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










Publication year range
1.
J Pediatric Infect Dis Soc ; 4(4): e143-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26582883

ABSTRACT

We conducted a phase I clinical trial (clinicaltrials.gov identifier, NCT00641017) of the experimental live-attenuated human parainfluenza virus type 1 (HPIV-1) vaccine rHPIV-1/84/del 170/942A sequentially in 3 groups: adults, HPIV-1-seropositive children, and HPIV-1-seronegative children, the target population for vaccination. rHPIV-1/84/del 170/942A was appropriately restricted in replication in adults and HPIV-1-seropositive children but was overattenuated (ie, insufficiently infectious and immunogenic) for HPIV-1-seronegative children.


Subject(s)
Parainfluenza Vaccines/therapeutic use , Paramyxoviridae Infections/prevention & control , Adult , Antibodies, Viral/blood , Child, Preschool , Double-Blind Method , Humans , Infant , Parainfluenza Virus 1, Human , Paramyxoviridae Infections/epidemiology , Vaccines, Attenuated/therapeutic use
2.
Blood ; 125(10): 1601-10, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25568348

ABSTRACT

The treatment of non-Hodgkin lymphomas has benefited enormously from the introduction of monoclonal antibody-based therapies. However, the efficacy of these treatments varies with lymphoma subtypes and typically decreases with subsequent relapses. Here, we report on antigen-armed antibodies (AgAbs) as a potential treatment of B-cell lymphoma. AgAbs include antigens from ubiquitous pathogens, such as Epstein-Barr virus (EBV), that persist in their host and elicit strong lifelong T-cell responses. They act as vectors by introducing antigen directly into tumor cells to induce an antigen-specific CD4(+) T-cell response against these cells. We have fused antibodies targeting human B-cell surface receptors (CD19-22) to immunodominant T-cell antigens from EBV proteins, including EBNA1, EBNA3B, and EBNA3C. Exposure of EBV-transformed B cells and of Burkitt lymphoma cells to AgAbs led to antigen presentation, T-cell recognition, and target cell killing. The efficiency of AgAb action paralleled the abundance of the targeted molecules on lymphoma cells as well as their HLA class II expression levels. AgAbs can also induce activation and proliferation of EBV-specific memory CD4(+) T cells ex vivo. These studies show the potential of AgAbs as an effective therapeutic strategy against B-cell lymphomas.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Antibodies, Neoplasm/genetics , Antigen Presentation , B-Lymphocytes/immunology , Cell Line, Transformed , Cell Line, Tumor , Epitopes, T-Lymphocyte , Herpesvirus 4, Human/immunology , Humans , Immunologic Memory , Lymphocyte Activation , Receptors, Antigen, B-Cell/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology
3.
Virology ; 433(2): 320-8, 2012 Nov 25.
Article in English | MEDLINE | ID: mdl-22959894

ABSTRACT

Human parainfluenza viruses (PIVs) cause acute respiratory illness in children, the elderly, and immunocompromised patients. PIV3 is a common cause of bronchiolitis and pneumonia, whereas PIV1 and 2 are frequent causes of upper respiratory tract illness and croup. To assess how PIV1, 2, and 3 differ with regard to replication and induction of type I interferons, interleukin-6, and relevant chemokines, we infected primary human airway epithelium (HAE) cultures from the same tissue donors and examined replication kinetics and cytokine secretion. PIV1 replicated to high titer yet did not induce cytokine secretion until late in infection, while PIV2 replicated less efficiently but induced an early cytokine peak. PIV3 replicated to high titer but induced a slower rise in cytokine secretion. The T cell chemoattractants CXCL10 and CXCL11 were the most abundant chemokines induced. Differences in replication and cytokine secretion might explain some of the differences in PIV serotype-specific pathogenesis and epidemiology.


Subject(s)
Bronchi/immunology , Bronchi/virology , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 2, Human/physiology , Parainfluenza Virus 3, Human/physiology , Trachea/immunology , Trachea/virology , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Cytokines/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Kinetics , Parainfluenza Virus 1, Human/classification , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Parainfluenza Virus 2, Human/classification , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 2, Human/pathogenicity , Parainfluenza Virus 3, Human/classification , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/pathogenicity , Polymorphism, Single Nucleotide , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Serotyping , Species Specificity , Virus Replication
4.
Expert Rev Respir Med ; 5(4): 515-26, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21859271

ABSTRACT

In children under 5 years of age, human parainfluenza viruses (HPIVs) as a group are the second most common etiology of acute respiratory illness leading to hospitalization, surpassed only by respiratory syncytial virus but ahead of influenza viruses. Using reverse genetics systems for HPIV serotypes 1, 2 and 3 (HPIV1, 2 and 3), several live-attenuated HPIVs have been generated and evaluated as intranasal vaccines in adults and in children. Two vaccines against HPIV3 were found to be well tolerated, infectious and immunogenic in Phase I trials in HPIV3-seronegative infants and children and should progress to proof-of-concept trials. Vaccines against HPIV1 and HPIV2 are less advanced and have just entered pediatric trials.


Subject(s)
Drug Design , Parainfluenza Vaccines/administration & dosage , Respirovirus Infections/prevention & control , Respirovirus/immunology , Acute Disease , Administration, Intranasal , Aerosols , Child, Preschool , Humans , Infant , Parainfluenza Vaccines/chemistry , Parainfluenza Vaccines/genetics , Respirovirus/genetics , Respirovirus Infections/epidemiology , Respirovirus Infections/immunology , Treatment Outcome , Vaccines, Attenuated/administration & dosage
5.
Herpesviridae ; 1(1): 6, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21429237

ABSTRACT

Genetic mutant organisms pervade all areas of Biology. Early on, herpesviruses (HV) were found to be amenable to genetic analysis using homologous recombination techniques in eukaryotic cells. More recently, HV genomes cloned onto a bacterial artificial chromosome (BAC) have become available. HV BACs can be easily modified in E.coli and reintroduced in eukaryotic cells to produce infectious viruses. Mutants derived from HV BACs have been used both to understand the functions of all types of genetic elements present on the virus genome, but also to generate mutants with potentially medically relevant properties such as preventative vaccines. Here we retrace the development of the BAC technology applied to the Epstein-Barr virus (EBV) and review the strategies available for the construction of mutants. We expand on the appropriate controls required for proper use of the EBV BACs, and on the technical hurdles researchers face in working with these recombinants. We then discuss how further technological developments might successfully overcome these difficulties. Finally, we catalog the EBV BAC mutants that are currently available and illustrate their contributions to the field using a few representative examples.

6.
Vaccine ; 28(3): 767-79, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19857454

ABSTRACT

A novel recombinant human parainfluenza virus type 1 (rHPIV1), rHPIV1-C+P, in which the overlapping open reading frames of the C and P genes were separated in order to introduce mutations into the C gene without affecting P, was generated. Infectious rHPIV1-C+P was readily recovered and replicated as efficiently as HPIV1 wild type (wt) in vitro and in African green monkeys (AGMs). rHPIV1-C+P expressed increased levels of C protein and, surprisingly, activated the type I IFN and apoptosis responses more strongly than HPIV1 wt. rHPIV1-C+P provided a useful backbone for recovering an attenuated P/C gene mutation (Delta 84-85), which was previously unrecoverable, likely due to detrimental effects of the deletion on the P protein. rHPIV1-C(Delta 84-85)+P and an additional mutant, rHPIV1-C(Delta 169-170)+P, were found to replicate to similar titers in vitro and to activate the type I IFN and apoptosis responses to a similar degree as rHPIV1-C+P. rHPIV1-C(Delta 84-85)+P was found to be highly attenuated in AGMs, and all viruses were immunogenic and effective in protecting AGMs against challenge with HPIV1 wt. rHPIV1-C(Delta 84-85)+P will be investigated as a potential live-attenuated vaccine candidate for HPIV1.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 1, Human/pathogenicity , Phosphoproteins/genetics , Sequence Deletion , Viral Proteins/genetics , Animals , Antibodies, Viral/blood , Apoptosis , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Interferon Type I/biosynthesis , Molecular Sequence Data , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load , Virulence , Virus Replication
7.
J Virol ; 83(4): 1892-910, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19052086

ABSTRACT

Human parainfluenza virus type 1 (HPIV1) is an important respiratory pathogen in children and the most common cause of viral croup. We performed a microarray-based analysis of gene expression kinetics to examine how wild-type (wt) HPIV1 infection altered gene expression in human respiratory epithelial cells and what role beta interferon played in this response. We similarly evaluated HPIV1-P(C-), a highly attenuated and apoptosis-inducing virus that does not express any of the four C proteins, and HPIV1-C(F170S), a less attenuated mutant that contains a single point mutation in C and, like wt HPIV1, does not efficiently induce apoptosis, to examine the role of the C proteins in controlling host gene expression. We also used these data to investigate whether the phenotypic differences between the two C mutants could be explained at the transcriptional level. Mutation or deletion of the C proteins of HPIV1 permitted the activation of over 2,000 cellular genes that otherwise would be repressed by HPIV1 infection. Thus, the C proteins profoundly suppress the response of human respiratory cells to HPIV1 infection. Cellular pathways targeted by the HPIV1 C proteins were identified and their transcriptional control was analyzed using bioinformatics. Transcription factor binding sites for IRF and NF-kappaB were overrepresented in some of the C protein-targeted pathways, but other pathways were dominated by less-known factors, such as forkhead transcription factor FOXD1. Surprisingly, the host responses to the P(C-) and C(F170S) mutants were very similar, and only subtle differences in the expression kinetics of caspase 3 and TRAIL receptor 2 were observed. Thus, changes in host cell transcription did not reflect the striking phenotypic differences observed between these two viruses.


Subject(s)
Host-Pathogen Interactions , Parainfluenza Virus 1, Human/physiology , Transcription, Genetic , Viral Proteins/metabolism , Cell Line , Epithelial Cells/virology , Gene Deletion , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Parainfluenza Virus 1, Human/genetics , Point Mutation , Respiratory Mucosa/virology , Viral Proteins/genetics
8.
J Virol ; 82(18): 8965-77, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18614629

ABSTRACT

Recombinant human parainfluenza virus type 1 (rHPIV1) was modified to create rHPIV1-P(C-), a virus in which expression of the C proteins (C', C, Y1, and Y2) was silenced without affecting the amino acid sequence of the P protein. Infectious rHPIV1-P(C-) was readily recovered from cDNA, indicating that the four C proteins were not essential for virus replication. Early during infection in vitro, rHPIV1-P(C-) replicated as efficiently as wild-type (wt) HPIV1, but its titer subsequently decreased coincident with the onset of an extensive cytopathic effect not observed with wt rHPIV1. rHPIV1-P(C-) infection, but not wt rHPIV1 infection, induced caspase 3 activation and nuclear fragmentation in LLC-MK2 cells, identifying the HPIV1 C proteins as inhibitors of apoptosis. In contrast to wt rHPIV1, rHPIV1-P(C-) and rHPIV1-C(F170S), a mutant encoding an F170S substitution in C, induced interferon (IFN) and did not inhibit IFN signaling in vitro. However, only rHPIV1-P(C-) induced apoptosis. Thus, the anti-IFN and antiapoptosis activities of HPIV1 were separable: both activities are disabled in rHPIV1-P(C-), whereas only the anti-IFN activity is disabled in rHPIV1-C(F170S). In African green monkeys (AGMs), rHPIV1-P(C-) was considerably more attenuated than rHPIV1-C(F170S), suggesting that disabling the anti-IFN and antiapoptotic activities of HPIV1 had additive effects on attenuation in vivo. Although rHPIV1-P(C-) protected against challenge with wt HPIV1, its highly restricted replication in AGMs and in primary human airway epithelial cell cultures suggests that it might be overattenuated for use as a vaccine. Thus, the C proteins of HPIV1 are nonessential but have anti-IFN and antiapoptosis activities required for virulence in primates.


Subject(s)
Apoptosis/drug effects , Interferons/antagonists & inhibitors , Parainfluenza Virus 1, Human/physiology , Parainfluenza Virus 1, Human/pathogenicity , Viral Proteins/metabolism , Virus Replication/drug effects , Animals , Base Sequence , Caspase 3/metabolism , Cell Line , Chlorocebus aethiops , Enzyme Activation , Humans , Molecular Sequence Data , Parainfluenza Virus 1, Human/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Recombination, Genetic , Vero Cells , Viral Proteins/genetics
9.
J Virol ; 82(16): 8059-70, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524813

ABSTRACT

Human parainfluenza virus type 1 (HPIV1) is a significant cause of pediatric respiratory disease in the upper and lower airways. An in vitro model of human ciliated airway epithelium (HAE), a useful tool for studying respiratory virus-host interactions, was used in this study to show that HPIV1 selectively infects ciliated cells within the HAE and that progeny virus is released from the apical surface with little apparent gross cytopathology. In HAE, type I interferon (IFN) is induced following infection with an HPIV1 mutant expressing defective C proteins with an F170S amino acid substitution, rHPIV1-C(F170S), but not following infection with wild-type HPIV1. IFN induction coincided with a 100- to 1,000-fold reduction in virus titer, supporting the hypothesis that the HPIV1 C proteins are critical for the inhibition of the innate immune response. Two recently characterized live attenuated HPIV1 vaccine candidates expressing mutant C proteins were also evaluated in HAE. The vaccine candidates, rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) and rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11), which contain temperature-sensitive (ts) attenuating (att) and non-ts att mutations, were highly restricted in growth in HAE at permissive (32 degrees C) and restrictive (37 degrees C) temperatures. The viruses grew slightly better at 37 degrees C than at 32 degrees C, and rHPIV1-C(R84G/Delta170)HN(T553A)L(Y942A) was less attenuated than rHPIV1-C(R84G/Delta170)HN(T553A)L(Delta1710-11). The level of replication in HAE correlated with that previously observed for African green monkeys, suggesting that the HAE model has potential as a tool for the preclinical evaluation of HPIV1 vaccines, although how these in vitro data will correlate with vaccine virus replication in seronegative human subjects remains to be seen.


Subject(s)
Bronchi/virology , Cilia/virology , Interferons/metabolism , Mutation , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/metabolism , Trachea/virology , Bronchi/metabolism , Humans , Interferon-alpha/metabolism , Interferon-beta/metabolism , Microscopy, Confocal , Models, Biological , Phenotype , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Temperature , Trachea/metabolism
10.
Virol J ; 4: 67, 2007 Jul 02.
Article in English | MEDLINE | ID: mdl-17605811

ABSTRACT

BACKGROUND: Two recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) mutant viruses have been developed, using a reverse genetics system, for evaluation as potential intranasal vaccine candidates. These rHPIV1 vaccine candidates have two non-temperature sensitive (non-ts) attenuating (att) mutations primarily in the P/C gene, namely CR84GHNT553A (two point mutations used together as a set) and CDelta170 (a short deletion mutation), and two ts att mutations in the L gene, namely LY942A (a point mutation), and LDelta1710-11 (a short deletion), the last of which has not been previously described. The latter three mutations were specifically designed for increased genetic and phenotypic stability. These mutations were evaluated on the HPIV1 backbone, both individually and in combination, for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). RESULTS: The rHPIV1 mutant bearing the novel LDelta1710-11 mutation was highly ts and attenuated in AGMs and was immunogenic and efficacious against HPIV1 wt challenge. The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates were highly ts, with shut-off temperatures of 38 degrees C and 35 degrees C, respectively, and were highly attenuated in AGMs. Immunization with rHPIV1-CR84G/Delta170HNT553ALY942A protected against HPIV1 wt challenge in both the upper and lower respiratory tracts. In contrast, rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 was not protective in AGMs due to over-attenuation, but it is expected to replicate more efficiently and be more immunogenic in the natural human host. CONCLUSION: The rHPIV1-CR84G/Delta170HNT553ALY942A and rHPIV1-CR84G/Delta170HNT553ALDelta1710-11 vaccine candidates are clearly highly attenuated in AGMs and clinical trials are planned to address safety and immunogenicity in humans.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, DNA/immunology , Viral Proteins/genetics , Administration, Intranasal , Animals , Attachment Sites, Microbiological/genetics , Base Sequence , Cell Line , Chlorocebus aethiops , Humans , Molecular Sequence Data , Mutation , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/physiology , Phosphoproteins/genetics , Phosphoproteins/immunology , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Treatment Outcome , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vero Cells , Viral Proteins/immunology , Virus Replication
11.
Vaccine ; 25(10): 1856-67, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17240000

ABSTRACT

Vaccines are urgently needed to elicit immunity to different influenza virus strains. DNA vaccines can elicit partial protective immunity, however their efficacy requires improvement. We assessed the capacity of individual type I IFN multigene family members as subtype transgenes to abrogate influenza virus replication in a vaccination/challenge mouse model. Differences in antiviral efficacy were found among the subtypes with IFNA5 and IFNA6 being most effective, while IFNA1 was the least effective in reducing lung virus replication. Mice vaccinated with combinatorial HA/IFNA6 or NP/IFNA6 showed reduced lung viral titres, clinical score, body weight loss, and pulmonary tissue damage compared to IFNA6, HA, or NP viral vaccination alone. In addition, IFNA6 increased IgG2a titres with upregulation of IFN-gamma response in the respiratory tract. We conclude that IFN-alpha 6 has antiviral and immunomodulatory effects, which improve efficacy of DNA vaccines for enhanced control of influenza.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Interferon-alpha/immunology , Nucleoproteins/immunology , RNA-Binding Proteins/immunology , Vaccines, DNA/immunology , Viral Core Proteins/immunology , Animals , Antibodies, Viral/blood , Body Weight , Disease Models, Animal , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/genetics , Interferon-alpha/classification , Interferon-alpha/genetics , Interferon-gamma/biosynthesis , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred BALB C , Nucleocapsid Proteins , Nucleoproteins/genetics , Orthomyxoviridae Infections/prevention & control , RNA-Binding Proteins/genetics , Vaccines, DNA/genetics , Viral Core Proteins/genetics
12.
Virology ; 352(1): 61-73, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16750233

ABSTRACT

Recombinant human parainfluenza virus type 1 (HPIV1) and mutants containing point and deletion (Delta) mutations in the P/C gene (r-CDelta10-15HNT553A, r-CR84G, r-CF170S and r-CDelta170), which have previously been evaluated as HPIV1 vaccine candidates, were evaluated for their effect on the type I interferon (IFN) response in vitro. HPIV1 wt infection inhibited the IFN response by inhibiting IFN regulatory factor-3 (IRF-3) activation and IFN production in A549 cells and IFN signaling in Vero cells. In contrast, r-CR84G, r-CF170S and r-CDelta170 were defective for inhibition of IRF-3 activation and IFN production and r-CF170S and r-CDelta170 did not inhibit IFN signaling. Thus, HPIV1 antagonizes the IFN response at both the level of induction and signaling, and antagonism at both levels was disrupted by mutations in the P/C gene. Because CF170S affects C and not P, the anti-IFN function can be attributed to the C proteins. These data, in the context of previous in vivo studies, suggest that the loss of antagonism of the IFN response at both the level of induction and signaling, observed with the P/C mutants, r-CF170S and r-CDelta170, was necessary for significant attenuation in African green monkeys (AGMs).


Subject(s)
Interferon Type I/metabolism , Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/pathogenicity , Phosphoproteins/genetics , Vaccines, Attenuated/immunology , Viral Proteins/genetics , Animals , Cell Line , Chlorocebus aethiops , Humans , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/immunology , Phosphoproteins/immunology , Point Mutation , Signal Transduction , Vaccines, Attenuated/genetics , Vero Cells , Viral Proteins/immunology
13.
Vaccine ; 24(14): 2674-84, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16364511

ABSTRACT

The P/C gene of human parainfluenza virus type 1 (HPIV1) encodes a nested set of related accessory C proteins, C'/C/Y1/Y2, which have been shown in other paramyxoviruses to have a role in evasion of the type I interferon (IFN) response following virus infection. We previously demonstrated that a set of two amino acid substitutions, CR84G/HNT553A, and a separate amino acid substitution, CF170S, are independently attenuating for HPIV1 in African green monkeys (AGMs). However, in each case the attenuation (att) phenotype is vulnerable to reversion by a single nucleotide change back to wild type. Using reverse genetics, recombinant HPIV1 (rHPIV1) vaccine candidates were generated that were designed for increased genetic and phenotypic stability by: (i) creating a two-amino acid deletion and substitution at the site of the CF170S mutation, yielding CDelta170; (ii) introducing a six amino acid deletion in the N-terminal region of C, CDelta10-15; and (iii) combining these stable deletion mutations with the att CR84G/HNT553A mutation. The resulting rHPIV1 vaccine candidates were evaluated for attenuation in hamsters and AGMs and for immunogenicity and protective efficacy in AGMs. The CDelta10-15 mutation was attenuating in hamsters but not in AGMs, and likely will be of limited value for an HPIV1 vaccine. Conversely, the CR84G/HNT553A mutation set was attenuating in AGMs but not in hamsters. Thus, these two mutations demonstrated reciprocal host range phenotypes involving different regions of C. The CDelta170 mutation conferred a significant level of attenuation in hamsters and AGMs that closely resembled that of CF170S and will be of particular utility for vaccine development because it involves a deletion of six nucleotides rendering it highly refractory to reversion. The combination of the CR84G/HNT553A mutation set and the CDelta170 deletion mutation yielded a virus, rCR84G/Delta170 HNT553A, that exhibited a satisfactory level of attenuation in hamsters and AGMs and was immunogenic and highly protective against HPIV1 wt challenge. This virus will be evaluated clinically as a live intranasal HPIV1 vaccine, one that can be further attenuated as necessary by the introduction of additional stabilized att mutations previously developed in the L protein.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, Attenuated/immunology , Animals , Cell Line , Cricetinae , Humans , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/pathogenicity , Point Mutation , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics
14.
Methods Mol Med ; 116: 207-19, 2005.
Article in English | MEDLINE | ID: mdl-16000864

ABSTRACT

Delivery of type I interferon (IFN) subtypes by intramuscular inoculation of mice with a recombinant mammalian expression vector encoding IFN stimulates the immune response. Such immunomodulation drives towards a Th1-like response. The degree of stimulation of the immune response was influenced by several parameters of the naked deoxyribonucleic acid (DNA) vaccination protocol. Pretreatment of mice with bupivacaine increased transgene expression in situ. The specific subtype gene of type I IFN, the DNA concentration, the combined use of two or more subtypes, and the timing of the DNA immunisations were all found to influence the level of efficacy of IFN gene therapy in a mouse model for cytomegalovirus (CMV) infection and disease. In addition, adjuvant therapy, using type I IFN genes, for DNA virus vaccination (CMV glycoprotein B) enhanced viral-specific immunity and reduced the severity of myocarditis in mice. Thus, type I IFN gene therapy has potent adjuvant properties when delivered as DNA and can be used to regulate virus infection and disease via pleiotropic actions in the stimulation of immune responses.


Subject(s)
Cytomegalovirus Infections , Gene Transfer Techniques , Genetic Therapy/methods , Interferons/therapeutic use , Protein Isoforms/therapeutic use , Anesthetics, Local/metabolism , Animals , Bupivacaine/metabolism , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/therapy , Humans , Interferons/genetics , Interferons/metabolism , Male , Mice , Mice, Inbred BALB C , Myocarditis/pathology , Myocarditis/virology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transgenes , Vaccines, DNA
15.
Vaccine ; 23(38): 4631-46, 2005 Sep 07.
Article in English | MEDLINE | ID: mdl-15951066

ABSTRACT

A set of recombinant, live attenuated human parainfluenza virus type 1 (rHPIV1) vaccine candidates was evaluated for attenuation, immunogenicity, and protective efficacy in African green monkeys (AGMs). Temperature sensitive (ts) and non-ts attenuating (att) mutations in the P/C and L genes were introduced individually or in various combinations into rHPIV1, including the C(R84G) and HN(T553A) mutations identified in the present work and the C(F170S), L(Y942A), and L(L992C) mutations identified previously. The rHPIV1 vaccine candidates exhibited a spectrum of attenuation in AGMs. One genetically and phenotypically stable vaccine candidate, rC(R84G/F170S)L(Y942A/L992C), was attenuated and efficacious in AGMs and is a promising live attenuated intranasal HPIV1 vaccine candidate suitable for clinical evaluation.


Subject(s)
Parainfluenza Vaccines/immunology , Parainfluenza Virus 1, Human/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Viral/blood , Disease Models, Animal , Humans , Macaca mulatta , Parainfluenza Vaccines/administration & dosage , Parainfluenza Vaccines/genetics , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 1, Human/pathogenicity , Point Mutation , Respirovirus Infections/prevention & control , Temperature , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Proteins/immunology
16.
Immunol Cell Biol ; 82(2): 119-26, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15061762

ABSTRACT

Gene therapy using DNA encoding type I IFN subtypes IFNA6, IFNA9 and IFNB suppresses murine cytomegalovirus (MCMV)-myocarditis, a predominantly cell-mediated disease in BALB/c mice. CD8(+) T cells are the principal cell type within the inflamed myocardium. As such, we investigated the effects of IFN subtype treatment on this T-cell subset and other cell types in the cardiac infiltrate. In the acute phase of disease, IFNA6 and IFNA9 treatments significantly reduced the number of CD8(+) T cells within the foci of cellular infiltration in the heart. During the chronic phase, which is primarily autoimmune in nature, IFNB treatment significantly reduced CD8(+) T cells. B-cell and neutrophil numbers in the cardiac infiltrate were also reduced following IFNB immunotherapy. Although early inflammatory responses are important for resolution of virus infection, high numbers of lymphocytes persisting in the myocardium may lead to exacerbation of disease. Our data suggests that type I IFN DNA therapy regulates cardiac cellular infiltration. Thus, treatment with IFN-beta administered prophylactically to high-risk patients in acquiring CMV infection may reduce the development of chronic autoimmune myocarditis.


Subject(s)
Autoimmune Diseases/therapy , Genetic Therapy , Interferon-alpha/genetics , Interferon-beta/genetics , Myocarditis/therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , DNA/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice , Myocarditis/immunology , Myocarditis/metabolism
17.
Biol Proced Online ; 5: 43-52, 2003.
Article in English | MEDLINE | ID: mdl-12734557

ABSTRACT

Type I interferon (IFN) gene therapy modulates the immune response leading to inflammatory heart disease following cytomegalovirus (CMV) infection in a murine model of post-viral myocarditis. Efficacy of different immunisation protocols for the IFN constructs was influenced by the dose of DNA, subtype choice, combination use, pre-medication, and timing of DNA administration. Optimal efficacy was found with bupivacaine treatment prior to DNA inoculation of 200mg IFN DNA 14 days prior to virus challenge. Maximal antiviral and antimyocarditic effects were achieved with this vaccination schedule. Furthermore, inoculation of synergistic IFN subtypes demonstrated enhanced efficacy when delivered either alone or with CMV gB DNA vaccination in the CMV model. Thus naked DNA delivery of IFN provides an avenue of immunotherapy for regulating herpesvirus-induced diseases.

18.
Blood ; 101(7): 2727-35, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12446459

ABSTRACT

Type I interferons (IFNs), pleiotropic cytokines with antiviral, antiproliferative, apoptotic, and immunoregulatory functions, are efficacious in the treatment of malignancies, viral infections, and autoimmune diseases. Binding of these cytokines to their cognate receptor leads to activation of the Jak-signal transducers and activators of transcription (STAT) signaling pathway and altered gene expression. This signal pathway has been intensely studied using human IFN-alpha 2 and IFN-beta. However, there are over 14 human IFN-alpha subtypes and over 10 murine IFN-alpha subtypes, with a single IFN-beta subtype in both species. J2E cells are immortalized at the proerythroblast stage of development and produce a rapid and fatal erythroleukemia in vivo. These cells retain the ability to respond to erythropoietin in vitro by proliferating, differentiating, and remaining viable in the absence of serum. Here, we show that J2E cells are also functionally regulated differentially by IFN subtype treatment in vitro. A novel finding was the selective activation of STAT and mitogen-activated protein kinase (MAPK) molecules by different subtypes binding the IFN receptor. These findings indicate distinct effects for individual type I IFN subtypes, which are able to differentially activate members of the STAT and MAPK family. Finally, we investigated the efficacy of IFN naked DNA therapy in treating J2E-induced erythroleukemia in athymic nude mice. IFN subtypes differentially regulated the onset of erythroleukemia with delayed onset and increased survival, possibly via a reduction in cell viability, and enhanced antiproliferative and apoptotic effects observed for IFNA6 and IFNA9 treatment, respectively. Moreover, these data highlight the necessity to choose the best IFN subtype in disease treatment.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon Type I/therapeutic use , Leukemia, Erythroblastic, Acute/therapy , Milk Proteins , Trans-Activators/metabolism , Animals , Cell Division/drug effects , DNA/administration & dosage , DNA/therapeutic use , DNA-Binding Proteins/drug effects , Genetic Therapy , Interferon Type I/genetics , Interferon Type I/pharmacology , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Protein Isoforms/pharmacology , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/drug effects , Survival Rate , Trans-Activators/drug effects , Treatment Outcome , Tumor Cells, Cultured
19.
Immunol Cell Biol ; 80(5): 425-35, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12225378

ABSTRACT

Delivery of type I IFN transgenes by naked DNA immunization can protect against cytomegalovirus infection and myocarditis. Here, we investigate IFN transgene expression, antiviral efficacy, and immunomodulation of myocarditis using various treatment regimes in a mouse CMV model. In vivo expression of the IFN transgene was observed in the sera for 35 days post-DNA inoculation. Prophylactic IFN-A6 and IFN-B DNA treatment for 14 days prior to murine cytomegalovirus (MCMV) infection was more efficacious in significantly reducing viral titres, than 2 days prior to or 2 days post-virus infection. Similarly, IFN-A6 DNA treatment commencing 14 days prior to virus infection was superior in suppressing both acute and chronic myocarditis. Furthermore, reduction of autoantibody titres was more pronounced when IFN was administered 14 days prior to viral infection. Combinational IFN gene therapy was assessed for synergy between IFN subtypes. Combination treatment with either IFN-A6/A9 or IFN-A6/B greatly reduced spleen viral titres while IFN-A6/B and IFN-A9/B reduced virus replication in the liver. Only IFN-A6/A9 and IFN-A9/B reduced acute viral myocarditis, whereas IFNA6/B treatment was most efficacious for autoimmune chronic myocarditis. Finally, treatment with IFN-A6 DNA 2 weeks post-MCMV infection proved effective at inhibiting the development of chronic autoimmune myocarditis. These findings suggest that immunomodulation of both antiviral and autoimmune responses by IFN DNA immunization may be an avenue for improved viral immunotherapy.


Subject(s)
Autoimmune Diseases/prevention & control , Cytomegalovirus Infections/prevention & control , Immunotherapy, Active , Interferon Type I/genetics , Interferon-beta/genetics , Muromegalovirus , Myocarditis/prevention & control , Vaccines, DNA/therapeutic use , Acute Disease , Animals , Autoantibodies/biosynthesis , Autoantibodies/immunology , Autoimmune Diseases/therapy , Autoimmune Diseases/virology , COS Cells , Chlorocebus aethiops , Chronic Disease , Cytokines/biosynthesis , Cytomegalovirus Infections/therapy , DNA, Complementary/genetics , Disease Models, Animal , Immunization Schedule , Injections, Intramuscular , Interferon Type I/physiology , Interferon-alpha , Interferon-beta/physiology , Mice , Mice, Inbred BALB C , Muromegalovirus/physiology , Muscle, Skeletal/physiology , Myocarditis/therapy , Myocarditis/virology , Regeneration , Specific Pathogen-Free Organisms , Spleen/virology , Transgenes , Vaccines, DNA/immunology , Virus Replication
20.
Immunology ; 106(3): 428-37, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12100732

ABSTRACT

Type I interferons (IFNs) are produced early in response to viral infection and modulate adaptive immunity. Previously we demonstrated localized protection against murine cytomegalovirus (MCMV) infection in IFN DNA-inoculated mice. Here we examine the effect of seven IFN subtypes (IFNA1, A2, A4, A5, A6, A9 and B), administered by DNA inoculation, on systemic MCMV infection and myocarditis. IFN transgene expression altered the pathogenesis of MCMV infection with regard to virus titre and myocarditis. IFNA6 treatment reduced MCMV replication whilst IFNA5 and A2 enhanced virus replication. IFNA6, A9, and B treatment inhibited acute myocarditis. A T helper type 1-like, antibody and cytokine, response correlated with decreased virus titre and myocarditis. In addition, IFNA6 was able to reduce chronic cardiac inflammation. This research into the effectiveness of seven type I IFNs, using DNA gene therapy, highlights the need for correct subtype usage in the treatment of disease. We demonstrate effective subtypes for treatment in both the acute and chronic phases of MCMV infection and the resultant development of myocarditis.


Subject(s)
Genetic Therapy/methods , Herpesviridae Infections/therapy , Interferon Type I/genetics , Muromegalovirus/immunology , Myocarditis/prevention & control , Acute Disease , Animals , Chronic Disease , Cytokines/biosynthesis , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Immunoglobulin G/biosynthesis , Interferon Type I/immunology , Male , Mice , Mice, Inbred BALB C , Muromegalovirus/physiology , Myocarditis/immunology , Myocarditis/virology , Myosins/immunology , Transgenes/immunology , Viral Load , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...