Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Vaccine ; 39(32): 4423-4428, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34210573

ABSTRACT

A correlate of protection (CoP) is urgently needed to expedite development of additional COVID-19 vaccines to meet unprecedented global demand. To assess whether antibody titers may reasonably predict efficacy and serve as the basis of a CoP, we evaluated the relationship between efficacy and in vitro neutralizing and binding antibodies of 7 vaccines for which sufficient data have been generated. Once calibrated to titers of human convalescent sera reported in each study, a robust correlation was seen between neutralizing titer and efficacy (ρ = 0.79) and binding antibody titer and efficacy (ρ = 0.93), despite geographically diverse study populations subject to different forces of infection and circulating variants, and use of different endpoints, assays, convalescent sera panels and manufacturing platforms. Together with evidence from natural history studies and animal models, these results support the use of post-immunization antibody titers as the basis for establishing a correlate of protection for COVID-19 vaccines.


Subject(s)
Antibodies, Neutralizing , COVID-19 , Animals , Antibodies, Viral , COVID-19/therapy , COVID-19 Vaccines , Humans , Immunization, Passive , SARS-CoV-2 , COVID-19 Serotherapy
2.
Vaccine ; 38(31): 4783-4791, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32507409

ABSTRACT

A novel coronavirus (CoV), Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in late 2019 in Wuhan, China and has since spread as a global pandemic. Safe and effective vaccines are thus urgently needed to reduce the significant morbidity and mortality of Coronavirus Disease 2019 (COVID-19) disease and ease the major economic impact. There has been an unprecedented rapid response by vaccine developers with now over one hundred vaccine candidates in development and at least six having reached clinical trials. However, a major challenge during rapid development is to avoid safety issues both by thoughtful vaccine design and by thorough evaluation in a timely manner. A syndrome of "disease enhancement" has been reported in the past for a few viral vaccines where those immunized suffered increased severity or death when they later encountered the virus or were found to have an increased frequency of infection. Animal models allowed scientists to determine the underlying mechanism for the former in the case of Respiratory syncytial virus (RSV) vaccine and have been utilized to design and screen new RSV vaccine candidates. Because some Middle East respiratory syndrome (MERS) and SARS-CoV-1 vaccines have shown evidence of disease enhancement in some animal models, this is a particular concern for SARS-CoV-2 vaccines. To address this challenge, the Coalition for Epidemic Preparedness Innovations (CEPI) and the Brighton Collaboration (BC) Safety Platform for Emergency vACcines (SPEAC) convened a scientific working meeting on March 12 and 13, 2020 of experts in the field of vaccine immunology and coronaviruses to consider what vaccine designs could reduce safety concerns and how animal models and immunological assessments in early clinical trials can help to assess the risk. This report summarizes the evidence presented and provides considerations for safety assessment of COVID-19 vaccine candidates in accelerated vaccine development.


Subject(s)
Antibodies, Viral/adverse effects , Antibodies, Viral/immunology , Betacoronavirus/immunology , Coronavirus Infections/immunology , Pneumonia, Viral/immunology , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Animals , Betacoronavirus/pathogenicity , COVID-19 , COVID-19 Vaccines , Clinical Trials as Topic , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Disease Models, Animal , Humans , Pandemics , Pneumonia, Viral/virology , Risk Assessment , SARS-CoV-2 , Severe Acute Respiratory Syndrome/immunology
3.
Curr Opin HIV AIDS ; 10(3): 129-34, 2015 May.
Article in English | MEDLINE | ID: mdl-25760933

ABSTRACT

PURPOSE OF REVIEW: We describe the history of passive immunization to provide context for the series of articles to follow. The history of passive immunization with antibodies to prevent or treat infectious diseases is a story of different eras. There was an extraordinary era of discovery and clinical implementation before the chemical nature of antibodies was even known. This empirical process provided the resources and reagents used to describe and characterize humoral immunity, better define the chemical properties and structure of antibodies, and extend the clinical use of immunoglobulin products to treat or prevent multiple viral and bacterial diseases over the ensuing several decades. The next distinct era came with the discovery of processes to produce monoclonal antibodies (mAbs), and development of more specific therapies. Interestingly, mAb technology resulted in many products to treat autoimmune and allergic diseases, but only one common infectious disease, respiratory syncytial virus, and only in a restricted population of high-risk infants. RECENT FINDINGS: The current era began in 2003 with a series of publications demonstrating processes for rapidly producing human mAbs. SUMMARY: This technology combined with new sequencing technology, advances in structural biology, atomic-level molecular design, and increased capacity for synthetic biology, promises new opportunities to apply passive immunization to the prevention and treatment of infectious diseases.


Subject(s)
Antibodies, Monoclonal , Communicable Diseases , Immunization, Passive , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Communicable Diseases/drug therapy , Communicable Diseases/immunology , Humans
4.
PLoS One ; 9(6): e100325, 2014.
Article in English | MEDLINE | ID: mdl-24956119

ABSTRACT

Chronic hepatitis C virus (HCV) infection is the most common cause of end-stage liver disease, often leading to liver transplantation, in which case circulating virions typically infect the transplanted liver within hours and viral concentrations can quickly exceed pre-transplant levels. MBL-HCV1 is a fully human monoclonal antibody recognizing a linear epitope of the HCV E2 envelope glycoprotein (amino acids 412-423). The ability of MBL-HCV1 to prevent HCV recurrence after liver transplantation was investigated in a phase 2 randomized clinical trial evaluating six MBL-HCV1-treated subjects and five placebo-treated subjects. MBL-HCV1 treatment significantly delayed time to viral rebound compared with placebo treatment. Here we report results from high-throughput sequencing on the serum of each of the eleven enrolled subjects prior to liver transplantation and after viral rebound. We further sequenced the sera of the MBL-HCV1-treated subjects at various interim time points to study the evolution of antibody-resistant viral variants. We detected mutations at one of two positions within the antibody epitope--mutations of N at position 415 to D, K or S, or mutation of N at position 417 to S. It has been previously reported that N415 is not glycosylated in the wild-type E2 protein, but N417S can lead to glycosylation at position 415. Thus N415 is a key position for antibody recognition and the only routes we identified for viral escape, within the constraints of HCV fitness in vivo, involve mutating or glycosylating this position. Evaluation of mutations along the entire E1 and E2 proteins revealed additional positions that changed moderately before and after MBL-HCV1 treatment for subsets of the six subjects, yet underscored the relative importance of position 415 in MBL-HCV1 resistance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Biological Evolution , Biomarkers/metabolism , Hepatitis C, Chronic/therapy , Liver Transplantation , Viral Envelope Proteins/immunology , Amino Acid Sequence , Antibodies, Viral/immunology , Double-Blind Method , Follow-Up Studies , Glycosylation , Hepatitis C, Chronic/immunology , High-Throughput Nucleotide Sequencing/methods , Humans , Molecular Sequence Data , Prognosis , RNA, Viral/blood , RNA, Viral/genetics , Recurrence , Sequence Homology, Amino Acid , Viral Envelope Proteins/antagonists & inhibitors
5.
PLoS One ; 8(4): e61210, 2013.
Article in English | MEDLINE | ID: mdl-23613814

ABSTRACT

Mutations in the gene encoding human SOD1 (hSOD1) can cause amyotrophic lateral sclerosis (ALS) yet the mechanism by which mutant SOD1 can induce ALS is not fully understood. There is currently no cure for ALS or treatment that significantly reduces symptoms or progression. To develop tools to understand the protein conformations present in mutant SOD1-induced ALS and as possible immunotherapy, we isolated and characterized eleven unique human monoclonal antibodies specific for hSOD1. Among these, five recognized distinct linear epitopes on hSOD1 that were not available in the properly-folded protein but were available on forms of protein with some degree of misfolding. The other six antibodies recognized conformation-dependent epitopes that were present in the properly-folded protein with two different recognition profiles: three could bind hSOD1 dimer or monomer and the other three were specific for hSOD1 dimer only. Antibodies with the capacity to bind hSOD1 monomer were able to prevent increased hydrophobicity when mutant hSOD1 was exposed to increased temperature and EDTA, suggesting that the antibodies stabilized the native structure of hSOD1. Two antibodies were tested in a G93A mutant hSOD1 transgenic mouse model of ALS but did not yield a statistically significant increase in overall survival. It may be that the two antibodies selected for testing in the mouse model were not effective for therapy or that the model and/or route of administration were not optimal to produce a therapeutic effect. Therefore, additional testing will be required to determine therapeutic potential for SOD1 mutant ALS and potentially some subset of sporadic ALS.


Subject(s)
Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Epitopes/immunology , Superoxide Dismutase/immunology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/enzymology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Epitopes/chemistry , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Immunoprecipitation , Mice , Mice, Transgenic , Mutant Proteins/metabolism , Protein Binding , Protein Conformation , Superoxide Dismutase/chemistry
6.
PLoS Pathog ; 8(8): e1002895, 2012.
Article in English | MEDLINE | ID: mdl-22952447

ABSTRACT

Hepatitis C virus (HCV) infection is a leading cause of liver transplantation and there is an urgent need to develop therapies to reduce rates of HCV infection of transplanted livers. Approved therapeutics for HCV are poorly tolerated and are of limited efficacy in this patient population. Human monoclonal antibody HCV1 recognizes a highly-conserved linear epitope of the HCV E2 envelope glycoprotein (amino acids 412-423) and neutralizes a broad range of HCV genotypes. In a chimpanzee model, a single dose of 250 mg/kg HCV1 delivered 30 minutes prior to infusion with genotype 1a H77 HCV provided complete protection from HCV infection, whereas a dose of 50 mg/kg HCV1 did not protect. In addition, an acutely-infected chimpanzee given 250 mg/kg HCV1 42 days following exposure to virus had a rapid reduction in viral load to below the limit of detection before rebounding 14 days later. The emergent virus displayed an E2 mutation (N415K/D) conferring resistance to HCV1 neutralization. Finally, three chronically HCV-infected chimpanzees were treated with a single dose of 40 mg/kg HCV1 and viral load was reduced to below the limit of detection for 21 days in one chimpanzee with rebounding virus displaying a resistance mutation (N417S). The other two chimpanzees had 0.5-1.0 log(10) reductions in viral load without evidence of viral resistance to HCV1. In vitro testing using HCV pseudovirus (HCVpp) demonstrated that the sera from the poorly-responding chimpanzees inhibited the ability of HCV1 to neutralize HCVpp. Measurement of antibody responses in the chronically-infected chimpanzees implicated endogenous antibody to E2 and interference with HCV1 neutralization although other factors may also be responsible. These data suggest that human monoclonal antibody HCV1 may be an effective therapeutic for the prevention of graft infection in HCV-infected patients undergoing liver transplantation.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hepacivirus/immunology , Hepatitis C Antibodies/therapeutic use , Hepatitis C, Chronic/therapy , Hepatitis C/prevention & control , Amino Acid Sequence , Animals , Cell Line , Disease Models, Animal , Hepatitis C/immunology , Hepatitis C/virology , Hepatitis C, Chronic/immunology , Humans , Liver Transplantation , Mutation , Neutralization Tests , Pan troglodytes , RNA, Viral/blood , Tetraspanin 28/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Load
7.
Antiviral Res ; 91(2): 187-94, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21693135

ABSTRACT

Replacement of polyclonal anti-rabies immunoglobulin (RIG) used in rabies post-exposure prophylaxis (PEP) with a monoclonal antibody will eliminate cost and availability constraints that currently exist using RIG in the developing world. The human monoclonal antibody RAB1 has been shown to neutralize all rabies street isolates tested; however for the laboratory-adapted fixed strain, CVS-11, mutation in the G glycoprotein of amino acid 336 from asparagine (N) to aspartic acid (D) resulted in resistance to neutralization. Interestingly, this same mutation in the G glycoprotein of a second laboratory-adapted fixed strain (ERA) did not confer resistance to RAB1 neutralization. Using cell surface staining and lentivirus pseudotyped with rabies virus G glycoprotein (RABVpp), we identified an amino acid alteration in CVS-11 (K346), not present in ERA (R346), which was required in combination with D336 to confer resistance to RAB1. A complete analysis of G glycoprotein sequences from GenBank demonstrated that no identified rabies isolates contain the necessary combination of G glycoprotein mutations for resistance to RAB1 neutralization, consistent with the broad neutralization of RAB1 observed in direct viral neutralization experiments with street isolates. All combinations of amino acids 336 and 346 reported in the sequence database were engineered into the ERA G glycoprotein and RAB1 was able to neutralize RABVpp bearing ERA G glycoprotein containing all known combinations at these critical residues. These data demonstrate that RAB1 has the capacity to neutralize all identified rabies isolates and a minimum of two distinct mutations in the G glycoprotein are required for abrogation of RAB1 neutralization.


Subject(s)
Amino Acid Substitution , Antibodies, Monoclonal/immunology , Antigens, Viral/immunology , Glycoproteins/immunology , Rabies virus/immunology , Viral Envelope Proteins/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/metabolism , Asparagine/metabolism , Binding Sites, Antibody , Cloning, Molecular , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Humans , Mutagenesis, Site-Directed/methods , Neutralization Tests , Point Mutation , Rabies virus/genetics , Rabies virus/metabolism , Sequence Analysis, Protein , Transfection , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
8.
N Engl J Med ; 362(3): 197-205, 2010 Jan 21.
Article in English | MEDLINE | ID: mdl-20089970

ABSTRACT

BACKGROUND: New therapies are needed to manage the increasing incidence, severity, and high rate of recurrence of Clostridium difficile infection. METHODS: We performed a randomized, double-blind, placebo-controlled study of two neutralizing, fully human monoclonal antibodies against C. difficile toxins A (CDA1) and B (CDB1). The antibodies were administered together as a single infusion, each at a dose of 10 mg per kilogram of body weight, in patients with symptomatic C. difficile infection who were receiving either metronidazole or vancomycin. The primary outcome was laboratory-documented recurrence of infection during the 84 days after the administration of monoclonal antibodies or placebo. RESULTS: Among the 200 patients who were enrolled (101 in the antibody group and 99 in the placebo group), the rate of recurrence of C. difficile infection was lower among patients treated with monoclonal antibodies (7% vs. 25%; 95% confidence interval, 7 to 29; P<0.001). The recurrence rates among patients with the epidemic BI/NAP1/027 strain were 8% for the antibody group and 32% for the placebo group (P=0.06); among patients with more than one previous episode of C. difficile infection, recurrence rates were 7% and 38%, respectively (P=0.006). The mean duration of the initial hospitalization for inpatients did not differ significantly between the antibody and placebo groups (9.5 and 9.4 days, respectively). At least one serious adverse event was reported by 18 patients in the antibody group and by 28 patients in the placebo group (P=0.09). CONCLUSIONS: The addition of monoclonal antibodies against C. difficile toxins to antibiotic agents significantly reduced the recurrence of C. difficile infection. (ClinicalTrials.gov number, NCT00350298.)


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antitoxins/therapeutic use , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Clostridioides difficile , Clostridium Infections/drug therapy , Enterotoxins/immunology , Adult , Aged , Aged, 80 and over , Antibodies/blood , Antibodies, Monoclonal/adverse effects , Antitoxins/adverse effects , Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Diarrhea/drug therapy , Diarrhea/microbiology , Double-Blind Method , Drug Therapy, Combination , Enterocolitis, Pseudomembranous/drug therapy , Enterotoxins/antagonists & inhibitors , Female , Humans , Male , Metronidazole/therapeutic use , Middle Aged , Secondary Prevention , Vancomycin/therapeutic use , Young Adult
9.
J Virol ; 83(23): 12473-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19759151

ABSTRACT

Nearly all livers transplanted into hepatitis C virus (HCV)-positive patients become infected with HCV, and 10 to 25% of reinfected livers develop cirrhosis within 5 years. Neutralizing monoclonal antibody could be an effective therapy for the prevention of infection in a transplant setting. To pursue this treatment modality, we developed human monoclonal antibodies (HuMAbs) directed against the HCV E2 envelope glycoprotein and assessed the capacity of these HuMAbs to neutralize a broad panel of HCV genotypes. HuMAb antibodies were generated by immunizing transgenic mice containing human antibody genes (HuMAb mice; Medarex Inc.) with soluble E2 envelope glycoprotein derived from a genotype 1a virus (H77). Two HuMAbs, HCV1 and 95-2, were selected for further study based on initial cross-reactivity with soluble E2 glycoproteins derived from genotypes 1a and 1b, as well as neutralization of lentivirus pseudotyped with HCV 1a and 1b envelope glycoproteins. Additionally, HuMAbs HCV1 and 95-2 potently neutralized pseudoviruses from all genotypes tested (1a, 1b, 2b, 3a, and 4a). Epitope mapping with mammalian and bacterially expressed proteins, as well as synthetic peptides, revealed that HuMAbs HCV1 and 95-2 recognize a highly conserved linear epitope spanning amino acids 412 to 423 of the E2 glycoprotein. The capacity to recognize and neutralize a broad range of genotypes, the highly conserved E2 epitope, and the fully human nature of the antibodies make HuMAbs HCV1 and 95-2 excellent candidates for treatment of HCV-positive individuals undergoing liver transplantation.


Subject(s)
Antibodies, Monoclonal/immunology , Hepatitis C Antibodies/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/isolation & purification , Conserved Sequence , Epitope Mapping , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Hepacivirus/immunology , Hepatitis C Antibodies/isolation & purification , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Neutralization Tests
10.
Clin Vaccine Immunol ; 14(3): 331-3, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17229882

ABSTRACT

Recombinant severe acute respiratory syndrome (SARS) nucleocapsid and spike protein-based immunoglobulin G immunoassays were developed and evaluated. Our assays demonstrated high sensitivity and specificity to the SARS coronavirus in sera collected from patients as late as 2 years postonset of symptoms. These assays will be useful not only for routine SARS coronavirus diagnostics but also for epidemiological and antibody kinetic studies.


Subject(s)
Antibodies, Viral/blood , Membrane Glycoproteins/immunology , Nucleocapsid Proteins/immunology , Severe Acute Respiratory Syndrome/diagnosis , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/immunology , Coronavirus Nucleocapsid Proteins , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/immunology , Sensitivity and Specificity , Serologic Tests , Spike Glycoprotein, Coronavirus
11.
Vaccine ; 25(15): 2800-10, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17240489

ABSTRACT

Rabies is a zoonosis that results in millions of human exposures worldwide each year. Human monoclonal antibodies (HuMAbs) that neutralize rabies virus may represent one viable strategy for post-exposure prophylaxis in humans, and have many advantages over current human or equine rabies immune globulin. Transgenic mice carrying human immunoglobulin genes were used to isolate human monoclonal antibodies that neutralized rabies virus. Several HuMAbs were identified that neutralized rabies virus variants from a broad panel of isolates of public health significance. HuMAb 17C7 was the most promising antibody identified because it neutralized all rabies virus isolates tested. HuMAb 17C7 recognizes a conformational epitope on the rabies virus glycoprotein which includes antigenic site III. HuMAb 17C7 protected hamsters from a lethal dose of rabies virus in a well-established in vivo model of post-exposure prophylaxis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Rabies virus/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Cell Line , Cricetinae , Glycoproteins/immunology , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Mephitidae , Mesocricetus , Mice , Mice, Transgenic , Rabies/immunology , Rabies/prevention & control , Rabies virus/genetics , Viral Proteins/immunology
12.
Infect Immun ; 74(11): 6339-47, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16966409

ABSTRACT

Clostridium difficile is the leading cause of nosocomial antibiotic-associated diarrhea, and recent outbreaks of strains with increased virulence underscore the importance of identifying novel approaches to treat and prevent relapse of Clostridium difficile-associated diarrhea (CDAD). CDAD pathology is induced by two exotoxins, toxin A and toxin B, which have been shown to be cytotoxic and, in the case of toxin A, enterotoxic. In this report we describe fully human monoclonal antibodies (HuMAbs) that neutralize these toxins and prevent disease in hamsters. Transgenic mice carrying human immunoglobulin genes were used to isolate HuMAbs that neutralize the cytotoxic effects of either toxin A or toxin B in cell-based in vitro neutralization assays. Three anti-toxin A HuMAbs (3H2, CDA1, and 1B11) could all inhibit the enterotoxicity of toxin A in mouse intestinal loops and the in vivo toxicity in a systemic mouse model. Four anti-toxin B HuMAbs (MDX-1388, 103-174, 1G10, and 2A11) could neutralize cytotoxicity in vitro, although systemic toxicity in the mouse could not be neutralized. Anti-toxin A HuMAb CDA1 and anti-toxin B HuMAb MDX-1388 were tested in the well-established hamster model of C. difficile disease. CDA1 alone resulted in a statistically significant reduction of mortality in hamsters; however, the combination treatment offered enhanced protection. Compared to controls, combination therapy reduced mortality from 100% to 45% (P<0.0001) in the primary disease hamster model and from 78% to 32% (P<0.0001) in the less stringent relapse model.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Clostridioides difficile/immunology , Enterocolitis, Pseudomembranous/mortality , Enterocolitis, Pseudomembranous/prevention & control , Enterotoxins/antagonists & inhibitors , Enterotoxins/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/isolation & purification , Bacterial Proteins/administration & dosage , Bacterial Toxins/administration & dosage , Cell Line , Cricetinae , Enterocolitis, Pseudomembranous/immunology , Enterotoxins/administration & dosage , Humans , Mice , Mice, Transgenic , Recurrence
13.
J Infect Dis ; 193(5): 685-92, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16453264

ABSTRACT

BACKGROUND: Immunotherapy with monoclonal antibodies (MAbs) offers safe interventions for the prevention of infection in patients after organ transplantation and for the treatment of cancers and autoimmune diseases. MAb 201 is a severe acute respiratory syndrome-associated coronavirus (SARS-CoV)-specific MAb that prevents establishment of viral replication in vitro and prevents viral replication in vivo when administered prophylactically. The efficacy of MAb 201 in the treatment of SARS was evaluated in golden Syrian hamsters, an animal model that supports SARS-CoV replication to high levels and displays severe pathological changes associated with infection, including pneumonitis and pulmonary consolidation. METHODS: Golden Syrian hamsters that were intranasally inoculated with SARS-CoV were treated with various doses of MAb 201 or an irrelevant MAb 24 h after inoculation. Two to 7 days after infection, the hamsters were killed, and their lungs were collected for evaluation of viral titers and pathological findings. RESULTS: Postexposure treatment with MAb 201 can alleviate the viral burden and associated pathological findings in a golden Syrian hamster model of SARS-CoV infection. After a hamster is treated with MAb 201, its viral burden is reduced by 102.4-103.9 50% tissue-culture infectious doses per gram of tissue, and the severity of associated pathological findings, including interstitial pneumonitis and consolidation, is also remarkably reduced. CONCLUSIONS: The demonstration of successful postexposure MAb 201 therapy in an animal model that demonstrates viral replication and associated pulmonary pathological findings suggests that MAb 201 may be useful in the arsenal of tools to combat SARS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Severe Acute Respiratory Syndrome/therapy , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Antibodies, Viral/blood , Cricetinae , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunotherapy , Lung/pathology , Lung/virology , Lung Diseases, Interstitial/pathology , Mesocricetus , Neutralization Tests , Severe acute respiratory syndrome-related coronavirus/physiology , Severe Acute Respiratory Syndrome/physiopathology , Severe Acute Respiratory Syndrome/virology , Viral Load
14.
J Virol Methods ; 128(1-2): 21-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15885812

ABSTRACT

Monoclonal antibodies (Mabs) against the Urbani strain of the SARS-associated coronavirus (SARS-CoV) were developed and characterized for reactivity to SARS-CoV and SARS-CoV S, N, M, and E proteins using enzyme-linked immunoabsorbent (ELISA), radioimmunoprecipitation, immunofluorescence, Western Blot and microneutralization assays. Twenty-six mAbs were reactive to SARS-CoV by ELISA, and nine were chosen for detailed characterization. Five mAbs reacted against the S protein, two against the M protein, and one each against the N and E proteins. Two of five S protein mAbs neutralized SARS-CoV infection of Vero E6 cells and reacted to an epitope within amino acids 490-510 in the S protein. While two of the three non-neutralizing antibodies recognized at second epitope within amino acids 270-350. The mAbs characterized should prove useful for developing SARS-CoV diagnostic assays and for studying the biology of infection and pathogenesis of disease.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antibody Specificity , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Structural Proteins/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Cell Line , Chlorocebus aethiops , Coronavirus M Proteins , Coronavirus Nucleocapsid Proteins , Epitope Mapping , Humans , Membrane Glycoproteins/immunology , Neutralization Tests , Nucleocapsid Proteins/immunology , Severe Acute Respiratory Syndrome/virology , Spike Glycoprotein, Coronavirus , Vero Cells , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Viroporin Proteins
15.
J Infect Dis ; 191(4): 507-14, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15655773

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome (SARS) remains a significant public health concern after the epidemic in 2003. Human monoclonal antibodies (MAbs) that neutralize SARS-associated coronavirus (SARS-CoV) could provide protection for exposed individuals. METHODS: Transgenic mice with human immunoglobulin genes were immunized with the recombinant major surface (S) glycoprotein ectodomain of SARS-CoV. Epitopes of 2 neutralizing MAbs derived from these mice were mapped and evaluated in a murine model of SARS-CoV infection. RESULTS: Both MAbs bound to S glycoprotein expressed on transfected cells but differed in their ability to block binding of S glycoprotein to Vero E6 cells. Immunoprecipitation analysis revealed 2 antibody-binding epitopes: one MAb (201) bound within the receptor-binding domain at aa 490-510, and the other MAb (68) bound externally to the domain at aa 130-150. Mice that received 40 mg/kg of either MAb prior to challenge with SARS-CoV were completely protected from virus replication in the lungs, and doses as low as 1.6 mg/kg offered significant protection. CONCLUSIONS: Two neutralizing epitopes were defined for MAbs to SARS-CoV S glycoprotein. Antibodies to both epitopes protected mice against SARS-CoV challenge. Clinical trials are planned to test MAb 201, a fully human MAb specific for the epitope within the receptor-binding region.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Immunization, Passive , Membrane Glycoproteins/immunology , Severe Acute Respiratory Syndrome/prevention & control , Severe acute respiratory syndrome-related coronavirus/immunology , Viral Envelope Proteins/immunology , Animals , Cells, Cultured , Disease Models, Animal , Epitope Mapping , Epitopes/immunology , Female , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutralization Tests , Protein Binding , Spike Glycoprotein, Coronavirus
16.
Proc Natl Acad Sci U S A ; 101(44): 15748-53, 2004 Nov 02.
Article in English | MEDLINE | ID: mdl-15496474

ABSTRACT

Angiotensin-converting enzyme 2 (ACE2) is a receptor for SARS-CoV, the novel coronavirus that causes severe acute respiratory syndrome [Li, W. Moore, M. J., Vasilieva, N., Sui, J., Wong, S. K., Berne, M. A., Somasundaran, M., Sullivan, J. L., Luzuriaga, K., Greenough, T. C., et al. (2003) Nature 426, 450-454]. We have identified a different human cellular glycoprotein that can serve as an alternative receptor for SARS-CoV. A human lung cDNA library in vesicular stomatitis virus G pseudotyped retrovirus was transduced into Chinese hamster ovary cells, and the cells were sorted for binding of soluble SARS-CoV spike (S) glycoproteins, S(590) and S(1180). Clones of transduced cells that bound SARS-CoV S glycoprotein were inoculated with SARS-CoV, and increases in subgenomic viral RNA from 1-16 h or more were detected by multiplex RT-PCR in four cloned cell lines. Sequencing of the human lung cDNA inserts showed that each of the cloned cell lines contained cDNA that encoded human CD209L, a C-type lectin (also called L-SIGN). When the cDNA encoding CD209L from clone 2.27 was cloned and transfected into Chinese hamster ovary cells, the cells expressed human CD209L glycoprotein and became susceptible to infection with SARS-CoV. Immunohistochemistry showed that CD209L is expressed in human lung in type II alveolar cells and endothelial cells, both potential targets for SARS-CoV. Several other enveloped viruses including Ebola and Sindbis also use CD209L as a portal of entry, and HIV and hepatitis C virus can bind to CD209L on cell membranes but do not use it to mediate virus entry. Our data suggest that the large S glycoprotein of SARS-CoV may use both ACE2 and CD209L in virus infection and pathogenesis.


Subject(s)
Cell Cycle Proteins/physiology , Membrane Proteins/physiology , Receptors, Virus/physiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Animals , Base Sequence , CHO Cells , Cell Cycle Proteins/genetics , Cell Line , Cricetinae , DNA, Complementary/genetics , Gene Library , Humans , Lung/metabolism , Lung/virology , Membrane Proteins/genetics , Receptors, Coronavirus , Receptors, Virus/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Transduction, Genetic
17.
J Virol ; 78(9): 4552-60, 2004 May.
Article in English | MEDLINE | ID: mdl-15078936

ABSTRACT

A novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV), has recently been identified as the causative agent of severe acute respiratory syndrome (SARS). SARS-CoV appears similar to other coronaviruses in both virion structure and genome organization. It is known for other coronaviruses that the spike (S) glycoprotein is required for both viral attachment to permissive cells and for fusion of the viral envelope with the host cell membrane. Here we describe the construction and expression of a soluble codon-optimized SARS-CoV S glycoprotein comprising the first 1,190 amino acids of the native S glycoprotein (S(1190)). The codon-optimized and native S glycoproteins exhibit similar molecular weight as determined by Western blot analysis, indicating that synthetic S glycoprotein is modified correctly in a mammalian expression system. S(1190) binds to the surface of Vero E6 cells, a cell permissive to infection, as demonstrated by fluorescence-activated cell sorter analysis, suggesting that S(1190) maintains the biologic activity present in native S glycoprotein. This interaction is blocked with serum obtained from recovering SARS patients, indicating that the binding is specific. In an effort to map the ligand-binding domain of the SARS-CoV S glycoprotein, carboxy- and amino-terminal truncations of the S(1190) glycoprotein were constructed. Amino acids 270 to 510 were the minimal receptor-binding region of the SARS-CoV S glycoprotein as determined by flow cytometry. We speculate that amino acids 1 to 510 of the SARS-CoV S glycoprotein represent a unique domain containing the receptor-binding site (amino acids 270 to 510), analogous to the S1 subunit of other coronavirus S glycoproteins.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Amino Acids/chemistry , Amino Acids/genetics , Animals , Cell Line , Chlorocebus aethiops , Codon , Flow Cytometry , Humans , Ligands , Membrane Glycoproteins/chemical synthesis , Membrane Glycoproteins/genetics , Mutation , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Spike Glycoprotein, Coronavirus , Structure-Activity Relationship , Vero Cells , Viral Envelope Proteins/chemical synthesis , Viral Envelope Proteins/genetics
18.
Blood ; 101(3): 831-6, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12393732

ABSTRACT

Patients undergoing hematopoietic cell transplantation (HCT) are at increased risk for infections with Streptococcus pneumoniae and have long-lasting, impaired antibody responses to pneumococcal polysaccharide vaccines. We examined whether donor immunization with a heptavalent pneumococcal conjugate vaccine (PCV7) would elicit protective antibody responses to additional doses of vaccine administered early after transplantation. Ninety-six patients scheduled to receive an allogeneic hematopoietic cell transplant were randomized with their donors to receive either a dose of PCV7 vaccine or no vaccine before transplantation. All patients received PCV7 at 3 months, 6 months, and 12 months following transplantation, and serotype-specific antibody concentrations were determined after each dose. Following HCT, geometric mean antibody concentrations of patients in the immunized donor group were significantly higher for 5 of the 7 vaccine serotypes after one dose (P <.05) and for 4 of the 7 serotypes after 2 doses of vaccine (P <.03). Sixty-seven percent of patients in the immunized donor group had presumed protective IgG concentrations more than or equal to 0.50 microg/mL to all 7 serotypes following the first dose of vaccine compared to 36% in the unimmunized donor group (P =.05). After the third dose of vaccine, both groups had more than 60% of patients with concentrations at least 0.50 microg/mL to all vaccine serotypes. Donor immunization enhances early antibody responses of patients undergoing HCT to pneumococcal conjugate vaccine. A 3-dose schedule of PCV7 vaccine at 3, 6, and 12 months is immunogenic in these patients regardless of donor immunization.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Immunization , Pneumococcal Vaccines/administration & dosage , Tissue Donors , Adolescent , Adult , Aged , Antibodies, Bacterial/blood , Antibody Formation/drug effects , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Pneumococcal Infections/etiology , Pneumococcal Infections/prevention & control , Pneumococcal Infections/therapy , Pneumococcal Vaccines/immunology , Time Factors , Transplantation, Homologous/adverse effects , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
19.
Vaccine ; 20(31-32): 3658-67, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12399193

ABSTRACT

As new vaccines are developed, novel adjuvants may play an important role in eliciting an effective immune response. We evaluated the safety and adjuvant properties of monophosphoryl lipid A (MPL in 129 healthy toddlers immunized with two doses of nine-valent pneumococcal-CRM(197) protein conjugate vaccine (PCV9) combined with 10, 25, or 50 micro g of MPL with or without alum (AlPO(4)). Vaccine-specific humoral and cell-mediated responses were examined following the second dose of study vaccine. All doses of MPL were well-tolerated and a dose-dependent effect of MPL on specific cellular responses was observed. The 10 micro g MPL dose significantly enhanced CRM(197)-specific T-cell proliferation (P=0.02) and interferon-gamma (INF-gamma) production (P=0.009) compared to responses of controls who received PCV9 with AlPO(4). In contrast, CRM(197)-specific T-cell proliferation and interferon-gamma production of the 50 micro g MPL/AlPO(4) group were decreased when compared to controls although these differences did not reach statistical significance. IL-5 and IL-13 responses after immunization showed a similar pattern with increased production in the 10 micro g MPL group and decreased production in the 50 micro g MPL/AlPO(4) group compared to controls. There were no differences in serum IgG antibody concentrations to the nine vaccine pneumococcal capsular polysaccharides and carrier protein between the MPL-containing and control vaccine groups. These findings demonstrate a dose-dependent effect of MPL on T-helper cell type 1 (TH-1) responses to the carrier protein and also suggest an effect on T-helper cell type 2 (TH-2) responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Proteins/administration & dosage , Lipid A/analogs & derivatives , Lipid A/administration & dosage , Polysaccharides, Bacterial/administration & dosage , T-Lymphocytes, Helper-Inducer/immunology , Adjuvants, Immunologic/adverse effects , Aluminum Compounds/administration & dosage , Aluminum Compounds/adverse effects , Aluminum Compounds/immunology , Aluminum Compounds/pharmacology , Antibodies, Bacterial/biosynthesis , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/adverse effects , Antigens, Bacterial/immunology , Bacterial Proteins/adverse effects , Bacterial Proteins/immunology , Child, Preschool , Cytokines/biosynthesis , Female , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunoglobulin G/biosynthesis , Infant , Lipid A/adverse effects , Lipid A/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Phosphates/administration & dosage , Phosphates/adverse effects , Phosphates/immunology , Phosphates/pharmacology , Polysaccharides, Bacterial/adverse effects , Polysaccharides, Bacterial/immunology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...