Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell ; 166(3): 596-608, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27453466

ABSTRACT

Influenza virus remains a threat because of its ability to evade vaccine-induced immune responses due to antigenic drift. Here, we describe the isolation, evolution, and structure of a broad-spectrum human monoclonal antibody (mAb), MEDI8852, effectively reacting with all influenza A hemagglutinin (HA) subtypes. MEDI8852 uses the heavy-chain VH6-1 gene and has higher potency and breadth when compared to other anti-stem antibodies. MEDI8852 is effective in mice and ferrets with a therapeutic window superior to that of oseltamivir. Crystallographic analysis of Fab alone or in complex with H5 or H7 HA proteins reveals that MEDI8852 binds through a coordinated movement of CDRs to a highly conserved epitope encompassing a hydrophobic groove in the fusion domain and a large portion of the fusion peptide, distinguishing it from other structurally characterized cross-reactive antibodies. The unprecedented breadth and potency of neutralization by MEDI8852 support its development as immunotherapy for influenza virus-infected humans.


Subject(s)
Alphainfluenzavirus/immunology , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Specificity , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/chemistry , Antibodies, Viral/isolation & purification , Binding Sites, Antibody , Crystallography, X-Ray , Epitopes/immunology , Ferrets , Humans , Influenza Vaccines , Mice , Orthomyxoviridae Infections/prevention & control , Protein Conformation
2.
Science ; 353(6301): 823-6, 2016 Aug 19.
Article in English | MEDLINE | ID: mdl-27417494

ABSTRACT

Zika virus (ZIKV), a mosquito-borne flavivirus with homology to Dengue virus (DENV), has become a public health emergency. By characterizing memory lymphocytes from ZIKV-infected patients, we dissected ZIKV-specific and DENV-cross-reactive immune responses. Antibodies to nonstructural protein 1 (NS1) were largely ZIKV-specific and were used to develop a serological diagnostic tool. In contrast, antibodies against E protein domain I/II (EDI/II) were cross-reactive and, although poorly neutralizing, potently enhanced ZIKV and DENV infection in vitro and lethally enhanced DENV disease in mice. Memory T cells against NS1 or E proteins were poorly cross-reactive, even in donors preexposed to DENV. The most potent neutralizing antibodies were ZIKV-specific and targeted EDIII or quaternary epitopes on infectious virus. An EDIII-specific antibody protected mice from lethal ZIKV infection, illustrating the potential for antibody-based therapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/therapeutic use , Antibody Specificity , Cross Reactions , Dengue Virus/immunology , Disease Models, Animal , Humans , Immunodominant Epitopes/immunology , Immunologic Memory , Protein Structure, Tertiary , T-Lymphocytes/immunology , Viral Envelope Proteins/immunology , Viral Nonstructural Proteins/immunology , Zika Virus Infection/prevention & control , Zika Virus Infection/therapy
3.
EMBO Mol Med ; 8(4): 407-21, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26992832

ABSTRACT

Currently available rabies post-exposure prophylaxis (PEP) for use in humans includes equine or human rabies immunoglobulins (RIG). The replacement of RIG with an equally or more potent and safer product is strongly encouraged due to the high costs and limited availability of existing RIG. In this study, we identified two broadly neutralizing human monoclonal antibodies that represent a valid and affordable alternative to RIG in rabies PEP. Memory B cells from four selected vaccinated donors were immortalized and monoclonal antibodies were tested for neutralizing activity and epitope specificity. Two antibodies, identified as RVC20 and RVC58 (binding to antigenic site I and III, respectively), were selected for their potency and broad-spectrum reactivity. In vitro, RVC20 and RVC58 were able to neutralize all 35 rabies virus (RABV) and 25 non-RABV lyssaviruses. They showed higher potency and breath compared to antibodies under clinical development (namely CR57, CR4098, and RAB1) and commercially available human RIG. In vivo, the RVC20-RVC58 cocktail protected Syrian hamsters from a lethal RABV challenge and did not affect the endogenous hamster post-vaccination antibody response.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Immunologic Factors/immunology , Post-Exposure Prophylaxis/methods , Rabies virus/immunology , Rabies/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/administration & dosage , Antibodies, Viral/isolation & purification , Disease Models, Animal , Humans , Immunization, Passive/methods , Immunologic Factors/administration & dosage , Immunologic Factors/isolation & purification , Mesocricetus , Survival Analysis , Treatment Outcome
4.
Science ; 351(6279): 1339-42, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26917593

ABSTRACT

Ebola virus disease in humans is highly lethal, with case fatality rates ranging from 25 to 90%. There is no licensed treatment or vaccine against the virus, underscoring the need for efficacious countermeasures. We ascertained that a human survivor of the 1995 Kikwit Ebola virus disease outbreak maintained circulating antibodies against the Ebola virus surface glycoprotein for more than a decade after infection. From this survivor we isolated monoclonal antibodies (mAbs) that neutralize recent and previous outbreak variants of Ebola virus and mediate antibody-dependent cell-mediated cytotoxicity in vitro. Strikingly, monotherapy with mAb114 protected macaques when given as late as 5 days after challenge. Treatment with a single human mAb suggests that a simplified therapeutic strategy for human Ebola infection may be possible.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adult , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Clinical Trials as Topic , Disease Outbreaks , Female , Hemorrhagic Fever, Ebola/epidemiology , Humans , Macaca , Male , Molecular Sequence Data , Survivors
5.
Proc Natl Acad Sci U S A ; 112(33): 10473-8, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26216974

ABSTRACT

Middle East Respiratory Syndrome (MERS) is a highly lethal pulmonary infection caused by a previously unidentified coronavirus (CoV), likely transmitted to humans by infected camels. There is no licensed vaccine or antiviral for MERS, therefore new prophylactic and therapeutic strategies to combat human infections are needed. In this study, we describe, for the first time, to our knowledge, the isolation of a potent MERS-CoV-neutralizing antibody from memory B cells of an infected individual. The antibody, named LCA60, binds to a novel site on the spike protein and potently neutralizes infection of multiple MERS-CoV isolates by interfering with the binding to the cellular receptor CD26. Importantly, using mice transduced with adenovirus expressing human CD26 and infected with MERS-CoV, we show that LCA60 can effectively protect in both prophylactic and postexposure settings. This antibody can be used for prophylaxis, for postexposure prophylaxis of individuals at risk, or for the treatment of human cases of MERS-CoV infection. The fact that it took only 4 mo from the initial screening of B cells derived from a convalescent patient for the development of a stable chinese hamster ovary (CHO) cell line producing neutralizing antibodies at more than 5 g/L provides an example of a rapid pathway toward the generation of effective antiviral therapies against emerging viruses.


Subject(s)
Antibodies, Monoclonal/immunology , Immunologic Memory , Middle East Respiratory Syndrome Coronavirus/drug effects , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Binding Sites , CHO Cells , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cricetinae , Cricetulus , Dipeptidyl Peptidase 4/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Molecular Conformation , Molecular Sequence Data , Mutation , Protein Binding , Sequence Homology, Amino Acid , Spike Glycoprotein, Coronavirus/chemistry , Viral Vaccines
6.
Nature ; 501(7467): 439-43, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23955151

ABSTRACT

Broadly neutralizing antibodies reactive against most and even all variants of the same viral species have been described for influenza and HIV-1 (ref. 1). However, whether a neutralizing antibody could have the breadth of range to target different viral species was unknown. Human respiratory syncytial virus (HRSV) and human metapneumovirus (HMPV) are common pathogens that cause severe disease in premature newborns, hospitalized children and immune-compromised patients, and play a role in asthma exacerbations. Although antisera generated against either HRSV or HMPV are not cross-neutralizing, we speculated that, because of the repeated exposure to these viruses, cross-neutralizing antibodies may be selected in some individuals. Here we describe a human monoclonal antibody (MPE8) that potently cross-neutralizes HRSV and HMPV as well as two animal paramyxoviruses: bovine RSV (BRSV) and pneumonia virus of mice (PVM). In its germline configuration, MPE8 is HRSV-specific and its breadth is achieved by somatic mutations in the light chain variable region. MPE8 did not result in the selection of viral escape mutants that evaded antibody targeting and showed potent prophylactic efficacy in animal models of HRSV and HMPV infection, as well as prophylactic and therapeutic efficacy in the more relevant model of lethal PVM infection. The core epitope of MPE8 was mapped on two highly conserved anti-parallel ß-strands on the pre-fusion viral F protein, which are rearranged in the post-fusion F protein conformation. Twenty-six out of the thirty HRSV-specific neutralizing antibodies isolated were also found to be specific for the pre-fusion F protein. Taken together, these results indicate that MPE8 might be used for the prophylaxis and therapy of severe HRSV and HMPV infections and identify the pre-fusion F protein as a candidate HRSV vaccine.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cross Reactions/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/virology , Paramyxoviridae/classification , Paramyxoviridae/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibody Specificity/immunology , Cattle , Epitopes/immunology , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Metapneumovirus/immunology , Mice , Models, Molecular , Molecular Sequence Data , Murine pneumonia virus/immunology , Paramyxoviridae Infections/prevention & control , Paramyxoviridae Infections/therapy , Pneumovirus Infections/immunology , Pneumovirus Infections/prevention & control , Pneumovirus Infections/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Bovine/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology
7.
J Clin Invest ; 122(10): 3781-90, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996451

ABSTRACT

Pemphigus vulgaris (PV) is an autoimmune blistering disease of skin and mucous membranes caused by autoantibodies to the desmoglein (DSG) family proteins DSG3 and DSG1, leading to loss of keratinocyte cell adhesion. To learn more about pathogenic PV autoantibodies, we isolated 15 IgG antibodies specific for DSG3 from 2 PV patients. Three antibodies disrupted keratinocyte monolayers in vitro, and 2 were pathogenic in a passive transfer model in neonatal mice. The epitopes recognized by the pathogenic antibodies were mapped to the DSG3 extracellular 1 (EC1) and EC2 subdomains, regions involved in cis-adhesive interactions. Using a site-specific serological assay, we found that the cis-adhesive interface on EC1 recognized by the pathogenic antibody PVA224 is the primary target of the autoantibodies present in the serum of PV patients. The autoantibodies isolated used different heavy- and light-chain variable region genes and carried high levels of somatic mutations in complementary-determining regions, consistent with antigenic selection. Remarkably, binding to DSG3 was lost when somatic mutations were reverted to the germline sequence. These findings identify the cis-adhesive interface of DSG3 as the immunodominant region targeted by pathogenic antibodies in PV and indicate that autoreactivity relies on somatic mutations generated in the response to an antigen unrelated to DSG3.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Desmoglein 3/immunology , Immunoglobulin G/immunology , Pemphigus/immunology , Somatic Hypermutation, Immunoglobulin , Amino Acid Sequence , Animals , Animals, Newborn , Antigen-Antibody Reactions , Autoantigens/chemistry , Cells, Cultured , Complementarity Determining Regions/immunology , Desmoglein 3/chemistry , Epitopes/chemistry , Epitopes/immunology , Humans , Immunization, Passive , Immunoglobulin Variable Region/genetics , Keratinocytes/immunology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
8.
Science ; 333(6044): 850-6, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21798894

ABSTRACT

The isolation of broadly neutralizing antibodies against influenza A viruses has been a long-sought goal for therapeutic approaches and vaccine design. Using a single-cell culture method for screening large numbers of human plasma cells, we isolated a neutralizing monoclonal antibody that recognized the hemagglutinin (HA) glycoprotein of all 16 subtypes and neutralized both group 1 and group 2 influenza A viruses. Passive transfer of this antibody conferred protection to mice and ferrets. Complexes with HAs from the group 1 H1 and the group 2 H3 subtypes analyzed by x-ray crystallography showed that the antibody bound to a conserved epitope in the F subdomain. This antibody may be used for passive protection and to inform vaccine design because of its broad specificity and neutralization potency.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A virus/immunology , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/isolation & purification , Antibody Specificity , Cells, Cultured , Cross Reactions , Crystallography, X-Ray , Epitopes/immunology , Ferrets , Glycosylation , Humans , Hydrophobic and Hydrophilic Interactions , Immunization, Passive , Immunoglobulin Variable Region/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza B virus/immunology , Influenza, Human/immunology , Mice , Models, Molecular , Molecular Sequence Data , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/therapy , Plasma Cells/immunology , Protein Multimerization , Protein Structure, Secondary
9.
PLoS One ; 6(3): e18207, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21483815

ABSTRACT

Neutralizing antibodies have been shown to protect macaques against SHIV challenge. However, genetically diverse HIV-1 clades have evolved, and a key question left unanswered is whether neutralizing antibodies can confer cross-clade protection in vivo. The novel human monoclonal antibody HGN194 was isolated from an individual infected with an HIV-1 clade AG recombinant circulating recombinant form (CRF). HGN194 targets an epitope in the third hypervariable loop (V3) of HIV-1 gp120 and neutralizes a range of relatively neutralization-sensitive and resistant viruses. We evaluated the potential of HGN194 to protect infant rhesus monkeys against a SHIV encoding a primary CCR5-tropic HIV-1 clade C envelope. After high-dose mucosal challenge, all untreated controls became highly viremic while all HGN194-treated animals (50 mg/kg) were completely protected. When HGN194 was given at 1 mg/kg, one out of two monkeys remained aviremic, whereas the other had delayed, lower peak viremia. Interestingly, all protected monkeys given high-dose HGN194 developed Gag-specific proliferative responses of both CD4+ and CD8+ T cells. To test whether generation of the latter involved cryptic infection, we ablated CD8+ cells after HGN194 clearance. No viremia was detected in any protected monkeys, thus ruling out virus reservoirs. Thus, induction of CD8 T-cell immunity may have resulted from transient "Hit and Run" infection or cross priming via Ag-Ab-mediated cross-presentation. Together, our data identified the HGN194 epitope as protective and provide proof-of-concept that this anti-V3 loop mAb can prevent infection with sterilizing immunity after challenge with virus of a different clade, implying that V3 is a potential vaccine target.


Subject(s)
Antibodies, Monoclonal/immunology , HIV Antibodies/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Lymphocytes/cytology , Lymphocytes/immunology , Macaca , Macaca mulatta , RNA, Viral/genetics
10.
PLoS Pathog ; 6(11): e1001195, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21124990

ABSTRACT

The human monoclonal antibody (mAb) HK20 neutralizes a broad spectrum of primary HIV-1 isolates by targeting the highly conserved heptad repeat 1 (HR1) of gp41, which is transiently exposed during HIV-1 entry. Here we present the crystal structure of the HK20 Fab in complex with a gp41 mimetic 5-Helix at 2.3 Å resolution. HK20 employs its heavy chain CDR H2 and H3 loops to bind into a conserved hydrophobic HR1 pocket that is occupied by HR2 residues in the gp41 post fusion conformation. Compared to the previously described HR1-specific mAb D5, HK20 approaches its epitope with a different angle which might favor epitope access and thus contribute to its higher neutralization breadth and potency. Comparison of the neutralization activities of HK20 IgG, Fab and scFv employing both single cycle and multiple cycle neutralization assays revealed much higher potencies for the smaller Fab and scFv over IgG, implying that the target site is difficult to access for complete antibodies. Nevertheless, two thirds of sera from HIV-1 infected individuals contain significant titers of HK20-inhibiting antibodies. The breadth of neutralization of primary isolates across all clades, the higher potencies for C-clade viruses and the targeting of a distinct site as compared to the fusion inhibitor T-20 demonstrate the potential of HK20 scFv as a therapeutic tool.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , HIV Antibodies/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/physiology , HIV-1/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Crystallization , Crystallography, X-Ray , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/metabolism , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mutation/genetics , Neutralization Tests , Protein Conformation , Surface Plasmon Resonance
11.
J Clin Invest ; 120(5): 1663-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20389023

ABSTRACT

The target of neutralizing antibodies that protect against influenza virus infection is the viral protein HA. Genetic and antigenic variation in HA has been used to classify influenza viruses into subtypes (H1-H16). The neutralizing antibody response to influenza virus is thought to be specific for a few antigenically related isolates within a given subtype. However, while heterosubtypic antibodies capable of neutralizing multiple influenza virus subtypes have been recently isolated from phage display libraries, it is not known whether such antibodies are produced in the course of an immune response to influenza virus infection or vaccine. Here we report that, following vaccination with seasonal influenza vaccine containing H1 and H3 influenza virus subtypes, some individuals produce antibodies that cross-react with H5 HA. By immortalizing IgG-expressing B cells from 4 individuals, we isolated 20 heterosubtypic mAbs that bound and neutralized viruses belonging to several HA subtypes (H1, H2, H5, H6, and H9), including the pandemic A/California/07/09 H1N1 isolate. The mAbs used different VH genes and carried a high frequency of somatic mutations. With the exception of a mAb that bound to the HA globular head, all heterosubtypic mAbs bound to acid-sensitive epitopes in the HA stem region. Four mAbs were evaluated in vivo and protected mice from challenge with influenza viruses representative of different subtypes. These findings reveal that seasonal influenza vaccination can induce polyclonal heterosubtypic neutralizing antibodies that cross-react with the swine-origin pandemic H1N1 influenza virus and with the highly pathogenic H5N1 virus.


Subject(s)
Antibodies, Neutralizing/chemistry , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Dogs , Epitope Mapping/methods , Female , Humans , Immunoglobulin G/metabolism , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred BALB C , Mutation , Orthomyxoviridae/metabolism , Protein Binding
12.
PLoS One ; 5(1): e8805, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20098712

ABSTRACT

BACKGROUND: The isolation of human monoclonal antibodies (mAbs) that neutralize a broad spectrum of primary HIV-1 isolates and the characterization of the human neutralizing antibody B cell response to HIV-1 infection are important goals that are central to the design of an effective antibody-based vaccine. METHODS AND FINDINGS: We immortalized IgG(+) memory B cells from individuals infected with diverse clades of HIV-1 and selected on the basis of plasma neutralization profiles that were cross-clade and relatively potent. Culture supernatants were screened using various recombinant forms of the envelope glycoproteins (Env) in multiple parallel assays. We isolated 58 mAbs that were mapped to different Env surfaces, most of which showed neutralizing activity. One mAb in particular (HJ16) specific for a novel epitope proximal to the CD4 binding site on gp120 selectively neutralized a multi-clade panel of Tier-2 HIV-1 pseudoviruses, and demonstrated reactivity that was comparable in breadth, but distinct in neutralization specificity, to that of the other CD4 binding site-specific neutralizing mAb b12. A second mAb (HGN194) bound a conserved epitope in the V3 crown and neutralized all Tier-1 and a proportion of Tier-2 pseudoviruses tested, irrespective of clade. A third mAb (HK20) with broad neutralizing activity, particularly as a Fab fragment, recognized a highly conserved epitope in the HR-1 region of gp41, but showed striking assay-dependent selectivity in its activity. CONCLUSIONS: This study reveals that by using appropriate screening methods, a large proportion of memory B cells can be isolated that produce mAbs with HIV-1 neutralizing activity. Three of these mAbs show unusual breadth of neutralization and therefore add to the current panel of HIV-1 neutralizing antibodies with potential for passive protection and template-based vaccine design.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , HIV Infections/immunology , Immunologic Memory , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Binding Sites, Antibody , CD4 Antigens/immunology , Epitopes/chemistry , HIV-1 , Humans , Molecular Sequence Data
13.
J Virol ; 84(2): 1005-13, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889756

ABSTRACT

Human cytomegalovirus (HCMV) is a widely circulating pathogen that causes severe disease in immunocompromised patients and infected fetuses. By immortalizing memory B cells from HCMV-immune donors, we isolated a panel of human monoclonal antibodies that neutralized at extremely low concentrations (90% inhibitory concentration [IC(90)] values ranging from 5 to 200 pM) HCMV infection of endothelial, epithelial, and myeloid cells. With the single exception of an antibody that bound to a conserved epitope in the UL128 gene product, all other antibodies bound to conformational epitopes that required expression of two or more proteins of the gH/gL/UL128-131A complex. Antibodies against gB, gH, or gM/gN were also isolated and, albeit less potent, were able to neutralize infection of both endothelial-epithelial cells and fibroblasts. This study describes unusually potent neutralizing antibodies against HCMV that might be used for passive immunotherapy and identifies, through the use of such antibodies, novel antigenic targets in HCMV for the design of immunogens capable of eliciting previously unknown neutralizing antibody responses.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Cytomegalovirus/immunology , Epitopes , Membrane Glycoproteins , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Cell Line , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Epitopes/chemistry , Epitopes/immunology , Female , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Molecular Sequence Data , Neutralization Tests , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...