567
Views
76
CrossRef citations to date
0
Altmetric
Review

Recent advances in the treatment of pathogenic infections using antibiotics and nano-drug delivery vehicles

, &
Pages 327-343 | Published online: 18 Jan 2019

Abstract

The worldwide misuse of antibiotics and the subsequent rise of multidrug-resistant pathogenic bacteria have prompted a paradigm shift in the established view of antibiotic and bacterial–human relations. The clinical failures of conventional antibiotic therapies are associated with lengthy detection methods, poor penetration at infection sites, disruption of indigenous microflora and high potential for mutational resistance. One of the most promising strategies to improve the efficacy of antibiotics is to complex them with micro or nano delivery materials. Such materials/vehicles can shield antibiotics from enzyme deactivation, increasing the therapeutic effectiveness of the drug. Alternatively, drug-free nanomaterials that do not kill the pathogen but target virulent factors such as adhesins, toxins, or secretory systems can be used to minimize resistance and infection severity. The main objective of this review is to examine the potential of the aforementioned materials in the detection and treatment of antibiotic-resistant pathogenic organisms.

Introduction

Antibiotics are compounds that either stop bacteria from growing or kill them outright, depending on their ability to inhibit critical bacterial cellular processes. Despite the profound success achieved by the use of broad-spectrum antibiotics in the treatment of complex bacterial diseases, new and recurrent bacterial infections continue to impose significant challenges on global health care.Citation1Citation5 Conventional broad-spectrum therapies kill native as well as pathogenic bacteria disrupting the indigenous microflora increasing the risk of infection recurrence and systemic infiltration of the host.Citation6Citation8 Recent studies suggest a number of pathogens such as Mycobacterium tuberculosis, Burkholderia pseudomallei, Staphylococcus aureus, and Listeria monocytogenes resist free form antibiotics by persisting in macrophages and erythrocytes.Citation9,Citation10 Some of these persistors remain dormant and can go undetected by standard culture methods.Citation11 For the last fifty years identification of antibiotic resistant strains relied on selective culturing (blood cultures) and enzyme linked immune-sorbent assays (ELISA).Citation12 Selective culturing for the diagnosis of pathogens takes from 18 to 48 hours.Citation13,Citation14 Unfortunately, these techniques are not economically feasible in low-resource settings where resistance rates exceed 80%.Citation15,Citation16 In 2012, the WHO published a list of critically important anti-microbials for human medicine (). Taken together, these challenges highlight the need for alternative antimicrobial detection and treatment strategies.

Table 1 WHO listing (third revision, 2012)Citation16 of critically important antimicrobials for human medicine

In the last few decades, many studies have focused on developing rapid detection and efficient drug delivery systems to meet the challenge of antibiotic resistance. These delivery vehicles can be nano or micron in size and composed of liposomes, dendrimers, peptides, polymer, and inorganic materials.Citation17,Citation18 Many of them are compatible with and have shown to enhance the sensitivities of conventional laboratory and field diagnostic tests by several orders of magnitude. The engineering of these vehicles is now so advanced that single bacteria can be routinely detected by point-of-care (POC) devices.Citation19

In addition to detection, nano-vehicles are also used to treat bacterial infections. A drug or drugs can be loaded to the exterior or interior of the vehicle via chemical conjugation, adsorption, and encapsulation and then delivered to the infection site. Antibiotics delivered in this way shield the drug from the resistant mechanisms of the pathogen. Moreover, if the vehicle itself exhibits antimicrobial activity, the concentration of antibiotic can be reduced further. Other advantages of this combinatorial approach include longer drug retention times, improved pharmacokinetic profiles, and minimization of adverse effects to the host. These advances come at a time when the dissemination of metallo-β-lactamases among Gram-negative bacteria is growing dramatically and the resistance to carbapenem and β-lactam antibiotics exceeds 50% in many patient groups.Citation20 As a result, several of these antibacterial vehicles are undergoing preclinical and clinical testing, while numerous others have recently been approved in the treatment of bacterial diseasesCitation21Citation30 by The US Food and Drug Administration such as silver and titanium nanoparticles (NPs) for antibacterial skin lotions and sunscreens that are in commercial use.

In this review, five areas where biocompatible nanomaterials can be used to combat antibiotic-resistant pathogenic bacteria are discussed. These areas include 1) detection of antibiotic-resistant pathogens, 2) targeted antibiotic treatments, 3) combinatorial antibiotic delivery, 4) NP vaccines, and 5) antibiotic repotentiation. Advances made in these areas bring us ever closer to real-time antibacterial therapies that can respond to changes in bacterial susceptibility, colonization resistance, infiltration of the host organism, and pathogenic virulence. In this review, we highlight the innovative materials currently being developed to detect and treat antibiotic-resistant pathogens.Citation31Citation36

Pathogenic detection

The rapid identification of pathogenic bacteria in the early stages of therapy is crucial in preventing drug resistance. Among the myriad of molecular biology detection techniques available in modern day laboratories, those based on nucleic acid amplification technology such as whole genome sequencing, modern PCR, multiplex PCR (mPCR), quantitative PCR (qPCR), and droplet PCR are routinely used to distinguish resistant pathogenic strains. Real-time PCR (RT-PCR) or qPCR is capable of continuously monitoring changes in bacterial susceptibility.Citation37 However, RT-PCR is currently too expensive to be employed in resource-limited settings where cost-effective qualitative tests dominate the field.Citation38 In this regard, NPs can improve the sensitivity, accuracy, and reliability of qualitative tests, leading to better patient outcomes.

The first stage in identification involves sample preparation. This can take several days using gold standard techniques, which may be too long for critically ill patients who require administration of a specific antibiotic within 24 hours. Immunomagnetic separation is a rapid and highly facile extraction process that allows the researchers to concentrate microbial organisms from various clinical samples within hours.Citation39 In the last decade, magnetic nanoparticles (MNPs) particularly superparamagnetic nanoparticles (SPMNPs) composed of magnetite (Fe3O4) or maghemite (γ-Fe2O3) are attracting a lot of attention due to their excellent magnetic properties, low cost, assay versatility, and higher capture efficiency.Citation40 In addition to sample preparation, SPMNPs can be used directly or as part of multifunctional composite to improve the sensitivity (eg, Fe3O4-Ag, FeO4-Au, and FePt-Ag) of disposable optical and electrochemical immu-noassays. For example, in 2017, Kearns et al combined lectin-functionalized silver-coated MNPs with optically active antibody-coated silver NPs to isolate and detect three bacterial pathogens including methicillin-resistant S. aureus (MRSA) in an Eppendorf tube.Citation41 In 2018, Cowger et al developed a novel susceptibility test called protein-adsorbed NP-mediated matrix-assisted laser desorption–ionization mass spectrometry. The whole procedure took <50 minutes and was able to distinguish between wild-type and drug-resistant strains of bacteria.Citation42 In addition, Wei et al reported a portable multiplexed bar-chart SpinChip (MB-SpinChip) integrated with NP-mediated magnetic aptasensors for the visual quantitative instrument-free detection of multiple pathogen. Using this MB-SpinChip, three major foodborne pathogens including Salmonella enterica, Escherichia coli, and L. monocytogenes were specifically quantified in apple juice with limits of detection of about 10 CFU/mL. Although significantly more expensive to construct than paper-based devices, MB-SpinChip may provide essential “real time” input during a potential pathogenic outbreak.Citation43

Antibody-labeled SPMNPs are routinely used with standard and miniaturized MRI systems found in large hospitals and research laboratories. Diagnostic assays using only magnetic fields are simpler to use and better suited for the detection of bacteria in optically opaque media.Citation44,Citation45 In 2013, Chung et al used a magnetic-DNA probe in combination with a miniaturized nuclear magnetic resonance to detect the bacterial RNA of 13 species from patient specimens. The miniaturized micronuclear magnetic resonance system had a sample volume of 2 mL and capable of supporting rapid, high-throughput operations in POC settings.Citation46 More recently, Park et al used a chip magnetic capture, culture, and detection assay for on-site detection of antigen-presenting cells exposed to S. enterica. The magnetic capture, culture, and detection achieved high sensitivity, detecting Salmonella-specific antibodies secreted from as little as 50 hybridoma cells/mL of blood. The assay took a minimum of 19 hours, 5 hours less than conventional tests and could be performed with unskilled labor.Citation47 A schematic diagram demonstrating the capture and culturing stages of the assay is shown in .

Figure 1 Checkerboard magnetic chip for cell capture.

Notes: (A) Checkerboard magnetic array with alternating polarity. (B) Magnetic field strength (Bz) between two checkerboard arrays. (C) Magnetic force simulation for 1 µm magnetic beads exposed to a simple two-pole system or checkerboard array. (D) Chip-array arrangement. (E) The effect of flow rate on the enrichment ratio. (F) Magnetic enrichment of hybridoma cells. Cells were labeled with CD45-specific magnetic beads; IgG2b beads were used as a control. Image reproduced with permission from Park KS, Kim H, Kim S, et al. Nanomagnetic system for rapid diagnosis of acute infection. ACS Nano. 2017;11(11):11425–11432. Copyright 2017 American Chemical Society.Citation47
Figure 1 Checkerboard magnetic chip for cell capture.

Using noninvasive imaging such as MRI to locate bacteria during the early stages of infection has the potential to reduce bacterial resistance and minimize organ damage to the host. Conventional MRI-based imaging has also been used to visualize S. aureus infections in endocarditis,Citation48 osteomyelitis,Citation49 and soft tissue infection models.Citation50,Citation51 Generally, these models detect inflammation rather than the causative agent of an infection. Although direct visualization of bacteria has been achieved using iron particle-labeled S. aureus,Citation52 this technique is frequently limited by the lack of specificity. Ongoing work to create specific molecular imaging probes includes using iron NPs conjugated with IgG to detect intracellular pathogens, and particles coated with M. tuberculosis surface antibody for the specific detection of extrapulmonary mycobacterial infectionCitation53,Citation54 have been reported.

For the past 3 decades, the optical and electrochemical properties of noble metal NPs, particularly AuNPs, have been extensively utilized by many fields of academic and industrial science.Citation55,Citation56 AuNPs exhibit a bright red maximum in the visible region of the optical spectrum; the maxima’s bandwidth and intensity can be tuned by varying the shape, composition, and the distance between particles.Citation57 Aggregation of NPs induces interparticle surface plasmon coupling that results in a blue shift in the maxima. This colorimetric change has been utilized in numerous biological and NP immunoassays. It should be noted that the sensitivity for colorimetric sensors is low compared with fluorescence-based sensors, although the associated instrumentation is significantly cheaper. Moreover, many types of colorimetric sensors are designed to detect clinically relevant bacterial concentrations such as those found on human skin, although more sensitive methodologies are sometimes employed when the individual is suspected to be infected with a hypervirulent pathogen. For a comprehensive understanding of surface plasmon resonance in gold NPs, the review by Amendola et al is recommended.Citation58

Pathogenic bacteria were first detected using AuNP aggregation by Elghanian et al in 1997.Citation59 Since then, aggregation and dispersion of AuNPs have been widely explored for the detection of bacteria-specific DNAs, proteins, and live cells. In 2004, Storhoff et al used AuNPs to detect the mecA gene in MRSA genomic DNA samples.Citation60 The approach was effective in discriminating MRSA from methicillin-sensitive S. aureus strains. More recently, Chan et al also used AuNPs for direct colorimetric PCR detection of MRSA in clinical specimens, a total of 72 clinical samples were tested.Citation61 The performance was comparable with real-time PCR assays but at lower cost per reaction. Similar technological approaches have been applied in the detection of multidrug-resistant M. tuberculosis and hemorrhagic E. coli.Citation62 Of particular interest is the application of these technologies in low-resource settings. In 2017, a working concept of an integrated paper microwell platform and an M. tuberculosis detection scheme based on AuNPs was demonstrated.Citation63 Moreover, continued improvements in micropatterned paper devices combined with digitized fluidics have led to an increase in the availability of affordable, accurate POC tests. There are several up-to-date reviews regarding the detection of antibiotic-resistant strains of pathogenic bacteria using POC.Citation64,Citation65 Unfortunately, the potential of AuNPs is yet to be realized in the clinic, as many nanodiagnostic tests are stuck in preclinical trials with few US Food and Drug Administration tests currently available to the patient.

In addition to iron-based and gold NPs, quantum dots (QDs), upconverting nanoparticles (UCNPs), and graphene oxide (GO) have also been used to detect bacteria.Citation66,Citation67 Fluorescent QDs and UCNPs exhibit greater sensitivity and specificity than nonfluorescent nanomaterials as only the signal originating from the nanomaterial is detected. Graphene can be combined with QDs to work as a quencher (silencing the fluorescence signal of QDs when both are in contact).Citation68 This property was utilized by Morales-Narváez et al in the fabrication of lateral flow biosensors.Citation69 Their system consisted of two lines of QDs deposited on paper (test and control). The first line was capable of capturing some bacteria (by means of antibodies attached on QDs). After dispensing the sample solution on the QD lines, a solution of GO was added. In the absence of bacteria, GO quenched the fluorescence of the lines. In comparison with traditional lateral flow biosensors, this simple referencing method prevented the formation of false positives as a negative sample always turned off both lines. More recently, Jin et al utilized the dissociation of UCNPs and fluorescence recovery to detect E. coli in food and water samples, exhibiting a detection range of 5–106 CFU/mL and detection limit of 3 CFU/mL, respectively.Citation70 The novel UCNP-based fluorescence resonance energy transfer (FRET) aptasensor could be used to detect a broad range of targets from whole cells to metal ions.

Targeted antibiotic therapies

The microenvironment of a bacterial infection site is rich in physiological and molecular cues that can be utilized to activate drug release or facilitate the binding of an NP or vehicle to bacteria. Passive targeting (natural barriers), physical– chemical gradients (surface charge and pH), and active targeting are three ways in which targeted drug delivery can be achieved.Citation71,Citation72 Passive targeting is possible due to the dysfunctional lymphatic drainage and elevated permeability that occurs at the infected site. This phenomenon can be exploited by nano and micron particles for targeted antibiotic delivery. In fact, the accumulation of both uncoated and PEGlyated liposomes at soft tissue infected by S. aureus has been shown to correlate closely with particle size.Citation73,Citation74

However, most infection sites are polymicrobial in nature.Citation75 For example, pulmonary infections often consist of combinations of Gram-negative and Gram-positive anaerobic or aerobic bacteria, such as Klebsiella pneumoniae, E. coli, Pseudomonas aeruginosa, and S. aureus including MRSA.Citation75,Citation76 Many of these bacteria present a negative surface charge under physiological pH and can be targeted using common ligands such as cationic or lipopeptides.Citation77 Cationic antimicrobial peptides and lipopeptides kill the microbes by interacting and disrupting bacterial cell membranes. Consequently, a range of vehicles incorporating natural and synthetic peptides have been developed for broad-spectrum applications.Citation79 Moreover, lipopeptides show remarkable growth inhibition activity of both Gram-positive and Gram-negative bacteria and fungus. It should be noted that there is a potential for electrostatic interactions and subsequent binding of cationic peptides with other charge modifying substances in circulation. Studies have shown potentially toxic in vitro and in vivo effects with the use of cationic lipids and polymer, including but not limited to cell shrinking and vacuolization of human cell lines.Citation80 Recently, Mitra et al showed that synthetic lipopeptides can exhibit good biocompatibility to different mammalian cell lines like HepG2, HeLa, and SiHa as well.Citation81

Utilizing surface negative charge is one way to target the infection site; however, some pathogens can negate this approach by secreting lactic and acetic acid into the microen-vironment.Citation82 To overcome this problem, several groups have developed surface charge switching polymer micelles and NPs that target the negatively charged cell wall of MRSA at low pH.Citation83,Citation84 Recently, a zwitterionic pH-sensitive polymer, poly (N′-citraconyl-2-(3-aminopropyl N, N dimethylam-monium) ethyl methacrylate) or P(CitAPDMAEMA), was used to inhibit the growth of E. coli and S. aureus under acidic conditions.Citation85 The polymer remained zwitterionic at physiological pH and exhibited low hemotoxicity and good biocompatibility. Surface charge switching micelles and NP have been synthesized containing pH-responsive poly-l-histidine,Citation84 beta-amino ester,Citation86 and numerous chemical functionalities.Citation87 In addition, polymers containing acid-degradable acetal/ketal functionalities have also shown promising results for pH-responsive drug delivery systems for anticancer drugs.Citation88 With this in mind, Kalhapure et al recently developed a pH-responsive solid lipid NP with an acetal linkage to deliver vancomycin and inhibit the growth of methicillin-sensitive S. aureus and MRSA. The in vivo study showed that the amount of MRSA remaining in the skin of encapsulated vancomycin-treated mice waŝ22-fold lower than those treated with the free form of the drug.Citation89

In addition to MRSA, many bacteria such as Helicobacter pylori, Listeria, Salmonella, Campylobacter rectus, E. coli, and Shigella flexneri are found in varying acidic conditions within the body (stomach [pH 1.0–2.0], vagina [pH 4.0–5.0], skin [pH 4.0–5.5], intestines [pH 5.0–8.0] and bladder [pH 4.5–8.0]).Citation84 The stomach mucosa is particularly vulnerable to the negative spiral bacterium H. pylori, which is etiologically associated with the development of gastritis, gastric ulcers, and associated carcinomas. The best results regarding the eradication of H. pylori have been achieved using a variety of antibiotic-loaded chitosan (CS) microparticles and NPs.Citation90Citation93 Despite these successes, the physical–chemical requirements that determine NP assembly on different nanostructured H. pylori surfaces are still not resolved.Citation94 Recent work by Westmeier et al attempted to address this issue by studying the behavior of a library of model NPs, varying in size, surface functionalization, shape, and material on H. pylori.Citation95 The study showed that NP–bacteria assembly did not require specific functionalization or negative surface charge and that assembly in interfering medium was significantly influenced by the impact of low pH on the bacterial surface. Moreover, assembly of nonbactericidal silica NPs (25% surface coverage) was shown to attenuate the pathogenesis of H. pylori by probable inhibition of the type IV secretion system ().

Figure 2 NP-coating attenuates Helicobacter pylori (H. pylori) pathobiology. Gastric cancer cells were infected with NP -H. pyloriGFP complexes estimated to maximally cover approximately 25% (Si30–25%: 1 × 108 bacteria, 600 µg mL−1 Si30; 10 min PBS) or 0.25% (Si30–0.25%: 1 × 108 bacteria, 60 µg mL−1 Si30, 10 min PBS) of the bacterial surface, respectively.

Notes: (A) Fluorescent microscope image demonstrating NP inhibition of the “hummingbird” phenotype. (B) Immunoblot analysis showing NP (Si30) attenuated type IV secretion system function and decreasing phosphorylated CagA levels. (C) β-Actin normalized phosphorylated CagA levels. (D) NP concentration-dependent decrease in IL-8 secretion. Westmeier D, Posselt G, Hahlbrock A, et al. Nanoparticle binding attenuates the pathobiology of gastric cancer-associated Helicobacter pylori. Nanoscale. 2018;10(3):1453–1463. Adapted by permission of The Royal Society of Chemistry.Citation95 Statistical significance was determined by using the Mann-Whitney test or paired t-test assuming significance at *P=0.05.
Abbreviations: Ctrl, control; NP, nanoparticle.
Figure 2 NP-coating attenuates Helicobacter pylori (H. pylori) pathobiology. Gastric cancer cells were infected with NP -H. pyloriGFP complexes estimated to maximally cover approximately 25% (Si30–25%: 1 × 108 bacteria, 600 µg mL−1 Si30; 10 min PBS) or 0.25% (Si30–0.25%: 1 × 108 bacteria, 60 µg mL−1 Si30, 10 min PBS) of the bacterial surface, respectively.

NPs have shown promise in treating stomach bacterial infections, but a significant challenge has been to develop antibacterial NPs that are suitable for the various pH gradients found in the gastrointestinal tract. One such material is the triblock copolymer poly(ethylene glycol)-block-poly (ami-nolated glycidyl methacrylate)-block-poly(2-(diisopropyl amino) ethyl methacrylate) (PEG-b-PAGA-b-PDPA), which was recently used as a pH-responsive micelle to target metastatic breast cancer.Citation96

In addition to negative charge and pH gradients, bacteria secrete many virulence factors including toxins, fibronectin proteins, collagen adhesins, and various enzymes (phosphatase, phospholipase, and lipase).Citation97 Thus, a simple way to target the infection site is to dope the surface of the antibiotic delivery vehicle with a complementary substrate or antibody. As the enzyme metabolizes the substrate, it perforates the vehicle releasing the antibiotic near the infection site. In the last decade, various lipase or phosphatase-sensitive polymeric triple-layered nanogels have been used for the synchronized delivery of antibiotics at infection sites.Citation98,Citation99 Lipases can also be used to shield AMPs from nonspecific interactions with the host. For example, the lipase substrate poly(caprolactone-b-ethylene glycol) (PCL-b-PEG) was recently used to mask the dendritic polycation G-2. The hybrid dendrimer successfully killed >99.9% of inoculated bacterial cells at ≤8 µg/mL, exhibiting good colloidal stability in the presence of serum and insignificant hemolytic toxicity even at ≥2,048 µg/mL.Citation100 The design of these hybrid vehicles is now so refined that it is possible to treat a specific pathogenic strain. For example, Yang et al recently reported a unique intracellular antibiotic delivery NP, in which mesoporous silica NPs loaded with gentamicin were coated with (bacterial toxin)-responsive lipid bilayer surface shell and bacteria-targeting peptide ubiquicidin (UBI29-41). The inhibition of S. aureus growth in both in vitro and in vivo of planktonic and intracellular infection, as well as a downregulation of inflammation-related gene expression in infected preosteoblasts or macrophages was observed.Citation101

If the obligate or facultative bacterium escapes the infection site, the risk of infection recurrence increases. It is now accepted that bacteria escape by hiding (at least survive) in osteoblasts, erythrocytes, and phagocytic cells.Citation9 These pathogens disrupt the microbicidal mechanisms of phagocytic cells by inhibiting the fusion of lysosomes with phagosomes prolonging their survival.Citation102 Recent studies suggest that several pathogenic bacteria such as M. tuberculosis, B. pseudomallei, S. aureus, and L. monocytogenes are capable of this.Citation10 Moreover, pathogens often use the cytoplasm of mammalian cells to exchange virulent and resistance genes.Citation103 One of the key mechanisms by which microbe’s requestor macrophages is by subverting the body’s most prolific nuclear receptors, the vitamin D receptor.Citation104 The impact of this subversion on melatonin homeostasis and the subsequent increase in intracellular infection are well known.Citation105 Unfortunately, the development of vitamin D receptor agonists such as olmesartan medoxomil has yet to be fully realized.Citation106

Once a macrophage becomes infected, the next step is to target the intracellular pathogen. As set out by Xiong et al,Citation20 the optimal activity of a given antibiotic against any bacterial-infected cell depends on the concentration of the active drug within the subcellular compartments, which is dependent on the drugs cellular retention, subcellular distribution, and activity. One way to achieve this is to deliver the antibiotic to the cell or infection site via a biodegradable vehicle. Over the past 2 decades, several biodegradable delivery systems composed of natural or synthetic polymers such as poly(lactide-co-glycolide) (PLGA),Citation18,Citation107,Citation108 poly(lactide) (PLA),Citation109 albumin, and CSCitation78 have demonstrated the potential of this approach. The major sites of intracellular infection are the mononuclear phagocytic system tissues found in the liver and spleen. This natural barrier brings together infected cells and antibiotic delivery vehicles amplifying the therapeutic effect of the antibiotic. This amplification is often reported when the mononuclear phagocytic system is the primary foci of infection in an experimental model. For example, ampicillin-loaded polyisohexylcyanoacrylate NPs demonstrated a therapeutic efficiency >2 orders better than the free form of the antibiotic in vivo.Citation110 Similar results have been obtained using gentamicin-loaded nanoplexes based on block copolymers of poly(ethylene oxide-b-sodium acrylate) and poly(ethylene oxide-b-sodium methacrylate).Citation111

Outside the liver and spleen, bacterial-infected macrophages are also found in the pulmonary cavities, eliminating these bacteria (active or dormant) remains one of the toughest challenges that developmental drug delivery systems currently face.Citation112 For example, current treatment of pulmonary tuberculosis (TB) infection involves prolonged oral administration of high systemic doses of combined frontline antibiotics like rifampicin (RFP), isoniazid, pyrazinamide, and ethambutol over a 6- to 8-month period.Citation113 M. tuberculosis can survive in alveolar macrophages by inhibiting phagosome maturation and phagosome–lysosome fusion. Delivery of RFP via PLGA microspheres to alveolar macrophages resulted in a 19-fold higher concentration of RFP in cells infected with TB compared with the administration of the free drug in solution.Citation114 Uptake of the antibiotic by the infected alveolar macrophages was dependent on the size and concentration of vehicles used and the population of macrophages encountered at the infection site. Using PLGA microspheres to extend the release of the drug to the primary site of infection is a promising treatment for TB as it can achieve the desired therapeutic effect in a shorter period.Citation115 Moreover, treatment time can be further reduced if the RFP-loaded PLGA microparticles are administered via intratracheal insufflation. When TB-infected guinea pigs were treated using the said procedure, their lungs showed significantly increased drug levels compared with intravenous and oral delivery of RFP.Citation116

The rapid development of targeted therapies for TB has benefited from the unique surfaces of mycobacteria, making the targeting of the infection site easier. However, differentiating between two Gram species is more difficult.Citation117 Yet, there are many common ligands (antibiotics included) that can be used to specifically target Gram-positive or Gram-negative pathogens.Citation118 In the last 2 decades, various antibiotics such as vancomycin and amoxicillin have been conjugated to the surface of gold NPs,Citation119,Citation120 iron oxide NPs,Citation121,Citation122 porous silica NPs,Citation123 and surface dendrimers, resulting in the preferential binding of Gram-positive bacteria. Moreover, other small molecules such as lectins, single domain antibodies, and aptamersCitation124 can be conjugated to the surface of the delivery vehicle to target Gram-negative pathogens. For example, mannose-specific or fucose-specific lectins showed enhanced binding affinity to the carbohydrate receptors on H. pylori, while bacterium-based aptamer selection techniques have been utilized to target NPs to Salmonella typhimurium. In addition to these techniques, other groups have utilized cell–pathogen adhesion mechanisms to target opportunistic pathogens.Citation125 Recently, Angsantikul et al utilized clarithromycin (CLR) loaded PLGA NPs (100 nm) coated with gastric epithelial cell membranes AGS-NP (CLR) to treat H. pylori infection in mice. After 6 days, the bacterial burden in a gram of stomach mice tissue treated with AGS-NP (CLR) was ~3.08 orders of magnitude lower than control ().Citation126

Figure 3 In vivo anti-Helicobacter pylori therapeutic efficacy of AGS-NP (CLR).

Notes: (A) H. pylori infection mouse model subject to inoculation, development, and treatment protocols. (B) Analysis of bacterial loads in mice treated with PBS, free CLR, PEG-NP (CLR), or AGS-NP (CLR) (n=6 per group). Bars represent median values. *P<0.05, **P<0.01, and ***P<0.001. Image reproduced from Coating nanopar-ticles with gastric epithelial cell membrane for targeted antibiotic delivery against Helicobacter pylori infection. Advanced Therapeutics. 2018;1(2):1800016. Copyright Wiley-VCH Verlag GmbH & Co. KGaA. Reproduced with permission.Citation126
Abbreviations: CFU, colony-forming unit; CLR, clarithromycin; NP, nanoparticle.
Figure 3 In vivo anti-Helicobacter pylori therapeutic efficacy of AGS-NP (CLR).

Another pathogen that resides within human macrophages is MRSA. Studies examining infected alveolar macrophages found that PEGylated and non-PEGylated liposomes loaded with vancomycin were capable of significant improvements in MRSA clearance.Citation127 Other types of delivery vehicles such as solid lipid NPs and PLGA loaded with vancomycin have shown similar improvements in MRSA clearance rates as well.Citation128 More recently, Pei et al combined the pH-sensitive properties of CS with PEGylated-PLGA to form a hybrid vehicle namely PpZEV. PpZEV NPs exhibited increased vancomycin release at acidic pH and significantly higher levels of uptake and MRSA clearance in infected macrophages when compared with free vancomycin and unmodified PLGA-NPs.Citation129 Macrophage uptake of antimicrobials can be further enhanced by attaching various ligands including O-stearoyl amylopectin, phosphatidylcholine, maleylated bovine serum albumin, and mannose onto the NP surface.Citation130 A summary of the various types of nanovehicles used to improve the efficacy of antibiotics and the treatment of bacterial infections is shown in .

Table 2 Targeted antibacterial therapies

Increasing microbial susceptibility: combinatorial applications

The accumulated misuse of free-form antibiotics has led to the emergence of multidrug-resistant MDR strains. The accepted definition of MDR is coresistance to three or more classes of antimicrobial drugs.Citation131 A serious concern is Enterobacteriaceae (avian E. coli), which is known to carry the New Delhi metallo-beta-lactamase gene (NDM-1), as well as genes that confer resistance to copper sulfate and quaternary ammonium disinfectants.Citation132 At present, most antibiotic-resistant mechanisms do not apply to NPs whose mode of action is direct contact with the cell wall. Thus, combining antibiotics with nanomaterials should reduce bacterial resistance and improve the pharmacokinetic profile of the drug.

Liposomes composed of phospholipids and cholesterols have been used to deliver antibacterial drugs for several decades.Citation133 As a drug delivery system, liposomes offer several advantages. Using mice and guinea pig TB-infected models, Deol et al showed that RMP, INH, and PZA-encapsulated liposomes were significantly more effective in reducing the recovery times than their free drug counterparts.Citation134 Due to the physical and chemical instability of liposomes, many researchers have employed niosomes to maintain the physical–chemical properties of antibiotic combinations.Citation135 Niosomal delivery allows for the delivery of combinatorial antibiotics that are too toxic in their free form. For example, drug compounds such as bismuth derivatives and gallium desferrioxamine interfere with iron absorption and reduction of alginate, LPS, and the secretion of adhesion factors of P. aeruginosa.Citation136 In a recent study by Mahdiun et al, the combinatorial effect of tobramycin and bismuth ethanedithiol-loaded niosomes (Nio-TOB) on the quorum sensing and biofilm production of P. aeruginosa was demonstrated. The minimum inhibitory concentration (MIC) of TOB and Nio-TOB for most strains was 16.64 and 1.3 mg/mL, respectively.Citation137 P. aeruginosa uses the same uptake mechanisms for both gallium and iron.Citation138 Thus, targeting bacterial iron metabolism can arrest P. aeruginosa virulence. Simple gallium siderophore complexes such as gallium citrate have shown good antibacterial activity (1–5 mg/mL MIC) against P. aeruginosa and low drug resistance in vivo. In addition, gallium protoporphyrin IX has shown 100- to 1,000-fold increase in potency against various MRSA strains compared with gallium nitrate.Citation139 However, relatively low aqueous solubility, moderate toxicity, and low synthetic yields of Ga-protoporphyrin IX may have dampened enthusiasm for further development. Antibiotics can also be conjugated with bacterial siderophores increasing their membrane permeability, which in turn increases their antimicrobial activity. In 2017, Oh et al reported on a novel siderophore-conjugated cephalosporin, LCB10-0200. In accordance with Clinical and Laboratory Standards Institute methods, LCB10-0200 showed potent antibacterial activity against P. aeruginosa clinical isolates, including β-lactamase producing strains. Moreover, LCB10-0200 was more effective than ceftazidime in treating systemic, respiratory tract, urinary tract, and thigh infections caused by antibiotic susceptible and resistant strains of P. aeruginosa in mouse models.Citation140

Until recently, first-line therapies for H. pylori eradication consist of proton pump inhibitors or ranitidine bismuth citrate and CLR, plus amoxicillin or metronidazole.Citation141,Citation142 Proton pump inhibitors and amoxicillin are known to increase the risk of secondary infections such as Clostridium difficile, while combinations of metronidazole are frequently used to treat C. difficile disease. Moreover, CLR resistance is now at a level where continued use of mainstream CLR-based triple therapy is not recommended.Citation143,Citation144 The clinical lifetime of CLR may have been prolonged if it had been encapsulated into a mucoadhesive particle. As early as 2008, several groups had tested various encapsulated triple combinations using NPs composed of CS, lectin-conjugated gliadin, and PGLA. Ramteke et al used lectin-conjugated NPs to simultaneously release amoxicillin, omeprazole, and CLR, resulting in a 94.83% eradication rate of H. pylori from infected rats.Citation145

As >40% of the global population harbor H. pylori, a new treatment strategy is urgently needed.Citation146 Fortunately, other types of eradication therapies such as liposomal linolenic acid (LipoLLA) have shown great potential in eradicating H. pylori. Unlike amoxicillin, LipoLLA kills both spiral and coccoid forms of the pathogen. However, a few studies suggested that certain carriers may cause adverse health effects by directly eliminating the gastrointestinal micro-biota or by alternating their functions. For example, silver NPs could damage the gut microbiota even though silver is generally considered to have low toxicity. In a recent paper by Li et al, the gut microbiota of H. pylori-infected mice exhibited minor alterations in bacteria populations after LipoLLA therapy with a notable boost in the relative abundance of Proteobacteria and Firmicutes along with a decrease in Verrucomicrobia and Bacteroidetes.Citation147 On the contrary, conventional triple therapy led to the growth of Enterobacteriaceae, Enterococcaceae, and Clostridiaceae species in the mice.

It is becoming increasingly obvious that the composition of the gut microbiome must be taken into consideration during the development and administration of future antibacterial treatments. Sparing the gut microbiome using narrow-spectrum antibiotics (fidaxomicin) has been shown to correlate with a reduction in C. difficile recurrence compared with broad-spectrum antibiotics.Citation148 However, TB resistance is increasing although narrow spectrum drugs are the mainstay of current treatments. Moreover the oral-pulmonary micro-biota is too small to act as a significant reservoir of antibiotic resistance, which suggests the administration of free form RFP, its rapid delocalization, and resulting gut metabolites have contributed significantly to an increase in its resistance.Citation149

Vaccination vehicles (toxins and antigens)

As well as killing bacteria, the large surface area of NPs can be used to capture bacterial toxins. Many pore-forming toxins (PFTs) such as listeriolysin O (LLO) L. monocytogenes and Aerolysin from Aeromonas hydrophila promote deacetylation and dephosphorylation of histones, resulting in subsets of immune genes, such as CXCL2, MKP2, or IFIT3, becoming repressed thus predisposing the host to secondary infections. A radical but simple solution to accelerate the removal of active toxins from the host is to use erythrocyte membrane-coated polymer NPs or nanosponges.Citation150 The membrane of red blood cells (RBCs) serves as perfect stealth coating with high biocompatibility and immune evasion ability.Citation151Citation153 Using the extrusion method, various nanomaterials such as PGLA, melatonin, inorganic NPs, gold nanocages, and QDs have been coated with RBC membranes. In all cases, the in vivo circulatory half-life of RBC-coated nanomaterials was superior to materials coated with PEG. In 2017, Zhang et al prepared RBC membrane-coated vancomycin nanogels using the cell membrane-templated polymerization method for the treatment of MRSA infections.Citation154 The redox-responsive hydrogel acted as the delivery vehicle for vancomycin, while the RBC membrane absorbed and neutralized the pore-forming toxin secreted by the bacteria. Compared with free antibiotics and nonresponsive nanogels, RBC-coated nanogels exhibited remarkable antibacterial activity in vitro. Recently, Chen et al developed a broad-spectrum nanosponge by coating 100 nm PLGA NPs with human RBC membranes.Citation155 When tested with PFTs, melittin, αhemolysin, LLO (L. monocytogenes), and streptolysin O (group A Streptococcus), the hRBC nanosponges completely inhibited toxin-induced hemolysis in a concentration-dependent manner. The inhibitory human nanosponge concentration (IC100) value for the four PFTs tested was 7.5, 16, 0.63, and 1.0 µg/mL, respectively. The investigation showed that nanosponge sequestered toxins were less toxic not only to cells but also to live animals. shows a schematic structure of human nanosponge and its mechanism of neutralizing PFTs. In addition to α-hemolysin, MRSA also secretes other membrane-damaging toxins, including gamma-toxin, leucocidins, and phantom–valentine leucocidin. Such toxins can also be impregnated onto the surface of RBC-coated NPs. In 2017, Wei et al demonstrated a multitoxin laden nanotoxoid formulation using the aforementioned cell membrane-cloaked NPs.Citation156 The formulation was nonhemolytic and showed no observable toxicity upon subcutaneous administration in mice. Immunization with the nanoformulation induced germinal center formation in the lymph node and increased antibody titers against three staphylococcal virulence factors. The vaccine was shown to be effective in reducing skin lesion formation and bacteria count in mouse model.

Figure 4 Structure of hNS.

Notes: (A) Schematic diagram depicting hNS and its mechanism of neutralizing PFTs. (B) Nanoparticle hydrodynamic size (diameter) and zeta potential (mV) (n=3) before and after membrane coating. (C) Transmission electron microscopy image of hNS. (D) Diameter stability of hNS over 5 days in different media (n=3). Image modified with permission from Chen Y, Chen M, Zhang Y, et al. Broad-spectrum neutralization of pore-forming toxins with human erythrocyte membrane-coated nanosponges. Adv Healthc Mater. 2018;7(13):e1701366. © 2018 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.Citation155
Abbreviations: hNS, human RBC nanosponge; PFTs, pore-forming toxins; PLGA, poly (lactide-co-glycolide); RBC, red blood cell.
Figure 4 Structure of hNS.

The unique capabilities of RBC membrane-coated NPs to detain toxins and induce an immunological response are well known.Citation157Citation160 However, Gram-specific and mycobacterial outer membrane vehicles (OMVs) 50–250 nm can also be coated onto NPs. Given their small size, proinflammatory pathogen-associated molecule patterns, suitability for antigen-presenting cells uptake, and efficient lymph node drainage, OMVs are well suited to bacterial vaccine development. By marrying the virtues of synthetic NPs and OMVs, E. coli OMV-coated 30 nm gold NPs have been shown to induce fast dendritic cell activation and maturation in the lymph nodes of mice. The hybrid vehicles generated higher antibody titers with stronger avidity than those elicited by OMVs alone.Citation161 In addition, the vaccinated mice showed elevated levels of IL-17 and interferon-gamma. The adjuvant activity of gold NPs has been used extensively in the development of antibacterial vaccines including L. monocytogenes, Yersinia pestis, P. aeruginosa, and Streptococcus pneumoniae.Citation31 Multishell gold NPs have the potential to deliver bacterial antigens either through encapsulation or surface association. However, such investigations are usually conducted using cheaper biodegradable polymer NPs, such as PLGA NPs. For example, PLGA NP-encapsulated ovalbumin induced a higher and sustained antibody response than liposomes of similar size. It was also noted that the PLGA vaccine elicited a protective immunity against listeria infection with the highest bacterial clearance compared with liposomes. Antigen-coated polymer NPs have been shown to induce a strong cytotoxic CD8+ T cell response, whereas antigen encapsulation preferentially activates CD4+ T cells. However, several studies have shown that particle-encapsulated antigens induce CD8+ T cell as well, clearly further investigations on cellular processing of antigens is needed.Citation162,Citation163

To obtain protective immunity, adjuvants that augment the immunostimulating activity are required. Adjuvants can be formulated in the same way as antigens by encapsulation, incorporation, or conjugation with a polymer NP. Akin to bacteria, adjuvants that activate pattern recognition receptors (PRRs) can be added to the surface of antigen-loaded NPs. There are three major families of PRRs: Toll-like receptors (TLRs), retinoic acid-inducible gene I-like receptors, and nucleotide-binding oligomerization domain-like receptors. The cooperation of multiple PRRs has been shown to contribute to host defense against bacteria. Critical to the control of S. typhimurium is the synergy between TLR2 and TLR4, whereas the cooperation between active TLR2 and TLR9 plays an important role in controlling TB infection.Citation164,Citation165

As previously highlighted, antibiotic administration via pulmonary insufflation has proved very effective in the treatment of TB. Insufflation was employed by Lu et al to deliver Ag85B antigen and trehalose-6-6-dibehenate-loaded PLGA NPs to TB-infected guinea pigs, resulting in anti-TB protection, albeit inferior to BCG vaccination.Citation166 A single toxin/antigen vaccine is often inadequate against MRSA or TB infection consequently researchers frequently adopt a triantigen approach. For example, three mycobacterial antigens, MPT-83, MPT-64, and Ag85B, were loaded into PLGA NPs by Cai et al and administered via intramuscular injection to mice. After vaccination, elevated cellular and humoral responses against all three antigens were observed. In particular, a single dose of the triantigenic formulation was comparable with the BCG vaccine.Citation167

Antibiotic re-potentiation and gene editing

NP coatings (Ag, Au, ZnO, CuO, TiO2, etc) have been used to inhibit the growth of bacterial films on various surfaces (implants, dental materials, and wound dressings) for decades.Citation168 Toxicity issues surrounding implant failure are beyond the scope of this review as are the coselective properties of ZnO, CuO, and TiO2 in animal feeds.Citation13,Citation23 In 2017, several in-depth reviews discussed the antibacterial mechanisms of free form inorganic NPs.Citation23,Citation34 More recently, work by Kadiyala et al showed that ZnO nanoparticles (ZnO-NPs) kill MRSA via ROS-independent mechanisms involving multiple metabolic pathways.Citation169 Many reports suggest that these inorganic nanomaterials are toxic to specific cell lines and organs.Citation170Citation173 However, when used at very low concentrations with antibiotics to treat infections they are not.Citation174 A recent study by Panáček et al demonstrated a strong synergistic effect of antibiotics combined with AgNPs at very low concentrations demonstrating no cytotoxic effect on mammalian cells. Moreover, restoration of the susceptibility of a resistant E. coli strain to ampicillin was observed when ampicillin was combined with AgNPs. In addition, the reduced impact of fewer NPs on gut DNAases using the synergistic approach makes the uptake of resistant plasmids and transposons by live bacteria in humans unlikely.Citation4

Much remains to be learnt about the exogenous and endogenous factors affecting antibiotic resistance. Changes in cell physiology can provide insight into the bacterial mechanisms contributing to antibiotic tolerance. Sugar potentiation of β-lactam antibiotics is now well established with arabinose, glucose, mannitol, ribose, and lactose capable of increasing the sensitivity of infectious strains of E. coli to carbenicillin and cefuroxime by several orders of magnitude.Citation175,Citation176 In addition to primary metabolite potentiation, nontoxic (human host) diarylamidines such as pentamidine have displayed synergy with antibiotics used to treat Gram-positive bacteria, yielding effective combinations in vitro to colistin-resistant bacteria.Citation177,Citation178 In addition, repotentiation of secondary metabolites (antibiotics) using metabolizable sugars and a diarylamidine was recently used to kill E. coli O157:H7 via a ROS mechanism while repressing Shiga toxin production.Citation179 More recently, Milton et alCitation180 presented a new urea-containing class of 2-aminoimidazole-based adjuvants that potentiated colistin activity against colistin-sensitive Acinetobacter baumannii. Lead compounds enabled 1,000-fold reduction in the MIC of colistin in vitro. It could be argued that there are many naturally occurring antibacterial polymers (CS) that can repotentiate last line antibiotics just as well. For example, recent work by Jamil et al synergistically enhanced the bactericidal activity of β-lactam antibiotic cefotaxime with antimicrobial biopolymer CS, rendering it more effective against biofilm producing MDR pathogens.Citation181 shows the superior antibiofilm activity of cefotaxime-loaded CS NPs compared with CS NPs. Finally, interfering RNAi has been shown to repotentiate current antibiotics by targeting resistance genes. Recently, Meng et al used lipid nanocarrier to deliver antisense oligonucleotides targeted to the mecA gene, which is known to play a role in β-lactam resistance. Upon testing, they found it was possible to restore MRSA susceptibility to oxacillin in vitro and in vivo.Citation182

Figure 5 CFU assay.

Note: Image reproduced from Jamil et alCitation181 with permission (Copyright 2016 Frontier Publishing).
Abbreviations: CFU, colony-forming unit; CSNP, chitosan nanoparticles; E. coli, Escherichia coli; KP, Klebsiella pneumoniae; MRSA, methicillin-resistant Staphylococcus aureus; NAB, nano-antibiotic; Pseudo, Pseudomonas aeruginosa.
Figure 5 CFU assay.

In the event that these combinatorial approaches fail to arrest antibiotic resistance in animal models, the last line of defense is gene editing. Using a combination of clustered regularly interspaced, short palindromic repeats (CRISPR) and Cas9 protein, this accurate method of gene editing was recently applied as an anti-MRSA strategy in a study by Bikard et al.Citation183 Using a delivery system known as a phagemid, it was possible to deliver the gene editing machinery to the MRSA resulting in a 104-fold reduction in the number of viable colonies in vitro. However, challenges arise due to the wildly varying size and structure of different phagemids; moreover, delivery of the CRISPR–Cas9 package to intracellular microbes could mitigate off-target effects at both host cell and bacterial level. Current delivery strategies involve in capsulation of the phagemid with biological inert polymers, lipids, silica, and DNA target.Citation184,Citation185 To the best of our knowledge, there are no reports of RBC membranes being used to deliver CRISPR/Cas9 machinery to antibiotic-resistant pathogens.

Conclusion

In the last decade, advances in bionanoengineering have contributed to the development of highly sensitive bacterial detection technologies and sensors. Major efforts in refining the interior and surface chemistry of organic and inorganic drug carriers such as CS, PLGA, and gold have demonstrated excellent outcomes via the detection treatment and prevention of bacterial diseases, by specific targeting of pathogenic bacteria, combinatorial delivery of antibiotics, and effective antibacterial vaccination strategies.

It is expected that advances in microbiology, digital microfluidics, and bionanoengineering will continue to bring improvements regarding the rapid isolation and detection of infectious pathogenic bacterial strains, improvements in cost-effective drug delivery systems, as well as tailored effective therapies in developed countries.

Unfortunately, the advantages of improved biodistribution and therapeutic outcomes using targeted nanomaterials have yet to be realized in developing countries where the need to combat antibiotic resistance is felt the strongest. Thus, the application of these advances will require communication and collaboration between scientists and clinicians from many different countries and fields of research.

Acknowledgments

This research was supported by the Gachon University Research Fund GCU-2016-0186.

Disclosure

The authors report no conflicts of interest in this work.

References

  • D’CostaVMKingCEKalanLAntibiotic resistance is ancientNature2011477736545746121881561
  • MartinezJLGeneral principles of antibiotic resistance in bacteriaDrug Discov Today Technol201411333924847651
  • McFall-NgaiMHadfieldMGBoschTCAnimals in a bacterial world, a new imperative for the life sciencesProc Natl Acad Sci U S A201311093229323623391737
  • van SchaikWThe human gut resistomePhilos Trans R Soc Lond B Biol Sci201537016702014008725918444
  • DaviesJDaviesDOrigins and evolution of antibiotic resistanceMicrobiol Mol Biol Rev201074341743320805405
  • PotgieterMBesterJKellDBPretoriusEThe dormant blood microbiome in chronic, inflammatory diseasesFEMS Microbiol Rev201539456759125940667
  • NajarMSSaldanhaCLBandayKAApproach to urinary tract infectionsIndian J Nephrol200919412913920535247
  • KellDPotgieterMPretoriusEIndividuality, phenotypic differentiation, dormancy and “persistence” in culturable bacterial systems: commonalities shared by environmental, laboratory, and clinical microbiologyF1000Res2015417926629334
  • PintoDSão-JoséCSantosMAChambelLCharacterization of two resuscitation promoting factors of Listeria monocytogenesMicrobiology2013159Pt 71390140123676438
  • LunguBRickeSCJohnsonMGGrowth, survival, proliferation and pathogenesis of Listeria monocytogenes under low oxygen or anaerobic conditions: a reviewAnaerobe2009151–271718926916
  • AudolyGFenollarFLagierJCLepidiHRaoultDDeglycosylation of Tropheryma whipplei biofilm and discrepancies between diagnostic results during Whipple’s disease progressionSci Rep2016612388327025850
  • KimESKimHBKimGKorea INfectious Diseases (KIND) study groupClinical and epidemiological factors associated with methicillin resistance in community-onset invasive Staphylococcus aureus infections: prospective multicenter cross-sectional study in KoreaPLoS One2014912e11412725485895
  • BuchanBWLedeboerNAEmerging technologies for the clinical microbiology laboratoryClin Microbiol Rev201427478382225278575
  • CohenJVincentJLAdhikariNKSepsis: a roadmap for future researchLancet Infect Dis201515558161425932591
  • LafleurLKBishopJDHeinigerEKA rapid, instrument-free, sample-to-result nucleic acid amplification testLab Chip201616193777378727549897
  • DoyleMPLoneraganGHScottHMSingerRSAntimicrobial resistance: challenges and perspectivesCompr Rev Food Sci Food Saf2013122234248
  • WinnickaKWroblewskaMWieczorekPSachaPTTryniszewskaEAThe effect of PAMAM dendrimers on the antibacterial activity of antibiotics with different water solubilityMolecules20131878607861723881050
  • GaoWThamphiwatanaSAngsantikulPZhangLNanoparticle approaches against bacterial infectionsWiley Interdiscip Rev Nanomed Nanobiotechnol20146653254725044325
  • HulmeJRecent advances in the detection of methicillin resistant Staphylococcus aureus (MRSA)BioChip J201711289100
  • XiongMHBaoYYangXZZhuYHWangJDelivery of antibiotics with polymeric particlesAdv Drug Deliv Rev201478637624548540
  • CouvreurPNanoparticles in drug delivery: past, present and futureAdv Drug Deliv Rev2013651212322580334
  • AbedNCouvreurPNanocarriers for antibiotics: a promising solution to treat intracellular bacterial infectionsInt J Antimicrob Agents201443648549624721232
  • WangLHuCShaoLThe antimicrobial activity of nanoparticles: present situation and prospects for the futureInt J Nanomedicine2017121227124928243086
  • PelgriftRYFriedmanAJNanotechnology as a therapeutic tool to combat microbial resistanceAdv Drug Deliv Rev20136513–141803181523892192
  • RaiMIngleAPPanditRBroadening the spectrum of small-molecule antibacterials by metallic nanoparticles to overcome microbial resistanceInt J Pharm2017532113914828870767
  • Torres-SangiaoEHolbanAGestalMCAdvanced nanobiomaterials: vaccines, diagnosis and treatment of infectious diseasesMolecules2016217867
  • CarabineiroSACApplications of gold nanoparticles in nanomedicine: recent advances in vaccinesMolecules2017225E85728531163
  • RizviSMDHussainTAhmedABFGold nanoparticles: a plausible tool to combat neurological bacterial infections in humansBiomed Pharmacother201810771830075371
  • AssoliniJPConcatoVMGonçalvesMDNanomedicine advances in toxoplasmosis: diagnostic, treatment, and vaccine applicationsParasitol Res201711661603161528477099
  • HemegHANanomaterials for alternative antibacterial therapyInt J Nanomedicine2017128211822529184409
  • BaptistaPVMcCuskerMPCarvalhoANano-strategies to fight multidrug resistant bacteria – “A Battle of the Titans”Front Microbiol20189144130013539
  • YangKHanQChenBAntimicrobial hydrogels: promising materials for medical applicationInt J Nanomedicine2018132217226329695904
  • GaoWChenYZhangYZhangQZhangLNanoparticle-based local antimicrobial drug deliveryAdv Drug Deliv Rev2018127465728939377
  • Dos Santos RamosMADa SilvaPBSpósitoLNanotechnology-based drug delivery systems for control of microbial biofilms: a reviewInt J Nanomedicine2018131179121329520143
  • HibbittsAO’LearyCEmerging nanomedicine therapies to counter the rise of methicillin-resistant Staphylococcus aureusMaterials (Basel)2018112E32129473883
  • KhuranaCChudasamaBNanoantibiotics: strategic assets in the fight against drug-resistant superbugsInt J Nanomedicine2018133629593387
  • LiuWdasJMephamAHNemrCRSargentEHKelleySOA fully-integrated and automated testing device for PCR-free viral nucleic acid detection in whole bloodLab Chip201818131928193529881833
  • ChengDYuMFuFDual recognition strategy for specific and sensitive detection of bacteria using aptamer-coated magnetic beads and antibiotic-capped gold nanoclustersAnal Chem201688182082526641108
  • SungYJSukHJSungHYLiTPooHKimMGNovel antibody/gold nanoparticle/magnetic nanoparticle nanocomposites for immunomagnetic separation and rapid colorimetric detection of Staphylococcus aureus in milkBiosens Bioelectron20134343243923370174
  • KumarNKulkarniKBeheraLVermaVPreparation and characterization of maghemite nanoparticles from mild steel for magnetically guided drug therapyJ Mater Sci Mater Med201728811628681216
  • KearnsHGoodacreRJamiesonLEGrahamDFauldsKSERS detection of multiple antimicrobial-resistant pathogens using nanosensorsAnal Chem20178923126661267328985467
  • CowgerTAYangYRinkDEProtein-adsorbed magnetic-nanoparticle-mediated assay for rapid detection of bacterial antibiotic resistanceBioconjug Chem201728489089628192992
  • WeiXZhouWSanjaySTMultiplexed instrument-free bar-chart spinchip integrated with nanoparticle-mediated magnetic aptasensors for visual quantitative detection of multiple pathogensAnal Chem201890169888989630028601
  • GuptaAKNaregalkarRRVaidyaVDGuptaMRecent advances on surface engineering of magnetic iron oxide nanoparticles and their biomedical applicationsNanomedicine (Lond)200721233917716188
  • CorcheroJLVillaverdeABiomedical applications of distally controlled magnetic nanoparticlesTrends Biotechnol200927846847619564057
  • ChungHJCastroCMImHLeeHWeisslederRA magneto-DNA nanoparticle system for rapid detection and phenotyping of bacteriaNat Nanotechnol20138536937523644570
  • ParkKSKimHKimSNanomagnetic system for rapid diagnosis of acute infectionACS Nano20171111114251143229121461
  • DiekemaDJBeachMLPfallerMAJonesRNSENTRY Participants GroupAntimicrobial resistance in viridans group streptococci among patients with and without the diagnosis of cancer in the USA, Canada and Latin AmericaClin Microbiol Infect20017315215711318814
  • Bubeck WardenburgJWilliamsWAMissiakasDHost defenses against Staphylococcus aureus infection require recognition of bacterial lipoproteinsProc Natl Acad Sci U S A200610337138311383616954184
  • KaimAHWischerTO’ReillyTMR imaging with ultrasmall superparamagnetic iron oxide particles in experimental soft-tissue infections in ratsRadiology2002225380881412461265
  • LiuJBaiRLiYMRI detection of bacterial brain abscesses and monitoring of antibiotic treatment using bacCESTMagn Reson Med201880266267129577382
  • HoerrVTuchscherrLHüveJBacteria tracking by in vivo magnetic resonance imagingBMC Biol20131116323714179
  • MoroLTuremisMMariniBIppodrinoRGiardiMTBetter together: strategies based on magnetic particles and quantum dots for improved biosensingBiotechnol Adv2017351516327923765
  • VeigasBFernandesARBaptistaPVAuNPs for identification of molecular signatures of resistanceFront Microbiol2014545525221547
  • LawJWAb MutalibNSChanKGLeeLHRapid methods for the detection of foodborne bacterial pathogens: principles, applications, advantages and limitationsFront Microbiol2014577025628612
  • KumarNHuYSinghSMizaikoffBEmerging biosensor platforms for the assessment of water-borne pathogensAnalyst2018143235937329271425
  • MocanTMateaCTPopTDevelopment of nanoparticle-based optical sensors for pathogenic bacterial detectionJ Nanobiotechnology20171512528359284
  • AmendolaVPilotRFrasconiMMaragòOMIatìMASurface plasmon resonance in gold nanoparticles: a reviewJ Phys Condens Matter2017292020300228426435
  • ElghanianRStorhoffJJMucicRCLetsingerRLMirkinCASelective colorimetric detection of polynucleotides based on the distance-dependent optical properties of gold nanoparticlesScience19972775329107810819262471
  • StorhoffJJMarlaSSBaoPGold nanoparticle-based detection of genomic DNA targets on microarrays using a novel optical detection systemBiosens Bioelectron200419887588315128107
  • ChanWSTangBSBoostMVChowCLeungPHDetection of methicillin-resistant Staphylococcus aureus using a gold nanoparticle-based colourimetric polymerase chain reaction assayBiosens Bioelectron20145310511124125759
  • WuS-YHulmeJAnSSARecent trends in the detection of pathogenic Escherichia coli O157:H7BioChip J201593173181
  • TsaiTTHuangCYChenCADiagnosis of tuberculosis using colorimetric gold nanoparticles on a paper-based analytical deviceACS Sens2017291345135428901134
  • CheonSAChoHHKimJLeeJKimHJParkTJRecent tuberculosis diagnosis toward the end TB strategyJ Microbiol Methods2016123516126853124
  • TangLLiJPlasmon-based colorimetric nanosensors for ultrasensitive molecular diagnosticsACS Sens20172785787528750528
  • FoubertABeloglazovaNVRajkovicABioconjugation of quantum dots: review and impact on future applicationTrAC Trends Anal Chem2016833148
  • GoldmanERClappARAndersonGPMultiplexed toxin analysis using four colors of quantum dot fluororeagentsAnal Chem200476368468814750863
  • BanerjeeRJaiswalARecent advances in nanoparticle-based lateral flow immunoassay as a point-of-care diagnostic tool for infectious agents and diseasesAnalyst201814391970199629645058
  • Morales-NarváezENaghdiTZorEMerkoçiAPhotoluminescent lateral-flow immunoassay revealed by graphene oxide: highly sensitive paper-based pathogen detectionAnal Chem201587168573857726205473
  • JinBWangSLinMUpconversion nanoparticles based FRET aptasensor for rapid and ultrasenstive bacteria detectionBiosens Bio-electron201790525533
  • DateAAHanesJEnsignLMNanoparticles for oral delivery: design, evaluation and state-of-the-artJ Control Release201624050452627292178
  • BrooksBDBrooksAETherapeutic strategies to combat antibiotic resistanceAdv Drug Deliv Rev201478142725450262
  • LavermanPDamsETStormGMicroscopic localization of PEG-liposomes in a rat model of focal infectionJ Control Release200175334735511489321
  • TranNHocquetMEonBNon-lamellar lyotropic liquid crystalline nanoparticles enhance the antibacterial effects of rifampicin against Staphylococcus aureusJ Colloid Interface Sci201851910711829486430
  • KalekoMBristolJAHubertSDevelopment of SYN-004, an oral beta-lactamase treatment to protect the gut microbiome from antibiotic-mediated damage and prevent Clostridium difficile infectionAnaerobe201641586727262694
  • DicksonRPErb-DownwardJRHuffnagleGBThe role of the bacterial microbiome in lung diseaseExpert Rev Respir Med20137324525723734647
  • SpicerCDJumeauxCGuptaBStevensMMPeptide and protein nanoparticle conjugates: versatile platforms for biomedical applicationsChem Soc Rev201847103574362029479622
  • ChenHLiMLiuZDesign of antibacterial peptide-like conjugated molecule with broad spectrum antimicrobial abilitySci China Chem2018611113117
  • ZhaoXPanFXuHMolecular self-assembly and applications of designer peptide amphiphilesChem Soc Rev20103993480349820498896
  • MitraRNShomeAPaulPDasPKAntimicrobial activity, biocompatibility and hydrogelation ability of dipeptide-based amphiphilesOrg Biomol Chem2009719410219081951
  • Radovic-MorenoAFLuTKPuscasuVAYoonCJLangerRFarokhzadOCSurface charge-switching polymeric nanoparticles for bacterial cell wall-targeted delivery of antibioticsACS Nano2012654279428722471841
  • CaoBXiaoFXingDHuXPolyprodrug antimicrobials: remarkable membrane damage and concurrent drug release to combat antibiotic resistance of methicillin-resistant Staphylococcus aureusSmall20181441e180200830118562
  • LiuPXuGPranantyoDXuLQNeohK-GKangE-TpH-sensitive zwitterionic polymer as an antimicrobial agent with effective bacterial targetingACS Biomater Sci Eng2018414046
  • MankociSKaiserRLSahaiNBartonHAJoyABactericidal peptidomimetic polyurethanes with remarkable selectivity against Escherichia coliACS Biomater Sci Eng201731025882597
  • JadhavMKalhapureRSRambharoseSNovel lipids with three C18-fatty acid chains and an amino acid head group for pH-responsive and sustained antibiotic deliveryChem Phys Lipids2018212122529305156
  • ChuLGaoHChengTA charge-adaptive nanosystem for prolonged and enhanced in vivo antibiotic deliveryChem Commun (Camb)201652376265626827077219
  • GilliesERFréchetJMpH-Responsive copolymer assemblies for controlled release of doxorubicinBioconjug Chem200516236136815769090
  • KalhapureRSSikwalDRRambharoseSEnhancing targeted antibiotic therapy via pH responsive solid lipid nanoparticles from an acid cleavable lipidNanomedicine20171362067207728434930
  • WangJByrneJDNapierMEDeSimoneJMMore effective nano-medicines through particle designSmall20117141919193121695781
  • HejaziRAmijiMStomach-specific anti-H. pylori therapy; part III: effect of chitosan microspheres crosslinking on the gastric residence and local tetracycline concentrations in fasted gerbilsInt J Pharm20042721–29910815019073
  • JingZWJiaYYWanNDesign and evaluation of novel pH-sensitive ureido-conjugated chitosan/TPP nanoparticles targeted to Helicobacter pyloriBiomaterials20168427628526851392
  • LuoMJiaYYJingZWConstruction and optimization of pH-sensitive nanoparticle delivery system containing PLGA and UCCs-2 for targeted treatment of Helicobacter pyloriColloids Surf B Biointerfaces2018164111929367052
  • KhutoryanskiyVVBeyond PEGylation: alternative surface-modification of nanoparticles with mucus-inert biomaterialsAdv Drug Deliv Rev201812414014928736302
  • HaydenSCZhaoGSahaKAggregation and interaction of cationic nanoparticles on bacterial surfacesJ Am Chem Soc2012134166920692322489570
  • WestmeierDPosseltGHahlbrockANanoparticle binding attenuates the pathobiology of gastric cancer-associated Helicobacter pyloriNanoscale20181031453146329303193
  • YuHGuoCFengBTriple-layered pH-responsive micelleplexes loaded with siRNA and cisplatin prodrug for NF-Kappa B targeted treatment of metastatic breast cancerTheranostics201661142726722370
  • RahmeLGStevensEJWolfortSFShaoJTompkinsRGAusubelFMCommon virulence factors for bacterial pathogenicity in plants and animalsScience19952685219189919027604262
  • SuYZhaoLMengFWangQYaoYLuoJSilver nanoparticles decorated lipase-sensitive polyurethane micelles for on-demand release of silver nanoparticlesColloids Surf B Biointerfaces201715223824428113126
  • ChenYLZhuSZhangLSmart conjugated polymer nanocarrier for healthy weight loss by negative feedback regulation of lipase activityNanoscale2016863368337526790821
  • XuLHeCHuiLBactericidal dendritic polycation cloaked with stealth material via lipase-sensitive intersegment acquires neutral surface charge without losing membrane-disruptive activityACS Appl Mater Interfaces2015750276022760726632646
  • YangSHanXYangYBacteria-targeting nanoparticles with microenvironment-responsive antibiotic release to eliminate intracellular Staphylococcus aureus and associated infectionACS Appl Mater Interfaces20181017142991431129633833
  • RezaeeRTalebrezaAZiariKBehnodVEmampourBFSDistribution of virulence factors and antimicrobial resistance properties of uropathogenic Escherichia coli isolated from diabetic and healthy males suffered from urinary tract infectionsBiosci Biotechnol Res Asia2016132931937
  • LimYMde GroofAJCBhattacharjeeMKFigurskiDHSchonEABacterial conjugation in the cytoplasm of mouse cellsInfect Immun200876115110511918765719
  • MarshallTGLeeREMarshallFECommon angiotensin receptor blockers may directly modulate the immune system via VDR, PPAR and CCR2bTheor Biol Med Model200631116403216
  • Pires-LapaMCarvalho-SousaCCeconEFernandesPMarkusRβ-adrenoceptors trigger melatonin synthesis in phagocytesInt J Mol Sci2018198E218230049944
  • ProalADAlbertPJMarshallTGBlaneyGPLindsethIAImmu-nostimulation in the treatment for chronic fatigue syndrome/myalgic encephalomyelitisImmunol Res2013562–339841223576059
  • AksungurPDemirbilekMDenkbaşEBVandervoortJLudwigAUnlüNDevelopment and characterization of Cyclosporine A loaded nanoparticles for ocular drug delivery: cellular toxicity, uptake, and kinetic studiesJ Control Release2011151328629421241752
  • MohammadiGNokhodchiABarzegar-JalaliMPhysicochemical and anti-bacterial performance characterization of clarithromycin nanoparticles as colloidal drug delivery systemColloids Surf B Biointerfaces2011881394421752610
  • GiannavolaCBucoloCMalteseAInfluence of preparation conditions on acyclovir-loaded poly-d,l-lactic acid nanospheres and effect of PEG coating on ocular drug bioavailabilityPharm Res200320458459012739765
  • CouvreurPFattalEAlphandaryHPuisieuxFAndremontAIntracellular targeting of antibiotics by means of biodegradable nanoparticlesJ Control Release1992191–3259267
  • RanjanAPothayeeNSeleemMDrug delivery using novel nanoplexes against a Salmonella mouse infection modelJ Nanopart Res2010123905914
  • SosnikACarcabosoAMGlisoniRJMorettonMAChiappettaDANew old challenges in tuberculosis: potentially effective nanotechnologies in drug deliveryAdv Drug Deliv Rev2010624–554755919914315
  • RawalTButaniSCombating tuberculosis infection: a forbidding challengeIndian J Pharm Sci201678181627168676
  • ChewNYChanHKUse of solid corrugated particles to enhance powder aerosol performancePharm Res200118111570157711758765
  • LawlorCKellyCO’LearySCellular targeting and trafficking of drug delivery systems for the prevention and treatment of MTbTuberculosis (Edinb)2011911939721237714
  • PandeyRSharmaAZahoorASharmaSKhullerGKPrasadBPoly (DL-lactide-co-glycolide) nanoparticle-based inhalable sustained drug delivery system for experimental tuberculosisJ Antimicrob Chemother200352698198614613962
  • FangGLiWShenXDifferential Pd-nanocrystal facets demonstrate distinct antibacterial activity against Gram-positive and Gram-negative bacteriaNat Commun20189112929317632
  • ChoiSKMycASilpeJEDendrimer-based multivalent vancomycin nanoplatform for targeting the drug-resistant bacterial surfaceACS Nano20137121422823259666
  • YuMWangHFuFDual-recognition Förster resonance energy transfer based platform for one-step sensitive detection of pathogenic bacteria using fluorescent vancomycin-gold nanoclusters and aptamer-gold nanoparticlesAnal Chem20178974085409028287715
  • SilveroCMJRoccaDMde La VillarmoisEASelective photo-induced antibacterial activity of amoxicillin-coated gold nanoparticles: from one-step synthesis to in vivo cytocompatibilityACS Omega2018311220123030023798
  • ZhuMLiuWLiuHConstruction of Fe3O4/Vancomycin/PEG magnetic nanocarrier for highly efficient pathogen enrichment and gene sensingACS Appl Mater Interfaces2015723128731288126005899
  • WangCZhangKZhouZVancomycin-modified Fe(3)O(4)@SiO(2)@Ag microflowers as effective antimicrobial agentsInt J Nanomedicine2017123077309428450783
  • CapelettiLBLoiolaLMDPiccoASda Silva LiberatoMCardosoMB8 – Silica nanoparticle applications in the biomedical fieldCiofaniGSmart Nanoparticles for BiomedicineElsevier2018115129
  • KimYParkEJNaDHRecent progress in dendrimer-based nano-medicine developmentArch Pharm Res201841657158229450862
  • WuSDuanNQiuYLiJWangZColorimetric aptasensor for the detection of Salmonella enterica serovar typhimurium using ZnFe2O4-reduced graphene oxide nanostructures as an effective peroxidase mimeticsInt J Food Microbiol2017261424828910678
  • AngsantikulPThamphiwatanaSZhangQCoating nanoparticles with gastric epithelial cell membrane for targeted antibiotic delivery against Helicobacter pylori infectionAdv Ther (Weinh)201812180001630320205
  • MuppidiKWangJBetageriGPumerantzASPEGylated liposome encapsulation increases the lung tissue concentration of vancomycinAntimicrob Agents Chemother201155104537454221788465
  • KalhapureRSSonawaneSJSikwalDRSolid lipid nanoparticles of clotrimazole silver complex: an efficient nano antibacterial against Staphylococcus aureus and MRSAColloids Surf B Biointerfaces201513665165826492156
  • PeiYMohamedMFSeleemMNYeoYParticle engineering for intracellular delivery of vancomycin to methicillin-resistant Staphylococcus aureus (MRSA)-infected macrophagesJ Control Release201726713314328797580
  • PuisneyCBaeza-SquibanABolandSMechanisms of uptake and translocation of nanomaterials in the lungAdv Exp Med Biol20181048213629453530
  • BerçotBPoirelLDortetLNordmannPIn vitro evaluation of antibiotic synergy for NDM-1-producing EnterobacteriaceaeJ Antimicrob Chemother201166102295229721807739
  • JohnsonTJSiekKEJohnsonSJNolanLKDNA sequence of a ColV plasmid and prevalence of selected plasmid-encoded virulence genes among avian Escherichia coli strainsJ Bacteriol2006188274575816385064
  • SercombeLVeeratiTMoheimaniFWuSYSoodAKHuaSAdvances and challenges of liposome assisted drug deliveryFront Pharmacol2015612728626648870
  • DeolPKhullerGKJoshiKTherapeutic efficacies of isoniazid and rifampin encapsulated in lung-specific stealth liposomes against Mycobacterium tuberculosis infection induced in miceAntimicrob Agents Chemother1997416121112149174172
  • RuckmaniKSankarVSivakumarMTissue distribution, pharmacokinetics and stability studies of zidovudine delivered by niosomes and proniosomesJ Biomed Nanotechnol201061435120499831
  • HuangCTStewartPSReduction of polysaccharide production in Pseudomonas aeruginosa biofilms by bismuth dimercaprol (BisBAL) treatmentJ Antimicrob Chemother199944560160510552975
  • MahdiunFMansouriSKhazaeliPMirzaeiRThe effect of tobramycin incorporated with bismuth-ethanedithiol loaded on niosomes on the quorum sensing and biofilm formation of Pseudomonas aeruginosaMicrob Pathog201710712913528323149
  • ChitambarCRGallium and its competing roles with iron in biological systemsBiochim Biophys Acta2016186382044205327150508
  • HammerNDSkaarEPMolecular mechanisms of Staphylococcus aureus iron acquisitionAnnu Rev Microbiol201165112914721639791
  • OhSHParkHSKimHSAntimicrobial activities of LCB10-0200, a novel siderophore cephalosporin, against the clinical isolates of Pseudomonas aeruginosa and other pathogensInt J Antimicrob Agents20175070070628668680
  • GoderskaKAgudo PenaSAlarconTHelicobacter pylori treatment: antibiotics or probioticsAppl Microbiol Biotechnol201810211729075827
  • TrifanAGirleanuICojocariuCPseudomembranous colitis associated with a triple therapy for Helicobacter pylori eradicationWorld J Gastroenterol201319427476747924259981
  • MiernykKMBulkowLRGoldBDPrevalence of Helicobacter pylori among Alaskans: factors associated with infection and comparison of urea breath test and anti-Helicobacter pylori IgG antibodiesHelicobacter2018233e1248229537130
  • WangYKKuoFCLiuCJDiagnosis of Helicobacter pylori infection: current options and developmentsWorld J Gastroenterol20152140112211123526523098
  • RamtekeSGaneshNBhattacharyaSJainNKTriple therapy-based targeted nanoparticles for the treatment of Helicobacter pyloriJ Drug Target200816969470518982518
  • CamargoMCGarcíaARiquelmeAThe problem of Helicobacter pylori resistance to antibiotics: a systematic review in Latin AmericaAm J Gastroenterol2014109448549524589670
  • LiXXShiSRongLFengMQZhongLThe impact of liposomal linolenic acid on gastrointestinal microbiota in miceInt J Nanomedicine2018131399140929563795
  • MullaneKFidaxomicin in Clostridium difficile infection: latest evidence and clinical guidanceTher Adv Chronic Dis201452698424587892
  • XuKLiangZCDingXNanomaterials in the prevention, diagnosis, and treatment of Mycobacterium tuberculosis infectionsAdv Healthc Mater2018711700509
  • AiXHuMWangZRecent advances of membrane-cloaked nanoplatforms for biomedical applicationsBioconjug Chem201829483885129509403
  • HuCMFangRHLukBTZhangLPolymeric nanotherapeutics: clinical development and advances in stealth functionalization strategiesNanoscale201461657524280870
  • EscajadilloTOlsonJLukBTZhangLNizetVA red blood cell membrane-camouflaged nanoparticle counteracts streptolysin O-mediated virulence phenotypes of invasive Group A StreptococcusFront Pharmacol2017847728769806
  • CharoenpholPOswaltKBishopCJTherapeutics incorporating blood constituentsActa Biomater201873648029626699
  • ZhangYZhangJChenWErythrocyte membrane-coated nanogel for combinatorial antivirulence and responsive antimicrobial delivery against Staphylococcus aureus infectionJ Control Release201726318519128087406
  • ChenYChenMZhangYBroad-spectrum neutralization of pore-forming toxins with human erythrocyte membrane-coated nanospongesAdv Healthc Mater2018713e170136629436150
  • WeiXGaoJWangFIn situ capture of bacterial toxins for antivirulence vaccinationAdv Mater201729331701644
  • FangRHLukBTHuCMZhangLEngineered nanoparticles mimicking cell membranes for toxin neutralizationAdv Drug Deliv Rev201590698025868452
  • JiangYFangRHZhangLBiomimetic nanosponges for treating antibody-mediated autoimmune diseasesBioconjug Chem201829487087729357234
  • YukSASanchez-RodriguezDATsifanskyMDYeoYRecent advances in nanomedicine for sepsis treatmentTher Deliv20189643545029722636
  • DasSAngsantikulPLeCNeutralization of cholera toxin with nanoparticle decoys for treatment of choleraPLoS Negl Trop Dis2018122e000626629470490
  • GaoWFangRHThamphiwatanaSModulating antibacterial immunity via bacterial membrane-coated nanoparticlesNano Lett20151521403140925615236
  • ShenHAckermanALCodyVEnhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticlesImmunology20061171788816423043
  • SongCNohYWLimYTPolymer nanoparticles for cross-presentation of exogenous antigens and enhanced cytotoxic T-lymphocyte immune responseInt J Nanomedicine2016113753376427540289
  • BaficaAScangaCAFengCGLeiferCCheeverASherATLR9 regulates Th1 responses and cooperates with TLR2 in mediating optimal resistance to Mycobacterium tuberculosisJ Exp Med2005202121715172416365150
  • WeissDSRaupachBTakedaKAkiraSZychlinskyAToll-like receptors are temporally involved in host defenseJ Immunol200417274463446915034062
  • LuDGarcia-ContrerasLXuDPoly (lactide-co-glycolide) microspheres in respirable sizes enhance an in vitro T cell response to recombinant Mycobacterium tuberculosis antigen 85BPharm Res200724101834184317657598
  • CaiHHuXDYuDHLiSXTianXZhuYXCombined DNA vaccine encapsulated in microspheres enhanced protection efficacy against Mycobacterium tuberculosis infection of miceVaccine200523324167417415908060
  • PriyadarsiniSMukherjeeSMishraMNanoparticles used in dentistry: a reviewJ Oral Biol Craniofac Res201881586729556466
  • KadiyalaUTurali-EmreESBahngJHKotovNAVanEppsJSUnexpected insights into antibacterial activity of zinc oxide nanoparticles against methicillin resistant Staphylococcus aureus (MRSA)Nanoscale201810104927493929480295
  • De MatteisVExposure to inorganic nanoparticles: routes of entry, immune response, biodistribution and in vitro/in vivo toxicity evaluationToxics201754E2929051461
  • BreznanDdasDDO’BrienJSDifferential cytotoxic and inflammatory potency of amorphous silicon dioxide nanoparticles of similar size in multiple cell linesNanotoxicology201711222323528142331
  • JainAKSenapatiVASinghDDubeyKMauryaRPandeyAKImpact of anatase titanium dioxide nanoparticles on mutagenic and genotoxic response in Chinese hamster lung fibroblast cells (V-79): the role of cellular uptakeFood Chem Toxicol201710512713928400324
  • VimbelaGVNgoSMFrazeCYangLStoutDAAntibacterial properties and toxicity from metallic nanomaterialsInt J Nanomedicine2017123941396528579779
  • PanáčekASmékalováMKilianováMStrong and nonspecific synergistic antibacterial efficiency of antibiotics combined with silver nanoparticles at very low concentrations showing no cytotoxic effectMolecules2015211E2626729075
  • ThorsingMBentinTGivskovMTolker-NielsenTGoltermannLThe bactericidal activity of β-lactam antibiotics is increased by metabolizable sugar speciesMicrobiology2015161101999200726243263
  • RahmanTYarnallBDoyleDAEfflux drug transporters at the forefront of antimicrobial resistanceEur Biophys J201746764765328710521
  • StokesJMMacNairCRIlyasBPentamidine sensitizes Gram-negative pathogens to antibiotics and overcomes acquired colistin resistanceNat Microbiol2017251702828263303
  • MelanderRJMelanderCThe challenge of overcoming antibiotic resistance: an adjuvant approach?ACS Infect Dis20173855956328548487
  • WuSYParkGYKimSHHulmeJAnSSADiminazene aceturate: an antibacterial agent for Shiga-toxin-producing Escherichia coli O157:H7Drug Des Devel Ther20161033633378
  • MiltonMEMinrovicBMHarrisDLRe-sensitizing multi-drug resistant bacteria to antibiotics by targeting bacterial response regulators: characterization and comparison of interactions between 2-aminoimidazoles and the response regulators BfmR from Acinetobacter baumannii and QseB from Francisella sppFront Mol Biosci201851529487854
  • JamilBHabibHAbbasiSAIhsanANasirHImranMDevelopment of cefotaxime impregnated chitosan as nano-antibiotics: de novo strategy to combat biofilm forming multi-drug resistant pathogensFront Microbiol2016733027047457
  • MengJHeGWangHReversion of antibiotic resistance by inhibiting mecA in clinical methicillin-resistant Staphylococci by antisense phosphorothioate oligonucleotideJ Antibiot201568315816425269464
  • BikardDEulerCWJiangWExploiting CRISPR-Cas nucleases to produce sequence-specific antimicrobialsNat Biotechnol201432111146115025282355
  • LuoMLLeenayRTBeiselCLCurrent and future prospects for CRISPR-based tools in bacteriaBiotechnol Bioeng2016113593094326460902
  • GiauVVLeeHShimKHBagyinszkyEAnSSAGenome-editing applications of CRISPR-Cas9 to promote in vitro studies of Alzheimer’s diseaseClin Interv Aging20181322123329445268