Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.360
Filter
1.
Vaccine ; 42(9): 2171-2180, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38461051

ABSTRACT

Tularemia is caused by subspecies of Francisella tularensis and can manifest in a variety of disease states, with the pneumonic presentation resulting in the greatest mortality. Despite decades of research, there are no approved vaccines against F. tularensis in the United States. Traditional vaccination strategies, such as live-attenuated or subunit vaccines, are not favorable due to inadequate protection or safety concerns. Because of this, novel vaccination strategies are needed to combat tularemia. Here we discuss the current state of and challenges to the tularemia vaccine field and suggest novel vaccine approaches going forward that might be better suited for protecting against F. tularensis infection.


Subject(s)
Francisella tularensis , Tularemia , Humans , Tularemia/prevention & control , Bacterial Vaccines/therapeutic use , Vaccines, Attenuated , Vaccination
2.
Front Immunol ; 14: 1126170, 2023.
Article in English | MEDLINE | ID: mdl-37090699

ABSTRACT

Syphilis is a sexually or vertically (mother to fetus) transmitted disease caused by the infection of Treponema pallidum subspecie pallidum (TPA). The incidence of syphilis has increased over the past years despite the fact that this bacterium is an obligate human pathogen, the infection route is well known, and the disease can be successfully treated with penicillin. As complementary measures to preventive campaigns and early treatment of infected individuals, development of a syphilis vaccine may be crucial for controlling disease spread and/or severity, particularly in countries where the effectiveness of the aforementioned measures is limited. In the last century, several vaccine prototypes have been tested in preclinical studies, mainly in rabbits. While none of them provided protection against infection, some prototypes prevented bacteria from disseminating to distal organs, attenuated lesion development, and accelerated their healing. In spite of these promising results, there is still some controversy regarding the identification of vaccine candidates and the characteristics of a syphilis-protective immune response. In this review, we describe what is known about TPA immune response, and the main mechanisms used by this pathogen to evade it. Moreover, we emphasize the importance of integrating this knowledge, in conjunction with the characterization of outer membrane proteins (OMPs), to expedite the development of a syphilis vaccine that can protect against TPA infection.


Subject(s)
Syphilis , Animals , Humans , Rabbits , Syphilis/epidemiology , Syphilis/prevention & control , Treponema pallidum , Immunity , Bacterial Vaccines/therapeutic use
3.
Mol Aspects Med ; 92: 101183, 2023 08.
Article in English | MEDLINE | ID: mdl-37018869

ABSTRACT

The gram-negative bacterium Helicobacter pylori is the most common chronic bacterial infection and the main cause of gastric cancer. Due to the increasing antimicrobial resistance of H. pylori, the development of an efficacious vaccine is a valid option to protect from disease or infection and ultimately prevent gastric cancer. However, despite more than 30 years of research, no vaccine has entered the market yet. This review highlights the most relevant previous preclinical and clinical studies to allow conclusions to be drawn on which parameters need special attention in the future to develop an efficacious vaccine against H. pylori and thus prevent gastric cancer.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Stomach Neoplasms/prevention & control , Stomach Neoplasms/drug therapy , Stomach Neoplasms/microbiology , Bacterial Vaccines/therapeutic use , Helicobacter Infections/complications , Helicobacter Infections/prevention & control , Vaccination
4.
Lancet Microbe ; 4(2): e113-e125, 2023 02.
Article in English | MEDLINE | ID: mdl-36528040

ABSTRACT

Vaccines can be highly effective tools in combating antimicrobial resistance as they reduce infections caused by antibiotic-resistant bacteria and antibiotic consumption associated with disease. This Review looks at vaccine candidates that are in development against pathogens on the 2017 WHO bacterial priority pathogen list, in addition to Clostridioides difficile and Mycobacterium tuberculosis. There were 94 active preclinical vaccine candidates and 61 active development vaccine candidates. We classified the included pathogens into the following four groups: Group A consists of pathogens for which vaccines already exist-ie, Salmonella enterica serotype Typhi, Streptococcus pneumoniae, Haemophilus influenzae type b, and M tuberculosis. Group B consists of pathogens with vaccines in advanced clinical development-ie, extra-intestinal pathogenic Escherichia coli, Salmonella enterica serotype Paratyphi A, Neisseria gonorrhoeae, and C difficile. Group C consists of pathogens with vaccines in early phases of clinical development-ie, enterotoxigenic E coli, Klebsiella pneumoniae, non-typhoidal Salmonella, Shigella spp, and Campylobacter spp. Finally, group D includes pathogens with either no candidates in clinical development or low development feasibility-ie, Pseudomonas aeruginosa, Acinetobacter baumannii, Staphylococcus aureus, Helicobacter pylori, Enterococcus faecium, and Enterobacter spp. Vaccines are already important tools in reducing antimicrobial resistance and future development will provide further opportunities to optimise the use of vaccines against resistance.


Subject(s)
Anti-Bacterial Agents , Enterococcus faecium , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Vaccines/therapeutic use , Escherichia coli , Drug Resistance, Bacterial
5.
Allergy Asthma Proc ; 43(6): 501-508, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36335411

ABSTRACT

Background: Orally or sublingually administered lysates of mixed respiratory pathogenic bacteria have been used in Europe, Asia, and other parts of the world, but not the United States, since the mid 1950s, first to prevent recurrent respiratory tract infections, later to prevent wheezing and asthma exacerbations associated with respiratory infections, and, more recently, for the treatment of allergic rhinitis. The apparent success of this treatment contrasts with the negative experience of treating with injections of similar mixed respiratory bacterial vaccines (MRBV or BV) to prevent asthma exacerbations associated with respiratory infections that was once common practice but abandoned ∼50 years ago. Methods: Textbooks and articles on the use of injected BVs to prevent asthma exacerbations associated with respiratory infections were reviewed, including a number of, randomized, double-blind, placebo controlled (RDBPC) studies the results of which were predominantly negative that contributed to the abandonment of this treatment. Also reviewed were more recent articles from Europe and China, which report both clinical and immunologic support for the use of the orally and sublingually administered mixed respiratory bacterial lysates (MRBL or BL). Results: A review of five RDBPC studies of the parenteral use of BVs for prevention of asthma exacerbations conducted by leading international allergists in the 1950s and 1960s showed, in a combined 532 patients, an overall reduction of asthma attacks by 4.9% over placebo. However, in five studies in 1126 patients of oral or sublingual treatment with BLs, the reduction in respiratory infections, wheezing episodes, and asthma exacerbations was 42.6% over placebo. Conclusion: Reported results with oral and sublingual BLs are far superior to the historical performance of injected BVs. Possible reasons for this difference are discussed, but none is clearly responsible for the difference in clinical results.


Subject(s)
Asthma , Respiratory Tract Infections , Rhinitis, Allergic , Humans , Respiratory Sounds , Asthma/drug therapy , Respiratory Tract Infections/prevention & control , Bacterial Vaccines/therapeutic use , Randomized Controlled Trials as Topic
6.
Curr Opin Allergy Clin Immunol ; 22(6): 380-386, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36305468

ABSTRACT

PURPOSE OF REVIEW: To discuss recently discovered mechanisms of action of some bacterial vaccines that may account for their clinical benefit in the prevention of recurrent wheezing and asthma exacerbations in infants and early childhood. RECENT FINDINGS: Trained immunity has been shown to confer innate immune cells with a quite long-term nonspecific protection against a broad spectrum of pathogens. Inducers of trained immunity include some bacterial vaccines. Trained immunity-based vaccines (TIbV) of bacterial origin have the capability to induce nonspecific responses to a variety of pathogens, including respiratory viruses, in addition to their nominal bacterial antigens. Clinical data, from epidemiological surveys to well designed randomized clinical trials, indicate that TIbV formulated with bacteria prevent respiratory tract infections of viral cause, such as those associated with recurrent wheezing or asthma exacerbation, in children. Administration of these vaccines by the mucosal route may be important for their outcome in respiratory infections. SUMMARY: Mucosal bacterial immunotherapy, including certain TIbV, confer protection against a broad spectrum of pathogens, such as viruses, through a mechanism mediated by trained immunity. Clinical studies on the use of these preparations against recurrent wheezing reflect these mechanistic effects. These findings open a new avenue for the development of new strategies for this condition.


Subject(s)
Asthma , Respiratory Tract Infections , Child , Infant , Child, Preschool , Humans , Respiratory Sounds , Adjuvants, Vaccine , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/drug therapy , Bacteria , Bacterial Vaccines/therapeutic use
8.
Proc Natl Acad Sci U S A ; 119(25): e2202059119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35714289

ABSTRACT

The bacterial genus Bartonella comprises numerous emerging pathogens that cause a broad spectrum of disease manifestations in humans. The targets and mechanisms of the anti-Bartonella immune defense are ill-defined and bacterial immune evasion strategies remain elusive. We found that experimentally infected mice resolved Bartonella infection by mounting antibody responses that neutralized the bacteria, preventing their attachment to erythrocytes and suppressing bacteremia independent of complement or Fc receptors. Bartonella-neutralizing antibody responses were rapidly induced and depended on CD40 signaling but not on affinity maturation. We cloned neutralizing monoclonal antibodies (mAbs) and by mass spectrometry identified the bacterial autotransporter CFA (CAMP-like factor autotransporter) as a neutralizing antibody target. Vaccination against CFA suppressed Bartonella bacteremia, validating CFA as a protective antigen. We mapped Bartonella-neutralizing mAb binding to a domain in CFA that we found is hypervariable in both human and mouse pathogenic strains, indicating mutational antibody evasion at the Bartonella subspecies level. These insights into Bartonella immunity and immune evasion provide a conceptual framework for vaccine development, identifying important challenges in this endeavor.


Subject(s)
Antibodies, Neutralizing , Antigens, Bacterial , Bacteremia , Bartonella Infections , Bartonella , Type V Secretion Systems , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacteremia/immunology , Bacteremia/microbiology , Bacteremia/prevention & control , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Bartonella/genetics , Bartonella/immunology , Bartonella Infections/immunology , Bartonella Infections/microbiology , Bartonella Infections/prevention & control , Cloning, Molecular , Immune Evasion , Mice , Type V Secretion Systems/immunology , Vaccination
9.
Urol Int ; 106(7): 730-736, 2022.
Article in English | MEDLINE | ID: mdl-35130558

ABSTRACT

INTRODUCTION: Recurrent urinary tract infections (rUTIs) affect 5-10% of women, resulting in an enormous healthcare and society burden. Uromune® is a polybacterial sublingual vaccine with an excellent clinical benefit in rUTI prophylaxis. This study assesses the impact of sublingual vaccination on healthcare resource use and expenditures associated with this pathology. METHODS: A quasi-experimental, pretest-posttest, single center study including women with rUTI and vaccinated with Uromune® in real-life clinical practice was performed. Variables were the need of healthcare resources, collected prospectively during two follow-up years, and the rUTI-associated expenditure, calculated using the micro-costing methodology; these were compared before and after vaccination. RESULTS: A total of 166 women {mean (standard deviation [SD]) urinary tract infection episodes/year 6.19 (2.15)} were included. After vaccination, annual consultations with a primary care physician (PCP) (43.9%), emergency room visits (71.8%), urinary analysis (90.0%), and ultrasound exams (35.6%) decreased compared to pre-vaccination (all p < 0.001). Per patient consumption in antibiotics, PCP consultations, emergency room visits, and complementary exams significantly decreased (all p < 0.02), resulting in a reduction in healthcare expenditure per patient/year from mean (SD) 1,001.1 (655.0) to 497.1 (444.4) EUR. CONCLUSION: Sublingual bacterial vaccination with Uromune® decreased healthcare resource use and associated expenditure in women with rUTI, representing an optimal strategy to reduce rUTI-associated healthcare and economic burden.


Subject(s)
Urinary Tract Infections , Anti-Bacterial Agents/therapeutic use , Bacterial Vaccines/therapeutic use , Cost-Benefit Analysis , Female , Humans , Recurrence , Urinary Tract Infections/drug therapy
10.
Int J Mol Sci ; 23(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35055181

ABSTRACT

The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell-cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/therapeutic use , Dysentery, Bacillary/prevention & control , Shigella/growth & development , Animals , Bacterial Vaccines/pharmacology , Disease Models, Animal , Drug Development , Dysentery, Bacillary/immunology , Humans , Microbial Viability/drug effects , Shigella/drug effects , Shigella/metabolism , Vaccination
11.
Vet Med Sci ; 8(2): 578-590, 2022 03.
Article in English | MEDLINE | ID: mdl-34687172

ABSTRACT

BACKGROUND: This field trial was designed to evaluate the efficacy of a new trivalent vaccine containing porcine circovirus type 2a and 2b (PCV2a/b), and Mycoplasma hyopneumoniae at three independent locations. METHODS: Three farms were selected based on their history of PCV2 and M. hyopneumoniae co-infection. Each farm housed a total of 60, 3-day-old pigs that were randomly allocated to one of three treatment groups. Pigs were administered the trivalent vaccine intramuscularly with either a 1.0 ml dose at 3 and 24 days of age or a 2.0 ml dose at 21 days of age in accordance with the manufacturer's recommendations. RESULTS: Clinically, the average daily weight gain of the one-dose and two-dose vaccinated groups within all three farms was significantly higher (p < 0.05) than those of unvaccinated animals during the growing (70-112 days of age), finishing (112-175 days of age) and overall (3-175 days of age) stages of production. One-dose and two-dose vaccinated animals elicited neutralizing antibodies and interferon-γ-secreting cells (IFN-γ-SC), which reduced the amount of PCV2 in terms of blood load and reduced the severity of lymphoid lesions when compared with unvaccinated animals. Similarly, one-dose and two-dose vaccinated animals elicited IFN-γ-SC, which reduced the amount of M. hyopneumoniae in terms of laryngeal load and reduced the severity of lung lesions. CONCLUSIONS: The intramuscular administration of either one or two doses of trivalent vaccine was not significantly different in any of the evaluated parameters. The results of field trial demonstrated that the trivalent vaccine was efficacious in the protection of swine herds where PCV2d and M. hyopneumoniae were in active circulation.


Subject(s)
Circoviridae Infections , Circovirus , Mycoplasma hyopneumoniae , Swine Diseases , Viral Vaccines , Animals , Bacterial Vaccines/therapeutic use , Circoviridae Infections/prevention & control , Circoviridae Infections/veterinary , Interferon-gamma , Swine , Swine Diseases/prevention & control
12.
Int Immunopharmacol ; 101(Pt A): 108184, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34601334

ABSTRACT

Trichinellosis is a foodborne zoonosis caused by Trichinella spiralis (T. spiralis) that not only causes considerable economic losses for the global pig breeding and food industries, but also seriously threats the health of human. Therefore, it is very necessary to develop an effective vaccine to prevent trichinellosis. In this study, the invasive Lactobacillus plantarum (L. plantarum) expressing fibronectin-binding protein A (FnBPA) was served as a live bacterial vector to deliver DNA to the host to produce a novel oral DNA vaccine. Co-expressing T. spiralis SS1 and murine interleukin-4 (mIL-4) of DNA vaccine were constructed and subsequently delivered to intestinal epithelial cells via invasive L. plantarum. At 10 days after the third immunization, the experimental mice were challenged with 350 T. spiralis infective larvae. The results found that the mice orally vaccinated with invasive L. plantarum harboring pValac-SS1/pSIP409-FnBPA not only stimulated the production of anti-SS1-specific IgG, Th1/Th2 cell cytokines, and secreted(s) IgA but also decreased worm burden and intestinal damage. However, the mice inoculated with invasive L. plantarum co-expressing SS1 and mIL-4 (pValac-SS1-IL-4/pSIP409-FnBPA) induced the highest protective immune response against T. spiralis infection. The DNA vaccine delivered by invasive L. plantarum provides a novel idea for the prevention of T. spiralis infection.


Subject(s)
Bacterial Vaccines/therapeutic use , Endodeoxyribonucleases/genetics , Helminth Proteins/genetics , Interleukin-4/genetics , Lactobacillus plantarum/immunology , Nucleic Acid-Based Vaccines/therapeutic use , Trichinella spiralis/immunology , Trichinellosis/prevention & control , Administration, Oral , Animals , Blotting, Western , Endodeoxyribonucleases/immunology , Fluorescent Antibody Technique , Helminth Proteins/immunology , Interleukin-4/immunology , Male , Mice , Mice, Inbred BALB C , Trichinellosis/immunology , Vaccines, Synthetic/therapeutic use
13.
Int Urol Nephrol ; 53(11): 2267-2272, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34499326

ABSTRACT

PURPOSE: To assess the efficacy of prophylaxis for urinary tract infections (UTI) in a two-year follow-up in women with StroVac compared to a therapy with Nitrofurantoin over three months. MATERIALS AND METHODS: All patients with documented recurrent urinary tract infections (rUTI) were offered vaccination with StroVac or therapy with three months Nitrofurantoin 100 mg once daily for three months at patient's choice. Only patients with a follow-up of at least 24 months were included. All episodes with signs of UTI were documented and urine culture was performed. Success was defined as one or none UTI per 12 months, documented by urine culture. StroVac booster injection was offered 12 months after primary vaccination at patient's choice. RESULTS: 173 patients were included in this study, 124 in the StroVac group, 49 chose Nitrofuratoin. In the first 12 months, 86.8% of patients in the StroVac group and 91.8% in Nitrofurantoin group were successful (p = 0.22). Side effects were noted in 2.3% in the StroVac group causing discontinuation of therapy, whereas in the Nitrofurantoin group 18.4% stopped medication premature, mostly due to mild diarrhoea. In the second year 79.3% of patients in the StroVac group were still successful, most of them had undergone booster injection. In contrast, in the Nitrofurantoin group only 59.2% of patients were still successful (p = 0.03). CONCLUSION: StroVac is an effective and lasting non-antibiotic prophylaxis for rUTI, easy to administer with low rates of adverse events and should be offered to patients with rUTI.


Subject(s)
Anti-Infective Agents, Urinary/therapeutic use , Bacterial Vaccines/therapeutic use , Nitrofurantoin/therapeutic use , Urinary Tract Infections/prevention & control , Adult , Aged , Bacterial Vaccines/immunology , Female , Humans , Middle Aged , Recurrence
14.
J Infect Dis ; 224(12 Suppl 2): S194-S203, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34469556

ABSTRACT

BACKGROUND: As part of the global Invasive Bacterial Vaccine-Preventable Diseases Surveillance Network, 12 African countries referred cerebrospinal fluid (CSF) samples to South Africa's regional reference laboratory. We evaluated the utility of real-time polymerase chain reaction (PCR) in detecting and serotyping/grouping Haemophilus influenzae, Neisseria meningitidis, and Streptococcus pneumoniae (HNS). METHODS: From 2008 to 2017, CSF samples collected from children <5 years old with suspected meningitis underwent routine microbiology testing in-country, and 11 680 samples were submitted for HNS PCR at the regional reference laboratory. Unconditional logistic regression, with adjustment for geographic location, was performed to identify factors associated with PCR positivity. RESULTS: The overall HNS PCR positivity rate for all countries was 10% (1195 of 11 626 samples). In samples with both PCR and culture results, HNS PCR positivity was 11% (744 of 6747 samples), and HNS culture positivity was 3% (207 of 6747). Molecular serotype/serogroup was assigned in 75% of PCR-positive specimens (762 of 1016). Compared with PCR-negative CSF samples, PCR-positive samples were more often turbid (adjusted odds ratio, 6.80; 95% confidence interval, 5.67-8.17) and xanthochromic (1.72; 1.29-2.28), had elevated white blood cell counts (6.13; 4.71-7.99) and high protein concentrations (5.80; 4.34-7.75), and were more often HNS culture positive (32.70; 23.18-46.12). CONCLUSION: PCR increased detection of vaccine-preventable bacterial meningitis in countries where confirmation of suspected meningitis cases is impeded by limited culture capacity.


Subject(s)
Haemophilus influenzae/genetics , Meningitis, Bacterial/diagnosis , Neisseria meningitidis/genetics , Real-Time Polymerase Chain Reaction/methods , Streptococcus pneumoniae/genetics , Africa, Eastern/epidemiology , Africa, Southern/epidemiology , Bacterial Vaccines/therapeutic use , Child, Preschool , Female , Haemophilus influenzae/isolation & purification , Humans , Infant , Infant, Newborn , Male , Meningitis, Bacterial/epidemiology , Meningitis, Bacterial/genetics , Molecular Diagnostic Techniques , Neisseria meningitidis/isolation & purification , Public Health Surveillance , Streptococcus pneumoniae/isolation & purification
15.
Biomolecules ; 11(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34203937

ABSTRACT

Bacterial secretory systems are essential for virulence in human pathogens. The systems have become a target of alternative antibacterial strategies based on small molecules and antibodies. Strategies to use components of the systems to design prophylactics have been less publicized despite vaccines being the preferred solution to dealing with bacterial infections. In the current review, strategies to design vaccines against selected pathogens are presented and connected to the biology of the system. The examples are given for Y. pestis, S. enterica, B. anthracis, S. flexneri, and other human pathogens, and discussed in terms of effectiveness and long-term protection.


Subject(s)
Bacteria , Bacterial Infections , Bacterial Proteins/immunology , Bacterial Vaccines , Bacteria/immunology , Bacteria/pathogenicity , Bacterial Infections/immunology , Bacterial Infections/prevention & control , Bacterial Secretion Systems , Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Humans , Virulence/immunology
16.
Elife ; 102021 07 13.
Article in English | MEDLINE | ID: mdl-34253291

ABSTRACT

Background: Vaccination is one of the most effective public health interventions. We investigate the impact of vaccination activities for Haemophilus influenzae type b, hepatitis B, human papillomavirus, Japanese encephalitis, measles, Neisseria meningitidis serogroup A, rotavirus, rubella, Streptococcus pneumoniae, and yellow fever over the years 2000-2030 across 112 countries. Methods: Twenty-one mathematical models estimated disease burden using standardised demographic and immunisation data. Impact was attributed to the year of vaccination through vaccine-activity-stratified impact ratios. Results: We estimate 97 (95%CrI[80, 120]) million deaths would be averted due to vaccination activities over 2000-2030, with 50 (95%CrI[41, 62]) million deaths averted by activities between 2000 and 2019. For children under-5 born between 2000 and 2030, we estimate 52 (95%CrI[41, 69]) million more deaths would occur over their lifetimes without vaccination against these diseases. Conclusions: This study represents the largest assessment of vaccine impact before COVID-19-related disruptions and provides motivation for sustaining and improving global vaccination coverage in the future. Funding: VIMC is jointly funded by Gavi, the Vaccine Alliance, and the Bill and Melinda Gates Foundation (BMGF) (BMGF grant number: OPP1157270 / INV-009125). Funding from Gavi is channelled via VIMC to the Consortium's modelling groups (VIMC-funded institutions represented in this paper: Imperial College London, London School of Hygiene and Tropical Medicine, Oxford University Clinical Research Unit, Public Health England, Johns Hopkins University, The Pennsylvania State University, Center for Disease Analysis Foundation, Kaiser Permanente Washington, University of Cambridge, University of Notre Dame, Harvard University, Conservatoire National des Arts et Métiers, Emory University, National University of Singapore). Funding from BMGF was used for salaries of the Consortium secretariat (authors represented here: TBH, MJ, XL, SE-L, JT, KW, NMF, KAMG); and channelled via VIMC for travel and subsistence costs of all Consortium members (all authors). We also acknowledge funding from the UK Medical Research Council and Department for International Development, which supported aspects of VIMC's work (MRC grant number: MR/R015600/1).JHH acknowledges funding from National Science Foundation Graduate Research Fellowship; Richard and Peggy Notebaert Premier Fellowship from the University of Notre Dame. BAL acknowledges funding from NIH/NIGMS (grant number R01 GM124280) and NIH/NIAID (grant number R01 AI112970). The Lives Saved Tool (LiST) receives funding support from the Bill and Melinda Gates Foundation.This paper was compiled by all coauthors, including two coauthors from Gavi. Other funders had no role in study design, data collection, data analysis, data interpretation, or writing of the report. All authors had full access to all the data in the study and had final responsibility for the decision to submit for publication.


Subject(s)
Bacterial Infections/prevention & control , Bacterial Vaccines/therapeutic use , COVID-19 , Global Health , Models, Biological , SARS-CoV-2 , Bacterial Infections/epidemiology , Humans
17.
Sci Rep ; 11(1): 14215, 2021 07 09.
Article in English | MEDLINE | ID: mdl-34244557

ABSTRACT

Clostridium difficile is a spore-forming gram-positive bacterium, recognized as the primary cause of antibiotic-associated nosocomial diarrhoea. Clostridium difficile infection (CDI) has emerged as a major health-associated infection with increased incidence and hospitalization over the years with high mortality rates. Contamination and infection occur after ingestion of vegetative spores, which germinate in the gastro-intestinal tract. The surface layer protein and flagellar proteins are responsible for the bacterial colonization while the spore coat protein, is associated with spore colonization. Both these factors are the main concern of the recurrence of CDI in hospitalized patients. In this study, the CotE, SlpA and FliC proteins are chosen to form a multivalent, multi-epitopic, chimeric vaccine candidate using the immunoinformatics approach. The overall reliability of the candidate vaccine was validated in silico and the molecular dynamics simulation verified the stability of the vaccine designed. Docking studies showed stable vaccine interactions with Toll-Like Receptors of innate immune cells and MHC receptors. In silico codon optimization of the vaccine and its insertion in the cloning vector indicates a competent expression of the modelled vaccine in E. coli expression system. An in silico immune simulation system evaluated the effectiveness of the candidate vaccine to trigger a protective immune response.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Vaccines/therapeutic use , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Clostridium Infections/immunology , Computational Biology/methods , Escherichia coli/metabolism , Humans
18.
ACS Appl Mater Interfaces ; 13(28): 32703-32715, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34251169

ABSTRACT

Drug resistance of Klebsiella pneumoniae severely threatens human health. Overcoming the mechanisms of K. pneumoniae resistance to develop novel vaccines against drug-resistant K. pneumoniae is highly desired. Here, we report a technology platform that uses high pressure to drive drug-resistant K. pneumoniae to pass through a gap, inducing the formation of stable artificial bacterial biomimetic vesicles (BBVs). These BBVs had little to no bacterial intracellular protein or nucleic acid and had high yields. BBVs were efficiently taken up by dendritic cells to stimulate their maturation. BBVs as K. pneumoniae vaccines had the dual functions of inducing bacteria-specific humoral and cellular immune responses to increase animals' survival rate and reduce pulmonary inflammation and bacterial loads. We believe that BBVs are new-generation technology for bacterial vesicle preparation. Establishment of this BBV vaccine platform can maximally expand preparation technology for vaccines against drug-resistant K. pneumoniae.


Subject(s)
Bacterial Vaccines/therapeutic use , Biomimetic Materials/therapeutic use , Extracellular Vesicles/immunology , Klebsiella Infections/therapy , Klebsiella pneumoniae/immunology , Animals , Bacterial Vaccines/chemical synthesis , Bacterial Vaccines/immunology , Bacterial Vaccines/toxicity , Biomimetic Materials/chemical synthesis , Biomimetic Materials/toxicity , Cell Fractionation/methods , Drug Resistance, Multiple, Bacterial/drug effects , Female , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Klebsiella pneumoniae/chemistry , Mice, Inbred C57BL , Mice, Inbred ICR , Pressure
19.
Sci Rep ; 11(1): 11197, 2021 05 27.
Article in English | MEDLINE | ID: mdl-34045649

ABSTRACT

Mycobacteroides abscessus (Previously Mycobacterium abscessus) is an emerging microorganism of the newly defined genera Mycobacteroides that causes mainly skin and tissue diseases in humans. The recent availability of total 34 fully sequenced genomes of different strains belonging to this species has provided an opportunity to utilize this genomics data to gain novel insights and guide the development of specific antimicrobial therapies. In the present study, we collected collectively 34 complete genome sequences of M. abscessus from the NCBI GenBank database. Pangenome analysis was conducted on these genomes to understand the genetic diversity and to obtain proteins associated with its core genome. These core proteins were then subjected to various subtractive filters to identify potential antigenic targets that were subjected to multi-epitope vaccine design. Our analysis projected the open pangenome of M. abscessus containing 3443 core genes. After applying various stepwise filtration steps on the core proteins, a total of four potential antigenic targets were identified. Utilizing their constituent CD4 and CD8 T-cell epitopes, a multi-epitope based subunit vaccine was computationally designed. Sequence-based analysis as well as structural characterization revealed the immunological effectiveness of this designed vaccine. Further molecular docking, molecular dynamics simulation and binding free energy estimation with Toll-like receptor 2 indicated strong structural associations of the vaccine with the immune receptor. The promising results are encouraging and need to be validated by additional wet laboratory studies for confirmation.


Subject(s)
Bacterial Vaccines/therapeutic use , Mycobacterium Infections, Nontuberculous/prevention & control , Mycobacterium abscessus/immunology , Vaccines, Subunit/therapeutic use , Vaccinology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Genome, Bacterial , Humans , Mycobacterium abscessus/genetics
20.
Chest ; 160(4): 1282-1291, 2021 10.
Article in English | MEDLINE | ID: mdl-33852919

ABSTRACT

BACKGROUND: Mycobacterium w (Mw), an immunomodulator, has been shown to resolve early organ failure in severe sepsis. RESEARCH QUESTION: Does Mw improve survival in patients with severe presumed gram-negative sepsis? STUDY DESIGN AND METHODS: This was a randomized, double-blind, placebo-controlled, parallel-group study conducted in ICUs of five tertiary care centers in India. We included consecutive patients (age ≥ 18 years) with presumed gram-negative sepsis in the study within 48 h of the first organ dysfunction. Patients in the treatment arm received 0.3 mL/d of Mw intradermally for 3 consecutive days, whereas the control arm received matching placebo. The primary outcome was 28-day all-cause mortality. The secondary outcomes were ventilator-free days, days receiving vasopressor therapy, ICU and hospital length of stay, nosocomial infection rate, antibiotic use duration, and delta Sequential Organ Failure Assessment (SOFA) score. RESULTS: We included 202 patients with severe sepsis (101 Mw, 101 placebo). The use of Mw significantly reduced the mortality (9/101 vs 20/101; estimate difference, 0.11 [95% CI, 0.01-0.21]; P = .04). We found no difference in ventilator-free days, days receiving vasopressor drugs, ICU length of stay, and the hospital length of stay. The time to mortality (median, 13 days vs 8.5 days) was significantly longer in the Mw than in the placebo arm. The delta SOFA score, rate of nosocomial infections, and antibiotic use duration were similar in the two arms. We found Mw to reduce significantly the odds (OR, 0.37 [95% CI, 0.15-0.9]) of mortality after adjusting for culture-positive sepsis, baseline SOFA score, age, and sex. INTERPRETATION: The use of Mw was associated with a significant reduction in mortality in patients with severe presumed gram-negative sepsis. Further studies are required to confirm our findings. TRIAL REGISTRY: ClinicalTrials.gov; No.: NCT02330432; URL: www.clinicaltrials.gov.


Subject(s)
Bacterial Vaccines/therapeutic use , Gram-Negative Bacterial Infections/therapy , Immunomodulating Agents/therapeutic use , Mortality , Sepsis/therapy , Adult , Anti-Bacterial Agents/therapeutic use , Double-Blind Method , Female , Fluid Therapy , Gram-Negative Bacterial Infections/immunology , Humans , Immunomodulation , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-2/immunology , Interleukin-4/immunology , Interleukin-6/immunology , Male , Middle Aged , Mycobacterium , Organ Dysfunction Scores , Respiration, Artificial , Sepsis/immunology , Th1-Th2 Balance , Tumor Necrosis Factor-alpha/immunology , Vasoconstrictor Agents/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...