1
|
Vance DJ, Rudolph MJ, Davis SA, Mantis NJ. Structural Basis of Antibody-Mediated Inhibition of Ricin Toxin Attachment to Host Cells. Biochemistry 2023; 62:3181-3187. [PMID: 37903428 DOI: 10.1021/acs.biochem.3c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Monoclonal antibodies, JB4 and SylH3, neutralize ricin toxin (RT) by inhibiting the galactose-specific lectin activity of the B subunit of the toxin (RTB), which is required for cell attachment and entry. It is not immediately apparent how the antibodies accomplish this feat, considering that RTB consists of two globular domains (D1, D2) each divided into three homologous subdomains (α, β, γ) with the two functional galactosyl-specific carbohydrate recognition domains (CRDs) situated on opposite poles (subdomains 1α and 2γ). Here, we report the X-ray crystal structures of JB4 and SylH3 Fab fragments bound to RTB in the context of RT. The structures revealed that neither Fab obstructed nor induced detectable conformational alterations in subdomains 1α or 2γ. Rather, JB4 and SylH3 Fabs recognize nearly identical epitopes within an ancillary carbohydrate recognition pocket located in subdomain 1β. Despite limited amino acid sequence similarity between SylH3 and JB4 Fabs, each paratope inserts a Phe side chain from the heavy (H) chain complementarity determining region (CDR3) into the 1β CRD pocket, resulting in local aromatic stacking interactions that potentially mimic a ligand interaction. Reconciling the fact that stoichiometric amounts of SylH3 and JB4 are sufficient to disarm RTB's lectin activity without evidence of allostery, we propose that subdomain 1β functions as a "coreceptor" required to stabilize glycan interactions principally mediated by subdomains 1α and 2γ. Further investigation into subdomain 1β will yield fundamental insights into the large family of R-type lectins and open novel avenues for countermeasures aimed at preventing toxin uptake into vulnerable tissues and cells.
Collapse
Affiliation(s)
- David J Vance
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| | - Michael J Rudolph
- New York Structural Biology Center, New York, New York 10027, United States
| | - Simon A Davis
- New York Structural Biology Center, New York, New York 10027, United States
| | - Nicholas J Mantis
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, New York 12208, United States
| |
Collapse
|
2
|
Chiang CY, Lane DJ, Zou Y, Hoffman T, Pan J, Hampton J, Maginnis J, Nayak BP, D'Oro U, Valiante N, Miller AT, Cooke M, Wu T, Bavari S, Panchal RG. A Novel Toll-Like Receptor 2 Agonist Protects Mice in a Prophylactic Treatment Model Against Challenge With Bacillus anthracis. Front Microbiol 2022; 13:803041. [PMID: 35369443 PMCID: PMC8965344 DOI: 10.3389/fmicb.2022.803041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/02/2022] [Indexed: 11/23/2022] Open
Abstract
Current therapies for anthrax include the use of antibiotics (i.e., doxycycline, and ciprofloxacin), an anthrax vaccine (BioThrax) and Bacillus anthracis-specific, monoclonal antibody (mAb) (i.e., Raxibacumab and obiltoxaximab). In this study, we investigated the activity of immunomodulators, which potentiate inflammatory responses through innate immune receptors. The rationale for the use of innate immune receptor agonists as adjunctive immunomodulators for infectious diseases is based on the concept that augmentation of host defense should promote the antimicrobial mechanism of the host. Our aim was to explore the anti-B. anthracis effector function of Toll-like receptor (TLR) agonists using a mouse model. Amongst the six TLR ligands tested, Pam3CSK4 (TLR1/2 ligand) was the best at protecting mice from lethal challenge of B. anthracis. We then evaluated the activity of a novel TLR2 ligand, DA-98-WW07. DA-98-WW07 demonstrated enhanced protection in B. anthracis infected mice. The surviving mice that received DA-98-WW07 when re-challenged with B. anthracis 20 days post the first infection showed increased survival rate. Moreover, ciprofloxacin, when treated in adjunct with a suboptimal concentration of DA-98-WW07 demonstrated augmented activity in protecting mice from B. anthracis infection. Taken together, we report the prophylactic treatment potential of DA-98-WW07 for anthrax and the utility of immunomodulators in combination with an antibiotic to treat infections caused by the B. anthracis bacterium.
Collapse
Affiliation(s)
- Chih-Yuan Chiang
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Douglas J Lane
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Yefen Zou
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Tim Hoffman
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Jianfeng Pan
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Janice Hampton
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Jillian Maginnis
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Bishnu P Nayak
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Ugo D'Oro
- Novartis Vaccines and Diagnostics, Siena, Italy
| | | | - Andrew T Miller
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Michael Cooke
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Tom Wu
- Genomics Institute of the Novartis Research Foundation, San Diego, CA, United States
| | - Sina Bavari
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| | - Rekha G Panchal
- Division of Molecular Biology, United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, United States
| |
Collapse
|
3
|
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: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
4
|
Tahir S, Bourquard T, Musnier A, Jullian Y, Corde Y, Omahdi Z, Mathias L, Reiter E, Crépieux P, Bruneau G, Poupon A. Accurate determination of epitope for antibodies with unknown 3D structures. MAbs 2021; 13:1961349. [PMID: 34432559 PMCID: PMC8405158 DOI: 10.1080/19420862.2021.1961349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MAbTope is a docking-based method for the determination of epitopes. It has been used to successfully determine the epitopes of antibodies with known 3D structures. However, during the antibody discovery process, this structural information is rarely available. Although we already have evidence that homology models of antibodies could be used instead of their 3D structure, the choice of the template, the methodology for homology modeling and the resulting performance still have to be clarified. Here, we show that MAbTope has the same performance when working with homology models of the antibodies as compared to crystallographic structures. Moreover, we show that even low-quality models can be used. We applied MAbTope to determine the epitope of dupilumab, an anti- interleukin 4 receptor alpha subunit therapeutic antibody of unknown 3D structure, that we validated experimentally. Finally, we show how the MAbTope-determined epitopes for a series of antibodies targeting the same protein can be used to predict competitions, and demonstrate the accuracy with an experimentally validated example. 3D: three-dimensionalRMSD: root mean square deviationCDR: complementary-determining regionCPU: central processing unitsVH: heavy chain variable regionVL: light chain variable regionscFv: single-chain variable fragmentsVHH: single-chain antibody variable regionIL4Rα: Interleukin 4 receptor alpha chainSPR: surface plasmon resonancePDB: protein data bankHEK293: Human embryonic kidney 293 cellsEDTA: Ethylenediaminetetraacetic acidFBS: Fetal bovine serumANOVA: Analysis of varianceEGFR: Epidermal growth factor receptorPE: PhycoerythrinAPC: AllophycocyaninFSC: forward scatterSSC: side scatterWT: wild type Keywords: MAbTope, Epitope Mapping, Molecular docking, Antibody modeling, Antibody-antigen docking
Collapse
Affiliation(s)
- Shifa Tahir
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France
| | - Thomas Bourquard
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais
| | - Astrid Musnier
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais
| | - Yann Jullian
- MAbSilico SAS, 1 Impasse Du Palais.,CaSciModOT, UFR De Sciences Et Techniques, Université De Tours
| | | | | | | | - Eric Reiter
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | - Pascale Crépieux
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | | | - Anne Poupon
- PRC, INRAE, CNRS, Université De Tours, Nouzilly, France.,MAbSilico SAS, 1 Impasse Du Palais.,France Inria, Inria Saclay-Île-de-France, Palaiseau, France.,Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| |
Collapse
|
5
|
Tsai MH, Chuang CC, Chen CC, Yen HJ, Cheng KM, Chen XA, Shyu HF, Lee CY, Young JJ, Kau JH. Nanoparticles assembled from fucoidan and trimethylchitosan as anthrax vaccine adjuvant: In vitro and in vivo efficacy in comparison to CpG. Carbohydr Polym 2020; 236:116041. [PMID: 32172855 DOI: 10.1016/j.carbpol.2020.116041] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/15/2019] [Accepted: 02/19/2020] [Indexed: 12/13/2022]
Abstract
Fucoidan/trimethylchitosan nanoparticles (FUC-TMC-NPs) have the potential to improve the immunostimulating efficiency of anthrax vaccine adsorbed (AVA). FUC-TMC-NPs with positive (+) or negative (-) surface charges were prepared via polyelectrolyte complexation, both charged NP types permitted high viability and presented no cytotoxicity on L929, A549 and JAWS II dendritic cells. Flow cytometry measurements indicated lower (+)-FUC-TMC-NPs internalization levels than (-)-FUC-TMC-NPs, yet produced high levels of pro-inflammatory cytokines IFN-γ, IL12p40, and IL-4. Moreover, fluorescence microscope images proved that both charged NP could deliver drugs into the nucleus. In vivo studies on A/J mice showed that (+)-FUC-TMC-NPs carrying AVA triggered an efficient response with a higher IgG anti-PA antibody titer than AVA with CpG oligodeoxynucleotides, and yielded 100 % protection when challenged with the anthracis spores. Furthermore, PA-specific IgG1 and IgG2a analysis confirmed that (+)-FUC-TMC-NPs strongly stimulated humoral immunity. In conclusion, (+)-FUC-TMC-NP is promising anthrax vaccine adjuvant as an alternative to CpG.
Collapse
Affiliation(s)
- Meng-Hung Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC; Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Chuan-Chang Chuang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC; Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Cheng-Cheung Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Hui-Ju Yen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Kuang-Ming Cheng
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Xin-An Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Huey-Fen Shyu
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Chia-Ying Lee
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC
| | - Jenn-Jong Young
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC.
| | - Jyh-Hwa Kau
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 11490, Taiwan, ROC; Institute of Preventive Medicine, National Defense Medical Center, New Taipei City 23742, Taiwan, ROC.
| |
Collapse
|
6
|
Omersa N, Podobnik M, Anderluh G. Inhibition of Pore-Forming Proteins. Toxins (Basel) 2019; 11:E545. [PMID: 31546810 PMCID: PMC6784129 DOI: 10.3390/toxins11090545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
Perforation of cellular membranes by pore-forming proteins can affect cell physiology, tissue integrity, or immune response. Since many pore-forming proteins are toxins or highly potent virulence factors, they represent an attractive target for the development of molecules that neutralize their actions with high efficacy. There has been an assortment of inhibitors developed to specifically obstruct the activity of pore-forming proteins, in addition to vaccination and antibiotics that serve as a plausible treatment for the majority of diseases caused by bacterial infections. Here we review a wide range of potential inhibitors that can specifically and effectively block the activity of pore-forming proteins, from small molecules to more specific macromolecular systems, such as synthetic nanoparticles, antibodies, antibody mimetics, polyvalent inhibitors, and dominant negative mutants. We discuss their mechanism of inhibition, as well as advantages and disadvantages.
Collapse
Affiliation(s)
- Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.
| |
Collapse
|
7
|
Human monoclonal anti-protective antigen antibody for the low-dose post-exposure prophylaxis and treatment of Anthrax. BMC Infect Dis 2018; 18:640. [PMID: 30526504 PMCID: PMC6288905 DOI: 10.1186/s12879-018-3542-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 11/23/2018] [Indexed: 12/21/2022] Open
Abstract
Background Disease caused by Bacillus anthracis is often accompanied by high mortality primarily due to toxin-mediated injury. In the early disease course, anthrax toxins are secreted; thus, antibiotic use is limited to the early stage. In this regard, antibodies against the toxin component, protective antigen (PA), play an important role in protecting against anthrax. Therefore, we developed PA21, a fully human anti-PA immunoglobulin G monoclonal antibody. Methods Combining human Fab was screened from a phage library with human heavy constant regions. Enzyme-linked immune sorbent assay, Western blot analysis and immunoprecipitation test evaluated the binding ability of PA21. Moreover, the affinity and neutralizing activity of the antibody was detected in vitro while the protective effectiveness in 60 rats was also examined in vivo. Results The Fischer 344 rats challenged with the lethal toxin can be protected by PA21 at a concentration of 0.067 mg/kg. All six rats remained alive although PA21 was injected 24 h before the toxin challenge. PA21 did not influence the binding of PA to cell receptors and that of a lethal factor to PA. Conclusion The PA21 monoclonal antibody against PA can be used for emergency prophylaxis and anthrax treatment.
Collapse
|
8
|
Bioinspired detoxification of blood: The efficient removal of anthrax toxin protective antigen using an extracorporeal macroporous adsorbent device. Sci Rep 2018; 8:7518. [PMID: 29760471 PMCID: PMC5951949 DOI: 10.1038/s41598-018-25678-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/26/2018] [Indexed: 12/28/2022] Open
Abstract
Whilst various remedial human monoclonal antibodies have been developed to treat the potentially life-threatening systemic complications associated with anthrax infection, an optimal and universally effective administration route has yet to be established. In the later stages of infection when antibody administration by injection is more likely to fail one possible route to improve outcome is via the use of an antibody-bound, adsorbent haemoperfusion device. We report here the development of an adsorbent macroporous polymer column containing immobilised B. anthracis exotoxin-specific antibodies, PANG (a non-glycosylated, version of a plant-produced human monoclonal antibody) and Valortim (a fully human monoclonal N-linked glycosylated antibody), for removal of anthrax protective antigen (PA) from freshly frozen human plasma and human whole blood. In addition, we have demonstrated that continuous extracorporeal blood recirculation through a Valortim-bound haemoperfusion column significantly reduced the blood plasma concentration of anthrax PA over 2 hours using an in vivo PA rat infusion model. This work provides proof-of-concept evidence to support the development of such alternative detoxification platforms.
Collapse
|
9
|
Henning LN, Carpenter S, Stark GV, Serbina NV. Development of Protective Immunity in New Zealand White Rabbits Challenged with Bacillus anthracis Spores and Treated with Antibiotics and Obiltoxaximab, a Monoclonal Antibody against Protective Antigen. Antimicrob Agents Chemother 2018; 62:e01590-17. [PMID: 29133571 PMCID: PMC5786786 DOI: 10.1128/aac.01590-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/03/2017] [Indexed: 02/08/2023] Open
Abstract
The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.
Collapse
|
10
|
Nagy CF, Mondick J, Serbina N, Casey LS, Carpenter SE, French J, Guttendorf R. Animal-to-Human Dose Translation of Obiltoxaximab for Treatment of Inhalational Anthrax Under the US FDA Animal Rule. Clin Transl Sci 2017; 10:12-19. [PMID: 27925405 PMCID: PMC5245108 DOI: 10.1111/cts.12433] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/07/2016] [Indexed: 12/14/2022] Open
Abstract
Obiltoxaximab, a monoclonal antibody against protective antigen (PA), is approved for treatment of inhalational anthrax under the US Food and Drug Administration's (FDA) Animal Rule. The human dose was selected and justified by comparing observed obiltoxaximab exposures in healthy and infected New Zealand White rabbits and cynomolgus macaques to observed exposures in healthy humans, to simulated exposures in healthy and infected humans, and to serum PA levels in infected animals. In humans, at 16 mg/kg intravenous, obiltoxaximab AUC was >2 times that in animals, while maximum serum concentrations were comparable to those in animals and were maintained in excess of the concentration required for PA neutralization in infected animals for 2-3 weeks. Obiltoxaximab 16 mg/kg in humans provided exposure beyond that of 16 mg/kg in animals, ensuring a sufficient duration of PA neutralization to allow for adaptive immunity development. Our approach to dose translation may be applicable to other agents being developed under the Animal Rule.
Collapse
Affiliation(s)
- CF Nagy
- Department of Clinical OperationsElusys Therapeutics, IncPine BrookNew JerseyUSA
| | - J Mondick
- Metrum Research Group LLCTariffvilleConnecticutUSA
| | - N Serbina
- Department of Research and Nonclinical Development, Elusys Therapeutics, IncPine BrookNew JerseyUSA
| | - LS Casey
- Department of Research and Nonclinical Development, Elusys Therapeutics, IncPine BrookNew JerseyUSA
| | - SE Carpenter
- Department of Research and Nonclinical Development, Elusys Therapeutics, IncPine BrookNew JerseyUSA
| | - J French
- Metrum Research Group LLCTariffvilleConnecticutUSA
| | - R Guttendorf
- Aclairo Pharmaceutical Development Group IncViennaVirginiaUSA
| |
Collapse
|
11
|
Efficacy Projection of Obiltoxaximab for Treatment of Inhalational Anthrax across a Range of Disease Severity. Antimicrob Agents Chemother 2016; 60:5787-95. [PMID: 27431222 PMCID: PMC5038317 DOI: 10.1128/aac.00972-16] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/06/2016] [Indexed: 01/14/2023] Open
Abstract
Inhalational anthrax has high mortality even with antibiotic treatment, and antitoxins are now recommended as an adjunct to standard antimicrobial regimens. The efficacy of obiltoxaximab, a monoclonal antibody against anthrax protective antigen (PA), was examined in multiple studies conducted in two animal models of inhalational anthrax. A single intravenous bolus of 1 to 32 mg/kg of body weight obiltoxaximab or placebo was administered to New Zealand White rabbits (two studies) and cynomolgus macaques (4 studies) at disease onset (significant body temperature increase or detection of serum PA) following lethal challenge with aerosolized Bacillus anthracis spores. The primary endpoint was survival. The relationship between efficacy and disease severity, defined by pretreatment bacteremia and toxemia levels, was explored. In rabbits, single doses of 1 to 16 mg/kg obiltoxaximab led to 17 to 93% survival. In two studies, survival following 16 mg/kg obiltoxaximab was 93% and 62% compared to 0% and 0% for placebo (P = 0.0010 and P = 0.0013, respectively). Across four macaque studies, survival was 6.3% to 78.6% following 4 to 32 mg/kg obiltoxaximab. In two macaque studies, 16 mg/kg obiltoxaximab reduced toxemia and led to survival rates of 31%, 35%, and 47% versus 0%, 0%, and 6.3% with placebo (P = 0.0085, P = 0.0053, P = 0.0068). Pretreatment bacteremia and toxemia levels inversely correlated with survival. Overall, obiltoxaximab monotherapy neutralized PA and increased survival across the range of disease severity, indicating clinical benefit of toxin neutralization with obiltoxaximab in both early and late stages of inhalational anthrax.
Collapse
|
12
|
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: 1.9] [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
|
13
|
Passive Immunotherapy Protects against Enteric Invasion and Lethal Sepsis in a Murine Model of Gastrointestinal Anthrax. Toxins (Basel) 2015; 7:3960-76. [PMID: 26426050 PMCID: PMC4626714 DOI: 10.3390/toxins7103960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 01/09/2023] Open
Abstract
The principal portal for anthrax infection in natural animal outbreaks is the digestive tract. Enteric exposure to anthrax, which is difficult to detect or prevent in a timely manner, could be exploited as an act of terror through contamination of human or animal food. Our group has developed a novel animal model of gastrointestinal (GI) anthrax for evaluation of disease pathogenesis and experimental therapeutics, utilizing vegetative Bacillus anthracis (Sterne strain) administered to A/J mice (a complement-deficient strain) by oral gavage. We hypothesized that a humanized recombinant monoclonal antibody (mAb) * that neutralizes the protective antigen (PA) component of B. anthracis lethal toxin (LT) and edema toxin (ET) could be an effective treatment. Although the efficacy of this anti-anthrax PA mAb has been shown in animal models of inhalational anthrax, its activity in GI infection had not yet been ascertained. We hereby demonstrate that passive immunotherapy with anti-anthrax PA mAb, administered at the same time as gastrointestinal exposure to B. anthracis, prevents lethal sepsis in nearly all cases (>90%), while a delay of up to forty-eight hours in treatment still greatly reduces mortality following exposure (65%). Moreover, passive immunotherapy protects against enteric invasion, associated mucosal injury and subsequent dissemination by gastrointestinal B. anthracis, indicating that it acts to prevent the initial stages of infection. * Expired raxibacumab being cycled off the Strategic National Stockpile; biological activity confirmed by in vitro assay.
Collapse
|
14
|
Abstract
Anthrax is caused by the spore-forming, gram-positive bacterium Bacillus anthracis. The bacterium's major virulence factors are (a) the anthrax toxins and (b) an antiphagocytic polyglutamic capsule. These are encoded by two large plasmids, the former by pXO1 and the latter by pXO2. The expression of both is controlled by the bicarbonate-responsive transcriptional regulator, AtxA. The anthrax toxins are three polypeptides-protective antigen (PA), lethal factor (LF), and edema factor (EF)-that come together in binary combinations to form lethal toxin and edema toxin. PA binds to cellular receptors to translocate LF (a protease) and EF (an adenylate cyclase) into cells. The toxins alter cell signaling pathways in the host to interfere with innate immune responses in early stages of infection and to induce vascular collapse at late stages. This review focuses on the role of anthrax toxins in pathogenesis. Other virulence determinants, as well as vaccines and therapeutics, are briefly discussed.
Collapse
Affiliation(s)
- Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; , , , ,
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; , , , ,
| | - Catherine Vrentas
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; , , , ,
| | - Andrei P Pomerantsev
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; , , , ,
| | - Shihui Liu
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892; , , , ,
| |
Collapse
|
15
|
Xiong S, Tang Q, Liang X, Zhou T, Yang J, Liu P, Chen Y, Wang C, Feng Z, Zhu J. A Novel Chimeric Anti-PA Neutralizing Antibody for Postexposure Prophylaxis and Treatment of Anthrax. Sci Rep 2015; 5:11776. [PMID: 26134518 PMCID: PMC4488766 DOI: 10.1038/srep11776] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 05/28/2015] [Indexed: 12/11/2022] Open
Abstract
Anthrax is a highly lethal infectious disease caused by the bacterium Bacillus anthracis, and the associated shock is closely related to the lethal toxin (LeTx) produced by the bacterium. The central role played by the 63 kDa protective antigen (PA63) region of LeTx in the pathophysiology of anthrax makes it an excellent therapeutic target. In the present study, a human/murine chimeric IgG mAb, hmPA6, was developed by inserting murine antibody variable regions into human constant regions using antibody engineering technology. hmPA6 expressed in 293F cells could neutralize LeTx both in vitro and in vivo. At a dose of 0.3 mg/kg, it could protect all tested rats from a lethal dose of LeTx. Even administration of 0.6 mg/kg hmPA6 48 h before LeTx challenge protected all tested rats. The results indicate that hmPA6 is a potential candidate for clinical application in anthrax treatment.
Collapse
Affiliation(s)
- Siping Xiong
- 1] Department of Pathology, Nanjing Medical University, Nanjing 210029, China [2] Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Qi Tang
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Xudong Liang
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 China
| | - Tingting Zhou
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Jin Yang
- Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Peng Liu
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Ya Chen
- 1] Department of Pathology, Nanjing Medical University, Nanjing 210029, China [2] Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Zhenqing Feng
- 1] Department of Pathology, Nanjing Medical University, Nanjing 210029, China [2] Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| | - Jin Zhu
- 1] Huadong Medical Institute of Biotechniques, Nanjing 210002, China [2] Key Laboratory of Antibody Technique of Ministry of Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
16
|
Diamant E, Torgeman A, Ozeri E, Zichel R. Monoclonal Antibody Combinations that Present Synergistic Neutralizing Activity: A Platform for Next-Generation Anti-Toxin Drugs. Toxins (Basel) 2015; 7:1854-81. [PMID: 26035486 PMCID: PMC4488679 DOI: 10.3390/toxins7061854] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/07/2015] [Accepted: 05/19/2015] [Indexed: 12/25/2022] Open
Abstract
Monoclonal antibodies (MAbs) are among the fastest-growing therapeutics and are being developed for a broad range of indications, including the neutralization of toxins, bacteria and viruses. Nevertheless, MAbs potency is still relatively low when compared to conventional polyclonal Ab preparations. Moreover, the efficacy of an individual neutralizing MAb may significantly be hampered by the potential absence or modification of its target epitope in a mutant or subtype of the infectious agent. These limitations of individual neutralizing MAbs can be overcome by using oligoclonal combinations of several MAbs with different specificities to the target antigen. Studies conducted in our lab and by others show that such combined MAb preparation may present substantial synergy in its potency over the calculated additive potency of its individual MAb components. Moreover, oligoclonal preparation is expected to be better suited to compensating for reduced efficacy due to epitope variation. In this review, the synergistic neutralization properties of combined oligoclonal Ab preparations are described. The effect of Ab affinity, autologous Fc fraction, and targeting a critical number of epitopes, as well as the unexpected contribution of non-neutralizing clones to the synergistic neutralizing effect are presented and discussed.
Collapse
Affiliation(s)
- Eran Diamant
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Amram Torgeman
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Eyal Ozeri
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| | - Ran Zichel
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel.
| |
Collapse
|
17
|
Ohanjanian L, Remy KE, Li Y, Cui X, Eichacker PQ. An overview of investigational toxin-directed therapies for the adjunctive management of Bacillus anthracis infection and sepsis. Expert Opin Investig Drugs 2015; 24:851-65. [PMID: 25920540 DOI: 10.1517/13543784.2015.1041587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis with Bacillus anthracis infection has a very high mortality rate despite appropriate antibiotic and supportive therapies. Over the past 15 years, recent outbreaks in the US and in Europe, coupled with anthrax's bioterrorism weapon potential, have stimulated efforts to develop adjunctive therapies to improve clinical outcomes. Since lethal toxin and edema toxin (LT and ET) make central contributions to the pathogenesis of B. anthracis, these have been major targets in this effort. AREAS COVERED Here, the authors review different investigative biopharmaceuticals that have been recently identified for their therapeutic potential as inhibitors of LT or ET. Among these inhibitors are two antibody preparations that have been included in the Strategic National Stockpile (SNS) and several more that have reached Phase I testing. Presently, however, many of these candidate agents have only been studied in vitro and very few tested in bacteria-challenged models. EXPERT OPINION Although a large number of drugs have been identified as potential therapeutic inhibitors of LT and ET, in most cases their testing has been limited. The use of the two SNS antibody therapies during a large-scale exposure to B. anthracis will be difficult. Further testing and development of agents with oral bioavailability and relatively long shelf lives should be a focus for future research.
Collapse
Affiliation(s)
- Lernik Ohanjanian
- National Institutes of Health, Clinical Center, Critical Care Medicine Department , Building 10, Room 2C145, Bethesda, MD 20892 , USA +1 301 402 2914 ; +1 301 402 1213 ;
| | | | | | | | | |
Collapse
|
18
|
Vance DJ, Rong Y, Brey RN, Mantis NJ. Combination of two candidate subunit vaccine antigens elicits protective immunity to ricin and anthrax toxin in mice. Vaccine 2015; 33:417-21. [PMID: 25475957 PMCID: PMC4274239 DOI: 10.1016/j.vaccine.2014.11.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/30/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
In an effort to develop combination vaccines for biodefense, we evaluated a ricin subunit antigen, RiVax, given in conjunction with an anthrax protective antigen, DNI. The combination led to high endpoint titer antibody response, neutralizing antibodies, and protective immunity against ricin and anthrax lethal toxin. This is a natural combination vaccine, since both antigens are recombinant subunit proteins that would be given to the same target population.
Collapse
Affiliation(s)
- David J Vance
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Yinghui Rong
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | | | - Nicholas J Mantis
- Division of Infectious Disease, Wadsworth Center, New York State Department of Health, Albany, NY, USA; Department of Biomedical Sciences, University at Albany, Albany, NY, USA.
| |
Collapse
|
19
|
Nixon AE, Sexton DJ, Ladner RC. Drugs derived from phage display: from candidate identification to clinical practice. MAbs 2014; 6:73-85. [PMID: 24262785 DOI: 10.4161/mabs.27240] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Phage display, one of today’s fundamental drug discovery technologies, allows identification of a broad range of biological drugs, including peptides, antibodies and other proteins, with the ability to tailor critical characteristics such as potency, specificity and cross-species binding. Further, unlike in vivo technologies, generating phage display-derived antibodies is not restricted by immunological tolerance. Although more than 20 phage display-derived antibody and peptides are currently in late-stage clinical trials or approved, there is little literature addressing the specific challenges and successes in the clinical development of phage-derived drugs. This review uses case studies, from candidate identification through clinical development, to illustrate the utility of phage display as a drug discovery tool, and offers a perspective for future developments of phage display technology.
Collapse
|
20
|
Preparation and evaluation of human-murine chimeric antibody against protective antigen of Bacillus anthracis. Int J Mol Sci 2014; 15:18496-507. [PMID: 25318053 PMCID: PMC4227228 DOI: 10.3390/ijms151018496] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/25/2014] [Accepted: 09/30/2014] [Indexed: 12/24/2022] Open
Abstract
The aim of this research is to develop a human/murine chimeric Fab antibody which neutralizes the anthrax toxin, protective antigen (PA). The chimeric Fab was constructed using variable regions of murine anti-PA monoclonal antibody in combination with constant regions of human IgG. The chimeric PA6-Fab was expressed in E. coli. BL21 and evaluated by ELISA and co-immunoprecipitation- mass spectra. The potency of PA6-Fab to neutralize LeTx was examined in J774A.1 cell viability in vitro and in Fisher 344 rats in vivo. The PA6-Fab did not have domain similarity corresponding to the current anti PA mAbs, but specifically bound to anthrax PA at an affinity of 1.76 nM, and was able to neutralize LeTx in vitro and protected 56.9% cells at 20 μg/mL against anthrax LeTx. One hundred μg PA6-Fab could neutralize 300 μg LeTx in vivo. The PA6-Fab has potential as a therapeutic mAb for treatment of anthrax.
Collapse
|
21
|
Chow SK, Smith C, MacCarthy T, Pohl MA, Bergman A, Casadevall A. Disease-enhancing antibodies improve the efficacy of bacterial toxin-neutralizing antibodies. Cell Host Microbe 2014; 13:417-28. [PMID: 23601104 DOI: 10.1016/j.chom.2013.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/14/2013] [Accepted: 02/13/2013] [Indexed: 12/25/2022]
Abstract
During infection, humoral immunity produces a polyclonal response with various immunoglobulins recognizing different epitopes within the microbe or toxin. Despite this diverse response, the biological activity of an antibody (Ab) is usually assessed by the action of a monoclonal population. We demonstrate that a combination of monoclonal antibodies (mAbs) that are individually disease enhancing or neutralizing to Bacillus anthracis protective antigen (PA), a component of anthrax toxin, results in significantly augmented protection against the toxin. This boosted protection is Fc gamma receptor (FcγR) dependent and involves the formation of stoichiometrically defined mAb-PA complexes that requires immunoglobulin bivalence and simultaneous interaction between PA and the two mAbs. The formation of these mAb-PA complexes inhibits PA oligomerization, resulting in protection. These data suggest that functional assessments of single Abs may inaccurately predict how the same Abs will operate in polyclonal preparations and imply that potentially therapeutic mAbs may be overlooked in single Ab screens.
Collapse
Affiliation(s)
- Siu-Kei Chow
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
22
|
Levy H, Glinert I, Weiss S, Sittner A, Schlomovitz J, Altboum Z, Kobiler D. Toxin-independent virulence of Bacillus anthracis in rabbits. PLoS One 2014; 9:e84947. [PMID: 24416317 PMCID: PMC3885664 DOI: 10.1371/journal.pone.0084947] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 11/14/2013] [Indexed: 12/24/2022] Open
Abstract
The accepted paradigm states that anthrax is both an invasive and toxinogenic disease and that the toxins play a major role in pathogenicity. In the guinea pig (GP) model we have previously shown that deletion of all three toxin components results in a relatively moderate attenuation in virulence, indicating that B. anthracis possesses an additional toxin-independent virulence mechanism. To characterize this toxin-independent mechanism in anthrax disease, we developed a new rabbit model by intravenous injection (IV) of B. anthracis encapsulated vegetative cells, artificially creating bacteremia. Using this model we were able to demonstrate that also in rabbits, B. anthracis mutants lacking the toxins are capable of killing the host within 24 hours. This virulent trait depends on the activity of AtxA in the presence of pXO2, as, in the absence of the toxin genes, deletion of either component abolishes virulence. Furthermore, this IV virulence depends mainly on AtxA rather than the whole pXO1. A similar pattern was shown in the GP model using subcutaneous (SC) administration of spores of the mutant strains, demonstrating the generality of the phenomenon. The virulent strains showed higher bacteremia levels and more efficient tissue dissemination; however our interpretation is that tissue dissemination per se is not the main determinant of virulence whose exact nature requires further elucidation.
Collapse
Affiliation(s)
- Haim Levy
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
- * E-mail:
| | - Itai Glinert
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Assa Sittner
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Josef Schlomovitz
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Zeev Altboum
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - David Kobiler
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| |
Collapse
|
23
|
Mapping the epitopes of a neutralizing antibody fragment directed against the lethal factor of Bacillus anthracis and cross-reacting with the homologous edema factor. PLoS One 2013; 8:e65855. [PMID: 23741517 PMCID: PMC3669279 DOI: 10.1371/journal.pone.0065855] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 05/03/2013] [Indexed: 11/19/2022] Open
Abstract
The lethal toxin (LT) of Bacillus anthracis, composed of the protective antigen (PA) and the lethal factor (LF), plays an essential role in anthrax pathogenesis. PA also interacts with the edema factor (EF, 20% identity with LF) to form the edema toxin (ET), which has a lesser role in anthrax pathogenesis. The first recombinant antibody fragment directed against LF was scFv 2LF; it neutralizes LT by blocking the interaction between PA and LF. Here, we report that scFv 2LF cross-reacts with EF and cross-neutralizes ET, and we present an in silico method taking advantage of this cross-reactivity to map the epitope of scFv 2LF on both LF and EF. This method identified five epitope candidates on LF, constituted of a total of 32 residues, which were tested experimentally by mutating the residues to alanine. This combined approach precisely identified the epitope of scFv 2LF on LF as five residues (H229, R230, Q234, L235 and Y236), of which three were missed by the consensus epitope candidate identified by pre-existing in silico methods. The homolog of this epitope on EF (H253, R254, E258, L259 and Y260) was experimentally confirmed to constitute the epitope of scFv 2LF on EF. Other inhibitors, including synthetic molecules, could be used to target these epitopes for therapeutic purposes. The in silico method presented here may be of more general interest.
Collapse
|
24
|
Affiliation(s)
| | - Sergey M. Bezrukov
- Program in Physical Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, U.S.A
| |
Collapse
|
25
|
Mechaly A, Levy H, Epstein E, Rosenfeld R, Marcus H, Ben-Arie E, Shafferman A, Ordentlich A, Mazor O. A novel mechanism for antibody-based anthrax toxin neutralization: inhibition of prepore-to-pore conversion. J Biol Chem 2012; 287:32665-73. [PMID: 22869370 DOI: 10.1074/jbc.m112.400473] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Protective antigen (PA), a key component of anthrax toxin, mediates the entry of lethal factor (LF) or edema factor (EF) through a membranal pore into target cells. We have previously reported the isolation and chimerization of cAb29, an anti-PA monoclonal antibody that effectively neutralizes anthrax toxin in an unknown mechanism. The aim of this study was to elucidate the neutralizing mechanism of this antibody in vitro and to test its ability to confer post-exposure protection against anthrax in vivo. By systematic evaluation of the steps taking place during the PA-based intoxication process, we found that cAb29 did not interfere with the initial steps of intoxication, namely its ability to bind to the anthrax receptor, the consecutive proteolytic cleavage to PA(63), oligomerization, prepore formation, or LF binding. However, the binding of cAb29 to the prepore prevented its pH-triggered transition to the transmembranal pore, thus preventing the last step of intoxication, i.e. the translocation of LF/EF into the cell. Epitope mapping, using a phage display peptide library, revealed that cAb29 binds the 2α(1) loop in domain 2 of PA, a loop that undergoes major conformational changes during pore formation. In vivo, we found that 100% of anthrax-infected rabbits survived when treated with cAb29 12 h after exposure. In conclusion, these experiments demonstrate that cAb29 exerts its potent neutralizing activity in a unique manner by blocking the prepore-to-pore conversion process.
Collapse
Affiliation(s)
- Adva Mechaly
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 74100, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|