Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 34(9): 1653-1666, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37682243

ABSTRACT

Coxiella burnetii is the causative agent of Q fever, for which there is yet to be an FDA-approved vaccine. This bacterial pathogen has both extra- and intracellular stages in its life cycle, and therefore both a cell-mediated (i.e., T lymphocyte) and humoral (i.e., antibody) immune response are necessary for effective eradication of this pathogen. However, most proposed vaccines elicit strong responses to only one mechanism of adaptive immunity, and some can either cause reactogenicity or lack sufficient immunogenicity. In this work, we aim to apply a nanoparticle-based platform toward producing both antibody and T cell immune responses against C. burnetii. We investigated three approaches for conjugation of the immunodominant outer membrane protein antigen (CBU1910) to the E2 nanoparticle to obtain a consistent antigen orientation: direct genetic fusion, high affinity tris-NTA-Ni conjugation to polyhistidine-tagged CBU1910, and the SpyTag/SpyCatcher (ST/SC) system. Overall, we found that the ST/SC approach yielded nanoparticles loaded with the highest number of antigens while maintaining stability, enabling formulations that could simultaneously co-deliver the protein antigen (CBU1910) and adjuvant (CpG1826) on one nanoparticle (CBU1910-CpG-E2). Using protein microarray analyses, we found that after immunization, antigen-bound nanoparticle formulations elicited significantly higher antigen-specific IgG responses than soluble CBU1910 alone and produced more balanced IgG1/IgG2c ratios. Although T cell recall assays from these protein antigen formulations did not show significant increases in antigen-specific IFN-γ production compared to soluble CBU1910 alone, nanoparticles conjugated with a CD4 peptide epitope from CBU1910 generated elevated T cell responses in mice to both the CBU1910 peptide epitope and whole CBU1910 protein. These investigations highlight the feasibility of conjugating antigens to nanoparticles for tuning and improving both humoral- and cell-mediated adaptive immunity against C. burnetii.


Subject(s)
Coxiella burnetii , Q Fever , Vaccines , Animals , Mice , Q Fever/prevention & control , Antigens, Bacterial , Antibodies , Epitopes
2.
Front Immunol ; 14: 1192821, 2023.
Article in English | MEDLINE | ID: mdl-37533862

ABSTRACT

Vaccines are among the most cost-effective public health measures for controlling infectious diseases. Coxiella burnetii is the etiological agent of Q fever, a disease with a wide clinical spectrum that ranges from mild symptoms, such as fever and fatigue, to more severe disease, such as pneumonia and endocarditis. The formalin-inactivated whole-cell vaccine Q-VAX® contains hundreds of antigens and confers lifelong protection in humans, but prior sensitization from infection or vaccination can result in deleterious reactogenic responses to vaccination. Consequently, there is great interest in developing non-reactogenic alternatives based on adjuvanted recombinant proteins. In this study, we aimed to develop a multivalent vaccine that conferred protection with reduced reactogenicity. We hypothesized that a multivalent vaccine consisting of multiple antigens would be more immunogenic and protective than a monovalent vaccine owing to the large number of potential protective antigens in the C. burnetii proteome. To address this, we identified immunogenic T and B cell antigens, and selected proteins were purified to evaluate with a combination adjuvant (IVAX-1), with or without C. burnetii lipopolysaccharide (LPS) in immunogenicity studies in vivo in mice and in a Hartley guinea pig intratracheal aerosol challenge model using C. burnetii strain NMI RSA 493. The data showed that multivalent vaccines are more immunogenic than monovalent vaccines and more closely emulate the protection achieved by Q-VAX. Although six antigens were the most immunogenic, we also discovered that multiplexing beyond four antigens introduces detectable reactogenicity, indicating that there is an upper limit to the number of antigens that can be safely included in a multivalent Q-fever vaccine. C. burnetii LPS also demonstrates efficacy as a vaccine antigen in conferring protection in an otherwise monovalent vaccine formulation, suggesting that its addition in multivalent vaccines, as demonstrated by a quadrivalent formulation, would improve protective responses.


Subject(s)
Coxiella burnetii , Humans , Guinea Pigs , Animals , Mice , Vaccines, Combined , Lipopolysaccharides , Bacterial Vaccines , Antigens , Adjuvants, Immunologic , Aerosols
3.
Front Immunol ; 14: 1196522, 2023.
Article in English | MEDLINE | ID: mdl-37215096

Subject(s)
Q Fever , Vaccines , Humans
4.
Front Immunol ; 12: 754712, 2021.
Article in English | MEDLINE | ID: mdl-34616410

ABSTRACT

Q-VAX®, a whole cell, formalin-inactivated vaccine, is the only vaccine licensed for human use to protect against Coxiella burnetii, the cause of Q fever. Although this vaccine provides long-term protection, local and systemic reactogenic responses are common in previously sensitized individuals which prevents its use outside of Australia. Despite the importance of preventing these adverse reactions to develop widely accepted, novel vaccines against C. burnetii, little is understood about the underlying cellular mechanisms. This is mostly attributed to the use of a guinea pig reactogenicity model where complex cellular analysis is limited. To address this, we compared three different mouse strains develop a model of C. burnetii whole cell vaccine reactogenic responses. SKH1 and C57Bl/6, but not BALBc mice, develop local granulomatous reactions after either infection- or vaccine-induced sensitization. We evaluated local and systemic responses by measuring T cell populations from the vaccination site and spleen during elicitation using flow cytometry. Local reaction sites showed influx of IFNγ+ and IL17a+ CD4 T cells in sensitized mice compared with controls and a reduction in IL4+ CD4 T cells. Additionally, sensitized mice showed a systemic response to elicitation by an increase in IFNγ+ and IL17a+ CD4 T cells in the spleen. These results indicate that local and systemic C. burnetii reactogenic responses are consistent with a Th1 delayed-type hypersensitivity. Our experiments provide insights into the pathophysiology of C. burnetii whole cell vaccine reactogenicity and demonstrate that C57Bl/6 and SKH1 mice can provide a valuable model for evaluating the reactogenicity of novel C. burnetii vaccine candidates.


Subject(s)
Bacterial Vaccines/adverse effects , Disease Models, Animal , Hypersensitivity, Delayed/immunology , Q Fever , Th1 Cells/immunology , Animals , Coxiella burnetii , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Q Fever/prevention & control , Vaccines, Inactivated/adverse effects
5.
Front Immunol ; 12: 653092, 2021.
Article in English | MEDLINE | ID: mdl-33815413

ABSTRACT

Q fever is caused by the obligate intracellular bacterium, Coxiella burnetii, a designated potential agent of bioterrorism because of its route of transmission, resistance to disinfectants, and low infectious dose. The only vaccine licensed for human use is Q-VAX® (Seqirus, licensed in Australia), a formalin-inactivated whole-cell vaccine, which produces severe local and systemic reactogenic responses in previously sensitized individuals. Accordingly, the U.S. Food and Drug Administration and other regulatory bodies around the world, have been reluctant to approve Q-VAX for widespread use. To obviate these adverse reactions, we prepared recombinant protein subunit vaccine candidates containing purified CBU1910, CBU0307, CBU0545, CBU0612, CBU0891, and CBU1398 proteins and TLR triagonist adjuvants. TLR triagonist adjuvants combine different TLR agonists to enhance immune responses to vaccine antigens. We tested both the protective efficacy and reactogenicity of our vaccine candidates in Hartley guinea pigs using intratracheal infection with live C. burnetii. While all of our candidates showed varying degrees of protection during challenge, local reactogenic responses were significantly reduced for one of our vaccine candidates when compared with a formalin-inactivated whole-cell vaccine. Our findings show that subunit vaccines combined with novel TLR triagonist adjuvants can generate protective immunity to C. burnetii infection while reducing reactogenic responses.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/pharmacology , Coxiella burnetii/immunology , Q Fever/prevention & control , Toll-Like Receptors/antagonists & inhibitors , Adjuvants, Immunologic/therapeutic use , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/pharmacology , Antigens, Bacterial/therapeutic use , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/therapeutic use , Disease Models, Animal , Guinea Pigs , Humans , Immunogenicity, Vaccine , Q Fever/immunology , Q Fever/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Vaccines, Subunit/genetics , Vaccines, Subunit/pharmacology , Vaccines, Subunit/therapeutic use , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology , Vaccines, Synthetic/therapeutic use
6.
Cell Rep Med ; 2(12): 100461, 2021 12 21.
Article in English | MEDLINE | ID: mdl-35028605

ABSTRACT

Q fever is caused by the intracellular bacterium Coxiella burnetii, for which there is no approved vaccine in the United States. A formalin-inactivated whole-cell vaccine (WCV) from virulent C. burnetii NMI provides single-dose long-lived protection, but concerns remain over vaccine reactogenicity. We therefore sought an alternate approach by purifying native C. burnetii antigens from the clonally derived avirulent NMII strain. A soluble bacterial extract, termed Sol II, elicits high-titer, high-avidity antibodies and induces a CD4 T cell response that confers protection in naive mice. In addition, Sol II protects against pulmonary C. burnetii challenge in three animal models without inducing hypersensitivity. An NMI-derived extract, Sol I, enhances protection further and outperforms the WCV gold standard. Collectively, these data represent a promising approach to design highly effective, non-reactogenic Q fever vaccines.


Subject(s)
Antigens, Bacterial/immunology , Coxiella burnetii/immunology , Hypersensitivity/immunology , Immunity , Q Fever/immunology , Q Fever/prevention & control , Aerosols , Animals , Antibody Affinity , Antigenic Variation , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Guinea Pigs , Immunization , Lipopolysaccharides , Lung/microbiology , Lung/pathology , Macaca mulatta , Male , Mice, Inbred C57BL , Q Fever/microbiology , Solubility
7.
Infect Immun ; 88(10)2020 09 18.
Article in English | MEDLINE | ID: mdl-32690632

ABSTRACT

Reproductive failure is the hallmark of brucellosis in animals. An uncommon but important complication in pregnant women who become acutely infected with Brucella melitensis is spontaneous pregnancy loss or vertical transmission to the fetus. Unfortunately, the mechanism behind reproductive failure is still obscure, partially due to the lack of a proper study model. Recently, it was demonstrated that intratracheal (IT) inoculation of nonpregnant guinea pigs would replicate features of clinical disease in humans. To determine if IT inoculation would induce reproductive disease, guinea pigs were infected at mid-gestation and monitored daily for fever and abortions. Fever developed between day 14 to 18 postinoculation, and by 3 weeks postinoculation, 75% of pregnant guinea pigs experienced stillbirths or spontaneous abortions mimicking natural disease. Next, to investigate the guinea pig as a model for evaluating vaccine efficacy during pregnancy, nonpregnant guinea pigs were vaccinated with S19, 16MΔvjbR + Quil-A, or 100 µl PBS + Quil-A (as control). Guinea pigs were bred and vaccinated guinea pigs were challenged at mid-gestation with B. melitensis IT inoculation and monitored for fever and abortions. Vaccination with both vaccines prevented fever and protected against abortion. Together, this study indicates that pregnant guinea pigs are an appropriate animal model to study reproductive disease and offer an improved model to evaluate the ability of vaccine candidates to protect against a serious manifestation of disease.


Subject(s)
Brucella Vaccine/administration & dosage , Brucella melitensis/immunology , Brucellosis/prevention & control , Disease Models, Animal , Pregnancy Complications, Infectious/prevention & control , Animals , Antibodies, Bacterial/blood , Brucella melitensis/pathogenicity , Brucellosis/microbiology , Brucellosis/pathology , Female , Guinea Pigs , Humans , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/pathology , Placenta/microbiology , Placenta/pathology , Pregnancy , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/pathology , Vaccination
8.
J Infect Dis ; 220(4): 624-634, 2019 07 19.
Article in English | MEDLINE | ID: mdl-30938819

ABSTRACT

BACKGROUND: Protective immunity against Coxiella burnetii infection is conferred by vaccination with virulent (PI-WCV), but not avirulent (PII-WCV) whole-cell inactivated bacterium. The only well-characterized antigenic difference between virulent and avirulent C. burnetii is they have smooth and rough lipopolysaccharide (LPS), respectively. METHODS: Mice were vaccinated with PI-WCV and PII-WCV. Humoral and cellular responses were evaluated using protein chip microarrays and ELISpots, respectively. Dendritic cell (DC) maturation after stimulation with PI-WVC and PII-WVC was evaluated using flow cytometry. Vaccine-challenge studies were performed to validate the importance of the receptor CCR7. RESULTS: Other than specific antibody response to PI-LPS, similar antibody profiles were observed but IgG titers were significantly higher after vaccination with PI-WCV. Furthermore, higher frequency of antigen-specific CD4+ T cells was detected in mice immunized with PI-WCV. PI-WCV-stimulated DCs displayed significantly higher levels of CCR7 and migratory ability to secondary lymphoid organs. Challenge-protection studies in wild-type and CCR7-deficient mice confirmed that CCR7 is critical for PI-WCV-induced cellular immunity. CONCLUSIONS: PI-WVC stimulates protective immunity to C. burnetii in mice through stimulation of migratory behavior in DCs for protective cellular immunity. Additionally, the humoral immune response to LPS is an important component of protective immunity.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Coxiella burnetii/immunology , Immunity, Cellular , Q Fever/immunology , Receptors, Chemokine/immunology , Animals , Antibody Formation , Dendritic Cells/immunology , Female , Humans , Lipopolysaccharides/immunology , Mice , Q Fever/microbiology , Q Fever/prevention & control , Vaccination
9.
Clin Vaccine Immunol ; 24(11)2017 11.
Article in English | MEDLINE | ID: mdl-28903988

ABSTRACT

Burkholderia pseudomallei is a Gram-negative, facultative intracellular pathogen that causes the disease melioidosis in humans and other mammals. Respiratory infection with B. pseudomallei leads to a fulminant and often fatal disease. It has previously been shown that glycoconjugate vaccines can provide significant protection against lethal challenge; however, the limited number of known Burkholderia antigens has slowed progress toward vaccine development. The objective of this study was to identify novel antigens and evaluate their protective capacity when incorporated into a nanoglycoconjugate vaccine platform. First, an in silico approach to identify antigens with strong predicted immunogenicity was developed. Protein candidates were screened and ranked according to predicted subcellular localization, transmembrane domains, adhesive properties, and ability to interact with major histocompatibility complex (MHC) class I and class II. From these in silico predictions, we identified seven "high priority" proteins that demonstrated seroreactivity with anti-B. pseudomallei murine sera and convalescent human melioidosis sera, providing validation of our methods. Two novel proteins, together with Hcp1, were linked to lipopolysaccharide (LPS) and incorporated with the surface of a gold nanoparticle (AuNP). Animals receiving AuNP glycoconjugate vaccines generated high protein- and polysaccharide-specific antibody titers. Importantly, immunized animals receiving the AuNP-FlgL-LPS alone or as a combination demonstrated up to 100% survival and reduced lung colonization following a lethal challenge with B. pseudomallei Together, this study provides a rational approach to vaccine design that can be adapted for other complex pathogens and provides a rationale for further preclinical testing of AuNP glycoconjugate in animal models of infection.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia pseudomallei/immunology , Glycoconjugates/immunology , Metal Nanoparticles/administration & dosage , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Female , Gold/immunology , Humans , Lipopolysaccharides/immunology , Melioidosis/immunology , Melioidosis/prevention & control , Mice , Mice, Inbred C57BL , Models, Animal , Vaccinology/methods
10.
J Infect Dis ; 215(10): 1580-1589, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27703037

ABSTRACT

Coxiella burnetii is a gram-negative bacterium that causes acute and chronic Q fever. Because of the severe adverse effect of whole-cell vaccination, identification of immunodominant antigens of C. burnetii has become a major focus of Q fever vaccine development. We hypothesized that secreted C. burnetii type IV secretion system (T4SS) effectors may represent a major class of CD8+ T-cell antigens, owing to their cytosolic localization. Twenty-nine peptides were identified that elicited robust CD8+ T-cell interferon γ (IFN-γ) recall responses from mice infected with C. burnetii. Interestingly, 22 of 29 epitopes were derived from 17 T4SS-related proteins, none of which were identified as immunodominant antigens by using previous antibody-guided approaches. These epitopes were expressed in an attenuated Listeria monocytogenes vaccine strain. Immunization with recombinant L. monocytogenes vaccines induced a robust CD8+ T-cell response and conferred measurable protection against C. burnetii infection in mice. These data suggested that T4SS effectors represent an important class of C. burnetii antigens that can induce CD8+ T-cell responses. We also showed that attenuated L. monocytogenes vaccine vectors are an efficient antigen-delivery platform that can be used to induce robust protective CD8+ T-cell immune responses against C. burnetii infection.


Subject(s)
Antigen Presentation/immunology , Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Coxiella burnetii/immunology , Epitopes, T-Lymphocyte/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/chemistry , Coxiella burnetii/chemistry , Epitopes, T-Lymphocyte/chemistry , Female , Interferon-gamma Release Tests , Listeria monocytogenes/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/immunology , Q Fever/immunology , Q Fever/microbiology , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/immunology , Vaccines, Attenuated/chemistry
11.
Nanomedicine ; 11(2): 447-56, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25194998

ABSTRACT

Burkholderia mallei are Gram-negative bacteria, responsible for the disease glanders. B. mallei has recently been classified as a Tier 1 agent owing to the fact that this bacterial species can be weaponised for aerosol release, has a high mortality rate and demonstrates multi-drug resistance. Furthermore, there is no licensed vaccine available against this pathogen. Lipopolysaccharide (LPS) has previously been identified as playing an important role in generating host protection against Burkholderia infection. In this study, we present gold nanoparticles (AuNPs) functionalised with a glycoconjugate vaccine against glanders. AuNPs were covalently coupled with one of three different protein carriers (TetHc, Hcp1 and FliC) followed by conjugation to LPS purified from a non-virulent clonal relative, B. thailandensis. Glycoconjugated LPS generated significantly higher antibody titres compared with LPS alone. Further, they improved protection against a lethal inhalation challenge of B. mallei in the murine model of infection. FROM THE CLINICAL EDITOR: Burkholderia mallei is associated with multi-drug resistance, high mortality and potentials for weaponization through aerosol inhalation. The authors of this study present gold nanoparticles (AuNPs) functionalized with a glycoconjugate vaccine against this Gram negative bacterium demonstrating promising results in a murine model even with the aerosolized form of B. Mallei.


Subject(s)
Bacterial Vaccines/administration & dosage , Burkholderia mallei/drug effects , Glanders/drug therapy , Metal Nanoparticles/administration & dosage , Administration, Inhalation , Animals , Bacterial Vaccines/chemistry , Burkholderia mallei/pathogenicity , Disease Models, Animal , Glanders/immunology , Glanders/microbiology , Glycoconjugates/administration & dosage , Glycoconjugates/chemistry , Gold/chemistry , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Metal Nanoparticles/chemistry , Mice
12.
Vaccine ; 33(5): 686-92, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25533326

ABSTRACT

The Gram-negative Burkholderia mallei is a zoonotic pathogen and the causative agent of glanders disease. Because the bacteria maintain the potential to be used as a biothreat agent, vaccine strategies are required for human glanders prophylaxis. A rhesus macaque (Macaca mulatta) model of pneumonic (inhalational) glanders was established and the protective properties of a nanoparticle glycoconjugate vaccine composed of Burkholderia thailandensis LPS conjugated to FliC was evaluated. An aerosol challenge dose of ∼1×10(4) CFU B. mallei produced mortality in 50% of naïve animals (n=2/4), 2-3 days post-exposure. Although survival benefit was not observed by vaccination with a glycoconjugate glanders vaccine (p=0.42), serum LPS-specific IgG titers were significantly higher on day 80 in 3 vaccinated animals who survived compared with 3 vaccinated animals who died. Furthermore, B. mallei was isolated from multiple organs of both non-vaccinated survivors, but not from any organs of 3 vaccinated survivors at 30 days post-challenge. Taken together, this is the first time a candidate vaccine has been evaluated in a non-human primate aerosol model of glanders and represents the initial step for consideration in pre-clinical studies.


Subject(s)
Bacterial Vaccines/immunology , Burkholderia mallei/immunology , Glanders/prevention & control , Gold/administration & dosage , Nanoparticles/administration & dosage , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Disease Models, Animal , Glycoconjugates/administration & dosage , Glycoconjugates/immunology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Macaca mulatta , Survival Analysis , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology
13.
Article in English | MEDLINE | ID: mdl-23532930

ABSTRACT

Vaccination has had a major impact on the control of infectious diseases. However, there are still many infectious diseases for which the development of an effective vaccine has been elusive. In many cases the failure to devise vaccines is a consequence of the inability of vaccine candidates to evoke appropriate immune responses. This is especially true where cellular immunity is required for protective immunity and this problem is compounded by the move toward devising sub-unit vaccines. Over the past decade nanoscale size (<1000 nm) materials such as virus-like particles, liposomes, ISCOMs, polymeric, and non-degradable nanospheres have received attention as potential delivery vehicles for vaccine antigens which can both stabilize vaccine antigens and act as adjuvants. Importantly, some of these nanoparticles (NPs) are able to enter antigen-presenting cells by different pathways, thereby modulating the immune response to the antigen. This may be critical for the induction of protective Th1-type immune responses to intracellular pathogens. Their properties also make them suitable for the delivery of antigens at mucosal surfaces and for intradermal administration. In this review we compare the utilities of different NP systems for the delivery of sub-unit vaccines and evaluate the potential of these delivery systems for the development of new vaccines against a range of pathogens.


Subject(s)
Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Vaccines, Subunit/administration & dosage , Antigen-Presenting Cells/immunology , Drug Carriers/pharmacokinetics , Immunity, Cellular , Th1 Cells/immunology , Vaccines, Subunit/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...