Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Sci Rep ; 12(1): 1325, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35079054

ABSTRACT

Pseudomonas aeruginosa as a common pathogen causing urinary tract infections (UTIs) has been resistant to different antibiotics and developing an effective vaccine can be an alternative strategy. In the present study, the immunogenicity and protection efficacy of formulations composed of a hybrid protein composed of P. aeruginosa V-antigen (PcrV) and exoenzyme S (ExoS) with alum and MPL were evaluated. The hybrid protein could increase the specific systemic and mucosal immune responses, as well as cellular responses as compared with control groups. Combining of alum or MPL adjuvant with the hybrid protein significantly improved the levels of IgG1, serum IgA, mucosal IgG, and IL-17 as compared to the ExoS.PcrV alone. After bladder challenge with a P. aeruginosa strain, the bacterial loads of bladder and kidneys were significantly decreased in mice received ExoS.PcrV admixed with alum and ExoS.PcrV admixed with MPL than controls. The present study indicated that immunization of mice with a hybrid protein composed of ExoS and PcrV could induce multifactorial immune responses and opsonize the bacteria and decrease the viable bacterial cells. Because P. aeruginosa have caused therapeutic challenges worldwide, our study proposed ExoS.PcrV + alum and ExoS.PcrV + MPL as promising candidates for the prevention of infections caused by P. aeruginosa.


Subject(s)
ADP Ribose Transferases/immunology , Adjuvants, Immunologic/pharmacology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections , Pseudomonas aeruginosa/drug effects , Animals , Mice , Mice, Inbred BALB C , Pseudomonas Infections/immunology , Pseudomonas Infections/prevention & control
2.
Front Immunol ; 12: 684823, 2021.
Article in English | MEDLINE | ID: mdl-34122448

ABSTRACT

HI, a fusion protein that consists of the alpha-toxin (Hla) and the N2 domain of iron surface determinant B (IsdB), is one of the antigens in the previously reported S. aureus vaccine rFSAV and has already entered phase II clinical trials. Previous studies revealed that HI is highly immunogenic in both mice and healthy volunteers, and the humoral immune response plays key roles in HI-mediated protection. In this study, we further investigated the protective efficacy of immunization with HI plus four different adjuvants in a mouse bacteremia model. Results showed that HI-mediated protection was altered in response to different adjuvants. Using antisera from immunized mice, we identified seven B-cell immunodominant epitopes on Hla and IsdB, including 6 novel epitopes (Hla1-18, Hla84-101, Hla186-203, IsdB342-359, IsdB366-383, and IsdB384-401). The immunodominance of B-cell epitopes, total IgG titers and the levels of IFN-γ and IL-17A from mice immunized with HI plus different adjuvants were different from each other, which may explain the difference in protective immunity observed in each immunized group. Thus, our results indicate that adjuvants largely affected the immunodominance of epitopes and the protective efficacy of HI, which may guide further adjuvant screening for vaccine development and optimization.


Subject(s)
Bacteremia/immunology , Bacterial Toxins/immunology , Cation Transport Proteins/immunology , Epitopes, B-Lymphocyte/immunology , Hemolysin Proteins/immunology , Immunodominant Epitopes/immunology , Staphylococcal Infections/prevention & control , Animals , Bacteremia/prevention & control , Disease Models, Animal , Female , Immunization, Passive , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Staphylococcal Infections/immunology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/immunology
3.
Curr Pharm Biotechnol ; 21(10): 973-979, 2020.
Article in English | MEDLINE | ID: mdl-32101119

ABSTRACT

BACKGROUND: Heat-Labile enterotoxin B subunit (LTB) produced by Escherichia coli, a non-toxic protein subunit with potential biological properties, is a powerful mucosal and parenteral adjuvant which can induce a strong immune response against co-administered antigens. OBJECTIVE: In the present study, LTB protein, encoded by the optimized ltb (also known synthetic ltb, s-ltb) gene in centella plant (Centella asiatica) for use as an antigen, has been discussed. METHODS: The s-ltb gene was cloned into a plant expression vector, pMYO51, adjacent to the CaMV 35S promoter and was then introduced into centella plant by biolistic transformation. PCR amplification was conducted to determine the presence of s-ltb gene in the transgenic centella plant. The expression of s-ltb gene was analyzed by immunoblotting and quantified by ELISA. In vitro activity of LTB protein was determined by GM1-ELISA. RESULTS: PCR amplification has found seven transgenic centella individuals. However, only five of them produced LTB protein. ELISA analysis showed that the highest amount of LTB protein detected in transgenic centella leaves was about 0.8% of the total soluble protein. GM1-ELISA assay indicated that plant LTB protein bound specifically to GM1-ganglioside, suggesting that the LTB subunits formed active pentamers. CONCLUSION: The s-ltb gene that was successfully transformed into centella plants by the biolistic method has produced a relatively high amount of plant LTB protein in the pentameric quaternary structure that has GM1-ganglioside binding affinity, a receptor on the intestinal epithelial membrane.


Subject(s)
Bacterial Toxins/genetics , Biolistics/methods , Centella/genetics , Enterotoxins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/genetics , Plants, Genetically Modified/genetics , Animals , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Centella/metabolism , Enterotoxins/chemistry , Enterotoxins/immunology , Enterotoxins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Gene Expression , Hot Temperature , Plant Extracts , Plants, Genetically Modified/metabolism , Triterpenes
4.
Lett Appl Microbiol ; 69(5): 385-390, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31529707

ABSTRACT

Clostridium perfringens is the main cause of sudden death in dogs and currently there is no vaccine to prevent it. In this study, a canine C. perfringens type A strain was used to prepare a vaccine. C. perfringens was inactivated by formaldehyde and adjuvants were added. The safety and immunological characteristics of the inactivated C. perfringens vaccine were evaluated in mice and dogs. The results showed that the C. perfringens vaccine was safe and had immunoprotective activity. The serum antibody titre of immunized mice reached up to 6·25 × 104 . Both single immunization of 4 ml and dual immunizations of 2 ml each provided good immune protection, with five of five immunized dogs surviving. This study also studied a detoxified crude α-toxin extract vaccine. The results showed that a single immunization with 0·5 ml of the detoxified crude α-toxin extract vaccine provided immune protection, with five of five immunized dogs surviving. The inactivated C. perfringens type A vaccine can be used to prevent canine C. perfringens infections. SIGNIFICANCE AND IMPACT OF THE STUDY: Clostridium perfringens is the main cause of sudden death in dogs and currently there is no vaccine to prevent it. In this study, an inactivated canine C. perfringens vaccine and a detoxified crude α-toxin vaccine were prepared. The safety and protective effects of these vaccines were evaluated using mouse and dog models. The vaccines were shown to be safe and to provide immune protection effects that can be used to prevent canine C. perfringens infection.


Subject(s)
Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Clostridium Infections/prevention & control , Clostridium perfringens/immunology , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Clostridium Infections/microbiology , Clostridium perfringens/genetics , Dogs , Drug Evaluation, Preclinical , Humans , Immunization , Mice , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology
5.
Int J Biol Macromol ; 137: 126-131, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31238071

ABSTRACT

Vibrio parahaemolyticus is the main etiological agent of human gastroenteritis by seafood consumption and some strains from this species causing the Acute Hepatopancreatic Necrosis Disease in shrimp have been recently reported. The PirA-like toxin from V. parahaemolyticus (ToxA) has been recently reported as an attractive antigen implicated in subunit vaccine development. Since plants are attractive hosts for the production and delivery of vaccines in the present study plants expressing ToxA were developed to account with a low cost platform for the production and oral delivery of ToxA. Tobacco plants were genetically engineered by Agrobacterium-mediated transformation to stably integrate the ToxA-coding gene into the nuclear genome. Transgenic lines were rescued in kanamycin-containing medium and analyzed by ELISA to determine ToxA yields observing levels up to 9 µg g-1 FW leaf tissues. Western blot analysis confirmed the presence of the ToxA protein in plant extracts. Immunogenicity assessment of the plant-made ToxA was performed in mice, comprising a 4-dose oral immunization scheme; revealing the induction of anti-ToxA humoral responses (IgG in serum and IgA in feces). This study opens the path for the development of low cost plant-based vaccines against Vibrio parahaemolyticus.


Subject(s)
Bacterial Toxins/biosynthesis , Bacterial Toxins/immunology , Nicotiana/genetics , Nicotiana/metabolism , Vibrio parahaemolyticus/genetics , Administration, Oral , Amino Acid Sequence , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Biotechnology , Immunity, Humoral/immunology , Mice , Plant Leaves/metabolism , Plants, Genetically Modified
6.
mSphere ; 4(3)2019 06 19.
Article in English | MEDLINE | ID: mdl-31217301

ABSTRACT

Inhalational anthrax caused by Bacillus anthracis, a spore-forming Gram-positive bacterium, is a highly lethal infection. Antibodies targeting the protective antigen (PA) binding component of the toxins have recently been authorized as an adjunct to antibiotics, although no conclusive evidence demonstrates that anthrax antitoxin therapy has any significant benefit. We discuss here the rational basis of anti-PA development regarding the pathogenesis of the disease. We argue that inductive reasoning may induce therapeutic bias. We identified anthrax animal model analysis as another bias. Further studies are needed to assess the benefit of anti-PA antibodies in the treatment of inhalational anthrax, while a clearer consensus should be established around what evidence should be proven in an anthrax model.


Subject(s)
Anthrax/immunology , Anthrax/therapy , Antibodies, Bacterial/therapeutic use , Bacillus anthracis/immunology , Immunotherapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Bacterial/immunology , Antitoxins/therapeutic use , Bacterial Toxins/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Humans
7.
Cancer Med ; 8(6): 3167-3181, 2019 06.
Article in English | MEDLINE | ID: mdl-30950210

ABSTRACT

Successful treatment of cancer remains a challenge, due to the unique pathophysiology of solid tumors, and the predictable emergence of resistance. Traditional methods for cancer therapy including radiotherapy, chemotherapy, and immunotherapy all have their own limitations. A novel approach is bacteriotherapy, either used alone, or in combination with conventional methods, has shown a positive effect on regression of tumors and inhibition of metastasis. Bacteria-assisted tumor-targeted therapy used as therapeutic/gene/drug delivery vehicles has great promise in the treatment of tumors. The use of bacteria only, or in combination with conventional methods was found to be effective in some experimental models of cancer (tumor regression and increased survival rate). In this article, we reviewed the major advantages, challenges, and prospective directions for combinations of bacteria with conventional methods for tumor therapy.


Subject(s)
Bacteria , Biological Therapy/methods , Neoplasms/therapy , Animals , Bacteria/genetics , Bacteria/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Biological Therapy/adverse effects , Clinical Studies as Topic , Combined Modality Therapy/methods , Drug Delivery Systems , Drug Evaluation, Preclinical , Enzymes/genetics , Enzymes/metabolism , Gene Transfer Techniques , Humans
8.
Toxins (Basel) ; 11(4)2019 04 15.
Article in English | MEDLINE | ID: mdl-30991691

ABSTRACT

BACKGROUND: Beta-toxin (CPB) is the major virulence factor of Clostridium perfringens type C, causing hemorrhagic enteritis in newborn pigs but also other animals and humans. Vaccines containing inactivated CPB are known to induce protective antibody titers in sow colostrum and neutralization of the CPB activity is thought to be essential for protective immunity in newborn piglets. However, no method is available to quantify the neutralizing effect of vaccine-induced antibody titers in pigs. (2) Methods: We developed a novel assay for the quantification of neutralizing anti-CPB antibodies. Sera and colostrum of sows immunized with a commercial C. perfringens type A and C vaccine was used to determine neutralizing effects on CPB induced cytotoxicity in endothelial cells. Antibody titers of sows and their piglets were determined and compared to results obtained by an ELISA. (3) Results: Vaccinated sows developed neutralizing antibodies against CPB in serum and colostrum. Multiparous sows developed higher serum and colostrum antibody titers after booster vaccinations than uniparous sows. The antibody titers of sows and those of their piglets correlated highly. Piglets from vaccinated sows were protected against intraperitoneal challenge with C. perfringens type C supernatant. (4) Conclusions: The test based on primary porcine endothelial cells quantifies neutralizing antibody activity in serum and colostrum of vaccinated sows and could be used to reduce and refine animal experimentation during vaccine development.


Subject(s)
Antibodies, Neutralizing/immunology , Bacterial Toxins/immunology , Bacterial Vaccines/administration & dosage , Colostrum/immunology , Animals , Antibodies, Neutralizing/blood , Bacterial Toxins/genetics , Biological Assay , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Recombinant Proteins/pharmacology , Swine , Vaccination
9.
J Infect Dis ; 220(3): 505-513, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30897198

ABSTRACT

BACKGROUND: Enterotoxigenic Escherichia coli (ETEC) commonly cause diarrhea in children living in developing countries and in travelers to those regions. ETEC are characterized by colonization factors (CFs) that mediate intestinal adherence. We assessed if bovine colostral IgG (bIgG) antibodies against a CF, CS17, or antibodies against CsbD, the minor tip subunit of CS17, would protect subjects against diarrhea following challenge with a CS17-expressing ETEC strain. METHODS: Adult subjects were randomized (1:1:1) to receive oral bIgG against CS17, CsbD, or placebo. Two days prior to challenge, subjects began dosing 3 times daily with the bIgG products (or placebo). On day 3, subjects ingested 5 × 109 cfu ETEC strain LSN03-016011/A in buffer. Subjects were assessed for diarrhea for 120 hours postchallenge. RESULTS: A total of 36 subjects began oral prophylaxis and 35 were challenged with ETEC. While 50.0% of the placebo recipients had watery diarrhea, none of the subjects receiving anti-CS17 had diarrhea (P = .01). In contrast, diarrhea rates between placebo and anti-CsbD recipients (41.7%) were comparable (P = 1.0). CONCLUSIONS: This is the first study to demonstrate anti-CS17 antibodies provide significant protection against ETEC expressing CS17. More research is needed to better understand why anti-CsbD was not comparably efficacious. Clinical Trials Registration. NCT00524004.


Subject(s)
Antibodies, Bacterial/immunology , Colostrum/immunology , Diarrhea/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Vaccines/immunology , Protective Agents/pharmacology , Adhesins, Bacterial/immunology , Adult , Animals , Bacterial Toxins/immunology , Cattle , Colostrum/microbiology , Diarrhea/microbiology , Double-Blind Method , Enterotoxins/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/immunology , Female , Humans , Immunoglobulin G/immunology , Male
10.
Sci Rep ; 9(1): 715, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679470

ABSTRACT

Sublingual immunization is emerging as an alternative to nasal immunization and induction of mucosal IgA responses. Using Bacillus anthracis edema toxin (EdTx) as an adjuvant, we previously showed that innate responses triggered after sublingual immunization could limit generation of IgA responses. We tested whether co-administration of a neutrophil elastase inhibitor (NEI) could rescue the ability of EdTx to induce broad antibody responses, including mucosal IgA. NEI supplementation of sublingual vaccines containing EdTx promoted antigen-specific serum IgA responses but also enhanced serum IgG1, and IgG2b responses. This enhancing effect of NEI did not extend to all antibody isotypes and IgG sublclasses, since NEI  reduced serum IgE responses and did not affect IgG2a/c and IgG3 responses. NEI supplementation also promoted anti-Bacillus anthracis protective antigen (PA) neutralizing antibodies and enhanced high affinity IgG1 and IgA antibodies. In addition to serum IgA, NEI supplementation stimulated antigen-specific mucosal IgA responses in the GI tract, and enhanced antigen-specific IgG responses in vaginal washes. Analysis of CD4+ T helper cell responses revealed that co-administration of NEI broadened the profile of cytokine responses, by stimulating Th1, Th2, Th17, and Tfh cytokines. We also noted that NEI had a higher stimulatory effect on IL-5, IL-10, IL-17 responses.


Subject(s)
Antibody Formation/immunology , Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Dietary Supplements , Immunity, Mucosal/immunology , Mucous Membrane/immunology , Proteinase Inhibitory Proteins, Secretory/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic , Administration, Sublingual , Animals , Antigens, Bacterial/administration & dosage , Bacterial Toxins/administration & dosage , Female , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer , Vaccination
11.
mBio ; 9(5)2018 10 16.
Article in English | MEDLINE | ID: mdl-30327445

ABSTRACT

Bacillus anthracis and Yersinia pestis, the causative agents of anthrax and plague, respectively, are two of the deadliest pathogenic bacteria that have been used as biological warfare agents. Although Biothrax is a licensed vaccine against anthrax, no Food and Drug Administration-approved vaccine exists for plague. Here, we report the development of a dual anthrax-plague nanoparticle vaccine employing bacteriophage (phage) T4 as a platform. Using an in vitro assembly system, the 120- by 86-nm heads (capsids) of phage T4 were arrayed with anthrax and plague antigens fused to the small outer capsid protein Soc (9 kDa). The antigens included the anthrax protective antigen (PA) (83 kDa) and the mutated (mut) capsular antigen F1 and the low-calcium-response V antigen of the type 3 secretion system from Y. pestis (F1mutV) (56 kDa). These viral nanoparticles elicited robust anthrax- and plague-specific immune responses and provided complete protection against inhalational anthrax and/or pneumonic plague in three animal challenge models, namely, mice, rats, and rabbits. Protection was demonstrated even when the animals were simultaneously challenged with lethal doses of both anthrax lethal toxin and Y. pestis CO92 bacteria. Unlike the traditional subunit vaccines, the phage T4 vaccine uses a highly stable nanoparticle scaffold, provides multivalency, requires no adjuvant, and elicits broad T-helper 1 and 2 immune responses that are essential for complete clearance of bacteria during infection. Therefore, phage T4 is a unique nanoparticle platform to formulate multivalent vaccines against high-risk pathogens for national preparedness against potential bioterror attacks and emerging infections.IMPORTANCE Following the deadly anthrax attacks of 2001, the Centers for Disease Control and Prevention (CDC) determined that Bacillus anthracis and Yersinia pestis that cause anthrax and plague, respectively, are two Tier 1 select agents that pose the greatest threat to the national security of the United States. Both cause rapid death, in 3 to 6 days, of exposed individuals. We engineered a virus nanoparticle vaccine using bacteriophage T4 by incorporating key antigens of both B. anthracis and Y. pestis into one formulation. Two doses of this vaccine provided complete protection against both inhalational anthrax and pneumonic plague in animal models. This dual anthrax-plague vaccine is a strong candidate for stockpiling against a potential bioterror attack involving either one or both of these biothreat agents. Further, our results establish the T4 nanoparticle as a novel platform to develop multivalent vaccines against pathogens of high public health significance.


Subject(s)
Anthrax Vaccines/immunology , Anthrax/prevention & control , Antigens, Bacterial/immunology , Bacteriophage T4 , Plague Vaccine/immunology , Plague/prevention & control , Respiratory Tract Infections/prevention & control , Animals , Antibodies, Bacterial/blood , Bacillus anthracis , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Capsid Proteins/immunology , Female , Male , Mice , Mice, Inbred BALB C , Nanoparticles , Pore Forming Cytotoxic Proteins/immunology , Rabbits , Rats , Th1 Cells/immunology , Th2 Cells/immunology , Yersinia pestis
12.
Clin Pharmacol Drug Dev ; 7(6): 652-660, 2018 08.
Article in English | MEDLINE | ID: mdl-29125719

ABSTRACT

Inhalational anthrax is a highly lethal infection caused by Bacillus anthracis and a serious bioterrorism threat. Protective antigen (PA) is a critical component required for the virulence of Bacillus anthracis. Obiltoxaximab, a high-affinity monoclonal antibody that neutralizes PA, is approved in the United States for intravenous use for the treatment of inhalational anthrax in combination with appropriate antibacterial drugs and for prophylaxis of inhalational anthrax when alternative therapies are not available or appropriate. Here, we explored the safety, pharmacokinetics (PK), and immunogenicity of obiltoxaximab administered by intramuscular injection at doses of 4, 8, 16, 20, and 24 mg/kg in healthy humans. Systemic exposures were approximately dose proportional, maximum serum concentrations were observed after 6-9 days, and terminal half-life ranged from 16 to 23 days. Average absolute intramuscular bioavailability was 64%. Obiltoxaximab was well tolerated, and local tolerability was acceptable up to 24 mg/kg intramuscularly, up to 6 injections per dose, and up to 5 mL per injection. No injection-site abscesses or hypersensitivity reactions occurred; no subjects developed treatment-emergent antitherapeutic antibodies over the study period of 71 days.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antitoxins/adverse effects , Drug Hypersensitivity/immunology , Anthrax/drug therapy , Anthrax/immunology , Antibodies, Monoclonal/administration & dosage , Antigens, Bacterial/immunology , Antitoxins/administration & dosage , Area Under Curve , Bacillus anthracis/drug effects , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Healthy Volunteers , Humans , Injections, Intramuscular , Male , Middle Aged , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology
13.
Vaccine ; 35(49 Pt B): 6858-6865, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29102330

ABSTRACT

Necrotic enteritis (NE) is a severe disease of chickens and turkeys caused by some strains of Clostridium perfringens type A. The disease is well controlled by the use of in-feed antibiotic growth promoters (AGPs). However, due to worldwide public and regulatory pressure to reduce the use of AGPs inter alia, there is an urgent need to develop non-antibiotic based preventative measures. Vaccination would be a suitable control measure, but currently there is no commercial vaccine. NetB (necrotic enteritis toxin B-like) is a pore-forming toxin produced by C. perfringens that has been reported as an important virulence factor in the pathogenesis of NE. The present study tests a non-virulent NetB producing strain of C. perfringens (nvNetB+), with or without adjuvants, as an orally administered live vaccine. Adjuvants used were Gel 01™, Cholera toxin (CT), Escherichia coli wild type heat-labile holotoxin (LT) and mutant E. coli LT (dmLT) (R192G/L211A). Several vaccine administration regimes were tested. All vaccination regimes elicited serum and mucosal antibody responses to alpha toxin and to secreted proteins of both nvNetB+ and a very virulent NetB positive (vvNetB+) strain (p<0.0001 to p<0.05). In some vaccinated groups, there was milder intestinal pathology upon disease challenge. 55% of birds vaccinated orally at days 2, 12 with nvNetB+ adjuvanted with CT did not develop any lesions of NE by 6 days post challenge, compared to a 100% incidence of NE lesions in the unvaccinated disease challenged group.


Subject(s)
Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Clostridium Infections/prevention & control , Enteritis/veterinary , Enterotoxins/immunology , Poultry Diseases/prevention & control , Adjuvants, Immunologic , Administration, Oral , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Bacterial Vaccines/administration & dosage , Calcium-Binding Proteins/immunology , Chickens , Clostridium Infections/immunology , Clostridium perfringens/immunology , Enteritis/prevention & control , Enteritis/virology , Immunity, Mucosal , Intestines/immunology , Intestines/virology , Poultry Diseases/virology , Type C Phospholipases/immunology , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Virulence
14.
Article in English | MEDLINE | ID: mdl-28784679

ABSTRACT

The fluorocycline TP-271 was evaluated in mouse and nonhuman primate (NHP) models of inhalational anthrax. BALB/c mice were exposed by nose-only aerosol to Bacillus anthracis Ames spores at a level of 18 to 88 lethal doses sufficient to kill 50% of exposed individuals (LD50). When 21 days of once-daily dosing was initiated at 24 h postchallenge (the postexposure prophylaxis [PEP] study), the rates of survival for the groups treated with TP-271 at 3, 6, 12, and 18 mg/kg of body weight were 90%, 95%, 95%, and 84%, respectively. When 21 days of dosing was initiated at 48 h postchallenge (the treatment [Tx] study), the rates of survival for the groups treated with TP-271 at 6, 12, and 18 mg/kg TP-271 were 100%, 91%, and 81%, respectively. No deaths of TP-271-treated mice occurred during the 39-day posttreatment observation period. In the NHP model, cynomolgus macaques received an average dose of 197 LD50 of B. anthracis Ames spore equivalents using a head-only inhalation exposure chamber, and once-daily treatment of 1 mg/kg TP-271 lasting for 14 or 21 days was initiated within 3 h of detection of protective antigen (PA) in the blood. No (0/8) animals in the vehicle control-treated group survived, whereas all 8 infected macaques treated for 21 days and 4 of 6 macaques in the 14-day treatment group survived to the end of the study (56 days postchallenge). All survivors developed toxin-neutralizing and anti-PA IgG antibodies, indicating an immunologic response. On the basis of the results obtained with the mouse and NHP models, TP-271 shows promise as a countermeasure for the treatment of inhalational anthrax.


Subject(s)
Anthrax/drug therapy , Anti-Bacterial Agents/therapeutic use , Bacillus anthracis/drug effects , Respiratory Tract Infections/drug therapy , Tetracyclines/therapeutic use , Animals , Anthrax/microbiology , Anthrax/mortality , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antigens, Bacterial/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Disease Models, Animal , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Post-Exposure Prophylaxis/methods , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/mortality , Spores, Bacterial , Survival Rate , Tetracyclines/pharmacokinetics
15.
Expert Opin Biol Ther ; 17(11): 1439-1445, 2017 11.
Article in English | MEDLINE | ID: mdl-28805081

ABSTRACT

INTRODUCTION: Clostridium difficile infection is a major economic and clinical burden, due to its high frequency of recurrence. Currently recommended treatments are not efficient for prevention and may contribute to the risk of recurrent infection. In recent years, research has focused on strategies to lessen this risk. Bezlotoxumab is a monoclonal antibody that prevents recurrences of C. difficile infection through the antagonism of toxin B. Areas covered: In this review, the authors discuss the burden of C. difficile infection and its recurrences, the mechanisms underlying the recurrences, and current C. difficile treatments. They subsequently analyze the strategic therapeutic rationale for bezlotoxumab use, as well as the supporting clinical evidence. Expert opinion: Bezlotoxumab is an attractive solution for reducing the unacceptable level of recurrence that occurs with the currently recommended C. difficile treatments and other alternative therapies under consideration. Even though bezlotoxumab has not been tested in large-scale trials exclusively in cases of already established recurrent C.difficile infection (rCDI), it has an advantage over current treatments in that it does not interfere with the patient's gut flora while directly neutralizing the key virulence factor. Although cost remains an important factor against its widespread use, simpler administration, fewer side-effects, and better social acceptability justify its consideration for treating rCDI.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Clostridium Infections/drug therapy , Anti-Bacterial Agents/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Broadly Neutralizing Antibodies , Clinical Trials as Topic , Clostridium Infections/pathology , Half-Life , Humans , Recurrence , Vancomycin/therapeutic use
16.
J Clin Invest ; 127(6): 2249-2261, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28463232

ABSTRACT

Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Bispecific , Antigens, Bacterial/immunology , Bacterial Load , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Complement System Proteins/metabolism , Drug Evaluation, Preclinical , Female , Kupffer Cells/microbiology , Lung/blood supply , Lung/microbiology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Microvessels/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections/immunology , Receptors, Fc/metabolism
17.
Toxins (Basel) ; 9(1)2016 12 23.
Article in English | MEDLINE | ID: mdl-28025545

ABSTRACT

Atopic dermatitis (AD) is a common allergic skin disease that is associated with chronic, recurrent eczematous and pruritic lesions at the flexural folds caused by interacting factors related to environmental and immune system changes. AD results in dry skin, and immunoglobulin E-mediated allergic reactions to foods and environmental allergens. While steroids and anti-histamines temporarily relieve the symptoms of AD, the possibility of side effects from pharmacological interventions remains. Despite intensive research, the underlying mechanisms for AD have not been clarified. A study of Staphylococcus aureus (S. aureus) established the role of its toxins in the pathogenesis of AD. Approximately 90% of patients with AD experience S. aureus colonization and up to 50%-60% of the colonizing S. aureus is toxin-producing. Any damage to the protective skin barrier allows for the entry of invading allergens and pathogens that further drive the pathogenesis of AD. Some natural toxins (or their components) that have therapeutic effects on AD have been studied. In addition, recent studies on inflammasomes as one component of the innate immune system have been carried out. Additionally, studies on the close relationship between the activation of inflammasomes and toxins in AD have been reported. This review highlights the literature that discusses the pathogenesis of AD, the role of toxins in AD, and the positive and negative effects of toxins on AD. Lastly, suggestions are made regarding the role of inflammasomes in AD.


Subject(s)
Bacterial Toxins/immunology , Dermatitis, Atopic/immunology , Allergens/immunology , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/microbiology , Dermatologic Agents/pharmacology , Disease Models, Animal , Humans , Plant Extracts/pharmacology , Protective Agents/pharmacology , Randomized Controlled Trials as Topic , Skin/immunology , Skin/microbiology , Staphylococcus aureus/immunology
18.
Clin Vaccine Immunol ; 23(12): 908-917, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27581434

ABSTRACT

Several candidate vaccines against Shigella spp. are in development, but the lack of a clear correlate of protection from challenge with the induction of adequate immune responses among the youngest age groups in the developing world has hampered Shigella vaccine development over the past several decades. Bioconjugation technology, exploited here for an Shigella flexneri 2a candidate vaccine, offers a novel and potentially cost-effective way to develop and produce vaccines against a major pathogen of global health importance. Flexyn2a, a novel S. flexneri 2a bioconjugate vaccine made of the polysaccharide component of the S. flexneri 2a O-antigen, conjugated to the exotoxin protein A of Pseudomonas aeruginosa (EPA), was evaluated for safety and immunogenicity among healthy adults in a single-blind, phase I study with a staggered randomization approach. Thirty subjects (12 receiving 10 µg Flexyn2a, 12 receiving Flexyn2a with aluminum adjuvant, and 6 receiving placebo) were administered two injections 4 weeks apart and were followed for 168 days. Flexyn2a was well-tolerated, independently of the adjuvant and number of injections. The Flexyn2a vaccine elicited statistically significant S. flexneri 2a lipopolysaccharide (LPS)-specific humoral responses at all time points postimmunization in all groups that received the vaccine. Elicited serum antibodies were functional, as evidenced by bactericidal activity against S. flexneri 2a. The bioconjugate candidate vaccine Flexyn2a has a satisfactory safety profile and elicited a robust humoral response to S. flexneri 2a LPS with or without inclusion of an adjuvant. Moreover, the bioconjugate also induced functional antibodies, showing the technology's features in producing a promising candidate vaccine. (This study has been registered at ClinicalTrials.gov under registration no. NCT02388009.).


Subject(s)
Antibodies, Bacterial/blood , Dysentery, Bacillary/prevention & control , Immunogenicity, Vaccine , Shigella Vaccines/adverse effects , Shigella Vaccines/immunology , Shigella flexneri/immunology , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , Adolescent , Adult , Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Dysentery, Bacillary/immunology , Exotoxins/genetics , Exotoxins/immunology , Female , Healthy Volunteers , Humans , Immunoglobulin A/immunology , Lipopolysaccharides/immunology , Male , Middle Aged , O Antigens/immunology , Shigella Vaccines/administration & dosage , Shigella sonnei/immunology , Single-Blind Method , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology , Virulence Factors/genetics , Virulence Factors/immunology , Young Adult , Pseudomonas aeruginosa Exotoxin A
19.
Vaccine ; 34(35): 4152-4160, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27395567

ABSTRACT

BACKGROUND: Extra-intestinal pathogenic Escherichia coli (ExPEC) are major human pathogens; however, no protective vaccine is currently available. We assessed in animal models the immunogenicity and safety of a 4-valent E. coli conjugate vaccine (ExPEC-4V, serotypes O1, O2, O6 and O25 conjugated to Exotoxin A from Pseudomonas aeruginosa (EPA)) produced using a novel in vivo bioconjugation method. METHODS: Three doses of ExPEC-4V (with or without aluminum hydroxide) were administered to rabbits (2µg or 20µg per O-antigen, subcutaneously), mice (0.2µg or 2µg per O-antigen, subcutaneously) and rats (0.4µg or 4µg per O-antigen, intramuscularly). Antibody persistence and boostability were evaluated in rats using O6-EPA monovalent conjugate (0.4µg O-antigen/dose, intramuscularly). Toxicity was assessed in rats (16µg total polysaccharide, intramuscularly). Serum IgG and IgM antibodies were measured by ELISA. RESULTS: Robust antigen-specific IgG responses were observed in all animal models, with increased responses in rabbits when administered with adjuvant. O antigen-specific antibody responses persisted up to 168days post-priming. Booster immunization induced a rapid recall response. Toxicity of ExPEC-4V when administered to rats was considered to be at the no observed adverse effect level. CONCLUSIONS: ExPEC-4V conjugate vaccine showed good immunogenicity and tolerability in animal models supporting progression to clinical evaluation.


Subject(s)
Escherichia coli Infections/prevention & control , Escherichia coli Vaccines/immunology , Immunogenicity, Vaccine , O Antigens/immunology , ADP Ribose Transferases/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Bacterial/blood , Bacterial Toxins/immunology , Escherichia coli , Exotoxins/immunology , Female , Immunization, Secondary , Mice , Mice, Inbred ICR , No-Observed-Adverse-Effect Level , Rabbits , Rats , Rats, Sprague-Dawley , Toxicity Tests , Vaccines, Conjugate/immunology , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
20.
BMC Immunol ; 17(1): 10, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27141950

ABSTRACT

BACKGROUND: In genetically modified (GM) crops there is a risk that the inserted genes may introduce new allergens and/or adjuvants into the food and feed chain. The MON810 maize, expressing the insecticidal Cry1Ab toxin, is grown in many countries worldwide. In animal models, intranasal and intraperitoneal immunisations with the purified Cry1Ab proteins have induced immune responses, and feeding trials with Cry1Ab-containing feed have revealed some altered immune responses. Previous investigations have primarily measured antibody responses to the protein, while investigations of clinical food allergy symptoms, or allergy promotion (adjuvant effect) associated with the Cry1Ab protein are largely missing. We aimed to investigate immunogenic, allergenic and adjuvant properties of purified Cry1Ab toxin (trypCry1Ab, i.e., trypsin activated Cry1Ab) in a mouse model of food allergy. METHOD: Female C3H/HeJ mice were immunized by intragastric gavage of 10 µg purified, trypsin activated Cry1Ab toxin (trypCry1Ab) alone or together with the food allergen lupin. Cholera toxin was added as a positive control for adjuvant effect to break oral tolerance. Clinical symptoms (anaphylaxis) as well as humoral and cellular responses were assessed. RESULTS: In contrast to results from previous airway investigations, we observed no indication of immunogenic properties of trypCry1Ab protein after repeated intragastric exposures to one dose, with or without CT as adjuvant. Moreover, the results indicated that trypCry1Ab given by the intragastric route was not able to promote allergic responses or anaphylactic reactions against the co-administered allergen lupin at the given dose. CONCLUSION: The study suggests no immunogenic, allergenic or adjuvant capacity of the given dose of trypCry1Ab protein after intragastric exposure of prime aged mice.


Subject(s)
Allergens/immunology , Cryptochromes/immunology , Food Hypersensitivity/immunology , Insect Proteins/immunology , Intestines/immunology , Plant Extracts/immunology , Zea mays/immunology , Animals , Bacterial Toxins/immunology , Cryptochromes/metabolism , Edible Grain , Female , Food, Genetically Modified , Immunoglobulin E/metabolism , Intestines/microbiology , Lupinus/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Organisms, Genetically Modified , Proteolysis , Trypsin/metabolism , Zea mays/genetics
SELECTION OF CITATIONS
SEARCH DETAIL