We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
PerspectiveOpen Accesscc iconby icon

Nanomedicine to overcome antimicrobial resistance: challenges and prospects

    Wei Zou

    *Author for correspondence:

    E-mail Address: wei.zou@nrc-cnrc.gc.ca

    Human Health Therapeutic Research Center, National Research Council Canada, Ottawa, ON, K1A 0R6, Canada

    ,
    Alyssa McAdorey

    Human Health Therapeutic Research Center, National Research Council Canada, Ottawa, ON, K1A 0R6, Canada

    Department of Chemistry & Centre for Biotechnology, Brock University, St Catharines, ON, L2S 3A1, Canada

    ,
    Hongbin Yan

    Department of Chemistry & Centre for Biotechnology, Brock University, St Catharines, ON, L2S 3A1, Canada

    &
    Wangxue Chen

    Human Health Therapeutic Research Center, National Research Council Canada, Ottawa, ON, K1A 0R6, Canada

    Department of Biological Science, Brock University, St Catharines, ON, L2S 3A1, Canada

    Published Online:https://doi.org/10.2217/nnm-2023-0022

    Abstract

    Translation of antibacterial nanoparticles into nanomedicine requires a deep understanding of the dynamic nature of nanoparticles and the ways they overcome immunological and biological barriers. Nanomedicines need prolonged serum stability by proper stealth coating or forming beneficial protein corona, to avoid rapid clearance by the mononuclear phagocytic system. A preferred nanoparticle formulation may include nonimmunogenic carbohydrates, which act both as a stealth coating and ligands of specific endothelium receptors to facilitate nanomedicines crossing the vascular barrier. This may lead to more rapid delivery and accumulation of nanomedicine at the infection site and provide broader and faster clinical responses than targeting specific bacterial surface receptors. Ideally, antibacterial nanomedicines should be able to penetrate biofilms through fusion and/or diffusion for targeted delivery.

    Tweetable abstract

    Challenges and prospects of using nanomedicine to overcome antimicrobial resistance.

    Antimicrobial resistance (AMR) has become a global threat to human health and economic development, and causes millions of deaths each year [1]. AMR is the result of a naturally evolving process through genetic mutation or by acquiring resistance genes from another microbe that could limit uptake of a drug, modify a drug target, inactivate a drug and/or actively efflux a drug [2]. These are often associated with improper use of the antimicrobial agents during the prevention and management of the infections and associated diseases. Additionally, the biofilms formed by AMR bacteria have significantly contributed to the multidrug resistance [3,4]. Although AMR have also been found in fungi, viruses and protozoa, the most significant and consequential challenge is the fact that more bacteria have developed multiantibiotic resistance. Multiantibiotic resistant bacteria are more difficult to treat, the choices of antibiotics are limited, and often higher doses are required. Alternative and more effective medications against AMR are urgently needed.

    The AMR threat may be overcome by introduction of effective vaccines and new antibiotics; however, these developments have encountered various challenges. The US FDA has only approved a small number of antibacterial agents in recent years [5], although intensive investigation has led some to late-stage clinical developments [6]. The WHO has listed 12 highly virulent and multidrug-resistant pathogens [7] that include Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. so-called ESKAPE bacteria, capable of ‘escaping’ the action of antimicrobial agents [8–10]. Currently, there is no effective vaccine for ESKAPE bacteria, and though a number of clinical trials have been conducted with most efforts on S. aureus vaccine candidates (Table 1, https://clinicaltrials.gov/), so far, the outcomes are rather disappointing [11]. On the other hand, ESKAPE bacteria have effectively developed multidrug resistance, through various mechanisms as summarized in Table 2 [2,12], including inhibition of cell wall synthesis, depolarizing cell membrane, inhibiting protein synthesis and nucleic acid synthesis, and interfering with other metabolic pathways through drug inactivation by bacterial enzymes, drug target mutation and modification, reduced permeability and elevated efflux [10,13]. These pathogens are also able to form biofilms that act to prevent immune response and antibiotic action, and become less sensitive to antibiotics: as a result they are responsible for many difficult-to-treat, reoccurring infections [14]. As time progresses, the number of antibiotics to treat ESKAPE pathogens declines. In response to those urgent needs, antibiotic combination has been widely studied [15,16] and a few are currently under clinical trials (see https://clinicaltrials.gov/). In addition, clinical trials, based on bacteriophage therapy [17], fecal microbial transplantation [18–20] and probiotic bacterial replacement [21] as alternative approaches, can also be found at https://clinicaltrials.gov/.

    Table 1. Clinical development of vaccine candidates against Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.
    BacteriaCurrent (discontinued)
    Phase IPhase IIPhase III
    E. faecium000
    S. aureus(4)2 (4)(1)
    K. pneumoniae010
    A. baumannii000
    P. aeruginosa001
    Enterobacter spp.000
    Table 2. Common antibiotics and bacterial resistance mechanisms.
    AntibioticsMechanism of actionMechanism of resistance
    β-Lactams
    – Carbapenems, cephalosporins, monobactams, penicillins
    Glycopeptides
    – Vancomycin
    Inhibiting cell wall synthesis by binding to proteins required for peptidoglycan crosslinkingProducing β-lactamases, modifying penicillin-binding proteins, replacing D-ala-D-ala with D-ala-D-lac or D-ala-D-ser to reduce vancomycin affinity, low permeability and increased efflux
    Lipopeptides
    – Daptomycin, polymyxin, colistin
    Depolarizing cell membrane and depleting ATPThickening of cell wall and increasing the positive charge in the cell wall
    Aminoglycosides
    – Gentamicin, streptomycin, kanamycin 
    Tetracyclines
    – Tigecycline, tetracycline
    Phenicols
    – Chloramphenicol
    Lincosamides
    – Clindamycin
    Macrolides
    – Azithromycin, erythromycin
    Oxazolidinones
    – Linezolid
    Inhibiting the translation of proteins by targeting subunits of rRNA, causing misreading and/or truncated proteins and cell deathAntibiotics modified by various enzymes, mutations in rRNA gene, and modification of rRNA, decreased influx and/or increased efflux
    Quinolones/fluoroquinolones
    – Ciprofloxacin
    Inhibiting nucleic acid synthesis by DNA gyrase and topoisomerase IVMutations in DNA gyrase or topoisomerase IV and increased efflux
    Sulfonamides
    – Sulfamethizole
    Pyrimidines
    – Trimethoprim
    Inhibiting dihydropteroate synthase and dihydrofolate reductaseMutations in the dihydropteroate synthase, modifications of dihydrofolate reductase and efflux of trimethoprim

    A high dose of antibiotics is often required to treat AMR bacteria. If the current antibiotics can be target-delivered to the infection site, an improved antibacterial efficacy and safety profile could be expected. Approaches with targeted delivery of broad-spectrum antibiotics include antibiotic–antibody conjugates (AACs) [22] and antibiotic nanoparticles, which may provide more effective treatment while minimizing pressure on other bacteria to become antibiotic resistant. These two strategies have been applied successfully as anticancer therapies, which have yielded FDA-approved antibody–drug conjugate (ADC) drugs [23] and anticancer nanomedicines [24]. Unlike the highly potent payloads used in ADC and uptake of ADC through endocytosis followed by drug release by lysosomal enzymes, resulting in effective cytotoxicity against cancer cells, AAC lacks all of those mechanisms. Therefore, AACs are less likely to become successful antibacterial drugs unless much more potent antibacterial agents are employed and rapid drug release is achieved, which would then provide the desired therapeutic effect. On the other hand, nanomedicines may provide an attractive alternative against the threat of AMR, since they are able to improve drug stability, prolong circulation, improve pharmacological properties and most importantly show efficacy as demonstrated by FDA-approved nanomedicines, for example, Doxil®, a doxrubincin-liposome (1995), DaunoXome®, a daunorubincin-liposome (1996), and Vyxeos®, a daunorubicin/cytarabine-liposome (2017) [25]. However, the clinical development of nanomedicine for AMR pathogens has met a few challenges which remain today. As an example, although quick uptake of nanoparticles by macrophages has provided a basis for the treatment of intracellular bacterial infections, the rapid clearance of nanoparticles by the mononuclear phagocytic system (MPS) and reticuloendothelial system is still a significant challenge. Other challenges for antibacterial nanomedicine include overcoming other biological barriers, including vascular barriers and microenvironmental barriers such as biofilm for targeted delivery.

    Adsorption of serum protein by nanoparticles forms a nanoparticle protein corona, which alters the size, shape and surface properties of nanoparticles, and often leads to rapid clearance. Some cancer nanomedicines are therefore formulated with PEGylated lipids to minimize their interaction with serum proteins to achieve a stealth effect thus providing more stability and longer circulation time. However, a new challenge has emerged since the discovery that anti-PEG antibodies could be generated, which can in turn interact with nanoparticles, therefore enhancing the rapid clearance by the immune response. Cancer nanomedicines overcome vascular barriers by taking advantage of highly porous capillaries between endothelial cells around abnormal tumor tissues for enhanced extravasation and sequestration into the tumor [26,27]. The stealth effect as well as the enhanced permeability and retention are believed to be essential characteristics for nanomedicine. Additionally, the diverse cellular, intracellular and microenvironmental barriers associated with patients are also found to be significant challenges. In order to address these challenges, personalized chemoimmunotherapy nanomedicines have been proposed as cancer treatment in the future [25,28,29].

    Antibacterial nanoparticles

    In order to be clinically successful, antibacterial nanomedicines need to be serum-stable to avoid rapid clearance by the immune system and be able to cross the vascular barrier effectively. Antimicrobial agents could then be efficiently delivered to the infection site. The approach needs the nanomedicine to penetrate biofilms, leading to antibiotic uptake by bacteria through fusion and/or diffusion, thus to achieve improved antibacterial activity and pharmacologic properties [30,31]. Serum-stable nanoparticles also provide a possibility to include those potent but toxic antimicrobial agents, so that the side effects can be minimized through targeted delivery without premature release [32]. In this respect, the combination of nanotechnology and antimicrobial agents is one of the most promising strategies to cope with AMR.

    Most successful antibacterial nanomedicines so far are delivered directly to proximity of the infection site, including the nanomedicine Arikayce™, an amikacin liposome inhalation suspension approved by the FDA for the treatment of Mycobacterium avium complex lung disease [33,34]. Further clinical trials were conducted on Arikayce™ against P. aeruginosa in cystic fibrosis patients, but this did not show benefits compared with the control group [35]. Other nanoagents, particularly silver nanoparticles, were also tested [31,36]. Table 3 lists those clinical developments (https://clinicaltrials.gov/). The progress and current status in the field have been extensively described [37–42], including applications of both organic (liposomes, lipid-based nanoparticles, polymeric micelles and polymeric nanoparticles) and inorganic (silver, silica, magnetic, zinc oxide, cobalt, selenium and cadmium) nanosystems. Studies on metallic nanoparticles as carriers for antibiotics – for example, glycopeptide antibiotics and daptomycin, to treat multidrug-resistant pathogens, particularly S. aureus, as well as E. coli, P. aeruginosa and A. baumannii – were also described by Berini et al. [43]. The metallic nanoparticles are often coated with biocompatible polymers like PEG, chitosan, polyvinylpyrrolidone and poly(vinyl alcohol), as well as surfactants, proteins (such as bovine serum albumin) and oligonucleotides to increase both the stability of the nanocomposite and the antibacterial activity [44,45]. Although some metallic nanoparticles, such as the most extensively studied silver nanoparticles, can act as broad-spectrum antibacterial agents (like SilvaSorb®), as well as carriers of antibiotics [36,46,47], the emergence of silver-resistant bacteria has been observed similar to the antibiotic resistance through the process of natural selection [48].

    Table 3. Nanoparticles tested in clinical trials against antibiotic-resistant bacteria.
    Trade nameNanoparticle typeActive agentTarget pathogens or infectionClinical trial phaseClinical trial number
    Arikayce™Liposomal (inhalation)AmikacinMycobacterium avium complexMarketedNCT02344004
    Arikayce™Liposomal (inhalation)AmikacinPseudomonas aeruginosa in cystic fibrosis patientsIII
    Without approval
    NCT01315678
    Bronchiectasis (P. aeruginosa)IINCT00775138
    Pulmaquin®Liposomal (inhalation)CiprofloxacinNon-CF Bronchiectasis (P. aeruginosa history)III
    Without approval
    NCT02104245
    (Linhaliq)NCT01515007
    NCT02104245
    SilvaSorb®AgNP gelSilverTopical infectionMarketedNCT00659204
    AgTive®AgNPSilverCentral venous catheter-related infectionIVNCT00337714
    NAAgNP (on nutrient agar)Silver + antibioticsStaphylococcus aureus and P. aeruginosa (in vitro)NANCT04775238 recruiting

    AgNP: Silver nanoparticle; NA: Not applicable.

    The ability of nanoparticles to penetrate biofilms, particularly those formed by ESKAPE bacteria, provides a possibility to overcome multidrug resistance with nanomedicine [49]. Antibiotics conjugated to nanocarriers [50] or encapsulated by a variety of nanoparticles all showed favorable antibacterial activities [36,44], by inhibiting biofilm formation and enhancing intracellular delivery. Despite clear evidence of their strong antibacterial efficacy, the metal oxide-based nanoparticles, which are only effective to address bacterial biofilms in wound dressing, surfaces of medical devices and dental materials [38], are unlikely to be applied to systematic infections due to safety concerns [51,52]. Meanwhile, the lipid-based nanoparticles have emerged not only as a topical treatment for bacterial infections [53–55], but have also been explored as antibacterial agents delivered systemically. Although the clinical outcome so far has been disappointing [56], hitherto, no lipid-based nanomedicine has been approved by the FDA for systematic treatment of bacterial infection.

    In a critical review, Crommelin et al. assessed the current status of clinical development of nanomedicines, and analyzed the nanomedicine research from academia and industry over two decades [57]. The most successful family within the broad field of nanomedicine so far were liposomes, while other families of nanosized drug-delivery systems have not met their expectations. Based on lessons learned from cancer nanomedicine development, antibacterial nanomedicine will likely follow a similar path; therefore, the ‘liposome experience’ should be valued in the antibacterial nanomedicine development. With the success in anticancer nanomedicine development and successful introduction of mRNA-liposome vaccines [58], lipid-based nanoparticles, including liposomes and solid lipid nanoparticles, could be favorable choices for anti-AMR nanomedicine. To enhance the interaction with specific targets, the liposome surface might be modified with proteins, antibodies, carbohydrates or immunoglobulin fragments, in addition to pH-sensitive and thermosensitive liposomal vesicles. Those modifications, however, need to be carefully executed with consideration of their burdens to immunological and biological barriers. The significant differences in microenvironments between cancer and bacterial infections also need to be properly addressed in the nanoparticle engineering and formulation. A few reviews on antibiotic liposomes are listed in Table 4, with their titles and highlights. The major challenges in antibacterial nanomedicine engineering are similar to those encountered in cancer nanomedicine development. In order to translate a promising technology into clinical reality, antibacterial nanoparticles, like liposomes, should be properly designed with deep understanding of the biological interactions involved, and meanwhile formulated with a simple approach. The following discussion provides some challenges and prospects on future anti-AMR nanomedicine development.

    Table 4. Selected reviews on liposome-delivery system against bacteria and bacterial infection.
    TitleHighlightsRef.
    Liposomes as antibiotic delivery systems: a promising nanotechnological strategy against antimicrobial resistanceThe review outlined various current antibiotic-liposome formulations, described increased antibiotic concentration in infection site, explained samples given on liposome overcoming antimicrobial resistance and biofilm formation, and concluded antibiotic-liposome as an effective therapeutic strategy for bacterial infections.[59]
    Liposomes for antibiotic encapsulation and deliveryThis review discussed the state of knowledge regarding the design of liposomes for encapsulation and delivery of antibiotics and provided insight into the challenges and promises of using liposomes for antibiotic delivery.
    The review also pointed out the need to understand more about the interactions of liposomes with planktonic bacteria and biofilms.
    [60]
    Liposomal delivery systems and their applications against Staphylococcus aureus and methicillin-resistant Staphylococcus aureusThe review discussed vancomycin, azithromycin, clarithromycin and other antibiotic agents in liposome formations against S. aureus and methicillin-resistant S. aureus; most reported better antibacterial activity compared with bare drug in in vitro or topical studies. Very few in vivo studies were described on the potential application of liposomal delivery systems.[61]
    A review of liposomes as a drug-delivery system: current status of approved products, regulatory environments, and future perspectivesThe review included a list of liposomal drug products including one against Mycobacterium avium complex lung disease approved by the US FDA and EMA, and discussed the relevant technologies applied in the marketed liposomal products, including the lipid excipient, manufacturing methods, nanosizing technique, drug-loading methods, etc. which can be applied to preparation of liposomal antibiotics.[62]
    Liposomes as delivery systems for antibioticsThis early review described the fundamentals on liposome and provided antibiotic-liposome examples in clinical development. Although most results were not satisfactory, liposomal amikacin for inhalation was capable of eradicating Pseudomonas aeruginosa cells from sputum samples of cystic fibrosis patients. This illustrates the importance of improving systematic delivery.[56]
    Lipid-based antimicrobial delivery systems for the treatment of bacterial infectionsMany Gram-negative and Gram-positive bacterial strains, resistant to a specific antibiotic free in solution, have been demonstrated to be more susceptible to these antibiotics when encapsulated in a liposomal nanocarrier. This may be a result from liposomal encapsulation which prevented enzymatic deactivation and from fusogenicity of liposomes which effectively delivered antibiotic to bacteria.[63]
    Systematic review on activity of liposomal encapsulated antioxidant, antibiotics, and antiviral agentsThe review showed that liposomal antimicrobial agent and liposomal antioxidants enhance the solubility, bioavailability and stability of antimicrobial agent and antioxidants, and make antioxidants more effective in protection against the damaging actions of reactive oxygen species.[64]

    Challenges & prospects

    Immunologic barrier

    Rapid clearance of nanoparticles by phagocytosis is a common problem faced during nanomedicine development [65]. Nanoparticles in circulation often adsorb various serum proteins on their surface to form coronas. If opsonin, immunoglobulins or complement protein are attached, their interaction with phagocytes will lead to a rapid clearance by MPS. Most nanoparticles are then taken up and accumulated in the spleen and liver. On the other hand, the adsorption of serum albumin may protect nanoparticles from phagocytic uptake, which could contribute to the stealth effect of nanoparticles [66–68]. Since the distinct corona protein may determine the outcome of serum stability of nanoparticles [67], it is plausible to engineer nanoparticles that are able to form a specific corona by refining surface charge and other surface properties. For example, it has been shown that opsonins interact more favorably with positively charged nanoparticles than those negatively charged, whereas neutral and slightly negative charged nanoparticles are observed with longer circulation time [69,70]. In addition, when a protein corona forms, the ligands on nanoparticle surface might be blocked as well, losing their targeting capability [71]. Furthermore, the nanoparticles of size larger than 200 nm can activate the complement system [72], which also leads to rapid clearance.

    One common method to improve serum stability of nanoparticles is to incorporate PEG as a stealth coating. However, nanoparticle PEGylation does not completely prevent the protein adsorption and the recognition by macrophages and other immune cells. Exposure to PEG also leads to the production of anti-PEG antibodies. Those anti-PEG antibodies can, in turn, aid the rapid clearance of PEGylated nanomedicine. Since PEGylated agents are commonly used in medicine or nonmedicine, pre-existing anti-PEG antibodies have been detected in humans. Additionally, PEGylated nanoparticles have been shown to cause severe allergic reactions and anaphylaxis in a small number of patients, suggesting PEGylation may not be a long-term solution. Mitigation approaches include use of cleavable PEG [73], and with formulations of chitosan [74] and dextran [75] included in nanoparticles. In the face of the observed drawbacks to PEGylated nanoparticles, the alternative hydrophilic and nonimmunogenic coating agents would be preferred in antibacterial nanomedicine. It is plausible that carbohydrates could play such a role as they are hydrophilic and, in many cases, poorly immunogenic. Indeed, bacteria are covered with carbohydrate with a purpose to evade the innate immune system. Since the nanoparticle as a carrier may augment the immunogenicity of antigens on its surface, it is important that only nonimmunogenic carbohydrates be incorporated. In fact, human glycoantigens, from either glycoprotein or glycolipid, have been introduced onto the nanoparticle surface as recently described by Singla et al., where ciprofloxacin-loaded liposomes functionalized with host cell glycans improved the delivery of antibiotics to the site of P. aeruginosa infection, likely due to increased retention of the liposomes in the circulation [75]. Another option is to optimize the surface properties of nanoparticles through the use of novel lipids and formulation, so that only serum albumin is selectively adsorbed as a stealth coating to avoid the uptake by phagocytes and rapid clearance from circulation.

    Since the interaction with serum proteins can bring significant changes to nanoparticles, such as size increase, surface charge neutralization, blocked ligands and even modified nanoparticle structure, one must be very mindful in the nanoparticle design by taking consideration of all those possibilities, and if possible, take advantage of those changes.

    Vascular barrier

    The nanoparticles circulating in the bloodstream need to cross the vascular barrier to achieve targeted delivery. It has been observed that the size, shape and surface properties all affect the attachment of nanoparticles to the blood vessel wall [76,77], and the complicated process is not fully predictable [78]. Optimized particle size (<200 nm) exhibited in vitro the greatest extent of vascular stability as well as the lowest extravasation [79]. Since the abnormal growth of tumors leads to formation of small pores between the endothelial cells [80], nanoparticles can traverse these pores and reach the interstitial sites in tumors and deliver the loaded drug to cancer cells.

    Similarly, during infection, bacteria produce toxins and other virulence factors that can disrupt the barrier function of the endothelium to enter and exit the bloodstream. Those damaged endothelial cells, weakened cytoskeleton and broken junctions between endothelial cells would passively allow nanoparticle extravasation. Additionally, endothelial cells overexpress adhesion molecules during infection, such as selectins that bind to certain carbohydrates presented on the leukocyte glycoprotein, which facilitates leukocyte to leave the blood vessel and enter the site of infection. Thus, in order to achieve active extravasation, nanoparticles may be modified with those carbohydrates, and it is known that sialylated and sulfated carbohydrates on glycoproteins, glycolipids and proteoglycans can be relevant ligands for selectins [81].

    In an early review by Forssen and Willis functional liposomes for targeted delivery against cancer were discussed, which included surface modifications with antibodies, peptides, carbohydrates, aptamers and cell surface receptors [82]. Folate receptors, adhesion molecules, extracellular matrix molecules and selectins were all suggested as targets for site-directed cancer therapy. In one example, Maruyama et al. [83] provided evidence that liposomes modified with a highly specific antibody to pulmonary endothelial cells and additional less than 10% GM1 ganglioside (immunoliposomes) resulted in slow MPS uptake and its improved accumulation in lungs. Kalita et al. [84] described how to harness glyconanoparticles' distinct optical, magnetic and electronic properties for disease diagnosis with improved magnetic resonance imaging, fluorescence imaging, etc., by adding carbohydrates on metal oxide nanoparticles. Discussion also included using glyconanoparticles to imitate viral particles (or host cells) to interfere with the virus–cell membrane interactions, thus protecting the host cells from infection. Under this context, properly designed nanoparticles with surface modification of carbohydrates may overcome not only the immunological barrier but also the vascular barrier. This could be a sensible approach to deliver nanoparticles to the infection site as illustrated in Figure 1.

    Figure 1. Carbohydrate coating of nanoparticles can be an alternative to PEGylation.

    In addition to resisting rapid clearance by immune system, the interaction of carbohydrates as ligands with overexpressed receptors on endothelial cell, due to inflammation caused by infection, facilitate nanoparticle crossing the vascular barrier and targeted delivery. The drug release at infection sites could be achieved through nanoparticle–bacterial fusion or diffusion if the nanoparticle is degraded by biofilm.

    Microenvironment & drug-taking-up barriers (heterogeneity & biofilm)

    Bacteria create a unique microenvironment and often form biofilms, particularly in chronic infections. In addition to infective endocarditis, gum and wound-related infections, biofilms have been found in many diseases such as lung, colon, urethra, eye and ear infections. Once the nanoparticles pass through the immunological and vascular barriers, they still need to overcome those additional obstacles in order to deliver antibiotics to bacteria. The microenvironment is heterogeneous, depending on the type of bacterial infections, and often slightly acidic. Bacterial biofilms contain various enzymes, toxins and other molecules released from bacteria, such as DNA, proteins and polysaccharides [85]. It is this extracellular matrix that makes biofilm communities highly resistant to antibiotic treatment, as observed that bacterial cultures in intact biofilms are up to a 1000-fold more resistant to antibiotics than those cultures growing as planktonic (free-floating) forms [86], by limiting the penetration of antibiotics to reach the bacterium. Additionally, biofilms may lead to physiological changes in the bacteria, such as a drastic slowing of the growth phase leading to a state close to dormancy, and in some cases, bacteria can develop a resistant phenotype by altering drug targets for antibiotic activity [87]. Biofilms, such as those found in urinary tract and lung infections, as well as those in implanted medical devices [76,79], are often caused by ESKAPE bacteria [88]. For example, P. aeruginosa biofilm contributes to chronic lung infections in cystic fibrosis patients [89,90], while S. aureus, K. pneumonia, A. baumannii and Enterobacter spp. are all responsible for upper respiratory and lung infections and all but S. aureus are also responsible in many cases for urinary tract infections.

    Certain nanoparticles, including liposomes, micelles and inorganic nanoparticles, have shown the ability to penetrate the biofilm and bacterial cell walls to kill bacteria by various mechanisms [91], which may help design and engineer more sophisticated nanomedicines against this adversary. The interaction between nanoparticles and biofilm is very complex and not fully understood, but it is believed that the interaction is primarily determined by their electrostatic characteristics. These features depend on the zeta potential of the nanoparticle after its corona formation and extravasation, and the charge of the biofilm matrix [49]. It is believed that the negatively charged matrix can interact with positively charged nanoparticles to allow further diffusion toward bacterial cell, such as those nanoparticles developed from metals or metal oxides, synthetic or natural polymers, or hybrids therein. However, since only lipid-based nanomedicines are clinically approved by the FDA, it will be more fruitful to pursue similar strategies through careful refinements of nanoparticle lipid compositions, the hydrodynamic size, surface charge and other properties, to ensure efficient biofilm targeting and matrix interactions, thereby enhancing their antibacterial efficacy. However, a delicate balance is required in nanoparticle formulation since negatively charged nanoparticles are preferred to overcome immunological barriers while positive charged nanoparticles interact better with biofilm. Although antibiotic-liposomes may be able to fuse with the bacterial membrane and release the drug directly into the bacteria [92], it should be remembered that the nanoparticles at this stage are no longer the same in both size and surface properties after passing through the immunological, vascular and other biological barriers. Thus, the consequent interactions with biofilm are very different from those observed in in vitro experiments. Lipid-based nanoparticles are attractive candidates for nanomedicines but improving their biofilm penetration is still a major challenge. Engineering antibiotic-liposomes which can be degraded within biofilm matrix could be one of the solutions. The approach results in release of antibiotics close to bacteria for their spontaneous diffusion.

    Payload encapsulation

    To date, only a few therapeutic agents have been formulated into lipid-based nanomedicines which provided clinical advantage by improving the circulation time and the pharmacology. The nanomedicines, for example, Doxil, Onivyde® and Onpattro®, are PEGylated stealth nanoparticles whereas DaunoXome, AmBiosome®, Marqibo®, Vyxeos and LEP-ETU are formulated as conventional liposomes (Table 5). Active ingredients are encapsulated within the aqueous chamber of liposomes [93,94] or carried in the lipid bilayer [95–98]. The encapsulation capacity of those nanomedicines, which is measured by lipid-to-drug weight ratio, varies from approximately 2.2:1 in Onivyde to 30:1 in LEP-ETU (Table 5). With an 8:1 lipid-to-drug ratio, Doxil's dosage for cancer treatments at 20–50 mg/m2 requires roughly 40–100 mg of doxorubicin or close to 0.36–0.9 g of nanomedicine per treatment.

    Table 5. Examples of lipid-based nanomedicines: formulation, size and drug encapsulation.
    NanomedicineDrugFormulationPEGylatingDiameter (nm)Lipid-to-drug weight ratio (w/w)Ref.
    Doxil®DoxorubicinLiposome+ca. 100ca. 8:1[93]
    DaunoXome®DaunorubicinLiposome-45–80ca. 18.7:1[94]
    AmBisome®Amphotericin BLiposome-ca. 100ca. 7:1[95]
    Marqibo®VincristineLiposome-ca. 100ca. 20:1[99]
    Onivyde®IrinotecanLiposome+ca. 110ca. 2.2:1[100]
    Vyxeos®Daunorubicin/cytarabine (1:5)Liposome (bilamellar)-ca. 110ca. 4.3:1[101,102]
    Onpattro®Patisiran (siRNA)LNP+ca. 100ca. 12:1[97,98]
    LEP-ETUPaclitaxelLiposome-ca. 150ca. 30:1[96]

    ca.: Circa.

    The cancer drugs encapsulated in the FDA-approved and experimental nanomedicines are highly potent, whereas antibiotics are often much less potent with variable sensitivity toward different bacteria (Table 6). This could pose a challenge for antibiotic liposomal nanomedicine, particularly if the efficiency of the antibiotic encapsulation is suboptimal. The encapsulation limitation may be addressed with novel lipid-based nanoparticles, formulated with biodegradable and biocompatible materials, instead of phospholipids. As choices of antibiotics have already been limited [103], antibacterial nanomedicines would likely include a combined drug regime to improve efficacy, reduce drug resistance and minimize toxicity. Additionally, the limitation on antibiotic payload could be mitigated if the nanoparticles have a more favourable serum stability and an effective ligand-directed extravasation to quickly reach and release antibiotics at infection site as we have discussed above (Figure 1).

    Table 6. MIC (mg/l) of antibiotics to antibiotic-resistant Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. bacteria.
    AntibioticE. faeciumS. aureusKl. pneumoniaA. baumanniiP. aeruginosaEnterobacter spp.
    Vancomycin 0.5–8    
    Penicillin≥160.25–32    
    Colistin   0.38 (MIC50)
    1 (MIC90)
    1 (MIC50)
    2 (MIC90)
     
    Ampicillin  >256   
    Piperazine  >16   
    Cephalothin  >32   
    Cefoxitin  >16   
    Clindamycin <0.0156–0.5    
    Ciprofloxacin   >32 0.006–0.023
    Tigecycline   2 (MIC50)
    4 (MIC90)
     0.5–1.5

    Data taken from references [104–109].

    Ligands for bacterial targeting or other receptors

    Early anticancer nanomedicines were formulated without specific ligands. The improved efficacy is largely due to improved circulation time and passive extravasation to cancer location due to the fenestrated blood vessels. More cancer nanomedicines with ligands such as specific monoclonal antibodies and other targeting moieties are believed to be able to enhance targeted delivery. Although ligand-aided liposomes delivery has been explored, and encouraging results were obtained in animal models, its potential benefits in patients have yet to be clinically established [110–112].

    Unlike cancer, bacterial infection progresses much more quickly and diversely, and it is not always possible to identify exactly the bacterium which causes infection in clinical settings. The lack of common receptors on bacterial surface and the presence of microbial biofilms have presented a serious challenge for bacteria-specific targeting, which seems difficult to overcome at this moment [113], even if bacteria-specific nanoparticles are developed with monoclonal and single domain antibodies, or other ligands by rapid assembly at bedside. When nanoparticle passes through the biofilm matrix, the ligand is no longer essential since the nanoparticle is already in the proximity of bacteria. Realistically, rather than directly targeting bacteria, the focus of targeted delivery should be more on enhancing the nanomedicine to reach the infection site through improving serum stability and active extravasation, which can be achieved, for example, through targeting adhesion molecules of the vascular endothelial cells (such as selectins), whose expression is often increased near the site of inflammation and infection. Nanoparticles that may degrade by interaction with biofilm matrix could also be considered as a means of targeted delivery.

    Conclusion

    Targeted delivery of therapeutic agents has been a long-sought necessity for the effective treatment of serious diseases with fewer side effects. Nanotechnology may provide such a platform to achieve this goal. Targeting cancer biomarkers, bacterial surface components and vascular adhesion molecules with functional nanoparticles is an attractive approach, but due to individual heterogeneity and changing disease states, optimal target selection would be a great challenge. Unlike cancer treatment, which is aided with precision diagnosis and regime optimization, antibacterial nanomedicine faces unique challenges. Acute bacterial infection must be dealt with quickly and decisively, often without knowing the nature of the bacterial infection. Therefore, the new paradigm in antibacterial nanomedicine should focus more on the engineering of stable, yet, biodegradable nanoparticles with stealth coating or the ability to acquire stealth through selective corona formation, such to resist the rapid clearance by immune system. Meanwhile the surface functional ligand which recognizes specific molecules on the vascular endothelium near the infection site remains a sound choice to facilitate vascular crossing and therapeutic delivery. This approach requires no information on what bacteria caused infection, and the nanomedicine with broad-spectrum antibiotics encapsulated may provide desired pharmacological properties and therapeutic outcome.

    However, after over three decades of intensive research and clinical development, only a limited number of nanoparticles have translated into nanomedicines to benefit patients. In addition to those challenges described above, the toxicity, manufacture and cost-associated concerns on nanoparticles have to be addressed [114]. More importantly, a deeper understanding of interplay between engineered nanomaterials and biological systems is required. The expectation in the translation of nanoparticles to nanomedicines can only be met by properly designed and engineered nanoparticles that could navigate through complex physical, immunological and biological barriers, and finally deliver desired doses of antibiotics to bacteria. Each step poses unique challenges, and a concerted overall strategy is required to achieve positive clinical outcomes.

    Future perspective

    Nanomedicine is designed as a vaccine and therapeutic delivery system, promising to provide improved efficacy and reduced side effects. However, only a few in clinical development so far have met the expectation. Unlike proposed future treatment for cancer by individualized nanomedicine, nanomedicine for AMR must be readily accessible and affordable. Simplicity must be a goal for anti-AMR nanomedicine, which can be gradually achieved through further understanding of the immune responses toward nanomedicine by the human body, and the biological interactions interplayed among nanomedicine, heterogeneous microbes and diverse human systems. A relationship between nanostructure and function may eventually emerge, which could guide nanomedicine design.

    In the near future, it is desired to engineer more robust nanoparticles with synthetic or natural biocompatible and biodegradable materials. They must provide better consistency and control in size and surface charges than current nanomedicines, with improved encapsulation, and be ready for in situ add-on as a multicomponent system for targeted delivery. More specifically, the antibacterial nanomedicine must overcome heterogeneous biofilm, the last and the most challenging barrier. As to clinical development, we believe that the focus of nanoparticle design should be more on taking advantage of the extracellular environment rather than on particular molecular structures. The delivery of therapeutics to bacteria can then be achieved through either fusion or diffusion.

    Finally, similar to virus-like particles as a promising vaccine platform against various diseases, nanoparticles, particularly the lipid-based nanoparticles, could be built through a combination of biosynthesis and chemical synthesis as microbe-like particles, which can be potentially explored as effective vaccines against AMR, other than as vaccine/immunogen delivery systems.

    Executive summary
    • Antimicrobial resistance poses a great challenge to human health and economic development.

    • Multiantibiotic-resistant bacteria – such as Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. in particular – need to be dealt with urgently.

    • Since the development of new antibiotics and vaccines has not yielded the desired outcome, alternative approaches have been explored, and antibacterial nanomedicine has emerged as one of the attractive solutions.

    Antibacterial nanoparticles

    • Extensive investigation on both organic (liposomes, lipid-based nanoparticles, polymeric micelles and polymeric nanoparticles) and inorganic (silver, silica, magnetic, zinc oxide, cobalt, selenium and cadmium) nanosystems is concisely summarized.

    • Liposome and lipid-based nanoparticles are more likely to become antibacterial nanomedicines due to the established safety and pharmacological profiles from their anticancer clinical applications.

    Challenges & prospects

    • Development of antibacterial nanomedicine is required to overcome challenges related to immunologic barriers (rapid clearance by immune system), and other biological barriers such as vascular barrier, heterogeneity, biofilm and drug-taking-up barriers.

    • Effective payload encapsulation and ligand selection for bacterial targeting are also challenges.

    • A preferred nanoparticle formulation is proposed to include nonimmunogenic carbohydrates, which act not only as a stealth coating to avoid rapid clearance by the immune system, but also as ligands of specific endothelium receptors to facilitate nanomedicines crossing the vascular barrier.

    • The successful translation of nanoparticle to nanomedicine can only be achieved by properly designed and engineered nanoparticles, which requires more knowledge on the dynamic nature of nanoparticles' physical, immunological and biological interactions with both host and bacteria.

    • A concerted overall strategy is required to reach positive clinical outcomes.

    Crown copyright

    This work is licensed under a Crown Copyright protection and licensed for use under the Open Government License unless otherwise indicated. Where any of the Crown copyright information in this work is republished or copied to others, the source of the material must be identified and the copyright status under the Open Government License acknowledged.

    Author contributions

    Conception: W Chen and W Zou. Initial manuscript drafting: A McAdorey and W Zou. Review and final manuscript preparation: H Yan, W Zou, A McAdorey and W Chen.

    Financial & competing interests disclosure

    The research in our laboratories has been supported by the National Research Council Canada (NRC) Ideation Small Team Project (National Program Office), by NRC intramural (A base) program under the Vaccine and Emerging Infection Research Initiative (Human Health Therapeutics) and by a Natural Sciences and Engineering Research Council of Canada (grant no. RGPIN-2020-07040). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    Refercences

    • 1. Murray CJ, Ikuta KS, Sharara F et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 399(10325), 629–655 (2022).
    • 2. Reygaert WC. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 4(3), 482–501 (2018).
    • 3. Uruén C, Chopo-Escuin G, Tommassen J, Mainar-Jaime RC, Arenas J. Biofilms as promoters of bacterial antibiotic resistance and tolerance. Antibiot. 10(1), 1–36 (2021).
    • 4. Hall CW, Mah TF. Molecular mechanisms of biofilm-based antibiotic resistance and tolerance in pathogenic bacteria. FEMS Microbiol. Rev. 41(3), 276–301 (2017).
    • 5. Andrei S, Droc G, Stefan G. FDA approved antibacterial drugs: 2018–2019. Discoveries 7(4), e102 (2019).
    • 6. Kaur R, Rani P, Atanasov A et al. Discovery and development of antibacterial agents: fortuitous and designed. Mini Rev. Med. Chem. 22(7), 984–1029 (2022).
    • 7. Tacconelli E, Carrara E, Savoldi A et al. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 18(3), 318–327 (2018).
    • 8. Rice LB. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J. Infect. Dis. 197(8), 1079–1081 (2008).
    • 9. Navidinia M. The clinical importance of emerging ESKAPE pathogens in nosocomial infections. Arch. Adv. Biosci. 7(3), 43–57 (2016).
    • 10. Mulani MS, Kamble EE, Kumkar SN, Tawre MS, Pardesi KR. Emerging strategies to combat ESKAPE pathogens in the era of antimicrobial resistance: a review. Front. Microbiol. 10, 539 (2019). • Review outlining alternative therapies for the treatment of Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. infections with special highlight on the importance of a combinatorial approach.
    • 11. Frost I, Sati H, Garcia-Vello P et al. The role of bacterial vaccines in the fight against antimicrobial resistance: an analysis of the preclinical and clinical development pipeline. Lancet Microbe 4(2), e113–e125 (2023). • Review of vaccine candidates against the 2017 WHO bacterial priority pathogen list.
    • 12. Darby EM, Trampari E, Siasat P et al. Molecular mechanisms of antibiotic resistance revisited. Nat. Rev. Microbiol. 21(5), 280–295 (2022).
    • 13. Santajit S, Indrawattana N. Mechanisms of antimicrobial resistance in ESKAPE pathogens. Biomed Res. Int. 2016, 2475067 (2016).
    • 14. Lewis K. Multidrug tolerance of biofilms and persister cells. In: Current Topics in Microbiology and Immunology, Bacterial Biofilms. Romero T (Ed.). Springer, Berlin, Heidelberg, Germany, 107–131 (2008).
    • 15. Wang N, Luo J, Deng F, Huang Y, Zhou H. Antibiotic combination therapy: a strategy to overcome bacterial resistance to aminoglycoside antibiotics. Front. Pharmacol. 13, 1–15 (2022).
    • 16. Coates ARM, Hu Y, Holt J, Yeh P. Antibiotic combination therapy against resistant bacterial infections: synergy, rejuvenation and resistance reduction. Expert Rev. Anti. Infect. Ther. 18(1), 5–15 (2020).
    • 17. Hatfull GF, Dedrick RM, Schooley RT. Phage therapy for antibiotic-resistant bacterial infections. Annu. Rev. Med. 73, 197–211 (2022).
    • 18. Woodworth MH, Hayden MK, Young VB, Kwon JH. The role of fecal microbiota transplantation in reducing intestinal colonization with antibiotic-resistant organisms: the current landscape and future directions. Open Forum Infect. Dis. 6(7), 1–9 (2019).
    • 19. Suetake I, Shinozaki F, Miyagawa J, Takeshima H, Tajima S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J. Biol. Chem. 279(26), 27816–27823 (2004).
    • 20. Gargiullo L, Del Chierico F, D'Argenio P, Putignani L. Gut microbiota modulation for multidrug-resistant organism decolonization: present and future perspectives. Front. Microbiol. 10, 1704 (2019).
    • 21. Montassier E, Valdés-Mas R, Batard E et al. Probiotics impact the antibiotic resistance gene reservoir along the human GI tract in a person-specific and antibiotic-dependent manner. Nat. Microbiol. 6(8), 1043–1054 (2021).
    • 22. Mariathasan S, Tan MW. Antibody–antibiotic conjugates: a novel therapeutic platform against bacterial infections. Trends Mol. Med. 23(2), 135–149 (2017).
    • 23. Baah S, Laws M. Antibody–drug conjugates – a tutorial review. Molecules 26(10), 2943 (2021).
    • 24. Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update. Bioeng. Transl. Med. 4(3), 1–16 (2019).
    • 25. Beltrán-Gracia E, López-Camacho A, Higuera-Ciapara I, Velázquez-Fernández JB, Vallejo-Cardona AA. Nanomedicine Review: Clinical developments in Liposomal Applications (Volume 10). Springer, Vienna, Austria (2019).
    • 26. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 91(1), 3–6 (2015).
    • 27. Das CGA, Kumar VG, Dhas TS, Karthick V, Kumar CMV. Nanomaterials in anticancer applications and their mechanism of action – a review. Nanomed. Nanotechnol. Biol. Med. 47(5), 2022 (2023).
    • 28. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 20(2), 101–124 (2021).
    • 29. Sudheesh MS, Pavithran K, Sabitha M. Revisiting the outstanding questions in cancer nanomedicine with a future outlook. Nanoscale Adv. 4(3), 634–653 (2022).
    • 30. Wang S, Gao Y, Jin Q, Ji J. Emerging antibacterial nanomedicine for enhanced antibiotic therapy. Biomater. Sci. 8(24), 6825–6839 (2020).
    • 31. Makabenta JMV, Nabawy A, Li CH, Schmidt-Malan S, Patel R, Rotello VM. Nanomaterial-based therapeutics for antibiotic-resistant bacterial infections. Nat. Rev. Microbiol. 19(1), 23–36 (2021). •• Review highlighting targeting mechanisms for nanomaterials, engineering design and properties.
    • 32. Singh S, Hussain A, Shakeel F et al. Recent insights on nanomedicine for augmented infection control. Int. J. Nanomed. 14, 2301–2325 (2019).
    • 33. Shirley M. Amikacin liposome inhalation suspension: a review in Mycobacterium avium complex lung disease. Drugs 79(5), 555–562 (2019).
    • 34. Griffith DE, Eagle G, Thomson R et al. Amikacin liposome inhalation suspension for treatment-refractory lung disease caused by Mycobacterium avium complex (CONVERT) a prospective, open-label, randomized study. Am. J. Respir. Crit. Care Med. 198(12), 1559–1569 (2018).
    • 35. Bilton D, Pressler T, Fajac I et al. Amikacin liposome inhalation suspension for chronic Pseudomonas aeruginosa infection in cystic fibrosis. J. Cyst. Fibros. 19(2), 284–291 (2020).
    • 36. Hawthorne GH, Bernuci MP, Bortolanza M, Issy AC, Del-Bel E. Chapter 29 – Clinical developments in antimicrobial nanomedicine: toward novel solutions. In: Nanostructures for Antimicrobial Therapy: Nanostructures in Therapeutic Medicine Series. Elsevier Inc., 653–668 (2017).
    • 37. Yacoby I, Benhar I. Antibacterial nanomedicine. Nanomed. 3(3), 329–341 (2008).
    • 38. Wang L, Hu C, Shao L. The antimicrobial activity of nanoparticles: present situation and prospects for the future. Int. J. Nanomed. 12, 1227–1249 (2017).
    • 39. Juneja M, Suthar T, Pardhi V, Ahmad J, Jain K. Emerging trends and promises of nanoemulsions in therapeutics of infectious diseases. Nanomed. (Lond.) 17(11), 793–812 (2022).
    • 40. Aderibigbe BA. Metal-based nanoparticles for the treatment of infectious diseases. Molecules 22(8), 1370 (2017).
    • 41. Vanamala K, Tatiparti K, Bhise K et al. Novel approaches for the treatment of methicillin-resistant Staphylococcus aureus: using nanoparticles to overcome multidrug resistance. Drug Discov. Today 26(1), 31–43 (2021).
    • 42. Eleraky NE, Allam A, Hassan SB, Omar MM. Nanomedicine fight against antibacterial resistance: an overview of the recent pharmaceutical innovations. Pharmaceutics 12(2), 142 (2020).
    • 43. Berini F, Orlandi V, Gornati R, Bernardini G, Marinelli F. Nanoantibiotics to fight multidrug resistant infections by Gram-positive bacteria: hope or reality? Biotechnol. Adv. 57, 107948 (2022). •• Review covering latest developments in nanoparticle use as carriers of frontline antibiotics on multidrug-resistant Gram-positive pathogens.
    • 44. Kvítek L, Panáček A, Soukupová J et al. Effect of surfactants and polymers on stability and antibacterial activity of silver nanoparticles (NPs). J. Phys. Chem. C 112(15), 5825–5834 (2008).
    • 45. Javed R, Zia M, Naz S, Aisida SO, Ain NU, Ao Q. Role of capping agents in the application of nanoparticles in biomedicine and environmental remediation: recent trends and future prospects. J. Nanobiotechnol. 18, 172 (2020).
    • 46. Lubkin A, Torres VJ. Bacteria and endothelial cells: a toxic relationship. Curr. Opin. Microbiol. 35, 58–63 (2017).
    • 47. Rai MK, Deshmukh SD, Ingle AP, Gade AK. Silver nanoparticles: the powerful nanoweapon against multidrug-resistant bacteria. J. Appl. Microbiol. 112(5), 841–852 (2012).
    • 48. McNeilly O, Mann R, Hamidian M, Gunawan C. Emerging concern for silver nanoparticle resistance in Acinetobacter baumannii and other bacteria. Front. Microbiol. 12, 652863 (2021).
    • 49. Shkodenko L, Kassirov I, Koshel E. Metal oxide nanoparticles against bacterial biofilms: perspectives and limitations. Microorganisms 8(10), 1545 (2020).
    • 50. Skwarecki AS, Milewski S, Schielmann M, Milewska MJ. Antimicrobial molecular nanocarrier–drug conjugates. Nanomedicine 12(8), 2215–2240 (2016).
    • 51. Wolfram J, Zhu M, Yang Y et al. Safety of nanoparticles in medicine. Curr. Drug Targets 16(14), 1671–1681 (2015).
    • 52. Najahi-Missaoui W, Arnold RD, Cummings BS. Safe nanoparticles: are we there yet? Int. J. Mol. Sci. 22(1), 1–22 (2021). •• Recent report summarizing nanoparticle toxicity and current approaches to minimizing toxicity.
    • 53. Rukavina Z, Šegvić Klarić M, Filipović-Grčić J, Lovrić J, Vanić Ž. Azithromycin-loaded liposomes for enhanced topical treatment of methicillin-resistant Staphyloccocus aureus (MRSA) infections. Int. J. Pharm. 553, 109–119 (2018).
    • 54. Castoldi A, Herr C, Niederstraßer J et al. Calcifediol-loaded liposomes for local treatment of pulmonary bacterial infections. Eur. J. Pharm. Biopharm. 118, 62–67 (2017).
    • 55. Vanić Ž, Rukavina Z, Manner S et al. Azithromycin-liposomes as a novel approach for localized therapy of cervicovaginal bacterial infections. Int. J. Nanomed. 14, 5957–5976 (2019).
    • 56. Drulis-Kawa Z, Dorotkiewicz-Jach A. Liposomes as delivery systems for antibiotics. Int. J. Pharm. 387, 187–198 (2010).
    • 57. Crommelin DJA, van Hoogevest P, Storm G. The role of liposomes in clinical nanomedicine development. What now? Now what? J. Control. Rel. 318, 256–263 (2020).
    • 58. Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid nanoparticles from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano 15(11), 16982–17015 (2021).
    • 59. Ferreira M, Ogren M, Dias JNR et al. Liposomes as antibiotic delivery systems: a promising nanotechnological strategy against antimicrobial resistance. Molecules 26(7), 2047 (2021).
    • 60. Gomez AG, Hosseinidoust Z. Liposomes for antibiotic encapsulation and delivery. ACS Infect. Dis. 6(5), 896–908 (2020).
    • 61. Nwabuife JC, Pant AM, Govender T. Liposomal delivery systems and their applications against Staphylococcus aureus and methicillin-resistant Staphylococcus aureus. Adv. Drug Deliv. Rev. 178, 113861 (2021).
    • 62. Liu P, Chen G, Zhang J. A review of liposomes as a drug delivery system: current status of approved products, regulatory environments, and future perspectives. Molecules 27(4), 1372 (2021).
    • 63. Wang DY, van der Mei HC, Ren Y, Busscher HJ, Shi L. Lipid-based antimicrobial delivery-systems for the treatment of bacterial infections. Front. Chem. 7, 872 (2020).
    • 64. K R R, Balakrishnan P, Gopi S. Systematic review on activity of liposomal encapsulated antioxidant, antibiotics, and antiviral agents. J. Liposome Res. 32(4), 340–353 (2022).
    • 65. Gustafson HH, Holt-Casper D, Grainger DW, Ghandehari H. Nanoparticle uptake: the phagocyte problem. Nano Today 10(4), 487–510 (2015).
    • 66. Walkey CD, Olsen JB, Guo H, Emili A, Chan WCW. Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake. J. Am. Chem. Soc. 134(4), 2139–2147 (2012).
    • 67. Ritz S, Schöttler S, Kotman N et al. Protein corona of nanoparticles: distinct proteins regulate the cellular uptake. Biomacromolecules 16(4), 1311–1321 (2015). • Study found a mixed protein corona forms as a result of the nanoparticles' physiochemical properties.
    • 68. Ogawara KI, Furumoto K, Nagayama S et al. Pre-coating with serum albumin reduces receptor-mediated hepatic disposition of polystyrene nanosphere: implications for rational design of nanoparticles. J. Control. Rel. 100(3), 451–455 (2004).
    • 69. Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat. Biotechnol. 33(9), 941–951 (2015).
    • 70. Kou L, Bhutia YD, Yao Q, He Z, Sun J, Ganapathy V. Transporter-guided delivery of nanoparticles to improve drug permeation across cellular barriers and drug exposure to selective cell types. Front. Pharmacol. 9, 27 (2018).
    • 71. Salvati A, Pitek AS, Monopoli MP et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nat. Nanotechnol. 8(2), 137–143 (2013).
    • 72. Soo Kim M, Haney M, Yuling Z et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 12(3), 655–664 (2016).
    • 73. Fang Y, Xue J, Gao S et al. Cleavable PEGylation: a strategy for overcoming the ‘PEG dilemma’ in efficient drug delivery. Drug Deliv. 24, 22–32 (2017).
    • 74. Silva NC, Silva S, Sarmento B, Pintado M. Chitosan nanoparticles for daptomycin delivery in ocular treatment of bacterial endophthalmitis. Drug Deliv. 22(7), 885–893 (2015).
    • 75. Singla A, Simbassa SB, Chirra B et al. Hetero-multivalent targeted liposomal drug delivery to treat Pseudomonas aeruginosa infections. ACS Appl. Mater. Interfaces 14(36), 40724–40737 (2022).
    • 76. Chauhan VP, Stylianopoulos T, Boucher Y, Jain RK. Delivery of molecular and nanoscale medicine to tumors: transport barriers and strategies. Annu. Rev. Chem. Biomol. Eng. 2, 281–298 (2011).
    • 77. Vu MN, Rajasekhar P, Poole DP et al. Rapid assessment of nanoparticle extravasation in a microfluidic tumor model. ACS Appl. Nano Mater. 2(4), 1844–1856 (2019).
    • 78. Sheth V, Wang L, Bhattacharya R, Mukherjee P, Wilhelm S. Strategies for delivering nanoparticles across tumor blood vessels. Adv. Funct. Mater. 31(8), doi:10.1002/adfm.202007363 (2021).
    • 79. Pegaz B, Debefve E, Ballini JP, Konan-Kouakou YN, van den Bergh H. Effect of nanoparticle size on the extravasation and the photothrombic activity of meso(p-tetracarboxyphenyl)porphyrin. J. Photochem. Photobiol. B Biol. 85(3), 216–222 (2006).
    • 80. Hobbs SK, Monsky WL, Yuan F et al. Regulation of transport pathways in tumor vessels: role of tumor type and microenvironment. Proc. Natl Acad. Sci. 95(8), 4607–4612 (1998).
    • 81. Varki A. Review selectin ligands. Proc. Natl Acad. Sci. 91(16), 7390–7397 (1994).
    • 82. Forssen E, Willis M. Ligand-targeted liposomes. Adv. Drug Deliv. Rev. 29, 249–271 (1998).
    • 83. Maruyama K, Kennel SJ, Huang L. Lipid composition is important for highly efficient target binding and retention of immunoliposomes. Proc. Natl Acad. Sci. 87(15), 5744–5748 (1990).
    • 84. Kalita M, Payne MM, Bossmann SH. Glyco-nanotechnology: a biomedical perspective. Nanomedicine 42, 102542 (2022).
    • 85. Sharma D, Misba L, Khan AU. Antibiotics versus biofilm: an emerging battleground in microbial communities. Antimicrob. Resist. Infect. Control 8, 76 (2019).
    • 86. Rossi-Fedele G, Roberts AP. A preliminary study investigating the survival of tetracycline resistant Enterococcus faecalis after root canal irrigation with high concentrations of tetracycline. Int. Endod. J. 40(10), 772–777 (2007).
    • 87. Donlan RM. Biofilm formation: a clinically relevant microbiological process. Clin. Infect. Dis. 33(8), 1387–1392 (2001).
    • 88. Nicolle LE, Evans G, Laverdieve M, Phillips P, Quan C, Rotstein C. Complicated urinary tract infection in adults. Can. J. Infect. Dis. Med. Microbiol. 16(6), 349–360 (2005).
    • 89. Hoiby N, Ciofu O, Bjarnsholt T. Pseudomonas aeruginosa biofilms in cystic fibrosis. Future Microbiol. 5(11), 1663–1674 (2010).
    • 90. Kolpen M, Kragh KN, Enciso JB et al. Bacterial biofilms predominate in both acute and chronic human lung infections. Thorax 77(10), 1015–1022 (2022).
    • 91. Li X, Chen D, Xie S. Current progress and prospects of organic nanoparticles against bacterial biofilm. Adv. Colloid Interface Sci. 294, 102475 (2021).
    • 92. Nisini R, Poerio N, Mariotti S, De Santis F, Fraziano M. The multirole of liposomes in therapy and prevention of infectious diseases. Front. Immunol. 9, 155 (2018).
    • 93. Doxorubicin Liposomal. (2022). www.drugs.com/pro/doxil.html
    • 94. Daunorubicin Liposomal. (2022). www.drug.com/pro/daunoxome.html
    • 95. Amphotericin B Liposomal. (2022). www.drug.com/pro/ambisome.html
    • 96. Slingerland M, Guchelaar HJ, Rosing H et al. Bioequivalence of liposome-entrapped paclitaxel easy-to-use (LEP-ETU) formulation and paclitaxel in polyethoxylated castor oil: a randomized, two-period crossover study in patients with advanced cancer. Clin. Ther. 35(12), 1946–1954 (2013).
    • 97. Patisiran. (2022). www.drug.com/pro/onpattro.html
    • 98. Akinc A, Maier MA, Manoharan M et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat. Nanotechnol. 14(12), 1084–1087 (2019).
    • 99. Vincristine Liposomal. (2022). www.drug.com/pro/mariqibo.html
    • 100. Irinotecan Liposomal. (2022). www.drug.com/pro/onivyde.html
    • 101. Dicko A, Kwak S, Frazier AA, Mayer LD, Liboiron BD. Biophysical characterization of a liposomal formulation of cytarabine and daunorubicin. Int. J. Pharm. 391, 248–259 (2010).
    • 102. Daunorubicin Liposomal/Cytarabine Liposomal. Merck Manuals, NJ, USA (2022). www.merckmanuals.com/professional/SearchResults?query=vyxeos
    • 103. Sriram A, Kalanxhi E, Kapoor G et al. State of the World's Antibiotics in 2021: A Global Analysis of Antimicrobial Resistance and Its Drivers Center for Disease Dynamics, Economics & Policy (CDDEP), Inc., Washington DC, USA (2021).
    • 104. Conceição N, da Silva LEP, da Costa Darini AL, Pitondo-Silva A, de Oliveira AG. Penicillin-resistant, ampicillin-susceptible Enterococcus faecalis of hospital origin: pbp4 gene polymorphism and genetic diversity. Infect. Genet. Evol. 28, 289–295 (2014).
    • 105. Hamed MI. Detection of methicillin-resistant and biofilm-producing Staphylococcus aureus in bovine mastitis. J. Adv. Vet. Res. 8(4), 95–100 (2018).
    • 106. Kim J, Jo A, Chukeatirote E, Ahn J. Assessment of antibiotic resistance in Klebsiella pneumoniae exposed to sequential in vitro antibiotic treatments. Ann. Clin. Microbiol. Antimicrob. 15, 60 (2016).
    • 107. Pourabbas B, Firouzi R, Pouladfar G. Characterization of carbapenem-resistant Acinetobacter calcoaceticus–baumannii complex isolates from nosocomial bloodstream infections in southern Iran. J. Med. Microbiol. 65(3), 235–239 (2016).
    • 108. Sader HS, Huband MD, Castanheira M, Flamm RK. Pseudomonas aeruginosa antimicrobial susceptibility results from four years (2012 to 2015) of the International Network for Optimal Resistance Monitoring Program in the United States. Antimicrob. Agents Chemother. 61(3), e02252–16 (2017).
    • 109. Landman D, Salamera J, Quale J. Irreproducible and uninterpretable polymyxin B MICs for Enterobacter cloacae and Enterobacter aerogenes. J. Clin. Microbiol. 51(12), 4106–4111 (2013).
    • 110. Wang D, Sun Y, Liu Y et al. Expert opinion on drug delivery clinical translation of immunoliposomes for cancer therapy: recent perspectives. Expert Opin. Drug Deliv. 15(9), 893–903 (2018).
    • 111. Belfiore L, Saunders DN, Ranson M, Thurecht KJ, Storm G, Vine KL. Towards clinical translation of ligand-functionalized liposomes in targeted cancer therapy: challenges and opportunities. J. Control. Rel. 277, 1–13 (2018).
    • 112. Xu Y, Fourniols T, Labrak Y, Beloqui A, Rieux A. Surface modification of lipid-based nanoparticles. ACS Nano 16, 7168–7196 (2022).
    • 113. Fulaz S, Vitale S, Quinn L, Casey E. Nanoparticle–biofilm interactions: the role of the EPS matrix. Trends Microbiol. 27(11), 915–926 (2019).
    • 114. Fadeel B, Alexiou C. Brave new world revisited: focus on nanomedicine. Biochem. Biophys. Res. Commun. 533(1), 36–49 (2020).