201
|
Wong AHY, Fukami Y, Sudo M, Kokubun N, Hamada S, Yuki N. Sialylated IgG-Fc: a novel biomarker of chronic inflammatory demyelinating polyneuropathy. J Neurol Neurosurg Psychiatry 2016; 87:275-9. [PMID: 25814494 DOI: 10.1136/jnnp-2014-309964] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/02/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Sialylation in Fc portion of IgG plays a crucial role in the pathogenesis of autoimmune diseases and the working mechanism of intravenous immunoglobulin (IVIG). We aim to test whether IgG-Fc sialylation is a biomarker of disease activity for chronic inflammatory demyelinating polyneuropathy (CIDP). METHODS By using specific lectins for sialylation, galactosylation and agalactosylation, lectin-enzyme assay and lectin blotting with pretreatment of IgG degradating enzyme of Streptococcus pyogenes were performed to compare the glycosylation levels of serum IgG-Fc (1) between patients of untreated CIDP (n=107) and normal control subjects (n=27), (2) among patients with untreated CIDP of different clinical severities and (3) before and after IVIG treatment of patients with CIDP (n=12). RESULTS Sialylation and galactosylation of IgG-Fc were significantly reduced in patients with CIDP than normal control subjects (p=0.003 and 0.033, respectively), whereas agalactosylation was increased in CIDP (p=0.21). Ratios of sialylated/agalactosylated IgG-Fc levels were significantly reduced in CIDP (p<0.001) and inversely related to disease severity (p=0.044). After IVIG treatment, levels of sialylated IgG-Fc significantly increased (p=0.003). CONCLUSIONS Sialylation of IgG-Fc is reduced in CIDP. Its level correlated with clinical severity and increased after IVIG treatment. Sialylated as well as ratio of sialylated/agalactosylated IgG-Fc could be new measures to monitor the disease severity and treatment status in CIDP.
Collapse
Affiliation(s)
- Anna Hiu Yi Wong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuki Fukami
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Makoto Sudo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Norito Kokubun
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Shinsuke Hamada
- Department of Neurology, Hokuyukai Neurological Hospital, Sapporo, Japan
| | - Nobuhiro Yuki
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
202
|
Antibody-based PET imaging of amyloid beta in mouse models of Alzheimer's disease. Nat Commun 2016; 7:10759. [PMID: 26892305 PMCID: PMC4762893 DOI: 10.1038/ncomms10759] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 01/19/2016] [Indexed: 12/18/2022] Open
Abstract
Owing to their specificity and high-affinity binding, monoclonal antibodies have potential as positron emission tomography (PET) radioligands and are currently used to image various targets in peripheral organs. However, in the central nervous system, antibody uptake is limited by the blood-brain barrier (BBB). Here we present a PET ligand to be used for diagnosis and evaluation of treatment effects in Alzheimer's disease. The amyloid β (Aβ) antibody mAb158 is radiolabelled and conjugated to a transferrin receptor antibody to enable receptor-mediated transcytosis across the BBB. PET imaging of two different mouse models with Aβ pathology clearly visualize Aβ in the brain. The PET signal increases with age and correlates closely with brain Aβ levels. Thus, we demonstrate that antibody-based PET ligands can be successfully used for brain imaging.
Collapse
|
203
|
Quast I, Keller CW, Weber P, Schneider C, von Gunten S, Lünemann JD. Protection from experimental autoimmune encephalomyelitis by polyclonal IgG requires adjuvant-induced inflammation. J Neuroinflammation 2016; 13:42. [PMID: 26893156 PMCID: PMC4758141 DOI: 10.1186/s12974-016-0506-x] [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: 11/23/2015] [Accepted: 02/08/2016] [Indexed: 11/11/2022] Open
Abstract
Background Intravenous immunoglobulin (IVIG) proved to be an efficient anti-inflammatory treatment for a growing number of neuroinflammatory diseases and protects against the development of experimental autoimmune encephalomyelitis (EAE), a widely used animal model for multiple sclerosis (MS). Methods The clinical efficacy of IVIG and IVIG-derived F(ab’)2 fragments, generated using the streptococcal cysteine proteinase Ide-S, was evaluated in EAE induced by active immunization and by adoptive transfer of myelin-specific T cells. Frequency, phenotype, and functional characteristics of T cell subsets and myeloid cells were determined by flow cytometry. Antibody binding to microbial antigen and cytokine production by innate immune cells was assessed by ELISA. Results We report that the protective effect of IVIG is lost in the adoptive transfer model of EAE and requires prophylactic administration during disease induction. IVIG-derived Fc fragments are not required for protection against EAE, since administration of F(ab’)2 fragments fully recapitulated the clinical efficacy of IVIG. F(ab’)2-treated mice showed a substantial decrease in splenic effector T cell expansion and cytokine production (GM-CSF, IFN-γ, IL-17A) 9 days after immunization. Inhibition of effector T cell responses was not associated with an increase in total numbers of Tregs but with decreased activation of innate myeloid cells such as neutrophils, monocytes, and dendritic cells. Therapeutically effective IVIG-derived F(ab’)2 fragments inhibited adjuvant-induced innate immune cell activation as determined by IL-12/23 p40 production and recognized mycobacterial antigens contained in Freund’s complete adjuvant which is required for induction of active EAE. Conclusions Our data indicate that F(ab’)2-mediated neutralization of adjuvant contributes to the therapeutic efficacy of anti-inflammatory IgG. These findings might partly explain the discrepancy of IVIG efficacy in EAE and MS.
Collapse
Affiliation(s)
- Isaak Quast
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | - Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | - Patrick Weber
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| | | | | | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
204
|
Spoerry C, Seele J, Valentin-Weigand P, Baums CG, von Pawel-Rammingen U. Identification and Characterization of IgdE, a Novel IgG-degrading Protease of Streptococcus suis with Unique Specificity for Porcine IgG. J Biol Chem 2016; 291:7915-25. [PMID: 26861873 DOI: 10.1074/jbc.m115.711440] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Indexed: 11/06/2022] Open
Abstract
Streptococcus suisis a major endemic pathogen of pigs causing meningitis, arthritis, and other diseases. ZoonoticS. suisinfections are emerging in humans causing similar pathologies as well as severe conditions such as toxic shock-like syndrome. Recently, we discovered an IdeS family protease ofS. suisthat exclusively cleaves porcine IgM and represents the first virulence factor described, linkingS. suisto pigs as their natural host. Here we report the identification and characterization of a novel, unrelated protease ofS. suisthat exclusively targets porcine IgG. This enzyme, designated IgdE forimmunoglobulinG-degradingenzyme ofS. suis, is a cysteine protease distinct from previous characterized streptococcal immunoglobulin degrading proteases of the IdeS family and mediates efficient cleavage of the hinge region of porcine IgG with a high degree of specificity. The findings that allS. suisstrains investigated possess the IgG proteolytic activity and that piglet serum samples contain specific antibodies against IgdE strongly indicate that the protease is expressedin vivoduring infection and represents a novel and putative important bacterial virulence/colonization determinant, and a thus potential therapeutic target.
Collapse
Affiliation(s)
- Christian Spoerry
- From the Department of Molecular Biology and Umeå Centre for Microbial Research, Umeå University, 90187 Umeå, Sweden
| | - Jana Seele
- the Institute for Microbiology, Center for Infection Medicine, University of Veterinary Medicine Hannover, 30173 Hannover, Germany, and
| | - Peter Valentin-Weigand
- the Institute for Microbiology, Center for Infection Medicine, University of Veterinary Medicine Hannover, 30173 Hannover, Germany, and
| | - Christoph G Baums
- the Institute for Microbiology, Center for Infection Medicine, University of Veterinary Medicine Hannover, 30173 Hannover, Germany, and the Institute for Bacteriology und Mycology, Centre for Infectious Diseases, College of Veterinary Medicine, University Leipzig, 04103 Leipzig, Germany
| | - Ulrich von Pawel-Rammingen
- From the Department of Molecular Biology and Umeå Centre for Microbial Research, Umeå University, 90187 Umeå, Sweden,
| |
Collapse
|
205
|
Resemann A, Jabs W, Wiechmann A, Wagner E, Colas O, Evers W, Belau E, Vorwerg L, Evans C, Beck A, Suckau D. Full validation of therapeutic antibody sequences by middle-up mass measurements and middle-down protein sequencing. MAbs 2016; 8:318-30. [PMID: 26760197 PMCID: PMC4966597 DOI: 10.1080/19420862.2015.1128607] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The regulatory bodies request full sequence data assessment both for innovator and biosimilar monoclonal antibodies (mAbs). Full sequence coverage is typically used to verify the integrity of the analytical data obtained following the combination of multiple LC-MS/MS datasets from orthogonal protease digests (so called “bottom-up” approaches). Top-down or middle-down mass spectrometric approaches have the potential to minimize artifacts, reduce overall analysis time and provide orthogonality to this traditional approach. In this work we report a new combined approach involving middle-up LC-QTOF and middle-down LC-MALDI in-source decay (ISD) mass spectrometry. This was applied to cetuximab, panitumumab and natalizumab, selected as representative US Food and Drug Administration- and European Medicines Agency-approved mAbs. The goal was to unambiguously confirm their reference sequences and examine the general applicability of this approach. Furthermore, a new measure for assessing the integrity and validity of results from middle-down approaches is introduced – the “Sequence Validation Percentage.” Full sequence data assessment of the 3 antibodies was achieved enabling all 3 sequences to be fully validated by a combination of middle-up molecular weight determination and middle-down protein sequencing. Three errors in the reference amino acid sequence of natalizumab, causing a cumulative mass shift of only −2 Da in the natalizumab Fd domain, were corrected as a result of this work.
Collapse
Affiliation(s)
- Anja Resemann
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | - Wolfgang Jabs
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | - Anja Wiechmann
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | - Elsa Wagner
- b Centre d'Immunologie Pierre , St Julien-en-Genevois, France
| | - Olivier Colas
- b Centre d'Immunologie Pierre , St Julien-en-Genevois, France
| | - Waltraud Evers
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | - Eckhard Belau
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | - Lars Vorwerg
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| | | | - Alain Beck
- b Centre d'Immunologie Pierre , St Julien-en-Genevois, France
| | - Detlev Suckau
- a Bruker Daltonics GmbH , Fahrenheitstr. 4, Bremen , Germany
| |
Collapse
|
206
|
Sjögren J, Olsson F, Beck A. Rapid and improved characterization of therapeutic antibodies and antibody related products using IdeS digestion and subunit analysis. Analyst 2016; 141:3114-25. [DOI: 10.1039/c6an00071a] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Antibody subunits LC, Fd and Fc/2, generated by IdeS digestion has been applied in analytical methodologies to characterize antibody quality attributes such as glycosylation, oxidation, deamidation, and identity.
Collapse
Affiliation(s)
| | | | - Alain Beck
- Centre d'Immunologie Pierre Fabre
- St Julien-en-Genevois
- France
| |
Collapse
|
207
|
Temel DB, Landsman P, Brader ML. Orthogonal Methods for Characterizing the Unfolding of Therapeutic Monoclonal Antibodies. Methods Enzymol 2016; 567:359-89. [DOI: 10.1016/bs.mie.2015.08.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
208
|
Könitzer JD, Sieron A, Wacker A, Enenkel B. Reformatting Rituximab into Human IgG2 and IgG4 Isotypes Dramatically Improves Apoptosis Induction In Vitro. PLoS One 2015; 10:e0145633. [PMID: 26713448 PMCID: PMC4694715 DOI: 10.1371/journal.pone.0145633] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/06/2015] [Indexed: 12/31/2022] Open
Abstract
The direct induction of cell death, or apoptosis, in target cells is one of the effector mechanisms for the anti CD20 antibody Rituximab. Here we provide evidence that Rituximab’s apoptotic ability is linked to the antibody IgG isotype. Reformatting Rituximab from the standard human IgG1 heavy chain into IgG2 or IgG4 boosted in vitro apoptosis induction in the Burkitt’s lymphoma B cell line Ramos five and four-fold respectively. The determinants for this behavior are located in the hinge region and CH1 domain of the heavy chain. By transplanting individual IgG2 or IgG4 specific amino acid residues onto otherwise IgG1 like backbones, thereby creating hybrid antibodies, the same enhancement of apoptosis induction could be achieved. The cysteines at position 131 of the CH1 domain and 219 in the hinge region, involved in IgG2 and IgG4 disulfide formation, were found to be of particular structural importance. Our data indicates that the hybrid antibodies possess a different CD20 binding mode than standard Rituximab, which appears to be key in enhancing apoptotic ability. The presented work opens up an interesting engineering route for enhancing the direct cytotoxic ability of therapeutic antibodies.
Collapse
Affiliation(s)
- Jennifer D. Könitzer
- Boehringer Ingelheim, Division Research Germany, Immune Modulation and Biotherapeutics Discovery, Biberach/Riß, Germany
- * E-mail:
| | - Annette Sieron
- Boehringer Ingelheim, Biopharma Operations Germany, Biberach/Riß, Germany
| | - Angelika Wacker
- Boehringer Ingelheim, Bioprocess and Pharmaceutical Development Germany, Biberach/Riß, Germany
| | - Barbara Enenkel
- Boehringer Ingelheim, Bioprocess and Pharmaceutical Development Germany, Biberach/Riß, Germany
| |
Collapse
|
209
|
Postnikov P, Krotov G, Mesonzhnik N, Efimova Y, Rodchenkov G. Fc-fragment removal allows the EPO-Fc fusion protein to be detected in blood samples by IEF-PAGE. Drug Test Anal 2015; 7:999-1008. [DOI: 10.1002/dta.1916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/02/2015] [Accepted: 10/06/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Pavel Postnikov
- Moscow Antidoping Centre; Russian Federation Moscow 105005 Elizavetinsky per., 10/1 Russia
| | - Grigory Krotov
- Moscow Antidoping Centre; Russian Federation Moscow 105005 Elizavetinsky per., 10/1 Russia
| | - Natalia Mesonzhnik
- Moscow Antidoping Centre; Russian Federation Moscow 105005 Elizavetinsky per., 10/1 Russia
| | - Yulia Efimova
- Moscow State university of fine chemical technologies; Russian Federation Moscow 119571 Vernadsky pr., 86 Russia
| | - Grigory Rodchenkov
- Moscow Antidoping Centre; Russian Federation Moscow 105005 Elizavetinsky per., 10/1 Russia
| |
Collapse
|
210
|
Järnum S, Bockermann R, Runström A, Winstedt L, Kjellman C. The Bacterial Enzyme IdeS Cleaves the IgG-Type of B Cell Receptor (BCR), Abolishes BCR-Mediated Cell Signaling, and Inhibits Memory B Cell Activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:5592-601. [PMID: 26553074 PMCID: PMC4671093 DOI: 10.4049/jimmunol.1501929] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/09/2015] [Indexed: 11/19/2022]
Abstract
Ag binding to the BCR is a critical step in B cell development and activation, initiating a cascade of signaling events ultimately leading to proliferation, differentiation, or cell death. A bacterial enzyme, IgG-degrading enzyme of Streptococcus pyogenes (IdeS), was shown to specifically cleave IgG molecules below the hinge region of soluble IgG and when IgG is bound to Ag, resulting in one F(ab')2 molecule and one homodimeric Fc fragment. Whether IdeS could also cleave the IgG molecule when it is present in the BCR attached to the B cell membrane in a complex with CD79a and CD79b is unknown. In this article, we present human in vitro and ex vivo data showing that IdeS cleaves the IgG present in the BCR complex and very efficiently blocks Ag binding to the BCR. As a consequence of IdeS cleaving the BCR, signaling cascades downstream of the BCR are blocked, and memory B cells are temporarily silenced, preventing them from responding to antigenic stimulation and their transition into Ab-producing cells.
Collapse
|
211
|
Persson H, Söderberg JJ, Vindebro R, Johansson BP, von Pawel-Rammingen U. Proteolytic processing of the streptococcal IgG endopeptidase IdeS modulates the functional properties of the enzyme and results in reduced immunorecognition. Mol Immunol 2015; 68:176-84. [DOI: 10.1016/j.molimm.2015.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 01/01/2023]
|
212
|
Barnett TC, Cole JN, Rivera-Hernandez T, Henningham A, Paton JC, Nizet V, Walker MJ. Streptococcal toxins: role in pathogenesis and disease. Cell Microbiol 2015; 17:1721-41. [PMID: 26433203 DOI: 10.1111/cmi.12531] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 08/13/2015] [Accepted: 09/02/2015] [Indexed: 12/15/2022]
Abstract
Group A Streptococcus (Streptococcus pyogenes), group B Streptococcus (Streptococcus agalactiae) and Streptococcus pneumoniae (pneumococcus) are host-adapted bacterial pathogens among the leading infectious causes of human morbidity and mortality. These microbes and related members of the genus Streptococcus produce an array of toxins that act against human cells or tissues, resulting in impaired immune responses and subversion of host physiological processes to benefit the invading microorganism. This toxin repertoire includes haemolysins, proteases, superantigens and other agents that ultimately enhance colonization and survival within the host and promote dissemination of the pathogen.
Collapse
Affiliation(s)
- Timothy C Barnett
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Jason N Cole
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Tania Rivera-Hernandez
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Anna Henningham
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia.,Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Victor Nizet
- Department of Pediatrics and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| | - Mark J Walker
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
213
|
Lood R, Waldetoft KW, Nordenfelt P. Localization-triggered bacterial pathogenesis. Future Microbiol 2015; 10:1659-68. [PMID: 26437846 DOI: 10.2217/fmb.15.89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Bacterial infections are becoming an increasing problem worldwide and there is a need for a deeper understanding of how bacteria turn pathogenic. Here, we suggest that one answer may be found by taking into account the localization of the bacteria, both at an anatomical level and at a microenvironment level. Both commensals and traditional pathogens alter their interaction with the human host depending on the local surroundings--turning either more or less virulent. These localization effects could derive from the characteristics of different anatomical sites but also from local differences within a microenvironment. In order to understand the adaptive functions of bacterial virulence factors, we need to study the bacteria in the environments where they have evolved.
Collapse
Affiliation(s)
- Rolf Lood
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| | | | - Pontus Nordenfelt
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, SE-221 84 Lund, Sweden
| |
Collapse
|
214
|
A rapid approach for characterization of thiol-conjugated antibody–drug conjugates and calculation of drug–antibody ratio by liquid chromatography mass spectrometry. Anal Biochem 2015; 485:34-42. [DOI: 10.1016/j.ab.2015.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/28/2015] [Accepted: 06/01/2015] [Indexed: 12/11/2022]
|
215
|
Takahashi R, Yuki N. Streptococcal IdeS: therapeutic potential for Guillain-Barré syndrome. Sci Rep 2015; 5:10809. [PMID: 26194472 PMCID: PMC4508529 DOI: 10.1038/srep10809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 04/28/2015] [Indexed: 01/13/2023] Open
Abstract
Plasma exchange and intravenous immunoglobulin are effective in treating Guillain–Barré syndrome (GBS) probably because the former removes IgG autoantibodies and complement and the latter inhibits complement activation subsequent to the autoantibody binding to peripheral nerve antigens. IgG degrading enzyme of Streptococcus pyogenes (IdeS) can cleave the pathogenic autoantibodies into F(ab’)2 and Fc. The purpose of this study is to show whether IdeS has novel therapeutic potential for GBS. Sera with anti-ganglioside IgG antibodies from 15 patients with GBS or Miller Fisher syndrome were used. We tested whether IdeS cleaved the anti-ganglioside IgG antibodies and inhibited deposition of activated complement component on ELISA plates. IdeS efficiently cleaved IgG and blocked complement activation mediated by anti-GM1, anti-GD1a and anti-GQ1b IgG antibodies. IdeS has therapeutic potential for GBS and related conditions.
Collapse
Affiliation(s)
- Ryo Takahashi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nobuhiro Yuki
- 1] Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore [2] Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
216
|
Winstedt L, Järnum S, Nordahl EA, Olsson A, Runström A, Bockermann R, Karlsson C, Malmström J, Palmgren GS, Malmqvist U, Björck L, Kjellman C. Complete Removal of Extracellular IgG Antibodies in a Randomized Dose-Escalation Phase I Study with the Bacterial Enzyme IdeS--A Novel Therapeutic Opportunity. PLoS One 2015; 10:e0132011. [PMID: 26177518 PMCID: PMC4503742 DOI: 10.1371/journal.pone.0132011] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 06/03/2015] [Indexed: 11/21/2022] Open
Abstract
IdeS is a streptococcal protease that cleaves IgG antibodies into F(ab’)2 and Fc fragments with a unique degree of specificity, thereby providing a novel treatment opportunity of IgG-driven autoimmune conditions and antibody mediated transplant rejection. Here we report the results from a first in man, double blinded and randomized study with single ascending doses of IdeS in healthy, male subjects. Twenty healthy subjects were given intravenous single ascending doses of IdeS. With impressive efficacy IdeS cleaved the entire plasma IgG-pool only minutes after dosing. IgG reached nadir 6-24 hours after dosing and then slowly recovered. The half-life of IdeS was 4.9 (±2.8) hours at 0.24 mg/kg with the main fraction eliminated during 24 hours. Already two hours after IdeS-dosing, the phagocytic capacity of IgG/IgG-fragments was reduced to background levels. Importantly, IdeS has the capacity to inactivate Fc-mediated effector function in vivo, was considered safe with no serious adverse events, and without dose limiting toxicity in this study. The complete, rapid, but temporary removal of IgG provides a new potent therapeutic opportunity in IgG-mediated pathogenic conditions.
Collapse
Affiliation(s)
- Lena Winstedt
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
| | - Sofia Järnum
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
| | | | - Andreas Olsson
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
| | - Anna Runström
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
| | - Robert Bockermann
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
| | - Christofer Karlsson
- Department of Clinical Sciences, Lund, Lund University, SE-221 84 Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences, Lund, Lund University, SE-221 84 Lund, Sweden
| | | | - Ulf Malmqvist
- Clinical Trials Unit, Skåne University Hospital, Lund, Sweden
| | - Lars Björck
- Department of Clinical Sciences, Lund, Lund University, SE-221 84 Lund, Sweden
- * E-mail: (CK); (LB)
| | - Christian Kjellman
- Hansa Medical AB, Scheelevägen 22, P.O. Box 785, SE-22007 Lund, Sweden
- * E-mail: (CK); (LB)
| |
Collapse
|
217
|
Sjögren J, Cosgrave EFJ, Allhorn M, Nordgren M, Björk S, Olsson F, Fredriksson S, Collin M. EndoS and EndoS2 hydrolyze Fc-glycans on therapeutic antibodies with different glycoform selectivity and can be used for rapid quantification of high-mannose glycans. Glycobiology 2015; 25:1053-63. [PMID: 26156869 PMCID: PMC4551147 DOI: 10.1093/glycob/cwv047] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 07/03/2015] [Indexed: 12/16/2022] Open
Abstract
Enzymes that affect glycoproteins of the human immune system, and thereby modulate defense responses, are abundant among bacterial pathogens. Two endoglycosidases from the human pathogen Streptococcus pyogenes, EndoS and EndoS2, have recently been shown to hydrolyze N-linked glycans of human immunoglobulin G. However, detailed characterization and comparison of the hydrolyzing activities have not been performed. In the present study, we set out to characterize the enzymes by comparing the activities of EndoS and EndoS2 on a selection of therapeutic monoclonal antibodies (mAbs), cetuximab, adalimumab, panitumumab and denosumab. By analyzing the glycans hydrolyzed by EndoS and EndoS2 from the antibodies using matrix-assisted laser desorption ionization time of flight, we found that both the enzymes cleaved complex glycans and that EndoS2 hydrolyzed hybrid and oligomannose structures to a greater extent compared with EndoS. A comparison of ultra-high-performance liquid chromatography (LC) profiles of the glycan pool of cetuximab hydrolyzed with EndoS and EndoS2 showed that EndoS2 hydrolyzed hybrid and oligomannose glycans, whereas these peaks were missing in the EndoS chromatogram. We utilized this difference in glycoform selectivity, in combination with the IdeS protease, and developed a LC separation method to quantify high mannose content in the Fc fragments of the selected mAbs. We conclude that EndoS and EndoS2 hydrolyze different glycoforms from the Fc-glycosylation site on therapeutic mAbs and that this can be used for rapid quantification of high mannose content.
Collapse
Affiliation(s)
- Jonathan Sjögren
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund 221 84, Sweden Genovis AB, Lund 200 07, Sweden
| | | | - Maria Allhorn
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund 221 84, Sweden
| | | | | | | | | | - Mattias Collin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund 221 84, Sweden
| |
Collapse
|
218
|
Bioanalytical approaches for characterizing catabolism of antibody–drug conjugates. Bioanalysis 2015; 7:1583-604. [DOI: 10.4155/bio.15.87] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The in vivo stability and catabolism of antibody–drug conjugates (ADCs) directly impact their PK, efficacy and safety, and metabolites of the cytotoxic or small molecule drug component of an ADC can further complicate these factors. This perspective highlights the importance of understanding ADC catabolism and the associated bioanalytical challenges. We evaluated different bioanalytical approaches to qualitatively and quantitatively characterize ADC catabolites. Here we review and discuss the rationale and experimental strategies used to design bioanalytical assays for characterization of ADC catabolism and supporting ADME studies during ADC clinical development. This review covers both large and small molecule approaches, and uses examples from Kadcyla® (T-DM1) and a THIOMAB™ antibody–drug conjugate to illustrate the process.
Collapse
|
219
|
Wehrenberg-Klee E, Redjal N, Leece A, Turker NS, Heidari P, Shah K, Mahmood U. PET imaging of glioblastoma multiforme EGFR expression for therapeutic decision guidance. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2015; 5:379-389. [PMID: 26269775 PMCID: PMC4529591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/06/2015] [Indexed: 06/04/2023]
Abstract
After initial therapy and total resection of glioblastoma multiforme (GBM), 80-90% of recurrences occur at the surgical margins. Insufficient sensitivity and specificity of current imaging techniques based on non-specific vascular imaging agents lead to delay in diagnosis of residual and/or recurrent disease. A tumor-specific imaging agent for GBM may improve detection of small residual disease in the post-operative period, and improve ability to distinguish tumor recurrence from its imaging mimics that can delay diagnosis. To this end, we developed an EGFR-targeted PET probe and assessed its ability to image EGFR WT (U87) and EGFRvIII (Gli36vIII) expressing GBMs in both murine intra-cranial xenografts and in a surgical-resection model. The developed imaging probe, (64)Cu-DOTAcetuximab-F(ab´)2, binds with a Kd of 11.2 nM to EGFR expressing GBM. (64)Cu-DOTA-cetuximab-F(ab´)2 specifically localized to intra-cranial tumor with a significant difference in SUVmean between tumor and contralateral brain for both Gli36vIII and U87 tumors (P<0.01 for both comparisons), with mean TBR of 22.5±0.7 for Gli36vIII tumors and 28.9±2.1 for U87 tumors (TBR±SEM). Tracer uptake by tumor was significantly inhibited by pre-injection with cetuximab (P<0.01 for both), with SUVmean reduced by 68% and 58% for Gli36vIII and U87 tumors, respectively. Surgical resection model PET-CT imaging demonstrates residual tumor and low nonspecific uptake in the resection site. We conclude that (64)Cu-DOTA-cetuximab-F(ab´)2 binds specifically to intracranial EGFR WT and EGFRvIII expressing GBM, demonstrates excellent TBR, and specifically images small residual tumor in a surgical model, suggesting future clinical utility in identifying true tumor recurrence.
Collapse
Affiliation(s)
- Eric Wehrenberg-Klee
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - Navid Redjal
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - Alicia Leece
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - N Selcan Turker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - Pedram Heidari
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - Khalid Shah
- Molecular Neurotherapy and Imaging Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| | - Umar Mahmood
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical SchoolBoston, MA
| |
Collapse
|
220
|
Brown EP, Normandin E, Osei-Owusu NY, Mahan AE, Chan YN, Lai JI, Vaccari M, Rao M, Franchini G, Alter G, Ackerman ME. Microscale purification of antigen-specific antibodies. J Immunol Methods 2015; 425:27-36. [PMID: 26078040 DOI: 10.1016/j.jim.2015.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/05/2015] [Accepted: 06/08/2015] [Indexed: 10/23/2022]
Abstract
Glycosylation of the Fc domain is an important driver of antibody effector function. While assessment of antibody glycoform compositions observed across total plasma IgG has identified differences associated with a variety of clinical conditions, in many cases it is the glycosylation state of only antibodies against a specific antigen or set of antigens that may be of interest, for example, in defining the potential effector function of antibodies produced during disease or after vaccination. Historically, glycoprofiling such antigen-specific antibodies in clinical samples has been challenging due to their low prevalence, the high sample requirement for most methods of glycan determination, and the lack of high-throughput purification methods. New methods of glycoprofiling with lower sample requirements and higher throughput have motivated the development of microscale and automatable methods for purification of antigen-specific antibodies from polyclonal sources such as clinical serum samples. In this work, we present a robot-compatible 96-well plate-based method for purification of antigen-specific antibodies, suitable for such population level glycosylation screening. We demonstrate the utility of this method across multiple antibody sources, using both purified plasma IgG and plasma, and across multiple different antigen types, with enrichment factors greater than 1000-fold observed. Using an on-column IdeS protease treatment, we further describe staged release of Fc and Fab domains, allowing for glycoprofiling of each domain.
Collapse
Affiliation(s)
- Eric P Brown
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Erica Normandin
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Nana Yaw Osei-Owusu
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH 03755, United States
| | - Alison E Mahan
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Ying N Chan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Jennifer I Lai
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States
| | - Monica Vaccari
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Mangala Rao
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | - Genoveffa Franchini
- Animal Models and Vaccine Section, National Cancer Institute, Bethesda, MD 20814, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA 02139, United States
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States.
| |
Collapse
|
221
|
Valenzuela NM, Trinh KR, Mulder A, Morrison SL, Reed EF. Monocyte recruitment by HLA IgG-activated endothelium: the relationship between IgG subclass and FcγRIIa polymorphisms. Am J Transplant 2015; 15:1502-18. [PMID: 25648976 PMCID: PMC4439339 DOI: 10.1111/ajt.13174] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/01/2014] [Accepted: 12/15/2014] [Indexed: 01/25/2023]
Abstract
It is currently unclear which donor specific HLA antibodies confer the highest risk of antibody-mediated rejection (AMR) and allograft loss. In this study, we hypothesized that two distinct features (HLA IgG subclass and Fcγ receptor [FcγR] polymorphisms) which vary from patient to patient, influence the process of monocyte trafficking to and macrophage accumulation in the allograft during AMR in an interrelated fashion. Here, we investigated the contribution of human IgG subclass and FcγR polymorphisms in monocyte recruitment in vitro by primary human aortic endothelium activated with chimeric anti-HLA I human IgG1 and IgG2. Both subclasses triggered monocyte adhesion to endothelial cells, via a two-step process. First, HLA I crosslinking by antibodies stimulated upregulation of P-selectin on endothelium irrespective of IgG subclass. P-selectin-induced monocyte adhesion was enhanced by secondary interactions of IgG with FcγRs, which was highly dependent upon subclass. IgG1 was more potent than IgG2 through differential engagement of FcγRs. Monocytes homozygous for FcγRIIa-H131 adhered more readily to HLA antibody-activated endothelium compared with FcγRIIa-R131 homozygous. Finally, direct modification of HLA I antibodies with immunomodulatory enzymes EndoS and IdeS dampened recruitment by eliminating antibody-FcγR binding, an approach that may have clinical utility in reducing AMR and other forms of antibody-induced inflammation.
Collapse
Affiliation(s)
- Nicole M. Valenzuela
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - K. Ryan Trinh
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA
| | - Arend Mulder
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Sherie L. Morrison
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| |
Collapse
|
222
|
Couchman EC, Browne HP, Dunn M, Lawley TD, Songer JG, Hall V, Petrovska L, Vidor C, Awad M, Lyras D, Fairweather NF. Clostridium sordellii genome analysis reveals plasmid localized toxin genes encoded within pathogenicity loci. BMC Genomics 2015; 16:392. [PMID: 25981746 PMCID: PMC4434542 DOI: 10.1186/s12864-015-1613-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 05/05/2015] [Indexed: 11/17/2022] Open
Abstract
Background Clostridium sordellii can cause severe infections in animals and humans, the latter associated with trauma, toxic shock and often-fatal gynaecological infections. Strains can produce two large clostridial cytotoxins (LCCs), TcsL and TcsH, related to those produced by Clostridium difficile, Clostridium novyi and Clostridium perfringens, but the genetic basis of toxin production remains uncharacterised. Results Phylogenetic analysis of the genome sequences of 44 strains isolated from human and animal infections in the UK, US and Australia placed the species into four clades. Although all strains originated from animal or clinical disease, only 5 strains contained LCC genes: 4 strains contain tcsL alone and one strain contains tcsL and tcsH. Four toxin-positive strains were found within one clade. Where present, tcsL and tcsH were localised in a pathogenicity locus, similar to but distinct from that present in C. difficile. In contrast to C. difficile, where the LCCs are chromosomally localised, the C. sordellii tcsL and tcsH genes are localised on plasmids. Our data suggest gain and loss of entire toxigenic plasmids in addition to horizontal transfer of the pathogenicity locus. A high quality, annotated sequence of ATCC9714 reveals many putative virulence factors including neuraminidase, phospholipase C and the cholesterol-dependent cytolysin sordellilysin that are highly conserved between all strains studied. Conclusions Genome analysis of C. sordellii reveals that the LCCs, the major virulence factors, are localised on plasmids. Many strains do not contain the LCC genes; it is probable that in several of these cases the plasmid has been lost upon laboratory subculture. Our data are consistent with LCCs being the primary virulence factors in the majority of infections, but LCC-negative strains may precipitate certain categories of infection. A high quality genome sequence reveals putative virulence factors whose role in virulence can be investigated. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1613-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edward C Couchman
- Department of Life Sciences, Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK.
| | | | - Matt Dunn
- Wellcome Trust Sanger Institute, Hinxton, UK.
| | | | - J Glenn Songer
- Department of Veterinary Science and Microbiology, University of Arizona, Tucson, USA.
| | - Val Hall
- Anaerobe Reference Laboratory, University Hospital of Wales, Cardiff, UK.
| | | | - Callum Vidor
- Department of Microbiology, Monash University, Clayton, VIC, 3800, Australia.
| | - Milena Awad
- Department of Microbiology, Monash University, Clayton, VIC, 3800, Australia.
| | - Dena Lyras
- Department of Microbiology, Monash University, Clayton, VIC, 3800, Australia.
| | - Neil F Fairweather
- Department of Life Sciences, Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
223
|
Seele J, Beineke A, Hillermann LM, Jaschok-Kentner B, von Pawel-Rammingen U, Valentin-Weigand P, Baums CG. The immunoglobulin M-degrading enzyme of Streptococcus suis, IdeSsuis, is involved in complement evasion. Vet Res 2015; 46:45. [PMID: 25928761 PMCID: PMC4404118 DOI: 10.1186/s13567-015-0171-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/05/2015] [Indexed: 12/20/2022] Open
Abstract
Streptococcus (S.) suis is one of the most important pathogens in pigs causing meningitis, arthritis, endocarditis and serositis. Furthermore, it is also an emerging zoonotic agent. In our previous work we identified a highly specific IgM protease in S. suis, designated Ide(Ssuis) . The objective of this study was to characterize the function of Ide(Ssuis) in the host-pathogen interaction. Edman-sequencing revealed that Ide(Ssuis) cleaves the heavy chain of the IgM molecule between constant domain 2 and 3. As the C1q binding motif is located in the C3 domain, we hypothesized that Ide(Ssuis) is involved in complement evasion. Complement-mediated hemolysis induced by porcine hyperimmune sera containing erythrocyte-specific IgM was abrogated by treatment of these sera with recombinant Ide(Ssuis) . Furthermore, expression of Ide(Ssuis) reduced IgM-triggered complement deposition on the bacterial surface. An infection experiment of prime-vaccinated growing piglets suggested attenuation in the virulence of the mutant 10Δide(Ssuis). Bactericidal assays confirmed a positive effect of Ide(Ssuis) expression on bacterial survival in porcine blood in the presence of high titers of specific IgM. In conclusion, this study demonstrates that Ide(Ssuis) is a novel complement evasion factor, which is important for bacterial survival in porcine blood during the early adaptive (IgM-dominated) immune response.
Collapse
Affiliation(s)
- Jana Seele
- Institute for Microbiology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30173, Hannover, Germany.
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, 30559, Hannover, Germany.
| | - Lena-Maria Hillermann
- Institute for Microbiology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30173, Hannover, Germany.
| | - Beate Jaschok-Kentner
- Department of Structure and Function of Proteins, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.
| | - Ulrich von Pawel-Rammingen
- Department of Molecular Biology and Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden.
| | - Peter Valentin-Weigand
- Institute for Microbiology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30173, Hannover, Germany.
| | - Christoph Georg Baums
- Institute for Bacteriology and Mycology, Centre of Infectious Diseases, College of Veterinary Medicine, University Leipzig, An den Tierkliniken 29, 04103, Leipzig, Germany.
| |
Collapse
|
224
|
Seele J, Hillermann LM, Beineke A, Seitz M, von Pawel-Rammingen U, Valentin-Weigand P, Baums CG. The immunoglobulin M-degrading enzyme of Streptococcus suis, IdeSsuis, is a highly protective antigen against serotype 2. Vaccine 2015; 33:2207-2212. [PMID: 25825330 DOI: 10.1016/j.vaccine.2015.03.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/03/2015] [Accepted: 03/17/2015] [Indexed: 11/26/2022]
Abstract
Streptococcus suis (S. suis) is a major porcine pathogen causing meningitis, arthritis and several other pathologies. Recently, we identified a highly specific immunoglobulin M degrading enzyme of S. suis, designated IdeSsuis, which is expressed by various serotypes. The objective of this work was to access the immunogenicity and protective efficacy of a recombinant vaccine including IdeSsuis. Vaccination with rIdeSsuis elicited antibodies efficiently neutralizing the IgM protease activity. Importantly, 18 piglets vaccinated with rIdeSsuis alone or in combination with bacterin priming were completely protected against mortality and severe morbidity after S. suis serotype 2 challenge. In contrast, 12 of the 17 piglets either treated with the placebo or primed with the bacterin only, succumbed to S. suis disease. Immunity against IdeSsuis was associated with increased killing of S. suis wt in porcine blood ex vivo leading to a tenfold difference in the bacterial survival factor in blood of placebo-treated and rIdeSsuis-vaccinated piglets. In conclusion, the results of this study indicate that rIdeSsuis is a highly protective antigen in pigs.
Collapse
Affiliation(s)
- Jana Seele
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Lena-Maria Hillermann
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maren Seitz
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Peter Valentin-Weigand
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Christoph G Baums
- Institute for Microbiology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Hannover, Germany; Institute for Bacteriology and Mycology, Centre of Infectious Diseases, College of Veterinary Medicine, University Leipzig, Leipzig, Germany.
| |
Collapse
|
225
|
Lamb LEM, Sriskandan S, Tan LKK. Bromine, bear-claw scratch fasciotomies, and the Eagle effect: management of group A streptococcal necrotising fasciitis and its association with trauma. THE LANCET. INFECTIOUS DISEASES 2015; 15:109-21. [PMID: 25541175 DOI: 10.1016/s1473-3099(14)70922-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Necrotising fasciitis is a rare, but potentially fatal, soft-tissue infection. Historical depictions of the disease have been described since classical times and were mainly recorded in wartime reports of battle injuries. Although several different species of bacteria can cause necrotising fasciitis, perhaps the most widely known is group A streptococcus (GAS). Infection control, early surgical debridement, and antibiotic therapy are now the central tenets of the clinical management of necrotising fasciitis; these treatment approaches all originate from those used in wars in the past 150 years. We review reports from the 19th century, early 20th century, and mid-20th century onwards to show how the management of necrotising fasciitis has progressed in parallel with prevailing scientific thought and medical practice. Historically, necrotising fasciitis has often, but not exclusively, been associated with penetrating trauma. However, along with a worldwide increase in invasive GAS disease, recent reports have cited cases of necrotising fasciitis following non-combat-related injuries or in the absence of antecedent events. We also investigate the specific association between GAS necrotising fasciitis and trauma. In the 21st century, molecular biology has improved our understanding of GAS pathogenesis, but has not yet affected attributable mortality.
Collapse
Affiliation(s)
- Lucy E M Lamb
- Department of Medicine, Imperial College London, Hammersmith Campus, Hammersmith Hospital, London, UK
| | - Shiranee Sriskandan
- Department of Medicine, Imperial College London, Hammersmith Campus, Hammersmith Hospital, London, UK
| | - Lionel K K Tan
- Department of Medicine, Imperial College London, Hammersmith Campus, Hammersmith Hospital, London, UK.
| |
Collapse
|
226
|
An Y, Zhang Y, Mueller HM, Shameem M, Chen X. A new tool for monoclonal antibody analysis: application of IdeS proteolysis in IgG domain-specific characterization. MAbs 2015; 6:879-93. [PMID: 24927271 DOI: 10.4161/mabs.28762] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Monoclonal antibody (mAb) products are extraordinarily heterogeneous due to the presence of a variety of enzymatic and chemical modifications, such as deamidation, isomerization, oxidation, glycosylation, glycation, and terminal cyclization. The modifications in different domains of the antibody molecule can result in different biological consequences. Therefore, characterization and routine monitoring of domain-specific modifications are essential to ensure the quality of the therapeutic antibody products. For this purpose, a rapid and informative methodology was developed to examine the heterogeneity of individual domains in mAb products. A recently discovered endopeptidase, IdeS, cleaves heavy chains below the hinge region, producing F(ab') 2 and Fc fragments. Following reduction of disulfide bonds, three antibody domains (LC, Fd, and Fc/2) can be released for further characterization. Subsequent analyses by liquid chromatography/mass spectrometry, capillary isoelectric focusing, and glycan mapping enable domain-specific profiling of oxidation, charge heterogeneity, and glycoform distribution. When coupled with reversed phase chromatography, the unique chromatographic profile of each molecule offers a simple strategy for an identity test, which is an important formal test for biopharmaceutical quality control purposes. This methodology is demonstrated for a number of IgGs of different subclasses (IgG1, IgG2, IgG4), as well as an Fc fusion protein. The presented technique provides a convenient platform approach for scientific and formal therapeutic mAb product characterization. It can also be applied in regulated drug substance batch release and stability testing of antibody and Fc fusion protein products, in particular for identity and routine monitoring of domain-specific modifications.
Collapse
Affiliation(s)
- Yan An
- Sterile Product and Analytical Development; Merck Research Laboratories; Kenilworth, NJ USA
| | - Ying Zhang
- Sterile Product and Analytical Development; Merck Research Laboratories; Kenilworth, NJ USA
| | - Hans-Martin Mueller
- Sterile Product and Analytical Development; Merck Research Laboratories; Kenilworth, NJ USA
| | - Mohammed Shameem
- Sterile Product and Analytical Development; Merck Research Laboratories; Kenilworth, NJ USA
| | - Xiaoyu Chen
- Sterile Product and Analytical Development; Merck Research Laboratories; Kenilworth, NJ USA
| |
Collapse
|
227
|
Henningham A, Döhrmann S, Nizet V, Cole JN. Mechanisms of group A Streptococcus resistance to reactive oxygen species. FEMS Microbiol Rev 2015; 39:488-508. [PMID: 25670736 PMCID: PMC4487405 DOI: 10.1093/femsre/fuu009] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/19/2014] [Indexed: 12/16/2022] Open
Abstract
Streptococcus pyogenes, also known as group A Streptococcus (GAS), is an exclusively human Gram-positive bacterial pathogen ranked among the ‘top 10’ causes of infection-related deaths worldwide. GAS commonly causes benign and self-limiting epithelial infections (pharyngitis and impetigo), and less frequent severe invasive diseases (bacteremia, toxic shock syndrome and necrotizing fasciitis). Annually, GAS causes 700 million infections, including 1.8 million invasive infections with a mortality rate of 25%. In order to establish an infection, GAS must counteract the oxidative stress conditions generated by the release of reactive oxygen species (ROS) at the infection site by host immune cells such as neutrophils and monocytes. ROS are the highly reactive and toxic byproducts of oxygen metabolism, including hydrogen peroxide (H2O2), superoxide anion (O2•−), hydroxyl radicals (OH•) and singlet oxygen (O2*), which can damage bacterial nucleic acids, proteins and cell membranes. This review summarizes the enzymatic and regulatory mechanisms utilized by GAS to thwart ROS and survive under conditions of oxidative stress. This review discusses the mechanisms utilized by the bacterial pathogen group A Streptococcus to detoxify reactive oxygen species and survive in the human host under conditions of oxidative stress.
Collapse
Affiliation(s)
- Anna Henningham
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA The School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia The Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Simon Döhrmann
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA Rady Children's Hospital, San Diego, CA 92123, USA
| | - Jason N Cole
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA The School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia The Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
228
|
Saleem RA, Affholter BR, Deng S, Campbell PC, Matthies K, Eakin CM, Wallace A. A chemical and computational approach to comprehensive glycation characterization on antibodies. MAbs 2015; 7:719-31. [PMID: 26030340 PMCID: PMC4622828 DOI: 10.1080/19420862.2015.1046663] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/15/2015] [Accepted: 04/22/2015] [Indexed: 01/10/2023] Open
Abstract
Non-enzymatic glycation is a challenging post-translational modification to characterize due to the structural heterogeneity it generates in proteins. Glycation has become increasingly recognized as an important product quality attribute to monitor, particularly for the biotechnology sector, which produces recombinant proteins under conditions that are amenable to protein glycation. The elucidation of sites of glycation can be problematic using conventional collision-induced dissociation (CID)-based mass spectrometry because of the predominance of neutral loss ions. A method to characterize glycation using an IgG1 monoclonal antibody (mAb) as a model is reported here. The sugars present on this mAb were derivatized using sodium borohydride chemistry to stabilize the linkage and identified using CID-based MS(2) mass spectrometry and spectral search engines. Quantification of specific glycation sites was then done using a targeted MS(1) based approach, which allowed the identification of a glycation hot spot in the heavy chain complementarity-determining region 3 of the mAb. This targeted approach provided a path forward to developing a structural understanding of the propensity of sites to become glycated on mAbs. Through structural analysis we propose a model in which the number and 3-dimensional distances of carboxylic acid amino acyl residues create a favorable environment for glycation to occur.
Collapse
Key Words
- BA, boronate affinity chromatography
- CDR3, complementary-determining region 3
- CEX, cation exchange chromatography
- CID, collision induced dissociation
- CV, coefficient of variation
- Da, daltons
- EIC, extracted ion chromatogram
- HC-CDR3, heavy chain complementary determining region 3
- HPLC, high performance liquid chromatography
- LC-MS2, liquid chromatography coupled with tandem mass spectrometry
- MS1, a mass to charge ratio survey scan
- MS2, tandem mass spectrometry - selected ions from MS1 are fragmented and fragment ion mass measured
- UPLC, ultrahigh performance liquid chromatography
- boronate affinity chromatography
- glycation
- mAb, monoclonal antibody
- structural modeling
- targeted mass spectrometry
- Å, angstroms
Collapse
Affiliation(s)
| | | | - Sihong Deng
- Drug Substance Development; Amgen Inc.; Seattle, WA, USA
| | | | - Kelli Matthies
- Functional Biocharacterization; Amgen Inc.; Thousand Oaks, CA, USA
| | | | - Alison Wallace
- Drug Substance Development; Amgen Inc.; Seattle, WA, USA
| |
Collapse
|
229
|
Formolo T, Ly M, Levy M, Kilpatrick L, Lute S, Phinney K, Marzilli L, Brorson K, Boyne M, Davis D, Schiel J. Determination of the NISTmAb Primary Structure. ACS SYMPOSIUM SERIES 2015. [DOI: 10.1021/bk-2015-1201.ch001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Trina Formolo
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Mellisa Ly
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Michaella Levy
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Lisa Kilpatrick
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Scott Lute
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Karen Phinney
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Lisa Marzilli
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Kurt Brorson
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Michael Boyne
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - Darryl Davis
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| | - John Schiel
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Mass Spectrometry and Biophysical Characterization, Analytical Research and Development, BioTherapeutics Pharmaceutical Sciences, Pfizer, Inc., Andover, Massachusetts 01810, United States
- Janssen Research and Development, LLC, Spring House, Pennsylvania 19002, United States
- Center for Drug Evaluation and Research, Office of Testing and Research, Division of Pharmaceutical Analysis, U.S. Food and Drug Administration, Saint Louis, Missouri 63110, United States
- Center for Drug Evaluation and Research, Office of Biotechnology Products, Division of Monoclonal Antibodies, U.S Food and Drug Administration, Silver Spring, Maryland 20993, United States
| |
Collapse
|
230
|
Michels DA, Ip AY, Dillon TM, Brorson K, Lute S, Chavez B, Prentice KM, Brady LJ, Miller KJ. Separation Methods and Orthogonal Techniques. ACS SYMPOSIUM SERIES 2015. [DOI: 10.1021/bk-2015-1201.ch005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- David A. Michels
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Anna Y. Ip
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Thomas M. Dillon
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Kurt Brorson
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Scott Lute
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Brittany Chavez
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Ken M. Prentice
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Lowell J. Brady
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| | - Karen J. Miller
- Department of Protein Analytical Chemistry, Genentech, South San Francisco, California 94080, United States
- Department of Process and Product Development, Amgen Inc., Thousand Oaks, California 91361, United States
- Division of Monoclonal Antibodies, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20903, United States
- Department of Process and Product Development, Amgen Inc., Seattle, Washington 98119, United States
- Global Analytical Sciences, Amgen Inc., Thousand Oaks, California 91320, United States
| |
Collapse
|
231
|
Fan X, Brezski RJ, Deng H, Dhupkar PM, Shi Y, Gonzalez A, Zhang S, Rycyzyn M, Strohl WR, Jordan RE, Zhang N, An Z. A Novel Therapeutic Strategy to Rescue the Immune Effector Function of Proteolytically Inactivated Cancer Therapeutic Antibodies. Mol Cancer Ther 2014; 14:681-91. [DOI: 10.1158/1535-7163.mct-14-0715] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022]
|
232
|
Brezski RJ, Kinder M, Grugan KD, Soring KL, Carton J, Greenplate AR, Petley T, Capaldi D, Brosnan K, Emmell E, Watson S, Jordan RE. A monoclonal antibody against hinge-cleaved IgG restores effector function to proteolytically-inactivated IgGs in vitro and in vivo. MAbs 2014; 6:1265-73. [PMID: 25517311 DOI: 10.4161/mabs.29825] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We report a chimeric monoclonal antibody (mAb) directed to a neo-epitope that is exposed in the IgG lower hinge following proteolytic cleavage. The mAb, designated 2095-2, displays specificity for IdeS-generated F(ab')₂ fragments, but not for full-length IgG or for closely-related F(ab')₂ fragments generated with other proteases. A critical component of the specificity is provided by the C-terminal amino acid of the epitope corresponding to gly-236 in the IgG1 (also IgG4) hinge. By its ability to bind to IdeS-cleaved anti-CD20 mAb, mAb 2095-2 fully restored antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against WIL2-S cells to the otherwise inactive anti-CD20 IgG1 F(ab')₂ fragment. Similarly, 2095-2 reinstated ADCC against MDA-MB-231 cells to an anti-CD142 IgG1 F(ab')₂ fragment. mAb 2095-2 was also capable of eliciting both CDC and ADCC to IgG4 F(ab')₂ fragments, an IgG subclass that has weaker ADCC and CDC when intact relative to intact IgG1. The in vitro cell-based efficacy of 2095-2 was extended to the in vivo setting using platelets as a cell clearance surrogate. In a canine model, the co-administration of 2095-2 together with IdeS-generated, platelet-targeting anti-CD41/61 F(ab')₂ fragment not only restored platelet clearance, but did so at a rate and extent of clearance that exceeded that of intact anti-CD41/61 IgG at comparable concentrations. To further explore this unexpected amplification effect, we conducted a rat study in which 2095-2 was administered at a series of doses in combination with a fixed dose of anti-CD41/61 F(ab')₂ fragments. Again, the combination, at ratios as low as 1:10 (w/w) 2095-2 to F(ab')₂, proved more effective than the anti-CD41/61 IgG1 alone. These findings suggest a novel mechanism for enhancing antibody-mediated cell-killing effector functions with potential applications in pathologic settings such as tumors and acute infections where protease activity is abundant.
Collapse
Affiliation(s)
- Randall J Brezski
- a Biologics Research, Janssen Research & Development, LLC , Spring House , PA , USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Wagner-Rousset E, Janin-Bussat MC, Colas O, Excoffier M, Ayoub D, Haeuw JF, Rilatt I, Perez M, Corvaïa N, Beck A. Antibody-drug conjugate model fast characterization by LC-MS following IdeS proteolytic digestion. MAbs 2014; 6:273-85. [PMID: 24135617 PMCID: PMC3929440 DOI: 10.4161/mabs.26773] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Here we report the design and production of an antibody-fluorophore conjugate (AFC) as a non-toxic model of an antibody-drug conjugate (ADC). This AFC is based on the conjugation of dansyl sulfonamide ethyl amine (DSEA )-linker maleimide on interchain cysteines of trastuzumab used as a reference antibody. The resulting AFC was first characterized by routine analytical methods (SEC, SDS-PAGE, CE-SDS, HIC and native MS), resulting in similar chromatograms,electropherograms and mass spectra to those reported for hinge Cys-linked ADCs. IdeS digestion of the AFC was then performed, followed by reduction and analysis by liquid chromatography coupled to mass spectrometry analysis. Dye loading and distribution on light chain and Fd fragments were calculated, as well as the average dye to antibody ratio (DAR) for both monomeric and multimeric species. In addition, by analyzing the Fc fragment in the same run, full glycoprofiling and demonstration of the absence of additional conjugation was easily achieved. As for naked antibodies and Fc-fusion proteins, IdeS proteolytic digestion may rapidly become a reference analytical method at all stages of ADC discovery, preclinical and clinical development. The method can be routinely used for comparability assays, formulation, process scale-up and transfer, and to define critical quality attributes in a quality-by-design approach.
Collapse
|
234
|
Abstract
PURPOSE OF REVIEW Increasing disease caused by beta-haemolytic streptococci indicates the need for improved understanding of pathogenesis. RECENT FINDINGS Streptococcus pyogenes, or group A Streptococcus (GAS), causes significant disease worldwide. The closely related Streptococcus dysgalactiae subspecies equisimilis (SDSE) is increasingly recognized as causing a similar disease spectrum. Whole-genome sequencing applied to the study of outbreaks may reveal factors that contribute to pathogenesis and changes in epidemiology. The role of quorum sensing in biofilm formation, and interspecies communication with other streptococci, is discussed. GAS has evolved multiple mechanisms to evade the humoral arm of innate immunity, including complement, which is well known in protecting the host from bacteria, and the coagulation-fibrinolytic system, which is increasingly recognized as an innate immune effector. SUMMARY Molecular biology has enhanced our understanding of the intricate balance of host-pathogen interactions that result in clearance or establishment of invasive streptococcal infection. Although the skin and oropharynx remain the usual ecological niche of GAS and SDSE, occasionally the bacteria find themselves within deeper tissues and blood. Recent research has armed us with better knowledge of bacterial adaptations to this alternative environment. However, the challenge is to translate this knowledge into clinical practice, through the development of novel therapeutic options and ultimately a vaccine against GAS.
Collapse
|
235
|
Badgujar SB, Mahajan RT. Nivulian-II a new milk clotting cysteine protease of Euphorbia nivulia latex. Int J Biol Macromol 2014; 70:391-8. [DOI: 10.1016/j.ijbiomac.2014.07.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/23/2014] [Accepted: 07/10/2014] [Indexed: 10/25/2022]
|
236
|
Saeki Y, Ishihara K. Infection-immunity liaison: pathogen-driven autoimmune-mimicry (PDAIM). Autoimmun Rev 2014; 13:1064-9. [PMID: 25182200 DOI: 10.1016/j.autrev.2014.08.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/26/2014] [Indexed: 12/19/2022]
Abstract
Autoimmunity causes pathological conditions resulting in autoimmune diseases (ADs). Although autoimmunity is a mystery, immunological dogma suggests that autoreactive cell reactivation (ACR) breaks self-tolerance and induces autoimmunity. Thus, ACR is a royal pathway for ADs. Cumulative evidence implicates environmental factors as secondary triggers of ADs in the genetically susceptible hosts. Infection is the most likely trigger. Although several mechanisms have been proposed to explain how infectious agents trigger ADs, ACR is assumed to be an essential pathway. Here, by showing some exemplary ADs, we propose two novel pathways, "molecular modification pathway" and "hyper-immune-inflammatory response pathway", which induce AD-like conditions directly by infectious agents without ACR. These AD-like conditions are actually not true "ADs" according to the current definition. Therefore, we define them as "pathogen-driven autoimmune-mimicry (PDAIM)". Confirming PDAIM will open perspectives in developing novel fundamental and non-immunosuppressive therapies for ADs. The idea should also provide novel insights into both the mechanisms of autoimmunity and the pathogenesis of ADs.
Collapse
Affiliation(s)
- Yukihiko Saeki
- Department of Clinical Research, National Hospital Organization (NHO) Osaka Minami Medical Center, 2-1 Kidohigashi-machi, Kawachinagano City, Osaka 586-8521, Japan.
| | - Katsuhiko Ishihara
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, 577 Matsushima, Kurashiki City, Okayama 701-0192, Japan
| |
Collapse
|
237
|
Klonoski JM, Hurtig HR, Juber BA, Schuneman MJ, Bickett TE, Svendsen JM, Burum B, Penfound TA, Sereda G, Dale JB, Chaussee MS, Huber VC. Vaccination against the M protein of Streptococcus pyogenes prevents death after influenza virus: S. pyogenes super-infection. Vaccine 2014; 32:5241-9. [PMID: 25077423 DOI: 10.1016/j.vaccine.2014.06.093] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 12/21/2022]
Abstract
Influenza virus infections are associated with a significant number of illnesses and deaths on an annual basis. Many of the deaths are due to complications from secondary bacterial invaders, including Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae, and Streptococcus pyogenes. The β-hemolytic bacteria S. pyogenes colonizes both skin and respiratory surfaces, and frequently presents clinically as strep throat or impetigo. However, when these bacteria gain access to normally sterile sites, they can cause deadly diseases including sepsis, necrotizing fasciitis, and pneumonia. We previously developed a model of influenza virus:S. pyogenes super-infection, which we used to demonstrate that vaccination against influenza virus can limit deaths associated with a secondary bacterial infection, but this protection was not complete. In the current study, we evaluated the efficacy of a vaccine that targets the M protein of S. pyogenes to determine whether immunity toward the bacteria alone would allow the host to survive an influenza virus:S. pyogenes super-infection. Our data demonstrate that vaccination against the M protein induces IgG antibodies, in particular those of the IgG1 and IgG2a isotypes, and that these antibodies can interact with macrophages. Ultimately, this vaccine-induced immunity eliminated death within our influenza virus:S. pyogenes super-infection model, despite the fact that all M protein-vaccinated mice showed signs of illness following influenza virus inoculation. These findings identify immunity against bacteria as an important component of protection against influenza virus:bacteria super-infection.
Collapse
Affiliation(s)
- Joshua M Klonoski
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Heather R Hurtig
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Brian A Juber
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Margaret J Schuneman
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Thomas E Bickett
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Joshua M Svendsen
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Brandon Burum
- Department of Chemistry, University of South Dakota, Vermillion, SD, United States
| | - Thomas A Penfound
- University of Tennessee Health Science Center and the Veterans Affairs Medical Center Research Service, Memphis, TN, United States
| | - Grigoriy Sereda
- Department of Chemistry, University of South Dakota, Vermillion, SD, United States
| | - James B Dale
- University of Tennessee Health Science Center and the Veterans Affairs Medical Center Research Service, Memphis, TN, United States
| | - Michael S Chaussee
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Victor C Huber
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States.
| |
Collapse
|
238
|
Bondt A, Rombouts Y, Selman MHJ, Hensbergen PJ, Reiding KR, Hazes JMW, Dolhain RJEM, Wuhrer M. Immunoglobulin G (IgG) Fab glycosylation analysis using a new mass spectrometric high-throughput profiling method reveals pregnancy-associated changes. Mol Cell Proteomics 2014; 13:3029-39. [PMID: 25004930 PMCID: PMC4223489 DOI: 10.1074/mcp.m114.039537] [Citation(s) in RCA: 200] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The N-linked glycosylation of the constant fragment (Fc) of immunoglobulin G has been shown to change during pathological and physiological events and to strongly influence antibody inflammatory properties. In contrast, little is known about Fab-linked N-glycosylation, carried by ∼ 20% of IgG. Here we present a high-throughput workflow to analyze Fab and Fc glycosylation of polyclonal IgG purified from 5 μl of serum. We were able to detect and quantify 37 different N-glycans by means of MALDI-TOF-MS analysis in reflectron positive mode using a novel linkage-specific derivatization of sialic acid. This method was applied to 174 samples of a pregnancy cohort to reveal Fab glycosylation features and their change with pregnancy. Data analysis revealed marked differences between Fab and Fc glycosylation, especially in the levels of galactosylation and sialylation, incidence of bisecting GlcNAc, and presence of high mannose structures, which were all higher in the Fab portion than the Fc, whereas Fc showed higher levels of fucosylation. Additionally, we observed several changes during pregnancy and after delivery. Fab N-glycan sialylation was increased and bisection was decreased relative to postpartum time points, and nearly complete galactosylation of Fab glycans was observed throughout. Fc glycosylation changes were similar to results described before, with increased galactosylation and sialylation and decreased bisection during pregnancy. We expect that the parallel analysis of IgG Fab and Fc, as set up in this paper, will be important for unraveling roles of these glycans in (auto)immunity, which may be mediated via recognition by human lectins or modulation of antigen binding.
Collapse
Affiliation(s)
- Albert Bondt
- From the ‡Department of Rheumatology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Yoann Rombouts
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; ¶Department of Rheumatology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Maurice H J Selman
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Paul J Hensbergen
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Karli R Reiding
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Johanna M W Hazes
- From the ‡Department of Rheumatology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Radboud J E M Dolhain
- From the ‡Department of Rheumatology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Manfred Wuhrer
- §Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; ‖Division of BioAnalytical Chemistry, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands; **Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|
239
|
LC-MS analysis of polyclonal IgGs using IdeS enzymatic proteolysis for oxidation monitoring. J Chromatogr B Analyt Technol Biomed Life Sci 2014; 961:1-4. [PMID: 24837162 DOI: 10.1016/j.jchromb.2014.04.053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/01/2014] [Accepted: 04/29/2014] [Indexed: 01/01/2023]
Abstract
Susceptibility of IgGs to oxidation is a significant issue for intravenous immunoglobulin preparations (IVIG) in liquid solution and raises both safety and efficacy concerns. Here we present an optimized chromatography method coupled to mass spectrometry (MS) to determine the oxidation of Fc/2 fragments derived from polyclonal IgGs after IdeS treatment. Separation of the four IgG subclasses was achieved using a diphenyl column and UV/MS detections were used for quantification and characterization. Several oxidized Fc/2 fragments generated by stress conditions were resolved and oxidized methionines were identified. This procedure can be used to monitor the oxidative status of IVIG preparations during formulation or stability studies.
Collapse
|
240
|
Dual-site phosphorylation of the control of virulence regulator impacts group a streptococcal global gene expression and pathogenesis. PLoS Pathog 2014; 10:e1004088. [PMID: 24788524 PMCID: PMC4006921 DOI: 10.1371/journal.ppat.1004088] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/10/2014] [Indexed: 11/20/2022] Open
Abstract
Phosphorylation relays are a major mechanism by which bacteria alter transcription in response to environmental signals, but understanding of the functional consequences of bacterial response regulator phosphorylation is limited. We sought to characterize how phosphorylation of the control of virulence regulator (CovR) protein from the major human pathogen group A Streptococcus (GAS) influences GAS global gene expression and pathogenesis. CovR mainly serves to repress GAS virulence factor-encoding genes and has been shown to homodimerize following phosphorylation on aspartate-53 (D53) in vitro. We discovered that CovR is phosphorylated in vivo and that such phosphorylation is partially heat-stable, suggesting additional phosphorylation at non-aspartate residues. Using mass spectroscopy along with targeted mutagenesis, we identified threonine-65 (T65) as an additional CovR phosphorylation site under control of the serine/threonine kinase (Stk). Phosphorylation on T65, as mimicked by the recombinant CovR T65E variant, abolished in vitro CovR D53 phosphorylation. Similarly, isoallelic GAS strains that were either unable to be phosphorylated at D53 (CovR-D53A) or had functional constitutive phosphorylation at T65 (CovR-T65E) had essentially an identical gene repression profile to each other and to a CovR-inactivated strain. However, the CovR-D53A and CovR-T65E isoallelic strains retained the ability to positively influence gene expression that was abolished in the CovR-inactivated strain. Consistent with these observations, the CovR-D53A and CovR-T65E strains were hypervirulent compared to the CovR-inactivated strain in a mouse model of invasive GAS disease. Surprisingly, an isoalleic strain unable to be phosphorylated at CovR T65 (CovR-T65A) was hypervirulent compared to the wild-type strain, as auto-regulation of covR gene expression resulted in lower covR gene transcript and CovR protein levels in the CovR-T65A strain. Taken together, these data establish that CovR is phosphorylated in vivo and elucidate how the complex interplay between CovR D53 activating phosphorylation, T65 inhibiting phosphorylation, and auto-regulation impacts streptococcal host-pathogen interaction. Group A Streptococcus (GAS) causes a variety of human diseases ranging from mild throat infections to deadly invasive infections. The capacity of GAS to cause infections at such diverse locations is dependent on its ability to precisely control the production of a broad variety of virulence factors. The control of virulence regulator (CovR) is a master regulator of GAS genes encoding virulence factors. It is known that CovR can be phosphorylated on aspartate-53 in vitro and that such phosphorylation increases its regulatory activity, but what additional factors influence CovR-mediated gene expression have not been established. Herein we show for the first time that CovR is phosphorylated in vivo and that phosphorylation of CovR on threonine-65 by the threonine/serine kinase Stk prevents aspartate-53 phosphorylation, thereby decreasing CovR regulatory activity. Further, while CovR-mediated gene repression is highly dependent on aspartate-53 phosphorylation, CovR-mediated gene activation proceeds via a phosphorylation-independent mechanism. Modifications in CovR phosphorylation sites significantly affected the expression of GAS virulence factors during infection and markedly altered the ability of GAS to cause disease in mice. These data establish that multiple inter-related pathways converge to influence CovR phosphorylation, thereby providing new insight into the complex regulatory network used by GAS during infection.
Collapse
|
241
|
Abstract
Acute rheumatic fever is an inflammatory sequela of Group A Streptococcal pharyngitis that affects multiple organ systems. The incidence of acute rheumatic fever has been declining even before the use of antibiotics became widespread, however the disease remains a significant cause of morbidity and mortality in children, particularly in developing countries and has been estimated to affect 19 per 100,000 children worldwide. Acute rheumatic fever is a clinical diagnosis, and therefore subject to the judgment of the clinician. Because of the variable presentation, the Jones criteria were first developed in 1944 to aid clinicians in the diagnosis of acute rheumatic fever. The Jones criteria have been modified throughout the years, most recently in 1992 to aid clinicians in the diagnosis of initial attacks of acute rheumatic fever and to minimize overdiagnosis of the disease. Diagnosis of acute rheumatic fever is based on the presence of documented preceding Group A Streptococcal infection, in addition to the presence of two major manifestations or one major and two minor manifestations of the Jones criteria. Without documentation of antecedent Group A Streptococcal infection, the diagnosis is much less likely except in a few rare scenarios. Carditis, polyarthritis and Sydenham's chorea are the most common major manifestations of acute rheumatic fever. However, despite the predominance of these major manifestations of acute rheumatic fever, there can be significant overlap with other disorders such as Lyme disease, serum sickness, drug reactions, and post-Streptococcal reactive arthritis. This overlap between disease processes has led to continued investigation of the pathophysiology as well as development of new biomarkers and laboratory studies to aid in the diagnosis of acute rheumatic fever and distinction from other disease processes.
Collapse
Affiliation(s)
- Rebecca J Burke
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Thomas Jefferson University, 1600 Rockland Road, Wilmington, DE 19803, United States
| | - Christopher Chang
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Thomas Jefferson University, 1600 Rockland Road, Wilmington, DE 19803, United States.
| |
Collapse
|
242
|
Kinder M, Greenplate AR, Grugan KD, Soring KL, Heeringa KA, McCarthy SG, Bannish G, Perpetua M, Lynch F, Jordan RE, Strohl WR, Brezski RJ. Engineered protease-resistant antibodies with selectable cell-killing functions. J Biol Chem 2013; 288:30843-54. [PMID: 23986451 DOI: 10.1074/jbc.m113.486142] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Molecularly engineered antibodies with fit-for-purpose properties will differentiate next generation antibody therapeutics from traditional IgG1 scaffolds. One requirement for engineering the most appropriate properties for a particular therapeutic area is an understanding of the intricacies of the target microenvironment in which the antibody is expected to function. Our group and others have demonstrated that proteases secreted by invasive tumors and pathological microorganisms are capable of cleaving human IgG1, the most commonly adopted isotype among monoclonal antibody therapeutics. Specific cleavage in the lower hinge of IgG1 results in a loss of Fc-mediated cell-killing functions without a concomitant loss of antigen binding capability or circulating antibody half-life. Proteolytic cleavage in the hinge region by tumor-associated or microbial proteases is postulated as a means of evading host immune responses, and antibodies engineered with potent cell-killing functions that are also resistant to hinge proteolysis are of interest. Mutation of the lower hinge region of an IgG1 resulted in protease resistance but also resulted in a profound loss of Fc-mediated cell-killing functions. In the present study, we demonstrate that specific mutations of the CH2 domain in conjunction with lower hinge mutations can restore and sometimes enhance cell-killing functions while still retaining protease resistance. By identifying mutations that can restore either complement- or Fcγ receptor-mediated functions on a protease-resistant scaffold, we were able to generate a novel protease-resistant platform with selective cell-killing functionality.
Collapse
Affiliation(s)
- Michelle Kinder
- From Biologics Research, Janssen Research and Development, LLC, Spring House, Pennsylvania 19477
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Watanabe S, Shimomura Y, Ubukata K, Kirikae T, Miyoshi-Akiyama T. Concomitant regulation of host tissue-destroying virulence factors and carbohydrate metabolism during invasive diseases induced by group g streptococci. J Infect Dis 2013; 208:1482-93. [PMID: 23901096 DOI: 10.1093/infdis/jit353] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Streptococcus dysgalactiae subsp. equisimilis (SDSE) has Lancefield group G or C antigens. Recent epidemiological studies reveal that invasive SDSE infections have been increasing in Asia, Europe, and the United States. The mechanisms and key virulence factors by which SDSE causes invasive diseases are poorly understood. METHODS We analyzed the SDSE transcriptome in vivo during intraperitoneal infection in mice. We also compared the abundance of streptolysin S (SLS) and streptolysin O (SLO) production between clinically dominant stG6792 strains and other clinical isolates. RESULTS Microarray data suggest that SDSE degraded host tissue polysaccharides by secreting poly/oligosaccharide lyases and simultaneously used the Entner-Doudoroff pathway to metabolize acquired carbohydrates. A global negative virulence gene regulator CsrRS of SDSE modulated the expression of genes encoding SLS and enzymes that metabolize carbohydrates. Moreover, a csrS-deficient mutant induced severe systemic hemolysis in mice. The most frequently isolated stG6792 strains secreted abundant SLS and SLO rather than other SDSE emm types, indicating the potential relationship between production of SLS and SLO and poor outcomes. CONCLUSIONS Our findings suggest that the concomitant regulation of virulence factors that destroy host tissues and metabolic enzymes might play an important role in invasive diseases induced by SDSE.
Collapse
Affiliation(s)
- Shinya Watanabe
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine
| | | | | | | | | |
Collapse
|
244
|
IgG protease Mac/IdeS is not essential for phagocyte resistance or mouse virulence of M1T1 group A Streptococcus. mBio 2013; 4:mBio.00499-13. [PMID: 23900173 PMCID: PMC3735186 DOI: 10.1128/mbio.00499-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The Mac/IdeS protein of group A Streptococcus (GAS) is a secreted cysteine protease with cleavage specificity for IgG and is highly expressed in the GAS serotype M1T1 clone, which is the serotype most frequently isolated from patients with life-threatening invasive infections. While studies of Mac/IdeS with recombinant protein have shown that the protein can potentially prevent opsonophagocytosis of GAS by neutrophils, the role of the protein in immune evasion as physiologically produced by the living organism has not been studied. Here we examined the contribution of Mac/IdeS to invasive GAS disease by generating a mutant lacking Mac/IdeS in the hyperinvasive M1T1 background. While Mac/IdeS was highly expressed and proteolytically active in the hyperinvasive strain, elimination of the bacterial protease did not significantly influence GAS phagocytic uptake, oxidative-burst induction, cathelicidin sensitivity, resistance to neutrophil or macrophage killing, or pathogenicity in pre- or postimmune mouse infectious challenges. We conclude that in the highly virulent M1T1 background, Mac/IdeS is not essential for either phagocyte resistance or virulence. Given the conservation of Mac/IdeS and homologues across GAS strains, it is possible that Mac/IdeS serves another important function in GAS ecology or contributes to virulence in other strain backgrounds. Group A Streptococcus (GAS) causes human infections ranging from strep throat to life-threatening conditions such as flesh-eating disease and toxic shock syndrome. Common disease-associated clones of GAS can cause both mild and severe infections because of a characteristic mutation and subsequent change in the expression of several genes that develops under host immune selection. One of these genes encodes Mac/IdeS, a protease that has been shown to cleave antibodies important to the immune defense system. In this study, we found that while Mac/IdeS is highly expressed in hypervirulent GAS, it does not significantly contribute to the ability of the bacteria to survive white blood cell killing or produce invasive infection in the mouse. These data underscore the importance of correlating studies on virulence factor function with physiologic expression levels and the complexity of streptococcal pathogenesis and contribute to our overall understanding of how GAS causes disease.
Collapse
|
245
|
|
246
|
Woodard J, Lau H, Latypov RF. Nondenaturing Size-Exclusion Chromatography-Mass Spectrometry to Measure Stress-Induced Aggregation in a Complex Mixture of Monoclonal Antibodies. Anal Chem 2013; 85:6429-36. [DOI: 10.1021/ac401455f] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jonathan Woodard
- Drug Product Development, Amgen Inc., Seattle, Washington 98119, United States
| | - Hollis Lau
- Drug Product Development, Amgen Inc., Seattle, Washington 98119, United States
| | - Ramil F. Latypov
- Drug Product Development, Amgen Inc., Seattle, Washington 98119, United States
| |
Collapse
|
247
|
Characterization of streptococcal platelet-activating factor acetylhydrolase variants that are involved in innate immune evasion. Infect Immun 2013; 81:3128-38. [PMID: 23774595 DOI: 10.1128/iai.00398-13] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human pathogen group A streptococcus (GAS) has developed mechanisms to subvert innate immunity. We recently reported that the secreted esterase produced by serotype M1 GAS (SsE(M1)) reduces neutrophil recruitment by targeting platelet-activating factor (PAF). SsE(M1) and SsE produced by serotype M28 GAS (SsE(M28)) have a 37% sequence difference. This study aims at determining whether SsE(M28) is also a PAF acetylhydrolase and participates in innate immune evasion. We also examined whether SsE evolved to target PAF by characterizing the PAF acetylhydrolase (PAF-AH) activity and substrate specificity of SsE(M1), SsE(M28), SeE, the SsE homologue in Streptococcus equi, and human plasma PAF-AH (hpPAF-AH). PAF incubated with SsE(M28) or SeE was converted into lyso-PAF. SsE(M1) and SsE(M28) had kcat values of 373 s(-1) and 467 s(-1), respectively, that were ≥ 30-fold greater than that of hpPAF-AH (12 s(-1)). The comparison of SsE(M1), SsE(M28), and hpPAF-AH in kcat and Km in hydrolyzing triglycerides, acetyl esters, and PAF indicates that the SsE proteins are more potent hydrolases against PAF and have high affinity for PAF. SsE(M28) possesses much lower esterase activities against triglycerides and other esters than SsE(M1) but have similar potency with SsE(M1) in PAF hydrolysis. Deletion of sse(M28) in a covS deletion mutant of GAS increased neutrophil recruitment and reduced skin infection, whereas in trans expression of SsE(M28) in GAS reduced neutrophil infiltration and increased skin invasion in subcutaneous infection of mice. These results suggest that the SsE proteins evolved to target PAF for enhancing innate immune evasion and skin invasion.
Collapse
|
248
|
Tradtrantip L, Asavapanumas N, Verkman AS. Therapeutic cleavage of anti-aquaporin-4 autoantibody in neuromyelitis optica by an IgG-selective proteinase. Mol Pharmacol 2013; 83:1268-75. [PMID: 23571414 PMCID: PMC3657102 DOI: 10.1124/mol.113.086470] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/09/2013] [Indexed: 02/06/2023] Open
Abstract
Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system caused by binding of pathogenic IgG autoantibodies (NMO-IgG) to astrocyte water channel aquaporin-4 (AQP4). Astrocyte damage and downstream inflammation require NMO-IgG effector function to initiate complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). Here, we evaluated the potential therapeutic utility of the bacterial enzyme IdeS (IgG-degrading enzyme of Streptococcus pyogenes), which selectively cleaves IgG antibodies to yield Fc and F(ab')(2) fragments. In AQP4-expressing cell cultures, IdeS treatment of monoclonal NMO-IgGs and NMO patient sera abolished CDC and ADCC, even when IdeS was added after NMO-IgG was bound to AQP4. Binding of NMO-IgG to AQP4 was similar to that of the NMO-F(ab')(2) generated by IdeS cleavage. NMO-F(ab')(2) competitively displaced pathogenic NMO-IgG, preventing cytotoxicity, and the Fc fragments generated by IdeS cleavage reduced CDC and ADCC. IdeS efficiently cleaved NMO-IgG in mice in vivo, and greatly reduced NMO lesions in mice administered NMO-IgG and human complement. IgG-selective cleavage by IdeS thus neutralizes NMO-IgG pathogenicity, and yields therapeutic F(ab')(2) and Fc fragments. IdeS treatment, by therapeutic apheresis or direct administration, may be beneficial in NMO.
Collapse
Affiliation(s)
- Lukmanee Tradtrantip
- Departments of Medicine and Physiology, University of California, San Francisco, CA, USA
| | | | | |
Collapse
|
249
|
Vindebro R, Spoerry C, von Pawel-Rammingen U. Rapid IgG heavy chain cleavage by the streptococcal IgG endopeptidase IdeS is mediated by IdeS monomers and is not due to enzyme dimerization. FEBS Lett 2013; 587:1818-22. [PMID: 23665032 DOI: 10.1016/j.febslet.2013.04.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 04/19/2013] [Accepted: 04/23/2013] [Indexed: 10/26/2022]
Abstract
Streptococcus pyogenes employs an IgG specific endopeptidase, IdeS, to counteract the effector functions of specific IgG. The physiological significant step in disarming specific IgG is the cleavage of one IgG heavy chain. So far, characterizations of IdeS enzymatic activity have employed techniques that failed to differentiate between the first and the second cleavage step. The present data demonstrate that IdeS is active as a monomer and that IdeS activity follows classical Michaelis-Menten kinetics arguing against the previously proposed formation of a functional IdeS dimer. Our results show that IdeS inactivates IgG 100-fold faster than previously reported.
Collapse
Affiliation(s)
- Reine Vindebro
- Department of Molecular Biology and Umeå Centre for Microbial Research, Umeå University, 90187 Umeå, Sweden
| | | | | |
Collapse
|
250
|
Potempa M, Potempa J. Protease-dependent mechanisms of complement evasion by bacterial pathogens. Biol Chem 2013; 393:873-88. [PMID: 22944688 DOI: 10.1515/hsz-2012-0174] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 05/06/2012] [Indexed: 12/11/2022]
Abstract
The human immune system has evolved a variety of mechanisms for the primary task of neutralizing and eliminating microbial intruders. As the first line of defense, the complement system is responsible for rapid recognition and opsonization of bacteria, presentation to phagocytes and bacterial cell killing by direct lysis. All successful human pathogens have mechanisms of circumventing the antibacterial activity of the complement system and escaping this stage of the immune response. One of the ways in which pathogens achieve this is the deployment of proteases. Based on the increasing number of recent publications in this area, it appears that proteolytic inactivation of the antibacterial activities of the complement system is a common strategy of avoiding targeting by this arm of host innate immune defense. In this review, we focus on those bacteria that deploy proteases capable of degrading complement system components into non-functional fragments, thus impairing complement-dependent antibacterial activity and facilitating pathogen survival inside the host.
Collapse
Affiliation(s)
- Michal Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland.
| | | |
Collapse
|