15-Vaccination against respiratory Pseudomonas aeruginosa infection - Enfermagem (2024)

Prévia do material em texto

Full Terms & Conditions of access and use can be found athttps://www.tandfonline.com/action/journalInformation?journalCode=khvi20Human Vaccines & ImmunotherapeuticsISSN: 2164-5515 (Print) 2164-554X (Online) Journal homepage: www.tandfonline.com/journals/khvi20Vaccination against respiratory Pseudomonasaeruginosa infectionKeith Grimwood, Jennelle M Kyd, Suzzanne J Owen, Helen M Massa & AllanW CrippsTo cite this article: Keith Grimwood, Jennelle M Kyd, Suzzanne J Owen, Helen M Massa & AllanW Cripps (2015) Vaccination against respiratory Pseudomonas aeruginosa infection, HumanVaccines & Immunotherapeutics, 11:1, 14-20, DOI: 10.4161/hv.34296To link to this article: https://doi.org/10.4161/hv.34296Copyright © 2015 Landes BiosciencePublished online: 01 Nov 2014.Submit your article to this journal Article views: 3479View related articles View Crossmark dataCiting articles: 13 View citing articles https://www.tandfonline.com/action/journalInformation?journalCode=khvi20https://www.tandfonline.com/journals/khvi20?src=pdfhttps://www.tandfonline.com/action/showCitFormats?doi=10.4161/hv.34296https://doi.org/10.4161/hv.34296https://www.tandfonline.com/action/authorSubmission?journalCode=khvi20&show=instructions&src=pdfhttps://www.tandfonline.com/action/authorSubmission?journalCode=khvi20&show=instructions&src=pdfhttps://www.tandfonline.com/doi/mlt/10.4161/hv.34296?src=pdfhttps://www.tandfonline.com/doi/mlt/10.4161/hv.34296?src=pdfhttp://crossmark.crossref.org/dialog/?doi=10.4161/hv.34296&domain=pdf&date_stamp=01 Nov 2014http://crossmark.crossref.org/dialog/?doi=10.4161/hv.34296&domain=pdf&date_stamp=01 Nov 2014https://www.tandfonline.com/doi/citedby/10.4161/hv.34296?src=pdfhttps://www.tandfonline.com/doi/citedby/10.4161/hv.34296?src=pdf©2014 Landes Bioscience. Do not distribute.Human Vaccines & Immunotherapeutics 11:1, 14–20; January 2015; © 2015 Landes Bioscience ReVIew14 Human Vaccines & Immunotherapeutics Volume 11 Issue 1ReVIewIntroductionPseudomonas aeruginosa is an oxidase positive, glucose non-fermenting Gram negative bacillus that normally inhabits soil and aquatic environments.1 Its large genome, adaptive regula-tory systems, metabolic versatility and high intrinsic antibiotic resistance, facilitates its survival in a diverse range of habitats.2 P. aeruginosa is also an opportunistic human pathogen, causing acute life-threatening infections in patients with a damaged epi-thelial barrier (e.g., burns, intravascular and urinary catheters, traumatic and surgical wound sites and endotracheal tubes) or impaired immune function (e.g., cancer, human immunodefi-ciency virus infection, bone marrow, and organ transplantation) and resulting in bacteraemia, urinary tract infection or pneu-monia.3 P. aeruginosa can also cause persistent infections within the lower respiratory tracts of patients with chronic pulmonary disorders. Chronic P. aeruginosa infection develops as a result of damaged or abnormal airway epithelium and compromised local pulmonary clearance mechanisms, and includes patients with cystic fibrosis (CF),4 non-CF bronchiectasis (nCFB),5 and chronic obstructive pulmonary disease (COPD).6 The organism possesses an impressive array of virulence factors that enable it to cause acute infections, and the metabolic versatility to facilitate its ongoing persistence within the lung microenvironment, both of which have been reviewed extensively elsewhere.7,8 This brief review examines the current status of the development of a vac-cine against P. aeruginosa.Why is a vaccine needed?The burden of disease caused by P. aeruginosa is substantial. In the United States, P. aeruginosa is the second most common pathogen causing acute healthcare-associated pneumonia in the critically ill, elderly and immunocompromised, while worldwide *Correspondence to: Allan W Cripps; Email: allan.cripps@griffith.edu.auSubmitted: 06/30/2014; Accepted: 08/07/2014; Published Online: 08/05/2014http://dx.doi.org/10.4161/hv.34296Vaccination against respiratory Pseudomonas aeruginosa infectionKeith Grimwood1,2,3, Jennelle M Kyd4, Suzzanne J Owen2,5, Helen M Massa2,5, and Allan w Cripps1,2,*1School of Medicine; Griffith University; Gold Coast, Queensland Australia; 2Griffith Health Institute; Griffith University; Gold Coast, Queensland Australia; 3Departments of Paediatrics and Infectious Diseases; Gold Coast University Hospital; Gold Coast, Queensland Australia; 4Swinburne University of Technology; Hawthorn, Victoria Australia; 5School of Medical Science; Griffith University; Gold Coast, Queensland AustraliaKeywords: P. aeruginosa, vaccine, oral immunisation, mucosal immunity, cystic fibrosis, chronic obstructive pulmonary disease, bronchiectasisAbbreviations: Ad, amidase; AHL, acyl hom*oserine lactones; BHL, butyryl L-hom*oserine lactone; BSA, bovine serum albumin; CF, cystic fibrosis; CFU, colony forming units; COPD, chronic obstructive pulmonary disease; HKPa, heat killed P. aeruginosa; IFA, incomplete Freund’s adjuvant; IT, intra-tracheal; KatA, catalase A; LPS, lipopolysaccharide; nCFB, non-cystic fibrosis bronchiectasis; NTHi, non-typeable Haemophilus influenzae; OdDHL, N-3-(oxododecanoyl)-L-hom*oserine lactone; OprF-I, Recombinant outer membrane fusion protein I of P. aeruginosa; PBS, phosphate buffered saline; QSSM, quorum sensing signalling moleculesRespiratory infections caused by Pseudomonas aeruginosa are a major clinical problem globally, particularly for patients with chronic pulmonary disorders, such as those with cystic fibrosis (CF), non-CF bronchiectasis (nCFB) and severe chronic obstructive pulmonary disease (COPD). In addition, critically ill and immunocompromised patients are also at significant risk of P. aeruginosa infection. For almost half a century, research efforts have focused toward development of a vaccine against infections caused by P. aeruginosa, but a licensed vaccine is not yet available. Significant advances in identifying potential vac-cine antigens have been made. Immunisations via both the mucosal and systemic routes have been trialled in animal mod-els and their effectiveness in clearing acute infections demon-strated. The challenge for translation of this research to human applications remains, since P. aeruginosa infections in the human respiratory tract can present both as an acute or chronic infection. In addition, immunisation prior to infection may not be possible for many patients with CF, nCFB or COPD. Therefore, development of a therapeutic vaccine provides an alternative approach for treatment of chronic infection. Preliminary ani-mal and human studies suggest that mucosal immunisation may be effective as a therapeutic vaccine against P. aeruginosa respiratory infections. Nevertheless, more research is needed to improve our understanding of the basic biology of P. aeruginosa and the mechanisms needed to upregulate the induction of host immune pathways to prevent infection. Recognition of variabil-ity in the host immune responses for a range of patient health conditions at risk from P. aeruginosa infection is also required to support development of a successful vaccine delivery strategy and vaccine. Activation of mucosal immune responses may pro-vide improved efficacy of vaccination for P. aeruginosa during both acute exacerbations and chronic infection.©2014 Landes Bioscience. Do not distribute.www.landesbioscience.com Human Vaccines & Immunotherapeutics 15it is second only to Staphylococcus aureus as a cause of infections within intensive care units.9,10 Treatment is complicated by the organism’s resistance to multiple antibiotics and its capacity to form aggregates and biofilms on mucosal membranes and medi-cal device surfaces, further increasing resistance to antibiotic action.11,12 For example, ventilator-associated P. aeruginosa pneu-monia, in particular has a highattributable case facility rate.13 A Canadian study reported the annual incidence of P. aeruginosa bacteraemia as 3.6/100,000 population, with only one in five cases being community-acquired.14 The overall mortality rate in this population-based study was 29% and the study identified important risk factors for bacteraemia, including that its fre-quency increased with advancing age, underlying chronic disease and male gender.The establishment of persistent P. aeruginosa infection in those with chronic pulmonary disorders is also of considerable importance. By adulthood, more than 70% of patients with CF will have a chronic P. aeruginosa infection, which leads to acceler-ated pulmonary decline, reduced quality of life and poorer sur-vival.15,16 Similarly, the organism also acts as a marker of severe disease and poor prognosis in the 10–30% of infected adult patients with nCFB and COPD.17,18 Chronic pulmonary condi-tions are the fourth leading cause of death globally, with more than four million people dying prematurely each year, and more than 200 million people diagnosed with COPD worldwide.19 Consequently, both acute and chronic P. aeruginosa infections are associated with significant morbidity, increased mortality and considerable cost to the health system and the community.Despite advances in managing the critically ill, mortality from acute P. aeruginosa infections, such as sepsis and pneumonia, remains high,20 while chronic infection in those with underlying pulmonary disorders and impaired airway clearance mechanisms is notoriously difficult to eradicate. The organism’s chromosom-ally encoded intrinsic resistance to antibiotics, its capacity to acquire resistance from mutations and horizontal gene transfer, and its capability to form self-protective biofilms, deep within the lower airways, are contributing factors to its persistence.11,21Overall, the development of effective therapeutic regimes for those at risk of infection from this opportunistic pathogen remains challenging,22 due to its multiple virulence mechanisms and the wide variation in the underlying causes of susceptibility to infection exhibited by various patient groups. Clearly, all indi-viduals vulnerable to P. aeruginosa infection could potentially benefit from a P. aeruginosa vaccine.Despite a substantial research effort over the past 50 years, a vaccine licensed for clinical use has not yet been delivered and several challenges remain to be addressed. For example, use of a prophylactic vaccine requires immunisation prior to P. aeru-ginosa colonisation of the airways and thus may be limited to the uninfected elderly and CF or nCFB patients who are already known to be at increased risk of P. aeruginosa infection. After P. aeruginosa colonisation and establishment of infection, a thera-peutic vaccine is required in order to clear the organism. It is cur-rently not known whether the same formulation will be effective for both scenarios. Furthermore, treatment of acute infections in patients resulting from mechanical ventilation, central vascular catheterization, chemotherapy-induced mucositis and neutropae-nia, or from severe trauma, including burns, are increasingly reli-ant upon a passive immunotherapeutic approach. These patients are usually critically ill, have impaired systemic immunity, and their responses to active immunisation may be delayed or diminished.23 In contrast, patients with chronic endobronchial infections may have impaired local immune responses and poor airway clearance, but retain relatively intact systemic immune responses.24The lung microenvironment in patients with CF provides an even greater challenge for successful vaccine development, due to its increased susceptibility to bacterial colonisation and acute exacerbation episodes during chronic infection. The lower air-ways of CF patients produce viscous mucus, while mucociliary clearance is decreased, effectively reducing the opportunity for pathogen clearance.25 Innate immunity within the CF lung is impaired (reduced pH and defensins),26 airway phagocytic func-tion is abnormal and there is apparent mucosal immune dysregu-lation.24 In addition to the opportunity for infection provided by the host’s compromised homeostasis, P. aeruginosa has an impressive armoury of weapons to facilitate successful coloni-sation and infection. For example, the organism produces sev-eral virulence factors that are able to impair or modulate local defenses (quorum sensing signaling molecules (QSSM), prote-ases and pyocyanin).27 Furthermore, using mutations and genetic rearrangements, it is also able to downregulate the expression of highly immunogenic virulence factors (O-antigen, type III secre-tion systems, flagella)28,29 and upregulate other factors, including pyoverdine,30 to produce aggregates and biofilms.7,12,28,29P. aeruginosa vaccine antigens and vaccine immune responsesSeveral P. aeruginosa antigens have been identified as poten-tial vaccine candidates, which, to date include lipopolysaccharide (LPS) O-antigen, polysaccharides, polysaccharide-protein conju-gates, outer membrane proteins F and I, the type III secretion system component PcrV, flagella, pili, attenuated P. aeruginosa S.enterica SL326, DNA and whole killed cells,31,32 in addition to a number of non-integral outer membrane protein candidate antigens.33It is increasingly clear that development of strong opsono-phagocytic antibody responses following immunisation is insuf-ficient for successful vaccine development and that stimulation of T cell responses, including IFN-γ, IL-17 and GM-CSF secre-tion, is essential. These observations highlight the possibility that different vaccine formulations may need to be considered for defined infections and patient populations.34,35 Furthermore, a Th17 T cell response may provide evidence of a potential mecha-nism for enhanced endobronchial clearance of non-typeable Hemophilus influenzae (NTHi) in COPD patients following oral immunisation with a whole killed cell NTHi oral vaccine.36 The proposed mechanism of this response is driven from oral vaccine stimulation of Peyer’s patches, followed by Th17 effector T cells circulating to the lungs via a mucosal immune network. Clearly, ©2014 Landes Bioscience. Do not distribute.16 Human Vaccines & Immunotherapeutics Volume 11 Issue 1significantly more basic research is required to determine the type of immune responses needed to provide or enhance immu-noprotection against P. aeruginosa infections and to encompass the diverse clinical presentations of infection caused by this organism, before an effective vaccine can be developed.Vaccine outcomes to dateCurrently, a licensed vaccine is not yet available against P. aeruginosa, despite many promising animal and preliminary clinical studies in humans. An excellent expert review by Priebe and Goldberg (2014) has recently addressed the “state-of-the-art” in vaccinology for P. aeruginosa and is recommended to the reader.35 That review, however, did not report on research scaf-folding the potential development of a mucosal vaccine against P. aeruginosa. For example, we have shown that a whole killed cell P. aeruginosa vaccine demonstrated considerable promise in an animal model of acute lung infection.37 These studies have been successfully expanded to demonstrate that mucosal immunisa-tion with cytosolic proteins can also lead to enhanced clearance from the lungs of animals acutely infected with P. aeruginosa.33 Two of these antigens, catalase A (KatA) and amidase (Ad) were further studied.P. aeruginosa possesses two heme-containing catalases,38 with KatA being the principal catalase, which provides the first line of defense against osmotic stress and H2O2 attack by phagocytes.39 KatA, the ‘isoenzyme A’ form, is located in the cytoplasm and periplasm and appears to also locate on the bacterialsurface.40 In the absence of KatA expression, biofilms are more easily killed by H2O241 and QSSMs mediate a significant component of biofilm resistance to H2O2.42 In contrast, Ad is an enzyme found in the periplasm and has no known relationship to bacte-rial virulence. The P. aeruginosa amidase is a 6x38-kDa enzyme that catalyzes the hydrolysis of a small range of short aliphatic amides and belongs to the nitrilases, a 13-branch superfamily of thiol enzymes involved in natural product biosynthesis and post-translational modification.43 Parenteral immunisation studies have demonstrated that KatA appears to be just as effective in mice as the hom*ologous killed P. aeruginosa whole cell vaccine, and slightly better than the leading vaccine candidate OprF-OprI (provided by Dr von Specht) (Fig. 1). We have also demonstrated that KatA com-bined with Ad is protective in a chronic lung infection model (Figs. 2 and 3), significantly reducing the number of bacteria recovered 4 h after acute challenge with P. aeruginosa. In this model, there was visually much less epithelial thickening in the bronchiole wall, less cellular infiltration, less alveolar wall damage and less lung consolidation in immunised animals compared with non-immunised controls (Fig. 4). Indeed, the extent of epithe-lial damage 4 h after bacterial challenge was still evident 24 h after administration of the bacterial challenge, with the alveoli exhibiting significant damage and bleeding in non-immunised animals. The presence of clear mucus in some smaller bronchi-oles provides evidence of localized mucosal responses within the KatA/Ad immunised animals. Most importantly, this study demonstrated that, parenteral immunisation was effective against an acute exacerbation of a chronic lung infection in mice when immunisation occurred after the infection was firmly established (Fig. 3). In most chronic pulmonary disorders, individuals suf-fer episodes of acute exacerbations. Thus, in this model, both the day 7 and day 35 P. aeruginosa challenges represent acute Figure 1. Clearance of P. aeruginosa from the acutely infected lung mouse model, 4 h post challenge with live bacteria. Mice (Balb/c) were parenterally immunised with 10 µg antigen (KatA or OprF-I) or 106 colony forming units (cfu) heat killed Pa in Incomplete Freund’s adju-vant (as indicated) on days 0 and 14. Acute P. aeruginosa lung infection involved direct intra-tracheal (IT) inoculation of live P. aeruginosa (106cfu) into the lungs while briefly sedated with alfaxan (12 mg/kg) on day 21. Clearance was assessed at 4 h post live challenge. *P < 0.05 compared with non-immune.Figure 2. Clearance of live P. aeruginosa administered IT in agar. Mice were parenterally immunised as described in Figure1, except Kat A was mixed equally with Ad (10 µg each). Live challenge with P. aeruginosa via the trachea was administered on day 21 with 2x104 colony forming units in 0.5% agar. ⎕ Non-immune group (SeM = ± 44%); ● KatA and Ad immunised group (SeM = ± 22%); ◆ heat-killed P. aeruginosa immunised group (SeM = ± 12%). Data presented as the mean of n = 5–6 mice per group. S.e.Ms were too small to show graphically and are given above in the legend text.*P < 0.05 compared with the non-immune.©2014 Landes Bioscience. Do not distribute.www.landesbioscience.com Human Vaccines & Immunotherapeutics 17exacerbation situations, with day 35 being a severe episode. These results show potential for candidate antigens, such as KatA and Ad, to enhance clearance of an acute P. aeruginosa-associated exacerbation of an established chronic P. aeruginosa infection.Despite the enhanced clearance of an acute experimental P. aeruginosa exacerbation, there remains the challenge to translate the findings to human clinical presentations. For example, during acute exacerbations caused by P. aeruginosa in CF patients with chronic P. aeruginosa infection, bacterial load is not necessarily increased before-hand44,45and while clinical improvement is asso-ciated with a fall in bacterial numbers45 this is not a requirement in those with advanced disease.46 Similarly, even if bacterial load is reduced by several orders of magnitude by the use of systemic and inhaled antibiotics, this effect can be independent of any clinical benefit in stable patients with chronic P. aeruginosa infec-tion in those with CF or nCFB.47,48 In contrast, a single clinical study in nCFB patients reported correlation of increased bacterial load during stable clinical states with increased inflammation, exacerbation risk and severity of exacerbation episode.49Chronic P. aeruginosa infection is difficult to clear using anti-biotics since P. aeruginosa can form aggregates or an organized exopolysaccharide biofilm matrix which can be up to 1000-fold more resistant to antibiotics than free-living planktonic cells.12,50,51 Biofilm formation and the expression of a range of associated virulence genes is controlled by a network of transcriptional regulators and their cognate ligands, small signaling molecules or autoinducers, including several acyl hom*oserine lactones (AHLs).52 This system has been termed “quorum sensing”. The importance of biofilm formation and quorum sensing in the pathogenesis of establishing P. aeru-ginosa infection has been demonstrated in a range of animal models where gene knockouts for one or more of the QSSMs or their receptors render the bacterium almost avirulent.53,54A novel approach to P. aeruginosa vaccine development would be to use QSSM carrier protein conjugates as vaccine formulations. In our mouse model of acute lung infection, we have demonstrated that the lasIRrhlIR knockout strain showed equal persistence to wild type parental PAO1, induced equal or greater neutrophil infiltration to the lungs, and induced similar levels of expression of inflammatory cytokines in the lungs and antibody responses, both in terms of magnitude and isotype.55 In contrast to previous reports, these results suggested that the lack of quorum sensing alone does not sig-nificantly affect the immunogenicity, infectiveness and persis-tence of P. aeruginosa. The most frequently produced QSSMs are N-3-(oxododecanoyl)-L-hom*oserine lactone (OdDHL) and butyryl L-hom*oserine lactone (BHL), collectively known as AHLs. It has been widely postulated that blocking quorum sensing and biofilm formation could have a therapeutic ben-efit by limiting the ability of P. aeruginosa to form biofilms. Together, these results suggest that aggregation of P. aerugi-nosa rather than biofilm formation may be of greater impor-tance for establishment of acute infection for this organism.Attempts to reduce the virulence of P. aeruginosa have used several strategies, including the use of structural analogs of QSSMs to block their function.56 Previously, it has been shown that an immune response to QSSMs can be generated, when they are chemically conjugated to a large protein. Conjugation of AHLs to bovine serum albumin (BSA) or keyhole limpet hemocyanin was used to produce monoclonal antibodies in mice,57 while immunisation of mice with BSA-conjugated OdDHL resulted in a partial protection against lung infec-tion.58 Recent studies in mice with OdDHL conjugated to KatA by Lazenby, Cooley and Kyd (unpublished data) also produced an effective immune response and together, these studies represent an ongoing direction for animal-based vac-cine development research.Figure 3. Vaccinating to clear an established chronic P. aeruginosa infection. This animal model was established in our laboratory to determine whether immunisation was effective in combating acute bacterial challenge in mice that had already been chronically infected with P. aeruginosa. (A) In this model, Balb/c mice received 104 colony forming units (cfu) of live P. aerugi-nosa in 0.5% agar intra-tracheally (IT) into the lungs on day 0.On day 7, they received a second dose of 104 cfu live P. aeruginosa in PBS (IT). One group of control non-immune mice ⎕, received only agar on day 0 and PBS on Day 7, prior to acute live P. aeruginosa challenge at day 35, this provided a control group not previously exposed to an infection. The mice were then immunised sub-cutaneously on days 14 and 28 with either PBS (control mice) or vaccines formulated in IFA. On day 35, mice received an acute infection of 106cfu live P. aeruginosa (IT). The mice were killed at 4 h post this infection and bronchial lavage samples collected for testing. (B) This figure illustrates the clearance of P. aeruginosa in the bronchial lavage following acute bacterial IT challenge, 106cfu live P. aeruginosa or PBS, at day 35 in each of the groups. Non-immune mice received PBS (IT) and were sham immunised. All the filled symbol groups received P. aeruginosa in agar (IT) at day 0. ◆Non-immunised control group (but did receive infections on days 0 and 7); ● KatA/Ad immunised group; ▲ heat-killed Pa. Kat A was mixed equally with Ad (10 µg each); and the heat killed P. aeruginosa (HKPa) group was immunised with heat-killed P. aerugi-nosa. Data presented as mean ± SeM of n = 5–6 mice per group.©2014 Landes Bioscience. Do not distribute.18 Human Vaccines & Immunotherapeutics Volume 11 Issue 1Expansion of animal research into mucosal vaccine develop-ment against P. aeruginosa in humans has been undertaken in the form of a safety and immunogenicity study, conducted on the whole killed cell P aeruginosa oral vaccine.59 Twenty-five normal healthy subjects were studied and no vaccine-attributable adverse events were observed clinically or on haematology and biochem-istry blood profiles. Increased serum P. aeruginosa specific antibodies and phago-cytic indices were observed post-immunisa-tion. A pilot open label study has also been conducted on nine patients with bronchiec-tasis.60 Results from these patients showed a significant reduction in total P. aeruginosa counts in sputum cultures (Fig. 5).Together, these animal and clinical studies demonstrate that mucosal immuni-sation holds promise for the development of a successful vaccine against P aeruginosa and that a range of antigen formulations are possible. Despite this progress, further studies on mucosal delivery systems for the protein antigens as well as the mechanisms of the immunity induced are needed. Much more developmental work is required, but if successful the rewards will be great for potentially millions of patients worldwide with chronic P. aeruginosa respiratory infections.Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.AcknowledgmentsThe authors wish to thank Ms Penny Chapman for her editorial assistance with this manuscript.Figure4. The histopathology of the lungs of Balb C mice immunised against Kat A/Ad, after acute exacerbation of chronic infection by P. aeruginosa. Immunised mice lungs showed reduced cellular recruitment, infiltration and lung consolidation and damage after the acute bacterial challenge com-pared with lungs from non-immunised, chronically infected control mice. (A) Shows the histological features of the lung in naïve mice, prior to chronic infection. Pulmonary bronchioles (PB) exhibit regular, intact epithelial surface and the epithelial walls of the alveolus (arrowed) encompass well defined expanded chambers, free of mucus. BV, blood vessel. (B) Illustrates the increased epithelial thickness (arrowed) and infiltration of nucleated cells adja-cent to the alveoli resulting from establishment of chronic infection, within the mouse lungs prior to immunisation. (C) Illustrates similar characteristics resulting from the establishment of chronic infection in the lung in non-immunised mice, prior to bacterial challenge. As observed in (B), chronic infec-tion has increased epithelial thickness (arrowed). (D) Shows the features of lung histology from chronically infected mice after immunisation against Kat A/Ad, prior to acute bacterial challenge. Although increased epithelial thickness is observed in the PB and there is evidence of localized cellular recruitment, the airways remain clear. (E) Demonstrates the damage induced in lung morphology from chronically infected mice in the absence of immunisation to Kat A/Ad, four hours after acute bacterial challenge. PB epithelia are thickened, alveolar walls and capillaries compromised lead-ing to extravasation of blood into airways (#), increased cellular recruitment and consolidation of the lung through collapse of alveolar structure. (F) Demonstrates, in comparison to (E), that 4 h post bacterial challenge, the lung morphology of mice immunised against Kat A/Ad does not exhibit the epithelial damage, alveolar destruction, cellular recruitment or leakage of blood into the airways seen in non-immunised animals. The airways remain relatively clear, with minimal epithelial thickening (arrow). There is some evidence of increased mucus secretion (*) in smaller airways that have retained the mucus during histological processing of the lung. (G and H) Illustrates the lung histology, 24 h post bacterial challenge in non-immunised (G) and animals immunised against Kat A/Ad. In both groups, clearance of the acute damage observed in (E and F), has occurred, however the epithelial thicken-ing, cellular infiltration and alveolar damage is more extensive in the lungs from non-immunised mice. Immunised mice still retain mucus within some of the airways however the extent of airway damage and consolidation appears less in this group.©2014 Landes Bioscience. Do not distribute.www.landesbioscience.com Human Vaccines & Immunotherapeutics 19References1. Kidd T, Whiley D, Bell S, Grimwood K. Pseudomonas. In: Liu D, ed. Molecular detection of human bacterial pathogens. Boca Raton, FL.: CRC Press, 2011:1009-21.2. Klockgether J, Cramer N, Wiehlmann L, Davenport CF, Tümmler B. Pseudomonas aeruginosa Genomic Structure and Diversity. Front Microbiol 2011; 2:150; PMID:21808635; http://dx.doi.org/10.3389/fmicb.2011.001503. Pier G, Ramphal R. Pseudomonas aeruginosa. In: Mandell G, Bennett J, Dolin R, eds. Mandell, Douglas and Bennett’s principles and practice of infectious diseases. Philadelphia: Churchill Livingstone, 2010:2835-60.4. Ciofu O, Hansen CR, Høiby N. Respiratory bacte-rial infections in cystic fibrosis. Curr Opin Pulm Med 2013; 19:251-8; PMID:23449384; http://dx.doi.org/10.1097/MCP.0b013e32835f1afc5. McShane PJ, Naureckas ET, Tino G, Strek ME. Non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med 2013; 188:647-56; PMID:238989226. Desai H, Eschberger K, Wrona C, Grove L, Agrawal A, Grant B, Yin J, Parameswaran GI, Murphy T, Sethi S. Bacterial colonization increases daily symp-toms in patients with chronic obstructive pulmo-nary disease. Ann Am Thorac Soc 2014; 11:303-9; PMID:24423399; http://dx.doi.org/10.1513/AnnalsATS.201310-350OC7. Gellatly SL, Hanco*ck RE. Pseudomonas aeruginosa: new insights into pathogenesis and host defenses. Pathog Dis 2013; 67:159-73; PMID:23620179; http://dx.doi.org/10.1111/2049-632X.120338. Crabbé A, Ledesma MA, Nickerson CA. Mimicking the host and its microenvironment in vitro for study-ing mucosal infections by Pseudomonas aeruginosa. Pathog Dis 2014; 71:1-19; PMID:24737619; http://dx.doi.org/10.1111/2049-632X.121809. Sievert DM, Ricks P, Edwards JR, Schneider A, Patel J, Srinivasan A, Kallen A, Limbago B, Fridkin S; National Healthcare Safety Network (NHSN) Team and Participating NHSN Facilities. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009-2010. Infect Control Hosp Epidemiol 2013; 34:1-14; PMID:23221186; http://dx.doi.org/10.1086/66877010. Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, Moreno R, Lipman J, Gomersall C, Sakr Y, et al.; EPIC II Group of Investigators. International study of the prevalence and outcomes of infection in intensive care units. JAMA 2009; 302:2323-9; PMID:19952319; http://dx.doi.org/10.1001/jama.2009.175411. Sun HY, Fujitani S, Quintiliani R, Yu VL. Pneumonia due to Pseudomonas aeruginosa: part II: antimicrobial resistance, pharmacodynamic con-cepts, and antibiotic therapy. Chest 2011; 139:1172-85; PMID:21540216; http://dx.doi.org/10.1378/chest.10-016712. Staudinger BJ, Muller JF, Halldórsson S, Boles B, Angermeyer A, Nguyen D, Rosen H, Baldursson O, Gottfreðsson M, Guðmundsson GH, et al. Conditions associated with the cystic fibrosis defect promote chronic Pseudomonas aeruginosa infec-tion. Am J Respir Crit Care Med 2014; 189:812-24; PMID:24467627; http://dx.doi.org/10.1164/rccm.201312-2142OC13. Rello J, Rué M, Jubert P, Muses G, Soñora R, Vallés J, Niederman MS. Survival in patients with nosocomial pneumonia: impact of the severity of illness and the etiologic agent. Crit Care Med 1997; 25:1862-7; PMID:9366771; http://dx.doi.org/10.1097/00003246-199711000-0002614. Parkins MD, Gregson DB, Pitout JD, Ross T, Laupland KB. Population-based study of the epi-demiology and the risk factors for Pseudomonas aeruginosa bloodstream infection. Infection 2010; 38:25-32; PMID:20012908; http://dx.doi.org/10.1007/s15010-009-9145-915. Sanders DB, Li Z, Laxova A, Rock MJ, Levy H, Collins J, Ferec C, Farrell PM. Risk factors for the progression of cystic fibrosis lung disease through-out childhood. Ann Am Thorac Soc 2014; 11:63-72; PMID:24261460; http://dx.doi.org/10.1513/AnnalsATS.201309-303OC16. Kerem E, Viviani L, Zolin A, MacNeill S, Hatziagorou E, Ellemunter H, Drevinek P, Gulmans V, Krivec U, Olesen H; ECFS Patient Registry Steering Group. Factors associated with FEV1 decline in cystic fibro-sis: analysis of the ECFS Patient Registry. Eur Respir J 2014; 43:125-33; PMID:23598952; http://dx.doi.org/10.1183/09031936.0016641217. Loebinger MR, Wells AU, Hansell DM, Chinyanganya N, Devaraj A, Meister M, Wilson R. Mortality in bronchiectasis: a long-term study assess-ing the factors influencing survival. Eur Respir J 2009; 34:843-9; PMID:1935715518. Almagro P, Salvadó M, Garcia-Vidal C, Rodríguez-Carballeira M, Cuchi E, Torres J, Heredia JL. Pseudomonas aeruginosa and mortality after hospital admission for chronic obstructive pulmonary disease. Respiration 2012; 84:36-43; PMID:2199655519. Ferkol T, Schraufnagel D. The global burden of respi-ratory disease. Ann Am Thorac Soc 2014; 11:404-6; PMID:24673696; http://dx.doi.org/10.1513/AnnalsATS.201311-405PS20. Williams BJ, Dehnbostel J, Blackwell TS. Pseudomonas aeruginosa: host defence in lung diseases. Respirology 2010; 15:1037-56; PMID:20723140; http://dx.doi.org/10.1111/j.1440-1843.2010.01819.x21. Høiby N. Recent advances in the treatment of Pseudomonas aeruginosa infections in cystic fibro-sis. BMC Med 2011; 9:32; PMID:21463524; http://dx.doi.org/10.1186/1741-7015-9-3222. Hurley MN, Cámara M, Smyth AR. Novel approaches to the treatment of Pseudomonas aeru-ginosa infections in cystic fibrosis. Eur Respir J 2012; 40:1014-23; PMID:22743672; http://dx.doi.org/10.1183/09031936.0004201223. Vincent JL. Vaccine development and passive immu-nization for Pseudomonas aeruginosa in critically ill patients: a clinical update. Future Microbiol 2014; 9:457-63; PMID:24810345; http://dx.doi.org/10.2217/fmb.14.1024. Cohen TS, Prince A. Cystic fibrosis: a mucosal immu-nodeficiency syndrome. Nat Med 2012; 18:509-19; PMID:22481418; http://dx.doi.org/10.1038/nm.271525. O’Sullivan BP, Freedman SD. Cystic fibrosis. Lancet 2009; 373:1891-904; PMID:19403164; http://dx.doi.org/10.1016/S0140-6736(09)60327-526. Hartl D, Gaggar A, Bruscia E, Hector A, Marcos V, Jung A, Greene C, McElvaney G, Mall M, Döring G. Innate immunity in cystic fibrosis lung disease. J Cyst Fibros 2012; 11:363-82; PMID:22917571; http://dx.doi.org/10.1016/j.jcf.2012.07.00327. Lavoie EG, Wangdi T, Kazmierczak BI. Innate immune responses to Pseudomonas aeruginosa infection. Microbes Infect 2011; 13:1133-45; PMID:21839853; http://dx.doi.org/10.1016/j.micinf.2011.07.01128. Yang L, Jelsbak L, Molin S. Microbial ecology and adaptation in cystic fibrosis airways. Environ Microbiol 2011; 13:1682-9; PMID:21429065; http://dx.doi.org/10.1111/j.1462-2920.2011.02459.x29. Folkesson A, Jelsbak L, Yang L, Johansen HK, Ciofu O, Høiby N, Molin S. Adaptation of Pseudomonas aeruginosa to the cystic fibrosis airway: an evolution-ary perspective. Nat Rev Microbiol 2012; 10:841-51; PMID:23147702; http://dx.doi.org/10.1038/nrmicro290730. Mayer-Hamblett N, Rosenfeld M, Gibson RL, Ramsey BW, Kulasekara HD, Retsch-Bogart GZ, Morgan W, Wolter DJ, Pope CE, Houston LS, et al. Pseudomonas aeruginosa in vitro phenotypes distin-guish cystic fibrosis infection stages and outcomes. Am J Respir Crit Care Med 2014; 190:289-97; PMID:24937177Figure5. Clinical trial results showing total sputum bacterial counts (calculated as the log10 colony forming units /mL sputum, mean ± SeM) for the nine patients with bronchiectasis from a pilot open label study. The mean sputum bacterial count is significantly lower (P < 0.05; unpaired t test) for days 56–84 compared with the pre-screening day (Day 28) and initial trial culture (Day 7). Oral immunisation commenced on Day 0, Day 28, or Day 56 as indicated by arrows and continued for a 3 d period. each subject received 2 × 1011 formaldehyde killed P. aeruginosa per day as an enteric coated capsule. All subjects were fasting for at least 6 h prior to the administration of the cap-sule.59 Reproduced with permission of the University of Sydney from: RL Clancy, G Pang, Dunkley M, and Aw Cripps Control of bacterial colonisation of the respiratory tract mucosa in man. In: Mucosal Solutions: Advances in mucosal immunology.Volume 1. editors: Alan J Husband, Kenneth w Beagley, Robert L Clancy, Andrew M Collins, Allan w Cripps and David L emery. University of Sydney, Sydney, Australia. pp 261–268. 1997©2014 Landes Bioscience. Do not distribute.20 Human Vaccines & Immunotherapeutics Volume 11 Issue 131. Döring G, Pier GB. Vaccines and immunotherapy against Pseudomonas aeruginosa. Vaccine 2008; 26:1011-24; PMID:18242792; http://dx.doi.org/10.1016/j.vaccine.2007.12.00732. 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; http://dx.doi.org/10.1517/14712598.5.7.96733. Thomas LD, Kyd JM, Bastin DA, Dunkley ML, Cripps AW. Immunisation with non-integral OMPs promotes pulmonary clearance of Pseudomonas aeruginosa. FEMS Immunol Med Microbiol 2003; 37:155-60; PMID:12832119; http://dx.doi.org/10.1016/S0928-8244(03)00073-734. Wu W, Huang J, Duan B, Traficante DC, Hong H, Risech M, Lory S, Priebe GP. Th17-stimulating pro-tein vaccines confer protection against Pseudomonas aeruginosa pneumonia. Am J Respir Crit Care Med 2012; 186:420-7; PMID:22723292; http://dx.doi.org/10.1164/rccm.201202-0182OC35. Priebe GP, Goldberg JB. Vaccines for Pseudomonas aeruginosa: a long and winding road. Expert Rev Vaccines 2014; 13:507-19; PMID:24575895; http://dx.doi.org/10.1586/14760584.2014.89005336. Clancy RL, Dunkley M. Acute exacerbations in COPD and their control with oral immunization with non-typeable haemophilus influenzae. Front Immunol 2011; 2:7; PMID:2256679837. Cripps AW, Dunkley ML, Clancy RL. Mucosal and systemic immunizations with killed Pseudomonas aeruginosa protect against acute respiratory infec-tion in rats. Infect Immun 1994; 62:1427-36; PMID:813234938. Macmillan H, Norimine J, Brayton KA, Palmer GH, Brown WC. Physical linkage of naturally com-plexed bacterial outer membrane proteins enhancesimmunogenicity. Infect Immun 2008; 76:1223-9; PMID:18086812; http://dx.doi.org/10.1128/IAI.01356-0739. Lee J-S, Heo Y-J, Lee JK, Cho Y-H. KatA, the major catalase, is critical for osmoprotection and virulence in Pseudomonas aeruginosa PA14. Infect Immun 2005; 73:4399-403; PMID:15972537; http://dx.doi.org/10.1128/IAI.73.7.4399-4403.200540. Thomas LD, Dunkley ML, Moore R, Reynolds S, Bastin DA, Kyd JM, Cripps AW. Catalase immuniza-tion from Pseudomonas aeruginosa enhances bacterial clearance in the rat lung. Vaccine 2000; 19:348-57; PMID:10930690; http://dx.doi.org/10.1016/S0264-410X(00)00146-841. Elkins JG, Hassett DJ, Stewart PS, Schweizer HP, McDermott TR. Protective role of catalase in Pseudomonas aeruginosa biofilm resistance to hydrogen peroxide. Appl Environ Microbiol 1999; 65:4594-600; PMID:1050809442. Hassett DJ, Ma J-F, Elkins JG, McDermott TR, Ochsner UA, West SEH, Huang CT, Fredericks J, Burnett S, Stewart PS, et al. Quorum sensing in Pseudomonas aeruginosa controls expression of cata-lase and superoxide dismutase genes and mediates biofilm susceptibility to hydrogen peroxide. Mol Microbiol 1999; 34:1082-93; PMID:10594832; http://dx.doi.org/10.1046/j.1365-2958.1999.01672.x43. Andrade J, Karmali A, Carrondo MA, Frazão C. Structure of amidase from Pseudomonas aerugi-nosa showing a trapped acyl transfer reaction inter-mediate state. J Biol Chem 2007; 282:19598-605; PMID:17442671; http://dx.doi.org/10.1074/jbc.M70103920044. Stressmann FA, Rogers GB, Marsh P, Lilley AK, Daniels TW, Carroll MP, Hoffman LR, Jones G, Allen CE, Patel N, et al. Does bacterial density in cystic fibrosis sputum increase prior to pulmo-nary exacerbation? J Cyst Fibros 2011; 10:357-65; PMID:21664196; http://dx.doi.org/10.1016/j.jcf.2011.05.00245. Reid DW, Latham R, Lamont IL, Camara M, Roddam LF. Molecular analysis of changes in Pseudomonas aeruginosa load during treatment of a pulmonary exacerbation in cystic fibrosis. J Cyst Fibros 2013; 12:688-99; PMID:23706827; http://dx.doi.org/10.1016/j.jcf.2013.03.00846. Deschaght P, Schelstraete P, Van Simaey L, Vanderkercken M, Raman A, Mahieu L, Van Daele S, De Baets F, Vaneechoutte M. Is the improvement of CF patients, hospitalized for pulmonary exacerba-tion, correlated to a decrease in bacterial load? PLoS One 2013; 8:e79010; PMID:24312174; http://dx.doi.org/10.1371/journal.pone.007901047. Tunney MM, Einarsson GG, Wei L, Drain M, Klem ER, Cardwell C, Ennis M, Boucher RC, Wolfgang MC, Elborn JS. Lung microbiota and bacterial abun-dance in patients with bronchiectasis when clinically stable and during exacerbation. Am J Respir Crit Care Med 2013; 187:1118-26; PMID:23348972; http://dx.doi.org/10.1164/rccm.201210-1937OC48. Quon BS, Goss CH, Ramsey BW. Inhaled antibiot-ics for lower airway infections. Ann Am Thorac Soc 2014; 11:425-34; PMID:24673698; http://dx.doi.org/10.1513/AnnalsATS.201311-395FR49. Chalmers JD, Smith MP, McHugh BJ, Doherty C, Govan JR, Hill AT. Short- and long-term antibiotic treatment reduces airway and systemic inflammation in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med 2012; 186:657-65; PMID:22744718; http://dx.doi.org/10.1164/rccm.201203-0487OC50. Drenkard E, Ausubel FM. Pseudomonas biofilm formation and antibiotic resistance are linked to phenotypic variation. Nature 2002; 416:740-3; PMID:11961556; http://dx.doi.org/10.1038/416740a51. Kyd JM, McGrath J, Krishnamurthy A. Mechanisms of bacterial resistance to antibiotics in infec-tions of COPD patients. Curr Drug Targets 2011; 12:521-30; PMID:21194403; http://dx.doi.org/10.2174/13894501179475151952. Schuster M, Greenberg EP. A network of networks: quorum-sensing gene regulation in Pseudomonas aeruginosa. Int J Med Microbiol 2006; 296:73-81; PMID:16476569; http://dx.doi.org/10.1016/j.ijmm.2006.01.03653. Pearson JP, Feldman M, Iglewski BH, Prince A. Pseudomonas aeruginosa cell-to-cell signaling is required for virulence in a model of acute pulmo-nary infection. Infect Immun 2000; 68:4331-4; PMID:10858254; http://dx.doi.org/10.1128/IAI.68.7.4331-4334.200054. Rumbaugh KP, Griswold JA, Iglewski BH, Hamood AN. Contribution of quorum sensing to the virulence of Pseudomonas aeruginosa in burn wound infections. Infect Immun 1999; 67:5854-62; PMID:1053124055. Lazenby JJ, Griffin PE, Kyd J, Whitchurch CB, Cooley MA. A quadruple knockout of lasIR and rhlIR of Pseudomonas aeruginosa PAO1 that retains wild-type twitching motility has equivalent infectivity and persistence to PAO1 in a mouse model of lung infec-tion. PLoS One 2013; 8:e60973; PMID:23593362; http://dx.doi.org/10.1371/journal.pone.006097356. Rasmussen TB, Givskov M. Quorum-sensing inhibi-tors as anti-pathogenic drugs. Int J Med Microbiol 2006; 296:149-61; PMID:16503194; http://dx.doi.org/10.1016/j.ijmm.2006.02.00557. Kaufmann GF, Sartorio R, Lee SH, Mee JM, Altobell LJ 3rd, Kujawa DP, Jeffries E, Clapham B, Meijler MM, Janda KD. Antibody interference with N-acyl hom*oserine lactone-mediated bacterial quorum sensing. J Am Chem Soc 2006; 128:2802-3; PMID:16506750; http://dx.doi.org/10.1021/ja057869858. Miyairi S, Tateda K, Fuse ET, Ueda C, Saito H, Takabatake T, Ishii Y, Horikawa M, Ishiguro M, Standiford TJ, et al. Immunization with 3-oxodo-decanoyl-L-hom*oserine lactone-protein conjugate protects mice from lethal Pseudomonas aeruginosa lung infection. J Med Microbiol 2006; 55:1381-7; PMID:17005787; http://dx.doi.org/10.1099/jmm.0.46658-059. Cripps AW, Peek K, Dunkley M, Vento K, Marjason JK, McIntyre ME, Sizer P, Croft D, Sedlak-Weinstein L. Safety and immunogenicity of an oral inactivated whole-cell pseudomonas aeruginosa vaccine admin-istered to healthy human subjects. Infect Immun 2006; 74:968-74; PMID:16428742; http://dx.doi.org/10.1128/IAI.74.2.968-974.200660. Clancy R, Pang G, Dunkley M, Cripps A. Control of bacterial colonisation of the respiratory tract mucosa in man. In: Alan J Husband, Kenneth W Beagley, Robert L Clancy, Andrew M Collins, Allan W Cripps, Emery DL, eds. Mucosal Solutions: Advances in mucosal immunology. Sydney, Australia: University of Sydney, 1997:261-8.
  • TCC- FINAL - MARINA SOARES DIAS (1)
  • jshsh
  • enfermagem
  • Enfermagem da Família
  • Metodologia do Trabalho Acadêmico
  • Obesidade na Gestação
  • Absenteísmo na Enfermagem
  • Relatório de Estágio em Enfermagem
  • 07. RELATORIO ATIVIDADE OBRIGATORIA - ROTEIRO
  • A AVENTURA DA CELULA - HPV
  • gabarito tecnico_de_enfermagem Marques ibade
  • Prova Técnico de Enfermagem
  • Preparação para exames
  • avaliação finalA pneumonia pode ser definida histologicamente como o preenchimento do espaço alveolar com um infiltrado inflamatório. É clinicament...
  • Tempo restante 0:59:41 Questão 1 Ainda não respondida Vale 1,00 ponto(s). Marcar questão Texto da questão Em relação à Consulta de Enfermagem: I. ...
  • Sobre os EquipamenO modelo de gestão adotado pelo CQH (Compromisso com a Qualidade Hospitalar) preconiza a análise de dados relativos ao desempenho...
  • ser definida como um conjunto de técnicas que podem ser aplicadas sobre um processo objetivando torná-lo mais eficiente, ou seja, maximizando a pro...
  • urante a avaliação pulmonar de um cliente, o enfermeiro detectou alteração na ausculta, relacionando ao estreitamento dos brônquios (broncoespasmo)...
  • Componente Específico estavam compreensíveis e objetivos?A Sim, todos.B Sim, a maioria.C Apenas cerca da metade.D Poucos.E Não, nenhum.
  • A respeito dessas asserções, assinale a opção correta.I. Os profissionais de enfermagem são expostos, no trabalho, a vários fatores estressores qu...
  • Considerando a prática de enfermeiros no PSE em relação à promoção da saúde mental e à atenção psicossocial, avalie as afirmacoes a seguir.I. O en...
  • Considerando os textos apresentados, avalie as afirmacoes a seguir.I. O CCC busca situar a criança em um lugar de protagonismo na escuta ativa e ...
  • Considerando as informações apresentadas, avalie as asserções a seguir e a relação proposta entre elas.I. Os profissionais de enfermagem são expo...
  • Considerando o contexto apresentado, é correto apenas o que se afirma emI. O enfermeiro deve estar atento aos sinais e sintomas que possam indicar...
  • Considerando os textos apresentados, avalie as afirmacoes a seguir.I. As equipes de atenção básica têm como foco de trabalho a programação e a im...
  • Considerando as informações do texto, assinale a opção correta, quanto à classificação de risco dos pacientes/usuários nos serviços de saúde.A O ...
  • Melatonina o que é para que serve e efeitos colaterais
  • Melhores remédios para cada tipo e intensidade de dor

Perguntas dessa disciplina

Sobre a Pseudomonas aeruginosa, é correto afirmar: A. Há a existência de Pseudomonas aeruginosa na água, porém ela não se manifesta na água, pois ...
2.Pseudomonas aeruginosa é uma bactéria gram-negativa, baciliforme e aeróbia. Está envolvida em vários processos infecciosos e apresenta resistênci...

UNIASSELVI

Sobre a bactéria Pseudomonas aeruginosa, classifique V para as sentenças verdadeiras e F para as falsas: ( ) O ácido nalidíxico tem atividade c...
As bactérias do gênero Pseudomonas são amplamente distribuídas no ambiente e podem ser encontradas no solo, na água, em plantas e em muitos outros ...

FECAF

15-Vaccination against respiratory Pseudomonas aeruginosa infection - Enfermagem (2024)

References

Top Articles
Latest Posts
Article information

Author: Amb. Frankie Simonis

Last Updated:

Views: 5496

Rating: 4.6 / 5 (56 voted)

Reviews: 95% of readers found this page helpful

Author information

Name: Amb. Frankie Simonis

Birthday: 1998-02-19

Address: 64841 Delmar Isle, North Wiley, OR 74073

Phone: +17844167847676

Job: Forward IT Agent

Hobby: LARPing, Kitesurfing, Sewing, Digital arts, Sand art, Gardening, Dance

Introduction: My name is Amb. Frankie Simonis, I am a hilarious, enchanting, energetic, cooperative, innocent, cute, joyous person who loves writing and wants to share my knowledge and understanding with you.