG, Bertoglio F, Russo G, Hust M. Developing Recombinant Antibodies by Phage Display Against Infectious Diseases and Toxins for Diagnostics and Therapy. Front Cell Infect Microbiol 2021; 11:697876. [PMID: 34307196 PMCID: PMC8294040 DOI: 10.3389/fcimb.2021.697876] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022] Open
Abstract
Antibodies are essential molecules for diagnosis and treatment of diseases caused by pathogens and their toxins. Antibodies were integrated in our medical repertoire against infectious diseases more than hundred years ago by using animal sera to treat tetanus and diphtheria. In these days, most developed therapeutic antibodies target cancer or autoimmune diseases. The COVID-19 pandemic was a reminder about the importance of antibodies for therapy against infectious diseases. While monoclonal antibodies could be generated by hybridoma technology since the 70ies of the former century, nowadays antibody phage display, among other display technologies, is robustly established to discover new human monoclonal antibodies. Phage display is an in vitro technology which confers the potential for generating antibodies from universal libraries against any conceivable molecule of sufficient size and omits the limitations of the immune systems. If convalescent patients or immunized/infected animals are available, it is possible to construct immune phage display libraries to select in vivo affinity-matured antibodies. A further advantage is the availability of the DNA sequence encoding the phage displayed antibody fragment, which is packaged in the phage particles. Therefore, the selected antibody fragments can be rapidly further engineered in any needed antibody format according to the requirements of the final application. In this review, we present an overview of phage display derived recombinant antibodies against bacterial, viral and eukaryotic pathogens, as well as microbial toxins, intended for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Esther Veronika Wenzel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Maximilian Ruschig
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Stephan Steinke
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Nora Langreder
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Philip Alexander Heine
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai-Thomas Schneider
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Rico Ballmann
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Viola Fühner
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | | | | | - Stefan Dübel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Giulio Russo
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| |
Collapse
|
12
|
Bertoglio F, Meier D, Langreder N, Steinke S, Rand U, Simonelli L, Heine PA, Ballmann R, Schneider KT, Roth KDR, Ruschig M, Riese P, Eschke K, Kim Y, Schäckermann D, Pedotti M, Kuhn P, Zock-Emmenthal S, Wöhrle J, Kilb N, Herz T, Becker M, Grasshoff M, Wenzel EV, Russo G, Kröger A, Brunotte L, Ludwig S, Fühner V, Krämer SD, Dübel S, Varani L, Roth G, Čičin-Šain L, Schubert M, Hust M. SARS-CoV-2 neutralizing human recombinant antibodies selected from pre-pandemic healthy donors binding at RBD-ACE2 interface. Nat Commun 2021; 12:1577. [PMID: 33707427 PMCID: PMC7952403 DOI: 10.1038/s41467-021-21609-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022] Open
Abstract
COVID-19 is a severe acute respiratory disease caused by SARS-CoV-2, a new recently emerged sarbecovirus. This virus uses the human ACE2 enzyme as receptor for cell entry, recognizing it with the receptor binding domain (RBD) of the S1 subunit of the viral spike protein. We present the use of phage display to select anti-SARS-CoV-2 spike antibodies from the human naïve antibody gene libraries HAL9/10 and subsequent identification of 309 unique fully human antibodies against S1. 17 antibodies are binding to the RBD, showing inhibition of spike binding to cells expressing ACE2 as scFv-Fc and neutralize active SARS-CoV-2 virus infection of VeroE6 cells. The antibody STE73-2E9 is showing neutralization of active SARS-CoV-2 as IgG and is binding to the ACE2-RBD interface. Thus, universal libraries from healthy human donors offer the advantage that antibodies can be generated quickly and independent from the availability of material from recovering patients in a pandemic situation.
Collapse
Affiliation(s)
- Federico Bertoglio
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Doris Meier
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Nora Langreder
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Stephan Steinke
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Ulfert Rand
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Luca Simonelli
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | - Philip Alexander Heine
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Rico Ballmann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Kai-Thomas Schneider
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Kristian Daniel Ralph Roth
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Maximilian Ruschig
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Peggy Riese
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kathrin Eschke
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Yeonsu Kim
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dorina Schäckermann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Mattia Pedotti
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland
| | | | | | | | | | | | - Marlies Becker
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Martina Grasshoff
- Helmholtz Centre for Infection Research, Research Group Innate Immunity and Infection, Braunschweig, Germany
| | - Esther Veronika Wenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Giulio Russo
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Andrea Kröger
- Helmholtz Centre for Infection Research, Research Group Innate Immunity and Infection, Braunschweig, Germany
| | - Linda Brunotte
- Westfälische Wilhelms-Universität Münster, Institut für Virologie (IVM), Münster, Germany
| | - Stephan Ludwig
- Westfälische Wilhelms-Universität Münster, Institut für Virologie (IVM), Münster, Germany
| | - Viola Fühner
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Luca Varani
- Institute for Research in Biomedicine (IRB), Università della Svizzera italiana (USI), Bellinzona, Switzerland.
| | | | - Luka Čičin-Šain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- Centre for Individualised Infection Medicine (CIIM), a joint venture of Helmholtz Centre for Infection Research and Medical School Hannover, Braunschweig, Germany.
| | - Maren Schubert
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| |
Collapse
|
13
|
Owens A, Iannuzzelli JA, Gu Y, Fasan R. MOrPH-PhD: An Integrated Phage Display Platform for the Discovery of Functional Genetically Encoded Peptide Macrocycles. ACS CENTRAL SCIENCE 2020; 6:368-381. [PMID: 32232137 PMCID: PMC7099587 DOI: 10.1021/acscentsci.9b00927] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Indexed: 05/04/2023]
Abstract
Macrocyclic peptides represent attractive scaffolds for targeting protein-protein interactions, making methods for the diversification and functional selection of these molecules highly valuable for molecular discovery purposes. Here, we report the development of a novel strategy for the generation and high-throughput screening of combinatorial libraries of macrocyclic peptides constrained by a nonreducible thioether bridge. In this system, spontaneous, posttranslational peptide cyclization by means of a cysteine-reactive noncanonical amino acid was integrated with M13 bacteriophage display, enabling the creation of genetically encoded macrocyclic peptide libraries displayed on phage particles. This platform, named MOrPH-PhD, was successfully applied to produce and screen 105- to 108-member libraries of peptide macrocycles against three different protein targets, resulting in the discovery of a high-affinity binder for streptavidin (K D: 20 nM) and potent inhibitors of the therapeutically relevant proteins Kelch-like ECH-associated protein 1 (K D: 40 nM) and Sonic Hedgehog (K D: 550 nM). This work introduces and validates an efficient and general platform for the discovery and evolution of functional, conformationally constrained macrocyclic peptides useful for targeting proteins and protein-mediated interactions.
Collapse
|
14
|
Burke CW, Froude JW, Rossi F, White CE, Moyer CL, Ennis J, Pitt ML, Streatfield S, Jones RM, Musiychuk K, Kervinen J, Zeitlin L, Yusibov V, Glass PJ. Therapeutic monoclonal antibody treatment protects nonhuman primates from severe Venezuelan equine encephalitis virus disease after aerosol exposure. PLoS Pathog 2019; 15:e1008157. [PMID: 31790515 PMCID: PMC6907853 DOI: 10.1371/journal.ppat.1008157] [Citation(s) in RCA: 19] [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: 07/31/2019] [Revised: 12/12/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022] Open
Abstract
There are no FDA licensed vaccines or therapeutics for Venezuelan equine encephalitis virus (VEEV) which causes a debilitating acute febrile illness in humans that can progress to encephalitis. Previous studies demonstrated that murine and macaque monoclonal antibodies (mAbs) provide prophylactic and therapeutic efficacy against VEEV peripheral and aerosol challenge in mice. Additionally, humanized versions of two neutralizing mAbs specific for the E2 glycoprotein, 1A3B-7 and 1A4A-1, administered singly protected mice against aerosolized VEEV. However, no studies have demonstrated protection in nonhuman primate (NHP) models of VEEV infection. Here, we evaluated a chimeric antibody 1A3B-7 (c1A3B-7) containing mouse variable regions on a human IgG framework and a humanized antibody 1A4A-1 containing a serum half-life extension modification (Hu-1A4A-1-YTE) for their post-exposure efficacy in NHPs exposed to aerosolized VEEV. Approximately 24 hours after exposure, NHPs were administered a single bolus intravenous mAb. Control NHPs had typical biomarkers of VEEV infection including measurable viremia, fever, and lymphopenia. In contrast, c1A3B-7 treated NHPs had significant reductions in viremia and lymphopenia and on average approximately 50% reduction in fever. Although not statistically significant, Hu-1A4A-1-YTE administration did result in reductions in viremia and fever duration. Delay of treatment with c1A3B-7 to 48 hours post-exposure still provided NHPs protection from severe VEE disease through reductions in viremia and fever. These results demonstrate that post-exposure administration of c1A3B-7 protected macaques from development of severe VEE disease even when administered 48 hours following aerosol exposure and describe the first evaluations of VEEV-specific mAbs for post-exposure prophylactic use in NHPs. Viral mutations were identified in one NHP after c1A3B-7 treatment administered 24 hrs after virus exposure. This suggests that a cocktail-based therapy, or an alternative mAb against an epitope that cannot mutate without resulting in loss of viral fitness may be necessary for a highly effective therapeutic.
Collapse
Affiliation(s)
- Crystal W. Burke
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Jeffery W. Froude
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Franco Rossi
- Center of Aerobiological Sciences, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Charles E. White
- Biostatisics Branch, US Army Medical Research Institute of Infectious Disease, Fort Detrick Maryland, United States of America
| | - Crystal L. Moyer
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - Jane Ennis
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - M. Louise Pitt
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| | - Stephen Streatfield
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Konstantin Musiychuk
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jukka Kervinen
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, California, United States of America
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Pamela J. Glass
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, United States of America
| |
Collapse
|
15
|
Laustsen AH. How can monoclonal antibodies be harnessed against neglected tropical diseases and other infectious diseases? Expert Opin Drug Discov 2019; 14:1103-1112. [PMID: 31364421 DOI: 10.1080/17460441.2019.1646723] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Monoclonal antibody-based therapies now represent the single-largest class of molecules undergoing clinical investigation. Although a handful of different monoclonal antibodies have been clinically approved for bacterial and viral indications, including rabies, therapies based on monoclonal antibodies are yet to fully enter the fields of neglected tropical diseases and other infectious diseases. Areas covered: This review presents the current state-of-the-art in the development and use of monoclonal antibodies against neglected tropical diseases and other infectious diseases, including viral, bacterial, and parasitic infections, as well as envenomings by animal bites and stings. Additionally, a short section on mushroom poisonings is included. Key challenges for developing antibody-based therapeutics are discussed for each of these fields. Expert opinion: Neglected tropical diseases and other infectious diseases represent a golden opportunity for academics and technology developers for advancing our scientific capabilities within the understanding and design of antibody cross-reactivity, use of oligoclonal antibody mixtures for multi-target neutralization, novel immunization methodologies, targeting of evasive pathogens, and development of fundamentally novel therapeutic mechanisms of action. Furthermore, a huge humanitarian and societal impact is to gain by exploiting antibody technologies for the development of biotherapies against diseases, for which current treatment options are suboptimal or non-existent.
Collapse
Affiliation(s)
- Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark , Kongens Lyngby , Denmark
| |
Collapse
|
16
|
Phelps AL, O’Brien LM, Eastaugh LS, Davies C, Lever MS, Ennis J, Zeitlin L, Nunez A, Ulaeto DO. Aerosol infection of Balb/c mice with eastern equine encephalitis virus; susceptibility and lethality. Virol J 2019; 16:2. [PMID: 30611287 PMCID: PMC6321726 DOI: 10.1186/s12985-018-1103-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Eastern equine encephalitis virus is an alphavirus that naturally cycles between mosquitoes and birds or rodents in Eastern States of the US. Equine infection occurs by being bitten by cross-feeding mosquitoes, with a case fatality rate of up to 75% in humans during epizootic outbreaks. There are no licensed medical countermeasures, and with an anticipated increase in mortality when exposed by the aerosol route based on anecdotal human data and experimental animal data, it is important to understand the pathogenesis of this disease in pursuit of treatment options. This report details the clinical and pathological findings of mice infected with EEEV by the aerosol route, and use as a model for EEEV infection in humans. METHODS Mice were exposed by the aerosol route to a dose range of EEEV to establish the median lethal dose. A pathogenesis study followed whereby mice were exposed to a defined dose of virus and sacrificed at time-points thereafter for histopathological analysis and virology. RESULTS Clinical signs of disease appeared within 2 days post challenge, culminating in severe clinical signs within 24 h, neuro-invasion and dose dependent lethality. EEEV was first detected in the lung 1 day post challenge, and by day 3 peak viral titres were observed in the brain, spleen and blood, corresponding with severe meningoencephalitis, indicative of encephalitic disease. Lethality follows severe neurological signs, and may be linked to a threshold level of virus replication in the brain. Effective medical countermeasures for EEEV may necessitate early inoculation to inhibit infection of the brain in zoonotic incidents, and be able to traverse the blood-brain barrier to sufficiently interrupt replication in the brain in cases of aerosol infection. CONCLUSIONS There is little human data on the hazard posed by aerosol infection with encephalitic alphaviruses, and use of EEEV as a bioweapon may be by the aerosol route. A well characterized model of aerosol exposure that recapitulates some of the most severe human clinical features is necessary to evaluate the efficacy of putative medical countermeasures, and to increase our understanding about how this route of infection induces such rapid neuro-invasion and resulting disease.
Collapse
Affiliation(s)
- Amanda L. Phelps
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| | - Lyn M. O’Brien
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| | - Lin S. Eastaugh
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| | - Carwyn Davies
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| | - Mark S. Lever
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| | - Jane Ennis
- Mapp Biopharmaceutical Inc, 6160 Lusk Blvd. #C105, San Diego, CA 92121 USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical Inc, 6160 Lusk Blvd. #C105, San Diego, CA 92121 USA
| | - Alejandro Nunez
- Pathology Unit, Animal and Plant Health Agency – Weybridge, Woodham Lane, New Haw, Addlestone, Surrey KT15 3NB UK
| | - David O. Ulaeto
- CBR Division, Defence Science and Technology Laboratory (Dstl), Room 201, Building 7a,, Porton Down, Salisbury, Wiltshire SP4 0JQ UK
| |
Collapse
|
17
|
Post-Exposure Protection in Mice against Sudan Virus by a Two Antibody Cocktail. Viruses 2018; 10:v10060286. [PMID: 29861435 PMCID: PMC6024315 DOI: 10.3390/v10060286] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 05/18/2018] [Accepted: 05/24/2018] [Indexed: 01/01/2023] Open
Abstract
Sudan virus (SUDV) and Ebola viruses (EBOV) are both members of the Ebolavirus genus and have been sources of epidemics and outbreaks for several decades. We present here the generation and characterization of cross-reactive antibodies to both SUDV and EBOV, which were produced in a cell-free system and protective against SUDV in mice. A non-human primate, cynomolgus macaque, was immunized with viral-replicon particles expressing the glycoprotein of SUDV-Boniface (8A). Two separate antibody fragment phage display libraries were constructed after four immunogen injections. Both libraries were screened first against the SUDV and a second library was cross-selected against EBOV-Kikwit. Sequencing of 288 selected clones from the two distinct libraries identified 58 clones with distinct VH and VL sequences. Many of these clones were cross-reactive to EBOV and SUDV and able to neutralize SUDV. Three of these recombinant antibodies (X10B1, X10F3, and X10H2) were produced in the scFv-Fc format utilizing a cell-free production system. Mice that were challenged with SUDV-Boniface receiving 100µg of the X10B1/X10H2 scFv-Fc combination 6 and 48-h post-exposure demonstrated partial protection individually and complete protection as a combination. The data herein suggests these antibodies may be promising candidates for further therapeutic development.
Collapse
|
18
|
Burke CW, Froude JW, Miethe S, Hülseweh B, Hust M, Glass PJ. Human-Like Neutralizing Antibodies Protect Mice from Aerosol Exposure with Western Equine Encephalitis Virus. Viruses 2018; 10:v10040147. [PMID: 29587363 PMCID: PMC5923441 DOI: 10.3390/v10040147] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/14/2018] [Accepted: 03/22/2018] [Indexed: 02/07/2023] Open
Abstract
Western equine encephalitis virus (WEEV) causes symptoms in humans ranging from mild febrile illness to life-threatening encephalitis, and no human medical countermeasures are licensed. A previous study demonstrated that immune serum from vaccinated mice protected against lethal WEEV infection, suggesting the utility of antibodies for pre- and post-exposure treatment. Here, three neutralizing and one binding human-like monoclonal antibodies were evaluated against WEEV aerosol challenge. Dose-dependent protection was observed with two antibodies administered individually, ToR69-3A2 and ToR68-2C3. In vitro neutralization was not a critical factor for protection in this murine model, as ToR69-3A2 is a strong neutralizing antibody, and ToR68-2C3 is a non-neutralizing antibody. This result highlights the importance of both neutralizing and non-neutralizing antibodies in the protection of mice from WEEV lethality.
Collapse
MESH Headings
- Aerosols
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/immunology
- Disease Models, Animal
- Encephalitis Virus, Western Equine/immunology
- Encephalomyelitis, Equine/immunology
- Encephalomyelitis, Equine/mortality
- Encephalomyelitis, Equine/prevention & control
- Encephalomyelitis, Equine/virology
- Immunization
- Mice
- Morbidity
- Mortality
Collapse
Affiliation(s)
- Crystal W Burke
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Jeffrey W Froude
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr.7, 38106 Braunschweig, Germany.
| | - Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS)-ABC-Schutz, Humboldtstr. 1, 29623 Munster, Germany.
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr.7, 38106 Braunschweig, Germany.
- YUMAB GmbH, Science Campus Braunschweig Süd, Inhoffenstr.7, 38124 Braunschweig, Germany.
| | - Pamela J Glass
- United States Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, USA.
| |
Collapse
|
19
|
Rasetti-Escargueil C, Avril A, Miethe S, Mazuet C, Derman Y, Selby K, Thullier P, Pelat T, Urbain R, Fontayne A, Korkeala H, Sesardic D, Hust M, Popoff MR. The European AntibotABE Framework Program and Its Update: Development of Innovative Botulinum Antibodies. Toxins (Basel) 2017; 9:toxins9100309. [PMID: 28974033 PMCID: PMC5666356 DOI: 10.3390/toxins9100309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 02/06/2023] Open
Abstract
The goal of the AntiBotABE Program was the development of recombinant antibodies that neutralize botulinum neurotoxins (BoNT) A, B and E. These serotypes are lethal and responsible for most human botulinum cases. To improve therapeutic efficacy, the heavy and light chains (HC and LC) of the three BoNT serotypes were targeted to achieve a synergistic effect (oligoclonal antibodies). For antibody isolation, macaques were immunized with the recombinant and non-toxic BoNT/A, B or E, HC or LC, followed by the generation of immune phage-display libraries. Antibodies were selected from these libraries against the holotoxin and further analyzed in in vitro and ex vivo assays. For each library, the best ex vivo neutralizing antibody fragments were germline-humanized and expressed as immunoglobulin G (IgGs). The IgGs were tested in vivo, in a standardized model of protection, and challenged with toxins obtained from collections of Clostridium strains. Protective antibody combinations against BoNT/A and BoNT/B were evidenced and for BoNT/E, the anti-LC antibody alone was found highly protective. The combination of these five antibodies as an oligoclonal antibody cocktail can be clinically and regulatorily developed while their high “humanness” predicts a high tolerance in humans.
Collapse
Affiliation(s)
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de Biotechnologie des Anticorps et Des Toxins, Cedex 38702 La Tronche, France.
- Institut de Recherche Biomédicale des Armées (IRBA), Département des Maladies Infectieuses, Unité Biothérapies anti-Infectieuses et Immunité, 1 Place du Général Valérie André, BP73, 91220 Brétigny-sur-Orge, France.
| | - Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany and YUMAB GmbH, Rebenring 33, Braunschweig 38106, Germany.
| | - Christelle Mazuet
- Institut Pasteur, Unité des Bactéries Anaérobies et Toxines, 25 Avenue du Docteur Roux, 75015 Paris, France.
| | - Yagmur Derman
- Department of Food Hygiene and Environmental Health, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| | - Katja Selby
- Department of Food Hygiene and Environmental Health, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de Biotechnologie des Anticorps et Des Toxins, Cedex 38702 La Tronche, France.
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de Biotechnologie des Anticorps et Des Toxins, Cedex 38702 La Tronche, France.
- BIOTEM, Parc d'activité Bièvre Dauphine 885, Rue Alphonse Gourju, 38140 Apprieu, France.
| | - Remi Urbain
- LFB Biotechnologies, Therapeutic Innovation Department, 59, Rue de Trévise, BP 2006-59011 Lille Cedex, France.
- Ecdysis Pharma, Bioincubateur Eurasanté, 70 Rue du Dr Yersin, 59120 Loos, France.
| | - Alexandre Fontayne
- LFB Biotechnologies, Therapeutic Innovation Department, 59, Rue de Trévise, BP 2006-59011 Lille Cedex, France.
| | - Hannu Korkeala
- Department of Food Hygiene and Environmental Health, University of Helsinki, P.O. Box 66, FI-00014 Helsinki, Finland.
| | - Dorothea Sesardic
- National Institute for Biological Standards and Control (NIBSC), a Center of the Medicines and Healthcare Products Regulatory Agency, Division of Bacteriology, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK.
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany and YUMAB GmbH, Rebenring 33, Braunschweig 38106, Germany.
| | - Michel R Popoff
- Institut Pasteur, Unité des Bactéries Anaérobies et Toxines, 25 Avenue du Docteur Roux, 75015 Paris, France.
| |
Collapse
|
20
|
Galán A, Comor L, Horvatić A, Kuleš J, Guillemin N, Mrljak V, Bhide M. Library-based display technologies: where do we stand? MOLECULAR BIOSYSTEMS 2017; 12:2342-58. [PMID: 27306919 DOI: 10.1039/c6mb00219f] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Over the past two decades, library-based display technologies have been staggeringly optimized since their appearance in order to mimic the process of natural molecular evolution. Display technologies are essential for the isolation of specific high-affinity binding molecules (proteins, polypeptides, nucleic acids and others) for diagnostic and therapeutic applications in cancer, infectious diseases, autoimmune, neurodegenerative, inflammatory pathologies etc. Applications extend to other fields such as antibody and enzyme engineering, cell-free protein synthesis and the discovery of protein-protein interactions. Phage display technology is the most established of these methods but more recent fully in vitro alternatives, such as ribosome display, mRNA display, cis-activity based (CIS) display and covalent antibody display (CAD), as well as aptamer display and in vitro compartmentalization, offer advantages over phage in library size, speed and the display of unnatural amino acids and nucleotides. Altogether, they have produced several molecules currently approved or in diverse stages of clinical or preclinical testing and have provided researchers with tools to address some of the disadvantages of peptides and nucleotides such as their low affinity, low stability, high immunogenicity and difficulty to cross membranes. In this review we assess the fundamental technological features and point out some recent advances and applications of display technologies.
Collapse
Affiliation(s)
- Asier Galán
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia.
| | - Lubos Comor
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Anita Horvatić
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia.
| | - Josipa Kuleš
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia.
| | - Nicolas Guillemin
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia.
| | - Vladimir Mrljak
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia.
| | - Mangesh Bhide
- ERA Chair FP7, Internal diseases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia. and Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia and Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
21
|
Froude JW, Pelat T, Miethe S, Zak SE, Wec AZ, Chandran K, Brannan JM, Bakken RR, Hust M, Thullier P, Dye JM. Generation and characterization of protective antibodies to Marburg virus. MAbs 2017; 9:696-703. [PMID: 28287337 DOI: 10.1080/19420862.2017.1299848] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) have been a source of epidemics and outbreaks for several decades. We present here the generation and characterization of the first protective antibodies specific for wild-type MARV. Non-human primates (NHP), cynomolgus macaques, were immunized with viral-replicon particles expressing the glycoproteins (GP) of MARV (Ci67 isolate). An antibody fragment (single-chain variable fragment, scFv) phage display library was built after four immunogen injections, and screened against the GP1-649 of MARV. Sequencing of 192 selected clones identified 18 clones with distinct VH and VL sequences. Four of these recombinant antibodies (R4A1, R4B11, R4G2, and R3F6) were produced in the scFv-Fc format for in vivo studies. Mice that were challenged with wild-type Marburg virus (Ci67 isolate) receiving 100 µg of scFv-Fc on days -1, 1 and 3 demonstrated protective efficacies ranging from 75-100%. The amino-acid sequences of the scFv-Fcs are similar to those of their human germline counterparts, sharing an identity ranging between 68 and 100% to human germline immunoglobulin. These results demonstrate for the first time that recombinant antibodies offer protection against wild-type MARV, and suggest they may be promising candidates for further therapeutic development especially due to their human homology.
Collapse
Affiliation(s)
- Jeffrey W Froude
- a US Army Medical Research Institute for Infectious Disease (USAMRIID) , Fort Detrick , MD , USA
| | - Thibaut Pelat
- b Unite de Biotechnologie des Anticorps, Institut de Recherche Biomedicale des Armees [IRBA-CRSSA] , La Tronche , France.,c BIOTEM, Apprieu , France
| | - Sebastian Miethe
- d Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik Braunschweig , Germany.,e YUMAB GmbH, Rebenring , Braunschweig , Germany
| | - Samantha E Zak
- a US Army Medical Research Institute for Infectious Disease (USAMRIID) , Fort Detrick , MD , USA
| | - Anna Z Wec
- f Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Kartik Chandran
- f Department of Microbiology and Immunology , Albert Einstein College of Medicine , Bronx , NY , USA
| | - Jennifer Mary Brannan
- a US Army Medical Research Institute for Infectious Disease (USAMRIID) , Fort Detrick , MD , USA
| | - Russell R Bakken
- a US Army Medical Research Institute for Infectious Disease (USAMRIID) , Fort Detrick , MD , USA
| | - Michael Hust
- d Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik Braunschweig , Germany
| | - Philippe Thullier
- b Unite de Biotechnologie des Anticorps, Institut de Recherche Biomedicale des Armees [IRBA-CRSSA] , La Tronche , France
| | - John M Dye
- a US Army Medical Research Institute for Infectious Disease (USAMRIID) , Fort Detrick , MD , USA
| |
Collapse
|
22
|
Abstract
Over the last 3 decades, monoclonal antibodies have become the most important class of therapeutic biologicals on the market. Development of therapeutic antibodies was accelerated by recombinant DNA technologies, which allowed the humanization of murine monoclonal antibodies to make them more similar to those of the human body and suitable for a broad range of chronic diseases like cancer and autoimmune diseases. In the early 1990s in vitro antibody selection technologies were developed that enabled the discovery of “fully” human antibodies with potentially superior clinical efficacy and lowest immunogenicity. Antibody phage display is the first and most widely used of the in vitro selection technologies. It has proven to be a robust, versatile platform technology for the discovery of human antibodies and a powerful engineering tool to improve antibody properties. As of the beginning of 2016, 6 human antibodies discovered or further developed by phage display were approved for therapy. In 2002, adalimumab (Humira®) became the first phage display-derived antibody granted a marketing approval. Humira® was also the first approved human antibody, and it is currently the best-selling antibody drug on the market. Numerous phage display-derived antibodies are currently under advanced clinical investigation, and, despite the availability of other technologies such as human antibody-producing transgenic mice, phage display has not lost its importance for the discovery and engineering of therapeutic antibodies. Here, we provide a comprehensive overview about phage display-derived antibodies that are approved for therapy or in clinical development. A selection of these antibodies is described in more detail to demonstrate different aspects of the phage display technology and its development over the last 25 years.
Collapse
Affiliation(s)
- André Frenzel
- a YUMAB GmbH , Rebenring , Braunschweig.,b Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie , Braunschweig , Germany
| | | | - Michael Hust
- b Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie , Braunschweig , Germany
| |
Collapse
|
23
|
Kuhn P, Fühner V, Unkauf T, Moreira GMSG, Frenzel A, Miethe S, Hust M. Recombinant antibodies for diagnostics and therapy against pathogens and toxins generated by phage display. Proteomics Clin Appl 2016; 10:922-948. [PMID: 27198131 PMCID: PMC7168043 DOI: 10.1002/prca.201600002] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/30/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022]
Abstract
Antibodies are valuable molecules for the diagnostic and treatment of diseases caused by pathogens and toxins. Traditionally, these antibodies are generated by hybridoma technology. An alternative to hybridoma technology is the use of antibody phage display to generate recombinant antibodies. This in vitro technology circumvents the limitations of the immune system and allows—in theory—the generation of antibodies against all conceivable molecules. Phage display technology enables obtaining human antibodies from naïve antibody gene libraries when either patients are not available or immunization is not ethically feasible. On the other hand, if patients or immunized/infected animals are available, it is common to construct immune phage display libraries to select in vivo affinity‐matured antibodies. Because the phage packaged DNA sequence encoding the antibodies is directly available, the antibodies can be smoothly engineered according to the requirements of the final application. In this review, an overview of phage display derived recombinant antibodies against bacterial, viral, and eukaryotic pathogens as well as toxins for diagnostics and therapy is given.
Collapse
Affiliation(s)
- Philipp Kuhn
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Viola Fühner
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Tobias Unkauf
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - André Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| | - Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany.
| |
Collapse
|
24
|
Nian S, Wu T, Ye Y, Wang X, Xu W, Yuan Q. Development and identification of fully human scFv-Fcs against Staphylococcus aureus. BMC Immunol 2016; 17:8. [PMID: 27129873 PMCID: PMC4850644 DOI: 10.1186/s12865-016-0146-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 04/19/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Staphylococcus aureus, a gram-positive pathogen, causes many human infections. Methicillin-resistant S. aureus (MRSA) is the most common drug-resistance bacteria. Nearly all MRSA bacteria are resistant to several drugs. Specific antibodies are the main components of the host's humoral immunity, and play a significant role in the process of the host's resistance to bacterial infection. RESULTS A single-chain variable fragment (scFv) library was constructed using mRNA from the peripheral blood mononuclear cells of S. aureus infected volunteers. After the scFv library DNA was transformed into Escherichia coli TG1, ~1.7 × 10(7) independent clones with full-length scFv inserts. The scFv library was screened by phage display for three rounds using S. aureus as an antigen. The single clones were chosen at random and the scFvs were expressed for enzyme-linked immunosorbent assay (ELISA) assessment. Approximately 50 % of the clones were positive with good binding activity to S. aureus. To improve the stability of scFvs, scFv-fragment crystallizable regions (-Fcs) were constructed and expressed in E. coli DH5α. The expressed scFv-Fcs were purified and identified by western blot. These antibodies were further characterized and analyzed for bioactivity. The results showed that the expression level and folding of scFv-Fcs induced at 25 °C without isopropyl β-D-1-thiogalactopyranoside (IPTG) were higher than that induced at 32 °C with 1.0 mmol/L IPTG. scFv-Fcs had good bioactivity and could specifically bind with S. aureus. CONCLUSION scFv-Fcs against S. aureus were successfully constructed and are good candidates for the development of future adjunctive therapy for severe S. aureus infections.
Collapse
Affiliation(s)
- Siji Nian
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China
| | - Tong Wu
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China
| | - Yingchun Ye
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China
| | - Xu Wang
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China
| | - Wenfeng Xu
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China
| | - Qing Yuan
- The School of Basic Medical Sciences, Sichuan medical university, Room 218, Hanguang building, No 319, Zhongshan road, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
25
|
Böldicke T, Miethe S, Fühner V, Schirrmann T, Frenzel A, Hust M. Generation of Recombinant Antibodies Against Toxins and Viruses by Phage Display for Diagnostics and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:55-76. [PMID: 27236552 PMCID: PMC7121732 DOI: 10.1007/978-3-319-32805-8_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antibody phage display is an in vitro technology to generate recombinant antibodies. In particular for pathogens like viruses or toxins, antibody phage display is an alternative to hybridoma technology, since it circumvents the limitations of the immune system. Phage display allows the generation of human antibodies from naive antibody gene libraries when either immunized patients are not available or immunization is not ethically feasible. This technology also allows the construction of immune libraries to select in vivo affinity matured antibodies if immunized patients or animals are available.In this review, we describe the generation of human and human-like antibodies from naive antibody gene libraries and antibodies from immune antibody gene libraries. Furthermore, we give an overview about phage display derived recombinant antibodies against viruses and toxins for diagnostics and therapy.
Collapse
Affiliation(s)
- Thomas Böldicke
- grid.7490.aRecombinant protein exprsn/Intrabdy unit, Helmholtz-Centre for Infection Rese, Braunschweig, Germany
| | - Sebastian Miethe
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Viola Fühner
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Thomas Schirrmann
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany
| | - André Frenzel
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany
| | - Michael Hust
- Abteilung Biotechnologie, Institut für Biochemie, Biotechnologie und Bioinformatik, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany.
| |
Collapse
|
26
|
Miethe S, Rasetti-Escargueil C, Avril A, Liu Y, Chahboun S, Korkeala H, Mazuet C, Popoff MR, Pelat T, Thullier P, Sesardic D, Hust M. Development of Human-Like scFv-Fc Neutralizing Botulinum Neurotoxin E. PLoS One 2015; 10:e0139905. [PMID: 26440796 PMCID: PMC4595074 DOI: 10.1371/journal.pone.0139905] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/17/2015] [Indexed: 11/18/2022] Open
Abstract
Background Botulinum neurotoxins (BoNTs) are considered to be the most toxic substances known on earth and are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food-poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNTs have been classified as category A agent by the Centers of Disease Control and Prevention (CDC) and are listed among the six agents with the highest risk to be used as bioweapons. Neutralizing antibodies are required for the development of effective anti-botulism therapies to deal with the potential risk of exposure. Results In this study, a macaque (Macaca fascicularis) was immunized with recombinant light chain of BoNT/E3 and an immune phage display library was constructed. After a multi-step panning, several antibody fragments (scFv, single chain fragment variable) with nanomolar affinities were isolated, that inhibited the endopeptidase activity of pure BoNT/E3 in vitro by targeting its light chain. Furthermore, three scFv were confirmed to neutralize BoNT/E3 induced paralysis in an ex vivo mouse phrenic nerve-hemidiaphragm assay. The most effective neutralization (20LD50/mL, BoNT/E3) was observed with scFv ELC18, with a minimum neutralizing concentration at 0.3 nM. Furthermore, ELC18 was highly effective in vivo when administered as an scFv-Fc construct. Complete protection of 1LD50 BoNT/E3 was observed with 1.6 ng/dose in the mouse flaccid paralysis assay. Conclusion These scFv-Fcs antibodies are the first recombinant antibodies neutralizing BoNT/E by targeting its light chain. The human-like nature of the isolated antibodies is predicting a good tolerance for further clinical development.
Collapse
Affiliation(s)
- Sebastian Miethe
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Christine Rasetti-Escargueil
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Yvonne Liu
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Siham Chahboun
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Hannu Korkeala
- University of Helsinki, Faculty of Veterinary Medicine, Centre of Excellence in Microbial Food Safety Research, Department of Food Hygiene and Environmental Health, P.O. Box 66 (Agnes Sjöbergin katu 2), 00014 Helsinki University, Helsinki, Finland
| | - Christelle Mazuet
- Unité des Bactéries anaérobies et Toxines, Institut Pasteur, 25 avenue du Docteur Roux, 75015, Paris, France
| | - Michel-Robert Popoff
- Unité des Bactéries anaérobies et Toxines, Institut Pasteur, 25 avenue du Docteur Roux, 75015, Paris, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA), Département de Microbiologie, Unité de biotechnologie des anticorps et des toxines, 24 avenue des Maquis du Grésivaudan, B.P. 87, 38702 La Tronche Cedex, France
| | - Dorothea Sesardic
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Spielmannstr. 7, 38106 Braunschweig, Germany
- * E-mail:
| |
Collapse
|
27
|
Rasetti-Escargueil C, Avril A, Chahboun S, Tierney R, Bak N, Miethe S, Mazuet C, Popoff MR, Thullier P, Hust M, Pelat T, Sesardic D. Development of human-like scFv-Fc antibodies neutralizing Botulinum toxin serotype B. MAbs 2015; 7:1161-77. [PMID: 26381852 PMCID: PMC4966489 DOI: 10.1080/19420862.2015.1082016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are responsible for human botulism, a life-threatening disease characterized by flaccid muscle paralysis that occurs naturally by food poisoning or colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNTs have been classified as category A agents by the Centers for Disease Control and Prevention. To date, 7 subtypes of BoNT/B were identified showing that subtypes B1 (16 strains) and B2 (32 strains) constitute the vast majority of BoNT/B strains. Neutralizing antibodies are required for the development of anti-botulism drugs to deal with the potential risk. In this study, macaques (Macaca fascicularis) were immunized with recombinant light chain (LC) or heavy chain (HC) of BoNT/B2, followed by the construction of 2 hyper-immune phage display libraries. The best single-chain variable fragments (scFvs) isolated from each library were selected according to their affinities and cross reactivity with BoNT/B1 toxin subtype. These scFvs against LC and HC were further analyzed by assessing the inhibition of in vitro endopeptidase activity of BoNT/B1 and B2 and neutralization of BoNT/B1 and B2 toxin-induced paralysis in the mouse ex vivo phrenic nerve assay. The antibodies B2–7 (against HC) and BLC3 (against LC) were produced as scFv-Fc, and, when tested individually, neutralized BoNT/B1 and BoNT/B2 in a mouse ex vivo phrenic nerve assay. Whereas only scFv-Fc BLC3 alone protected mice against BoNT/B2-induced paralysis in vivo, when B2–7 and BLC3 were combined they exhibited potent synergistic protection. The present study provided an opportunity to assess the extent of antibody-mediated neutralization of BoNT/B1 and BoNT/B2 subtypes in ex vivo and in vitro assays, and to confirm the benefit of the synergistic effect of antibodies targeting the 2 distinct functional domains of the toxin in vivo. Notably, the framework regions of the most promising antibodies (B2–7 and BLC3) are close to the human germline sequences, which suggest that they may be well tolerated in potential clinical development.
Collapse
Affiliation(s)
- Christine Rasetti-Escargueil
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Arnaud Avril
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Siham Chahboun
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Rob Tierney
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Nicola Bak
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| | - Sebastian Miethe
- c Institut für Biochemie, Biotechnologie und Bioinformatik; Technische Universität Braunschweig ; Braunschweig , Germany
| | - Christelle Mazuet
- d Unité des Bactéries anaérobies et Toxines; Institut Pasteur ; Paris , France
| | - Michel R Popoff
- d Unité des Bactéries anaérobies et Toxines; Institut Pasteur ; Paris , France
| | - Philippe Thullier
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Michael Hust
- c Institut für Biochemie, Biotechnologie und Bioinformatik; Technische Universität Braunschweig ; Braunschweig , Germany
| | - Thibaut Pelat
- b Département des Maladies Infectieuses ; Unité Interaction Hôte-Pathogène; Institut de Recherche Biomédicale des Armées (IRBA) ; Brétigny-sur-Orge , France
| | - Dorothea Sesardic
- a Division of Bacteriology; National Institute for Biological Standards and Control (NIBSC), a centre of Medicines and Healthcare products Regulatory Agency ; Hertfordshire UK
| |
Collapse
|
28
|
Isolation of nanomolar scFvs of non-human primate origin, cross-neutralizing botulinum neurotoxins A1 and A2 by targeting their heavy chain. BMC Biotechnol 2015; 15:86. [PMID: 26382731 PMCID: PMC4574468 DOI: 10.1186/s12896-015-0206-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 09/11/2015] [Indexed: 12/14/2022] Open
Abstract
Background Botulism is a naturally occurring disease, mainly caused by the ingestion of food contaminated by the botulinum neurotoxins (BoNTs). Botulinum neurotoxins are the most lethal. They are classified among the six major biological warfare agents by the Centers for Disease Control. BoNTs act on the cholinergic motoneurons, where they cleave proteins implicated in acetylcholine vesicle exocytosis. This exocytosis inhibition induces a flaccid paralysis progressively affecting all the muscles and generally engendering a respiratory distress. BoNTs are also utilized in medicine, mainly for the treatment of neuromuscular disorders, preventing large scale vaccination. Botulism specific treatment requires injections of antitoxins, usually of equine origin and thus poorly tolerated. Therefore, development of human or human-like neutralizing antibodies is of a major interest, and it is the subject of the European framework project called “AntiBotABE”. Results In this study, starting from a macaque immunized with the recombinant heavy chain of BoNT/A1 (BoNT/A1-HC), an immune antibody phage-display library was generated and antibody fragments (single chain Fragment variable) with nanomolar affinity were isolated and further characterized. The neutralization capacities of these scFvs were analyzed in the mouse phrenic nerve-hemidiaphragm assay. Conclusions After a three-round panning, 24 antibody fragments with affinity better than 10 nM were isolated. Three of them neutralized BoNT/A1 efficiently and two cross-neutralized BoNT/A1 and BoNT/A2 subtypes in the mouse phrenic nerve-hemidiaphragm assay. These are the first monoclonal human-like antibodies cross-neutralizing both BoNT/A1 and BoNT/A2. The antibody A1HC38 was selected for further development, and could be clinically developed for the prophylaxis and treatment of botulism. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0206-0) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Droste P, Frenzel A, Steinwand M, Pelat T, Thullier P, Hust M, Lashuel H, Dübel S. Structural differences of amyloid-β fibrils revealed by antibodies from phage display. BMC Biotechnol 2015; 15:57. [PMID: 26084577 PMCID: PMC4472244 DOI: 10.1186/s12896-015-0146-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 04/20/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Beside neurofibrillary tangles, amyloid plaques are the major histological hallmarks of Alzheimer's disease (AD) being composed of aggregated fibrils of β-amyloid (Aβ). During the underlying fibrillogenic pathway, starting from a surplus of soluble Aβ and leading to mature fibrils, multiple conformations of this peptide appear, including oligomers of various shapes and sizes. To further investigate the fibrillization of β-amyloid and to have tools at hand to monitor the distribution of aggregates in the brain or even act as disease modulators, it is essential to develop highly sensitive antibodies that can discriminate between diverse aggregates of Aβ. RESULTS Here we report the generation and characterization of a variety of amyloid-β specific human and human-like antibodies. Distinct fractions of monomers and oligomers of various sizes were separated by size exclusion chromatography (SEC) from Aβ42 peptides. These antigens were used for the generation of two Aβ42 specific immune scFv phage display libraries from macaque (Macaca fascicularis). Screening of these libraries as well as two naïve human phage display libraries resulted in multiple unique binders specific for amyloid-β. Three of the obtained antibodies target the N-terminal part of Aβ42 although with varying epitopes, while another scFv binds to the α-helical central region of the peptide. The affinities of the antibodies to various Aβ42 aggregates as well as their ability to interfere with fibril formation and disaggregation of preformed fibrils were determined. Most significantly, one of the scFv is fibril-specific and can discriminate between two different fibril forms resulting from variations in the acidity of the milieu during fibrillogenesis. CONCLUSION We demonstrated that the approach of animal immunization and subsequent phage display based antibody selection is applicable to generate highly specific anti β-amyloid scFvs that are capable of accurately discriminating between minute conformational differences.
Collapse
Affiliation(s)
- Patrick Droste
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Spielmannstr.7, 38106, Braunschweig, Germany. .,Current address: Celerion Switzerland AG, Allmendstrasse 32, 8320, Fehraltorf, Switzerland.
| | - André Frenzel
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Spielmannstr.7, 38106, Braunschweig, Germany. .,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany.
| | - Miriam Steinwand
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Spielmannstr.7, 38106, Braunschweig, Germany. .,Current address: Delenex Therapeutics AG, Wagistrasse 27, 8952, Schlieren, Switzerland.
| | - Thibaut Pelat
- Institut de recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines, La Tronche Cedex, France. .,Current address: BIOTEM Parc d'Activités Bièvre Dauphine, 885, rue Alphonse Gourju, 38140, Apprieu, France.
| | - Philippe Thullier
- Institut de recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines, La Tronche Cedex, France.
| | - Michael Hust
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Spielmannstr.7, 38106, Braunschweig, Germany.
| | - Hilal Lashuel
- SV-BMI, Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Station 19, 1015, Lausanne, Switzerland.
| | - Stefan Dübel
- Technische Universität Braunschweig, Institute of Biochemistry, Biotechnology and Bioinformatics, Spielmannstr.7, 38106, Braunschweig, Germany.
| |
Collapse
|
30
|
Leishmania tarentolae: an alternative approach to the production of monoclonal antibodies to treat emerging viral infections. Infect Dis Poverty 2015; 4:8. [PMID: 26191408 PMCID: PMC4506428 DOI: 10.1186/2049-9957-4-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/13/2015] [Indexed: 11/10/2022] Open
Abstract
Background Monoclonal antibody therapy has an important role to play as a post-exposure prophylactic and therapeutic for the treatment of viral infections, including emerging infections. For example, several patients of the present Ebola virus outbreak in West Africa were treated with ZMapp, a cocktail of three monoclonal antibodies which are expressed in Nicotiana benthamiana. Discussion The majority of monoclonal antibodies in clinical use are expressed in mammalian cell lines which offer native folding and glycosylation of the expressed antibody. Monoclonal antibody expression in vegetal systems offers advantages over expression in mammalian cell lines, including improved potential for scale up and reduced costs. In this paper, I highlight the advantages of an upcoming protozoal system for the expression of recombinant antibody formats. Leishmania tarentolae offers a robust, economical expression of proteins with mammalian glycosylation patterns expressed in stable cell lines and grown in suspension culture. Several advantages of this system make it particularly suited for use in developing contexts. Summary Given the potential importance of monoclonal antibody therapy in the containment of emerging viral infections, novel and alternative strategies to improve production must be explored. Electronic supplementary material The online version of this article (doi:10.1186/2049-9957-4-8) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
Kügler J, Wilke S, Meier D, Tomszak F, Frenzel A, Schirrmann T, Dübel S, Garritsen H, Hock B, Toleikis L, Schütte M, Hust M. Generation and analysis of the improved human HAL9/10 antibody phage display libraries. BMC Biotechnol 2015; 15:10. [PMID: 25888378 PMCID: PMC4352240 DOI: 10.1186/s12896-015-0125-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/09/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Antibody phage display is a proven key technology that allows the generation of human antibodies for diagnostics and therapy. From naive antibody gene libraries - in theory - antibodies against any target can be selected. Here we describe the design, construction and characterization of an optimized antibody phage display library. RESULTS The naive antibody gene libraries HAL9 and HAL10, with a combined theoretical diversity of 1.5×10(10) independent clones, were constructed from 98 healthy donors using improved phage display vectors. In detail, most common phagemids employed for antibody phage display are using a combined His/Myc tag for detection and purification. We show that changing the tag order to Myc/His improved the production of soluble antibodies, but did not affect antibody phage display. For several published antibody libraries, the selected number of kappa scFvs were lower compared to lambda scFvs, probably due to a lower kappa scFv or Fab expression rate. Deletion of a phenylalanine at the end of the CL linker sequence in our new phagemid design increased scFv production rate and frequency of selected kappa antibodies significantly. The HAL libraries and 834 antibodies selected against 121 targets were analyzed regarding the used germline V-genes, used V-gene combinations and CDR-H3/-L3 length and composition. The amino acid diversity and distribution in the CDR-H3 of the initial library was retrieved in the CDR-H3 of selected antibodies showing that all CDR-H3 amino acids occurring in the human antibody repertoire can be functionally used and is not biased by E. coli expression or phage selection. Further, the data underline the importance of CDR length variations. CONCLUSION The highly diverse universal antibody gene libraries HAL9/10 were constructed using an optimized scFv phagemid vector design. Analysis of selected antibodies revealed that the complete amino acid diversity in the CDR-H3 was also found in selected scFvs showing the functionality of the naive CDR-H3 diversity.
Collapse
Affiliation(s)
- Jonas Kügler
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,mAb-factory GmbH, Gelsenkirchenstr. 5, 38108, Braunschweig, Germany.
| | - Sonja Wilke
- mAb-factory GmbH, Gelsenkirchenstr. 5, 38108, Braunschweig, Germany.
| | - Doris Meier
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - Florian Tomszak
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - André Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany.
| | - Thomas Schirrmann
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany. .,YUMAB GmbH, Rebenring 33, 38106, Braunschweig, Germany.
| | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| | - Henk Garritsen
- Klinikum Braunschweig g GmbH, Institut für Klinische Transfusionsmedizin, Celler Str. 38, 38114, Braunschweig, Germany. .,Department Vaccinology, Helmholtz-Zentrum für Infektionsforschung, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| | | | | | | | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Spielmannstr. 7, 38106, Braunschweig, Germany.
| |
Collapse
|
32
|
Exposure of epitope residues on the outer face of the chikungunya virus envelope trimer determines antibody neutralizing efficacy. J Virol 2014; 88:14364-79. [PMID: 25275138 DOI: 10.1128/jvi.01943-14] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Chikungunya virus (CHIKV) is a reemerging alphavirus that causes a debilitating arthritic disease and infects millions of people and for which no specific treatment is available. Like many alphaviruses, the structural targets on CHIKV that elicit a protective humoral immune response in humans are poorly defined. Here we used phage display against virus-like particles (VLPs) to isolate seven human monoclonal antibodies (MAbs) against the CHIKV envelope glycoproteins E2 and E1. One MAb, IM-CKV063, was highly neutralizing (50% inhibitory concentration, 7.4 ng/ml), demonstrated high-affinity binding (320 pM), and was capable of therapeutic and prophylactic protection in multiple animal models up to 24 h postexposure. Epitope mapping using a comprehensive shotgun mutagenesis library of 910 mutants with E2/E1 alanine mutations demonstrated that IM-CKV063 binds to an intersubunit conformational epitope on domain A, a functionally important region of E2. MAbs against the highly conserved fusion loop have not previously been reported but were also isolated in our studies. Fusion loop MAbs were broadly cross-reactive against diverse alphaviruses but were nonneutralizing. Fusion loop MAb reactivity was affected by temperature and reactivity conditions, suggesting that the fusion loop is hidden in infectious virions. Visualization of the binding sites of 15 different MAbs on the structure of E2/E1 revealed that all epitopes are located at the membrane-distal region of the E2/E1 spike. Interestingly, epitopes on the exposed topmost and outer surfaces of the E2/E1 trimer structure were neutralizing, whereas epitopes facing the interior of the trimer were not, providing a rationale for vaccine design and therapeutic MAb development using the intact CHIKV E2/E1 trimer. IMPORTANCE CHIKV is the most important alphavirus affecting humans, resulting in a chronic arthritic condition that can persist for months or years. In recent years, millions of people have been infected globally, and the spread of CHIKV to the Americas is now beginning, with over 100,000 cases occurring in the Caribbean within 6 months of its arrival. Our study reports on seven human MAbs against the CHIKV envelope, including a highly protective MAb and rarely isolated fusion loop MAbs. Epitope mapping of these MAbs demonstrates how some E2/E1 epitopes are exposed or hidden from the human immune system and suggests a structural mechanism by which these MAbs protect (or fail to protect) against CHIKV infection. Our results suggest that the membrane-distal end of CHIKV E2/E1 is the primary target for the humoral immune response to CHIKV, and antibodies targeting the exposed topmost and outer surfaces of the E2/E1 trimer determine the neutralizing efficacy of this response.
Collapse
|
33
|
Farajnia S, Ahmadzadeh V, Tanomand A, Veisi K, Khosroshahi SA, Rahbarnia L. Development trends for generation of single-chain antibody fragments. Immunopharmacol Immunotoxicol 2014; 36:297-308. [DOI: 10.3109/08923973.2014.945126] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
34
|
Hülseweh B, Rülker T, Pelat T, Langermann C, Frenzel A, Schirrmann T, Dübel S, Thullier P, Hust M. Human-like antibodies neutralizing Western equine encephalitis virus. MAbs 2014; 6:718-27. [PMID: 24518197 PMCID: PMC4011916 DOI: 10.4161/mabs.28170] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
This study describes the development of the first neutralizing antibodies against Western equine encephalitis virus (WEEV), a member of the genus Alphavirus. WEEV is transmitted by mosquitoes and can spread to the human central nervous system, causing symptoms ranging from mild febrile reactions to life-threatening encephalitis. WEEV has been classified as a biological warfare agent by the US Centers for Disease Control and Prevention. No anti-WEEV drugs are currently commercially available. Neutralizing antibodies are useful for the pre- and post-exposure treatment of WEEV infections. In this study, two immune antibody gene libraries were constructed from two macaques immunized with inactivated WEEV. Four antibodies were selected from these libraries and recloned as scFv-Fc, with a human Fc part. These antibodies bound WEEV specifically in ELISA with little or no cross-reaction with other alphaviruses. They were further analyzed by immunohistochemistry. All binders were suitable for the intracellular detection of WEEV particles. Neutralizing activity was determined in vitro. Three of the four antibodies were found to be neutralizing; about 1 ng/mL of the best antibody (ToR69–3A2) neutralized 50% of 5x104 TCID50/mL. Due to its human-like nature with a germinality index of 89% (VH) and 91% (VL), the ToR69–3A2 antibody is a promising candidate for future passive vaccine development.
Collapse
Affiliation(s)
- Birgit Hülseweh
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Torsten Rülker
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Claudia Langermann
- Wehrwissenschaftliches Institut für Schutztechnologien (WIS); ABC-Schutz; Munster, Germany
| | - Andrè Frenzel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Stefan Dübel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines; La Tronche, France
| | - Michael Hust
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie und Bioinformatik; Braunschweig, Germany
| |
Collapse
|
35
|
Miethe S, Rasetti-Escargueil C, Liu Y, Chahboun S, Pelat T, Avril A, Frenzel A, Schirrmann T, Thullier P, Sesardic D, Hust M. Development of neutralizing scFv-Fc against botulinum neurotoxin A light chain from a macaque immune library. MAbs 2014; 6:446-59. [PMID: 24492304 PMCID: PMC3984333 DOI: 10.4161/mabs.27773] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Botulinum toxins (BoNTs) are among the most toxic substances on earth, with serotype A toxin being the most toxic substance known. They are responsible for human botulism, a disease characterized by flaccid muscle paralysis that occurs naturally through food poisoning or the colonization of the gastrointestinal tract by BoNT-producing clostridia. BoNT has been classified as a category A agent by the Centers for Disease Control, and it is one of six agents with the highest potential risk of use as bioweapons. Human or human-like neutralizing antibodies are thus required for the development of anti-botulinum toxin drugs to deal with this possibility. In this study, Macaca fascicularis was hyperimmunized with a recombinant light chain of BoNT/A. An immune phage display library was constructed and, after multistep panning, several scFv with nanomolar affinities that inhibited the endopeptidase activity of BoNT/A1 in vitro as scFv-Fc, with a molar ratio (ab binding site:toxin) of up to 1:1, were isolated. The neutralization of BoNT/A-induced paralysis by the SEM120-IID5, SEM120-IIIC1 and SEM120-IIIC4 antibodies was demonstrated in mouse phrenic nerve-hemidiaphragm preparations with the holotoxin. The neutralization observed is the strongest ever measured in the phrenic nerve-hemidiaphragm assay for BoNT/A1 for a monoclonal antibody. Several scFv-Fc inhibiting the endopeptidase activity of botulinum neurotoxin A were isolated. For SEM120-IID5, SEM120-IIIC1, and SEM120-IIIC4, inhibitory effects in vitro and protection against the toxin ex vivo were observed. The human-like nature of these antibodies makes them promising lead candidates for further development of immunotherapeutics for this disease.
Collapse
Affiliation(s)
- Sebastian Miethe
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Christine Rasetti-Escargueil
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Yvonne Liu
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Siham Chahboun
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Thibaut Pelat
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Arnaud Avril
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - André Frenzel
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Thomas Schirrmann
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| | - Philippe Thullier
- Institut de Recherche Biomédicale des Armées (IRBA-CRSSA); Département de Microbiologie; Unité de biotechnologie des anticorps et des toxines ; La Tronche Cedex, France
| | - Dorothea Sesardic
- National Institute for Biological Standards and Control (NIBSC); Medicines and Healthcare Products Regulatory Agency; Division of Bacteriology; Potters Bar, UK
| | - Michael Hust
- Technische Universität Braunschweig; Institut für Biochemie, Biotechnologie, und Bioinformatik; Abteilung Biotechnologie; Braunschweig, Germany
| |
Collapse
|
36
|
Possible future monoclonal antibody (mAb)-based therapy against arbovirus infections. BIOMED RESEARCH INTERNATIONAL 2013; 2013:838491. [PMID: 24058915 PMCID: PMC3766601 DOI: 10.1155/2013/838491] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 07/05/2013] [Accepted: 07/11/2013] [Indexed: 11/17/2022]
Abstract
More than 150 arboviruses belonging to different families are known to infect humans, causing endemic infections as well as epidemic outbreaks. Effective vaccines to limit the occurrence of some of these infections have been licensed, while for the others several new immunogens are under development mostly for their improvements concerning safety and effectiveness profiles. On the other hand, specific and effective antiviral drugs are not yet available, posing an urgent medical need in particular for emergency cases. Neutralizing monoclonal antibodies (mAbs) have been demonstrated to be effective in the treatment of several infectious diseases as well as in preliminary in vitro and in vivo models of arbovirus-related infections. Given their specific antiviral activity as well-tolerated molecules with limited side effects, mAbs could represent a new therapeutic approach for the development of an effective treatment, as well as useful tools in the study of the host-virus interplay and in the development of more effective immunogens. However, before their use as candidate therapeutics, possible hurdles (e.g., Ab-dependent enhancement of infection, occurrence of viral escape variants) must be carefully evaluated. In this review are described the main arboviruses infecting humans and candidate mAbs to be possibly used in a future passive immunotherapy.
Collapse
|
37
|
Taylor KG, Paessler S. Pathogenesis of Venezuelan equine encephalitis. Vet Microbiol 2013; 167:145-50. [PMID: 23968890 DOI: 10.1016/j.vetmic.2013.07.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/03/2013] [Accepted: 07/10/2013] [Indexed: 10/26/2022]
Abstract
Equine encephalids have high mortality rates and represent a significant zoonotic public health threat. Of these the most pathogenic viruses to equids are the alphaviruses in the family Togaviridae. The focus of this review Venezualen equine encephalitis virus (VEEV) has caused the most widespread and recent epidemic outbreaks of disease. Circulation in naturally occuring rodent-mosquito cycles, results in viral spread to both human and equine populations. However, equines develop a high titer viremia and can transmit the virus back to mosquito populations. As such, the early recognition and control of viral infection in equine populations is strongly associated with prevention of epidemic spread of the virus and limiting of disease incidence in human populations. This review will address identification and pathogenesis of VEEV in equids vaccination and treatment options, and current research for drug and vaccine development.
Collapse
Affiliation(s)
- Katherine G Taylor
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77550, United States.
| | | |
Collapse
|
38
|
Nagata LP, Wong JP, Hu WG, Wu JQ. Vaccines and therapeutics for the encephalitic alphaviruses. Future Virol 2013. [DOI: 10.2217/fvl.13.42] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This article is a review of vaccines and therapeutics in development for the encephalitic alphaviruses, which includes eastern equine encephalitis virus, western equine encephalitis virus and Venezuelan equine encephalitis virus. The encephalitic alphaviruses are endemic within regions in North and South America. Hosts are normally exposed after being bitten by infectious mosquitoes, and infection can develop into encephalitis in equines and humans with severe rates of morbidity and mortality. These viruses are also potential biological threat agents, being highly infectious via an aerosol route of exposure. In humans, equine encephalitis virus and western equine encephalitis virus are neurotropic viruses targeting the CNS and causing encephalitis. Mortality rates are 50 and 10%, respectively, for these viruses. On the other hand, Venezuelan equine encephalitis virus produces a systemic influenza-like illness with pathogenesis in the lungs and lymphoid tissue in adults and older children. The incidence of encephalitis is less than 5% in younger children with a case–mortality rate of 1%. The host response to virus infectivity is briefly discussed, along with a number of promising therapeutic and prophylactic approaches. These approaches can be broadly classified as: virus-specific, including vaccines, antibody therapy and gene-silencing oligonucleotides; or broad-spectrum, including interferon and activation of the host‘s innate immunity.
Collapse
Affiliation(s)
- Les P Nagata
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Jonathan P Wong
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Wei-gang Hu
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| | - Josh Q Wu
- BioThreat Defence Section, Defence Research & Development Canada, PO Box 4000, Medicine Hat, AB T1A 8K6, Canada
| |
Collapse
|
39
|
Jäger V, Büssow K, Wagner A, Weber S, Hust M, Frenzel A, Schirrmann T. High level transient production of recombinant antibodies and antibody fusion proteins in HEK293 cells. BMC Biotechnol 2013; 13:52. [PMID: 23802841 PMCID: PMC3699382 DOI: 10.1186/1472-6750-13-52] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/24/2013] [Indexed: 12/02/2022] Open
Abstract
Background The demand of monospecific high affinity binding reagents, particularly monoclonal antibodies, has been steadily increasing over the last years. Enhanced throughput of antibody generation has been addressed by optimizing in vitro selection using phage display which moved the major bottleneck to the production and purification of recombinant antibodies in an end-user friendly format. Single chain (sc)Fv antibody fragments require additional tags for detection and are not as suitable as immunoglobulins (Ig)G in many immunoassays. In contrast, the bivalent scFv-Fc antibody format shares many properties with IgG and has a very high application compatibility. Results In this study transient expression of scFv-Fc antibodies in human embryonic kidney (HEK) 293 cells was optimized. Production levels of 10-20 mg/L scFv-Fc antibody were achieved in adherent HEK293T cells. Employment of HEK293-6E suspension cells expressing a truncated variant of the Epstein Barr virus (EBV) nuclear antigen (EBNA) 1 in combination with production under serum free conditions increased the volumetric yield up to 10-fold to more than 140 mg/L scFv-Fc antibody. After vector optimization and process optimization the yield of an scFv-Fc antibody and a cytotoxic antibody-RNase fusion protein further increased 3-4-fold to more than 450 mg/L. Finally, an entirely new mammalian expression vector was constructed for single step in frame cloning of scFv genes from antibody phage display libraries. Transient expression of more than 20 different scFv-Fc antibodies resulted in volumetric yields of up to 600 mg/L and 400 mg/L in average. Conclusion Transient production of recombinant scFv-Fc antibodies in HEK293-6E in combination with optimized vectors and fed batch shake flasks cultivation is efficient and robust, and integrates well into a high-throughput recombinant antibody generation pipeline.
Collapse
|
40
|
Production and characterization of a CD25-specific scFv-Fc antibody secreted from Pichia pastoris. Appl Microbiol Biotechnol 2012; 97:3855-63. [PMID: 23250227 DOI: 10.1007/s00253-012-4632-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/29/2012] [Accepted: 12/01/2012] [Indexed: 02/05/2023]
Abstract
Antibodies against CD25 would be novel tools for the diagnosis and treatment of adult T cell leukemia lymphoma (ATLL) and many other immune disorders. In our previous work, we successfully produced the single-chain fragment of a variable antibody against CD25, the Dmab(scFv) antibody, using Pichia pastoris. Here, we describe a novel form of an antibody against CD25, the Dmab(scFv)-Fc antibody, also produced by P. pastoris. To construct the Dmab(scFv)-Fc antibody, the Dmab(scFv) antibody was genetically fused to the Fc fragment of a human IgG1 antibody. A fusion gene encoding Dmab(scFv)-Fc antibody was cloned into the pPIC9K plasmid and expressed at high levels, 60-70 mg/l, by P. pastoris under optimized conditions. The Dmab(scFv)-Fc antibody was similar to the Dmab(scFv) antibody in its binding specificity but different in its molecular form and Fc-mediated effector functions. The Dmab(scFv)-Fc antibody and the Dmab(scFv) antibody both bound to CD25-positive MJ cells but not to CD25-negative K562 cells. The Dmab(scFv)-Fc antibody existed as a dimer whereas the Dmab(scFv) antibody was a monomer because it lacks the Fc fragment. The Dmab(scFv)-Fc antibody enhanced the antibody-dependent cellular cytotoxicity of CD25-positive cancer cells, whereas the Dmab(scFv) antibody was inactive in the antibody-dependent cellular cytotoxicity assays. In addition, compared to the Dmab(scFv) antibody, the Dmab(scFv)-Fc antibody showed stronger immunosuppressive activity in the Con A-stimulated lymphocyte proliferation system and in the mixed lymphocyte reaction system. These results demonstrate that the Dmab(scFv)-Fc antibody produced in P. pastoris is functional, and therefore it might be developed as a novel diagnostic and therapeutic tool for ATLL and other immune disorders.
Collapse
|