Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
PLoS Pathog ; 20(4): e1012134, 2024 Apr.
Article En | MEDLINE | ID: mdl-38603762

Monoclonal antibodies (mAbs) are an important class of antiviral therapeutics. MAbs are highly selective, well tolerated, and have long in vivo half-life as well as the capacity to induce immune-mediated virus clearance. Their activities can be further enhanced by integration of their variable fragments (Fvs) into bispecific antibodies (bsAbs), affording simultaneous targeting of multiple epitopes to improve potency and breadth and/or to mitigate against viral escape by a single mutation. Here, we explore a bsAb strategy for generation of pan-ebolavirus and pan-filovirus immunotherapeutics. Filoviruses, including Ebola virus (EBOV), Sudan virus (SUDV), and Marburg virus (MARV), cause severe hemorrhagic fever. Although there are two FDA-approved mAb therapies for EBOV infection, these do not extend to other filoviruses. Here, we combine Fvs from broad ebolavirus mAbs to generate novel pan-ebolavirus bsAbs that are potently neutralizing, confer protection in mice, and are resistant to viral escape. Moreover, we combine Fvs from pan-ebolavirus mAbs with those of protective MARV mAbs to generate pan-filovirus protective bsAbs. These results provide guidelines for broad antiviral bsAb design and generate new immunotherapeutic candidates.


Antibodies, Bispecific , Antibodies, Viral , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Mice , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Antibodies, Viral/immunology , Humans , Filoviridae/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Monoclonal/immunology , Female , Mice, Inbred BALB C , Filoviridae Infections/immunology , Filoviridae Infections/therapy , Filoviridae Infections/prevention & control
2.
Nat Microbiol ; 8(6): 1051-1063, 2023 06.
Article En | MEDLINE | ID: mdl-37188812

Human monoclonal antibodies (mAbs) that target the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein have been isolated from convalescent individuals and developed into therapeutics for SARS-CoV-2 infection. However, therapeutic mAbs for SARS-CoV-2 have been rendered obsolete by the emergence of mAb-resistant virus variants. Here we report the generation of a set of six human mAbs that bind the human angiotensin-converting enzyme-2 (hACE2) receptor, rather than the SARS-CoV-2 spike protein. We show that these antibodies block infection by all hACE2 binding sarbecoviruses tested, including SARS-CoV-2 ancestral, Delta and Omicron variants at concentrations of ~7-100 ng ml-1. These antibodies target an hACE2 epitope that binds to the SARS-CoV-2 spike, but they do not inhibit hACE2 enzymatic activity nor do they induce cell-surface depletion of hACE2. They have favourable pharmacology, protect hACE2 knock-in mice against SARS-CoV-2 infection and should present a high genetic barrier to the acquisition of resistance. These antibodies should be useful prophylactic and treatment agents against any current or future SARS-CoV-2 variants and might be useful to treat infection with any hACE2-binding sarbecoviruses that emerge in the future.


COVID-19 , Severe acute respiratory syndrome-related coronavirus , Humans , Animals , Mice , SARS-CoV-2 , COVID-19/prevention & control , Antibodies, Monoclonal/pharmacology
3.
JCI Insight ; 7(5)2022 03 08.
Article En | MEDLINE | ID: mdl-35260532

Phosphopeptides derived from dysregulated protein phosphorylation in cancer cells can be processed and presented by MHC class I and class II molecules and, therefore, represent an untapped class of tumor-specific antigens that could be used as widely expressed "public" cancer neoantigens (NeoAgs). We generated a TCR mimic (TCRm) mAb, 6B1, specific for a phosphopeptide derived from insulin receptor substrate 2 (pIRS2) presented by HLA-A*02:01. The pIRS2 epitope's presentation by HLA-A*02:01 was confirmed by mass spectrometry. The TCRm 6B1 specifically bound to pIRS2/HLA-A2 complex on tumor cell lines that expressed pIRS2 in the context of HLA-A*02:01. Bispecific mAbs engaging CD3 of T cells were able to kill tumor cell lines in a pIRS2- and HLA-A*02:01-restricted manner. Structure modeling shows a prerequisite for an arginine or lysine at the first position to bind mAb. Therefore, 6B1 could recognize phosphopeptides derived from various phosphorylated proteins with similar amino acid compositions. This raised the possibility that a TCRm specific for the pIRS2/HLA-A2 complex could target a range of phosphopeptides presented by HLA-A*02:01 in various tumor cells. This is the first TCRm mAb to our knowledge targeting a phosphopeptide/MHC class I complex; the potential of this class of agents for clinical applications warrants further investigation.


HLA-A2 Antigen , Phosphopeptides , Antibodies, Monoclonal/metabolism , Insulin Receptor Substrate Proteins/metabolism , Phosphopeptides/metabolism , Receptors, Antigen, T-Cell/metabolism
4.
Cell Chem Biol ; 29(5): 811-823.e7, 2022 05 19.
Article En | MEDLINE | ID: mdl-35231399

Zika virus (ZIKV) is a flavivirus that can cause severe disease, but there are no approved treatments or vaccines. A complication for flavivirus vaccine development is the potential of immunogens to enhance infection via antibody-dependent enhancement (ADE), a process mediated by poorly neutralizing and cross-reactive antibodies. Thus, there is a great need to develop immunogens that minimize the potential to elicit enhancing antibodies. Here we utilized structure-based protein engineering to develop "resurfaced" (rs) ZIKV immunogens based on E glycoprotein domain III (ZDIIIs), in which epitopes bound by variably neutralizing antibodies were masked by combinatorial mutagenesis. We identified one resurfaced ZDIII immunogen (rsZDIII-2.39) that elicited a protective but immune-focused response. Compared to wild type ZDIII, immunization with resurfaced rsZDIII-2.39 protein nanoparticles produced fewer numbers of ZIKV EDIII antigen-reactive B cells and elicited serum that had a lower magnitude of induced ADE against dengue virus serotype 1 (DENV1) Our findings enhance our understanding of the structural and functional determinants of antibody protection against ZIKV.


Dengue Virus , Nanoparticles , Zika Virus Infection , Zika Virus , Antibodies, Neutralizing , Antibodies, Viral , Dengue Virus/chemistry , Humans , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Zika Virus Infection/prevention & control
5.
Front Immunol ; 12: 729851, 2021.
Article En | MEDLINE | ID: mdl-34721393

Multiple agents in the family Filoviridae (filoviruses) are associated with sporadic human outbreaks of highly lethal disease, while others, including several recently identified agents, possess strong zoonotic potential. Although viral glycoprotein (GP)-specific monoclonal antibodies have demonstrated therapeutic utility against filovirus disease, currently FDA-approved molecules lack antiviral breadth. The development of broadly neutralizing antibodies has been challenged by the high sequence divergence among filovirus GPs and the complex GP proteolytic cleavage cascade that accompanies filovirus entry. Despite this variability in the antigenic surface of GP, all filoviruses share a site of vulnerability-the binding site for the universal filovirus entry receptor, Niemann-Pick C1 (NPC1). Unfortunately, this site is shielded in extracellular GP and only uncovered by proteolytic cleavage by host proteases in late endosomes and lysosomes, which are generally inaccessible to antibodies. To overcome this obstacle, we previously developed a 'Trojan horse' therapeutic approach in which engineered bispecific antibodies (bsAbs) coopt viral particles to deliver GP:NPC1 interaction-blocking antibodies to their endo/lysosomal sites of action. This approach afforded broad protection against members of the genus Ebolavirus but could not neutralize more divergent filoviruses. Here, we describe next-generation Trojan horse bsAbs that target the endo/lysosomal GP:NPC1 interface with pan-filovirus breadth by exploiting the conserved and widely expressed host cation-independent mannose-6-phosphate receptor for intracellular delivery. Our work highlights a new avenue for the development of single therapeutics protecting against all known and newly emerging filoviruses.


Antibodies, Bispecific/pharmacology , Antiviral Agents/pharmacology , Broadly Neutralizing Antibodies/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Lysosomes/drug effects , Niemann-Pick C1 Protein/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Antibodies, Bispecific/genetics , Broadly Neutralizing Antibodies/genetics , Ebolavirus/immunology , Ebolavirus/pathogenicity , Epitopes , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions , Humans , Ligands , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/virology , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/immunology , Niemann-Pick C1 Protein/metabolism , Protein Engineering , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , THP-1 Cells , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism
6.
Chembiochem ; 22(24): 3443-3451, 2021 12 10.
Article En | MEDLINE | ID: mdl-34605595

With the emergence of novel viruses, the development of new antivirals is more urgent than ever. A key step in human immunodeficiency virus type 1 (HIV-1) infection is six-helix bundle formation within the envelope protein subunit gp41. Selective disruption of bundle formation by peptides has been shown to be effective; however, these drugs, exemplified by T20, are prone to rapid clearance from the patient. The incorporation of non-natural amino acids is known to improve these pharmacokinetic properties. Here, we evaluate a peptide inhibitor in which a critical Ile residue is replaced by fluorinated analogues. We characterized the influence of the fluorinated analogues on the biophysical properties of the peptide. Furthermore, we show that the fluorinated peptides can block HIV-1 infection of target cells at nanomolar levels. These findings demonstrate that fluorinated amino acids are appropriate tools for the development of novel peptide therapeutics.


Anti-HIV Agents/pharmacology , HIV Envelope Protein gp41/antagonists & inhibitors , HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Peptides/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , HIV Envelope Protein gp41/metabolism , HIV Fusion Inhibitors/chemical synthesis , HIV Fusion Inhibitors/chemistry , HIV Infections/metabolism , HIV-1/metabolism , Halogenation , Humans , Microbial Sensitivity Tests , Peptides/chemical synthesis , Peptides/chemistry
7.
Cell ; 184(13): 3486-3501.e21, 2021 06 24.
Article En | MEDLINE | ID: mdl-34077751

Crimean-Congo hemorrhagic fever virus (CCHFV) is a World Health Organization priority pathogen. CCHFV infections cause a highly lethal hemorrhagic fever for which specific treatments and vaccines are urgently needed. Here, we characterize the human immune response to natural CCHFV infection to identify potent neutralizing monoclonal antibodies (nAbs) targeting the viral glycoprotein. Competition experiments showed that these nAbs bind six distinct antigenic sites in the Gc subunit. These sites were further delineated through mutagenesis and mapped onto a prefusion model of Gc. Pairwise screening identified combinations of non-competing nAbs that afford synergistic neutralization. Further enhancements in neutralization breadth and potency were attained by physically linking variable domains of synergistic nAb pairs through bispecific antibody (bsAb) engineering. Although multiple nAbs protected mice from lethal CCHFV challenge in pre- or post-exposure prophylactic settings, only a single bsAb, DVD-121-801, afforded therapeutic protection. DVD-121-801 is a promising candidate suitable for clinical development as a CCHFV therapeutic.


Antibodies, Neutralizing/immunology , Hemorrhagic Fever, Crimean/immunology , Survivors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigens, Viral/metabolism , Biophysical Phenomena , Chlorocebus aethiops , Epitope Mapping , Epitopes/metabolism , Female , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/prevention & control , Humans , Immunoglobulin G/metabolism , Male , Mice , Neutralization Tests , Protein Binding , Protein Engineering , Recombinant Proteins/immunology , Vero Cells , Viral Proteins/chemistry
8.
PLoS Pathog ; 15(11): e1008061, 2019 11.
Article En | MEDLINE | ID: mdl-31697791

Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes persistent arthritis in a subset of human patients. We report the isolation and functional characterization of monoclonal antibodies (mAbs) from two patients infected with CHIKV in the Dominican Republic. Single B cell sorting yielded a panel of 46 human mAbs of diverse germline lineages that targeted epitopes within the E1 or E2 glycoproteins. MAbs that recognized either E1 or E2 proteins exhibited neutralizing activity. Viral escape mutations localized the binding epitopes for two E1 mAbs to sites within domain I or the linker between domains I and III; and for two E2 mAbs between the ß-connector region and the B-domain. Two of the E2-specific mAbs conferred protection in vivo in a stringent lethal challenge mouse model of CHIKV infection, whereas the E1 mAbs did not. These results provide insight into human antibody response to CHIKV and identify candidate mAbs for therapeutic intervention.


Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Chikungunya Fever/immunology , Chikungunya virus/immunology , Epitopes/immunology , Glycoproteins/immunology , Viral Envelope Proteins/immunology , Adult , Animals , Antibodies, Neutralizing/immunology , Chikungunya Fever/virology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR
9.
Nature ; 563(7732): 559-563, 2018 11.
Article En | MEDLINE | ID: mdl-30464266

The zoonotic transmission of hantaviruses from their rodent hosts to humans in North and South America is associated with a severe and frequently fatal respiratory disease, hantavirus pulmonary syndrome (HPS)1,2. No specific antiviral treatments for HPS are available, and no molecular determinants of in vivo susceptibility to hantavirus infection and HPS are known. Here we identify the human asthma-associated gene protocadherin-1 (PCDH1)3-6 as an essential determinant of entry and infection in pulmonary endothelial cells by two hantaviruses that cause HPS, Andes virus (ANDV) and Sin Nombre virus (SNV). In vitro, we show that the surface glycoproteins of ANDV and SNV directly recognize the outermost extracellular repeat domain of PCDH1-a member of the cadherin superfamily7,8-to exploit PCDH1 for entry. In vivo, genetic ablation of PCDH1 renders Syrian golden hamsters highly resistant to a usually lethal ANDV challenge. Targeting PCDH1 could provide strategies to reduce infection and disease caused by New World hantaviruses.


Cadherins/metabolism , Orthohantavirus/physiology , Virus Internalization , Animals , Cadherins/chemistry , Cadherins/deficiency , Cadherins/genetics , Endothelial Cells/virology , Female , Orthohantavirus/pathogenicity , Hantavirus Pulmonary Syndrome/virology , Haploidy , Host-Pathogen Interactions/genetics , Humans , Lung/cytology , Male , Mesocricetus/virology , Protein Domains , Protocadherins , Sin Nombre virus/pathogenicity , Sin Nombre virus/physiology
10.
J Biol Chem ; 293(16): 6201-6211, 2018 04 20.
Article En | MEDLINE | ID: mdl-29500195

Filoviruses (family Filoviridae) include five ebolaviruses and Marburg virus. These pathogens cause a rapidly progressing and severe viral disease with high mortality rates (generally 30-90%). Outbreaks of filovirus disease are sporadic and, until recently, were limited to less than 500 cases. However, the 2013-2016 epidemic in western Africa, caused by Ebola virus (EBOV), illustrated the potential of filovirus outbreaks to escalate to a much larger scale (over 28,000 suspected cases). mAbs against the envelope glycoprotein represent a promising therapeutic platform for managing filovirus infections. However, mAbs that exhibit neutralization or protective properties against multiple filoviruses are rare. Here we examined a panel of engineered bi- and trispecific antibodies, in which variable domains of mAbs that target epitopes from multiple filoviruses were combined, for their capacity to neutralize viral infection across filovirus species. We found that bispecific combinations targeting EBOV and Sudan virus (another ebolavirus), provide potent cross-neutralization and protection in mice. Furthermore, trispecific combinations, targeting EBOV, Sudan virus, and Marburg virus, exhibited strong neutralization potential against all three viruses. These results provide important insights into multispecific antibody engineering against filoviruses and will inform future immunotherapeutic discoveries.


Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Glycoproteins/immunology , Marburgvirus/immunology , Protein Engineering , Viral Proteins/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/genetics , Epitopes/immunology , Female , Hemorrhagic Fever, Ebola/immunology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/genetics
11.
Immunol Lett ; 190: 289-295, 2017 10.
Article En | MEDLINE | ID: mdl-28890093

The Sudan virus (SUDV), an ebolavirus, causes severe hemorrhagic fever with human case fatality rates of ∼50%. Previous work from our lab demonstrated the synthetic antibody F4 potently inhibits viral entry and protects against lethal virus challenge in mice [Chen et al., ACS Chem. Biol., 2014, 9, 2263-2273]. Here, we explore mechanistic requirements as well as contribution of the Fc region and function on neutralization and in vivo protection. Live cell imaging demonstrates that the antibody colocalizes with vesicular stomatitis virus particles containing the Sudan virus glycoprotein (VSV-GPSUDV) and that the antibody is rapidly degraded within cellular endosomes. A viral escape mutant contained substitutions on the N-heptad repeat (NHR) segment of GP2, the fusion subunit. Truncation studies indicated that the size of the Fc impacts virus neutralization potential. Finally, we examined the protective efficacy of Fc-null mutants in mice, and found that Fc function was not required for high levels of protection. Altogether, these results indicate that neutralization of SUDV GP-mediated cell entry likely involves blockade of viral membrane fusion within endosomes, and that inhibition of viral entry is the likely mechanism of in vivo protection.


Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Immunoglobulin Fc Fragments/genetics , Animals , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , HEK293 Cells , Humans , Membrane Fusion , Mice , Mice, Knockout , Mutation/genetics , Viral Envelope Proteins/immunology , Virus Internalization
12.
Cell ; 169(5): 878-890.e15, 2017 May 18.
Article En | MEDLINE | ID: mdl-28525755

Experimental monoclonal antibody (mAb) therapies have shown promise for treatment of lethal Ebola virus (EBOV) infections, but their species-specific recognition of the viral glycoprotein (GP) has limited their use against other divergent ebolaviruses associated with human disease. Here, we mined the human immune response to natural EBOV infection and identified mAbs with exceptionally potent pan-ebolavirus neutralizing activity and protective efficacy against three virulent ebolaviruses. These mAbs recognize an inter-protomer epitope in the GP fusion loop, a critical and conserved element of the viral membrane fusion machinery, and neutralize viral entry by targeting a proteolytically primed, fusion-competent GP intermediate (GPCL) generated in host cell endosomes. Only a few somatic hypermutations are required for broad antiviral activity, and germline-approximating variants display enhanced GPCL recognition, suggesting that such antibodies could be elicited more efficiently with suitably optimized GP immunogens. Our findings inform the development of both broadly effective immunotherapeutics and vaccines against filoviruses.


Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Ebola Vaccines/immunology , Hemorrhagic Fever, Ebola/immunology , Survivors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Chlorocebus aethiops , Cross Reactions , Ebolavirus/classification , Ebolavirus/immunology , Female , Ferrets , Hemorrhagic Fever, Ebola/virology , Humans , Kinetics , Mice , Mice, Inbred BALB C , Models, Molecular , Sequence Alignment , Vero Cells
13.
Hum Vaccin Immunother ; 13(4): 836-842, 2017 04 03.
Article En | MEDLINE | ID: mdl-27786606

Bispecific antibody engineering, in which binding specificities toward 2 distinct epitopes are combined into a single molecule, can greatly enhance immunotherapeutic properties of monoclonal antibodies. While the bispecific antibody approach has been applied widely to targets for indications such as cancer and inflammation, the development of such agents for viral immunotherapy is only now emerging. Here, we review recent advances in the development of bispecific antibodies for viral immunotherapy, highlighting promising in vitro and in vivo results.


Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Immunologic Factors/immunology , Immunotherapy/methods , Virus Diseases/therapy , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Drug Discovery/trends , Humans , Immunologic Factors/administration & dosage , Mice
14.
Science ; 354(6310): 350-354, 2016 10 21.
Article En | MEDLINE | ID: mdl-27608667

There is an urgent need for monoclonal antibody (mAb) therapies that broadly protect against Ebola virus and other filoviruses. The conserved, essential interaction between the filovirus glycoprotein, GP, and its entry receptor Niemann-Pick C1 (NPC1) provides an attractive target for such mAbs but is shielded by multiple mechanisms, including physical sequestration in late endosomes. Here, we describe a bispecific-antibody strategy to target this interaction, in which mAbs specific for NPC1 or the GP receptor-binding site are coupled to a mAb against a conserved, surface-exposed GP epitope. Bispecific antibodies, but not parent mAbs, neutralized all known ebolaviruses by coopting viral particles themselves for endosomal delivery and conferred postexposure protection against multiple ebolaviruses in mice. Such "Trojan horse" bispecific antibodies have potential as broad antifilovirus immunotherapeutics.


Antibodies, Bispecific/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Carrier Proteins/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Membrane Glycoproteins/immunology , Receptors, Virus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Binding Sites/immunology , Cell Line, Tumor , Endosomes/virology , Hemorrhagic Fever, Ebola/therapy , Humans , Immunotherapy/methods , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Niemann-Pick C1 Protein , Virus Internalization
15.
Cell Rep ; 15(7): 1514-1526, 2016 05 17.
Article En | MEDLINE | ID: mdl-27160900

Previous efforts to identify cross-neutralizing antibodies to the receptor-binding site (RBS) of ebolavirus glycoproteins have been unsuccessful, largely because the RBS is occluded on the viral surface. We report a monoclonal antibody (FVM04) that targets a uniquely exposed epitope within the RBS; cross-neutralizes Ebola (EBOV), Sudan (SUDV), and, to a lesser extent, Bundibugyo viruses; and shows protection against EBOV and SUDV in mice and guinea pigs. The antibody cocktail ZMapp™ is remarkably effective against EBOV (Zaire) but does not cross-neutralize other ebolaviruses. By replacing one of the ZMapp™ components with FVM04, we retained the anti-EBOV efficacy while extending the breadth of protection to SUDV, thereby generating a cross-protective antibody cocktail. In addition, we report several mutations at the base of the ebolavirus glycoprotein that enhance the binding of FVM04 and other cross-reactive antibodies. These findings have important implications for pan-ebolavirus vaccine development and defining broadly protective antibody cocktails.


Antibodies, Monoclonal/therapeutic use , Ebolavirus/physiology , Epitopes/immunology , Glycoproteins/metabolism , Hemorrhagic Fever, Ebola/immunology , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/ultrastructure , Antibodies, Neutralizing , Antibodies, Viral/chemistry , Binding Sites , Disease Models, Animal , Female , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/ultrastructure , Guinea Pigs , HEK293 Cells , Humans , Kinetics , Mice, Inbred BALB C , Models, Molecular , Mutation/genetics , Negative Staining , Neutralization Tests , Treatment Outcome
16.
Sci Rep ; 6: 19193, 2016 Jan 13.
Article En | MEDLINE | ID: mdl-26758505

Filoviruses (Ebola and Marburg) cause severe hemorrhagic fever. There are five species of ebolavirus; among these, the Ebola (Zaire) and Sudan viruses (EBOV and SUDV, respectively) are highly pathogenic and have both caused recurring, large outbreaks. However, the EBOV and SUDV glycoprotein (GP) sequences are 45% divergent and thus antigenically distinct. Few antibodies with cross-neutralizing properties have been described to date. We used antibody engineering to develop novel bispecific antibodies (Bis-mAbs) that are cross-reactive toward base epitopes on GP from EBOV and SUDV. These Bis-mAbs exhibit potent neutralization against EBOV and SUDV GP pseudotyped viruses as well as authentic pathogens, and confer a high degree (in one case 100%) post-exposure protection of mice from both viruses. Our studies show that a single agent that targets the GP base epitopes is sufficient for protection in mice; such agents could be included in panfilovirus therapeutic antibody cocktails.


Antibodies, Bispecific/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Post-Exposure Prophylaxis , Animals , Antibodies, Bispecific/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibody Affinity/immunology , Cell Line , Cross Reactions , Disease Models, Animal , Ebolavirus/classification , Humans , Mice , Neutralization Tests , Protein Binding , Protein Engineering
17.
J Virol ; 90(1): 266-78, 2016 01 01.
Article En | MEDLINE | ID: mdl-26468533

UNLABELLED: The unprecedented 2014-2015 Ebola virus disease (EVD) outbreak in West Africa has highlighted the need for effective therapeutics against filoviruses. Monoclonal antibody (MAb) cocktails have shown great potential as EVD therapeutics; however, the existing protective MAbs are virus species specific. Here we report the development of pan-ebolavirus and pan-filovirus antibodies generated by repeated immunization of mice with filovirus glycoproteins engineered to drive the B cell responses toward conserved epitopes. Multiple pan-ebolavirus antibodies were identified that react to the Ebola, Sudan, Bundibugyo, and Reston viruses. A pan-filovirus antibody that was reactive to the receptor binding regions of all filovirus glycoproteins was also identified. Significant postexposure efficacy of several MAbs, including a novel antibody cocktail, was demonstrated. For the first time, we report cross-neutralization and in vivo protection against two highly divergent filovirus species, i.e., Ebola virus and Sudan virus, with a single antibody. Competition studies indicate that this antibody targets a previously unrecognized conserved neutralizing epitope that involves the glycan cap. Mechanistic studies indicated that, besides neutralization, innate immune cell effector functions may play a role in the antiviral activity of the antibodies. Our findings further suggest critical novel epitopes that can be utilized to design effective cocktails for broad protection against multiple filovirus species. IMPORTANCE: Filoviruses represent a major public health threat in Africa and an emerging global concern. Largely driven by the U.S. biodefense funding programs and reinforced by the 2014 outbreaks, current immunotherapeutics are primarily focused on a single filovirus species called Ebola virus (EBOV) (formerly Zaire Ebola virus). However, other filoviruses including Sudan, Bundibugyo, and Marburg viruses have caused human outbreaks with mortality rates as high as 90%. Thus, cross-protective immunotherapeutics are urgently needed. Here, we describe monoclonal antibodies with cross-reactivity to several filoviruses, including the first report of a cross-neutralizing antibody that exhibits protection against Ebola virus and Sudan virus in mice. Our results further describe a novel combination of antibodies with enhanced protective efficacy. These results form a basis for further development of effective immunotherapeutics against filoviruses for human use. Understanding the cross-protective epitopes are also important for rational design of pan-ebolavirus and pan-filovirus vaccines.


Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Filoviridae/immunology , Hemorrhagic Fever, Ebola/prevention & control , Immunization, Passive , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/administration & dosage , Cross Protection , Disease Models, Animal , Epitopes/immunology , Female , Mice, Inbred BALB C , Treatment Outcome
18.
ACS Infect Dis ; 1(1): 42-52, 2015 Jan 09.
Article En | MEDLINE | ID: mdl-25984565

The Ebolaviruses are members of the family Filoviridae ("filoviruses") and cause severe hemhorragic fever with human case fatality rates as high as 90%. Infection requires attachment of the viral particle to cells and triggering of membrane fusion between the host and viral membranes, a process that occurs in the host endosome and is facilitated by the envelope glycoprotein (GP). One potential strategy for therapeutic intervention is the development of agents (antibodies, peptides, and small molecules) that can interfere with viral entry aspects such as attachment, uptake, priming, or membrane fusion. This paper highlights recent developments in the discovery and evaluation of therapeutic entry inhibitors and identifies opportunities moving forward.

19.
J Chem Inf Model ; 55(3): 495-500, 2015 Mar 23.
Article En | MEDLINE | ID: mdl-25648076

Involved in numerous key biological functions, protein helix-helix interactions follow a well-defined intermolecular recognition pattern. The characteristic structure of the α-helical coiled-coil allows for the specific randomization of clearly defined interaction partners within heteromeric systems. In this work, a rationally designed heterodimeric coiled-coil was used to investigate potential factors influencing the sequence selectivity in interhelical interactions.


Bacteriophages/metabolism , Peptide Library , Proteins/chemistry , Circular Dichroism , Models, Molecular , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Proteins/metabolism , Tyrosine/chemistry
20.
ACS Med Chem Lett ; 5(12): 1300-3, 2014 Dec 11.
Article En | MEDLINE | ID: mdl-25516788

Since peptides are vital for cellular and pathogenic processes, much effort has been put into the design of unnatural oligomers that mimic natural peptide structures, also referred to as foldamers. However, to enable the specific application of foldamers, a thorough characterization of their interaction profiles in native protein environments is required. We report here the application of phage display for the identification of suitable helical environments for a sequence comprising an alternating set of ß- and γ-amino acids. In vitro selected sequences show that an increase in the hydrophobic surface area at the helical interface as well as the incorporation of a polar H-bond donor functionality can significantly improve interhelical interactions involving backbone-extended amino acids. Thus, our data provide insight into the principles of the rational design of foldameric inhibitors for protein-protein interactions.

...