Abstract
Bacterial infections of the lung frequently occur as a secondary infection to many respiratory viral infections and conditions, including influenza, COVID-19, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF). Currently, clinical standard treats bacterial infections of the lung with antibiotic drugs. However, the use of broad-spectrum antibiotics can disrupt host microbiomes, lead to patient discomfort, and current clinical settings face the constantly increasing threat of drug-resistant bacteria. Biofilms further obstruct effective treatment due to their protective matrix layer, which shields bacteria from both the host immune system and antimicrobial drugs and subsequently promotes drug resistance. Alternative antimicrobial agents, including bacteriophages and antimicrobial peptides, have been utilized to treat drug-resistant bacteria. However, these antimicrobial agents have significant limitations pertaining to their ability to arrive at infection sites without compromised function and ability to persist over an extended period to fully treat infections. Enhanced delivery strategies present great promise in addressing these issues by using micro/nanoparticle carriers that shield antimicrobial agents in transit and result in sustained release, enhancing subsequent therapeutic effect and can even be modulated to be multi-functional to further improve recovery following bacterial infection.
Keywords: Biomaterials, Lung infection, Biofilms, Drug-resistant infection, Microparticles, Nanoparticles
1. Background
Bacterial infections of the lung frequently occur as a secondary infection to respiratory viral infections such as influenza [1] and COVID-19 [2,3] and conditions such as chronic obstructive pulmonary disease (COPD) [4] and cystic fibrosis (CF) [5]. Common causal bacteria include Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa), with additional species documented [6]. To treat such infections, the current standard of clinical care is to employ antimicrobial drugs (e.g., antibiotics). In the current clinical setting, however, drug resistance in bacteria has become an ever-increasing problem, which complicates the use of antimicrobial drugs. To address this issue, clinical care strategies recommend administering empirical antibiotic regimens, which entails the use of multiple antimicrobial drugs [6,7]. However, this strategy not only has the potential to further exacerbate the ability of bacteria to resist drugs but also leads to side effects resulting from non-selective killing of human gut and lung flora. In turn, these effects can lead to additional discomfort for patients and can lead to imbalances in the body including the promotion of opportunistic infections [8].
Under normal conditions, the lung contains its own microbiome with significant variations between the upper and lower respiratory tracts that are thought to arise from bacterial species travelling through the nose and the passages connected to it [[9], [10], [11]]. Studies also support that the lung microbiome is connected to the gut microbiome, and that these microbiomes are critical for maintaining homeostasis and directing immune responses [9,12]. When a bacterial infection occurs, the microbiome becomes disrupted and exhibits a decreased diversity in bacterial presence (i.e.: fewer different species of bacteria present) [9,11]. With these findings in mind, a sweeping antibiotic treatment may not only result in discomfort associated with gut flora changes, but also potential exacerbation of the respiratory system condition after treatment [9,13], prompting the need for treatment options with greater specificity, be it through limiting range of affected bacteria or more localized delivery of antibiotic agents.
1.1. Significance of biofilms and development of antibiotic resistance
Importantly, bacteria can form biofilms, presenting another obstacle to effective treatment by antimicrobial drugs. Formation of a biofilm begins after planktonic bacteria proliferate and attach to a surface. From there, these bacteria create microcolonies, consisting of bacteria aggregates covered in a matrix, which can then further proliferate and turn into a biofilm. At this point, the biofilm consists mostly of a highly protective matrix layer consisting of extracellular polymeric substances (referred to as EPS), such as polysaccharides and proteins [14,15]. This matrix serves to shield bacteria from threats, such as the immune system and antimicrobial drugs. The EPS can also serve as a diffusional barrier (in addition to physical), causing antibiotic drugs that are administered to arrive at bacteria at reduced, nonlethal concentrations over prolonged periods time which promotes drug tolerance [16]. This drug tolerance is the result of the reduced concentration antibiotic drugs causing selection towards bacteria with mutations that extend the period of time where bacteria grow in size but not in number, subsequently promoting the rise of phenotypes that allow bacteria to be in a more dormant or quiescent state (known as persister bacteria) and evade drugs that target actively proliferating cells [14,17]. As these persister bacteria proliferate, genetic transfer of genes for antibiotic drug resistance is greatly enabled, leading to the development of biofilms with highly drug resistant bacteria [14,18,19].
Although the emergence of drug-resistant bacteria is problematic, these bacteria still have weaknesses that can be exploited. Drug resistance in bacteria partially relies on the rise of randomly emerging genetic mutations, which occur at low frequency due to low error rate with replication of bacterial genomes. As a consequence of this reliance on low probability, it is unlikely for bacteria to acquire resistance to multiple antibiotic agents (including unresisted antibiotic drugs) when they are administered simultaneously [20,21]. Drug-resistant bacteria can still be affected by changes in population dynamics, which means that disrupting this balance—such as by probiotic strategies—can disrupt the ability of drug-resistant pathogenic bacteria to colonize [20,22]. In healthy conditions, the lung has a mucosal layer that protects the epithelial lining and provides a diffusion barrier. This mucosal layer is also circulated and cleaned of foreign material and pathogens by the beating of the cilia of the epithelial layer. However, under diseased conditions (e.g., COPD and CF), the mucus viscosity increases and the motion of the cilia can be inhibited, causing any bacteria in the mucosal layer to be trapped as opposed to transported for expulsion from the body. Thus, in context of the lung, it is these trapped bacteria that often proliferate and develop into biofilms [14,18]. Additionally, insertion of some medical devices (such as endotracheal tubes) leads to the device becoming coated with host proteins, which can then promote the attachment of bacteria to the device as well as the damaged tissue and introduce biofilm formation. Planktonic bacteria from the device biofilm can then spread and attach to nearby biologic surfaces, including the mucosal layer of the lung [18].
1.2. Alternative antimicrobial strategies
Because of the drug-evading and drug-resistant properties associated with biofilms, there is a great need for either alternative targets for antimicrobial agents (to eliminate quiescent bacteria in addition to active bacteria) or enhanced delivery to penetrate the biofilm matrix and promote earlier elimination of biofilm bacteria to prevent development of antibiotic resistance (Fig. 1). To deal with the drug-circumvention ability of persister bacteria, alternative antimicrobial agents have been sought out. Most notable of these alternative antimicrobial agents are bacteriophages—bacteria-targeting viruses—and antimicrobial peptides (AMPs).
Fig. 1.
Overview of biofilm environment and notable targets for improving treatment. Biofilms have several properties that make them difficult to treat. Biofilms have a protective matrix layer, which provides obstacle to effective delivery of antibiotic drugs to the bacteria within. Additionally, some of the bacteria involved (highlighted as planktonic bacteria and bacteria in biofilm in this image) can be of a quiescent or persister phenotype, which diminishes the effect of drugs targeting actively replicating bacteria and thus creates a need for antibiotic agents with alternative targets.
1.2.1. Bacteriophages
Bacteriophages are viruses that can target and kill bacteria. Since the mechanism by which bacteriophages cause bacterial death is different from antibiotic drugs, bacteriophages can be used to treat drug-resistant strains. Bacteriophages that undergo lytic cycle (as opposed to lysogenic phages) are typically used as antimicrobial agents. To proliferate, bacteriophages will attach to target host bacteria and inject their genetic material. The host bacterium then replicates the bacteriophage's genetic material and synthesizes bacteriophage components, which then self-assemble. As this process proceeds, increasing amounts of phage specific proteins are synthesized. These phage specific proteins can then cause lysis of bacteria and release more bacteriophages that can repeat this cycle [3,23,24].
Bacteriophages target bacteria with high specificity and often target only one or a few species [25,26]. This high specificity indicates that bacteriophages will not affect human cells or gut flora and thus have low off-targeting effects. Bacteriophages can also be quickly isolated and produced against newly emerging drug-resistant strains, meaning that they are antimicrobial agents that can evolve alongside their target bacterial strains. Furthermore, bacteriophages have seen use in patients both historically (in the former Soviet Union) and currently (in Poland, Russia, and Georgia) without notable side effects directly related to bacteriophages [23,27]. Because of these properties, bacteriophages represent an attractive source of treatment for drug resistant bacteria.
However, despite their promising properties, bacteriophages also have some significant drawbacks. Due to the high degree of specificity, bacteriophages need to be implemented in cocktails of different bacteriophage strains. In addition, the required bacteriophage cocktail can vary according to infection site even when the target bacteria species is the same, which can limit where in the body a particular strain of bacteriophages can be used. As a consequence of the need for multiple strains of bacteriophages in a treatment, time needed to create these cocktails and associated financial costs can increase significantly [23,28]. Furthermore, the host immune system can inactivate bacteriophages and bacteria are able to develop resistance to bacteriophages, which limits how often bacteriophages can be used to treat their target drug-resistant strains [29,30].
1.2.2. Antimicrobial peptides (AMPs)
AMPs are peptides that have potent antimicrobial activity. AMPs have different mechanisms of antimicrobial action, which enable AMPs to induce bacterial death and thus are able to treat drug-resistant bacteria [31,32]. The mechanism of antimicrobial activity varies across different kinds of AMPs, with direct action typically occurring through causing disruption of bacterial cell membrane or disruption of intracellular function. Important to the direct activity of AMPs is the presence of a positive charge, which directs AMPs selectively towards anionic bacterial membranes [31,32], and the presence of a hydrophobic component, which is needed to interact with the hydrophobic interior of bacterial membranes. After reaching a certain concentration, AMPs can then induce pore formation and enter bacteria, consequently disrupting organelle membranes [[33], [34], [35]]. Some AMPs are reported to inhibit bacterial protein and nucleic acid synthesis, block vital bacterial enzyme activity, or induce apoptosis-like death through generating reactive oxidative species [[36], [37], [38]]. AMPs also have indirect antimicrobial effect by directing the immune host response through the promotion of inflammation—improving nucleic acid recognition—and activation of immune cells [39,40].
AMPs have been reported to affect quiescent bacterial phenotypes and interfere with EPS production as well, which makes AMPs an appealing agent for biofilm prevention [[41], [42], [43]]. Additionally, bacterial resistance to AMPs does not develop quickly, which means that AMPs can remain an effective antimicrobial agent for greater number of uses than antibiotic drugs [31,32,44]. AMPs are associated with additional beneficial properties that can help with recovery from infection, such as wound-healing, which can help repair tissue damage associated with infection, and anti-inflammatory properties, which can help reduce patient pain and discomfort [31,32,45,46]. Examples of AMPs that have been investigated for treatment of respiratory infections include: amphibian AMP Esculentin (1–21) which was reported to significantly affect planktonic and biofilm-forming P. aeruginosa when administered intra-tracheal in a mouse model of pulmonary infection at 5 mg/kg animal [31,47], and Tachyplesin III which was reported to prolong survival of mice infected with co-infection of multi-drug resistant P. aeruginosa and Acinetobacter baumannii and reported to significantly reduce bacterial presence in bronchoalveolar lavage fluid when administered intravenously at 10 mg/kg intravenously [32,48].
Although AMPs are a promising approach to treat drug-resistant strains of bacteria while bestowing relevant recovery enhancing properties—such as wound-healing and anti-inflammatory properties—there are still notable limitations. First, AMPs isolated from natural sources are associated with challenges balancing antimicrobial activity and mammalian cell toxicity. To address this issue, investigators have modulated or synthetically created AMPs to enhance antimicrobial ability [31,49]. However, this workaround demonstrates that additional steps may be required for effective treatment by AMPs. Furthermore, some AMPs are reported to not be stable in biological fluids, which can impede activity on the way to the infection site [31,32]. Additionally, high concentrations, often to the point of mammalian cell cytotoxicity, are needed for situations where multiple bacteria species are involved, which is common for clinical cases of bacterial biofilms, and development of AMP therapies can entail high financial costs [32]. However, efforts have been made in the field to address these issues including the cyclization of AMP molecules to reduce protease and peptidase interaction, modulation of physical and chemical properties (including size, sequence, structure/shape, charge, and hydrophobicity) to reduce associated toxicity and hemolytic properties, and recombinant biosynthesis in bacteria or fungi (as opposed to isolation from animals) [31,32,50].
1.2.3. Limitations in delivery of current alternative antimicrobial strategies
Although these alternative antimicrobial agents show great promise in treating drug-resistant bacteria, further constraints remain when seeking to treat infections in the lung. Multiple routes of delivery have been utilized for treatment of lung infections including oral, systemic/intravenous, and parenteral routes. However, these methods of delivery generally result in inaccurate concentration of antimicrobial agents at the ultimate site of delivery [51,52]. For this reason, pulmonary delivery has been considered as an alternative for treating infection of the lung. The pulmonary route entails inhalation of treatment agents, aiming to directly deliver treatments to the epithelial layer of the lung, which reduces how much treatment agents get metabolized compared to oral and other routes of administration. This route also touts being a needle-free technique, enabling greater patient comfort, and having high surface exchange area—thanks to the alveoli and bronchioles—enabling quicker and more effective drug absorption [53,54]. This delivery target is appropriate for systemic drugs and has been reported to enhance the therapeutic effect in the context of treating lung infections [53,55]. In summary, pulmonary delivery affords efficient delivery of anti-microbial agents at the site of infection. As such, this localized delivery addresses limitations of systemic (i.e., intravascular, oral) delivery by (i) reducing the overall dose and systemic distribution as well as associated off-target effects and (ii) increasing local concentrations and persistence time of the therapeutic. However, the lung is designed to expel and/or clear any foreign material present, which presents further challenges to the delivery and persistence of treatment agents [53,56].
Because of the issues presented here, there is a need for either improving the ability of antimicrobial agents to reach bacteria or improving the ability of antimicrobial agents to persist long enough to achieve significant antimicrobial effect. In other words, there is a great need for improved delivery of antimicrobial agents overall. Biomaterials provide promising strategies to address these issues through a variety of means. In this review, we discuss applications of biomaterial-based strategies to improve delivery of treatments against drug-resistant bacteria and biofilms in the lung. In particular, micro/nanoparticle-based strategies for enhanced delivery by pulmonary route are highlighted (Fig. 2).
Fig. 2.
Overview of frequently used biomaterial strategies for treatment of lung infections. This review is focused on biomaterial strategies for enhanced delivery (Highlighted as the overlaps in the biomaterial types presented in the image).
1.3. Enhanced delivery by micro/nanoparticles
Biomaterial strategies focus on the use of carriers to enable enhanced delivery through obstacles, such as biofilms, or sustained release of antimicrobial agents at the site of infection. Enhanced delivery biomaterial strategies can also protect or shield antimicrobial agents from degradation in the in vivo environment, which can preserve the activity of antimicrobial agents during transit to infection site and thus enhance subsequent therapeutic effects [51,[57], [58], [59]]. Additionally, these strategies can be used to address other issues associated with antimicrobial agents and drugs, such as in vivo solubility [51,60] (Table 1).
Table 1.
Summary of pros and cons of currently used antimicrobial agents.
| Antimicrobial Agent | Description/Examples | Pros | Cons |
|---|---|---|---|
| Antibiotic Drugs | Commercially available antibiotic drugs. Multiple drugs are typically administered simultaneously to treat infection by drug-resistant strains of bacteria | +Clinical standard |
|
| Example Administrations: | +Readily available |
|
|
| Vancomycin, 15 mg/kg IV |
|
||
| Ciprofloxacin, 400 mg IV | |||
| Tobramycin, 5–7 mg/kg IV | |||
| Bacteriophages | Bacteria-targeting viruses | +High selectivity towards target bacteria |
|
| +Low off-target effects |
|
||
| +Can evolve alongside emerging strains of drug resistant bacteria |
|
||
| +Reported effective use in humans → potential for lower regulatory barrier |
|
||
| Antimicrobial Peptides (AMPs) | Peptides with the ability to kill bacteria | +Wide range of mechanisms for antimicrobial effect |
|
| Examples: | +Can interfere with biofilm protective matrix |
|
|
| Esculentin (1–21), 5 mg/kg intra-tracheal | +Associated with wound healing and anti-inflammatory properties → helps with recovery outcomes |
|
|
| Tachyplesin III, 10 mg/kg IV | +Bacteria do not rapidly develop resistance to AMPs |
1.3.1. Commonly used materials for micro/nanoparticle carriers
Micro/nanoparticles for enhanced delivery have been made of many different materials. Frequently reported is the use of polymeric particles, both of natural and synthetic origin. Polymeric particles are popular in the field as they allow easy encapsulation methodologies, can slow the absorption of drugs, and enable sustained release. Natural polymers frequently used are chitosan and alginate, although other materials such as dextran have been reported [61]. Natural polymers have been favored owing to greater cytocompatibility and biodegradability. Furthermore, both chitosan and alginate have been reported to result in improved aerosolization [51,[62], [63], [64], [65], [66]], enabling easier preparation for pulmonary delivery. Alginate is also attractive because of its relatively lower costs. However, these natural polymers have limitations. Safety issues in the lung have been reported with the use of chitosan and chitosan derivatives [51,67] while drug delivery with alginate involves rapid release, which is not ideal in treatment of persistent infections [51,68]. Furthermore, the ability to tune the physicochemical properties of these materials, which impacts drug release and clearance rates, is limited. As an alternative to naturally-sourced materials, synthetic polymers provide greater control can be exercised over delivery profile and release kinetics. Because of this aspect, synthetic polymers can therefore have more effective drug release over time for sustained delivery [51,69]. As a result, a variety of synthetic polymeric particles have been used to deliver antibiotic agents including poly(2-ethyl-2-oxazoline) (PEtOX), poly(lactic-co-glycolic) acid (PLGA), poly(lactic) acid (PLA), polyvinyl alcohol (PVA) and other biodegradable polymers [51,57,59,70]. Mesoporous-silica based nanoparticles have been used to deliver the antibiotic drug rifampin to the lung as well [71,72]. Despite the great benefit of significant control over particle properties, there are notable downsides. Many synthetic processes involve harsh reagents and/or solvents, which makes toxic residue a significant concern, as well as the possibility of reducing the bioactivity of the therapeutic agent [51,73]. Furthermore, high working temperatures for methods such as spray-drying can lead to thermal degradation of particular materials, which can affect the efficacy of encapsulation [51,74].
1.3.2. Micro/nanoparticle-based enhanced delivery strategies for antibiotic drugs
The current clinical standard for the treatment of lung infections is to administer an empirical regimen of antibiotic drugs. However, in addition to contributing to the development of drug-resistant strains of bacteria, many antibiotic drugs are primarily delivered by oral and systemic administration can significantly change the final concentration in the lung. Furthermore, some antibiotic drugs are associated with lack of water-solubility, making it difficult to absorb into the body. To address these issues, micro/nanoparticles have been used as delivery vehicles. Antibiotic drugs that have been encapsulated using micro/nanoparticles include ciprofloxacin, rifampin, isoniazid, and ofloxacin [53,57,71,72].
Biomaterial-based delivery strategies for antibiotic drugs enable improved solubility and enables sustained release. For example, Sabuj et al. created PEtOX nanoparticles loaded with ciprofloxacin to address its issue of poor in vivo solubility and poor delivery when administered orally and intravenously. These ciprofloxacin-loaded particles exhibited sustained release over the course of 14 days, significantly extending the persistence ability of ciprofloxacin [57]. By increasing the persistence of antibiotic drugs at effective concentrations, enhanced delivery enables improved antimicrobial activity against more persistent bacteria, such as those involved in biofilms. However, certain negative micro/nanoparticulate properties, particularly potential toxicity (be it associated with fabrication process or otherwise), need to be kept in consideration.
1.3.3. Micro/nanoparticle-based enhanced delivery strategies for alternative antimicrobial agents
Biomaterials have also been exploited for enhanced delivery of alternative antimicrobial agents such as bacteriophages and AMPs. For example, Agarwal et al. used inhalable porous PLGA microparticles to enhance bacteriophage delivery in terms of tissue distribution and persistence for the treatment of infections caused by P. aeruginosa and reported reduced bacterial presence in healthy and CF mouse in vivo models of infection. Notably, it was also reported that the antimicrobial effect of the phage-loaded microparticles did not diminish even when mice experienced previous exposure to the phage-loaded microparticles, indicating that these antimicrobial particles did not generate neutralizing immunity and suggesting the potential for multiple dosing interventions [70]. Building off these findings, Kalelkar et al. tested these PLGA microparticles for treating infections caused by drug-resistant S. aureus and co-infection of S. aureus and P. aeruginosa and showed reduced bacterial presence with in vivo microparticle delivery of bacteriophages. Additionally, mice treated with bacteriophage-loaded particles had higher concentrations of bacteriophages recovered after takedown, indicating longer sustained release of bacteriophages and ability of the phages to infect and replicate in their host bacteria (compared to free bacteriophages) [59]. These studies demonstrate that biomaterial-based delivery of bacteriophages enables sustained release and ensures that bacteriophages remain at a sufficient concentration to achieve effective antimicrobial effects. For AMPs, delivery using biomaterials maintains prolonged presence at concentrations high enough for significant antimicrobial effects while also shielding AMPs from degradation. Additionally, the shielding of AMPs means that non-target tissues and cells experience less exposure to AMPs, thus reducing the associated toxicity. For example, Falciani et al. has developed dextran-based nanoparticles that were loaded with AMPs and reported significant killing of Pseudomonas aeruginosa with prolonged in vivo presence and tolerable levels of toxicity [61].
Overall, biomaterial-based delivery strategies for alternative antimicrobial agents enable sustained release of these agents and, in the case of AMPs, reduced toxicity. However, for encapsulation of AMPs, there is a general trend of needing to compromise efficacy with toxicity. Additionally, while these strategies do improve some of the limitations of alternative antimicrobial agents, they do not address issues associated with the time-intensive and high-cost production methods for these alternative antimicrobial agents.
1.3.4. Hybrid micro/nanoparticle strategies for enhanced delivery
Notably, micro/nanoparticles can also be enabled with additional functionalities to improve other infection recovery outcomes (such as reduced inflammation) or additional control for antimicrobial agent release. As an example, Zhao et al. created polymeric nanoparticles with bacterial targeting motifs that were loaded with drugs and photosensitive agents to enable phototherapy for suppression of inflammatory response caused by bacteria. These nanoparticles, when exposed to near infrared light, disrupted biofilms in vitro and significantly reduced bacterial presence and inflammatory response in a mouse model of pneumonia [75]. In a similar manner, Gao et al. created size and charge adaptive nanoparticles made of poly(amidoamine) dendrimer and 2,3-dimethyl maleic anhydride-modified poly(ethylene glycol)-block-polylysine (PEG-b-Plys) and coupled them to an antibiotic drug. Due to the charge adaptive aspect, the nanoparticles were able to directly attach to bacteria and increase membrane permeability in addition to having controlled release, resulting in long circulation time with low toxicity towards mammalian cells [76]. Notably, Yang et al. created multi-functional gold/silver composite nanocages with AMPs and hyaluronic acid (HA). The nanometal component enables phototherapy, AMPs provide enhanced targeting and bactericidal effects, and HA suppresses immune responses and provides nontoxic coating of nanometal particles. As a result of the combination of these components, following exposure to near-infrared light, the nanocages significantly reduced bacterial presence at 3 days after treatment in a mouse model of pneumonia while demonstrating low mammalian cell toxicity low blood toxicity and decreased inflammatory responses [77].
Overall, these hybridized delivery strategies allow for the existence of alternative treatments that not only can be used to enhance antimicrobial effects, but also can be used to improve other related outcomes, such as inflammatory response, as well. Additionally, the ability to combine multiple therapeutic strategies indicates a potential for high degree of modulation, meaning that significant control can be exerted over multiple functions. However, the increased complexity of these strategies can increase the complication of fabrication as well as associated time and financial costs as well as regulatory burden.
1.4. Outlook
Biomaterials have provided powerful and versatile platforms to address current issues in the treatment of lung infections. Biomaterial-based strategies for enhanced delivery have been crucial to not only enhancing therapeutic effects of current antibiotic drugs, but also to addressing the shortcomings of alternative antimicrobial agents as well, including bacteriophages and AMPs. In particular, micro/nanoparticles can shield antimicrobial agents from degradation en route to infection sites and allow controllable and sustained release of antimicrobial agents. As a result of these properties, enhanced delivery strategies enable greater therapeutic effects against drug resistant infections and biofilms and even enables additional administration methods for some antimicrobial agents.
Whereas enhanced delivery biomaterials present promising applications towards addressing the problem of respiratory infections caused by drug-resistant bacteria, limitations still need to be addressed (Table 2). Although enhanced delivery biomaterials can improve delivery to the lung, there are still physiological constraints on the effectiveness of these biomaterials. Due to the lung's tendency to exhale/clear foreign material [53], it is important that particle-based delivery vehicles are in a particular size range to reduce clearance. In addition to mucus expulsion, host immune cells can phagocytize the micro/nanoparticles [78]. In other words, nano/microparticles need to be small enough to be inhaled and deposit deeply into the lung while also being large enough to stem degradation by host defense mechanisms and ensure that enough of the delivered antimicrobial agent is present for intended therapeutic effect [51,78]. However, a method of dealing with this constraint has been developed via the use of porous microparticles. By adding pores to the microparticles, microparticle density is reduced and thus enables larger sized microparticles to be able to deposit into the lung while also resisting degradation by host defense mechanisms [70,79].
Table 2.
Summary of micro/nanoparticle-based strategies for enhanced delivery of antimicrobial agents to the lung.
| Strategy type | Description | Pros | Cons |
|---|---|---|---|
| Enhanced delivery of antibiotic drugs | Micro/nanoparticle carriers encapsulating/loaded with antibiotic drugs | +Sustained release allows antibiotic drug to be present at effective concentration at infection site |
|
| +Delivery by MP/NP circumvents poor solubility by directly delivering drug to lung epithelial layer for direct absorption |
|
||
| Enhanced delivery of alternative antimicrobial agents | Micro/nanoparticle carriers encapsulating or loaded with bacteriophages or AMPs | +Sustained release allows greater persistence of effective concentration at infection site |
|
| +Helps reduce toxicity for AMPs |
|
||
| |||
| |||
| Hybridized strategies of enhanced delivery | Hybrid micro/nanoparticle based strategies that add extra control mechanism for antimicrobial agent release or additional functionalities not related to bacterial killing | +Multi-functional |
|
| +Enables combination of multiple treatment options to address multiple aspects related to infection (e.g.: phototherapy + drug delivery) |
|
While promising delivery vehicles for anti-microbial agents, micro/nanoparticles are but a component to a device. To administer these enhanced delivery materials, aerosolization or nebulization is relied upon, resulting in administration through devices such as inhalers. Enhanced delivery strategies have also seen several clinical trials and commercialization efforts, particularly in strategies based around drug encapsulation. Pulmosphere is a commercialized inhalable microparticle treatment that has been loaded with antibiotic drugs to treat lung infections, distributed in the form of an inhaler [54,80]. It has seen an approved clinical trial for delivery of tobramycin with clinically significant reduction of sputum bacterial presence at 112 mg of inhalable treatment, twice daily [81] and completed clinical trials to assess safety in healthy people [82] and for delivery of ciprofloxacin in COPD patients, including pediatric patients [83].
Other enhanced delivery therapeutics that have been commercialized include liposomal nanoparticles encasing amikacin for inhalation and nebulized tobramycin [84]. Arikayce is a commercialized version of liposomal nanoparticles encasing amikacin that has been evaluated in clinical trials, treating infections of P. aeruginosa in COPD patients [84]. In one clinical trial, a 560 mg dose resulted in improved lung function and clinically significant reduction in sputum P. aeruginosa [84,85]. These nanoparticles have also been tested in patients in treating Mycobacterium abscessus with 590 mg dose and reported to have no presence of M. abscessus in sputum with stabilized/improved pulmonary function [86].
Other micro/nanoparticle encapsulated drugs have been developed. Pulmaquin (Linhaliq) is a dual release formulation of nanoliposomal ciprofloxacin that includes both free ciprofloxacin and encapsulated ciprofloxacin that is administered as an aerosol. The product has seen a phase 2 clinical trial that resulted in clinically significant reduction (log 4.2 reduction) of P. aeruginosa after 28 days of treatment in non-CF patients with bronchiectasis [87]. Two parallel phase 3 clinical trials were also conducted and demonstrated reduced exacerbation frequency and P. aeruginosa sputum density with the treatment reported as safe and well tolerated [88].
In conclusion, biomaterial-based strategies for enhanced delivery of antimicrobials demonstrate great potential for not only enhancing existing therapeutic strategies but also for expanding the range of antimicrobial agents available for treatment of lung infections. However, in order to reach full potential, there are still a number of obstacles that need to be overcome including challenges related to the delivery site's physiology as well as process associated issues such as concerns about toxicity from residual organic solvents used in the manufacturing process for polymeric micro/nanoparticles. Overall, this area in the field of biomaterials is full of promise for addressing the challenges of treating the lung in an environment rife with drug resistant bacteria but still requires further development and optimization. However, some of these enhanced delivery strategies have seen commercialization and successful clinical trials, demonstrating the potent potential these strategies have in the fight against drug resistant infection of the lung.
CRediT authorship contribution statement
Eunice Chee: Writing – original draft, Conceptualization, Visualization. Andrés J. García: Writing – review & editing, Funding acquisition, Supervision.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
This work was supported by a grant from the National Institutes of Health (NIH R01 AR062920). Figures were created using Biorender.
Contributor Information
Eunice Chee, Email: echee6@gatech.edu.
Andrés J. García, Email: andres.garcia@me.gatech.edu.
Data availability
No data was used for the research described in the article.
References
- 1.Centers for Disease Control and Prevention . 2022. Causes of pneumonia.https://www.cdc.gov/pneumonia/causes.html 2022. [Google Scholar]
- 2.Zhou F.Y. u T., Du R., Fan G., Liu Y., Liu Z., et al. Clinical course and risk factors for mortality of adult inpatients with COVID19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054–1062. doi: 10.1016/S0140-6736(20)30566-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Manohar P., Loh B., Athira S., et al. Secondary bacterial infections during pulmonary viral disease: phage therapeutics as alternatives to antibiotics? Front Microbiol. 2020;11:1434. doi: 10.3389/fmicb.2020.01434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Clinic Mayo. 2020. COPD symptoms & causes.https://www.mayoclinic.org/diseases-conditions/copd/symptoms-causes/syc-20353679 2022. [Google Scholar]
- 5.Clinic Mayo. 2021. Cystic fibrosis: symptoms & causes.https://www.mayoclinic.org/diseases-conditions/cystic-fibrosis/symptoms-causes/syc-20353700#:~:text=Cystic%20fibrosis%20(CF)%20is%20an,are%20normally%20thin%20and%20slippery 2022. [Google Scholar]
- 6.Metlay J.P., Waterer G.W., Long A.C., et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official clinical practice guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Respir Crit Care Med. 2019;200(7):e45–e67. doi: 10.1164/rccm.201908-1581ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kalil A.C., Metersky M.L., Klompas M., et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):e61–e111. doi: 10.1093/cid/ciw353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yang L., Bajinka O., Jarju P.O., Tan Y., Taal A.M., Ozdemir G. The varying effects of antibiotics on gut microbiota. Amb Express. 2021;11(1):1–13. doi: 10.1186/s13568-021-01274-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Fabbrizzi A., Amedei A., Lavorini F., Renda T., Fontana G. The lung microbiome: clinical and therapeutic implications. Internal and emergency medicine. 2019;14(8):1241–1250. doi: 10.1007/s11739-019-02208-y. [DOI] [PubMed] [Google Scholar]
- 10.Charlson E.S., Bittinger K., Haas A.R., et al. Topographical continuity of bacterial populations in the healthy human respiratory tract. Am J Respir Crit Care Med. 2011;184(8):957–963. doi: 10.1164/rccm.201104-0655OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Dickson R.P., Erb-Downward J.R., Prescott H.C., et al. Analysis of culture-dependent versus culture-independent techniques for identification of bacteria in clinically obtained bronchoalveolar lavage fluid. J Clin Microbiol. 2014;52(10):3605–3613. doi: 10.1128/JCM.01028-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Dang A.T., Marsland B.J. Microbes, metabolites, and the gut–lung axis. Mucosal Immunol. 2019;12(4):843–850. doi: 10.1038/s41385-019-0160-6. [DOI] [PubMed] [Google Scholar]
- 13.Huang Y.J., Sethi S., Murphy T., Nariya S., Boushey H.A., Lynch S.V. Airway microbiome dynamics in exacerbations of chronic obstructive pulmonary disease. J Clin Microbiol. 2014;52(8):2813–2823. doi: 10.1128/JCM.00035-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Domingue J.C., Drewes J.L., Merlo C.A., Housseau F., Sears C.L. Host responses to mucosal biofilms in the lung and gut. Mucosal Immunol. 2020;13(3):413–422. doi: 10.1038/s41385-020-0270-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Flemming H.-C., Wingender J. The biofilm matrix. Nat Rev Microbiol. 2010;8(9):623–633. doi: 10.1038/nrmicro2415. [DOI] [PubMed] [Google Scholar]
- 16.Levin-Reisman I., Ronin I., Gefen O., Braniss I., Shoresh N., Balaban N.Q. Antibiotic tolerance facilitates the evolution of resistance. Science. 2017;355(6327):826–830. doi: 10.1126/science.aaj2191. [DOI] [PubMed] [Google Scholar]
- 17.Fisher R.A., Gollan B., Helaine S. Persistent bacterial infections and persister cells. Nat Rev Microbiol. 2017;15(8):453–464. doi: 10.1038/nrmicro.2017.42. [DOI] [PubMed] [Google Scholar]
- 18.Maurice N.M., Bedi B., Sadikot R.T. Pseudomonas aeruginosa biofilms: host response and clinical implications in lung infections. Am J Respir Cell Mol Biol. 2018;58(4):428–439. doi: 10.1165/rcmb.2017-0321TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chakraborty P., Bajeli S., Kaushal D., Radotra B.D., Kumar A. Biofilm formation in the lung contributes to virulence and drug tolerance of Mycobacterium tuberculosis. Nat Commun. 2021;12(1):1–17. doi: 10.1038/s41467-021-21748-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cook M.A., Wright G.D. The past, present, and future of antibiotics. Sci Transl Med. 2022;14(657) doi: 10.1126/scitranslmed.abo7793. [DOI] [PubMed] [Google Scholar]
- 21.Wright G.D. Antibiotic adjuvants: rescuing antibiotics from resistance. Trends Microbiol. 2016;24(11):862–871. doi: 10.1016/j.tim.2016.06.009. [DOI] [PubMed] [Google Scholar]
- 22.Goldenberg J.Z., Yap C., Lytvyn L., et al. Probiotics for the prevention of Clostridium difficile‐associated diarrhea in adults and children. Cochrane Database Syst Rev. 2017;(12) doi: 10.1002/14651858.CD006095.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nikolich M.P., Filippov A.A. Bacteriophage therapy: developments and directions. Antibiotics. 2020;9(3):135. doi: 10.3390/antibiotics9030135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Payne R.J., Jansen V.A. Pharmacokinetic principles of bacteriophage therapy. Clin Pharmacokinet. 2003;42(4):315–325. doi: 10.2165/00003088-200342040-00002. [DOI] [PubMed] [Google Scholar]
- 25.Abd El-Aziz A.M., Elgaml A., Ali Y.M. Bacteriophage therapy increases complement-mediated lysis of bacteria and enhances bacterial clearance after acute lung infection with multidrug-resistant Pseudomonas aeruginosa. J Infect Dis. 2019;219(9):1439–1447. doi: 10.1093/infdis/jiy678. [DOI] [PubMed] [Google Scholar]
- 26.Nobrega F.L., Costa A.R., Kluskens L.D., Azeredo J. Revisiting phage therapy: new applications for old resources. Trends Microbiol. 2015;23(4):185–191. doi: 10.1016/j.tim.2015.01.006. [DOI] [PubMed] [Google Scholar]
- 27.Międzybrodzki R., Hoyle N., Zhvaniya F., et al. Current updates from the long-standing phage research centers in Georgia, Poland, and Russia. Bacteriophage: Biology, Technology, Therapy. 2021:921–951. [Google Scholar]
- 28.Loc-Carrillo C., Abedon S.T. Pros and cons of phage therapy. Bacteriophage. 2011;1(2):111–114. doi: 10.4161/bact.1.2.14590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Oechslin F. Resistance development to bacteriophages occurring during bacteriophage therapy. Viruses. 2018;10(7):351. doi: 10.3390/v10070351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Azam A.H., Tanji Y. Bacteriophage-host arm race: an update on the mechanism of phage resistance in bacteria and revenge of the phage with the perspective for phage therapy. Appl Microbiol Biotechnol. 2019;103(5):2121–2131. doi: 10.1007/s00253-019-09629-x. [DOI] [PubMed] [Google Scholar]
- 31.Luong H.X., Thanh T.T., Tran T.H. Antimicrobial peptides–Advances in development of therapeutic applications. Life Sci. 2020;260 doi: 10.1016/j.lfs.2020.118407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Batoni G., Maisetta G., Esin S. Therapeutic potential of antimicrobial peptides in polymicrobial biofilm-associated infections. Int J Mol Sci. 2021;22(2):482. doi: 10.3390/ijms22020482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shai Y. Mode of action of membrane active antimicrobial peptides. Peptide Science: Original Research on Biomolecules. 2002;66(4):236–248. doi: 10.1002/bip.10260. [DOI] [PubMed] [Google Scholar]
- 34.Huang Y., Huang J., Chen Y. Alpha-helical cationic antimicrobial peptides: relationships of structure and function. Protein & cell. 2010;1(2):143–152. doi: 10.1007/s13238-010-0004-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jiang Z., Vasil A.I., Hale J.D., Hancock R.E., Vasil M.L., Hodges R.S. Effects of net charge and the number of positively charged residues on the biological activity of amphipathic α‐helical cationic antimicrobial peptides. Peptide Science. 2008;90(3):369–383. doi: 10.1002/bip.20911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yan J., Wang K., Dang W., et al. Two hits are better than one: membrane-active and DNA binding-related double-action mechanism of NK-18, a novel antimicrobial peptide derived from mammalian NK-lysin. Antimicrob Agents Chemother. 2013;57(1):220–228. doi: 10.1128/AAC.01619-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Park C.B., Kim H.S., Kim S.C. Mechanism of action of the antimicrobial peptide buforin II: buforin II kills microorganisms by penetrating the cell membrane and inhibiting cellular functions. Biochem Biophys Res Commun. 1998;244(1):253–257. doi: 10.1006/bbrc.1998.8159. [DOI] [PubMed] [Google Scholar]
- 38.Lee B., Hwang J., Lee D. Antibacterial action of lactoferricin B like peptide against Escherichia coli: reactive oxygen species‐induced apoptosis‐like death. J Appl Microbiol. 2020;129(2):287–295. doi: 10.1111/jam.14632. [DOI] [PubMed] [Google Scholar]
- 39.Hancock R.E., Sahl H.-G. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol. 2006;24(12):1551–1557. doi: 10.1038/nbt1267. [DOI] [PubMed] [Google Scholar]
- 40.Zhang L-j, Gallo R.L. Antimicrobial peptides. Curr Biol. 2016;26(1):R14–R19. doi: 10.1016/j.cub.2015.11.017. [DOI] [PubMed] [Google Scholar]
- 41.Grassi L., Di Luca M., Maisetta G., et al. Generation of persister cells of Pseudomonas aeruginosa and Staphylococcus aureus by chemical treatment and evaluation of their susceptibility to membrane-targeting agents. Front Microbiol. 2017;8:1917. doi: 10.3389/fmicb.2017.01917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.de Breij A., Riool M., Cordfunke R.A., et al. The antimicrobial peptide SAAP-148 combats drug-resistant bacteria and biofilms. Sci Transl Med. 2018;10(423) doi: 10.1126/scitranslmed.aan4044. [DOI] [PubMed] [Google Scholar]
- 43.Batoni G., Maisetta G., Esin S. Antimicrobial peptides and their interaction with biofilms of medically relevant bacteria. Biochim Biophys Acta Biomembr. 2016;1858(5):1044–1060. doi: 10.1016/j.bbamem.2015.10.013. [DOI] [PubMed] [Google Scholar]
- 44.Volejníková A., Melicherčík P., Nešuta O., et al. Antimicrobial peptides prevent bacterial biofilm formation on the surface of polymethylmethacrylate bone cement. J Med Microbiol. 2019;68(6):961–972. doi: 10.1099/jmm.0.001000. [DOI] [PubMed] [Google Scholar]
- 45.Mangoni M.L., McDermott A.M., Zasloff M. Antimicrobial peptides and wound healing: biological and therapeutic considerations. Exp Dermatol. 2016;25(3):167–173. doi: 10.1111/exd.12929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Grassi L., Pompilio A., Kaya E., et al. The anti-microbial peptide (Lin-SB056-1) 2-K reduces pro-inflammatory cytokine release through interaction with Pseudomonas aeruginosa lipopolysaccharide. Antibiotics. 2020;9(9):585. doi: 10.3390/antibiotics9090585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Luca V., Stringaro A., Colone M., Pini A., Mangoni M.L. Esculentin (1-21), an amphibian skin membrane-active peptide with potent activity on both planktonic and biofilm cells of the bacterial pathogen Pseudomonas aeruginosa. Cell Mol Life Sci. 2013;70:2773–2786. doi: 10.1007/s00018-013-1291-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Qi J., Gao R., Liu C., et al. Potential role of the antimicrobial peptide Tachyplesin III against multidrug-resistant P. aeruginosa and A. baumannii coinfection in an animal model. Infect Drug Resist. 2019:2865–2874. doi: 10.2147/IDR.S217020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li B., Ouyang X., Ba Z., et al. Novel β-hairpin antimicrobial peptides containing the β-turn sequence of-RRRF-having high cell selectivity and low incidence of drug resistance. J Med Chem. 2022;65(7):5625–5641. doi: 10.1021/acs.jmedchem.1c02140. [DOI] [PubMed] [Google Scholar]
- 50.Rončević T., Puizina J., Tossi A. Antimicrobial peptides as anti-infective agents in pre-post-antibiotic era? Int J Mol Sci. 2019;20(22):5713. doi: 10.3390/ijms20225713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sabuj M.Z.R., Islam N. Inhaled antibiotic-loaded polymeric nanoparticles for the management of lower respiratory tract infections. Nanoscale Adv. 2021;3(14):4005–4018. doi: 10.1039/d1na00205h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Stanojevic S., McDonald A., Waters V., et al. Effect of pulmonary exacerbations treated with oral antibiotics on clinical outcomes in cystic fibrosis. Thorax. 2017;72(4):327–332. doi: 10.1136/thoraxjnl-2016-208450. [DOI] [PubMed] [Google Scholar]
- 53.Pramanik S., Mohanto S., Manne R., et al. Nanoparticle-based drug delivery system: the magic bullet for the treatment of chronic pulmonary diseases. Mol Pharm. 2021;18(10):3671–3718. doi: 10.1021/acs.molpharmaceut.1c00491. [DOI] [PubMed] [Google Scholar]
- 54.Jain H., Bairagi A., Srivastava S., Singh S.B., Mehra N.K. Recent advances in the development of microparticles for pulmonary administration. Drug Discov Today. 2020;25(10):1865–1872. doi: 10.1016/j.drudis.2020.07.018. [DOI] [PubMed] [Google Scholar]
- 55.Newman S.P. Drug delivery to the lungs: challenges and opportunities. Ther Deliv. 2017;8(8):647–661. doi: 10.4155/tde-2017-0037. [DOI] [PubMed] [Google Scholar]
- 56.Pramanik S., Sali V. Connecting the dots in drug delivery: a tour d'horizon of chitosan-based nanocarriers system. Int J Biol Macromol. 2021;169:103–121. doi: 10.1016/j.ijbiomac.2020.12.083. [DOI] [PubMed] [Google Scholar]
- 57.Sabuj M.Z.R., Dargaville T.R., Nissen L., Islam N. Inhaled ciprofloxacin-loaded poly (2-ethyl-2-oxazoline) nanoparticles from dry powder inhaler formulation for the potential treatment of lower respiratory tract infections. PLoS One. 2021;16(12) doi: 10.1371/journal.pone.0261720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Markwalter C.E., Pagels R.F., Hejazi A.N., et al. Sustained release of peptides and proteins from polymeric nanocarriers produced by inverse Flash NanoPrecipitation. J Contr Release. 2021;334:11–20. doi: 10.1016/j.jconrel.2021.04.002. [DOI] [PubMed] [Google Scholar]
- 59.Kalelkar P.P., Moustafa D.A., Riddick M., Goldberg J.B., McCarty N.A., García A.J. Bacteriophage‐loaded poly (lactic‐co‐glycolic acid) microparticles mitigate Staphylococcus aureus infection and cocultures of Staphylococcus aureus and Pseudomonas aeruginosa. Advanced Healthcare Materials. 2022 doi: 10.1002/adhm.202102539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jeong Y.-I., Na H.-S., Seo D.-H., et al. Ciprofloxacin-encapsulated poly (DL-lactide-co-glycolide) nanoparticles and its antibacterial activity. Int J Pharm. 2008;352(1–2):317–323. doi: 10.1016/j.ijpharm.2007.11.001. [DOI] [PubMed] [Google Scholar]
- 61.Falciani C., Zevolini F., Brunetti J., et al. Antimicrobial peptide-loaded nanoparticles as inhalation therapy for Pseudomonas aeruginosa infections. Int J Nanomed. 2020;15:1117. doi: 10.2147/IJN.S218966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Garg T., Rath G., Goyal A. Inhalable chitosan nanoparticles as antitubercular drug carriers for an effective treatment of tuberculosis. Artif Cell Nanomed Biotechnol. 2016;44:997–1001. doi: 10.3109/21691401.2015.1008508. [DOI] [PubMed] [Google Scholar]
- 63.Manca M.L., Manconi M., Valenti D., et al. Liposomes coated with chitosan–xanthan gum (chitosomes) as potential carriers for pulmonary delivery of rifampicin. J Pharmaceut Sci. 2012;101(2):566–575. doi: 10.1002/jps.22775. [DOI] [PubMed] [Google Scholar]
- 64.Osman R., Kan P.L., Awad G., Mortada N., Abd-Elhameed E.-S., Alpar O. Spray dried inhalable ciprofloxacin powder with improved aerosolisation and antimicrobial activity. Int J Pharm. 2013;449(1–2):44–58. doi: 10.1016/j.ijpharm.2013.04.009. [DOI] [PubMed] [Google Scholar]
- 65.Pourshahab P.S., Gilani K., Moazeni E., Eslahi H., Fazeli M.R., Jamalifar H. Preparation and characterization of spray dried inhalable powders containing chitosan nanoparticles for pulmonary delivery of isoniazid. J Microencapsul. 2011;28(7):605–613. doi: 10.3109/02652048.2011.599437. [DOI] [PubMed] [Google Scholar]
- 66.Zhou Q.T., Leung S.S.Y., Tang P., Parumasivam T., Loh Z.H., Chan H.-K. Inhaled formulations and pulmonary drug delivery systems for respiratory infections. Adv Drug Deliv Rev. 2015;85:83–99. doi: 10.1016/j.addr.2014.10.022. [DOI] [PubMed] [Google Scholar]
- 67.Islam N., Ferro V. Recent advances in chitosan-based nanoparticulate pulmonary drug delivery. Nanoscale. 2016;8(30):14341–14358. doi: 10.1039/c6nr03256g. [DOI] [PubMed] [Google Scholar]
- 68.Abasalizadeh F., Moghaddam S.V., akbari Alizadeh E.E., Kashani E., Fazljou S.M.B., Torbati M., Akbarzadeh A. J Biol Eng. 2020;14(8) doi: 10.1186/s13036-020-0227-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Rytting E., Nguyen J., Wang X., Kissel T. Biodegradable polymeric nanocarriers for pulmonary drug delivery. Expet Opin Drug Deliv. 2008;5(6):629–639. doi: 10.1517/17425247.5.6.629. [DOI] [PubMed] [Google Scholar]
- 70.Agarwal R., Johnson C.T., Imhoff B.R., Donlan R.M., McCarty N.A., García A.J. Inhaled bacteriophage-loaded polymeric microparticles ameliorate acute lung infections. Nature biomedical engineering. 2018;2(11):841–849. doi: 10.1038/s41551-018-0263-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.García-Fernández A., Sancenón F., Martínez-Máñez R. Mesoporous silica nanoparticles for pulmonary drug delivery. Adv Drug Deliv Rev. 2021;177 doi: 10.1016/j.addr.2021.113953. [DOI] [PubMed] [Google Scholar]
- 72.Hwang A.A., Lee B.Y., Clemens D.L., Dillon B.J., Zink J.I., Horwitz M.A. pH‐responsive isoniazid‐loaded nanoparticles markedly improve tuberculosis treatment in mice. Small. 2015;11(38):5066–5078. doi: 10.1002/smll.201500937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pooja D., Tunki L., Kulhari H., Reddy B.B., Sistla R. Optimization of solid lipid nanoparticles prepared by a single emulsification-solvent evaporation method. Data Brief. 2016;6:15–19. doi: 10.1016/j.dib.2015.11.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ali M.E., Lamprecht A. Spray freeze drying for dry powder inhalation of nanoparticles. Eur J Pharm Biopharm. 2014;87(3):510–517. doi: 10.1016/j.ejpb.2014.03.009. [DOI] [PubMed] [Google Scholar]
- 75.Zhao Y., Yu C., Yu Y., et al. Bioinspired heteromultivalent ligand-decorated nanotherapeutic for enhanced photothermal and photodynamic therapy of antibiotic-resistant bacterial pneumonia. ACS Appl Mater Interfaces. 2019;11(43):39648–39661. doi: 10.1021/acsami.9b15118. [DOI] [PubMed] [Google Scholar]
- 76.Gao Y., Wang J., Chai M., et al. Size and charge adaptive clustered nanoparticles targeting the biofilm microenvironment for chronic lung infection management. ACS Nano. 2020;14(5):5686–5699. doi: 10.1021/acsnano.0c00269. [DOI] [PubMed] [Google Scholar]
- 77.Yang J., Zhao Y., Cao J., et al. Hyaluronic acid and antimicrobial peptide-modified gold/silver hybrid nanocages to combat bacterial multidrug resistance. Int J Pharm. 2020;586 doi: 10.1016/j.ijpharm.2020.119505. [DOI] [PubMed] [Google Scholar]
- 78.Champion J.A., Walker A., Mitragotri S. Role of particle size in phagocytosis of polymeric microspheres. Pharmaceut Res. 2008;25(8):1815–1821. doi: 10.1007/s11095-008-9562-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Edwards D.A., Hanes J., Caponetti G., et al. Large porous particles for pulmonary drug delivery. Science. 1997;276(5320):1868–1872. doi: 10.1126/science.276.5320.1868. [DOI] [PubMed] [Google Scholar]
- 80.Geller D.E., Weers J., Heuerding S. Development of an inhaled dry-powder formulation of tobramycin using PulmoSphere™ technology. J Aerosol Med Pulm Drug Deliv. 2011;24(4):175–182. doi: 10.1089/jamp.2010.0855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Galeva I., Konstan M.W., Higgins M., et al. Tobramycin inhalation powder manufactured by improved process in cystic fibrosis: the randomized EDIT trial. Curr Med Res Opin. 2013;29(8):947–956. doi: 10.1185/03007995.2013.805122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Stass H., Nagelschmitz J., Willmann S., Delesen H., Gupta A., Baumann S. Inhalation of a dry powder ciprofloxacin formulation in healthy subjects: a phase I study. Clin Drug Invest. 2013;33:419–427. doi: 10.1007/s40261-013-0082-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Canton R., Cobos N., De Gracia J., et al. Antimicrobial therapy for pulmonary pathogenic colonisation and infection by Pseudomonas aeruginosa in cystic fibrosis patients. Clinical microbiology and infection. 2005;11(9):690–703. doi: 10.1111/j.1469-0691.2005.01217.x. [DOI] [PubMed] [Google Scholar]
- 84.Doroudian M., MacLoughlin R., Poynton F., Prina-Mello A., Donnelly S.C. Nanotechnology based therapeutics for lung disease. Thorax. 2019;74(10):965–976. doi: 10.1136/thoraxjnl-2019-213037. [DOI] [PubMed] [Google Scholar]
- 85.Clancy J., Dupont L., Konstan M., et al. Phase II studies of nebulised Arikace in CF patients with Pseudomonas aeruginosa infection. Thorax. 2013;68(9):818–825. doi: 10.1136/thoraxjnl-2012-202230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Caimmi D., Martocq N., Trioleyre D., et al. Paper presented at: open forum infectious diseases. 2018. Positive effect of liposomal amikacin for inhalation on Mycobacterium abcessus in cystic fibrosis patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Serisier D.J., Bilton D., De Soyza A., et al. Inhaled, dual release liposomal ciprofloxacin in non-cystic fibrosis bronchiectasis (ORBIT-2): a randomised, double-blind, placebo-controlled trial. Thorax. 2013;68(9):812–817. doi: 10.1136/thoraxjnl-2013-203207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Haworth C., Wanner A., Froehlich J., et al. B14. Clinical trials across pulmonary disease. American Thoracic Society; 2017. Inhaled liposomal ciprofloxacin in patients with bronchiectasis and chronic Pseudomonas aeruginosa infection: results from two parallel phase III trials (ORBIT-3 and-4) A7604-A7604. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.


