1
|
Ben Debba L, Derreumaux D, Lonnet G, Taddei L, Scherbakov M. Clinical and economic burden of otitis media in children under 5 years of age in the United States: A retrospective study. Hum Vaccin Immunother 2024; 20:2409510. [PMID: 39422213 PMCID: PMC11492675 DOI: 10.1080/21645515.2024.2409510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Reductions in all-cause otitis media (OM) following widespread pneumococcal conjugate vaccine use have plateaued. Granular burden of disease data are needed to guide evaluation and implementation of new measures targeting OM prevention. We conducted a retrospective study to assess the incidence and treatment costs of OM, tympanostomy tube placement (TTP), and hearing loss in children aged <5 years in the United States (US). OM episodes and TTP between 2016 and 2017 were identified in IBM MarketScan Commercial Claims and Encounters, Medicare Supplemental and Coordination of Benefits, and Multi-Medicaid databases using diagnosis codes (ICD-10). The incidence rate per 100,000 person-years (IR) of OM in <5-year-olds was 62,726 in Commercial/Medicare databases and 55,874 in Medicaid. IRs peaked at 9-<12 months (115,552 and 110,960, respectively). Approximately 5% and 4% of OM episodes in the respective databases had TTP (IR 3233 and 2404). Around 2% of children with OM had hearing loss (IR 1468 and 1109, respectively), of whom 41% had TTP. We estimated that there were 11.1 million OM episodes in 2020 costing USD 4.8 billion. 243,618 children were estimated to have OM with hearing loss at a direct total cost of USD 637 million, or 13% of the overall cost. The clinical and economic burden attributable to OM in the US was high in children aged <5 years during the study period. Novel approaches are needed to improve and broaden vaccine-induced protection against OM and its complications. The study results could guide policymakers considering age-specific interventions to reduce the OM burden.
Collapse
|
2
|
Breivik TJ, Gjermo P, Gundersen Y, Opstad PK, Murison R, Hugoson A, von Hörsten S, Fristad I. Microbiota-immune-brain interactions: A new vision in the understanding of periodontal health and disease. Periodontol 2000 2024; 96:20-41. [PMID: 39233381 PMCID: PMC11579829 DOI: 10.1111/prd.12610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
This review highlights the significance of interactions between the microbiota, immune system, nervous and hormonal systems, and the brain on periodontal health and disease. Microorganisms in the microbiota, immune cells, and neurons communicate via homeostatic nervous and hormonal systems, regulating vital body functions. By modulating pro-inflammatory and anti-inflammatory adaptive immune responses, these systems control the composition and number of microorganisms in the microbiota. The strength of these brain-controlled responses is genetically determined but is sensitive to early childhood stressors, which can permanently alter their responsiveness via epigenetic mechanisms, and to adult stressors, causing temporary changes. Clinical evidence and research with humans and animal models indicate that factors linked to severe periodontitis enhance the responsiveness of these homeostatic systems, leading to persistent hyperactivation. This weakens the immune defense against invasive symbiotic microorganisms (pathobionts) while strengthening the defense against non-invasive symbionts at the gingival margin. The result is an increased gingival tissue load of pathobionts, including Gram-negative bacteria, followed by an excessive innate immune response, which prevents infection but simultaneously destroys gingival and periodontal tissues. Thus, the balance between pro-inflammatory and anti-inflammatory adaptive immunity is crucial in controlling the microbiota, and the responsiveness of brain-controlled homeostatic systems determines periodontal health.
Collapse
Affiliation(s)
- Torbjørn Jarle Breivik
- Department of Periodontology, Faculty of Dentistry, Institute of Clinical OdontologyUniversity of OsloOsloNorway
- Division for ProtectionNorwegian Defence Research EstablishmentKjellerNorway
| | - Per Gjermo
- Department of Periodontology, Faculty of Dentistry, Institute of Clinical OdontologyUniversity of OsloOsloNorway
| | - Yngvar Gundersen
- Division for ProtectionNorwegian Defence Research EstablishmentKjellerNorway
| | - Per Kristian Opstad
- Division for ProtectionNorwegian Defence Research EstablishmentKjellerNorway
| | - Robert Murison
- Department of Biological and Medical Psychology, Faculty of PsychologyUniversity of BergenBergenNorway
| | - Anders Hugoson
- Department of Periodontology, Institute of OdontologyThe Sahlgrenska Academy at University of Gothenburg and School of Health and WelfareGothenburgSweden
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Hospital Erlangen, Preclinical Experimental CenterFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Inge Fristad
- Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenBergenNorway
| |
Collapse
|
3
|
Da Costa RM, Rooke JL, Wells TJ, Cunningham AF, Henderson IR. Type 5 secretion system antigens as vaccines against Gram-negative bacterial infections. NPJ Vaccines 2024; 9:159. [PMID: 39218947 PMCID: PMC11366766 DOI: 10.1038/s41541-024-00953-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Infections caused by Gram-negative bacteria are leading causes of mortality worldwide. Due to the rise in antibiotic resistant strains, there is a desperate need for alternative strategies to control infections caused by these organisms. One such approach is the prevention of infection through vaccination. While live attenuated and heat-killed bacterial vaccines are effective, they can lead to adverse reactions. Newer vaccine technologies focus on utilizing polysaccharide or protein subunits for safer and more targeted vaccination approaches. One promising avenue in this regard is the use of proteins released by the Type 5 secretion system (T5SS). This system is the most prevalent secretion system in Gram-negative bacteria. These proteins are compelling vaccine candidates due to their demonstrated protective role in current licensed vaccines. Notably, Pertactin, FHA, and NadA are integral components of licensed vaccines designed to prevent infections caused by Bordetella pertussis or Neisseria meningitidis. In this review, we delve into the significance of incorporating T5SS proteins into licensed vaccines, their contributions to virulence, conserved structural motifs, and the protective immune responses elicited by these proteins.
Collapse
Affiliation(s)
- Rochelle M Da Costa
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jessica L Rooke
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
4
|
Fantoni G, Boccadifuoco G, Verdirosa F, Molesti E, Manenti A, Montomoli E. Current challenges and improvements in assessing the immunogenicity of bacterial vaccines. Front Microbiol 2024; 15:1404637. [PMID: 39044946 PMCID: PMC11263209 DOI: 10.3389/fmicb.2024.1404637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
The increase in antimicrobial-resistant bacterial strains has highlighted the need for a new vaccine strategy. The primary goal of a candidate vaccine is to prevent disease, by inducing a persistent immunologic memory, through the activation of pathogen-specific immune response. Antibody titer is the main parameter used to assess the immunogenicity of bacterial vaccine candidates and it is the most widely used as a correlate of protection. On the other hand, the antibody titer alone cannot provide complete information on all the activity mediated by antibodies which can only be assessed by functional assays, like the serum bactericidal assay and the opsonophagocytosis assay. However, due to the involvement of many biological factors, these assays are difficult to standardize. Some improvements have been achieved in recent years, but further optimizations are needed to minimize inter- and intra-laboratories variability and to allow the applicability of these functional assays for the vaccine immunogenicity assessment on a larger scale.
Collapse
Affiliation(s)
- Giulia Fantoni
- VisMederi S.r.l., Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | | | | | | | - Emanuele Montomoli
- VisMederi S.r.l., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
5
|
Doherty TM, Di Pasquale A, Finnegan G, Lele J, Philip RK. Sustaining the momentum for adult vaccination post-COVID-19 to leverage the global uptake of life-course immunisation: A scoping review and call to action. Int J Infect Dis 2024; 142:106963. [PMID: 38354849 DOI: 10.1016/j.ijid.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/22/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024] Open
Abstract
OBJECTIVES The COVID-19 pandemic changed the adult vaccination landscape, possibly permanently. This review attempts to quantitate the magnitude of those changes. METHODS PubMed was searched for studies on adult / life-course vaccination between 1 January 2020 until 8 November 2022. RESULTS Twenty-one articles were identified and observations summarised as positive developments/impediments to life-course immunisation, and areas needing policy and structural reform. Unprecedented funding, international co-operation and technical advances led to COVID-19 vaccines authorised in record time. Investments in infrastructure and an expanded healthcare workforce streamlined vaccine delivery to adults. Constant media coverage and targeted messaging have improved health literacy. Conversely, the speed of vaccine development was perceived as a safety risk, and an 'infodemic' of misinformation propagated through social media negatively influenced vaccine uptake. Vaccine access and affordability remains inequitable among older adults and minority groups. CONCLUSIONS The COVID pandemic led to an opportunity to permanently change policies, attitudes, and systems for vaccine delivery to adults to establish a global life-course approach to immunisation. This is a call for action to sustain the momentum triggered by the COVID-19 pandemic. Addressing inequalities, improving health literacy and optimally using social media are critical to sustain adult vaccinations in post-COVID-19 era.
Collapse
Affiliation(s)
| | - Alberta Di Pasquale
- Takeda Pharmaceuticals International AG Singapore Branch, Singapore, Singapore
| | | | - Jayesh Lele
- Indian Medical Association, National Hospital Board of India, Mumbai, India
| | - Roy K Philip
- University Maternity Hospital Limerick, University of Limerick School of Medicine, Limerick, Ireland
| |
Collapse
|
6
|
Hibbert T, Krpetic Z, Latimer J, Leighton H, McHugh R, Pottenger S, Wragg C, James CE. Antimicrobials: An update on new strategies to diversify treatment for bacterial infections. Adv Microb Physiol 2024; 84:135-241. [PMID: 38821632 DOI: 10.1016/bs.ampbs.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Ninety-five years after Fleming's discovery of penicillin, a bounty of antibiotic compounds have been discovered, modified, or synthesised. Diversification of target sites, improved stability and altered activity spectra have enabled continued antibiotic efficacy, but overwhelming reliance and misuse has fuelled the global spread of antimicrobial resistance (AMR). An estimated 1.27 million deaths were attributable to antibiotic resistant bacteria in 2019, representing a major threat to modern medicine. Although antibiotics remain at the heart of strategies for treatment and control of bacterial diseases, the threat of AMR has reached catastrophic proportions urgently calling for fresh innovation. The last decade has been peppered with ground-breaking developments in genome sequencing, high throughput screening technologies and machine learning. These advances have opened new doors for bioprospecting for novel antimicrobials. They have also enabled more thorough exploration of complex and polymicrobial infections and interactions with the healthy microbiome. Using models of infection that more closely resemble the infection state in vivo, we are now beginning to measure the impacts of antimicrobial therapy on host/microbiota/pathogen interactions. However new approaches are needed for developing and standardising appropriate methods to measure efficacy of novel antimicrobial combinations in these contexts. A battery of promising new antimicrobials is now in various stages of development including co-administered inhibitors, phages, nanoparticles, immunotherapy, anti-biofilm and anti-virulence agents. These novel therapeutics need multidisciplinary collaboration and new ways of thinking to bring them into large scale clinical use.
Collapse
Affiliation(s)
- Tegan Hibbert
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Zeljka Krpetic
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Joe Latimer
- School of Science, Engineering, and Environment, University of Salford, Salford, UK
| | - Hollie Leighton
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Rebecca McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Sian Pottenger
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Charlotte Wragg
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool, UK
| | - Chloë E James
- School of Science, Engineering, and Environment, University of Salford, Salford, UK.
| |
Collapse
|
7
|
Naveed M, Waseem M, Mahkdoom I, Ali N, Asif F, Hassan JU, Jamil H. Transient comparison of techniques to counter multi-drug resistant bacteria: prime modules in curation of bacterial infections. FRONTIERS IN ANTIBIOTICS 2024; 2:1309107. [PMID: 39816650 PMCID: PMC11732137 DOI: 10.3389/frabi.2023.1309107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2025]
Abstract
Multidrug-resistant organisms are bacteria that are no longer controlled or killed by specific drugs. One of two methods causes bacteria multidrug resistance (MDR); first, these bacteria may disguise multiple cell genes coding for drug resistance to a single treatment on resistance (R) plasmids. Second, increased expression of genes coding for multidrug efflux pumps, which extrude many drugs, can cause MDR. Antibiotic resistance is a big issue since some bacteria may withstand almost all antibiotics. These bacteria can cause serious sickness, making them a public health threat. Methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus (VRE), Multidrug resistant Mycobacterium tuberculosis (TB), and CRE are gut bacteria that resist antibiotics. Antimicrobial resistance is rising worldwide, increasing clinical and community morbidity and mortality. Superbugs have made antibiotic resistance in some environmental niches even harder to control. This study introduces new medicinal plants, gene-editing methods, nanomaterials, and bacterial vaccines that will fight MDR bacteria in the future.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Muhammad Waseem
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Izma Mahkdoom
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Nouman Ali
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Farrukh Asif
- National Institute of Biotechnology and Genetic Engineering, Faisalabad, Punjab, Pakistan
| | - Jawad ul Hassan
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Hamza Jamil
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
8
|
Mayer RL, Mechtler K. Immunopeptidomics in the Era of Single-Cell Proteomics. BIOLOGY 2023; 12:1514. [PMID: 38132340 PMCID: PMC10740491 DOI: 10.3390/biology12121514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
Immunopeptidomics, as the analysis of antigen peptides being presented to the immune system via major histocompatibility complexes (MHC), is being seen as an imperative tool for identifying epitopes for vaccine development to treat cancer and viral and bacterial infections as well as parasites. The field has made tremendous strides over the last 25 years but currently still faces challenges in sensitivity and throughput for widespread applications in personalized medicine and large vaccine development studies. Cutting-edge technological advancements in sample preparation, liquid chromatography as well as mass spectrometry, and data analysis, however, are currently transforming the field. This perspective showcases how the advent of single-cell proteomics has accelerated this transformation of immunopeptidomics in recent years and will pave the way for even more sensitive and higher-throughput immunopeptidomics analyses.
Collapse
Affiliation(s)
- Rupert L. Mayer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, 1030 Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, 1030 Vienna, Austria
- Gregor Mendel Institute of Molecular Plant Biology (GMI), Austrian Academy of Sciences, Vienna BioCenter (VBC), 1030 Vienna, Austria
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter (VBC), 1030 Vienna, Austria
| |
Collapse
|
9
|
Chiu FF, Tu LL, Chen W, Zhou H, Liu BS, Liu SJ, Leng CH. A broad-spectrum pneumococcal vaccine induces mucosal immunity and protects against lethal Streptococcus pneumoniae challenge. Emerg Microbes Infect 2023; 12:2272656. [PMID: 37855122 PMCID: PMC10606790 DOI: 10.1080/22221751.2023.2272656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Pneumococcal disease is a major threat to public health globally, impacting individuals across all age groups, particularly infants and elderly individuals. The use of current vaccines has led to unintended consequences, including serotype replacement, leading to a need for a new approach to combat pneumococcal disease. A promising solution is the development of a broad-spectrum pneumococcal vaccine. In this study, we present the development of a broad-spectrum protein-based pneumococcal vaccine that contains three pneumococcal virulence factors: rlipo-PsaA (lipidated form), rPspAΔC (truncated form), and rPspCΔC (truncated form). Intranasal immunization with rlipo-PsaA, rPspAΔC, and rPspCΔC (LAAC) resulted in significantly higher IgG titres than those induced by administration of nonlipidated rPsaA, rPspAΔC, and rPspCΔC (AAC). Furthermore, LAAC immunization induced the production of higher IgA titres in vaginal washes, feces, and sera in mice, indicating that LAAC can induce systemic mucosal immunity. In addition, administration of LAAC also induced Th1/Th17-biased immune responses and promoted opsonic phagocytosis of Streptococcus pneumoniae strains of various serotypes, implying that the immunogenicity of LAAC immunization provides a protective effect against pneumococcal infection. Importantly, challenge data showed that the LAAC-immunized mice had a reduced bacterial load not only for several serotypes of the 13-valent conjugate pneumococcal vaccine (PCV13) but also for selected non-PCV13 serotypes. Consistently, LAAC immunization increased the survival rate of mice after bacterial challenge with both PCV13 and non-PCV13 serotypes. In conclusion, our protein-based pneumococcal vaccine provides protective effects against a broad spectrum of Streptococcus pneumoniae serotypes.
Collapse
Affiliation(s)
- Fang-Feng Chiu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ling Tu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Wangxue Chen
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Canada
| | - Hongyan Zhou
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Canada
| | - Bing-Sin Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hsiang Leng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
10
|
Martin Aispuro P, Bottero D, Zurita ME, Gaillard ME, Hozbor DF. Impact of maternal whole-cell or acellular pertussis primary immunization on neonatal immune response. Front Immunol 2023; 14:1192119. [PMID: 37435078 PMCID: PMC10330814 DOI: 10.3389/fimmu.2023.1192119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
With the introduction of pertussis immunization for pregnant women in many countries, there has been renewed interest in the impact of whole-cell pertussis vaccine (wP) versus acellular vaccine (aP) on disease control, particularly regarding the best approach for priming. To gather evidence on this topic, we analyzed the impact of aP or wP priming on aP vaccination during pregnancy (aPpreg) in mice. Two-mother vaccination schemes were employed (wP-wP-aPpreg and aP-aP-aPpreg), and the immune response in the mothers and their offspring, as well as the protection of the offspring against Bordetella pertussis challenge, were assessed. Pertussis toxin (PTx)-specific IgG responses were detected in mothers after both the second and third doses, with higher titers after the third dose, regardless of the vaccination schedule. However, a significant reduction in PTx-IgG levels was observed after 22 weeks post aPpreg immunization in mothers with the aP-aP-aPpreg scheme but not in the wP-wP-aPpreg immunized mothers. The aP-aP-aPpreg schedule triggered a murine antibody response mainly to a Th2-profile, while wP-wP-aPpreg induced a Th1/Th2 mixed profile. Both immunization schemes administered to the mothers protected the offspring against pertussis, but the wP-wP-aPpreg vaccination conferred offspring protection in all pregnancies at least up to 20 weeks after receiving the aPpreg-dose. In contrast, the immunity induced by aP-aP-aPpreg began to decline in births that occurred 18 weeks after receiving the aPpreg dose. For the aP-aP-aPpreg scheme, pups born from gestations furthest from aPpreg (+22 weeks) had lower PTx-specific IgG levels than those born closer to the application of the dose during pregnancy. In contrast, for pups born to wP-wP-aPpreg vaccinated mothers, the PTx-specific IgG levels were maintained over time, even for those born at the longest time studied (+22 weeks). It is noteworthy that only the pups born from mothers with aP-aP-aPpreg and receiving a neonatal dose of either aP or wP were more susceptible to B. pertussis infection than mice with only maternal immunity, suggesting interference with the induced immunity (p<0.05). However, it should be noted that mice with maternal immunity, whether vaccinated or not with neonatal doses, are better protected against colonization with B. pertussis than mice without maternal immunity but vaccinated with aP or wP.
Collapse
|
11
|
Chen X, Gula H, Pius T, Ou C, Gomozkova M, Wang LX, Schneewind O, Missiakas D. Immunoglobulin G subclasses confer protection against Staphylococcus aureus bloodstream dissemination through distinct mechanisms in mouse models. Proc Natl Acad Sci U S A 2023; 120:e2220765120. [PMID: 36972444 PMCID: PMC10083571 DOI: 10.1073/pnas.2220765120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/03/2023] [Indexed: 03/29/2023] Open
Abstract
Antibodies bind target molecules with exquisite specificity. The removal of these targets is mediated by the effector functions of antibodies. We reported earlier that the monoclonal antibody (mAb) 3F6 promotes opsonophagocytic killing of Staphylococcus aureus in blood and reduces bacterial replication in animals. Here, we generated mouse immunoglobulin G (mIgG) subclass variants and observed a hierarchy in protective efficacy 3F6-mIgG2a > 3F6-mIgG1 ≥ 3F6-mIgG2b >> 3F6-mIgG3 following bloodstream challenge of C57BL/6J mice. This hierarchy was not observed in BALB/cJ mice: All IgG subclasses conferred similar protection. IgG subclasses differ in their ability to activate complement and interact with Fcγ receptors (FcγR) on immune cells. 3F6-mIgG2a-dependent protection was lost in FcγR-deficient, but not in complement-deficient C57BL/6J animals. Measurements of the relative ratio of FcγRIV over complement receptor 3 (CR3) on neutrophils suggest the preferential expression of FcγRIV in C57BL/6 mice and of CR3 in BALB/cJ mice. To determine the physiological significance of these differing ratios, blocking antibodies against FcγRIV or CR3 were administered to animals before challenge. Correlating with the relative abundance of each receptor, 3F6-mIgG2a-dependent protection in C57BL/6J mice showed a greater reliance for FcγRIV while protection in BALB/cJ mice was only impaired upon neutralization of CR3. Thus, 3F6-based clearance of S. aureus in mice relies on a strain-specific contribution of variable FcγR- and complement-dependent pathways. We surmise that these variabilities are the result of genetic polymorphism(s) that may be encountered in other mammals including humans and may have clinical implications in predicting the efficacy of mAb-based therapies.
Collapse
Affiliation(s)
- Xinhai Chen
- Department of Microbiology, Howard Taylor Ricketts Laboratory, The University of Chicago, Lemont, IL60439
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen518132, China
| | - Haley Gula
- Department of Microbiology, Howard Taylor Ricketts Laboratory, The University of Chicago, Lemont, IL60439
| | - Tonu Pius
- Department of Microbiology, Howard Taylor Ricketts Laboratory, The University of Chicago, Lemont, IL60439
| | - Chong Ou
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD20742
| | - Margaryta Gomozkova
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD20742
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD20742
| | - Olaf Schneewind
- Department of Microbiology, Howard Taylor Ricketts Laboratory, The University of Chicago, Lemont, IL60439
| | - Dominique Missiakas
- Department of Microbiology, Howard Taylor Ricketts Laboratory, The University of Chicago, Lemont, IL60439
| |
Collapse
|
12
|
Islam MS, Rahman MT. A Comprehensive Review on Bacterial Vaccines Combating Antimicrobial Resistance in Poultry. Vaccines (Basel) 2023; 11:vaccines11030616. [PMID: 36992200 DOI: 10.3390/vaccines11030616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Bacterial vaccines have become a crucial tool in combating antimicrobial resistance (AMR) in poultry. The overuse and misuse of antibiotics in poultry farming have led to the development of AMR, which is a growing public health concern. Bacterial vaccines are alternative methods for controlling bacterial diseases in poultry, reducing the need for antibiotics and improving animal welfare. These vaccines come in different forms including live attenuated, killed, and recombinant vaccines, and they work by stimulating the immune system to produce a specific response to the target bacteria. There are many advantages to using bacterial vaccines in poultry, including reduced use of antibiotics, improved animal welfare, and increased profitability. However, there are also limitations such as vaccine efficacy and availability. The use of bacterial vaccines in poultry is regulated by various governmental bodies and there are economic considerations to be taken into account, including costs and return on investment. The future prospects for bacterial vaccines in poultry are promising, with advancements in genetic engineering and vaccine formulation, and they have the potential to improve the sustainability of the poultry industry. In conclusion, bacterial vaccines are essential in combating AMR in poultry and represent a crucial step towards a more sustainable and responsible approach to poultry farming.
Collapse
Affiliation(s)
- Md Saiful Islam
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Md Tanvir Rahman
- Department of Microbiology and Hygiene, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| |
Collapse
|
13
|
Helmy YA, Taha-Abdelaziz K, Hawwas HAEH, Ghosh S, AlKafaas SS, Moawad MMM, Saied EM, Kassem II, Mawad AMM. Antimicrobial Resistance and Recent Alternatives to Antibiotics for the Control of Bacterial Pathogens with an Emphasis on Foodborne Pathogens. Antibiotics (Basel) 2023; 12:274. [PMID: 36830185 PMCID: PMC9952301 DOI: 10.3390/antibiotics12020274] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Antimicrobial resistance (AMR) is one of the most important global public health problems. The imprudent use of antibiotics in humans and animals has resulted in the emergence of antibiotic-resistant bacteria. The dissemination of these strains and their resistant determinants could endanger antibiotic efficacy. Therefore, there is an urgent need to identify and develop novel strategies to combat antibiotic resistance. This review provides insights into the evolution and the mechanisms of AMR. Additionally, it discusses alternative approaches that might be used to control AMR, including probiotics, prebiotics, antimicrobial peptides, small molecules, organic acids, essential oils, bacteriophage, fecal transplants, and nanoparticles.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Department of Veterinary Science, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Khaled Taha-Abdelaziz
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - Hanan Abd El-Halim Hawwas
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Samar Sami AlKafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31511, Egypt
| | | | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Issmat I. Kassem
- Centre for Food Safety, Department of Food Science and Technology, University of Georgia, Griffin, GA 30609, USA
| | - Asmaa M. M. Mawad
- Department of Biology, College of Science, Taibah University, Madinah 42317, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
14
|
Deng JZ, Kuster N, Drumheller A, Lin M, Ansbro F, Grozdanovic M, Samuel R, Zhuang P. Antibody enhanced HPLC for serotype-specific quantitation of polysaccharides in pneumococcal conjugate vaccine. NPJ Vaccines 2023; 8:2. [PMID: 36690697 PMCID: PMC9869843 DOI: 10.1038/s41541-022-00584-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 11/25/2022] [Indexed: 01/24/2023] Open
Abstract
Bacterial infection remains as one of the major healthcare issues, despite significant scientific and medical progress in this field. Infection by Streptococcus Pneumoniae (S. Pneumoniae) can cause pneumonia and other serious infectious diseases, such as bacteremia, sinusitis and meningitis. The pneumococcal capsular polysaccharides (CPS) that constitute the outermost layer of the bacterial cell are the main immunogens and protect the pathogen from host defense mechanisms. Over 90 pneumococcal CPS serotypes have been identified, among which more than 30 can cause invasive pneumococcal diseases that could lead to morbidity and mortality. Multivalent pneumococcal vaccines have been developed to prevent diseases caused by S. Pneumoniae. These vaccines employ either purified pneumococcal CPSs or protein conjugates of these CPSs to generate antigen-specific immune responses for patient protection. Serotype-specific quantitation of these polysaccharides (Ps) antigen species are required for vaccine clinical dosage, product release and quality control. Herein, we have developed an antibody-enhanced high-performance liquid chromatography (HPLC) assay for serotype-specific quantitation of the polysaccharide contents in multivalent pneumococcal conjugate vaccines (PCVs). A fluorescence-labeled multiplex assay format has also been developed. This work laid the foundation for a serotype-specific antigen assay format that could play an important role for future vaccine research and development.
Collapse
Affiliation(s)
- James Z. Deng
- grid.417993.10000 0001 2260 0793Vaccine Analytical Research & Development, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Nathan Kuster
- grid.417993.10000 0001 2260 0793Vaccine Analytical Research & Development, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Ashley Drumheller
- grid.417993.10000 0001 2260 0793Vaccine Analytical Research & Development, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Mingxiang Lin
- grid.417993.10000 0001 2260 0793Analytical External Capabilities, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Frances Ansbro
- grid.417993.10000 0001 2260 0793Cell-Based Sciences, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Milica Grozdanovic
- grid.417993.10000 0001 2260 0793Cell-Based Sciences, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Rachelle Samuel
- grid.417993.10000 0001 2260 0793Cell-Based Sciences, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| | - Ping Zhuang
- grid.417993.10000 0001 2260 0793Vaccine Analytical Research & Development, Analytical Research & Development, MRL, Merck & Co., Inc., Rahway, NJ USA
| |
Collapse
|
15
|
Koizumi R, Kusama Y, Asai Y, Tsuzuki S, Aoyagi K, Ishikane M, Muraki Y, Ohmagari N. Effects of population age structure on parenteral antimicrobial use estimations. Sci Rep 2023; 13:840. [PMID: 36646845 PMCID: PMC9841941 DOI: 10.1038/s41598-023-27769-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Antimicrobial use (AMU) is conventionally reported as unadjusted defined daily doses (DDDs) or population-adjusted DDDs per 1000 inhabitants per day (DID). DID is frequently used to monitor national AMU trends, this metric does not intrinsically take temporal changes in population age structure into account. We examined the effects of population age structure on DID estimates of parenteral AMU in Japan, and predicted future trends in DDDs based on population projections. Parenteral AMU data from 2013 to 2018 were acquired from a national claims database. We assessed temporal trends in parenteral AMU by age group (children aged < 15 years, working-age persons aged 15-64 years, and older persons aged ≥ 65 years) using both DID and DDDs. In addition, we modeled DDD predictions based on age-specific population projections from 2019 to 2030. DID values for older persons were 8.08-10.15 times and 5.43-5.63 times higher than in children and working-age persons, respectively. DID was stable, but DDDs increased in older persons. The prediction models showed that DDDs will continue to increase until 2030 if DID remains steady or decreases. DID estimates were skewed by the older population. More rigorous antimicrobial stewardship efforts targeting geriatric care are needed to counter the aging-associated increase in AMU.
Collapse
Affiliation(s)
- Ryuji Koizumi
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Yoshiki Kusama
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan. .,Department of Pediatric Infectious Diseases, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo, Japan.
| | - Yusuke Asai
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Shinya Tsuzuki
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kensuke Aoyagi
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Masahiro Ishikane
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Yuichi Muraki
- Department of Clinical Pharmacoepidemiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Norio Ohmagari
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| |
Collapse
|
16
|
Laupèze B, Doherty TM. Maintaining a 'fit' immune system: the role of vaccines. Expert Rev Vaccines 2023; 22:256-266. [PMID: 36864769 DOI: 10.1080/14760584.2023.2185223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
INTRODUCTION Conventionally, vaccines are thought to induce a specific immune response directed against a target pathogen. Long recognized but poorly understood nonspecific benefits of vaccination, such as reduced susceptibility to unrelated diseases or cancer, are now being investigated and may be due in part to "trained immunity'. AREAS COVERED We discuss 'trained immunity' and whether vaccine-induced 'trained immunity' could be leveraged to prevent morbidity due to a broader range of causes. EXPERT OPINION The prevention of infection i.e. maintaining homeostasis by preventing the primary infection and resulting secondary illnesses, is the pivotal strategy used to direct vaccine design and may have long-term, positive impacts on health at all ages. In the future, we anticipate that vaccine design will change to not only prevent the target infection (or related infections) but to generate positive modifications to the immune response that could prevent a wider range of infections and potentially reduce the impact of immunological changes associated with aging. Despite changing demographics, adult vaccination has not always been prioritized. However, the SARS-CoV-2 pandemic has demonstrated that adult vaccination can flourish given the right circumstances, demonstrating that harnessing the potential benefits of life-course vaccination is achievable for all.
Collapse
|
17
|
Ikuta KS, Swetschinski LR, Robles Aguilar G, Sharara F, Mestrovic T, Gray AP, Davis Weaver N, Wool EE, Han C, Gershberg Hayoon A, Aali A, Abate SM, Abbasi-Kangevari M, Abbasi-Kangevari Z, Abd-Elsalam S, Abebe G, Abedi A, Abhari AP, Abidi H, Aboagye RG, Absalan A, Abubaker Ali H, Acuna JM, Adane TD, Addo IY, Adegboye OA, Adnan M, Adnani QES, Afzal MS, Afzal S, Aghdam ZB, Ahinkorah BO, Ahmad A, Ahmad AR, Ahmad R, Ahmad S, Ahmad S, Ahmadi S, Ahmed A, Ahmed H, Ahmed JQ, Ahmed Rashid T, Ajami M, Aji B, Akbarzadeh-Khiavi M, Akunna CJ, Al Hamad H, Alahdab F, Al-Aly Z, Aldeyab MA, Aleman AV, Alhalaiqa FAN, Alhassan RK, Ali BA, Ali L, Ali SS, Alimohamadi Y, Alipour V, Alizadeh A, Aljunid SM, Allel K, Almustanyir S, Ameyaw EK, Amit AML, Anandavelane N, Ancuceanu R, Andrei CL, Andrei T, Anggraini D, Ansar A, Anyasodor AE, Arabloo J, Aravkin AY, Areda D, Aripov T, Artamonov AA, Arulappan J, Aruleba RT, Asaduzzaman M, Ashraf T, Athari SS, Atlaw D, Attia S, Ausloos M, Awoke T, Ayala Quintanilla BP, Ayana TM, Azadnajafabad S, Azari Jafari A, B DB, Badar M, Badiye AD, Baghcheghi N, Bagherieh S, Baig AA, Banerjee I, Barac A, Bardhan M, Barone-Adesi F, Barqawi HJ, Barrow A, Baskaran P, Basu S, Batiha AMM, Bedi N, Belete MA, Belgaumi UI, Bender RG, Bhandari B, Bhandari D, Bhardwaj P, Bhaskar S, Bhattacharyya K, Bhattarai S, Bitaraf S, Buonsenso D, Butt ZA, Caetano dos Santos FL, Cai J, Calina D, Camargos P, Cámera LA, Cárdenas R, Cevik M, Chadwick J, Charan J, Chaurasia A, Ching PR, Choudhari SG, Chowdhury EK, Chowdhury FR, Chu DT, Chukwu IS, Dadras O, Dagnaw FT, Dai X, Das S, Dastiridou A, Debela SA, Demisse FW, Demissie S, Dereje D, Derese M, Desai HD, Dessalegn FN, Dessalegni SAA, Desye B, Dhaduk K, Dhimal M, Dhingra S, Diao N, Diaz D, Djalalinia S, Dodangeh M, Dongarwar D, Dora BT, Dorostkar F, Dsouza HL, Dubljanin E, Dunachie SJ, Durojaiye OC, Edinur HA, Ejigu HB, Ekholuenetale M, Ekundayo TC, El-Abid H, Elhadi M, Elmonem MA, Emami A, Engelbert Bain L, Enyew DB, Erkhembayar R, Eshrati B, Etaee F, Fagbamigbe AF, Falahi S, Fallahzadeh A, Faraon EJA, Fatehizadeh A, Fekadu G, Fernandes JC, Ferrari A, Fetensa G, Filip I, Fischer F, Foroutan M, Gaal PA, Gadanya MA, Gaidhane AM, Ganesan B, Gebrehiwot M, Ghanbari R, Ghasemi Nour M, Ghashghaee A, Gholamrezanezhad A, Gholizadeh A, Golechha M, Goleij P, Golinelli D, Goodridge A, Gunawardane DA, Guo Y, Gupta RD, Gupta S, Gupta VB, Gupta VK, Guta A, Habibzadeh P, Haddadi Avval A, Halwani R, Hanif A, Hannan MA, Harapan H, Hassan S, Hassankhani H, Hayat K, Heibati B, Heidari G, Heidari M, Heidari-Soureshjani R, Herteliu C, Heyi DZ, Hezam K, Hoogar P, Horita N, Hossain MM, Hosseinzadeh M, Hostiuc M, Hostiuc S, Hoveidamanesh S, Huang J, Hussain S, Hussein NR, Ibitoye SE, Ilesanmi OS, Ilic IM, Ilic MD, Imam MT, Immurana M, Inbaraj LR, Iradukunda A, Ismail NE, Iwu CCD, Iwu CJ, J LM, Jakovljevic M, Jamshidi E, Javaheri T, Javanmardi F, Javidnia J, Jayapal SK, Jayarajah U, Jebai R, Jha RP, Joo T, Joseph N, Joukar F, Jozwiak JJ, Kacimi SEO, Kadashetti V, Kalankesh LR, Kalhor R, Kamal VK, Kandel H, Kapoor N, Karkhah S, Kassa BG, Kassebaum NJ, Katoto PDMC, Keykhaei M, Khajuria H, Khan A, Khan IA, Khan M, Khan MN, Khan MAB, Khatatbeh MM, Khater MM, Khayat Kashani HR, Khubchandani J, Kim H, Kim MS, Kimokoti RW, Kissoon N, Kochhar S, Kompani F, Kosen S, Koul PA, Koulmane Laxminarayana SL, Krapp Lopez F, Krishan K, Krishnamoorthy V, Kulkarni V, Kumar N, Kurmi OP, Kuttikkattu A, Kyu HH, Lal DK, Lám J, Landires I, Lasrado S, Lee SW, Lenzi J, Lewycka S, Li S, Lim SS, Liu W, Lodha R, Loftus MJ, Lohiya A, Lorenzovici L, Lotfi M, Mahmoodpoor A, Mahmoud MA, Mahmoudi R, Majeed A, Majidpoor J, Makki A, Mamo GA, Manla Y, Martorell M, Matei CN, McManigal B, Mehrabi Nasab E, Mehrotra R, Melese A, Mendoza-Cano O, Menezes RG, Mentis AFA, Micha G, Michalek IM, Micheletti Gomide Nogueira de Sá AC, Milevska Kostova N, Mir SA, Mirghafourvand M, Mirmoeeni S, Mirrakhimov EM, Mirza-Aghazadeh-Attari M, Misganaw AS, Misganaw A, Misra S, Mohammadi E, Mohammadi M, Mohammadian-Hafshejani A, Mohammed S, Mohan S, Mohseni M, Mokdad AH, Momtazmanesh S, Monasta L, Moore CE, Moradi M, Moradi Sarabi M, Morrison SD, Motaghinejad M, Mousavi Isfahani H, Mousavi Khaneghah A, Mousavi-Aghdas SA, Mubarik S, Mulita F, Mulu GBB, Munro SB, Muthupandian S, Nair TS, Naqvi AA, Narang H, Natto ZS, Naveed M, Nayak BP, Naz S, Negoi I, Nejadghaderi SA, Neupane Kandel S, Ngwa CH, Niazi RK, Nogueira de Sá AT, Noroozi N, Nouraei H, Nowroozi A, Nuñez-Samudio V, Nutor JJ, Nzoputam CI, Nzoputam OJ, Oancea B, Obaidur RM, Ojha VA, Okekunle AP, Okonji OC, Olagunju AT, Olusanya BO, Omar Bali A, Omer E, Otstavnov N, Oumer B, P A M, Padubidri JR, Pakshir K, Palicz T, Pana A, Pardhan S, Paredes JL, Parekh U, Park EC, Park S, Pathak A, Paudel R, Paudel U, Pawar S, Pazoki Toroudi H, Peng M, Pensato U, Pepito VCF, Pereira M, Peres MFP, Perico N, Petcu IR, Piracha ZZ, Podder I, Pokhrel N, Poluru R, Postma MJ, Pourtaheri N, Prashant A, Qattea I, Rabiee M, Rabiee N, Radfar A, Raeghi S, Rafiei S, Raghav PR, Rahbarnia L, Rahimi-Movaghar V, Rahman M, Rahman MA, Rahmani AM, Rahmanian V, Ram P, Ranjha MMAN, Rao SJ, Rashidi MM, Rasul A, Ratan ZA, Rawaf S, Rawassizadeh R, Razeghinia MS, Redwan EMM, Regasa MT, Remuzzi G, Reta MA, Rezaei N, Rezapour A, Riad A, Ripon RK, Rudd KE, Saddik B, Sadeghian S, Saeed U, Safaei M, Safary A, Safi SZ, Sahebazzamani M, Sahebkar A, Sahoo H, Salahi S, Salahi S, Salari H, Salehi S, Samadi Kafil H, Samy AM, Sanadgol N, Sankararaman S, Sanmarchi F, Sathian B, Sawhney M, Saya GK, Senthilkumaran S, Seylani A, Shah PA, Shaikh MA, Shaker E, Shakhmardanov MZ, Sharew MM, Sharifi-Razavi A, Sharma P, Sheikhi RA, Sheikhy A, Shetty PH, Shigematsu M, Shin JI, Shirzad-Aski H, Shivakumar KM, Shobeiri P, Shorofi SA, Shrestha S, Sibhat MM, Sidemo NB, Sikder MK, Silva LMLR, Singh JA, Singh P, Singh S, Siraj MS, Siwal SS, Skryabin VY, Skryabina AA, Socea B, Solomon DD, Song Y, Sreeramareddy CT, Suleman M, Suliankatchi Abdulkader R, Sultana S, Szócska M, Tabatabaeizadeh SA, Tabish M, Taheri M, Taki E, Tan KK, Tandukar S, Tat NY, Tat VY, Tefera BN, Tefera YM, Temesgen G, Temsah MH, Tharwat S, Thiyagarajan A, Tleyjeh II, Troeger CE, Umapathi KK, Upadhyay E, Valadan Tahbaz S, Valdez PR, Van den Eynde J, van Doorn HR, Vaziri S, Verras GI, Viswanathan H, Vo B, Waris A, Wassie GT, Wickramasinghe ND, Yaghoubi S, Yahya GATY, Yahyazadeh Jabbari SH, Yigit A, Yiğit V, Yon DK, Yonemoto N, Zahir M, Zaman BA, Zaman SB, Zangiabadian M, Zare I, Zastrozhin MS, Zhang ZJ, Zheng P, Zhong C, Zoladl M, Zumla A, Hay SI, Dolecek C, Sartorius B, Murray CJL, Naghavi M. Global mortality associated with 33 bacterial pathogens in 2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 2022; 400:2221-2248. [PMID: 36423648 PMCID: PMC9763654 DOI: 10.1016/s0140-6736(22)02185-7] [Citation(s) in RCA: 590] [Impact Index Per Article: 196.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/13/2022] [Accepted: 11/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Reducing the burden of death due to infection is an urgent global public health priority. Previous studies have estimated the number of deaths associated with drug-resistant infections and sepsis and found that infections remain a leading cause of death globally. Understanding the global burden of common bacterial pathogens (both susceptible and resistant to antimicrobials) is essential to identify the greatest threats to public health. To our knowledge, this is the first study to present global comprehensive estimates of deaths associated with 33 bacterial pathogens across 11 major infectious syndromes. METHODS We estimated deaths associated with 33 bacterial genera or species across 11 infectious syndromes in 2019 using methods from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2019, in addition to a subset of the input data described in the Global Burden of Antimicrobial Resistance 2019 study. This study included 343 million individual records or isolates covering 11 361 study-location-years. We used three modelling steps to estimate the number of deaths associated with each pathogen: deaths in which infection had a role, the fraction of deaths due to infection that are attributable to a given infectious syndrome, and the fraction of deaths due to an infectious syndrome that are attributable to a given pathogen. Estimates were produced for all ages and for males and females across 204 countries and territories in 2019. 95% uncertainty intervals (UIs) were calculated for final estimates of deaths and infections associated with the 33 bacterial pathogens following standard GBD methods by taking the 2·5th and 97·5th percentiles across 1000 posterior draws for each quantity of interest. FINDINGS From an estimated 13·7 million (95% UI 10·9-17·1) infection-related deaths in 2019, there were 7·7 million deaths (5·7-10·2) associated with the 33 bacterial pathogens (both resistant and susceptible to antimicrobials) across the 11 infectious syndromes estimated in this study. We estimated deaths associated with the 33 bacterial pathogens to comprise 13·6% (10·2-18·1) of all global deaths and 56·2% (52·1-60·1) of all sepsis-related deaths in 2019. Five leading pathogens-Staphylococcus aureus, Escherichia coli, Streptococcus pneumoniae, Klebsiella pneumoniae, and Pseudomonas aeruginosa-were responsible for 54·9% (52·9-56·9) of deaths among the investigated bacteria. The deadliest infectious syndromes and pathogens varied by location and age. The age-standardised mortality rate associated with these bacterial pathogens was highest in the sub-Saharan Africa super-region, with 230 deaths (185-285) per 100 000 population, and lowest in the high-income super-region, with 52·2 deaths (37·4-71·5) per 100 000 population. S aureus was the leading bacterial cause of death in 135 countries and was also associated with the most deaths in individuals older than 15 years, globally. Among children younger than 5 years, S pneumoniae was the pathogen associated with the most deaths. In 2019, more than 6 million deaths occurred as a result of three bacterial infectious syndromes, with lower respiratory infections and bloodstream infections each causing more than 2 million deaths and peritoneal and intra-abdominal infections causing more than 1 million deaths. INTERPRETATION The 33 bacterial pathogens that we investigated in this study are a substantial source of health loss globally, with considerable variation in their distribution across infectious syndromes and locations. Compared with GBD Level 3 underlying causes of death, deaths associated with these bacteria would rank as the second leading cause of death globally in 2019; hence, they should be considered an urgent priority for intervention within the global health community. Strategies to address the burden of bacterial infections include infection prevention, optimised use of antibiotics, improved capacity for microbiological analysis, vaccine development, and improved and more pervasive use of available vaccines. These estimates can be used to help set priorities for vaccine need, demand, and development. FUNDING Bill & Melinda Gates Foundation, Wellcome Trust, and Department of Health and Social Care, using UK aid funding managed by the Fleming Fund.
Collapse
|
18
|
Santos KR, Souza FN, Ramos-Sanchez EM, Batista CF, Reis LC, Fotoran WL, Heinemann MB, Cunha AF, Rocha MC, Faria AR, Andrade HM, Cerqueira MMOP, Gidlund M, Goto H, Della Libera AMMP. Staphylococcus aureus-Cure-Associated Antigens Elicit Type 3 Immune Memory T Cells. Antibiotics (Basel) 2022; 11:1831. [PMID: 36551488 PMCID: PMC9774748 DOI: 10.3390/antibiotics11121831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is one of the most frequently major mastitis pathogens that cause clinical and subclinical mastitis worldwide. Current antimicrobial treatments are usually ineffective, and the commercially available vaccines lack proven effectiveness. The immunological response elicited by the recombinant S. aureus-cure-associated proteins phosphoglycerate kinase (PGK), enolase (ENO), and elongation factor-G (EF-G) in combination with the granulocyte-macrophage colony-stimulating factor (GM-CSF) DNA vaccination was studied in this work. METHODS Here, twenty-three C57BL/6 mice were divided into four groups and vaccinated with: G1: none (control); G2: GM-CSF DNA plasmid DNA vaccine; G3: the combination of EF-G+ENO+PGK; and G4: the combinations of EF-G+ENO+PGK proteins plus GM-CSF plasmid DNA vaccine. After 44 days, spleen cells were collected for immunophenotyping and lymphocyte proliferation evaluation by flow cytometry upon S. aureus stimulus. RESULTS Immunization with the three S. aureus recombinant proteins alone resulted in a higher percentage of IL-17A+ cells among CD8+ T central memory cells, as well as the highest intensity of IL-17A production by overall lymphocytes indicating that the contribution of the combined lymphocyte populations is crucial to sustaining a type 3 cell immunity environment. CONCLUSION The immunization with three S. aureus-cure-associated recombinant proteins triggered type 3 immunity, which is a highly interesting path to pursue an effective bovine S. aureus mastitis vaccine.
Collapse
Affiliation(s)
- Kamila R. Santos
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - Fernando N. Souza
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
- Programa de Pós-Graduação em Ciência Animal, Universidade Federal da Paraíba, Areia 58397-000, Brazil
| | - Eduardo M. Ramos-Sanchez
- Programa de Pós-Graduação em Ciência Animal, Universidade Federal da Paraíba, Areia 58397-000, Brazil
- Laboratório de Soroloepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
- Departamento de Salud Publica, Facultad de Ciencias de La Salud, Universidad Nacional Torino Rodriguez de Mendonza de Amazonas, Chachapoyas 01001, Peru
| | - Camila F. Batista
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - Luiza C. Reis
- Laboratório de Soroloepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Wesley L. Fotoran
- Laboratório de Genética, Instituto Butantã, Universidade de São Paulo, São Paulo 05503-900, Brazil
| | - Marcos B. Heinemann
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - Adriano F. Cunha
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| | - Mussya C. Rocha
- Laboratório de Soroloepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
| | - Angélica R. Faria
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
- Laboratório de Parasitologia Clínica, Faculdade de Ciências Farmacêuticas, Universidade Federal de Alfenas, Alfenas 37130-000, Brazil
| | - Hélida M. Andrade
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Mônica M. O. P. Cerqueira
- Departamento de Tecnologia e Inspeção de Produtos de Origem Animal, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte 31270-010, Brazil
| | - Magnus Gidlund
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Hiro Goto
- Laboratório de Soroloepidemiologia e Imunobiologia, Instituto de Medicina Tropical, Universidade de São Paulo, São Paulo 05403-000, Brazil
- Departamento de Medicina Preventiva, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Alice Maria M. P. Della Libera
- Veterinary Clinical Immunology Research Group, Departamento de Clínica Médica, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| |
Collapse
|
19
|
Shivatare SS, Shivatare VS, Wong CH. Glycoconjugates: Synthesis, Functional Studies, and Therapeutic Developments. Chem Rev 2022; 122:15603-15671. [PMID: 36174107 PMCID: PMC9674437 DOI: 10.1021/acs.chemrev.1c01032] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glycoconjugates are major constituents of mammalian cells that are formed via covalent conjugation of carbohydrates to other biomolecules like proteins and lipids and often expressed on the cell surfaces. Among the three major classes of glycoconjugates, proteoglycans and glycoproteins contain glycans linked to the protein backbone via amino acid residues such as Asn for N-linked glycans and Ser/Thr for O-linked glycans. In glycolipids, glycans are linked to a lipid component such as glycerol, polyisoprenyl pyrophosphate, fatty acid ester, or sphingolipid. Recently, glycoconjugates have become better structurally defined and biosynthetically understood, especially those associated with human diseases, and are accessible to new drug, diagnostic, and therapeutic developments. This review describes the status and new advances in the biological study and therapeutic applications of natural and synthetic glycoconjugates, including proteoglycans, glycoproteins, and glycolipids. The scope, limitations, and novel methodologies in the synthesis and clinical development of glycoconjugates including vaccines, glyco-remodeled antibodies, glycan-based adjuvants, glycan-specific receptor-mediated drug delivery platforms, etc., and their future prospectus are discussed.
Collapse
Affiliation(s)
- Sachin S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Vidya S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
20
|
Gupta S, Sharma N, Naorem LD, Jain S, Raghava GP. Collection, compilation and analysis of bacterial vaccines. Comput Biol Med 2022; 149:106030. [DOI: 10.1016/j.compbiomed.2022.106030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 11/03/2022]
|
21
|
Chakraborty N, Jha D, Roy I, Kumar P, Gaurav SS, Marimuthu K, Ng OT, Lakshminarayanan R, Verma NK, Gautam HK. Nanobiotics against antimicrobial resistance: harnessing the power of nanoscale materials and technologies. J Nanobiotechnology 2022; 20:375. [PMID: 35953826 PMCID: PMC9371964 DOI: 10.1186/s12951-022-01573-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Given the spasmodic increment in antimicrobial resistance (AMR), world is on the verge of “post-antibiotic era”. It is anticipated that current SARS-CoV2 pandemic would worsen the situation in future, mainly due to the lack of new/next generation of antimicrobials. In this context, nanoscale materials with antimicrobial potential have a great promise to treat deadly pathogens. These functional materials are uniquely positioned to effectively interfere with the bacterial systems and augment biofilm penetration. Most importantly, the core substance, surface chemistry, shape, and size of nanomaterials define their efficacy while avoiding the development of AMR. Here, we review the mechanisms of AMR and emerging applications of nanoscale functional materials as an excellent substitute for conventional antibiotics. We discuss the potential, promises, challenges and prospects of nanobiotics to combat AMR.
Collapse
Affiliation(s)
- Nayanika Chakraborty
- Department of Chemistry, University of Delhi, New Delhi, 110007, India.,Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India
| | - Diksha Jha
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Indrajit Roy
- Department of Chemistry, University of Delhi, New Delhi, 110007, India
| | - Pradeep Kumar
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi University Campus, 110007, New Delhi, India
| | - Shailendra Singh Gaurav
- Department of Genetics and Plant Breeding, Faculty of Agriculture, Chaudhary Charan Singh University, Meerut, 250004, India
| | - Kalisvar Marimuthu
- National Centre for Infectious Diseases (NCID), Singapore, 308442, Singapore.,Tan Tock Seng Hospital (TTSH), 308433, Singapore, Singapore
| | - Oon-Tek Ng
- National Centre for Infectious Diseases (NCID), Singapore, 308442, Singapore.,Tan Tock Seng Hospital (TTSH), 308433, Singapore, Singapore
| | - Rajamani Lakshminarayanan
- Ocular Infections and Anti-Microbials Research Group, Singapore Eye Research Institute, The Academia, 20 College Road, Singapore, 169856, Singapore. .,Department of Pharmacy, National University of Singapore, Singapore, 117543, Singapore. .,Academic Clinical Program in Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore, 169857, Singapore.
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore, 308232, Singapore. .,National Skin Centre, Singapore, 308205, Singapore.
| | - Hemant K Gautam
- Department of Immunology and Infectious Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, New Delhi, 110025, India.
| |
Collapse
|
22
|
Izeli Portilho A, Araujo Correa V, Dos Santos Cirqueira C, De Gaspari E. Intranasal and Intramuscular Immunization with Outer Membrane Vesicles from Serogroup C Meningococci Induced Functional Antibodies and Immunologic Memory. Immunol Invest 2022; 51:2066-2085. [PMID: 35950702 DOI: 10.1080/08820139.2022.2107931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Immunization is the key to prevent invasive meningococcal disease (IMD), caused by Neisseria meningitidis. Outer membrane vesicles (OMVs) can be used as meningococcal antigens. METHODS Isogenic mice A/Sn (H2a) were immunized with low antigenic doses of OMVs of an N. meningitidis C:2a:P1.5 strain, via intranasal/intramuscular route, adjuvanted by cholera toxin subunit B (CTB) or via intramuscular route only, adjuvanted by aluminium hydroxide (AH). Mice were followed until old age and humoral and cellular responses were assessed by ELISA, Immunoblotting, Dot-blot, Serum-bactericidal assay, Immunohistochemistry and ELISpot. RESULTS OMV+CTB and OMV+AH groups presented statistically higher antibodies titers, which persisted until middle and old ages. IgG isotypes point to a Th2 type of response. Avidity indexes were considered high, regardless of adjuvant use, but only groups immunized with OMVs and adjuvants (OMV+CTB and OMV+AH) presented bactericidal activity. The antibodies recognized antigens of molecular weights attributed to porin and cross-reactivity proteins. Although the spleen of old mice did not present differences in immunohistochemistry marking of CD68+, CD4+, CD79+ and CD25+ cells, splenocytes of immune groups secreted IL-4 and IL-17 when stimulated with OMVs and meningococcal C polysaccharide. CONCLUSION We concluded that both adjuvants, CTB and AH, improved the immunogenicity of low doses of OMVs and contributed to a persistent immune response. Even though AH is well established in the vaccinology area, CTB seems to be a promising adjuvant candidate for meningococcal vaccines: it is suitable for mucosal delivery and supports a Th2 type of response. Therefore, OMVs are still a relevant vaccine platform.
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil.,Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Victor Araujo Correa
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil.,Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | | | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil.,Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Tobuse AJ, Ang CW, Yeong KY. Modern vaccine development via reverse vaccinology to combat antimicrobial resistance. Life Sci 2022; 302:120660. [PMID: 35642852 DOI: 10.1016/j.lfs.2022.120660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 10/18/2022]
Abstract
With the continuous evolution of bacteria, the global antimicrobial resistance health threat is causing millions of deaths yearly. While depending on antibiotics as a primary treatment has its merits, there are no effective alternatives thus far in the pharmaceutical market against some drug-resistant bacteria. In recent years, vaccinology has become a key topic in scientific research. Combining with the growth of technology, vaccine research is seeing a new light where the process is made faster and more efficient. Although less discussed, bacterial vaccine is a feasible strategy to combat antimicrobial resistance. Some vaccines have shown promising results with good efficacy against numerous multidrug-resistant strains of bacteria. In this review, we aim to discuss the findings from studies utilizing reverse vaccinology for vaccine development against some multidrug-resistant bacteria, as well as provide a summary of multi-year bacterial vaccine studies in clinical trials. The advantages of reverse vaccinology in the generation of new bacterial vaccines are also highlighted. Meanwhile, the limitations and future prospects of bacterial vaccine concludes this review.
Collapse
Affiliation(s)
- Asuka Joy Tobuse
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| | - Chee Wei Ang
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia.
| |
Collapse
|
24
|
Correa VA, Portilho AI, De Gaspari E. Immunological Effects of Dimethyldioctadecylammonium Bromide and Saponin as Adjuvants for Outer Membrane Vesicles from Neisseria meningitidis. Diseases 2022; 10:diseases10030046. [PMID: 35892740 PMCID: PMC9326571 DOI: 10.3390/diseases10030046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/13/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022] Open
Abstract
The meningococcal disease is a global health threat, but is preventable through vaccination. Adjuvants improve meningococcal vaccines and are able to trigger different aspects of the immune response. The present work evaluated the immune response of mice against Neisseria meningitidis outer membrane vesicles (OMV) complexed with the adjuvants aluminium hydroxide (AH), via subcutaneous route; and dimethyldioctadecylammonium bromide (DDA) or Saponin (Sap), via intranasal/subcutaneous routes. ELISA demonstrated that all adjuvants increased IgG titers after the booster dose, remaining elevated for 18 months. Additionally, adjuvants increased the avidity of the antibodies and the bactericidal titer: OMVs alone were bactericidal until 1:4 dilution but, when adjuvanted by Alum, DDA or Sap, it increased to 1/32. DDA and Sap increased all IgG isotypes, while AH improved IgG1 and IgG2a levels. Thus, Sap led to the recognition of more proteins in Immunoblot, followed by DDA and AH. Sap and AH induced higher IL-4 and IL-17 release, respectively. The use of adjuvants improved both cellular and humoral immune response, however, each adjuvant contributed to particular parameters. This demonstrates the importance of studying different adjuvant options and their suitability to stimulate different immune mechanisms, modulating the immune response.
Collapse
Affiliation(s)
- Victor Araujo Correa
- Immunology Center, Adolfo Lutz Institute, Av. Dr. Arnaldo, 355, 11th Floor, Room 1116, Cerqueira César, São Paulo 01246-902, SP, Brazil; (V.A.C.); (A.I.P.)
- Graduate Program Interunits in Biotechnology, Biomedical Sciences Institute, São Paulo University, Av. Prof. Lineu Prestes, 2415, ICB Hall III, Cidade Universitária, São Paulo 05508-900, SP, Brazil
| | - Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, Av. Dr. Arnaldo, 355, 11th Floor, Room 1116, Cerqueira César, São Paulo 01246-902, SP, Brazil; (V.A.C.); (A.I.P.)
- Graduate Program Interunits in Biotechnology, Biomedical Sciences Institute, São Paulo University, Av. Prof. Lineu Prestes, 2415, ICB Hall III, Cidade Universitária, São Paulo 05508-900, SP, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, Av. Dr. Arnaldo, 355, 11th Floor, Room 1116, Cerqueira César, São Paulo 01246-902, SP, Brazil; (V.A.C.); (A.I.P.)
- Graduate Program Interunits in Biotechnology, Biomedical Sciences Institute, São Paulo University, Av. Prof. Lineu Prestes, 2415, ICB Hall III, Cidade Universitária, São Paulo 05508-900, SP, Brazil
- Correspondence: ; Tel.: +55-11-3068-2898
| |
Collapse
|
25
|
van
der Put RMF, Smitsman C, de Haan A, Hamzink M, Timmermans H, Uittenbogaard J, Westdijk J, Stork M, Ophorst O, Thouron F, Guerreiro C, Sansonetti PJ, Phalipon A, Mulard LA. The First-in-Human Synthetic Glycan-Based Conjugate Vaccine Candidate against Shigella. ACS CENTRAL SCIENCE 2022; 8:449-460. [PMID: 35559427 PMCID: PMC9088300 DOI: 10.1021/acscentsci.1c01479] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 05/12/2023]
Abstract
Shigella, the causative agent of shigellosis, is among the main causes of diarrheal diseases with still a high morbidity in low-income countries. Relying on chemical synthesis, we implemented a multidisciplinary strategy to design SF2a-TT15, an original glycoconjugate vaccine candidate targeting Shigella flexneri 2a (SF2a). Whereas the SF2a O-antigen features nonstoichiometric O-acetylation, SF2a-TT15 is made of a synthetic 15mer oligosaccharide, corresponding to three non-O-acetylated repeats, linked at its reducing end to tetanus toxoid by means of a thiol-maleimide spacer. We report on the scale-up feasibility under GMP conditions of a high yielding bioconjugation process established to ensure a reproducible and controllable glycan/protein ratio. Preclinical and clinical batches complying with specifications from ICH guidelines, WHO recommendations for polysaccharide conjugate vaccines, and (non)compendial tests were produced. The obtained SF2a-TT15 vaccine candidate passed all toxicity-related criteria, was immunogenic in rabbits, and elicited bactericidal antibodies in mice. Remarkably, the induced IgG antibodies recognized a large panel of SF2a circulating strains. These preclinical data have paved the way forward to the first-in-human study for SF2a-TT15, demonstrating safety and immunogenicity. This contribution discloses the yet unreported feasibility of the GMP synthesis of conjugate vaccines featuring a unique homogeneous synthetic glycan hapten fine-tuned to protect against an infectious disease.
Collapse
Affiliation(s)
| | | | - Alex de Haan
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Martin Hamzink
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | | | | | - Janny Westdijk
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Michiel Stork
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Olga Ophorst
- Intravacc, P.O. Box 450, 3720 AL Bilthoven, The Netherlands
| | - Françoise Thouron
- Institut
Pasteur, U1202 Inserm, Unité
de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Catherine Guerreiro
- Institut
Pasteur, Université Paris Cité, CNRS UMR3523, Unité de Chimie des Biomolécules, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Philippe J. Sansonetti
- Institut
Pasteur, U1202 Inserm, Unité
de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
- Chaire
de Microbiologie et Maladies Infectieuses, Collège de France, 11, place Marcelin Berthelot, 75005 Paris, France
| | - Armelle Phalipon
- Institut
Pasteur, U1202 Inserm, Unité
de Pathogénie Microbienne Moléculaire, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Laurence A. Mulard
- Institut
Pasteur, Université Paris Cité, CNRS UMR3523, Unité de Chimie des Biomolécules, 28 rue du Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
26
|
Mutalik VK, Arkin AP. A Phage Foundry Framework to Systematically Develop Viral Countermeasures to Combat Antibiotic-Resistant Bacterial Pathogens. iScience 2022; 25:104121. [PMID: 35402883 PMCID: PMC8983348 DOI: 10.1016/j.isci.2022.104121] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
At its current rate, the rise of antimicrobial-resistant (AMR) infections is predicted to paralyze our industries and healthcare facilities while becoming the leading global cause of loss of human life. With limited new antibiotics on the horizon, we need to invest in alternative solutions. Bacteriophages (phages)-viruses targeting bacteria-offer a powerful alternative approach to tackle bacterial infections. Despite recent advances in using phages to treat recalcitrant AMR infections, the field lacks systematic development of phage therapies scalable to different applications. We propose a Phage Foundry framework to establish metrics for phage characterization and to fill the knowledge and technological gaps in phage therapeutics. Coordinated investment in AMR surveillance, sampling, characterization, and data sharing procedures will enable rational exploitation of phages for treatments. A fully realized Phage Foundry will enhance the sharing of knowledge, technology, and viral reagents in an equitable manner and will accelerate the biobased economy.
Collapse
Affiliation(s)
- Vivek K. Mutalik
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Adam P. Arkin
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
| |
Collapse
|
27
|
Noori Goodarzi N, Bolourchi N, Fereshteh S, Soltani Shirazi A, Pourmand MR, Badmasti F. Investigation of novel putative immunogenic targets against Staphylococcus aureus using a reverse vaccinology strategy. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105149. [PMID: 34801756 DOI: 10.1016/j.meegid.2021.105149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/31/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND The emergence of methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant S. aureus (VRSA) strains is a significant public health concern. Considering the high morbidity and mortality of invasive S. aureus infections and multi-drug resistant strains, there is an urgent need for non-antibiotic immune-based approaches to cure these infections. Despite all efforts, vaccine candidates targeting S. aureus failed in human clinical trials, and no approved vaccine is available against this pathogen. Therefore, this study aimed to introduce suitable candidates for immunization against S. aureus using a comprehensive reverse vaccinology approach. METHODS In this study, we retrieved putative immunogenic targets from three different levels (literature review, automated reverse vaccinology, and manual reverse vaccinology) and evaluated them using several immunoinformatics analyses including antigenicity, allergenicity, PSI-BLAST to human proteome, physiochemical properties, B-cell, and T-cell epitopes. In the next step, the quartile method scoring was used to the shortlisted proteins. Finally, the molecular docking and immune simulation of immunogenic targets were performed. RESULTS This study presents 12 vaccine candidates, including three enzymatic proteins (WP_000222271.1, WP_001170274, and WP_000827736.1), three cell wall-associated proteins (WP_001125631.1, WP_000731642, and WP_000751265.1), two hemolysins (WP_000594517.1, and WP_000916697.1), one secretion involved protein (WP_000725226.1), one heme‑iron binding protein (WP_001041573.1), one superantigen like protein (WP_000668994.1) and one hypothetical proteins (WP_000737711.1). CONCLUSION Through quartile scoring method, immune simulation and molecular docking, four promising targets including lytic transglycosylase IsaA, HlgA, secretory antigen precursor SsaA, and heme uptake protein IsdB were selected as the shortlisted proteins. It seems that a polarized immunization (Th1/Th17) response is needed for protection against this bacterium. An optimized formulation based on these putative immunogenic proteins and a wisely adjuvant selection may drive the immune system toward a full protection.
Collapse
Affiliation(s)
- Narjes Noori Goodarzi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Bolourchi
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Mohammad Reza Pourmand
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
28
|
Noori Goodarzi N, Bolourchi N, Fereshteh S, Badmasti F. Introduction of novel putative immunogenic targets against Proteus mirabilis using a reverse vaccinology approach. INFECTION GENETICS AND EVOLUTION 2021; 95:105045. [PMID: 34428568 DOI: 10.1016/j.meegid.2021.105045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 11/15/2022]
Abstract
Multi-drug resistance of Proteus mirabilis, a frequent cause of catheter-associated urinary tract infections, renders ineffective treatment. Therefore, new advanced strategies are needed to overcome it. In the meantime, vaccination may be the most effective and promising method. In this study antigenicity, allergenicity, subcellular localization, human homology, B-cell epitopes and MHC-II binding sites, conserved domains and protein-protein interactions were predicted using different reverse vaccinology methods and bioinformatics databases to find new putative immunogenic targets against P. mirabilis. Finally, 5 putative immunogenic targets against P. mirabilis were identified. Considering all criteria, QKQ94350.1 (Cell envelope opacity-associated protein A), QKQ94681.1 (Porin), QKQ95001.1 (TonB-dependent hemoglobin/ transferrin/ lactoferrin family receptor), QKQ95221.1 (AsmA) and QKQ94335.1 (N-acetylmuramoyl-L-alanine amidase) are excellent putative immunogenic targets. Finally, a multi-epitope vaccine was designed using the conserved linear epitopes of two OMPs (QKQ94681.1 and QKQ95001.1) and N-acetylmuramoyl-L-alanine amidase (QKQ94335.1), which have promising properties for immunization. These findings can simplify the development of efficient vaccines against P. mirabilis.
Collapse
Affiliation(s)
- Narjes Noori Goodarzi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Negin Bolourchi
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
29
|
Badmasti F, Habibi M, Firoozeh F, Fereshteh S, Bolourchi N, Goodarzi NN. The combination of CipA and PBP-7/8 proteins contribute to the survival of C57BL/6 mice from sepsis of Acinetobacter baumannii. Microb Pathog 2021; 158:105063. [PMID: 34166729 DOI: 10.1016/j.micpath.2021.105063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
Due to the emergence of multi-drug resistant Acinetobacter baumannii strains, there is an urgent need to develop several new strategies to control this bacterium. In this context, vaccination may be the best approach to reduce the morbidity and mortality associated with MDR isolates in vulnerable groups. Serum resistance factors have a key role in the pathogenesis of A. baumannii and can be considered as potential vaccine candidates. This project aimed to evaluate the immunological reactivity of CipA and PBP-7/8 as two serum resistance factors in a combination form against sepsis infections of A. baumannii. Recombinant proteins were obtained and immunological evaluations were performed against sepsis infection in the C57BL/6 mouse model. The data showed a statistically significant increase in total IgG levels in all three immunization regimens (CipA, PBP-7/8, and CipA + PBP-7/8) compared to the control group. The ratios of IgG2c/IgG1 in the CipA, PBP-7/8, and CipA + PBP-7/8 schedules were 8.7, 46.50, and 33.29, respectively. It appears that the immunization schedules developed a strong polarized Th1 response. The cytokine profiles of the three plans showed that IFN-γ was highly concentrated in the combination plan. However, the highest concentration of IL-17 belonged to the PBP-7/8 plan. In conclusion, the data of total IgG, survival rates and splenic bacterial loads showed that the CipA + PBP-7/8 plan was more effective than each protein individually.
Collapse
Affiliation(s)
- Farzad Badmasti
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Iran.
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzaneh Firoozeh
- Department of Microbiology, School of Medicine, Alborz University of Medical Science, Karaj, Iran
| | | | - Negin Bolourchi
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| | - Narjes Noori Goodarzi
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Mayer RL, Impens F. Immunopeptidomics for next-generation bacterial vaccine development. Trends Microbiol 2021; 29:1034-1045. [PMID: 34030969 DOI: 10.1016/j.tim.2021.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
Antimicrobial resistance is an increasing global threat and alternative treatments substituting failing antibiotics are urgently needed. Vaccines are recognized as highly effective tools to mitigate antimicrobial resistance; however, the selection of bacterial antigens as vaccine candidates remains challenging. In recent years, advances in mass spectrometry-based proteomics have led to the development of so-called immunopeptidomics approaches that allow the untargeted discovery of bacterial epitopes that are presented on the surface of infected cells. Especially for intracellular bacterial pathogens, immunopeptidomics holds great promise to uncover antigens that can be encoded in viral vector- or nucleic acid-based vaccines. This review provides an overview of immunopeptidomics studies on intracellular bacterial pathogens and considers future directions and challenges in advancing towards next-generation vaccines.
Collapse
Affiliation(s)
- Rupert L Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|