2,917
Views
21
CrossRef citations to date
0
Altmetric
Review

Facing the challenges of multidrug-resistant Acinetobacter baumannii: progress and prospects in the vaccine development

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 3784-3794 | Received 03 Feb 2021, Accepted 05 May 2021, Published online: 09 Jun 2021

ABSTRACT

In 2017, the World Health Organization (WHO) named A. baumannii as one of the three antibiotic-resistant bacterial species on its list of global priority pathogens in dire need of novel and effective treatment. With only polymyxin and tigecycline antibiotics left as last-resort treatments, the need for novel alternative approaches to the control of this bacterium becomes imperative. Vaccines against numerous bacteria have had impressive records in reducing the burden of the respective diseases and addressing antimicrobial resistance; as in the case of Haemophilus influenzae type b . A similar approach could be appropriate for A. baumannii. Toward this end, several potentially protective antigens against A. baumannii were identified and evaluated as vaccine antigen candidates. A licensed vaccine for the bacteria, however, is still not in sight. Here we explore and discuss challenges in vaccine development against A. baumannii and the promising approaches for improving the vaccine development process.

Introduction

Acinetobacter species are mostly free-living saprophytes with different genus inhabiting different habitats including soil, water, and sewage.Citation1 Among the Acinetobacters, A. baumannii, A. haemolyticus, and A. calcoaceticus have been reported as the species with high clinical importance.Citation2 Acinetobacter baumannii in particular, is most commonly found within the healthcare facility environments.Citation1 . baumannii, a Gram-negative coccobacillus was previously regarded as a low-grade pathogen of humans. It has in recent decades emerged as a leading cause of health-care-associated infections (HAIs).Citation3 The bacteria causes a wide range of infections including skin and soft-tissue infections associated with trauma, urinary tract infection, pneumonia, meningitis, and septicemia.Citation4,Citation5 Early on before the 1970s, infections by A. baumannii were easily treatable with beta-lactams and sulfonamide antibiotics.Citation3 Within a few decades, however, the bacteria acquired extensive resistance to the antibiotics which led to the eventual emergence of the multidrug resistance (MDR) A. baumannii strains. A. baumannii was a major concern in conflict regions such as in Iraq and Afghanistan, where a high incidence of MDR A. baumannii bloodstream infections and battlefield trauma in injured military personnel were reported; which earned it the moniker “Iraqibacter”.Citation6–8 An increasing trend in the prevalence of the bacterium in long-term care facilities and nursing homes suggests that Acinetobacter strains are no longer strictly a healthcare facility-associated pathogen.Citation9,Citation10 Community-acquired infections caused by A. baumannii are increasingly being reported, especially in the tropical or subtropical regions of the world.Citation11,Citation12

Common infections with A. baumannii could lead to mortality if left untreated or ineffectively treated when such infections involved antibiotic-resistant (ABR) bacterial strains. The ABR as human pathogens not only poses a detrimental impact on human health but also on economic wellbeing.Citation13 Specifically for A.baumannii, infection which resulted in admission into an intensive care unit (ICU), lead to increase length of hospital stay (LOS), thus, escalating the cost of treatment (COT).Citation14 Economically, the LOS and mortality are proportionately related to the productivity losses for a country that directly contributing to the economic burden.Citation15

Antibiotic resistance of A. baumannii has been linked to the “resistome”, a well-coordinated network between antibiotic resistance genes, and other genetic elements such as those associated with critical bacterial metabolism.Citation16 The MDR A. baumannii isolates have been reported worldwide and they are resistant to at least three classes of antimicrobial agents, viz. penicillins and cephalosporins, fluoroquinolones and aminoglycosides.Citation17–19 Owing to its serious health concerns and global spread, the MDR A. baumannii was listed by the World Health Organization (WHO) as amongst the 12 bacterial families for which new drugs are urgently needed.Citation19 In addition, an independent Review on Antimicrobial Resistance, jointly supported by the UK Government and Wellcome Trust, issued specific recommendations to reduce dependence on antibiotics and called for the development of new vaccines, or greater use of existing vaccines against the bacterial infections.Citation20,Citation21 Use of vaccines would reduce the usage of antibiotics,Citation21 hence, decrease the incidence of antimicrobial resistance.Citation22 Vaccines could also play a significant role as a long-term approach in preventing bacterial infections and lowering the overall disease burden.Citation23

Toward vaccine development: identifying virulence factors and understanding the pathogenesis of A. baumannii

Over the past decades, our understanding of the pathogenesis of A. baumannii, including its virulence factors, antimicrobial resistance mechanisms, and quorum sensing have increased tremendously.Citation3,Citation24 The numerous factors underlying the success of the bacterium in causing disease have led to the identification of candidate antigens for the development of novel vaccines.Citation3,Citation25–27 Among the factors that are essential for the successful transmission of A. baumannii are those associated with the membrane proteins and cell surface adaptations, antibiotic tolerance, mechanism of nutrient acquisition, and bacterial community interactions.Citation24 Some of these factors have been demonstrated to work in concert with others.

One of the striking features of A. baumannii is its resistance to most antibiotics, including the last resort antimicrobials; carbapenems, colistin, and tigecycline.Citation28 Resistance is associated with environmental stress conditions,Citation29 as well as the bacterial cell surface structure which consisted of the lipopolysaccharides (LPS) as the major component of the outer leaflet of the outer membrane (OM), capsular polysaccharides (CPS), glycosylated proteins, and the peptidoglycans.Citation30 These bacterial components provide strong structural stabilization of the bacteria and contribute to cellular homeostasis under changing environmental conditions.Citation31 Additionally, the biofilm which is a complex extracellular surface structure formation is among the virulence factors of A. baumannii that is also associated with antimicrobial resistance.Citation27 The biofilm-associated proteins Bap and Blp1 have been reported to enhance attachment to host cells or abiotic surfaces.Citation32 Expression of other proteins within the bacterium such as autotransporter (Ata) and the extracellular poly-N-acetyl-β-(1-6)-glucosamine (PNAG) are shown to mediate attachment and adhesion of the cells in the biofilm.Citation24 The biofilm not only protects the bacterium from antibiotics but also facilitates attachment to abiotic surfaces as well as the host epithelial cells.Citation33 The key role of the cell surface structures, however, is in mediating immune evasion from the host, e.g. K1 CPS, which augments the bacterium’s ability to survive in human serum and contribute to causing disease within the host.Citation34,Citation35

The extraordinary balance and plasticity of the A. baumannii genome enable the bacterium to adapt for survival, contributing to its superbug status. Further investigation into the genetics of antibiotic resistance revealed that antibiotic resistance is not dependent on an individual characteristic, but rather on a repertoire of traits including enzymatic determinants (e.g. β-lactamase), genetic manipulations (where genes are recombined, acquired, or lost), as well as efflux pumps that extrude the antibiotics.Citation35 A. baumannii possesses representatives of each of the six efflux pump families associated with resistance to various antimicrobials; major facilitator superfamily (MFS) pumps (tetracycline), multidrug and toxic compound extrusion family (MATE; aminoglycosides, quinolones, and chloramphenicol), small multidrug resistance (SMR) superfamily (erythromycin and novobiocin), resistance-nodulation-division family (RND; aminoglycosides, fluoroquinolones, tetracycline, trimethoprim, and β-lactams), ATP-binding cassette (ABC) family, and proteobacterial antimicrobial compound efflux family.Citation28,Citation35 While these efflux pumps have been well-described, the full understanding of their underlying mechanisms is still limited.Citation28,Citation35 A transcriptomic study of multidrug resistance in A. baumannii showed that the regulation of antibiotic resistance occurred at the level of operons, which involved different responses of the strains to different classes of antibiotics. The study also revealed that multiple antibiotic resistance genes were necessary to confer antibiotic resistance in MDR strains so that the broad resistance was not dependent on any single antibiotic gene.Citation36 It is hypothesized that the upregulation of efflux pumps responsible for antibiotic tolerance could represent a form of cellular adaptation under nutrient limiting conditions.Citation35

A. baumannii can produce various low molecular weight compounds known as siderophores that have an extremely high affinity toward metals.Citation37,Citation38 Metals, especially iron, are an essential nutrient for all living organisms as they are needed for various biochemical and physiological functions. They are also crucial for the survival of A. baumanii within the host. Regardless of the abundance of metals in the biological systems, the bioavailability of certain active metals such as ferric iron is relatively low due to sequestration by compounds like heme, lactoferrin, and transferrin.Citation28,Citation39 A. baumannii produces three classes of siderophore; namely acinetobactin,Citation40 fimsbactins,Citation41 and baumannoferrins,Citation42 which are transported by their respective TonB-dependent transporters.Citation43 This metal acquisition system, particularly those involving iron, contributes to the ability of A. baumannii to survive and causes infection in humans.Citation44 It is suggested that, under conditions of low iron concentration, the RND efflux pump genes known as adeABC, are co-transcribed as a single adeABC operon leading to overexpression of the adeABC pump.Citation45 Additionally, the mutation of the TonB-dependent copper receptor in A. baumannii leads to a reduction in the biofilm formation, underlining the additional role of copper, besides iron, in the regulation of its virulence.Citation25 Iron limitation has also been reported to regulate the biofilm formation via the induction of N-acyl-homoserine lactone;Citation46 a signal that mediates the communication between neighboring bacteria (quorum sensing), thus, enables the bacteria to survive under limited micronutrients supply while forming the biofilms.Citation46 Considering this, it is plausible that interfering with iron or other essential metal transport mechanism could be the basis of a new strategy toward a broadly efficacious therapeutic or vaccine.

More virulence factors are now being reported and they offer great potential for discovering novel anti-virulence or vaccine targets against the bacterium. Nevertheless, the number of identified virulence factors is still scarce in comparison to factors identified for other Gram-negative pathogens. A few of the identified virulence factors of A. baumannii, however, are also bacterial strain-specific, rendering them less promising for the development of broadly efficacious therapeutic or vaccine.Citation3

Progress in A. baumannii vaccine development

Vaccines are now beginning to be recognized as potentially highly effective tools to mitigate antimicrobial resistance (AMR).Citation22 Several studies reporting the potential of vaccines against A. baumannii have recently emerged,Citation47,Citation48 and various types of vaccine for this bacterium have been evaluated (). The earlier vaccines for A. baumannii were based on the whole cells, either grown without antibioticCitation47 or under antibiotic-exposed conditionCitation49 followed by inactivation with formalin, known as inactivated whole-cell (IWC) vaccine. IWC-based vaccines have been demonstrated to induce specific antibody responses against the A. baumannii and confer protection against subsequent sepsis challenges with the live bacterium.Citation47 Low production cost adds to the attraction of this approach.Citation49 As an alternative to formalin-IWC, A. baumannii ghosts (ABGs) also hold promise. ABGs, like other bacterial ghosts, comprise the cellular surface antigens without the cytoplasm and its constituent.Citation50 In studies, where rats were vaccinated with the ABGs via oral, subcutaneous, intramuscular and intraperitoneal routes, all routes demonstrated full protection following challenge with a lethal dose of the live bacteria, except those vaccinated orally which showed 67% protection.Citation50 Nonetheless, safety remains a primary concern for the use of IWCs or ABGs, as the risk of incomplete inactivation/treatment could potentially result in undesirable side effects. For example, IWCs or ABGs containing pathogen-associated molecular patterns (PAMPs), which are known to be pro-inflammatory molecules, can lead to reactogenicity in hosts.Citation68 to Citation51

Table 1. List of developed A. baumannii vaccine evaluated in animal models

Apart from the whole bacterium, the bacterial cell outer membrane complexes (OMCs)Citation52 and outer membrane vesicles (OMVs)Citation53,Citation54 have also been explored for vaccine development. These cell components exhibited promising outcomes as they elicited antibodies that recognized antigens from diverse A. baumannii strains.Citation52 Application of these cell components as vaccines in humans, however, has been precluded owing to the possibility of contamination with pyrogenic LPS during vaccine preparation.Citation54,Citation55 To overcome this concern, LPS-deficient IWC was developed by introducing mutation to the lpxD gene essential for lipid A biosynthesis. Without the lipid A, the bacteria become LPS-deficient. The LPS-deficient IWC was shown to evoke robust immune responses similar to that of the wild-type bacterium.Citation56 With the success of LPS-deficient IWC, the cell component vaccines using the LPS-deficient bacteria were developed and called LPS-free OMCs and LPS-free OMVs, respectively.Citation55,Citation57 While these LPS-free candidate vaccines exhibit low levels of endotoxins activity, they are less immunogenic in comparison to the wild type OMCs and OMVs. This is shown in the findings whereby LPS-free OMCs and LPS-free OMVs vaccines were demonstrated to provide only partial protection against sepsis infection when challenged with a lethal dose of live bacteria.Citation55,Citation57 Total protection against infection was achieved only in mice immunized with a high dose of LPS-free OMVs.Citation57 While concern over endotoxin contamination is lessened with LPS-deficient IWC, another concern arises from incomplete inactivation of the strain. Incomplete inactivation of LPS-deficient strain has the potential to result in dissemination of this strain among the vaccinee and possibly into the community. This would be a worrisome predicament as it has been demonstrated that loss of LPS in A. baumannii strains leads to resistance to colistin, among the last option antibiotic for the treatment of A. baumannii infections.Citation58 In this regard, it is uncertain if the absence of LPS components within LPS-free OMCs and LPS-free OMVs could potentially evoke similar negative effects as those observed in LPS-deficient IWC. It might be argued that, unlike the IWCs, the OMCs and OMVs are only sub-cellular components, thus, not associated with the same risk as LPS-deficient IWC. This is reflected in the licensure of vaccines utilizing the Neisseria meningitidis OMVs, such as VA-MENGOC-BC, MenBvac, MeNZB and Bexero.Citation59 It is noteworthy that although some of the earlier described A. baumannii candidate vaccines displayed high immunoefficiency, their applications are limited by safety and potential regulatory concerns. Thus, the discovery of other antigens that can offer significant protective immune response, safety and ease in the manufacturing processes remains a priority.

Bacterial surface polysaccharides, particularly capsular polysaccharides (CPS), display immunomodulatory effects that make them a potential target for vaccine development.Citation60 The bacterial polysaccharides are covalently linked to carrier proteins, and these glycoconjugates are employed to overcome the limitation of unconjugated polysaccharides in failing to evoke immunological memory.Citation61 Several glycoconjugate vaccines have been licensed, such as those for the prevention of Haemophilus influenza type b (Hib), Streptococcus pneumoniae (23 serotypes) and Neisseria meningitides,Citation73 with more under development.Citation60 Specifically for A. baumannii, polyclonal K1 capsule antisera that have been developed for immunotherapy exhibited bactericidal activity against bacterial challenge in a rat soft tissue infection model. Nevertheless, the effect has only been observed within the K1-positive bacterial strains (13 out of 100 tested clinical isolates).Citation62 There are nearly 40 serovars identified and information on seroprevalence of clinical isolates is currently limited.Citation63 Since there are multiple serovars of the CPS, perhaps the utilization of K1 CPS could be applied as a component of a multivalent vaccine for active vaccination, as in the case of Streptococcus pneumoniae.Citation62 Another important common constituent of extracellular polysaccharides in A. baumannii reported to significantly influence the pathogenicity of the bacterium is the PNAG.Citation64 Inoculation with antiserum to PNAG; 9Glc-NH2-TT, induced opsonic and protective antibodies and passive immunization with the antisera reduced the bacterial burden in pneumonia and bacteremia challenges.Citation48 Based on these findings, it is suggested that PNAG is a potential component vaccine for both active and passive immunization with broad serotype coverage.Citation48,Citation63 Passive antibody immunotherapy could particularly be useful for the treatment of acute infections especially at the time of an outbreak.Citation65 Although passive immunization with antisera has potential therapeutic value, some drawbacks could render its wider application unlikely, such as (i) the high cost of producing the antisera, (ii) the lack of heterologous protection as observed for antisera against K1 CPS, (iii) the lack of protection against septic shock and (iv) immediate but short-lived protection.Citation65 In contrast to passive immunization, active immunization (prophylactic vaccines) offers long-lasting and possibly lifelong protection.Citation66

Among other potential vaccine targets include the outer membrane proteins (OMPs) of A. baumannii, especially OmpA. It has been proposed as a potential vaccine candidate owing to its important role in the pathogenesis of the bacteria.Citation67 Efforts are underway for the vaccine development using OmpA as vaccine target via different technologies, either as a protein-based vaccineCitation68,Citation69 or DNA vaccine (encoding OmpA aloneCitation70 or combination of OmpA with other proteinCitation71). All these vaccines are shown to elicit antibodies and to induce promising protective immunity against the bacterium infection.Citation68,Citation69,Citation70,Citation71 Polymorphism of OmpA that resulted in five major bacterium variants, however, may limit the effectiveness of vaccine from a single OmpA variant.Citation72 In addition to OmpA, other OMPs that have been evaluated for their potential as vaccine candidates are Omp22Citation73 and BamA.Citation74 These target proteins have been demonstrated as highly conserved among the A. baumannii strains (95–99%) and displayed promising potential for active and passive immunization.Citation73,Citation74 Study on anti-OmpA MAbs, however, showed that the binding of the antibody was prevented by CPS of A. baumannii, which in turn inhibited the activation of macrophages and complement-dependent bactericidal activity. This raised concern that not only OmpA, but other OMPs may not be viable vaccine targets as these epitopes are shielded by the CPS.Citation75 Hence, the surface-exposed proteins or surface-associated proteins that penetrate the capsule layer may offer better success as candidate vaccine targets.Citation32 Some of these surface-exposed proteins are involved in the formation of biofilm that contributes to the adhesion of the bacterium to the host cell. Among the evaluated proteins are the trimeric autotransporter protein (Ata)Citation76 and Blp1 protein.Citation32 Antibodies to Ata possess opsonophagocytic killing and bactericidal properties, while the antisera to Ata mediates both phagocyte-dependent and phagocyte-independent killing of A. baumannii.Citation76 However, ata gene is present in only 78% of the assessed A. baumannii strains.Citation77 This suggests that a single valent Ata protein may not be sufficient to protect against the strains without Ata, and a combination of proteins might be needed to obtain broader protective efficacy. On the other hand, Blp1 protein encoded in all the examined clinical A. baumannii strains of globally spread clonal lineages I and II (IC I and IC II) could be an alternative.Citation32 The vaccine incorporating Blp1 protein elicited efficient protection against lethal A. baumannii infection in a sepsis murine model, achieving 60% and 100% in the active and passive immunization, respectively.Citation32 The opsonophagocytic killing properties were observed in the antisera against Blp1-specific antigen.Citation32 Although the Blp1 was observed in all of the evaluated A. baumannii strains, differences in the characterization and function of Blp1 protein between IC strains in biofilm formation and adhesion to epithelial cells require further investigation.

Other bacterial components that have been evaluated as vaccine candidates were the outer membrane lipoprotein A, small protein A (smpA), and phospholipase D (PLD).Citation78 Not much is currently known on the function of smpA in A. baumannii, but a study on its homolog in Pseudomonas aeruginosa showed that the lipoprotein is important in the formation and maintenance of cell membrane structure and integrity.Citation78 PLD on the other hand, has been demonstrated to be involved in epithelial cell invasion and in vivo pathogenesis.Citation24,Citation26 The smpA and PLD have been evaluated as a single and multi-component vaccine.Citation78 Mice immunized with these proteins as a single component exhibited lower pro-inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) and serum, and experienced reduced lung infiltration and bacterial load.Citation78 Nevertheless, the combined components did not display any advantages in reducing the bacterial burden, while the protection conferred by passive immunization with the anti-sera provides a slight increase in the survival against bacterial challenge in a pneumonia model.Citation78 As different bacterial strains may elicit different immune responses, perhaps lipoprotein A, smpA, and PLD, as well as K1 CPS could be included among the targets in multicomponent vaccines against A. baumannii.Citation78 In the search for vaccine antigens that provide broad protective coverage, ZnuD has been seen as a promising candidate in Neisseria meningitidis.Citation79 In A. baumannii, ZnuD is necessary for the internalization of zinc in low zinc conditions. In this regard, zinc acquisition plays a crucial role in bacterium growth in the human host.Citation80 Recently, a hybrid antigen displaying a surface loop of ZnuD from A. baumannii consisting of an individual loop (2, 5, 7, and 11) or the loops together in a single hybrid has been explored as a potential vaccine for A. baumannii.Citation80 The study included the refolded ZnuD protein as a control. It is reported that, when evaluated in an active immunization study, the hybrid ZnuD proteins provided better protection from lethal dose challenge in the sepsis model in comparison to the refolded ZnuD.Citation80 This suggested that the structure of ZnuD may need to be displayed in its native structure. Although the refolding process resulted in soluble protein, their native protein structure is possibly affected.

More recently, advancements in molecular technologies and high throughput full genome sequencing have contributed to the establishment of huge databases, allowing for the rational design of vaccines using a process known as “reverse vaccinology” (RV).Citation81,Citation82 RV has led to the discovery of previously unknown antigens and has added greatly to our understanding of the biology of several pathogens.Citation83 It has also contributed to the development of universal vaccines for several pathogens, exemplified by the meningococcus B vaccine.Citation84 RV has already been applied to the development of vaccines for several bacterial pathogens including but not limited to Bacillus anthracis, Streptococcus pneumoniae, Mycobacterium tuberculosis, and Cryptosporidium hominis.Citation85

In the development of A. baumannii vaccines, various RV approaches have been employed to identify potential novel vaccine candidates. These methodologies include direct RV,Citation82,Citation86 antibiotic-resistant determinant-focused RV,Citation85 virulome-based RV,Citation87 and a combination of subtractive proteomics and RV.Citation88 These different RV methods were attempted to identify novel targets that have the potential to elicit broad protective immunoresponse. Although several potential candidates have been identified using these techniques, only a few have been validated in animal models; they include FilF, BamA, OmpK, FK1B and OmpP1.Citation74,Citation82,Citation86 FilF and OmpP1 have been shown to decrease the bacterial load in immunized and challenged mice in pneumonia model of A. baumannii, and increased their survival rate by up to 50%.Citation47,Citation51 However, that level of protection is not considered potent enough against this evolving pathogen. As for OmpK and FK1B, they did not confer protection against pneumonia bacterial challenge despite eliciting a strong IgG response.Citation49 These observations demonstrate possible discordance between in silico and the in vivo findings, emphasizing the importance of further validation.

Prospects for A. baumannii vaccine development

Validation of antigenic protein

To date, no A. baumannii vaccine candidate has advanced to the clinical stage although the application of RV has demonstrated great potential to accelerate the identification of possible candidates. Furthermore, only a few have been experimentally evaluated and most of those selected were associated with the outer membrane of A. baumannii and were generally proteins related to antibiotic-resistance development, transport (efflux), or metabolism. A number of the candidates have been deprioritized due to their high molecular mass, since they may present challenges for efficient protein expression, purification and process development. More efforts to validate these antigens either in vitro and/or in vivo are needed. With the development of better in vitro cell-based or cell-free expression technologies, high-throughput platforms to screen and select the vaccine antigen candidates will become available, including those that screen for toxicity. This should greatly accelerate the vaccine development process.Citation89,Citation90

Alternative vaccine expression host

Many expression platforms for the production of recombinant proteins are now available, and these consist of the eukaryotic, plant or microbial expression systems. The microbial expression system remained a popular choice and widely used mainly because of the short production time, scalable, and low production cost.Citation91 Among this expression system, Escherichia coli has been dominantly used for over 30 years. This could be contributed by the rapid growth kinetics of E. coli, the easy transformation process, and the wide choices of vectors and other commercially available tools for DNA manipulation.Citation92,Citation93 However, this expression system also has several disadvantages, particularly for the development of A. baumannii vaccines. Most highly antigenic A. baumannii proteins such as BamA and FilF are only expressed as insoluble inclusion bodies in E. coli.Citation74,Citation82 Therefore, large-scale vaccine antigen production would require more complex purification and refolding processes, typically leading to a reduced yield of the final antigen product.Citation74,Citation93 To overcome this limitation, in silico vaccine design strategy such as that designed for a soluble Omp34 immunogen with increased antigenicity was described.Citation94 In the study, based on the predicted topology of Omp34, the exposed linear B-cell epitope was predicted. A peptide loop that was considered “inappropriate” was removed and replaced with a more compatible sequence predicted by in silico analysis. This modification, however, has yet to be evaluated in vitro as well as in vivo. Although the modification may increase the solubility of the antigenic protein, it may also have detrimental effects on the immunogenicity,Citation95 such as those reported for the ZnuD mentioned earlier.Citation80 As an alternative to E. coli, expression in Gram-positive bacteria, such as Bacillus and Corynebacterium has been proposed.Citation96,Citation97 Among the Gram-positive bacteria, the spore-forming Gram-positive Bacillus sp. i.e. Bacillus subtilis, has the potential to be used as an expression vehicle in bio-factories for vaccine production. The bacteria are nonpathogenic and with a safe record in human and animal use as probiotics and food additives.Citation98 The supplementation of B. subtilis as a probiotic has been demonstrated as a safe and effective way to stimulate systemic and mucosal responses.Citation99 This further affirms the potential of B. subtilis as a live bacterial vector that could both serve as an expression and delivery vehicle, which also possesses the much desired adjuvant properties.Citation100,Citation101 Amenability to oral delivery in itself constitutes an additional advantage as a delivery platform for the A. baumannii vaccine. The idea of formulating A. baumannii vaccines for mucosal delivery is appealing since the A. baumannii usually gain access into the body via mucosal sites to affect a wide range of tissues. Mucosal vaccines would be expected to elicit both the systemic and mucosal antibody responses.Citation69 Studies on mucosal delivery using B. subtilis spore-displaying antigenic protein have demonstrated the ability to elicit protection and immunogenicity responses.Citation98,Citation102–104 With the advancement of expression systems such as B. subtilis, validation of potential vaccine antigen candidates for A. baumannii is anticipated.

Vaccine delivery

The elucidation of the immune pathways that are critical in defense against A. baumannii infection will hasten the discovery of a novel vaccine.Citation105 What is currently known is that LPS is a potent stimulator of TLR-4, which leads to activation of NF-κB. That, in turn, induces secretion of macrophage inflammatory protein 2 (MIP-2) and KC/IL-8,Citation105 and stimulates the tumor necrosis factor-alpha (TNF-α) release.Citation106 These cytokines, which have chemo-attractant properties, lead to the recruitment and activation of neutrophils and macrophages, in addition to a range of other host factors.Citation24,Citation106 Apart from TLR-4, IL-17 has been shown to stimulate the production of CXC chemokines, which ultimately clears the bacteria during primary infection with Gram-negative bacteria.Citation105 With more evidence showing the critical role played by the Th17 cells (CD4+ IL-17 producing cells) in the host defense against many bacteria, a similar pathway could be important in protecting against A. baumanni infection. Most Th17 cells reside in the barrier tissues such as the intestinal and respiratory tracts, and the skin. The signature cytokines of Th17 cells (include IL-17A, IL-17F, IL-22, and IL-26) besides innate lymphoid cells are critical in regulating tissue homeostasis.Citation105,Citation107 It was reported that Th17 effector cytokines IL-17 and IL-22 induced an array of antimicrobial peptides such as β-defensin-2, lipocalin 2, and the S100 family proteins from barrier tissues, such as gut, skin, and lung.Citation105,Citation107 Based on the identified immune pathways against A. baumannii¸ it appears that the mucosal route for vaccination could be better for the elicitation of an effective and protective immune responses against A. baumannii. However, most of the earlier described A. baumannii vaccine candidates, focused on the parenteral route of administration as it was more challenging to formulate vaccines for mucosal delivery. Currently, the availability of mucosal vaccines in general is still limited. Most of the licensed oral vaccines are based on live attenuated platforms (bacteria or viruses). In mucosal vaccine delivery, the main challenges are to overcome the low immunogenicity and instability of antigens in the harsh mucosal environment.Citation108 Mucosal delivery vehicles that protect and preserve the structural integrity of the target antigen while simultaneously increasing the production of local and systemic neutralizing immune responses are highly needed. Numerous studies have shown that spores of B. subtilis possess these characteristics.Citation98,Citation100,Citation103,Citation109 Other examples of mucosal delivery vehicles being studied include virus-like particles (VLP), immunostimulatory complexes (ISCOMs), polymeric and inorganic nanoparticles, emulsions, pollen grains, and live bacterial vector.Citation59,Citation98

Animal models and bacterial strains

In vaccine studies, preclinical assessment in animal models is one of the prerequisites before the clinical phase in human subjects. As most of the A. baumannii strains do not infect immunocompetent mice commonly used in vaccine studies, such as BALB/c or ICR, there is an urgent need to search for better mammalian models, especially animal model of sepsis and pneumonia. For the establishment of a pneumonia model, infection performed via intra-tracheal,Citation110 intranasal,Citation111 inhalation through nebulizerCitation112 and oral aspiration are feasible.Citation113 As for the sepsis model, infections through intraperitonealCitation114 and intravenous via tail veinCitation113 have been performed. The effects of these different routes of infection on the biomarkers and the elicited host-immune responses, are yet to be fully evaluated. Improved mammalian models that serve as surrogate models for the assessment of vaccines for A. baumannii are urgently needed.Citation114

Currently, to increase the infectivity of A. baumannii, mice to be infected are engineered to be immunocompromised, either by inducing diabetes or administrating the alkylating agent cyclophosphamide (CYP).Citation112 Alternatively, increased virulence of the bacterium is achieved by adding porcine mucin to the challenge bacteria.Citation115 These models nevertheless, are still limited in mimicking human infections.Citation116 Special mouse strains; A/J and C3HeB/FeJ are noted to be more susceptible to A. baumannii infection in comparison to the commonly used mice strains, hence, should be explored as a better model.Citation117 The susceptibility of A/J mice could be attributed to the delayed neutrophil recruitment to the lung caused by diminished CXC chemokine responses to the bacteria, while the reason for susceptibility of C3HeB/FeJ has yet to be explained.Citation117 In addition to mice, rats could represent a better animal model.Citation118 The rat pneumonia model demonstrates many characteristics that mimic human infection without altering the immune profile or requiring the use of porcine mucine.Citation118

Apart from selecting the right animal model, it would be advantageous if the bacterial strains employed for the animal challenge are genotypically characterized. Small genomic changes within the bacteria could lead to significant differences in virulence that could then present a different drug resistance profile.Citation24 Hypervirulent A. baumannii strains have been demonstrated to persist in the blood at high bacterial densities for up to 24 hours and are less susceptible to complement-mediated killing and macrophage uptake.Citation117 Among the hypervirulent strains, HUMC-1,Citation112 AB5075,Citation110 LAC-4,Citation111,Citation114 and VA-AB41Citation113 have been evaluated for their ability to cause lung infections and bacteremia in mice. Among these strains, only LAC-4 strain has been shown to elicit lethal and sublethal pneumonia and sepsis infections, in immunocompetent BALB/c and C57BL/6 mice without any neutropenic rendering procedure.Citation111,Citation114

Vaccine target population

The ultimate goal of vaccine development is a successful product licensed for use. Although WHO listed A. baumannii as among the bacteria that are in critical need of novel antimicrobial agents, the commercial interest may not match the urgency. This is partly due to the low prevalence of the infection and the difficulty in establishing vaccine programs for target populations.Citation119 It is envisioned that three populations would benefit from vaccination against A. baumannii. The first population consists of hospitalized patients or those who will require prolonged treatment in the hospital due to health issues. For this population, vaccination could be incorporated into the pre-surgical care plan.Citation119 Although the prevalence of A. baumannii infection is relatively lower in comparison to other bacterial infections such as those caused by Escherichia coli, Staphylococcus aureus, and Clostridium difficile, the significant burden of the cost associated with the HAIs to the healthcare agency and the patient are still enormous.Citation14 On the other hand, vaccination of those in the ICU could be challenging as the target population may be already immunosuppressed. Because of this difficulty, passive immunization with monoclonal antibodies is perhaps a viable strategy.Citation119

The second population that could benefit from A. baumannii vaccination is the elderly. The global population is rapidly aging. By 2050, it is estimated that the number of persons older than 60 years of age is expected to double.Citation120 Aging is a significant risk factor for A. baumannii infection and the severity of infection is higher in the elderly due to immunosenescence.Citation121 With the increasing prevalence of MDR-A. baumannii and limiting avenues for effective antimicrobial treatment, infection with the bacteria is foreseen to pose more burden clinically and economically, hence, preventive and treatment measures are desperately needed. The elderly population that would be suited as target population would be those above the ages of 50, or perhaps 65, and those with underlying diseases; this would be similar to the study group recommended for Streptococcus pneumoniae.Citation120 As immunosenescence in the elderly could increase susceptibility to infections, it could at the same time pose a challenge for vaccination as the capacity to respond toward vaccine is also compromised.Citation122 Several potential strategies to improve immunogenicity in the elderly have been suggested. These include the use of stronger adjuvants, higher antigen doses or more boosters, and different or novel routes of administration.Citation120–122

The third population that could benefit from A. baumannii vaccination comprises the healthcare workers (HCWs) as they are in general at higher risk of contracting the infection through occupational transmission. A broader grouping could also include the military, volunteers, and humanitarian personnel who visit conflict-affected areas. Potential responses to infection, however, vary in this population group. Although most healthy people may not develop symptoms from A. baumannii infection, they could transmit the bacteria to cause serious health problems to the vulnerable population or they become pre-disposed following tissue injuries.Citation123,Citation124

Summary and prospects

The widespread presence of antibiotic-resistance A. baumannii strains and the lack of new antimicrobials on the horizon, limit the treatment options against the bacterium. Vaccination against the bacterium as an option to tackle the drug-resistance issue has increasingly been emphasized. Significant progress has been made toward it but to date, there is no viable candidate vaccine that has reached the clinical trial phase. The progress in A. baumannii vaccine development is hampered by several challenges. The complex interaction between the virulence factors and the pathogenesis of A. baumannii remains to be fully discovered. The adaptability and plasticity of the genome conferring the multidrug resistance posed a challenge in the selection of the vaccine targets. It may, therefore, be necessary to identify factors that affect the overall function of the bacterium, potentially one that is crucial for its metabolism. This would impact the bacterial replication within the host, allowing the host immune response to overcome the infection, thus, reducing the severity of the infections. While vaccines derived from the bacterial cells are more immunogenic in comparison to the subunit vaccine, the latter may be preferred as it would raise a lesser safety concern. With the successful licensure of menB vaccines, RV presents a highly feasible and attractive approach to identify novel antigenic proteins of A. baumannii for the development of a broadly efficacious vaccine. The application of the RV approach in combination with high throughput in silico screening is foreseen to accelerate the vaccine target selection. Currently, most of the studied A. baumannii vaccines are formulated and tested for administration through the parenteral route. Our understanding of the immune responses against A. baumannii, suggests mucosal route inoculation could provide a better protective response. However, mucosal-based vaccines have several challenges including potentially weak immunogenicity and stability of antigens in the mucosal environment. This could be overcome with a stable expression system such as the spore-based Bacillus subtilis delivery method. Additionally, this platform has a high safety profile, can potentially stimulate mucosal as well as systemic responses, and needle-free route of administration. Moreover, the platform would provide fewer manufacturing and delivery challenges, which could translate into wider availability of the vaccines. Although the recombinant spore platform has yet to be licensed as a candidate vaccine, its potential is worth exploring.

More reliable pneumonia and sepsis animal models are also needed for the development of the A. baumannii vaccine. The current widely used inbred mice model has its limitation, thus, genetic modification is performed to render the mice susceptible to the infection. However, this may alter the critical pathways in the host-pathogen interaction,Citation125 which may not reflect the natural immune responses conferred by the vaccine in humans. Alternatively, rats displayed better characteristics as the animal model for challenge study with live A. baumannii, as it mimics human infection, hence, should be rigorously explored.

Despite the significant threat posed by A. baumannii to global public health, vaccine development for this bacterium still lags behind other Gram-negative pathogens. Adapting the successful approach in identifying the vaccine targets of other bacterial vaccines (eg. 4CMenB vaccine for Neisseria meningitidis) and exploring alternative expression systems and delivery platforms, such as B. subtilis spore, could hasten the vaccine development that would eventually lead to approved A. baumannii vaccine. To ensure success, conscientious and continuous endeavor coordinated and harmonized efforts between the government agencies, academia, and industries are required to accelerate the discovery of alternative treatment to thwart the challenges posed by MDR A. baumannii.

Author contributions

N.M.R., L.H-Y., U.S., and S.A conceived, wrote and edited the manuscript.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Acknowledgments

N.M.R. would like to thank the Director-General of Health, Ministry of Health Malaysia for the Postgraduate Scholarship (Hadiah Latihan Persekutuan).

Additional information

Funding

This work was supported by the Universiti Malaya Tropical Infectious Diseases Research and Education Centre (TIDREC) Malaysia – U.S. Neglected Tropical Infectious Diseases Initiative grant; Ministry of Higher Education Malaysia for niche area research under the Higher Institution Centre of Excellence (HICoE) program [MO002-2019].

References

  • Vallenet D, Nordmann P, Barbe V, Poirel L, Mangenot S, Bataille E, Dossat C, Gas S, Kreimeyer A, Lenoble P, et al. Comparative analysis of acinetobacters: three genomes for three lifestyles. PLoS One. 2008;3(3):e1805. doi:10.1371/journal.pone.0001805.
  • Doughari HJ, Ndakidemi PA, Human IS, Benade S. The ecology, biology and pathogenesis of Acinetobacter spp.: an overview. Microbes Environ. 2011;26(2):101–12. doi:10.1264/jsme2.ME10179.
  • Lee CR, Lee JH, Park M, Park KS, Bae IK, Kim YB, Cha CJ, Jeong BC, Lee SH. Biology of Acinetobacter baumannii: pathogenesis, antibiotic resistance mechanisms, and prospective treatment options. Front Cell Infect Microbiol. 2017;7:55. 2235-2988 (Electronic).
  • Roca I, Espinal P, Vila-Farres X, Vila J. The Acinetobacter baumannii oxymoron: commensal hospital dweller turned pan-drug-resistant menace. Front Microbiol. 2012;3:148. 1664-302X (Electronic). doi:10.3389/fmicb.2012.00148.
  • Hiraki Y, Yoshida M, Masuda Y, Inoue D, Tsuji Y, Kamimura H, Karube Y, Takaki K, Kawano F. Successful treatment of skin and soft tissue infection due to carbapenem-resistant Acinetobacter baumannii by ampicillin-sulbactam and meropenem combination therapy. Int J Infect Dis. 2013;17(12):e1234–e1236. doi:10.1016/j.ijid.2013.05.002.
  • Fournier P-E, Vallenet D, Barbe V, Audic S, Ogata H, Poirel L, Richet H, Robert C, Mangenot S, Abergel C, et al. Comparative genomics of multidrug resistance in Acinetobacter baumannii. PLoS Genet. 2006;2(1):e7. doi:10.1371/journal.pgen.0020007.
  • Howard A, O’Donoghue M, Feeney A, Sleator RD. Acinetobacter baumannii. Virulence. 2012;3(3):243–50. doi:10.4161/viru.19700.
  • Peleg AY, Seifert H, Paterson DL. Acinetobacter baumannii: emergence of a successful pathogen. Clin Microbiol Rev. 2008;21:538–82.
  • Mortensen E, Trivedi KK, Rosenberg J, Cody SH, Long J, Jensen BJ, Vugia DJ. Multidrug-resistant Acinetobacter baumannii infection, colonization, and transmission related to a long-term care facility providing subacute care. Infect Control Hospit Epidemiol. 2014;35(4):406–11. doi:10.1086/675612.
  • Sengstock M, Thyagarajan R, Apalara J, Mira A, Chopra T, Kaye KS. Multidrug-resistant Acinetobacter baumannii: an emerging pathogen among older adults in community hospitals and nursing homes. Clin Infect Dis. 2010;50(12):1611–16. doi:10.1086/652759.
  • Meumann EM, Anstey NM, Currie BJ, Piera KA, Kenyon JJ, Hall RM, Davis JS, Sarovich DS. Genomic epidemiology of severe community-onset Acinetobacter baumannii infection. Microbial Genomics. 2019;5:5. 2057-5858 (Electronic). doi:10.1099/mgen.0.000258.
  • CDC. Antibiotic Resistance Threats in the United States, 2019. Atlanta (GA): U.S. Department of Health and Human Services, CDC; 2019.
  • Jit M, Ng DHL, Luangasanatip N, Sandmann F, Atkins KE, Robotham JV, Pouwels KB. Quantifying the economic cost of antibiotic resistance and the impact of related interventions: rapid methodological review, conceptual framework and recommendations for future studies. BMC Med. 2020;18(1):38–38. doi:10.1186/s12916-020-1507-2.
  • Lee BY, McGlone SM, Doi Y, Bailey RR, Harrison LH. Economic impact of Acinetobacter baumannii infection in the intensive care unit. Infect Control Hospit Epidemiol. 2010;31:1087–89.
  • Shrestha P, Cooper BS, Coast J, Oppong R, Do Thi Thuy N, Phodha T, Celhay O, Guerin PJ, Wertheim H, Lubell Y. Enumerating the economic cost of antimicrobial resistance per antibiotic consumed to inform the evaluation of interventions affecting their use. Antimicrob Resist Infect Control. 2018;7(1):98. doi:10.1186/s13756-018-0384-3.
  • Liu L, Cui Y, Zheng B, Jiang S, Yu W, Shen P, Ji J, Li L, Qin N, Xiao Y. Analysis of tigecycline resistance development in clinical Acinetobacter baumannii isolates through a combined genomic and transcriptomic approach. Sci Rep. 2016 May 31;6:26930. doi:10.1038/srep26930.
  • Manchanda V, Sanchaita S, Singh N. Multidrug resistant Acinetobacter. J Glob Infect Dis. 2010;2(3):291–304. doi:10.4103/0974-777X.68538.
  • Olaitan AO, Berrazeg M, Fagade OE, Adelowo OO, Alli JA, Rolain JM. Emergence of multidrug-resistant Acinetobacter baumannii producing oxa-23 carbapenemase, Nigeria. Int J Infect Dis. 2013;17(6):e469–e470. doi:10.1016/j.ijid.2012.12.008.
  • WorldHealthOrganization. WHO priority pathogens list of  R&D of new antibiotics. In: Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics; Essential Medicines and Health Products Information Portal: World Health Organization; 2017.
  • Wellcome Trust. Tackling drug-resistant infections globally: final report and recommendations / the Review on Antimicrobial Resistance chaired by Jim O'Neill. Wellcome Collection. Attribution 4.0 International (CC BY 4.0).
  • Makri A. Progress lags on vaccines to beat antimicrobial resistance. Lancet. 2019;394(10211):1793–94. doi:10.1016/S0140-6736(19)32758-8.
  • Jansen KU, Knirsch C, Anderson AS. The role of vaccines in preventing bacterial antimicrobial resistance. Nat Med. 2018;24(1):10–19. doi:10.1038/nm.4465.
  • Laxminarayan R, Matsoso P, Pant S, Brower C, Rottingen JA, Klugman K, Davies S. Access to effective antimicrobials: a worldwide challenge. Lancet. 2016;387:168–75. 1474-547X (Electronic). doi:10.1016/S0140-6736(15)00474-2.
  • Morris FC, Dexter C, Kostoulias X, Uddin MI, Peleg AY. The mechanisms of disease caused by Acinetobacter baumannii. Front Microbiol. 2019;10:1601. doi:10.3389/fmicb.2019.01601.
  • Abdollahi S, Rasooli I, Mousavi Gargari SL. The role of tonb-dependent copper receptor in virulence of Acinetobacter baumannii. Infect Genet Evol. 2018;60:181–90. 1567-7257 (Electronic). doi:10.1016/j.meegid.2018.03.001.
  • Stahl J, Bergmann H, Gottig S, Ebersberger I, Averhoff B. Acinetobacter baumannii virulence is mediated by the concerted action of three phospholipases d. PLoS One. 2015;10(9):e0138360. 1932-6203 (Electronic). doi:10.1371/journal.pone.0138360.
  • Dhabaan GN, AbuBakar S, Cerqueira GM, Al-Haroni M, Pang SP, Hassan H. Imipenem treatment induces expression of important genes and phenotypes in a resistant Acinetobacter baumannii isolate. Antimicrob Agents Chemother. 2015;60(3):1370–76. doi:10.1128/AAC.01696-15.
  • Asif M, Alvi IA, Rehman SU. Insight into Acinetobacter baumannii: pathogenesis, global resistance, mechanisms of resistance, treatment options, and alternative modalities. Infect Drug Resist. 2018;11:1249–60. doi:10.2147/IDR.S166750.
  • Gebhardt MJ, Gallagher LA, Jacobson RK, Usacheva EA, Peterson LR, Zurawski DV, Shuman HA. Joint transcriptional control of virulence and resistance to antibiotic and environmental stress in Acinetobacter baumannii. mBio. 2015;6(6):e01660–01615. doi:10.1128/mBio.01660-15.
  • Harding CM, Hennon SW, Feldman MF. Uncovering the mechanisms of Acinetobacter baumannii virulence. Nat Rev Microbiol. 2018;16(2):91–102. doi:10.1038/nrmicro.2017.148.
  • Silhavy TJ, Kahne D, Walker S. The bacterial cell envelope. Cold Spring Harb Perspect Biol. 2010;2(5):a000414. doi:10.1101/cshperspect.a000414.
  • Skerniskyte J, Karazijaite E, Deschamps J, Krasauskas R, Armalyte J, Briandet R, Suziedeliene E. Blp1 protein shows virulence-associated features and elicits protective immunity to Acinetobacter baumannii infection. BMC Microbiol. 2019;19(1):259. doi:10.1186/s12866-019-1615-3.
  • Lee HW, Koh YM, Kim J, Lee JC, Lee YC, Seol SY, Cho DT, Kim J. Capacity of multidrug-resistant clinical isolates of Acinetobacter baumannii to form biofilm and adhere to epithelial cell surfaces. Clin Microbiol Infect. 2008;14(1):49–54. doi:10.1111/j.1469-0691.2007.01842.x.
  • Skerniskyte J, Krasauskas R, Pechoux C, Kulakauskas S, Armalyte J, Suziedeliene E. Surface-related features and virulence among Acinetobacter baumannii clinical isolates belonging to international clones i and ii. Front Microbiol. 2019;9:3116. 1664-302X (Print). doi:10.3389/fmicb.2018.03116.
  • Blanchard C, Barnett P, Perlmutter J, Dunman PM. Identification of Acinetobacter baumannii serum-associated antibiotic efflux pump inhibitors. Antimicrob Agents Chemother. 2014;58(11):6360–70. doi:10.1128/AAC.03535-14.
  • Qin H, Lo NW-S, Loo JF-C, Lin X, Yim AK-Y, Tsui SK-W, Lau TC-K, Ip M, Chan T-F. Comparative transcriptomics of multidrug-resistant Acinetobacter baumannii in response to antibiotic treatments. Sci Rep. 2018;8(1):3515. doi:10.1038/s41598-018-21841-9.
  • Mortensen BL, Rathi S, Chazin WJ, Skaar EP. Acinetobacter baumannii response to host-mediated zinc limitation requires the transcriptional regulator zur. J Bacteriol. 2014;196(14):2616–26. doi:10.1128/JB.01650-14.
  • Gaddy JA, Arivett BA, McConnell MJ, Lopez-Rojas R, Pachon J, Actis LA. Role of acinetobactin-mediated iron acquisition functions in the interaction of Acinetobacter baumannii strain atcc 19606t with human lung epithelial cells, galleria mellonella caterpillars, and mice. Infect Immun. 2012;80(3):1015–24. doi:10.1128/IAI.06279-11.
  • Mortensen BL, Skaar EP. The contribution of nutrient metal acquisition and metabolism to Acinetobacter baumannii survival within the host. Front Cell Infect Microbiol. 2013;3:3. 2235-2988 (Print). doi:10.3389/fcimb.2013.00095.
  • Yamamoto S, Okujo N, Sakakibara Y. Isolation and structure elucidation of acinetobactin, a novel siderophore from Acinetobacter baumannii. Arch Microbiol. 1994;162(4):249–54. doi:10.1007/BF00301846.
  • Proschak A, Lubuta P, Grun P, Lohr F, Wilharm G, De Berardinis V, Bode HB. Structure and biosynthesis of fimsbactins a-f, siderophores from Acinetobacter baumannii and acinetobacter baylyi. Chembiochem. 2013;14(5):633–38. doi:10.1002/cbic.201200764.
  • Penwell WF, DeGrace N, Tentarelli S, Gauthier L, Gilbert CM, Arivett BA, Miller AA, Durand-Reville TF, Joubran C, Actis LA. Discovery and characterization of new hydroxamate siderophores, baumannoferrin a and b, produced by Acinetobacter baumannii. Chembiochem. 2015;16(13):1896–904. doi:10.1002/cbic.201500147.
  • Moynié L, Serra I, Scorciapino MA, Oueis E, Page MG, Ceccarelli M, Naismith JH. Preacinetobactin not acinetobactin is essential for iron uptake by the baua transporter of the pathogen Acinetobacter baumannii. eLife. 2018;7:e42270. doi:10.7554/eLife.42270.
  • Runci F, Gentile V, Frangipani E, Rampioni G, Leoni L, Lucidi M, Visaggio D, Harris G, Chen W, Stahl J, et al. Contribution of active iron uptake to Acinetobacter baumannii pathogenicity. Infect Immun. 2019;87(4). doi:10.1128/IAI.00755-18.
  • Modarresi F, Azizi O, Shakibaie MR, Motamedifar M, Valibeigi B, Mansouri S. Effect of iron on expression of efflux pump (adeABC) and quorum sensing (luxi, luxr) genes in clinical isolates of Acinetobacter baumannii. APMIS. 2015;123(11):959–68. doi:10.1111/apm.12455.
  • Tiwari V, Rajeswari MR, Tiwari M. Proteomic analysis of iron-regulated membrane proteins identify fhue receptor as a target to inhibit siderophore-mediated iron acquisition in Acinetobacter baumannii. Int J Biol Macromol. 2019;125:1156–67. doi:10.1016/j.ijbiomac.2018.12.173.
  • McConnell MJ, Pachon J. Active and passive immunization against Acinetobacter baumannii using an inactivated whole cell vaccine. Vaccine. 2010;29:1–5. 1873-2518 (Electronic). doi:10.1016/j.vaccine.2010.10.052.
  • Bentancor LV, O’Malley JM, Bozkurt-Guzel C, Gb P, Maira-Litrán T. Poly-n-acetyl-beta-(1-6)-glucosamine is a target for protective immunity against Acinetobacter baumannii infections. Infect Immun. 2012;80:651–56. 1098-5522 (Electronic). doi:10.1128/IAI.05653-11.
  • Shu MH, MatRahim N, NorAmdan N, Pang SP, Hashim SH, Phoon WH, AbuBakar S. An inactivated antibiotic-exposed whole-cell vaccine enhances bactericidal activities against multidrug-resistant Acinetobacter baumannii. Sci Rep. 2016;6:22332. 2045-2322 (Electronic). doi:10.1038/srep22332.
  • Sheweita SA, Batah AM, Ghazy AA, Hussein A, Amara AA. A new strain of Acinetobacter baumannii and characterization of its ghost as a candidate vaccine. J Infect Public Health. 2019;12(6):831–42. doi:10.1016/j.jiph.2019.05.009.
  • Das S, Mohakud NK, Suar M, Sahu BR. Vaccine development for enteric bacterial pathogens: where do we stand? Pathog Dis. 2018;76(5). doi:10.1093/femspd/fty057.
  • McConnell MJ, Dominguez-Herrera J, Smani Y, Lopez-Rojas R, Docobo-Perez F, Pachon J. Vaccination with outer membrane complexes elicits rapid protective immunity to multidrug-resistant Acinetobacter baumannii. Infect Immun. 2011;79(1):518–26. doi:10.1128/IAI.00741-10.
  • McConnell MJ, Rumbo C, Bou G, Pachón J. Outer membrane vesicles as an acellular vaccine against Acinetobacter baumannii. Vaccine. 2011;29:5705–10.
  • Huang W, Yao Y, Long Q, Yang X, Sun W, Liu C, Jin X, Li Y, Chu X, Chen B, et al. Immunization against multidrug-resistant Acinetobacter baumannii effectively protects mice in both pneumonia and sepsis models. PLoS One. 2014;9:e100727. 1932-6203 (Electronic). doi:10.1371/journal.pone.0100727.
  • Pulido MR, Garcia-Quintanilla M, Pachon J, McConnell MJ. Immunization with lipopolysaccharide-free outer membrane complexes protects against Acinetobacter baumannii infection. Vaccine. 2018;36(29):4153–56. doi:10.1016/j.vaccine.2018.05.113.
  • Garcia-Quintanilla M, Pulido MR, Pachon J, McConnell MJ. Immunization with lipopolysaccharide-deficient whole cells provides protective immunity in an experimental mouse model of Acinetobacter baumannii infection. PLoS One. 2014;9(12):e114410. doi:10.1371/journal.pone.0114410.
  • Pulido MR, Garcia-Quintanilla M, Pachon J, McConnell MJ. A lipopolysaccharide-free outer membrane vesicle vaccine protects against Acinetobacter baumannii infection. Vaccine. 2020;38(4):719–24. doi:10.1016/j.vaccine.2019.11.043.
  • Moffatt JH, Harper M, Harrison P, Hale JD, Vinogradov E, Seemann T, Henry R, Crane B, Michael FS, Cox AD, et al. Colistin resistance in Acinetobacter baumannii is mediated by complete loss of lipopolysaccharide production. Antimicrob Agents Chemother. 2010;54:4971–77. 1098-6596 (Electronic). doi:10.1128/AAC.00834-10.
  • Miquel-Clopes A, Bentley EG, Stewart JP, Carding SR. Mucosal vaccines and technology. Clin Exp Immunol. 2019;196(2):205–14. doi:10.1111/cei.13285.
  • Micoli F, Costantino P, Adamo R. Potential targets for next generation antimicrobial glycoconjugate vaccines. FEMS Microbiol Rev. 2018;42:388–423.
  • Micoli F, Del Bino L, Alfini R, Carboni F, Romano MR, Adamo R. Glycoconjugate vaccines: current approaches towards faster vaccine design. Expert Rev Vaccines. 2019;18(9):881–95. doi:10.1080/14760584.2019.1657012.
  • Russo TA, Beanan JM, Olson R, MacDonald U, Cox AD, Michael FS, Vinogradov EV, Spellberg B, Luke-Marshall NR, Campagnari AA. The k1 capsular polysaccharide from Acinetobacter baumannii is a potential therapeutic target via passive immunization. Infect Immun. 2013;81:915–22. 1098-5522 (Electronic). doi:10.1128/IAI.01184-12.
  • Chen W. Current advances and challenges in the development of acinetobacter vaccines. Hum Vaccin Immunother. 2015;11(10):2495–500. doi:10.1080/21645515.2015.1052354.
  • Singh JK, Adams FG, Brown MH. Diversity and function of capsular polysaccharide in Acinetobacter baumannii. Front Microbiol. 2018;9:3301. doi:10.3389/fmicb.2018.03301.
  • Ahmad TA, Tawfik DM, Sheweita SA, Haroun M, El-Sayed LH. Development of immunization trials against Acinetobacter baumannii. Trials Vaccinol. 2016;5:53–60. doi:10.1016/j.trivac.2016.03.001.
  • Baxter D. Active and passive immunity, vaccine types, excipients and licensing. Occup Med (Lond). 2007;57(8):552–56. doi:10.1093/occmed/kqm110.
  • Nie D, Hu Y, Chen Z, Li M, Hou Z, Luo X, Mao X, Xue X. Outer membrane protein a (ompa) as a potential therapeutic target for Acinetobacter baumannii infection. J Biomed Sci. 2020;27(1):26. doi:10.1186/s12929-020-0617-7.
  • Luo G, Lin L, Ibrahim AS, Baquir B, Pantapalangkoor P, Bonomo RA, Doi Y, Adams MD, Russo TA, Spellberg B. Active and passive immunization protects against lethal, extreme drug resistant-Acinetobacter baumannii infection. PLoS One. 2012;7:e29446.
  • Zhang X, Yang T, Cao J, Sun J, Dai W, Zhang L. Mucosal immunization with purified OmpA elicited protective immunity against infections caused by multidrug-resistant Acinetobacter baumannii. Microb Pathog. 2016;96:20–25. 1096-1208 (Electronic). doi:10.1016/j.micpath.2016.04.019.
  • Ansari H, Tahmasebi-Birgani M, Bijanzadeh M, Doosti A, Kargar M. Study of the immunogenicity of outer membrane protein a (OmpA) gene from Acinetobacter baumannii as DNA vaccine candidate in vivo. Iran J Basic Med Sci. 2019;22(6):669–75. doi:10.22038/ijbms.2019.30799.7427.
  • Lei L, Yang F, Zou J, Jing H, Zhang J, Xu W, Zou Q, Zhang J, Wang X. DNA vaccine encoding ompa and pal from Acinetobacter baumannii efficiently protects mice against pulmonary infection. Mol Biol Rep. 2019;46(5):5397–408. doi:10.1007/s11033-019-04994-2.
  • Viale AM, Evans BA. Microevolution in the major outer membrane protein OmpA of Acinetobacter baumannii. Microb Genom. 2020:6(6).
  • Huang W, Yao Y, Wang S, Xia Y, Yang X, Long Q, Sun W, Liu C, Li Y, Chu X, et al. Immunization with a 22-kda outer membrane protein elicits protective immunity to multidrug-resistant Acinetobacter baumannii. Sci Rep. 2016;6:20724. 2045-2322 (Electronic). doi:10.1038/srep20724.
  • Singh R, Capalash N, Sharma P. Immunoprotective potential of bama, the outer membrane protein assembly factor, against MDR Acinetobacter baumannii. Sci Rep. 2017;7(1):12411. doi:10.1038/s41598-017-12789-3.
  • Wang-Lin SX, Olson R, Beanan JM, MacDonald U, Balthasar JP, Russo TA. The capsular polysaccharide of Acinetobacter baumannii is an obstacle for therapeutic passive immunization strategies. Infect Immun. 2017;85(12). doi:10.1128/IAI.00591-17.
  • Bentancor LV, Routray A, Bozkurt-Guzel C, Camacho-Peiro A, Pier GB, Maira-Litrán T. Evaluation of the trimeric autotransporter ata as a vaccine candidate against Acinetobacter baumannii infections. Infect Immun. 2012;80:3381–88. 1098-5522 (Electronic). doi:10.1128/IAI.06096-11.
  • Weidensdorfer M, Ishikawa M, Hori K, Linke D, Djahanschiri B, Iruegas R, Ebersberger I, Riedel-Christ S, Enders G, Leukert L, et al. The acinetobacter trimeric autotransporter adhesin ata controls key virulence traits of Acinetobacter baumannii. Virulence. 2019;10(1):68–81. doi:10.1080/21505594.2018.1558693.
  • Li H, Tan H, Hu Y, Pan P, Su X, Hu C. Small protein a and phospholipase d immunization serves a protective role in a mouse pneumonia model of Acinetobacter baumannii infection. Mol Med Rep. 2017;16(2):1071–78. doi:10.3892/mmr.2017.6688.
  • Hubert K, Devos N, Mordhorst I, Tans C, Baudoux G, Feron C, Goraj K, Tommassen J, Vogel U, Poolman JT, et al. Znud, a potential candidate for a simple and universal neisseria meningitidis vaccine. Infect Immun. 2013;81(6):1915–27. doi:10.1128/IAI.01312-12.
  • Qamsari MM, Rasooli I, Chaudhuri S, Astaneh SDA, Schryvers AB. Hybrid antigens expressing surface loops of znud from Acinetobacter baumannii is capable of inducing protection against infection. Front Immunol. 2020;11:158–158. doi:10.3389/fimmu.2020.00158.
  • Rappuoli R. Reverse vaccinology. Curr Opin Microbiol. 2000;3:445–50. 1369-5274 (Print). doi:10.1016/S1369-5274(00)00119-3.
  • Singh R, Garg N, Shukla G, Capalash N, Sharma P. Immunoprotective efficacy of Acinetobacter baumannii outer membrane protein, filf, predicted in silico as a potential vaccine candidate. Front Microbiol. 2016;7. 1664-302X (Print). doi:10.3389/fmicb.2016.00158.
  • Sette A, Rappuoli R. Reverse vaccinology: developing vaccines in the era of genomics. Immunity. 2010;33(4):530–41. doi:10.1016/j.immuni.2010.09.017.
  • Serruto D, Bottomley MJ, Ram S, Giuliani MM, Rappuoli R. The new multicomponent vaccine against meningococcal serogroup b, 4cmenb: immunological, functional and structural characterization of the antigens. Vaccine. 2012;30(Suppl 2):B87–97. 1873-2518 (Electronic). doi:10.1016/j.vaccine.2012.01.033.
  • Ni Z, Chen Y, Ong E, He Y. Antibiotic resistance determinant-focused Acinetobacter baumannii vaccine designed using reverse vaccinology. Int J Mol Sci. 2017;18:E458. 1422-0067 (Electronic). doi:10.3390/ijms18020458.
  • Chiang MH, Sung WC, Lien SP, Chen YZ, Lo AFY, Huang JH, Kuo SC, Chong P. Identification of novel vaccine candidates against Acinetobacter baumannii using reverse vaccinology. Hum Vaccin Immunother. 2015;11:1065–73. 2164-554X (Electronic). doi:10.1080/21645515.2015.1010910.
  • Ahmad S, Azam SS. A novel approach of virulome based reverse vaccinology for exploring and validating peptide-based vaccine candidates against the most troublesome nosocomial pathogen: Acinetobacter baumannii. J Mol Graph Model. 2018;83:1–11. 1873-4243 (Electronic). doi:10.1016/j.jmgm.2018.04.020.
  • Solanki V, Tiwari V. Subtractive proteomics to identify novel drug targets and reverse vaccinology for the development of chimeric vaccine against Acinetobacter baumannii. Sci Rep. 2018;8(1):9044. doi:10.1038/s41598-018-26689-7.
  • Tapia-Calle G, Stoel M, de Vries-idema J, Huckriede A. Distinctive responses in an in vitro human dendritic cell-based system upon stimulation with different influenza vaccine formulations. Vaccine. 2017;5:E21. 2076-393X (Print).
  • Stech M, Nikolaeva O, Thoring L, Stocklein WFM, Wustenhagen DA, Hust M, Dubel S, Kubick S. Cell-free synthesis of functional antibodies using a coupled in vitro transcription-translation system based on cho cell lysates. Sci Rep. 2017;7:12030. 2045-2322 (Electronic). doi:10.1038/s41598-017-12364-w.
  • Dilworth MV, Piel MS, Bettaney KE, Ma P, Luo J, Sharples D, Poyner DR, Gross SR, Moncoq K, Henderson PJF, et al. Microbial expression systems for membrane proteins. Methods. 2018;147:3–39. doi:10.1016/j.ymeth.2018.04.009.
  • Fakruddin M, Mohammad Mazumdar R, Bin Mannan KS, Chowdhury A, Hossain MN. Critical factors affecting the success of cloning, expression, and mass production of enzymes by recombinant E. coli. ISRN Biotechnol. 2013;2013:590587. doi:10.5402/2013/590587.
  • Rosano GL, Ceccarelli EA. Recombinant protein expression in escherichia coli: advances and challenges. Front Microbiol. 2014;5:172. 1664-302X (Print). doi:10.3389/fmicb.2014.00172.
  • Jahangiri A, Rasooli I, Owlia P, Fooladi AAI, Salimian J. An integrative in silico approach to the structure of omp33-36 in Acinetobacter baumannii. Comput Biol Chem. 2018;72:77–86. 1476–928X (Electronic). doi:10.1016/j.compbiolchem.2018.01.003.
  • Gefen T, Vaya J, Khatib S, Rapoport I, Lupo M, Barnea E, Admon A, Heller ED, Aizenshtein E, Pitcovski J. The effect of haptens on protein-carrier immunogenicity. Immunology. 2015;144(1):116–26. doi:10.1111/imm.12356.
  • Meyer H, Schmidhalter D. Microbial Expression Systems and Manufacturing from a Market and Economic Perspective, Innovations in Biotechnology. In: Dr. Eddy C. Agbo editor. InTech. ISBN: 978-953-51- 0096-6. Available from: http://www.intechopen.com/books/innovations-in-biotechnology/microbialexpression-systems-and-manufacturing-from-a-market-and-economic-perspective.
  • White J. Protein expression in corynebacterium glutamicum. Bioprocess J. 2011;9:53–55. doi:10.12665/J92.White.
  • Tavares Batista M, Souza RD, Paccez JD, Luiz WB, Ferreira EL, Cavalcante RCM, Ferreira RCC, Ferreira LCS. Gut adhesive bacillus subtilis spores as a platform for mucosal delivery of antigens. Infect Immun. 2014;82(4):1414–23. doi:10.1128/IAI.01255-13.
  • Lefevre M, Racedo SM, Ripert G, Housez B, Cazaubiel M, Maudet C, Justen P, Marteau P, Urdaci MC. Probiotic strain bacillus subtilis cu1 stimulates immune system of elderly during common infectious disease period: a randomized, double-blind placebo-controlled study. Immun Ageing. 2015;12:24. doi:10.1186/s12979-015-0051-y.
  • Rosales-Mendoza S, Angulo C. Bacillus subtilis comes of age as a vaccine production host and delivery vehicle. Expert Rev Vaccines. 2015;4:1135–48. 1744–8395 (Electronic).
  • Wenzel M, Muller A, Siemann-Herzberg M, Altenbuchner J. Self-inducible bacillus subtilis expression system for reliable and inexpensive protein production by high-cell-density fermentation. Appl Environ Microbiol. 2011;77(18):6419–25. doi:10.1128/AEM.05219-11.
  • Copland A, Diogo GR, Hart P, Harris S, Tran AC, Paul MJ, Singh M, Cutting SM, Reljic R. Mucosal delivery of fusion proteins with bacillus subtilis spores enhances protection against tuberculosis by Bacillus calmette-guérin. Front Immunol. 2018;9:346. doi:10.3389/fimmu.2018.00346.
  • Sun H, Lin Z, Zhao L, Chen T, Shang M, Jiang H, Tang Z, Zhou X, Shi M, Zhou L, et al. Bacillus subtilis spore with surface display of paramyosin from clonorchis sinensis potentializes a promising oral vaccine candidate. Parasit Vectors. 2018;11(1):156. doi:10.1186/s13071-018-2757-0.
  • Sibley L, Reljic R, Radford DS, Huang J-M, Hong HA, Cranenburgh RM, Cutting SM. Recombinant bacillus subtilis spores expressing mpt64 evaluated as a vaccine against tuberculosis in the murine model. FEMS Microbiol Lett. 2014;358(2):170–79. doi:10.1111/1574-6968.12525.
  • Yan Z, Yang J, Hu R, Hu X, Chen K. Acinetobacter baumannii infection and il-17 mediated immunity. Mediators Inflamm. 2016;2016:9834020. doi:10.1155/2016/9834020.
  • Erridge C, Moncayo-Nieto OL, Morgan R, Young M, Poxton IR. Acinetobacter baumannii lipopolysaccharides are potent stimulators of human monocyte activation via toll-like receptor 4 signalling. J Med Microbiol. 2007;56:165–71. 0022-2615 (Print). doi:10.1099/jmm.0.46823-0.
  • Peck A, Mellins ED. Precarious balance: Th17 cells in host defense. Infect Immun. 2010;78(1):32–38. doi:10.1128/IAI.00929-09.
  • Wang S, Liu H, Zhang X, Qian F. Intranasal and oral vaccination with protein-based antigens: advantages, challenges and formulation strategies. Protein Cell. 2015;6(7):480–503. doi:10.1007/s13238-015-0164-2.
  • Rhayat L, Maresca M, Nicoletti C, Perrier J, Brinch KS, Christian S, Devillard E, Eckhardt E. Effect of bacillus subtilis strains on intestinal barrier function and inflammatory response. Front Immunol. 2019;10:564. doi:10.3389/fimmu.2019.00564.
  • Jacobs AC, Thompson MG, Black CC, Kessler JL, Clark LP, McQueary CN, Gancz HY, Corey BW, Moon JK, Si Y, et al. Ab5075, a highly virulent isolate of Acinetobacter baumannii, as a model strain for the evaluation of pathogenesis and antimicrobial treatments. mBio. 2014;5(3):e01076–01014. doi:10.1128/mBio.01076-14.
  • Harris G, Kuo Lee R, Lam CK, Kanzaki G, Patel GB, Xu HH, Chen W. A mouse model of Acinetobacter baumannii-associated pneumonia using a clinically isolated hypervirulent strain. Antimicrob Agents Chemother. 2013;57(8):3601–13. doi:10.1128/AAC.00944-13.
  • Luo G, Spellberg B, Gebremariam T, Bolaris M, Lee H, Fu Y, French SW, Ibrahim AS. Diabetic murine models for Acinetobacter baumannii infection. J Antimicrob Chemother. 2012;67(6):1439–45. doi:10.1093/jac/dks050.
  • Luna BM, Yan J, Reyna Z, Moon E, Nielsen TB, Reza H, Lu P, Bonomo R, Louie A, Drusano G, et al. Natural history of Acinetobacter baumannii infection in mice. PLOS ONE. 2019;14(7):e0219824. doi:10.1371/journal.pone.0219824.
  • Harris G, KuoLee R, Xu HH, Chen W. Acute intraperitoneal infection with a hypervirulent Acinetobacter baumannii isolate in mice. Sci Rep. 2019;9(1):6538. doi:10.1038/s41598-019-43000-4.
  • McConnell MJ, Actis L, Pachon J. Acinetobacter baumannii: human infections, factors contributing to pathogenesis and animal models. FEMS Microbiol Rev. 2013;37(2):130–55. doi:10.1111/j.1574-6976.2012.00344.x.
  • Qiu H, KuoLee R, Harris G, Chen W. High susceptibility to respiratory Acinetobacter baumannii infection in A/J mice is associated with a delay in early pulmonary recruitment of neutrophils. Microbes Infect. 2009;11(12):946–55. doi:10.1016/j.micinf.2009.06.003.
  • Wong D, Nielsen TB, Bonomo RA, Pantapalangkoor P, Luna B, Spellberg B. Clinical and pathophysiological overview of acinetobacter infections: a century of challenges. Clin Microbiol Rev. 2017;30:409–47.
  • Russo TA, Beanan JM, Olson R, MacDonald U, Luke NR, Gill SR, Campagnari AA. Rat pneumonia and soft-tissue infection models for the study of Acinetobacter baumannii biology. Infect Immun. 2008;76(8):3577–86. doi:10.1128/IAI.00269-08.
  • WellcomeTrust, BostonConsultingGroup. Vaccines to tackle drug-resistant infections: An evaluation of R&D opportunities.[Online]. https://vaccinesforamr.org/wp-content/uploads/2018/09/Vaccines_for_AMR.pdf
  • Weinberger B. Vaccines for the elderly: current use and future challenges. Immun Ageing. 2018;15:3. doi:10.1186/s12979-017-0107-2.
  • Gu H, Liu D, Zeng X, Peng LS, Yuan Y, Chen ZF, Zou QM, Shi Y. Aging exacerbates mortality of Acinetobacter baumannii pneumonia and reduces the efficacies of antibiotics and vaccine. Aging. 2018;10(7):1597–608. doi:10.18632/aging.101495.
  • Chen WH, Kozlovsky BF, Effros RB, Grubeck-Loebenstein B, Edelman R, Sztein MB. Vaccination in the elderly: an immunological perspective. Trends Immunol. 2009;30(7):351–59. doi:10.1016/j.it.2009.05.002.
  • Whitman TJ, Qasba SS, Timpone JG, Babel BS, Kasper MR, English JF, Sanders JW, Hujer KM, Hujer AM, Endimiani A, et al. Occupational transmission of Acinetobacter baumannii from a united states serviceman wounded in iraq to a health care worker. Clin Infect Dis. 2008;47(4):439–43. doi:10.1086/589247.
  • Sacadura-Leite E, Mendonça-Galaio L, Shapovalova O, Pereira I, Rocha R, Sousa-Uva A. Biological hazards for healthcare workers: occupational exposure to vancomycin-resistant staphylococcus aureus as an example of a new challenge. Portuguese J Public Health. 2018;36(1):26–31. doi:10.1159/000487746.
  • Gruenheid S, Gros P. Forward genetic dissection of innate response to infection in inbred mouse strains: selected success stories. Clin Exp Immunol. 2010;162(3):393–401. doi:10.1111/j.1365-2249.2010.04249.x.