Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 304
Filter
1.
J Virol ; 86(1): 420-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013033

ABSTRACT

La Crosse virus (LACV) and Jamestown Canyon virus (JCV), family Bunyaviridae, are mosquito-borne viruses that are endemic in North America and recognized as etiologic agents of encephalitis in humans. Both viruses belong to the California encephalitis virus serogroup, which causes 70 to 100 cases of encephalitis a year. As a first step in creating live attenuated viral vaccine candidates for this serogroup, we have generated a recombinant LACV expressing the attachment/fusion glycoproteins of JCV. The JCV/LACV chimeric virus contains full-length S and L segments derived from LACV. For the M segment, the open reading frame (ORF) of LACV is replaced with that derived from JCV and is flanked by the untranslated regions of LACV. The resulting chimeric virus retained the same robust growth kinetics in tissue culture as observed for either parent virus, and the virus remains highly infectious and immunogenic in mice. Although both LACV and JCV are highly neurovirulent in 21 day-old mice, with 50% lethal dose (LD50) values of 0.1 and 0.5 log10 PFU, respectively, chimeric JCV/LACV is highly attenuated and does not cause disease even after intracerebral inoculation of 10³ PFU. Parenteral vaccination of mice with 10¹ or 10³ PFU of JCV/LACV protected against lethal challenge with LACV, JCV, and Tahyna virus (TAHV). The chimeric virus was infectious and immunogenic in rhesus monkeys and induced neutralizing antibodies to JCV, LACV, and TAHV. When vaccinated monkeys were challenged with JCV, they were protected against the development of viremia. Generation of highly attenuated yet immunogenic chimeric bunyaviruses could be an efficient general method for development of vaccines effective against these pathogenic viruses.


Subject(s)
Chimera/immunology , Encephalitis Virus, California/genetics , Encephalitis, California/prevention & control , Gene Expression , La Crosse virus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Chimera/genetics , Chimera/growth & development , Encephalitis, California/immunology , Encephalitis, California/virology , Humans , La Crosse virus/genetics , La Crosse virus/growth & development , Macaca mulatta , Mice , Vaccination , Viral Envelope Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
2.
J Fish Biol ; 76(3): 538-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20666895

ABSTRACT

Initial relative mass (W(R), low v. high) and energetic trajectory in time (starved v. fed) were experimentally manipulated in bluegill Lepomis macrochirus. Fed fish starting at low W(R) grew more and gained more W(R) than fed fish starting at high W(R). Similarly, starved fish starting at high W(R) lost more mass and W(R) than did starved fish starting at low W(R). Temporal changes in other variables did not consistently match that of W(R), but, by the end of the experiment, proximate composition showed a high correlation to W(R). Regression slopes of W(R) on proximate composition increased with time in the laboratory. Differences between wild and laboratory fish appeared to result from relaxation of environmental stress. When excess resources are available such that L. macrochirus grow, condition indices will increase, but individual response will depend on initial values and thus past environmental experience.


Subject(s)
Body Composition , Environment , Nutritional Status , Perciformes/growth & development , Perciformes/physiology , Animals , Body Size
3.
Gene Ther ; 14(4): 304-15, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17024103

ABSTRACT

Single-stranded oligodeoxynucleotide (ssODN) gene targeting may facilitate animal model creation and gene repair therapy. Lipofection of ssODN can introduce point mutations into target genes. However, typical efficiencies in mouse embryonic stem cells (ESC) are <10(-4), leaving corrections too rare to effectively identify. We developed ESC lines with an integrated mutant neomycin resistance gene (Tyr22Ter). After targeting with ssODN, repaired cells survive selection in G418. Correction efficiencies varied with different lipofection procedures, clonal lines, and ssODN designs, ranging from 1 to 100 corrections per million cells plated. Uptake studies using cell sorting of Cy5-labelled ssODN showed 40% of the corrections concentrated in the best transfected 22% of cells. Four different basepair mismatches were tested and results show that the base-specificity of the mismatch is critical. Dual mismatch ssODN also showed mismatch preferences. These ESC lines may facilitate development of improved ssODN targeting technologies for either animal production or ex vivo gene therapy.


Subject(s)
DNA, Single-Stranded , Embryonic Stem Cells/metabolism , Gene Targeting/methods , Genetic Engineering , Animals , Base Sequence , Chimera , DNA Repair , Drug Resistance/genetics , Female , Liposomes , Mice , Mice, Transgenic , Models, Animal , Molecular Sequence Data , Neomycin , Transfection/methods
4.
Ann Oncol ; 17(1): 29-34, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16303863

ABSTRACT

PURPOSE: The synergic combination of oxaliplatin and capecitabine has demonstrated activity against various gastrointestinal cancers, including colon cancer. We therefore undertook this phase II study to test this first-line combination in patients with metastatic adenocarcinoma of the esophagus, gastroesophageal junction and gastric cardia. PATIENTS AND METHODS: Forty-three patients with histologic or cytologic confirmation of the above malignancy were recruited. The cohort had Eastern Cooperative Oncology Group performance statuses of 0, 1 and 2 in 47%, 51%, and 2%, respectively. Median age was 61 years (range 32-80). All had adequate organ function. Initially, patients were prescribed 130 mg/m2 intravenously on day 1 and capecitabine 1000 mg/m2 orally twice a day, on days 1-14 of a 21-day cycle. Four treatment-related deaths in the first 24 patients led to a reduction in capecitabine to 850 mg/m2 orally twice a day, days 1-14, for the remainder of the cohort. RESULTS: The tumor response rate was 35% [95% confidence intervals (CI) 23% to 50%]. All responses were partial; seven of 24 occurred before the capecitabine dose reduction, and eight of 19 after. Median time to tumor progression was 4 months (95% CI 3.1-4.6), and median survival 6.4 months (95% CI 4.6-10). To date, there have been 36 deaths. Four were treatment-related (one infection, two myocardial infarctions, one respiratory failure), and all occurred before the capecitabine dose reduction. Notable grade 4 events from the entire cohort included diarrhea (two patients), vomiting (three), dyspnea (one), thrombosis (two) and anorexia (two). Grade 3 events included nausea (12 patients), diarrhea (12), fatigue (10), abdominal pain (seven), vomiting (six), dyspnea (six), hypokalemia (six), dehydration (five), hypokalemia (five) and infection (four). CONCLUSIONS: Oxaliplatin and capecitabine in combination demonstrates activity in metastatic adenocarcinoma of the esophagus, gastroesophageal junction and gastric cardia. The lower dose (capecitabine 850 mg/m2 orally twice a day, days 1-14, and oxaliplatin 130 mg/m2 intravenously on day 1) yielded an acceptable toxicity profile and merits further study.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/secondary , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cardia/drug effects , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/drug effects , Adult , Aged , Aged, 80 and over , Capecitabine , Cardia/pathology , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Dose-Response Relationship, Drug , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Female , Fluorouracil/analogs & derivatives , Humans , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Treatment Outcome
5.
Avian Dis ; 47(3 Suppl): 1127-30, 2003.
Article in English | MEDLINE | ID: mdl-14575127

ABSTRACT

H9N2 subtype avian influenza viruses have been identified in avian species worldwide, and infections in pigs were confirmed in Hong Kong in 1998. Subsequently, H9N2 viruses were isolated from two children in Hong Kong in 1999, and five human infections were reported from China, raising the possibility that H9N2 viruses pose a potential pandemic threat for humans. These events prompted us to develop a vaccine candidate to protect humans against this subtype of influenza A viruses. Reassortant H1N1 and H3N2 human influenza A viruses with the six internal gene segments of A/Ann Arbor/6/60 (H2N2)(AA) cold-adapted (ca) virus have been tested extensively in humans and have proved to be attenuated and safe as live virus vaccines. Using classical genetic reassortment, we generated a reassortant that contains the hemagglutinin and neuraminidase genes from A/chicken/Hong Kong/G9/97 (H9N2) and six internal gene segments from the AAca virus. The G9/AAca reassortant virus exhibits the ca phenotype and the temperature-sensitive phenotypes of the AAca virus and was attenuated in mice. The reassortant virus was immunogenic and protected mice from wild-type H9N2 virus challenge. The G9/AAca virus bears the in vitro and in vivo phenotypes specified by the AAca virus and will be evaluated as a potential vaccine candidate in humans.


Subject(s)
Influenza A Virus, H9N2 Subtype/immunology , Influenza Vaccines , Reassortant Viruses/immunology , Acclimatization , Animals , Antigens, Viral/analysis , Child , Cold Temperature , Ferrets , Hong Kong , Humans , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza A Virus, H9N2 Subtype/physiology , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Reassortant Viruses/physiology , Reverse Transcriptase Polymerase Chain Reaction , Swine/virology
6.
Vaccine ; 21(27-30): 4430-6, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14505926

ABSTRACT

H9N2 subtype influenza A viruses have been identified in avian species worldwide and were isolated from humans in 1999, raising concerns about their pandemic potential and prompting the development of candidate vaccines to protect humans against this subtype of influenza A virus. Reassortant H1N1 and H3N2 human influenza A viruses with the internal genes of the influenza A/Ann Arbor/6/60 (H2N2) (AA) cold-adapted (ca) virus have proven to be attenuated and safe as live virus vaccines in humans. Using classical genetic reassortment, we generated a reassortant virus (G9/AA ca) that contains the hemagglutinin and neuraminidase genes from influenza A/chicken/Hong Kong/G9/97 (H9N2) (G9) and six internal gene segments from the AA ca virus. When administered intranasally, the reassortant virus was immunogenic and protected mice from subsequent challenge with wild-type H9N2 viruses, although it was restricted in replication in the respiratory tract of mice. The G9/AA ca virus bears properties that are desirable in a vaccine for humans and is available for clinical evaluation and use, should the need arise.


Subject(s)
Adaptation, Physiological/physiology , Influenza A Virus, H9N2 Subtype , Influenza A virus/immunology , Influenza Vaccines/immunology , Animals , Chickens/immunology , Cold Temperature , Female , Ferrets/immunology , Influenza A virus/genetics , Mice , Mice, Inbred BALB C , Phenotype , Vaccines, Attenuated/immunology , Virus Replication
7.
Med Oncol ; 20(3): 283-90, 2003.
Article in English | MEDLINE | ID: mdl-14514978

ABSTRACT

No effective treatment exists for primary amyloidosis, a plasma cell dyscrasia characterized by deposition of amyloid fibrils consisting of monoclonal light chains in various organs. TNF-alpha has been implicated in other amyloid disorders; therefore, we used etanercept to treat patients with advanced amyloidosis who had failed other therapies or were ineligible for other treatment regimens. Sixteen patients with amyloidosis that included patients with severe cardiac or multiple organ involvement were treated with etanercept and evaluated every 4-6 wk for evidence of toxicity and clinical response. Patients were treated with etanercept for a median of 42 wk. Eight of 16 patients (50%) experienced objective improvements and 14 patients (88%) experienced subjective improvements in symptoms. Only one patient experienced an adverse effect attributable to etanercept. For the entire group, improvement in performance status was statistically significant (p = 0.001), estimated median survival is 24.2 mo, 8 of whom are still alive with a median survival is 26.6 mo. The 12 patients with any cardiac involvement had an estimated median survival of 24.2 mo. Six of those 12 patients are still alive, with a median survival is 26.6 mo. The group of eight patients with severe cardiac involvement showed an estimated median survival of 13.2 mo, three of whom are still alive with a median survival is 25.9 mo. The clinical observations in this group of advanced and relapsed/refractory patients are highly encouraging. For the group as a whole, median survival was 24.2 mo and improvement in performance status was highly significant. Median survival for the patients with severe cardiac involvement was 13.2 mo with 3/8 patients are alive with a median survival of 25+ mo. Moreover, there was a statistically significant improvement in patients' performance status. These results, even though in a small group of patients, suggest that etanercept may provide a new therapeutic option for the management of amyloidosis that should be studied further.


Subject(s)
Amyloidosis/drug therapy , Antirheumatic Agents/therapeutic use , Immunoglobulin G/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , Adult , Aged , Aged, 80 and over , Antirheumatic Agents/administration & dosage , Etanercept , Female , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Humans , Immunoglobulin G/administration & dosage , Male , Middle Aged , Receptors, Tumor Necrosis Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Salvage Therapy , Survival Rate , Treatment Outcome
8.
Arch Virol ; 148(5): 999-1006, 2003 May.
Article in English | MEDLINE | ID: mdl-12721805

ABSTRACT

Using reverse genetics, it is possible to readily add well-defined attenuating mutations to the genome of wild type or incompletely attenuated dengue (DEN) viruses to generate vaccine candidates that exhibit the desired balance between attenuation and immunogenicity. Here, we describe the identification of eight temperature sensitive missense mutations distributed in four non-structural protein genes that specify a 60- to 10,000-fold range of restricted replication in the suckling mouse brain compared to wild type recombinant DEN4 virus. The usefulness of such mutations in flavivirus vaccine design and development is discussed.


Subject(s)
Brain/virology , Dengue Virus/genetics , Dengue Virus/physiology , Genes, Viral/genetics , Mutation, Missense/genetics , Viral Nonstructural Proteins/genetics , Virus Replication , Animals , Animals, Suckling , Humans , Mice , Phenotype , Temperature
9.
J Infect Dis ; 186(6): 834-8, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12198619

ABSTRACT

Thirty-two patients undergoing allogeneic hematopoietic stem-cell transplantation were given respiratory syncytial virus (RSV) immune globulin (RSVIG) at the time of transplantation and again 3 weeks later. Antibody titers to RSV, human parainfluenza virus 3, measles, and influenza H1N1, H3N2, and B were measured prior to administration of RSVIG and 6 more times over the course of the subsequent 6 weeks. Baseline antiviral titers and increases in antibody after administration of RSVIG were extremely variable for all the viruses. In 18 patients in whom the baseline titers of antibody titers to RSV-F protein were 1:640-1:2048, there was a 7.7-fold initial increase in these titers after the first dose of RSVIG, compared with a 2.1-fold increase in 14 patients with baseline titers of 1:4096-1:20,840; increases in titers of antibody against the other viruses after the first dose of RSVIG reflected similar variability. The subset of patients with the lowest titers appear to receive the greatest benefit from administration of RSVIG.


Subject(s)
Antibodies, Viral/immunology , Hematopoietic Stem Cell Transplantation , Immunization, Passive , Immunoglobulin G/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Antibodies, Viral/administration & dosage , Humans , Immunoglobulin G/administration & dosage , Immunoglobulins, Intravenous/administration & dosage , Immunoglobulins, Intravenous/immunology , Prospective Studies
10.
Am J Trop Med Hyg ; 65(5): 405-13, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11716091

ABSTRACT

The recombinant dengue virus type-4 vaccine candidate 2AA30 was attenuated in rhesus monkeys due to an engineered 30-nucleotide deletion in the 3'-untranslated region of the viral genome. A clinical trial to evaluate the safety and immunogenicity of a single dose of 2Adelta30 was conducted with 20 adult human volunteers. The vaccine candidate was well tolerated and did not cause systemic illness in any of the 20 volunteers. Viremia was detectable in 14 volunteers at a mean level of 1.6 log10 plaque-forming units/ml of serum, although all 20 volunteers seroconverted with a seven-fold or greater increase in serum neutralizing antibody titer on day 28 post-vaccination (mean titer = 1:580). A mild, asymptomatic, macular rash developed in 10 volunteers, and a transient elevation in the serum level of alanine aminotransferase was noted in five volunteers. The low level of reactogenicity and high degree of immunogenicity of this vaccine candidate warrant its further evaluation and its use to create chimeric vaccine viruses expressing the structural genes of dengue virus types 1, 2, and 3.


Subject(s)
3' Untranslated Regions/physiology , Dengue Virus/immunology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adult , Animals , Dengue Virus/genetics , Dengue Virus/physiology , Humans , Immunization , Macaca mulatta , Vaccines, Attenuated/immunology , Virus Replication
11.
Am J Trop Med Hyg ; 65(5): 414-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11716092

ABSTRACT

2Adelta30 is a live dengue-4 virus vaccine candidate with a 30-nucleotide deletion in its 3'-untranslated region. To assess the transmissibility of 2Adelta30 by mosquitoes, we compared its in vivo replication in mosquitoes with that of its wild type DEN-4 parent. Both the vaccine candidate and wild type virus were equally able to infect the mosquito Toxorhynchites splendens after intrathoracic inoculation. Relative to its wild type parent, 2Adelta30 was slightly restricted in its ability to infect the midgut of Aedes aegypti mosquitoes fed on an artificial blood meal and was even more restricted in its ability to disseminate from the midgut to the salivary glands. Thus, the 30-nucleotide deletion rendered the vaccine candidate more sensitive than its wild type parent to the mosquito midgut escape barrier. Most significantly, 2Adelta30 was not transmitted to 352 Ae. albopictus mosquitoes fed on 10 vaccinees, all of whom were infected with the vaccine candidate.


Subject(s)
Aedes/virology , Dengue Virus/immunology , Dengue/transmission , Insect Vectors/virology , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adult , Animals , Dengue Virus/physiology , Female , Humans , Vaccination , Vaccines, Attenuated/immunology , Virus Replication
12.
J Virol ; 75(24): 12128-40, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11711604

ABSTRACT

An obstacle to developing a vaccine against human respiratory syncytial virus (RSV) is that natural infection typically does not confer solid immunity to reinfection. To investigate methods to augment the immune response, recombinant RSV (rRSV) was constructed that expresses murine granulocyte-macrophage colony-stimulating factor (mGM-CSF) from a transcription cassette inserted into the G-F intergenic region. Replication of rRSV/mGM-CSF in the upper and lower respiratory tracts of BALB/c mice was reduced 23- to 74- and 5- to 588-fold, respectively, compared to that of the parental rRSV. Despite this strong attenuation of replication, the level of RSV-specific serum antibodies induced by rRSV/mGM-CSF was comparable to, or marginally higher than, that of the parental rRSV. The induction of RSV-specific CD8(+) cytotoxic T cells was moderately reduced during the initial infection, which might be a consequence of reduced antigen expression. Mice infected with rRSV/mGM-CSF had elevated levels of pulmonary mRNA for gamma interferon (IFN-gamma) and interleukin 12 (IL-12) p40 compared to animals infected by wild-type rRSV. Elevated synthesis of IFN-gamma could account for the restriction of RSV replication, as was observed previously with an IFN-gamma-expressing rRSV. The accumulation of total pulmonary mononuclear cells and total CD4(+) T lymphocytes was accelerated in animals infected with rRSV/mGM-CSF compared to that in animals infected with the control virus, and the level of IFN-gamma-positive or IL-4-positive pulmonary CD4(+) cells was elevated approximately twofold. The number of pulmonary lymphoid and myeloid dendritic cells and macrophages was increased up to fourfold in mice infected with rRSV/mGM-CSF compared to those infected with the parental rRSV, and the mean expression of major histocompatibility complex class II molecules, a marker of activation, was significantly increased in the two subsets of dendritic cells. Enhanced antigen presentation likely accounts for the maintenance of a strong antibody response despite reduced viral replication and would be a desirable property for a live attenuated rRSV vaccine.


Subject(s)
Antigen-Presenting Cells/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lung/immunology , Respiratory Syncytial Virus Vaccines/immunology , Vaccines, Synthetic/immunology , Virus Replication , Animals , Antibodies, Viral/biosynthesis , Female , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred BALB C , Recombinant Proteins/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Attenuated/immunology
13.
J Virol ; 75(21): 10498-504, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11581420

ABSTRACT

The chimeric recombinant virus rHPIV3-N(B), a version of human parainfluenza virus type 3 (HPIV3) that is attenuated due to the presence of the bovine PIV3 nucleocapsid (N) protein open reading frame (ORF) in place of the HPIV3 ORF, was modified to encode the measles virus hemagglutinin (HA) inserted as an additional, supernumerary gene between the HPIV3 P and M genes. This recombinant, designated rHPIV3-N(B)HA, replicated like its attenuated rHPIV3-N(B) parent virus in vitro and in the upper and lower respiratory tracts of rhesus monkeys, indicating that the insertion of the measles virus HA did not further attenuate rHPIV3-N(B) in vitro or in vivo. Monkeys immunized with rHPIV3-N(B)HA developed a vigorous immune response to both measles virus and HPIV3, with serum antibody titers to both measles virus (neutralizing antibody) and HPIV3 (hemagglutination inhibiting antibody) of over 1:500. An attenuated HPIV3 expressing a major protective antigen of measles virus provides a method for immunization against measles by the intranasal route, a route that has been shown with HPIV3 and respiratory syncytial virus vaccines to be relatively refractory to the neutralizing and immunosuppressive effects of maternally derived virus-specific serum antibodies. It should now be possible to induce a protective immune response against measles virus in 6-month-old infants, an age group that in developing areas of the world is not responsive to the current measles virus vaccine.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Parainfluenza Virus 3, Human/genetics , Respirovirus/genetics , Vaccines, Synthetic/immunology , Virus Replication , Animals , Antibodies, Viral/blood , Cattle , Chimera , Humans , Immunization , Macaca mulatta , Parainfluenza Virus 3, Human/growth & development , Parainfluenza Virus 3, Human/immunology , Respiratory System/virology , Vaccines, Attenuated/immunology
14.
J Virol ; 75(20): 9731-40, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11559806

ABSTRACT

A recombinant live attenuated dengue virus type 4 (DEN4) vaccine candidate, 2ADelta30, was found previously to be generally well tolerated in humans, but a rash and an elevation of liver enzymes in the serum occurred in some vaccinees. 2ADelta30, a non-temperature-sensitive (non-ts) virus, contains a 30-nucleotide deletion (Delta30) in the 3' untranslated region (UTR) of the viral genome. In the present study, chemical mutagenesis of DEN4 was utilized to generate attenuating mutations which may be useful in further attenuation of the 2ADelta30 candidate vaccine. Wild-type DEN4 2A virus was grown in Vero cells in the presence of 5-fluorouracil, and a panel of 1,248 clones were isolated. Twenty ts mutant viruses were identified that were ts in both simian Vero and human liver HuH-7 cells (n = 13) or only in HuH-7 cells (n = 7). Each of the 20 ts mutant viruses possessed an attenuation phenotype, as indicated by restricted replication in the brains of 7-day-old mice. The complete nucleotide sequence of the 20 ts mutant viruses identified nucleotide substitutions in structural and nonstructural genes as well as in the 5' and 3' UTRs, with more than one change occurring, in general, per mutant virus. A ts mutation in the NS3 protein (nucleotide position 4995) was introduced into a recombinant DEN4 virus possessing the Delta30 deletion, thereby creating rDEN4Delta30-4995, a recombinant virus which is ts and more attenuated than rDEN4Delta30 virus in the brains of mice. We are assembling a menu of attenuating mutations that should be useful in generating satisfactorily attenuated recombinant dengue vaccine viruses and in increasing our understanding of the pathogenesis of dengue virus.


Subject(s)
Dengue Virus/drug effects , Dengue/virology , Fluorouracil/pharmacology , Mutagens/pharmacology , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Animals, Suckling , Brain/virology , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Dengue Virus/genetics , Dengue Virus/physiology , Genes, Viral , Genome, Viral , Humans , Mice , Molecular Sequence Data , RNA Helicases , Recombination, Genetic , Serine Endopeptidases , Temperature , Vaccines, Attenuated/genetics , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Vaccines/genetics , Virus Replication
15.
J Immunol ; 167(7): 3910-8, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564809

ABSTRACT

A respiratory syncytial virus (RSV) vaccine will need to be administered by 1 mo of age to protect young infants; therefore, it will need to be effective in the presence of maternally acquired RSV Abs. In the present study, the immunogenicity and efficacy of two live attenuated RSV vaccine candidates of different level of attenuation were evaluated in mice passively immunized with varying quantities of RSV Abs. The replication of the RSV vaccines was suppressed in the lower, but not the upper, respiratory tract of the passively immunized mice. Immunization with either vaccine candidate was highly efficacious against challenge with wild-type RSV in both passively immunized and control mice. Nonetheless, a high level of immunity was seen even in passively/actively immunized animals that failed to develop a humoral immune response, suggesting that T cells mediated the immunity. Depletion of CD4+ and CD8+ T cells in passively/actively immunized and control animals at the time of challenge with wild-type RSV demonstrated that CD4+ and CD8+ T cells made significant independent contributions to the restriction of replication of RSV challenge virus in both the upper and lower respiratory tracts. Although passively acquired serum RSV Abs suppressed the primary systemic and mucosal Ab responses of IgM, IgG, and IgA isotypes, B lymphocytes were nevertheless primed for robust secondary Ab responses. Thus, immunity mediated by CD4+ and CD8+ T cells and Abs can be readily induced in mice by live RSV vaccine candidates in the presence of physiologic levels of RSV neutralizing Abs.


Subject(s)
Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Respiratory Syncytial Virus Infections/therapy , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes/immunology , Animals , Cell Line , Immunity, Mucosal , Immunization, Passive , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Vaccines, Attenuated/immunology , Virus Replication
16.
Vaccine ; 19(27): 3620-31, 2001 Jun 14.
Article in English | MEDLINE | ID: mdl-11395195

ABSTRACT

PIV1 and PIV2 are important agents of pediatric respiratory tract disease. We are developing live-attenuated vaccines against these viruses. We earlier constructed a PIV3/PIV1 antigenic chimeric virus, designated rPIV3-1, in which the hemagglutinin-neuraminidase (HN) and fusion (F) proteins of wild type rPIV3 were replaced by their PIV1 counterparts. In the present study, rPIV3-1 was used as a vector to express the HN protein of PIV2 to generate a single virus capable of inducing immunity to both PIV1 and PIV2. The PIV2 HN open reading frame was expressed from an extra gene cassette, under the control of PIV3 cis-acting transcription signals, inserted between the F and HN genes of rPIV3-1. The recombinant derivative, designated rPIV3-1.2HN, was readily recovered and exhibited a level of temperature sensitivity and in vitro growth similar to that of its parental virus. The rPIV3-1.2HN virus was restricted in replication in both the upper and lower respiratory tracts of hamsters compared with rPIV3-1, identifying an attenuating effect of the PIV2 HN insert in hamsters. rPIV3-1.2HN elicited serum antibodies to both PIV1 and PIV2 and induced resistance against challenge with wild type PIV1 or PIV2. Thus, rPIV3-1.2HN, a virus attenuated solely by the insertion of the PIV2 HN gene, functioned as a live attenuated bivalent vaccine candidate against both PIV1 and PIV2.


Subject(s)
Parainfluenza Vaccines/biosynthesis , Parainfluenza Virus 1, Human/immunology , Parainfluenza Virus 2, Human/immunology , Vaccines, DNA/biosynthesis , Animals , Cell Line , Cricetinae , Humans , Parainfluenza Vaccines/genetics , Parainfluenza Vaccines/therapeutic use , Parainfluenza Virus 1, Human/genetics , Parainfluenza Virus 2, Human/genetics , Respirovirus Infections/prevention & control , Vaccines, Attenuated/biosynthesis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/therapeutic use , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/therapeutic use
18.
J Virol ; 75(10): 4594-603, 2001 May.
Article in English | MEDLINE | ID: mdl-11312329

ABSTRACT

Recombinant bovine/human parainfluenza virus type 3 (rB/HPIV3), a recombinant bovine PIV3 (rBPIV3) in which the F and HN genes were replaced with their HPIV3 counterparts, was used to express the major protective antigens of respiratory syncytial virus (RSV) in order to create a bivalent mucosal vaccine against RSV and HPIV3. The attenuation of rB/HPIV3 is provided by the host range restriction of the BPIV3 backbone in primates. RSV G and F open reading frames (ORFs) were placed under the control of PIV3 transcription signals and inserted individually into the rB/HPIV3 genome in the promoter-proximal position preceding the nucleocapsid protein gene. The recombinant PIV3 expressing the RSV G ORF (rB/HPIV3-G1) was not restricted in its replication in vitro, whereas the virus expressing the RSV F ORF (rB/HPIV3-F1) was eightfold restricted compared to its rB/HPIV3 parent. Both viruses replicated efficiently in the respiratory tract of hamsters, and each induced RSV serum antibody titers similar to those induced by RSV infection and anti-HPIV3 titers similar to those induced by HPIV3 infection. Immunization of hamsters with rB/HPIV3-G1, rB/HPIV3-F1, or a combination of both viruses resulted in a high level of resistance to challenge with RSV or HPIV3 28 days later. These results describe a vaccine strategy that obviates the technical challenges associated with a live attenuated RSV vaccine, providing, against the two leading viral agents of pediatric respiratory tract disease, a bivalent vaccine whose attenuation phenotype is based on the extensive host range sequence differences of BPIV3.


Subject(s)
Antigens, Viral/immunology , Genetic Vectors/immunology , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/immunology , Respirovirus Infections/prevention & control , Respirovirus/immunology , Vaccines, Synthetic/immunology , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology , Viral Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Antigens, Viral/genetics , Base Sequence , Cattle , Cell Line , Cricetinae , DNA, Viral , Gene Expression , Genetic Vectors/genetics , Genetic Vectors/physiology , Humans , Immunity, Mucosal , Macaca mulatta , Molecular Sequence Data , Mutagenesis, Insertional , Open Reading Frames , Parainfluenza Vaccines/genetics , Parainfluenza Virus 3, Human/genetics , Recombination, Genetic , Respiratory System/metabolism , Respirovirus/genetics , Respirovirus/physiology , Tumor Cells, Cultured , Vaccination , Vaccines, Synthetic/genetics , Viral Envelope Proteins/genetics , Viral Fusion Proteins/genetics , Viral Proteins/genetics , Virus Replication
19.
J Natl Cancer Inst ; 93(4): 284-92, 2001 Feb 21.
Article in English | MEDLINE | ID: mdl-11181775

ABSTRACT

BACKGROUND: Studies in animal models have shown that systemic immunization with a papillomavirus virus-like particle (VLP) vaccine composed of L1, a major structural viral protein, can confer protection against subsequent experimental challenge with the homologous virus. Here we report results of a double-blind, placebo-controlled, dose-escalation trial to evaluate the safety and immunogenicity of a human papillomavirus (HPV) type 16 (HPV16) L1 VLP vaccine in healthy adults. METHODS: Volunteers were given intramuscular injections with placebo or with 10- or 50-microg doses of HPV16 L1 VLP vaccine given without adjuvant or with alum or MF59 as adjuvants at 0, 1, and 4 months. All vaccine recipients were monitored for clinical signs and symptoms for 7 days after each inoculation. Immune responses were measured by an HPV16 L1 VLP-based enzyme-linked immunosorbent assay (ELISA) and by an HPV16 pseudovirion neutralization assay. The antibody titers were given as the reciprocals of the highest dilution showing positive reactivity in each assay. All statistical tests were two-sided. RESULTS: The prevaccination geometric mean ELISA titer for six seropositive individuals was 202 (range, 40--640). All vaccine formulations were well tolerated, and all subjects receiving vaccine seroconverted. Serum antibody responses at 1 month after the third injection were dose dependent in recipients of vaccine without adjuvant or with MF59 but were similar at both doses when alum was the adjuvant. With the higher dose, the geometric means of serum ELISA antibody titers (95% confidence intervals) to purified VLP 1 month after the third injection were as follows: 10,240 (1499 to 69 938) without adjuvant, 10,240 (1114 to 94 145) with MF59, and 2190 (838 to 5723) with alum. Responses of subjects within each group were similar. Neutralizing and ELISA antibody titers were highly correlated (Spearman correlation =.85), confirming that ELISA titers are valid proxies for neutralizing antibodies. CONCLUSIONS: The HPV16 L1 VLP vaccine is well tolerated and is highly immunogenic even without adjuvant, with the majority of the recipients achieving serum antibody titers that were approximately 40-fold higher than what is observed in natural infection.


Subject(s)
Antibodies, Viral/blood , Papillomaviridae/immunology , Papillomavirus Vaccines , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adult , Alum Compounds/administration & dosage , Baculoviridae , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization Schedule , Immunoglobulins/blood , Injections, Intramuscular , Male , Polysorbates/administration & dosage , Recombinant Proteins , Reference Values , Squalene/administration & dosage , Viral Vaccines/administration & dosage
20.
J Virol ; 74(23): 11017-26, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11069997

ABSTRACT

The intergenic sequences (IGS) between the first nine genes of human respiratory syncytial virus (RSV) vary in length from 1 to 56 nucleotides and lack apparent conserved sequence motifs. To investigate their influence on sequential transcription and viral growth, recombinant RSV strain A2, from which the SH gene had been deleted to facilitate manipulation, was further modified to contain an M-G IGS of 16, 30, 44, 58, 65, 72, 86, 100, 120, 140, or 160 nucleotides. All of the viruses were viable. For viruses with an M-G IGS of 100 nucleotides or more, plaque size decreased with increasing IGS length. In this same length range, increasing IGS length was associated with modest attenuation during single-step, but not multistep, growth in HEp-2 cells. Surprisingly, Northern blot analysis of the accumulation of six different mRNAs indicated that there was little or no change in transcription with increasing IGS length. Thus, the RSV polymerase apparently can readily cross IGS of various lengths, including unnaturally long ones, with little or no effect on the efficiency of termination and reinitiation. This finding supports the view that the IGS do not have much effect on sequential transcription and provides evidence from infectious virus that IGS length is not an important regulatory feature. To evaluate replication in vivo, BALB/c mice were infected intranasally with RSV containing an M-G IGS of 65, 140, or 160 nucleotides. Replication of the latter two viruses was decreased up to 5- and 25-fold in the upper and lower respiratory tracts, respectively, on day 3 following infection. However, the level of replication at both sites on days 4 and 5 was very similar to that of the virus with an IGS of 65 nucleotides. Thus, the modest attenuation in vivo associated with the longer IGS was additive to that conferred by deletion of the SH gene and might be useful to incrementally increase the level of attenuation of a live-attenuated vaccine virus.


Subject(s)
Respiratory Syncytial Viruses/genetics , Transcription, Genetic , Virus Replication , Animals , Base Sequence , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Respiratory Syncytial Viruses/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...