Open access peer-reviewed chapter

The Battle against Antibiotic Resistance: Novel Therapeutic Options for Acinetobacter baumannii

Written By

Amir Emami, Neda Pirbonyeh and Fatemeh Javanmardi

Submitted: 27 August 2023 Reviewed: 28 August 2023 Published: 23 November 2023

DOI: 10.5772/intechopen.1003617

From the Edited Volume

Acinetobacter baumannii - The Rise of a Resistant Pathogen

Karyne Rangel and Salvatore Giovanni De-Simone

Chapter metrics overview

77 Chapter Downloads

View Full Metrics

Abstract

Undoubtedly, Acinetobacter baumannii stands out as one of the most effective bacteria responsible for nosocomial infections within the healthcare system. Due to its multidrug-resistant nature and the frequency of outbreaks that it causes the treatment of infections caused by this bacterium is challenging, antimicrobial combination therapy has been utilized to treat multidrug resistance Gram-negatives when monotherapy is ineffective. In contrast to antibiotics or short peptides, which possess only the capacity to bind and regulate a specific target, antibodies exhibit supplementary properties attributed to their Fc region, including opsonophagocytic activity, the agglutination process, and activation of the complement system. The criticality of antibodies is exemplified in triggering immunity against A. baumannii, stimulating protective mechanisms, preventing bacterial attachment to epithelial cells, opsonization, and complement-dependent bacterial destruction. Given antibodies’ significant role in humoral immunity, monoclonal antibodies (mAbs) may be generated to specifically bind to certain targets, thereby providing supplemental defense as a form of immunotherapy or passive immunization. Many encouraging tactics, ranging from phage therapy to immunotherapy, are being scrutinized for their efficacy in treating infectious diseases, thus shaping the future treatment landscape.

Keywords

  • antimicrobial peptides
  • bacteriophage therapy
  • drug repurposing
  • nanoparticles
  • MDR

1. Introduction

Bacterial infections are the leading cause of death worldwide. Although the discovery of antibiotics successfully controlled bacterial infections, overuse and misuse of antimicrobial agents exacerbated the selection of multidrug-resistant (MDR) organisms. Acinetobacter baumannii is a bacteria that increases infection and mortality in vulnerable patients due to its ability to escape from antibiotic treatments effectively.

The increasing prevalence of nosocomial A. baumannii infections can be largely attributed to the remarkable ability of A. baumannii to colonize and form biofilms. Treatment options for these highly resistant pathogens are very limited. Because of this, clinicians are forced to resort to last-resort antibiotics, including colistin, which may induce nephrotoxicity, and select colistin-resistant A. baumannii.

To effectively treat and limit the spread of MDR A. baumannii (MDR-AB), a thorough understanding of the bacterial virulence factors and host-pathogen interactions is crucial. Therefore, before dealing with the new methods of treating A. baumannii, there is a need for a brief explanation to clarify the interactions between the host and the pathogen.

Advertisement

2. Virulence factors

A. baumannii, a highly antibiotic-resistant pathogen, possesses several virulence factors contributing to its pathogenesis and high mortality rates. Several recent studies have investigated virulence factors associated with the pathogenesis of A. baumannii and could thus serve as novel therapeutic targets.

These factors include the capsular polysaccharide (k-type), a major virulence factor [1]. The prevalent capsular types of A. baumannii include KL2, KL10, KL14, KL22, and KL52, with KL2 being associated with higher drug resistance and virulence [2].

Other virulence factors of A. baumannii include outer membrane proteins (Omps), lipopolysaccharide (LPS), capsular polysaccharide (CPS), phospholipase, nutrient-acquisition systems, efflux pumps, protein secretion systems, quorum sensing, and biofilm production (Table 1) [3]. Understanding these virulence factors is crucial for developing novel therapeutic targets and strategies to combat this multidrug-resistant pathogen [4, 5].

Virulence factorFunctionsModulation
ompsInduce cell apoptosis, complement resistance, biofilm formation, cell invasion, and OMV biogenesis.Unknown
CPSComplement resistance and biofilm formationUp-regulated upon antibiotic or ROS exposure
OMVsTransferring OmpA and toxin deliveryUp-regulated upon antibiotic exposure
LPSMembrane integrity, induce cell apoptosis, and antibiotic resistanceLoss during colistin resistance development
T6SSInterspecies competitionActivate upon contact with competing bacteria
Micronutrient acquisition systemsNutrient acquisitionUp-regulated under nutrient-deprived conditions
Type IV piliTwitching motilityUp-regulated during growth in human serum
BapBiofilm formationUp-regulated while growing under low iron conditions
Csu PiliBiofilm formationAntibiotic exposure

Table 1.

A. baumannii components, functions, and regulation conditions of them.

2.1 The mechanisms of A. Baumannii to promote self-survival

A. baumannii outer membrane proteins (Omps) play a versatile role in promoting bacterial survival. Omps in A. baumannii facilitate bacterial acclimatization to antibiotic- and host-induced stresses, aiding in immune evasion, stress tolerance, and resistance to antibiotics and antibacterial [6]. The ability of A. baumannii to adhere to abiotic surfaces and form biofilms, facilitated by Omps, and helps the bacteria survive in harsh environmental conditions such as desiccation, nutrient deficiency, and antibiotic treatment [7]. Additionally, A. baumannii outer membrane vesicles (OMVs), which contain Omps, contribute to the delivery of virulence factors to host cells, enhancing bacterial survival, nutrient acquisition, biofilm formation, and pathogenesis. A. baumannii strains that produce more abundant Omps, such as MDR-AB, exhibit more powerful cytotoxicity, stronger innate immune responses, and contain more virulence factors, potentially leading to worse outcomes [8].

LPS is the main component on the extracellular membrane of Gram-negative bacteria [9]. Mutations in the lipid A biosynthetic pathway can lead to changes in the structure of LPS in A. baumannii, resulting in colistin resistance. LPS-deficient A. baumannii strains show altered activation of the host innate immune inflammatory response, indicating the importance of LPS in interacting with host immune system. In addition, LPS-deficient A. baumannii can have alterations in their lipid A composition, such as the addition of phosphoethanolamine (pEtN) and galactosamine (GalN), which can affect the binding affinity of colistin. Loss of LPS in A. baumannii can lead to the upregulation of lipoproteins and the accumulation of the capsular polysaccharide poly-β-1,6-N-acetylglucosamine as compensatory mechanisms for membrane stabilization.

Capsular polysaccharides (CPS) in A. baumannii play a crucial role in bacterial virulence and survival [10]. The CPS structures in A. baumannii are diverse and can vary between strains [5]. These CPS structures often include rare sugars and branched oligosaccharide repeating units. The CPS biosynthesis gene encodes glycosyltransferases that are responsible for the synthesis of CPS structures. The presence of specific CPS structures, such as KL2, has been associated with antibiotic resistance and clinical outcomes in A. baumannii infections. Understanding the CPS structures and the genetics involved in their synthesis is important for developing targeted treatment strategies against A. baumannii infections [11].

Phospholipase functions of A. baumannii play a crucial role in promoting bacterial survival. Multiple studies have identified phospholipases as virulence factors that contribute to the pathogenicity of A. baumannii. The phospholipases are involved in the growth of phosphatidyl choline as a carbon source. These phospholipases involve various processes, such as hemolytic and cytolytic activities [12]. The phospholipases enable A. baumannii to adapt to different host niches and environments, enhance resistance to antimicrobial peptides, and facilitate the invasion of host cells [13].

Nutrient acquisition systems are often crucial for pathogen growth and survival during infection and represent attractive therapeutic targets. The pathogen utilizes various mechanisms to acquire essential nutrients from the host, such as heme and zinc.

A. baumannii has metal homeostatic systems that regulate the levels of essential nutrient metals in bacteria, particularly iron and zinc, that are important for colonizing different tissues and growth within vertebrates [14]. These systems, such as siderophores, heme uptake systems, and zinc uptake systems, enable the bacteria to overcome host-imposed zinc limitation by aiding in zinc uptake into the cells. The hemO locus, including the heme-degrading enzyme and scavenger, is required for high-affinity heme acquisition from host hemoglobin and serum albumin [15]. Additionally, A. baumannii possesses a Zn uptake (Znu) system consisting of an inner membrane ABC transporter and an outer membrane TonB-dependent receptor, which allows the pathogen to overcome host-imposed Zn limitation [16]. The TonB-dependent receptor HphR is an important component of the heme uptake system in A. baumannii and is involved in iron acquisition and cellular processes contributing to virulence [17].

Efflux pumps, such as the RND-type efflux pumps AdeABC and AdeIJK, contribute to resistance against antibiotics and biocides [18]. They are involved in extruding hazardous substances, including antibiotics, from within the bacterial cells [19]. The overexpression of these efflux pumps, particularly AdeABC, has been found to enhance the survival of A. baumannii when exposed to residual concentrations of biocides [20]. Additionally, efflux pump genes, such as adeABC, have been associated with tigecycline resistance in A. baumannii [21]. Efflux pumps have broad substrate specificity and are widely distributed among bacterial species, making them a major contributor to multidrug resistance in A. baumannii [22].

Secretion systems have recently been demonstrated to be involved in the pathogenic process, and five types of secretion systems out of the currently known six from Gram-negative bacteria have been found in A. baumannii. They can promote the bacteria’s fitness and pathogenesis by releasing various effectors. Additionally, antibiotic resistance is found to be related to some types of secretion systems [23]. The type VI secretion system (T6SS) is one such system found in A. baumannii, which is involved in bacterial competition and the delivery of toxic effector proteins [2425]. The T6SS in A. baumannii is highly diverse, with significant diversity in the range of encoded T6SS VgrG and effector proteins. There are multiple VgrG genes in A. baumannii strains, with most strains encoding between two and four different VgrG proteins. T6SS structural components of A. baumannii are distinctive from other Gram-negative pathogens, as evidenced by the presence of the Acinetobacter genus-specific protein AsaA. The T6SS in A. baumannii is involved in bacterial competition and secretion of T6SS effectors, such as Hcp is associated, which acts as a virulence factor, transporter of effectors, and chaperone [26]. The putative T6SS effectors in A. baumannii have diverse functions, including peptidoglycan hydrolases, lipases, nucleases, and nucleic acid deaminases [27].

Quorum sensing (QS) in A. baumannii plays a crucial role in bacterial survival and pathogenicity [28]. The QS system coordinates the behavior of individual bacteria in a population by mediating the synthesis, secretion, and binding of auto-inducer signals. The deletion of the auto-inducer synthase gene abaI in A. baumannii resulted in a decrease in biofilm formation and pathogenicity [28]. Additionally, the QS system regulates important virulence-related phenotypes, such as surface-associated motility and biofilm formation [29]. The antibacterial peptide octopromycin inhibited biofilm formation and surface movements in A. baumannii, demonstrating its anti-quorum sensing activity [30]. Furthermore, the abaI/abaR QS system was found to affect growth characteristics, morphology, biofilm formation, resistance, motility, and virulence in A. baumannii [31].

Targeting virulence factors can be an effective strategy for combating A. baumannii and other multidrug-resistant bacteria. Additionally, the development of innovative strategies, such as using bacteriophages and antibiotics in combination, has shown increased efficacy in eradicating biofilms formed by antibiotic-resistant A. baumannii strains. So, targeting virulence factors and biofilm formation can be an effective approach for designing drugs to combat multidrug-resistant bacteria.

Advertisement

3. Antimicrobial drug resistance and overcoming its problems

Antimicrobial drug resistance is one of the three major global threats to public health identified by the World Health Organization (WHO) in the twenty-first century [32]. A. baumannii is one of the main and most successful pathogens responsible for hospital-acquired infections in the modern healthcare system, associated with high mortality rates [33]. According to the reports of the WHO, about 80% of MDR or extensively drug-resistant (XDR) microbes have occurred due to the misuse and overuse of antibiotics, and these infections are associated with severe side effects [34]. Due to the prevalence of infections and outbreaks caused by A. baumannii drug resistance, few antibiotics are effective for treating infections caused by this pathogen [35]. Due to the spread of MDR bacteria and other resistant pathogens, there are limited treatment and prevention options, the failure of most antibiotics necessitates the search for better treatment options, and the need for alternative treatment options to treat these microbial pathogens (Figure 1).

Figure 1.

Newer approaches to tackle of MDR A. baumannii.

3.1 Combined treatment

Antibiotics, such as colistin, carbapenems, and tigecycline, have been widely used to treat A. baumannii [36]. However, the emergence of this bacterium’s multidrug-resistant strains has limited these drugs’ effectiveness. Therefore, new treatment options like combination therapy are an urgent need. The idea behind combination therapy is to use two or more antibiotics to kill the bacteria, which works in different ways [37]. In fact, in this method, two antibiotics with other mechanisms of action are used, such as a β-lactam antibiotic and an aminoglycoside, or two antibiotics that have the same means of action but work on different targets, such as two different carbapenems [38].

There are several notable advantages to using combination therapy to treat A. baumannii. One of these advantages is increasing the effectiveness of treatment [39]. Bacteria are attacked from different angles, making it more difficult for bacteria to develop resistance using multiple drugs. Another advantage is that combination therapy can reduce the risk of treatment failure [40]. Since A. baumannii is very resistant to antibiotics, using one antibiotic may not be effective in treating the infection, using multiple antibiotics reduces the chance of treatment failure, using lower doses of each antibiotic can reduce the risk of side effects in patients.

For example, pairing β-lactam antibiotics with β-lactamase inhibitors has proven effective in combating resistant strains. Additionally, combining antibiotics with different mechanisms of action can target multiple bacterial pathways, increasing treatment efficacy [41].

Combination therapy, including colistin/imipenem, colistin/meropenem, colistin/rifampicin, colistin/teicoplanin, colistin/sulbactam, colistin/tigecycline, and imipenem/sulbactam has been widely studied [42].

Combination therapy has been explored as a potential treatment option for A. baumannii infections. Various combinations have been studied, including polymyxins, rifampicin, fosfomycin, sulbactam, and avibactam. Polymyxin-based combinations, such as with cell-wall acting agents, rifamycins, and fosfomycin, have been extensively studied [43, 44]. Berberine hydrochloride (BBH) has shown synergistic effects with antibiotics against MDR A. baumannii (MDR-AB), including tigecycline, sulbactam, meropenem, and ciprofloxacin [45]. High-dose sulbactam, combined with additional antibacterial agents, including colistin, has shown promise in treating MDR-AB or XDR A. baumannii (XDR-AB) infections [46]. Fosfomycin has also been explored as a potential component of combination therapy against carbapenem-resistant A. baumannii (CR-AB) conditions [47, 48].

The combination of colistin and tigecycline is effective in the treatment of A. baumannii infection that causes pneumonia by ventilator, and the combined therapy of colistin, meropenem, and ampicillin-sulbactam in A. baumannii infection in patients. It was effective in treating blood malignancy [49, 50, 51]. The combined treatment of colistin/rifampicin and ampicillin/sulbactam/carbapenem combination therapy is effective for the treatment of A. baumannii MDR bacteria, causing carbapenem-resistant skin and soft tissue infections [52].

3.2 Repurposing

Repurposing existing drugs is also considered a strategy for treating MDR bacterial infections. Repurposing drugs, drug repositioning, or therapeutic switching is like giving a second life to medication previously used for different purposes [53].

Instead of starting from scratch, drug repurposing allows researchers to tap into a vast library of already approved drugs, saving time and resources in drug development [54]. Moreover, low risk of failure, shorter time frame cycles, high success rates, and less investment are the practicalities of drug repurposing. These drugs have undergone rigorous safety and efficacy testing, making them attractive candidates for new applications [55].

Drug repurposing has emerged as a promising approach to combating drug-resistant A. baumannii infections. Several FDA-approved drugs have shown potential for repurposing in treating A. baumannii. Etoposide and genistein inhibit the synthesis of polyphosphates, a virulence factor in A. baumannii (Table 2) [56, 57, 58]. 5-fluorouracil (5-FU), fluspirilene, and Bay 11–7082 were identified as drugs that resensitize A. baumannii to azithromycin and colistin in combination [59, 60]. Erythromycin, levamisole, chloroquine, and propranolol inhibit quorum sensing and virulence factors in A. baumannii [61, 62]. Tyrothricin, typically active against Gram-positive bacteria, exhibited antimicrobial activity against drug-resistant A. baumannii [63, 64].

CompoundActivity-alone or in combination withApproved use or known as
Central Nervous SystemCitalopramPolymyxin BAntidepressant
FluspirileneColistinAntipsychotic
InfectiologyApramycinAloneAntibacterial
NiclosamideColistinAnti-helminthic
OxyclozanideAloneAnti-helminthic
RafoxanideAloneAnti-helminthic
IvermectinAloneAnti-parasitic
ZidovudineAloneAntiretroviral
CiclopiroxAloneAntifungal
MetabolismEbselenAloneAnti-inflammatory
Bay 11–7082ColistinAnti-inflammatory
Natural CompoundResveratrolColistinStilbene
OncologyMitomycin CAloneAnti-tumor
TamoxifenAloneBreast cancer
5-Fluorouracil (5-FU)ZidovudineAntineoplastic (Colon Cancer)
MitotanePolymyxin BAntineoplastic
GalliumAloneAntineoplastic
ToremifeneAloneBreast cancer

Table 2.

Relevant repurposing reports for MDR-AB.

Apramycin, Niclosamide, Oxyclozanide, Rafoxanide, and Ciclopirox are antibacterial, antifungal, and anthelmintic agents that have a therapeutic effect on MDR-AB.

Apramycin is an aminoglycoside approved for veterinary use. Apramycin can potentially be used against highly drug-resistant pathogens [65]. Niclosamide is an anthelmintic drug that has been commercially available in some countries since the 1960s. Niclosamide is usually administered orally and is well absorbed by the intestinal mucosa. High doses of this drug are associated with serious side effects. This drug has recently been suggested to treat other diseases, such as cancer [66].

Niclosamide alone has no antibacterial activity against A. baumannii, but a synergistic interaction between Niclosamide and colistin has been observed against CR-AB. This drug interacts with colistin-resistant strains negatively charged outer membrane, leading to a synergistic effect with colistin.

Oxyclozanide is used in veterinary medicine to treat fluke infections in ruminants. Oxyclozanide enhances the effect of colistin on colistin-sensitive and resistant isolates of A. baumannii, P. aeruginosa, and K. pneumoniae. This effect may be due to disrupting of the bacterial cell envelope [67].

Rafoxanide at a suitable dose had histidine kinase-antagonistic activities, which disrupted the abilities of MDR bacterial and fungal cells to adapt to stress conditions [68].

Ciclopirox, an antifungal drug, has bacteriostatic activity against E. coli, K. pneumonia, and A. baumannii of MDR strains [69]. The ciclopirox mechanism of action is the effect on the galactose and LPS salvage pathways [70].

Mitomycin C, tamoxifen, 5-FU, mitotane, and gallium include anti-tumor and antineoplastic drug agents that have a therapeutic effect on MDR-AB. The anticancer drug Mitomycin C can kill A. baumannii exponential-phase, stationary-phase, and biofilm cells [71].

The tamoxifen metabolites were active against MDR Gram-negative bacilli and might be potential antimicrobial agents to treat infections by these pathogens [72]. Colistin combination therapy with selective estrogen receptor modulators (SERM) as tamoxifen, raloxifene, and toremifen also exhibited good activity against polymyxin-resistant P. aeruginosa, K. pneumonia, and A. baumannii [73].

5-FU, another anticancer drug, despite the overall safety of 5-FU, is toxic in some cases, with toxicities including gastrointestinal (e.g., diarrhea, nausea, vomiting, mucositis/stomatitis, anorexia), hematological (e.g., neutropenia, thrombocytopenia, anemia), and dermal (e.g., hand-foot syndrome) symptoms [74]. The combination of 5-FU with azithromycin was effective against CR-AB; this combination, possibly reducing 5-FU toxicity, has also been found to inhibit the growth of bacterial pathogens and reduce the production of virulence factors [66].

Mitotane, an antineoplastic agent approved for cancer treatment, acts with polymyxin B on carbapenem- or polymyxin-resistant GNB in vitro. These efflux pump inhibitors alone did not affect the bacteria, but their activity was restored when combined with an antibiotic [70].

Gallium’s antibacterial activity dates back many years, but this drug was originally used as an anticancer agent. Due to its chemical similarity to iron, gallium inhibits the reactions or redox pathways of iron and the growth of bacteria [75]. Therefore, gallium compounds show broad-spectrum antibacterial activity and inhibit the growth of important bacterial pathogens such as A. baumannii, P. aeruginosa, S. aureus, K. pneumoniae, and E. coli [76].

3.3 New antibiotics

Acinetobacter is one of the ESKAPE pathogens known for their ability to escape commonly used antibacterial treatments. With the rise of antibiotic resistance and the limited efficacy of current therapeutic options, exploring novel antibiotics offers hope in combating A. baumannii infections. New antibiotics have been developed and tested for treating MDR bacterial strains. These include cephalosporins and carbapenems in combination with new β-lactamase inhibitors, tetracycline derivatives, fourth-generation fluoroquinolones, new combinations of β-lactam and β-lactamase inhibitors, siderophore cephalosporins, and new aminoglycosides that have been approved or are in clinical development [77, 78].

Novel siderophore cephalosporins antibiotics, such as cefiderocol (CFDC) [79] and GT-1 (LCB10–0200), have shown promise for the treatment of A. baumannii infections. CFDC demonstrates strong activity against MDR-AB isolates with lower minimum inhibitory concentration (MIC) values than other Gram-negative agents [80]. GT-1, combined with a β-lactamase inhibitor GT-055, has shown efficacy against many multidrug-resistant pathogens, including A. baumannii [81]. These novel siderophore cephalosporins utilize a “trojan-horse approach” to evade resistance mechanisms in Gram-negative bacteria [82]. However, available clinical data for cefiderocol are conflicting, leaving infectious disease specialists uncertain about its optimal use in clinical practice.

New tetracycline antibiotics, such as eravacycline and TP-6076, have shown promise for treating Acinetobacter infections. Eravacycline has demonstrated higher potency than tigecycline and has been effective against XDR-AB in vitro [83]. It has also been found to have a low propensity to induce Clostridioides difficile infection (CDI) [84]. TP-6076, a fully synthetic fluorocycline, has shown greater activity than other tetracycline-class antimicrobials against CR-AB isolates [85]. These novel tetracyclines can be valuable additions to the limited armamentarium of drugs targeting Acinetobacter [86].

Non-β-lactam β-lactamase inhibitors antibiotics have shown promise for the treatment of Acinetobacter infections. The etx2514/sulbactam combination has demonstrated efficacy against MDR-AB isolates, including those producing class D β-lactamases [87]. The zidebactam/cefepime combination has shown in vitro activity against CR-AB [88]. Wck 4234/meropenem combination has exhibited broad-spectrum activity against MDR Enterobacteriaceae, including NDM, KPC, OXA, CTX-M, SHV, and TEM enzyme-producing isolates [89]. Ln-1-255/meropenem-imipenem combination has demonstrated decreased resistance rates against CR-AB isolates [90]. However, non-β-lactam β-lactamase inhibitors for treating Acinetobacter infections are still ongoing research and development. Further, clinical data is needed to support the efficacy of these inhibitors, and gaps still exist in the treatment of infections caused by MDR Acinetobacter spp.

Novel β-lactam antibiotics, such as AIC-499 and FSI-1671, combined with sulbactam have shown promise for treating Acinetobacter infections. AIC-499 is a member of the diazabicyclooctane class of β-lactamase inhibitors with broad-spectrum activity against Ambler class A, C, and D serine β-lactamases [87]. Sulbactam, a first-generation β-lactamase inhibitor, has limited action against Acinetobacter spp. due to susceptibility to cleavage by β-lactamases [91]. However, when combined with durlobactam, the activity of sulbactam is effectively restored against MDR Acinetobacter strains [92]. FSI-1671, in combination with sulbactam, has also shown enhanced antimicrobial activity against A. baumannii clinical strains in China, with cefoperazone-sulbactam as the most potent compound.

Novel polymyxin B-derived molecules, such as SPR741 and FADDI-287, have shown potential for treating A. baumannii. SPR741 has been found to potentiate several large scaffold antibiotics in Gram-negative pathogens by interacting predominantly with the outer membrane (OM) [93]. FADDI-287, on the other hand, has been shown to induce significant perturbation in glycerophospholipid metabolism and histidine degradation pathways, leading to synergistic bacterial killing in both polymyxin-susceptible and resistant A. baumannii [94]. These findings suggest that these novel polymyxin B-derived molecules can overcome resistance mechanisms and enhance the efficacy of antibiotics against A. baumannii.

A new aminoglycoside called apramycin (EBL-1003) has promising potential for treating A. baumannii infections. It has proven to have wide antibacterial action against A. baumannii strains resistant to various drugs, including standard-of-care aminoglycosides [95, 96]. Because of its distinct chemical makeup, apramycin can circumvent resistance mechanisms frequently present in clinical isolates that produce carbapenemase [97]. Apramycin is quickly bactericidal against A. baumannii according to in vitro experiments [98]. In a mouse lung infection model, apramycin was also discovered to have a high likelihood of target attainment and robust in vivo activity [99]. According to these findings, apramycin may be a potent therapeutic alternative for treating CR-AB lung infections linked to high mortality rates and few therapeutic options.

3.4 Bacteriophage

A. baumannii infections may be treated using bacteriophage therapy, particularly when there is MDR. Bacteriophages are viruses that can target and eradicate particular types of bacteria. The effectiveness and safety of phage therapy against A. baumannii infections have been demonstrated in several studies [91, 100, 101]. Other Gram-negative and Gram-positive bacteria, such as S. aureus, P. aeruginosa, K. pneumoniae, Enterococcus, and Salmonella, have been demonstrated to be sensitive to phage therapy. The effectiveness of phages can be assessed by evaluating their host range, adsorption rate, and growth curve from various sources, such as sewage or wastewater [102]. Bacteriophages, particularly lytic ones, have shown promise as anti-A. baumannii therapeutics [103]. Both options are using phages in monophage therapy, phage cocktails, or in conjunction with antibiotics [104]. Bacteriophages can be administered parenterally, orally, topically, or by inhalation [105]. Endolysins and depolymerases, two phage-derived enzymes, have also been investigated for use against A. baumannii [106]. Antimicrobial therapy can use specific lytic bacteriophages or enzymes generated from phages to treat infections brought on by strains of A. baumannii that are incredibly drug-resistant.

Bacteriophages have multiple mechanisms of action against Acinetobacter. Some phages produce depolymerase, such as the tail spike proteins of phages Fri1, AS12, BS46, and AP22, which specifically recognize and digest the capsular polysaccharide (CPS) of A. baumannii. Other phages can cause mutations in genes that alter the architecture of the bacterial envelope, leading to phage resistance but also increased sensitivity to antibiotics, such as colistin [107]. Additionally, phages can degrade biofilms formed by A. baumannii, as demonstrated by the bacteriophage AB3 and its endolysin LysAB3 [106]. Further, phage therapy can target specific mechanisms of antimicrobial resistance, such as efflux pumps, by using efflux pump inhibitors or phage steering [108]. So, phage therapy, including phage cocktails and combination therapy with antibiotics, as well as phage-derived enzymes, such as endolysins and depolymerase, shows promise in combating MDR-AB [109].

But, bacteriophage therapy for Acinetobacter infections has limitations that must be addressed. Firstly, there needs to be more reliable safety and efficacy data for phage therapy due to the heterogeneity in previously published studies [110]. Secondly, the systemic effects of phage therapy need to be better understood [111]. Thirdly, the optimal application protocol for phage therapy, including the route of administration, frequency of administration, treatment duration, and phage titer, is still [112]. Additionally, the concurrent ecological and evolutionary interplay between phages and host bacteria requires further research to utilize the potential of bacteriophage therapy [103].

3.5 Antimicrobial peptides

Antimicrobial peptides (AMPs) have been demonstrated to prevent the MDR bacteria A. baumannii from growing. AMPs with a strong antibacterial action against A. baumannii strains include PapMA-3, MSI-78, h-Lf1–11, magainin-2, and LL-37. These peptides exhibit potential as antibacterial agents for treating bacteria that are resistant to antibiotics and have a broad spectrum of antimicrobial action [113].

Multiple mechanisms explain how AMPs work against Acinetobacter. The bacterial membrane may be damaged by AMPs, which will cause cell lysis [114, 115]. Additionally, they can infiltrate bacterial cells and engage with internal elements [116]. The mode of action of AMPs can be identified using experimental biophysical methods with model membranes and bacterial cells [117]. AMPs can cause the Acinetobacter membrane to become permeable [113]. The membrane’s interaction with AMPs and lipids may result in soft supramolecular configurations, which can thin and lyse the membrane [118].

A hybrid peptide called PapMA-3 showed low cytotoxicity and strong bacterial selectivity against carbapenem-resistant bacteria [119]. A. baumannii was also significantly resistant to the antibacterial effects of MSI-78, h-Lf1–11, magainin-2, and LL-37 [120, 121]. The anti-A. baumannii potency of Melittin, Histatin-8, Omega76, AM-CATH36, Hymenochirin, (this peptide showed moderate activity against Gram-negative bacteria), and Mastoparan was the highest [113]. In animal models of A. baumannii-induced pneumonia, the AMP derivatives dN4 and dC4 have shown therapeutic effectiveness [119, 120, 121, 122]. Furthermore, it has been demonstrated that larger doses of dN4 and dC4 can suppress and/or remove Acinetobacter biofilms [119].

A. baumannii, the cyclic peptide ZY4, exhibited little potential to induce resistance and outstanding efficacy against A. baumannii, including MDR strains [123]. According to these results, the antimicrobial peptides PapMA-3, MSI-78, h-Lf1–11, magainin-2, LL-37, Melittin, Histatin-8, Omega76, AM-CATH36, Hymenochirin, Mastoparan, dN4, and dC4 are efficient in treating Acinetobacter infections.

For the treatment of Acinetobacter infections, AMPs have limitations. Their toxicity and stability in vivo are two drawbacks that restrict their use [124]. Another drawback is their inherent limitations as peptides, such as stability, cytotoxicity, and bioavailability [125]. Natural AMPs are only useful for topical applications due to their pharmacological characteristics [126]. However, efforts are being made to get around these restrictions by creating novel AMPs and peptidomimetics by clever chemical changes [127]. Despite these drawbacks, AMPs hold potential as alternative therapies for focusing on bacterial infections, such as Acinetobacter, in both extracellular and intracellular contexts [113].

3.6 Monoclonal antibodies

Monoclonal antibodies (MAbs) are synthetic proteins, replicating the immune system’s defense against pathogens like bacteria and cancer cells. These antibodies target molecules on the pathogen’s surface known as antigens. The distinguishing quality of MAbs is their specificity, which enables them to recognize and bind to a certain target with extreme accuracy [128].

Heavy chains and light chains, two different protein chains, make up mAbs. These chains come together to form a Y-shaped structure. The antibody’s variable region, found at the end of each Y-shaped arm and binds to the particular antigen [129]. On the other hand, the antibody’s constant region controls its effector actions, such as triggering the immune system or obstructing the pathogen’s activity [130, 131].

MAbs can use different pathways to exert their therapeutic effects. The process of neutralization, in which antibodies bind to the pathogen and stop it from infecting host cells, is a typical one. In order to enlist immune cells in the fight against the disease, antibodies can potentially trigger antibody-dependent cellular cytotoxicity (ADCC). Furthermore, MAbs can influence the immune system’s response, enhancing the body’s ability to eliminate the infection.

MAbs have shown promise as novel therapeutics for Acinetobacter infections. These antibodies targeting outer membrane protein A (OmpA) of A. baumannii have improved opsonophagocytic killing of the bacteria [132]. Another MAb targeting the capsule of A. baumannii has been found to enhance macrophage opsonophagocytosis and reduce pro-inflammatory cytokines, leading to improved survival in mouse models [133]. A second MAb, developed through hybridoma technology, has also been shown to enhance macrophage opsonophagocytosis and improve survival in murine models of A. baumannii infection alone and combination with antibiotics [134]. Furthermore, MAbs have the advantage of being a narrow spectrum, targeting only the pathogenic species and potentially avoiding microbiome disruption. Additionally, a human MAb targeting a DNABII epitope has demonstrated efficacy in disrupting biofilms formed by Gram-positive and Gram-negative bacteria, including A. baumannii [135]. These findings suggest that MAbs have the potential to be effective therapeutic options for Acinetobacter infections, either alone or in combination with antibiotics.

Modulation of pro- and anti-inflammatory cytokines, such as IL-1, IL-6, TNF, and IL-10, was necessary for MAb treatment to be effective [136]. The FDA has approved three antibacterial MAb medicines, and numerous others are undergoing clinical studies [137].

Treating A. baumannii infections has led to the development of the MAbs C8 and 65. In deadly bacteremic sepsis and aspiration pneumonia models of XDR-AB infection, MAb C8 improves opsonophagocytosis by focusing on the capsular carbohydrate on the bacterial surface [134]. On the other hand, MAb 65 is extremely powerful and effective while expanding the coverage of immunotherapeutic strains. Combined with antibiotics, such as colistin, it improves macrophage opsonophagocytosis and results [135]. These MAbs have demonstrated the ability to decrease cytokine production, blood bacterial density, and sepsis biomarkers, showing their therapeutic potential [132, 133]. Antibacterial MAb treatment works by regulating pro- and anti-inflammatory cytokines and improving germ clearance by opsonophagocytosis.

AR401-mAb is a monoclonal antibody developed for the treatment of A. baumannii infections. It is highly effective against a broad range of clinical isolates and has been shown to improve outcomes when combined with antibiotics [134]. AR401- MAb is synergistic with colistin, a commonly used antibiotic, further enhancing its protective effects [138]. These findings suggest that using MAbs, such as AR-401, in treating A. baumannii infections may effectively improve outcomes and reduce bacterial burden.

Another study produced MAbs against the outer membrane protein A (OmpA) of A. baumannii, with one MAb, 1G1-E7, showing high reactivity and opsonophagocytic killing activity [132].

3.7 Nanoparticles

Nanoparticles are exceedingly small particles, usually measuring between 1 and 100 nanometers. Various substances, including metals, metal oxides, lipids, and polymers, can be used to create them. Nanoparticles differ from their bulk counterparts in multiple ways due to their small size and frequently show improved reactivity and physical features.

Due to their unique characteristics, nanoparticles hold considerable potential for infection control. The ability of nanoparticles to carry antimicrobial drugs or to naturally have antimicrobial features makes them useful in fighting drug-resistant bacteria. They also interact with bacterial cells well due to their large surface area-to-volume ratio, strengthening their antimicrobial actions.

The bacterial cell membrane can be damaged by nanoparticle interaction, which results in cell death. They can pierce bacterial membranes, resulting in structural damage and cellular component release. This condition impairs the bacteria’s capacity to continue performing essential tasks and ultimately results in their death. Acinetobacter is typically treated with nanoparticles by various mechanisms, including membrane damage, ROS production, efflux pump inhibition, and disruption of bacterial growth and biofilm formation. Copper sulfide nanoparticles, gallate-polyvinylpyrrolidone-capped hybrid silver nanoparticles, metal nanoparticles (such as silver, copper, gold, and aluminum), and zinc oxide nanoparticles are the specific types of nanoparticles utilized in treating Acinetobacter. These nanoparticles can potentially be potent therapeutic agents since they have demonstrated antibacterial activity against drug-resistant strains of Acinetobacter.

Nanoparticles, especially silver nanoparticles (AgNPs) and copper sulfide nanoparticles (cN16E-CuS), have shown promise in treating A. baumannii infections.

Silver nanoparticles (AgNPs) prevent the growth of drug-resistant strains by damaging bacterial membranes and producing reactive oxygen species (ROS). Biologically synthesized AgNPs also show efflux pump inhibitory activity, contributing to their antibacterial effect against MDR-AB. In addition, silver nanoparticles can induce apoptosis, inhibit the synthesis of new DNA in bacteria, and contribute to their antibacterial products. The antimicrobial activity of AgNPs is concentration-dependent and effective against extracellular and intracellular A. baumannii. Therefore, silver nanoparticles (AgNPs) have shown potential in treating A. baumannii infection. AgNPs showed good inhibitory activity against MDR-AB isolates, both alone and in combination with certain antibiotics. The combination of AgNPs with colistin, meropenem, or tigecycline significantly increased the sensitivity of MDR-AB to these antibiotics. In addition, silver nanoparticles inhibited the growth of A. baumannii and showed anti-biofilm activity, especially against weak biofilm producers. AgNPs have been found to interfere with the development of A. baumannii and disrupt biofilm formation, leading to a decrease in the expression of virulence and biofilm genes. Biogenic silver nanoparticles (Bio-AgNPs) synthesized by Fusarium oxysporum have also demonstrated antibacterial activity against CR-AB. Combined with polymyxin B, they showed synergistic effects, reducing the viable A. baumannii cells.

Another study reported using cationic antimicrobial lipid-stabilized copper sulfide nanoparticles (cN16E-CuS) for treating CR-AB. cN16E-CuS exhibited excellent antimicrobial activity against A. baumannii, producing excess reactive oxygen species and damaging bacterial membranes [139]. These findings suggest that nanoparticles, such as AgNPs and cN16E-CuS, can be used as alternative treatments for A. baumannii infections.

3.8 Gene editing

The DNA of creatures, including bacteria, can be changed using groundbreaking gene editing. It entails precise genetic manipulations such as adding, deleting, or changing particular genes. By concentrating on and interrupting the genes important for antibiotic resistance and other virulence factors, this method holds enormous potential for fighting bacterial infections.

Several gene editing tools have been developed to target bacterial pathogens, including A. baumannii. These tools include zinc finger nucleases (ZFNs) [140], transcription activator-like effector nucleases (TALENs) [141], and clustered regularly interspaced short palindromic repeats (CRISPR) [142] systems. Each tool offers unique advantages and can be tailored to target specific genes or regions within the bacterial genome.

CRISPR/Cas systems have shown potential as gene-editing tools for treating A. baumannii infections [143]. Genetic manipulation methods for studying A. baumannii pathogenesis and drug-resistance mechanisms are time-consuming and inefficient [144]. However, a detailed protocol for genetic manipulation in A. baumannii, including gene deletion, insertion, and point mutation, has been provided [145]. This protocol can aid in developing more innovative approaches to diagnosing and treating A. baumannii infections [146]. CRISPR/Cas systems can provide useful information about the functions of genes in A. baumannii and help identify potential targets for antimicrobials [140].

The Cas9 enzyme, which functions as molecular scissors, and a tiny RNA molecule known as a guide RNA, which points the Cas9 enzyme to the precise target spot in the bacterial genome, make up the CRISPR-Cas9 system.

Researchers have successfully applied the CRISPR-Cas9 system to target and edit the genes in A. baumannii. By designing appropriate guide RNA molecules, specific genes involved in antibiotic resistance or biofilm formation can be disrupted or modified, rendering the bacteria susceptible to existing antibiotics or impeding their ability to form biofilms.

CRISPR-Cas9 has been used for genetic manipulation in A. baumannii to study pathogenesis and drug-resistance mechanisms [145], which allowed for investigating drug-resistant mechanisms [147]. Additionally, a method for deleting drug-resistant genes in A. baumannii using CRISPR-Cas9 has been developed, providing a novel approach for preventing the spread of drug-resistant genes and treating drug-resistant bacteria [148].

3.9 Other

LpxC inhibitors have shown potential for the treatment of MDR-AB infections. Inhibiting LpxC, an enzyme involved in lipid biosynthesis, can reduce the toxicity of lipopolysaccharide (LPS) and enhance the efficacy of antibiotics [149, 150]. Compounds, such as LpxC-2 and LpxC-4, are synergistic with iron chelators (2,2′-bipyridyl and deferiprone) and gallium nitrate, significantly reducing bacterial counts.

The lipid A production is inhibited by LpxC inhibitors, such PF-5081090, which also increase cell permeability and improve resistance to a range of antibiotics such as rifampin, vancomycin, azithromycin, imipenem, and amikacin [72]. Additionally, LpxC inhibitors can prevent the activation of the Toll-like receptor 4 (TLR4) by A. baumannii LPS, which increases the opsonophagocytic death of the bacterium and decreases inflammation [150]. According to these results, LpxC inhibitors might be a different type of treatment for A. baumannii infections resistant to many drugs [151].

RX-P873, a novel antibiotic from the Pyrrolocytosine series, has shown high binding affinity for the bacterial ribosome and broad-spectrum antibiotic properties. It has demonstrated in vitro activity against MDR Gram-negative and Gram-positive strains of bacteria, including A. baumannii. In a study, RX-P873 was found to be highly active against A. baumannii isolates, with a MIC90 value of 1 μg/ml, which was two-fold more active than colistin and four-fold more active than tigecycline [152]. Additionally, a case report described the successful treatment of XDR-AB peritoneal dialysis-associated peritonitis with combination antibiotics, including intraperitoneal polymyxin B, without the need for catheter removal or switch to hemodialysis [153]. A study on RX-P873’s activity against extracellular and intracellular forms of infection by A. baumannii, and other bacteria found that RX-P873 may be a useful alternative for disorders involving intracellular bacteria, especially Gram-negative species [154]. Therefore, RX-P873 shows potential as a treatment for Acinetobacter infections.

Advertisement

4. Conclusion

Novel therapeutic strategies for antimicrobial therapy of Acinetobacter baumannii include combination therapy, drug repurposing, novel antibiotics, bacteriophage therapy, antimicrobial peptides (AMPs), human monoclonal antibodies (Hu-mAbs), nanoparticles, and gene editing. These strategies aim to overcome drug resistance and improve the efficacy of treatment against extensively drug-resistant Acinetobacter baumannii.

Advertisement

Acknowledgments

We are grateful to the financial support by Burn Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.

Advertisement

Conflict of interest

The authors declare that they have no conflict of interest.

References

  1. 1. Neto S, Vieira A, Oliveira H, Espiña B. Assessing Acinetobacter baumannii virulence and treatment with a bacteriophage using zebrafish embryos. The FASEB Journal. 2023;37(7):e23013. DOI: 10.1096/fj.202300385R
  2. 2. Chen J, Li G, Wan F, Liu P, Du F, Shao Y, et al. Virulence characteristics and drug resistance of the prevalent capsule types in Acinetobacter baumannii. Microbial Drug Resistance. 2023;29(7):274-279. DOI: 10.1089/mdr.2022.0310
  3. 3. Zhou JX, Feng DY, Li X, Zhu JX, Wu WB, Zhang TT. Advances in research on virulence factors of Acinetobacter baumannii and their potential as novel therapeutic targets. Journal of Applied Microbiology. 2023;134(2):lxac089. DOI: 10.1093/jambio/lxac089
  4. 4. Shadan A, Pathak A, Ma Y, Pathania R, Singh RP. Deciphering the virulence factors, regulation, and immune response to Acinetobacter baumannii infection. Frontiers in Cellular and Infection Microbiology. 2023;13:1053968. DOI: 10.3389/fcimb.2023.1053968
  5. 5. Dehbanipour R, Ghalavand Z. Acinetobacter baumannii: Pathogenesis, virulence factors, novel therapeutic options and mechanisms of resistance to antimicrobial agents with emphasis on tigecycline. Journal of Clinical Pharmacy and Therapeutics. 2022;47(11):1875-1884. DOI: 10.1111/jcpt.13787
  6. 6. Uppalapati SR, Sett A, Pathania R. The outer membrane proteins OmpA, CarO, and OprD of Acinetobacter baumannii confer a two-pronged defense in facilitating its success as a potent human pathogen. Frontiers in Microbiology. 2020;11:589234. DOI: 10.3389/fmicb.2020.589234
  7. 7. Li ZT, Zhang RL, Bi XG, Xu L, Fan M, Xie D, et al. Outer membrane vesicles isolated from two clinical Acinetobacter baumannii strains exhibit different toxicity and proteome characteristics. Microbial Pathogenesis. 2015;81:46-52. DOI: 10.1016/j.micpath.2015.03.009
  8. 8. Zahn M, Bhamidimarri SP, Baslé A, Winterhalter M, van den Berg B. Structural insights into outer membrane permeability of Acinetobacter baumannii. Structure. 2016;24(2):221-231. DOI: 10.1016/j.str.2015.12.009
  9. 9. Kamoshida G, Akaji T, Takemoto N, Suzuki Y, Sato Y, Kai D, et al. Lipopolysaccharide-deficient Acinetobacter baumannii due to colistin resistance is killed by neutrophil-produced lysozyme. Frontiers in Microbiology. 2020;11:573. DOI: 10.3389/fmicb.2020.00573
  10. 10. Popova AV, Shneider MM, Arbatsky NP, Kasimova AA, Senchenkova SN, Shashkov AS, et al. Specific interaction of novel friunavirus phages encoding tailspike depolymerases with corresponding Acinetobacter baumannii capsular types. Journal of Virology. 2021;95(5):1-23. DOI: 10.1128/jvi.01714-20
  11. 11. Yang JL, Yang CJ, Chuang YC, Sheng WH, Chen YC, Chang SC. Association of capsular polysaccharide locus 2 with prognosis of Acinetobacter baumannii bacteraemia. Emerging Microbes & Infections. 2022;11(1):83-90. DOI: 10.1080/22221751.2021.2011624
  12. 12. Fiester SE, Arivett BA, Schmidt RE, Beckett AC, Ticak T, Carrier MV, et al. Iron-regulated phospholipase C activity contributes to the cytolytic activity and virulence of Acinetobacter baumannii. PLoS One. 2016;11(11):e0167068. DOI: 10.1371/journal.pone.0167068
  13. 13. Stahl J, Bergmann H, Göttig S, Ebersberger I, Averhoff B. Acinetobacter baumannii virulence is mediated by the concerted action of three phospholipases D. PLoS One. 2015;10(9):e0138360. DOI: 10.1371/journal.pone.0138360
  14. 14. Mortensen BL, Skaar EP. The contribution of nutrient metal acquisition and metabolism to Acinetobacter baumannii survival within the host. Frontiers in Cellular and Infection Microbiology. 2013;3:95. DOI: 10.3389/fcimb.2013.00095
  15. 15. Bateman TJ, Shah M, Ho TP, Shin HE, Pan C, Harris G, et al. A slam-dependent hemophore contributes to heme acquisition in the bacterial pathogen Acinetobacter baumannii. Nature Communications. 2021;12(1):6270. DOI: 10.1038/s41467-021-26545-9
  16. 16. Hesse LE, Lonergan ZR, Beavers WN, Skaar EP. The Acinetobacter baumannii Znu system overcomes host-imposed nutrient zinc limitation. Infection and Immunity. 2019;87(12):e00746-e00719. DOI: 10.1128/iai.00746-19
  17. 17. Zimbler DL, Arivett BA, Beckett AC, Menke SM, Actis LA. Functional features of TonB energy transduction systems of Acinetobacter baumannii. Infection and Immunity. 2013;81(9):3382-3394. DOI: 10.1128/iai.00540-13
  18. 18. Meyer C, Lucaβen K, Gerson S, Xanthopoulou K, Wille T, Seifert H, et al. Contribution of RND-type efflux pumps in reduced susceptibility to biocides in Acinetobacter baumannii. Antibiotics (Basel). 2022;11(11):1635. DOI: 10.3390/antibiotics11111635
  19. 19. Sharma S, Kaushik V, Kulshrestha M, Tiwari V. Different efflux pump systems in Acinetobacter baumannii and their role in multidrug resistance. Advances in Experimental Medicine and Biology. 2023;1370:155-168. DOI: 10.1007/5584_2023_771
  20. 20. Mahapatra A, Pati A, Gupta K. Role of efflux pumps in reduced susceptibility to tigecycline among clinical isolates of Acinetobacter baumannii. International Journal of Applied & Basic Medical Research. 2022;12(4):243-248. DOI: 10.4103/ijabmr.ijabmr_219_22
  21. 21. Verma P, Tiwari M, Tiwari V. Efflux pumps in multidrug-resistant Acinetobacter baumannii: Current status and challenges in the discovery of efflux pumps inhibitors. Microbial Pathogenesis. 2021;152:104766. DOI: 10.1016/j.micpath.2021.104766
  22. 22. Su CC, Morgan CE, Kambakam S, Rajavel M, Scott H, Huang W, et al. Cryo-electron microscopy structure of an Acinetobacter baumannii multidrug efflux pump. MBio. 2019;10(4):e01295-e01219. DOI: 10.1128/mBio.01295-19
  23. 23. Li P, Zhang S, Wang J, Al-Shamiri MM, Han B, Chen Y, et al. Uncovering the secretion systems of Acinetobacter baumannii: Structures and functions in pathogenicity and antibiotic resistance. Antibiotics (Basel). 2023;12(2):195. DOI: 10.3390/antibiotics12020195
  24. 24. Li L, Wang YN, Jia HB, Wang P, Dong JF, Deng J, et al. The type VI secretion system protein AsaA in Acinetobacter baumannii is a periplasmic protein physically interacting with TssM and required for T6SS assembly. Scientific Reports. 2019;9(1):9438. DOI: 10.1038/s41598-019-45875-9
  25. 25. Lewis JM, Deveson Lucas D, Harper M, Boyce JD. Systematic identification and analysis of Acinetobacter baumannii type VI secretion system effector and immunity components. Frontiers in Microbiology. 2019;10:2440. DOI: 10.3389/fmicb.2019.02440
  26. 26. Fitzsimons TC, Lewis JM, Wright A, Kleifeld O, Schittenhelm RB, Powell D, et al. Identification of novel Acinetobacter baumannii type VI secretion system antibacterial effector and immunity pairs. Infection and Immunity. 2018;86(8):e00297-e00218. DOI: 10.1128/iai.00297-18
  27. 27. Ruiz FM, Santillana E, Spínola-Amilibia M, Torreira E, Culebras E, Romero A. Crystal structure of hcp from Acinetobacter baumannii: A component of the type VI secretion system. PLoS One. 2015;10(6):e0129691. DOI: 10.1371/journal.pone.0129691
  28. 28. Xiong L, Yi F, Yu Q , Huang X, Ao K, Wang Y, et al. Transcriptomic analysis reveals the regulatory role of quorum sensing in the Acinetobacter baumannii ATCC 19606 via RNA-seq. BMC Microbiology. 2022;22(1):198. DOI: 10.1186/s12866-022-02612-z
  29. 29. Rajapaksha DC, Edirisinghe SL, Nikapitiya C, Whang I, De Zoysa M. The antimicrobial peptide octopromycin suppresses biofilm formation and quorum sensing in Acinetobacter baumannii. Antibiotics (Basel). 2023;12(3):623. DOI: 10.3390/antibiotics12030623
  30. 30. Mayer C, Muras A, Parga A, Romero M, Rumbo-Feal S, Poza M, et al. Quorum sensing as a target for controlling surface associated motility and biofilm formation in Acinetobacter baumannii ATCC(®) 17978(TM). Frontiers in Microbiology. 2020;11:565548. DOI: 10.3389/fmicb.2020.565548
  31. 31. Sun X, Ni Z, Tang J, Ding Y, Wang X, Li F. The abaI/abaR quorum sensing system effects on pathogenicity in Acinetobacter baumannii. Frontiers in Microbiology. 2021;12:679241. DOI: 10.3389/fmicb.2021.679241
  32. 32. Prestinaci F, Pezzotti P, Pantosti A. Antimicrobial resistance: A global multifaceted phenomenon. Pathogens and Global Health. 2015;109(7):309-318. DOI: 10.1179/2047773215y.0000000030
  33. 33. Peleg AY, Seifert H, Paterson DL. Acinetobacter baumannii: Emergence of a successful pathogen. Clinical Microbiology Reviews. 2008;21(3):538-582. DOI: 10.1128/cmr.00058-07
  34. 34. Uddin TM, Chakraborty AJ, Khusro A, Zidan BRM, Mitra S, Emran TB, et al. Antibiotic resistance in microbes: History, mechanisms, therapeutic strategies and future prospects. Journal of Infection and Public Health. 2021;14(12):1750-1766. DOI: 10.1016/j.jiph.2021.10.020
  35. 35. Lin MF, Lan CY. Antimicrobial resistance in Acinetobacter baumannii: From bench to bedside. World Journal of Clinical Cases. 2014;2(12):787-814. DOI: 10.12998/wjcc.v2.i12.787
  36. 36. Oikonomou O, Sarrou S, Papagiannitsis CC, Georgiadou S, Mantzarlis K, Zakynthinos E, et al. Rapid dissemination of colistin and carbapenem resistant Acinetobacter baumannii in Central Greece: Mechanisms of resistance, molecular identification and epidemiological data. BMC Infectious Diseases. 2015;15:559. DOI: 10.1186/s12879-015-1297-x
  37. 37. Zhao X, Drlica K. Restricting the selection of antibiotic-resistant mutants: A general strategy derived from fluoroquinolone studies. Clinical Infectious Diseases. 2001;33(Suppl. 3):S147-S156. DOI: 10.1086/321841
  38. 38. Wang N, Luo J, Deng F, Huang Y, Zhou H. Antibiotic combination therapy: A strategy to overcome bacterial resistance to aminoglycoside antibiotics. Frontiers in Pharmacology. 2022;13:839808. DOI: 10.3389/fphar.2022.839808
  39. 39. Adeniji OO, Nontongana N, Okoh JC, Okoh AI. The potential of antibiotics and nanomaterial combinations as therapeutic strategies in the management of multidrug-resistant infections: A review. International Journal of Molecular Sciences. 2022;23(23):15038. DOI: 10.3390/ijms232315038
  40. 40. Dufour JF, Caussy C, Loomba R. Combination therapy for non-alcoholic steatohepatitis: Rationale, opportunities and challenges. Gut. 2020;69(10):1877-1884. DOI: 10.1136/gutjnl-2019-319104
  41. 41. Narendrakumar L, Chakraborty M, Kumari S, Paul D. Das B.β-lactam potentiators to re-sensitize resistant pathogens: Discovery, development, clinical use and the way forward. Frontiers in Microbiology. 2022;13:1092556. DOI: 10.3389/fmicb.2022.1092556
  42. 42. Liepiņš M, Krūmiņa A, Meistere I, Kosjkina D, Ķibilds J, Valciņa O, et al. Retrospective study of genetic diversity of Acinetobacter baumannii-resistant strains isolated from patients in Rīga east University Hospital in Latvia. Proceedings of the Latvian Academy of Sciences. 2021;75(2):142-148. DOI: 10.2478/prolas-2021-0022
  43. 43. Scudeller L, Righi E, Chiamenti M, Bragantini D, Menchinelli G, Cattaneo P, et al. Systematic review and meta-analysis of in vitro efficacy of antibiotic combination therapy against carbapenem-resistant gram-negative bacilli. International Journal of Antimicrobial Agents. 2021;57(5):106344. DOI: 10.1016/j.ijantimicag.2021.106344
  44. 44. Ardebili A, Izanloo A, Rastegar M. Polymyxin combination therapy for multidrug-resistant, extensively-drug resistant, and difficult-to-treat drug-resistant gram-negative infections: Is it superior to polymyxin monotherapy? Expert Review of Anti-Infective Therapy. 2023;21(4):387-429. DOI: 10.1080/14787210.2023.2184346
  45. 45. Li X, Song Y, Wang L, Kang G, Wang P, Yin H, et al. A potential combination therapy of berberine hydrochloride with antibiotics against multidrug-resistant Acinetobacter baumannii. Frontiers in Cellular and Infection Microbiology. 2021;11:660431. DOI: 10.3389/fcimb.2021.660431
  46. 46. Butler DA, Biagi M, Tan X, Qasmieh S, Bulman ZP, Wenzler E. Multidrug resistant Acinetobacter baumannii: Resistance by any other name would still be hard to treat. Current Infectious Disease Reports. 2019;21(12):46. DOI: 10.1007/s11908-019-0706-5
  47. 47. Mazzitelli M, Gregori D, Sasset L, Trevenzoli M, Scaglione V, Lo Menzo S, et al. Cefiderocol-based versus Colistin-based regimens for severe carbapenem-resistant Acinetobacter baumannii infections: A propensity score-weighted, retrospective cohort study during the first two years of the COVID-19 pandemic. Microorganisms. 2023;11(4):984. DOI: 10.3390/microorganisms11040984
  48. 48. Mohd Sazlly Lim S, Heffernan AJ, Roberts JA, Sime FB. Semi-mechanistic PK/PD modelling of fosfomycin and sulbactam combination against carbapenem-resistant Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2023;65(5):e02472-e02420. DOI: 10.1128/aac.02472-20
  49. 49. Falcone M, Tiseo G, Leonildi A, Della Sala L, Vecchione A, Barnini S, et al. Cefiderocol-compared to colistin-based regimens for the treatment of severe infections caused by carbapenem-resistant Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2022;66(5):e0214221. DOI: 10.1128/aac.02142-21
  50. 50. Russo A, Bruni A, Gullì S, Borrazzo C, Quirino A, Lionello R, et al. Efficacy of cefiderocol-vs colistin-containing regimen for treatment of bacteraemic ventilator-associated pneumonia caused by carbapenem-resistant Acinetobacter baumannii in patients with COVID-19. International Journal of Antimicrobial Agents. 2023;62(1):106825. DOI: 10.1016/j.ijantimicag.2023.106825
  51. 51. Liu J, Shu Y, Zhu F, Feng B, Zhang Z, Liu L, et al. Comparative efficacy and safety of combination therapy with high-dose sulbactam or colistin with additional antibacterial agents for multiple drug-resistant and extensively drug-resistant Acinetobacter baumannii infections: A systematic review and network meta-analysis. Journal of Global Antimicrobial Resistance. 2021;24:136-147. DOI: 10.1016/j.jgar.2020.08.021
  52. 52. Gautam D, Dolma KG, Khandelwal B, Mitsuwan W, Mahboob T, Pereira ML, et al. Acinetobacter baumannii: An overview of emerging multidrug-resistant pathogen. The Medical Journal of Malaysia. 2022;77(3):357-370
  53. 53. Sonaye HV, Sheikh RY, Doifode CA. Drug repurposing: Iron in the fire for older drugs. Biomedicine & Pharmacotherapy. 2021;141:111638. DOI: 10.1016/j.biopha.2021.111638
  54. 54. Choudhury C, Arul Murugan N, Priyakumar UD. Structure-based drug repurposing: Traditional and advanced AI/ML-aided methods. Drug Discovery Today. 2022;27(7):1847-1861. DOI: 10.1016/j.drudis.2022.03.006
  55. 55. Krishnamurthy N, Grimshaw AA, Axson SA, Choe SH, Miller JE. Drug repurposing: A systematic review on root causes, barriers and facilitators. BMC Health Services Research. 2022;22(1):970. DOI: 10.1186/s12913-022-08272-z
  56. 56. Gautam LK, Sharma P, Capalash N. Attenuation of Acinetobacter baumannii virulence by inhibition of polyphosphate kinase 1 with repurposed drugs. Microbiological Research. 2021;242:126627. DOI: 10.1016/j.micres.2020.126627
  57. 57. Law SKK, Tan HS. The role of quorum sensing, biofilm formation, and iron acquisition as key virulence mechanisms in Acinetobacter baumannii and the corresponding anti-virulence strategies. Microbiological Research. 2022;260:127032. DOI: 10.1016/j.micres.2022.127032
  58. 58. Terzioğlu E, Arslan M, Balaban BG, Çakar ZP. Microbial silver resistance mechanisms: Recent developments. World Journal of Microbiology and Biotechnology. 2022;38(9):158. DOI: 10.1007/s11274-022-03341-1
  59. 59. Cheng YS, Sun W, Xu M, Shen M, Khraiwesh M, Sciotti RJ, et al. Repurposing screen identifies unconventional drugs with activity against multidrug resistant Acinetobacter baumannii. Frontiers in Cellular and Infection Microbiology. 2018;8:438. DOI: 10.3389/fcimb.2018.00438
  60. 60. Ayon NJ. High-throughput screening of natural product and synthetic molecule libraries for antibacterial drug discovery. Metabolites. 2023;13(5):625. DOI: 10.3390/metabo13050625
  61. 61. Seleem NM, Abd El Latif HK, Shaldam MA, El-Ganiny A. Drugs with new lease of life as quorum sensing inhibitors: For combating MDR Acinetobacter baumannii infections. European Journal of Clinical Microbiology & Infectious Diseases. 2020;39(9):1687-1702. DOI: 10.1007/s10096-020-03882-z
  62. 62. Zhong S, He S. Quorum sensing inhibition or quenching in Acinetobacter baumannii: The novel therapeutic strategies for new drug development. Frontiers in Microbiology. 2021;12:558003. DOI: 10.3389/fmicb.2021.558003
  63. 63. Kundu R. Cationic amphiphilic peptides: Synthetic antimicrobial agents inspired by nature. Chem Med Chem. 2020;15(20):1887-1896. DOI: 10.1002/cmdc.202000301
  64. 64. Han Y, Zhang M, Lai R, Zhang Z. Chemical modifications to increase the therapeutic potential of antimicrobial peptides. Peptides. 2021;146:170666. DOI: 10.1016/j.peptides.2021.170666
  65. 65. Yong Y, Zhou Y, Liu K, Liu G, Wu L, Fang B. Exogenous citrulline and glutamine contribute to reverse the resistance of Salmonella to apramycin. Frontiers in Microbiology. 2021;12:759170. DOI: 10.3389/fmicb.2021.759170
  66. 66. Gontijo AVL, Pereira SL, de Lacerda BH. Can drug repurposing be effective against carbapenem-resistant Acinetobacter baumannii? Current Microbiology. 2021;79(1):13. DOI: 10.1007/s00284-021-02693-5
  67. 67. Ayerbe-Algaba R, Gil-Marqués ML, Miró-Canturri A, Parra-Millán R, Pachón-Ibáñez ME, Jiménez-Mejías ME, et al. The anthelmintic oxyclozanide restores the activity of colistin against colistin-resistant gram-negative bacilli. International Journal of Antimicrobial Agents. 2019;54(4):507-512. DOI: 10.1016/j.ijantimicag.2019.07.006
  68. 68. Bendary MM, Abd El-Hamid MI, Abousaty AI, Elmanakhly AR, Alshareef WA, Mosbah RA, et al. Therapeutic switching of rafoxanide: A new approach to fighting drug-resistant bacteria and fungi. Microbiology Spectrum. 2023;11(4):e0267922. DOI: 10.1128/spectrum.02679-22
  69. 69. Carlson-Banning KM, Chou A, Liu Z, Hamill RJ, Song Y, Zechiedrich L. Toward repurposing ciclopirox as an antibiotic against drug-resistant Acinetobacter baumannii, Escherichia coli, and Klebsiella pneumoniae. PLoS One. 2013;8(7):e69646. DOI: 10.1371/journal.pone.0069646
  70. 70. Peyclit L, Baron SA, Rolain JM. Drug repurposing to fight colistin and carbapenem-resistant bacteria. Frontiers in Cellular and Infection Microbiology. 2019;9:193. DOI: 10.3389/fcimb.2019.00193
  71. 71. Cruz-Muñiz MY, López- Jacome LE, Hernández-Durán M, Franco-Cendejas R, Licona-Limón P, Ramos-Balderas JL, et al. Repurposing the anticancer drug mitomycin C for the treatment of persistent Acinetobacter baumannii infections. International Journal of Antimicrobial Agents. 2017;49(1):88-92. DOI: 10.1016/j.ijantimicag.2016.08.022
  72. 72. Miró-Canturri A, Vila-Domínguez A, Caretero-Ledesma M, Ayerbe-Algaba R, Pachón J, Jiménez-Mejías ME, et al. Repurposing of the tamoxifen metabolites to treat methicillin-resistant Staphylococcus epidermidis and vancomycin-resistant enterococcus faecalis infections. Microbiology Spectrum. 2021;9(2):e0040321. DOI: 10.1128/Spectrum.00403-21
  73. 73. Hussein M, Han ML, Zhu Y, Schneider-Futschik EK, Hu X, Zhou QT, et al. Mechanistic insights from global metabolomics studies into synergistic bactericidal effect of a polymyxin B combination with tamoxifen against cystic fibrosis MDR Pseudomonas aeruginosa. Computational and Structural Biotechnology Journal. 2018;16:587-599. DOI: 10.1016/j.csbj.2018.11.001
  74. 74. Brutcher E, Christensen D, Hennessey Smith M, Koutlas JB, Sellers JB, Timmons T, et al. 5-fluorouracil and capecitabine: Assessment and treatment of uncommon early-onset severe toxicities associated with administration. Clinical Journal of Oncology Nursing. 2018;22(6):627-634. DOI: 10.1188/18.cjon.627-634
  75. 75. Antunes LC, Imperi F, Minandri F, Visca P. In vitro and in vivo antimicrobial activities of gallium nitrate against multidrug-resistant Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2012;56(11):5961-5970. DOI: 10.1128/aac.01519-12
  76. 76. Hijazi S, Visaggio D, Pirolo M, Frangipani E, Bernstein L, Visca P. Antimicrobial activity of gallium compounds on ESKAPE pathogens. Frontiers in Cellular and Infection Microbiology. 2018;8:316. DOI: 10.3389/fcimb.2018.00316
  77. 77. Wagenlehner FME. New antibiotics for the treatment of urinary tract infections. Urologie. 2023;62(7):705-710. DOI: 10.1007/s00120-023-02121-5
  78. 78. Terreni M, Taccani M, Pregnolato M. New antibiotics for multidrug-resistant bacterial strains: Latest research developments and future perspectives. Molecules. 2021;26(9):2671. DOI: 10.3390/molecules26092671
  79. 79. Abdul-Mutakabbir JC, Nguyen L, Maassen PT, Stamper KC, Kebriaei R, Kaye KS, et al. In vitro antibacterial activity of cefiderocol against multidrug-resistant Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2021;65(9):e0264620. DOI: 10.1128/aac.02646-20
  80. 80. Halasohoris SA, Scarff JM, Pysz LM, Lembirik S, Lemmon MM, Biek D, et al. In vitro and in vivo activity of GT-1, a novel siderophore cephalosporin, and GT-055, a broad-spectrum β-lactamase inhibitor, against biothreat and ESKAPE pathogens. Journal of Antibiotics (Tokyo). 2021;74(12):884-892. DOI: 10.1038/s41429-021-00472-9
  81. 81. Abdul-Mutakabbir JC, Alosaimy S, Morrisette T, Kebriaei R, Rybak MJ. Cefiderocol: A novel siderophore cephalosporin against multidrug-resistant gram-negative pathogens. Pharmacotherapy. 2020;40(12):1228-1247. DOI: 10.1002/phar.2476
  82. 82. Nguyen LP, Park CS, Pinto NA, Lee H, Seo HS, Vu TN, et al. In vitro activity of a novel siderophore-cephalosporin LCB10-0200 (GT-1), and LCB10-0200/avibactam, against carbapenem-resistant Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa strains at a tertiary hospital in Korea. Pharmaceuticals (Basel). 2021;14(4):370. DOI: 10.3390/ph14040370
  83. 83. Yin T, Lai JJ, Huang WC, Kuo SC, Chiang TT, Yang YS. In vitro and in vivo comparison of eravacycline- and tigecycline-based combination therapies for tigecycline-resistant Acinetobacter baumannii. Journal of Chemotherapy. 2022;34(3):166-172. DOI: 10.1080/1120009x.2021.2005755
  84. 84. Buckley AM, Altringham J, Clark E, Bently K, Spittal W, Ewin D, et al. Eravacycline, a novel tetracycline derivative, does not induce clostridioides difficile infection in an in vitro human gut model. The Journal of Antimicrobial Chemotherapy. 2021;76(1):171-178. DOI: 10.1093/jac/dkaa386
  85. 85. Van Hise N, Petrak RM, Skorodin NC, Fliegelman RM, Anderson M, Didwania V, et al. A real-world assessment of clinical outcomes and safety of eravacycline: A novel fluorocycline. Infectious Disease and Therapy. 2020;9(4):1017-1028. DOI: 10.1007/s40121-020-00351-0
  86. 86. Seifert H, Stefanik D, Olesky M, Higgins PG. In vitro activity of the novel fluorocycline TP-6076 against carbapenem-resistant Acinetobacter baumannii. International Journal of Antimicrobial Agents. 2020;55(1):105829. DOI: 10.1016/j.ijantimicag.2019.10.010
  87. 87. Shapiro AB, Moussa SH, McLeod SM, Durand-Réville T, Miller AA. Durlobactam, a new diazabicyclooctane β-lactamase inhibitor for the treatment of Acinetobacter infections in combination with sulbactam. Frontiers in Microbiology. 2021;12:709974. DOI: 10.3389/fmicb.2021.709974
  88. 88. Yahav D, Giske CG, Grāmatniece A, Abodakpi H, Tam VH, Leibovici L. New β-lactam-β-lactamase inhibitor combinations. Clinical Microbiology Reviews. 2020;34(1):e00115-e00120. DOI: 10.1128/cmr.00115-20
  89. 89. Barnes MD, Kumar V, Bethel CR, Moussa SH, O'Donnell J, Rutter JD, et al. Targeting multidrug-resistant Acinetobacter spp.: Sulbactam and the diazabicyclooctenone β-lactamase inhibitor ETX2514 as a novel therapeutic agent. MBio. 2019;10(2):e00159-e00119. DOI: 10.1128/mBio.00159-19
  90. 90. Monogue ML, Giovagnoli S, Bissantz C, Zampaloni C, Nicolau DP. In vivo efficacy of meropenem with a novel non-β-lactam-β-lactamase inhibitor, nacubactam, against gram-negative organisms exhibiting various resistance mechanisms in a murine complicated urinary tract infection model. Antimicrobial Agents and Chemotherapy. 2018;62(9):e02596-e02517. DOI: 10.1128/aac.02596-17
  91. 91. Wang L, Chen Y, Han R, Huang Z, Zhang X, Hu F, et al. Sulbactam enhances in vitro activity of β-lactam antibiotics against Acinetobacter baumannii. Infection and Drug Resistance. 2021;14:3971-3977. DOI: 10.2147/idr.s332160
  92. 92. Lasarte-Monterrubio C, Vázquez-Ucha JC, Maneiro M, Arca-Suárez J, Alonso I, Guijarro-Sánchez P, et al. Activity of Imipenem, meropenem, cefepime, and sulbactam in combination with the β-lactamase inhibitor LN-1-255 against Acinetobacter spp. Antibiotics (Basel). 2021;10(2):210. DOI: 10.3390/antibiotics10020210
  93. 93. French S, Farha M, Ellis MJ, Sameer Z, Côté JP, Cotroneo N, et al. Potentiation of antibiotics against gram-negative bacteria by polymyxin B analogue SPR741 from unique perturbation of the outer membrane. ACS Infectious Diseases. 2020;6(6):1405-1412. DOI: 10.1021/acsinfecdis.9b00159
  94. 94. Tran TB, Bergen PJ, Creek DJ, Velkov T, Li J. Synergistic killing of polymyxin B in combination with the antineoplastic drug mitotane against polymyxin-susceptible and -resistant Acinetobacter baumannii: A metabolomic study. Frontiers in Pharmacology. 2018;9:359. DOI: 10.3389/fphar.2018.00359
  95. 95. Becker K, Aranzana-Climent V, Cao S, Nilsson A, Shariatgorji R, Haldimann K, et al. Efficacy of EBL-1003 (apramycin) against Acinetobacter baumannii lung infections in mice. Clinical Microbiology and Infection. 2021;27(9):1315-1321. DOI: 10.1016/j.cmi.2020.12.004
  96. 96. Juhas M, Widlake E, Teo J, Huseby DL, Tyrrell JM, Polikanov YS, et al. In vitro activity of apramycin against multidrug-, carbapenem- and aminoglycoside-resistant Enterobacteriaceae and Acinetobacter baumannii. The Journal of Antimicrobial Chemotherapy. 2019;74(4):944-952. DOI: 10.1093/jac/dky546
  97. 97. Kang AD, Smith KP, Berg AH, Truelson KA, Eliopoulos GM, McCoy C, et al. Efficacy of Apramycin against multidrug-resistant Acinetobacter baumannii in the murine neutropenic thigh model. Antimicrobial Agents and Chemotherapy. 2018;62(4):e02585-e02517. DOI: 10.1128/aac.02585-17
  98. 98. Smani Y, Pachón-Ibáñez ME, Pachón J. New molecules and adjuvants in the treatment of infections by Acinetobacter baumannii. Expert Opinion on Pharmacotherapy. 2016;17(9):1207-1214. DOI: 10.1080/14656566.2016.1176144
  99. 99. Kang AD, Smith KP, Eliopoulos GM, Berg AH, McCoy C, Kirby JE. Invitro apramycin activity against multidrug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa. Diagnostic Microbiology and Infectious Disease. 2017;88(2):188-191. DOI: 10.1016/j.diagmicrobio.2017.03.006
  100. 100. Yan W, Banerjee P, Liu Y, Mi Z, Bai C, Hu H, et al. Development of thermosensitive hydrogel wound dressing containing Acinetobacter baumannii phage against wound infections. International Journal of Pharmaceutics. 2021;602:120508. DOI: 10.1016/j.ijpharm.2021.120508
  101. 101. Broncano-Lavado A, Santamaría-Corral G, Esteban J, García-Quintanilla M. Advances in bacteriophage therapy against relevant multi drug-resistant pathogens. Antibiotics (Basel). 2021;10(6):672. DOI: 10.3390/antibiotics10060672
  102. 102. Malik S, Nehra K, Rana JS. Bacteriophage cocktail and phage antibiotic synergism as promising alternatives to conventional antibiotics for the control of multi-drug-resistant uropathogenic Escherichia coli. Virus Research. 2021;302:198496. DOI: 10.1016/j.virusres.2021.198496
  103. 103. Rai S, Kumar A. Bacteriophage therapeutics to confront multidrug-resistant Acinetobacter baumannii—A global health menace. Environmental Microbiology Reports. 2022;14(3):347-364. DOI: 10.1111/1758-2229.12988
  104. 104. Düzgüneş N, Sessevmez M, Yildirim M. Bacteriophage therapy of bacterial infections: The rediscovered frontier. Pharmaceuticals (Basel). 2021;14(1):34. DOI: 10.3390/ph14010034
  105. 105. Barber OW, Miramontes IM, Jain M, Ozer EA, Hartmann EM. The future of bacteriophage therapy will promote antimicrobial susceptibility. mSystems. 2021;6(4):e0021821. DOI: 10.1128/mSystems.00218-21
  106. 106. Junghans S, Rojas SV, Skusa R, Püschel A, Grambow E, Kohlen J, et al. Bacteriophages for the treatment of graft infections in cardiovascular medicine. Antibiotics (Basel). 2021;10(12):1446. DOI: 10.3390/antibiotics10121446
  107. 107. Liu D, Van Belleghem JD, de Vries CR, Burgener E, Chen Q , Manasherob R, et al. The safety and toxicity of phage therapy: A review of animal and clinical studies. Viruses. 2021;13(7):1268. DOI: 10.3390/v13071268
  108. 108. Jeon J, Park JH, Yong D. Efficacy of bacteriophage treatment against carbapenem-resistant Acinetobacter baumannii in galleria mellonella larvae and a mouse model of acute pneumonia. BMC Microbiology. 2019;19(1):70. DOI: 10.1186/s12866-019-1443-5
  109. 109. Tan Y, Su J, Fu M, Zhang H, Zeng H. Recent advances in phage-based therapeutics for multi-drug resistant Acinetobacter baumannii. Bioengineering (Basel). 2022;10(1):35. DOI: 10.3390/bioengineering10010035
  110. 110. Bhargava K, Nath G, Bhargava A, Aseri GK, Jain N. Phage therapeutics: From promises to practices and prospectives. Applied Microbiology and Biotechnology. 2021;105(24):9047-9067. DOI: 10.1007/s00253-021-11695-z
  111. 111. Mu A, McDonald D, Jarmusch AK, Martino C, Brennan C, Bryant M, et al. Assessment of the microbiome during bacteriophage therapy in combination with systemic antibiotics to treat a case of staphylococcal device infection. Microbiome. 2021;9(1):92. DOI: 10.1186/s40168-021-01026-9
  112. 112. Onsea J, Uyttebroek S, Chen B, Wagemans J, Lood C, Van Gerven L, et al. Bacteriophage therapy for difficult-to-treat infections: The implementation of a multidisciplinary phage task force (the PHAGEFORCE study protocol). Viruses. 2021;13(8):1543. DOI: 10.3390/v13081543
  113. 113. Neshani A, Sedighian H, Mirhosseini SA, Ghazvini K, Zare H, Jahangiri A. Antimicrobial peptides as a promising treatment option against Acinetobacter baumannii infections. Microbial Pathogenesis. 2020;146:104238. DOI: 10.1016/j.micpath.2020.104238
  114. 114. Luo Y, Song Y. Mechanism of antimicrobial peptides: Antimicrobial, anti-inflammatory and antibiofilm activities. International Journal of Molecular Sciences. 2021;22(21):11401. DOI: 10.3390/ijms222111401
  115. 115. Zhang QY, Yan ZB, Meng YM, Hong XY, Shao G, Ma JJ, et al. Antimicrobial peptides: Mechanism of action, activity and clinical potential. Military Medical Research. 2021;8(1):48. DOI: 10.1186/s40779-021-00343-2
  116. 116. Drayton M, Deisinger JP, Ludwig KC, Raheem N, Müller A, Schneider T, et al. Host defense peptides: Dual antimicrobial and immunomodulatory action. International Journal of Molecular Sciences. 2021;22(20):11172. DOI: 10.3390/ijms222011172
  117. 117. Benfield AH, Henriques ST. Mode-of-action of antimicrobial peptides: Membrane disruption vs. intracellular mechanisms. Frontiers in Medical Technology. 2020;2:610997. DOI: 10.3389/fmedt.2020.610997
  118. 118. Wadhwani P, Sekaran S, Strandberg E, Bürck J, Chugh A, Ulrich AS. Membrane interactions of Latarcins: Antimicrobial peptides from spider venom. International Journal of Molecular Sciences. 2021;22(18):10156. DOI: 10.3390/ijms221810156
  119. 119. Choi J, Jang A, Yoon YK, Kim Y. Development of novel peptides for the antimicrobial combination therapy against carbapenem-resistant Acinetobacter baumannii infection. Pharmaceutics. 2021;13(11):1800. DOI: 10.3390/pharmaceutics13111800
  120. 120. Denardi LB, de Arruda TP, Weiblen C, Ianiski LB, Stibbe PC, Pinto SC, et al. In vitro activity of the antimicrobial peptides h-Lf1-11, MSI-78, LL-37, fengycin 2B, and magainin-2 against clinically important bacteria. Brazilian Journal of Microbiology. 2022;53(1):171-177. DOI: 10.1007/s42770-021-00645-6
  121. 121. Ridyard KE, Overhage J. The potential of human peptide LL-37 as an antimicrobial and anti-biofilm agent. Antibiotics (Basel). 2021;10(6):650. DOI: 10.3390/antibiotics10060650
  122. 122. Rima M, Rima M, Fajloun Z, Sabatier JM, Bechinger B, Naas T. Antimicrobial peptides: A potent alternative to antibiotics. Antibiotics (Basel). 2021;10(9):1095. DOI: 10.3390/antibiotics10091095
  123. 123. Mwangi J, Yin Y, Wang G, Yang M, Li Y, Zhang Z, et al. The antimicrobial peptide ZY4 combats multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii infection. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(52):26516-26522. DOI: 10.1073/pnas.1909585117
  124. 124. Jung CJ, Liao YD, Hsu CC, Huang TY, Chuang YC, Chen JW, et al. Identification of potential therapeutic antimicrobial peptides against Acinetobacter baumannii in a mouse model of pneumonia. Scientific Reports. 2021;11(1):7318. DOI: 10.1038/s41598-021-86844-5
  125. 125. Amiss AS, Henriques ST, Lawrence N. Antimicrobial peptides provide wider coverage for targeting drug-resistant bacterial pathogens. Peptide Science. 2022;114(2):e24246. DOI: 10.1002/pep2.24246
  126. 126. Wang C, Hong T, Cui P, Wang J, Xia J. Antimicrobial peptides towards clinical application: Delivery and formulation. Advanced Drug Delivery Reviews. 2021;175:113818. DOI: 10.1016/j.addr.2021.05.028
  127. 127. Ioannou P, Baliou S, Kofteridis DP. Antimicrobial peptides in infectious diseases and beyond—A narrative review. Life (Basel). 2023;13(8):1651. DOI: 10.3390/life13081651
  128. 128. Horenstein AL, Chillemi A, Quarona V, Zito A, Mariani V, Faini AC, et al. Antibody mimicry, receptors and clinical applications. Human Antibodies. 2017;25(3-4):75-85. DOI: 10.3233/hab-160305
  129. 129. Santoriello D, Markowitz GS. Heavy and light chains all at once: A new immunofluorescence technique to evaluate monoclonal immunoglobulin deposits. Kidney International. 2021;100(1):22-24. DOI: 10.1016/j.kint.2021.04.022
  130. 130. Melani RD, Srzentić K, Gerbasi VR, McGee JP, Huguet R, Fornelli L, et al. Direct measurement of light and heavy antibody chains using ion mobility and middle-down mass spectrometry. MAbs. 2019;11(8):1351-1357. DOI: 10.1080/19420862.2019.1668226
  131. 131. Posner J, Barrington P, Brier T, Datta-Mannan A. Monoclonal antibodies: Past, present and future. Handbook of Experimental Pharmacology. 2019;260:81-141. DOI: 10.1007/164_2019_323
  132. 132. Yeganeh O, Shabani M, Pakzad P, Mosaffa N, Hashemi A. Production and characterization of novel monoclonal antibodies against outer membrane protein a (OmpA) of Acinetobacter baumannii. Journal of Immunological Methods. 2021;499:113169. DOI: 10.1016/j.jim.2021.113169
  133. 133. Nielsen TB, Yan J, Luna BM, Talyansky Y, Slarve M, Bonomo RA, et al. Monoclonal antibody requires immunomodulation for efficacy against Acinetobacter baumannii infection. The Journal of Infectious Diseases. 2021;224(12):2133-2147. DOI: 10.1093/infdis/jiab265
  134. 134. Nielsen TB, Yan J, Slarve M, Lu P, Li R, Ruiz J, et al. Monoclonal antibody therapy against Acinetobacter baumannii. Infection and Immunity. 2021;89(10):e0016221. DOI: 10.1128/iai.00162-21
  135. 135. Nielsen TB, Pantapalangkoor P, Luna BM, Bruhn KW, Yan J, Dekitani K, et al. Monoclonal antibody protects against Acinetobacter baumannii infection by enhancing bacterial clearance and evading sepsis. The Journal of Infectious Diseases. 2017;216(4):489-501. DOI: 10.1093/infdis/jix315
  136. 136. Ouyang W. O'Garra a.IL-10 family cytokines IL-10 and IL-22: From basic science to clinical translation. Immunity. 2019;50(4):871-891. DOI: 10.1016/j.immuni.2019.03.020
  137. 137. Wang-Lin SX, Balthasar JP. Pharmacokinetic and pharmacodynamic considerations for the use of monoclonal antibodies in the treatment of bacterial infections. Antibodies (Basel). 2018;7(1):5. DOI: 10.3390/antib7010005
  138. 138. Skariyachan S, Taskeen N, Ganta M, Venkata KB. Recent perspectives on the virulent factors and treatment options for multidrug-resistant Acinetobacter baumannii. Critical Reviews in Microbiology. 2019;45(3):315-333. DOI: 10.1080/1040841x.2019.1600472
  139. 139. Karnan Singaravelu D, Bala Subramaniyan S, Tharunya MP, Veerappan A. Antimicrobial lipid capped copper sulfide nanoparticles display enhanced bactericidal effect against carbapenem-resistant Acinetobacter baumannii. Materials Letters. 2022;306:130985. DOI: 10.1016/j.matlet.2021.130985
  140. 140. Serajian S, Ahmadpour E, Oliveira SMR, Pereira ML, Heidarzadeh S. CRISPR-Cas technology: Emerging applications in clinical microbiology and infectious diseases. Pharmaceuticals (Basel). 2021;14(11):1171. DOI: 10.3390/ph14111171
  141. 141. Thompson MK, Sobol RW, Prakash A. Exploiting DNA endonucleases to advance mechanisms of DNA repair. Biology (Basel). 2021;10(6):530. DOI: 10.3390/biology10060530
  142. 142. Nargesi S, Kaboli S, Thekkiniath J, Heidari S, Keramati F, Seyedmousavi S, et al. Recent advances in genome editing tools in medical mycology research. Journal of Fungi (Basel). 2021;7(4):257. DOI: 10.3390/jof7040257
  143. 143. Liu W, Li L, Jiang J, Wu M, Lin P. Applications and challenges of CRISPR-Cas gene-editing to disease treatment in clinics. Precision Clinical Medicine. 2021;4(3):179-191. DOI: 10.1093/pcmedi/pbab014
  144. 144. Malech HL. Treatment by CRISPR-Cas9 gene editing—A proof of principle. The New England Journal of Medicine. 2021;384(3):286-287. DOI: 10.1056/NEJMe2034624
  145. 145. Wang Y, Wang Z. Ji Q.CRISPR-Cas9-based genome editing and cytidine base editing in Acinetobacter baumannii. STAR Protocols. 2020;1(1):100025. DOI: 10.1016/j.xpro.2020.100025
  146. 146. Zhang B. CRISPR/Cas gene therapy. Journal of Cellular Physiology. 2021;236(4):2459-2481. DOI: 10.1002/jcp.30064
  147. 147. Wang Y, Wang Z, Chen Y, Hua X, Yu Y, Ji Q. A highly efficient CRISPR-Cas9-based genome engineering platform in Acinetobacter baumannii to understand the H(2)O(2)-sensing mechanism of OxyR. Cell Chemistry & Biology. 2019;26(12):1732-42.e5. DOI: 10.1016/j.chembiol.2019.09.003
  148. 148. Ruegsegger L, Xiao J, Naziripour A, Kanumuambidi T, Brown D, Williams F, et al. Multidrug-resistant gram-negative bacteria in burn patients. Antimicrobial Agents and Chemotherapy. 2022;66(9):e0068822. DOI: 10.1128/aac.00688-22
  149. 149. Gong Z, Li J, Luo H, Zhan D, Liu X, Gao C, et al. Low-temperature laminar flow ward for the treatment of multidrug resistance Acinetobacter baumannii pneumonia. European Journal of Clinical Microbiology & Infectious Diseases. 2020;39(5):877-887. DOI: 10.1007/s10096-019-03790-x
  150. 150. García-Quintanilla M, Caro-Vega JM, Pulido MR, Moreno-Martínez P, Pachón J, McConnell MJ. Inhibition of LpxC increases antibiotic susceptibility in Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2016;60(8):5076-5079. DOI: 10.1128/aac.00407-16
  151. 151. Lin L, Tan B, Pantapalangkoor P, Ho T, Baquir B, Tomaras A, et al. Inhibition of LpxC protects mice from resistant Acinetobacter baumannii by modulating inflammation and enhancing phagocytosis. MBio. 2012;3(5):e00312-e00312. DOI: 10.1128/mBio.00312-12
  152. 152. Flamm RK, Rhomberg PR, Jones RN, Farrell DJ. In vitro activity of RX-P873 against Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii. Antimicrobial Agents and Chemotherapy. 2015;59(4):2280-2285. DOI: 10.1128/aac.04840-14
  153. 153. Morrissey I, Magnet S, Genet E, Jeandey P, Marra A, Hawser S, et al. Activity of RX-P873, a novel pyrrolocytosine, against gram-negative bacteria. International Journal of Antimicrobial Agents. 2015;46(3):352-354. DOI: 10.1016/j.ijantimicag.2015.06.001
  154. 154. Fitzpatrick MA, Esterly JS, Postelnick MJ, Sutton SH. Successful treatment of extensively drug-resistant Acinetobacter baumannii peritoneal dialysis peritonitis with intraperitoneal polymyxin B and ampicillin-sulbactam. The Annals of Pharmacotherapy. 2012;46(7-8):e17. DOI: 10.1345/aph.1R086

Written By

Amir Emami, Neda Pirbonyeh and Fatemeh Javanmardi

Submitted: 27 August 2023 Reviewed: 28 August 2023 Published: 23 November 2023