Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Vaccine ; 32(9): 1113-20, 2014 Feb 19.
Article in English | MEDLINE | ID: mdl-24342254

ABSTRACT

Pneumococcal infections impose a large burden of disease on the human population, mainly in developing countries, and the current pneumococcal vaccines offer serotype-specific protection, but do not cover all pathogenic strains, leaving populations vulnerable to disease caused by non-vaccine serotypes. The pneumococcal whole cell vaccine is a low-cost strategy based on non-capsular antigens common to all strains, inducing serotype-independent immunity. Therefore, we developed the process for the cGMP production of this cellular vaccine. Initially, three engineering runs and two cGMP runs were performed in 60-L bioreactors, demonstrating the consistency of the production process, as evaluated by the growth curves, glucose consumption and metabolite formation (lactate and acetate). Cell recovery by tangential filtration was 92 ± 13 %. We optimized the conditions for beta-propiolactone (BPL) inactivation of the bacterial suspensions, establishing a maximum cell density of OD600 between 27 and 30, with a BPL concentration of 1:4000 (v/v) at 150 rpm and 4 °C for 30 h. BPL was hydrolyzed by heating for 2h at 37 °C. The criteria and methods for quality control were defined using the engineering runs and the cGMP Lots passed all specifications. cGMP vaccine Lots displayed high potency, inducing between 80 and 90% survival in immunized mice when challenged with virulent pneumococci. Sera from mice immunized with the cGMP Lots recognized several pneumococcal proteins in the extract of encapsulated strains by Western blot. The cGMP whole cell antigen bulk and whole cell vaccine product lots were shown to be stable for up to 12 and 18 months, respectively, based upon survival assays following i.p. challenge. Our results show the consistency and stability of the cGMP whole cell pneumococcal vaccine lots and demonstrate the feasibility of production in a developing country setting.


Subject(s)
Bioreactors , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/biosynthesis , Propiolactone/pharmacology , Animals , Antibodies, Bacterial/blood , Female , Fermentation , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Microbial Viability , Pneumococcal Vaccines/immunology , Quality Control
2.
J Immune Based Ther Vaccines ; 9: 5, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21801401

ABSTRACT

BACKGROUND: Streptococcus pneumoniae causes widespread morbidity and mortality. Current vaccines contain free polysaccharides or protein-polysaccharide conjugates, and do not induce protection against serotypes that are not included in the vaccines. An affordable and broadly protective vaccine is very desirable. The goal of this study was to determine the optimal formulation of a killed whole cell pneumococcal vaccine with aluminum-containing adjuvants for intramuscular injection. METHODS: Four aluminium-containing adjuvants were prepared with different levels of surface phosphate groups resulting in different adsorptive capacities and affinities for the vaccine antigens. Mice were immunized three times and the antigen-specific antibody titers and IL-17 responses in blood were analyzed. RESULTS: Although all adjuvants induced significantly higher antibody titers than antigen without adjuvant, the vaccine containing aluminum phosphate adjuvant (AP) produced the highest antibody response when low doses of antigen were used. Aluminum hydroxide adjuvant (AH) induced an equal or better antibody response at high doses compared with AP. Vaccines formulated with AH, but not with AP, induced an IL-17 response. The vaccine formulated with AH was stable and retained full immunogenicity when stored at 4°C for 4 months. CONCLUSIONS: Antibodies are important for protection against systemic streptococcal disease and IL-17 is critical in the prevention of nasopharyngeal colonization by S. pneumoniae in the mouse model. The formulation of the whole killed bacterial cells with AH resulted in a stable vaccine that induced both antibodies and an IL-17 response. These experiments underscore the importance of formulation studies with aluminium containing adjuvants for the development of stable and effective vaccines.

3.
Vaccine ; 28(47): 7468-75, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-20858450

ABSTRACT

Mucosal immunization with a killed whole-cell pneumococcal vaccine, given with enterotoxin-related adjuvants, has been shown to confer multi-serotype protection against colonization of the nasopharynx and middle ear in mice. However, because novel mucosal immunization strategies may be difficult to implement, here we evaluated subcutaneous injection. Strain RM200 was engineered to be capsule-negative, autolysin-negative, and to express a non-toxic mutant pneumolysoid. Liter-scale and 60-l Good Manufacturing Practice (GMP) cultures were grown in bovine-free soy-based medium, killed with chloroform or beta-propiolactone, and injected into C57Bl/6 mice without or with aluminum adjuvant. The adjuvant Al(OH)(3) strongly increased responses, particularly if pre-treated with phosphate. Protection was found in several tested model infections: nasal colonization with a serotype 6B strain and fatal aspiration-sepsis with strains of serotype 3 and 5. Protection against colonization was mechanistically dependent on the presence of CD4+ T cells at the time of challenge; in contrast, in the type 3 aspiration-sepsis model, CD4+ T cells were not required for protection at the time of challenge, suggesting that antibody alone was sufficient to protect against death in this model. Rabbits receiving sequential intramuscular injections in a pilot toxicity study displayed local reactogenicity at injection sites but no clinical signs. The rabbit antiserum thus produced was active in an in vitro phagocytic killing assay and passively protected mice in the type 3 aspiration-sepsis model. Approval is being sought for human trials of this vaccine.


Subject(s)
Nasopharynx/microbiology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/administration & dosage , Sepsis/prevention & control , Adjuvants, Immunologic/administration & dosage , Alum Compounds/administration & dosage , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Vaccines/immunology , Propiolactone/administration & dosage , Rabbits , Sepsis/immunology , Sepsis/microbiology
4.
Clin Vaccine Immunol ; 17(6): 1005-12, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20427625

ABSTRACT

We previously reported that ethanol-killed cells of a noncapsulated strain of Streptococcus pneumoniae, given intranasally with cholera toxin as an adjuvant, protect rats against pneumonia and mice against colonization of the nasopharynx and middle ear by capsulated pneumococci of various serotypes. The acceleration of pneumococcal clearance from the nasopharynx in mice is CD4+ T cell-dependent and interleukin 17A (IL-17A) mediated and can be antibody independent. Here, anticipating human studies, we have demonstrated protection with a new vaccine strain expressing a nonhemolytic derivative of pneumolysin and grown in bovine-free culture medium. Killing the cells with chloroform, trichloroethylene, or beta-propiolactone--all used without postinactivation washing--produced more-potent immunogens than ethanol, and retention of soluble components released from the cells contributed to protection. Two sequential intranasal administrations of as little as 1 microg of protein (total of cellular and soluble combined) protected mice against nasopharyngeal challenge with pneumococci. Nontoxic single and double mutants of Escherichia coli heat-labile toxin were effective as mucosal adjuvants. Protection was induced by the sublingual and buccal routes, albeit requiring larger doses than when given intranasally. Protection was likewise induced transdermally with sonicates of the killed-cell preparation. Thus, this whole-cell antigen can be made and administered in a variety of ways to suit the manufacturer and the vaccination program and is potentially a solution to the need for a low-cost vaccine to reduce the burden of childhood pneumococcal disease in low-income countries.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines , Streptococcus pneumoniae/immunology , Streptolysins , Vaccines, Inactivated , Administration, Buccal , Administration, Intranasal , Administration, Sublingual , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Cholera Toxin/genetics , Cholera Toxin/immunology , Enterotoxins/genetics , Enterotoxins/immunology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Humans , Mice , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/administration & dosage , Pneumococcal Vaccines/genetics , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/cytology , Streptolysins/administration & dosage , Streptolysins/genetics , Streptolysins/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology
5.
Vaccine ; 28(47): 7468-7475, 2010.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1068348

ABSTRACT

Mucosal immunization with a killed whole-cell pneumococcal vaccine, given with enterotoxin-related adjuvants, has been shown to confer multi-serotype protection against colonization of the nasopharynx and middle ear in mice. However, because novel mucosal immunization strategies may be difficult to implement, here we evaluated subcutaneous injection. Strain RM200 was engineered to be capsule-negative, autolysin-negative, and to express a non-toxic mutant pneumolysoid. Liter-scale and 60-l Good Manufacturing Practice (GMP) cultures were grown in bovine-free soy-based medium, killed with chloroform or beta-propiolactone, and injected into C57Bl/6 mice without or with aluminum adjuvant. The adjuvant Al(OH)3 strongly increased responses, particularly if pre-treated with phosphate. Protection was found in several tested model infections: nasal colonization with a serotype 6B strain and fatal aspiration-sepsis with strains of serotype 3 and 5. Protection against colonization was mechanistically dependent on the presence of CD4+ T cells at the time of challenge; in contrast, in the type 3 aspiration-sepsis model, CD4+ T cells were not required for protection at the time of challenge, suggesting that antibody alone was sufficient to protect against death in this model. Rabbits receiving sequential intramuscular injections in a pilot toxicity study displayed local reactogenicity at injection sites but no clinical signs. The rabbit antiserum thus produced was active in an in vitro phagocytic killing assay and passively protected mice in the type 3 aspiration-sepsis model. Approval is being sought for human trials of this vaccine.


Subject(s)
Humans , Animals , Rats , Streptococcus pneumoniae/immunology , Vaccines
6.
FEMS Immunol Med Microbiol ; 55(2): 258-70, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19281568

ABSTRACT

Chlamydia trachomatis infection is the most common sexually transmitted bacterial infection worldwide, with over 91 million cases estimated annually. An effective subunit vaccine against Chlamydia may require a multivalent subunit cocktail of antigens in a single formulation for broad coverage of a heterogeneous major histocompatibility complex population. Herein, we describe the identification of novel C. trachomatis antigens by CD4+ and CD8+ T-cell expression cloning, serological expression cloning, and an in silico analysis of the C. trachomatis genome. These antigens elicited human CD4+ T-cell responses, and a subset proved to be immunogenic and protective when administered as immunoprophylactic vaccines against C. trachomatis challenge. Candidate vaccines consisting of the prioritized C. trachomatis antigens adjuvanted in a GlaxoSmithKline proprietary AS01B adjuvant were prioritized based on induction of solid protection against challenge in C57BL/6 and BALB/c mice with C. trachomatis. Some of the vaccines prevented bacterial shedding and colonization of the upper genital tract to varying degrees by mechanisms that may include CD4+ T cells.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Genital Diseases, Female/prevention & control , Recombinant Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Chlamydia Infections/immunology , Colony Count, Microbial , Female , Genital Diseases, Female/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccines, Synthetic/immunology
7.
Microbiology (Reading) ; 152(Pt 12): 3667-3681, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17159220

ABSTRACT

Despite accumulating data implicating Propionibacterium acnes in a variety of diseases, its precise role in infection remains to be determined. P. acnes antigen-specific CD4(+) T cells are present in early inflamed acne lesions and may be involved in the inflammatory response; however, little is known about the specific antigens involved. In this study, B cell and T cell antigens from P. acnes expression libraries were cloned and evaluated and the four predominant proteins identified were investigated. Two of these antigens share some homology with an M-like protein of Streptococcus equi and have dermatan-sulphate-binding activity (PA-25957 and 5541). The remaining two antigens, PA-21693 and 4687, are similar to the product of the Corynebacterium diphtheriae htaA gene from the hmu ABC transport locus, although only one of these (PA-21693) is encoded within an hmu-like operon and conserved amongst a range of clinical isolates. All four proteins contain an LPXTG motif, although only PA-21693 contains a characteristic sortase-sorting signal. Variation in the expression of PA-4687, 25957 and 5541 is evident amongst clinical isolates and is generated both by frameshifts associated with the putative signal peptide and by variable numbers of repeat regions toward the carboxy-terminus, potentially generating heterogeneity of molecular mass and antigenic variation. In addition, in the case of PA-25957, a frameshift in a C-rich region at the extreme carboxy-terminus eliminates the LPXTG motif in some isolates. For the dermatan-sulphate-binding PA-25957, IgG1 antibody in serum from acne-positive donors was shown to be specific for the amino-terminal region of the protein, which also contains a CD4(+) T cell epitope. In contrast, serum from acne-negative donors shows an IgG2 and IgG3 antibody subclass response to the carboxy-terminal region. These data have implications for the potential role of P. acnes in inflammatory acne and other diseases.


Subject(s)
Acne Vulgaris/immunology , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Propionibacterium acnes/immunology , Propionibacterium acnes/metabolism , Amino Acid Sequence , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Carrier Proteins/genetics , Cloning, Molecular , Conserved Sequence , Corynebacterium diphtheriae/genetics , Dermatan Sulfate/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Frameshift Mutation , Gene Expression , Genetic Variation , Humans , Immunoblotting , Immunoglobulin G/immunology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Propionibacterium acnes/genetics , Protein Binding , Protein Sorting Signals/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sequence Homology, Amino Acid , Streptococcus equi/genetics
8.
Microbiology (Reading) ; 151(Pt 5): 1369-1379, 2005 May.
Article in English | MEDLINE | ID: mdl-15870447

ABSTRACT

Analysis of the draft genome sequence of the opportunistic pathogen Propionibacterium acnes type strain NCTC 737 (=ATCC 6919) revealed five genes with sequence identity to the co-haemolytic Christie-Atkins-Munch-Peterson (CAMP) factor of Streptococcus agalactiae. The predicted molecular masses for the expressed proteins ranged from 28 to 30 kDa. The genes were present in each of the three recently identified recA-based phylogenetic groupings of P. acnes (IA, IB and II), as assessed by PCR amplification. Conserved differences in CAMP factor gene sequences between these three groups were also consistent with their previous phylogenetic designations. All type IA, IB and II isolates were positive for the co-haemolytic reaction on sheep blood agar. Immunoblotting and silver staining of SDS-PAGE gels, however, revealed differential protein expression of CAMP factors amongst the different groups. Type IB and II isolates produced an abundance of CAMP factor 1, detectable by specific antibody labelling and silver staining of SDS-PAGE gels. In contrast, abundant CAMP factor production was lacking in type IA isolates, although larger amounts of CAMP factor 2 were detectable by immunoblotting compared with type II isolates. While the potential role of the abundant CAMP factor 1 in host colonization or virulence remains to be determined, it should be noted that the type strain of P. acnes used in much of the published literature is a type IA isolate and is, therefore, lacking in this attribute.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Propionibacterium acnes/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Base Sequence , DNA, Bacterial/analysis , Hemolysin Proteins , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction/methods , Propionibacterium acnes/classification , Propionibacterium acnes/genetics , Propionibacterium acnes/growth & development , Sequence Analysis, DNA , Streptococcus agalactiae/genetics , Streptococcus agalactiae/metabolism
9.
J Clin Microbiol ; 43(1): 326-34, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15634990

ABSTRACT

Although two phenotypes of the opportunistic pathogen Propionibacterium acnes (types I and II) have been described, epidemiological investigations of their roles in different infections have not been widely reported. Using immunofluorescence microscopy with monoclonal antibodies (MAbs) QUBPa1 and QUBPa2, specific for types I and II, respectively, we investigated the prevalences of the two types among 132 P. acnes isolates. Analysis of isolates from failed prosthetic hip implants (n = 40) revealed approximately equal numbers of type I and II organisms. Isolates from failed prosthetic hip-associated bone (n = 6) and tissue (n = 38) samples, as well as isolates from acne (n = 22), dental infections (n = 8), and skin removed during surgical incision (n = 18) were predominately of type I. A total of 11 (8%) isolates showed atypical MAb labeling and could not be conclusively identified. Phylogenetic analysis of P. acnes by nucleotide sequencing revealed the 16S rRNA gene to be highly conserved between types I and II. In contrast, sequence analysis of recA and a putative hemolysin gene (tly) revealed significantly greater type-specific polymorphisms that corresponded to phylogenetically distinct cluster groups. All 11 isolates with atypical MAb labeling were identified as type I by sequencing. Within the recA and tly phylogenetic trees, nine of these isolates formed a cluster distinct from other type I organisms, suggesting a further phylogenetic subdivision within type I. Our study therefore demonstrates that the phenotypic differences between P. acnes types I and II reflect deeper differences in their phylogeny. Furthermore, nucleotide sequencing provides an accurate method for identifying the type status of P. acnes isolates.


Subject(s)
Gram-Positive Bacterial Infections/microbiology , Hemolysin Proteins/genetics , Phylogeny , Propionibacterium acnes/classification , RNA, Ribosomal, 16S/genetics , Rec A Recombinases/genetics , Animals , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Bacterial Typing Techniques , DNA, Ribosomal/analysis , Genes, rRNA , Gram-Positive Bacterial Infections/epidemiology , Humans , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Molecular Sequence Data , Propionibacterium acnes/genetics , Propionibacterium acnes/immunology , Sequence Analysis, DNA
10.
Infect Immun ; 72(12): 7231-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557648

ABSTRACT

Chlamydia trachomatis is an obligate intracellular gram-negative bacterium responsible for a wide spectrum of diseases in humans. Both genital and ocular C. trachomatis infections are associated with tissue inflammation and pathology. Dendritic cells (DC) play an important role in both innate and adaptive immune responses to microbial pathogens and are a source of inflammatory cytokines. To determine the potential contribution of DC to the inflammatory process, human DC were infected with C. trachomatis serovar E or L2. Both C. trachomatis serovars were found to infect and replicate in DC. Upon infection, DC up-regulated the expression of costimulatory (B7-1) and cell adhesion (ICAM-1) molecules. Furthermore, chlamydial infection induced the secretion of interleukin-1beta (IL-1beta), IL-6, IL-8, IL-12p70, IL-18, and tumor necrosis factor alpha (TNF-alpha). The mechanisms involved in Chlamydia-induced IL-1beta and IL-18 secretion differed from those of the other cytokines. Chlamydia-induced IL-1beta and IL-18 secretion required infection with viable bacteria and was associated with the Chlamydia-induced activation of caspase-1 in infected host cells. In contrast, TNF-alpha and IL-6 secretion did not require that the Chlamydia be viable, suggesting that there are at least two mechanisms involved in the Chlamydia-induced cytokine secretion in DC. Interestingly, an antibody to Toll-like receptor 4 inhibited Chlamydia-induced IL-1beta, IL-6, and TNF-alpha secretion. The data herein demonstrate that DC can be infected by human C. trachomatis serovars and that chlamydial components regulate the secretion of various cytokines in DC. Collectively, these data suggest that DC play a role in the inflammatory processes caused by chlamydial infections.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , Cytokines/biosynthesis , Dendritic Cells/physiology , Caspase 1/metabolism , Cell Adhesion Molecules/biosynthesis , Dendritic Cells/microbiology , Enzyme Activation , Humans , Interleukin-1/biosynthesis , Interleukin-18/biosynthesis , Membrane Glycoproteins , Receptors, Cell Surface , Toll-Like Receptor 4 , Toll-Like Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...