Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Host Microbe ; 30(1): 41-52.e5, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34879230

ABSTRACT

Respiratory syncytial virus (RSV) infection is a major cause of respiratory illness in infants and the elderly. Although several vaccines have been developed, none have succeeded in part due to our incomplete understanding of the correlates of immune protection. While both T cells and antibodies play a role, emerging data suggest that antibody-mediated mechanisms alone may be sufficient to provide protection. Therefore, to map the humoral correlates of immunity against RSV, antibody responses across six different vaccines were profiled in a highly controlled nonhuman primate-challenge model. Viral loads were monitored in both the upper and lower respiratory tracts, and machine learning was used to determine the vaccine platform-agnostic antibody features associated with protection. Upper respiratory control was associated with virus-specific IgA levels, neutralization, and complement activity, whereas lower respiratory control was associated with Fc-mediated effector mechanisms. These findings provide critical compartment-specific insights toward the rational development of future vaccines.


Subject(s)
Primates/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Vaccination , Animals , Antibodies, Neutralizing , Antibodies, Viral/blood , Biomarkers/blood , Chlorocebus aethiops , Humans , Immunity, Innate , Immunoglobulin A/blood , Lung/virology , Respiratory Syncytial Virus Infections/virology , Viral Load
2.
NPJ Vaccines ; 5(1): 16, 2020.
Article in English | MEDLINE | ID: mdl-32128257

ABSTRACT

The RSV Fusion (F) protein is a target for neutralizing antibody responses and is a focus for vaccine discovery; however, the process of RSV entry requires F to adopt a metastable prefusion form and transition to a more stable postfusion form, which displays less potent neutralizing epitopes. mRNA vaccines encode antigens that are translated by host cells following vaccination, which may allow conformational transitions similar to those observed during natural infection to occur. Here we evaluate a panel of chemically modified mRNA vaccines expressing different forms of the RSV F protein, including secreted, membrane associated, prefusion-stabilized, and non-stabilized structures, for conformation, immunogenicity, protection, and safety in rodent models. Vaccination with mRNA encoding native RSV F elicited antibody responses to both prefusion- and postfusion-specific epitopes, suggesting that this antigen may adopt both conformations in vivo. Incorporating prefusion stabilizing mutations further shifts the immune response toward prefusion-specific epitopes, but does not impact neutralizing antibody titer. mRNA vaccine candidates expressing either prefusion stabilized or native forms of RSV F protein elicit robust neutralizing antibody responses in both mice and cotton rats, similar to levels observed with a comparable dose of adjuvanted prefusion stabilized RSV F protein. In contrast to the protein subunit vaccine, mRNA-based vaccines elicited robust CD4+ and CD8+ T-cell responses in mice, highlighting a potential advantage of the technology for vaccines requiring a cellular immune response for efficacy.

3.
Nat Commun ; 10(1): 4153, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515478

ABSTRACT

Respiratory syncytial virus (RSV) infection is the leading cause of hospitalization and infant mortality under six months of age worldwide; therefore, the prevention of RSV infection in all infants represents a significant unmet medical need. Here we report the isolation of a potent and broadly neutralizing RSV monoclonal antibody derived from a human memory B-cell. This antibody, RB1, is equipotent on RSV A and B subtypes, potently neutralizes a diverse panel of clinical isolates in vitro and demonstrates in vivo protection. It binds to a highly conserved epitope in antigenic site IV of the RSV fusion glycoprotein. RB1 is the parental antibody to MK-1654 which is currently in clinical development for the prevention of RSV infection in infants.


Subject(s)
Antibodies, Viral/immunology , Broadly Neutralizing Antibodies/immunology , Conserved Sequence , Glycoproteins/immunology , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/immunology , Animals , Antibodies, Monoclonal/isolation & purification , B-Lymphocytes/immunology , Binding Sites , Disease Models, Animal , Epitopes/immunology , Female , Humans , Immunologic Memory , Models, Molecular , Protein Binding , Sigmodontinae
4.
PLoS Pathog ; 15(6): e1007716, 2019 06.
Article in English | MEDLINE | ID: mdl-31170257

ABSTRACT

There is still no safe and effective vaccine against dengue virus infection. Epidemics of dengue virus infection are increasingly a threat to human health around the world. Antibodies generated in response to dengue infection have been shown to impact disease development and effectiveness of dengue vaccine. In this study, we investigated monoclonal antibody responses to an experimental dengue vaccine in rhesus macaques. Variable regions of both heavy chain (VH) and light chain (VL) were cloned from single antibody-secreting B cells. A total of 780 monoclonal antibodies (mAbs) composed of paired VH and VL were characterized. Results show that the vaccination induces mAbs with diverse germline sequences and a wide range of binding affinities. Six potent neutralizing mAbs were identified among 130 dengue envelope protein binders. Critical amino acids for each neutralizing antibody binding to the dengue envelope protein were identified by alanine scanning of mutant libraries. Diverse epitopes were identified, including epitopes on the lateral ridge of DIII, the I-III hinge, the bc loop adjacent to the fusion loop of DII, and the ß-strands and loops of DI. Significantly, one of the neutralizing mAbs has a previously unknown epitope in DII at the interface of the envelope and membrane protein and is capable of neutralizing all four dengue serotypes. Taken together, the results of this study not only provide preclinical validation for the tested experimental vaccine, but also shed light on a potential application of the rhesus macaque model for better dengue vaccine evaluation and design of vaccines and immunization strategies.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neutralizing , Antibodies, Viral , Dengue Vaccines , Epitopes , Immunoglobulin Heavy Chains , Immunoglobulin Light Chains , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Dengue Vaccines/genetics , Dengue Vaccines/immunology , Dengue Virus/immunology , Epitopes/genetics , Epitopes/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Macaca mulatta
5.
J Virol Methods ; 263: 88-95, 2019 01.
Article in English | MEDLINE | ID: mdl-30381239

ABSTRACT

Viral plaque assays are important tools in the development and evaluation of new antiviral drugs or vaccines in both preclinical and clinical research. While plaque assays are the standard tools to measure infectious virus, the methodology is time-consuming and requires experience in recognizing plaques. The assays are also prone to variation among analysts due to plaque recognition and manual counting errors. Here we describe the development of two simplified plaque assays for measuring RSV virus titers and anti-RSV antibody neutralization titers using 96 well plate formats. First, we evaluated multiple parameters to build up a quantitative plaque assay to measure infectious RSV. We then optimized the assay conditions to assess the fundamental changes from the traditional plaque assay, which were elimination of overnight pre-seeding host cells and addition of a centrifugation step after viral infection of the cells. We designed DoE to refine four key parameters within one experiment for host cell density, host cell volume, viral inoculum volume, host cell and viral mixture incubation time to make this assay more robust. We have also adapted these conditions into a second assay, which was an automated plaque reduction neutralization assay (PRNT) to determine neutralization titers of anti-RSV antibodies. Both assays utilize immune fluorescence staining to detect viral plaques. The images of the immuno-stained wells are captured by the PerkinElmer EnSight instrument and show clear visualization of plaques harvesting on day 3. Software algorithm was specifically designed for automatic counting of these fluorescent "objects". The quantitative plaque assay provided titers of RSV similar to those obtained from the traditional plaque assay. The method has been successfully utilized to screen multiple vaccine candidates in viral shedding efficacy studies. The automated PRNT assay provided antibody neutralizing titers that matched with published data. This automated 96 well plaque assay has made it possible to screen RSV samples in a higher throughput manner, and can be extended to other infectious organisms that form plaques for vaccine or drug evaluation.


Subject(s)
High-Throughput Screening Assays/methods , Optical Imaging , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Viral Plaque Assay/methods , Algorithms , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation , Female , Humans , Neutralization Tests , Reproducibility of Results , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Sigmodontinae/immunology , Sigmodontinae/virology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
6.
PLoS One ; 12(11): e0187642, 2017.
Article in English | MEDLINE | ID: mdl-29121080

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of serious lower respiratory tract disease in young children and older adults throughout the world. Prevention of severe RSV disease through active immunization is optimal but no RSV vaccine has been licensed so far. Immune mechanisms of protection against RSV infection in humans have not been fully established, thus a comprehensive characterization of virus-specific immune responses in a relevant animal model will be beneficial in defining correlates of protection. In this study, we infected juvenile naive AGMs with RSV A2 strain and longitudinally assessed virus-specific humoral and cellular immune responses in both peripheral blood and the respiratory tract. RSV viral loads at nasopharyngeal surfaces and in the lung peaked at around day 5 following infection, and then largely resolved by day 10. Low levels of neutralizing antibody titers were detected in serum, with similar kinetics as RSV fusion (F) protein-binding IgG antibodies. RSV infection induced CD8+, but very little CD4+, T lymphocyte responses in peripheral blood. Virus-specific CD8+ T cell frequencies were ~10 fold higher in bronchoaveolar lavage (BAL) compared to peripheral blood and exhibited effector memory (CD95+CD28-) / tissue resident memory (CD69+CD103+) T (TRM) cell phenotypes. The kinetics of virus-specific CD8+ T cells emerging in peripheral blood and BAL correlated with declining viral titers, suggesting that virus-specific cellular responses contribute to the clearance of RSV infection. RSV-experienced AGMs were protected from subsequent exposure to RSV infection. Additional studies are underway to understand protective correlates in these seropositive monkeys.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Immunologic Memory , Lung/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antigens, CD/blood , Antigens, CD/immunology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chlorocebus aethiops , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lung/metabolism , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Viruses/metabolism
7.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28298602

ABSTRACT

Human respiratory syncytial virus (RSV) is a common cause of severe respiratory disease among infants, immunocompromised individuals, and the elderly. No licensed vaccine is currently available. In this study, we evaluated two parainfluenza virus 5 (PIV5)-vectored vaccines expressing RSV F (PIV5/F) or G (PIV5/G) protein in the cotton rat and African green monkey models for their replication, immunogenicity, and efficacy of protection against RSV challenge. Following a single intranasal inoculation, both animal species shed the vaccine viruses for a limited time but without noticeable clinical symptoms. In cotton rats, the vaccines elicited RSV F- or G-specific serum antibodies and conferred complete lung protection against RSV challenge at doses as low as 103 PFU. Neither vaccine produced the enhanced lung pathology observed in animals immunized with formalin-inactivated RSV. In African green monkeys, vaccine-induced serum and mucosal antibody responses were readily detected, as well. PIV5/F provided nearly complete protection against RSV infection in the upper and lower respiratory tract at a dose of 106 PFU of vaccine. At the same dose levels, PIV5/G was less efficacious. Both PIV5/F and PIV5/G were also able to boost neutralization titers in RSV-preexposed African green monkeys. Overall, our data indicated that PIV5/F is a promising RSV vaccine candidate.IMPORTANCE A safe and efficacious respiratory syncytial virus (RSV) vaccine remains elusive. We tested the recombinant parainfluenza virus 5 (PIV5) vectors expressing RSV glycoproteins for their immunogenicity and protective efficacy in cotton rats and African green monkeys, which are among the best available animal models to study RSV infection. In both species, a single dose of intranasal immunization with PIV5-vectored vaccines was able to produce systemic and local immunity and to protect animals from RSV challenge. The vaccines could also boost RSV neutralization antibody titers in African green monkeys that had been infected previously. Our data suggest that PIV5-vectored vaccines could potentially protect both the pediatric and elderly populations and support continued development of the vector platform.


Subject(s)
Parainfluenza Virus 5/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Chlorocebus aethiops , Disease Models, Animal , Genetic Vectors , Lung/virology , Rats , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/genetics , Respiratory Syncytial Virus, Human/genetics , Sigmodontinae , Vaccination , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vero Cells , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics
8.
J Biol Chem ; 292(1): 278-291, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27879316

ABSTRACT

A major goal for HIV-1 vaccine development is an ability to elicit strong and durable broadly neutralizing antibody (bNAb) responses. The trimeric envelope glycoprotein (Env) spikes on HIV-1 are known to contain multiple epitopes that are susceptible to bNAbs isolated from infected individuals. Nonetheless, all trimeric and monomeric Env immunogens designed to date have failed to elicit such antibodies. We report the structure-guided design of HIV-1 cyclically permuted gp120 that forms homogeneous, stable trimers, and displays enhanced binding to multiple bNAbs, including VRC01, VRC03, VRC-PG04, PGT128, and the quaternary epitope-specific bNAbs PGT145 and PGDM1400. Constructs that were cyclically permuted in the V1 loop region and contained an N-terminal trimerization domain to stabilize V1V2-mediated quaternary interactions, showed the highest homogeneity and the best antigenic characteristics. In guinea pigs, a DNA prime-protein boost regimen with these new gp120 trimer immunogens elicited potent neutralizing antibody responses against highly sensitive Tier 1A isolates and weaker neutralizing antibody responses with an average titer of about 115 against a panel of heterologous Tier 2 isolates. A modest fraction of the Tier 2 virus neutralizing activity appeared to target the CD4 binding site on gp120. These results suggest that cyclically permuted HIV-1 gp120 trimers represent a viable platform in which further modifications may be made to eventually achieve protective bNAb responses.


Subject(s)
Antibodies, Neutralizing/blood , Drug Design , HIV Antibodies/blood , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/immunology , HIV-1/immunology , Animals , Antibodies, Neutralizing/immunology , Binding Sites , Crystallography, X-Ray , Epitopes/immunology , Guinea Pigs , HIV Antibodies/immunology , HIV Infections/blood , HIV Infections/virology , Humans , Protein Binding , Protein Conformation , Protein Multimerization
9.
PLoS One ; 11(6): e0156798, 2016.
Article in English | MEDLINE | ID: mdl-27258388

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in infants, the elderly and in immunosuppressed populations. The vast majority of neutralizing antibodies isolated from human subjects target the RSV fusion (F) glycoprotein, making it an attractive target for the development of vaccines and therapeutic antibodies. Currently, Synagis® (palivizumab) is the only FDA approved antibody drug for the prevention of RSV infection, and there is a great need for more effective vaccines and therapeutics. Phage display is a powerful tool in antibody discovery with the advantage that it does not require samples from immunized subjects. In this study, Morphosys HuCAL GOLD® phage libraries were used for panning against RSV prefusion and postfusion F proteins. Panels of human monoclonal antibodies (mAbs) against RSV F protein were discovered following phage library panning and characterized. Antibodies binding specifically to prefusion or postfusion F proteins and those binding both conformations were identified. 3B1 is a prototypic postfusion F specific antibody while 2E1 is a prototypic prefusion F specific antibody. 2E1 is a potent broadly neutralizing antibody against both RSV A and B strains. Epitope mapping experiments identified a conformational epitope spanning across three discontinuous sections of the RSV F protein, as well as critical residues for antibody interaction.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Glycoproteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Viruses/immunology , Respiratory Syncytial Viruses/pathogenicity , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Surface Display Techniques , Epitope Mapping , Epitopes/immunology , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/drug effects
10.
MAbs ; 8(1): 129-40, 2016.
Article in English | MEDLINE | ID: mdl-26491897

ABSTRACT

Monitoring antigen-specific memory B cells and the antibodies they encode is important for understanding the specificity, breadth and duration of immune response to an infection or vaccination. The antibodies isolated could further help design vaccine antigens for raising relevant protective immune responses. However, developing assays to measure and isolate antigen-specific memory B cells is technically challenging due to the low frequencies of these cells that exist in the circulating blood. Here, we describe a flow cytometry method to identify and isolate dengue envelope-specific memory B cells using a labeled dengue envelope protein. We enumerated dengue-envelope specific memory B cells from a cohort of dengue seropositive donors using this direct flow cytometry assay. A more established and conventional assay, the cultured B ELISPOT, was used as a benchmark comparator. Furthermore, we were able to confirm the single-sorted memory B-cell specificity by culturing B cells and differentiating them into plasma cells using cell lines expressing CD40L. The culture supernatants were assayed for antigen binding and the ability of the antibodies to neutralize the cognate dengue virus. Moreover, we successfully isolated the heavy and light Ig sequences and expressed them as full-length recombinant antibodies to reproduce the activity seen in culture supernatants. Mapping of these antibodies revealed a novel epitope for dengue 2 virus serotype. In conclusion, we established a reproducible methodology to enumerate antigen-specific memory B cells and assay their encoded antibodies for functional characterization.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Dengue Virus/immunology , Immunologic Memory , Viral Envelope Proteins/immunology , B-Lymphocytes/cytology , Cell Culture Techniques , Female , Flow Cytometry , Humans , Male
11.
Antimicrob Agents Chemother ; 58(3): 1652-63, 2014.
Article in English | MEDLINE | ID: mdl-24379202

ABSTRACT

Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are a mainstay of therapy for treating human immunodeficiency type 1 virus (HIV-1)-infected patients. MK-1439 is a novel NNRTI with a 50% inhibitory concentration (IC50) of 12, 9.7, and 9.7 nM against the wild type (WT) and K103N and Y181C reverse transcriptase (RT) mutants, respectively, in a biochemical assay. Selectivity and cytotoxicity studies confirmed that MK-1439 is a highly specific NNRTI with minimum off-target activities. In the presence of 50% normal human serum (NHS), MK-1439 showed excellent potency in suppressing the replication of WT virus, with a 95% effective concentration (EC95) of 20 nM, as well as K103N, Y181C, and K103N/Y181C mutant viruses with EC95 of 43, 27, and 55 nM, respectively. MK-1439 exhibited similar antiviral activities against 10 different HIV-1 subtype viruses (a total of 93 viruses). In addition, the susceptibility of a broader array of clinical NNRTI-associated mutant viruses (a total of 96 viruses) to MK-1439 and other benchmark NNRTIs was investigated. The results showed that the mutant profile of MK-1439 was superior overall to that of efavirenz (EFV) and comparable to that of etravirine (ETR) and rilpivirine (RPV). Furthermore, E138K, Y181C, and K101E mutant viruses that are associated with ETR and RPV were susceptible to MK-1439 with a fold change (FC) of <3. A two-drug in vitro combination study indicated that MK-1439 acts nonantagonistically in the antiviral activity with each of 18 FDA-licensed drugs for HIV infection. Taken together, these in vitro data suggest that MK-1439 possesses the desired properties for further development as a new antiviral agent.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Pyridones/pharmacology , Triazoles/pharmacology , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Drug Synergism , HIV Infections/drug therapy , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , Humans , In Vitro Techniques , Macrophages/drug effects , Monocytes/drug effects , Pyridones/adverse effects , Triazoles/adverse effects , Virus Replication/drug effects
12.
J Virol ; 88(4): 2000-10, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284325

ABSTRACT

A prophylactic vaccine for genital herpes disease remains an elusive goal. We report the results of two studies performed collaboratively in different laboratories that assessed immunogenicity and vaccine efficacy in herpes simplex virus 1 (HSV-1)-seropositive guinea pigs immunized and subsequently challenged intravaginally with HSV-2. In study 1, HSV-2 glycoproteins C (gC2) and D (gD2) were produced in baculovirus and administered intramuscularly as monovalent or bivalent vaccines with CpG and alum. In study 2, gD2 was produced in CHO cells and given intramuscularly with monophosphoryl lipid A (MPL) and alum, or gC2 and gD2 were produced in glycoengineered Pichia pastoris and administered intramuscularly as a bivalent vaccine with Iscomatrix and alum to HSV-1-naive or -seropositive guinea pigs. In both studies, immunization boosted neutralizing antibody responses to HSV-1 and HSV-2. In study 1, immunization with gC2, gD2, or both immunogens significantly reduced the frequency of genital lesions, with the bivalent vaccine showing the greatest protection. In study 2, both vaccines were highly protective against genital disease in naive and HSV-1-seropositive animals. Comparisons between gD2 and gC2/gD2 in study 2 must be interpreted cautiously, because different adjuvants, gD2 doses, and antigen production methods were used; however, significant differences invariably favored the bivalent vaccine. Immunization of naive animals with gC2/gD2 significantly reduced the number of days of vaginal shedding of HSV-2 DNA compared with that for mock-immunized animals. Surprisingly, in both studies, immunization of HSV-1-seropositive animals had little effect on recurrent vaginal shedding of HSV-2 DNA, despite significantly reducing genital disease.


Subject(s)
Herpes Genitalis/prevention & control , Herpesvirus 1, Human/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/pharmacology , Analysis of Variance , Animals , Antibodies, Neutralizing/immunology , Baculoviridae , CHO Cells , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Female , Guinea Pigs , Injections, Intramuscular , Lipid A/analogs & derivatives , Pichia , Real-Time Polymerase Chain Reaction , Viral Vaccines/administration & dosage
13.
Vaccine ; 31(42): 4888-93, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23941913

ABSTRACT

There is a need for novel rabies vaccines suitable for short course, pre- and post-exposure prophylactic regimens which require reduced doses of antigen to address the current worldwide supply issue. We evaluated in rhesus macaques the immunogenicity of a quarter-dose of a standard rabies vaccine formulated with Merck's amorphous aluminum hydroxylphosphate sulfate adjuvant, the saponin-based ISCOMATRIX™ adjuvant, or a synthetic TLR9 agonist. All adjuvants significantly increased the magnitude and durability of the humoral immune response as measured by rapid fluorescent focus inhibition test (RFFIT). Several three-dose vaccine regimens resulted in adequate neutralizing antibody of ≥ 0.5 IU/ml earlier than the critical day seven post the first dose. Rabies vaccine with ISCOMATRIX™ adjuvant given at days 0 and 3 resulted in neutralizing antibody titers which developed faster and were up to one log10 higher compared to WHO-recommended intramuscular and intradermal regimens and furthermore, passive administration of human rabies immunoglobulin did not interfere with immunogenicity of this reduced dose, short course vaccine regimen. Adjuvantation of whole-killed rabies vaccine for intramuscular injection may therefore be a viable alternative to intradermal application of non-adjuvanted vaccine for both pre- and post-exposure regimens.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Compounds/administration & dosage , Cholesterol/administration & dosage , Phospholipids/administration & dosage , Rabies Vaccines/administration & dosage , Rabies Vaccines/immunology , Saponins/administration & dosage , Toll-Like Receptor 9/agonists , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Drug Combinations , Female , Injections, Intramuscular , Macaca mulatta , Male , Neutralization Tests , Phosphates/administration & dosage , Rabies/prevention & control , Sulfates/administration & dosage , Toll-Like Receptor 9/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
14.
Hum Vaccin ; 7(11): 1118-23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22048265

ABSTRACT

BACKGROUND: Introduction of rotavirus vaccines in the United States beginning in 2006 led to a rapid decline in the frequency of acute rotavirus gastroenteritis necessitating medical attention. We examined whether serotype replacement was occurring as a result of vaccine use. METHODS: Children with gastroenteritis presenting to CHOP have been tested for rotavirus antigen in the stool. Commencing with the 1999-2000 season, positive specimens were genotyped to establish the G (VP7) and P (VP4) type. RESULTS: In 2009-2010, 4 hospital-acquired and 18 community-acquired cases of rotavirus gastroenteritis were identified at CHOP. For the third consecutive full season since the introduction of rotavirus vaccines, the proportion of annual G3 cases was higher than in the prevaccine era. Although G3 strains caused 50% of the community cases in 2009-10, the absolute number of G3 cases actually dropped from 15 in 2007-08 to 8 and 9 in the 2008-09 and 2009-10 seasons, respectively. P[8] accounted for > 90% of cases seen at CHOP in each of the last 3 seasons, including 20/22 (91%) cases during the 2009-10 season. CONCLUSIONS: Findings to date provide suggestive but still inconclusive evidence for vaccine-driven serotype replacement. Given the increased proportion of G3 cases in the new vaccine era despite the overall marked reduction in rotavirus gastroenteritis, continued surveillance is prudent.


Subject(s)
Community-Acquired Infections/epidemiology , Cross Infection/epidemiology , Gastroenteritis/epidemiology , Hospitals, Pediatric/statistics & numerical data , Rotavirus Infections/epidemiology , Rotavirus Vaccines/administration & dosage , Rotavirus/isolation & purification , Adolescent , Antigens, Viral/analysis , Capsid Proteins/analysis , Child , Child, Preschool , Community-Acquired Infections/virology , Cross Infection/virology , Epidemics , Feces/virology , Gastroenteritis/virology , Genotype , Humans , Infant , Philadelphia/epidemiology , Rotavirus/classification , Rotavirus/genetics , Rotavirus Infections/virology , Rotavirus Vaccines/immunology
15.
J Med Chem ; 54(22): 7920-33, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-21985673

ABSTRACT

Highly active antiretroviral therapy (HAART) significantly reduces human immunodeficiency virus (HIV) viral load and has led to a dramatic decrease in acquired immunodeficiency syndrome (AIDS) related mortality. Despite this success, there remains a critical need for new HIV therapies to address the emergence of drug resistant viral strains. Next generation NNRTIs are sought that are effective against these mutant forms of the HIV virus. The bound conformations of our lead inhibitors, MK-1107 (1) and MK-4965 (2), were divergent about the oxymethylene linker, and each of these conformations was rigidified using two isomeric cyclic constraints. The constraint derived from the bioactive conformation of 2provided novel, highly potent NNRTIs that possess broad spectrum antiviral activity and good pharmacokinetic profiles. Systematic SAR led to the identification of indazole as the optimal conformational constraint to provide MK-6186 (3) and MK-7445 (6). Despite their reduced flexibility, these compounds had potency comparable to that of the corresponding acyclic ethers in both recombinant enzyme and cell based assays against both the wild-type and the clinically relevant mutant strains.


Subject(s)
Anti-HIV Agents/chemical synthesis , Imidazoles/chemical synthesis , Indazoles/chemical synthesis , Pyrazoles/chemical synthesis , Reverse Transcriptase Inhibitors/chemical synthesis , Triazoles/chemical synthesis , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Cells, Cultured , Dogs , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/genetics , HIV-1/isolation & purification , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Indazoles/pharmacokinetics , Indazoles/pharmacology , Models, Molecular , Molecular Conformation , Mutation , Nitriles/chemical synthesis , Nitriles/pharmacokinetics , Nitriles/pharmacology , Nitrobenzenes/chemical synthesis , Nitrobenzenes/pharmacokinetics , Nitrobenzenes/pharmacology , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship , Thermodynamics , Triazoles/pharmacokinetics , Triazoles/pharmacology
16.
J Virol ; 85(20): 10472-86, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813597

ABSTRACT

Attempts to develop a vaccine to prevent genital herpes simplex virus 2 (HSV-2) disease have been only marginally successful, suggesting that novel strategies are needed. Immunization with HSV-2 glycoprotein C (gC-2) and gD-2 was evaluated in mice and guinea pigs to determine whether adding gC-2 to a gD-2 subunit vaccine would improve protection by producing antibodies that block gC-2 immune evasion from complement. Antibodies produced by gC-2 immunization blocked the interaction between gC-2 and complement C3b, and passive transfer of gC-2 antibody protected complement-intact mice but not C3 knockout mice against HSV-2 challenge, indicating that gC-2 antibody is effective, at least in part, because it prevents HSV-2 evasion from complement. Immunization with gC-2 also produced neutralizing antibodies that were active in the absence of complement; however, the neutralizing titers were higher when complement was present, with the highest titers in animals immunized with both antigens. Animals immunized with the gC-2-plus-gD-2 combination had robust CD4+ T-cell responses to each immunogen. Multiple disease parameters were evaluated in mice and guinea pigs immunized with gC-2 alone, gD-2 alone, or both antigens. In general, gD-2 outperformed gC-2; however, the gC-2-plus-gD-2 combination outperformed gD-2 alone, particularly in protecting dorsal root ganglia in mice and reducing recurrent vaginal shedding of HSV-2 DNA in guinea pigs. Therefore, the gC-2 subunit antigen enhances a gD-2 subunit vaccine by stimulating a CD4+ T-cell response, by producing neutralizing antibodies that are effective in the absence and presence of complement, and by blocking immune evasion domains that inhibit complement activation.


Subject(s)
Ganglia, Spinal/immunology , Herpes Genitalis/prevention & control , Herpes Zoster Vaccine/immunology , Herpes Zoster/prevention & control , Herpesvirus 2, Human/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , Female , Guinea Pigs , Herpes Genitalis/immunology , Herpes Zoster/immunology , Herpes Zoster Vaccine/administration & dosage , Immunization/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Secondary Prevention , Vagina/virology , Virus Shedding
17.
Diagn Microbiol Infect Dis ; 70(2): 218-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21398071

ABSTRACT

Most rotavirus gastroenteritis is caused by G1P[8] strains. When G2 infections are encountered, the P type has most often been reported to be P[4]. The purpose of our study was to describe an unusual outbreak of G2P[6] cases. Children presenting to The Children's Hospital of Philadelphia with acute gastroenteritis have been monitored for rotavirus antigen in stool by ELISA (with G-typing if ELISA positive) since 1994-1995. Compared to the last 12 rotavirus seasons before the 2006 introduction of a new rotavirus vaccine, the 2005-2006 season had by far the highest number of evaluable rotavirus infections [n = 275 from September 2005 through June 2006, of which 261 (95%) were G typed] and the greatest number of G2 cases (n = 101, 39% of typed strains). Only 16% of G2 strains were associated with P[4], whereas genotype G2P[6] was responsible for 83% of the G2 infections. Eighty-eight percent of the 84 G2P[6] cases occurred in the 60% of patients who were African-Americans, most of whom were urban residents. Among 157 African-American patients, G2 cases (n = 80; 52%) predominated, including 74 due to G2P[6]. Children <6 months old accounted for 27% of cases overall, but 36% of the G2P[6] cases. G2 rotaviruses caused over a third of the community-acquired rotavirus cases in children presenting to CHOP in 2005-2006, attesting to the potential impact of G2 strains during some epidemics. The large majority of G2 strains had the rare P[6] genotype. Urban African-American children under 6 months of age were disproportionately affected.


Subject(s)
Disease Outbreaks , Gastroenteritis/epidemiology , Rotavirus Infections/epidemiology , Rotavirus/classification , Antigens, Viral/analysis , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Feces/virology , Female , Gastroenteritis/virology , Genotype , Humans , Infant , Infant, Newborn , Male , Molecular Epidemiology , Molecular Typing , Philadelphia/epidemiology , Rotavirus/genetics , Rotavirus/isolation & purification , Rotavirus Infections/virology
19.
Bioorg Med Chem Lett ; 20(22): 6754-7, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20869872

ABSTRACT

Optimization studies using an HIV RNase H active site inhibitor containing a 1-hydroxy-1,8-naphthyridin-2(1H)-one core identified 4-position substituents that provided several potent and selective inhibitors. The best compound was potent and selective in biochemical assays (IC(50)=0.045 µM, HIV RT RNase H; 13 µM, HIV RT-polymerase; 24 µM, HIV integrase) and showed antiviral efficacy in a single-cycle viral replication assay in P4-2 cells (IC(50)=0.19 µM) with a modest window with respect to cytotoxicity (CC(50)=3.3 µM).


Subject(s)
Anti-HIV Agents/pharmacology , Enzyme Inhibitors/pharmacology , HIV-1/enzymology , Ribonuclease H/antagonists & inhibitors , Anti-HIV Agents/chemistry , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Naphthyridines/chemistry , Naphthyridines/pharmacology
20.
Bioorg Med Chem Lett ; 20(15): 4328-32, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20609585

ABSTRACT

Biaryl ethers were recently reported as potent NNRTIs. Herein, we disclose a detailed effort to modify the previously reported compound 1. We have designed and synthesized a series of novel pyrazole derivatives as a surrogate for pyrazolopyridine motif that were potent inhibitors of HIV-1 RT with nanomolar intrinsic activity on the WT and key mutant enzymes and potent antiviral activity in infected cells.


Subject(s)
Anti-HIV Agents/chemistry , Ethers/chemistry , HIV Reverse Transcriptase/antagonists & inhibitors , Pyrazoles/chemistry , Pyridines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Allosteric Regulation , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacokinetics , Dogs , Ethers/chemical synthesis , Ethers/pharmacokinetics , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Mutation , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Rats , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/pharmacokinetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...