1
|
Santamarina-Fernández R, Fuentes-Valverde V, Silva-Rodríguez A, García P, Moscoso M, Bou G. Pseudomonas aeruginosa Vaccine Development: Lessons, Challenges, and Future Innovations. Int J Mol Sci 2025; 26:2012. [PMID: 40076637 PMCID: PMC11900337 DOI: 10.3390/ijms26052012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen with a multidrug-resistant profile that has become a critical threat to global public health. It is one of the main causes of severe nosocomial infections, including ventilator-associated pneumonia, chronic infections in patients with cystic fibrosis, and bloodstream infections in immunosuppressed individuals. Development of vaccines against P. aeruginosa is a major challenge owing to the high capacity of this bacterium to form biofilms, its wide arsenal of virulence factors (including secretion systems, lipopolysaccharides, and outer membrane proteins), and its ability to evade the host immune system. This review provides a comprehensive historical overview of vaccine development efforts targeting this pathogen, ranging from early attempts in the 1970s to recent advancements, including vaccines based on novel proteins and emerging technologies such as nanoparticles and synthetic conjugates. Despite numerous promising preclinical developments, very few candidates have progressed to clinical trials, and none have achieved final approval. This panorama highlights the significant scientific efforts undertaken and the inherent complexity of successfully developing an effective vaccine against P. aeruginosa.
Collapse
Affiliation(s)
- Rebeca Santamarina-Fernández
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Víctor Fuentes-Valverde
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Área de Medicamentos Biológicos, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), 28022 Madrid, Spain
| | - Alis Silva-Rodríguez
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Patricia García
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miriam Moscoso
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Universidad de A Coruña, 15006 A Coruña, Spain
| |
Collapse
|
2
|
Inoue K, Kinoshita M, Muranishi K, Ohara J, Sudo K, Kawaguchi K, Shimizu M, Naito Y, Moriyama K, Sawa T. Effect of a Novel Trivalent Vaccine Formulation against Acute Lung Injury Caused by Pseudomonas aeruginosa. Vaccines (Basel) 2023; 11:1088. [PMID: 37376477 DOI: 10.3390/vaccines11061088] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
An effective vaccine against Pseudomonas aeruginosa would benefit people susceptible to severe infection. Vaccination targeting V antigen (PcrV) of the P. aeruginosa type III secretion system is a potential prophylactic strategy for reducing P. aeruginosa-induced acute lung injury and acute mortality. We created a recombinant protein (designated POmT) comprising three antigens: full-length PcrV (PcrV#1-#294), the outer membrane domain (#190-342) of OprF (OprF#190-#342), and a non-catalytic mutant of the carboxyl domain (#406-613) of exotoxin A (mToxA#406-#613(E553Δ)). In the combination of PcrV and OprF, mToxA, the efficacy of POmT was compared with that of single-antigen vaccines, two-antigen mixed vaccines, and a three-antigen mixed vaccine in a murine model of P. aeruginosa pneumonia. As a result, the 24 h-survival rates were 79%, 78%, 21%, 7%, and 36% in the POmT, PcrV, OprF, mTox, and alum-alone groups, respectively. Significant improvement in acute lung injury and reduction in acute mortality within 24 h after infection was observed in the POmT and PcrV groups than in the other groups. Overall, the POmT vaccine exhibited efficacy comparable to that of the PcrV vaccine. The future goal is to prove the efficacy of the POmT vaccine against various P. aeruginosa strains.
Collapse
Affiliation(s)
- Keita Inoue
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kentaro Muranishi
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka 814-0180, Japan
| | - Junya Ohara
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ken Kawaguchi
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masaru Shimizu
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshifumi Naito
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Mitaka 181-8611, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
3
|
Wang X, Liu C, Rcheulishvili N, Papukashvili D, Xie F, Zhao J, Hu X, Yu K, Yang N, Pan X, Liu X, Wang PG, He Y. Strong immune responses and protection of PcrV and OprF-I mRNA vaccine candidates against Pseudomonas aeruginosa. NPJ Vaccines 2023; 8:76. [PMID: 37231060 DOI: 10.1038/s41541-023-00672-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Pseudomonas aeruginosa (PA) is a leading cause of hospital-acquired and ventilator-associated pneumonia. The multidrug-resistance (MDR) rate of PA is increasing making the management of PA a global challenge. Messenger RNA (mRNA) vaccines represent the most promising alternative to conventional vaccines and are widely studied for viral infection and cancer immunotherapy while rarely studied for bacterial infections. In this study, two mRNA vaccines encoding PcrV- the key component of the type III secretion system in Pseudomonas and the fusion protein OprF-I comprising outer membrane proteins OprF and OprI were constructed. The mice were immunized with either one of these mRNA vaccines or with the combination of both. Additionally, mice were vaccinated with PcrV, OprF, or the combination of these two proteins. Immunization with either mRNA-PcrV or mRNA-OprF-I elicited a Th1/Th2 mixed or slighted Th1-biased immune response, conferred broad protection, and reduced bacterial burden and inflammation in burn and systemic infection models. mRNA-PcrV induced significantly stronger antigen-specific humoral and cellular immune responses and higher survival rate compared with the OprF-I after challenging with all the PA strains tested. The combined mRNA vaccine demonstrated the best survival rate. Moreover, the mRNA vaccines showed the superiority over protein vaccines. These results suggest that mRNA-PcrV as well as the mixture of mRNA-PcrV and mRNA-OprF-I are promising vaccine candidates for the prevention of PA infection.
Collapse
Affiliation(s)
- Xingyun Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Department of Critical Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Cong Liu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Nino Rcheulishvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Dimitri Papukashvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Fengfei Xie
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jiao Zhao
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xing Hu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Kaiwei Yu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Nuo Yang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xuehua Pan
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xueyan Liu
- Department of Critical Medicine, Shenzhen People's Hospital, First Affiliated Hospital of Southern University of Science and Technology, Second Clinical Medicine College of Jinan University, Shenzhen, China.
| | - Peng George Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Yunjiao He
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
4
|
Killough M, Rodgers AM, Ingram RJ. Pseudomonas aeruginosa: Recent Advances in Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10071100. [PMID: 35891262 PMCID: PMC9320790 DOI: 10.3390/vaccines10071100] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Pseudomonas aeruginosa is an important opportunistic human pathogen. Using its arsenal of virulence factors and its intrinsic ability to adapt to new environments, P. aeruginosa causes a range of complicated acute and chronic infections in immunocompromised individuals. Of particular importance are burn wound infections, ventilator-associated pneumonia, and chronic infections in people with cystic fibrosis. Antibiotic resistance has rendered many of these infections challenging to treat and novel therapeutic strategies are limited. Multiple clinical studies using well-characterised virulence factors as vaccine antigens over the last 50 years have fallen short, resulting in no effective vaccination being available for clinical use. Nonetheless, progress has been made in preclinical research, namely, in the realms of antigen discovery, adjuvant use, and novel delivery systems. Herein, we briefly review the scope of P. aeruginosa clinical infections and its major important virulence factors.
Collapse
Affiliation(s)
- Matthew Killough
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK;
| | - Aoife Maria Rodgers
- Department of Biology, The Kathleen Lonsdale Institute for Human Health Research, Maynooth University, R51 A021 Maynooth, Ireland;
| | - Rebecca Jo Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, UK;
- Correspondence:
| |
Collapse
|
5
|
Development of Multi-epitope Subunit Vaccine Against Pseudomonas aeruginosa Using OprF/OprI and PopB Proteins. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2021. [DOI: 10.5812/archcid.118243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The emerging problem of antibiotic resistance in Pseudomonas aeruginosa is a global health concern; hence, revealing innovative therapeutic approaches (such as designing an immunogenic vaccine candidate) is needed. There is no evidence of the availability of an effective vaccine that can combat the infection caused by this microorganism. Objectives: This research was conducted to develop a potential chimeric vaccine against P. aeruginosa using reverse vaccinology approaches. Methods: The present vaccine candidate comprised outer membrane protein F and I (OprF/OprI) and PopB with appropriate linkers. After applying meticulous immune-informatics investigation, the multi-epitope vaccine was created, including helper T lymphocyte (HTL), cytotoxic T lymphocyte (CTL), interferon gamma (IFN-γ), and interleukin 4 (IL-4) epitopes. Then, the physicochemical characteristics, allergenicity, toxicity, and antigenicity were analyzed. After investigating the secondary structure, the tertiary structure (3D) model was generated, refined, and validated via computational methods. Besides, the strong protein-ligand interaction and stability between the vaccine candidate and toll-like receptor 4 (TLR4) were determined via molecular docking and dynamics analyses. Moreover, in silico cloning accompanied by pET-22b (+) was used to achieve high translation efficiency. Results: Our results presumed that the chimeric-designed vaccine was thermostable and contained optimal physicochemical properties. This vaccine candidate was nontoxic and highly soluble and had stable protein and TLR4 interaction, adequately overexpressed in Escherichia coli. Overall, it could induce immune responses and repress this microorganism. Conclusions: Therefore, to inhibit Pseudomonas infections experimentally, the efficacy and safety of the vaccine design need to be validated.
Collapse
|
6
|
Khaledi M, Afkhami H, Matouri RN, Dezfuli AAZ, Bakhti S. Effective Strategies to Deal With Infection in Burn Patient. J Burn Care Res 2021; 43:931-935. [PMID: 34935044 DOI: 10.1093/jbcr/irab226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Treatment of bacterial infection is difficult. Treatment protocol of burned patient is hard. Furthermore, treatment in burned patients is accompanied with problems such as complexity in diagnosis of infection's agent, multiple infections, being painful, and involving with different organelles. There are different infections of Gram-positive and Gram-negative bacteria in burned patients. From important bacteria can be noted to Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus that have high range of morbidity and mortality. Treatment of those bacterial infections is extremely important. Hence, many studies about methods of treatment of bacterial infections have published. Herein, we have suggested practical methods for example ant virulence therapies, nanotechnology, vaccine, and photodynamic therapy in treatment of bacterial infections. Those methods have been done in many researches and had good effect.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Hamed Afkhami
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Raed Nezhad Matouri
- Department of Medical Library and Information Sciences, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | | | - Shahriar Bakhti
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
7
|
Quest for Novel Preventive and Therapeutic Options Against Multidrug-Resistant Pseudomonas aeruginosa. Int J Pept Res Ther 2021; 27:2313-2331. [PMID: 34393689 PMCID: PMC8351238 DOI: 10.1007/s10989-021-10255-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 11/20/2022]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a critical healthcare challenge due to its ability to cause persistent infections and the acquisition of antibiotic resistance mechanisms. Lack of preventive vaccines and rampant drug resistance phenomenon has rendered patients vulnerable. As new antimicrobials are in the preclinical stages of development, mining for the unexploited drug targets is also crucial. In the present study, we designed a B- and T-cell multi-epitope vaccine against P. aeruginosa using a subtractive proteomics and immunoinformatics approach. A total of five proteins were shortlisted based on essentiality, extracellular localization, virulence, antigenicity, pathway association, hydrophilicity, and low molecular weight. These include two outer membrane porins; OprF (P13794) and OprD (P32722), a protein activator precursor pra (G3XDA9), a probable outer membrane protein precursor PA1288 (Q9I456), and a conserved hypothetical protein PA4874 (Q9HUT9). These shortlisted proteins were further analyzed to identify immunogenic and antigenic B- and T-cell epitopes. The best scoring epitopes were then further subjected to the construction of a polypeptide multi-epitope vaccine and joined with cholera toxin B subunit adjuvant. The final chimeric construct was docked with TLR4 and confirmed by normal mode simulation studies. The designed B- and T-cell multi-epitope vaccine candidate is predicted immunogenic in nature and has shown strong interactions with TLR-4. Immune simulation predicted high-level production of B- and T-cell population and maximal expression was ensured in E. coli strain K12. The identified drug targets qualifying the screening criteria were: UDP-2-acetamido-2-deoxy-d-glucuronic acid 3-dehydrogenase WbpB (G3XD23), aspartate semialdehyde dehydrogenase (Q51344), 2-amino-4-hydroxy-6-hydroxymethyldihydropteridine pyrophosphokinase (Q9HV71), 3-deoxy-D-manno-octulosonic-acid transferase (Q9HUH7), glycyl-tRNA synthetase alpha chain (Q9I7B7), riboflavin kinase/FAD synthase (Q9HVM3), aconitate hydratase 2 (Q9I2V5), probable glycosyltransferase WbpH (G3XD85) and UDP-3-O-[3-hydroxylauroyl] glucosamine N-acyltransferase (Q9HXY6). For druggability and pocketome analysis crystal and homology structures of these proteins were retrieved and developed. A sequence-based search was performed in different databases (ChEMBL, Drug Bank, PubChem and Pseudomonas database) for the availability of reported ligands and tested drugs for the screened targets. These predicted targets may provide a basis for the development of reliable antibacterial preventive and therapeutic options against P. aeruginosa.
Collapse
|
8
|
Mayeux G, Gayet L, Liguori L, Odier M, Martin DK, Cortès S, Schaack B, Lenormand JL. Cell-free expression of the outer membrane protein OprF of Pseudomonas aeruginosa for vaccine purposes. Life Sci Alliance 2021; 4:4/6/e202000958. [PMID: 33972378 PMCID: PMC8127326 DOI: 10.26508/lsa.202000958] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022] Open
Abstract
Production of recombinant proteoliposomes containing OprF from P. aeruginosa promotes the active open conformation of the porin exposing native epitopes. These OprF proteoliposomes were used as vaccines to protect mice against a P. aeruginosa acute pulmonary infection model. Pseudomonas aeruginosa is the second-leading cause of nosocomial infections and pneumonia in hospitals. Because of its extraordinary capacity for developing resistance to antibiotics, treating infections by Pseudomonas is becoming a challenge, lengthening hospital stays, and increasing medical costs and mortality. The outer membrane protein OprF is a well-conserved and immunogenic porin playing an important role in quorum sensing and in biofilm formation. Here, we used a bacterial cell-free expression system to reconstitute OprF under its native forms in liposomes and we demonstrated that the resulting OprF proteoliposomes can be used as a fully functional recombinant vaccine against P. aeruginosa. Remarkably, we showed that our system promotes the folding of OprF into its active open oligomerized state as well as the formation of mega-pores. Our approach thus represents an easy and efficient way for producing bacterial membrane antigens exposing native epitopes for vaccine purposes.
Collapse
Affiliation(s)
- Géraldine Mayeux
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Landry Gayet
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | - Lavinia Liguori
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Maison Familiale Rurale Moirans, Moirans, France
| | - Marine Odier
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,Catalent Pharma Solutions, Eberbach, Germany
| | - Donald K Martin
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| | | | - Béatrice Schaack
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France.,University Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Jean-Luc Lenormand
- TheREx and Synabi, University Grenoble Alpes, CNRS, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble Institut Polytechnique (INP), Translational Innovation in Medicine and Complexity (TIMC), Grenoble, France
| |
Collapse
|
9
|
López-Siles M, Corral-Lugo A, McConnell MJ. Vaccines for multidrug resistant Gram negative bacteria: lessons from the past for guiding future success. FEMS Microbiol Rev 2021; 45:fuaa054. [PMID: 33289833 DOI: 10.1093/femsre/fuaa054] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance is a major threat to global public health. Vaccination is an effective approach for preventing bacterial infections, however it has not been successfully applied to infections caused by some of the most problematic multidrug resistant pathogens. In this review, the potential for vaccines to contribute to reducing the burden of disease of infections caused by multidrug resistant Gram negative bacteria is presented. Technical, logistical and societal hurdles that have limited successful vaccine development for these infections in the past are identified, and recent advances that can contribute to overcoming these challenges are assessed. A synthesis of vaccine technologies that have been employed in the development of vaccines for key multidrug resistant Gram negative bacteria is included, and emerging technologies that may contribute to future successes are discussed. Finally, a comprehensive review of vaccine development efforts over the last 40 years for three of the most worrisome multidrug resistant Gram negative pathogens, Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa is presented, with a focus on recent and ongoing studies. Finally, future directions for the vaccine development field are highlighted.
Collapse
Affiliation(s)
- Mireia López-Siles
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andrés Corral-Lugo
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Michael J McConnell
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
10
|
Sabzehali F, Rahimi H, Goudarzi H, Goudarzi M, Yoosefi Izad MH, Salimi Chirani A, Jalali SA, Faghihloo E. Functional engineering of OprF-OprI-PopB as a chimeric immunogen and its cross-protective evaluation with GM-CSF against Pseudomonas aeruginosa: A comprehensive immunoinformatics evaluation. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
11
|
Sainz-Mejías M, Jurado-Martín I, McClean S. Understanding Pseudomonas aeruginosa-Host Interactions: The Ongoing Quest for an Efficacious Vaccine. Cells 2020; 9:cells9122617. [PMID: 33291484 PMCID: PMC7762141 DOI: 10.3390/cells9122617] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022] Open
Abstract
Pseudomonas aeruginosa is a leading cause of chronic respiratory infections in people with cystic fibrosis (CF), bronchiectasis or chronic obstructive pulmonary disease (COPD), and acute infections in immunocompromised individuals. The adaptability of this opportunistic pathogen has hampered the development of antimicrobial therapies, and consequently, it remains a major threat to public health. Due to its antimicrobial resistance, vaccines represent an alternative strategy to tackle the pathogen, yet despite over 50 years of research on anti-Pseudomonas vaccines, no vaccine has been licensed. Nevertheless, there have been many advances in this field, including a better understanding of the host immune response and the biology of P. aeruginosa. Multiple antigens and adjuvants have been investigated with varying results. Although the most effective protective response remains to be established, it is clear that a polarised Th2 response is sub-optimal, and a mixed Th1/Th2 or Th1/Th17 response appears beneficial. This comprehensive review collates the current understanding of the complexities of P. aeruginosa-host interactions and its implication in vaccine design, with a view to understanding the current state of Pseudomonal vaccine development and the direction of future efforts. It highlights the importance of the incorporation of appropriate adjuvants to the protective antigen to yield optimal protection.
Collapse
|
12
|
Blears E, Sommerhalder C, Toliver-Kinsky T, Finnerty CC, Herndon DN. Current problems in burn immunology. Curr Probl Surg 2020; 57:100779. [PMID: 32507131 DOI: 10.1016/j.cpsurg.2020.100779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Elizabeth Blears
- Department of Surgery, University of Texas Medical Branch, Galveston, TX
| | | | - Tracy Toliver-Kinsky
- Department of Anesthesiology, Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX.
| | - Celeste C Finnerty
- Department of Surgery, University of Texas Medical Branch, Galveston, TX; Shriners Hospitals for Children, Galveston, TX
| | | |
Collapse
|
13
|
Ranjbar M, Behrouz B, Norouzi F, Mousavi Gargari SL. Anti-PcrV IgY antibodies protect against Pseudomonas aeruginosa infection in both acute pneumonia and burn wound models. Mol Immunol 2019; 116:98-105. [PMID: 31634816 DOI: 10.1016/j.molimm.2019.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly acquires antibiotic resistance; thus, developing an effective therapeutic approach is the most promising strategy for combating infection. Type III secretion system (T3SS) translocates bacterial toxins into the cytosol of the targeted eukaryotic cells, which plays important roles in the virulence of P. aeruginosa infections in both acute pneumonia and burn wound models. The PcrV protein, a T3SS translocating protein, is required for T3SS function and is a well-validated target in animal models of immunoprophylactic strategies targeting P. aeruginosa. In the present study, we evaluated the protective efficacy of chicken egg yolk antibodies (IgY) raised against recombinant PcrV (r-PcrV) in both acute pneumonia and burn wound models. R-PcrV protein was generated by expressing the pcrV gene (cloned in pET-28a vector) in E. coli BL-21. Anti-PcrV IgY was obtained by immunization of hen. Anti-PcrV IgY induced greater protection in P. aeruginosamurine acute pneumonia and burn wound models than control IgY (C-IgY) and PBS groups. Anti-PcrV IgY improved opsonophagocytic killing and inhibition of bacterial invasion of host cells. Taken together, our data provide evidence that anti-PcrV IgY can be a promising therapeutic candidate for combating P. aeruginosa infections.
Collapse
Affiliation(s)
- Mahya Ranjbar
- Department of Microbiology, Shahed University, Faculty of Medical Sciences, Tehran, Iran; Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | - Bahador Behrouz
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | - Fatemeh Norouzi
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | | |
Collapse
|
14
|
Hashemi FB, Behrouz B, Irajian G, Laghaei P, Korpi F, Fatemi MJ. A trivalent vaccine consisting of "flagellin A+B and pilin" protects against Pseudomonas aeruginosa infection in a murine burn model. Microb Pathog 2019; 138:103697. [PMID: 31465785 DOI: 10.1016/j.micpath.2019.103697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/22/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly achieves antibiotic resistance, and thus, developing an effective vaccine is critically important for combating P. aeruginosa infection. Flagella and pili play important roles in colonization of P. aeruginosa at the burn wound site and its subsequent dissemination to deeper tissue and organs. In the present study, we evaluated protective efficacy of a trivalent vaccine containing flagellins A and B (FlaA + FlaB) + pilin (PilA) in a murine burn model of infection. "FlaA + FlaB + PilA" induced greater protection in P. aeruginosa murine burn model than the single components alone, and it showed broad immune protection against P. aeruginosa strains. Immunization with "FlaA + FlaB + PilA" induced strong opsonophagocytic antibodies and resulted in reduced bacterial loads, systemic IL-12/IL-10 cytokine expression, and increased survival after challenge with three times lethal dose fifty (LD50) of P. eruginosa strains. Moreover, the protective efficacy of "FlaA + FlaB + PilA" vaccination was largely attributed to specific antibodies. Taken together, these data further confirm that the protective effects of "FlaA + FlaB + PilA" vaccine significantly enhance efficacy compared with antibodies against either mono or divalent antigen, and that the former broadens the coverage against P. eruginosa strains that express two of the three antigens.
Collapse
Affiliation(s)
- Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Baker SM, Pociask D, Clements JD, McLachlan JB, Morici LA. Intradermal vaccination with a Pseudomonas aeruginosa vaccine adjuvanted with a mutant bacterial ADP-ribosylating enterotoxin protects against acute pneumonia. Vaccine 2019; 37:808-816. [PMID: 30638799 DOI: 10.1016/j.vaccine.2018.12.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 12/20/2022]
Abstract
Respiratory infections are a leading cause of morbidity and mortality globally. This is partially due to a lack of effective vaccines and a clear understanding of how vaccination route and formulation influence protective immunity in mucosal tissues such as the lung. Pseudomonas aeruginosa is an opportunistic pathogen capable of causing acute pulmonary infections and is a leading cause of hospital-acquired and ventilator-associated pneumonia. With multidrug-resistant P. aeruginosa infections on the rise, the need for a vaccine against this pathogen is critical. Growing evidence suggests that a successful P. aeruginosa vaccine may require mucosal antibody and Th1- and Th17-type CD4+ T cells to prevent pulmonary infection. Intradermal immunization with adjuvants, such as the bacterial ADP-Ribosylating Enterotoxin Adjuvant (BARE) double mutant of E. coli heat-labile toxin (dmLT), can direct protective immune responses to mucosal tissues, including the lungs. We reasoned that intradermal immunization with P. aeruginosa outer membrane proteins (OMPs) adjuvanted with dmLT could drive neutralizing antibodies and migration of CD4+ T cells to the lungs and protect against P. aeruginosa pneumonia in a murine model. Here we show that mice immunized with OMPs and dmLT had significantly more antigen-specific IgG and Th1- and Th17-type CD4+ memory T cells in the pulmonary environment compared to control groups of mice. Furthermore, OMPs and dmLT immunized mice were significantly protected against an otherwise lethal lung infection. Protection was associated with early IFN-γ and IL-17 production in the lungs of immunized mice. These results indicate that intradermal immunization with dmLT can drive protective immunity to the lung mucosa and may be a viable vaccination strategy for a multitude of respiratory pathogens.
Collapse
Affiliation(s)
- Sarah M Baker
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - Derek Pociask
- Department of Medicine, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - John D Clements
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA.
| |
Collapse
|
16
|
Rafiezadeh Shahi H, Vahedian M, Movahedi M, Bahaadinbeigy K, Hashemian M, Mirafzal A. Measuring serum albumin levels at 0 and 24 h: Effect on the accuracy of clinical evaluations in the prediction of burn-related mortality. Burns 2018; 44:709-717. [DOI: 10.1016/j.burns.2017.10.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 11/27/2022]
|
17
|
Protective Efficacy of the Trivalent Pseudomonas aeruginosa Vaccine Candidate PcrV-OprI-Hcp1 in Murine Pneumonia and Burn Models. Sci Rep 2017. [PMID: 28638106 PMCID: PMC5479855 DOI: 10.1038/s41598-017-04029-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Pseudomonas aeruginosa is a formidable pathogen that is responsible for a diverse spectrum of human infectious diseases, resulting in considerable annual mortality rates. Because of biofilm formation and its ability of rapidly acquires of resistance to many antibiotics, P. aeruginosa related infections are difficult to treat, and therefore, developing an effective vaccine is the most promising method for combating infection. In the present study, we designed a novel trivalent vaccine, PcrV28-294-OprI25-83-Hcp11-162 (POH), and evaluated its protective efficacy in murine pneumonia and burn models. POH existed as a dimer in solution, it induced better protection efficacy in P. aeruginosa lethal pneumonia and murine burn models than single components alone when formulated with Al(OH)3 adjuvant, and it showed broad immune protection against several clinical isolates of P. aeruginosa. Immunization with POH induced strong immune responses and resulted in reduced bacterial loads, decreased pathology, inflammatory cytokine expression and inflammatory cell infiltration. Furthermore, in vitro opsonophagocytic killing assay and passive immunization studies indicated that the protective efficacy mediated by POH vaccination was largely attributed to POH-specific antibodies. Taken together, these data provided evidence that POH is a potentially promising vaccine candidate for combating P. aeruginosa infection in pneumonia and burn infections.
Collapse
|
18
|
Rello J, Krenn CG, Locker G, Pilger E, Madl C, Balica L, Dugernier T, Laterre PF, Spapen H, Depuydt P, Vincent JL, Bogár L, Szabó Z, Völgyes B, Máñez R, Cakar N, Ramazanoglu A, Topeli A, Mastruzzo MA, Jasovich A, Remolif CG, Del Carmen Soria L, Andresen Hernandez MA, Ruiz Balart C, Krémer I, Molnár Z, von Sonnenburg F, Lyons A, Joannidis M, Burgmann H, Welte T, Klingler A, Hochreiter R, Westritschnig K. A randomized placebo-controlled phase II study of a Pseudomonas vaccine in ventilated ICU patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:22. [PMID: 28159015 PMCID: PMC5291979 DOI: 10.1186/s13054-017-1601-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/04/2017] [Indexed: 11/10/2022]
Abstract
Background Currently, no vaccine against Pseudomonas is available. IC43 is a new, recombinant, protein (OprF/I)-based vaccine against the opportunistic pathogen, Pseudomonas aeruginosa, a major cause of serious hospital-acquired infections. IC43 has proven immunogenicity and tolerability in healthy volunteers, patients with burns, and patients with chronic lung diseases. In order to assess the immunogenicity and safety of IC43 in patients who are most at risk of acquiring Pseudomonas infections, it was evaluated in mechanically ventilated ICU patients. Methods We conducted a randomized, placebo-controlled, partially blinded study in mechanically ventilated ICU patients. The immunogenicity of IC43 at day 14 was determined as the primary endpoint, and safety, efficacy against P. aeruginosa infections, and all-cause mortality were evaluated as secondary endpoints. Vaccinations (100 μg or 200 μg IC43 with adjuvant, or 100 μg IC43 without adjuvant, or placebo) were given twice in a 7-day interval and patients were followed up for 90 days. Results Higher OprF/I IgG antibody titers were seen at day 14 for all IC43 groups versus placebo (P < 0.0001). Seroconversion (≥4-fold increase in OprF/I IgG titer from days 0 to 14) was highest with 100 μg IC43 without adjuvant (80.6%). There were no significant differences in P. aeruginosa infection rates, with a low rate of invasive infections (pneumonia or bacteremia) in the IC43 groups (11.2-14.0%). Serious adverse events (SAEs) considered possibly related to therapy were reported by 2 patients (1.9%) in the group of 100 µg IC43 with adjuvant. Both SAEs resolved and no deaths were related to study treatment. Local tolerability symptoms were mild and rare (<5% of patients), a low rate of treatment-related treatment-emergent adverse events (3.1–10.6%) was observed in the IC43 groups. Conclusion This phase II study has shown that IC43 vaccination of ventilated ICU patients produced a significant immunogenic effect. P. aeruginosa infection rates did not differ significantly between groups. In the absence of any difference in immune response following administration of 100 μg IC43 without adjuvant compared with 200 μg IC43 with adjuvant, the 100 μg dose without adjuvant was considered for further testing of its possible benefit of improved outcomes. There were no safety or mortality concerns. Trial registration ClinicalTrials.gov, NCT00876252. Registered on 3 April 2009. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1601-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jordi Rello
- Hospital Universitari Joan XXIII, C. Dr. Mallafrè Guasch 4, 43007, Tarragona, Spain. .,CIBERES, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119, 08035, Barcelona, Spain. .,Universitat Autonoma de Barcelona, Barcelona, Spain.
| | - Claus-Georg Krenn
- Medical University of Vienna, Intensive Care 13C1, Währinger Gürtel 18 - 20, 1090, Vienna, Austria
| | - Gottfried Locker
- Department of Internal Medicine I, Medical University of Vienna, Intensive Care 13I2, Währinger Gürtel 18 - 20, 1090, Vienna, Austria
| | - Ernst Pilger
- Intensive Care, Department of Internal Medicine, University Hospital Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Christian Madl
- Department of Internal Medicine III, Intensive Care 13H1, Medical University of Vienna, Währinger Gürtel 18 - 20, 1090, Vienna, Austria
| | - Laura Balica
- Emergency Clinical Hospital Bucharest, Toxicology - ICU, 8 Floreasca Street, 01446, Bucharest, Romania
| | - Thierry Dugernier
- Clinique St. Pierre, Intensive Care Department, Avenue Reine Fabiola 9, 1340, Ottignies, Belgium
| | - Pierre-Francois Laterre
- Department of CCM, St. Luc University Hospital UCL, Université Catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Herbert Spapen
- University Hospital Vrije Universiteit Brussels, Laarbeeklaan 101, 1090, Brussels, Belgium
| | | | - Jean-Louis Vincent
- Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Lajos Bogár
- University of Pécs Anesthesiology and Intensive Care Department, Ifjúság ut 13, 7624, Pécs, Hungary
| | | | - Barbara Völgyes
- Bajcsy Zsilinszky Hospital and Polyclinic, Intensive Care Unit, Maglodi út 89-91, 1106, Budapest, Hungary
| | - Rafael Máñez
- Department for Critical Care Medicine, Bellvitge University Hospital, Feixa Llarga s/n, 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Nahit Cakar
- Department of Anesthesiology and Reanimation, Istanbul University Capa Medical Faculty, 34390, Istanbul, Turkey
| | - Atilla Ramazanoglu
- Department of Anesthesiology, Dumlupinar Bulvari Kampus Antalya, Akdeniz University, Faculty of Medicine Hospital, 07070, Antalya, Turkey
| | - Arzu Topeli
- Department of Internal Medicine, Intensive Care Unit, Hacettepe University Hospital, 06100, Ankara, Turkey
| | - Maria A Mastruzzo
- Hospital Dr. Carlos Bocalandro, Ruta 8 No. 9100, B1657BHD Loma Hermosa, Partido 3 de Febrero, Buenos Aires, Argentina
| | - Abel Jasovich
- Sanatorio Güemes, Av. Roque Sanchez Pena 811 5°C, C1035AAP, Buenos Aires, Argentina
| | - Christian G Remolif
- Hospital "Heroes de Malvinas", Av. Ricardo Balbín 1910, B1721FJN Merlo, Buenos Aires, Argentina
| | | | - Max A Andresen Hernandez
- Hospital Clinico, Facultad de Medicina Pontificia, Universidad Católica de Chile, Marcoleta 367, Santiago, Chile
| | - Carolina Ruiz Balart
- Hospital Dr. Sótero del Rio, Unidad de Cuidado Intensivo, Departamento de Medicina Intensiva, Escuela de Medicina, Pontificia Universidad Católica de Chile, Avenida Concha y Toro, 3459, Puente Alto, Santiago, Chile
| | - Ildikó Krémer
- Flor Ferenc County Hospital, Semmelweis tér 1, 2143, Kistarcsa, Hungary
| | - Zsolt Molnár
- Department of Anaesthesia and Intensive Care, University of Szeged, Semmelweis u. 6, 6720, Szeged, Hungary
| | - Frank von Sonnenburg
- Department of Infectious Diseases and Tropical Medicine, University of Munich, Georgenstr. 5, 80799, Munich, Germany
| | - Arthur Lyons
- Clinical Research Department, Division of Virus Diseases, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Michael Joannidis
- Department of Internal Medicine, Division of Intensive Care and Emergency Medicine, Medical University of Innsbruck, Anichstr. 35, 6020, Innsbruck, Austria
| | - Heinz Burgmann
- Department of Internal Medicine I, Division of Infectious Diseases, Medical University of Vienna, Währinger Gürtel 18 - 20, 1090, Vienna, Austria
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Anton Klingler
- Assign Data Management and Biostatistics GmbH, Stadlweg 23, 6020, Innsbruck, Austria
| | - Romana Hochreiter
- Valneva Austria GmbH, Campus Vienna Biocenter 3, 1030, Vienna, Austria
| | | |
Collapse
|
19
|
Ramírez-Estrada S, Borgatta B, Rello J. Pseudomonas aeruginosa ventilator-associated pneumonia management. Infect Drug Resist 2016; 9:7-18. [PMID: 26855594 PMCID: PMC4725638 DOI: 10.2147/idr.s50669] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ventilator-associated pneumonia is the most common infection in intensive care unit patients associated with high morbidity rates and elevated economic costs; Pseudomonas aeruginosa is one of the most frequent bacteria linked with this entity, with a high attributable mortality despite adequate treatment that is increased in the presence of multiresistant strains, a situation that is becoming more common in intensive care units. In this manuscript, we review the current management of ventilator-associated pneumonia due to P. aeruginosa, the most recent antipseudomonal agents, and new adjunctive therapies that are shifting the way we treat these infections. We support early initiation of broad-spectrum antipseudomonal antibiotics in present, followed by culture-guided monotherapy de-escalation when susceptibilities are available. Future management should be directed at blocking virulence; the role of alternative strategies such as new antibiotics, nebulized treatments, and vaccines is promising.
Collapse
Affiliation(s)
| | - Bárbara Borgatta
- Critical Care Department, Vall d’Hebron University Hospital, Barcelona, Spain
- CRIPS, Vall d’Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Jordi Rello
- Department of Medicine, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedad Respiratoria – CIBERES, Madrid, Spain
| |
Collapse
|
20
|
Chatterjee M, Anju C, Biswas L, Anil Kumar V, Gopi Mohan C, Biswas R. Antibiotic resistance in Pseudomonas aeruginosa and alternative therapeutic options. Int J Med Microbiol 2016; 306:48-58. [DOI: 10.1016/j.ijmm.2015.11.004] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 01/05/2023] Open
|
21
|
Farsani HH, Rasooli I, Gargari SLM, Nazarian S, Astaneh SDA. Recombinant outer membrane protein F-B subunit of LT protein as a prophylactic measure against Pseudomonas aeruginosa burn infection in mice. World J Methodol 2015; 5:230-237. [PMID: 26713284 PMCID: PMC4686421 DOI: 10.5662/wjm.v5.i4.230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 08/05/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To study immunogenicity of outer membrane protein F (OprF) fused with B subunit of LT (LTB), against Pseudomonas aeruginosa (P. aeruginosa).
METHODS: The OprF, a major surface exposed outer membrane protein that is antigenically conserved in various strains of P. aeruginosa, is a promising immunogen against P. aeruginosa. In the present study recombinant OprF and OprF-LTB fusion gene was cloned, expressed and purified. BALB/c mice and rabbits were immunized using recombinant OprF and OprF-LTB and challenged at the burn site with P. aeruginosa lethal dose of 104 CFU. The protective efficacy of rabbit anti OprF IgG against P. aeruginosa burn infection was investigated by passive immunization.
RESULTS: It has been well established that the LTB is a powerful immunomodulator with strong adjuvant activity. LTB as a bacterial adjuvant enhanced immunogenicity of OprF and anti OprF IgG titer in serum was increased. Experimental findings showed significantly higher average survival rate in burned mice immunized with OprF-LTB than immunized with OprF or the control group. Rabbits anti OprF IgG brought about 75% survival of mice following challenge with P. aeruginosa. Post challenge hepatic and splenic tissues of mice group immunized with OprF-LTB had significantly lower bacterial load than those immunized with OprF or the control groups.
CONCLUSION: These results demonstrate that LTB-fused OprF might be a potential candidate protein for a prophylactic measure against P. aeruginosa in burn infection.
Collapse
|
22
|
Cui Z, Han D, Sun X, Zhang M, Feng X, Sun C, Gu J, Tong C, Lei L, Han W. Mannose-modified chitosan microspheres enhance OprF-OprI-mediated protection of mice against Pseudomonas aeruginosa infection via induction of mucosal immunity. Appl Microbiol Biotechnol 2014; 99:667-80. [PMID: 25381907 DOI: 10.1007/s00253-014-6147-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 01/22/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that localizes to and colonizes mucosal tissue. Thus, vaccines that elicit a strong mucosal response against P. aeruginosa should be superior to other vaccination strategies. In this study, to stimulate rapid and enhanced mucosal immune responses, mannose-modified chitosan microspheres loaded with the recombinant outer membrane protein OprF190-342-OprI21-83 (FI) (FI-MCS-MPs) of P. aeruginosa were developed as a potent subunit vaccine for mucosal delivery. FI-MCS-MPs were successfully obtained via the tripolyphosphate ionic crosslinking method. Confocal and immunohistochemical analyses indicated that FI-MCS-MPs exhibited the ability to bind the macrophage mannose receptor (MMR, CD206) in vitro and in vivo. After intranasal immunization of mice with FI-MCS-MPs, FI-specific humoral immune responses were detected, measured as local IgM antibody titers in lung tissue slurry; IgA antibody titers in nasal washes, bronchoalveolar lavage (BAL), and intestinal lavage; and systemic IgA and IgG antibody titers in serum. FI-MCS-MPs induced early and high mucosal and systemic humoral antibody responses comparable to those in the group vaccinated with unmodified mannose. High levels of IFN-γ and IL-4 in addition to T lymphocyte subsets induced a mixed Th1/Th2 response in mice immunized with FI-MCS-MPs, resulting in the establishment of cellular immunity. Additionally, when immunized mice were challenged with P. aeruginosa via the nasal cavity, FI-MCS-MPs demonstrated 75 % protective efficacy. Together, these data indicate that mannose-modified chitosan microspheres are a promising subunit delivery system for vaccines against P. aeruginosa infection.
Collapse
Affiliation(s)
- Ziyin Cui
- College of Veterinary Medicine, Jilin University, Xi'an Road 5333#, Changchun, 130062, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Westritschnig K, Hochreiter R, Wallner G, Firbas C, Schwameis M, Jilma B. A randomized, placebo-controlled phase I study assessing the safety and immunogenicity of a Pseudomonas aeruginosa hybrid outer membrane protein OprF/I vaccine (IC43) in healthy volunteers. Hum Vaccin Immunother 2013; 10:170-83. [PMID: 24064511 DOI: 10.4161/hv.26565] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION IC43 is a recombinant outer membrane protein-based vaccine against Pseudomonas aeruginosa (P. aeruginosa) consisting of OprF- and OprI- epitopes (Opr, outer membrane protein; OprF/I, OprF/OprI hybrid vaccine) with an N-terminal His 6 tag (Met-Ala-(His)6-OprF190-342-OprI21-83). OBJECTIVES The study aimed to confirm the optimal dose of IC43 in adults with regard to immunogenicity, safety, and tolerability after vaccination with three different dosages of IC43, compared with placebo, and to investigate a potential immune-enhancing effect of the adjuvant, aluminum hydroxide. Subjects were randomly allocated in a 1:1:1:1:1 ratio to one of five treatment groups: 50, 100, or 200 µg IC43 with adjuvant, 100 µg IC43 without adjuvant, or placebo (0.9% sodium chloride) and two intramuscular injections were given in the deltoid region 7 d apart. RESULTS The primary immunogenicity analysis of OprF/I-specific IgG antibody titers on day 14 demonstrated statistically significant differences among treatment groups (P<0.0001), with a significantly higher immune response detected in each IC43 treatment group compared with placebo. From day 0 to day 14, a ≥4-fold increase in OprF/I-specific immunoglobulin G (IgG) antibody titers were observed in>90% of subjects in all IC43 treatment groups in the per-protocol (PP) and intention-to-treat (ITT) populations; a ≥50-fold titer increase was observed in 42.6% subjects including all IC43 treatment groups. On day 90, OprF/I-specific IgGs started to decline in all IC43 treatment groups but remained significantly higher until 6 mo compared with placebo. Assessment of functional antibody induction by opsonophagocytic assay (OPA) followed a similar pattern compared with OprF/I-specific IgG kinetics. At day 14, a ≥2-fold increase in OPA titer was observed in 54.5% subjects within all IC43 treatment groups. An increase in antibody avidity index was observed after the second vaccination. At day 14, >96% of subjects in each IC43 treatment group had detectable OprF/I-specific IgG antibodies. Anti-histidine IgG antibody titers peaked on day 14 and were reduced on day 90 in all IC43 treatment groups. OprF/I-specific IgG secreted by antibody-secreting cell (ASC) was detected in all IC43 groups by B-cell ELIspot after the second vaccination and up to 6 mo. All vaccinations were safe and well tolerated up to the maximum cumulative dosage of 400 µg IC43. CONCLUSION IC43 doses equal to or greater than 50 µg were sufficient to induce a plateau of IgG antibody responses in healthy volunteers. Higher doses, whether adjuvanted or non-adjuvanted, were not more effective. METHODS In this phase I, randomized, placebo-controlled, observer-blinded, multicenter clinical trial, 163 healthy volunteers (18-65 y) were randomly assigned to five treatment groups (1:1:1:1:1). Three groups received IC43 with adjuvant: 50 µg (n=32), 100 µg (n=33), or 200 µg (n=33). One group received IC43 100 µg without adjuvant (n=32), and one group received placebo (0.9% sodium chloride) (n=33). Each subject received two intramuscular vaccinations, separated by a 7-d interval (days 0 and 7) (Fig. 1). Humoral immune response was assessed by measurement of outer membrane protein F/I (OprF/I)-specific antibodies determined by enzyme-linked immunosorbent assay (ELISA), anti-histidine antibodies determined by ELISA, and functional antibody activity determined by opsonophagocytic assay (OPA), up to 6 mo post-vaccination. Antibody avidity was measured on days 7 and 14 from samples that had detectable vaccine antibody-specific immunoglobulin G (IgG) antibody titers. At the Austrian site only, the B-cell ELIspot assay was used to determine specific ASC responses. Safety was assessed using adverse event monitoring and clinical laboratory tests. Local and systemic tolerability was recorded in a subject diary for 7 d after each vaccination and by investigators up to 6 mo post-vaccination.
Collapse
Affiliation(s)
| | | | | | - Christa Firbas
- Department of Clinical Pharmacology Medical University of Vienna; Währinger Gürtel; Vienna, Austria
| | - Michael Schwameis
- Department of Clinical Pharmacology Medical University of Vienna; Währinger Gürtel; Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology Medical University of Vienna; Währinger Gürtel; Vienna, Austria
| |
Collapse
|
24
|
Confer AW, Ayalew S. The OmpA family of proteins: Roles in bacterial pathogenesis and immunity. Vet Microbiol 2013; 163:207-22. [DOI: 10.1016/j.vetmic.2012.08.019] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/23/2012] [Indexed: 10/27/2022]
|
25
|
Kaloshin AA, Gatypova EV, Mikhailova NA. Obtaining recombinant forms of the outer membrane protein F of Pseudomonas aeruginosa and assessment of their immunogenic properties. APPL BIOCHEM MICRO+ 2011. [DOI: 10.1134/s0003683811080060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Sharma A, Krause A, Worgall S. Recent developments for Pseudomonas vaccines. HUMAN VACCINES 2011; 7:999-1011. [PMID: 21941090 DOI: 10.4161/hv.7.10.16369] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infections with Pseudomonas aeruginosa are a major health problem for immune-compromised patients and individuals with cystic fibrosis. A vaccine against: P. aeruginosa has long been sought after, but is so far not available. Several vaccine candidates have been assessed in experimental animals and humans, which include sub-cellular fractions, capsule components, purified and recombinant proteins. Unique characteristics of the host and the pathogen have complicated the vaccine development. This review summarizes the current state of vaccine development for this ubiquitous pathogen, in particular to provide mucosal immunity against infections of the respiratory tract in susceptible individuals with cystic fibrosis.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | | | | |
Collapse
|
27
|
Abstract
OBJECTIVE Multidrug-resistant Acinetobacter baumannii is associated with a wide spectrum of infectious diseases ranging from nosocomial, community-acquired infections to those acquired following war or natural disaster. Especially to military personnel with war wounds, Acinetobacter infection is a formidable threat. The treatment has become exceedingly difficult, not only because the bacterium can develop extensive antimicrobial resistance but because it also forms biofilms that are resistant to host defense and antimicrobial treatment. Such causative factors as biofilm formation and resistance are highly likely to lead to nonhealing wounds. This review, although focused on A baumannii infections, aims to summarize what is known about immunization protection against wound biofilm infections and to apply such understanding in exploring the unknown area of war-wound infection protection. DATA SOURCES AND STUDY SELECTION Publications were searched and selected through http://www.pubmed.gov by using the key words Acinetobacter baumannii, bacteria, war wounds, burn wounds, wound infections, biofilms, vaccines, and immunization. The literature selected was categorized according to the subheadings within this article. CONCLUSIONS It is imperative to develop such effective measures as active and passive immunization to control multidrug-resistant and tenacious A baumannii infections and to prevent nonhealing wounds. The authors' understanding in immunization against burn wound-related infections by the model bacteria will facilitate research progress in the poorly explored area of immunization against war-wound biofilm infections.
Collapse
|
28
|
Ding B, von Specht BU, Li Y. OprF/I-vaccinated sera inhibit binding of human interferon-gamma to Pseudomonas aeruginosa. Vaccine 2010; 28:4119-22. [PMID: 20433804 DOI: 10.1016/j.vaccine.2010.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 04/01/2010] [Accepted: 04/13/2010] [Indexed: 11/17/2022]
Abstract
The recombinant outer membrane protein OprF/I has been demonstrated in previous studies to protect against Pseudomonas aeruginosa infection through a mechanism of enhanced antibody-mediated opsonophagocytosis. Recent evidence indicates that P. aeruginosa enhances its virulence phenotype as a consequence of binding to human IFN-gamma through an outer membrane protein, OprF. In this study, we demonstrate that a single boost injection of OprF/I vaccine elicited a strong OprF/I-specific antibody response in individuals who were previously vaccinated with OprF/I in a clinical trial. The OprF/I-vaccinated sera inhibit P. aeruginosa binding to IFN-gamma, suggesting an alternative mechanism by which the OprF/I vaccine confers protection against P. aeruginosa infection.
Collapse
Affiliation(s)
- Bin Ding
- Centre for Clinical Research, University of Freiburg, 79106 Freiburg, Germany
| | | | | |
Collapse
|
29
|
Hare NJ, Cordwell SJ. Proteomics of bacterial pathogens: Pseudomonas aeruginosa infections in cystic fibrosis - a case study. Proteomics Clin Appl 2010; 4:228-48. [PMID: 21137046 DOI: 10.1002/prca.200900144] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Revised: 09/09/2009] [Accepted: 09/30/2009] [Indexed: 12/20/2022]
Abstract
Technology development in the high throughput sciences of genomics, transcriptomics and proteomics, has been driven by bacteriological research. These organisms are excellent models for testing new methodology due to their comparatively small genome size, the relative ease of culturing large amounts of material, and the inherent biomedical, environmental and biotechnological interest in their underlying biology. Techniques developed in prokaryotes have since become applicable to higher organisms and human disease, opening vast research opportunities for understanding complex molecular processes. Pseudomonas aeruginosa is an excellent example of a microbe with fascinating properties suitable for stretching the boundaries of technology, and with underlying biology that remains poorly understood. P. aeruginosa is an opportunistic pathogen in humans and contains one of the largest genetic capabilities for a single-celled organism (approximately 5500 genes), which allows it to encode a wide variety of surface-associated and secreted virulence factors, as well as adapt to harsh environments, forming resistance to an array of antibacterial agents. While it is a major threat as a nosocomial pathogen, and particularly in the immunocompromised, it is also the most significant cause of mortality in patients suffering from the genetic disorder, cystic fibrosis. This review examines the role of proteomics in gaining a better understanding of the molecular basis of P. aeruginosa infection and persistence in the lungs of cystic fibrosis patients.
Collapse
Affiliation(s)
- Nathan J Hare
- School of Molecular and Microbial Biosciences, The University of Sydney, Sydney, Australia
| | | |
Collapse
|
30
|
Bumann D, Behre C, Behre K, Herz S, Gewecke B, Gessner JE, von Specht BU, Baumann U. Systemic, nasal and oral live vaccines against Pseudomonas aeruginosa: A clinical trial of immunogenicity in lower airways of human volunteers. Vaccine 2010; 28:707-13. [DOI: 10.1016/j.vaccine.2009.10.080] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2008] [Revised: 09/30/2009] [Accepted: 10/14/2009] [Indexed: 10/20/2022]
|
31
|
Kaufmann SHE, Meinke AL, von Gabain A. [Novel vaccination concepts on the basis of modern insights into immunology]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2009; 52:1069-82. [PMID: 19838650 DOI: 10.1007/s00103-009-0951-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Since their introduction more than 200 years ago, vaccines have prevented millions of deaths caused by infectious diseases. This progress was possible because these vaccines protect through antibodies, which are relatively easily stimulated. In the meantime, we understand that diseases such as AIDS, tuberculosis, malaria and hepatitis C cannot be tackled by these conventional approaches. Recent insights into immunology provide the basis for the development of custom-tailored vaccines to successfully combat these threatening infections. These new generation vaccines comprise components that modulate the mediators of immunity (B cells, T cells, antigen-presenting cells and cytokines) in such a way that the best possible immune response develops. Alternative application methods offer the possibility to further improve the immune response. Thus, hope remains that the remarkable increase in knowledge in the areas of immunology and infectious disease research will help to successfully control infectious diseases.
Collapse
Affiliation(s)
- S H E Kaufmann
- Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117, Berlin.
| | | | | |
Collapse
|
32
|
Weimer ET, Ervin SE, Wozniak DJ, Mizel SB. Immunization of young African green monkeys with OprF epitope 8-OprI-type A- and B-flagellin fusion proteins promotes the production of protective antibodies against nonmucoid Pseudomonas aeruginosa. Vaccine 2009; 27:6762-9. [PMID: 19744586 DOI: 10.1016/j.vaccine.2009.08.080] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 08/18/2009] [Accepted: 08/22/2009] [Indexed: 02/01/2023]
Abstract
There is currently no approved vaccine against Pseudomonas aeruginosa, the major cause of morbidity and mortality in cystic fibrosis (CF) patients and a major pathogen in ventilated and burn patients. In a previous study, we demonstrated the immunization of mice with OprF(311-341)-OprI-type A- and B-flagellin fusion proteins dramatically enhanced clearance of nonmucoid P. aeruginosa. The goal of the current study was to evaluate the ability of OprF(311-341)-OprI-flagellins to elicit the production of protective IgG in young (4-6 months old) African green monkeys. Intramuscular immunization of African green monkeys with 1, 3, 10, or 30mug of OprF(311-341)-OprI-flagellins generated robust antigen-specific IgG responses. In addition, immunization with OprF(311-341)-OprI-flagellins elicited high-affinity anti-flagellins, OprI, and OprF IgG that individually promoted extensive deposition of complement component C3 on P. aeruginosa and synergized to facilitate maximal C3 deposition. Passive immunization of mice with plasma from OprF(311-341)-OprI-flagellins immunized monkeys significantly reduced lung bacterial burden three days post-challenge compared to mice that received pre-immunization plasma. Based on our results, OprF(311-341)-OprI-A- and B-flagellin fusion proteins are highly effective in mice and nonhuman primates and thus merit additional development as a potential vaccine for use in humans.
Collapse
Affiliation(s)
- Eric T Weimer
- Department of Microbiology & Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
33
|
A fusion protein vaccine containing OprF epitope 8, OprI, and type A and B flagellins promotes enhanced clearance of nonmucoid Pseudomonas aeruginosa. Infect Immun 2009; 77:2356-66. [PMID: 19349426 DOI: 10.1128/iai.00054-09] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although chronic Pseudomonas aeruginosa infection is the major cause of morbidity and mortality in cystic fibrosis (CF) patients, there is no approved vaccine for human use against P. aeruginosa. The goal of this study was to establish whether a multivalent vaccine containing P. aeruginosa type A and B flagellins as well as the outer membrane proteins OprF and OprI would promote enhanced clearance of P. aeruginosa. Intramuscular immunization with flagellins and OprI (separate) or OprI-flagellin fusion proteins generated significant antiflagellin immunoglobulin G (IgG) responses. However, only the fusions of OprI with type A and type B flagellins generated OprI-specific IgG. Immunization with a combination of OprF epitope 8 (OprF(311-341)), OprI, and flagellins elicited high-affinity IgG antibodies specific to flagellins, OprI, and OprF that individually promoted extensive deposition of C3 on P. aeruginosa. Although these antibodies exhibited potent antibody-dependent complement-mediated killing of nonmucoid bacteria, they were significantly less effective with mucoid isolates. Mice immunized with the OprF(311-341)-OprI-flagellin fusion had a significantly lower bacterial burden three days postchallenge and cleared the infection significantly faster than control mice. In addition, mice immunized with the OprF(311-341)-OprI-flagellin fusion had significantly less inflammation and lung damage throughout the infection than OprF-OprI-immunized mice. Based on our results, OprF(311-341)-OprI-flagellin fusion proteins have substantial potential as components of a vaccine against nonmucoid P. aeruginosa, which appears to be the phenotype of the bacterium that initially colonizes CF patients.
Collapse
|
34
|
Immune responses in the airways by nasal vaccination with systemic boosting against Pseudomonas aeruginosa in chronic lung disease. Vaccine 2009; 27:2755-9. [PMID: 19366571 DOI: 10.1016/j.vaccine.2009.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 02/26/2009] [Accepted: 03/04/2009] [Indexed: 02/01/2023]
Abstract
RATIONALE Pneumonia caused by Pseudomonas (P.) aeruginosa is a leading cause of morbidity and mortality in patients with chronic lung diseases. Systemic vaccination in patients with cystic fibrosis has been only successful in part. Mucosal vaccination could lead to enhanced airway immunogenicity. Pathogen specific secretory IgA antibodies could prevent bacterial invasion into the lung mucosa. OBJECTIVES A phase 1-2 mucosal vaccination trial with an intranasal P. aeruginosa vaccine was performed. METHODS 12 patients with chronic lung diseases (8 COPD, 2 cystic fibrosis, 1 bronchiectasis, 1 histiocytosis X) were vaccinated three times intranasally followed by a systemic booster vaccination with a recombinant hybrid protein encompassing the main protective epitopes of two outer membrane proteins of P. aeruginosa. Mucosal and systemic antibody responses were measured after boosting and after a half-year follow-up compared to a representative control cohort. MEASUREMENTS Specific IgG and IgA antibodies in the patient's sera, saliva and sputum were determined by enzyme-linked immunosorbent assay (ELISA) and IgG subclass distributions were defined with monoclonal mouse antibodies. RESULTS Both forms of vaccination were well tolerated. Significant elevated IgA and IgG antibodies could be measured in sputum, saliva and in the sera of 11/12 patients. CONCLUSIONS Mucosal vaccination followed by systemic boost with an outer membrane protein vaccine against P. aeruginosa leads to airway immunogenicity against the pathogen. Further clinical trials should elucidate the protective efficacy of this vaccination method.
Collapse
|
35
|
A recombinant hybrid outer membrane protein OprF–OprH from Pseudomonas aeruginosa and its immunogenicity. J Biotechnol 2008. [DOI: 10.1016/j.jbiotec.2008.07.430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Vaccines and immunotherapy against Pseudomonas aeruginosa. Vaccine 2008; 26:1011-24. [PMID: 18242792 DOI: 10.1016/j.vaccine.2007.12.007] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 11/28/2007] [Accepted: 12/05/2007] [Indexed: 11/21/2022]
|
37
|
Protective immunity to pseudomonas aeruginosa induced with a capsid-modified adenovirus expressing P. aeruginosa OprF. J Virol 2007; 81:13801-8. [PMID: 17942539 DOI: 10.1128/jvi.01246-07] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study focuses on the development of a new clinical vaccine candidate (AdOprF.RGD.Epi8) against Pseudomonas aeruginosa using an E1(-) E3(-) adenovirus (Ad) vector expressing OprF (AdOprF.RGD.Epi8) and modifications of the Ad genome providing two capsid changes: (i) modification of the Ad hexon gene to incorporate an immune-dominant OprF epitope (Epi8) into loop 1 of the hexon, enabling repeat administration to boost the anti-OprF immune response, and (ii) modification of the fiber gene to incorporate an integrin-binding RGD sequence to enhance gene delivery to antigen-presenting cells. Western analysis confirmed that AdOprF.RGD.Epi8 expresses OprF, contains Epi8 in the hexon protein, and enhances gene transfer to dendritic cells compared to AdOprF, a comparable Ad vector expressing OprF with an unmodified capsid. Intramuscular immunization of C57BL/6 mice with AdOprF.RGD.Epi8 resulted in the generation of anti-OprF antibodies at comparable levels to those induced following immunization with AdOprF, but immunization with AdOprF.RGD.Epi8 was associated with increased CD4 and CD8 gamma interferon T-cell responses against OprF as well as increased survival against lethal pulmonary challenge with agar-encapsulated P. aeruginosa. Importantly, repeat administration of AdOprF.RGD.Epi8 resulted in boosting of the humoral anti-OprF response as well as increased protection, whereas no boosting could be achieved with repeat administration of AdOprF. This suggests that the capsid-modified AdOprF.RGD.Epi8 vector is a more effective immunogen compared to a comparable wild-type Ad capsid, making it a good candidate for an anti-P. aeruginosa vaccine.
Collapse
|
38
|
Baumann U, Göcke K, Gewecke B, Freihorst J, von Specht BU. Assessment of pulmonary antibodies with induced sputum and bronchoalveolar lavage induced by nasal vaccination against Pseudomonas aeruginosa: a clinical phase I/II study. Respir Res 2007; 8:57. [PMID: 17683588 PMCID: PMC1973076 DOI: 10.1186/1465-9921-8-57] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Accepted: 08/05/2007] [Indexed: 02/06/2023] Open
Abstract
Background Vaccination against Pseudomonas aeruginosa is a desirable albeit challenging strategy for prevention of airway infection in patients with cystic fibrosis. We assessed the immunogenicity of a nasal vaccine based on the outer membrane proteins F and I from Pseudomonas aeruginosa in the lower airways in a phase I/II clinical trial. Methods N = 12 healthy volunteers received 2 nasal vaccinations with an OprF-OprI gel as a primary and a systemic (n = 6) or a nasal booster vaccination (n = 6). Antibodies were assessed in induced sputum (IS), bronchoalveolar lavage (BAL), and in serum. Results OprF-OprI-specific IgG and IgA antibodies were found in both BAL and IS at comparable rates, but differed in the predominant isotype. IgA antibodies in IS did not correlate to the respective serum levels. Pulmonary antibodies were detectable in all vaccinees even 1 year after the vaccination. The systemic booster group had higher IgG levels in serum. However, the nasal booster group had the better long-term response with bronchial antibodies of both isotypes. Conclusion The nasal OprF-OprI-vaccine induces a lasting antibody response at both, systemic and airway mucosal site. IS is a feasible method to non-invasively assess bronchial antibodies. A further optimization of the vaccination schedule is warranted.
Collapse
Affiliation(s)
- Ulrich Baumann
- Department of Paediatric Pulmonology and Neonatalogy, Hanover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Kerstin Göcke
- Department of Paediatric Pulmonology and Neonatalogy, Hanover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Britta Gewecke
- Department of Paediatric Pulmonology and Neonatalogy, Hanover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Joachim Freihorst
- Department of Paediatric Pulmonology and Neonatalogy, Hanover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
- Pediatric Hospital, Ostalb-Klinikum, 73430 Aalen, Germany
| | - Bernd Ulrich von Specht
- Centre for Clinical Research, Freiburg University, Breisacherstr.66, 79106 Freiburg, Germany
| |
Collapse
|
39
|
Sedlak-Weinstein E, Cripps AW, Kyd JM, Foxwell AR. Pseudomonas aeruginosa: the potential to immunise against infection. Expert Opin Biol Ther 2005; 5:967-82. [PMID: 16018741 DOI: 10.1517/14712598.5.7.967] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pseudomonas aeruginosa remains a serious pathogen for specific cohorts of patients where chronic infection is a poor prognostic indicator, such as those with cystic fibrosis, burn wounds or those who are immunocompromised. Significant disease burden is associated with a diverse spectrum of both nosocomial and community-acquired infections. To date, vaccines against P. aeruginosa have shown limited and often conflicting efficacy data, especially against heterologous strains, which are increasingly identified as co-colonisers of biofilms. While few studies have gone beyond Phase II clinical trials, a particular concern is the ability of P. aeruginosa to evade the immune system while provoking an immune response that contributes to the destructive nature of infection. Therefore, vaccine development needs to focus on preventing attachment and colonisation, as well as preventing conversion to a mucoid phenotype that is characteristic of the chronic condition that promotes pathology.
Collapse
Affiliation(s)
- E Sedlak-Weinstein
- Griffith University Gold Coast Campus, School of Medicine, PMB 50, Gold Coast Mail Centre, Queensland 9726, Australia
| | | | | | | |
Collapse
|
40
|
Gillis RJ, White KG, Choi KH, Wagner VE, Schweizer HP, Iglewski BH. Molecular basis of azithromycin-resistant Pseudomonas aeruginosa biofilms. Antimicrob Agents Chemother 2005; 49:3858-67. [PMID: 16127063 PMCID: PMC1195439 DOI: 10.1128/aac.49.9.3858-3867.2005] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Pseudomonas aeruginosa biofilms are extremely recalcitrant to antibiotic treatment. Treatment of cystic fibrosis patients with azithromycin (AZM) has shown promise. We used DNA microarrays to identify differentially expressed transcripts in developing P. aeruginosa biofilms exposed to 2 mug/ml AZM. We report that transcripts for multiple restriction-nodulation-cell division (RND) efflux pumps, known to be involved in planktonic antibiotic resistance, and transcripts involved in type III secretion were upregulated in the resistant biofilms that developed in the presence of AZM. Interestingly, the MexAB-OprM and MexCD-OprJ efflux pumps, but not type III secretion, appear to be integral to biofilm formation in the presence of AZM, as evidenced by the fact that a mutant deleted in both mexAB-oprM and mexCD-oprJ was unable to form a biofilm in the presence of AZM. A mutant deleted in type III secretion was still able to form biofilms in the presence of drug. Furthermore, single mexAB-oprM- and mexCD-oprJ-null mutants were able to form a biofilm in the presence of drug, indicating that either of the pumps can confer resistance to AZM during biofilm development. In contrast to planktonically grown cells, where no mexC expression was detectable regardless of the presence of AZM, biofilms exhibited induction of mexC expression from the outset of their formation, but only in the presence of AZM. mexA, which is constitutively expressed in planktonic cells, was uniformly expressed in biofilms regardless of the presence of AZM. These data indicate that the MexCD-OprJ pump acts as a biofilm-specific mechanism for AZM resistance.
Collapse
Affiliation(s)
- Richard J Gillis
- Department of Microbiology and Immunology, Box 672, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
41
|
Wagner VE, Gillis RJ, Iglewski BH. Transcriptome analysis of quorum-sensing regulation and virulence factor expression in Pseudomonas aeruginosa. Vaccine 2005; 22 Suppl 1:S15-20. [PMID: 15576196 DOI: 10.1016/j.vaccine.2004.08.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The opportunistic pathogen Pseudomonas aeruginosa possesses two well-studied quorum-sensing (QS) systems (las and rhl) that are important in the production of virulence factors, antibiotic sensitivity, and biofilm development. High-density oligonucleotide microarrays were used to further characterize the las QS system and to investigate the effect of environment (planktonic or biofilm mode of growth, absence or presence of oxygen) and nutritional conditions on detection of transcripts encoding QS-regulated virulence factors. Transcriptome results indicate that the QS system is far more complex than previously proposed. Interestingly, we found that many QS-regulated genes encoding virulence products were expressed in all conditions investigated.
Collapse
Affiliation(s)
- Victoria E Wagner
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Box 672, Rochester, NY 14642, USA
| | | | | |
Collapse
|
42
|
Baumann U, Mansouri E, von Specht BU. Recombinant OprF-OprI as a vaccine against Pseudomonas aeruginosa infections. Vaccine 2004; 22:840-7. [PMID: 15040936 DOI: 10.1016/j.vaccine.2003.11.029] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A vaccine against Pseudomonas aeruginosa based on recombinant outer membranes has been developed. After intramuscularly injecting into patients with severe burns, antibodies against P. aeruginosa were induced. Vaccination was well tolerated. Intranasal application of the vaccine into volunteers, induced specific s-IgA antibodies. We conclude that the newly developed vaccine may be suitable for protection of the main risk groups of P. aeruginosa infections. In particular, for the protection of burn patients and patients with cystic fibrosis.
Collapse
Affiliation(s)
- U Baumann
- Medizinische Hochschule Hannover, Abteilung Pädiatrische Pneumonologie und Neonatalogie, 30623 Hannover, Germany
| | | | | |
Collapse
|
43
|
Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2004; 13:49-64. [PMID: 14971123 DOI: 10.1002/pds.914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|